Fabrizia Bonacina (Italy)

University of Milan Department of Pharmacological and Biomolecular science
Dr. Bonacina obtained the degree in Pharmacy at the University of Milan in 2012 under the supervision of Prof. Alberico Luigi Catapano and Giuseppe Danilo Norata and from 2012 to 2016 she attended the Doctoral School in Pharmacological Sciences of the University of Milan, discussing a thesis entitled: "Role of the long pentraxin 3 (PTX3) in cardiometabolic disease", shifting her scientific interest towards the mechanisms responsible for the alteration of immune-inflammatory response in the context of cardio-metabolic diseases. Thanks to a research experience of about a year in the group of Prof. Federica Marelli-Berg, at the William Harvey Research Institute of Queen Mary University in London, and collaborations in the field of immunology, Dr. Bonacina deepened her expertise in immunological research. From 2016, Dr. Bonacina is a Post-doc Research Fellow in the lab of Lipoprotein, Immunity and Atherosclerosis, under the supervision of Prof. Alberico Luigi Catapano and Giuseppe Danilo Norata. Main current research interests of Dr. Bonacina are related to the study of the connection between functional and metabolic adapatations of immune cells in the context of cardiometabolic diseases.

Author Of 2 Presentations

O004 - Role of the Interleukin-1 Receptor/Toll Like Receptor TIR8/SIGIRR in experimental atherosclerosis (ID 1235)

Session Type
Late Breaking Sessions
Session Time
11:00 - 12:30
Date
Mon, 31.05.2021
Room
Hall A (Live Q&A)
Lecture Time
11:21 - 11:28

Abstract

Background and Aims

TIR8/SIGIRR dampens the excessive activation mediated by ILRs and TLRs agonism and thus is a key regulator of inflammation. Aim of this study was to investigate the role of TIR8 in atherosclerosis.

Methods

8 weeks old-LDLR KO and TIR8/LDLR double KO (DKO) male mice were fed with standard diet (STD) or cholesterol-enriched diet (WTD) for 12 weeks. Plasma lipid profiling, extensive immunophenotyping and histological analysis of the atherosclerotic plaques were then performed.

Results

TIR8 deletion in STD-fed LDLR KO mice impacts circulating immune cell profile: decreased percentage of T lymphocytes (-29%, p<0.001) and increased percentage of B cells (+14%, p<0.05) were observed compared to LDLR KO mice, as well as increased mature Natural Killer cells (+13%, p<0.0001), as already described in the TIR8 KO mouse model. When fed a cholesterol rich diet for 12 weeks to induce atherosclerosis, in addition to changes observed on STD, also circulating levels of monocytes increased in DKO mice compared to LDL-R KO mice (mean 1464 vs 910 cells/ul, p<0.05). These changes in immune profile, however, did not affect atherosclerotic plaque area or stability. Similarly, no differences in plasma lipid profile were observed.

Conclusions

TIR8 deficiency in LDLR KO mice increases NKs and monocytes blood levels compared to LDL-R KO mice. Changes in these immune subsets, however, do not impact the development of atherosclerosis.

Hide

O023 - The low-density lipoprotein receptor (LDL-R) is an immune-metabolic checkpoint during CD8 T lymphocytes activation (ID 1251)

Session Type
Vascular Biology
Session Time
16:00 - 17:30
Date
Mon, 31.05.2021
Room
Hall E
Lecture Time
16:30 - 16:38

Abstract

Background and Aims

Activation of T lymphocytes combines functional to metabolic rewiring of cell machinery, including cholesterol homeostasis. Here we evaluated the role of LDLR on T cell biology

Methods

Immunophenotypic characterization of CD8T cells from WT and LDLR KO mice was performed in vitro (anti-CD3/CD28) and in vivo (vaccination, homeostatic proliferation) coupled to proteomic and confocal analysis on isolated CD8T cells. In parallel, T cells from FH were tested.

Results

LDLR deficiency dampened CD8+ proliferation (-35%, p<0.01) paralleled by a reduction in INFγ production (-39.6%, p<0.01). In vivo antigen-specific activation by ovalbumin vaccination, but not homeostatic proliferation, resulted in a decreased proliferation and cytokines production (↓IFNγ p<0.001,↓IL13 p<0.01,↓perforin p<0.05) in CD8+ of KO mice. Incubation with LDL significantly increased the proliferation in WT but not KO CD8T cells (+11%,p<0.01), a phenotype that was compensated by a reprogramming of de-novo synthesis in KO CD8T cells. Proteomic analysis revealed an impairment in glycolysis and OXPHOS associated to downregulation of pathways downstream to mTORC1 activation, possibly link to its reduced lysosomal localization observed in KO CD8T cells.

CD8+ T cells from FH subjects proliferated less (-36%, p>0.05) compared to sex- and age-matched controls and presented a decreased granzyme production (-60.3%, p<0.01) when CD8 memory response was tested in vitro with virus-derived peptides in seasonal influenza vaccinated matched controls and FH subjects.

Conclusions

LDLR plays a critical role in regulating the immunometabolic responses in CD8+ Tcells, and thus might represent a checkpoint linking cellular cholesterol metabolism to adaptive immune response.

Hide

Presenter of 1 Presentation

O023 - The low-density lipoprotein receptor (LDL-R) is an immune-metabolic checkpoint during CD8 T lymphocytes activation (ID 1251)

Session Type
Vascular Biology
Session Time
16:00 - 17:30
Date
Mon, 31.05.2021
Room
Hall E
Lecture Time
16:30 - 16:38

Abstract

Background and Aims

Activation of T lymphocytes combines functional to metabolic rewiring of cell machinery, including cholesterol homeostasis. Here we evaluated the role of LDLR on T cell biology

Methods

Immunophenotypic characterization of CD8T cells from WT and LDLR KO mice was performed in vitro (anti-CD3/CD28) and in vivo (vaccination, homeostatic proliferation) coupled to proteomic and confocal analysis on isolated CD8T cells. In parallel, T cells from FH were tested.

Results

LDLR deficiency dampened CD8+ proliferation (-35%, p<0.01) paralleled by a reduction in INFγ production (-39.6%, p<0.01). In vivo antigen-specific activation by ovalbumin vaccination, but not homeostatic proliferation, resulted in a decreased proliferation and cytokines production (↓IFNγ p<0.001,↓IL13 p<0.01,↓perforin p<0.05) in CD8+ of KO mice. Incubation with LDL significantly increased the proliferation in WT but not KO CD8T cells (+11%,p<0.01), a phenotype that was compensated by a reprogramming of de-novo synthesis in KO CD8T cells. Proteomic analysis revealed an impairment in glycolysis and OXPHOS associated to downregulation of pathways downstream to mTORC1 activation, possibly link to its reduced lysosomal localization observed in KO CD8T cells.

CD8+ T cells from FH subjects proliferated less (-36%, p>0.05) compared to sex- and age-matched controls and presented a decreased granzyme production (-60.3%, p<0.01) when CD8 memory response was tested in vitro with virus-derived peptides in seasonal influenza vaccinated matched controls and FH subjects.

Conclusions

LDLR plays a critical role in regulating the immunometabolic responses in CD8+ Tcells, and thus might represent a checkpoint linking cellular cholesterol metabolism to adaptive immune response.

Hide