Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 68
Filter
1.
Nat Immunol ; 23(9): 1330-1341, 2022 09.
Article in English | MEDLINE | ID: mdl-35999392

ABSTRACT

Fibroblasts, the most abundant structural cells, exert homeostatic functions but also drive disease pathogenesis. Single-cell technologies have illuminated the shared characteristics of pathogenic fibroblasts in multiple diseases including autoimmune arthritis, cancer and inflammatory colitis. However, the molecular mechanisms underlying the disease-associated fibroblast phenotypes remain largely unclear. Here, we identify ETS1 as the key transcription factor governing the pathological tissue-remodeling programs in fibroblasts. In arthritis, ETS1 drives polarization toward tissue-destructive fibroblasts by orchestrating hitherto undescribed regulatory elements of the osteoclast differentiation factor receptor activator of nuclear factor-κB ligand (RANKL) as well as matrix metalloproteinases. Fibroblast-specific ETS1 deletion resulted in ameliorated bone and cartilage damage under arthritic conditions without affecting the inflammation level. Cross-tissue fibroblast single-cell data analyses and genetic loss-of-function experiments lent support to the notion that ETS1 defines the perturbation-specific fibroblasts shared among various disease settings. These findings provide a mechanistic basis for pathogenic fibroblast polarization and have important therapeutic implications.


Subject(s)
Arthritis, Rheumatoid , Fibroblasts , Proto-Oncogene Protein c-ets-1 , Arthritis, Rheumatoid/metabolism , Arthritis, Rheumatoid/pathology , Fibroblasts/metabolism , Fibroblasts/pathology , Humans , Matrix Metalloproteinases/metabolism , Proto-Oncogene Protein c-ets-1/genetics , Proto-Oncogene Protein c-ets-1/metabolism , RANK Ligand/genetics , Transcription Factors/metabolism
2.
Nat Immunol ; 21(8): 892-901, 2020 08.
Article in English | MEDLINE | ID: mdl-32601470

ABSTRACT

Autoreactive T cells are eliminated in the thymus to prevent autoimmunity by promiscuous expression of tissue-restricted self-antigens in medullary thymic epithelial cells. This expression is dependent on the transcription factor Fezf2, as well as the transcriptional regulator Aire, but the entire picture of the transcriptional program has been obscure. Here, we found that the chromatin remodeler Chd4, also called Mi-2ß, plays a key role in the self-antigen expression in medullary thymic epithelial cells. To maximize the diversity of self-antigen expression, Fezf2 and Aire utilized completely distinct transcriptional mechanisms, both of which were under the control of Chd4. Chd4 organized the promoter regions of Fezf2-dependent genes, while contributing to the Aire-mediated induction of self-antigens via super-enhancers. Mice deficient in Chd4 specifically in thymic epithelial cells exhibited autoimmune phenotypes, including T cell infiltration. Thus, Chd4 plays a critical role in integrating Fezf2- and Aire-mediated gene induction to establish central immune tolerance.


Subject(s)
Autoantigens/immunology , Central Tolerance/physiology , Gene Expression Regulation/immunology , Mi-2 Nucleosome Remodeling and Deacetylase Complex/immunology , Animals , Autoantigens/biosynthesis , DNA Helicases/immunology , DNA Helicases/metabolism , HEK293 Cells , Humans , Mi-2 Nucleosome Remodeling and Deacetylase Complex/metabolism , Mice , Mice, Inbred C57BL , Transcription Factors/immunology , Transcription Factors/metabolism , AIRE Protein
3.
Cell ; 168(6): 1000-1014.e15, 2017 Mar 09.
Article in English | MEDLINE | ID: mdl-28283057

ABSTRACT

Super-enhancers are an emerging subclass of regulatory regions controlling cell identity and disease genes. However, their biological function and impact on miRNA networks are unclear. Here, we report that super-enhancers drive the biogenesis of master miRNAs crucial for cell identity by enhancing both transcription and Drosha/DGCR8-mediated primary miRNA (pri-miRNA) processing. Super-enhancers, together with broad H3K4me3 domains, shape a tissue-specific and evolutionarily conserved atlas of miRNA expression and function. CRISPR/Cas9 genomics revealed that super-enhancer constituents act cooperatively and facilitate Drosha/DGCR8 recruitment and pri-miRNA processing to boost cell-specific miRNA production. The BET-bromodomain inhibitor JQ1 preferentially inhibits super-enhancer-directed cotranscriptional pri-miRNA processing. Furthermore, super-enhancers are characterized by pervasive interaction with DGCR8/Drosha and DGCR8/Drosha-regulated mRNA stability control, suggesting unique RNA regulation at super-enhancers. Finally, super-enhancers mark multiple miRNAs associated with cancer hallmarks. This study presents principles underlying miRNA biology in health and disease and an unrecognized higher-order property of super-enhancers in RNA processing beyond transcription.


Subject(s)
Enhancer Elements, Genetic , MicroRNAs/metabolism , Animals , Azepines/pharmacology , Gene Expression Regulation , Histone Code , Humans , Mice , Neoplasms/genetics , Organ Specificity , RNA Processing, Post-Transcriptional/drug effects , Transcription Factors/metabolism , Transcription, Genetic , Triazoles/pharmacology
4.
Cell ; 171(5): 1138-1150.e15, 2017 Nov 16.
Article in English | MEDLINE | ID: mdl-29056342

ABSTRACT

Despite its success in several clinical trials, cancer immunotherapy remains limited by the rarity of targetable tumor-specific antigens, tumor-mediated immune suppression, and toxicity triggered by systemic delivery of potent immunomodulators. Here, we present a proof-of-concept immunomodulatory gene circuit platform that enables tumor-specific expression of immunostimulators, which could potentially overcome these limitations. Our design comprised de novo synthetic cancer-specific promoters and, to enhance specificity, an RNA-based AND gate that generates combinatorial immunomodulatory outputs only when both promoters are mutually active. These outputs included an immunogenic cell-surface protein, a cytokine, a chemokine, and a checkpoint inhibitor antibody. The circuits triggered selective T cell-mediated killing of cancer cells, but not of normal cells, in vitro. In in vivo efficacy assays, lentiviral circuit delivery mediated significant tumor reduction and prolonged mouse survival. Our design could be adapted to drive additional immunomodulators, sense other cancers, and potentially treat other diseases that require precise immunological programming.


Subject(s)
Gene Regulatory Networks , Immunotherapy/methods , Ovarian Neoplasms/therapy , Animals , Female , Humans , Immunomodulation , Mice , Ovarian Neoplasms/immunology , Promoter Regions, Genetic , Receptors, Antigen, T-Cell/metabolism , T-Lymphocytes, Cytotoxic/immunology
5.
Mol Cell ; 69(4): 648-663.e7, 2018 02 15.
Article in English | MEDLINE | ID: mdl-29398447

ABSTRACT

Regulation of RNA polymerase II (Pol II) elongation is a critical step in gene regulation. Here, we report that U1 snRNP recognition and transcription pausing at stable nucleosomes are linked through premature polyadenylation signal (PAS) termination. By generating RNA exosome conditional deletion mouse embryonic stem cells, we identified a large class of polyadenylated short transcripts in the sense direction destabilized by the RNA exosome. These PAS termination events are enriched at the first few stable nucleosomes flanking CpG islands and suppressed by U1 snRNP. Thus, promoter-proximal Pol II pausing consists of two processes: TSS-proximal and +1 stable nucleosome pausing, with PAS termination coinciding with the latter. While pausing factors NELF/DSIF only function in the former step, flavopiridol-sensitive mechanism(s) and Myc modulate both steps. We propose that premature PAS termination near the nucleosome-associated pause site represents a common transcriptional elongation checkpoint regulated by U1 snRNP recognition, nucleosome stability, and Myc activity.


Subject(s)
Gene Expression Regulation , Nucleosomes/physiology , Polyadenylation , RNA Polymerase II/metabolism , Ribonucleoprotein, U1 Small Nuclear/metabolism , Spliceosomes/metabolism , Transcription Elongation, Genetic , Animals , Cells, Cultured , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Exosome Multienzyme Ribonuclease Complex/genetics , Exosome Multienzyme Ribonuclease Complex/metabolism , HEK293 Cells , Humans , Mice , Promoter Regions, Genetic , RNA Polymerase II/genetics , Ribonucleoprotein, U1 Small Nuclear/genetics , Spliceosomes/genetics , Transcription Factors
6.
BMC Biol ; 20(1): 248, 2022 11 11.
Article in English | MEDLINE | ID: mdl-36357926

ABSTRACT

BACKGROUND: Combinatorial gene regulation by multiple microRNAs (miRNAs) is widespread and closely spaced target sites often act cooperatively to achieve stronger repression ("neighborhood" miRNA cotargeting). While miRNA cotarget sites are suggested to be more conserved and implicated in developmental control, the pathological significance of miRNA cotargeting remains elusive. RESULTS: Here, we report the pathogenic impacts of combinatorial miRNA regulation on inflammation in systemic lupus erythematosus (SLE). In the SLE mouse model, we identified the downregulation of two miRNAs, miR-128 and miR-148a, by TLR7 stimulation in plasmacytoid dendritic cells. Functional analyses using human cell lines demonstrated that miR-128 and miR-148a additively target KLF4 via extensively overlapping target sites ("seed overlap" miRNA cotargeting) and suppress the inflammatory responses. At the transcriptome level, "seed overlap" miRNA cotargeting increases susceptibility to downregulation by two miRNAs, consistent with additive but not cooperative recruitment of two miRNAs. Systematic characterization further revealed that extensive "seed overlap" is a prevalent feature among broadly conserved miRNAs. Highly conserved target sites of broadly conserved miRNAs are largely divided into two classes-those conserved among eutherian mammals and from human to Coelacanth, and the latter, including KLF4-cotargeting sites, has a stronger association with both "seed overlap" and "neighborhood" miRNA cotargeting. Furthermore, a deeply conserved miRNA target class has a higher probability of haplo-insufficient genes. CONCLUSIONS: Our study collectively suggests the complexity of distinct modes of miRNA cotargeting and the importance of their perturbations in human diseases.


Subject(s)
Lupus Erythematosus, Systemic , MicroRNAs , Humans , Animals , Mice , MicroRNAs/genetics , MicroRNAs/metabolism , Gene Expression Regulation , Lupus Erythematosus, Systemic/genetics , Lupus Erythematosus, Systemic/metabolism , Down-Regulation , Transcriptome , Mammals/genetics
7.
Cancer Sci ; 113(2): 382-391, 2022 Feb.
Article in English | MEDLINE | ID: mdl-34865286

ABSTRACT

Understanding the characteristics of cancer cells is essential for the development of improved diagnosis and therapeutics. From a gene regulation perspective, the super-enhancer concept has been introduced to systematically understand the molecular mechanisms underlying the identities of various cell types and has been extended to the analysis of cancer cells and cancer genome alterations. In addition, several characteristic features of super-enhancers have led to the recognition of the link between gene regulation and biomolecular condensates, which is often mediated by liquid-liquid phase separation. Several lines of evidence have suggested molecular and biophysical principles and their alterations in cancer cells, which are particularly associated with gene regulation and cell signaling (" transcriptional" and "signaling" condensates). These findings collectively suggest that the modification of biomolecular condensates represents an important mechanism by which cancer cells acquire various cancer hallmark traits and establish functional innovation for cancer initiation and progression. The condensate model also provides the molecular basis of the vulnerability of cancer cells to transcriptional perturbation and further suggests the possibility of therapeutic targeting of condensates. This review summarizes recent findings regarding the relationships between super-enhancers and biomolecular condensate models, multiple scenarios of condensate alterations in cancers, and the potential of the condensate model for therapeutic development.


Subject(s)
Biomolecular Condensates/pathology , Neoplasms/pathology , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Biomolecular Condensates/drug effects , Biomolecular Condensates/genetics , Enhancer Elements, Genetic , Gene Expression Regulation, Neoplastic/drug effects , Humans , Intrinsically Disordered Proteins/genetics , MicroRNAs/genetics , Neoplasms/drug therapy , Neoplasms/genetics , Signal Transduction/genetics , Transcription, Genetic/drug effects
8.
Cancer Sci ; 113(11): 3932-3946, 2022 Nov.
Article in English | MEDLINE | ID: mdl-35789143

ABSTRACT

Small cell lung cancer (SCLC) is a highly aggressive neuroendocrine tumor with dismal prognosis. Recently, molecular subtypes of SCLC have been defined by the expression status of ASCL1, NEUROD1, YAP1, and POU2F3 transcription regulators. ASCL1 is essential for neuroendocrine differentiation and is expressed in the majority of SCLC. Although previous studies investigated ASCL1 target genes in SCLC cells, ASCL1-mediated regulation of miRNAs and its relationship to molecular subtypes remain poorly explored. Here, we performed genome-wide profiling of chromatin modifications (H3K27me3, H3K4me3, and H3K27ac) by CUT&Tag assay and ASCL1 knockdown followed by RNA sequencing and miRNA array analyses in SCLC cells. ASCL1 could preferentially regulate genes associated with super-enhancers (SEs) defined by enrichment of H3K27ac marking. Moreover, ASCL1 positively regulated several SE-associated miRNAs, such as miR-7, miR-375, miR-200b-3p, and miR-429, leading to repression of their targets, whereas ASCL1 suppressed miR-455-3p, an abundant miRNA in other molecular subtypes. We further elucidated unique patterns of SE-associated miRNAs in different SCLC molecular subtypes, highlighting subtype-specific miRNA networks with functional relevance. Notably, we found apparent de-repression of common target genes of different miRNAs following ASCL1 knockdown, suggesting combinatorial action of multiple miRNAs underlying molecular heterogeneity of SCLC (e.g., co-targeting of YAP1 by miR-9 and miR-375). Our comprehensive analyses provide novel insights into SCLC pathogenesis and a clue to understanding subtype-dependent phenotypic differences.


Subject(s)
Lung Neoplasms , MicroRNAs , Small Cell Lung Carcinoma , Humans , Small Cell Lung Carcinoma/metabolism , Lung Neoplasms/pathology , Gene Expression Regulation, Neoplastic , Cell Line, Tumor , MicroRNAs/genetics , MicroRNAs/metabolism , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism
9.
RNA ; 25(10): 1291-1297, 2019 10.
Article in English | MEDLINE | ID: mdl-31289130

ABSTRACT

Argonaute (Ago) proteins interact with various binding partners and play a pivotal role in microRNA (miRNA)-mediated silencing pathways. By utilizing immunoprecipitation followed by mass spectrometry to determine cytoplasmic Ago2 protein complexes in mouse embryonic stem cells (mESCs), we identified a putative RNA-binding protein FAM120A (also known as OSSA/C9ORF10) as an Ago2 interacting protein. Individual nucleotide resolution cross-linking and immunoprecipitation (iCLIP) analysis revealed that FAM120A binds to homopolymeric tracts in 3'-UTRs of about 2000 mRNAs, particularly poly(G) sequences. Comparison of FAM120A iCLIP and Ago2 iCLIP reveals that greater than one-third of mRNAs bound by Ago2 in mESCs are co-bound by FAM120A. Furthermore, such FAM120A-bound Ago2 target genes are not subject to Ago2-mediated target degradation. Reporter assays suggest that the 3'-UTRs of several FAM120A-bound miRNA target genes are less sensitive to Ago2-mediated target repression than those of FAM120A-unbound miRNA targets and FAM120A modulates them via its G-rich target sites. These findings suggest that Ago2 may exist in multiple protein complexes with varying degrees of functionality.


Subject(s)
Argonaute Proteins/metabolism , MicroRNAs/metabolism , Nerve Tissue Proteins/metabolism , RNA-Binding Proteins/metabolism , 3' Untranslated Regions , Animals , Cells, Cultured , Embryonic Stem Cells/metabolism , Immunoprecipitation , Mice
10.
Proc Natl Acad Sci U S A ; 115(47): E11053-E11060, 2018 11 20.
Article in English | MEDLINE | ID: mdl-30404915

ABSTRACT

A hallmark of biological systems is that particular functions and outcomes are realized in specific contexts, such as when particular signals are received. One mechanism for mediating specificity is described by Fisher's "lock and key" metaphor, exemplified by enzymes that bind selectively to a particular substrate via specific finely tuned interactions. Another mechanism, more prevalent in multicellular organisms, relies on multivalent weak cooperative interactions. Its importance has recently been illustrated by the recognition that liquid-liquid phase transitions underlie the formation of membraneless condensates that perform specific cellular functions. Based on computer simulations of an evolutionary model, we report that the latter mechanism likely became evolutionarily prominent when a large number of tasks had to be performed specifically for organisms to function properly. We find that the emergence of weak cooperative interactions for mediating specificity results in organisms that can evolve to accomplish new tasks with fewer, and likely less lethal, mutations. We argue that this makes the system more capable of undergoing evolutionary changes robustly, and thus this mechanism has been repeatedly positively selected in increasingly complex organisms. Specificity mediated by weak cooperative interactions results in some useful cross-reactivity for related tasks, but at the same time increases susceptibility to misregulation that might lead to pathologies.


Subject(s)
Biological Evolution , Cell Physiological Phenomena , Computer Simulation , Models, Biological , Animals , Humans , Protein Domains/physiology , Proteins/metabolism
11.
Int J Mol Sci ; 22(24)2021 Dec 13.
Article in English | MEDLINE | ID: mdl-34948199

ABSTRACT

The genome is pervasively transcribed across various species, yielding numerous non-coding RNAs. As a counterbalance for pervasive transcription, various organisms have a nuclear RNA exosome complex, whose structure is well conserved between yeast and mammalian cells. The RNA exosome not only regulates the processing of stable RNA species, such as rRNAs, tRNAs, small nucleolar RNAs, and small nuclear RNAs, but also plays a central role in RNA surveillance by degrading many unstable RNAs and misprocessed pre-mRNAs. In addition, associated cofactors of RNA exosome direct the exosome to distinct classes of RNA substrates, suggesting divergent and/or multi-layer control of RNA quality in the cell. While the RNA exosome is essential for cell viability and influences various cellular processes, mutations and alterations in the RNA exosome components are linked to the collection of rare diseases and various diseases including cancer, respectively. The present review summarizes the relationships between pervasive transcription and RNA exosome, including evolutionary crosstalk, mechanisms of RNA exosome-mediated RNA surveillance, and physiopathological effects of perturbation of RNA exosome.


Subject(s)
Exosome Multienzyme Ribonuclease Complex/physiology , RNA Stability/physiology , Transcription, Genetic/genetics , Animals , Exosome Multienzyme Ribonuclease Complex/genetics , Exosome Multienzyme Ribonuclease Complex/metabolism , Genome/genetics , Humans , RNA/genetics , RNA/metabolism , RNA Stability/genetics , RNA, Nuclear/genetics , RNA, Nuclear/metabolism
12.
Am J Respir Cell Mol Biol ; 63(6): 831-842, 2020 12.
Article in English | MEDLINE | ID: mdl-32946266

ABSTRACT

Fibroblasts provide a structural framework for multiple organs and are essential for wound repair and fibrotic processes. Here, we demonstrate functional roles of FOXL1 (forkhead box L1), a transcription factor that characterizes the pulmonary origin of lung fibroblasts. We detected high FOXL1 transcripts associated with DNA hypomethylation and super-enhancer formation in lung fibroblasts, which is in contrast with fibroblasts derived from other organs. RNA in situ hybridization and immunohistochemistry in normal lung tissue indicated that FOXL1 mRNA and protein are expressed in submucosal interstitial cells together with airway epithelial cells. Transcriptome analysis revealed that FOXL1 could control a broad array of genes that potentiate fibroblast function, including TAZ (transcriptional coactivator with PDZ-binding motif)/YAP (Yes-associated protein) signature genes and PDGFRα (platelet-derived growth factor receptor-α). FOXL1 silencing in lung fibroblasts attenuated cell growth and collagen gel contraction capacity, underscoring the functional importance of FOXL1 in fibroproliferative reactions. Of clinical importance, increased FOXL1 mRNA expression was found in fibroblasts of idiopathic pulmonary fibrosis lung tissue. Our observations suggest that FOXL1 regulates multiple functional aspects of lung fibroblasts as a key transcription factor and is involved in idiopathic pulmonary fibrosis pathogenesis.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Fibroblasts/metabolism , Fibrosis/metabolism , Forkhead Transcription Factors/metabolism , Cell Proliferation/physiology , Forkhead Transcription Factors/genetics , Gene Expression Regulation/physiology , Humans , Idiopathic Pulmonary Fibrosis/pathology , Lung/metabolism , Lung/pathology
13.
Mol Cell ; 44(3): 424-36, 2011 Nov 04.
Article in English | MEDLINE | ID: mdl-22055188

ABSTRACT

MicroRNAs (miRNAs) are versatile regulators of gene expression and undergo complex maturation processes. However, the mechanism(s) stabilizing or reducing these small RNAs remains poorly understood. Here we identify mammalian immune regulator MCPIP1 (Zc3h12a) ribonuclease as a broad suppressor of miRNA activity and biogenesis, which counteracts Dicer, a central ribonuclease in miRNA processing. MCPIP1 suppresses miRNA biosynthesis via cleavage of the terminal loops of precursor miRNAs (pre-miRNAs). MCPIP1 also carries a vertebrate-specific oligomerization domain important for pre-miRNA recognition, indicating its recent evolution. Furthermore, we observed potential antagonism between MCPIP1 and Dicer function in human cancer and found a regulatory role of MCPIP1 in the signaling axis comprising miR-155 and its target c-Maf. These results collectively suggest that the balance between processing and destroying ribonucleases modulates miRNA biogenesis and potentially affects pathological miRNA dysregulation. The presence of this abortive processing machinery and diversity of MCPIP1-related genes may imply a dynamic evolutional transition of the RNA silencing system.


Subject(s)
DEAD-box RNA Helicases/metabolism , Gene Expression Regulation, Neoplastic , MicroRNAs/biosynthesis , Neoplasms/enzymology , Neoplasms/genetics , RNA Precursors/metabolism , RNA Stability , Ribonuclease III/metabolism , Transcription Factors/metabolism , Base Sequence , DEAD-box RNA Helicases/genetics , HEK293 Cells , Hep G2 Cells , Humans , Jurkat Cells , MicroRNAs/chemistry , MicroRNAs/metabolism , Molecular Sequence Data , Nucleic Acid Conformation , Protein Multimerization , Proto-Oncogene Proteins c-maf/metabolism , RNA Interference , Ribonuclease III/genetics , Ribonucleases , Time Factors , Transcription Factors/chemistry , Transcription Factors/genetics , Transfection
14.
Int J Mol Sci ; 21(1)2019 Dec 24.
Article in English | MEDLINE | ID: mdl-31878193

ABSTRACT

MicroRNAs (miRNAs) are approximately 22-nucleotide-long, small non-coding RNAs that post-transcriptionally regulate gene expression. The biogenesis of miRNAs involves multiple steps, including the transcription of primary miRNAs (pri-miRNAs), nuclear Drosha-mediated processing, cytoplasmic Dicer-mediated processing, and loading onto Argonaute (Ago) proteins. Further, miRNAs control diverse biological and pathological processes via the silencing of target mRNAs. This review summarizes recent findings regarding the quantitative aspects of miRNA homeostasis, including Drosha-mediated pri-miRNA processing, Ago-mediated asymmetric miRNA strand selection, and modifications of miRNA pathway components, as well as the roles of RNA modifications (epitranscriptomics), epigenetics, transcription factor circuits, and super-enhancers in miRNA regulation. These recent advances have facilitated a system-level understanding of miRNA networks, as well as the improvement of RNAi performance for both gene-specific targeting and genome-wide screening. The comprehensive understanding and modeling of miRNA biogenesis and function have been applied to the design of synthetic gene circuits. In addition, the relationships between miRNA genes and super-enhancers provide the molecular basis for the highly biased cell type-specific expression patterns of miRNAs and the evolution of miRNA-target connections, while highlighting the importance of alterations of super-enhancer-associated miRNAs in a variety of human diseases.


Subject(s)
MicroRNAs/metabolism , Animals , Argonaute Proteins/metabolism , Humans , MicroRNAs/genetics , RNA, Messenger/metabolism , Synthetic Biology
15.
Int J Mol Sci ; 20(20)2019 Oct 10.
Article in English | MEDLINE | ID: mdl-31658594

ABSTRACT

Aging is broadly defined as the functional decline that occurs in all body systems. The accumulation of senescent cells is considered a hallmark of aging and thought to contribute to the aging pathologies. Transforming growth factor-ß (TGF-ß) is a pleiotropic cytokine that regulates a myriad of cellular processes and has important roles in embryonic development, physiological tissue homeostasis, and various pathological conditions. TGF-ß exerts potent growth inhibitory activities in various cell types, and multiple growth regulatory mechanisms have reportedly been linked to the phenotypes of cellular senescence and stem cell aging in previous studies. In addition, accumulated evidence has indicated a multifaceted association between TGF-ß signaling and aging-associated disorders, including Alzheimer's disease, muscle atrophy, and obesity. The findings regarding these diseases suggest that the impairment of TGF-ß signaling in certain cell types and the upregulation of TGF-ß ligands contribute to cell degeneration, tissue fibrosis, inflammation, decreased regeneration capacity, and metabolic malfunction. While the biological roles of TGF-ß depend highly on cell types and cellular contexts, aging-associated changes are an important additional context which warrants further investigation to better understand the involvement in various diseases and develop therapeutic options. The present review summarizes the relationships between TGF-ß signaling and cellular senescence, stem cell aging, and aging-related diseases.


Subject(s)
Aging/physiology , Cellular Senescence/physiology , Signal Transduction/physiology , Transforming Growth Factor beta/metabolism , Alzheimer Disease/metabolism , Cell Proliferation , Fibrosis , Hematopoietic Stem Cells , Homeostasis , Inflammation , Ligands , Mesenchymal Stem Cells , Muscular Atrophy/metabolism , Obesity/metabolism , Stem Cells
16.
Am J Physiol Lung Cell Mol Physiol ; 314(1): L177-L191, 2018 01 01.
Article in English | MEDLINE | ID: mdl-28971975

ABSTRACT

Lung fibroblasts participate in the pathogenesis of respiratory diseases, including lung cancer and pulmonary fibrosis. Although fibroblasts are ubiquitous constituents of various organs, their cellular diversity among different organs has been poorly characterized. Here, we aimed to investigate the distinct gene signature of lung fibroblasts that represents its pulmonary origin and the underlying gene regulatory networks. Promoter-level differential expression analysis by cap analysis of gene expression (CAGE) sequencing revealed distinct gene expression patterns of fibroblasts derived from different anatomical sites and identified 88 coding genes with higher expression in lung fibroblasts relative to other fibroblasts. Multiple key transcription factors important for lung mesenchyme development, including the T-box transcription factors TBX2, TBX4, and TBX5 were enriched in this lung-specific signature and were associated with super-enhancers. TBX4 showed highly specific expression in lung fibroblasts and was required for cell proliferation and collagen gel contraction capacity. Transcriptome analysis revealed that TBX4 could broadly regulate fibroblast-related pathways and partly contribute to super-enhancer-mediated transcriptional programs. Of pathological importance, lung fibroblast-specific genes were globally downregulated in lung cancer-associated fibroblasts (CAFs). Notably, TBX2, TBX4, and TBX5 were downregulated and hypermethylated in lung CAFs, suggesting an association between epigenetic silencing of these factors and phenotypic alteration of lung fibroblasts in cancer. Our study highlights the importance of T-box transcription factors, especially TBX4, and super-enhancers in the roles of lung fibroblasts in pulmonary physiology and pathogenesis.


Subject(s)
Biomarkers/metabolism , Fibroblasts/metabolism , Gene Expression Regulation, Developmental , Lung/metabolism , T-Box Domain Proteins/metabolism , Cells, Cultured , Fibroblasts/cytology , Gene Expression Profiling , Humans , Lung/cytology , Regulatory Sequences, Nucleic Acid , T-Box Domain Proteins/genetics , Transcription Factors/genetics , Transcription Factors/metabolism , Transcription, Genetic
17.
Int J Mol Sci ; 19(7)2018 06 28.
Article in English | MEDLINE | ID: mdl-29958433

ABSTRACT

Transcriptional and post-transcriptional regulation shapes the transcriptome and proteome changes induced by various cellular signaling cascades. MicroRNAs (miRNAs) are small regulatory RNAs that are approximately 22 nucleotides long, which direct the post-transcriptional regulation of diverse target genes and control cell states. Transforming growth factor (TGF)-ß family is a multifunctional cytokine family, which plays many regulatory roles in the development and pathogenesis of diverse diseases, including fibrotic disease, cardiovascular disease and cancer. Previous studies have shown that the TGF-ß pathway includes the miRNA pathway as an important component of its downstream signaling cascades. Multiple studies of epithelial⁻mesenchymal transition (EMT)-related miRNAs have highlighted that miRNAs constitute the intrinsic bistable molecular switches of cell states by forming double negative feedback loops with EMT-inducing transcription factors. This may be important for understanding the reversibility of EMT at the single-cell level, the presence of distinct EMT transition states and the intra- and inter-tumor heterogeneity of cancer cell phenotypes. In the present review, I summarize the connection between TGF-ß signaling and the miRNA pathway, placing particular emphasis on the regulation of miRNA expression by TGF-ß signaling, the modulation of TGF-ß signaling by miRNAs, the miRNA-mediated modulation of EMT and endothelial⁻mesenchymal transition as well as the crosstalk between miRNA and TGF-ß pathways in the tumor microenvironment.


Subject(s)
MicroRNAs/genetics , Neoplasms/genetics , Transcriptome/genetics , Transforming Growth Factor beta/genetics , Epithelial-Mesenchymal Transition/genetics , Gene Expression Regulation, Neoplastic , Humans , Neoplasms/pathology , Signal Transduction/genetics , Tumor Microenvironment/genetics
18.
Genes Cells ; 21(1): 99-116, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26663584

ABSTRACT

Transforming growth factor-ß (TGF-ß) plays central roles in endothelial-mesenchymal transition (EndMT) involved in development and pathogenesis. Although EndMT and epithelial-mesenchymal transition are similar processes, roles of microRNAs in EndMT are largely unknown. Here, we report that constitutively active microRNA-31 (miR-31) is a positive regulator of TGF-ß-induced EndMT. Although the expression is not induced by TGF-ß, miR-31 is required for induction of mesenchymal genes including α-SMA, actin reorganization and MRTF-A activation during EndMT. We identified VAV3, a regulator of actin remodeling and MRTF-A activity, as a miR-31 target. Global transcriptome analysis further showed that miR-31 positively regulates EndMT-associated unique secretory phenotype (EndMT-SP) characterized by induction of multiple inflammatory chemokines and cytokines including CCL17, CX3CL1, CXCL16, IL-6 and Angptl2. As a mechanism for this phenomenon, TGF-ß and miR-31 suppress Stk40, a negative regulator of NF-κB pathway. Interestingly, TGF-ß induces alternative polyadenylation (APA)-coupled miR-31-dependent Stk40 suppression without concomitant miR-31 induction, and APA-mediated exclusion of internal poly(A) sequence in Stk40 3'UTR enhances target efficiency of Stk40. Finally, miR-31 functions as a molecular hub to integrate TGF-ß and TNF-α signaling to enhance EndMT. These data confirm that constitutively active microRNAs, as well as inducible microRNAs, serve as phenotypic modifiers interconnected with transcriptome dynamics during EndMT.


Subject(s)
Endothelium/drug effects , Epithelial-Mesenchymal Transition/drug effects , Mesoderm/drug effects , Mesoderm/metabolism , MicroRNAs/metabolism , Secretory Pathway/drug effects , Transforming Growth Factor beta/pharmacology , 3' Untranslated Regions/genetics , Actins/metabolism , Animals , Base Sequence , Cell Line , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Endothelium/cytology , Endothelium/metabolism , Epithelial-Mesenchymal Transition/genetics , Gene Expression Regulation/drug effects , Gene Regulatory Networks/drug effects , Mesoderm/cytology , Mice , MicroRNAs/genetics , Molecular Sequence Data , Phenotype , Polyadenylation/drug effects , Proto-Oncogene Proteins c-vav/metabolism , Trans-Activators/metabolism , Transcriptome/drug effects , Transcriptome/genetics , Tumor Necrosis Factor-alpha/pharmacology
19.
Cancer Sci ; 107(12): 1755-1766, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27627196

ABSTRACT

Small cell lung cancer (SCLC) is a highly aggressive and metastatic malignancy that shows rapid development of chemoresistance and a high rate of recurrence. Recent genome and transcriptome studies have provided the whole landscape of genomic alterations and gene expression changes in SCLC. In light of the inter-individual heterogeneity of SCLC, subtyping of SCLC might be helpful for prediction of therapeutic response and prognosis. Based on the transcriptome data of SCLC cell lines, we undertook transcriptional network-defined SCLC classification and identified a unique SCLC subgroup characterized by relatively high expression of Hippo pathway regulators Yes-associated protein (YAP) and transcriptional coactivator with PDZ-binding motif (TAZ) (YAP/TAZ subgroup). The YAP/TAZ subgroup displayed adherent cell morphology, lower expression of achaete-scute complex homolog 1 (ASCL1) and neuroendocrine markers, and higher expression of laminin and integrin. YAP knockdown caused cell morphological alteration reminiscent of floating growth pattern in many SCLC cell lines, and microarray analyses revealed a subset of genes regulated by YAP, including Ajuba LIM protein (AJUBA). AJUBA also contributed to cell morphology regulation. Of clinical importance, SCLC cell lines of the YAP/TAZ subgroup showed unique patterns of drug sensitivity. Our findings shed light on a subtype of SCLC with YAP and TAZ expression, and delineate molecular networks underlying the heterogeneity of SCLC.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Phenotype , Phosphoproteins/metabolism , Small Cell Lung Carcinoma/metabolism , Small Cell Lung Carcinoma/pathology , Adaptor Proteins, Signal Transducing/genetics , Antineoplastic Agents/pharmacology , Cell Adhesion/genetics , Cell Line, Tumor , Cluster Analysis , Drug Resistance, Neoplasm/genetics , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Humans , Intracellular Signaling Peptides and Proteins/genetics , LIM Domain Proteins/genetics , LIM Domain Proteins/metabolism , Lung Neoplasms/genetics , Phosphoproteins/genetics , Small Cell Lung Carcinoma/genetics , Topotecan/pharmacology , Trans-Activators , Transcription Factors , Transcriptional Coactivator with PDZ-Binding Motif Proteins , Transcriptome , YAP-Signaling Proteins
20.
Am J Pathol ; 185(11): 2923-38, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26456580

ABSTRACT

Pulmonary fibrosis (PF) is an intractable disorder with a poor prognosis. Lung macrophages have been reported to regulate both progression and remission of bleomycin-induced diffuse PF. However, it remains unclear how macrophages contribute to silica-induced progressive nodular PF and the associated tissue cell responses in vivo. We found that lack of monocyte-derived macrophages results in the formation of diffuse PF after silica instillation. We found that the proportion and the number of monocyte-derived macrophages were persistently higher in silica-induced progressive PF compared with bleomycin-induced PF. Surprisingly, in Ccr2(-/-) mice, in which monocyte-derived macrophage infiltration is impaired, silica administration induced diffuse PF with loose nodule formation and greater activation of tissue cells. In the diffuse lesions, the distribution of epithelial cells, distribution of myofibroblasts, and architecture of the basement membrane were disrupted. Consistent with the development of diffuse lesions, genes that were differentially expressed in CD45(-) tissue cells from the lung of wild-type and Ccr2(-/-) mice were highly enriched in human diffuse, progressive PF. In gene ontology network analyses, many of these genes were associated with tissue remodeling and included genes not previously associated with PF, such as Mmp14, Thbs2, and Fgfr4. Overall, these results indicate that monocyte-derived macrophages prevent transition from nodular to diffuse silica-induced PF, potentially by regulating tissue cell responses.


Subject(s)
Macrophages, Alveolar/pathology , Pulmonary Fibrosis/pathology , Receptors, CCR2/metabolism , Animals , Bleomycin/adverse effects , Disease Models, Animal , Epithelial Cells/pathology , Gene Expression Profiling , Humans , Hydroxyproline/analysis , Lung/drug effects , Lung/pathology , Macrophages, Alveolar/drug effects , Mice , Mice, Inbred C57BL , Monocytes , Oligonucleotide Array Sequence Analysis , Pulmonary Fibrosis/chemically induced , Receptors, CCR2/genetics , Silicon Dioxide/adverse effects
SELECTION OF CITATIONS
SEARCH DETAIL