Network


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

Hotspot


Dive into the research topics where Patrick Matthys is active.

Publication


Featured researches published by Patrick Matthys.


Nature Medicine | 2002

Revascularization of ischemic tissues by PlGF treatment, and inhibition of tumor angiogenesis, arthritis and atherosclerosis by anti-Flt1

Aernout Luttun; Marc Tjwa; Lieve Moons; Yan Wu; Anne Angelillo-Scherrer; Fang Liao; Janice A. Nagy; Andrea T. Hooper; Josef Priller; Bert De Klerck; Veerle Compernolle; Evis Daci; Peter Bohlen; Mieke Dewerchin; Jean Marc Herbert; Roy A. Fava; Patrick Matthys; Geert Carmeliet; Desire Collen; Harold F. Dvorak; Daniel J. Hicklin; Peter Carmeliet

The therapeutic potential of placental growth factor (PlGF) and its receptor Flt1 in angiogenesis is poorly understood. Here, we report that PlGF stimulated angiogenesis and collateral growth in ischemic heart and limb with at least a comparable efficiency to vascular endothelial growth factor (VEGF). An antibody against Flt1 suppressed neovascularization in tumors and ischemic retina, and angiogenesis and inflammatory joint destruction in autoimmune arthritis. Anti-Flt1 also reduced atherosclerotic plaque growth and vulnerability, but the atheroprotective effect was not attributable to reduced plaque neovascularization. Inhibition of VEGF receptor Flk1 did not affect arthritis or atherosclerosis, indicating that inhibition of Flk1-driven angiogenesis alone was not sufficient to halt disease progression. The anti-inflammatory effects of anti-Flt1 were attributable to reduced mobilization of bone marrow–derived myeloid progenitors into the peripheral blood; impaired infiltration of Flt1-expressing leukocytes in inflamed tissues; and defective activation of myeloid cells. Thus, PlGF and Flt1 constitute potential candidates for therapeutic modulation of angiogenesis and inflammation.


Journal of Leukocyte Biology | 2001

Modes of action of Freund’s adjuvants in experimental models of autoimmune diseases

Alfons Billiau; Patrick Matthys

Freund’s adjuvants are irreplaceable components of induction protocols of many experimental animal models of autoimmune disease. Apart from the early studies done in the 1950s and 1960s, no further direct investigation on the mode of action of these adjuvants has been undertaken. It is generally assumed that incomplete (IFA) and complete Freund’s adjuvant (CFA) act by prolonging the lifetime of injected autoantigen, by stimulating its effective delivery to the immune system and by providing a complex set of signals to the innate compartment of the immune system, resulting in altered leukocyte proliferation and differentiation. Here, we review evidence collected from various types of studies that provide more insight in the specific alterations of the immune response caused by IFA and CFA. Early events include rapid uptake of adjuvant components by dendritic cells, enhanced phagocytosis, secretion of cytokines by mononuclear phagocytes, and transient activation and proliferation of CD4+ lymphocytes. The mycobacterial components within CFA signal T lymphocytes to assume a Th1 profile so that strong delayed‐type hypersensitivity against autoantigens develops. In the absence of mycobacteria, T‐lymphocyte differentiation tends to assume a Th2 profile with strong antibody production only. The mycobacterial component also accounts for a morphologic and functional remodeling of the haemopoietic system that develops over a period of several weeks and that is characterized by a drastic expansion of Mac‐1+ immature myeloid cells. These cells have been found to be associated with enhanced disease in some models but with reduced disease in others. Thus, in experimental autoimmune diseases, CFA‐mediated activation of the innate immune compartment is important not only by regulating the early induction phase but also by providing a surplus of effector and regulator cells in the late phase.


Cytokine & Growth Factor Reviews | 2009

Interferon-γ: A historical perspective

Alfons Billiau; Patrick Matthys

This article reviews the main lines of thinking and exploration that have led to our current conception of the role of IFN-gamma in immune defense and autoimmunity. In 1965 the first report appeared describing production of an interferon-like virus inhibitor in cultured human leukocytes following exposure to the mitogen phytohemagglutinin. In the early 1970s the active principle became recognized as being distinct from classical virus-induced interferons, leading to its designation as immune interferon or Type II interferon, and eventually IFN-gamma. Up to that point interest in the factor had come almost exclusively from virologists, in particular those among them who were believers in interferon. Evidence first coming forward in the 1980s that IFN-gamma is indistinguishable from macrophage-activating factor (MAF), then a prototype lymphokine, was the signal for immunologists at large to become interested. Today IFN-gamma ranks among the most important endogenous regulators of immune responses.


Journal of Immunology | 2001

AMD3100, a potent and specific antagonist of the stromal cell-derived factor-1 chemokine receptor CXCR4, inhibits autoimmune joint inflammation in IFN-gamma receptor-deficient mice.

Patrick Matthys; Sigrid Hatse; Kurt Vermeire; Anja Wuyts; Gary J. Bridger; Geoffrey W. Henson; Erik De Clercq; Alfons Billiau; Dominique Schols

Autoimmune collagen-induced arthritis (CIA) in IFN-γR-deficient DBA/1 mice was shown to be reduced in severity by treatment with the bicyclam derivative AMD3100, a specific antagonist of the interaction between the chemokine stromal cell-derived factor-1 (SDF-1) and its receptor CXCR4. The beneficial effect of the CXCR4 antagonist was demonstrable when treatment was initiated between the time of immunization and appearance of the first symptoms. Treatment also reduced the delayed-type hypersensitivity response to the autoantigen, collagen type II. These observations are indicative of an action on a late event in the pathogenesis, such as chemokine-mediated attraction of leukocytes toward joint tissues. The notion of SDF-1 involvement was further supported by the observation that exogenous SDF-1 injected in periarthritic tissue elicited an inflammatory response that could be inhibited by AMD3100. The majority of leukocytes harvested from inflamed joints of mice with CIA were found to be Mac-1+ and CXCR4+, and AMD3100 was demonstrated to interfere specifically with chemotaxis and Ca2+ mobilization induced in vitro by SDF-1 on Mac-1+/CXCR4+ splenocytes. We conclude that SDF-1 plays a central role in the pathogenesis of murine CIA, by attracting Mac-1+/CXCR4+ cells to the inflamed joints.


Arthritis & Rheumatism | 2009

Proinflammatory role of the Th17 cytokine interleukin-22 in collagen-induced arthritis in C57BL/6 mice.

Lies Geboes; Laure Dumoutier; Hilde Kelchtermans; Evelien Schurgers; Tania Mitera; Jean-Christophe Renauld; Patrick Matthys

OBJECTIVE To investigate the role of interleukin-22 (IL-22) in collagen-induced arthritis (CIA), an animal model of rheumatoid arthritis. METHODS C57BL/6 mice were immunized with type II collagen (CII) in Freunds incomplete adjuvant with added Mycobacterium tuberculosis, and levels of IL-22 and its specific receptor, IL-22 receptor type I (IL-22RI), were measured in sera and tissue by enzyme-linked immunosorbent assay and real-time quantitative polymerase chain reaction analysis. Clinical and histologic signs of arthritis were recorded and compared with those in C57BL/6 mice deficient in the IL-22 gene (IL-22(-/-)). Humoral and cellular immune responses against CII were analyzed. In vitro osteoclastogenesis assays were performed on splenocytes. RESULTS Upon immunization with CII in Freunds incomplete adjuvant plus heat-killed Mycobacterium tuberculosis, sera from C57BL/6 mice were found to contain high levels of IL-22, and the specific IL-22RI was expressed in lymphoid tissue, including splenocytes. IL-22(-/-) mice were less susceptible to CIA than were wild-type mice, as evidenced by their decreased incidence of arthritis and decreased pannus formation. Remarkably, the less severe form of arthritis in IL-22(-/-) mice was associated with increased production of CII-specific and total IgG antibodies, whereas cellular CII responses were unchanged. In vitro, IL-22 was found to promote osteoclastogenesis, a process that might contribute to its proinflammatory activity in CIA. CONCLUSION Endogenous IL-22 plays a proinflammatory role in CIA in C57BL/6 mice. Our data also indicate that IL-22 promotes osteoclastogenesis and regulates antibody production.


Nature Genetics | 2011

A20 (TNFAIP3) deficiency in myeloid cells triggers erosive polyarthritis resembling rheumatoid arthritis

Mourad Matmati; Peggy Jacques; Jonathan Maelfait; Eveline Verheugen; Mirjam Kool; Mozes Sze; Lies Geboes; Els Louagie; Conor Mc Guire; Lars Vereecke; Yuanyuan Chu; Louis Boon; Steven Staelens; Patrick Matthys; Bart N. Lambrecht; Marc Schmidt-Supprian; Manolis Pasparakis; Dirk Elewaut; Rudi Beyaert; Geert van Loo

A20 (TNFAIP3) is a protein that is involved in the negative feedback regulation of NF-κB signaling in response to specific proinflammatory stimuli in different cell types and has been suggested as a susceptibility gene for rheumatoid arthritis. To define the contribution of A20 to rheumatoid arthritis pathology, we generated myeloid-specific A20-deficient mice and show that specific ablation of Tnfaip3 in myeloid cells results in spontaneous development of a severe destructive polyarthritis with many features of rheumatoid arthritis. Myeloid-A20–deficient mice have high levels of inflammatory cytokines in their serum, consistent with a sustained NF-κB activation and higher TNF production by macrophages. Destructive polyarthritis in myeloid A20 knockout mice was TLR4-MyD88 and IL-6 dependent but was TNF independent. Myeloid A20 deficiency also promoted osteoclastogenesis in mice. Together, these observations indicate a critical and cell-specific function for A20 in the etiology of rheumatoid arthritis, supporting the idea of developing A20 modulatory drugs as cell-targeted therapies.


Arthritis Research & Therapy | 2005

Defective CD4+CD25+ regulatory T cell functioning in collagen-induced arthritis: an important factor in pathogenesis, counter-regulated by endogenous IFN-γ

Hilde Kelchtermans; Bert De Klerck; Tania Mitera; Maarten Van Balen; Dominique Bullens; Alfons Billiau; Georges Leclercq; Patrick Matthys

Mice with a deficiency in IFN-γ or IFN-γ receptor (IFN-γR) are more susceptible to collagen-induced arthritis (CIA), an experimental autoimmune disease that relies on the use of complete Freunds adjuvant (CFA). Here we report that the heightened susceptibility of IFN-γR knock-out (KO) mice is associated with a functional impairment of CD4+CD25+ Treg cells. Treatment of wild-type mice with depleting anti-CD25 antibody after CFA-assisted immunisation with collagen type II (CII) significantly accelerated the onset of arthritis and increased the severity of CIA. This is an indication of a role of Treg cells in the effector phase of CIA. IFN-γR deficiency did not affect the number of CD4+CD25+ T cells in the central and peripheral lymphoid tissues. In addition, CD4+CD25+ T cells isolated from naive IFN-γR KO mice had a normal potential to suppress T cell proliferation in vitro. However, after immunisation with CII in CFA, the suppressive activity of CD4+CD25+ T cells became significantly more impaired in IFN-γR-deficient mice. Moreover, expression of the mRNA for Foxp3, a highly specific marker for Treg cells, was lower. We further demonstrated that the effect of endogenous IFN-γ, which accounts for more suppressive activity in wild-type mice, concerns both Treg cells and accessory cells. Our results demonstrate that the decrease in Treg cell activity in CIA is counter-regulated by endogenous IFN-γ.


Nature Immunology | 2013

Antiapoptotic Mcl-1 is critical for the survival and niche-filling capacity of Foxp3 + regulatory T cells

Wim Pierson; Bénédicte Cauwe; Antonia Policheni; Susan M. Schlenner; Dean Franckaert; Julien Berges; Stéphanie Humblet-Baron; Susann Schönefeldt; Marco J. Herold; David A. Hildeman; Andreas Strasser; Li-Fan Lu; Patrick Matthys; Antonio A. Freitas; Rita J. Luther; Casey T. Weaver; James Dooley; Daniel Gray; Adrian Liston

Foxp3+ regulatory T (Treg) cells are a crucial immunosuppressive population of CD4+ T cells, yet the homeostatic processes and survival programs that maintain the Treg cell pool are poorly understood. Here we report that peripheral Treg cells markedly alter their proliferative and apoptotic rates to rapidly restore numerical deficit through an interleukin 2–dependent and costimulation-dependent process. By contrast, excess Treg cells are removed by attrition, dependent on the Bim-initiated Bak- and Bax-dependent intrinsic apoptotic pathway. The antiapoptotic proteins Bcl-xL and Bcl-2 were dispensable for survival of Treg cells, whereas Mcl-1 was critical for survival of Treg cells, and the loss of this antiapoptotic protein caused fatal autoimmunity. Together, these data define the active processes by which Treg cells maintain homeostasis via critical survival pathways.


Trends in Immunology | 2008

How interferon-γ keeps autoimmune diseases in check

Hilde Kelchtermans; Alfons Billiau; Patrick Matthys

Interferon-gamma (IFN-gamma) is regarded traditionally as a proinflammatory factor and as the signature cytokine of Th1-dominated autoimmune processes. Early evidence indicative of an opposite, protective role has recently received further attention from reports revealing an increasing number of pathways by which IFN-gamma can counteract harmful inflammation in Th1-associated autoimmune diseases. Here, we review evidence for IFN-gammas anti-inflammatory effects primarily from the perspective of one experimental model, collagen-induced arthritis (CIA), and question the classic proinflammatory role of IFN-gamma and also the Th1-Th2 paradigm as a basis for explaining the regulation of autoimmune diseases. We conclude that endogenous production of IFN-gamma during inflammatory and autoimmune diseases should be considered as a process with bidirectional immunoregulatory consequences, often resulting in moderation of pathology.


European Journal of Cancer and Clinical Oncology | 1991

Anti-interferon-γ antibody treatment, growth of Lewis lung tumours in mice and tumour-associated cachexia

Patrick Matthys; Hubertine Heremans; Ghislain Opdenakker; Alfons Billiau

C57BL/6N mice bearing Lewis lung tumours were treated with anti-gamma-interferon (IFN-gamma) monoclonal antibodies. Early, but not late, treatment inhibited tumour growth, suggesting that endogenous IFN-gamma promotes initial tumour cell proliferation. Tumour development was associated with failure to gain weight or with progressive weight loss. Anti-IFN-gamma given early or late counteracted this wasting syndrome, which indicates that IFN-gamma production subsists during tumour growth and is directly or indirectly responsible for tumour-associated cachexia. Studies of body composition in cachectic mice revealed fat tissue to be particularly affected. Fat loss was enhanced by IFN-gamma and antagonised by anti-IFN-gamma. Tumour-bearing mice were also hypersensitive to the lethal effect of endotoxin; anti-IFN-gamma was unable to mitigate this sensitisation, suggesting that IFN-gamma does not exert its cachexia-inducing effect through augmentation of the host response to an endogenous endotoxin source.

Collaboration


Dive into the Patrick Matthys's collaboration.

Top Co-Authors

Avatar

Alfons Billiau

Katholieke Universiteit Leuven

View shared research outputs
Top Co-Authors

Avatar

Tania Mitera

Katholieke Universiteit Leuven

View shared research outputs
Top Co-Authors

Avatar

Hubertine Heremans

Katholieke Universiteit Leuven

View shared research outputs
Top Co-Authors

Avatar

Kurt Vermeire

Rega Institute for Medical Research

View shared research outputs
Top Co-Authors

Avatar

Carine Wouters

Centre Hospitalier de Luxembourg

View shared research outputs
Top Co-Authors

Avatar

Anneleen Avau

Katholieke Universiteit Leuven

View shared research outputs
Top Co-Authors

Avatar

Ellen Brisse

Katholieke Universiteit Leuven

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Karen Put

Rega Institute for Medical Research

View shared research outputs
Top Co-Authors

Avatar

Stéphanie Put

Katholieke Universiteit Leuven

View shared research outputs
Researchain Logo
Decentralizing Knowledge