Beatrice Biferali’s research while affiliated with San Raffaele Scientific Institute and other places

What is this page?


This page lists works of an author who doesn't have a ResearchGate profile or hasn't added the works to their profile yet. It is automatically generated from public (personal) data to further our legitimate goal of comprehensive and accurate scientific recordkeeping. If you are this author and want this page removed, please let us know.

Publications (8)


Long non-coding RNAs and their role in muscle regeneration
  • Literature Review

April 2024

·

15 Reads

·

1 Citation

Current Topics in Developmental Biology

Beatrice Biferali

·

·

·

Davide Gabellini

Fig. 1. DUX4 and DUX4-r associate to and activate different gene sets. (A) Heatmap showing the 500 most variable genes in REH cells expressing EV, wt DUX4, DUX4-IGH, or DUX4-del50. (B) Venn diagram displaying (left) overlap between genes up-regulated in the present study and in DUX4-r B-ALL patients (EGAS00001000654) and (right) overlap between genes down-regulated in the present study and in DUX4-r B-ALL patients (EGAS00001000654). Twotailed Fisher's exact test. (C) Functional families enriched in the overlap between: genes positively (left) or negatively (right) regulated in DUX4-r B-ALL patients and by DUX4-r in REH cells. Dot size is proportional to the percentage of modulated genes in each pathway. (D) Heatmap showing the 500 most variable repetitive elements in REH cells expressing EV, wt DUX4, DUX4-IGH, or DUX4-del50. (E) Top: Venn diagram displaying the overlap between CUT&Tag peaks in wt DUX4-, DUX4-IGH-, or DUX4-del50-expressing REH cells. Bottom: CUT&Tag profile plots of wt DUX4 (red) and DUX4-IGH (cyan) peaks on wt DUX4 (left) or DUX4-r (right) direct transcriptional targets. (F) De novo motif calling of peaks selectively associated to wt DUX4 (DUX4 only) or DUX4-IGH and DUX4-del50 (DUX4-IGH/DUX4-del50 overlap). (G) CUT&Tag profile plot of H3K27Ac at wt DUX4 (maroon/red) or DUX4-IGH (blue/ teal) direct targets in the absence (left) or presence (right) of Dox. Wilcoxon rank sum test to compare the signal of H3K27Ac signal between −Dox and +Dox. (H) Cleavage Under Targeted Accessible Chromatin (CUTAC) profile plot at wt DUX4 (maroon/red) or DUX4-IGH (blue/teal) peaks on direct targets in the absence (left) or presence (right) of Dox. Wilcoxon rank sum test to compare the CUTAC signal between −Dox and +Dox conditions. See also fig. S1.
Fig. 2. DUX4-r activates cell adhesion and migration, promoting cell proliferation. (A) HS5 bone marrow stroma adhesion (top) and transwell migration assays (bottom) schematics. (B) Adhesion of REH cells expressing EV, wt DUX4, or DUX4-r variants in −Dox/+Dox. Bar plots represent the means ± SD of three biological replicates (n = 3). Student's t test. ***P ≤ 0.001; ****P ≤ 0.0001. (C) Migration of REH cells expressing EV, wt DUX4, or DUX4-r variants in −Dox/ +Dox. Bar plots represent the means ± SD of three independent biological replicates (n = 3). Student's t test. **P ≤ 0.01 and ****P ≤ 0.0001. (D) Western blot validation of DUX4-r knockdown in NALM6 cells using vinculin as a loading control. (E) Adhesion (top) and migration (bottom) of control (shCTRL) or DUX4-r (shDUX4-r) knockdown NALM6 cells. Bar plots represent the means ± SD of three independent biological replicates (n = 3). Student's t test. **P ≤ 0.01; ****P ≤ 0.0001. (F) Real-time tumor spheroid quantification of REH cells expressing the indicated transgene. Curves represent the means ± SD of six biological replicates (n = 6). Two-way analysis of variance (ANOVA) with Bonferroni's correction to compare curves at the 45-hour time point. ****P ≤ 0.0001. (G) Real-time tumor spheroid quantification of control (shCTRL) or DUX4-r (shDUX4-r) knockdown NALM6 cells. Curves represent the means ± SD of six biological replicates (n = 6). Two-way ANOVA with Bonferroni's correction to compare curves at the 45-hour time point. ****P ≤ 0.0001. (H) Proliferation of REH cells expressing EV, wt DUX4, DUX4-IGH, or DUX4-del50. Curves represent the means ± SD of three independent biological replicates (n = 3). One-way ANOVA. **P ≤ 0.01; ***P ≤ 0.001. (I) Proliferation assay of control (shCTRL) or DUX4-r (shDUX4-r) knockdown NALM6 cells. Curves represent the means ± SD of three independent biological replicates (n = 3). One-way ANOVA. *P ≤ 0.05. See also fig. S2. ns, not significant; PBS, phosphatebuffered saline; FACS, fluorescence-activated cell sorting.
Fig. 3. DUX4-r activity is prominent in B-ALL cells. (A) RT-qPCR expression analysis of the indicated DUX4-r (top) or wt DUX4 (bottom, right) direct target genes in REH cells expressing EV, wt DUX4, DUX4-IGH, or DUX4-del50. Relative levels of wt DUX4/DUX4-r are also shown (bottom, left). Relative mRNA levels were normalized to glyceraldehyde-3-phosphate dehydrogenase (GAPDH). Bar plots represent the average of three independent biological replicates (n = 3). Error bars represent means ± SD. One-way ANOVA with multiple comparisons. *P ≤ 0.05; **P ≤ 0.01; ***P ≤ 0.001; ****P ≤ 0.0001. (B) RT-qPCR analysis of the same genes as in (A) in HEK cells expressing EV, wt DUX4, DUX4-IGH, or DUX4-del50. Relative mRNA levels were normalized to GAPDH. Bar plots represent the average of three independent biological replicates (n = 3). Error bars represent means ± SD. One-way ANOVA with multiple comparisons. ***P ≤ 0.001; ****P ≤ 0.0001. (C) RT-qPCR analysis of the same genes as in (A) and (B) in Jurkat cells expressing EV, wt DUX4, DUX4-IGH, or DUX4-del50. Relative mRNA levels were normalized to GAPDH. Bar plots represent the average of three independent biological replicates (n = 3). Error bars represent means ± SD. One-way ANOVA with multiple comparisons. ***P ≤ 0.001; ****P ≤ 0.0001. (D) CUT&Tag profile plot of genomic enrichment at (left) wt DUX4 on its direct transcriptional targets in human embryonic kidney 293 (HEK) (dark red) and REH (red) cells and (right) DUX4-IGH on its direct transcriptional targets in HEK (blue) and REH (teal) cells. Wilcoxon rank sum test was performed to compare the signal of wt DUX4 or DUX4-IGH between HEK and REH cells.
Fig. 4. DUX4-r selectively interacts with GTF2I. (A) Heatmap showing DUX4-r-selective interactors which are more expressed in REH cells. Protein levels were expressed as MS spectral counts. The heatmap reports unscaled MS/MS counts. (B) Tandem affinity purification followed by immunoblotting validation of selective GTF2I interaction with DUX4-IGH and DUX4-del50. Five percent of input and first elution and 10% of second elution were loaded and incubated with antibodies specific for GTF2I or wt DUX4 and DUX4-r. (C) Western blot validation of nuclear protein levels of GTF2I in REH, Jurkat, or REH cells using GAPDH as a loading control. (D) Densitometric analysis of signals in Fig. 3D using the ImageLab software (Bio-Rad, ver. 6.1). Bar plots represent the mean of three independent biological replicates (n = 3). Error bars represent ± SD. One-way ANOVA with multiple comparisons against REH. *P ≤ 0.05; ***P ≤ 0.001. (E) CUT&Tag profile plot of GTF2I enrichment on DUX4-r direct targets in HEK EV (red), REH EV (blue), or REH DUX4-IGH (teal) cells. Wilcoxon ranks sum test was performed to compare the signal of GTF2I between HEK EV, REH EV, and REH DUX4-IGH cells. (F) Heatmap showing the expression levels (FPKM, fragments per kilobase of transcript per million fragments mapped) of DUX4-r core genes in different DUX4-r B-ALL patients ordered based on decreasing GTF2I expression levels. The heatmap reports Z scores scaled by row. (G) Pearson's correlation plot showing the positive correlation between GTF2I expression levels and the mean activation of DUX4-r core genes in different DUX4-r B-ALL patients. The correlation coefficient (R) and P value are indicated. See also fig. S3.
Fig. 5. GTF2I is required for DUX4-r biological activity in vitro and in vivo. (A) RT-qPCR (left) and immunoblot (right) of GTF2I knockdown in REH DUX4-IGH (top) or NALM6 cells (bottom). Bar plots show means ± SD (n = 3). Student's t test. *P ≤ 0.05 and ****P ≤ 0.0001. (B) RT-qPCR of the indicated direct DUX4-r targets in REH DUX4-IGH (top) and NALM6 (bottom) CTRL or GTF2I knockdown. Bar plots show means ± SD (n = 3). One-way ANOVA with multiple comparisons. **P ≤ 0.01; ***P ≤ 0.001; ****P ≤ 0.0001. (C) Adhesion (top) and migration (bottom) of REH DUX4-IGH (left) and NALM6 (right) in shCTRL or shGTF2I cells. Bar plots show means ± SD (n = 3). Student's t test. **P ≤ 0.01; ***P ≤ 0.001; ****P ≤ 0.0001. (D) Real-time tumor spheroid quantification of REH DUX4-IGH plus CTRL or GTF2I knockdown. Curves show means ± SD (n = 6). Two-way ANOVA with Bonferroni's correction to compare curves at the 45-hour time point. ****P ≤ 0.0001. (E) Proliferation of REH DUX4-IGH plus CTRL or GTF2I knockdown. Curves represent the means ± SD (n = 3). One-way ANOVA. *P ≤ 0.05. (F) Apoptosis in CTRL or GTF2I knockdown REH DUX4-IGH (left) and NALM6 cells (right). Bar plots show means ± SD (n = 4). Student's t test. ***P ≤ 0.001 (r.l.u., relative luminescence unit). (G) RT-qPCR for GTF2I and the indicated DUX4-r direct targets in HEK cells expressing EV, wt DUX4, or DUX4-IGH plus EV or GTF2I. Bar plots show means ± SD (n = 3). One-way ANOVA with multiple comparisons was performed. **P ≤ 0.01; ***P ≤ 0.001. (H) CUT&Tag profile plot of DUX4-IGH on its direct targets in HEK DUX4-IGH cells transfected with EV (blue) or GTF2I (teal). Wilcoxon ranks sum test to compare the signal of DUX4-IGH between EV and GTF2I conditions. (I) Leukemia expansion in peripheral blood of NSG mice 7 and 14 days after NALM6-Lucia cell transplantation. Bar plots show the mean Luciferase signal ± SD of shCTRL or shGTF2I NALM6-Lucia mice. (n = 5). Two-way ANOVA. **P ≤ 0.01. (J) Kaplan-Meier survival analysis of the above mice. *P ≤ 0.05. See also fig. S4.
DUX4-r exerts a neomorphic activity that depends on GTF2I in acute lymphoblastic leukemia
  • Article
  • Full-text available

September 2023

·

45 Reads

Science Advances

·

Mara Salomé

·

Beatrice Biferali

·

[...]

·

Davide Gabellini

Translocations producing rearranged versions of the transcription factor double homeobox 4 (DUX4-r) are one of the most frequent causes of B cell acute lymphoblastic leukemia (B-ALL). DUX4-r retains the DNA binding domain of wild-type DUX4 but is truncated on the C-terminal transcription activation domain. The precise mechanism through which DUX4-r causes leukemia is unknown, and no targeted therapy is currently available. We found that the rearrangement leads to both a loss and a gain of function in DUX4-r. Loss of CBP/EP300 transcriptional coactivator interaction leads to an inability to bind and activate repressed chromatin. Concurrently, a gain of interaction with the general transcription factor 2 I (GTF2I) redirects DUX4-r toward leukemogenic targets. This neomorphic activity exposes an Achilles’ heel whereby DUX4-r–positive leukemia cells are exquisitely sensitive to GTF2I targeting, which inhibits DUX4-r leukemogenic activity. Our work elucidates the molecular mechanism through which DUX4-r causes leukemia and suggests a possible therapeutic avenue tailored to this B-ALL subtype.

Download

Prdm16-mediated H3K9 methylation controls fibro-adipogenic progenitors identity during skeletal muscle repair

June 2021

·

377 Reads

·

44 Citations

Science Advances

H3K9 methylation maintains cell identity orchestrating stable silencing and anchoring of alternate fate genes within the heterochromatic compartment underneath the nuclear lamina (NL). However, how cell type–specific genomic regions are specifically targeted to the NL is still elusive. Using fibro-adipogenic progenitors (FAPs) as a model, we identified Prdm16 as a nuclear envelope protein that anchors H3K9-methylated chromatin in a cell-specific manner. We show that Prdm16 mediates FAP developmental capacities by orchestrating lamina-associated domain organization and heterochromatin sequestration at the nuclear periphery. We found that Prdm16 localizes at the NL where it cooperates with the H3K9 methyltransferases G9a/GLP to mediate tethering and silencing of myogenic genes, thus repressing an alternative myogenic fate in FAPs. Genetic and pharmacological disruption of this repressive pathway confers to FAP myogenic competence, preventing fibro-adipogenic degeneration of dystrophic muscles. In summary, we reveal a druggable mechanism of heterochromatin perinuclear sequestration exploitable to reprogram FAPs in vivo.


Epigenetic regulation of Wnt7b expression by the cis-acting long noncoding RNA Lnc-Rewind in muscle stem cells

January 2021

·

105 Reads

·

25 Citations

eLife

Skeletal muscle possesses an outstanding capacity to regenerate upon injury due to the adult muscle stem cells (MuSCs) activity. This ability requires the proper balance between MuSCs expansion and differentiation which is critical for muscle homeostasis and contributes, if deregulated, to muscle diseases. Here, we functionally characterize a novel chromatin-associated lncRNA, Lnc-Rewind, which is expressed in murine MuSCs and conserved in human. We find that, in mouse, Lnc-Rewind acts as an epigenetic regulator of MuSCs proliferation and expansion by influencing the expression of skeletal muscle genes and several components of the WNT (Wingless-INT) signalling pathway. Among them, we identified the nearby Wnt7b gene as a direct Lnc-Rewind target. We show that Lnc-Rewind interacts with the G9a histone lysine methyltransferase and mediates the in cis repression of Wnt7b by H3K9me2 deposition. Overall, these findings provide novel insights into the epigenetic regulation of adult muscle stem cells fate by lncRNAs.


Epigenetic regulation of Wnt7b expression by the cis-acting long noncoding RNA lnc-Rewind in muscle stem cells

January 2020

·

82 Reads

Skeletal muscle possesses an outstanding capacity to regenerate upon injury due to the adult muscle stem cells (MuSCs) activity. This ability requires the proper balance between MuSCs expansion and differentiation which is critical for muscle homeostasis and contributes, if deregulated, to muscle diseases. Here, we functionally characterize a novel chromatin-associated lncRNA, lnc-Rewind, which is expressed in murine MuSCs and conserved in human. We find that, in mouse, lnc-Rewind acts as an epigenetic regulator of MuSCs proliferation and expansion by influencing the expression of skeletal muscle genes and several components of the WNT (Wingless-INT) signalling pathway. Among them, we identified the nearby Wnt7b gene as a direct lnc-Rewind target. We show that lnc- Rewind interacts with the G9a histone lysine methyltransferase and mediates the in cis repression of Wnt7b by H3K9me2 deposition. Overall, these findings provide novel insights into the epigenetic regulation of adult muscle stem cells fate by lncRNAs.


Statins interfere with the attachment of S. cerevisiae mtDNA to the inner mitochondrial membrane

November 2019

·

248 Reads

·

9 Citations

The 3-hydroxy-3-methylglutaryl-CoA reductase, a key enzyme of the mevalonate pathway for the synthesis of cholesterol in mammals (ergosterol in fungi), is inhibited by statins, a class of cholesterol lowering drugs. Indeed, statins are in a wide medical use, yet statins treatment could induce side effects as hepatotoxicity and myopathy in patients. We used Saccharomyces cerevisiae as a model to investigate the effects of statins on mitochondria. We demonstrate that statins are active in S.cerevisiae by lowering the ergosterol content in cells and interfering with the attachment of mitochondrial DNA to the inner mitochondrial membrane. Experiments on murine myoblasts confirmed these results in mammals. We propose that the instability of mitochondrial DNA is an early indirect target of statins.


Schematic illustration showing the known mediators that govern the interaction between FAPs, muscle stem cells (MuSCs), and the different immune cells in skeletal muscle homeostasis. Green arrows (TGF-β, IL-13, IL-4, and IL-15) indicate the molecules that positively regulate FAPs expansion. Blue arrows (IL-33, IL-6, Follistatin, IL-10, WISP1, MMP-14, and BMP-1) represent the molecules secreted by FAPs that act on the different cell targets. Red lines (TNF-α, IL-4, IL-13, and IL-15) show the factors that inhibit the fibro-adipogenic differentiation of FAPs.
Fibro–Adipogenic Progenitors Cross-Talk in Skeletal Muscle: The Social Network

August 2019

·

451 Reads

·

181 Citations

Skeletal muscle is composed of a large and heterogeneous assortment of cell populations that interact with each other to maintain muscle homeostasis and orchestrate regeneration. Although satellite cells (SCs) – which are muscle stem cells – are the protagonists of functional muscle repair following damage, several other cells such as inflammatory, vascular, and mesenchymal cells coordinate muscle regeneration in a finely tuned process. Fibro–adipogenic progenitors (FAPs) are a muscle interstitial mesenchymal cell population, which supports SCs differentiation during tissue regeneration. During the first days following muscle injury FAPs undergo massive expansion, which is followed by their macrophage-mediated clearance and the re-establishment of their steady-state pool. It is during this critical time window that FAPs, together with the other cellular components of the muscle stem cell niche, establish a dynamic network of interactions that culminate in muscle repair. A number of different molecules have been recently identified as important mediators of this cross-talk, and its alteration has been associated with different muscle pathologies. In this review, we will focus on the soluble factors that regulate FAPs activity, highlighting their roles in orchestrating the inter-cellular interactions between FAPs and the other cell populations that participate in muscle regeneration.


Citations (6)


... 126,127 Finally, the increasing knowledge regarding the key role of non-coding RNAs (e.g., miRNAs, circular RNAs, and long non-coding RNAs) on the post-transcriptional and epigenetic regulation of normal and abnormal muscle physiology, vastly enlarges the number of candidate targets for CRISPR-based gene modulation interventions. 128 ...

Reference:

On RNA-programmable gene modulation as a versatile set of principles targeting muscular dystrophies
Long non-coding RNAs and their role in muscle regeneration
  • Citing Chapter
  • April 2024

Current Topics in Developmental Biology

... Moreover, G9a plays a crucial role in myogenesis, and G9a inhibits myoblast differentiation through the methylation of MyoD (myogenic differentiation) at Lys104 (Ling et al. 2012) and of MEF2D (myocyte enhancer factor 2D) at Lys26 in vitro (Zhang et al. 2016). In vivo, global inhibition of G9a enzyme activity promoted muscle regeneration (Biferali et al. 2021), whereas Ehmt2 floxed Myod cre (Ehmt2 ΔmyoD ) mice presented normal skeletal muscle development (Zhang et al. 2016), possibly because myogenesis is a complicated process coordinated by multiple cell types, and G9a expressed in cell types other than myocytes also contributes. In this work, we show that G9a exacerbates muscular atrophy in both the aging model and denervation models (Figs. 3 and 6). ...

Prdm16-mediated H3K9 methylation controls fibro-adipogenic progenitors identity during skeletal muscle repair

Science Advances

... AsRNAs have been recognized as regulators of gene expression in multiple biological processes [7][8][9][10], and their aberrant expression/ function is associated with tumorigenesis [11,12]. AsRNAs may thus represent a rich and yet underexplored environment for the identification of cancer-relevant biomarkers and therapeutic targets. ...

Epigenetic regulation of Wnt7b expression by the cis-acting long noncoding RNA Lnc-Rewind in muscle stem cells

eLife

... We tested the toxicity of the compounds in S. cerevisiae W303 cells with a range of the concentrations. In Fig. 6, the effect of the tested compounds at the highest concentration (400 μM) for this model system is shown [42,43]. After treatment, the cultures' serial dilutions were spotted on YPD medium and grown at 28 • C. ...

Statins interfere with the attachment of S. cerevisiae mtDNA to the inner mitochondrial membrane

... FAPs have been identified as CD31 − /CD45 − /SM/C-2.6 − /PDGFR + or CD31 − /CD45 − /Integrin-7 − /Sca-1 + skeletal muscle resident cells that are normally quiescent but become activated upon injury, [31,32] and are a promising population to include in a construct due to their significant involvement in skeletal muscle regeneration. [33][34][35] Although FAPs are known to be one of the main contributors to fibrosis and fatty infiltration of muscle, [31,32,36] they are also a source of pro-myogenic and wound healing signaling. Recently, we showed that unsorted human myogenic progenitor cells (differentiated from human embryonic stem cells) which contained a population identified as FAPs exhibited better survival when implanted in TEMGs in vivo than the Pax-7-sorted myogenic population, [16] implicating FAPs potential benefits when included in a TEMG. ...

Fibro–Adipogenic Progenitors Cross-Talk in Skeletal Muscle: The Social Network

... Lastly, Yiweitang components, apart from increasing antioxidant enzymes, collagen I, and hydroxyproline levels [182], manifest DNA methylation and lncRNA and miRNA regulatory effects [185,187]. Although our study presents novel findings within its specific field, it aligns with existing research that explores how epigenetic alterations contribute to accelerated aging, such as microglia and muscle satellite cells (MuSCs) [196,197]. The microglial study mapped the transcriptional and epigenetic profiles, identified age-dependent microglia (ADEM) genes during the aging process, and correlated microglial behaviors with brain aging [196]. ...

Epigenetic Regulation of Muscle Stem Cells During Skeletal Muscle Regeneration and Disease
  • Citing Chapter
  • January 2019