Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Paola Casarosa is active.

Publication


Featured researches published by Paola Casarosa.


Journal of Virology | 2002

Kaposi's sarcoma-associated herpesvirus-encoded G protein-coupled receptor ORF74 constitutively activates p44/p42 MAPK and Akt via G(i) and phospholipase C-dependent signaling pathways

Martine J. Smit; Dennis Verzijl; Paola Casarosa; Marjon Navis; Henk Timmerman; Rob Leurs

ABSTRACT The G protein-coupled receptor encoded by Kaposis sarcoma-associated herpesvirus, also referred to as ORF74, has been shown to stimulate oncogenic and angiogenic signaling pathways in a constitutively active manner. The biochemical routes linking ORF74 to these signaling pathways are poorly defined. In this study, we show that ORF74 constitutively activates p44/p42 mitogen-activated protein kinase (MAPK) and Akt via Gi- and phospholipase C (PLC)-mediated signaling pathways. Activation of Akt by ORF74 appears to be phosphatidylinositol 3-kinase (PI3-K) dependent but, interestingly, is also mediated by activation of protein kinase C (PKC) and p44/p42 MAPK. ORF74 may signal to Akt via p44/p42 MAPK, which can be activated by Gi, through activation of PI3-K or through PKC via the PLC pathway. Signaling of ORF74 to these proliferative and antiapoptotic signaling pathways can be further modulated positively by growth-related oncogene (GROα/CXCL1) and negatively by human gamma interferon-inducible protein 10 (IP-10/CXCL10), thus acting as an agonist and an inverse agonist, respectively. Despite the ability of the cytomegalovirus-encoded chemokine receptor US28 to constitutively activate PLC, this receptor does not increase phosphorylation of p44/p42 MAPK or Akt in COS-7 cells. Hence, ORF74 appears to signal through a larger diversity of G proteins than US28, allowing it to couple to proliferative and antiapoptotic signaling pathways. ORF74 can therefore be envisioned as an attractive target for novel treatment of Kaposis sarcoma.


Journal of Biological Chemistry | 2003

Human cytomegalovirus chemokine receptor US28-induced smooth muscle cell migration is mediated by focal adhesion kinase and Src

Daniel N. Streblow; Jennifer Vomaske; Patsy Smith; Ryan Melnychuk; Laurel Hall; Dora Pancheva; Martine Smit; Paola Casarosa; David D. Schlaepfer; Jay A. Nelson

The human cytomegalovirus-encoded chemokine receptor US28 induces arterial smooth muscle cell (SMC) migration; however, the underlying mechanisms involved in this process are unclear. We have previously shown that US28-mediated SMC migration occurs by a ligand-dependent process that is sensitive to protein-tyrosine kinase inhibitors. We demonstrate here that US28 signals through the non-receptor protein-tyrosine kinases Src and focal adhesion kinase (FAK) and that this activity is necessary for US28-mediated SMC migration. In the presence of RANTES (regulated on activation normal T cell expressed and secreted), US28 stimulates the production of a FAK·Src kinase complex. Interestingly, Src co-immunoprecipitates with US28 in a ligand-dependent manner. This association occurs earlier than the formation of the FAK·Src kinase complex, suggesting that US28 activates Src before FAK. US28 binding to RANTES also promotes the formation of a Grb2·FAK complex, which is sensitive to treatment with the Src inhibitor PP2, further highlighting the critical role of Src in US28 activation of FAK. Human cytomegalovirus US28-mediated SMC migration is inhibited by treatment with PP2 and through the expression of either of two dominant negative inhibitors of FAK (F397Y and NH2-terminal amino acids 1–401). These findings demonstrate that activation of FAK and Src plays a critical role in US28-mediated signaling and SMC migration.


Journal of Biological Chemistry | 2003

Identification of the First Nonpeptidergic Inverse Agonist for a Constitutively Active Viral-encoded G Protein-coupled Receptor

Paola Casarosa; Wiro M. P. B. Menge; Rosalba Minisini; Claas Otto; Jane van Heteren; Aldo Jongejan; Henk Timmerman; Barbara Moepps; Frank Kirchhoff; Thomas Mertens; Martine J. Smit; Rob Leurs

Human cytomegalovirus (HCMV) encodes a G protein-coupled receptor (GPCR), named US28, which shows homology to chemokine receptors and binds several chemokines with high affinity. US28 induces migration of smooth muscle cells, a feature essential for the development of atherosclerosis, and may serve as a co-receptor for human immunodeficiency virus-type 1 entry into cells. Previously, we have shown that HCMV-encoded US28 displays constitutive activity, whereas its mammalian homologs do not. In this study we have identified a small nonpeptidergic molecule (VUF2274) that inhibits US28-mediated phospholipase C activation in transiently transfected COS-7 cells and in HCMV-infected fibroblasts. Moreover, VUF2274 inhibits US28-mediated HIV entry into cells. In addition, VUF2274 fully displaces radiolabeled RANTES (regulated on activation normal T cell expressed and secreted) binding at US28, apparently with a noncompetitive behavior. Different analogues of VUF2274 have been synthesized and pharmacologically characterized, to understand which features are important for its inverse agonistic activity. Finally, by means of mutational analysis of US28, we have identified a glutamic acid in transmembrane 7 (TM 7), which is highly conserved among chemokine receptors, as a critical residue for VUF2274 binding to US28. The identification of a full inverse agonist provides an important tool to investigate the relevance of US28 constitutive activity in viral pathogenesis.


Journal of Virology | 2002

The Rat Cytomegalovirus R33-Encoded G Protein-Coupled Receptor Signals in a Constitutive Fashion

Yvonne K. Gruijthuijsen; Paola Casarosa; Suzanne J.F. Kaptein; Jos L. V. Broers; Rob Leurs; Cathrien A. Bruggeman; Martine J. Smit; Cornelis Vink

ABSTRACT The rat cytomegalovirus (RCMV) R33 gene is conserved among all betaherpesviruses and encodes a protein (pR33) that shows sequence similarity with chemokine-binding G protein-coupled receptors (GPCRs). Previously, the physiological significance of the R33 gene was demonstrated by the finding that an RCMV strain with R33 deleted is severely attenuated in vivo and is unable to either enter or replicate in the salivary glands of infected rats. Here, we report that RCMV pR33 is expressed as a functional GPCR that signals in an agonist-independent manner in both COS-7 and Rat2 cells. Transient expression of pR33 in COS-7 cells results in constitutive activation of phospholipase C (PLC) due to coupling to G proteins of the Gq class. Interestingly, PLC activation is partially inhibited by cotransfection with Gα-transducin subunits, which indicates the involvement of Gβγ as well as Gα subunits in pR33-mediated signaling. Surprisingly, PLC activation is also partially inhibited by addition of pertussis toxin (PTX), suggesting that pR33 activates not only Gq but also Gi/0 proteins. The constitutive activation of Gi/0 proteins by pR33 is further demonstrated by the PTX-sensitive decrease of CRE-mediated transcription and the PTX-sensitive increase of both NF-κB- and SRE-mediated transcription. In contrast to its homolog of human herpesvirus 6B (pU12), pR33 does not bind RANTES.


FEBS Journal | 2005

The human cytomegalovirus-encoded chemokine receptor US28 induces caspase-dependent apoptosis

Olivier Pleskoff; Paola Casarosa; L. Verneuil; Fadela Aïnoun; Patrick S. Beisser; Martine J. Smit; Rob Leurs; Pascal Schneider; Susan Michelson; Jean Claude Ameisen

Viral subversion of apoptosis regulation plays an important role in the outcome of host/virus interactions. Although human cytomegalovirus (HCMV) encodes several immediate early (IE) antiapoptotic proteins (IE1, IE2, vMIA and vICA), no proapoptotic HCMV protein has yet been identified. Here we show that US28, a functional IE HCMV‐encoded chemokine receptor, which may be involved in both viral dissemination and immune evasion, constitutively induces apoptosis in several cell types. In contrast, none of nine human cellular chemokine receptors, belonging to three different subfamilies, induced any significant level of apoptosis. US28‐induced cell death involves caspase 10 and caspase 8 activation, but does not depend on the engagement of cell‐surface death receptors of the tumour necrosis factor receptor/CD95 family. US28 cell‐death induction is prevented by coexpression of C‐FLIP, a protein that inhibits Fas‐associated death domain protein (FADD)‐mediated activation of caspase 10 and caspase 8, and by coexpression of the HCMV antiapoptotic protein IE1. The use of US28 mutants indicated that the DRY sequence of its third transmenbrane domain, required for constitutive G‐protein signalling, and the US28 intracellular terminal domain required for constitutive US28 endocytosis, are each partially required for cell‐death induction. Thus, in HCMV‐infected cells, US28 may function either as a chemokine receptor, a phospholipase C activator, or a proapoptotic factor, depending on expression levels of HCMV and/or cellular antiapoptotic proteins.


Journal of Biological Chemistry | 2004

Constitutively active Gq/11-coupled Receptors Enable Signaling by Co-expressed Gi/o-coupled Receptors

Remko A. Bakker; Paola Casarosa; Henk Timmerman; Martine J. Smit; Rob Leurs

Co-expression of guanine nucleotide-binding regulatory (G) protein-coupled receptors (GPCRs), such as the Gi/o-coupled human 5-hydroxytryptamine receptor 1B (5-HT1BR), with the Gq/11-coupled human histamine 1 receptor (H1R) results in an overall increase in agonist-independent signaling, which can be augmented by 5-HT1BR agonists and inhibited by a selective inverse 5-HT1BR agonist. Interestingly, inverse H1R agonists inhibit constitutively H1R-mediated as well as 5-HT1BR agonist-induced signaling in cells co-expressing both receptors. This phenomenon is not solely characteristic of 5-HT1BR; it is also evident with muscarinic M2 and adenosine A1 receptors and is mimicked by mastoparan-7, an activator of Gi/o proteins, or by over-expression of Gβγ subunits. Likewise, expression of the Gq/11-coupled human cytomegalovirus (HCMV)-encoded chemokine receptor US28 unmasks a functional coupling of Gi/o-coupled CCR1 receptors that is mediated via the constitutive activity of receptor US28. Consequently, constitutively active Gq/11-coupled receptors, such as the H1R and HCMV-encoded chemokine receptor US28, constitute a regulatory switch for signal transduction by Gi/o-coupled receptors, which may have profound implications in understanding the role of both constitutive GPCR activity and GPCR cross-talk in physiology as well as in the observed pathophysiology upon HCMV infection.


Journal of Immunology | 2009

Significance of N-Terminal Proteolysis of CCL14a to Activity on the Chemokine Receptors CCR1 and CCR5 and the Human Cytomegalovirus-Encoded Chemokine Receptor US28

Rudolf Richter; Paola Casarosa; Ludger Ständker; Jan Münch; Jean-Yves Springael; Saskia Nijmeijer; Wolf-Georg Forssmann; Henry F. Vischer; Jalal Vakili; Michel Detheux; Marc Parmentier; Rob Leurs; Martine J. Smit

The CC chemokine CCL14a is constitutively expressed in a large variety of tissues and its inactive proform CCL14a(1–74) circulates in high concentrations in plasma. CCL14a(1–74) is converted into CCL14a(9–74) by the proteases urokinase-type plasminogen activator and plasmin and is a highly active agonist for the chemokine receptors CCR1 and CCR5. In this study, a new CCL14a analog, CCL14a(12–74), was isolated from blood filtrate. To elucidate the functional role of the N terminus, a panel of N-terminally truncated CCL14a analogs were tested on the receptors CCR1 to CCR5 and on the human cytomegalovirus (HCMV)-encoded chemokine receptor US28. The rank order of binding affinity to these receptors and of the activation of CCR1 and CCR5-mediated intracellular Ca2+ concentration mobilization is CCL14a(6–74)<(7–74)<(8–74)≪(9–74) = (10–74)≫(11–74)≫(12–74). The almost identical affinities of CCL14a(7–74), CCL14a(9–74), and CCL14a(10–74) for the US28 receptor and the inhibition of US28-mediated HIV infection of 293T cells by all of the N-terminally truncated CCL14a analogs support the promiscuous nature of the viral chemokine receptor US28. In high concentrations, CCL14a(12–74) did reveal antagonistic activity on intracellular Ca2+ concentration mobilization in CCR1- and CCR5-transfected cells, which suggests that truncation of Tyr11 might be of significance for an efficient inactivation of CCL14a. A putative inactivation pathway of CCL14a(9–74) to CCL14a(12–74) may involve the dipeptidase CD26/dipeptidyl peptidase IV (DPPIV), which generates CCL14a(11–74), and the metalloprotease aminopeptidase N (CD13), which displays the capacity to generate CCL14a(12–74) from CCL14a(11–74). Our results suggest that the activity of CCL14a might be regulated by stringent proteolytic activation and inactivation steps.


International Congress Series | 2003

Modulation of constitutive GPCR activity: a way of life?

Rob Leurs; Remko A. Bakker; Paola Casarosa; Dennis Verzijl; Henk Timmerman; Martine J. Smit

G-protein coupled receptors (GPCRs) are highly versatile signalling modules and, as such, one of the most important drug targets in many therapeutic areas. In recent years, the view on GPCR activation has been firmly challenged and it is now clear that GPCRs do not always need external ligands to modulate intracellular signal transduction. The histamine H3 receptor shows a high level of constitutive activity both in vitro and in vivo. Constitutive H3 receptor activity might be modulated by biological (receptor isoforms) and chemical means (H3 receptor ligands). Furthermore, we recently found that constitutive activity seems to be a general characteristic of viral GPCRs, suggesting that the concept of constitutive GPCR activity is successfully exploited by a variety of pathogenic herpes viruses (e.g. HCMV and KSHV). As for most GPCRs, also for constitutively active viral GPCRs (endogenous) and synthetic ligands, can be found that shut down agonist-independent signalling, thereby acting as potential new anti-viral therapeutics.


Molecular Pharmacology | 2018

Identifying Functional Hotspot Residues for Biased Ligand Design in G-Protein-Coupled Receptors

Anita K. Nivedha; Christofer S. Tautermann; Sangbae Lee; Paola Casarosa; Ines Kollak; Tobias Kiechle; Nagarajan Vaidehi

G-protein-coupled receptors (GPCRs) mediate multiple signaling pathways in the cell, depending on the agonist that activates the receptor and multiple cellular factors. Agonists that show higher potency to specific signaling pathways over others are known as “biased agonists” and have been shown to have better therapeutic index. Although biased agonists are desirable, their design poses several challenges to date. The number of assays to identify biased agonists seems expensive and tedious. Therefore, computational methods that can reliably calculate the possible bias of various ligands ahead of experiments and provide guidance, will be both cost and time effective. In this work, using the mechanism of allosteric communication from the extracellular region to the intracellular transducer protein coupling region in GPCRs, we have developed a computational method to calculate ligand bias ahead of experiments. We have validated the method for several β-arrestin–biased agonists in β2-adrenergic receptor (β2AR), serotonin receptors 5-HT1B and 5-HT2B and for G-protein–biased agonists in the κ-opioid receptor. Using this computational method, we also performed a blind prediction followed by experimental testing and showed that the agonist carmoterol is β-arrestin–biased in β2AR. Additionally, we have identified amino acid residues in the biased agonist binding site in both β2AR and κ-opioid receptors that are involved in potentiating the ligand bias. We call these residues functional hotspots, and they can be used to derive pharmacophores to design biased agonists in GPCRs.


Journal of Biological Chemistry | 2001

Constitutive signaling of the Human Cytomegalovirus-encoded Chemokine receptor US28

Paola Casarosa; Remko A. Bakker; Dennis Verzijl; Marjon Navis; Henk Timmerman; Rob Leurs; Martine J. Smit

Collaboration


Dive into the Paola Casarosa's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Rob Leurs

VU University Amsterdam

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Jay A. Nelson

Scripps Research Institute

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge