Bio


I am a highly motivated and devoted scientist, deeply committed to advancing our understanding of the molecular foundations of cardiovascular disease while spearheading innovative therapeutic approaches and drug discovery. As a Postdoctoral Research Fellow at the Stanford Cardiovascular Institute, under the guidance of Dr. Joseph C. Wu, I am privileged to contribute to cutting-edge research. My work spans various disciplines, including multi-omics, molecular and cellular biology, tissue engineering, biochemistry, structural biology, and state-of-the-art imaging technologies. This holistic approach and sharp critical thinking equips me to untangle the complexities of cardiac diseases and innovate novel therapeutic strategies, particularly for rare cardiac conditions such as Transthyretin Cardiac Amyloidosis.

Honors & Awards


  • Postdoctoral Fellowship - Research Supplement to Promote Diversity in Science, American Heart Association (AHA) (2022-2024)
  • Vice-presidency of Education, Information, and Communication (VPEIC-FIOCRUZ), Oswaldo Cruz Foundation (FIOCRUZ) (2018-2021)
  • CAPES Fellowship, Higher Education Personnel Scholarship (CAPES) (2016-2018)

Boards, Advisory Committees, Professional Organizations


  • Member of the Council on Genomic and Precision Medicine (Early Career Committee), American Heart Association (AHA) (2022 - Present)
  • Member, American Heart Association (AHA) (2022 - Present)
  • Member, International Society of Amyloidosis (ISA) (2023 - Present)
  • Member, Sigma Xi (2024 - Present)

Stanford Advisors


All Publications


  • Unveiling Polysomal Long Non-Coding RNA Expression on the First Day of Adipogenesis and Osteogenesis in Human Adipose-Derived Stem Cells. International journal of molecular sciences Bonilauri, B., Ribeiro, A. L., Spangenberg, L., Dallagiovanna, B. 2024; 25 (4)

    Abstract

    Understanding the intricate molecular mechanisms governing the fate of human adipose-derived stem cells (hASCs) is essential for elucidating the delicate balance between adipogenic and osteogenic differentiation in both healthy and pathological conditions. Long non-coding RNAs (lncRNAs) have emerged as key regulators involved in lineage commitment and differentiation of stem cells, operating at various levels of gene regulation, including transcriptional, post-transcriptional, and post-translational processes. To gain deeper insights into the role of lncRNAs' in hASCs' differentiation, we conducted a comprehensive analysis of the lncRNA transcriptome (RNA-seq) and translatome (polysomal-RNA-seq) during a 24 h period of adipogenesis and osteogenesis. Our findings revealed distinct expression patterns between the transcriptome and translatome during both differentiation processes, highlighting 90 lncRNAs that are exclusively regulated in the polysomal fraction. These findings underscore the significance of investigating lncRNAs associated with ribosomes, considering their unique expression patterns and potential mechanisms of action, such as translational regulation and potential coding capacity for microproteins. Additionally, we identified specific lncRNA gene expression programs associated with adipogenesis and osteogenesis during the early stages of cell differentiation. By shedding light on the expression and potential functions of these polysome-associated lncRNAs, we aim to deepen our understanding of their involvement in the regulation of adipogenic and osteogenic differentiation, ultimately paving the way for novel therapeutic strategies and insights into regenerative medicine.

    View details for DOI 10.3390/ijms25042013

    View details for PubMedID 38396700

    View details for PubMedCentralID PMC10888724

  • Generation of two induced pluripotent stem cell lines from hereditary amyloidosis patients with polyneuropathy carrying heterozygous transthyretin (TTR) mutation. Stem cell research Melesio, J., Bonilauri, B., Li, A., Pang, P. D., Liao, R., Witteles, R. M., Wu, J. C., Sallam, K. 2023; 74: 103265

    Abstract

    Hereditary transthyretin amyloidosis with polyneuropathy (ATTR-PN) results from specific TTR gene mutations. In this study, we generated two induced pluripotent stem cell (iPSC) lines derived from ATTR-PN patients with heterozygous TTR gene mutations (Ala97Ser and Phe64Leu). These iPSC lines exhibited normal morphology, karyotype, high pluripotency marker expression, and differentiation into cells representing all germ layers. The generation of these iPSC lines serve as a valuable tool for investigating the mechanisms of ATTR-PN across various cell types and facilitating patient-specific in vitro amyloidosis modeling.

    View details for DOI 10.1016/j.scr.2023.103265

    View details for PubMedID 38100909

  • Generation of two induced pluripotent stem cell lines from patients with cardiac amyloidosis carrying heterozygous transthyretin (TTR) mutation. Stem cell research Bonilauri, B., Shin, H. S., Htet, M., Yan, C. D., Witteles, R. M., Sallam, K., Wu, J. C. 2023; 72: 103215

    Abstract

    Specific mutations in the TTR gene are responsible for the development of variant (hereditary) ATTR amyloidosis. Here, we generated two human induced pluripotent stem cell (iPSC) lines from patients diagnosed with Transthyretin Cardiac Amyloidosis (ATTR-CM) carrying heterozygous mutation in the TTR gene (i.e., p.Val30Met). The patient-derived iPSC lines showed expression of high levels of pluripotency markers, trilineage differentiation capacity, and normal karyotype. The generation of these iPSC lines represents a great tool for modeling patient-specific amyloidosis in vitro, allowing the investigation of the pathological mechanisms related to the disease in different cell types and tissues.

    View details for DOI 10.1016/j.scr.2023.103215

    View details for PubMedID 37788558

  • Microproteins in skeletal muscle: hidden keys in muscle physiology. Journal of cachexia, sarcopenia and muscle Bonilauri, B., Dallagiovanna, B. 2022; 13 (1): 100-113

    Abstract

    Recent advances in the transcriptomics, translatomics, and proteomics have led us to the exciting new world of functional endogenous microproteins. These microproteins have a small size and are derived from small open reading frames (smORFs) of RNAs previously annotated as non-coding (e.g. lncRNAs and circRNAs) as well as from untranslated regions and canonical mRNAs. The presence of these microproteins reveals a much larger translatable portion of the genome, shifting previously defined dogmas and paradigms. These findings affect our view of organisms as a whole, including skeletal muscle tissue. Emerging evidence demonstrates that several smORF-derived microproteins play crucial roles during muscle development (myogenesis), maintenance, and regeneration, as well as lipid and glucose metabolism and skeletal muscle bioenergetics. These microproteins are also involved in processes including physical activity capacity, cellular stress, and muscular-related diseases (i.e. myopathy, cachexia, atrophy, and muscle wasting). Given the role of these small proteins as important key regulators of several skeletal muscle processes, there are rich prospects for the discovery of new microproteins and possible therapies using synthetic microproteins.

    View details for DOI 10.1002/jcsm.12866

    View details for PubMedID 34850602

    View details for PubMedCentralID PMC8818594

  • Non-target molecular network and putative genes of flavonoid biosynthesis in Erythrina velutina Willd., a Brazilian semiarid native woody plant. Frontiers in plant science Chacon, D. S., Santos, M. D., Bonilauri, B., Vilasboa, J., da Costa, C. T., da Silva, I. B., Torres, T. d., de Araujo, T. F., Roque, A. d., Pilon, A. C., Selegatto, D. M., Freire, R. T., Reginaldo, F. P., Voigt, E. L., Zuanazzi, J. A., Scortecci, K. C., Cavalheiro, A. J., Lopes, N. P., Ferreira, L. D., Dos Santos, L. V., Fontes, W., de Sousa, M. V., Carvalho, P. C., Fett-Neto, A. G., Giordani, R. B. 2022; 13: 947558

    Abstract

    Erythrina velutina is a Brazilian native tree of the Caatinga (a unique semiarid biome). It is widely used in traditional medicine showing anti-inflammatory and central nervous system modulating activities. The species is a rich source of specialized metabolites, mostly alkaloids and flavonoids. To date, genomic information, biosynthesis, and regulation of flavonoids remain unknown in this woody plant. As part of a larger ongoing research goal to better understand specialized metabolism in plants inhabiting the harsh conditions of the Caatinga, the present study focused on this important class of bioactive phenolics. Leaves and seeds of plants growing in their natural habitat had their metabolic and proteomic profiles analyzed and integrated with transcriptome data. As a result, 96 metabolites (including 43 flavonoids) were annotated. Transcripts of the flavonoid pathway totaled 27, of which EvCHI, EvCHR, EvCHS, EvCYP75A and EvCYP75B1 were identified as putative main targets for modulating the accumulation of these metabolites. The highest correspondence of mRNA vs. protein was observed in the differentially expressed transcripts. In addition, 394 candidate transcripts encoding for transcription factors distributed among the bHLH, ERF, and MYB families were annotated. Based on interaction network analyses, several putative genes of the flavonoid pathway and transcription factors were related, particularly TFs of the MYB family. Expression patterns of transcripts involved in flavonoid biosynthesis and those involved in responses to biotic and abiotic stresses were discussed in detail. Overall, these findings provide a base for the understanding of molecular and metabolic responses in this medicinally important species. Moreover, the identification of key regulatory targets for future studies aiming at bioactive metabolite production will be facilitated.

    View details for DOI 10.3389/fpls.2022.947558

    View details for PubMedID 36161018

  • Proteogenomic Analysis Reveals Proteins Involved in the First Step of Adipogenesis in Human Adipose-Derived Stem Cells. Stem cells international Bonilauri, B., Camillo-Andrade, A. C., Santos, M. D., Fischer, J. d., Carvalho, P. C., Dallagiovanna, B. 2021; 2021: 3168428

    Abstract

    Obesity is characterized as a disease that directly affects the whole-body metabolism and is associated with excess fat mass and several related comorbidities. Dynamics of adipocyte hypertrophy and hyperplasia play an important role in health and disease, especially in obesity. Human adipose-derived stem cells (hASC) represent an important source for understanding the entire adipogenic differentiation process. However, little is known about the triggering step of adipogenesis in hASC. Here, we performed a proteogenomic approach for understanding the protein abundance alterations during the initiation of the adipogenic differentiation process.hASC were isolated from adipose tissue of three donors and were then characterized and expanded. Cells were cultured for 24 hours in adipogenic differentiation medium followed by protein extraction. We used shotgun proteomics to compare the proteomic profile of 24 h-adipogenic, differentiated, and undifferentiated hASC. We also used our previous next-generation sequencing data (RNA-seq) of the total and polysomal mRNA fractions of hASC to study posttranscriptional regulation during the initial steps of adipogenesis.We identified 3420 proteins out of 48,336 peptides, of which 92 proteins were exclusively identified in undifferentiated hASC and 53 proteins were exclusively found in 24 h-differentiated cells. Using a stringent criterion, we identified 33 differentially abundant proteins when comparing 24 h-differentiated and undifferentiated hASC (14 upregulated and 19 downregulated, respectively). Among the upregulated proteins, we shortlisted several adipogenesis-related proteins. A combined analysis of the proteome and the transcriptome allowed the identification of positive correlation coefficients between proteins and mRNAs.These results demonstrate a specific proteome profile related to adipogenesis at the beginning (24 hours) of the differentiation process in hASC, which advances the understanding of human adipogenesis and obesity. Adipogenic differentiation is finely regulated at the transcriptional, posttranscriptional, and posttranslational levels.

    View details for DOI 10.1155/2021/3168428

    View details for PubMedID 34956370

    View details for PubMedCentralID PMC8702357

  • Long Non-Coding RNAs Associated with Ribosomes in Human Adipose-Derived Stem Cells: From RNAs to Microproteins. Biomolecules Bonilauri, B., Holetz, F. B., Dallagiovanna, B. 2021; 11 (11)

    Abstract

    Ribosome profiling reveals the translational dynamics of mRNAs by capturing a ribosomal footprint snapshot. Growing evidence shows that several long non-coding RNAs (lncRNAs) contain small open reading frames (smORFs) that are translated into functional peptides. The difficulty in identifying bona-fide translated smORFs is a constant challenge in experimental and bioinformatics fields due to their unconventional characteristics. This motivated us to isolate human adipose-derived stem cells (hASC) from adipose tissue and perform a ribosome profiling followed by bioinformatics analysis of transcriptome, translatome, and ribosome-protected fragments of lncRNAs. Here, we demonstrated that 222 lncRNAs were associated with the translational machinery in hASC, including the already demonstrated lncRNAs coding microproteins. The ribosomal occupancy of some transcripts was consistent with the translation of smORFs. In conclusion, we were able to identify a subset of 15 lncRNAs containing 35 smORFs that likely encode functional microproteins, including four previously demonstrated smORF-derived microproteins, suggesting a possible dual role of these lncRNAs in hASC self-renewal.

    View details for DOI 10.3390/biom11111673

    View details for PubMedID 34827671

    View details for PubMedCentralID PMC8615451

  • Linking long noncoding RNAs (lncRNAs) and doping detection. Drug testing and analysis Bonilauri, B., Dallagiovanna, B. 2021; 13 (5): 1068-1071

    Abstract

    In the fight against doping, efficient methods for detecting substances or biomarkers are still being improved. Indirect methods are an interesting alternative for the detection of substances misuse longitudinally. Here we shed lights the long non-coding RNAs (lncRNAs) as a possible biomarkers due to their characteristics such as tissue-specific expression and strict regulation.

    View details for DOI 10.1002/dta.2952

    View details for PubMedID 33119947

  • The impact of blood-processing time on the proteome of human peripheral blood mononuclear cells. Biochimica et biophysica acta. Proteins and proteomics Bonilauri, B., Santos, M. D., Camillo-Andrade, A. C., Bispo, S., Nogueira, F. C., Carvalho, P. C., Zanchin, N. I., Fischer, J. d. 2021; 1869 (3): 140581

    Abstract

    Human peripheral blood mononuclear cells (PBMC) are key to several diagnostics assays and basic science research. Blood pre-analytical variations that occur before obtaining the PBMC fraction can significantly impact the assays results, including viability, composition, integrity, and gene expression changes of immune cells. With this as motivation, we performed a quantitative shotgun proteomics analysis using Isobaric Tag for Relative and Absolute Quantitation (iTRAQ 8plex) labeling to compare PBMC obtained from 24 h-stored blood at room temperature versus freshly isolated. We identified a total of 3195 proteins, of which 245 were differentially abundant (101 upregulated and 144 downregulated). Our results revealed enriched pathways of downregulated proteins related to exocytosis, localization, vesicle-mediated transport, cell activation, and secretion. In contrast, pathways related to exocytosis, neutrophil degranulation and activation, granulocyte activation, leukocyte degranulation, and myeloid leukocyte activation involved in immune response were enriched in upregulated proteins, which may indicate probable granulocyte contamination and activation due to blood storage time and temperature. Examples of upregulated proteins in the 24 h-PBMC samples are CAMP, S100A8, LTA4H, RASAL3, and S100A6, which are involved in an adaptive immune system and antimicrobial activity, proinflammatory mediation, aminopeptidase activities, and naïve T cells survival. Moreover, examples of downregulated proteins are NDUFA5, TAGLN2, H3C1, TUBA8, and CCT2 that are related to the cytoskeleton, cell junction, mitochondrial respiratory chain. In conclusion, the delay in blood-processing time directly impacts the proteomic profile of human PBMC, possibly through granulocyte contamination and activation.

    View details for DOI 10.1016/j.bbapap.2020.140581

    View details for PubMedID 33301959

  • Long Non-coding RNAs Are Differentially Expressed After Different Exercise Training Programs. Frontiers in physiology Bonilauri, B., Dallagiovanna, B. 2020; 11: 567614

    Abstract

    Molecular regulation related to the health benefits of different exercise modes remains unclear. Long non-coding RNAs (lncRNAs) have emerged as an RNA class with regulatory functions in health and diseases. Here, we analyzed the expression of lncRNAs after different exercise training programs and their possible modes of action related to physical exercise adaptations.Public high-throughput RNA-seq data (skeletal muscle biopsies) were downloaded, and bioinformatics analysis was performed. We primarily analyzed data reports of 12 weeks of resistance training (RT), high-intensity interval training (HIIT), and combined (CT) exercise training. In addition, we analyzed data from 8 weeks of endurance training (ET). Differential expression analysis of lncRNAs was performed, and an adjusted P-value < 0.1 and log2 (fold change) ≥0.5 or ≤-0.5 were set as the cutoff values to identify differentially expressed lncRNAs (DELs).We identified 204 DELs after 12 weeks of HIIT, 43 DELs after RT, and 15 DELs after CT. Moreover, 52 lncRNAs were differentially expressed after 8 weeks of ET. The lncRNA expression pattern after physical exercise was very specific, with distinct expression profiles for the different training programs, where few lncRNAs were common among the exercise types. LncRNAs may regulate molecular responses to exercise, such as collagen fibril organization, extracellular matrix organization, myoblast and plasma membrane fusion, skeletal muscle contraction, synaptic transmission, PI3K and TORC regulation, autophagy, and angiogenesis.For the first time, we show that lncRNAs are differentially expressed in skeletal muscle after different physical exercise programs, and these lncRNAs may act in various biological processes related to physical activity adaptations.

    View details for DOI 10.3389/fphys.2020.567614

    View details for PubMedID 33071823

    View details for PubMedCentralID PMC7533564

  • Data describing the experimental design and quality control of RNA-Seq of human adipose-derived stem cells undergoing early adipogenesis and osteogenesis. Data in brief Marcon, B. H., Spangenberg, L., Bonilauri, B., Robert, A. W., Angulski, A. B., Cabo, G. C., Cofré, A. R., Bettes, P. S., Dallagiovanna, B., Shigunov, P. 2020; 28: 105053

    Abstract

    An important tool to study the regulation of gene expression is the sequencing and the analysis of different RNA fractions: total, ribosome-free, monosomal and polysomal. By comparing these different populations, it is possible to identity which genes are differentially expressed and to get information on how transcriptional and translational regulation modulates cellular function. Therefore, we used this strategy to analyze the regulation of gene expression of human adipose-derived stem cells during the triggering of the adipogenic and osteogenic differentiation. Here, we have focused on analyzing the differential expression of mRNAs during early adipogenic and osteogenic differentiation, and presented the detailed data concerning the experimental design, the RNA-Seq quality data, the raw data obtained and the RT-qPCR validation data. This information is important to confirm the accuracy of the data considering a future reuse of the data provided. Moreover, this study may be used as groundwork for future characterization of the transcriptome and the translatome regulation of different cell types.

    View details for DOI 10.1016/j.dib.2019.105053

    View details for PubMedID 31989002

    View details for PubMedCentralID PMC6970145