Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 130
1.
Nature ; 623(7988): 803-813, 2023 Nov.
Article En | MEDLINE | ID: mdl-37938781

Patients with autoimmune polyendocrinopathy syndrome type 1 (APS-1) caused by autosomal recessive AIRE deficiency produce autoantibodies that neutralize type I interferons (IFNs)1,2, conferring a predisposition to life-threatening COVID-19 pneumonia3. Here we report that patients with autosomal recessive NIK or RELB deficiency, or a specific type of autosomal-dominant NF-κB2 deficiency, also have neutralizing autoantibodies against type I IFNs and are at higher risk of getting life-threatening COVID-19 pneumonia. In patients with autosomal-dominant NF-κB2 deficiency, these autoantibodies are found only in individuals who are heterozygous for variants associated with both transcription (p52 activity) loss of function (LOF) due to impaired p100 processing to generate p52, and regulatory (IκBδ activity) gain of function (GOF) due to the accumulation of unprocessed p100, therefore increasing the inhibitory activity of IκBδ (hereafter, p52LOF/IκBδGOF). By contrast, neutralizing autoantibodies against type I IFNs are not found in individuals who are heterozygous for NFKB2 variants causing haploinsufficiency of p100 and p52 (hereafter, p52LOF/IκBδLOF) or gain-of-function of p52 (hereafter, p52GOF/IκBδLOF). In contrast to patients with APS-1, patients with disorders of NIK, RELB or NF-κB2 have very few tissue-specific autoantibodies. However, their thymuses have an abnormal structure, with few AIRE-expressing medullary thymic epithelial cells. Human inborn errors of the alternative NF-κB pathway impair the development of AIRE-expressing medullary thymic epithelial cells, thereby underlying the production of autoantibodies against type I IFNs and predisposition to viral diseases.


Autoantibodies , Genetic Predisposition to Disease , Interferon Type I , NF-kappa B , Humans , Autoantibodies/immunology , COVID-19/genetics , COVID-19/immunology , Gain of Function Mutation , Heterozygote , I-kappa B Proteins/deficiency , I-kappa B Proteins/genetics , Interferon Type I/antagonists & inhibitors , Interferon Type I/immunology , Loss of Function Mutation , NF-kappa B/deficiency , NF-kappa B/genetics , NF-kappa B p52 Subunit/deficiency , NF-kappa B p52 Subunit/genetics , Pneumonia, Viral/genetics , Pneumonia, Viral/immunology , Thymus Gland/abnormalities , Thymus Gland/immunology , Thymus Gland/pathology , Thyroid Epithelial Cells/metabolism , Thyroid Epithelial Cells/pathology , AIRE Protein , NF-kappaB-Inducing Kinase
2.
Cells ; 10(8)2021 08 10.
Article En | MEDLINE | ID: mdl-34440811

NF-κB (nuclear factor kappa B) belongs to a family of transcription factors known to regulate a broad range of processes such as immune cell function, proliferation and cancer, neuroprotection, and long-term memory. Upcoming fields of NF-κB research include its role in stem cells and developmental processes. In the present review, we discuss one role of NF-κB in development in Drosophila, Xenopus, mice, and humans in accordance with the concept of evo-devo (evolutionary developmental biology). REL domain-containing proteins of the NF-κB family are evolutionarily conserved among these species. In addition, we summarize cellular phenotypes such as defective B- and T-cell compartments related to genetic NF-κB defects detected among different species. While NF-κB proteins are present in nearly all differentiated cell types, mouse and human embryonic stem cells do not contain NF-κB proteins, potentially due to miRNA-dependent inhibition. However, the mesodermal and neuroectodermal differentiation of mouse and human embryonic stem cells is hampered upon the repression of NF-κB. We further discuss NF-κB as a crucial regulator of differentiation in adult stem cells such as neural crest-derived and mesenchymal stem cells. In particular, c-REL seems to be important for neuronal differentiation and the neuroprotection of human adult stem cells, while RELA plays a crucial role in osteogenic and mesodermal differentiation.


NF-kappa B/metabolism , Stem Cells/metabolism , Adult Stem Cells/cytology , Adult Stem Cells/metabolism , Animals , Cell Differentiation , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Humans , MicroRNAs/metabolism , NF-kappa B/deficiency , NF-kappa B/genetics , Signal Transduction , Stem Cells/cytology , Transcription Factor RelA/metabolism
3.
Front Immunol ; 11: 580974, 2020.
Article En | MEDLINE | ID: mdl-33262759

Novel adjuvants, such as Toll-like receptors (TLRs) agonists, are needed for the development of new formulations able to circumvent limitations of current vaccines. Among TLRs, TLR7/8 agonists represent promising candidates, as they are well described to enhance antigen-specific antibody responses and skew immunity toward T helper (TH) 1 responses. We find here that the incorporation of the synthetic TLR7/8 ligand 3M-052 in a cationic DOEPC-based liposome formulation shifts immunity toward TH1 responses and elicits strong and long-lasting germinal center and follicular T helper cell responses in adult mice. This reflects the prolonged recruitment of innate cells toward the site of immunization and homing of activated antigen-loaded monocytes and monocyte-derived dendritic cells toward draining lymph nodes. We further show that this adjuvanticity is independent of type I IFN but NF-κB-dependent. Overall, our data identify TLR7/8 agonists incorporated in liposomes as promising and effective adjuvants to enhance TH1 and germinal center responses.


Adjuvants, Immunologic/administration & dosage , Membrane Glycoproteins/agonists , Monocytes/immunology , Toll-Like Receptor 7/agonists , Toll-Like Receptor 8/agonists , Animals , B-Lymphocytes/immunology , Dendritic Cells/immunology , Drug Compounding , Germinal Center/immunology , Heterocyclic Compounds, 3-Ring/administration & dosage , Immunity, Innate , Interferon Type I/immunology , Ligands , Liposomes/administration & dosage , Mice , Mice, Inbred C57BL , Mice, Knockout , NF-kappa B/deficiency , NF-kappa B/genetics , NF-kappa B/immunology , Phosphatidylcholines/administration & dosage , Receptor, Interferon alpha-beta/deficiency , Receptor, Interferon alpha-beta/genetics , Receptor, Interferon alpha-beta/immunology , Signal Transduction/immunology , Stearic Acids/administration & dosage , Th1 Cells/immunology
4.
Artif Cells Nanomed Biotechnol ; 47(1): 3677-3686, 2019 Dec.
Article En | MEDLINE | ID: mdl-31841039

AMP-activated alpha 1 catalytic subunit (PRKAA1) is one of the subunits of the mammalian 5'-AMP-activated protein kinase (AMPK) playing an important role in maintaining intracellular energy metabolism and associating with the risk of gastric cancer (GC). This paper aims to uncover the influences of PRKAA1 on the tumorigenesis of GC, as well as the underlying mechanisms. We found that Helicobacter pylori (H. pylori) infection markedly increased p-NF-κBp50 and NF-κBp50 expression, along with the PRKAA1 expression, which was inhibited by NF-κBp50 knockdown. NF-κBp50 and PRKAA1 expression were lower in non-tumor gastric tissues compared with that in GC tumor tissues. Up-regulation of PRKAA1 expression was correlated with poor survival in GC patients. MKN-45 and BGC-823 cells stably knockdown of PRKAA1 were transplanted into nude mice and observed the decreased cell metastasis in the lungs. PRKAA1 knockdown in GC cells showed significant decreases in the cell invasion and migration and inhibited MMP-2 expression and NF-κB activation, whereas PRKAA1 involved in NF-κBp50 mediated GC cell invasion and migration. In conclusion, our findings suggest the involvement of NF-κBp50 in the regulation of PRKAA1 in GC tumorigenesis.


AMP-Activated Protein Kinases/metabolism , Carcinogenesis , NF-kappa B/metabolism , Signal Transduction , Stomach Neoplasms/pathology , Animals , Cell Line, Tumor , Cell Movement , Cell Transformation, Neoplastic , Female , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Humans , Male , Mice , Middle Aged , NF-kappa B/deficiency , NF-kappa B/genetics , Neoplasm Invasiveness , Neoplasm Metastasis , Stomach Neoplasms/metabolism
5.
Curr Opin Infect Dis ; 32(4): 330-336, 2019 08.
Article En | MEDLINE | ID: mdl-31116133

PURPOSE OF REVIEW: The current review gives a concise and updated overview of the relative new field of anticytokine autoantibodies (ACAA) and associated infections with a focus on recent findings regarding clinical manifestions, diagnostic and treatments. RECENT FINDINGS: Several recent case reports of unusual presentations of patients with neutralizing autoantibodies to IFN-γ and granulocyt macrophage colony-stimulating factor and expand the spectrum of clinical manifestations and suggest that anticytokine-mediated acquired immunodeficiency causing susceptibility to infection may be underdiagnosed. There is an expanding geographical distribution of antigranulocyt macrophage colony-stimulating factor associated Cryptococcus gattii infection. The spectrum of identified infections in patients with neutralizing antibodies to IFN-γ has a strong endemic component. Rituximab or cyclophophamide in addition to antimycobacterials could be a treatment options in refractory cases. NF-κB2 deficiency may be associated with a complex pattern of high titre neutralizing ACAA similar to autoimmune polyglandular syndrome type I and Thymoma. New technique for the detection of anticytokine antibodies are presented. Quantiferon testing, which is widely available for TB-diagnostic, may be repurposed to detect anti-IFN-γ autoantibodies. We propose that this test could be as well used to show if they are neutralizing. SUMMARY: ACAA are an emerging cause of acquired immunodeficiency which is likely underdiagnosed. Recent case reports document expanding spectra of clinical manifestations. NF-κB2 deficiency may be associated with a complex anti cytokine autoantibody pattern.


Autoantibodies/adverse effects , Cytokines/antagonists & inhibitors , Disease Susceptibility , Infections/diagnosis , Infections/etiology , Infections/therapy , Antibodies, Neutralizing/immunology , Autoantibodies/immunology , Cytokines/immunology , Disease Susceptibility/immunology , Genetic Predisposition to Disease , Granulocyte Colony-Stimulating Factor/antagonists & inhibitors , Granulocyte Colony-Stimulating Factor/immunology , Humans , Interferon-gamma/antagonists & inhibitors , Interferon-gamma/immunology , Mycobacterium Infections/diagnosis , Mycobacterium Infections/etiology , Mycobacterium Infections/therapy , NF-kappa B/deficiency , Opportunistic Infections/diagnosis , Opportunistic Infections/etiology , Opportunistic Infections/therapy , Severity of Illness Index
6.
Nature ; 568(7751): 249-253, 2019 04.
Article En | MEDLINE | ID: mdl-30894749

The non-canonical NF-κB signalling cascade is essential for lymphoid organogenesis, B cell maturation, osteoclast differentiation, and inflammation in mammals1,2; dysfunction of this system is associated with human diseases, including immunological disorders and cancer3-6. Although expression of NF-κB-inducing kinase (NIK, also known as MAP3K14) is the rate-limiting step in non-canonical NF-κB pathway activation2,7, the mechanisms by which transcriptional responses are regulated remain largely unknown. Here we show that the sine oculis homeobox (SIX) homologue family transcription factors SIX1 and SIX2 are integral components of the non-canonical NF-κB signalling cascade. The developmentally silenced SIX proteins are reactivated in differentiated macrophages by NIK-mediated suppression of the ubiquitin proteasome pathway. Consequently, SIX1 and SIX2 target a subset of inflammatory gene promoters and directly inhibit the trans-activation function of the transcription factors RELA and RELB in a negative feedback circuit. In support of a physiologically pivotal role for SIX proteins in host immunity, a human SIX1 transgene suppressed inflammation and promoted the recovery of mice from endotoxic shock. In addition, SIX1 and SIX2 protected RAS/P53-driven non-small-cell lung carcinomas from inflammatory cell death induced by SMAC-mimetic chemotherapeutic agents (small-molecule activators of the non-canonical NF-κB pathway). Our findings identify a NIK-SIX signalling axis that fine-tunes inflammatory gene expression programs under both physiological and pathological conditions.


Homeodomain Proteins/metabolism , Inflammation/metabolism , NF-kappa B/deficiency , NF-kappa B/metabolism , Nerve Tissue Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Signal Transduction , Animals , Antineoplastic Agents/pharmacology , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Tumor , Female , Fibroblasts , Gene Expression Regulation/drug effects , HEK293 Cells , Homeodomain Proteins/immunology , Humans , Inflammation/genetics , Listeria monocytogenes/immunology , Male , Mice , NF-kappa B/genetics , Nerve Tissue Proteins/immunology , Promoter Regions, Genetic , Shigella flexneri/immunology , Signal Transduction/drug effects , Signal Transduction/genetics , Transcription Factor RelA/metabolism , Transcription Factor RelB/metabolism , NF-kappaB-Inducing Kinase
7.
Cell Death Differ ; 26(3): 455-469, 2019 03.
Article En | MEDLINE | ID: mdl-29795336

Intrahepatocytic accumulation of misfolded α1-antitrypsin Z variant (ATZ) is responsible for liver disease in some individuals with α1-antitrypsin deficiency (ATD), characterized by fibrosis/cirrhosis and predisposition to carcinogenesis. Previous results showing that accumulation of ATZ in model systems activates the NFκB signaling pathway have led us to hypothesize that downstream targets of NFκB are elements of a proteostasis response network for this type of proteinopathy. Here we show that only a subset of downstream targets within the NFκB transcriptomic repertoire are activated in model systems of this proteinopathy. Breeding of the PiZ mouse model of ATD to two different mouse models with NFκB deficiency led to greater intrahepatocytic accumulation of ATZ, more severe hepatic fibrosis, decreased autophagy and hyperproliferation of hepatocytes with massive ATZ inclusions. Specific downstream targets of NFκB could be implicated in each pathological effect. These results suggest a new role for NFκB signaling in which specific downstream targets of this pathway mediate an integrated program of proteostatic responses designed to mitigate the pathologic effects of proteinopathy, including autophagic disposal of misfolded protein, degradation of collagen and prevention of hyperproliferation.


NF-kappa B/metabolism , alpha 1-Antitrypsin Deficiency/metabolism , alpha 1-Antitrypsin Deficiency/pathology , Animals , Autophagy , Down-Regulation , Early Growth Response Protein 1/genetics , Early Growth Response Protein 1/metabolism , HeLa Cells , Humans , Liver/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , NF-kappa B/biosynthesis , NF-kappa B/deficiency , NF-kappa B/genetics , Protein Folding , Proteostasis , Transcriptome , alpha 1-Antitrypsin , alpha 1-Antitrypsin Deficiency/genetics
8.
Aging Cell ; 18(1): e12882, 2019 02.
Article En | MEDLINE | ID: mdl-30468013

Increased activation of the major pro-inflammatory NF-κB pathway leads to numerous age-related diseases, including chronic liver disease (CLD). Rapamycin, an inhibitor of mTOR, extends lifespan and healthspan, potentially via suppression of inflammaging, a process which is partially dependent on NF-κB signalling. However, it is unknown if rapamycin has beneficial effects in the context of compromised NF-κB signalling, such as that which occurs in several age-related chronic diseases. In this study, we investigated whether rapamycin could ameliorate age-associated phenotypes in a mouse model of genetically enhanced NF-κB activity (nfκb1-/- ) characterized by low-grade chronic inflammation, accelerated aging and CLD. We found that, despite showing no beneficial effects in lifespan and inflammaging, rapamycin reduced frailty and improved long-term memory, neuromuscular coordination and tissue architecture. Importantly, markers of cellular senescence, a known driver of age-related pathology, were alleviated in rapamycin-fed animals. Our results indicate that, in conditions of genetically enhanced NF-κB, rapamycin delays aging phenotypes and improves healthspan uncoupled from its role as a suppressor of inflammation.


Inflammation/pathology , Longevity/physiology , NF-kappa B/deficiency , Sirolimus/pharmacology , Animals , Biomarkers/metabolism , Longevity/drug effects , Mice, Inbred C57BL , Mice, Knockout , Mitochondria/drug effects , Mitochondria/metabolism
9.
Protein Cell ; 9(11): 945-965, 2018 11.
Article En | MEDLINE | ID: mdl-29968158

Vascular cell functionality is critical to blood vessel homeostasis. Constitutive NF-κB activation in vascular cells results in chronic vascular inflammation, leading to various cardiovascular diseases. However, how NF-κB regulates human blood vessel homeostasis remains largely elusive. Here, using CRISPR/Cas9-mediated gene editing, we generated RelA knockout human embryonic stem cells (hESCs) and differentiated them into various vascular cell derivatives to study how NF-κB modulates human vascular cells under basal and inflammatory conditions. Multi-dimensional phenotypic assessments and transcriptomic analyses revealed that RelA deficiency affected vascular cells via modulating inflammation, survival, vasculogenesis, cell differentiation and extracellular matrix organization in a cell type-specific manner under basal condition, and that RelA protected vascular cells against apoptosis and modulated vascular inflammatory response upon tumor necrosis factor α (TNFα) stimulation. Lastly, further evaluation of gene expression patterns in IκBα knockout vascular cells demonstrated that IκBα acted largely independent of RelA signaling. Taken together, our data reveal a protective role of NF-κB/RelA in modulating human blood vessel homeostasis and map the human vascular transcriptomic landscapes for the discovery of novel therapeutic targets.


Blood Vessels/cytology , CRISPR-Cas Systems , Gene Knockout Techniques , Homeostasis , NF-kappa B/metabolism , Transcription Factor RelA/metabolism , Blood Vessels/metabolism , Embryonic Stem Cells/cytology , Humans , NF-kappa B/deficiency , Transcription Factor RelA/deficiency
10.
Immunity ; 48(3): 570-583.e8, 2018 03 20.
Article En | MEDLINE | ID: mdl-29562203

Polymorphisms in NFKB1 that diminish its expression have been linked to human inflammatory diseases and increased risk for epithelial cancers. The underlying mechanisms are unknown, and the link is perplexing given that NF-κB signaling reportedly typically exerts pro-tumorigenic activity. Here we have shown that NF-κB1 deficiency, even loss of a single allele, resulted in spontaneous invasive gastric cancer (GC) in mice that mirrored the histopathological progression of human intestinal-type gastric adenocarcinoma. Bone marrow chimeras revealed that NF-κB1 exerted tumor suppressive functions in both epithelial and hematopoietic cells. RNA-seq analysis showed that NF-κB1 deficiency resulted in aberrant JAK-STAT signaling, which dysregulated expression of effectors of inflammation, antigen presentation, and immune checkpoints. Concomitant loss of STAT1 prevented these immune abnormalities and GC development. These findings provide mechanistic insight into how polymorphisms that attenuate NFKB1 expression predispose humans to epithelial cancers, highlighting the pro-tumorigenic activity of STAT1 and identifying targetable vulnerabilities in GC.


Gene Expression Regulation, Neoplastic , Inflammation/genetics , Inflammation/metabolism , NF-kappa B/deficiency , STAT1 Transcription Factor/metabolism , Stomach Neoplasms/genetics , Stomach Neoplasms/metabolism , Animals , Antigen Presentation/immunology , Bone Marrow Cells/metabolism , Bone Marrow Cells/pathology , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Disease Models, Animal , Epithelial Cells/metabolism , Epithelial Cells/pathology , Gene Regulatory Networks , Humans , Inflammation/pathology , Mice , Mice, Knockout , STAT1 Transcription Factor/deficiency , Stomach Neoplasms/immunology , Stomach Neoplasms/pathology
11.
Int J Biochem Cell Biol ; 92: 141-147, 2017 11.
Article En | MEDLINE | ID: mdl-28987523

BACKGROUND AND OBJECTIVE: Pulmonary Hypertension (pH) is a chronic progressive disease. Endothelial cells (EC) play a central and critical role in the initiation and progression of pH. The NF-κB family (NF-κB1 (p50/p105), NF-κB2 (p52/p100), RelA (p65), RelB, and C-Rel) regulates a wide array of genes involved in inflammatory responses, cell proliferation, and survival. The involvement of specific NF-κB family members in the pathogenesis of hypoxia-induced pH remains to be determined. The objective of this study was to assess the specific role of individual NF-κB family members in mediating endothelial cell responses to hypoxia and its downstream effect on smooth muscle cell proliferation. METHODS AND RESULTS: NF-κB family members' expression were selectively reduced by siRNA in human pulmonary microvascular endothelial cells. Cells were then exposed to hypoxia (1%) for 24h. Endothelin1, ICAM1 gene expression and Stat1 and Stat3 phosphorylation were assessed. Smooth muscle cells (SMC) proliferation was assessed by culturing them with EC conditioned media. Reduction of either NF-κB2 or RelA in EC, led to a significant decrease in Endothelin1 and ICAM1 gene expression. C-Rel knockdown resulted in a significant increase in phosphorylated STAT1; both C-Rel and RelA knockdown significantly decreased phosphorylated STAT3 in EC. There was a significant reduction in SMC proliferation, and AKT/ERK phosphorylation in SMC, when cultured in RelA knockdown, EC conditioned media. CONCLUSION: RelA in EC plays crucial role in hypoxia induced vascular remodeling and development of pH. Targeting RelA in EC alleviates SMC proliferation as well as inflammation related processes.


Lung/blood supply , NF-kappa B/metabolism , Vascular Remodeling , Animals , Cell Hypoxia , Cell Proliferation , Endothelial Cells/cytology , Endothelin-1/metabolism , Gene Expression Regulation , Gene Knockdown Techniques , Hydrogen-Ion Concentration , Intercellular Adhesion Molecule-1/metabolism , Lung/cytology , Male , Mice , Mice, Inbred C57BL , Myocytes, Smooth Muscle/cytology , NF-kappa B/deficiency , NF-kappa B/genetics
12.
Nat Cell Biol ; 19(9): 1116-1129, 2017 Sep.
Article En | MEDLINE | ID: mdl-28846096

Apoptosis represents a key anti-cancer therapeutic effector mechanism. During apoptosis, mitochondrial outer membrane permeabilization (MOMP) typically kills cells even in the absence of caspase activity. Caspase activity can also have a variety of unwanted consequences that include DNA damage. We therefore investigated whether MOMP-induced caspase-independent cell death (CICD) might be a better way to kill cancer cells. We find that cells undergoing CICD display potent pro-inflammatory effects relative to apoptosis. Underlying this, MOMP was found to stimulate NF-κB activity through the downregulation of inhibitor of apoptosis proteins. Strikingly, engagement of CICD displays potent anti-tumorigenic effects, often promoting complete tumour regression in a manner dependent on intact immunity. Our data demonstrate that by activating NF-κB, MOMP can exert additional signalling functions besides triggering cell death. Moreover, they support a rationale for engaging caspase-independent cell death in cell-killing anti-cancer therapies.


Caspases/metabolism , Colonic Neoplasms/enzymology , Inflammation Mediators/metabolism , Mitochondria/enzymology , Mitochondrial Membranes/enzymology , NF-kappa B/metabolism , Aniline Compounds/pharmacology , Animals , Antineoplastic Agents/pharmacology , Apoptosis , Colonic Neoplasms/drug therapy , Colonic Neoplasms/immunology , Colonic Neoplasms/pathology , Genotype , HeLa Cells , Humans , Inhibitor of Apoptosis Proteins/metabolism , Macrophage Activation , Mice, Inbred BALB C , Mice, Inbred NOD , Mice, Knockout , Mice, SCID , Mitochondria/drug effects , Mitochondria/immunology , Mitochondria/pathology , Mitochondrial Membranes/drug effects , Mitochondrial Membranes/immunology , Mitochondrial Membranes/pathology , NF-kappa B/deficiency , Necrosis , Permeability , Phenotype , Protein Serine-Threonine Kinases/metabolism , RNA Interference , Receptors, Tumor Necrosis Factor, Type I/genetics , Receptors, Tumor Necrosis Factor, Type I/metabolism , Signal Transduction , Sulfonamides/pharmacology , Time Factors , Transfection , Tumor Necrosis Factor-alpha/metabolism , NF-kappaB-Inducing Kinase
13.
Am J Hematol ; 92(11): 1119-1130, 2017 Nov.
Article En | MEDLINE | ID: mdl-28699284

Elaboration of tumor necrosis factor (TNF) is a very early event in development of ischemia/reperfusion injury pathophysiology. Therefore, TNF may be a prominent mediator of endothelial cell and vascular wall dysfunction in sickle cell anemia, a hypothesis we addressed using NY1DD, S+SAntilles , and SS-BERK sickle transgenic mice. Transfusion experiments revealed participation of abnormally activated blood monocytes exerting an endothelial activating effect, dependent upon Egr-1 in both vessel wall and blood cells, and upon NFκB(p50) in a blood cell only. Involvement of TNF was identified by beneficial impact from TNF blockers, etanercept and infliximab, with less benefit from an IL-1 blocker, anakinra. In therapeutic studies, etanercept ameliorated multiple disturbances of the murine sickle condition: monocyte activation, blood biomarkers of inflammation, low platelet count and Hb, vascular stasis triggered by hypoxia/reoxygenation (but not if triggered by hemin infusion), tissue production of neuro-inflammatory mediators, endothelial activation (monitored by tissue factor and VCAM-1 expression), histopathologic liver injury, and three surrogate markers of pulmonary hypertension (perivascular inflammatory aggregates, arteriolar muscularization, and right ventricular mean systolic pressure). In aggregate, these studies identify a prominent-and possibly dominant-role for an abnormal monocyte-TNF-endothelial activation axis in the sickle context. Its presence, plus the many benefits of etanercept observed here, argue that pilot testing of TNF blockade should be considered for human sickle cell anemia, a challenging but achievable translational research goal.


Anemia, Sickle Cell/metabolism , Endothelial Cells/metabolism , Monocytes/metabolism , Signal Transduction , Tumor Necrosis Factor-alpha/metabolism , Anemia, Sickle Cell/diagnosis , Anemia, Sickle Cell/drug therapy , Anemia, Sickle Cell/genetics , Animals , Antibodies, Monoclonal/pharmacology , Biomarkers , Bone Marrow Transplantation , Cell Aggregation/genetics , Cell Aggregation/immunology , Disease Models, Animal , Early Growth Response Protein 1/genetics , Early Growth Response Protein 1/metabolism , Endothelium, Vascular/metabolism , Etanercept/pharmacology , Etanercept/therapeutic use , Heart Function Tests , Humans , Inflammation Mediators , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/metabolism , Mice , Mice, Knockout , Mice, Transgenic , Molecular Targeted Therapy , Monocytes/drug effects , Monocytes/immunology , NF-kappa B/deficiency , NF-kappa B/genetics , Phenotype , Protein Kinase Inhibitors/pharmacology , Signal Transduction/drug effects , Thromboplastin/metabolism , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Vascular Cell Adhesion Molecule-1/metabolism
14.
Muscle Nerve ; 56(4): 759-767, 2017 Oct.
Article En | MEDLINE | ID: mdl-27997693

INTRODUCTION: Duchenne muscular dystrophy (DMD) is a genetic muscle disease characterized by dystrophin deficiency. Beyond gene replacement, the question of whether ablation of the p65 gene of nuclear factor-kappa B (NF-κB) in DMD can improve muscle physiology function is unknown. In this study, we investigated muscle physiological improvement in mdx mice (DMD model) with a genetic reduction of NF-κB. METHODS: Muscle physiological function and histology were studied in 2-month-old mdx/p65+/- , wild-type, mdx, and human minidystrophin gene transgenic mdx (TghΔDys/mdx) mice. RESULTS: Improved muscle physiological function was found in mdx/p65+/- mice when compared with mdx mice; however, it was similar to TghΔDys/mdx mice. The results indicate that genetic reduction of p65 levels diminished chronic inflammation in dystrophic muscle, thus leading to amelioration of muscle pathology and improved muscle physiological function. CONCLUSIONS: The results show that inhibition of NF-κB may be a promising therapy when combined with gene therapy for DMD. Muscle Nerve 56: 759-767, 2017.


Gene Deletion , Muscle, Skeletal/physiology , Muscular Dystrophy, Duchenne/genetics , NF-kappa B/genetics , Protein Subunits/genetics , Animals , Humans , Mice , Mice, Inbred mdx , Mice, Transgenic , Muscle, Skeletal/pathology , Muscular Dystrophy, Duchenne/pathology , Muscular Dystrophy, Duchenne/physiopathology , NF-kappa B/deficiency , Protein Subunits/deficiency
15.
Diabetes ; 65(8): 2367-79, 2016 08.
Article En | MEDLINE | ID: mdl-27217485

The nuclear factor-κB protein c-Rel plays a critical role in controlling autoimmunity. c-Rel-deficient mice are resistant to streptozotocin-induced diabetes, a drug-induced model of autoimmune diabetes. We generated c-Rel-deficient NOD mice to examine the role of c-Rel in the development of spontaneous autoimmune diabetes. We found that both CD4(+) and CD8(+) T cells from c-Rel-deficient NOD mice showed significantly decreased T-cell receptor-induced IL-2, IFN-γ, and GM-CSF expression. Despite compromised T-cell function, c-Rel deficiency dramatically accelerated insulitis and hyperglycemia in NOD mice along with a substantial reduction in T-regulatory (Treg) cell numbers. Supplementation of isogenic c-Rel-competent Treg cells from prediabetic NOD mice reversed the accelerated diabetes development in c-Rel-deficient NOD mice. The results suggest that c-Rel-dependent Treg cell function is critical in suppressing early-onset autoimmune diabetogenesis in NOD mice. This study provides a novel natural system to study autoimmune diabetes pathogenesis and reveals a previously unknown c-Rel-dependent mechanistic difference between chemically induced and spontaneous diabetogenesis. The study also reveals a unique protective role of c-Rel in autoimmune diabetes, which is distinct from other T-cell-dependent autoimmune diseases such as arthritis and experimental autoimmune encephalomyelitis, where c-Rel promotes autoimmunity.


Diabetes Mellitus, Type 1/metabolism , NF-kappa B/deficiency , NF-kappa B/metabolism , Proto-Oncogene Proteins c-rel/deficiency , Proto-Oncogene Proteins c-rel/metabolism , Animals , CD4-Positive T-Lymphocytes/metabolism , Cell Proliferation/genetics , Cell Proliferation/physiology , Cytokines/metabolism , Diabetes Mellitus, Type 1/genetics , Diabetes Mellitus, Type 1/immunology , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, Knockout , NF-kappa B/genetics , Pancreas/metabolism , Proto-Oncogene Proteins c-rel/genetics , Real-Time Polymerase Chain Reaction , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism
16.
J Biomed Sci ; 22: 40, 2015 Jun 10.
Article En | MEDLINE | ID: mdl-26059504

BACKGROUND: The NF-κB signaling pathway plays a role in local and remote tissue damage following ischemia-reperfusion (I/R) injury to skeletal muscles. Evidence suggests that exosomes can act as intercellular communicators by transporting active proteins to remote cells and may play a role in regulating inflammatory processes. This study aimed to profile the exosomal protein expression in the serum of NF-κB knockout mice following skeletal muscle ischemia-reperfusion injury. RESULTS: To investigate the potential changes in protein expression mediated by NF-κB in secreted exosomes in the serum following I/R injury, the levels of circulating exosomal proteomes in C57BL/6 and NF-κB(-/-) mice were compared using two dimensional differential in-gel electrophoresis (2-DE), liquid chromatography tandem mass spectrometry (LC-MS/MS), and proteomic analysis. In C57BL/6 mice, the levels of circulating exosomal proteins, including complement component C3 prepropeptide, PK-120 precursor, alpha-amylase one precursor, beta-enolase isoform 1, and adenylosuccinate synthetase isozyme 1, increased following I/R injury. However, in the NF-κB(-/-) mice, the expression of the following was upregulated in the exosomes: protease, serine 1; glyceraldehyde-3-phosphate dehydrogenase-like isoform 1; glyceraldehyde-3-phosphate dehydrogenase; and pregnancy zone protein. In contrast, the expression of apolipoprotein B, complement component C3 prepropeptide, and immunoglobulin kappa light chain variable region was downregulated in NF-κB(-/-) mice. Bioinformatic annotation using the Protein Analysis Through Evolutionary Relationships (PANTHER) database revealed that the expression of the exosomal proteins that participate in metabolic processes and in biological regulation was lower in NF-κB(-/-) mice than in C57BL/6 mice, whereas the expression of proteins that participate in the response to stimuli, in cellular processes, and in the immune system was higher. CONCLUSIONS: The data presented in this study suggest that NF-κB might regulate exosomal protein expression at a remote site via circulation following I/R injury.


Muscle, Skeletal/metabolism , NF-kappa B/deficiency , Proteome , Reperfusion Injury/physiopathology , Signal Transduction , Animals , Exosomes/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , NF-kappa B/genetics
17.
J Immunol ; 194(5): 2380-9, 2015 Mar 01.
Article En | MEDLINE | ID: mdl-25624452

Engagement of promoters with distal elements in long-range looping interactions has been implicated in regulation of Ig class switch recombination (CSR). The principles determining the spatial and regulatory relationships among Igh transcriptional elements remain poorly defined. We examined the chromosome conformation of C region (CH) loci that are targeted for CSR in a cytokine-dependent fashion in mature B lymphocytes. Germline transcription (GLT) of the γ1 and ε CH loci is controlled by two transcription factors, IL-4-inducible STAT6 and LPS-activated NF-κB. We showed that although STAT6 deficiency triggered loss of GLT, deletion of NF-κB p50 abolished both GLT and γ1 locus:enhancer looping. Thus, chromatin looping between CH loci and Igh enhancers is independent of GLT production and STAT6, whereas the establishment and maintenance of these chromatin contacts requires NF-κB p50. Comparative analysis of the endogenous γ1 locus and a knock-in heterologous promoter in mice identified the promoter per se as the interactive looping element and showed that transcription elongation is dispensable for promoter/enhancer interactions. Interposition of the LPS-responsive heterologous promoter between the LPS-inducible γ3 and γ2b loci altered GLT expression and essentially abolished direct IgG2b switching while maintaining a sequential µâ†’γ3→γ2b format. Our study provides evidence that promoter/enhancer looping interactions can introduce negative constraints on distal promoters and affect their ability to engage in germline transcription and determine CSR targeting.


B-Lymphocytes/immunology , Chromatin/chemistry , Immunoglobulin Class Switching/genetics , Immunoglobulins/chemistry , V(D)J Recombination/genetics , Animals , B-Lymphocytes/cytology , B-Lymphocytes/drug effects , Chromatin/immunology , Chromosomes, Mammalian , Gene Expression Regulation/immunology , Genetic Loci , Immunoglobulins/genetics , Immunoglobulins/immunology , Interleukin-4/genetics , Interleukin-4/immunology , Lipopolysaccharides/pharmacology , Mice , Mice, Inbred C57BL , Mice, Knockout , NF-kappa B/deficiency , NF-kappa B/genetics , NF-kappa B/immunology , Promoter Regions, Genetic , STAT6 Transcription Factor/deficiency , STAT6 Transcription Factor/genetics , STAT6 Transcription Factor/immunology , Signal Transduction , Spleen/cytology , Spleen/drug effects , Spleen/immunology , Transcription Elongation, Genetic
18.
Folia Neuropathol ; 53(4): 367-76, 2015.
Article En | MEDLINE | ID: mdl-26785371

Amyotrophic lateral sclerosis (ALS) is a fatal incurable neurodegenerative disease whose etiology is unknown and pathogenesis is still not fully understood. A great majority of its cases are sporadic. Clinical ALS signs are caused by damage and dying-out of the lower and upper motor neurons. This study was aimed at identifying possible sporadic ALS-associated aberrations in the spinal cord expression of the transcription nuclear factor κ light-chain-enhancer of activated B cells (NF-κB). NF-κB is widely distributed among various cell types, including those specific for the central nervous system (CNS), and is involved in the control of many physiological and pathological processes, including, inter alia, inflammatory response, proliferation, angiogenesis, and cell survival and death. It is constitutively expressed and its inactive form resides in the cytoplasm. After activation, it enters the cell nucleus and promotes the transcription of target genes. NF-κB is a dimer and its most common form is a heterodimer made of subunits p50 and p65. In this study, we estimated and compared by immunohistochemical means the contents of these subunits in spinal cord motoneurons in a few archival cases of sporadic ALS of varying disease duration and the respective age-matched control cases with no CNS pathology. The major goal of the study was to seek possible changes in the expression of these proteins in the course of the disease. The control cases showed a strong expression of both p50 and p65 in spinal cord motoneurons, with both cytoplasmic and nuclear localization. In contrast, the ALS cases studies revealed a considerably lower and varying intensity of specific immunohistochemical staining for the two subunits, which suggested an increased deficit of their expression linked to longer disease duration. Moreover, there was an apparent shift toward mostly cytoplasmic localization of the two subunits. These preliminary data suggest that the changes in the expression of theses NF-κB subunits may be involved in pathogenesis of sporadic ALS.


Amyotrophic Lateral Sclerosis/metabolism , Amyotrophic Lateral Sclerosis/pathology , Motor Neurons/metabolism , Motor Neurons/pathology , NF-kappa B/deficiency , Aged , Female , Humans , Male , Middle Aged , Pilot Projects , Spinal Cord/metabolism , Spinal Cord/pathology
19.
Stem Cells ; 31(7): 1383-95, 2013 Jul.
Article En | MEDLINE | ID: mdl-23553791

An inflammatory microenvironment may cause organ degenerative diseases and malignant tumors. However, the precise mechanisms of inflammation-induced diseases are not fully understood. Here, we show that the proinflammatory cytokines interferon-γ (IFN-γ) and tumor necrosis factor α (TNF-α) synergistically impair self-renewal and differentiation of mesenchymal stem cells (MSCs) via nuclear factor κB (NFκB)-mediated activation of mothers against decapentaplegic homolog 7 (SMAD7) in ovariectomized (OVX) mice. More interestingly, a long-term elevated levels of IFN-γ and TNF-α result in significantly increased susceptibility to malignant transformation in MSCs through NFκB-mediated upregulation of the oncogenes c-Fos and c-Myc. Depletion of either IFN-γ or TNF-α in OVX mice abolishes MSC impairment and the tendency toward malignant transformation with no NFκB-mediated oncogene activation. Systemic administration of aspirin, which significantly reduces the levels of IFN-γ and TNF-α, results in blockage of MSC deficiency and tumorigenesis by inhibition of NFκB/SMAD7 and NFκB/c-FOS and c-MYC pathways in OVX mice. In summary, this study reveals that inflammation factors, such as IFN-γ and TNF-α, synergistically induce MSC deficiency via NFκB/SMAD7 signaling and tumorigenesis via NFκB-mediated oncogene activation.


Interferon-gamma/metabolism , Mesenchymal Stem Cells/metabolism , NF-kappa B/metabolism , Tumor Necrosis Factor-alpha/metabolism , Animals , Aspirin/pharmacology , Carcinogenesis , Cell Differentiation/physiology , Female , Genes, fos , Genes, myc , Interferon-gamma/deficiency , Interferon-gamma/genetics , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Knockout , Mice, Nude , NF-kappa B/antagonists & inhibitors , NF-kappa B/deficiency , NF-kappa B/genetics , Signal Transduction , Smad7 Protein/metabolism , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Tumor Necrosis Factor-alpha/deficiency , Tumor Necrosis Factor-alpha/genetics
20.
Cell Biochem Biophys ; 66(3): 431-41, 2013 Jul.
Article En | MEDLINE | ID: mdl-23238643

Intermittent hypoxia (IH) is a hallmark feature in obstructive sleep apnea (OSA) which is increasingly recognized as an independent risk factor for atherosclerosis. Oxidative stress, inflammation, and cell apoptosis are major pathological events initiating or accelerating atherogenesis. This study addressed whether IH would affect these proatherogenic factors in endothelial cells and the mechanistic pathways involved. EA.hy926 cells were exposed to intermittent normoxia or IH for different numbers of cycles (32, 64, or 96). IH exposure time-dependently raised cellular GSSG/GSH ratio, increased production of IL-6 and IL-8, and accelerated cell apoptosis and death, concurrent with activation of NF-κB and inhibition of Nrf2/HO-1 pathways. At 64 cycles, inhibition of NF-κB attenuated IH-induced cellular oxidative stress and accumulation of inflammatory cytokines in cell culture medium but aggravated IH-induced cell apoptosis, while stimulation of HO-1 suppressed IH-induced cellular oxidative stress and cell apoptosis without affecting accumulation of inflammatory cytokines in cell culture medium. We demonstrated that early stage of exposure to IH-induced oxidative and inflammatory stresses leading to acceleration of cell apoptosis via NF-κB and Nrf2/HO-1 pathways in endothelial cells, suggesting the potential mechanisms for IH-induced vascular pathogenesis, in resemblance to OSA.


Endothelial Cells/cytology , Heme Oxygenase-1/metabolism , NF-kappa B/metabolism , Apoptosis/drug effects , Basic-Leucine Zipper Transcription Factors/deficiency , Basic-Leucine Zipper Transcription Factors/genetics , Cell Hypoxia/drug effects , Cell Line , Culture Media/chemistry , Cytokines/metabolism , Endothelial Cells/drug effects , Fanconi Anemia Complementation Group Proteins/deficiency , Fanconi Anemia Complementation Group Proteins/genetics , Gene Knockdown Techniques , Hemin/pharmacology , Humans , NF-E2-Related Factor 2/metabolism , NF-kappa B/deficiency , NF-kappa B/genetics , Oxidative Stress/drug effects , Signal Transduction/drug effects , Thiophenes/pharmacology
...