Projects:

TitleManagerType
Aging-associated, specific pathomechanisms of endothelial dysfunction in sepsis: pathway analysis and experimental therapy [more]S. ChłopickiNCN, Weave-UNISONO
Metabolic reprogramming in age-dependent endothelial dysfunction and vascular stiffness; novel mechanism of “inflamm-ageing” [more]S. ChłopickiNCN, MAESTRO
LDL-mimetic liposomal contrast agent for MRI-based detection of increased endothelial permeability in the early phase of endothelial dysfunction in vivo [more]A. BarNCN, OPUS 20
In search of the unexplored aspects of ferric heme protein-dependent regulation of nitric oxide signaling in the erythrocyte and vessel wall [more]J. DybaśNCN, OPUS 21
Normalization of energy metabolism as a way to develop innovative antithrombotic therapies [more]P. KaczaraNCN, OPUS 21
The role of MCPIP1 RNase in disrupted endothelial homeostasis accompanying non-alcoholic fatty liver disease [more] J. KotlinowskiNCN, OPUS, 205024
Breast cancer metastasis-induced endothelial-mesenchymal t ransition alongside ageing; implications for therapy [more]M. SmędaNCN, OPUS 21
CPL and RROA probes in the study of protein structure – induction of chirality and new methods of amplifying the chiroptical signal [more]G. ZającNCN,OPUS 18
Ecto-enzymes in interactions of vascular endothelium with blood circulating cells in physiology, pathology and therapy; could cells exchange their ecto-enzymes? [more]B. Kutryb-ZającNCN, SONATA 15
Impairment of coronary microcirculation in a mouse model of a heart failure [more]G. KwiatkowskiNCN, SONATA 16
From the structure of fenestrations in live Liver Sinusoidal Endothelial Cells to real-time in vitro pharmacology [more]B. ZapotocznyNCN, SONATA 15
Endothelial inflammation studied in situ within functional blood vessels using Raman imaging, atomic force microscopy and imaged cytometry [more] M. PaciaNCN, SONATINA 1
The role of lactate dehydrogenase A in development of endothelial dysfunction in murine model of heart failure [more]A. KaraśNCN, PRELUDIUM 20
S. MosiołekNCN, PRELUDIUM 22
Novel strategies for pharmacological modulation of platelet-derived microparticles – effects on endothelium [more] A. PełeszNCN, PRELUDIUM19
PDMS-based specially designed microfluidic channel lined with primary endothelial cells: a novel tool for experimental pharmacology of endothelium.S. RocchettiNCN, PRELUDIUM
Cross-talk between endothelium and perivascular adipose tissue upon inflammation: high-resolution imaging of adipocytes in co-culture with endothelial cells [more]E. StanekNCN, PRELUDIUM 22
Function and phenotype of liver sinusoidal endothelial cells (LSECs) and their role in liver dysfunction development in murine model of heart failure [more]K. Wojnar-LasońNCN, PRELUDIUM 18
Circularly polarized luminescence and other chiroptical methods towards detection of protein alterationsG. ZającNAWA

Finished projects:

TitleManagerType
Prostacyclin, nitric oxide and carbon monoxide-based pharmacotherapy of endothelial dysfunction and platelet activation-a novel strategy to inhibit cancer metastasis [more] S. Chłopicki NCBiR, STRATEGMED, 233226
Jagiellonian Centre for Experimental Therapeutics – JCET [more] S. ChłopickiNCBiR, POIG, DZIAŁANIE 2.2
Vascular endothelium in life-style diseases: from basic research to the offer of an innovative endothelium-targeted therapeutic [more] S. ChłopickiNCBiR, POIG, DZIAŁANIE 1.1.2
Endothelial profiling to predict therapeutic activity or vascular toxicity of compounds in early preclinical research [more]S. ChłopickiTEAM TECH
DoReMi – Low Dose Research towards Multidisciplinary Integration [more] S. ChłopickiFP7
Molecular logic lab-on-a-vesicle for intracellular diagnostics [more]M. BarańskaH2020-MSCA-ITN-2017
Development of a combination of natural compounds comprehensively improving endothelial function [more] S. Chłopicki (One part)NCBiR, PBS
Pharmacotherapeutic mechanisms, spectroscopic signatures, and nanomechanics of dysfunctional endothelium in liver steatosis and heart failure; towards organ-specyfic mechanisms [more] S. Chłopicki NCN, SYMFONIA3, 282784
Towards novel mechanisms of endothelial dysfunction in E3L.CETP mice displaying hyperlipidaemia with human-like lipoprotein profile [more]S. ChłopickiNCN, OPUS
The role of nicotinamide N-methyltransferase (NNMT) and mitochondrial mechanisms in endothelial dysfunction [lider IBD PAN] [more] S. Chłopicki NCN,OPUS 10
RS-SRS-MED: Spontaneous and stimulated Raman scattering microscopy for sensitive and ultrafast imaging of cells upon development of life style diseases [more]M. Barańska oraz S. ChłopickiNCN, OPUS
The role of nicotinamide N-methyltransferase and mitochondrial activity during endothelial dysfunction [more] K. Zabłocki (IBD PAN)NCN, OPUS
Studies on antiplatelet and antithrombotic activity of CO-relasing molecules (CO-RM) [more] S. ChłopickiNCN, HARMONIA4, 212354
The development of spectroscopic analysis of lipid droplets in vitro: biochemistry, and their localisation in relation to the biologic [more]M. BarańskaNCN, HARMONIA
Diagnostics of the biochemical alterations in packed human red blood cells (PRBCs) with the use of vibrational spectroscopy [more] K. MarzecNCBiR, LIDER
Lipid unsaturation degree in perivascular adipose tissue – a new biomarker of vascular wall inflammation? Raman studies of murine caridovascular models and primary isolated adipocytes [more]A. Kaczor (Wydział Chemii UJ/JCET)NCN, OPUS
Induced chirality in “guest-host” supramolecular assemblies – developement of new methods of resonance Raman ptical Activity [more] A. KaczorNCN, OPUS
To catch lifestyle diseases red-handed – in vivo Raman spectroscopy [more] A. KaczorNCN, OPUS 13
Development of EPR oximetry methodology for evaluation of endothelial action of drugs [more] P. KaczaraNCN, SONATA3, 184181
In search of biochemical, mechanical and functional fingerprints of oxidative stress in RBCs [more] K. Marzec NCN, OPUS
Structural studies and chemical imaging of vitamins A, E and their metabolites in healthy and pathologically altered animal tissue [more] K. MarzecNCN, SONATA4, 205024
Spectral histopathology, FTIR and immunoSERS, for the recognition of biochemical status of pre-metastatic niche and micrometastasis in murine model of breast cancer [more]K. Małek (Wydział Chemii UJ)NCN, OPUS12
Design of SERS nanosensors for ex vivo detection of inflammatory state in blood vessels [more] K. MałekNCN, OPUS
Raman spectroscopy in vitro studies of chemotherapeutic impact on endothelial cells [more]K. Majzner (Wydział Chemii UJ)NCN, SONATA11
In search for endothelial mechanisms of TPO-induced neuroprotection. Marie Skłodowska-Curie Individual Fellowships (IF), Reintegration Fellowship (H2020-MSCA-IF-2014)  [more] M. AdamskiHORYZONT 2020
Towards experimental pharmacology of endothelium in vivo based on multiparameter method of assessment of the vascular endothelial function in mice with the use of MR imaging techniques [more] A. BarNCN, PRELUDIUM12
In search of spectroscopic markers of autophagy based on pharmacologically induced phospholipidosis in endothelial cells [more]E. BikNCN, PRELUDIUM 18
Description of platelets role in metastasis and evaluation of anti-platelet treatment efficacy in mice model of metastatic breast cancer [more] E. BuczekNCN, PRELUDIUM 8
Aptamer-based biosensor for simultaneous detection of thrombin and von Willebrand factor in plasma [more] K. DerszniakNCN, PRELUDIUM11
Aptamers in the study of thrombotic mechanisms dependent on platelets and erythrocytes functions by microfluidic devices [more]K. Derszniak NCN, ETIUDA 8
Resonance Raman spectroscopy studies of the in vitro and ex vivo haemoglobin adducts with gasotransmitters in biological systems [more] J. DybaśNCN, PRELUDIUM12
Molecular spectroscopy studies on chosen hemoprotein adducts and their changes in biological systems [more] K. DybaśNCN, ETIUDA
Effects of physicochemical properties of drugs on their distribution in endotehlial cell compartment and their interactions with plasma proteins [more] A. A. Gonciarz-DytmanNCN, PRELUDIUM7, 258846
Physicochemical tests of cell membrane and intracellular compartment of endothelial cells [more] A. JaworskaNCN, PRELUDIUM3, 185400
Bioanalitycal and pharmacological studies on the compensatory mechanism dependent on MNA/COX-2/PGI2 pathway in NO-deficient mice [more] A. KijNCN, PRELUDIUM
Pharmacokinetic profiling to understand differences in biological activity of two novel liver-selective NO-releasing compounds V-PYRRO/NO and V-PROLI/NO in the treatment of experimental model of NAFLD in mice [more] K. KuśNCN, PRELUDIUM6, 241590
From sympthoms to treatment: comprehensive characteristics of pathological changes in the experimental model of fatty liver disease by oscillatory spectroscopy [more] K. N. KochanNCN, PRELUDIUM5, 220775
The anti-inflammatory effect of vitamin K – Mechanism of action [more]A. Kierońska-RudekNCN, PRELUDIUM
Studies on biologically relevant systems in conditions of resonance Raman optical activity [more]E. MachalskaNCN, PRELUDIUM
Perivascular adipose tissue versus gut microbiota: Raman imaging study [more]Z. Majka
NCN, PRELUDIUM 20
Comparative evaluation of the effects of tyrosine kinase inhibitors (TKIs) on endothelial function in vivo in mice [more]B. MarczykNCN, PRELUDIUM
Role of angiotensin 1-12 and ACE-independent generation of Angiotensin II in peripheral endothelial dysfunction in heart failure [more]T. MohaissenNCN, PRELUDIUM
Raman spectroscopy in studies on cellular uptake of sinusoidal endothelial cells in vitros [more] E. Matuszyk (Szafraniec) NCN, PRELUDIUM
Raman spectroscopy in the study on non-alcoholic fatty liver disease using isolated liver cells and in vitro cell models [more] E. Matuszyk (Szafraniec) NCN, ETIUDA
Glycocalyx damage – the first stage of pathomechanism of endothelial dysfunction; measurements using capillary electrophoresis [more] K. Matyjaszczyk-Gwarda NCN, PRELUDIUM
Label-free imaging of subcellular structures of endothelium using Confocal Raman Spectroscopy supported by FT-IR Spectroscopy, AFM and Fluorescence Microscopy [more] K. B. MajznerNCN, PRELUDIUM5, 216588
The role of reactive oxygen species (ROS) generated during a maximal exercise by endothelial NOX2 i NOX4 in the regulation of post-exercise oxidant stress, postexercise haemostasis and exercise capacity [more] K. PrzyborowskiNCN, PRELUDIUM 13
Small scale, great significance: Raman spectroscopy, 3D imaging, atomic force microscopy and immunohistochemical staining in study of diabetes [more] M.Pacia (Pilarczyk)NCN, PRELUDIUM4, 203097
Towards mechanisms underlying antiplatelet effects of CO-releasing compounds: the role of CO kinetics release, antiadhesive function and effects on metabolism [more] B. SitekNCN, PRELUDIUM11
Vibrational spectroscopy in analysis of biochemical status of blood that mirrors vascular wall pathology in animals with systemic and pulmonary hypertension [more] E. A. StaniszewskaNCN, PRELUDIUM5, 217395
Spectroscopic characteristics of perivascular adipose tissue in the progression of atherosclerosis [more] K. CzamaraNCN, MINIATURA 2
The role of PGI2 and NO, produced by liver sinusoidal endothelial cells (LSEC), in the regulation of hapatic glycogenolysis and gluconeogenesis at an early stage of NAFLD development – studies using a unique isolated perfused liver system [more] I. Czyżyńska-CichońNCN, MINIATURA
Analysis of the molecular mechanisms of metastasis in murine model of breast cancer via the comprehensive study on protein expression changes in lungs [more] A. KurpińskaNCN, MINIATURA
Changes in paracrine signaling of liver sinusoidal endothelial cells stimulated with fatty acids and lipid droplets formation in hepatocytes – studies with use of a platform for three-dimensional co-culture under micro-flow condition, OrganoPlate [more] E. Kuś (Maślak)NCN, MINIATURA
Non-invasive MRI assessment of microvascular nitric oxide synthase dysfunctionand permeability in the lung endothelium in a mouse model of a chronic heartfailure [more] G. KwiatkowskiNCN, MINIATURA
Effect of visfatin on endothelial bioenergetics and phenotype [more] L. MateuszukNCN, MINIATURA
Application of a new immunohistochemical technique for imaging of DMPO spin trap to image oxidative stress in cardiac muscle in mice [more] B. ProniewskiNCN, MINIATURA
Multiparametric analysis of pulmonary endothelial dysfunction in the murine model along the progression of breast cancer [more] M. SmędaNCN, MINIATURA
The PGI2 / SIRT1 pathway is a chance to revolutionize pharmacotherapy of cardiovascular disease [more]M. SternakNCN, MINIATURA
Toward pharmacology of cancer cells adhesion and transendothelial migration using unique 3D microfluidic platforms [more] M. StojakNCN, MINIATURA
Proteomic diagnostics of endothelial-dependent mechanisms of endotoxemia development in young and aging mice.J.Suraj-PrażmowskaNCN, MINIATURA
The effect of the activators of Nrf2 transcription factor on endothelial permeability- ex vivo studies on B6.129X1-Nfe2l2tm1Ywk / J knockout mice [more] E. Szcęsny-MałysiakNCN, MINIATURA
Scholarship of the Minister of Science and Higher Education for outstanding young scientists [more]M. PaciaMNiSW
The role of endothelial Akt1 and Akt2 in the modulation of angiotensin II-induced endothelial dysfunction; a possible upstream mechanism of non-classical angiotensin pathway [more]M. SternakNAWA

Aging-associated, specific pathomechanisms of endothelial dysfunction in sepsis: pathway analysis and experimental therapy 

Funding Agency: National Science Centre, Poland, Program: Weave-UNISONO
Project leader: Prof. Stefan Chłopicki (JCET, UJ)

This project should shed light on the pharmacotherapeutic mechansisms of age-associated worsening of endothelial dysfunction in sepsis. This topic represent an important challenge in medicine given the fact that as yet there are no effective pharmacological approaches for the therapy of circulatory shock and multiorgan failure of sepsis.

The central hypotheses of the present application are that (1) accelerated endothelial dysfunction is a key contributor to multiple organ failure and mortality in aging animals during sepsis and (2) in aging blood vessels, unique cellular and molecular mechanisms operate that render them extremely vulnerable to the oxidative stress-associated endothelial dysfunction during sepsis. 


Towards novel mechanisms of endothelial dysfunction in E3L.CETP mice displaying hyperlipidaemia with human-like lipoprotein profile

Funding Agency: National Science Centre, Poland, Program: OPUS
Project leader: Prof. Stefan Chłopicki (JCET, UJ)

Vitamin K is a group of fat-soluble vitamin involved in the regulation of coagulation and bone and calcium homeostasis via vitamin K-dependent (VKD) proteins. Recent epidemiological evidence suggest that intake of vitamin K2 (exerting mainly extrahepatic activity), but not vitamin K1 (exerting mainly hepatic activity) reduce cardiovascular and total mortality. This effect cannot be explained by profile of of action of vitamin K2 described so far. Based on these unpublished results we claim that impairment of endogenous synthesis of vitamin K2 in endothelium contribute to the development of endothelial dysfunction in atherosclerosis and supplementation with vitamin K2 reverse the endogenous deficit of vitamin K2 in the endothelium with subsequent improvement of carboxylation status in endothelium and endothelial function. Altogether, our preliminary results suggest that vitamin K2 plays an important role in the regulation of endothelial function that has been not revealed so far. Surprisingly, despite long-term history of vitamin K research, knowledge on the role of vitamin K in the regulation of endothelium in the context of physiology, biochemistry, pathophysiology and pharmacology of endothelium and various diseases related to endothelial dysfunction is virtually absent. To complement this knowledge within the project, it may bring not only a new understanding of the mechanisms of exogenous vitamin K2, and regulation of endothelium by this vitamin, but also possibly open new therapeutic avenues for many diseases associated with vascular endothelial dysfunction. In this project we will use E3L.CETP mice representing a clinically-relevant and unique model of slowly progressing endothelial dysfunction induced by mild hyperlipoproteinaemia proceeding atherosclerosis development. The better understanding of the mechanisms of endothelial dysfunction in this model may foster mechanistic studies on novel pharmacotherapeutic mechanisms of endothelial dysfunction in vivo.


Molecular logic lab-on-a-vesicle for intracellular diagnostics

Funding Agency: European Commission, Research Executive Agency, HORIZON 2020, LOGIC LAB
Copartner: Prof. Malgorzata Baranska (JCET, UJ)

A dysfunction of cells lining the inner walls of blood vessels, i.e. the endothelium, is the primary cause of many lifestyle related diseases. According to the WHO, those diseases accounted for 60% of all deaths worldwide in 2005. Tailor-made diagnostic tools for early and reliable identification of endothelial dysfunction are urgently needed both in fundamental research and clinical routine, respectively.

The Marie Skłodowska-Curie action LOGIC LAB objects to develop and characterize innovative molecular logic gates that can be applied as advanced diagnostic tools for parallel analyte sensing in live mammalian cells. Thereby, providing a unique method to discover endothelial dysfunction and the onset of diseases much easier and earlier than so far.

LOGIC LAB creates a multi-faceted and multi-sectoral research environment for the next generation of scientists in order to establish a novel type of molecular logic sensors that reliably operate in biological media – a crucial requirement for their application i.e. as rapid and easy-to-handle tools for intracellular diagnostics.

With excellent cross-disciplinary scientific and complementary training provided in the network, we aim to educate highlyskilled young scientists in the fields of chemistry, physics and biology, who will significantly strengthen the international.


Endothelial profiling to predict therapeutic activity or vascular toxicity of compounds in early preclinical research

Funding Agency: Foundation for Polish Science, Poland, Program: TEAM TECH Core Facility
Project leader: Prof. Stefan Chłopicki (JCET, UJ)

Cardiovascular safety liabilities remain the major cause of drug attrition during preclinical and clinical development. Lack of rationally-designed preclinical endothelial profiling of novel compounds also limits the possibility to discover vasoprotective activity of novel compounds. We assert that endothelial profiling may improve drug attrition related to endothelial toxicity, hopefully avoiding a repeat of the rofecoxib, torcetrapib, bardoxolone and omeprazol cases, and pave the way for the design of “endothelium-safe” therapeutics. Endothelial profiling will also provide an innovative tool to predict therapeutic efficacy that may direct further development of novel compounds. We present a proposal to develop and validate a panel of in vitro and vivo endothelial profiling that will be offered to the biotech/pharma industry. This approach predicting vascular toxicity and uncovering therapeutic efficacy of novel compounds, will present a real asset in the preclinical drug development.


Metabolic reprogramming in age-dependent endothelial dysfunction and vascular stiffness; novel mechanism of “inflamm-ageing”

Funding Agency: National Science Centre, Poland, Program: MAESTRO
Project leader: Prof. Stefan Chłopicki (JCET, UJ)

Inflamm-ageing is defined as the state of chronic sterile low-grade inflammation associated with ageing and is considered the major risk factor for cardiovascular diseases. Mechanisms of inflamm-ageing proposed so far, cannot fully explain age-related increase in inflammatory tone. Systemic endothelial dysfunction and increased stiffness of the large arteries represent two specific age-associated arterial phenotypes that are induced by inflamm-ageing and predict CVD-related morbidity and mortality. We hypothesize that age-related increase in inflammatory tone is linked to reprogrammed metabolic mechanisms of endothelium and vascular wall.

To the best of our knowledge, this project has a pioneering nature, takes advantage of state-of-theart methodologies and unique murine models, as well as offers a translational perspective. Given the fact that the systemic endothelial dysfunction and increased stiffness of the large arteries (dysfunctional vasculature) represent two specific age-associated arterial phenotypes that predict CVD-related morbidity and mortality, this project providing novel insight into metabolic reprogramming of dysfunctional vasculature in inflamm-ageing may open novel therapeutic opportunities.


The development of spectroscopic analysis of lipid droplets in vitro: biochemistry, and their localisation in relation to the biological function

Funding Agency: National Science Centre, Poland, Program: HARMONIA 8
Project leader: Prof. Malgorzata Baranska (JCET, UJ)

Lipids play an important role in the body, they are present in cells as the membrane component and in its interior, where, for example they store energy or transmit signals. These clusters are called intracellular lipid droplets, and despite the widespread presence in the cells of almost all organisms, knowledge of their functions, composition and the mechanism is still negligible. Excess of lipids, both in the circulation and at the level of tissues contribute to the disorder known as endothelial dysfunction, which can cause a number of metabolic diseases such as obesity, atherosclerosis
and diabetes and its cardiovascular complications.

As the role and composition of lipid droplets in the endothelium is not fully known hence the objective of the project is the complete, comprehensive study of these cellular organelles, in normal conditions and stimulating stress and disease. As the droplet size ranges from 20-40 nm to 100 microns, in situ analysis of cells requires the use of complex methods. This condition meets the vibrational spectroscopy in the form of several techniques. Not all are available in Poland, hence the need for cooperation with foreign partners who have experience in the study of lipids in different cellular models.


RS-SRS-MED: Spontaneous and stimulated Raman scattering microscopy for sensitive and ultrafast imaging of cells upon development of life style diseases

Funding Agency: National Science Centre, Poland, Program: OPUS
Project leader: Prof. Malgorzata Baranska (JCET, UJ)

Lifestyle diseases are a growing problem in aging societies. Although they are studied by an army of scientists around the world, many mechanisms of their development are still unclear. Better diagnostics and successful therapy could be based on the knowledge gathered from studies on biochemical processes associated progress of pathologies and their treatment at the cellular level, but it is very challenging since requires really fast methods of high sensitivity and selectivity. There are several biological models available to study biochemical processes, e.g. animals (in vivo), tissues (ex vivo) and cell cultures (in vitro). In the project, we plan to use animal models of civilization diseases and carry out measurements on tissues and cells. In particular, we are interested in the endothelium, whose dysfunction may lead to the development of many diseases (in some cases we observe secondary endothelial dysfunction as a result of these diseases). In this project we declare to develop a new methodology to study fast process at the subcellular level, including cellular transport of macromolecules. Transport of macromolecules across vascular endothelium and its modification, by e.g. fluid mechanical forces, are important for normal tissue function and in the development of various pathologies. Using spectroscopy, i.e. a method in which we examine the interaction of radiation with a sample, information on the biochemical status of samples can be obtained in a non-destructive and comprehensive manner. We plan to study biochemical changes at the cellular level using spectroscopic methods, first of all – Raman imaing, a classical approach (RS, Raman scattering microscopy) and non-linear one (SRS, stimulated Raman scattering microscopy), and biological models – cells of specific organs (e.g. vessels, heart, liver and brain) of animal models, mimicking atherosclerosis, fatty liver and Alzheimer’s disease (MEDical samples). RS-SRS-MED microscopy is not yet available in Poland; so we plan to design, build and test this approach, and for the first time to use the potential of SRS microscopy together with other techniques for analysis of cells of various organs. We strongly believe that RS-SRS-MED microscopy can be used as a tool to get new knowledge and to find new markers of lifestyle diseases, that will have an important societal impact, also related to alternative diagnostics and therapy of these diseases.


LDL-mimetic liposomal contrast agent for MRI-based detection of increased endothelial permeability in the early phase of endothelial dysfunction in vivo

Funding Agency: National Science Centre, Poland, Program: OPUS 20
Project leader: Anna Bar, PhD (JCET, UJ)

The overall aim of this project is: firstly, to develop optimized MRI-based methodology for the endothelial permeability assessment in murine models in vivo and secondly to use this optimized method to characterize endothelial permeability changes induced by disease development and in response to pharmacotherapy.

Endothelial cells form the largest endocrine organ in the body, that cover huge, inner surfaces of the entire cardiovascular system.In turn, endothelial dysfunction (ED) is a hallmark of various diseases and may be regarded as a barometer of cardiovascular risk.Moreover, studies on ED development in mice in vivo, constituting a basic model in preclinical studies, are essential for the better understanding of the role of endothelial cells in the disease progression, for indication the drugs with detrimental or beneficial effects on the endothelium or for identification of sensitive parameters of ED.Currently, various tests are used for the endothelial phenotype assessment, but most of them are not sensitive to detect the early phase of ED. However, based on our preliminary results, we hypothesize that among many methods of ED measurements, assessment of changes in permeabilityin vivo may provide one of the most sensitive approaches to detect changes in endothelial phenotype, but this avenue was not explored further and presents the subject of this grant proposal.The number of publications describing preclinical studies on endothelial permeability in vivoin mice is limited, due to requirement of using specialized tools and methodology. One of the methods allowing detection of endothelial permeability changes in vivo is magnetic resonance imaging (MRI)-based assessment of the gadolinium contrast agent (CA) accumulation in the vessel wall.However, there is still lack of sensitive CAs, which are uptaken by endothelium in a similar way as low-density lipoproteins (LDLs) to mimetic pathophysiological events, occurring in the early stage of increased endothelial permeability, in atherosclerosis.
The accumulation of LDL in the intimal layer of arteries represents a pivotal step in atherosclerosis development, therefore in the present project, the optimization of CA will rely on the development of LDL-mimetic liposomal CA, in order to increase the local concentration of CA in the vessel wall,uptakenin a similar way as LDL during increased endothelial permeability, in the very early phase of ED. In particular, liposome physicochemical parameters including size, composition, surface charge or surface accessibility, will be modified. Moreover, in the frame of optimization, liposomes will be decorated with apolipoprotein B100-mimetic peptide (the primary protein in LDL).During this studies the mechanism of the CA uptake in the vessel wall will be also identified.Alternatively to the MR imaging of increased endothelial permeability detected as an increased signal from accumulated in the vessel wall gadolinium-loaded liposomes, the imaging of 19-fluorine (19F)-loaded liposomes will also be used, what will might provide a more sensitive way for detection of changes in endothelial permeability, because of the lack of any 19F background in the body.In the second part of the project, optimized methodology will be used to assess the progression of increased endothelial permeability in E3L.CETP mice, the unique mice model for mild hyperlipidemia that displays human-like lipoprotein metabolism and representing a clinically-relevant model of ED.In E3L.CETP mice long term development of ED recapitulates slow progression of ED in humans with age-dependent changes in endothelium accelerated by mild hyperlipidemia. Our approach will allow for determining whether increased endothelial permeability assessment is suitable for the earliest ED detection, preceding other features of ED. Optimized MRI-based methodology for endothelial permeability assessmentin vivo, will be also used to perform proof-of-concept study on effects of nuclear factor 2 (Nrf2) activators on endothelial permeability, which despite beneficial effects on the endothelium, evidenced by a decrease in reactive oxygen species production, displayed a differential effect on endothelial barrier function in human microvascular endothelium. Moreover, the role of endothelin-1 and Nrf2 pathways involvement in the regulation of endothelial permeability by Nrf2 activators will be studied, what has not been performed to this date.


In search of the unexplored aspects of ferric heme protein-dependent regulation of nitric oxide signaling in the erythrocyte and vessel wall 

Funding Agency: National Science Centre, Poland, Program: OPUS 21
Project leader: Jakub Dybaś, PhD (JCET, UJ)

Nitric oxide (NO) is one of the major transmitters involved in maintaining of the proper vascular homeostasis in the circulatory system through contribution in lowering blood pressure and vasodilating smooth muscle cells. Bioactivity of NO is controlled, among the others, by heme proteins which participate in both – promoting and suppression of NO vasodilating property. Significance of heme proteins depend on the oxidation state of porphyrin-bound iron ion – ferrous heme proteins (with Fe2+ iron ion) scavenge and attenuate NO while ferric heme proteins (with Fe3+ iron ion) interact reversibly with NO allowing its diffusion and permitting bioactivity. However, precise role of ferric heme proteins remain elusive due to the lack of methodology allowing for simultaneous detection, differentiation and spatial distribution characterization of ferrous and ferric heme proteins. The aim of this project is to design the unique imaging methodology based on resonance Raman (rR) spectroscopy allowing examination of unexplored aspects of NO bioactivity regulation by ferric heme proteins and better understanding of mechanisms regulating NO signaling in erythrocyte and vessel wall.


Normalization of energy metabolism as a way to develop innovative antithrombotic therapies 

Funding Agency: National Science Centre, Poland, Program: OPUS 21
Project leader: Patrycja Kaczara,PhD (JCET, UJ)

The aim of the project is to investigate the relationship between the reactivity of blood platelets and their energy metabolism. The development of thrombosis is accompanied by increased platelet reactivity, caused, among others, by diseases such as diabetes or atherosclerosis, both accompanied by disturbances of metabolic processes. The hypothesis of the project assumes that the pharmacological normalization of metabolic processes in platelets can lead to an improvement of the effectiveness of commonly used antiplatelet drugs. The results of the project can be used to formulate dietary guidelines to help slow the development of thrombosis and to demonstrate the potential of biochemical pathways to develop new antiplatelet therapies to prevent or reduce the serious and life-threatening effects of thrombosis.


Lipid unsaturation degree in perivascular adipose tissue – a new biomarker of vascular wall inflammation? Raman studies of murine caridovascular models and primary isolated adipocytes

Funding Agency: National Science Centre, Poland, Program: OPUS 17
Project leader: Agnieszka Kaczor, PhD, D.Sc. (JCET, UJ)

Perivascular adipose tissue (pVAT) surrounds all blood vessels except for the brain vasculature. Since some time it has become clear that pVAT shows tremendous endocrine activity and can exert significant influence on the inner layers of the vascular vessels via releasing nitric oxide, angiotensin, leptin, adiponectin and an enigmatic adipocyte derived relaxing factor. A dysfunctional pVAT produces a plethora of pro-inflammatory adipokines, cytokines and chemokines and is considerably involved in progression of cardiovascular diseases. Therefore, it is not surprising that pVAT is a potential novel therapeutic target of the prevention and treatment of cardiovascular diseases. Yet, chemical changes in pVAT under development of cardiovascular pathologies have not been investigated, partially due to the lack of an adequate methodology. The fundamental objective of the project is investigation of chemical changes of pVAT in various vascular beds under development of model murine cardiovascular pathologies in order to find the chemical marker(s) of pVAT/vascular inflammation for potential cardiovascular risk prediction. We hypothesize that the lipid unsaturation degree may be a plausible marker of pVAT dysfunction with a potential toward diagnostic applications.


Spectral histopathology, FTIR and immunoSERS, for the recognition of biochemical status of pre-metastatic niche and micrometastasis in murine model of breast cancer

Funding Agency: National Science Centre, Poland, Program: OPUS 12
Project leader: Kamilla Malek, PhD, D.Sc. (JCET, UJ)

The project entitled ” Spectral histopathology, FTIR and immunoSERS, for the recognition of biochemical status of pre-metastatic niche and micrometastasis in murine model of breast cancer ” aims to “take fingerprint” of cancer metastasis before it fully develops. As is generally known, cancer is the most deadly disease in the world and despite the continuous development of new diagnostic and therapeutic strategies, the number of new cases and deaths continues to rise. It is predicted that 50% of new cases among women and men will be affected by breast/prostate, lung, bronchial and colorectum cancers, of which 20% is attributed to breast cancer. But the primary tumor is not the main cause of death, only its metastasis. This process occurs through intravasation of cancer cells into blood and lymphatic vessels, their transport via systemic circulation to distant organs and then the formation of micrometastases proliferating to macrometastasis. It is also known that the metastatic site is not accidental but an organ “receives” the chemical signal from the primary tumor to “prepare” its tissue matrix to accept the unwelcome guest and to create so-called the pre-metastatic niche. And this project is dedicated to the recognition the pre-metastatic niche and micrometastases by advanced imaging techniques of vibrational motion of all the componentspresent in a biological system, i.e. by infrared absorption spectroscopy and Raman spectroscopy using specially designed nanosensors (surface-enhanced Raman spectroscopy, SERS). Vibrations of molecules are specific for each of them and provides a unique “fingerprint” which is described in terms of chemical composition. If we combine such as detection with spatial measurement we get an image of chemical information strictly specific for a sample. Based on such research technology, we will image tissues and cells from lungs to that metastasis occurs in breast cancer. Designed research requires collection of biological material at well-defined stages of metastasis and therefore we will carry out our studies on animal models of cancers.


The role of MCPIP1 RNase in disrupted endothelial homeostasis accompanying non-alcoholic fatty liver disease

Funding Agency: National Science Centre, Poland, Program: OPUS
Project leader: Jerzy Kotlinowski, Ph.D. (UJ, JCET)

The main goal of our project is to determine role of RNase called MCPIP1 (MonocyteChemoattractant Protein-Induced Protein 1) in paracrine activation of endothelial cells. MCPIP1 is one of the protein involved in a negative regulation of inflammation. MCPIP1 binds to mRNAs and digests it – in the similar way to ‘Pacman’ from the classic PC computer game. Such activity of MCPIP1 defines it as an enzyme called “endoribonuclease”. MCPIP1 shortens life of selected transcripts leading, as a result, to reduced protein amount of IL-1β and IL-6 for example. Both of above-mentioned proteins are important in the development of inflammation. Development of inflammation is tightly linked to the function of endothelial cells. Endothelium plays a pivotal role in maintaining homeostatic balance of the organism. In normal conditions it acts like a selective permeable barrier between organs, blood and immune cells. However, under pathological state, e.g. exaggerated inflammation, endothelium becomes activated or damaged what leads to its loss of function:

  1. increased expression of adhesion molecules,
  2. enhanced permeability,
  3. diminished production of NO,
  4. migration of leukocytes to the surrounding tissue.

In this project we want to test what impact will have proinflammatory activation of hepatocytes and leucocytes, related to Mcpip1 depletion in these cells. We hypothesize, that lack of Mcpip1 in hepatocytes and leukocytes of the myeloid lineage will lead to enhanced production of proinflammatory mediators by this cells. As a result, we will be able to demonstrate activation or damage of endothelium by Mcpip1 acting in a cells’ extrinsic way. For analysis of Mcpip1 in activation of hepatocytes and myeloid leukocytes leading to altered paracrine stimulation of endothelium we will use genetically modified mice that do not have Mcpip1 protein in hepatocytes and in leukocytes of myeloid origin. In our opinion, demonstration of Mcpip1 involvement in regulation of endothelial cell biology in a cells’ extrinsic way can contribute to the discovery of new medical therapies. Compounds that inhibit degradation of Mcpip1 might be used in the future for designing new drugs used in the treatment of chronic inflammation. Since Mcpip1 inhibits inflammation, its local activation should help to restore homeostasis unbalanced by unresolved or untamed inflammatory processes.


Breast cancer metastasis-induced endothelial-mesenchymal transition alongside ageing; implications for therapy

Funding Agency: National Science Centre, Poland, Program: OPUS 21
Project leader: PhD Marta Smęda (JCET, UJ)

Ageing results in progressive vascular dysfunction. The cornerstone mechanism that may lay behind age- and/or cancer-dependent deterioration of endothelial function is mesenchymal transformation of endothelial cells (EndMT) that may determine the endothelium status and, thus, the outcome of breast cancer. Progression of EndMT and its consequences in ageing remain largely unknown. Both cancer and ageing alter platelet phenotype. Therefore, although circulating platelets of healthy individuals support endothelium function, age- and/or cancer-altered platelets may act in the opposite way: drive EndMT and disrupt endothelial barrier.

The project is based on the assumption that mesenchymal transformation forms the basis for age-related endothelial dysfunction and its advancement determines endothelium status as well as the outcomes of breast cancer. The aim of the project is to investigate mesenchymal transformation in the systemic circulation (aorta), and pulmonary circulation (the lungs) alongside ageing as well as during cancer metastasis. It will be also investigated if age- and cancer-altered platelets can affect EndMT progression and endothelial barrier. Finally, it is also planed to seek candidate proteins released from platelets which can determine this process.


CPL and RROA probes in the study of protein structure – induction of chirality and new methods of amplifying the chiroptical signal

Funding Agency: National Science Centre, Poland, Program: OPUS 18
Project leader: PhD Grzegorz Zając (JCET, UJ)

Protein structural alterations play a key role in many neurodegenerative and civilization diseases, including Alzheimer, Parkinson disease, atherosclerosis, diabetes and other diseases that are related to the endothelium dysfunction. Typical protein structural changes upon disease state is the chemical modifications (phosphorylation, methylation, oxidation, peroxidation etc.), mutation of the primary structure, modification of the secondary structure (α-helix, β-sheet, etc.), as well as aggregation into fibril form. Spectroscopic methods sensitive to the chiral samples, have been recently proved to be very sensitive to the three dimensional structure, and structure alterations of the chiral biological systems, including proteins, and their aggregates, showing additional structural information, not reachable by any other method. However due to its nature, those methods usually hamper from the low intensity of the obtained signal, thus they were unsuitable for study of biomolecules in their natural, low concentrated, environment. As it recently turns, some of that methods, ROA (Raman Optical Activity) and VCD (Vibrational Circular Dichroism) can be enhanced due to the aggregation increasing of their sensitivity. Furthermore, another spectroscopic method: CPL (circularly polarized luminescence), and CPL based signaling probes, could also increase not only the intensity of the signal, but also sensitivity to structural changes. The main scientific goal of the project is the spectroscopic screening of the structural modification of the amino acids, peptides and proteins in the model and biological systems, upon the influence of various physical-chemical conditions, as well as study of the intermolecular interaction of signaling probes with protein based, molecular and aggregated systems.  To clarify additional structural information from spectroscopic measurements, the theoretical calculations of the spectroscopic properties of various molecular and supramolecular models will be also performed.


Raman spectroscopy in vitro studies of chemotherapeutic impact on endothelial cells

Funding Agency: National Science Centre, Poland, Program: SONATA 11
Project leader: Katarzyna Majzner, PhD. (JCET, UJ)

The project involves application of multi-parameter methodology with the use of confocal Raman spectroscopy, infrared absorption spectroscopy, atomic force microscopy (AFM) combined with biochemical tests to simultaneously evaluate changes in endothelial cells upon chemotherapeutic exposition in the level of morphology biochemistry and physicochemistry. Research hypothesis of the project is presence of spectroscopic markers (biomarkers) indicating early changes in biochemical profile of endothelial cells which are results of chemotherapeutic impact leading to toxic side effects for cardiovascular system. To confirm research hypothesis planned is what following:

  1. endothelial profiling of anticancer drug via viability tests, measurements of prostacyclin and NO generation, and reactive oxygen species for confirmation of oxidative stress presence,
  2. spectroscopic measurements leading to investigating of biochemical composition of endothelial cells stimulated with anticancer drugs and supported with AFM measurements,
  3. developing of complex method based on detection and analysis of biomarkers, including spectroscopic markers. Planned researches are multidisciplinary linking application of spectroscopic method with pharmacological drug profiling.

In addition, examination of cells using Raman spectroscopy will be supported by atomic force microscopy (AFM), which allows for simultaneous examination of the biochemical composition and topography of the analyzed sample. Applying a combination of Raman spectroscopy and AFM to study these same areas of the cells it is possible to obtain complete information about the biochemical composition of endothelial cells in vitro and the distribution of the individual components and investigate physical and mechanical properties of the sample.

To the expected effects of the project include getting the information about composition and distribution such chemical components like DNA, lipids, proteins or carbohydrates. Although the results have no direct potential application, they will contribute to new information and reassessment of existing scientific data. The results of the project will be presented at the conference forum and published in pre-reviewed papers in an international chemical or chemistry-related journals. Experience of the Raman Imaging Group Faculty of Chemistry UJ in terms of Raman spectroscopy (measurements and advanced spectral analysis), as well JCET research groups in in vitro pharmacological and biological studies, and the availability of necessary equipment for the project allow for the statement that the risk of this project is low.


Ecto-enzymes in interactions of vascular endothelium with blood circulating cells in physiology, pathology and therapy; could cells exchange their ecto-enzymes?

Funding Agency: National Science Centre, Poland, Program: SONATA 15
Project leader: Karbara Kutryb-Zając,PhD. (MUG), collaboration (JCET, UJ)

The goal of the awarded project is to demonstrate the role of ecto-enzymes in direct contact between cells. Ecto-enzymes are proteins located on the surface of cells, metabolizing a wide spectrum of substrates, which mediate intercellular communication in physiology and pathology. As part of the project, changes in ecto-enzyme activity induced by the interaction of vascular endothelium with blood cells that can stimulate the formation of an adverse endothelial cell phenotype will be demonstrated. In addition, the role of ecto-enzymes as adhesion molecules and the therapeutic possibilities of compounds modulating their properties in vascular pathologies and cancer will be investigated.


Impairment of coronary microcirculation in a mouse model of a heart failure

Funding Agency: National Science Centre, Poland, Program: SONATA 16
Project leader: Grzegorz Kwiatkowski,PhD (JCET, UJ)

In this project we aim to apply new protocols using advanced Magnetic Resonance Imaging methods to directly and indirectly observe changes in microvascular status occurring in coronary microcirculatory system and the corresponding impairment of the cardiac blood flow supply (so-called perfusion) using a murine model of heart failure. For this purpose, we plan to use a special breed of mice that over their life-span develop a cardiac impairment which is very similar to what we observe in human patients with congestive heart failure. We plan to probe the changes in coronary microcirculation during the whole progression of heart failure in animals and later to use the newly acquire knowledge to assess the effect of pharmacotherapy with several medication groups, commonly used to treat heart failure in humans.


From the structure of fenestrations in live Liver Sinusoidal Endothelial Cells to real-time in vitro pharmacology

Funding Agency: National Science Centre, Poland, Program: SONATA 15
Project leader: Bartłomiej Zapotoczny,PhD. (IFJ PAN), collaboration (JCET, UJ)

For nearly 50 years since the discovery of fenestrae, the speed of processes associated with the changes in the number and size of fenestrae remained speculative. With the development of atomic force microscopy (AFM) imaging methodology, new, so far out of reach, methods of investigating the porosity of LSEC cells appeared. The current literature research, including ours which is based on AFM imaging, indicate the huge role of the cytoskeleton in the structure of fenestrae. The aim of this project is to understand the role of the cytoskeleton in fenestrae formation and maintenance, as well as the influence of vasoprotective mediators on the porosity of live primary, freshly isolated murine LSECs in vitro. The experiments will be conducted in three models of defenestration in LSECs. The research will provide information on the phenotypic changes in the cytoskeleton in real time. The interactions between cytoskeletal elements in living cells, both intact and exposed to pharmacological tools, can be translated into other cell types. This will allow you to learn about the dynamics of the LSEC cytoskeleton and to understand the mechanisms regulating fenestrae, which is of direct importance for the treatment of liver diseases.


Endothelial inflammation studied in situ within functional blood vessels using Raman imaging, atomic force microscopy and imaged cytometry

Funding Agency: National Science Centre, Poland, Program: SONATINA 1
Project leader: Marta Z. Pacia, PhD. (JCET, UJ)

The vascular endothelium forms the innermost layer of all blood and lymphatic vessels. Together with the underlying basement membrane it forms the tunica intima. This inner layer is surrounded by the tunica media which, depending on local mechanical and metabolic needs, predominantly consists of vascular smooth muscle cells and elastin fibres. Endothelium is a thin, unicellular layer of highly specialized cells lining the interior surface of blood and lymphatic vessels. It is not only a physical barrier between blood and tissue, but it also plays a plethora of essential functions in the organism. It regulates vascular tone, permeability of the vessel wall, smooth muscle cell proliferation and migration, trafficking of inflammatory cells between blood and underlying tissue, angiogenesis, innate and adaptive immunity organ regeneration, fibrinolysis and inflammation.

This project involves the measurement of in situ endothelial cells within isolated functional blood vessels. This means that after dissection of the blood vessel from the mice body, the split-open vessel wall is kept in the special medium supporting the functionality of endothelial cells. The project involves the development of comprehensive methodology including high resolution Raman imaging, Raman spectroscopy via Raman fiber probes, AFM imaging (topography, phase and amplitude), force distance curve measurements and biological functional assays for analysis of in situ endothelial cells.

Enlargement of background knowledge about influence of the inflammation state on in situ endothelial cell seems to be important step toward specifying the markers of inflammation state in the endothelium. The results of chemical alterations (e.g. creation of lipid bodies measured by Raman imaging), physical changes (e.g. increasing stiffness checked by AFM force distance curves measurements) or reduced amount of NO (biological assays) upon stimulation by pro-inflammatory agents will be characterized and compared. The next step of the project assumes application of combination of methods to pharmacology of endothelial cells within functional isolated blood vessel. In the selected model of inflammation, the effectiveness of endothelium-protective agents like statins or angiotensin converting enzyme inhibitors on depicting markers of inflammation will be evaluated.


Scholarship of the Minister of Science and Higher Education for outstanding young scientists

Funding Agency: Ministry of Science and Higher Education, Scholarship
Project leader: Marta Z. Pacia, PhD. (JCET, UJ)

Scholarship of the Minister of Science and Higher Education for outstanding young scientists showing significant achievements in scientific activity in 2018.


In search of spectroscopic markers of autophagy based on pharmacologically induced phospholipidosis in endothelial cells

Funding Agency: National Science Centre, Poland, Program: PRELUDIUM 18
Project leader: Ewelina Bik, M.Sc.,  Ph.D. student

It has been observed that patients taking psychotropic drugs from the CADs (cationic amphiphilic drugs) group have an increased risk of developing the cardiovascular diseases. These drugs can cause phospholipidosis in the cells. I hypothesize that the drug-induced phospholipidosis mechanism is the result of modulation of autophagy in endothelium, which is a barrier between vessel wall and blood. In addition, the research hypothesis to be verified here is the possibility of finding spectroscopic markers of autophagy in endothelial cells. The participation of autophagy in drug-induced phospholipidosis and lipid metabolism in the endothelium is still an unexplored area. Spectroscopic methods, such as the main analytical method proposed here – Raman microscopy, allow for non-destructive testing of biological material without the use of labels. Thus, it is possible to identify lipids, their content and distribution in cells, simultaneously studying other cellular components and drugs that cells are treated with. The project plan is to study phospholipidosis, which occurs when phospholipids accumulate excessively in cells and tissues. CADs, i.e. those used to treat depression, psychosis, malaria and arrhythmias can induce phospholipidosis in cells. The aim of the project is to find specific spectroscopic markers of autophagy by studying in vitro the effect of selected CADs drugs on endothelial cells using Raman spectroscopy, fluorescence microscopy, and biological tests. In particular, the emphasis will be put on searching for these spectroscopic markers, that will allow identifying early biochemical changes in endothelial cells caused by drug-induced phospholipidosis. From the point of view of endothelial pharmacology, the term and understanding the mechanism of phospholipidosis along with its identification, may be a response to the growing concerns about the CADs administration.


The role of lactate dehydrogenase A in development of endothelial dysfunction in murine model of heart failure

Funding Agency: National Science Centre, Poland, Program: PRELUDIUM 20
Project leader: Agnieszka Karaś, M.Sc.,  Ph.D. student

The aim of this project is to evaluate the link between vascular metabolism and endothelial dysfunction during the development of heart failure. Heart failure (HF) is currently a leading cause of mortality worldwide, moreover, in Poland, the morbidity and mortality of HF are one of the highest in Europe. Interestingly, systemic endothelial dysfunction is observed in patients even with mild HF and is an important factor predicting poor patient outcome and increased risk of cardiac death. Endothelium, the single layer of cells lining the interior surface of blood vessels, plays a crucial role in maintaining healthy vessel function. To date, the mechanisms of endothelial dysfunction development in HF are not clear. We hypothesise that alterations in cellular bioenergetics in the vessel wall may contribute to vascular pathology in the progression of HF. Therefore, this study is set out to understand the connection between endothelial dysfunction and metabolic changes focusing on the role of lactate dehydrogenase A (LDHA), an enzyme responsible for the production of lactate, which is not only an energy source but also a crucial regulatory factor. This project will be performed using a murine model of HF. Results of this study will shed new light on vascular pathophysiology in HF and may contribute to novel therapeutic strategies improving patient prognosis.


The anti-inflammatory effect of vitamin K – Mechanism of action

Funding Agency: National Science Centre, Poland, Program: PRELUDIUM
Project leader: Anna Kierońska-Rudek, M.Sc.,  Ph.D. student

Research hypothesis Recent studies demonstrate that vitamin K functions goes beyond coagulation and calcification including inter alia anti-inflammatory effects but the mechanisms of this action is not fully understood. Our primary studies in murine macrophages indicate that the anti-inflammatory effect of vitamin K encompasses a wider spectrum of effects than has been describing until now. We postulate that vitamin K regulate a wide scope of pathways during inflammation in macrophages. We will comprehensively characterize these effects in macrophages and examine the involvement of vitamin K- dependent carboxylation, regulation of NFkB signaling pathway and mitochondrial functions in the anti-inflammatory mechanisms of vitamin K (K1, K3, K2MK4, K2MK5, K2MK7, K2MK9) action in these cells.

The benefits for science in terms of basic research carried out by this project are as follows:

  • Defining the anti-inflammatory profile of action of a wide range of commonly used and new representatives of the vitamin K group (K1, K3, K2MK4, K2MK5, K2MK7, K2MK9).
  • Determination of the influence of commonly used anticoagulants – vitamin K inhibitors (warfarin, acenocoumarol) on the anti-inflammatory effect of vitamin K
  • Proteomic analysis of the presence of vitamin K dependent proteins in M1 / M2 macrophage subpopulations
  • Characterization of a broad profile of changes in inflammatory markers in response to treatment with vitamin K in the M1 / M2 macrophage subgroups.
  • Comprehensive assessment of the carboxylation share of the anti-inflammatory effect of vitamin K in macrophages, including M1 / M2 polarization
  • Determination of the involvement of the NFkB signaling pathway in the anti- inflammatory effect of vitamin K in M1 / M2 macrophages
  • Determination of the effect of vitamin K on the improvement of mitochondrial function in M1 / M2 macrophages

Studies on biologically relevant systems in conditions of resonance Raman optical activity

Funding Agency: National Science Centre, Poland, Program: PRELUDIUM 17
Project leader: Ewa Machalska, M.Sc.,  Ph.D. student

The aim of the project is to study the structure and chirality of biologically important molecules as well as a development of Raman optical activity methods in the resonance conditions. Experimental approach will be supported, to some extent, by theoretical calculations for investigation of resonantly enhanced Raman optically active systems, such as vitamin B12 and its modifications, and amphotericin B (AmB). Apart from Raman optical activity (ROA) spectroscopy other chiroptical techniques, i.e. electronic circular dichroism (ECD) and vibrational circular dichroism (VCD) will be used. Vitamin B12 (CNCbl) was selected as a main object because it shows strong chirality due to ten stereogenic centres in the chromophoric corrin ring. Moreover, enzymatically active forms of vitamin B12 play an essential role in various biological processes. Therefore, the 1st goal of the project is to investigate exhaustively the complex structure and strong optical activity of corrinoid molecules (vitamin B12) to check the structure-activity relation and to understand better a key role of these compounds. ROA spectroscopy seems to be a powerful tool to investigate chiral features of molecules, however, the method is hampered by its low sensitivity, so the 2nd goal is a development of resonantly enhanced methods providing ROA significant signal enhancement. The 3rd goal is to explore the impact of modifications of vitamin B12 on its structure, its ability to interact with proteins/drugs and hence biological activity. Another research object is AmB exhibiting strong chirality due to nineteen chiral centres. It is known that AmB has an unusual ability to form molecular aggregates and thus it can be an example of experimental observation of aggregation induced pre-resonance Raman Optical Activity (AIRROA).


Perivascular adipose tissue versus gut microbiota: Raman imaging study

Funding Agency: National Science Centre, Poland, Program: PRELUDIUM 20
Project leader: Zuzanna Majka, M.Sc.,  Ph.D. student

For many years, perivascular adipose tissue (PVAT) has been overlooked by scientists studying human anatomy. Until the 1990s, PVAT was routinely removed during the preparation of blood vessels for their further examination. However, scientists proved the effect of PVAT on blood vessels and thus ushered in a new direction of research in 1991. Nowadays it is known that PVAT as a paracrine tissue actively participates in vascular homeostasis and its dysfunctions are inherent in atherosclerosis, obesity and insulin resistance. Gut microbiota has also become a top of top topic among scientists dealing with cardiometabolic diseases. It is estimated that 40,000 different species of bacteria live in the human intestine, which absorbs about 10% of daily nutrients. Indigestible carbohydrates, such as fiber or starch, are used by gut microbiota to produce short chain fatty acids. Metabolites of the gut microbiota, including most often acetic, propionic and butyric acid, affect, among others functioning of the liver, pancreas or adipose tissue by improving lipid and glucose metabolism, thus protecting against obesity and insulin resistance.

The aim of the project is to investigate the effect of gut microbiota on perivascular adipose tissuefrom the point of view of the development of cardiometabolic diseases.In our multifaceted research, both murine models and the in situ functional PVAT model will be used.The main research technique used in the study will be Raman spectroscopy. This technique is based on inelastic light scattering and is more and more often applied in biochemical studies.The state of the gut microbiota will be investigated using Next Generation Sequencing (NGS). We suppose that gut microbiota via SCFA will impact PVAT and will (at least partially) reverse the negative influence of obesity and insulin resistance caused by a HFD.


Comparative evaluation of the effects of tyrosine kinase inhibitors (TKIs) on endothelial function in vivo in mice

Funding Agency: National Science Centre, Poland, Program: PRELUDIUM 17
Project leader: Brygida Marczyk, M.Sc.,

TKIs block the action of tyrosine kinases that regulate many cellular functions, including cell signalling, growth and division. These enzymes may be overactive in some types of cancer cells, promoting cell growth, which in turn may be effectively inhibited by the use of TKIs. The introduction of TKIs radically changed the treatment of various cancers, including chronic myeloid leukaemia (CML). However, long-term use of TKIs is associated with unwanted vascular and procoagulant effects, which has a significant impact on the morbidity and mortality of patients due to adverse vascular events. The first introduced TKI was imatinib, however, due to the observed resistance to this drug, TKI of 2nd generation (nilotinib, dasatinib and bosutinib) and then of 3rd generation TKI (ponatinib) were introduced. While unwanted vascular effects have been observed many times with ponatinib and nilotinib treatment, imatinib treatment is considered to be free from significant adverse vascular and prothrombotic effects. These clinical observations suggest distinct effects of various TKIs on the cardiovascular system and vascular endothelium in particular. As there are no studies that compare in experimental conditions in vivo toxicity profile of various TKIs in animals we plan to characterize the effects of TKIs, in vivo mice. In particular, we plan to assess whether TKIs, with good and limited safety profile as suggested in clinical studies in humans, would have a similar or different effect on the endothelial function in physiological and pathophysiological conditions in mice (healthy mice and mice with atherosclerosis, respectively). Accordingly, the aim of the study is to comprehensively compare the effects of tyrosine kinases inhibitors (TKIs) on endothelial function. A state-of-the-art methodology based on magnetic resonance imaging (MRI) will be used to assess the effects of TKI on endothelial-dependent responses and changes of endothelial permeability in murine models in vivo. The assessment in vivo will be based on the detection of impaired vasodilatation induced by acetylcholine, which is related with impaired generation of endothelial nitric oxide (NO) or paradoxical vasoconstriction in response to acetylcholine in advanced stages of endothelial dysfunction. In addition, the endothelial function assessment will be complemented with the assessment of vasodilation resulting from increased blood flow after short-term occlusion (flow mediated-dilatation, FMD), which is the gold standard technique to assess endothelial function in humans. Finally, MR imaging will be also used to assess endothelial permeability with a contrast agent. The above MRI-based methods for the assessment of endothelial phenotype in vivo will be supplemented by classical methods of measurements of endothelial function using isolated vessels and biochemical measurements of NO production in ex vivo vessels. We claim that this project will demonstrate whether TKIs (1st, 2nd, 3rd generation) with apparently distinct vascular safety profile in humans have a different effect on endothelium in healthy mice or in murine models of atherosclerosis. As such this project will help to better understand the mechanisms of vascular toxicity of TKIs and should open the novel methodological approach to assess vascular toxicity of compounds in preclinical studies based on the comprehensive assessment of endothelial function in vivo using unique MRI-based approach.


Role of angiotensin 1-12 and ACE-independent generation of Angiotensin II in peripheral endothelial dysfunction in heart failure

Funding Agency: National Science Centre, Poland, Program: PRELUDIUM 15
Project leader: Tasnim Mohaissen, M.Sc.,  Ph.D. student

The renin-angiotensin-aldosterone system (RAAS) plays fundamental role in cardiovascular diseases including heart failure. In particular, primary detrimental mediator of this system angiotensin II (Ang II), contributes to cardiac remodeling, arrhythmias, fibrosis and endothelial dysfunction. Current pharmacotherapy of heart failure is mainly based on minimizing the pathogenic effects of Ang II, however, recent studies show that tissue pathways generating Ang II are resistant to angiotensin-converting enzyme inhibitor (ACEI) representing a mainstay of heart failure treatment. Accordingly, it is a critical need to understand alternative pathways for generating Ang II in vascular wall and as well as in other tissues and to design a novel pharmacotherapeutic approach to inhibit effects of Ang II in tissues.

The main purpose of this project is to assess the involvement of ACE-independent pathways in the development of peripheral endothelial dysfunction in the course of pathology development of HF in a unique mouse model of heart failure (Tgαq*44). Methods which will be used in this study are a combination of analytical, pharmacological and biochemical research that allow to obtain high quality results. This study will characterize both; the development of peripheral endothelial dysfunction using functional and biochemical measurements, and the alteration of alternative Ang II formatting pathways- with their enzymes, using functional and liquid chromatography /mass spectroscopy method. Based on the results of the research we can obtain a unique model for the study new compounds in heart failure disease, which will allow us to understand the role of alternative pathways in the formation of Ang II and the development of cardiovascular disease.


Novel strategies for pharmacological modulation of platelet-derived microparticles – effects on endothelium

Funding Agency: National Science Centre, Poland, Program: PRELUDIUM 19
Project leader: Agnieszka Pełesz  M.Sc.

Currently, antiplatelet therapy plays an important role in prevention of arterial thrombosis and is thought also to be effective in preventing metastasis spread. Yet, we still have gaps in our understanding how antiplatelet agents affect cross-talk mechanisms between cells. The main intercellular communication mechanism of activated platelets involves extracellular vesicles transfer. Thus, extracellular vesicles act as important messengers in health and disease. Platelet-derived extracellular vesicles (PEVs) exhibit a wide range of pro-thrombotic, pro-inflammatory properties as well as can afford endothelial barrier protection. Therefore, it seems crucial to know how applied anti-platelet treatment affects modulation of PEVs release and their properties as messengers. To date, limited studies are available considering this topic, despite the fact that the repertoire of antiplatelet agents is still increasing. In particular, it is not known how antiplatelet agents modulate PEVs release and their effects on endothelial cells’ phenotype in healthy conditions as compared with atherosclerosis.

In the current project, we will take the advantage of unique methods that have been developed in JCET for comprehensive endothelial profiling in vitro and ex vivo. Combining them with methods that will be developed within the frame of this project, we aim to broaden the knowledge about effects of antiplatelet pharmacology on extracellular vesicles release and function and their cross-talk with healthy endothelium or dysfunctional endothelium in atherosclerosis. Importantly, the research can have its continuation also in a context of different disease entities, where antiplatelet treatment effectiveness is reported biased. By complement this knowledge within the project, possibly new therapeutic avenues for many disease entities that are associated with vascular endothelial dysfunction can become opened.


Cross-talk between endothelium and perivascular adipose tissue upon inflammation: high-resolution imaging of adipocytes in co-culture with endothelial cells

Funding Agency: National Science Centre, Poland, Program: PRELUDIUM 22
Project leader: Ewa Stanek  M.Sc.

The role of perivascular adipose tissue (PVAT) in the progression of vascular pathology, including endothelial dysfunction, is not yet well understood. Hence the overarching goal of this research is to understand the interaction of endothelial cells (ECs) and PVAT adipocytes, under normal cell development as well as under pathological conditions caused by a high-fat diet (HFD, obesity model) and pro-inflammatory factors connected with obesity. Due to the lack of commercial PVAT cell lines, this project will be the first to identify new in vitro models to study this tissue which will contribute to the field of vascular biology and a better understanding of related diseases.

The project will focus on two models of cell co-cultures based on direct or indirect interactions between PVAT adipocytes and ECs in vitro. PVAT progenitor cells from healthy and obese mice (fed with HFD) will be differentiated into mature adipocytes to analyze the effects of obesity on PVAT chemical composition and its impact on ECs. Additionally, the murine cell line of primary aortic ECs will be exposed to pro-inflammatory agents, i.e. tumor necrosis factor (TNF), to examine how inflamed ECs impact the regulation of PVAT. To assess the involvement of PVAT in vascular metabolism as well as to uncover markers of inflammation resulting from changes in the biomolecular composition of lipid droplets, several methods will be used, including high-spatial resolution Raman microscopy.


Function and phenotype of liver sinusoidal endothelial cells (LSECs) and their role in liver dysfunction development in murine model of heart failure.

Funding Agency: National Science Centre, Poland, Program: PRELUDIUM 18
Project leader: Kamila Wojnar-Lasoń, M.Sc., Ph.D. student

Heart failure (HF) occurs when the heart is unable to pump sufficiently to sustain the blood flow required to meet body’s metabolic demands. A number of cardiac and systemic pathophysiological factors involved in HF have been well-defined but the mechanisms of the progression of chronic heart failure are still not entirely understood. In particular, a large proportion of chronic heart failure patients display liver dysfunction, a condition that adversely affects the prognosis of HF, complicating the management of HF. In fact, liver diseases can cause cardiac dysfunction and failure in the absence of other cardiovascular impairments and the mechanisms of intricate connections between the heart condition and the progressive deterioration of the liver function in HF are still not elucidated. Liver sinusoidal endothelial cells (LSECs) are highly specialized endothelial cells with a unique morphology and function. LSECs are present only in the liver lining the hepatic sinusoids and represent a permeable barier allowing active uptake and degradation of molecules. LSECs produce number of vaso- and hepato-protective mediators and impaired LSECs activity is considered to be the cause of many liver diseases e.g. nonalcoholic fatty liver disease, liver steatosis, liver fibrosis and others. There is a convincing evidence that LSECs play a prominent role in maintaining proper liver function, but LSECs response to the progression of HF has not been studied yet. The aim of this project is to characterize the alterations in liver function during progression of HF in Tgαq*44 mice, which are considered a reliable and unique model for studying the progression of this disease. In fact, Tgαq*44 mice mimic pathophysiology of human chronic HF. A particular attention will be dedicated to the role of LSECs, as they are a master regulator of the hepatic circulation, and could be responsible for the activation of mechanisms involved in development of the liver dysfunction accompanying HF.


The role of endothelial Akt1 and Akt2 in the modulation of angiotensin II-induced endothelial dysfunction; a possible upstream mechanism of non-classical angiotensin pathway

Funding Agency: The Polish National Agency for Academic Exchange
Project leader: Magdalena Sternak, M.Sc., Ph.D. 

The role of selective endothelial protein kinase Akt in the regulation of endothelial dysfunction is not clear. Here, we analyze the role of Akt1 and Akt2 in the regulation of NO/ROS and ACE/ACE2 balance in vascular wall as well as endothelial permeability changes and development of endothelial senescence in response to angiotensin II by using the unique, endothelial selective Akt-deficient mice. Moreover, we verify whether Akt is involved in the novel upstream regulation of non-classical angiotensyn pathway towards effective Akt-dependent pharmacology.

Budget: 84000 PLN, Time: 6 months


Spectroscopic characteristics of perivascular adipose tissue in the progression of atherosclerosis

Funding: National Research Centre (funding agency), Program MINIATURA
Project leader: Krzysztof Czamara, PhD

The project is planned for original studies of perivascular adipose tissue (PVAT) of mouse model of atherosclerosis. The aim of the project is to define the chemical composition of PVAT and to determine the chemical changes in the progression of atherosclerosis using Raman spectroscopy techniques, including the use of Raman fibre optic probes. Atherosclerosis is a process that is quite well understood in the context of the influence of the endothelium layer on its pathogenesis, however, the recent discovery of the paracrine interaction of PVAT opens up a new field for the study of this disease. The chosen direction of research seems to be an interesting and a good starting point for the future early diagnosis of atherosclerosis, aimed at the prevention and treatment of vascular diseases. This type of research can also contribute to the understanding of the mechanisms of action of PVAT on the wall of blood vessels.

FINISHED PROJECTS

Prostacyclin, nitric oxide and carbon monoxide-based pharmacotherapy of endothelial dysfunction and platelet activation-a novel strategy to inhibit cancer metastasis

Funding: Polish Strategical Framework Program STRATEGMED
Partners:

Jagiellonian University in Kraków – JCET (project leader)
Medical University of Łódź;
Medical University of Gdańsk;
Medical University of Białystok;
Institute of Immunology and Experimental Therapy of Polish Academy of Sciences in Wrocław
Latvian Institute of Organic Synthesis

Our own preliminary studies indicate that endothelium-targeted pharmacotherapy bears potential to inhibit cancer metastasis. Aims of this project are:

  1. to characterize the endothelium related pharmacotherapeutic mechanisms that throught its vasoprotective, anti–platelet action may provide the novel strategy for prevention of cancer metastasis;
  2. to develop a novel rationally-designed, endothelium-targeted compounds effective in preventing of endothelial dysfunction, displaying anti–platelet, anti-thrombotic activity and thus inhibiting cancer metastasis, and finally
  3. to develop the novel panel of biomarkers of endothelial dysfunction, platelet activation and cancer metastasis, and its application to assess the efficacy of endothelium/platelet targeted pharmacotherapy in prevention of cancer metastasis in mice as well as its validation in humans to assess its usefulness as markers of metastasis.

This project will develop endothelium-targeted innovative pharmacotherapy for cancer metastasis. We will provide extensive preclinical dossier for novel molecules with endothelium-based anti-metastatic properties and prove their mechanisms of action, efficacy, profile of pharmacokinetics and safety.

The consortium of 6 partners (5 from Poland and one foreign), represents dynamic and interdisciplinary group with complementary research expertise.


Studies on antiplatelet and antithrombotic activity of CO-relasing molecules (CO-RM)

Funding: National Research Centre (funding agency), Program HARMONIA
Partner: Dr Roberto Motterlini (University Paris-Est, INSERM U955, Faculty of Medicine)

Purpose of research / hypothesis Carbon monoxide (CO) has significant vasodilator, antiplatelet and antithrombotic action, but the mechanism underlying the pharmacological activity of CO still remains unexplored. Until now it was impossible to separate the antiplatelet and antithrombotic effects of CO with vasodilator and toxic effects of CO. Our previous studies conducted in collaboration with Dr. Motterlini team showed that the release kinetics of the CO donor has a significant impact on the anti-platelet and anti-thrombotic effects of CO. These results constitute the background for the current collaborative project.

Scientific objectives of the project is to investigate the kinetics of release of CO from the CO releasing molecules in terms of its regulation by the oxidative stress, to characterize the effects of novel CO-RMs on platelets and endothelial cells in normoxia and hypoxia with particular emphasis on the mitochondria mechanisms and the role of guanylate cyclase in the pharmacological activity of CO. Furthermore, our aim is to examine the antithrombotic properties of CO-RMs, in the classical model of arterial thrombosis in the rat and in the state-of the art thrombosis model with confocal imaging in mice.

These studies should help not only to better understand the antiplatelet effect of carbon monoxide, but also may allow to point out a possible molecular target of CO in platelets.


logo Diagnostics of the biochemical alterations in packed human red blood cells (PRBCs) with the use of vibrational spectroscopy

Funding Agency: National Centre for Research and Development “Lider VIII”
Project leader: Katarzyna Marzec, Ph.D.

LIDER2A large number of patients worldwide are in need of periodic, therapeutic transfusions of packed red blood cells (PRBCs). This has led to an increased demand for an efficient technology providing information on the PRBCs alterations, which may affect their quality. The goal of this project is the development of the spectroscopic methodology for the Raman (RS) and Infrared (IR) diagnosis of the pre-transfusion quality of RBCs. In this project we are planning to study PRBCs fractions with the focus on biochemical changes of RBCs due to donor characteristics, age and characteristics of PRBCs, level of oxidative stress and damage in RBCs (steps I and II). RS and IR spectroscopies will provide fast determination of the RBCs condition before transfusions. To understand the biochemistry behind all changes observed by IR and RS, the RBCs samples will be studied also by other classical techniques. We can include here absorption spectrophotometry, blood gas analysis, flow cytometry, atomic force microcopy, ektocytometry, staining’s and the measurement of the chosen biochemical parameters. The focus on comprehensive assessment of PRBCs will provide the knowledge about biochemical fingerprints of RBCs condition. The last step of project (step III), which goal is an evaluation of the obtained SPECDIAG-RED methodology, will be focused on the studies of previously chosen spectroscopic biomarkers of biochemical state of PRBCs for a specific group of patient (patients with acute myeloid or/and acute lymphoblastic leukemia). The project will not affect the medical procedures performed routinely in hospital as the IR/RS measurement will be carry out on the small sample of PRBCs (1 mL) or by the direct measurement through the PRBCs wall. The PRBCs state will be assessed with the use of SPEC-DIAG-RED methodology and combine with the feedback from the clinics about patients condition before and after transfusion what will allow for the final evaluation of results.

See more: Red Blood Cells Spectroscopy Research Team


Vascular endothelium in life-style diseases: from basic research to the offer of an innovative endothelium-targeted therapeutic

Funding: European Regional Development Fund, Operational Program – Innovative Economy, Action 1.1.2
Partners:

Jagiellonian University in Kraków – JCET (project leader) 7 research teams from the following faculties: Faculty of Medicine; Faculty of Pharmacy; Faculty of Chemistry; Faculty of Biochemistry, Biophysics and Biotechnology; Faculty of Physics, Astronomy and Applied Computer Science;
Lodz University of Technology;
Nuclear Physics Institute of Polish Academy of Sciences in Kraków;
University of Agriculture in Kraków;
University School of Physical Education in Kraków;
Medical University of Łódź;
Nencki Institute of Experimental Biology of Polish Academy of Sciences in Warsaw;
Medical University of Gdańsk;
Medical University of Białystok;
Ludwik Hirszfeld Institute of Immunology and Experimental Therapy of Polish Academy of Sciences in Wrocław

Project period: 01.03.2009—30.06.2015
Project Budget: 63,5 mln PLN

The aim of the project is to carry out interdisciplinary research and development work on the vascular endothelium and its role in the development of lifestyle diseases, and, in particularly, development of original interdisciplinary research on the role of endothelium in the development of lifestyle diseases, with a particular interest in the new area of this knowledge; Development of interdisciplinary studies on synthesis,mechanisms of action and therapeutic and pharmacokinetic properties of the original chemical compounds (natural and synthetic) affecting the endothelium, which may be the basis for new therapies of lifestyle diseases; Tightening the collaboration between research units and the industry in the field of endothelial studies. The strategic aim of the project is the development of novel research strategies in the field of assessment endothelial dysfunction, as well as development of original, interdisciplinary studies on endothelium-targeted therapeutics including:

  • a development of a unique specialized technology of a comprehension assessment of endothelial dysfunction and panel of assays for detection of endothelial activity of chemical compounds;
  • an offer of innovative endothelium-targeted therapeutic.

Project fits into the main aim of the Operative Program – Innovative Economy, which is undertaking the actions assuring the increase in the competitiveness of Polish science and in a further perspective also the increase of the innovation of the industrial companies.


Development of a combination of natural compounds comprehensively improving endothelial function

Funding: The National Centre for Research and Development
Partners:

Endo Medical Sp. z o.o.  (project leader)
Jagiellonian University, Jagiellonian Centre for Experimental Therapeutics (JCET)
Poznan University of Medical Sciences 

Endothelial dysfunction associated with reduced synthesis and bioavailability of nitric oxide (NO) is the main cause ofthe development of hypertension, cardiovascular diseases, type 2 diabetes, dyslipidemia, overweight and obesity,known also as the metabolic syndrome. The analysis of mechanisms of development of endothelial dysfunction andimpaired NO bioavailability revealed that the administration of a combination of nitrates or L-arginine and antioxidantseliminate these phenomena. The aim of the project is to develop a combined preparation of the above ingredients withverified results in improving the functioning of the vascular endothelium and nitric oxide synthesis. Developedpreparation will be the basis for the subsequent development of products (currently not available on the market) likefood supplements and/or medical food with indications for use in people with hypertension, prediabetes or at risk ofdevelopment of metabolic syndrome.


Pharmacotherapeutic mechanisms, spectroscopic signatures, and nanomechanics of dysfunctional endothelium in liver steatosis and heart failure; towards organ-specyfic mechanisms

Funding: National Research Centre (funding agency), Program SYMFONIA
Parties:

Prof. Stefan Chłopicki (JCET, UJ)
Prof. Malgorzata Baranska (JCET, UJ)
Prof. Marek Szymoński (Faculty of Physics, Astronomy and Applied Computer Science, UJ)

Impairment of endothelial function is a primary cause or a result of many human diseases. Although the mechanisms causing the primary endothelial dysfunction are known (i.e. those leading to the development and progression of atherosclerotic plaque), still there are many diseases in which the secondary endothelial dysfunction occurs, but underlying mechanisms remains unknown. Particularly a dysfunction of liver sinusoidal endothelial cells (LSEC) mediated by alterations in hepatocyte-derived paracrine signals is essential for the development of liver steatosis, while dysfunction of cardiac microvascular endothelial cell (CMEC) mediated by changes in cardiomyocyte–derived paracrine signals is an important determinant of the development of heart failure. We hypothesize, that secondary endothelial dysfunction that develops in liver steatosis, is mediated by a decrease in the activity of hepatocyte-derived VEGF signaling that may be associated with increased sFLT activity, and results in impaired LCES-derived NO production. In turn, we claim that in the heart failure alterations in cardiomyocyte-derived PDGF signaling and excessive activity of myocardial angiotensin II-aldosterone pathway results in the impairment of NO production in CMEC. The aim of the project is to understand the signals that governs intracellular communication between hepatocytes and LSECs and between cardiomyocytes and CMECs, since these microvascular bed-specific mechanisms determine the phenotype of the endothelium in an organ-specific way and may constitute the pathophysiological basis for the hepato-selective and cardio-selective therapy of endothelial dysfunction.

In the presented project we aim to examine the progression of changes of various aspects of endothelial phenotype (structure, function, elasticity, glycocalix, biochemical profile, vasoprotective, pro-thrombotic and pro-inflammatory mechanisms) in LSECs in the course of the development of liver steatosis in mice (induced with high-fat diet), and in CMECs in the course of the development of heart failure in Tgg*44 mice in both cases in the relation to the comprehensive description of the development of these pathologies. In particular, we aim to investigate the role of intracellular signaling homeostasis depending on a) VEGFNO in the liver, and b) PDGF/Aldosterone –NO in the myocardium in the development of the secondary endothelial dysfunction in the above mentioned diseases.


The role of nicotinamide N-methyltransferase (NNMT) and mitochondrial mechanisms in endothelial dysfunction

Funding Agency: National Science Centre, Poland, Program: OPUS
Project leader: Lider: IBD PAN, Prof. Stefan Chłopicki (JCET, UJ) (one part)

An impairment of the vascular endothelium plays a role in most, if not all, of human diseases, either as a primary cause or as a result of organ damage. Endothelial dysfunction in diabetes leads to macro-and microangiopathies of the cardiovascular system leading to myocardial infarction, nephropathy, or diabetic retinopathy. Despite intensive research biochemical and molecular mechanisms underlying endothelial dysfunction are not fully explained. One of the factors affecting normal endothelial functions can be nicotinamide N-methyltransferase (NNMT) and 1-methylonicotinamide (MNA), which is the product of nicotinamide methylation catalyzed by this enzyme. Exogenously administered MNA exhibits anticoagulant, anti-inflammatory and vascular protecting properties. However, a function of endogenous MNA synthetized in endothelial cells has remained unknown, so far. NNMT utilizes, nicotinamide (NA), which also is a precursor of NAD +, thus this enzyme affects cellular pool of nicotinamideadenine dinucleotides, and therefore may indirectly influence mitochondrial energy metabolism and other NAD+-dependent cellular processes e.g sirtuin-cartalyzed reaction. We hypothesize that increased activity of NNMT in endothelial cells under stress conditions have cytoprotective effect due to stabilization (stimulation) of specific sirtuin activity. The aim of this project is to explore the role of NNMT in response of endothelial cells to different types (models) of cellular stress, including palmitate induced insulin resistance, TNFα – induced inflammation, exposition to genotoxic factors and under metabolic and oxidative stress conditions.


Jagiellonian Centre for Experimental Therapeutics – JCET

Funding: European Regional Development Fund, Operational Program – Innovative Economy, Action 2.2
Partners:

Jagiellonian University in Kraków – JCET (project leader)
Lodz University of Technology
Henryk Niewodniczański Nuclear Physics Institute of Polish Academy of Sciences in Kraków

Project period: 02.01.2008—30.09.2012
Project Budget:  48 mln PLN

The main aim of the project is to create a modern research infrastructure by creating laboratories of the Jagiellonian Centre for Experimental Therapeutics (JCET) at the Jagiellonian University and equipping and modernization of the rooms of the Laboratory of Chemical Synthesis of the Lodz University of Technology as well as purchase of MRI for in vivo animal imaging and modernization of the rooms necessary for the equipment installation in the In vivo Imaging Laboratory of the Nuclear Physics Institute of Polish Academy of Sciences in Kraków.

Jagiellonian University has bought, within the project, research equipment worth approximately 22 million PLN; moreover JCET has allocated approximately 5.5 mln PLN for JCET laboratory adaptation. Lodz University of Technology acquired research equipment worth 3 mln PLN and already adapted the starting the Laboratory of Chemical Synthesis in the Institute of Radiation Technology of the Faculty o Chemistry. Nuclear Physics Institute of Polish Academy of Sciences in Kraków has bought the MRI equipment for almost 10 mln PLN and adapted rooms for In vivo Imaging Laboratory in the Department of Magnetic Resonance Imaging, necessary for setting-up the modern MRI equipment.

The purchase of research-scientific equipment made within the project executed by the consortium of units under the supervision of the Jagiellonian Centre for Experimental Therapeutics (JCET) was funded by structural fund of the European Union within European Regional Development Fund from the Operative Program – Innovative Economy.We invite research groups from national and international research units and companies interested in JCET offer to collaborate and take the opportunities to use available equipment and unique expertise of JCET research teams.


The role of nicotinamide N-methyltransferase and mitochondrial activity during endothelial dysfunction

Funding: National Research Centre (funding agency), Program OPUS
Project leader: Prof. Krzysztof Olaf Zabłocki (Nencki Institute of Experimental Biology Polish Academy of Sciences)
Copartner: Prof. Stefan Chłopicki (JCET, UJ)

mong many possible factors maintaining the healthy endothelial phenotype, the activity of nicotinamide N-methyltransferase (NNMT) still needs further examination. NNMT catalyses nicotinamide (NA) methylation, releasing 1-methylnicotinamide (MNA), which anti-inflammatory, anti-thrombotic and vasoprotective action has been reported in many animal models. Although beneficial effect of MNA treatment has been demonstrated in several studies, it is still little known about the role of endogenous MNA in dysfunctional endothelium, where NA methylation by NNMT seems to be linked with cellular bioenergetics. In our preliminary studies we demonstrated the upregulation of NNMT in endothelial cells stimulated with TNFα. According to the hypothesis, activation of endothelial NNMT stabilizes NAD- dependent sirtuins, histone deacetylases involved in genome expression and stability. The main aim of this project is to examine the role of NNMT and endogenous MNA during endothelial insulin resistance and endothelial dysfunction, triggered by different stressors: oxidative, metabolic, hypoxic and genotoxic. This project could provide necessary data to verify the hypothesis about vasoprotective, MNA-dependent effect of NNMT on mitochondrial sirtuin 3 and nuclear sirtuin 1 regulation, which is probably linked to biogenesis of mitochondria. The second aim of the project is to examine the cross-talk between NNMT activity, mitochondrial metabolism and endoplasmic reticulum, as well as potential MNA protective effect on above-mentioned organelles.


In search for endothelial mechanisms of TPO-induced neuroprotection

Funding: European Commission, Research Executive Agency, HORIZON2020, TONEURENDO
Project leader: Mateusz Adamski, PhD

Ageing is recognized as one of the greatest social and economical challenges of the 21century for European societies. With,,Vascular Cognitive Impairment (VCI) being one of the leading causes of age-related cognitive impairment and one of the major causes of disability in the elderly. Although, the concept of VCI was introduced in 1993, current treatment is limited to management of vascular risks and symptomatic  harmacotherapy targeting vascular dementia.

The overall objective of this project is to fill the significant gap in early detection, prevention and treatment of VCI. This will be achieved by explaining microvascular mechanism of protective effects of  hrombopoietin (TPO) in a novel unique mixed-risk animal model of VCI- specific to hypertension plus carotid-artery hypoperfusion (HH-VCI).

Given increased number of progenitor endothelial cells after TPO treatment, it is hypothesizes that protective effect of TPO is mediated by endothelium. Furthermore, protective effect of TPO is expected to be caused by activation of neoangiogenesis, anti-inflammatory and vasoprotective mechanisms driven by TPO action on endothelial. This hypothesis will be tested in two stages first, in-vitro and second, in-vivo. In-vitro models, will be used to investigate endothelial response to TPO in terms of its modulation of inflammatory response angiogenic potential and vasoprotective mechanisms. In-vivo models of TPOR KO mice and HH-VCI mice will be used to validate and confirm mechanisms identified in in vitro stage of experiments.


Induced chirality in “guest-host” supramolecular assemblies – developement of new methods of resonance Raman Optical Activity

Funding Agency: National Science Centre, Poland, Program: OPUS 13
Project leader: Agnieszka Kaczor, PhD, D.Sc. (JCET, UJ)

The project is devoted to studies of chirality induction in supramolecular ”host-guest” aggregates using chiroptical methods, in particular Raman Optical Activity (ROA) and Vibrational Circular Dichroism (VCD). The aim of the project is to synthesize and characterize supramolecular ”host-guest” systems of different structure and applications, but designed in such a way that they exhibit resonance ROA and enable induction of chiral information in these systems. Analysis of “guest-host” interactions and conformational effects in generated aggregates will be undertaken in order to propose optimal ”guest” and ”host” molecules, designed to create universal and efficient systems for inducing chirality, and thus the possibility of indirect detection of achiral molecules. Development of methods of resonantly enhanced Raman Optical Activity is a side, nonetheless relevant effect of the study. Analysis of mechanisms of chirality induction can become crucial in designing chiral sensors and understanding of chiral recognition processes. Thus, the submitted project brings significant cognitive potential and can provide a conceptual basis for application of supramolecular entities in controlled transfer of chiral information.


To catch lifestyle diseases red-handed – in vivo Raman spectroscopy

Funding Agency: National Science Centre, Poland, Program: OPUS 9
Project leader: Agnieszka Kaczor, PhD, D.Sc. (JCET, UJ)
Participants: Marta Pacia, Krzysztof Czamara, Ewelina Bik, Zuzanna Majka

Raman spectroscopy is successfully used to study biochemical changes in tissues and cells in in vitro and ex vivo models. These models, although undoubtedly informative, are based on several assumptions, for instance negligibility of the lack of the “tissue environment” (in case of in vitro cellular models), negligible influence of fixation procedures, transferability of conclusions based on studying animals on humans or various other limitations of the model systems that are often highly homogenous, gene knock-out or simplified, one-factor activated models. All these limitations can be eliminated with the application of the of fiber optic probes and in vivo approach, in which the tissue of interest is studied in the living organism. According to my best knowledge, in vivo Raman spectroscopy is not developed in Poland, although it is the method tested since a decade or two in studies of various pathologies. In the light of growing risk of various diseases of affluence, application of in vivo Raman spectroscopy to study these pathologies is an important factor that may help to contribute to their prevention. Therefore, the objective of this project is the application of the Raman fiber optic probe system to investigate and characterize biochemical changes in the organisms in vivo (in real physiological environment) upon development and progression of chosen lifestyle diseases.


Development of EPR oximetry methodology for evaluation of endothelial action of drugs

Funding: National Research Centre (funding agency), Program SONATA 3
Project leader: Patrycja Kaczara, PhD

The project is planned for original studies of perivascular adipose tissue The main goal of the project was to develop a methodology for measurements of endothelial cells respiration. It was necessary for better understanding of mechanisms, which underlie oxygen consumption and production of intracellular energy versus generation of reactive oxygen species, which can participate in variety of signaling processes, beneficial or detrimental for the cell. Endothelial cells are postulated to cover majority of energy demands not by oxidative phosphorylation in electron transport chain, but by aerobic glycolysis. These bioenergetics processes may be helpful in evaluation of endothelial cell activity in physiological and pathological conditions under treatment with newly developed pharmacological compounds.


In search of biochemical, mechanical and functional fingerprints of oxidative stress in RBCs

Funding Agency: National Science Centre, Poland, Program: OPUS
Project leader: Katarzyna Marzec, Ph.D.

The reactive oxygen species (ROS), reactive nitrogen species (RNS) as well as other free radical species may have damaging impact on our bodies on the cellular level, however their production is also an integral process necessary for many metabolic reactions. Blood play a special role in oxidative stress mechanism, as it is not only a connective tissue which supply oxygen and nutrients to tissues but also removes many wastes including oxidative species. Therefore all blood cells are exposed during oxidative stress to oxidative environments rich in ROS, NOS and free radical species. The foundation of the project is to investigate RBCs alternations. This is untypical approach, as previous studies were mostly focused on other blood fractions, mainly white blood cells. We believe, that biological understanding of biochemical changes due to oxidative stress in situ in isolated RBCs as well as RBCs in whole blood could expand or knowledge about oxidative stress action. RBCs exhibit oxidative damage through characteristic changes in the biochemical content of the hemoporphirins and cell membrane, the size and shape of cell morphology as well as mechanical properties such as for example stiffness and functional parameters. All those changes have impact or are connected with changes in other blood fractions and general blood properties like increased
blood viscosity and impaired flow.

In this project we focus on comprehensive assessment of oxidative stress in red blood cells (RBCs) with the use of innovative technology. The use of combination of Raman spectroscopy (RS), Fourier transform infrared spectroscopy (FT-IR) and atomic force microscopy (AFM) supported with referenced techniques (UV-Vis, biochemical parameters, stainings, blood analysis and ektocytometry) will provide the knowledge about biochemical, mechanical and functional fingerprints of level of oxidative stress in RBCs. The recognition of such fingerprints due to oxidative stress in situ in RBCs can be a biomarker of general oxidative stress status. Project will allow to create the innovative methodology for sensitive and fast diagnosis of the oxidative stress level in RBCs which will be tested to be apply to in vitro/ex vivo description of oxidative stress level in RBCs obtained from mice models of different diseases of affluence. Such diseases as atherosclerosis, or diabetes share etiologic pathways wherein oxidative stress plays the contributing role.

To the main aims of the project we may include:

  1. In situ characterization of oxidative stress chemically induced in isolated RBCs from healthy mice with the focus on haemoglobin structure; alternations of the lipids and proteins inside membranes of RBCs; mechanical properties of RBCs such as stiffness, topography, adhesion; functional properties such as deformability and aggregation; biochemical content of the macrovesicles produced by RBCs in oxidative stress conditions.
  2. Definition and description of the possible biochemical, mechanical and functional fingerprints of oxidative stress in RBCs will be carry out depending on type of chemically induce oxidative damage/pathology, type of RBCs and comparison between results obtained for isolated RBCs versus whole blood.
  3. Selection and testing of chosen fingerprints obtained with the use of IR or Raman or AFM for in vitro/ex vivo detection of the oxidative stress level in RBCs obtained from the different mice models of human disease of affluence which share etiologic pathways wherein oxidative stress plays the contributing role.

We believe that the planed in our studies use of Raman, IR and AFM techniques allows not only for detection of oxidative stress level of RBCs but maybe also a potential new set of techniques for detection of some RBCs alternations in the first place. Raman technique uses the advantages of immersion confocal spectroscopy as well as provides information about the biochemical changes of the sample on the molecular level. IR has additional advantage of not only detection but also easy quantification of many biochemical changes. It was already shown that both, IR and RS have huge diagnostic potential also in RBCs studies. If specific molecular change of RBCs has specific marker bands the diagnosis could take even up to several second what gives additional possibility of the fast in vitro/ex vivo determination of the oxidative stress in RBCs. AFM can provide a mechanical fingerprint of a single-cell pathological changes also for RBCs what makes this a promising technique in cell alternation diagnosis. Moreover, those techniques allow for an analysis of the molecular and mechanical changes in a single RBC in small quantity of blood without previous staining or fixation. Obtained data (the set of Raman/IR/AFM results) which keep the molecular information, can be easily stored for future analysis.

It is expected that the new methodology used in this project will be developed for characterization of the oxidative stress level in RBCs. Results of the project will give physical output such as formation of unique research group which uses new applications of imaging biospectroscopy and AFM to new concept of RBCs investigation. Moreover, results will be published in international journals with a high Impact Factor, present on international and Polish conferences. The project contribution to a higher level goal is creation of the methodology of RS/IR/AFM diagnosis of the oxidation damage in RBCs which can be apply in studies on animal models of different human diseases of affluence.

See more: Red Blood Cells Spectroscopy Research Team


Structural studies and chemical imaging of vitamins A, E and their metabolites in healthy and pathologically altered animal tissue

Funding: National Research Centre (funding agency), Program SONATA 4
Project leader: Katarzyna M. Marzec, PhD

The project was focused on finding the spectroscopic differences (fingerprints, connected with different molecular structures) between the title vitamins A, E and their metabolites. The structural studies were carried out on the basis of various spectroscopic techniques (infrared (IR) and Raman (RS) spectroscopy) as well as theoretical vibrational analysis based on DFT calculations. This allowed first recognizing and imaging the compounds in situ in the biological samples and second, location and study of the distribution of the title compounds in situ inside tissues of healthy and altered mice obtained from different organs such as liver, lungs, aorta, kidneys, brain and sight organs. The results were published in six manuscripts and presented in the form of nine poster presentations.


Design of SERS nanosensors for ex vivo detection of inflammatory state in blood vessels

Funding Agency: National Science Centre, Poland, Program: OPUS 11
Project leader: Kamilla Malek, PhD, D.Sc. (JCET, UJ)

Optical chemical- and bio-sensing is one of the most promising directions of diagnostic sensors development nowadays, offering advantages of high sensitivity, combined with non-invasive nature of analysis, portable devices suitable for in-point care analysis. The principle aim of the current project is to develop novel sensitive detection technologies combining a highly selective immunoassay approach with unique spectral signatures of Surface Enhanced Raman Spectroscopy (SERS) for detection of markers of inflammatory state. SERS is a highly sensitive Raman detection technique based on gold nanostructured substrates fabricated in chemical reaction (nanoparticles like nanostars, nanoshells) and in processes like anodization and photoreduction giving periodic in shape nano-sized structures. SERS has far-reaching applications in fields as diverse as biomedical diagnostics and biochemical studies and shows great potential for multiplex analysis, making it particularly suitable for the simultaneous diagnosis of multiple disease markers.

The general objectives of the project is to design two types of biosensing assays to detect ex vivo inflammatory vascular disease in tissue cross-section and blood plasma. One of them will provide multiplex staining combined with imaging of few antigens in arteries in atherosclerotic plaque while the second SERS biosensor will be a sandwich-type bioassay for quantitative detection of pro-inflammatory cytokines in blood plasma. Effectiveness and function of designed sensors will be validated in murine models expressing vascular inflammation (lipopolysaccharide-induced endotexia – LPS model and atherosclerosis – model ApoE/LDLR-/-) which perfectly reproduce human diseases. Their choice is dictated by the fact that elevated levels of several inflammatory mediators among apparently healthy men and women have proven to have predictive value for future vascular events. Actually, inflammation is a beginning stage of almost all diseases, including heart attack, stroke, cardiovascular complications of diabetes, cance, etc. The planned research are multidisciplinary linking of nanotechnology, extremely sensitive spectroscopic technique and medical technology supported by three groups of specialists in the field of the project.

The motivation of this research is a continuing need to develop advantages of SERS biosensing and solve pitfalls limiting translation of this diagnostic technology into clinics. The main beneficiaries of the project results will be nanotechnologists, spectroscopists and clinical scientists who will receive a potential novel tool and deeper insight into designing new ones for  nvestigation of biological systems of clinical significance. It seems, therefore, that the project is innovative because of the technology used and the object of study. Due to the fact that this kind of research is pioneering in Poland it should be highlighted that the project will have an impact on our country’s growing importance in world-wide biomedical research.


Low Dose Research towards Multidisciplinary Integration (DoReMi)

Funding: European Commission, Research Executive Agency, 7PR
Project leader: Prof. Stefan Chłopicki (JCET, UJ)

Coronary artery disease and atherothrombosis are preceded by and associated with the development of endothelial dysfunction that promotes vascular inflammation and thrombosis. It has been demonstrated that
exposures at low doses of low-LET radiation during various stages of atherosclerosis protect against the progression of the disease in genetically-modified mice. Recently, it has been demonstrated that exposures to low-level low-LET ionizing radiation attenuate the development and progression of various diabetic complications such as nephropathy, vascular and cardiac inflammation, inhibited proliferation of neurons, and impaired wound healing. Since, in contrast to high-dose exposures, irradiations at low doses of Xor γ-rays have been shown to stimulate anti-oxidant functions in various cells and tissues, it can be hypothesized that such beneficial effects of the low-level exposures result from the radiation-induced suppression of the oxidative damage to the endothelial lining of micro- and macrovasculature. Thus far, no direct evidence has been provided on the effects of low-level exposures to ionizing radiation on the development or reversal of the impaired function of endothelium in diabetes.
We presume that endothelial effects of low doses of low-LET radiation will afford protection against atherothrombotic complications of diabetes. The scientific objectives of the proposal include evaluation of the effects of low doses of X-rays on the structure and function of vascular endothelium in mice before and during the development of hyperglycaemia – a cause of the vascular complications in diabetes.


Towards experimental pharmacology of endothelium in vivo based on multiparameter method of assessment of the vascular endothelial function in mice with the use of MR imaging techniques

Funding: National Research Centre (funding agency), Program PRELUDIUM 12
Project leader: Anna Bar, PhD

Endothelial dysfunction accompanies development of many disease, not only diseases of the cardiovascular system and has pathophysiological, prognostic and therapeutic significance. Therefore, development of fast and reliable methods of assessment of endothelial phenotype in vivo, in mice models, constituting a basic model in preclinical studies, is essential for the better understanding research development on of the role of endothelial cells in the progression of disease and on the efficacy of novel research on the pharmacotherapeutic mechanisms in the treatment of endothelial dysfunction in vivo.

The aim of this study is: firstly, to develop and validate a sensitive method for endothelial imaging with using magnetic-resonance techniques (MR) in vivo, allowing for an effective assessment of impaired, endothelial-dependent vasodilatation response and permeability of endothelium, which are the main features of endothelial dysfunction; and secondly, to use the developed methodology to assess the effects of various endothelial pharmacotherapies in murine model of atherosclerosis (ApoE/LDLR-/- mice).

The novel methodology developed in this project, will allow for a comprehensive assessment of the main features of endothelial dysfunction in murine models in vivo, including evaluation of: a/ endothelial-dependent response to Ach in various vessels available for MR imaging, b/ FMD in mice, contributing to the development of preclinical studies of pharmacotherapeutic mechanism of endothelium, based on the technique used in humans, opening the way for translational research, c/ changes in endothelial permeability using a unique formulation of gadolinium contained in a liposome, which is a significant addition to assessment of the vasodilatation response of endothelium. Performed assessment of endothelial dysfunction progression in ApoE/LDLR-/- mice, and the effect of ACE-I and PPI and vitamin K2 on endothelial function in vivo, in comparison to the EPR measurements, which are the most sensitive method for disorders of NO production detection in a blood vessel ex vivo, altogether will open up a new perspectives for research on pharmacotherapy of endothelium and on profiling of drugs effects on endothelium in murine models in vivo.

To sum up, on the one hand the objective of this project is to develop and validate new and a unique methodology for measuring the phenotype of endothelial cells in vivo using MR imaging and on the other hand to characterize mechanisms of the vitamin K2 effects on the endothelium function in murine models of atherosclerosis in vivo.


Description of platelets role in metastasis and evaluation of anti-platelet treatment efficacy in mice model of metastatic breast cancer

Funding Agency: National Science Centre, Poland, Program: PRELUDIUM
Project leader: Elżbieta Buczek, M.Sc.,  Ph.D. student

The main purpose of the project is to investigate the role of platelets in the development of metastases in the lungs in experimental model of murine 4T1 mammary adenocarcinoma. This project allows for a comprehensive assessment of changes in platelet activation and reactivity in mice at different stages of metastases development using three complementary methods:

  • a flow cytometry to examine platelet activation markers
  • evaluation of thromboxane B2 generation in whole blood ex vivo
  • assessment of the platelet adhesion and aggregation.

It is also relevant to prove that 4T1 breast cancer is a proper model to test antimetastatic potential of new substances. It is planned to assess the efficacy of antiplatelet therapy with different mechanisms of action for inhibition of the development of primary tumor and metastasis in this model.


Aptamer-based biosensor for simultaneous detection of thrombin and von Willebrand factor in plasma

Funding: National Research Centre (funding agency), Program PRELUDIUM 11
Project leader: Katarzyna Derszniak, PhD Student

The aim of the project is to develop an aptamer-based biosensor, with aptamers as molecules for capturing ligands, for the simultaneous detection of von Willebrand factor and thrombin in biological samples, including plasma. It turns out, that the detection of disorders of the coagulation system may help to predict the development of both atherosclerosis and cancer diseases. Thrombin, as a key component of the coagulation system, favors the development of atherosclerotic plaque and has influence on rising risk of thrombo-embolic incidents which are complications of atherosclerosis. It can also stimulate the proliferation of vascular endothelial cells and tumor cells. What is more, thrombin also promotes intravascular clotting and tumor-cell – platelet interactions. Moreover, it can cause that the endothelial cells are more sensitive to the action of Vascular Endothelial Growth Factor (VEGF). It leads with increasing of endothelial cells mitosis and cell migration. The occurrence of Venous Thromboembolism (VTE) among cancer patients is further evidence of the correlations between dysfunction of coagulation system and cancer diseases. One of the identified risk factors for VTE among cancer patients is hypercoagulable state, which results from increased thrombin generation. The von Willebrand factor is an another important component of the coagulation system which is released by activated endothelial cells, promoting the development of thrombosis and tumor metastasis. Therefore, developing a method to study the condition of the coagulation system is important to assess the state of cancer development and cardiovascular pathologies.


Aptamers in the study of thrombotic mechanisms dependent on platelets and erythrocytes functions by microfluidic devices

Funding Agency: National Science Centre, Poland, Program: ETIUDA 8
Project leader: Katarzyna Derszniak, PhD Student

The mechanism of erythrocytes and platelets adhesion to the vessel wall will be studied by the application of aptamers and the microfluidic vascularized 3D platform formed by selfassembling cells and developed by the team of Professor Christopher C. W. Hughes (University of California, Irvine, USA).


Resonance Raman spectroscopy studies of the in vitro and ex vivo haemoglobin adducts with gasotransmitters in biological systems

Funding: National Research Centre (funding agency), Program PRELUDIUM 12
Project leader: Jakub Dybaś, PhD

The aim of the project is to study an influence of endothelial gaseous mediators, nitric oxide (NO), carbon monoxide (CO) and hydrogen sulfide (H2S) on porphyrins in various human- and murine-derived biological samples. Especially, the interaction between the iron ion in hemoglobin of red blood cells and NO, CO and H2S will be reevaluated, as well as with detection of the formed Hb-X adducts, which could have a potential importance in these gasotransmitters bioavailability. The principal assumption of the project is to carry out experiments ex vivo (whole human blood and isolated human red blood cells, murine isolated Kupffer cells) and in vitro (murine macrophage cell line) on biological samples instead on standard compounds. Due to the used methods (resonance Raman spectroscopy and spectrophotometry UV-Vis), it will be possible to deeply inquire into the changes both on molecular level and on quite bigger, macroscopic scale of studied systems. All the measurements will be done in pH and temperature controlled environment to the best reproduction of physiological conditions.

Obtained results should shed additional light towards better understanding the role, which formed adducts plays in the context of NO, CO and H2S bioavailability.


Molecular spectroscopy studies on chosen hemoprotein adducts and their changes in biological systems

Funding: National Research Centre (funding agency), Program ETIUDA 5
Project leader: Jakub Dybaś, PhD

As a part of this project, Jakub spent 6 months in Saint Louis University (SLU), where his main aims were to enhance knowledge about resonance Raman (rR) application to heme studies and to spectroscopically characterize cytochrome c (Cc) with the special attention on rR spectral response to changes in oxidation and spin states marker band positions as well as detection and differentiation of the modes arising from the peripheral substituents with the use of various excitation wavelengths.During this internship, Jakub performed also rR measurements of myoglobin, hemoglobin andcytochromes c adducts with isotopically labeled O2, CO, NO and CN– ligands to deeply inquire their molecular structure and biophysical properties. He was trained in use of highly specialized Raman instrumentation containing a high resolution 1250 M spectrometer (Horiba Scientific) equipped with a Princeton Instruments liquid nitrogen cooled array detector (PYLON CCD, Princeton Instruments), a Coherent Innova 300C Kr+ ion laser and a KimmonHe:Cd laser, as well as all necessary optics, including holographic Notch filters covering many laser lines in the range of 350 to 440 nm.


Bioanalitycal and pharmacological studies on the compensatory mechanism dependent on MNA/COX-2/PGI2 pathway in NO-deficient mice

Funding Agency: National Science Centre, Poland, Program: PRELUDIUM 9
Project leader: Agnieszka Kij, M.Sc. 

The goal of this project is to assess the compensatory role of MNA/COX-2/PGI2 pathway in the development of endothelial dysfunction associated with the lack of NO bioavailability as well as the evaluation of pharmacological effects of upregulation of this pathway on platelets activity in NO-deficient mice.

We know that, exogenous MNA (1-methylnicotinamide) exerts anti-thrombotic and anti-inflammatory effect through activation of COX-2/PGI2 pathway. The concentration of MNA in inflammatory states such as atherosclerosis or hepatitis is elevated what indicates that this molecule can be responsible for the counteract of endothelial dysfunction associated with those pathologies. We hypothesize, that endogenous MNA is a regulator of COX-2/PGI2 pathway activity, thus activation of the compensatory effect in the lack of the endothelium-derived NO can be attributed to MNA/COX-2/PGI2 mechanism. This project aims to verify this hypothesis using NO-deficient mice with hypertension induced by L-NAME which is an inhibitor of all isoforms of NO synthases. To confirm our thesis, we will evaluate the influence of impaired NO bioactivity on the action of MNA/COX-2/PGI2 system by the quantification of MNA and its metabolites, evaluation of eicosanoids profile in urine and plasma specimens and also by platelets activity analysis. All measurements will be performed in different stages of hypertension before as well as after administration of MNA donor (MNACl) or MNA and NO source (MNANO2, MNANO3) to NO-deficient mice. The changes in platelets activity analysed by ex vivo dynamic TXB2 generation assay in whole blood combined with alternation in MNA and PGI2 synthesis in NO-deficient mice during progression of hypertension and after pharmacotherapy using MNA or MNA and NO donors will allow to assess the importance of MNA/COX-2/PGI2 pathway in the regulation of platelets activity in different stages of endothelium-derived NO bioavailability impairment. The project is also concentrated on the development of bioanalytical method for quantification of selected eicosanoids in mice urine and plasma applying LC/MS/MS technique as well as method validation. Moreover, based on the results we will attempt to evaluate the PK/PD model encompassing the integration of pharmacokinetics, biochemical and pharmacodynamics parameters which describe the biological effect of studied substances and antiplatelet activity of two considered pathways – NO and COX-2/PGI2.


Pharmacokinetic profiling to understand differences in biological activity of two novel liver-selective NO-releasing compounds V-PYRRO/NO and V-PROLI/NO in the treatment of experimental model of NAFLD in mice

Funding: National Research Centre (funding agency), Program PRELUDIUM 6
Project leader: Kamil Kuś, PhD

The main goal of the proposed project is to analyze in comprehensive way pharmacokinetic profile of two NO-releasing hepatoselective prodrugs: V-PYRRO/NO and V-PROLI/NO, that exert differential pharmacological effects in the murine model of NAFLD (Non-Alcoholic Fatty Liver Disease). In our preliminary experiments V-PYRRO/NO, but not V-PROLI/NO proved to be effective in inhibiting liver steatosis, improving insulin sensitivity and modifying hepatotoxic lipid profile in the liver. Based on the results of this studies, we presume that pharmacokinetic differences between these two agents are responsible for the differences in their pharmacological efficacy. Therefore in-depth examination of PK profile of studied compounds will help to understand the differences in their pharmacological activities. The specific objectives of the project are as follows: – (1) to optimize the methodology, including the development and validation of LC/MS/MS bioanalitical methods. – (2) to characterize pharmacokinetic profile of V-PYRRO/NO and V-PROLI/NO in vivo – (3) to study liver elimination process of V-PYRRO/NO and V-PROLI/NO ex vivo – (4) to study metabolism of V-PYRRO/NO and V-PROLI/NO using mouse liver microsomes and primary mouse liver hepatocytes in vitro – (5) to explain differences in V-PYRRO/NO and V-PROLI/NO pharmacological efficacy, based on pharmacokinetics studies planned in the current project.


Glycocalyx damage – the first stage of pathomechanism of endothelial dysfunction; measurements using capillary electrophoresis

Funding Agency: National Science Centre, Poland, Program: PRELUDIUM 16
Project leader: Karolina Matyjaszczyk-Gwarda, M.Sc.,  Ph.D.

An endothelial dysfunction is a crucial element of the pathophysiology of many different diseases, including atherosclerosis, diabetes and neurodegenerative diseases. The proper function of the vascular endothelium is closely associated with the correct function and structure of the glycocalyx (GLX) – a carbohydrate-rich ‘jelly-like’ layer covering the endothelium. There is more and more evidence that damage of GLX precedes the development of endothelial dysfunction, therefore the knowledge about changes in the quantitative and qualitative structure of GLX related to different pathological states may be extremely helpful in the understanding of the role of the particular glycosaminoglycans (GAG) in physiological processes. So far, published studies represent only a fraction of knowledge about quantitative and qualitative changes in GLX that occur during the development of endothelial dysfunction. The available analytical methods for GLX measurements have limitations: they can be used only for ex vivo material (immunohistochemistry, atomic force microscopy), they are based on the quantitative analysis of only one of the GLX components (enzyme immunoenzymatic methods, e.g. ELISA), or they are not sensitive enough.

To our knowledge, no studies have been carried out to explain how individual classes of glycosaminoglycans (GAG) – polysaccharides, which are one of the GLX building blocks, behave during subsequent stages of GLX damage. We suppose that the study of differences in the levels of GAG in plasma (heparan sulphate/ heparin, dermatan sulfate/chondroitin sulfate, keratan sulfate, hyaluronan) can reflect in more comprehensive way the complexity of the GLX damage process and consequently become a more sensitive biomarker of GLX degradation compared to currently used (ELISA, proteoglycan-based tests, or only one of GAG). The use of capillary electrophoresis (CE) in the proposed project is a unique approach that allows the simultaneous determination of several different GAG. By applying quantitative method, developed based on the special possibilities offered by CE, we can expect results that will answer the question of how changes in the entire GAG profile, look, during the subsequent stages of GLX damage leading to endothelial dysfunction. The developed methodology will be used to study GLX in an in vitro and in vivo model, in the acute GLX injury induced by LPS (endotoxaemia model).

The results obtained from in vitro experiments will be used to create a scheme of how GLX behaves under the influence of more or less specific enzymes and what changes in GAG levels are most characteristic of individual enzymes. This formula will serve as a basis for investigating whether specific stages can be expected in in vivo models correlated with changes representative of a particular GAG. The proposed project may become the first step towards the diagnosis of endothelial dysfunction at a very early stage based on the diagnosis of GLX injury. The use of CE, which is a high-throughput and relatively cheap technique, can introduce a new quality among the available methods and become a “gold standard” in the diagnosis of early vascular endothelial damage.


Raman spectroscopy in studies on cellular uptake of sinusoidal endothelial cells in vitros

Funding Agency: National Science Centre, Poland, Program: PRELUDIUM 14
Project leader: Ewelina Matuszyk (Szafraniec), M.Sc., Ph.D. student

The project is focused on application of Raman spectroscopy to study the process of cellular uptake (mainly by endocytosis) that takes place in Liver Sinusoidal Endothelial Cells (LSECs). The aim of the project is to develop an analytical method allowing to follow the uptake of molecules and to determine the kinetics of this process, taking an advantage of high chemical specificity and resolving power of Raman imaging technique. The outcomes will be referred to results achieved with the use of fluorescence microcopy, which will serve as a reference technique. The crucial stage of the project will be the validation of the developed method, which involves the inhibition of the cellular uptake by the use of appropriate inhibitors. This stage is intended to confirm the participation of known mechanisms mediating the uptake process. LSEC cells are characterized by high endocytic capacity allowing for blood purification from potentially dangerous molecules, while being a significant ‘player’ in maintaining liver homeostasis and immunity. Dysfunction of LSECs is a key event in multiple liver disorders, having a straight correlation with the loss of endocytic capacity.

The planned research will provide a new insight into the molecules uptake by LSECs. In addition, the use of Raman spectroscopy is an innovative approach in this field. This enhances the application potential of spectroscopic methods that offer research capabilities not available to conventional techniques used in subcellular investigations.


Raman spectroscopy in the study on non-alcoholic fatty liver disease using isolated liver cells and in vitro cell models

Funding Agency: National Science Centre, Poland, Program: ETIUDA
Project leader: Ewelina Matuszyk (Szafraniec), M.Sc., Ph.D. student

The 21st century is a period of increased incidence of related civilization diseaseswith an unhealthy, sedentary lifestyle. Many of these disorders are associated with endothelial dysfunction lining all blood vessels. This also applies to liver dysfunction such as non-alcoholic fatty liver disease (NAFLD). Despite the growing incidence, the exact mechanism of the emergence and development of this disease is still unknown. The ongoing work focuses on understanding the processes associated with the so-called secondary liver sinusoidal endothelial cell dysfunction (LSECs), which appears to be crucial in the development of NAFLD and contribute to the further progression of this disease towards hepatitis and cirrhosis. The work uses a unique approach involving the study of isolated liver cells derived from a mouse model of non-alcoholic fatty liver resulting from the use of a high-fat diet. The key aspect of the work is to find spectroscopic markers that allow to identify the appearance of dysfunction and track its development during disease progression. For this purpose, research was carried out at an early stage(2 weeks high-fat diet) and late (15-20 weeks) development of fatty liver. In addition, the purpose of the work is to explore the processes of lipid uptake and metabolism in LSECs and hepatocytes. Raman spectroscopy has become a valuable tool in research at the cellular and subcellular level, enabling detection and localization of biochemical changes that occur during the development of dysfunction, as well as to study various cellular processes. The use of this technique creates new research opportunities and at the same time, strengthens the application potential of vibrational spectroscopy methods.


Label-free imaging of subcellular structures of endothelium using Confocal Raman Spectroscopy supported by FT-IR Spectroscopy, AFM and Fluorescence Microscopy

Funding: National Research Centre (funding agency), Program PRELUDIUM 5
Project leader: Katarzyna B. Majzner, PhD

The project involves the development of novel methods with the use of confocal Raman microspectroscopy, infrared absorption spectroscopy, atomic force microscopy (AFM) and fluorescence microscopy to simultaneously evaluate changes in the level of intracellular morphological, biochemical and physicochemical, with particular emphasis on research formation and the role of lipid bodies endothelium. Lipid droplets (LDs), sometimes referred to in the literature lipid bodies are organelles generally known in the neutrophils, eosinophils, or tumor cells. The presence of LDs in the endothelium represents a new aspect of the research, especially that the role of LDs in cells is unexplored. Thus naturally comes the overall objective of the project, i.e. the development of a comprehensive research methodology to study the effect of supplementation with arachidonic acid on the presence of LDs in endothelial cells. Using the developed research methodology for study of LDs in the endothelium is the basis for a better understanding of the biological role of the LDs, including study the formation these structures and their functions.

The hypothesis of the potential protective role of lipid bodies, as a reservoir of arachidonic acid available for the production of prostacyclin (PGI2) will be verified using the methodology developed in the project.

Leading research method planned in the project is Raman spectroscopy. This is dictated by several factors:

  • The opportunity to study the biochemical composition of cell at subcellular level in a non-destructive and sensitive way (possibility of a live cells studies),
  • the possibility of rapid and unambiguous identification of LDs in a cell based on the unique spectroscopic “signature” of these organelles,
  • the ease of obtaining information about the distribution of the main biochemical components with high spatial resolution (up to ca. 300 nm),
  • using a confocal Raman imaging allow for the 3D reconstruction and tracking the distribution, shape and size of LDs.

In addition, examination of cells using Raman spectroscopy will be supported by AFM, what allows the simultaneous examination of the biochemical composition and topography of the analyzed sample. Applying a combination of Raman spectroscopy and AFM to study these same areas of the cells is possible to obtain complete information about the biochemical composition of endothelial cells in vitro and the distribution of the individual components and investigate the physical and mechanical properties of the sample. Knowledge about the changes taking place in the cell as a result of pharmacological stimulation will be supplemented by the results obtained from the use of infrared absorption spectroscopy and fluorescence microscopy (immunocytochemical staining).

Described project is particularly important because of the proposed research methodology taking into account the combination of vibrational spectroscopy, atomic force microscopy and fluorescent staining. This approach to research will allow for a significant increase in both knowledge of the biochemical changes associated with pharmacological stimulation of endothelial cells, the role of lipids in endothelial bodies, as well as presenting new research methodology that can be successfully applied to other systems. It should be noted that this project is focused on the theme, brand new in the context of research on the endothelium, because knowledge about lipid bodies observed in the endothelium is negligible. Although the results have no direct potential application, they will contribute to new information and reassessment of existing scientific data.

The results of the project will be incorporated to the PhD thesis of the Applicant. They will be also presented at the conference forum and published in pre-reviewed papers in the international chemical or chemistry-related journals. Experience of the Raman Imaging Group Faculty of Chemistry UJ in terms of Raman spectroscopy (measurements and advanced spectral analysis), as well JCET research groups in in vitro pharmacological and biological studies, and the availability of the necessary equipment for the project allow for the statement that the risk of this project is low.


Small scale, great significance: Raman spectroscopy, 3D imaging, atomic force microscopy and immunohistochemical staining in study of diabetes

Funding: National Research Centre (funding agency), Program PRELUDIUM 4
Project leader: Marta Z. Pacia (Pilarczyk), Ph.D.

The main aim of the project is to find spectroscopic changes in the vascular wall in the healthy and diabetic (db/db model) mice. The proposed study is aimed to determine the biochemical composition of vascular wall samples, to investigate the effect of diabetes on the biochemical composition and structure of blood vessel walls, find differences between healthy and diabetic tissue and explain them on the biochemical ground. In order to achieve the objectives of the project, an innovative and unique approach is offered, i.e. Raman scattering spectroscopy technique with an emphasis on Raman imaging, 3D imaging correlated well with other techniques: atomic force microscopy (AFM) and immunohistochemical staining (IHC). This project involves increasing the knowledge and awareness of changes taking place in tissue (vascular wall of mice) in study of diabetes using combination of Raman/AFM spectroscopy.

The subject of the research will be vascular wall samples of healthy and diabetic mice (model db/db). Two different types of preparations: the aortic cross-section of example thickness of 5 mm and aorta en face will be applied. Measurements of en face samples allows for studying of the inner side of aorta after opening (aorta is cut along and opened). The basic technique used in project is Raman imaging allowing for studying of the biochemical composition of the vascular wall and distribution of the individual components of the selected areas of tissue. Especially, for en face samples it seems to be a valuable idea to perform a series of Raman images for the same area of a tissue and visualize changes in the biochemical composition in the depth profile of tissue to obtain 3D Raman image. The project proposes a new approach using a combination of Raman spectroscopy, AFM and immunohistochemical staining to study of exactly the same areas in the sample. Multimodal approach of Raman imaging, combined with atomic force microscopy (AFM), immunohistochemical staining (IHC) and probably with scanning near-field optical microscopy (SNOM) provides complex information about chemical, physical and mechanical properties of the vascular wall with emphasis on the changes between healthy and diabetic mice.

Diabetes as a civilization disease is a major health problem for many people around the world. Currently, a lot of work focuses on developing methods to prevent and treat diabetes. This project is focused on understanding of the exact biochemical changes in vascular wall of diabetic subjects (db/db mouse model) compared with healthy animals. The results obtained in the project will expand the knowledge on the impact of diabetes on biochemical composition of vascular wall. Although the results are not directly applicable, they will contribute find new information about diabetes and reassess of existing scientific data. Application of Raman imaging combined with AFM and aided by SNOM and IHC staining to analyze the vascular wall is an innovative approach that allows obtaining multidimensional information about the studied system. The reached conclusions will help to increase knowledge about the mechanisms of vascular wall inflammation in diabetes, that is of the prime importance from the point of view of diabetes treatment and prevention.The results will be published in scientific journals and presented at conferences in Poland and abroad, which allows for their popularization.


The role of reactive oxygen species (ROS) generated during a maximal exercise by endothelial NOX2 i NOX4 in the regulation of post-exercise oxidant stress, postexercise haemostasis and exercise capacity

Funding Agency: National Science Centre, Poland, Program: PRELUDIUM 13
Project leader: Kamil Przyborowski Ph.D. 

Oxidative stress, resulting from overproduction of reactive oxygen species (ROS), may lead to damage of cellular structures, proteins, lipids and DNA and has been involved in a wide range of pathologies such as neurodegeneration, cardiovascular diseases and cancer. On the other hand, ROS play an important role as signaling molecules in regulation of physiological processes, including cell proliferation and differentiation, inflammation, innate immune response, vascular tone and endothelium phenotype.

Physical exercise results in excessive generation of ROS and could trigger oxidative stress resulting in impaired skeletal muscle contractility, muscular fatigue and endothelial dysfunction due to NO inactivation. On the other hand, there is a growing number of evidence that ROS generated during exercise could improve insulin sensitivity, enhance physical fitness and extend the lifespan. Currently, there is no data clearly defining the beneficial and detrimental effects of ROS generated during a single bout of acute exercise that could depend on the different enzymatic source of ROS.

The aim of this project is to investigate the role of ROS generated during a single bout of maximal exercise by endothelial NOX2 and NOX4 enzymes (Nicotinamide Adenine Dinucleotide Phosphate (NADPH) Oxidase) in post-exercise oxidant stress and adaptation/maladaptation of endothelial cells, and in regulation of exercise capacity and post-exercise haemostatic/thrombotic parameters using unique animal models of endothelial-specific NOX knock-out mice.

The identification of beneficial or detrimental role of ROS generated in response to exercise depending on their enzymatic sources in endothelium may contribute to development of innovative pharmacological endothelium-targeted strategy which could improve exercise tolerance and protect against dangerous consequences of acute intense exercise in patients with endothelial dysfunction.


Vibrational spectroscopy in analysis of biochemical status of blood that mirrors vascular wall pathology in animals with systemic and pulmonary hypertension

Funding: National Research Centre (funding agency), Program PRELUDIUM 5
Project leader: Emilia Staniszewska, PhD

The main aim of the project is to determine the overall biochemical profile of blood components in animal models of systemic hypertension and pulmonary hypertension by using Fourier Transform Infrared (FTIR) and Raman spectroscopy. The study will be carried out for various stages of pathological changes in both models of hypertension. Planned research and defined methodology will allow developing a new strategy for predicting the pathology of blood vessel wall and related risks on the basis of fingerprint induced by hypertension in biochemical profiles of blood recorded in vibrational spectra.

The object of the studies will be platelet poor- and rich- plasma, serum, erythrocytes and leukocytes isolated from healthy animals (mice BL/6J C57 and rats type Wistar) and from animals with induced systemic hypertension (mouse model after 8-week administration of L-Name) and pulmonary hypertension (rat model after single administration of monocrotaline). A tool of investigations will be two techniques of vibrational spectroscopy: Infrared and Raman spectroscopy. Searching of spectral markers will be supported by chemometric methods (cluster analysis (CA), principal components analysis (PCA), etc.), and next spectral analysis will be compared with structural , biochemical, and functional markers of pathology of blood vessel wall.


The role of PGI2 and NO, produced by liver sinusoidal endothelial cells (LSEC), in the regulation of hapatic glycogenolysis and gluconeogenesis at an early stage of NAFLD development – studies using a unique isolated perfused liver system

Funding: National Research Centre (funding agency), Program MINIATURA
Project leader: Izabela Czyżyńska-Cichoń, PhD

The aim of the project is to assess the role of prostacyclin (PGI2) and nitric oxide (NO) produced by liver sinusoidal endothelial cells (LSEC) in paracrine regulation of hepatic glycogenolysis and gluconeogenesis at an early stage of the development of non-alcoholic fatty liver disease (NAFLD). This research may answer the questions: if LSEC mediators can regulate glucose metabolism in hepatocytes and whether LSEC-dependent glucose metabolism regulation is impaired during NAFLD? These studies will use the unique ex vivo method of isolated liver perfusion, which enables biochemical and functional profiling of the organ avoiding the influence of other tissues.


Analysis of the molecular mechanisms of metastasis in murine model of breast cancer via the comprehensive study on protein expression changes in lungs

Funding: National Research Centre (funding agency), Program MINIATURA 1
Project leader: Anna Kurpińska, PhD

Breast cancer is one of the most common cancers. It is characterized by a robust ability to metastasize, specifically to the lungs and consequently results in high mortality. Despite significant progress in diagnosis and treatment still not fully understood mechanisms of tumorigenesis and metastasis form a barrier for the selection of an effective and selective drugs.

The aim of this project was to analyse the mechanisms underlying dysfunction of the lungs in the course of cancer progression, via the search for biomarkers characteristic for early metastatic processes in the lungs and description of the metabolic shift in late phase of the cancer progression.

Application of the proteomic techniques enabled broadening the knowledge in the field of cancer metastasis, characterization of the molecular changes and to analyze the communication and adaptation mechanisms of the metastases development. An important result of the presented studies is the answer to the question on how lung tissue is remodeled in the early stages of cancer progression, when the metastatic cells are not observed or only few cancer cells are present in the lungs. The obtained results clearly indicate very early stimulation from cells forming the primary tumor and activation of mechanisms that prepare the tissue for the invasion of cancer cells.


Non-invasive MRI assessment of microvascular nitric oxide synthase dysfunctionand permeability in the lung endothelium in a mouse model of a chronic heartfailure

Funding: National Research Centre (funding agency), Program MINIATURA
Project leader: Grzegorz Kwiatkowski, PhD

Endothelial dysfunction is a key characteristic of several cardiovascular diseases including diabetics, atherosclerosis and hypertension. In particular, global endothelial status holds a prognostic value in the development of heart failure. Several reports shown endothelial dysfunction developed over the course heart failure including impairment ofnitricoxide synthesis. Moreover, at the advanced stage of heart failure, the endothelial impairment was also observed in the lungs. There is, however, very little known on a relationship between lung microvascular endothelial status and heart failure progression. In particular, it is not known whether the lung endothelium exhibits adaptive or maladaptive mechanisms towards heart failure progression. The lung endothelium covers up to 30% of the total endothelial surface and as such is potentially an important treatment target.

Recent developments in the Magnetic Resonance Imaging offer novel, dedicated methods to study microcirculation but have not been yet applied to the lungs. We plan to employed state-of-the-art MRI protocols to characterize changes in the lung endothelium phenotype during the progression of heart failure in a mouse model of this disease.


Changes in paracrine signaling of liver sinusoidal endothelial cells stimulated with fatty acids and lipid droplets formation in hepatocytes – studies with use of a platform for three-dimensional co-culture under micro-flow condition, OrganoPlate

Funding: National Research Centre (funding agency), Program MINIATURA 1
Project leader: Edyta Kuś (Maślak), PhD

The goal of the project was to investigate the effect of fatty acids on the paracrine signaling of liver sinusoidal endothelial cells (LSEC) and its contribution to the formation of lipid drops in hepatocytes. Research were conducted using a unique platform for three-dimensional co-cultures in micro-flow, OrganoPlate mimicking liver physiological environment, i.e. static conditions in the extracellular matrix for hepatocytes and controlled microflow for LSEC with maintained continuous communication between cells. The project aims to better understand the role of LSEC in the development of fatty liver disease.


Effect of visfatin on endothelial bioenergetics and phenotype

Funding: National Research Centre (funding agency), Program MINIATURA
Project leader: Łukasz Mateuszuk, PhD

Nicotinamide phosphoribosyltransferase (NAMPT) and 5’ectonucleotidase (CD73) are involved in nicotinamide (NA) metabolic pathway, leading to the synthesis of nicotinamide adenine dinucleotide (NAD), an important cofactor of redox reactions and substrate for sirtuins and poly-ADP-ribose polymerases (PARPs). Intracellular NAMPT is responsible for nicotinamide mononucleotide (NMN) synthesis from NA, which is a direct precursor of NAD. Extracellular form of NAMPT is known as visfatin (Vis), an adipokine with pro-inflammatory properties, which high plasma concentration has been previously detected in obesity, type 2 diabetes and metabolic syndrome. Vis increases IL1, IL6 and TNFα production, decreases endothelial nitric oxide synthesis and most probably is also released by dysfunctional endothelium, which may lead to vascular inflammation usually preceding the symptoms of cardiovascular diseases. Both NAMPT and Vis are encoded by the same gene, but Vis protein molecule is slightly bigger, which suggests post-translational modification of polypeptide chain. Although adipokine activity of Vis has been documented during previous studies, the extracellular enzymatic properties of this protein still requires additional studies. By ribosyl-phosphorylation of extracellular NA, Vis may act as an alternative source of NMN. The main aim of this project was to assess endothelial expression of Vis and CD73 in basal conditions and during endothelial disfunction, as well as examining the phosphoribosiltransferase potential of visfatin in a presence of NA.


Application of a new immunohistochemical technique for imaging of DMPO spin trap to image oxidative stress in cardiac muscle in mice

Funding: National Research Centre (funding agency), Program MINIATURA
Project leader: Bartosz Proniewski, PhD

Reactive oxygen species (ROS) serve as important physiological transmitters, however in inflammation and other pathologies, they can be overproduced and overpower the antioxidant capacity, leading to the so called oxidative stress. Clinical effects of oxidative stress have been reported in heart failure and in various mouse models of this disease, including earlier studies using the Tgαq*44 mouse model, where increased superoxide production was seen within the heart. Methods typically used to study oxidative stress are either unspecific (fluorescent probes) or require sample homogenization. What is more, in the literature often just the increased production of a reactive oxygen species (e.g. superoxide) is considered indicative of oxidative stress, whereas in fact it’s definition encompasses an increased ROS production that is unbalanced by the inefficient antioxidant defense.

Here, DMPO immuno-spin trapping (IST) has been used to image oxidative stress in the heart of mice, based on to the high reactivity of DMPO spin trap against protein/lipid radicals and the utilization of specific anti-DMPO antibodies, allowing the visualization of oxidative modifications within the hearts of transgenic Tgαq*44 mice, which develop heart failure over time due to a cardiomyocyte-specific overexpression of the Gαq protein. It was demonstrated that these detrimental changes appear much later, then increased superoxide production is detected, but parallel to the first functional symptoms of heart failure, as seen using in vivo imaging (USG/MRI). This is due to effective antioxidative mechanism that are activated in the studied mouse model. The results suggest, that the IST method in fact shows the “end effect” of oxidative stress, in contrast to measurements based solely on free radical detection, without monitoring of enzymatic activity of the antioxidative pathways in parallel.

Fluorescent imaging of DMPO-nitrone adducts in slices of the isolated myocardium allowed for determination that the ventricles are most affected by oxidative modifications, especially the left ventricle. Moreover, using a double staining procedure with lectin antibody to visualize the coronary endothelium, it was shown that it is not only the cardiomyocyte being affected. In fact, both large coronary vessels and small capillaries are under oxidative stress. These oxidative modifications seen in the endothelium suggest that there has to be a mechanism by which oxidative stress is propagated from the cardiomyocytes, where it originates based on the Gq protein modification, to the endothelium, and it’s identification and pharmacological modulation may lead to the design of novel, effective therapeutic route in heart failure.


Multiparametric analysis of pulmonary endothelial dysfunction in the murine model along the progression of breast cancer

Funding: National Research Centre (funding agency), Program MINIATURA 1
Project leader: Marta Smęda, PhD

Healthy endothelium constitutes an “impermeable barrier” for cancer cells. The aim of the project is to perform multiparametric characterization of endothelial dysfunction in the metastatic lungs throughout the progression of breast cancer that will comprise measurement of pulmonary NO production and eNOS activation, assessment of mesenchymal transformation of pulmonary endothelium (EndMT) as well as pulmonary endothelium permeability and activation. The study will be conducted in the orthotopic model allowing for spontaneous metastasis as well as in the model of experimental metastasis after intravenous inoculation of cancer cells. The study will contribute significantly to the field of experimental research regarding mechanisms of metastasis.


The PGI2 / SIRT1 pathway is a chance to revolutionize pharmacotherapy of cardiovascular disease

Funding: National Research Centre (funding agency), Program MINIATURA 2
Project leader: Magdalena Sternak, PhD

SIRT 1 is attributed to participation in the activation of repair mechanisms in endothelial dysfunction. Despite the extensive literature on sirtuins and in the context of the non-specificity of SIRT1 activators (Pacholec et al., 2010), their mechanism of action, and in particular the contribution of the PGI2/COX2 pathway to the SIRT1 regulation mechanism, remains unclear. There are speculations that the use of SIRT1-targeted drugs by acting on endogenous endothelial mediators can be a promising therapy for the pathogenesis of human diseases, which is one of the major challenges of medicine. The aim of the planned research in the project is to investigate the role of prostacyclin PGI2 in defense mechanisms associated with SIRT1 activity in improving the vascular endothelium function in the mouse transgenic model (endothelial CDK5R1 (EC-p25) with the overexpression of the EC-p25 protein. The EC-p25 mouse model that was used to implement the project is an innovative transgenic model, which may contribute to the explanation of important mechanisms of endothelial dysfunction, and may also contribute to finding effective therapy in the treatment of diseases based on interference in the pathway associated with SIRT1 and thus lead to the development of clinical trials.

Budget: 49 500 PLN, Time: 12 months,


Toward pharmacology of cancer cells adhesion and transendothelial migration using unique 3D microfluidic platforms

Funding: National Research Centre (funding agency), Program MINIATURA 1
Project leader: Marta Stojak, PhD

The main goal of the project was to learn and implement the unique approach of 3D microfluidic platforms (VMT, vascularized microtumors) during the internship in the laboratory of Professor Christopher C. W. Hughes (Department of Molecular Biology and Biochemistry, University of California, Irvine, USA) and the use of this methodology to study the adhesion and transendothelial migration of cancer cells. The main problem in understanding cancer cells interactions with the endothelium is lack of good in vitro models. Classical models put on simplicity and convenience, at the same time ignoring complex interplay between tumor cells and microenvironment. In the Hughes Lab, an innovative 3D microphysiological system was developed which ensures physiological flow and transport of nutrients and metabolites. In addition, the VMT platform is transparent, which allows non-invasive imaging of tissues or cells. All together VMT devices have great potential and advantage over other commercially available microfluidic devices as well as on the classic system I have used in previous studies.


The effect of the activators of Nrf2 transcription factor on endothelial permeability- ex vivo studies on B6.129X1-Nfe2l2tm1Ywk / J knockout mice

Funding: National Research Centre (funding agency), Program MINIATURA
Project leader: Ewa Szczęsny-Małysiak, PhD

The aim of the project is to investigate the role of the activation of Nrf2 transcription factor in changes in aortic endothelial cell permeability (which is a measure of endothelial dysfunction). Experiments will be performed ex vivo on blood vessels taken from knockout mice B6.129X1-Nfe2l2tm1Ywk / J Nrf2 characterized by impaired expression of Nrf2 factor and wild-type C57BL / 6J mice. Beforehand, the blood vessels will be treated with drugs – activators of factor Nrf2: methyl bardoxolone, dimethyl fumarate and L-sulforaphane. Showing differences in the action of individual substances will allow to determine whether the observed effects depend on the activation of Nrf2 or whether it will be necessary to determine another molecular mechanism of the occurring phenomena.