Network


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

Hotspot


Dive into the research topics where Jörg Hamann is active.

Publication


Featured researches published by Jörg Hamann.


Blood | 2013

Genetic programs expressed in resting and IL-4 alternatively activated mouse and human macrophages: similarities and differences.

Fernando O. Martinez; Laura Helming; Ronny Milde; Audrey Varin; Barbro N. Melgert; Christina Draijer; Benjamin Thomas; Marco Fabbri; Anjali Crawshaw; Ling-Pei Ho; Nick H. T. ten Hacken; Viviana Cobos Jiménez; Neeltje A. Kootstra; Jörg Hamann; David R. Greaves; Massimo Locati; Alberto Mantovani; Siamon Gordon

The molecular repertoire of macrophages in health and disease can provide novel biomarkers for diagnosis, prognosis, and treatment. Th2-IL-4–activated macrophages (M2) have been associated with important diseases in mice, yet no specific markers are available for their detection in human tissues. Although mouse models are widely used for macrophage research, translation to the human can be problematic and the human macrophage system remains poorly described. In the present study, we analyzed and compared the transcriptome and proteome of human and murine macrophages under resting conditions (M0) and after IL-4 activation (M2). We provide a resource for tools enabling macrophage detection in human tissues by identifying a set of 87 macrophage-related genes. Furthermore, we extend current understanding of M2 activation in different species and identify Transglutaminase 2 as a conserved M2 marker that is highly expressed by human macrophages and monocytes in the prototypic Th2 pathology asthma.


Journal of Immunological Methods | 2012

Systematic validation of specific phenotypic markers for in vitro polarized human macrophages

Carmen Ambarus; Sarah Krausz; M. van der Eijk; Jörg Hamann; Timothy R. D. J. Radstake; Kris A. Reedquist; Paul P. Tak; Dominique Baeten

BACKGROUND Polarization of macrophages by specific micro-environmental conditions impacts upon their function following subsequent activation. This study aimed to systematically validate robust phenotypic markers for in vitro polarized human macrophages in order to facilitate the study of macrophage subsets in vivo. METHODS Human peripheral blood monocytes were polarized in vitro with IFN-γ, IL-4, or IL-10. Similar experiments were performed with TNF, IL-13, dexamethasone, M-CSF and GM-CSF as polarizing stimuli. Phenotypic markers were assessed by flow cytometry and qPCR. RESULTS IFN-γ polarized macrophages (MΦ(IFN-γ)) specifically enhanced membrane expression of CD80 and CD64, IL-4 polarized macrophages (MΦ(IL-4)) mainly upregulated CD200R and CD206, and downregulated CD14 levels, and IL-10 polarized macrophages (MΦ(IL-10)) selectively induced CD163, CD16, and CD32. The expression profiles of the most specific markers were confirmed by qPCR, dose-response experiments, and the use of alternative polarizing factors for each macrophage subset (TNF, IL-13, and dexamethasone, respectively). GM-CSF polarized macrophages (MΦ(GM-CSF)) upregulated CD80 but not CD64 expression, showing a partial phenotypic similarity with MΦ(IFN-γ), and also upregulated the expression of the alternative activation marker CD206. M-CSF polarized macrophages (MΦ(M-CSF)) not only expressed increased levels of CD163 and CD16, resembling MΦ(IL-10,) but also displayed high levels of CD64. The phenotype of MΦ(M-CSF) could be further modulated by additional polarization with IFN-γ, IL-4, or IL-10, whereas MΦ(GM-CSF) showed less phenotypic plasticity. CONCLUSION This study validated CD80 as the most robust phenotypic marker for human MΦ(IFN-γ), whereas CD200R was upregulated and CD14 was specifically downregulated on MΦ(IL-4). CD163 and CD16 were found to be specific markers for MΦ(IL-10). The GM-CSF/M-CSF differentiation model showed only a partial phenotypic similarity with the IFN-γ/IL-4/IL-10 induced polarization.


Science Signaling | 2013

Sticky Signaling—Adhesion Class G Protein–Coupled Receptors Take the Stage

Tobias Langenhan; Gabriela Aust; Jörg Hamann

Emerging insight on the structural and functional versatility of Adhesion-GPCRs draws a fascinating picture of the multiple signals these molecules transmit. Gloss Adhesion-type heterotrimeric guanine nucleotide–binding protein (G protein)–coupled receptors (Adhesion-GPCRs) comprise a class of noncanonical seven-transmembrane spanning (7TM) receptors present on almost any cell type. Despite their abundance, remarkable size, and intriguing two-unit structure facilitating cell-cell and cell-matrix contacts in various organ systems, Adhesion-GPCRs are by far the most poorly understood 7TM receptors. Data indicate that a juxtamembrane GPCR autoproteolysis-inducing domain containing the GPCR proteolysis site (GPS) figures prominently in the biological function of Adhesion-GPCRs. This review, which has five figures, five tables, and 216 references, discusses the currently available data regarding the major structural elements of Adhesion-GPCRs in the context of receptor function and signaling. Adhesion-type heterotrimeric guanine nucleotide–binding protein (G protein)–coupled receptors (Adhesion-GPCRs) comprise a class of widely distributed seven-transmembrane spanning (7TM) receptors with unusual layout and properties. The key to understanding the function of Adhesion-GPCRs appears to be their hybrid architecture: They have an extracellular domain containing an extended array of protein folds fit for interactions, and structural elements of GPCRs with a 7TM and an intracellular domain. If and how these distinct protein portions interact is currently under intense investigation. Intriguingly, all Adhesion-GPCRs have a juxtamembrane GPCR autoproteolysis-inducing domain that, in many homologs, facilitates the autocatalytic processing into an N-terminal fragment (NTF) and a C-terminal fragment (CTF), which subsequently remain attached at the cell surface. The NTF provides the ability for combinatorial engagement with cellular or matrix-associated molecules facilitating cell adhesion, orientation, and positioning during development, immune responses, and tumor growth. The CTF, like in canonical GPCRs, initiates interactions with different types of signaling molecules, including heterotrimeric G proteins, small guanosine triphosphatases, and transmembrane protein partners, yet the agonistic potential of most known Adhesion-GPCR ligands is uncertain. Studies with truncated receptors suggest that the NTF and CTF of Adhesion-GPCRs may function as autonomous adhesive and signaling units, respectively, but other studies in nonvertebrates demonstrating NTF-CTF interplay challenge this view. We discuss the available data concerning the main structural elements of Adhesion-GPCRs in the context of receptor function and signaling.


Molecular Immunology | 2010

Serine proteases of the human immune system in health and disease.

Kirstin M. Heutinck; Ineke J. M. ten Berge; C. Erik Hack; Jörg Hamann; Ajda T. Rowshani

Serine proteases form a large family of protein-cleaving enzymes that play an essential role in processes like blood coagulation, apoptosis and inflammation. Immune cells express a wide variety of serine proteases such as granzymes in cytotoxic lymphocytes, neutrophil elastase, cathepsin G and proteinase 3 in neutrophils and chymase and tryptase in mast cells. Regulation of proteolysis induced by these serine proteases is essential to prevent self-induced damage. Hence, there are specialized serine protease inhibitors, serpins, which are broadly distributed. Here, we discuss the function of human serine proteases in inflammation, apoptosis and tissue remodeling. Furthermore, we address their impact on development and progression of immune mediated-diseases. Understanding the mode of action of serine proteases will help to unravel molecular processes involved in immunological disorders and will facilitate the identification of new therapeutic targets.


Journal of Immunology | 2008

CD27 Defines Phenotypically and Functionally Different Human NK Cell Subsets

Mireille T. M. Vossen; Mourad Matmati; Kirsten M. L. Hertoghs; Paul A. Baars; Mi-Ran Gent; Georges Leclercq; Jörg Hamann; Taco W. Kuijpers; René A. W. van Lier

The absence of the TNF-receptor family member CD27 marks the stable acquisition of cytolytic effector functions by both CD4+ and CD8+ T cells. We found that the majority of circulating human NK cells was CD27−. These cells were largely CD56dim, contained high levels of perforin and granzyme B, and were able to exert strong cytotoxic activity. In contrast, circulating CD27+ NK cells were mostly CD56dim/bright, had significant lower levels of perforin and granzyme B, and had a low cytolytic potential. Primary and secondary lymphoid organs were markedly enriched for CD27+ NK cells. When correlating the expression of CD27 to recently defined developmental stages of NK cells in tonsil, we observed that CD27 was exclusively found on mature CD94+, stage 4 NK cells. On these cells, regulation of CD27 expression appeared to be controlled by the common γ-chain cytokine IL-15, and down-regulation of CD27 was specifically induced by its ligand, CD70. Thus, the absence of CD27 expression allows the definition of cytotoxic effector cells within the known mature NK cell subsets in humans.


Arthritis & Rheumatism | 1999

Expression of the activation antigen CD97 and its ligand CD55 in rheumatoid synovial tissue

Jörg Hamann; Jérôme O. Wishaupt; René A. W. van Lier; Tom J. M. Smeets; Ferdinand C. Breedveld; Paul P. Tak

OBJECTIVE Fibroblast-like synoviocytes (FLS) express decay-accelerating factor (CD55) at high levels. Recently, it was found that CD55 is a specific cellular ligand for the 7-span transmembrane receptor CD97. The objective of this study was to define the expression of this receptor-ligand pair in synovial tissue (ST) to provide more insight into the interaction between FLS and surrounding cells. METHODS Antibodies against CD97 and CD55 were used for immunohistologic analysis of synovial biopsy specimens from 16 patients with rheumatoid arthritis (RA) and 15 patients with osteoarthritis (OA). In addition, an enzyme-linked immunosorbent assay system was used to determine the expression of soluble CD97 (sCD97) in synovial fluid (SF) from 30 patients with RA, 13 with OA, and 10 with reactive arthritis (ReA). RESULTS In both RA and OA ST sections, strong expression of CD55 was confirmed on FLS in the intimal lining layer, where it was also found that all macrophages expressed CD97. The percentage of macrophages that expressed CD97 was lower in the synovial sublining (P = 0.005). The mean levels of sCD97 in SF were significantly higher in RA patients than in patients with OA or ReA (P < 0.0001). CONCLUSION These results suggest that FLS are able to interact with macrophages via the CD97/CD55 receptor-ligand system. In this respect, the CD97/CD55 pair may account for the specific architecture of the intimal lining layer and may be of primary importance in maintaining and amplifying synovial inflammation. The specific increase in sCD97 levels in RA SF might be related to the presence of activated proteolytic systems or to the increase in synovial mass, rather than a consequence of local receptor-ligand interaction.


Pharmacological Reviews | 2015

International Union of Basic and Clinical Pharmacology. XCIV. Adhesion G Protein–Coupled Receptors

Jörg Hamann; Gabriela Aust; Demet Araç; Felix B. Engel; Caroline J. Formstone; Robert Fredriksson; Randy A. Hall; Breanne L. Harty; Christiane Kirchhoff; Barbara Knapp; Arunkumar Krishnan; Ines Liebscher; Hsi-Hsien Lin; David C. Martinelli; Kelly R. Monk; Miriam C. Peeters; Xianhua Piao; Simone Prömel; Torsten Schöneberg; Thue W. Schwartz; Kathleen Singer; Martin Stacey; Yuri A. Ushkaryov; Mario Vallon; Uwe Wolfrum; Mathew W. Wright; Lei Xu; Tobias Langenhan; Helgi B. Schiöth

The Adhesion family forms a large branch of the pharmacologically important superfamily of G protein–coupled receptors (GPCRs). As Adhesion GPCRs increasingly receive attention from a wide spectrum of biomedical fields, the Adhesion GPCR Consortium, together with the International Union of Basic and Clinical Pharmacology Committee on Receptor Nomenclature and Drug Classification, proposes a unified nomenclature for Adhesion GPCRs. The new names have ADGR as common dominator followed by a letter and a number to denote each subfamily and subtype, respectively. The new names, with old and alternative names within parentheses, are: ADGRA1 (GPR123), ADGRA2 (GPR124), ADGRA3 (GPR125), ADGRB1 (BAI1), ADGRB2 (BAI2), ADGRB3 (BAI3), ADGRC1 (CELSR1), ADGRC2 (CELSR2), ADGRC3 (CELSR3), ADGRD1 (GPR133), ADGRD2 (GPR144), ADGRE1 (EMR1, F4/80), ADGRE2 (EMR2), ADGRE3 (EMR3), ADGRE4 (EMR4), ADGRE5 (CD97), ADGRF1 (GPR110), ADGRF2 (GPR111), ADGRF3 (GPR113), ADGRF4 (GPR115), ADGRF5 (GPR116, Ig-Hepta), ADGRG1 (GPR56), ADGRG2 (GPR64, HE6), ADGRG3 (GPR97), ADGRG4 (GPR112), ADGRG5 (GPR114), ADGRG6 (GPR126), ADGRG7 (GPR128), ADGRL1 (latrophilin-1, CIRL-1, CL1), ADGRL2 (latrophilin-2, CIRL-2, CL2), ADGRL3 (latrophilin-3, CIRL-3, CL3), ADGRL4 (ELTD1, ETL), and ADGRV1 (VLGR1, GPR98). This review covers all major biologic aspects of Adhesion GPCRs, including evolutionary origins, interaction partners, signaling, expression, physiologic functions, and therapeutic potential.


Journal of Immunology | 2004

The Epidermal Growth Factor-Seven Transmembrane (EGF-TM7) Receptor CD97 Is Required for Neutrophil Migration and Host Defense

Jaklien C. Leemans; Anje A. te Velde; Sandrine Florquin; Roelof J. Bennink; Kora de Bruin; René A. W. van Lier; Tom van der Poll; Jörg Hamann

The epidermal growth factor-seven transmembrane (EGF-TM7) family is a group of seven-span transmembrane receptors predominantly expressed by cells of the immune system. Family members CD97, EGF module-containing mucin-like receptor (EMR) 1, EMR2, EMR3, EMR4, and EGF-TM7-latrophilin-related protein are characterized by an extended extracellular region with a variable number of N-terminal EGF-like domains. EGF-TM7 receptors bind cellular ligands as demonstrated by the interaction of CD97 with decay accelerating factor (CD55) and dermatan sulfate. Investigating the effect of newly generated mAb on the migration of neutrophilic granulocytes, we here report for the first time in vivo data on the function of CD97. In dextran sulfate sodium-induced experimental colitis, we show that homing of adoptively transferred neutrophils to the colon was significantly delayed when cells were preincubated with CD97 mAb. The consequences of this defect in neutrophil migration for host defense are demonstrated in a murine model of Streptococcus pneumoniae-induced pneumonia. Mice treated with CD97 mAb to EGF domain 1 (1B2) and EGF domain 3 (1C5) displayed a reduced granulocytic inflammatory infiltrate at 20 h after inoculation. This was associated with a significantly enhanced outgrowth of bacteria in the lungs at 44 h and a strongly diminished survival. Together, these findings indicate an essential role for CD97 in the migration of neutrophils.


European Journal of Immunology | 1998

Characterization of the CD55 (DAF)-binding site on the seven-span transmembrane receptor CD97

Jörg Hamann; Catalijne Stortelers; Endre Kiss-Toth; Björn Vogel; Wolfram Eichler; René A. W. van Lier

CD97 is an activation‐induced antigen on leukocytes which belongs to a new group of seven‐span transmembrane (7‐TM) molecules, designated EGF‐TM7 family. Family members, including EMR1 and F4/80, are characterized by an extended extracellular region with several N‐terminal epidermal growth factor‐like (EGF) domains. Alternative splicing of CD97 results in isoforms possessing either three (EGF1, 2, 5), four (EGF1, 2, 3, 5) or five EGF domains (EGF1, 2, 3, 4, 5). We recently identified decay accelerating factor (DAF, CD55), a regulatory protein of the complement cascade, as a cellular ligand of the smallest isoform. Employing mutants of CD97(EGF1, 2, 5) in which the EGF domains have been systematically deleted, we here demonstrate the necessity of at least three tandemly linked EGF domains for the interaction with CD55. Consistent with the involvement of different EGF domains, monoclonal antibodies directed against the first EGF domain as well as the removal of Ca2+ , for which binding sites exist in the second and fifth EGF domain, blocked binding to CD55. Compared to CD97(EGF1, 2,5) the larger isoforms CD97(EGF1, 2, 3, 5) and CD97(EGF1, 2, 3, 4, 5) have a significantly lower affinity for CD55. Thus, alternative splicing may regulate the ligand specificity of CD97 and probably other members of the EGF‐TM7 family.


American Journal of Pathology | 2002

Expression and Regulation of CD97 in Colorectal Carcinoma Cell Lines and Tumor Tissues

Matthias Steinert; Manja Wobus; Carsten Boltze; Alexander Schütz; Mandy Wahlbuhl; Jörg Hamann; Gabriela Aust

The expression of CD97, a member of the EGF-TM7 family with adhesive properties, is proportional to the aggressiveness and lymph node involvement in thyroid tumors. CD97 has never been systematically investigated in other tumors. First, we examined colorectal carcinoma cell lines (n = 18) for CD97 expression and regulation. All cell lines were CD97-positive. The level of CD97 in each line correlated with migration and invasion in vitro. This result was confirmed in CD97-inducible Tet-off HT1080 cells. Transforming growth factor-beta, which inhibits proliferation in transforming growth factor-beta-sensitive LS513 and LS1034 cells, down-regulated CD97 in these cell lines. Examining CD97 during sodium butyrate-induced cell differentiation of Caco-2 cells, we could demonstrate a CD97-decreasing effect. Second, we screened 81 colorectal adenocarcinomas by immunohistology for expression of CD97. Normal colorectal epithelium is CD97-negative. Seventy-five of 81 of the carcinomas expressed CD97. The strongest staining for CD97 occurred in scattered tumor cells at the invasion front compared to cells located within solid tumor formations of the same tumor. Carcinomas with more strongly CD97-stained scattered tumor cells showed a poorer clinical stage as well as increased lymph vessel invasion compared to cases with uniform CD97 staining. In summary, CD97 expression correlates with dedifferentiation, migration, and invasion in colorectal tumor cell lines. Moreover, more strongly CD97-stained tumor cells at the invasion front of colorectal carcinomas indicate the involvement of the molecule in tumor migration and invasion.

Collaboration


Dive into the Jörg Hamann's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Paul P. Tak

University of 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

Robert M. Hoek

Netherlands Institute for Neuroscience

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge