Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 103
Filtrar
1.
Nucleus ; 15(1): 2350182, 2024 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-38738760

RESUMO

Long noncoding RNAs (LncRNAs) are key regulators of gene expression and can mediate their effects in both the nucleus and cytoplasm. Some of the best-characterized lncRNAs are localized within the nucleus, where they modulate the nuclear architecture and influence gene expression. In this review, we discuss the role of lncRNAs in nuclear architecture in the context of their gene regulatory functions in innate immunity. Here, we discuss various approaches to functionally characterize nuclear-localized lncRNAs and the challenges faced in the field.


Assuntos
Núcleo Celular , RNA Longo não Codificante , RNA Longo não Codificante/genética , RNA Longo não Codificante/metabolismo , Humanos , Núcleo Celular/metabolismo , Núcleo Celular/genética , Animais , Imunidade Inata , Regulação da Expressão Gênica
2.
Proc Natl Acad Sci U S A ; 121(22): e2322524121, 2024 May 28.
Artigo em Inglês | MEDLINE | ID: mdl-38781216

RESUMO

Long noncoding RNAs (lncRNAs) account for the largest portion of RNA from the transcriptome, yet most of their functions remain unknown. Here, we performed two independent high-throughput CRISPRi screens to understand the role of lncRNAs in monocyte function and differentiation. The first was a reporter-based screen to identify lncRNAs that regulate TLR4-NFkB signaling in human monocytes and the second screen identified lncRNAs involved in monocyte to macrophage differentiation. We successfully identified numerous noncoding and protein-coding genes that can positively or negatively regulate inflammation and differentiation. To understand the functional roles of lncRNAs in both processes, we chose to further study the lncRNA LOUP [lncRNA originating from upstream regulatory element of SPI1 (also known as PU.1)], as it emerged as a top hit in both screens. Not only does LOUP regulate its neighboring gene, the myeloid fate-determining factor SPI1, thereby affecting monocyte to macrophage differentiation, but knockdown of LOUP leads to a broad upregulation of NFkB-targeted genes at baseline and upon TLR4-NFkB activation. LOUP also harbors three small open reading frames capable of being translated and are responsible for LOUP's ability to negatively regulate TLR4/NFkB signaling. This work emphasizes the value of high-throughput screening to rapidly identify functional lncRNAs in the innate immune system.


Assuntos
Diferenciação Celular , Inflamação , Macrófagos , Monócitos , RNA Longo não Codificante , Transdução de Sinais , RNA Longo não Codificante/genética , RNA Longo não Codificante/metabolismo , Humanos , Macrófagos/metabolismo , Macrófagos/citologia , Diferenciação Celular/genética , Monócitos/metabolismo , Monócitos/citologia , Inflamação/genética , Inflamação/metabolismo , Receptor 4 Toll-Like/metabolismo , Receptor 4 Toll-Like/genética , NF-kappa B/metabolismo , Transativadores/metabolismo , Transativadores/genética , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas/genética , Sistemas CRISPR-Cas , Regulação da Expressão Gênica
3.
bioRxiv ; 2024 Mar 29.
Artigo em Inglês | MEDLINE | ID: mdl-38585942

RESUMO

Long noncoding RNAs (lncRNAs) are a newer class of noncoding transcripts identified as key regulators of biological processes. Here we aimed to identify novel lncRNA targets that play critical roles in major human respiratory viral infections by systematically mining large-scale transcriptomic datasets. Using bulk RNA-sequencing (RNA-seq) analysis, we identified a previously uncharacterized lncRNA, named virus inducible lncRNA modulator of interferon response (VILMIR), that was consistently upregulated after in vitro influenza infection across multiple human epithelial cell lines and influenza A virus subtypes. VILMIR was also upregulated after SARS-CoV-2 and RSV infections in vitro. We experimentally confirmed the response of VILMIR to influenza infection and interferon-beta (IFN-ß) treatment in the A549 human epithelial cell line and found the expression of VILMIR was robustly induced by IFN-ß treatment in a dose and time-specific manner. Single cell RNA-seq analysis of bronchoalveolar lavage fluid (BALF) samples from COVID-19 patients uncovered that VILMIR was upregulated across various cell types including at least five immune cells. The upregulation of VILMIR in immune cells was further confirmed in the human T cell and monocyte cell lines, SUP-T1 and THP-1, after IFN-ß treatment. Finally, we found that knockdown of VILMIR expression reduced the magnitude of host transcriptional responses to IFN-ß treatment in A549 cells. Together, our results show that VILMIR is a novel interferon-stimulated gene (ISG) that regulates the host interferon response and may be a potential therapeutic target for human respiratory viral infections upon further mechanistic investigation.

4.
Cell ; 187(9): 2030-2051, 2024 Apr 25.
Artigo em Inglês | MEDLINE | ID: mdl-38670064

RESUMO

Over the past 50 years in the field of immunology, something of a Copernican revolution has happened. For a long time, immunologists were mainly concerned with what is termed adaptive immunity, which involves the exquisitely specific activities of lymphocytes. But the other arm of immunity, so-called "innate immunity," had been neglected. To celebrate Cell's 50th anniversary, we have put together a review of the processes and components of innate immunity and trace the seminal contributions leading to the modern state of this field. Innate immunity has joined adaptive immunity in the center of interest for all those who study the body's defenses, as well as homeostasis and pathology. We are now entering the era where therapeutic targeting of innate immune receptors and downstream signals hold substantial promise for infectious and inflammatory diseases and cancer.


Assuntos
Imunidade Inata , Humanos , Animais , História do Século XX , História do Século XXI , Imunidade Adaptativa , Alergia e Imunologia/história
5.
Proc Natl Acad Sci U S A ; 121(9): e2312587121, 2024 Feb 27.
Artigo em Inglês | MEDLINE | ID: mdl-38381785

RESUMO

To ensure a robust immune response to pathogens without risking immunopathology, the kinetics and amplitude of inflammatory gene expression in macrophages need to be exquisitely well controlled. There is a growing appreciation for stress-responsive membraneless organelles (MLOs) regulating various steps of eukaryotic gene expression in response to extrinsic cues. Here, we implicate the nuclear paraspeckle, a highly ordered biomolecular condensate that nucleates on the Neat1 lncRNA, in tuning innate immune gene expression in murine macrophages. In response to a variety of innate agonists, macrophage paraspeckles rapidly aggregate (0.5 h poststimulation) and disaggregate (2 h poststimulation). Paraspeckle maintenance and aggregation require active transcription and MAPK signaling, whereas paraspeckle disaggregation requires degradation of Neat1 via the nuclear RNA exosome. In response to lipopolysaccharide treatment, Neat1 KO macrophages fail to properly express a large cohort of proinflammatory cytokines, chemokines, and antimicrobial mediators. Consequently, Neat1 KO macrophages cannot control replication of Salmonella enterica serovar Typhimurium or vesicular stomatitis virus. These findings highlight a prominent role for MLOs in orchestrating the macrophage response to pathogens and support a model whereby dynamic assembly and disassembly of paraspeckles reorganizes the nuclear landscape to enable inflammatory gene expression following innate stimuli.


Assuntos
Paraspeckles , RNA Longo não Codificante , Humanos , Animais , Camundongos , RNA Longo não Codificante/genética , RNA Longo não Codificante/metabolismo , Macrófagos/metabolismo
6.
Stem Cells ; 41(5): 520-539, 2023 05 15.
Artigo em Inglês | MEDLINE | ID: mdl-36945732

RESUMO

Epigenetic mechanisms regulate the multilineage differentiation capacity of hematopoietic stem cells (HSCs) into a variety of blood and immune cells. Mapping the chromatin dynamics of functionally defined cell populations will shed mechanistic insight into 2 major, unanswered questions in stem cell biology: how does epigenetic identity contribute to a cell type's lineage potential, and how do cascades of chromatin remodeling dictate ensuing fate decisions? Our recent work revealed evidence of multilineage gene priming in HSCs, where open cis-regulatory elements (CREs) exclusively shared between HSCs and unipotent lineage cells were enriched for DNA binding motifs of known lineage-specific transcription factors. Oligopotent progenitor populations operating between the HSCs and unipotent cells play essential roles in effecting hematopoietic homeostasis. To test the hypothesis that selective HSC-primed lineage-specific CREs remain accessible throughout differentiation, we used ATAC-seq to map the temporal dynamics of chromatin remodeling during progenitor differentiation. We observed epigenetic-driven clustering of oligopotent and unipotent progenitors into distinct erythromyeloid and lymphoid branches, with multipotent HSCs and MPPs associating with the erythromyeloid lineage. We mapped the dynamics of lineage-primed CREs throughout hematopoiesis and identified both unique and shared CREs as potential lineage reinforcement mechanisms at fate branch points. Additionally, quantification of genome-wide peak count and size revealed overall greater chromatin accessibility in HSCs, allowing us to identify HSC-unique peaks as putative regulators of self-renewal and multilineage potential. Finally, CRISPRi-mediated targeting of ATACseq-identified putative CREs in HSCs allowed us to demonstrate the functional role of selective CREs in lineage-specific gene expression. These findings provide insight into the regulation of stem cell multipotency and lineage commitment throughout hematopoiesis and serve as a resource to test functional drivers of hematopoietic lineage fate.


Assuntos
Cromatina , Hematopoese , Cromatina/genética , Cromatina/metabolismo , Hematopoese/genética , Células-Tronco Hematopoéticas/metabolismo , Diferenciação Celular/genética , Linhagem da Célula/genética
7.
Nat Rev Mol Cell Biol ; 24(6): 430-447, 2023 06.
Artigo em Inglês | MEDLINE | ID: mdl-36596869

RESUMO

Genes specifying long non-coding RNAs (lncRNAs) occupy a large fraction of the genomes of complex organisms. The term 'lncRNAs' encompasses RNA polymerase I (Pol I), Pol II and Pol III transcribed RNAs, and RNAs from processed introns. The various functions of lncRNAs and their many isoforms and interleaved relationships with other genes make lncRNA classification and annotation difficult. Most lncRNAs evolve more rapidly than protein-coding sequences, are cell type specific and regulate many aspects of cell differentiation and development and other physiological processes. Many lncRNAs associate with chromatin-modifying complexes, are transcribed from enhancers and nucleate phase separation of nuclear condensates and domains, indicating an intimate link between lncRNA expression and the spatial control of gene expression during development. lncRNAs also have important roles in the cytoplasm and beyond, including in the regulation of translation, metabolism and signalling. lncRNAs often have a modular structure and are rich in repeats, which are increasingly being shown to be relevant to their function. In this Consensus Statement, we address the definition and nomenclature of lncRNAs and their conservation, expression, phenotypic visibility, structure and functions. We also discuss research challenges and provide recommendations to advance the understanding of the roles of lncRNAs in development, cell biology and disease.


Assuntos
RNA Longo não Codificante , RNA Longo não Codificante/genética , Núcleo Celular/genética , Cromatina/genética , Sequências Reguladoras de Ácido Nucleico , RNA Polimerase II/genética
8.
Am J Respir Cell Mol Biol ; 68(5): 511-522, 2023 05.
Artigo em Inglês | MEDLINE | ID: mdl-36657060

RESUMO

Cigarette smoke (CS) exposure is a risk factor for many chronic diseases, including chronic obstructive pulmonary disease, but the mechanism by which smoke exposure can alter homeostasis and bring about chronic inflammation is poorly understood. Here, we showcase a novel role for smoke in regulating long noncoding RNAs, showing that it activates lincRNA-Cox2, which we previously characterized as functional in inflammatory regulation. Exposing lincRNA-Cox2 murine models to smoke in vivo confirmed lincRNA-Cox2 as a regulator of inflammatory gene expression in response to smoke both systemically and within the lung. We also report that lincRNA-Cox2 negatively regulates genes in smoked bone marrow-derived macrophages exposed to LPS stimulation. In addition to the effects on long noncoding RNAs, we also report dysregulated transcription and splicing of inflammatory protein-coding genes in the bone marrow niche after CS exposure in vivo. Collectively, this work provides insights into how innate immune signaling from gene expression to splicing is altered after in vivo exposure to CS and highlights an important new role for lincRNA-Cox2 in regulating immune genes after smoke exposure.


Assuntos
Doença Pulmonar Obstrutiva Crônica , RNA Longo não Codificante , Camundongos , Animais , RNA Longo não Codificante/genética , Macrófagos/metabolismo , Inflamação/metabolismo , Pulmão/metabolismo , Doença Pulmonar Obstrutiva Crônica/metabolismo
9.
Trends Immunol ; 43(9): 741-756, 2022 09.
Artigo em Inglês | MEDLINE | ID: mdl-35965152

RESUMO

Next-generation sequencing (NGS) technologies have greatly expanded the size of the known transcriptome. Many newly discovered transcripts are classified as long noncoding RNAs (lncRNAs) which are assumed to affect phenotype through sequence and structure and not via translated protein products despite the vast majority of them harboring short open reading frames (sORFs). Recent advances have demonstrated that the noncoding designation is incorrect in many cases and that sORF-encoded peptides (SEPs) translated from these transcripts are important contributors to diverse biological processes. Interest in SEPs is at an early stage and there is evidence for the existence of thousands of SEPs that are yet unstudied. We hope to pique interest in investigating this unexplored proteome by providing a discussion of SEP characterization generally and describing specific discoveries in innate immunity.


Assuntos
Peptídeos , RNA Longo não Codificante , Imunidade Inata , Fases de Leitura Aberta , Transcriptoma
10.
J Immunol ; 208(8): 1886-1900, 2022 04 15.
Artigo em Inglês | MEDLINE | ID: mdl-35365562

RESUMO

Our respiratory system is vital to protect us from the surrounding nonsterile environment; therefore, it is critical for a state of homeostasis to be maintained through a balance of inflammatory cues. Recent studies have shown that actively transcribed noncoding regions of the genome are emerging as key regulators of biological processes, including inflammation. lincRNA-Cox2 is one such example of an inflammatory inducible long intergenic noncoding RNA functioning to fine-tune immune gene expression. Using bulk and single-cell RNA sequencing, in addition to FACS, we find that lincRNA-Cox2 is most highly expressed in the lung and is most upregulated after LPS-induced lung injury (acute lung injury [ALI]) within alveolar macrophages, where it functions to regulate inflammation. We previously reported that lincRNA-Cox2 functions to regulate its neighboring protein Ptgs2 in cis, and in this study, we use genetic mouse models to confirm its role in regulating gene expression more broadly in trans during ALI. Il6, Ccl3, and Ccl5 are dysregulated in the lincRNA-Cox2-deficient mice and can be rescued to wild type levels by crossing the deficient mice with our newly generated lincRNA-Cox2 transgenic mice, confirming that this gene functions in trans. Many genes are specifically regulated by lincRNA-Cox2 within alveolar macrophages originating from the bone marrow because the phenotype can be reversed by transplantation of wild type bone marrow into the lincRNA-Cox2-deficient mice. In conclusion, we show that lincRNA-Cox2 is a trans-acting long noncoding RNA that functions to regulate immune responses and maintain homeostasis within the lung at baseline and on LPS-induced ALI.


Assuntos
Lesão Pulmonar Aguda , Ciclo-Oxigenase 2 , Inflamação , Macrófagos Alveolares , RNA Longo não Codificante , Lesão Pulmonar Aguda/induzido quimicamente , Lesão Pulmonar Aguda/genética , Lesão Pulmonar Aguda/metabolismo , Animais , Ciclo-Oxigenase 2/genética , Ciclo-Oxigenase 2/metabolismo , Modelos Animais de Doenças , Inflamação/genética , Inflamação/metabolismo , Lipopolissacarídeos/farmacologia , Macrófagos Alveolares/metabolismo , Camundongos , RNA Longo não Codificante/genética , RNA Longo não Codificante/metabolismo
11.
Adv Exp Med Biol ; 1363: 3-8, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35220562

RESUMO

As sequencing technologies improved, new classes of genes were uncovered. Initially, many of these were considered non-functional given their low protein-coding potential but have now emerged as important regulators of biological processes. One of the new classes of genes are called long noncoding RNAs (lncRNAs). LncRNAs are the largest group of transcribed RNA. As their name suggests, they are non-protein coding genes. To differentiate them from other smaller, noncoding RNAs, lncRNAs are transcripts whose length are greater than 200 nucleotides. According to GENCODE Release 38, there are approximately 18,000 lncRNAs, of which only 4% have a known function. Of the lncRNAs characterized, many of them play regulatory roles in many biological processes, including regulation of gene expression, alternative splicing, chromatin modification, protein activity, and posttranscriptional mechanisms. Compared to protein coding genes, lncRNAs show high cell type specificity. Many lncRNAs have been shown to be expressed in distinct immune cell populations and play RNA-mediated immune-regulatory roles. Many aspects of the immune response, including the duration, magnitude, and subsequent return to homeostasis are carefully controlled. Dysregulation of lncRNAs can result in an uncontrolled immune response, which can lead to a variety of immune-related diseases. This introduction aims to summarize the chapters highlighting the discovery of lncRNAs, their role in the immune response, and their functional characterization, either through interaction with DNA, RNA, and/or proteins in distinct immune cell populations or cells implicated in immune-related diseases. Additionally, the immune regulatory role of lncRNAs will be covered, and how lncRNA localization, sequence and secondary structure can inform function. Delving into this largely unexplored field can identify lncRNAs as potential therapeutic targets.


Assuntos
RNA Longo não Codificante , Análise de Sequência de RNA , Processamento Alternativo , Imunidade , RNA Longo não Codificante/genética , RNA Longo não Codificante/metabolismo
12.
Adv Exp Med Biol ; 1363: 179-183, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35220571

RESUMO

Until somewhat recently, the complexity of the human genome has not been well understood. With advancements in sequencing technology, we now know that nearly the whole genome is transcribed but a very small portion of those transcripts code for proteins. As the research of non-coding genes and transcripts has evolved rapidly in the last decade, it has become clear that many of them serve important biological functions in many previously well-studied cell processes. As the previous chapters in this book have reviewed, the field of noncoding RNA research has provided new insights into specific disease states, especially those driven by inflammation. Understanding the basic mechanisms of non-coding RNAs in the context of inflammation has led to prospective therapeutics that may overcome many of the challenges faced in diagnosing and treating inflammatory diseases. In this final chapter we discuss the current state of the field of non-coding RNA therapeutics and how it may evolve to overcome the short cummings we currently face with diagnosing and treating inflammatory diseases.


Assuntos
RNA Longo não Codificante , Humanos , Inflamação/genética , RNA Longo não Codificante/genética , RNA Longo não Codificante/metabolismo , RNA não Traduzido/genética
13.
Nat Commun ; 13(1): 14, 2022 01 10.
Artigo em Inglês | MEDLINE | ID: mdl-35013241

RESUMO

Type I interferons (IFNs) are critical for anti-viral responses, and also drive autoimmunity when dysregulated. Upon viral sensing, monocytes elicit a sequential cascade of IFNß and IFNα production involving feedback amplification, but how exactly this cascade is regulated in human cells is incompletely understood. Here we show that the PYHIN protein myeloid cell nuclear differentiation antigen (MNDA) is required for IFNα induction in monocytes. Unlike other PYHINs, this is not due to a pathogen sensing role, but rather MNDA regulated expression of IRF7, a transcription factor essential for IFNα induction. Mechanistically, MNDA is required for recruitment of STAT2 and RNA polymerase II to the IRF7 gene promoter, and in fact MNDA is itself recruited to the IRF7 promoter after type I IFN stimulation. These data implicate MNDA as a critical regulator of the type I IFN cascade in human myeloid cells and reveal a new role for human PYHINs in innate immune gene induction.


Assuntos
Antígenos de Diferenciação Mielomonocítica/metabolismo , Imunidade Inata , Interferon Tipo I/metabolismo , Células Mieloides/metabolismo , Fatores de Transcrição/metabolismo , Linhagem Celular , Regulação da Expressão Gênica , Humanos , Imunidade Inata/genética , Imunidade Inata/fisiologia , Monócitos/metabolismo
14.
Immunol Rev ; 305(1): 9-28, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34747035

RESUMO

For years, we have taken a reductionist approach to understanding gene regulation through the study of one gene in one cell at a time. While this approach has been fruitful it is laborious and fails to provide a global picture of what is occurring in complex situations involving tightly coordinated immune responses. The emergence of whole-genome techniques provides a system-level view of a response and can provide a plethora of information on events occurring in a cell from gene expression changes to splicing changes and chemical modifications. As with any technology, this often results in more questions than answers, but this wealth of knowledge is providing us with an unprecedented view of what occurs inside our cells during an immune response. In this review, we will discuss the current RNA-sequencing technologies and what they are helping us learn about the innate immune system.


Assuntos
Regulação da Expressão Gênica , Imunidade Inata , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Sistema Imunitário , Imunidade Inata/genética , Tecnologia
15.
Elife ; 102021 05 28.
Artigo em Inglês | MEDLINE | ID: mdl-34047695

RESUMO

Determining the layers of gene regulation within the innate immune response is critical to our understanding of the cellular responses to infection and dysregulation in disease. We identified a conserved mechanism of gene regulation in human and mouse via changes in alternative first exon (AFE) usage following inflammation, resulting in changes to the isoforms produced. Of these AFE events, we identified 95 unannotated transcription start sites in mice using a de novo transcriptome generated by long-read native RNA-sequencing, one of which is in the cytosolic receptor for dsDNA and known inflammatory inducible gene, Aim2. We show that this unannotated AFE isoform of Aim2 is the predominant isoform expressed during inflammation and contains an iron-responsive element in its 5'UTR enabling mRNA translation to be regulated by iron levels. This work highlights the importance of examining alternative isoform changes and translational regulation in the innate immune response and uncovers novel regulatory mechanisms of Aim2.


Assuntos
Processamento Alternativo , Proteínas de Ligação a DNA/genética , Éxons , Imunidade Inata/genética , Inflamação/genética , Macrófagos/metabolismo , Regiões 5' não Traduzidas , Animais , Células Cultivadas , Proteínas de Ligação a DNA/metabolismo , Perfilação da Expressão Gênica , Humanos , Inflamação/imunologia , Inflamação/metabolismo , Macrófagos/imunologia , Camundongos , Regiões Promotoras Genéticas , Transcriptoma
16.
J Biol Chem ; 296: 100784, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34000296

RESUMO

RNA-seq is routinely used to measure gene expression changes in response to cell perturbation. Genes upregulated or downregulated following some perturbation are designated as genes of interest, and their most expressed isoform(s) would then be selected for follow-up experimentation. However, because of its need to fragment RNA molecules, RNA-seq is limited in its ability to capture gene isoforms and their expression patterns. This lack of isoform-specific data means that isoforms would be selected based on annotation databases that are incomplete, not tissue specific, or do not provide key information on expression levels. As a result, minority or nonexistent isoforms might be selected for follow-up, leading to loss in valuable resources and time. There is therefore a great need to comprehensively identify gene isoforms along with their corresponding levels of expression. Using the long-read nanopore-based R2C2 method, which does not fragment RNA molecules, we generated an Isoform-level transcriptome Atlas of Macrophage Activation that identifies full-length isoforms in primary human monocyte-derived macrophages. Macrophages are critical innate immune cells important for recognizing pathogens through binding of pathogen-associated molecular patterns to toll-like receptors, culminating in the initiation of host defense pathways. We characterized isoforms for most moderately-to-highly expressed genes in resting and toll-like receptor-activated monocyte-derived macrophages, identified isoforms differentially expressed between conditions, and validated these isoforms by RT-qPCR. We compiled these data into a user-friendly data portal within the UCSC Genome Browser (https://genome.ucsc.edu/s/vollmers/IAMA). Our atlas represents a valuable resource for innate immune research, providing unprecedented isoform information for primary human macrophages.


Assuntos
Ativação de Macrófagos , Transcriptoma , Células Cultivadas , Perfilação da Expressão Gênica , Humanos , Macrófagos/imunologia , Macrófagos/metabolismo , Isoformas de Proteínas/genética
17.
PLoS One ; 16(4): e0249117, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33836009

RESUMO

Macrophages and dendritic cells (DCs) are innate immune cells that play a key role in defense against pathogens. In vitro cultures of bone marrow-derived macrophages (BMDMs) and dendritic cells (BMDCs) are well-established and valuable methods for immunological studies. Typically, commercially available recombinant GM-CSF is utilized to generate BMDCs and is also used to culture alveolar macrophages. We have generated a new HEK-293T cell line expressing murine GM-CSF that secretes high levels of GM-CSF (~180 ng/ml) into complete media as an alternative to commercial GM-CSF. Differentiation of dendritic cells and expression of various markers were kinetically assessed using the GM-CSF HEK293T cell line, termed supGM-CSF and compared directly to purified commercial GMCSF. After 7-9 days of cell culture the supGM-CSF yielded twice as many viable cells compared to the commercial purified GM-CSF. In addition to differentiating BMDCs, the supGM-CSF can be utilized to culture functionally active alveolar macrophages. Collectively, our results show that supernatant from our GM-CSF HEK293T cell line supports the differentiation of mouse BMDCs or alveolar macrophage culturing, providing an economical alternative to purified GM-CSF.


Assuntos
Exocitose , Fator Estimulador de Colônias de Granulócitos e Macrófagos/metabolismo , Animais , Técnicas de Cultura de Células/métodos , Diferenciação Celular , Meios de Cultivo Condicionados/metabolismo , Meios de Cultivo Condicionados/farmacologia , Células Dendríticas/citologia , Células Dendríticas/efeitos dos fármacos , Fator Estimulador de Colônias de Granulócitos e Macrófagos/genética , Células HEK293 , Humanos , Camundongos , Transgenes
18.
Proc Natl Acad Sci U S A ; 118(7)2021 02 16.
Artigo em Inglês | MEDLINE | ID: mdl-33568531

RESUMO

Recent studies have identified thousands of long noncoding RNAs (lncRNAs) in mammalian genomes that regulate gene expression in different biological processes. Although lncRNAs have been identified in a variety of immune cells and implicated in immune response, the biological function and mechanism of the majority remain unexplored, especially in sepsis. Here, we identify a role for a lncRNA-gastric adenocarcinoma predictive long intergenic noncoding RNA (GAPLINC)-previously characterized for its role in cancer, now in the context of innate immunity, macrophages, and LPS-induced endotoxic shock. Transcriptome analysis of macrophages from humans and mice reveals that GAPLINC is a conserved lncRNA that is highly expressed following macrophage differentiation. Upon inflammatory activation, GAPLINC is rapidly down-regulated. Macrophages depleted of GAPLINC display enhanced expression of inflammatory genes at baseline, while overexpression of GAPLINC suppresses this response. Consistent with GAPLINC-depleted cells, Gaplinc knockout mice display enhanced basal levels of inflammatory genes and show resistance to LPS-induced endotoxic shock. Mechanistically, survival is linked to increased levels of nuclear NF-κB in Gaplinc knockout mice that drives basal expression of target genes typically only activated following inflammatory stimulation. We show that this activation of immune response genes prior to LPS challenge leads to decreased blood clot formation, which protects Gaplinc knockout mice from multiorgan failure and death. Together, our results identify a previously unknown function for GAPLINC as a negative regulator of inflammation and uncover a key role for this lncRNA in modulating endotoxic shock.


Assuntos
Imunidade Inata , Choque Séptico/imunologia , Animais , Células Cultivadas , Feminino , Humanos , Lipopolissacarídeos/toxicidade , Masculino , Camundongos , Camundongos Endogâmicos C57BL , NF-kappa B/metabolismo , RNA Longo não Codificante/genética , RNA Longo não Codificante/metabolismo , Choque Séptico/etiologia , Choque Séptico/genética , Células THP-1 , Transcriptoma
19.
Cell Rep ; 33(13): 108541, 2020 12 29.
Artigo em Inglês | MEDLINE | ID: mdl-33378675

RESUMO

Macrophages are critical effector cells of the immune system, and understanding genes involved in their viability and function is essential for gaining insights into immune system dysregulation during disease. We use a high-throughput, pooled-based CRISPR-Cas screening approach to identify essential genes required for macrophage viability. In addition, we target 3' UTRs to gain insights into previously unidentified cis-regulatory regions that control these essential genes. Next, using our recently generated nuclear factor κB (NF-κB) reporter line, we perform a fluorescence-activated cell sorting (FACS)-based high-throughput genetic screen and discover a number of previously unidentified positive and negative regulators of the NF-κB pathway. We unravel complexities of the TNF signaling cascade, showing that it can function in an autocrine manner in macrophages to negatively regulate the pathway. Utilizing a single complex library design, we are capable of interrogating various aspects of macrophage biology, thus generating a resource for future studies.


Assuntos
Citometria de Fluxo/métodos , Ensaios de Triagem em Larga Escala/métodos , Inflamação/genética , Inflamação/metabolismo , Macrófagos/fisiologia , NF-kappa B/fisiologia , Fator de Necrose Tumoral alfa/fisiologia , Regiões 3' não Traduzidas , Animais , Sistemas CRISPR-Cas , Linhagem Celular , Sobrevivência Celular , Repetições Palindrômicas Curtas Agrupadas e Regularmente Espaçadas , Regulação da Expressão Gênica , Células HEK293 , Humanos , Camundongos , RNA Guia de Cinetoplastídeos/genética , Transdução de Sinais
20.
Cell Rep ; 32(2): 107881, 2020 07 14.
Artigo em Inglês | MEDLINE | ID: mdl-32668250

RESUMO

Developing strategies that promote the resolution of vascular inflammation and atherosclerosis remains a major therapeutic challenge. Here, we show that exosomes produced by naive bone marrow-derived macrophages (BMDM-exo) contain anti-inflammatory microRNA-99a/146b/378a that are further increased in exosomes produced by BMDM polarized with IL-4 (BMDM-IL-4-exo). These exosomal microRNAs suppress inflammation by targeting NF-κB and TNF-α signaling and foster M2 polarization in recipient macrophages. Repeated infusions of BMDM-IL-4-exo into Apoe-/- mice fed a Western diet reduce excessive hematopoiesis in the bone marrow and thereby the number of myeloid cells in the circulation and macrophages in aortic root lesions. This also leads to a reduction in necrotic lesion areas that collectively stabilize atheroma. Thus, BMDM-IL-4-exo may represent a useful therapeutic approach for atherosclerosis and other inflammatory disorders by targeting NF-κB and TNF-α via microRNA cargo delivery.


Assuntos
Aterosclerose/genética , Aterosclerose/patologia , Exossomos/metabolismo , Hematopoese/genética , Inflamação/genética , Inflamação/patologia , Macrófagos/metabolismo , MicroRNAs/metabolismo , Animais , Apolipoproteínas E/deficiência , Apolipoproteínas E/metabolismo , Polaridade Celular , Exossomos/ultraestrutura , Edição de Genes , Humanos , Interleucina-4/metabolismo , Macrófagos/ultraestrutura , Camundongos Endogâmicos C57BL , MicroRNAs/genética , Células Mieloides/metabolismo , NF-kappa B/metabolismo , Transdução de Sinais , Distribuição Tecidual , Fator de Necrose Tumoral alfa/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...