Institute of molecular biology and pathology (IBPM)

Expertise

Collectively, the skills and expertise available at IBPM cover a broad range of disciplines:

Bioinformatics and computational biology
o Protein structure modeling.
o Design of proteins and bioactive peptides.
o Computational analysis of protein and nucleic acid sequences and their 3D structures.
o Genomes and big data analysis.
o IBPM is part of Elixir, the European network for the development of bioinformatics in Life Sciences (https://www.elixir-europe.org/)

Chemistry and molecular biology
o Synthesis of chiral catalysts and their immobilization on nanoparticles. Synthesis and purification of bioactive compounds, and their chemical/physical characterization by: Gas Chromatography coupled Mass Spectrometry (GC/MS), High Performance Liquid Chromatography (HPLC), Liquid Chromatography coupled to Mass Spectrometry (LC-MS), Nuclear Magnetic Resonance (NMR), Infrared Spectroscopy (IR), polarimetry.
o Engineering and purification of peptides, proteins and nucleic acids.
o Analysis of protein dynamics, enzymatic catalysis and reaction mechanisms by both static and time-resolved UV-visible absorption spectroscopy, fluorescence and circular dichroism (stopped-flow, temperature jump, continuous flow). NO amperometry.
o Protein structure analysis by X-ray crystallography (BioCrystal Facility, see 'Services').
o Through the BioCrystal Facility, IBPM has access to synchrotron facilities in Grenoble (France), Berlin (Germany) and Trieste (Italy).

Advanced Molecular Techniques
o Analysis of the interaction between DNA, RNA, proteins and small molecules, by analytic ultracentrifugation, Surface Plasmon Resonance (SPR), Differential Scanning Fluorimetry (DSF), Isothermal Titration Calorimetry (ITC), RNA ImmunoPrecipitation (RIP), Cross Linking ImmunoPrecipitation (CLIP), RNA pull-down, Two-hybrid system, Phage display.
o 'Omics' techniques applied to the study of proteins, DNA, RNA and their interactions (microarrays, Next Generation Sequencing, ChIP, standard and redox proteomics)
o Genome editing and gene expression reprogramming: CRISPR-Cas9 technology, adeno-associated viral (AAV) vectors and regulatory genes creation.
o Synthesis and characterization of protein nanovectors for drug delivery applications.

Cellular biology, cytofluorimetry and imaging.
o Animal and plant cell cultures, cancer and mesenchymal stem cells, cellular models.
o Isolation of organelles and cell structures, and their functional characterization.
o Transient and stable transfection. Transformation of protoplasts from plant cells. Viral infection and microinjection.
o Multidimensional cytofluorimetry for the analysis of membrane and intracellular molecules, and for the study of cell proliferation and survival.
o In situ hybridization techniques for the analysis of gene expression in both animal and plant tissues. Immunohistochemical and immunocytochemical techniques. In situ Proximity Ligation Assay (PLA) for the visualization of specific protein-protein interactions inside the cell.
o Cytological analysis of chromosome squashes from meiotic and mitotic cells and fluorescent in situ hybridization (FISH) for the study of genome instability.
o Molecular microscopy (TEM/AFM) of nucleic acids and chromatin.
o Profiling of cell energy metabolism by high-resolution respirometry and ATP synthesis assays.
o In vitro and in vivo assays for the evaluation of the angiogenic properties of synthetic compounds or proteins of interest.
o Wide field and confocal microscopy, both in bright field and fluorescent. Methods of video recording of living cells (time lapse) for the analysis of dynamic cellular processes. Image analysis; development of software tools for both qualitative and quantitative analysis (Microscopy Platform, see 'Services').

Model organisms and systems
o Model organisms include bacteria (Escherichia coli, Shigella flexneri), Saccharomyces cerevisiae, Giardia intestinalis, Drosophila melanogaster, Arabidopsis thaliana, Pteris vittata. In addition, immune response, neuropathologies, neuromuscular pathologies and oncological pathologies are studied through the use of genetically modified mouse lines.
o Genetic manipulation of model organisms. Gene expression reprogramming in preclinical mouse models (AAV and artificial regulatory genes). Transgenic plants production (Arabidopsis thaliana).
o Cellular models include: cancer and mesenchymal stem cell from patients, myoblasts derived from patients affected by muscular pathologies, neural human progenitors from pluripotent cells, mouse embryonic stem cells, muscular and neuronal cell differentiation models, rodent primary neuronal cultures and human and mouse leukocyte lines (natural killer lymphocytes, antigen-specific T lymphocytes, dendritic cells).

Genetic analyses and techniques
o Formal genetic analysis in Drosophila melanogaster, Saccharomyces cerevisiae e Arabidopsis thaliana.
o Collections of genetic control mutants.
o Analysis of human genetic variability: analysis of genetic polymorphisms (SNPs) aimed at the disease prediction and to study the human evolution.