Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 167
Filter
1.
Mol Immunol ; 171: 22-35, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38749236

ABSTRACT

OBJECTIVES: Systemic lupus erythematosus (SLE) is a multi-organ autoimmune disease, of which the pathogens is remains obscure. Viral infection, particularly Epstein Barr viru (EBV) infection, has been considered a common pathogenic factor. This study suggests that c-Maf may be an important target in T cell differentiation during SLE progression, providing a potentially new perspective on the role of viral infection in the pathogenesis of autoimmune diseases. METHODS: Cytokines of EBV-infected SLE patients were measured by ELISA and assessed in conjunction with their clinical data. IFN-α, c-Maf, and the differentiation of Th17/Treg cells in SLE patients and MRL/LPR mice were analyzed using FCM, WB, RT-PCR, etc. Following the infection of cells and mice with EBV or viral mimic poly (dA:dT), the changes of the aforementioned indicators were investigated. The relationship among IFN-α, STAT3, c-Maf and Th17 cells was determined by si-RNA technique. RESULTS: Many SLE patients are found to be complicated by viral infections; Further, studies have demonstrated that viral infection, especially EBV, is involved in SLE development. This study showed that viral infections might promote IFN-α secretion, inhibit c-Maf expression by activating STAT3, increase Th17 cell differentiation, and lead to the immune imbalance of Th17/Treg cells, thus playing a role in the onset and progression of SLE. CONCLUSION: This study demonstrates that EBV infections may contribute to SLE development by activating STAT3 through IFN-α, inhibiting c-Maf, and causing Th17/Treg immune imbalance. Our work provided a new insight into the pathogenesis and treatment of SLE.


Subject(s)
Epstein-Barr Virus Infections , Herpesvirus 4, Human , Interferon-alpha , Lupus Erythematosus, Systemic , Mice, Inbred MRL lpr , Proto-Oncogene Proteins c-maf , T-Lymphocytes, Regulatory , Th17 Cells , Lupus Erythematosus, Systemic/immunology , Lupus Erythematosus, Systemic/virology , Th17 Cells/immunology , Humans , Animals , Epstein-Barr Virus Infections/immunology , Epstein-Barr Virus Infections/virology , Epstein-Barr Virus Infections/complications , T-Lymphocytes, Regulatory/immunology , Mice , Interferon-alpha/immunology , Interferon-alpha/metabolism , Female , Adult , Herpesvirus 4, Human/immunology , Proto-Oncogene Proteins c-maf/immunology , Proto-Oncogene Proteins c-maf/genetics , Male , Cell Differentiation/immunology , Disease Progression , Middle Aged , STAT3 Transcription Factor/metabolism , STAT3 Transcription Factor/immunology , Young Adult
2.
Nat Immunol ; 25(5): 886-901, 2024 May.
Article in English | MEDLINE | ID: mdl-38609547

ABSTRACT

Intestinal immune responses to microbes are controlled by the cytokine IL-10 to avoid immune pathology. Here, we use single-cell RNA sequencing of colon lamina propria leukocytes (LPLs) along with RNA-seq and ATAC-seq of purified CD4+ T cells to show that the transcription factors Blimp-1 (encoded by Prdm1) and c-Maf co-dominantly regulate Il10 while negatively regulating proinflammatory cytokines in effector T cells. Double-deficient Prdm1fl/flMaffl/flCd4Cre mice infected with Helicobacter hepaticus developed severe colitis with an increase in TH1/NK/ILC1 effector genes in LPLs, while Prdm1fl/flCd4Cre and Maffl/flCd4Cre mice exhibited moderate pathology and a less-marked type 1 effector response. LPLs from infected Maffl/flCd4Cre mice had increased type 17 responses with increased Il17a and Il22 expression and an increase in granulocytes and myeloid cell numbers, resulting in increased T cell-myeloid-neutrophil interactions. Genes over-expressed in human inflammatory bowel disease showed differential expression in LPLs from infected mice in the absence of Prdm1 or Maf, revealing potential mechanisms of human disease.


Subject(s)
Colitis , Helicobacter hepaticus , Mice, Knockout , Positive Regulatory Domain I-Binding Factor 1 , Proto-Oncogene Proteins c-maf , Animals , Positive Regulatory Domain I-Binding Factor 1/genetics , Positive Regulatory Domain I-Binding Factor 1/metabolism , Mice , Proto-Oncogene Proteins c-maf/genetics , Colitis/immunology , Colitis/genetics , Humans , Helicobacter hepaticus/immunology , Helicobacter Infections/immunology , Mice, Inbred C57BL , Intestinal Mucosa/immunology , Intestinal Mucosa/pathology , Intestinal Mucosa/microbiology , Inflammatory Bowel Diseases/immunology , Inflammatory Bowel Diseases/genetics , Gene Expression Regulation , Disease Models, Animal
4.
Nat Cell Biol ; 25(12): 1833-1847, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37945904

ABSTRACT

MAF amplification increases the risk of breast cancer (BCa) metastasis through mechanisms that are still poorly understood yet have important clinical implications. Oestrogen-receptor-positive (ER+) BCa requires oestrogen for both growth and metastasis, albeit by ill-known mechanisms. Here we integrate proteomics, transcriptomics, epigenomics, chromatin accessibility and functional assays from human and syngeneic mouse BCa models to show that MAF directly interacts with oestrogen receptor alpha (ERα), thereby promoting a unique chromatin landscape that favours metastatic spread. We identify metastasis-promoting genes that are de novo licensed following oestrogen exposure in a MAF-dependent manner. The histone demethylase KDM1A is key to the epigenomic remodelling that facilitates the expression of the pro-metastatic MAF/oestrogen-driven gene expression program, and loss of KDM1A activity prevents this metastasis. We have thus determined that the molecular basis underlying MAF/oestrogen-mediated metastasis requires genetic, epigenetic and hormone signals from the systemic environment, which influence the ability of BCa cells to metastasize.


Subject(s)
Breast Neoplasms , Epigenesis, Genetic , Estrogen Receptor alpha , Gene Amplification , Proto-Oncogene Proteins c-maf , Animals , Female , Humans , Mice , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Line, Tumor , Chromatin , Estrogen Receptor alpha/genetics , Estrogen Receptor alpha/metabolism , Estrogens , Histone Demethylases/genetics , Histone Demethylases/metabolism , Proto-Oncogene Proteins c-maf/genetics
5.
Biochem Biophys Res Commun ; 684: 149135, 2023 12 03.
Article in English | MEDLINE | ID: mdl-37879249

ABSTRACT

Multiple myeloma displays the clonal B cell expansion and the overproduction of monoclonal immunoglobulins. Genetic translocations at 14q32, particularly with partners like 16q23, lead to the dysregulation of oncogene expression, including the significant enhancement of c-Maf. This aberrant expression of c-Maf has prompted research into strategies for targeting this transcription factor as a potential therapeutic avenue for multiple myeloma treatment. In this study, we introduce a screening pipeline to test small compounds for their ability to inhibit c-Maf. Using a luciferase indicator driven by the Ccl8 gene promoter, we identified two small compounds that inhibit transcriptional activity of c-Maf. These molecules impede the proliferation of c-Maf-expressing myeloma cells, and repress the expression of c-Maf target genes such as ITGB7 and CCR1. Importantly, these molecules target c-Maf-expressing multiple myeloma cells, but not c-Maf-negative myeloma cells, showing potential for tailoring therapeutic intervention. In conclusion, our screening pipeline is effective to explore leads for a novel c-Maf inhibitor for multiple myeloma therapy.


Subject(s)
Multiple Myeloma , Humans , Multiple Myeloma/drug therapy , Multiple Myeloma/genetics , Multiple Myeloma/metabolism , Proto-Oncogene Proteins c-maf/genetics , Proto-Oncogene Proteins c-maf/metabolism , B-Lymphocytes/metabolism , Gene Expression Regulation , Cell Proliferation
6.
Am J Med Genet A ; 188(10): 2888-2898, 2022 10.
Article in English | MEDLINE | ID: mdl-36097645

ABSTRACT

Pathogenic variants in the v-maf avian musculoaponeurotic fibrosarcoma oncogene homologue (MAF) encoding a transcription factor (from a unique subclass of basic leucine zipper transcription factors) are associated with isolated congenital cataracts (CCs) and Aymé-Gripp syndrome (AYGRPS). We collected detailed disease histories from, and performed comprehensive ophthalmic and systemic examinations in 269 patients with CCs; we then performed whole-exome sequencing. Pathogenicity assessments were evaluated using multiple predictive tools. The clinical validities of the reported gene-disease relationships for MAF genes (MAF-CCs and MAF-AYGRPS) were assessed using the ClinGen gene curation framework. We identified two novel (c.173C>A, p.Thr58Asn and c.947T>C, p. Leu316Pro) variants and one known (c.173C>T, p.Thr58Ile) MAF missense variant in three patients. We described novel phenotypes including cleft palate, macular hypoplasia, and retinal neovascularization in the peripheral avascular area and analyzed the genotype-phenotype correlations. We demonstrated associations of variants in the MAF C-terminal DNA-binding domain with CCs and associations of variants in the N-terminal transactivation domain of MAF with AYGRPS. We thus expand the genotypic and phenotypic spectrum of the MAF gene. The ClinGen gene curation framework results suggested that variants in different domains of MAF are associated with different diseases.


Subject(s)
Cataract , Proto-Oncogene Proteins c-maf , Cataract/pathology , China , Facies , Genotype , Growth Disorders , Hearing Loss, Sensorineural , Humans , Intellectual Disability , Phenotype , Proto-Oncogene Proteins c-maf/genetics
7.
J Exp Med ; 219(12)2022 12 05.
Article in English | MEDLINE | ID: mdl-36121416

ABSTRACT

The primary function of the small intestine (SI) is to absorb nutrients to maintain whole-body energy homeostasis. Enterocytes are the major epithelial cell type facilitating nutrient sensing and uptake. However, the molecular regulators governing enterocytes have remained undefined. Here, we identify c-Maf as an enterocyte-specific transcription factor within the SI epithelium. c-Maf expression was determined by opposing Noggin/BMP signals and overlapped with the zonated enrichment of nutrient transporters in the mid-villus region. Functionally, enterocytes required c-Maf to appropriately differentiate along the villus axis. Specifically, gene programs controlling carbohydrate and protein absorption were c-Maf-dependent. Consequently, epithelial cell-specific c-Maf deletion resulted in impaired enterocyte maturation and nutrient uptake, including defects in the adaptation to different nutrient availability. Concomitantly, intraepithelial lymphocytes were less abundant, while commensal epithelial cell-attaching SFB overgrew in a c-Maf-deficient environment, highlighting the close interdependence between the intestinal epithelium, immune system, and microbiota. Collectively, our data identified c-Maf as a key regulator of SI enterocyte differentiation and function, essential for nutrient, immune, and microbial homeostasis.


Subject(s)
Enterocytes , Intestines , Proto-Oncogene Proteins c-maf , Animals , Carbohydrates , Enterocytes/metabolism , Mice , Nutrients , Proto-Oncogene Proteins c-maf/genetics , Proto-Oncogene Proteins c-maf/metabolism , Transcription Factors/metabolism
8.
Front Immunol ; 13: 818814, 2022.
Article in English | MEDLINE | ID: mdl-35359922

ABSTRACT

Regulatory B cells (Bregs) have been highlighted in very different pathology settings including autoimmune diseases, allergy, graft rejection, and cancer. Improving tools for the characterization of Bregs has become the main objective especially in humans. Transitional, mature B cells and plasma cells can differentiate into IL-10 producing Bregs in both mice and humans, suggesting that Bregs are not derived from unique precursors but may arise from different competent progenitors at unrestricted development stages. Moreover, in addition to IL-10 production, regulatory B cells used a broad range of suppressing mechanisms to modulate the immune response. Although Bregs have been consistently described in the literature, only a few reports described the molecular aspects that control the acquisition of the regulatory function. In this manuscript, we detailed the latest reports describing the control of IL-10, TGFß, and GZMB production in different Breg subsets at the molecular level. We focused on the understanding of the role of the transcription factors STAT3 and c-MAF in controlling IL-10 production in murine and human B cells and how these factors may represent an important crossroad of several key drivers of the Breg response. Finally, we provided original data supporting the evidence that MAF is expressed in human IL-10- producing plasmablast and could be induced in vitro following different stimulation cocktails. At steady state, we reported that MAF is expressed in specific human B-cell tonsillar subsets including the IgD+ CD27+ unswitched population, germinal center cells and plasmablast.


Subject(s)
Autoimmune Diseases , B-Lymphocytes, Regulatory , Animals , Autoimmune Diseases/pathology , Interleukin-10 , Lymphocyte Count , Mice , Plasma Cells , Proto-Oncogene Proteins c-maf/genetics
9.
Clin Transl Med ; 12(3): e781, 2022 03.
Article in English | MEDLINE | ID: mdl-35343082

ABSTRACT

Systemic lupus erythematosus (SLE) is a chronic autoimmune disease that involves T follicular helper (TFH ) cell-mediated humoral immune responses. Absent in melanoma 2 (human AIM2 and murine Aim2) is a well-known component of the inflammasome in the innate immune system. Surprisingly, we observed that in SLE patients, upregulated levels of AIM2 expression were found in peripheral blood and skin lesions, with the highest levels detected in TFH -like cells. In the CD4cre Aim2fl/fl conditional knockout mice, a markedly reduced TFH cell response was observed, with significantly lower levels of serum autoantibodies and proteinuria, as well as profoundly reduced renal IgG deposition in pristane-induced lupus mice. Mechanistically, IL-21 was found to recruit hydroxymethyltransferase ten-eleven translocation 2 (TET2) to the AIM2 promoter, resulting in DNA demethylation and increased transcription of AIM2. In addition, AIM2 could regulate c-MAF expression to enhance IL-21 production, which consequently promoted TFH cell differentiation. Our results have identified a role of AIM2 in promoting the TFH cell response and further revealed that the IL-21-TET2-AIM2-c-MAF signalling pathway is dysregulated in lupus pathogenesis, which provides a potential therapeutic target for SLE.


Subject(s)
Dioxygenases , Lupus Erythematosus, Systemic , Animals , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Humans , Interleukins/genetics , Lupus Erythematosus, Systemic/genetics , Lupus Erythematosus, Systemic/metabolism , Mice , Proto-Oncogene Proteins c-maf/genetics , T Follicular Helper Cells
10.
Mucosal Immunol ; 15(3): 480-490, 2022 03.
Article in English | MEDLINE | ID: mdl-35169232

ABSTRACT

Immunosuppressive Interleukin (IL)-10 production by pro-inflammatory CD4+ T cells is a central self-regulatory function to limit aberrant inflammation. Still, the molecular mediators controlling IL-10 expression in human CD4+ T cells are largely undefined. Here, we identify a Notch/STAT3 signaling-module as a universal molecular switch to induce IL-10 expression across human naïve and major effector CD4+ T cell subsets. IL-10 induction was transient, jointly controlled by the transcription factors Blimp-1/c-Maf and accompanied by upregulation of several co-inhibitory receptors, including LAG-3, CD49b, PD-1, TIM-3 and TIGIT. Consistent with a protective role of IL-10 in inflammatory bowel diseases (IBD), effector CD4+ T cells from Crohn's disease patients were defective in Notch/STAT3-induced IL-10 production and skewed towards an inflammatory Th1/17 cell phenotype. Collectively, our data identify a Notch/STAT3-Blimp-1/c-Maf axis as a common anti-inflammatory pathway in human CD4+ T cells, which is defective in IBD and thus may represent an attractive therapeutic target.


Subject(s)
Crohn Disease , Inflammatory Bowel Diseases , Animals , Crohn Disease/metabolism , Humans , Inflammatory Bowel Diseases/metabolism , Interleukin-10/metabolism , Mice , Mice, Knockout , Proto-Oncogene Proteins c-maf/genetics , Proto-Oncogene Proteins c-maf/metabolism , STAT3 Transcription Factor/metabolism , Th1 Cells/metabolism
11.
Biochim Biophys Acta Mol Cell Res ; 1869(5): 119222, 2022 05.
Article in English | MEDLINE | ID: mdl-35093454

ABSTRACT

The activation of Nuclear Factor, Erythroid 2 Like 2 - Kelch Like ECH Associated Protein 1 (NRF2-KEAP1) signaling pathway plays a critical dual role by either protecting or promoting the carcinogenesis process. However, its activation or nuclear translocation during hepatocellular carcinoma (HCC) progression has not been addressed yet. This study characterizes the subcellular localization of both NRF2 and KEAP1 during diethylnitrosamine-induced hepatocarcinogenesis in the rat. NRF2-KEAP1 pathway was continuously activated along with the increased expression of its target genes, namely Nqo1, Hmox1, Gclc, and Ptgr1. Similarly, the nuclear translocation of NRF2, MAF, and KEAP1 increased in HCC cells from weeks 12 to 22 during HCC progression. Likewise, colocalization of NRF2 with KEAP1 was higher in the cell nuclei of HCC neoplastic nodules than in surrounding cells. Moreover, immunofluorescence analyses revealed that the interaction of KEAP1 with filamentous Actin was disrupted in HCC cells. This disruption may be contributing to the release and nuclear translocation of NRF2 since the cortical actin cytoskeleton serves as anchoring of KEAP1. In conclusion, this evidence indicates that NRF2 is progressively activated and promotes the progression of experimental HCC.


Subject(s)
Carcinoma, Hepatocellular/pathology , Kelch-Like ECH-Associated Protein 1/metabolism , Liver Neoplasms/pathology , NF-E2-Related Factor 2/metabolism , Actin Cytoskeleton/metabolism , Animals , Carcinoma, Hepatocellular/chemically induced , Carcinoma, Hepatocellular/veterinary , Cell Nucleus/metabolism , Cyclooxygenase 1/genetics , Cyclooxygenase 1/metabolism , Diethylnitrosamine/toxicity , Disease Progression , Kelch-Like ECH-Associated Protein 1/genetics , Liver Neoplasms/chemically induced , Liver Neoplasms/veterinary , Membrane Proteins/genetics , Membrane Proteins/metabolism , NF-E2-Related Factor 2/genetics , Proto-Oncogene Proteins c-maf/genetics , Proto-Oncogene Proteins c-maf/metabolism , Rats , Rats, Inbred F344
12.
Neurobiol Aging ; 111: 95-106, 2022 03.
Article in English | MEDLINE | ID: mdl-34852950

ABSTRACT

The genetic locus containing the WWOX and MAF genes was implicated as a clinical Alzheimer's disease (AD) risk locus in two recent large meta-analytic genome wide association studies (GWAS). In a prior GWAS, we identified a variant in WWOX as a suggestive risk allele for hippocampal sclerosis. We hypothesized that the WWOX/MAF locus may be preferentially associated with non-plaque- and non-tau-related neuropathological changes (NC). Data from research participants with GWAS and autopsy measures from the National Alzheimer's Coordinating Center and the Religious Orders Study and the Rush Memory and Aging Project were meta-analyzed. Notably, no variants in the locus were significantly associated with ADNC. However, several WWOX/MAF variants had significant adjusted associations with limbic-predominant age-related TDP-43 encephalopathy NC (LATE-NC), HS, and brain arteriolosclerosis. These associations remained largely unchanged after adjustment for ADNC (operationalized with standard semiquantitative staging), suggesting that these associations are independent of ADNC. Thus, WWOX genetic variants were associated pathologically with LATE-NC, not ADNC.


Subject(s)
Alzheimer Disease/genetics , Dementia/genetics , Genetic Variation/genetics , Genome-Wide Association Study/methods , Phenotype , Proto-Oncogene Proteins c-maf/genetics , TDP-43 Proteinopathies/genetics , Tumor Suppressor Proteins/genetics , WW Domain-Containing Oxidoreductase/genetics , Aged , Aged, 80 and over , Female , Humans , Male
13.
Int Immunopharmacol ; 101(Pt A): 108264, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34715493

ABSTRACT

Topoisomerase (TOP) inhibitors were commonly used as chemotherapeutic agents in the treatment of cancers. In our present study, we found that etoposide (ETO), a topoisomerase 2 (TOP2) inhibitor, upregulated the production of Interleukin 10 (IL-10) in lipopolysaccharide (LPS)-stimulated macrophages. Besides, other TOP2 inhibitors including doxorubicin hydrochloride (DOX) and teniposide (TEN) were also able to augment IL-10 production. Meanwhile, the expression levels of pro-inflammatory factors, for example IL-6 and TNF-α, were also decreased accordingly by the treatment of the TOP2 inhibitors. Of note, ETO facilitated IL-10 secretion, which might be regulated by transcription factor Maf via PI3K/AKT pathway, as pharmaceutic blockage of kinase PI3K or AKT attenuated ETO-induced Maf and IL-10 expression. Further, in LPS-induced mice sepsis model, the enhanced generation of IL-10 was observed in ETO-treated mice, whereas pro-inflammatory cytokines were decreased, which significantly reduced the mortality of mice from LPS-induced lethal cytokine storm. Taken together, these results indicated that ETO may exhibit an anti-inflammatory role by upregulating the alteration of transcription factor Maf and promoting subsequential IL-10 secretion via PI3K/Akt pathway in LPS-induced macrophages. Therefore, ETO may serve as a potential anti-inflammatory agent and employed to severe pro-inflammatory diseases including COVID-19.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Etoposide/pharmacology , Interleukin-10/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins c-maf/genetics , Topoisomerase II Inhibitors/pharmacology , Animals , Anti-Inflammatory Agents/therapeutic use , Cell Line , Disease Models, Animal , Down-Regulation/drug effects , Etoposide/therapeutic use , Female , Interleukin-10/genetics , Interleukin-6/genetics , Interleukin-6/metabolism , Lipopolysaccharides/toxicity , Macrophages/drug effects , Mice , Mice, Inbred C57BL , Proto-Oncogene Proteins c-maf/metabolism , Shock, Septic/chemically induced , Shock, Septic/drug therapy , Topoisomerase II Inhibitors/therapeutic use , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism , Up-Regulation/drug effects , COVID-19 Drug Treatment
14.
Int J Mol Sci ; 22(17)2021 Sep 06.
Article in English | MEDLINE | ID: mdl-34502552

ABSTRACT

Immune response control is critical as excessive cytokine production can be detrimental and damage the host. Interleukin-10 (Il-10), an anti-inflammatory cytokine produced primarily by macrophages, is a key regulator that counteracts and controls excessive inflammatory response. Il-10 expression is regulated through the transcription factor c-Maf. Another regulator of Il-10 production is p35, an activator of the cyclin-dependent kinase 5 (Cdk5), which decreases Il-10 production in macrophages, thus increasing inflammation. However, Cdk5 regulation of c-Maf and the involvement of Il-10 production in macrophages has not yet been investigated. We used in vitro primary bone marrow-derived macrophages (BMDMs) lacking Cdk5, stimulated them with lipopolysaccharid (LPS) and observed increased levels of c-Maf and Il-10. In an in vivo mouse model of LPS-induced endotoxemia, mice lacking Cdk5 in macrophages showed increased levels of c-Maf and elevated levels of Il-10 in lungs as well as in plasma, resulting in ameliorated survival. Taken together, we identified Cdk5 as a potential novel regulator of Il-10 production through c-Maf in macrophages under inflammatory conditions. Our results suggest that inhibition of Cdk5 enhances the c-Maf-Il-10 axis and thus potentiates improvement of anti-inflammatory therapy.


Subject(s)
Cyclin-Dependent Kinase 5/genetics , Endotoxemia/genetics , Inflammation/genetics , Interleukin-10/genetics , Macrophages/metabolism , Proto-Oncogene Proteins c-maf/genetics , Animals , Cells, Cultured , Chemokines/genetics , Chemokines/metabolism , Cyclin-Dependent Kinase 5/metabolism , Cytokines/genetics , Cytokines/metabolism , Endotoxemia/chemically induced , Endotoxemia/metabolism , Gene Expression Regulation , Inflammation/metabolism , Interleukin-10/metabolism , Lipopolysaccharides , Lung/metabolism , Lung/pathology , Mice, Inbred C57BL , Mice, Knockout , Proto-Oncogene Proteins c-maf/metabolism
15.
Nat Commun ; 12(1): 5450, 2021 09 14.
Article in English | MEDLINE | ID: mdl-34521827

ABSTRACT

Multiple myeloma is a genetically heterogeneous cancer of the bone marrow plasma cells (PC). Distinct myeloma transcriptome profiles are primarily driven by myeloma initiating events (MIE) and converge into a mutually exclusive overexpression of the CCND1 and CCND2 oncogenes. Here, with reference to their normal counterparts, we find that myeloma PC enhanced chromatin accessibility combined with paired transcriptome profiling can classify MIE-defined genetic subgroups. Across and within different MM genetic subgroups, we ascribe regulation of genes and pathways critical for myeloma biology to unique or shared, developmentally activated or de novo formed candidate enhancers. Such enhancers co-opt recruitment of existing transcription factors, which although not transcriptionally deregulated per se, organise aberrant gene regulatory networks that help identify myeloma cell dependencies with prognostic impact. Finally, we identify and validate the critical super-enhancer that regulates ectopic expression of CCND2 in a subset of patients with MM and in chronic lymphocytic leukemia.


Subject(s)
Carcinogenesis/genetics , Cyclin D1/genetics , Cyclin D2/genetics , Gene Expression Regulation, Neoplastic , Leukemia, Lymphocytic, Chronic, B-Cell/genetics , Multiple Myeloma/genetics , Transcriptome , Bone Marrow Cells/metabolism , Bone Marrow Cells/pathology , Carcinogenesis/metabolism , Carcinogenesis/pathology , Case-Control Studies , Cell Line, Tumor , Chromatin/chemistry , Chromatin/metabolism , Cyclin D1/metabolism , Cyclin D2/metabolism , Enhancer Elements, Genetic , Gene Expression Profiling , Gene Regulatory Networks , Histone-Lysine N-Methyltransferase/genetics , Histone-Lysine N-Methyltransferase/metabolism , Humans , Leukemia, Lymphocytic, Chronic, B-Cell/metabolism , Leukemia, Lymphocytic, Chronic, B-Cell/mortality , Leukemia, Lymphocytic, Chronic, B-Cell/pathology , Multiple Myeloma/metabolism , Multiple Myeloma/mortality , Multiple Myeloma/pathology , Plasma Cells/metabolism , Plasma Cells/pathology , Proto-Oncogene Proteins c-maf/genetics , Proto-Oncogene Proteins c-maf/metabolism , Repressor Proteins/genetics , Repressor Proteins/metabolism , Survival Analysis
16.
Biomolecules ; 11(7)2021 07 15.
Article in English | MEDLINE | ID: mdl-34356658

ABSTRACT

MAF is a transcription factor that may act either as a tumor suppressor or as an oncogene, depending on cell type. We have shown previously that the overexpressed miR-1290 influences MAF protein levels in LSCC (laryngeal squamous cell carcinoma) cell lines. In this study, we shed further light on the interaction between miR-1290 and MAF, as well as on cellular MAF protein localization in LSCC. We confirmed the direct interaction between miR-1290 and MAF 3'UTR by a dual-luciferase reporter assay. In addition, we used immunohistochemistry staining to analyze MAF protein distribution and observed loss of MAF nuclear expression in 58% LSCC samples, of which 10% showed complete absence of MAF, compared to nuclear and cytoplasmatic expression in 100% normal mucosa. Using TCGA data, bisulfite pyrosequencing and CNV analysis, we excluded the possibility that loss-of-function mutations, promoter region DNA methylation or CNV are responsible for MAF loss in LSCC. Finally, we identified genes involved in the regulation of apoptosis harboring the MAF binding motif in their promoter region by applied FIMO and DAVID GO analysis. Our results highlight the role of miR-1290 in suppressing MAF expression in LSCC. Furthermore, MAF loss or mislocalization in FFPE LSCC tumor samples might suggest that MAF acts as a LSCC tumor suppressor by regulating apoptosis.


Subject(s)
Head and Neck Neoplasms/genetics , MicroRNAs/genetics , Proto-Oncogene Proteins c-maf/genetics , Squamous Cell Carcinoma of Head and Neck/genetics , 3' Untranslated Regions , Aged , Cell Line, Tumor , Cell Nucleus/genetics , Cell Nucleus/metabolism , DNA Methylation , Female , Gene Dosage , Gene Expression Regulation, Neoplastic , Head and Neck Neoplasms/pathology , Humans , Male , Middle Aged , Mutation , Promoter Regions, Genetic , Proto-Oncogene Proteins c-maf/metabolism , Squamous Cell Carcinoma of Head and Neck/pathology
17.
JNCI Cancer Spectr ; 5(4)2021 08.
Article in English | MEDLINE | ID: mdl-34377934

ABSTRACT

Background: The Adjuvant Zoledronic Acid (ZA) study in early breast cancer (AZURE) showed correlation between a nonamplified MAF gene in the primary tumor and benefit from adjuvant ZA. Adverse ZA outcomes occurred in MAF-amplified patients. NSABP B-34 is a validation study. Methods: A retrospective analysis of MAF gene status in NSABP B-34 was performed. Eligible patients were randomly assigned to standard adjuvant systemic treatment plus 3 years oral clodronate (1600 mg/daily) or placebo. Tumors were tested for MAF gene amplification and analyzed for their relationship to clodronate for disease-free survival (DFS) and overall survival (OS) in MAF nonamplified patients. All statistical tests were 2-sided . Results: MAF status was assessed in 2533 available primary tumor samples from 3311 patients. Of these, 37 withdrew consent; in 77 samples, no tumor was found; 536 assays did not meet quality standards, leaving 1883 (77.8%) evaluable for MAF assay by fluorescence in situ hybridization (947 from placebo and 936 from clodronate arms). At 5 years, in MAF nonamplified patients receiving clodronate, DFS improved by 30% (hazard ratio = 0.70, 95% confidence interval = 0.51 to 0.94; P = .02). OS improved at 5 years (hazard ratio = 0.59, 95% confidence interval = 0.37 to 0.93; P = .02) remaining statistically significant for clodronate throughout study follow-up. Conversely, adjuvant clodronate in women with MAF-amplified tumors was not associated with benefit but rather possible harm in some subgroups. Association between MAF status and menopausal status was not seen. Conclusions: Nonamplified MAF showed statistically significant benefits (DFS and OS) with oral clodronate, supporting validation of the AZURE study.


Subject(s)
Bone Density Conservation Agents/administration & dosage , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Clodronic Acid/administration & dosage , Gene Amplification , Proto-Oncogene Proteins c-maf/genetics , Administration, Oral , Bone Density Conservation Agents/adverse effects , Breast Neoplasms/mortality , Breast Neoplasms/pathology , Chemotherapy, Adjuvant , Confidence Intervals , Disease-Free Survival , Double-Blind Method , Female , Humans , In Situ Hybridization, Fluorescence , Injections, Intravenous , Middle Aged , Placebos/administration & dosage , Retrospective Studies , Zoledronic Acid/administration & dosage , Zoledronic Acid/adverse effects
18.
Exp Eye Res ; 209: 108663, 2021 08.
Article in English | MEDLINE | ID: mdl-34119483

ABSTRACT

Age is a major risk factor for cataract (ARC). However, the influence of aging on the lens transcriptome is under studied. Lens epithelial (LEC) and fiber cells (LFC) were isolated from young (3 month old) and aged (24 month old) C57BL/6J mice, and the transcriptome elucidated via RNAseq. EdgeR estimated differential gene expression in pairwise contrasts, and Advaita's Ipathway guide and custom R scripts were used to evaluate the potential biological significance of differentially expressed genes (DEGs). This analysis revealed age-dependent decreases in lens differentiation marker expression in both LECs and LFCs, with gamma crystallin transcripts downregulating nearly 50 fold in aged LFCs. The expression of the transcription factors Hsf4 and Maf, which are known to activate lens fiber cell preferred genes, are downregulated, while FoxE3, which represses gamma crystallin expression, is upregulated in aged fibers. Aged LECs upregulate genes controlling the immune response, complement pathways, and cellular stress responses, including glutathione peroxidase 3 (Gpx3). Aged LFCs exhibit broad changes in the expression of genes regulating cell communication, and upregulate genes involved in antigen processing/presentation and cholesterol metabolism, while changes in the expression of mitochondrial respiratory chain genes are consistent with mitochondrial stress, including upregulation of NDufa4l2, which encodes an alternate electron transport chain protein. However, age did not profoundly affect the response of LECs to injury as both young and aged LECs upregulate inflammatory gene signatures at 24 h post injury to similar extents. These RNAseq profiles provide a rich data set that can be mined to understand the genetic regulation of lens aging and how this impinges on the pathophysiology of age related cataract.


Subject(s)
Aging/genetics , Cataract/genetics , Forkhead Transcription Factors/genetics , Gene Expression Regulation , Heat Shock Transcription Factors/genetics , Proto-Oncogene Proteins c-maf/genetics , Transcriptome/genetics , Animals , Cataract/metabolism , Disease Models, Animal , Female , Forkhead Transcription Factors/biosynthesis , Heat Shock Transcription Factors/biosynthesis , Heat-Shock Proteins , Lens, Crystalline/metabolism , Male , Mice , Mice, Inbred C57BL , Proto-Oncogene Proteins c-maf/biosynthesis , RNA/genetics , gamma-Crystallins/biosynthesis , gamma-Crystallins/genetics
19.
Biol Reprod ; 105(4): 958-975, 2021 10 11.
Article in English | MEDLINE | ID: mdl-34007995

ABSTRACT

Testis differentiation is initiated when Sry in pre-Sertoli cells directs the gonad toward a male-specific fate. Sertoli cells are essential for testis development, but cell types within the interstitial compartment, such as immune and endothelial cells, are also critical for organ formation. Our previous work implicated macrophages in fetal testis morphogenesis, but little is known about genes underlying immune cell development during organogenesis. Here, we examine the role of the immune-associated genes Mafb and Maf in mouse fetal gonad development, and we demonstrate that deletion of these genes leads to aberrant hematopoiesis manifested by supernumerary gonadal monocytes. Mafb; Maf double knockout embryos underwent initial gonadal sex determination normally, but exhibited testicular hypervascularization, testis cord formation defects, Leydig cell deficit, and a reduced number of germ cells. In general, Mafb and Maf alone were dispensable for gonad development; however, when both genes were deleted, we observed significant defects in testicular morphogenesis, indicating that Mafb and Maf work redundantly during testis differentiation. These results demonstrate previously unappreciated roles for Mafb and Maf in immune and vascular development and highlight the importance of interstitial cells in gonadal differentiation.


Subject(s)
MafB Transcription Factor/genetics , Myeloid Cells/metabolism , Organogenesis/genetics , Proto-Oncogene Proteins c-maf/genetics , Testis/embryology , Animals , Embryo, Mammalian/embryology , MafB Transcription Factor/metabolism , Male , Mice , Proto-Oncogene Proteins c-maf/metabolism
20.
Cell Commun Signal ; 19(1): 24, 2021 02 24.
Article in English | MEDLINE | ID: mdl-33627137

ABSTRACT

BACKGROUND: The oncogenic transcript factor c-Maf is stabilized by the deubiquitinase Otub1 and promotes myeloma cell proliferation and confers to chemoresistance. Inhibition of the Otub1/c-Maf axis is a promising therapeutic target, but there are no inhibitors reported on this specific axis. METHODS: A luciferase assay was applied to screen potential inhibitors of Otub1/c-Maf. Annexin V staining/flow cytometry was applied to evaluate cell apoptosis. Immunoprecipitation was applied to examine protein ubiquitination and interaction. Xenograft models in nude mice were used to evaluate anti-myeloma activity of AVT. RESULTS: Acevaltrate (AVT), isolated from Valeriana glechomifolia, was identified based on a bioactive screen against the Otub1/c-Maf/luciferase system. AVT disrupts the interaction of Otub1/c-Maf thus inhibiting Otub1 activity and leading to c-Maf polyubiquitination and subsequent degradation in proteasomes. Consistently, AVT inhibits c-Maf transcriptional activity and downregulates the expression of its target genes key for myeloma growth and survival. Moreover, AVT displays potent anti-myeloma activity by triggering myeloma cell apoptosis in vitro and impairing myeloma xenograft growth in vivo but presents no marked toxicity. CONCLUSIONS: The natural product AVT inhibits the Otub1/c-Maf axis and displays potent anti-myeloma activity. Given its great safety and efficacy, AVT could be further developed for MM treatment. Video Abstract.


Subject(s)
Antineoplastic Agents, Phytogenic/therapeutic use , Cysteine Endopeptidases/metabolism , Cysteine Proteinase Inhibitors/therapeutic use , Iridoids/therapeutic use , Multiple Myeloma/drug therapy , Proto-Oncogene Proteins c-maf/antagonists & inhibitors , Animals , Antineoplastic Agents, Phytogenic/pharmacology , Apoptosis/drug effects , Cell Line , Cell Survival/drug effects , Cysteine Endopeptidases/genetics , Cysteine Proteinase Inhibitors/pharmacology , Female , Humans , Iridoids/pharmacology , Mice, Inbred BALB C , Mice, Nude , Multiple Myeloma/metabolism , Multiple Myeloma/pathology , Proto-Oncogene Proteins c-maf/genetics , Proto-Oncogene Proteins c-maf/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...