Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 581
Filter
1.
Proc Natl Acad Sci U S A ; 119(36): e2206327119, 2022 09 06.
Article in English | MEDLINE | ID: mdl-36037380

ABSTRACT

Cerebral malaria (CM) is a life-threatening form of Plasmodium falciparum infection caused by brain inflammation. Brain endothelium dysfunction is a hallmark of CM pathology, which is also associated with the activation of the type I interferon (IFN) inflammatory pathway. The molecular triggers and sensors eliciting brain type I IFN cellular responses during CM remain largely unknown. We herein identified the stimulator of interferon response cGAMP interactor 1 (STING1) as the key innate immune sensor that induces Ifnß1 transcription in the brain of mice infected with Plasmodium berghei ANKA (Pba). This STING1/IFNß-mediated response increases brain CXCL10 governing the extent of brain leukocyte infiltration and blood-brain barrier (BBB) breakdown, and determining CM lethality. The critical role of brain endothelial cells (BECs) in fueling type I IFN-driven brain inflammation was demonstrated in brain endothelial-specific IFNß-reporter and STING1-deficient Pba-infected mice, which were significantly protected from CM lethality. Moreover, extracellular particles (EPs) released from Pba-infected erythrocytes activated the STING1-dependent type I IFN response in BECs, a response requiring intracellular acidification. Fractionation of the EPs enabled us to identify a defined fraction carrying hemoglobin degradation remnants that activates STING1/IFNß in the brain endothelium, a process correlated with heme content. Notably, stimulation of STING1-deficient BECs with heme, docking experiments, and in vitro binding assays unveiled that heme is a putative STING1 ligand. This work shows that heme resultant from the parasite heterotrophic activity operates as an alarmin, triggering brain endothelial inflammatory responses via the STING1/IFNß/CXCL10 axis crucial to CM pathogenesis and lethality.


Subject(s)
Brain , Heme , Interferon-beta , Malaria, Cerebral , Membrane Proteins , Animals , Brain/parasitology , Endothelial Cells/immunology , Endothelial Cells/metabolism , Endothelial Cells/parasitology , Endothelium/immunology , Endothelium/parasitology , Heme/metabolism , Interferon-beta/immunology , Malaria, Cerebral/immunology , Malaria, Cerebral/parasitology , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Plasmodium berghei/metabolism , Transcriptional Activation/immunology
2.
J Immunol ; 208(2): 247-256, 2022 01 15.
Article in English | MEDLINE | ID: mdl-35017214

ABSTRACT

IFNs are comprised of three families of cytokines that confer protection against pathogen infection and uncontrolled cellular proliferation. The broad role IFNs play in innate and adaptive immune regulation has placed them under heavy scrutiny to position them as "friend" or "foe" across pathologies. Genetic lesions in genes involving IFN synthesis and signaling underscore the disparate outcomes of aberrant IFN signaling. Abrogation of the response leads to susceptibility to microbial infections whereas unabated IFN induction underlies a variety of inflammatory diseases and tumor immune evasion. Type I and III IFNs have overlapping roles in antiviral protection, yet the mechanisms by which they are induced and promote the expression of IFN-stimulated genes and inflammation can distinguish their biological functions. In this review, we examine the molecular factors that shape the shared and distinct roles of type I and III IFNs in immunity.


Subject(s)
Interferon Type I/immunology , Interferons/immunology , Virus Diseases/immunology , Gene Expression Regulation/genetics , Gene Expression Regulation/immunology , Humans , Inflammation/immunology , Interferon Type I/metabolism , Interferon Type I/therapeutic use , Interferons/metabolism , Interferons/therapeutic use , Signal Transduction/immunology , Transcriptional Activation/genetics , Transcriptional Activation/immunology , Interferon Lambda
3.
Inflammation ; 45(1): 308-330, 2022 Feb.
Article in English | MEDLINE | ID: mdl-34536158

ABSTRACT

Immunosuppression developed by cancer cells remains a leading cause of treating failure of immunotherapies. This study aimed to explore the function of human endogenous retrovirus-H long terminal repeat-associating 2 (HHLA2), an immune checkpoint molecule from the B7 family, in the immune escape in hepatocellular carcinoma (HCC). Mouse models with primary HCC or with xenograft tumors were established. The portion of tumor-associated macrophages (TAMs) and the level of PD-L1 in the tumor tissues were examined. THP-1 cells were treated with PMA to obtain a macrophage-like phenotype. The PMA-treated THP-1 cells were co-cultured with the HCC cells in Transwell chambers to examine the function of HHLA2 in chemotactic migration and polarization of macrophages. HHLA2 expression was correlated with infiltration of immune cells, especially macrophages, and was linked to poor prognosis of patients with HCC. HHLA2 knockdown reduced incidence rate of primary HCC in mice. It also reduced tumor metastasis, the portion of M2 macrophages, and the expression of PD-L1 in primary and xenograft tumors. In vitro, HHLA2 upregulation increased expression of PD-L1 in HCC cells indirectly by inducing M2 polarization and chemotactic migration of macrophages. Interferon gamma (IFNG) enhanced expression of interferon regulatory factor 1 (IFR1) in HCC cells, and IFR1 bound to the promoter region of HHLA2 to activate HHLA2 expression. This study suggested that the IFNG/IFR1/HHLA2 axis in HCC induces M2 polarization and chemotactic migration of macrophages, which leads to immune escape and development of HCC.


Subject(s)
Carcinoma, Hepatocellular/immunology , Immunoglobulins/immunology , Interferon Regulatory Factor-1/immunology , Interferon-gamma/immunology , Liver Neoplasms/immunology , Tumor Escape/immunology , Animals , B7-H1 Antigen/immunology , B7-H1 Antigen/metabolism , Carcinoma, Hepatocellular/metabolism , Humans , Immunoglobulins/metabolism , Interferon Regulatory Factor-1/metabolism , Interferon-gamma/metabolism , Liver Neoplasms/metabolism , Male , Mice , Mice, Inbred BALB C , THP-1 Cells , Transcriptional Activation/immunology
4.
J Invest Dermatol ; 142(2): 390-401, 2022 02.
Article in English | MEDLINE | ID: mdl-34293351

ABSTRACT

Psoriasis is a chronic immune-mediated disease characterized by excessive proliferation of epidermal keratinocytes and increased immune cell infiltration to the skin. Although it is well-known that psoriasis pathogenesis is driven by aberrant production of proinflammatory cytokines, the mechanisms underlying the imbalance between proinflammatory and anti-inflammatory cytokine expression are incompletely understood. In this study, we report that the transcriptional coregulators CtBP1 and 2 can transactivate a common set of proinflammatory genes both in the skin of imiquimod-induced mouse psoriasis model and in human keratinocytes and macrophages stimulated by imiquimod. We find that mice overexpressing CtBP1 in epidermal keratinocytes display severe skin inflammation phenotypes with increased expression of T helper type 1 and T helper type 17 cytokines. We also find that the expression of CtBPs and CtBP-target genes is elevated both in human psoriatic lesions and in the mouse imiquimod psoriasis model. Moreover, we were able to show that topical treatment with a peptidic inhibitor of CtBP effectively suppresses the CtBP-regulated proinflammatory gene expression and thus attenuates psoriatic inflammation in the imiquimod mouse model. Together, our findings suggest to our knowledge previously unreported strategies for therapeutic modulation of the immune response in inflammatory skin diseases.


Subject(s)
Alcohol Oxidoreductases/antagonists & inhibitors , Anti-Inflammatory Agents/pharmacology , DNA-Binding Proteins/antagonists & inhibitors , Psoriasis/drug therapy , Alcohol Oxidoreductases/genetics , Alcohol Oxidoreductases/metabolism , Animals , Anti-Inflammatory Agents/therapeutic use , Cell Proliferation/drug effects , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Disease Models, Animal , HaCaT Cells , Humans , Imiquimod/immunology , Keratinocytes/drug effects , Keratinocytes/immunology , Keratinocytes/pathology , Mice , Mice, Transgenic , Psoriasis/genetics , Psoriasis/immunology , Psoriasis/pathology , Transcriptional Activation/drug effects , Transcriptional Activation/immunology
5.
Front Immunol ; 12: 727861, 2021.
Article in English | MEDLINE | ID: mdl-34659213

ABSTRACT

Inflammatory response is a host-protective mechanism against tissue injury or infections, but also has the potential to cause extensive immunopathology and tissue damage, as seen in many diseases, such as cardiovascular diseases, neurodegenerative diseases, metabolic syndrome and many other infectious diseases with public health concerns, such as Coronavirus Disease 2019 (COVID-19), if failure to resolve in a timely manner. Recent studies have uncovered a superfamily of endogenous chemical molecules that tend to resolve inflammatory responses and re-establish homeostasis without causing excessive damage to healthy cells and tissues. Among these, the monocyte chemoattractant protein-induced protein (MCPIP) family consisting of four members (MCPIP-1, -2, -3, and -4) has emerged as a group of evolutionarily conserved molecules participating in the resolution of inflammation. The focus of this review highlights the biological functions of MCPIP-1 (also known as Regnase-1), the best-studied member of this family, in the resolution of inflammatory response. As outlined in this review, MCPIP-1 acts on specific signaling pathways, in particular NFκB, to blunt production of inflammatory mediators, while also acts as an endonuclease controlling the stability of mRNA and microRNA (miRNA), leading to the resolution of inflammation, clearance of virus and dead cells, and promotion of tissue regeneration via its pleiotropic effects. Evidence from transgenic and knock-out mouse models revealed an involvement of MCPIP-1 expression in immune functions and in the physiology of the cardiovascular system, indicating that MCPIP-1 is a key endogenous molecule that governs normal resolution of acute inflammation and infection. In this review, we also discuss the current evidence underlying the roles of other members of the MCPIP family in the regulation of inflammatory processes. Further understanding of the proteins from this family will provide new insights into the identification of novel targets for both host effectors and microbial factors and will lead to new therapeutic treatments for infections and other inflammatory diseases.


Subject(s)
Gene Expression Regulation/genetics , Inflammation Mediators/metabolism , Inflammation/immunology , Ribonucleases/immunology , SARS-CoV-2/immunology , Transcription Factors/immunology , Animals , Apoptosis/genetics , COVID-19/immunology , Humans , Inflammation/pathology , Mice , NF-kappa B/metabolism , RNA Processing, Post-Transcriptional/genetics , Transcriptional Activation/immunology , Ubiquitination
6.
Cells ; 10(10)2021 09 26.
Article in English | MEDLINE | ID: mdl-34685526

ABSTRACT

Psoriasis is a chronic inflammatory skin disease that is mediated by complex crosstalk between immune cells and keratinocytes (KCs). Emerging studies have showed a specific psoriatic microRNAs signature, in which miR-21 is one of the most upregulated and dynamic miRNAs. In this study, we focused our investigations on the passenger miR-21-3p strand, which is poorly studied in skin and in psoriasis pathogenesis. Here, we showed the upregulation of miR-21-3p in an IMQ-induced psoriasiform mouse model. This upregulation was correlated with IL-22 expression and functionality, both in vitro and in vivo, and it occurred via STAT3 and NF-κB signaling. We identified a network of differentially expressed genes involved in abnormal proliferation control and immune regulatory genes implicated in the molecular pathogenesis of psoriasis in response to miR-21-3p overexpression in KCs. These results were confirmed by functional assays that validated the proliferative potential of miR-21-3p. All these findings highlight the importance of miR-21-3p, an underestimated miRNA, in psoriasis and provide novel molecular targets for therapeutic purposes.


Subject(s)
Inflammation/immunology , Interleukins/metabolism , MicroRNAs/genetics , Psoriasis/metabolism , Animals , Cell Proliferation/genetics , Cell Proliferation/physiology , Down-Regulation , Keratinocytes/metabolism , Mice , MicroRNAs/metabolism , Psoriasis/drug therapy , Skin/metabolism , Transcriptional Activation/immunology , Up-Regulation , Interleukin-22
7.
J Immunol Res ; 2021: 8483938, 2021.
Article in English | MEDLINE | ID: mdl-34222497

ABSTRACT

Follicular dendritic cells are important stromal components of the germinal center (GC) and have pivotal roles in maintaining the GC microenvironment for high-affinity antibody production. Tumor necrosis factor-α (TNFα) is essential for the development and functions of follicular dendritic cells. Despite the importance of follicular dendritic cells in humoral immunity, their molecular control mechanisms have yet to be fully elucidated due to the lack of an adequate investigation system. Here, we have used a unique human primary follicular dendritic cell-like cell (FDCLC) to demonstrate that the migration of these cells is enhanced by TNFα-mediated metalloproteinase 3 (MMP3) expression. MMP3 was found to be highly expressed in normal human GCs and markedly upregulated in human primary FDCLCs by TNFα. TNFα induced ERK1/2 phosphorylation and the transcription of MMP3 through AP1. TNFα treatment increased FDCLC migration, and a knockdown of MMP3 significantly reduced the TNFα-induced migration of FDCLCs. Overall, we have newly identified a control mechanism for the expression of MMP3 in FDCLCs that modulates their migration and may indicate an important role in GC biology. Since GCs are observed in the lesions of autoimmune diseases and lymphomas, targeting the MMP3/TNFα-mediated migration of stromal cells in the B cell follicle may have great potential as a future therapeutic modality against aberrant GC-associated disorders.


Subject(s)
Dendritic Cells, Follicular/immunology , Germinal Center/immunology , Matrix Metalloproteinase 3/genetics , Transcription Factor AP-1/metabolism , Tumor Necrosis Factor-alpha/metabolism , Cell Movement/genetics , Cells, Cultured , Dendritic Cells, Follicular/metabolism , Gene Knockdown Techniques , Germinal Center/cytology , Germinal Center/metabolism , Humans , MAP Kinase Signaling System/immunology , Matrix Metalloproteinase 3/metabolism , Phosphorylation/immunology , Primary Cell Culture , Transcriptional Activation/immunology
8.
Nat Commun ; 12(1): 3182, 2021 06 01.
Article in English | MEDLINE | ID: mdl-34075041

ABSTRACT

Interleukin 9 (IL-9)-producing helper T (Th9) cells are essential for inducing anti-tumor immunity and inflammation in allergic and autoimmune diseases. Although transcription factors that are essential for Th9 cell differentiation have been identified, other signaling pathways that are required for their generation and functions are yet to be explored. Here, we identify that Epidermal Growth Factor Receptor (EGFR) is essential for IL-9 induction in helper T (Th) cells. Moreover, amphiregulin (Areg), an EGFR ligand, is critical for the amplification of Th9 cells induced by TGF-ß1 and IL-4. Furthermore, our data show that Areg-EGFR signaling induces HIF1α, which binds and transactivates IL-9 and NOS2 promoters in Th9 cells. Loss of EGFR or HIF1α abrogates Th9 cell differentiation and suppresses their anti-tumor functions. Moreover, in line with its reliance on HIF1α expression, metabolomics profiling of Th9 cells revealed that Succinate, a TCA cycle metabolite, promotes Th9 cell differentiation and Th9 cell-mediated tumor regression.


Subject(s)
ErbB Receptors/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Interleukin-9/genetics , Melanoma, Experimental/therapy , Skin Neoplasms/therapy , T-Lymphocytes, Helper-Inducer/immunology , Amphiregulin/metabolism , Animals , Cell Differentiation/immunology , Female , HEK293 Cells , Healthy Volunteers , Humans , Immunotherapy, Adoptive/methods , Melanoma, Experimental/immunology , Mice , Mice, Knockout , Nitric Oxide Synthase Type II/genetics , Primary Cell Culture , RNA-Seq , Signal Transduction/genetics , Signal Transduction/immunology , Skin Neoplasms/immunology , Succinic Acid/metabolism , T-Lymphocytes, Helper-Inducer/transplantation , Transcriptional Activation/immunology
9.
Eur J Pharmacol ; 906: 174270, 2021 Sep 05.
Article in English | MEDLINE | ID: mdl-34171392

ABSTRACT

In addition to its pivotal role in purine metabolism, xanthine oxidoreductase (XOR) is one of the key enzymes involved in superoxide radical generation. Oxidative stress has been implicated in the etiology of colorectal cancer, but the contribution of XOR remains unclear. Here we investigated the role of XOR in colitis-associated colorectal cancer (CAC) and the underlying mechanisms. Using clinical samples, we demonstrated that XOR up-regulation was an early event in colonic carcinogenesis. Pharmacological inhibition of XOR effectively delayed the progression of CAC. Moreover, XOR activity positively correlated with tumor necrosis factor-alpha (TNFα) protein levels. Mechanistically, TNFα may activate XOR transcription via activator protein-1 and, thus, promote endogenous hydrogen peroxide generation, resulting in oxidative DNA damage in colon cancer cells. On the other hand, XOR may regulate the TNFα mRNA transcripts by mediating LPS-induced macrophage M1 polarization. Collectively, XOR promotes tumor development by programming the tumor microenvironment and stimulates CAC progression via DNA damage-induced genetic instability.


Subject(s)
Colitis-Associated Neoplasms/immunology , DNA Damage/immunology , Macrophages/immunology , Oxidative Stress/immunology , Xanthine Dehydrogenase/metabolism , Animals , Carcinogenesis/chemically induced , Carcinogenesis/immunology , Cell Line, Tumor , Colitis-Associated Neoplasms/genetics , Colitis-Associated Neoplasms/pathology , Colon/immunology , Colon/pathology , Disease Models, Animal , Disease Progression , Gene Expression Profiling , Gene Expression Regulation, Neoplastic/immunology , Humans , Macrophages/metabolism , Male , Transcriptional Activation/immunology , Tumor Microenvironment/immunology , Tumor Necrosis Factor-alpha/metabolism , Up-Regulation , Xanthine Dehydrogenase/genetics
10.
Mol Cells ; 44(5): 342-355, 2021 May 31.
Article in English | MEDLINE | ID: mdl-33972476

ABSTRACT

The microphthalmia-associated transcription factor family (MiT family) proteins are evolutionarily conserved transcription factors that perform many essential biological functions. In mammals, the MiT family consists of MITF (microphthalmia-associated transcription factor or melanocyte-inducing transcription factor), TFEB (transcription factor EB), TFE3 (transcription factor E3), and TFEC (transcription factor EC). These transcriptional factors belong to the basic helix-loop-helix-leucine zipper (bHLH-LZ) transcription factor family and bind the E-box DNA motifs in the promoter regions of target genes to enhance transcription. The best studied functions of MiT proteins include lysosome biogenesis and autophagy induction. In addition, they modulate cellular metabolism, mitochondria dynamics, and various stress responses. The control of nuclear localization via phosphorylation and dephosphorylation serves as the primary regulatory mechanism for MiT family proteins, and several kinases and phosphatases have been identified to directly determine the transcriptional activities of MiT proteins. In different immune cell types, each MiT family member is shown to play distinct or redundant roles and we expect that there is far more to learn about their functions and regulatory mechanisms in host defense and inflammatory responses.


Subject(s)
Phosphorylation/immunology , Transcription Factors/immunology , Transcriptional Activation/immunology , Amino Acid Sequence , Humans
11.
Mol Immunol ; 135: 268-275, 2021 07.
Article in English | MEDLINE | ID: mdl-33940513

ABSTRACT

The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infections have triggered global pandemic that continue to impact adversely human health. New understanding has emerged about the innate and adaptive immune responses elicited in SARS-CoV-2 infection. The understanding of innate immune responses generated in hosts early in SARS-CoV-2 infection is vital for treatment efforts. Antiviral cytokines are released by innate immune cells in response to viral infections that play a pivotal role in limiting viral replication, pathology and generating optimal adaptive immune responses alongside the long-term memory responses against reinfections. One aspect of innate immune response generated against SARS-CoV-2 in vivo and which has received much attention has been high proinflammatory cytokine release in COVID-19 patients. Another vital discovery has been that the antiviral cytokine type I Interferon (IFN) family IFN-α mediates upregulation of angiotensin converting enzyme 2 (ACE2) membrane protein in airway epithelial cells. ACE2 is a receptor that SARS-CoV-2 binds to infect host cells. New understanding has emerged about the mechanism of SARS-CoV-2 induced exaggerated proinflammatory cytokine release as well as transcriptional regulation of ACE2. This review discusses various mechanisms underlying SARS-CoV-2 induced exaggerated proinflammatory cytokine response as well as transcriptional regulation of ACE2 receptor. We further elaborate on adaptive and memory responses generated against SARS-CoV-2.


Subject(s)
COVID-19/immunology , Cytokines/immunology , Immunity, Innate , Immunologic Memory , SARS-CoV-2/immunology , Angiotensin-Converting Enzyme 2/immunology , COVID-19/pathology , Humans , Transcriptional Activation/immunology
12.
Viruses ; 13(2)2021 Jan 20.
Article in English | MEDLINE | ID: mdl-33498196

ABSTRACT

The nuclear factor κB (NF-κB) family are the master transcription factors that control cell proliferation, apoptosis, the expression of interferons and proinflammatory factors, and viral infection. During viral infection, host innate immune system senses viral products, such as viral nucleic acids, to activate innate defense pathways, including the NF-κB signaling axis, thereby inhibiting viral infection. In these NF-κB signaling pathways, diverse types of ubiquitination have been shown to participate in different steps of the signal cascades. Recent advances find that viruses also modulate the ubiquitination in NF-κB signaling pathways to activate viral gene expression or inhibit host NF-κB activation and inflammation, thereby facilitating viral infection. Understanding the role of ubiquitination in NF-κB signaling during viral infection will advance our knowledge of regulatory mechanisms of NF-κB signaling and pave the avenue for potential antiviral therapeutics. Thus, here we systematically review the ubiquitination in NF-κB signaling, delineate how viruses modulate the NF-κB signaling via ubiquitination and discuss the potential future directions.


Subject(s)
Interferons/immunology , NF-kappa B/metabolism , Ubiquitination , Ubiquitins/immunology , Virus Diseases/immunology , Animals , Humans , Protein Binding , Signal Transduction/immunology , Structure-Activity Relationship , Transcriptional Activation/immunology , Virus Diseases/virology
13.
J Invest Dermatol ; 141(3): 596-606.e7, 2021 03.
Article in English | MEDLINE | ID: mdl-32853659

ABSTRACT

IL-33 is constitutively expressed in the skin. Psoriasis is a common skin inflammatory disease. The roles of IL-33 in psoriasis have not been well-elucidated. We identified that keratinocytes (KCs) are the predominant cells expressing IL-33 and its receptor, suppression of tumorigenicity 2, in the skin. KCs actively released IL-33 on psoriasis inflammatory stimuli and induced psoriasis-related cytokine, chemokine, and inflammatory molecules genes transcription in KCs in an autocrine manner. IL-33‒specific deficiency in KCs ameliorated imiquimod-induced psoriatic dermatitis. In addition, intradermal injection of recombinant IL-33 alone induced psoriasis-like dermatitis, which is attributed to the transcriptional upregulation of genes enriched in IL-17, TNF, and chemokine signaling pathway in KCs on recombinant IL-33 stimulation. Our data demonstrate that the autocrine circuit of IL-33 in KCs promotes the progression of psoriatic skin inflammation, and IL-33 is a potential therapeutic target for psoriasis.


Subject(s)
Interleukin-33/metabolism , Keratinocytes/metabolism , Psoriasis/immunology , Adult , Animals , Autocrine Communication/immunology , Biopsy , Case-Control Studies , Disease Models, Animal , Disease Progression , Healthy Volunteers , Humans , Imiquimod/immunology , Injections, Intradermal , Interleukin-1 Receptor-Like 1 Protein/metabolism , Interleukin-33/administration & dosage , Interleukin-33/genetics , Keratinocytes/immunology , Male , Mice , Middle Aged , Psoriasis/diagnosis , Psoriasis/genetics , Psoriasis/pathology , Recombinant Proteins/administration & dosage , Recombinant Proteins/metabolism , Severity of Illness Index , Signal Transduction/genetics , Signal Transduction/immunology , Skin/immunology , Skin/pathology , Transcriptional Activation/immunology , Up-Regulation/immunology
14.
Carcinogenesis ; 42(3): 344-356, 2021 04 17.
Article in English | MEDLINE | ID: mdl-33146712

ABSTRACT

Recently, we identified that the atypical protein kinase C isoform ι (PKCι) enhances the expression of Yes-associated protein 1 (YAP1) to promote the tumorigenesis of pancreatic adenocarcinoma harboring mutant KRAS (mu-KRAS). To advance our understanding about underlying mechanisms, we analyze the transcription of YAP1 in pancreatic cancer cells and reveal that transcription factor specificity protein 1 (Sp1) is upregulated by PKCι and subsequently binds to multiple sites in YAP1 promoter to drive the transactivation of YAP1 in pancreatic cancer cells carrying mu-KRAS. The bioinformatics analysis further substantiates that the expression of PKCι, Sp1 and YAP1 is correlated and associated with the stages and prognosis of pancreatic tumors. Moreover, our apoptotic detection data demonstrate that combination of PKCι and Sp1 inhibitors at subtoxic doses displays synergistic effects on inducing apoptosis and reversing the immunosuppression of pancreatic cancer cells, establishing the combination of PKCι and Sp1 inhibitors as a promising novel therapeutic approach, or an adjuvant strategy to potentiate the antitumor effects of other immunotherapeutic agents in pancreatic cancer treatment.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Isoenzymes/metabolism , Pancreatic Neoplasms/genetics , Protein Kinase C/metabolism , Sp1 Transcription Factor/genetics , Transcription Factors/genetics , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Apoptosis/drug effects , Apoptosis/genetics , Apoptosis/immunology , Carcinogenesis/drug effects , Carcinogenesis/genetics , Carcinogenesis/immunology , Cell Line, Tumor , Computational Biology , Datasets as Topic , Drug Synergism , Gene Expression Regulation, Neoplastic/drug effects , Gene Expression Regulation, Neoplastic/immunology , Humans , Isoenzymes/antagonists & inhibitors , Mutation , Pancreas/immunology , Pancreas/pathology , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/immunology , Pancreatic Neoplasms/pathology , Prognosis , Promoter Regions, Genetic/genetics , Protein Kinase C/antagonists & inhibitors , Proto-Oncogene Proteins p21(ras)/genetics , RNA-Seq , Sp1 Transcription Factor/antagonists & inhibitors , Sp1 Transcription Factor/metabolism , Transcriptional Activation/drug effects , Transcriptional Activation/immunology , Tumor Escape/drug effects , Tumor Escape/genetics , Up-Regulation/drug effects , Up-Regulation/immunology , YAP-Signaling Proteins
15.
FEBS Open Bio ; 11(1): 105-113, 2021 01.
Article in English | MEDLINE | ID: mdl-33159419

ABSTRACT

Interferon (IFN)-inducible 44 like (IFI44L) is an IFN-stimulated gene (ISG), which is located on the same chromosome as the known antiviral ISG IFI44. Expression of IFI44L is induced by IFN and HIV-1 infection. However, the mechanism by which IFN-I induces IFI44L production has not yet been determined. In this study, we analyzed transcriptional regulation of IFI44L via cloning of the IFI44L promoter. We found that IFI44L has two IFN-stimulated response elements (ISRE), which are necessary for the basal level of IFI44L transcription. IFN-I and IFN-II can activate the IFI44L promoter through one of the two ISREs. IFN regulatory factor (IRF)-1 can activate transcription of IFI44L by binding to one of the ISREs. Additionally, co-transfection of the IFI44L promoter with an HIV-1 infectious clone or HIV-1 infection activated IFI44L promoter transcription, but did not upregulate IFI44L expression via ISREs. These findings will help to understand the interaction between IFI44L and HIV-1, and aid in elucidation of the role of IFI44L in the antiviral innate immune response.


Subject(s)
HIV-1/immunology , Interferon Regulatory Factor-1/metabolism , Tumor Suppressor Proteins/genetics , Cloning, Molecular , HEK293 Cells , HeLa Cells , Humans , Immunity, Innate/genetics , Promoter Regions, Genetic , Transcriptional Activation/immunology
16.
Theranostics ; 10(26): 12011-12025, 2020.
Article in English | MEDLINE | ID: mdl-33204326

ABSTRACT

Calcium oxalate (CaOx) crystal can trigger kidney injury, which contributes to the pathogenesis of nephrocalcinosis. The phenotypes of infiltrating macrophage may impact CaOx-mediated kidney inflammatory injury as well as crystal deposition. How aryl hydrocarbon receptor (AhR) regulates inflammation and macrophage polarization is well understood; however, how it modulates CaOx nephrocalcinosis remains unclear. Methods: Mice were intraperitoneally injected with glyoxylate to establish CaOx nephrocalcinosis model with or without the treatment of AhR activator 6-formylindolo(3,2-b)carbazole (FICZ). Positron emission tomography computed tomography (PET-CT) imaging, Periodic acid-Schiff (PAS) staining, and polarized light optical microscopy were used to evaluate kidney injury and crystal deposition in mice kidney. Western blotting, immunofluorescence, chromatin immunoprecipitation, microRNA-fluorescence in situ hybridization, and luciferase reporter assays were applied to analyze polarization state and regulation mechanism of macrophage. Results: AhR expression was significantly upregulated and negatively correlated with interferon-regulatory factor 1 (IRF1) and hypoxia inducible factor 1-alpha (HIF-1α) levels in a murine CaOx nephrocalcinosis model following administration of FICZ. Moreover, AhR activation suppressed IRF1 and HIF-1α levels and decreased M1 macrophage polarization in vitro. In terms of the mechanism, bioinformatics analysis and chromatin immunoprecipitation assay confirmed that AhR could bind to miR-142a promoter to transcriptionally activate miR-142a. In addition, luciferase reporter assays validated that miR-142a inhibited IRF1 and HIF-1α expression by directly targeting their 3'-untranslated regions. Conclusions: Our results indicated that AhR activation could diminish M1 macrophage polarization and promote M2 macrophage polarization to suppress CaOx nephrocalcinosis via the AhR-miR-142a-IRF1/HIF-1α pathway.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Calcium Oxalate/metabolism , Macrophages/immunology , MicroRNAs/genetics , Nephrocalcinosis/immunology , Receptors, Aryl Hydrocarbon/metabolism , 3' Untranslated Regions/genetics , Animals , Basic Helix-Loop-Helix Transcription Factors/agonists , Basic Helix-Loop-Helix Transcription Factors/analysis , Carbazoles/administration & dosage , Case-Control Studies , Cells, Cultured , Computational Biology , Disease Models, Animal , Epithelial Cells , Glyoxylates/administration & dosage , Glyoxylates/toxicity , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Interferon Regulatory Factor-1/genetics , Kidney/diagnostic imaging , Kidney/drug effects , Kidney/pathology , Kidney/surgery , Macrophage Activation , Macrophages/metabolism , Male , Mice , MicroRNAs/metabolism , Nephrocalcinosis/chemically induced , Nephrocalcinosis/diagnosis , Nephrocalcinosis/surgery , Nephrolithotomy, Percutaneous , Positron Emission Tomography Computed Tomography , Primary Cell Culture , Receptors, Aryl Hydrocarbon/agonists , Receptors, Aryl Hydrocarbon/analysis , Transcriptional Activation/immunology , Up-Regulation/immunology
17.
Proc Natl Acad Sci U S A ; 117(47): 29823-29831, 2020 11 24.
Article in English | MEDLINE | ID: mdl-33168738

ABSTRACT

Nuclear factor-ĸB (NF-ĸB) transcription factor is a family of essential regulators of the immune response and cell proliferation and transformation. A typical factor is a heterodimer made of either p50 or p52, which are limited processing products of either p105 or p100, respectively, and a member of the Rel family of proteins, typically p65. The transcriptional program of NF-ĸB is tightly regulated by the composition of the dimers. In our previous work, we demonstrated that the ubiquitin ligase KPC1 is involved in ubiquitination and proteasomal processing of p105 to generate p50. Its overexpression and the resulting high level of p50 stimulates transcription of a broad array of tumor suppressors. Here we demonstrate that additional mechanisms are involved in the p50-mediated tumor-suppressive effect. p50 down-regulates expression of a major immune checkpoint inhibitor, the programmed cell death-ligand 1 (PD-L1), both in cells and in tumors. Importantly, the suppression is abrogated by overexpression of p65. This highlights the importance of the cellular quantities of the two different subunits of NF-ĸB which determine the composition of the dimer. While the putative p50 homodimer is tumor-suppressive, the "canonical" p50p65 heterodimer is oncogenic. We found that an additional mechanism is involved in the tumor-suppressive phenomenon: p50 up-regulates expression of the proinflammatory chemokines CCL3, CCL4, and CCL5, which in turn recruit into the tumors active natural killer (NK) cells and macrophages. Overall, p50 acts as a strong tumor suppressor via multiple mechanisms, including overexpression of tumor suppressors and modulation of the tumor microenvironment by recruiting active immune cells.


Subject(s)
B7-H1 Antigen/metabolism , Gene Expression Regulation, Neoplastic/immunology , NF-kappa B p50 Subunit/metabolism , Neoplasms/genetics , Ubiquitin-Protein Ligases/metabolism , Adoptive Transfer , Animals , B7-H1 Antigen/immunology , Cell Line, Tumor , Chemokines/immunology , Chemokines/metabolism , HEK293 Cells , Humans , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Killer Cells, Natural/transplantation , Macrophages/immunology , Macrophages/metabolism , Mice , Neoplasms/immunology , Neoplasms/pathology , Primary Cell Culture , Transcription Factor RelA/metabolism , Transcriptional Activation/immunology , Tumor Microenvironment/genetics , Tumor Microenvironment/immunology , Ubiquitination/genetics , Ubiquitination/immunology , Up-Regulation , Xenograft Model Antitumor Assays
18.
J Immunol ; 205(11): 2953-2958, 2020 12 01.
Article in English | MEDLINE | ID: mdl-33139491

ABSTRACT

Transcription factor Foxp3 specifies and maintains regulatory T cell (Treg) identity. During Treg differentiation, a CpG-rich Foxp3 intronic enhancer, conserved noncoding sequence 2 (CNS2), is activated via DNA demethylation to establish epigenetic memory of Foxp3 expression to protect Treg identity. However, it is unclear how this epigenetic memory of Foxp3 expression is established, as CNS2 is thought to be demethylated independently of Foxp3 expression. In this article, we uncover an unexpected causal relationship between Foxp3-transcriptional activation and CNS2 demethylation in mice. CRISPR/dCas9-mediated Foxp3-transcriptional activation elicits CNS2 demethylation. Sustaining Foxp3-transcriptional activation in induced Tregs also promotes CNS2 demethylation, enhancing Treg lineage stability and suppressive function. Importantly, CRISPR-mediated silencing of Foxp3 transcription, but not protein expression, abolishes CNS2 demethylation. The novel finding that Foxp3-transcriptional activation promotes CNS2 demethylation may facilitate the development of Treg-based therapies and represent a general mechanism for the establishment of epigenetic memory of immune gene expression.


Subject(s)
Clustered Regularly Interspaced Short Palindromic Repeats/genetics , Epigenesis, Genetic/genetics , Forkhead Transcription Factors/genetics , Regulatory Sequences, Nucleic Acid/genetics , T-Lymphocytes, Regulatory/immunology , Transcription, Genetic/genetics , Animals , Clustered Regularly Interspaced Short Palindromic Repeats/immunology , Conserved Sequence/genetics , Conserved Sequence/immunology , DNA Methylation/genetics , DNA Methylation/immunology , Epigenesis, Genetic/immunology , Epigenomics/methods , Forkhead Transcription Factors/immunology , Gene Expression/genetics , Gene Expression/immunology , Gene Expression Regulation/genetics , Gene Expression Regulation/immunology , Mice , Regulatory Sequences, Nucleic Acid/immunology , Transcription, Genetic/immunology , Transcriptional Activation/genetics , Transcriptional Activation/immunology
19.
Sci Rep ; 10(1): 18170, 2020 10 23.
Article in English | MEDLINE | ID: mdl-33097782

ABSTRACT

Stroke is caused by obstructed blood flow (ischaemia) or unrestricted bleeding in the brain (haemorrhage). Global brain ischaemia occurs after restricted cerebral blood flow e.g. during cardiac arrest. Following ischaemic injury, restoration of blood flow causes ischaemia-reperfusion (I/R) injury which worsens outcome. Secondary injury mechanisms after any stroke are similar, and encompass inflammation, endothelial dysfunction, blood-brain barrier (BBB) damage and apoptosis. We developed a new model of transient global forebrain I/R injury (dual carotid artery ligation; DCAL) and compared the manifestations of this injury with those in a conventional I/R injury model (middle-cerebral artery occlusion; MCAo) and with intracerebral haemorrhage (ICH; collagenase model). MRI revealed that DCAL produced smaller bilateral lesions predominantly localised to the striatum, whereas MCAo produced larger focal corticostriatal lesions. After global forebrain ischaemia mice had worse overall neurological scores, although quantitative locomotor assessment showed MCAo and ICH had significantly worsened mobility. BBB breakdown was highest in the DCAL model while apoptotic activity was highest after ICH. VCAM-1 upregulation was specific to ischaemic models only. Differential transcriptional upregulation of pro-inflammatory chemokines and cytokines and TLRs was seen in the three models. Our findings offer a unique insight into the similarities and differences in how biological processes are regulated after different types of stroke. They also establish a platform for analysis of therapies such as endothelial protective and anti-inflammatory agents that can be applied to all types of stroke.


Subject(s)
Cerebrovascular Circulation/physiology , Hemorrhagic Stroke/pathology , Ischemic Stroke/pathology , Prosencephalon/blood supply , Reperfusion Injury/pathology , Animals , Anti-Inflammatory Agents/therapeutic use , Apoptosis/immunology , Blood-Brain Barrier/diagnostic imaging , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/pathology , Carotid Arteries/physiopathology , Cerebrovascular Circulation/drug effects , Collagenases/administration & dosage , Collagenases/adverse effects , Cytokines/genetics , Cytokines/immunology , Disease Models, Animal , Endothelium, Vascular/drug effects , Endothelium, Vascular/pathology , Hemorrhagic Stroke/drug therapy , Hemorrhagic Stroke/immunology , Hemorrhagic Stroke/physiopathology , Humans , Ischemic Stroke/drug therapy , Ischemic Stroke/immunology , Ischemic Stroke/physiopathology , Ligation , Locomotion/physiology , Magnetic Resonance Imaging , Male , Mice , Middle Cerebral Artery/physiopathology , Prosencephalon/diagnostic imaging , Prosencephalon/drug effects , Prosencephalon/pathology , Protective Agents/therapeutic use , Reperfusion Injury/drug therapy , Reperfusion Injury/immunology , Reperfusion Injury/physiopathology , Toll-Like Receptors/genetics , Transcriptional Activation/immunology
20.
J Immunol ; 205(10): 2606-2617, 2020 11 15.
Article in English | MEDLINE | ID: mdl-33046502

ABSTRACT

Sensors that detect dsRNA stimulate IFN responses as a defense against viral infection. IFN responses are also well documented in a variety of human autoimmune diseases, including relapsing-remitting multiple sclerosis (MS), in which increased IFN responses result from increased levels of double-stranded endogenous Alu RNAs. Mechanisms underlying increases in double-stranded Alu RNAs in MS are obscure. We find widespread loss of adenosine-to-inosine editing of Alu RNAs in MS. Unedited Alu RNAs are potent activators of both IFN and NF-κB responses via the dsRNA sensors, RIG-I, and TLR3. Minor editing of highly active Alu elements abrogates the ability to activate both transcriptional responses. Thus, adenosine-to-inosine editing may also represent an important defense against autoimmune diseases such as MS.


Subject(s)
Alu Elements/immunology , Multiple Sclerosis, Relapsing-Remitting/genetics , RNA Editing/immunology , RNA, Double-Stranded/immunology , Transcriptional Activation/immunology , Adenosine/genetics , Alu Elements/genetics , DEAD Box Protein 58/metabolism , Datasets as Topic , HEK293 Cells , Humans , Inflammation/genetics , Inflammation/immunology , Inosine/genetics , Interferons/metabolism , Multiple Sclerosis, Relapsing-Remitting/blood , Multiple Sclerosis, Relapsing-Remitting/immunology , NF-kappa B/metabolism , RNA, Double-Stranded/genetics , RNA, Double-Stranded/metabolism , RNA-Seq , Receptors, Immunologic/metabolism , Signal Transduction/genetics , Signal Transduction/immunology , THP-1 Cells , Toll-Like Receptor 3/metabolism , Whole Genome Sequencing
SELECTION OF CITATIONS
SEARCH DETAIL
...