Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 69
Filter
1.
Ageing Res Rev ; 82: 101765, 2022 12.
Article in English | MEDLINE | ID: mdl-36270606

ABSTRACT

Cellular changes that are linked to aging in humans include genomic instability, telomere attrition, epigenetic alterations, mitochondrial dysfunction, cellular senescence, and altered intercellular communications. The extent of the changes in these aging hallmarks and their interactions with each other are part of the human aging. However, the molecular mechanisms through which the aging hallmarks interact with each other remain unclear. Studies have indicated a potential role for the type I interferon (IFN) and p53-inducible IFI16 proteins in interactions with the aging hallmarks. The IFI16 proteins are members of the PYHIN protein family. Proteins in the family share a DNA-binding domain (the HIN domain) and a protein-protein interaction pyrin domain (PYD). IFI16 proteins are needed for cytosolic DNA-induced activation of the cGAS-STING pathway for type I IFN (IFN-ß) expression. The pathway plays an important role in aging-related inflammation (inflammaging). Further, increased levels of the IFI16 proteins potentiate the cell growth inhibitory functions of the p53 and pRb tumor suppressors proteins. Moreover, IFI16 proteins are needed for most aging hallmarks. Therefore, here we discuss how an improved understanding of the role of the IFI16 proteins in integration of the aging hallmarks has potential to improve the human health and lifespan.


Subject(s)
Phosphoproteins , Tumor Suppressor Protein p53 , Humans , Tumor Suppressor Protein p53/metabolism , Phosphoproteins/genetics , Phosphoproteins/metabolism , Cellular Senescence/genetics , Aging/genetics , DNA/metabolism , Nuclear Proteins/genetics , Nuclear Proteins/metabolism
2.
Medicine (Baltimore) ; 101(3): e28645, 2022 Jan 21.
Article in English | MEDLINE | ID: mdl-35060553

ABSTRACT

ABSTRACT: The peripheral immune system has a key pathophysiologic role in Frontotemporal degeneration (FTD). We sought a comprehensive transcriptome-wide evaluation of gene expression alterations unique to the peripheral immune system in FTD compared to healthy controls and amyotrophic lateral sclerosis.Nineteen subjects with FTD with 19 matched healthy controls and 9 subjects with amyotrophic lateral sclerosis underwent isolation of peripheral blood mononuclear cells (PBMCs) which then underwent bulk ribonucleic acid sequencing.There was increased expression in genes associated with CD19+ B-cells, CD4+ T-cells, and CD8+ T-cells in FTD participants compared to healthy controls. In contrast, there was decreased expression in CD33+ myeloid cells, CD14+ monocytes, BDCA4+ dendritic cells, and CD56+ natural killer cells in FTD and healthy controls. Additionally, there was decreased expression is seen in associated with 2 molecular processes: autophagy with phagosomes and lysosomes, and protein processing/export. Significantly downregulated in PBMCs of FTD subjects were genes involved in antigen processing and presentation as well as lysosomal lumen formation compared to healthy control PBMCs.Our findings that the immune signature based on gene expression in PBMCs of FTD participants favors adaptive immune cells compared to innate immune cells. And decreased expression in genes associated with phagosomes and lysosomes in PBMCs of FTD participants compared to healthy controls.


Subject(s)
Amyotrophic Lateral Sclerosis , Frontotemporal Dementia , Neurodegenerative Diseases , Aged , Atrophy , Case-Control Studies , Female , Gene Expression , Humans , Leukocytes, Mononuclear , Male , Middle Aged , Transcriptome
3.
Life (Basel) ; 11(4)2021 Apr 20.
Article in English | MEDLINE | ID: mdl-33923931

ABSTRACT

Increased levels of type I (T1) interferon (IFN)-inducible POP3 protein in myeloid cells inhibit activation of the AIM2 inflammasome and production of IL-1ß and IL-18 proinflammatory cytokines. The AIM2 mRNA levels were significantly higher in benign prostate hyperplasia (BPH) than the normal prostate. Further, human normal prostate epithelial cells (PrECs), upon becoming senescent, activated an inflammasome. Because in aging related BPH senescent PrECs accumulate, we investigated the role of POP3 and AIM2 proteins in pre-senescent and senescent PrECs. Here we report that the basal levels of the POP3 mRNA and protein were lower in senescent (versus young or old) PrECs that exhibited activation of the T1 IFN response. Further, treatment of PrECs and a BPH cell line (BPH-1) that expresses the androgen receptor (AR) with the male sex hormone dihydrotestosterone (DHT) increased the basal levels of POP3 mRNA and protein, but not AIM2, and inhibited activation of the AIM2 inflammasome. Of interest, a stable knockdown of POP3 protein expression in the BPH-1 cell line increased cytosolic DNA-induced activation of AIM2 inflammasome. These observations suggest a potential role of POP3 protein in aging-related prostatic inflammation.

4.
Cytokine ; 132: 154682, 2020 08.
Article in English | MEDLINE | ID: mdl-30904426

ABSTRACT

Systemic lupus erythematosus (SLE) is a complex autoimmune disease that exhibits a strong female bias (female-to-male ratio 9:1) in patients. Further, 40-60% SLE patients develop lupus nephritis (LN), which significantly increases the mortality rates. The failure of current therapies to adequately treat LN in patients reflects an incomplete understanding of the disease pathogenesis. Notably, a chronic increase in serum interferon-α (IFN-α) activity is a heritable risk factor to develop SLE. Accordingly, blood cells from most SLE patients with an active disease exhibit an increase in the expression of the type I IFN (IFN-α/ß)-stimulated genes (ISGs, also referred to as "IFN-signature"), a type I IFN response. Further, LN patients during renal flares also exhibit an "IFN-signature" in renal biopsies. Therefore, an improved understanding of the regulation of type I IFNs expression is needed. Basal levels of the IFN-ß through "priming" of IFN-α producing cells augment the expression of the IFN-α genes. Of interest, recent studies have indicated a role for the type I IFN-inducible Absent in Melanoma 2 proteins (the murine Aim2 and human AIM2) in the negative regulation of the type I IFN response through inflammasome-dependent and independent mechanisms. Further, an increase in the expression of Aim2 and AIM2 proteins in kidney and renal macrophages associated with the development of nephritis. Therefore, we discuss the role of Aim2/AIM2 proteins in the regulation of type I IFNs and LN. An improved understanding of the mechanisms by which the Absent in Melanoma 2 proteins suppress the type I IFN response and modulate nephritis is key to identify novel therapeutic targets to treat a group of LN patients.


Subject(s)
DNA-Binding Proteins/physiology , Interferon Type I/metabolism , Lupus Nephritis/metabolism , Animals , B-Cell Activating Factor/metabolism , DNA/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Exodeoxyribonucleases/genetics , Female , Gene Expression Regulation , Humans , Interferon Regulatory Factors/genetics , Interferon Regulatory Factors/metabolism , Interferon-alpha/metabolism , Interferon-beta/metabolism , Male , Membrane Proteins/genetics , Mice , Phosphoproteins/genetics , Sex Characteristics
5.
J Neuroinflammation ; 16(1): 236, 2019 Nov 26.
Article in English | MEDLINE | ID: mdl-31771614

ABSTRACT

Cumulative evidence indicates that activation of innate immune responses in the central nervous system (CNS) induces the expression of type 1 interferons (T1 IFNs), a family of cytokines. The T1 IFNs (IFN-α/ß), through activation of the JAK/STAT-signaling in microglia, astrocytes, and neurons, induce the expression of IFN-inducible proteins, which mediate the pro- and anti-inflammatory functions of IFNs. Accordingly, T1 IFN-inducible Absent in Melanoma 2 proteins (murine Aim2 and human AIM2) negatively regulate the expression of TI IFNs and, upon sensing higher levels of cytosolic DNA, assemble the Aim2/AIM2 inflammasome, resulting in activation of caspase-1, pyroptosis, and the secretion of pro-inflammatory cytokines (e.g., IL-1ß and IL-18). Of interest, studies have indicated a role for the Aim2/AIM2 proteins in neuroinflammation and neurodegenerative diseases, including Alzheimer's disease (AD). The ability of Aim2/AIM2 proteins to exert pro- and anti-inflammatory effects in CNS may depend upon age, sex hormones, cell-types, and the expression of species-specific negative regulators of the Aim2/AIM2 inflammasome. Therefore, we discuss the role of Aim2/AIM2 proteins in the development of AD. An improved understanding of the role of Absent in Melanoma 2 proteins in AD could identify new approaches to treat patients.


Subject(s)
Alzheimer Disease/metabolism , Brain/metabolism , DNA-Binding Proteins/metabolism , Inflammation/metabolism , Neuroglia/metabolism , Neurons/metabolism , Alzheimer Disease/genetics , Animals , Cytokines/metabolism , DNA-Binding Proteins/genetics , Humans , Inflammasomes/metabolism , Inflammation/genetics , Signal Transduction/physiology
6.
Clin Immunol ; 176: 42-48, 2017 03.
Article in English | MEDLINE | ID: mdl-28062222

ABSTRACT

Type I interferons (IFN-α/ß)-inducible PYRIN and HIN domain-containing protein family includes Absent in Melanoma 2 (murine Aim2 and human AIM2), murine p202, and human PYRIN-only protein 3 (POP3). The generation of Aim2-deficient mice indicated that the Aim2 protein is essential for inflammasome activation, resulting in the secretion of interleukin-1ß (IL-1ß) and IL-18 and cell death by pyroptosis. Further, Aim2-deficiency also increased constitutive expression of the IFN-ß and expression of the p202 protein. Notably, an increased expression of p202 protein in female mice associated with the development of systemic lupus erythematosus (SLE). SLE in patients is characterized by a constitutive increase in serum levels of IFN-α and an increase in the expression IFN-stimulated genes. Recent studies indicate that p202 and POP3 proteins inhibit activation of the Aim2/AIM2 inflammasome and promote IFN-ß expression. Therefore, we discuss the role of Aim2/AIM2 proteins in the suppression of type I IFNs production and lupus susceptibility.


Subject(s)
DNA-Binding Proteins/metabolism , Lupus Erythematosus, Systemic/metabolism , Melanoma/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Animals , Humans , Interferon Type I/metabolism , Interleukin-18/metabolism , Interleukin-1beta/metabolism , Tumor Suppressor p53-Binding Protein 1/metabolism
7.
Cell Mol Life Sci ; 73(23): 4383-4395, 2016 12.
Article in English | MEDLINE | ID: mdl-27328971

ABSTRACT

Recent studies utilizing chemical-induced colitis-associated and sporadic colon cancer in mouse models indicated a protective role for absent in melanoma 2 (Aim2) in colon epithelial cells. Accordingly, mutations in the human AIM2 gene have been found in colorectal cancer (CRC), and reduced expression of AIM2 in CRC is associated with its progression. Furthermore, the overexpression of AIM2 protein in human cancer cell lines inhibits cell proliferation. Interferon-inducible Aim2 and AIM2 are members of the PYHIN (PYRIN and HIN domain-containing) protein family and share ~57 % amino acid identity. The family also includes murine p202, human PYRIN-only protein 3, and IFI16, which negatively regulate Aim2/AIM2 functions. Because the CRC incidence and mortality rates are higher among men compared with women and the expression of Aim2/AIM2 proteins and their regulators is dependent upon age, gender, and sex hormones, we discuss the potential roles of Aim2/AIM2 in the development of cancer. An improved understanding of the biological functions of the AIM2 in the development of CRC will likely identify new therapeutic approaches to treat patients.


Subject(s)
Carcinogenesis/metabolism , Carcinogenesis/pathology , DNA-Binding Proteins/metabolism , Neoplasms/metabolism , Neoplasms/pathology , Animals , Cell Proliferation , Colitis/pathology , Humans , Protein Multimerization
8.
Toxicol In Vitro ; 35: 24-30, 2016 Sep.
Article in English | MEDLINE | ID: mdl-27179668

ABSTRACT

Zinc is both an essential and potentially toxic metal. It is widely believed that oral zinc supplementation can reduce the effects of the common cold; however, there is strong clinical evidence that intranasal (IN) zinc gluconate (ZG) gel treatment for this purpose causes anosmia, or the loss of the sense of smell, in humans. Using the rat olfactory neuron cell line, Odora, we investigated the molecular mechanism by which zinc exposure exerts its toxic effects on olfactory neurons. Following treatment of Odora cells with 100 and 200µM ZG for 0-24h, RNA-seq and in silico analyses revealed up-regulation of pathways associated with zinc metal response, oxidative stress, and ATP production. We observed that Odora cells recovered from zinc-induced oxidative stress, but ATP depletion persisted with longer exposure to ZG. ZG exposure increased levels of NLRP3 and IL-1ß protein levels in a time-dependent manner, suggesting that zinc exposure may cause an inflammasome-mediated cell death, pyroptosis, in olfactory neurons.


Subject(s)
Gluconates/toxicity , Neurons/drug effects , Olfactory Mucosa/cytology , Acetophenones/pharmacology , Acetylcysteine/pharmacology , Adenosine Triphosphate/metabolism , Animals , Antioxidants/pharmacology , Apoptosis/drug effects , Caspase 1/metabolism , Caspase Inhibitors/pharmacology , Cell Line , Cell Survival/drug effects , Glutathione/metabolism , Glutathione Disulfide/metabolism , Hydrogen Peroxide/metabolism , Interleukin-1beta/metabolism , NADPH Oxidases/antagonists & inhibitors , NG-Nitroarginine Methyl Ester/pharmacology , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Neurons/metabolism , Nitric Oxide Synthase/antagonists & inhibitors , Rats
9.
Ageing Res Rev ; 28: 27-36, 2016 07.
Article in English | MEDLINE | ID: mdl-27063514

ABSTRACT

DNA-damage induces a DNA-damage response (DDR) in mammalian cells. The response, depending upon the cell-type and the extent of DNA-damage, ultimately results in cell death or cellular senescence. DDR-induced signaling in cells activates the ATM-p53 and ATM-IKKα/ß-interferon (IFN)-ß signaling pathways, thus leading to an induction of the p53 and IFN-inducible IFI16 gene. Further, upon DNA-damage, DNA accumulates in the cytoplasm, thereby inducing the IFI16 protein and STING-dependent IFN-ß production and activation of the IFI16 inflammasome, resulting in the production of proinflammatory cytokines (e.g., IL-1ß and IL-18). Increased expression of IFI16 protein in a variety of cell-types promotes cellular senescence. However, reduced expression of IFI16 in cells promotes cell proliferation. Because expression of the IFI16 gene is induced by activation of DNA-damage response in cells and increased levels of IFI16 protein in cells potentiate the p53-mediated transcriptional activation of genes and p53 and pRb-mediated cell cycle arrest, we discuss how an improved understanding of the role of IFI16 protein in cellular senescence and associated inflammatory secretory phenotype is likely to identify the molecular mechanisms that contribute to the development of aging-associated human inflammatory diseases and a failure to cancer therapy.


Subject(s)
Aging/metabolism , Cellular Senescence/genetics , DNA Damage/genetics , Nuclear Proteins/metabolism , Phosphoproteins/metabolism , Tumor Suppressor Protein p53/metabolism , Aging/genetics , Humans , Inflammasomes/metabolism , Signal Transduction , Transcriptional Activation
10.
Oncotarget ; 7(19): 28183-94, 2016 May 10.
Article in English | MEDLINE | ID: mdl-27058421

ABSTRACT

The molecular mechanisms by which hypoxia contributes to prostatic chronic inflammation (PCI) remain largely unknown. Because hypoxia stimulates the transcriptional activity of NF-κB, which "primes" cells for inflammasome activation by inducing the expression of NLRP3 or AIM2 receptor and pro-IL-1ß, we investigated whether hypoxia could activate the NLRP3 and AIM2 inflammasome in human normal prostate epithelial cells (PrECs) and cancer cell lines. Here we report that hypoxia (1% O2) treatment of PrECs, prostate cell lines, and a macrophage cell line (THP-1) increased the levels of NLRP3, AIM2, and pro-IL-1ß. Further, hypoxia in cells potentiated activation of the NLRP3 and AIM2 inflammasome activity. Notably, hypoxia "primed" cells for NLRP3 and AIM2 inflammasome activation through stimulation of the NF-κB activity. Our observations support the idea that hypoxia in human prostatic tumors contributes to PCI, in part, by priming cells for the activation of NLRP3 and AIM2 inflammasome.


Subject(s)
Epithelial Cells/pathology , Inflammasomes/metabolism , Prostate/pathology , Prostatic Neoplasms/pathology , Cell Hypoxia/physiology , Cell Line, Tumor , DNA-Binding Proteins/metabolism , Epithelial Cells/metabolism , Humans , Male , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Prostatic Neoplasms/complications , Prostatic Neoplasms/metabolism , Prostatitis/etiology , Prostatitis/metabolism , Prostatitis/pathology
12.
Mol Cell Endocrinol ; 415: 45-55, 2015 Nov 05.
Article in English | MEDLINE | ID: mdl-26277401

ABSTRACT

Environmental factors contribute to the development of autoimmune diseases, including systemic lupus erythematosus (SLE), which exhibits a strong female bias (female-to-male ratio 9:1). However, the molecular mechanisms remain largely unknown. Because a feedforward loop between the female sex hormone estrogen (E2) and type I interferon (IFN-α/ß)-signaling induces the expression of certain p200-family proteins (such as murine p202 and human IFI16) that regulate innate immune responses and modify lupus susceptibility, we investigated whether treatment of myeloid cells with bisphenol A (BPA), an environmental estrogen, could regulate the p200-family proteins and activate innate immune responses. We found that treatment of murine bone marrow-derived cells (BMCs) and human peripheral blood mononuclear cells with BPA induced the expression of ERα and IFN-ß, activated the IFN-signaling, and stimulated the expression of the p202 and IFI16 proteins. Further, the treatment increased levels of the NLRP3 inflammasome and stimulated its activity. Accordingly, BPA-treatment of BMCs from non lupus-prone C57BL/6 and the lupus-prone (NZB×NZW)F1 mice activated the type I IFN-signaling, induced the expression of p202, and activated an inflammasome activity. Our study demonstrates that BPA-induced signaling in the murine and human myeloid cells stimulates the type I IFN-signaling that results in an induction of the p202 and IFI16 innate immune sensors for the cytosolic DNA and activates an inflammasome activity. These observations provide novel molecular insights into the role of environmental BPA exposures in potentiating the development of certain autoimmune diseases such as SLE.


Subject(s)
Benzhydryl Compounds/pharmacology , Estrogen Receptor alpha/genetics , Inflammasomes/metabolism , Interferons/genetics , Myeloid Cells/drug effects , Phenols/pharmacology , Animals , Cell Line , Female , Gene Expression Regulation/drug effects , Humans , Immunity, Innate/drug effects , Inflammasomes/drug effects , Lupus Erythematosus, Systemic/genetics , Male , Mice , Myeloid Cells/cytology , Signal Transduction/drug effects
13.
Toxicol Lett ; 237(3): 228-36, 2015 Sep 17.
Article in English | MEDLINE | ID: mdl-26149760

ABSTRACT

We previously reported that NR2E3, an orphan nuclear receptor, plays an important role in maintaining the basal expression of estrogen receptor α (ER) and that the NR2E3 level is highly correlated with the relapse-free survival of breast cancer patients. Here, we investigated the role of NR2E3 in benzo(a)pyrene (BaP)-mediated cell injury. BaP treatment reduced NR2E3 homo-dimer formation and expression and subsequently decreased ER expression. The chromatin immunoprecipitation assay results showed that the treatment of MCF-7 breast cancer cells and the mouse liver with BaP released NR2E3 from the ER promoter to transform the transcriptionally active histone modification status into a repressive state. NR2E3 depletion in MCF-7 cells also induced a similar inactive epigenetic status in the ER promoter region, indicating that NR2E3 is an essential epigenetic player that maintains basal ER expression. Interestingly, these negative effects of BaP on the expression levels of NR2E3 and ER were rescued by antioxidant treatment. Collectively, our study provides novel evidence to show that BaP-induced oxidative stress decreases ER expression, in part by regulating NR2E3 function, which modulates the epigenetic status of the ER promoter. NR2E3 is likely an essential epigenetic player that maintains basal ER expression to protect cells from BaP-induced oxidative injury.


Subject(s)
Benzo(a)pyrene/toxicity , Orphan Nuclear Receptors/metabolism , Oxidative Stress/drug effects , Promoter Regions, Genetic , Receptors, Estrogen/genetics , Animals , Antioxidants/pharmacology , Down-Regulation , Epigenesis, Genetic , Gene Expression Regulation, Neoplastic , Hep G2 Cells , Histones/genetics , Histones/metabolism , Humans , MCF-7 Cells , Male , Mice , Mice, Inbred C57BL , Orphan Nuclear Receptors/genetics
14.
Int J Toxicol ; 34(3): 242-9, 2015.
Article in English | MEDLINE | ID: mdl-25878193

ABSTRACT

Aryl hydrocarbon receptor (AhR) is a ligand-activated transcription factor that regulates multiple cellular processes. The anticancer drug doxorubicin (DOX) can activate AhR-mediated transcription of target genes. Because DOX in cells activates a DNA damage response involving ataxia telangiectasia-mutated (ATM)-mediated activation of p53, we investigated whether the activation of the p53 in cells by DNA-damaging agents such as DOX or bleomycin could regulate the AhR levels. Here we report that activation of p53 by DNA-damaging agents in human cells increased levels of AhR through a posttranscriptional mechanism. Accordingly, fibroblasts from ATM patients, which are defective in p53 activation, expressed reduced constitutive levels of AhR and treatment of cells with bleomycin did not appreciably increase the AhR levels. Further, activation of p53 in cells stimulated the expression of AhR target genes. In murine cells, activation of p53 reduced the levels of AhR messenger RNA and protein and reduced the expression of AhR target genes. Our observations revealed that activation of p53 in human and murine cells differentially regulates AhR levels.


Subject(s)
Antibiotics, Antineoplastic/pharmacology , Basic Helix-Loop-Helix Transcription Factors/agonists , Basic Helix-Loop-Helix Transcription Factors/antagonists & inhibitors , Mutagens/toxicity , Receptors, Aryl Hydrocarbon/agonists , Receptors, Aryl Hydrocarbon/antagonists & inhibitors , Teratogens/toxicity , Tumor Suppressor Protein p53/agonists , Animals , Antibiotics, Antineoplastic/adverse effects , Basic Helix-Loop-Helix Transcription Factors/chemistry , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Line , Cell Line, Transformed , Cell Line, Tumor , Fibroblasts/drug effects , Fibroblasts/metabolism , Gene Expression Regulation/drug effects , Humans , Ligands , Macrophages/drug effects , Macrophages/metabolism , Mice , Mutant Proteins/agonists , Mutant Proteins/metabolism , Osteosarcoma/drug therapy , Osteosarcoma/metabolism , Protein Stability/drug effects , Receptors, Aryl Hydrocarbon/chemistry , Receptors, Aryl Hydrocarbon/genetics , Receptors, Aryl Hydrocarbon/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Species Specificity , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
15.
Nat Rev Rheumatol ; 10(12): 740-51, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25155581

ABSTRACT

Sexual dimorphism is evident in the risk and expression of several human autoimmune diseases. Differences in disease manifestations observed between sexes are likely to involve immunomodulation by sex steroids, nonhormonal factors encoded by genes on the X and Y chromosomes, and immunological phenomena unique to pregnancy. In systemic lupus erythematosus (SLE), and perhaps other autoantibody-mediated diseases, oestrogen seems to increase the risk of disease in genetically predisposed women by targeting key immune pathways, including the type 1 interferon (IFN) response, differentiation of CD4(+) T helper cells and survival of autoreactive B cells. By contrast, progesterone seems to reduce the risk of SLE by counteracting the effects of oestrogen on some of these same pathways, which suggests that the balance between oestrogen and progesterone can determine disease expression. In this Review we focus on the roles of the sex steroid hormones oestrogen and progesterone in modulating the risk and expression of SLE and rheumatoid arthritis. Intensive research in this area promises to identify novel therapeutic strategies and improve understanding of the immunological requirements and complications of pregnancy, and is expected to define the mechanisms behind sexual dimorphism in autoimmunity, immunity and other aspects of human health--a newly announced directive of the NIH.


Subject(s)
Arthritis, Rheumatoid/etiology , Autoimmune Diseases/etiology , Estrogens/physiology , Lupus Erythematosus, Systemic/etiology , Progesterone/physiology , Autoantibodies/physiology , B-Lymphocytes/physiology , Humans , Interferons/physiology , T-Lymphocytes/physiology
16.
Innate Immun ; 20(7): 751-9, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24131791

ABSTRACT

A host type I IFN response is induced by cytosolic sensing of the bacterial second messenger cyclic-di-GMP (c-di-GMP) by STING (stimulator of IFN genes). Because the STING, an adaptor protein, links the cytosolic detection of DNA by the cytosolic DNA sensors such as the IFN-inducible human IFI16 and murine p202 proteins to the TBK1/IRF3 axis, we investigated whether c-di-GMP-induced signaling could regulate expression of IFI16 and p202 proteins. Here, we report that activation of c-di-GMP-induced signaling in human and murine cells increased steady-state levels of IFI16 and p202 proteins. The increase was c-di-GMP concentration- and time-dependent. Unexpectedly, treatment of cells with type I IFN decreased levels of the adaptor protein STING. Therefore, we investigated whether the IFI16 or p202 protein could regulate the expression of STING and activation of the TBK1/IRF3 axis. We found that constitutive knockdown of IFI16 or p202 expression in cells increased steady-state levels of STING. Additionally, the knockdown of IFI16 resulted in activation of the TBK1/IRF3 axis. Accordingly, increased levels of the IFI16 or p202 protein in cells decreased STING levels. Together, our observations identify a novel negative feedback loop between c-di-GMP-induced levels of IFI16 and p202 cytosolic DNA sensors and the adaptor protein STING.


Subject(s)
Cyclic GMP/analogs & derivatives , Cytosol/metabolism , DNA/metabolism , Feedback, Physiological/drug effects , Intracellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/drug effects , Nuclear Proteins/drug effects , Phosphoproteins/drug effects , Animals , Cell Line , Cyclic GMP/pharmacology , Gene Knockdown Techniques , Humans , Intracellular Signaling Peptides and Proteins/drug effects , Intracellular Signaling Peptides and Proteins/genetics , Mice , Mice, Inbred C57BL , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Phosphoproteins/genetics , Phosphoproteins/metabolism , Signal Transduction/drug effects , Tumor Suppressor p53-Binding Protein 1
17.
Mol Cancer Res ; 11(10): 1193-202, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23864729

ABSTRACT

UNLABELLED: Close links have been noted between chronic inflammation of the prostate and the development of human prostatic diseases such as benign prostate hyperplasia (BPH) and prostate cancer. However, the molecular mechanisms that contribute to prostatic inflammation remain largely unexplored. Recent studies have indicated that the IFN-inducible AIM2 protein is a cytosolic DNA sensor in macrophages and keratinocytes. Upon sensing DNA, AIM2 recruits the adaptor ASC and pro-CASP1 to assemble the AIM2 inflammasome. Activation of the AIM2 inflammasome cleaves pro-interleukin (IL)-1ß and pro-IL-18 and promotes the secretion of IL-1ß and IL-18 proinflammatory cytokines. Given that human prostatic infections are associated with chronic inflammation, the development of BPH is associated with an accumulation of senescent cells with a proinflammatory phenotype, and the development of prostate cancer is associated with the loss of IFN signaling, the role of AIM2 in mediating the formation of prostatic diseases was investigated. It was determined that IFNs (α, ß, or γ) induced AIM2 expression in human prostate epithelial cells and cytosolic DNA activated the AIM2 inflammasome. Steady-state levels of the AIM2 mRNA were higher in BPH than in normal prostate tissue. However, the levels of AIM2 mRNA were significantly lower in clinical tumor specimens. Accordingly, constitutive levels of AIM2 mRNA and protein were lower in a subset of prostate cancer cells as compared with BPH cells. Further, the cytosolic DNA activated the AIM2 inflammasome in the androgen receptor-negative PC3 prostate cancer cell line, suggesting that AIM2-mediated events are independent of androgen receptor status. IMPLICATIONS: The AIM2 inflammasome has a fundamental role in the generation of human prostatic diseases.


Subject(s)
Adenocarcinoma/metabolism , Cell Transformation, Neoplastic , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Prostatic Hyperplasia/metabolism , Prostatic Neoplasms/metabolism , Adenocarcinoma/genetics , Adenocarcinoma/pathology , Cell Line, Tumor , Cytosol/metabolism , DNA/metabolism , DNA-Binding Proteins , Gene Expression Regulation, Neoplastic/drug effects , Humans , Inflammasomes/drug effects , Inflammasomes/genetics , Inflammasomes/metabolism , Interferon-gamma/pharmacology , Male , Prostatic Hyperplasia/genetics , Prostatic Hyperplasia/pathology , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology
18.
Cell Rep ; 4(2): 327-39, 2013 Jul 25.
Article in English | MEDLINE | ID: mdl-23850291

ABSTRACT

Mouse p202 containing two hemopoietic expression, interferon inducibility, nuclear localization (HIN) domains antagonizes AIM2 inflammasome signaling and potentially modifies lupus susceptibility. We found that only HIN1 of p202 binds double-stranded DNA (dsDNA), while HIN2 forms a homotetramer. Crystal structures of HIN1 revealed that dsDNA is bound on face opposite the site used in AIM2 and IFI16. The structure of HIN2 revealed a dimer of dimers, the face analogous to the HIN1 dsDNA binding site being a dimerization interface. Electron microscopy imaging showed that HIN1 is flexibly linked to HIN2 in p202, and tetramerization provided enhanced avidity for dsDNA. Surprisingly, HIN2 of p202 interacts with the AIM HIN domain. We propose that this results in a spatial separation of the AIM2 pyrin domains, and indeed p202 prevented the dsDNA-dependent clustering of apoptosis-associated speck-like protein containing caspase recruitment domain (ASC) and AIM2 inflammasome activation. We hypothesize that while p202 was evolutionarily selected to limit AIM2-mediated inflammation in some mouse strains, the same mechanism contributes to increased interferon production and lupus susceptibility.


Subject(s)
Inflammasomes/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Nuclear Proteins/metabolism , Amino Acid Sequence , Animals , DNA-Binding Proteins , Enzyme Activation , Humans , Intracellular Signaling Peptides and Proteins/genetics , Mice , Models, Molecular , Molecular Sequence Data , Nuclear Proteins/genetics , Protein Structure, Tertiary , Signal Transduction
19.
Int Immunol ; 25(9): 521-9, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23728775

ABSTRACT

The endoplasmic reticulum transmembrane protein, Unc93b1, is essential for trafficking of endosomal TLRs from the endoplasmic reticulum to endosomes. A genetic defect in the human UNC93B1 gene is associated with immunodeficiency. However, systemic lupus erythematosus (SLE) patients express increased levels of the UNC93B1 protein in B cells. Because SLE in patients and certain mouse models exhibits a sex bias and increased serum levels of type I interferons in patients are associated with the disease activity, we investigated whether the female sex hormone estrogen (E2) or type I interferon signaling could up-regulate the expression of the murine Unc93b1 gene. We found that steady-state levels of Unc93b1 mRNA and protein were measurably higher in immune cells (CD3(+), B220(+), CD11b(+) and CD11c(+)) isolated from C57BL/6 (B6) females than age-matched males. Moreover, treatment of CD11b(+) and B220(+) cells with E2 or interferons (IFN-α, IFN-ß or IFN-γ) significantly increased the levels of Unc93b1 mRNA and protein. Accordingly, a deficiency of estrogen receptor-α or STAT1 expression in immune cells decreased the expression levels of the Unc93b1 protein. Interestingly, levels of Unc93b1 protein were appreciably higher in B6.Nba2 lupus-prone female mice compared with age-matched B6 females. Furthermore, increased expression of the interferon- and E2-inducible p202 protein in a murine macrophage cell line (RAW264.7) increased the levels of the Unc93b1 protein, whereas knockdown of p202 expression reduced the levels. To our knowledge, our observations demonstrate for the first time that activation of interferon and estrogen signaling in immune cells up-regulates the expression of murine Unc93b1.


Subject(s)
Autoimmunity , Estrogens/metabolism , Interferons/immunology , Membrane Transport Proteins/genetics , Sex Characteristics , Signal Transduction , Up-Regulation , Animals , Female , Male , Membrane Transport Proteins/biosynthesis , Mice , Mice, Inbred C57BL
20.
Mol Immunol ; 53(1-2): 15-23, 2013 Jan.
Article in English | MEDLINE | ID: mdl-22784990

ABSTRACT

Systemic lupus erythematosus (SLE) in patients and certain mouse models exhibits a strong sex bias. Additionally, in most patients, increased serum levels of type I interferon (IFN-α) are associated with severity of the disease. Because increased levels of B cell activating factor (BAFF) in SLE patients and mouse models are associated with the development of SLE, we investigated whether the female sex hormone estrogen (E2) and/or IFNs (IFN-α or γ) could regulate the expression of murine BAFF. We found that steady-state levels of BAFF mRNA and protein were measurably higher in immune cells (CD11b(+), CD11c(+), and CD19(+)) isolated from C57BL/6 females than the age-matched male mice. Treatment of immune cells with IFN or E2 significantly increased levels of BAFF mRNA and protein and a deficiency of estrogen receptor-α, IRF5, or STAT1 expression in splenic cells decreased expression of BAFF. Moreover, treatment of RAW264.7 macrophage cells with IFN-α, IFN-γ, or E2 induced expression of BAFF. Interestingly, increased expression of p202, an IFN and estrogen-inducible protein, in RAW264.7 cells significantly increased the expression levels of BAFF and also stimulated the activity of the BAFF-luc-reporter. Accordingly, the increased expression of the p202 protein in lupus-prone B6.Nba2-ABC than non lupus-prone C57BL/6 and B6.Nba2-C female mice was associated with increased expression levels of BAFF. Together, our observations demonstrated that estrogen and IFN-induced increased levels of the p202 protein in immune cells contribute to sex bias in part through up-regulation of BAFF expression.


Subject(s)
Autoimmunity/immunology , B-Cell Activating Factor/immunology , Estrogens/immunology , Interferons/immunology , Intracellular Signaling Peptides and Proteins/immunology , Sex Characteristics , Animals , Autoimmune Diseases/immunology , Autoimmune Diseases/metabolism , B-Cell Activating Factor/metabolism , Female , Immunoblotting , Intracellular Signaling Peptides and Proteins/metabolism , Male , Mice , Mice, Inbred C57BL , RNA, Messenger/analysis , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/immunology , Up-Regulation
SELECTION OF CITATIONS
SEARCH DETAIL
...