Veličina fonta
Sivi ton
Kontrast
Disleksija

Studenti koji imaju simptome infekcije ili dokazanu bolest COVID-19 ili su bili u bliskom kontaktu s oboljelim osobama moraju se istoga dana obvezno javiti elektroničkom poštom na adresu: covid-mef@mef.hr

Projekt HRZZ: MEFRA

Projekt Hrvatske zaklade za znanost br. 7406

Molecular mediators of Fas-driven osteoresorption in arthritis

ACRONIM: MEFRA
Principal Investigator (PI) – prof.dr.sc. Nataša Kovačić
Project duration in months: 48 (2.11.2015-31.10.2019)
Project funding: 1.000.000 HRK (250.000 HRK / year)

Project synopsis:

The inflammatory arthritides are chronic joint diseases, and a major health problem because of progressive disability. The most common form is rheumatoid arthritis (RA) characterized by bone and cartilage damage leading to deformity and permanent disability. Arthritis is also a common in systemic lupus erythematosus (SLE), mostly as a non-erosive form.

In RA, inflammatory and immune cells create a microenvironment that favors osteoresorption by increasing osteoclast, and suppressing osteoblast differentiation and activity. Current treatment options such as biological therapeutics may modify the course of disease, but are not able to reverse osteoresorption.

The Fas/Fas ligand system is important regulator of immune system homeostasis, and there is accumulating evidence for the role of this system in the regulation of bone homeostasis. Inactivation of Fas has a bone-sparing effect, preventing bone loss upon estrogen withdrawal, as well as in inflammatory arthritides, but the exact lineages and signaling pathways mediating this effect are not defined.

Using the murine model of antigen-induced arthritis (AIA) in mice deficient for Fas, and synovial fluid samples from patients with RA and SLE, we aim to identify cell populations responsible for inhibition of osteoresorption, as well as functional alterations in these populations by analysing their gene expression profiles. In addition, we expect to confirm the role of identified genes/molecules by their inactivation in vivo in the murine model of AIA, and the role of a lineage using conditional lineage-specific models of Fas-inactivation. The analysis of human samples will allow for the translation to the clinical setting.

Our approach aims to identify novel cellular/molecular drivers of bone resorption in arthritis, on a lineage-specific level. Proposed studies will provide new targets for preventing, reducing, and potentially reversing the osteoresorptive process which is a consequence of inflammatory events.

DETALJNIJE INFORMACIJE O PROJEKTU

Pathogenesis of inflammatory arthritis

The inflammatory arthritides are chronic joint diseases, which represent a major health problem because of progressive disability and only partially successful therapy (Pisetsky DS. Best Pract Res Clin Rheumatol2012). The most common form of inflammatory arthritis is rheumatoid arthritis (RA),which occurrs in 1-2% of the population (Scott DL. Lancet 2010), followed by a spondyloarthritis (SpA), occurring in 0.3–0.6% of the population (Helmick CG. Arthritis Rheum2008; Akkoc N. Curr RheumatolRep2008). Arthritis is also a common feature, occurring in 60%–90% of patients with systemic lupus erythematosus (SLE), and in the majority of cases presents as non-deforming and non-erosive arthritis, whereas only a minor proportion of patients develop deforming or erosive arthritis (Kakumanu P. J Rheumatol 2009).

The main characteristic of inflammatory arthritides is synovial infiltration by lymphocytes, macrophages and neutrophils, which all produce cytokines and inflammatory mediators (McInnes IB. NatRevImmunol 2007; Feldmann M.Cell 1996). Another important feature of inflammatory arthritisis synovial thickening, which develops as a result of uncontrolled fibroblast-like synoviocyte (FLS) proliferation, and is responsible for the destruction of underlying cartilage and bone (Ospelt C. Best PractRes ClinRheumatol 2008).

Infiltrating cells as well as proliferating FLS in arthritis produce pro-inflammatory  mediators, such as tumor necrosis factor (TNF)-α, interleukin (IL)-1, -6, -7, -8, -15, -17, -18, -32, -35,and interferons (IFN)(McInnes IB. Nat Rev Immunol 2007; Furuzawa-Carballeda J. Autoimmun Rev 2007; Lotito AP. Autoimmun Rev 2007; Malemud CJ.Open Access Rheumatol: Res Rev 2009; Malemud CJ. J Clin Cell Immunol 2011; Hartgring SA. Ann Rheum Dis 2006; Petrovic-Rackov L. Clin Rheumatol 2006; Joosten LA. Proc Natl Acad Sci USA 2006; Collison LW. Nature 2007; Bottini N. Nat Rev Rheumatol 2012). These cytokines trigger multiple signaling pathways, such as JAK/STAT, SAPK/MAPK and PI-3K/AKT/mTOR, responsible for the increase of matrix metalloproteinase (MMP) gene expression, further downstream activation of nuclear factor (NF)-κB, prolonged survival and resistance to apoptosis of immune-cells and/or FLSs, as well as the regulation of neo-angiogenesis, and thus the perpetuation of inflammation (Malemud CJ. J Clin Cell Immunol 2013).In addition to the intra-articular processes, the underlying bone marrow is also infiltrated with T and B lymphocytes, plasma cells and macrophages, and contains increased numbers of active osteoclasts contributing to subchondral bone loss (Bugatti S. Arthritis Res Ther 2012). All of these point to a great complexity in the pathogenesis of the disease involving multiple cell lineages and signaling pathways, which accounts for a heterogeneity in clinical presentations and prognoses.

Apoptotic signaling and the Fas/Fas ligand system in arthritis

Pathogenesis of arthritic disease is regulated not only by the activity of infiltrating cells, but also by their lifespan, which is controlled by apoptosis. Apoptosis may be triggered internally, in response to cellular stress, or externally, via death receptors, characterized by the intracytoplasmatic death domain, which then triggers caspase signaling (Aggarwal BB. Nat Rev Immunol 2003). Fas (CD95, Apo-1) is a typical death receptor first described as the regulator of immune system homeostasis, particularly through the phenomenon of activation induced cell death (Askenasy N. Blood 2005). However, it is ubiquitously expressed on different cells and tissues (French LE. Cell Death Differ 2003). Upon activation by Fas ligand, the intracellular part of Fas recruits and activates caspases-8 and -10, which further activate caspases-3, -6 and -7, effectors of apoptosis (Siegel RM. Nat Immunol 2000). In addition to apoptosis, caspase-8 is essential for the normal function of myeloid and B lymphocyteprogenitors in bone marrow, and is able to inhibit the activity of NF-κB signaling pathway (Kang TB. J Immunol 2004; Frelin C. Cell Death Diff2008), whereas caspase-3 is involved in the regulation of osteoblast differentiation of bone marrow stromal cells (Miura M. J Clin Invest 2004).

Clearly, the end result of Fas activation depends on the cell type, as well as on its stage of activity or differentiation. For example, Fas signaling can be modified by inhibitors of apoptosis, such as X-chromosome-linked IAP (XIAP), whose elevated levels of may be responsible for low caspase-3 activity and apoptosis-resistance of FLSin RA (Lewis AC. Recent Res Devel Pharmacol 2011).Fas-like inhibitory protein (FLIP) can block caspase-8 activation by Fas and induction of apoptosis, so cells expressing high levels of FLIP had low caspase-8 activity and increased resistance to Fas-induced apoptosis(Malemud CJ. Curr Rheum Rev 2005; Malemud CJ. J Clin Cell Immunol 2011).Another mechanism for resistance of FLS to Fas-induced apoptosis involves the activation of PI-3K/AKT/mTOR-dependent-SphK1 pathway (Wisler BA. Curr Signal Transduct Ther 2011). Under such conditions, death receptor activation may result in non-apoptotic signaling which is becoming increasingly recognized. Activation of Fas has been even shown to promote cell growth (Chen L. Nature 2010).

A substantial amount of data on the physiological role of the Fas/Fas ligand system is obtained by studies in mice with spontaneous mutations in the Fas or Fas ligand genes, which results in the lymphoproliferative syndrome (lpr) or generalized lymphoproliferative disorder (gld), both characterized by lymphadenopathy, splenomegaly and systemic autoimmunity (Roths JB. J Exp Med 1984; Ramsdell F. Eur J Immunol 1994; Watanabe-Fukunaga R. J Immunol 1992). The same, even more pronounced phenotype is observed in knock-out mouse strains (Fas -/-, Adachi M. Nat Genet 1995; and FasL -/-, Karray S. J Immunol 2004). The severity of the phenotype is strain-specific, with the MRL strain developing the most severe phenotype, resembling human SLE, and is often (~20% of animals)accompanied by arthritis resembling human RA (Ratkay LG. J Immunol 1993). Mutations in human Fas/Fas ligand genes, or genes coding their downstream signaling molecules similarly produce the severe autoimmune lymphoproliferative syndrome (ALPS; Rieux-Laucat F. Science 1995; Fisher GH. Cell 1995). More data on the cell lineages required for the autoimmune phenotype come from studies in mice with conditional inactivation of Fas by Cre recombination, where an inactivation of Fas in T lymphocytes, B lymphocytes or both cell types in C57BL6 strain failed to induce a lymphoproliferative phenotype (Hao Z. J Exp Med 2004). On the other side, selective Fas-dependent depletion of M2 (M-CSF-dependent) macrophages in “Mafia” (macrophage Fas-induced apoptosis), resulted in lymphadenopathy and splenomegaly (Burnet SH. J Leukoc Biol 2004; Fleetwood AJ. J Leukoc Biol 2009), suggesting myeloid cells as important players in immune system homeostasis regulated by Fas.

As an immune regulator, the Fas/Fas ligand system has already been attributed to pathogenesis of RA and inflammatory arthritides. It acts either to suppress inflammatory responses via activation-induced cell death of auto reactive T lymphocytes, inflammatory myeloid cells and/or FLS (Parihar A. J Innate Immun 2010), or to mediate direct tissue damage through apoptosis of bone/cartilage cells. However, the exact role of Fas in arthritis is still unclear, and there are contradicting reports on the beneficial, as well as detrimental roles in the pathogenic process (Peng SL. Rheumatology (Oxford) 2006). Fas/Fas ligand system seems inappropriate for systemic targeting, not only because of multiple effects on the different cell lineages in the inflamed joints, but also due to lethal consequences of systemic Fas blockade through induction of liver failure (Ogasawara J. Nature 1993). Revealing molecular mediators of Fas signaling and dissection of their lineage-specific effects may offer a more specific targets, applicable in clinical setting.

Bone loss in arthritis

As mentioned above, bone loss is often a consequence of inflammatory arthritis, leading to irreversible structural changes of the joint. Current treatment options such as biological therapeutics to block pro-inflammatory cytokines (ie IL-1, TNF, IL-6, IL-12/IL-23), have been shown to modify the disease course, but are not able to reverse osteoresorption, indicating the need for further studies aimed to increase the reparatory processes (Lundewe R. Arthritis Rheum 2006; Furst DE. Clin Exp Rheumatol 2010).

Bone loss occurs due to predomination of bone resorption by osteoclasts over bone formation by osteoblasts (WalshNC. Immunol Rev 2010; Schett G. Arthritis Res Ther 2009). Differentiation of osteoclasts is triggered by activation of receptor activator of NF-κΒ (RANK) signaling in osteoclast progenitors (OCPs) in the presence of macrophage colony-stimulating factor (M-CSF) (Boyce BF. Arch Bichem Biophys 2008). RANK ligand (RANKL) is expressed on osteoblast lineage cells, hypertrophying chondrocytes, activated T lymphocytes and FLS, and can be stimulated by a number of local and systemic inflammatory mediators. RANKL binding is inhibited by osteoprotegerin (OPG), which acts as a soluble decoy receptor for RANKL (Boyce BF. Arch Bichem Biophys 2008). Besides acting on the RANK/OPG system, the inflammatory process affects migration, homing, commitment and activation of different myeloid lineage populations, containing OCPs.

In addition to increased bone resorption by osteoclasts, differentiation of mesenchymal progenitors into bone forming cells(osteoblasts) is also affected, thus contributing to bone loss.Osteoblasts develop from mesenchymal stem cells (MSC) under the control of transcription factor Runx2 (Ducy P. Cell 1997). MSC reside inbone marrow and other tissues including synovia. Inflammatory cells canaffect the survival, the differentiation and/or apoptosis of mesenchymal lineage cells via secreted cytokines and surface cellular receptors and ligands,inducing various responses such as proliferation, differentiation or cell death. Generally, mesenchymal cells within the hyperplastic synovia are characterized by a reduced ability to differentiate into mature mesenchymal lineage cells (osteoblasts, chondroblasts, and adipocytes) due to an increased expression of NF-κB which promotes their proliferation and sustains them in an undifferentiated state (Li X. Proc Natl Acad Sci USA 2006; Lazic E. Arthritis Res Ther 2012).

There is increasing evidence of an involvement of the Fas/Fas ligand system in bone homeostasis (Kovacic N. Expert Opinion Ther Targets 2011). Estrogen deficiency, a major cause of osteoporosis, is characterized byan increased expression of Fas on cells of the osteoblast lineage (Garcia-Moreno C. Bone 2004; Kovacic N. Lab Invest 2010), or Fas ligand on the cells of the osteoclast lineage (Nakamura T. Cell2007). Absence of Fas ligand in vivo results in an increased bone mass (Katavić V. J Immunol 2003), and a deficiency of either Fas or Fas ligand is able to protect mice from estrogen withdrawal-induced bone loss (Katavić V. J Immunol 2003; Kovacic N. Lab Invest 2010). Effects of systemic Fas inactivation in the prevention of inflammation-induced osteoresorption in arthritis have been shown in the Fas deficient lpr strain on a DBA/J1 background, which exerts the attenuated form of the collagen-induced arthritis (CIA, a murine model of RA), with fewer bone erosions (Hoang TR. Arthritis Res Ther 2004). Our preliminary data on the murine model of antigen induced arthritis (AIA) induced by methylated bovine serum albumin (mBSA) in Fas -/- mouse strain on the C57BL6 background also revealed milder arthritis and less subchondral bone loss compared to a wild-type mice.

From all this, it is clear that Fas inactivation can alter the course of arthritis, having a bone-sparing effect.Further investigation of the altered signaling molecules in distinct lineages involved in the pathogenesis of arthritis could define novel signaling molecules involved in the cross-talk and may offer new targets to pharmacologically modulate osteoresorption in arthritic disease.

Scientific objectives

  • Analyze the cellular composition of the synovial compartment, and subchondral bone marrow in the model of AIA attenuated by Fas deficiency, to determine whichlineages and lineage subpopulations could contribute to the attenuation of arthritis and bone-sparing effects of Fas ina
  • Analyze the cellular composition of SF and synovial tissue from patients with RA (osteoresorptive form) and non-resorptive arthritides or control patients.
  • Define the critical molecular mediators in the affected subpopulations in murine and human samples by comparisons of expression profiles of sorted synovial and subchondral bone marrow-derived myeloid/mesenchymal/lymphoid lineage cells from wild-type and Fas -/- mice with AIA, as well as sorted synovial fluid-derived myeloid/mesenchymal/lymphoid lineage cells from patients with RA and non-resorptive arthritis/control patients.
  • Confirm the importance of altered genes/proteins by in vivo functional studies assessing the development and course of AIA after inactivation/inhibition of selected molecules.
  • Confirm the importance of specific lineages in Fas-dependent attenuation of arthritis by assessing the development of AIA in the murine model of conditional Fas inactivation by Cre recombination driven by myeloid/mesenchymal/lymphoid lineage specific promoters.

Hypothesis

Various clinical and pathophysiological forms of arthritides are the result of pathogenic sequence characterized by participation of specific lineage cell subpopulations, which also determines bone destruction by inflammatory processes. Inactivation of Fas affects these sequences and prevents osteoresorption, by molecular and cellular mechanisms which are not yet fully defined. Osteoresorptive processes are main feature of human RA and uncommon in SLE-related arthritis.
An in depth analysis of frequencies of various cell subpopulations in osteoresorptive and non-osteoresorptive forms of murine and human arthritides, and their gene expression signatures, will determine the critical cellular and molecular regulators of bone resorption in arthritic diseases and provide new molecular targets for preventing and treating inflammation-induced osteoresorption.

PRELIMINIRANE STUDIJE I OČEKIVANI REZLTATI

Preliminary results

We have previously shown that mice with a spontaneous mutation in the Fas ligand gene have increased cancellous bone volume, trabecular number and thickness, and decreased trabecular spacing (Katavic V. J Immunol 2003). These mice also had increased osteoblast differentiation from bone marrow progenitors, and increased osteoblastic production of OPG. When we systematically analyzed the expression of Fas and Fas ligand on bone marrow-derived osteoblasts, we found low expressions of both Fas and Fas ligand in immature osteoblastogenic cultures, and a significant increase in Fas expression with osteoblast maturation (Kovacic J. Immunol 2007). However, exogenous Fas ligand was able to induce apoptosis of only a minor proportion of cells in mature osteoblastogenic cultures, but specifically reduced the number of osteoblast colonies, suggesting an effect on differentiation (Kovacic J. Immunol 2007). This mechanism was dependent on the caspase-8 activation, since inhibition of caspase-8 rescued the inhibitory effect of Fas ligand on osteoblast differentiation.

Our preliminary data showed that osteoblasts derived from synovial mesenchymal progenitors also express Fas, and expression was increased in AIA (Fig 1), suggesting that osteoblastogenesis can be suppressed by Fas in arthritis. We also confirmed that osteoblastogenesis from synovial progenitors of patients with juvenile idiopathic arthritis (JIA) was inversely correlated with the inflammatory activity, and that osteoblasts from more aggressive poliarticular JIA expressed more Fas than osteoblasts from patients with oligoarticular form of the disease (Lazic E. Arthritis Res Ther 2012).   Fas -/- mice have a more robust osteoblastogenesis, but basally an unchanged bone mass. However, we were interested whether systemic inactivation of Fas could have protective effect in conditions where excessive bone turnover resulted in bone loss, such as estrogen deficiency and local or systemic inflammation. We found that an absence of Fas protects mice from bone loss induced by ovariectomy and was related to increased osteoblastogenesis in response to estrogen withdrawal (Kovacic N. Lab Invest 2010). 3 weeks post AIA induction, Fas –/– mice revealed milder joint swelling, less synovial infiltration and thickening, and less cartilage and bone damage compared to wild-type mice and no loss of femoral metaphyseal trabecular bone.

How proposed research will improve development of research field and group

Our research group is focused on the functional interactions between bone and immune cells. We have confirmed that Fas /Fas ligand system is an important regulator of bone-cell differentiation and function (Katavic V. J Immuol 2003; Lukic IK. Clin Exp Immunol 2005; Kovacic N. J Immunol 2007; Kovacic N. Lab Invest 2010). We have extended our research to several human diseases, including arthritis (Lukic IK. Inflamm Allergy Drug Targets 2009; Lazic E. Arth Res Ther 2012; Ikic M Int Orthop 2014). In patients with different forms of arthritis, we have identified the expression profile of selected cytokines, growth and apoptotic factors that may serve as diagnostic markers (Grcevic D. J Rhumatol 2010). We have defined the role of TNF in endochondral bone formation, the role of B lymphocytes in bone formation, and subpopulations of murine OCPs and their responses to inflammatory stimuli in murine models and patient samples (Marusic A. Lab Invest 2000; Lukic IK. Clin Exp Immunol 2005, Cvija H. Inflammation 2012; Ikic M. Int Orthop 2014; Sucur A Int Orthop 2014). The proposed project will use the model of attenuated osteoresorption in arthritic disease which is a result of Fas deficiency to further elucidate cellular and molecular mechanisms of inflammation-induced bone destruction. We will expand the method set used in laboratory, by introducing microarray analysis which will provide a global insight in the molecular signature favoring osteoresorption within specific cell lineages. In addition, introducing conditional knockout models, which have not been previously applied in our laboratory, we will expand the ability to more precisely define the role of specific lineages involved in the immune-mediated regulation of bone metabolism.

The unique contribution of this research

Our study will provide the basis for further investigation of targets for prevention, reduction and potentially reversal of bone destructive process, which is a consequence of inflammatory arthritis.

It is clearly evident that systemic Fas inactivation hasan osteoprotective effect, so by using this model we aim to identify key cellular/molecular drivers of bone resorption in arthritis, on a lineage-specific level. Using SF samples from patients will enable translation to clinical setting.

Expected results

We expect to:

  • Confirm differences in the proportions and absolute numbers of myeloid, lymphoid and mesenchymal subpopulations in synovial compartment and subchondral bone marrow from wild-type and Fas -/- mice with AIA, and the synovial fluid patients with RA and control patients.
  • Identify key molecules mediating the attenuation of arthritis and preventing osteoresorption by analyzing differences in gene expression patterns among different lineages separated from the synovial compartment and subchondral bone marrow of the wild-type and Fas -/- mice with AIA, and synovial fluid samples from patients with RA and control patients.
  • Confirm the functional importance of identified genes/proteins by in vivo functional blocking of gene (siRNA) and/or protein (antibodies, enzyme inhibitors) function,

Confirm the functional importance of a specific lineage by conditional inactivation of Fas by Cre recombination in the murine model of AIA.

METODOLOGIJA I FINANCIJSKI PLAN

Methodology

The project will use mouse model of AIA and synovial samples of patients with RA and control patients. In vivo model of AIA in C57BL/6 mice, immunophenotyping by flow cytometry, cell sorting and RNA extraction and analysis, as well as analysis of human samples are already in use in the involved laboratories, so we do not expect major technical difficulties to apply them in the proposed experimental design. Preliminary studies on the mouse AIA model confirmed the proposed methodology as suitable to test the research hypothesis. The novel steps are separation of discrete cell populations from the inflamed joints, and obtaining sufficient amount of high quality RNA for subsequent gene expression analysis, and optimization of functional studies (siRNA, protein blocking and inhibition).

Financial plan

Total project budget: 1000000.00 HRK (250000.00/year)

SUDIONICI PROJEKTA I OPREMA

Scientific project leader

NATASA KOVACIC, M.D., Ph.D., associate professor

Department of Anatomy, University of Zagreb School of Medicine, Croatia

Croatian Institute for Brain Research; Laboratory for Molecular Immunology, Zagreb, Croatia

Project team members

DANKA GRCEVIC, M.D., PhD, professor, Department of Physiology and Immunology, University of Zagreb School of Medicine, Croatia

TOMISLAV KELAVA, M.D., PhD, assistant professor, Department of Physiology and Immunology, University of Zagreb School of Medicine, Croatia

NINA LUKAC, M.D., HRZZ project associate, PhD student, Department of Anatomy, University of Zagreb School of Medicine, Croatia

ALAN SUCUR, M.D., PhD, senior assistant, Department of Physiology and Immunology, University of Zagreb School of Medicine, Croatia

FRANE PAIC, molecular biologist, PhD, assistant professor, Department of Medical Biology, University of Zagreb School of Medicine, Zagreb, Croatia.

BRANIMIR ANIC, M.D., PhD, professor, Chair, Department for Clinical Immunology, University hospital centre, University of Zagreb School of Medicine, Croatia

MISLAV CEROVEC, M.D., PhD, assistant professor, Department for Clinical Immunology, University hospital centre, University of Zagreb School of Medicine, Croatia

MISLAV JELIC, M.D., PhD, associate professor, Department for Orthopedics, University of Zagreb School of Medicine, Croatia

KATARINA BARBARIC, M.D., Department for Orthopedics, University of Zagreb School of Medicine, Croatia

KATERINA ZRINSKI PETROVIC, laboratory technician, Laboratory for Molecular Immunology , University of Zagreb School of Medicine, Croatia

MARTINA FADLJEVIC, student, University of Zagreb School of Medicine, Croatia

IGOR RADANOVIC, student, University of Zagreb School of Medicine, Croatia

ELVIRA LAZIC MOSLER, M.D., Ph.D., dermatovenerologist, Clinical hospital Ivo Pedisic Sisak, Croatia

PETER IAN CROUCHER, M.D., Ph.D., associate professor, Bone Biology Division, Garvan Institute of Medical Research, Conjoint Professor, Faculty of Medicine, University of New South Wales, Australia.

NIVES ZIMMERMANN, M.D., associate professor, University of Cincinnati, Cincinnati Children’s Hospital, Ohio, USA

Facilities

Laboratory for Molecular Immunology, Croatian Institute for Brain Research within ZUSM, consists of two laboratory rooms and a cabinet. The first laboratory is used for general laboratory procedures, histological techniques, ELISA, Western blot, cell labeling and protein extraction. The second laboratory is used for cell culture and gene expression analysis. Rooms are equipped with CO2 incubators (Heracell 150, Haereus), laminar flow chamber (Aura-V, Ehret), spectrophotometer (NanoDrop 1000, Thermo Scientific), cooling centrifuges (Hettich Universal 32R and Hettich Micro 120), vacuum centrifuge (Concentrator 5301, Eppendorf) and a real-time PCR system (SDS750, Applied Biosystems by LT). Rooms are also fully equipped with common small laboratory instruments. Cabinet contains a fluorescent invert microscope (Axiovert 200, Carl Zeiss), with camera and software for image analysis (AxioVision, Carl Zeiss), and with a large-filed microscope (AxioZoom V8, Carl Zeiss). Core facilities and capital equipment at the Institute include dark room, molecular core with electroporation based transfection system (Neon, Invitrogen by LT), two-laser flow-cytometer (Attune, Applied Biosystems by LT), ultracentrifuge, cryostat (Leica CM-3050S), ELISA reader (Model 680, Bio-Rad Laboratories), -80°C freezers, autoclave and precise balance. Animal Facility is certified for breeding and maintaining of the experimental mice, provides appropriate conditions to conduct AIA model, and includes laboratory for in vivo manipulation, cabinets for mice during follow-up, and other services for animal work.

Laboratory for flow cytometry and cell sorting at the Croatian Institute for Brain Research (established in 2017), equipped with cell sorter containing three lasers (FACSAria I, BDB), and FACS Calibur instrument with two lasers.

Laboratory for Microscopy and Morphometry, Department of Anatomy, ZUSM, is equipped for general histology with rotational microtome (Leica) and a fluorescent microscope (AxioImager, Carl Zeiss) with camera and software for image analysis and reconstruction (Osteomeasure, Ostoemtrix). Preclinical magnetic resonance technology (Preclinical MRI BioSpec 7T, Bruker; still under installation) and micro-CT instrument (SkyScan1076, SkyScan) are also placed at the Department of Anatomy.

Centre for functional genomics, within ZUSM, has all necessary equipment for Affymetrix hybridization and analysis, and sequencing.

REZLTATI PROJEKTA

1st Year

Sažetak: Elvira Lazic Mosler, Darja Flegar, Alan Sucur, Martina Fadljevic, Tomislav Kelava, Antonio Markotic, Sanja Ivcevic, Katerina Zrinski-Petrovic, Vedran Katavic, Danka Grcevic & Natasa Kovacic. Identifying cell populations coupling inflammation to osteoresorption in arthritis. Bone Abstracts (2016) 5 P10

Identifying cell populations coupling inflammation to osteoresorption in arthritis

Sažetak: Martina Fadljević, Nina Lukač, Igor Radanović, Elvira Lazić Mosler, Darja Flegar, Alan Šućur, Tomislav Kelava, Vedran Katavić, Danka Grčević, Nataša Kovačić. Alterations in the synovial cellular composition associated with osteoresorption in antigen-induced arthritis. Annual Meeting Of The Croatian Immunological Society abstract book. 2016.

HID2016

2nd Year

Poster: Croatian Student Summit 13, Medicinski fakultet Sveučilišta u Zagrebu, Martina Fadljević i Igor Radanović.

CROSS

Rektorova nagrada: Martina Fadljević i Igor Radanović, Procjena učinka imunizacije na generalizirani i subkondralni gubitak kosti u tijeku artritisa potaknutog antigenom.

Rektorova nagrada

Sažetak: Igor Radanović, Martina Fadljević, Nina Lukač, Vedran Katavić, Nataša Kovačić. Assessment of the systemic and local bone loss in the model of antigen induced arthritis. Annual Meeting Of The Croatian Immunological Society with EFIS on Tour abstract book. 2017.

LINK

Poster: Igor Radanović, Martina Fadljević, Nina Lukač, Vedran Katavić, Nataša Kovačić. Assessment of the systemic and local bone loss in the model of antigen induced arthritis. Annual Meeting Of The Croatian Immunological Society with EFIS on Tour poster. 2017.

HID2017

Usmeno izlaganje: Nina Lukač, Uloga populacija sinovijalnih stanica u upalnom oštećenju subkondralne kosti u artritisu. Memorijal akademika Draga Perovića i akademkinje Jelene Krmpotić‐Nemanić. Medicinski fakultet Sveučilišta u Zagrebu.

Uloga populacija sinovijalnih stanica u upalnom oštećenju subkondralne kosti u artritisu

Pozvano predavanje: Nives Zimmerman. The life and death of eosinophils. Medicinski fakultet Sveučilišta u Zagrebu.

The life and death of eosinophils

Publikacija: Sucur A, Jajic Z, Artukovic M, Ikic Matijasevic M, Anic B, Flegar D, i sur. Chemokine signals are crucial for enhanced homing and differentiation of circulating osteoclast progenitor cells. Arthritis Res Ther. 2017;19(1):142.

Chemokine signals are crucial for enhanced homing and differentiation of circulating osteoclast progenitor cells

3rd Year

PUBLIKACIJA: Elvira Lazić Mosler, Nina Lukač, Darja Flegar, Martina Fadljević, Igor Radanović, Hrvoje Cvija, Tomislav Kelava, Sanja Ivčević, Alan Šućur, Antonio Markotić, Vedran Katavić, Ana Marušić, Danka Grčević, Nataša Kovačić. Fas receptor induces apoptosis of synovial bone and cartilage progenitor populations and promotes bone loss in antigen-induced arthritis. The FASEB Journal. Prihvaćeno za objavu u listopadu 2018. (Manuscript No. 201801426)

FASEB

PUBLIKACIJA: Grčević D, Sironi M, Valentino S, Deban L, Cvija H, Inforzato A, Kovačić N, Katavić V, Kelava T, Kalajzić I, Mantovani A, Bottazzi B. The Long Pentraxin 3 Plays a Role in Bone Turnover and Repair. Front Immunol. 2018 Mar 5;9:417.

PTX3

POZVANO PREDAVANJE: Nataša Kovačić. Cellular and molecular targets of Fas-dependent inflammation and bone resorption in arthritis. 2018 Annual Meeting of the Croatian Immunological Society. Str. 17.

HID2018

SAŽETAK I POSTER:  N. Lukač, M. Fadljevic, I. Radanovic, E. Lazic Mosler, A. Sucur, D. Flegar, T. Kelava, V. Katavic, D. Grcevic, N. Kovacic. Myeloid-specific molecular mediators of subchondral bone damage in antigen-induced arthritis. 5th European Congress of Immunology Abstract Book. 2018:326. (P.C1.08.11)

ECI2018
ECInina

SAŽETAK I POSTER: K. Barbaric Starcevic, N. Lukac, M. Jelic, A. Sucur, D. Grcevic, N. Kovacic. Synovial osteoprogenitor phenotype in patients with rheumatoid arthritis. 5th European Congress of Immunology Abstract Book. 2018:413. (P.C6.02.13)

ECI2018
ECIbarbara

SAŽETAK: Kottyan LC, Maddox A, Hoffman K, Ray L, Forney C, Eby M, Mingler MK, Kaufman K, Dexheimer P, Lukac N, Caldwell JM, Rochman M, Shoda T, Wen T, Wheeler JC, Hogan SP, Mukkada VA, Putnam PE, Abonia PP, Genta RM, Martin LJ, Dellon ES, Rothenberg ME. X chromosomal linkage to eosinophilic esophagitis susceptibility. J Allergy Clin Immunol. 2018;141:AB225.

JACI

SAŽETAK I POSTER: K. Barbarić Starčević, N. Lukač, M.Jelić, A. Šućur, D. Grčević, N. Kovačić. Synovial osteoprogenitor phenotype in patients with rheumatoid arthritis. Annual Meeting of the Croatian Immunological Society abstract book. 2018:40

HID2018

SAŽETAK I POSTER: N. Lukač, M. Fadljević, I. Radanović, A. Šućur, D. Flegar, T. Kelava, K. Zrinski Petrović, V. Katavić, D. Grčević, N. Kovačić. Transcriptional profiling of synovial myeloid cells to determine molecular mediators of bone resorption in antigen–induced arthritis. Annual Meeting of the Croatian Immunological Society abstract book. 2018:52.

HID2018

DAN DOKTORATA: Nina Lukač. Myeloid-specific molecular mediators of subchondral bone damage in antigen-induced arthritis. 2018:15

DanD2018
DanDnina

4th Year

USMENO IZLAGANJE: N. Lukač. Transcriptome analysis of small synovial stromal populations in a murine model of rheumatoid arthritis. Seminar “Agilent Genomics Solutions: getting the optimal results from your samples”, Zagreb, 22.10.

Aglient

PUBLIKACIJA: A. Inforzato, R. Parente, C. Sobacchi, B. Bottazzi, A. Mantovani, D. Grcevic. The long pentraxin PTX3 in bone homeostasis and pathology. Frontiers in Immunology. 2019.

DankaFrontiers

PUBLIKACIJA: A. Sucur, Z. Jajic, M. Ikic Matijasevic, A. Stipic Markovic, D. Flegar, N. Lukac, T. Kelava, N. Kovacic, D. Grcevic. Combined manual and automated immunophenotypisation identified disease-specific immune subpopulations in rheumatoid arthritis, ankylosing spondylitis and psoriatic arthritis. Clin Exp Rheumatol. 2019.

SucurCER

SAŽETAK I POSTER: N. Lukač. Ubiquitin E3 ligase Mid1 mediates subchondral bone resorption in a mouse model of rheumatoid arthritis. MefZg PhD day 2019 – Abstract book. 2019:12

NinaPhD2019poster
NinaPhD2019abstractbook

SAŽETAK, POSTER I USMENO IZLAGANJE: N. Lukač, A. Šućur, D. Flegar, T. Kelava, K. Zrinski Petrović, D. Šisl, V. Katavić, D. Grčević, N. Kovačić. Mid1 is a novel mediator of subchondral bone resorption in antigen-induced arthritis. ECTS 2019. Calcif Tissue Int 2019:S39

NinaECT2019ppt
NinaECT2019poster
NinaECT2019abstract

SAŽETAK I POSTER: A. Sucur, Z. Jajic, M. Ikic Matijasevic, M. Artukovic, D. Flegar, N. Lukac, T. Kelava, A. Markotic, N. Kovacic, D. Grcevic. Association of T-cell and B-cell aberrancies with disease activity in rheumatoid arthritis, psoriatic arthritis and ankylosing spondylitis. ECTS 2019. Calcif Tissue Int 2019:S113

AlanECTS2019poster
AlanECTS2019abstract

SAŽETAK: N. Lukač, D. Flegar, T. Kelava, A. Šućur, K. Zrinski Petrović, D. Šisl, M. Filipović, V. Katavić, D. Grčević, N. Kovačić. The role of Mid1-PP2A axis in inflammation-induced joint damage in antigen-induced arthritis. 2019 Annual Meeting of the Croatian Immunological Society abstract book. 2019:52

NinaHID2019abstract

SAŽETAK I USMENO IZLAGANJE: D. Flegar, A. Šućur, A. Markotić, M. Filipović, N. Lukač, D. Šisl, N. Kovačić, T. Kelava, V. Katavić, K. Zrinski Petrović, D. Grčević. The effect of CCL2/CCR2 signaling blocade on bone resorption and osteoclast progenitors in collagen induced arthritis. 2019 Annual Meeting of the Croatian Immunological Society abstract book. 2019:24

DarjaHID2019abstract

SAŽETAK: A. Špehar Uroić, M. Filipović, D. Flegar, A. Šućur, N. Kovačić, D. Grčević. Immunophenotyping of chemokine receptors on peripheral blood mononuclear cells in children with type 1 diabetes. 2019 Annual Meeting of the Croatian Immunological Society abstract book. 2019:51

MasaHID2019abstract

SAŽETAK I USMENO IZLAGANJE: D. Flegar, A. Šućur, A. Markotić, N. Lukač, D. Šisl, N. Kovačić, T. Kelava, V. Katavić, K. Zrinski Petrović, D. Grčević. The effect of inhibition of CCL2/CCR2 signaling on myeloid lineage cells and osteoclast progenitor subpopulation in collagen induced arthritis.  5th International Conferenceof translational medicineon pathogenesis and therapy of immunomediated diseases (2019, Milano)

DarjaSIICA2019poster

SAŽETAK I USMENO IZLAGANJE: N. Lukač, A. Šućur, D. Flegar, T. Kelava, K. Zrinski Petrović, V. Katavić, D. Grčević, N. Kovačić. Mijeloidno-specifični molekularni posrednici subhondralnog koštanog razaranja u mišjem modelu reumatoidnog artritisa. 1. Simpozij Perović – Krmpotić Nemanić: Knjiga sažetaka. 2019:12

NinaKrmpotic2019poster
NinaKrmpotic2019abstract

SAŽETAK I USMENO IZLAGANJE: M. Fadljević, I. Radanović, N. Lukač, N. Kovačić. Povezanost imunizacijskog protokola i doze sa sustavnim i subkondralnim gubitkom kosti u tijeku artritisa potaknutog antigenom. 1. Simpozij Perović – Krmpotić Nemanić: Knjiga sažetaka. 2019:11

MartinaKrmpotic2019posterr
MartinaKrmpotic2019abstract