Isora V Sernandez's research while affiliated with Spanish National Centre for Cardiovascular Research and other places

What is this page?


This page lists the scientific contributions of an author, who either does not have a ResearchGate profile, or has not yet added these contributions to their profile.

It was automatically created by ResearchGate to create a record of this author's body of work. We create such pages to advance our goal of creating and maintaining the most comprehensive scientific repository possible. In doing so, we process publicly available (personal) data relating to the author as a member of the scientific community.

If you're a ResearchGate member, you can follow this page to keep up with this author's work.

If you are this author, and you don't want us to display this page anymore, please let us know.

Publications (10)


Fig 1. Characterization of R26 +/AID Cd19 +/Cre mice. (A) Increased expression of Aicda mRNA in B cells from R26 +/AID Cd19 +/Cre mice. mRNA levels of R26-AID and total Aicda was measured by RT-qPCR in naïve (left) and in LPS+IL4 activated B (right) cells from R26 +/AID Cd19 +/Cre (red bars) and R26 +/+ Cd19 +/Cre control mice (blue bars). Values are relative to levels in naïve (left) or activated (right) B cells from R26 +/AID Cd19 +/Cre mice; mean of two experiments is shown; n = 6 per group. (B) Increased AID protein in B cells from R26 +/AID Cd19 +/Cre mice. Total lysates of naïve or LPS+IL4 activated B cells from R26 +/AID Cd19 +/Cre and R26 +/+ Cd19 +/Cre mice were analysed by western blot with a monoclonal anti-AID antibody. Incubation with an anti-tubulin antibody is shown as loading control. (C) Quantification of AID protein in LPS+IL4 activated B cells by western blot. Total lysates of activated B cells from 4 individual mice of each genotype, R26 +/AID Cd19 +/Cre and R26 +/+ Cd19 +/Cre and 1 Aicda -/-were analyzed by western blot as shown in (B). Values are relative to R26 +/+ Cd19 +/Cre and normalized to actin or tubulin. (D) Increased CSR in R26 +/AID Cd19 +/Cre mice. Representative FACS analysis of CSR to IgG1 in LPS+IL4 activated B cells from R26 +/AID Cd19 +/Cre and R26 +/+ Cd19 +/Cre mice (left panel). Quantification is shown on the right. Each dot represents an individual mouse (n = 11 mice per group). (E) Increased immunization response in R26 +/AID Cd19 +/Cre mice. Representative FACS analysis of GC (Fas+GL7+) B cells (upper panels) and CSR to IgG1 (lower panels) in non-immunized R26 +/+ Cd19 +/Cre (control, PBS) and SRBC-immunized R26 +/ + Cd19 +/Cre and R26 +/AID Cd19 +/Cre mice after B220+ gating. (F) Quantification of Fas+GL7+ GC B cells (upper panel) and CSR to IgG1 (lower graphs) on B220+-gated (lower left) and B220+Fas+GL7+-gated B cells (lower right) from the immunization experiment shown in (E). Each dot represents an individual mouse (n = 2-5 mice per group). � p�0.05; �� p�0.005; ���� p�0.0001; two-tailed t-test.
Fig 2. R26 +/AID Cd19 +/Cre mice do not develop lymphoma. Kaplan-Meier survival analysis (left) and representative H&E staining of spleen sections (right) of (A) R26 +/+ Cd19 +/Cre (n = 51) and R26 +/AID Cd19 +/Cre mice (n = 44) mice and (B) R26 +/+ Vav +/cre (n = 23) and R26 +/AID Vav +/cre (n = 35) mice. (C) Kaplan-Meier survival analysis of R26 +/+ Cd19 +/Cre Tp53 -/-(n = 40) and R26 +/AID Cd19 +/Cre Tp53 -/-(n = 49) and representative H&E staining of thymus T cell lymphomas found in both groups of mice. Mice were followed for 80 weeks (A-B) or at humane endpoint (C). p>0.05 in all cases; log-rank Mantel-Cox test. Magnification is 5x (left) and 20x (right); scale bar is 200μm.
Fig 3. Increased SHM in BER deficient mice. Mutation analysis of IgH Sμ region in naïve and GC B cells from R26 +/+ Cd19 +/cre Ung +/-, R26 +/AID Cd19 +/cre Ung +/-, R26 +/+ Cd19 +/cre Ung -/-, R26 +/AID Cd19 +/cre Ung -/-and Aicda -/-mice by PCR-Seq. (A) Total mutation frequency and (B) frequency of transition and transversion mutations at C/G lying in WRCY/RGYW hotspots. (n = 2, each composed of a pool of 3 mice; � p � 0.05; two-tailed t-test; error bars represent SD).
Fig 5. AID contributes to intratumor heterogeneity. (A) Average transition mutation frequency at total C/G pairs and at C/G within WRC/GYW hotspots in tumors from R26 +/+ Cd19 +/cre Ung -/-and R26 +/AID Cd19 +/cre Ung -/-mice as measured by whole exome sequencing (Two-tailed γ 2 test; all comparisons -Total C/G vs WRC/GYW within the same genotype; Total C/G AID WT vs AID KI; WRC/GYW AID WT vs AID KI-, p <2.2x10 -16 ). (B) Number (upper panel) and proportion (lower panel) of clonal and subclonal variants in each of the seven R26 +/+ Cd19 +/cre Ung -/-(WT) and eight R26 +/AID Cd19 +/cre Ung -/-(KI) tumors analyzed. Piecharts depict the proportion of clonal and subclonal variants of all the tumors analyzed pooled by genotype (Two-tailed Fisher test; ���� p �10 −4 ). Inset panels show a representative example of the intratumor heterogeneity in each genotype. Clusters as defined by SciClone are depicted in different colors. Clonal variants are those belonging to the cluster with the highest variant allele frequency (VAF; orange clusters in inset panels), usually between 40-50%. Subclonal variants are those belonging to clusters with a lower VAF than that of the clonal cluster. Density plots depict the posterior predictive density, a surrogate of the accuracy of the prediction model assigning variants to the different clusters (green lines represent the diploid model; grey continuous and discontinuous lines represent the model fit of each cluster and each variant, respectively). Tumor coverage refers to the number of reads supporting each variant. Genes that are frequently mutated in human lymphoma tumors are indicated in black. https://doi.org/10.1371/journal.pgen.1008960.g005
Fig 6. AID broadens the functional impact of oncogenic events in UNG deficient tumors. (A) Cancer-related genes found mutated in tumors from R26 +/+ Cd19 +/cre Ung -/-and R26 +/AID Cd19 +/cre Ung -/-mice. Upper panel shows total number of variants identified in all cancer-related genes in each of the tumors; color code defines the functional impact of the variants as annotated by Variant Effect Predictor (VEP; see methods section for details). Lower panel represents presence (colored box) or absence (empty box) of variants in each of the cancer-related genes for each of the analyzed tumors. For simplicity, only mutated genes found in > = 2 tumors per group are shown; a full list is included in S3 Table. Green indicates genes exclusively mutated in R26 +/+ Cd19 +/cre Ung -/-tumors; orange indicates genes exclusively mutated in R26 +/AID Cd19 +/cre Ung -/-tumors; blue indicates genes mutated in both. (B) Circos plot representation of the biological functions associated to all mutated cancer-related genes (Gene Ontology biological process database) in R26 +/+ Cd19 +/cre Ung -/-(left) and R26 +/AID Cd19 +/cre Ung -/-(right) tumors. (C) Number of genes (left) and p-value of the enrichment (right, EASE score -modified Fisher exact test-as provided by DAVID, see methods for details) of the common biological functions affected in both R26 +/
Interplay between UNG and AID governs intratumoral heterogeneity in mature B cell lymphoma
  • Article
  • Full-text available

December 2020

·

146 Reads

·

3 Citations

PLOS Genetics

PLOS Genetics

·

Ángel F. Álvarez-Prado

·

·

[...]

·

Most B cell lymphomas originate from B cells that have germinal center (GC) experience and bear chromosome translocations and numerous point mutations. GC B cells remodel their immunoglobulin (Ig) genes by somatic hypermutation (SHM) and class switch recombination (CSR) in their immunoglobulin (Ig) genes. Activation Induced Deaminase (AID) initiates CSR and SHM by generating U:G mismatches on Ig DNA that can then be processed by Uracyl-N-glycosylase (UNG). AID promotes collateral damage in the form of chromosome translocations and off-target SHM, however, the exact contribution of AID activity to lymphoma generation and progression is not completely understood. Here we show using a conditional knock-in strategy that AID supra-activity alone is not sufficient to generate B cell transformation. In contrast, in the absence of UNG, AID supra-expression increases SHM and promotes lymphoma. Whole exome sequencing revealed that AID heavily contributes to lymphoma SHM, promoting subclonal variability and a wider range of oncogenic variants. Thus, our data provide direct evidence that UNG is a brake to AID-induced intratumoral heterogeneity and evolution of B cell lymphoma.

Download
Share

Interplay between UNG and AID governs intratumoral heterogeneity in mature B cell lymphoma

June 2020

·

93 Reads

Most B cell lymphomas originate from B cells that have germinal center (GC) experience and bear chromosome translocations and numerous point mutations. GCs B cells remodel their immunoglobulin (Ig) genes by somatic hypermutation (SHM) and class switch recombination (CSR) in their immunoglobulin (Ig) genes. Activation Induced Deaminase (AID) initiates CSR and SHM by generating U:G mismatches on Ig DNA that can then be processed by Uracyl-N-glycosylase (UNG). AID promotes collateral damage in the form of chromosome translocations and off-target SHM, however, the exact contribution of AID activity to lymphoma generation and progression is not completely understood. Here we show using a conditional knock-in strategy that AID supra-activity alone is not sufficient to generate B cell transformation. In contrast, in the absence of UNG, AID supra-expression increases SHM and promotes lymphoma. Whole exome sequencing revealed that AID heavily contributes to lymphoma SHM, promoting subclonal variability and a wider range of oncogenic variants. Thus, our data provide direct evidence that UNG is a brake to AID-induced intratumoral heterogeneity and evolution of B cell lymphoma.




Table 1. Analysis of AID mutagenic activity by Sanger sequencing. 
Figure 1. Inflammation-induced AID expression does not contribute to carcinogenesis. A-C AID expression was analyzed in colon and pancreatic human cell lines and in pancreas explants from C57BL/6 mice. Samples were treated as indicated with 50 ng/ml TNF-a. (A) qRT-PCR analysis of AID expression in LoVo and SW480 colon cell lines. n = 5 (LoVo); 3 (SW480). **P-value: LoVo: 0.0017; SW480: 0.0079. (B) qRT-PCR analysis of AID expression in AsPC and PaTU-8988S pancreatic cell lines (n = 2). *P-value: AsPC: 0.0369; PaTU-8988S: 0.0119. (C) qRT-PCR analysis of AID expression in pancreatic explants from wild-type mice (n = 3). *P = 0.0242. D AID À/À or AID +/À mice were treated with 3% DSS for 10 cycles, and colonic sections were analyzed by histologic inspection after H/E staining. Graphs represent mean frequency values of adenoma and adenocarcinoma lesions of five independent experiments. n = 28 (AID À/À males); 35 (females); 23 (AID +/À males); 25 (females). P-value: male: 0.8; female: 0.246. Data information: All data are mean values AE SEM. Statistical differences were analyzed by two-tailed unpaired Student's t-test.
Figure 2. Heterologous AID expression does not promote carcinoma development.  
AID-expressing epithelium is protected from oncogenic transformation by an NKG2D surveillance pathway

August 2015

·

90 Reads

·

6 Citations

EMBO Molecular Medicine

Activation-induced deaminase (AID) initiates secondary antibody diversification in germinal center B cells, giving rise to higher affinity antibodies through somatic hypermutation (SHM) or to isotype-switched antibodies through class switch recombination (CSR). SHM and CSR are triggered by AID-mediated deamination of cytosines in immunoglobulin genes. Importantly, AID activity in B cells is not restricted to Ig loci and can promote mutations and pro-lymphomagenic translocations, establishing a direct oncogenic mechanism for germinal center-derived neoplasias. AID is also expressed in response to inflammatory cues in epithelial cells, raising the possibility that AID mutagenic activity might drive carcinoma development. We directly tested this hypothesis by generating conditional knock-in mouse models for AID overexpression in colon and pancreas epithelium. AID overexpression alone was not sufficient to promote epithelial cell neoplasia in these tissues, in spite of displaying mutagenic and genotoxic activity. Instead, we found that heterologous AID expression in pancreas promotes the expression of NKG2D ligands, the recruitment of CD8(+) T cells, and the induction of epithelial cell death. Our results indicate that AID oncogenic potential in epithelial cells can be neutralized by immunosurveillance protective mechanisms. © 2015 The Authors. Published under the terms of the CC BY 4.0 license.



Estrogen directly activates AID transcription and function

February 2009

·

376 Reads

·

127 Citations

The immunological targets of estrogen at the molecular, humoral, and cellular level have been well documented, as has estrogen's role in establishing a gender bias in autoimmunity and cancer. During a healthy immune response, activation-induced deaminase (AID) deaminates cytosines at immunoglobulin (Ig) loci, initiating somatic hypermutation (SHM) and class switch recombination (CSR). Protein levels of nuclear AID are tightly controlled, as unregulated expression can lead to alterations in the immune response. Furthermore, hyperactivation of AID outside the immune system leads to oncogenesis. Here, we demonstrate that the estrogen-estrogen receptor complex binds to the AID promoter, enhancing AID messenger RNA expression, leading to a direct increase in AID protein production and alterations in SHM and CSR at the Ig locus. Enhanced translocations of the c-myc oncogene showed that the genotoxicity of estrogen via AID production was not limited to the Ig locus. Outside of the immune system (e.g., breast and ovaries), estrogen induced AID expression by >20-fold. The estrogen response was also partially conserved within the DNA deaminase family (APOBEC3B, -3F, and -3G), and could be inhibited by tamoxifen, an estrogen antagonist. We therefore suggest that estrogen-induced autoimmunity and oncogenesis may be derived through AID-dependent DNA instability.



Estrogen directly activates AID transcription and function

January 2009

·

210 Reads

·

217 Citations

The immunological targets of estrogen at the molecular, humoral, and cellular level have been well documented, as has estrogen's role in establishing a gender bias in autoimmunity and cancer. During a healthy immune response, activation-induced deaminase (AID) deaminates cytosines at immunoglobulin (Ig) loci, initiating somatic hypermutation (SHM) and class switch recombination (CSR). Protein levels of nuclear AID are tightly controlled, as unregulated expression can lead to alterations in the immune response. Furthermore, hyperactivation of AID outside the immune system leads to oncogenesis. Here, we demonstrate that the estrogen–estrogen receptor complex binds to the AID promoter, enhancing AID messenger RNA expression, leading to a direct increase in AID protein production and alterations in SHM and CSR at the Ig locus. Enhanced translocations of the c-myc oncogene showed that the genotoxicity of estrogen via AID production was not limited to the Ig locus. Outside of the immune system (e.g., breast and ovaries), estrogen induced AID expression by >20-fold. The estrogen response was also partially conserved within the DNA deaminase family (APOBEC3B, -3F, and -3G), and could be inhibited by tamoxifen, an estrogen antagonist. We therefore suggest that estrogen-induced autoimmunity and oncogenesis may be derived through AID-dependent DNA instability.


Haploinsufficiency of Activation-Induced Deaminase for Antibody Diversification and Chromosome Translocations both In Vitro and In Vivo

February 2008

·

163 Reads

·

55 Citations

The humoral immune response critically relies on the secondary diversification of antibodies. This diversification takes places through somatic remodelling of the antibody genes by two molecular mechanisms, Class Switch Recombination (CSR) and Somatic Hypermutation (SHM). The enzyme Activation Induced Cytidine Deaminase (AID) initiates both SHM and CSR by deaminating cytosine residues on the DNA of immunoglobulin genes. While crucial for immunity, AID-catalysed deamination is also the triggering event for the generation of lymphomagenic chromosome translocations. To address whether restricting the levels of AID expression in vivo contributes to the regulation of its function, we analysed mice harbouring a single copy of the AID gene (AID(+/-)). AID(+/-) mice express roughly 50% of normal AID levels, and display a mild hyperplasia, reminiscent of AID deficient mice and humans. Moreover, we found that AID(+/-) cells have an impaired competence for CSR and SHM, which indicates that AID gene dose is limiting for its physiologic function. We next evaluated the impact of AID reduction in AID(+/-) mice on the generation of chromosome translocations. Our results show that the frequency of AID-promoted c-myc/IgH translocations is reduced in AID(+/-) mice, both in vivo and in vitro. Therefore, AID is haploinsufficient for antibody diversification and chromosome translocations. These findings suggest that limiting the physiologic levels of AID expression can be a regulatory mechanism that ensures an optimal balance between immune proficiency and genome integrity.

Citations (5)


... Thus, deaminases acting on DNA strongly contribute to the generation of genetically distinct subclonal populations within a tumour. Examples include, among others, APOBEC3B in breast cancer [63] or AID in B cell lymphoma [64]. Furthermore, changes in the deaminase-associated mutational signatures during tumour progression have been identified and proposed as a model of tumour growth [65]. ...

Reference:

Unifying Different Cancer Theories in a Unique Tumour Model: Chronic Inflammation and Deaminases as Meeting Points
Interplay between UNG and AID governs intratumoral heterogeneity in mature B cell lymphoma
PLOS Genetics

PLOS Genetics

... To address the contribution of AID activity to B cell lymphomagenesis we made use of a conditional mouse model for AID expression [52]. A cassette containing the Aicda and EGFP cDNAs separated by an internal ribosomal entry site (IRES) was placed into the endogenous Rosa26 locus preceded by a floxed transcriptional stop sequence (R26 +/AID mice, S1A Fig, [52]). ...

AID-expressing epithelium is protected from oncogenic transformation by an NKG2D surveillance pathway

EMBO Molecular Medicine

... E2 sup ports differentiation of HSCs [13][14][15] , lymphoid and myeloid immune cells, including marginal zone (MZ) B cells, follicular helper T (T FH ) cells, germinal center (GC) B cells, MBCs and granulocytes, all expressing estrogen receptors ERα and ERβ [13][14][15][16][17][18][19][20][21][22][23][24][25][26] . E2 also boosts B cell AID and BLIMP1 expression, enabling SHM/CSR and PC differentiation [27][28][29][30] . THX mice reconstitute a human immune system, including peripheral lymph nodes (LNs), Peyer's patches and human thymic epithelial cells (huTECs). ...

Estrogen directly activates AID transcription and function
Journal of Experimental Medicine (JEM)

Journal of Experimental Medicine (JEM)

... Another explanation is hormonal: hormones, such as estrogen, which females produce in larger quantities than males, help to defend against coronaviruses like MERS-CoV, SARS-CoV and SARS-CoV-2. There are various possible explanations of estrogen protective action mechanism (19)(20)(21)(22)(23)(24). Among the effects of estrogen in the immune defence, there is an influence on adaptive immunity fighters, T-cells and B-cells, by impairing negative selection of high affinity auto-reactive B cells, modulating B cell function and leading to Th2 response (25), and induction of T cell homing by enhancing the expression of CCR5, a homing marker Please note that this is an unedited version of the manuscript that has been accepted for publication. ...

Estrogen directly activates AID transcription and function
Journal of Cell Biology (JCB)

Journal of Cell Biology (JCB)

... In order for antibody isotype differentiation to efficiently occur, the two copies of the Aicda gene need to be expressed and the resulting AID protein has to target the S regions at the Igh locus. Therefore, Aicda haploinsufficiency and reduced AID expression or targeting result in CSR deficiency 255,256 . To first exclude the possibility that ZMYND8 is involved in Aicda transcription, Aicda mRNA levels in primary B cells were quantified by RT-qPCR (quantitative reverse transcription PCR). ...

Haploinsufficiency of Activation-Induced Deaminase for Antibody Diversification and Chromosome Translocations both In Vitro and In Vivo
PLOS ONE

PLOS ONE