Network


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

Hotspot


Dive into the research topics where Matthias Corbascio is active.

Publication


Featured researches published by Matthias Corbascio.


Journal of Clinical Investigation | 1999

Asialo GM1+ CD8+ T cells play a critical role in costimulation blockade–resistant allograft rejection

Joel Trambley; Adam W. Bingaman; Angello Lin; Eric T. Elwood; Seung Yeun Waitze; Jong-Won Ha; Megan M. Durham; Matthias Corbascio; Shannon R. Cowan; Thomas C. Pearson; Christian P. Larsen

Simultaneous blockade of the CD40 and CD28 costimulatory pathways is an effective treatment strategy to promote allograft acceptance but does not lead to indefinite allograft survival. The immune mechanisms responsible for costimulation-independent rejection are not defined. Here we have studied the rejection responses of murine C57BL/6 recipients, which we show to be relatively resistant to inhibition by combined CD40/CD28 blockade. We demonstrate that asialo GM1(+) CD8(+) cells play a critical role in this costimulation blockade-resistant rejection. These results provide new insights into the costimulatory requirements for T-cell subsets and demonstrate for the first time that combined blockade of the CD40 and CD28 pathways does not adequately inhibit CD8-mediated skin allograft rejection. Furthermore, we provide evidence that asialo GM1 is a potentially important therapeutic target for CD8-dependent immune responses.


The Lancet | 2016

Percutaneous coronary angioplasty versus coronary artery bypass grafting in treatment of unprotected left main stenosis (NOBLE): a prospective, randomised, open-label, non-inferiority trial

Timo H. Mäkikallio; Niels R. Holm; Mitchell Lindsay; Mark S. Spence; Andrejs Erglis; Ian Ba Menown; Thor Trovik; Markku Eskola; Hannu Romppanen; Thomas Kellerth; Jan Ravkilde; Lisette Okkels Jensen; Gintaras Kalinauskas; Rikard Linder; Markku O. Pentikäinen; Anders Hervold; Adrian P. Banning; Azfar Zaman; Jamen Cotton; Erlend Eriksen; Sulev Margus; Henrik Toft Sørensen; Per Hostrup Nielsen; Matti Niemelä; Kari Kervinen; Jens Flensted Lassen; Michael Maeng; Keith G. Oldroyd; Geoff Berg; Simon Walsh

BACKGROUND Coronary artery bypass grafting (CABG) is the standard treatment for revascularisation in patients with left main coronary artery disease, but use of percutaneous coronary intervention (PCI) for this indication is increasing. We aimed to compare PCI and CABG for treatment of left main coronary artery disease. METHODS In this prospective, randomised, open-label, non-inferiority trial, patients with left main coronary artery disease were enrolled in 36 centres in northern Europe and randomised 1:1 to treatment with PCI or CABG. Eligible patients had stable angina pectoris, unstable angina pectoris, or non-ST-elevation myocardial infarction. Exclusion criteria were ST-elevation myocardial infarction within 24 h, being considered too high risk for CABG or PCI, or expected survival of less than 1 year. The primary endpoint was major adverse cardiac or cerebrovascular events (MACCE), a composite of all-cause mortality, non-procedural myocardial infarction, any repeat coronary revascularisation, and stroke. Non-inferiority of PCI to CABG required the lower end of the 95% CI not to exceed a hazard ratio (HR) of 1·35 after up to 5 years of follow-up. The intention-to-treat principle was used in the analysis if not specified otherwise. This trial is registered with ClinicalTrials.gov identifier, number NCT01496651. FINDINGS Between Dec 9, 2008, and Jan 21, 2015, 1201 patients were randomly assigned, 598 to PCI and 603 to CABG, and 592 in each group entered analysis by intention to treat. Kaplan-Meier 5 year estimates of MACCE were 29% for PCI (121 events) and 19% for CABG (81 events), HR 1·48 (95% CI 1·11-1·96), exceeding the limit for non-inferiority, and CABG was significantly better than PCI (p=0·0066). As-treated estimates were 28% versus 19% (1·55, 1·18-2·04, p=0·0015). Comparing PCI with CABG, 5 year estimates were 12% versus 9% (1·07, 0·67-1·72, p=0·77) for all-cause mortality, 7% versus 2% (2·88, 1·40-5·90, p=0·0040) for non-procedural myocardial infarction, 16% versus 10% (1·50, 1·04-2·17, p=0·032) for any revascularisation, and 5% versus 2% (2·25, 0·93-5·48, p=0·073) for stroke. INTERPRETATION The findings of this study suggest that CABG might be better than PCI for treatment of left main stem coronary artery disease. FUNDING Biosensors, Aarhus University Hospital, and participating sites.


Transplantation | 1998

Prolonged acceptance of concordant and discordant xenografts with combined CD40 and CD28 pathway blockade

Eric T. Elwood; Christian P. Larsen; Hong Rae Cho; Matthias Corbascio; Shannon C. Ritchie; Diane Z. Alexander; Carol Tucker-Burden; Peter S. Linsley; Alejandro Aruffo; Diane Hollenbaugh; Kevin J. Winn; Thomas C. Pearson

BACKGROUND The prompt and vigorous immune response to xenogenic tissue remains a significant barrier to clinical xenotransplantation. Simultaneous blockade of the CD28 and CD40 costimulatory pathways has been shown to dramatically inhibit the immune response to alloantigen. METHODS . In this study, we investigated the ability of simultaneous blockade of the CD28 and CD40 pathways to inhibit the immune response to xenoantigen in the rat-to-mouse and pig-to-mouse models. RESULTS Simultaneous blockade of the CD28 and CD40 pathways produced marked inhibition of the cellular response to xenoantigen in vivo and produced long-term acceptance of xenogeneic cardiac and skin grafts (rat-to-mouse), and markedly suppressed an evoked antibody response to xenoantigen. In addition, this strategy significantly prolonged the survival of pig skin on recipient mice. CONCLUSIONS Long-term hyporesponsiveness to xenoantigen across both a concordant and discordant species barrier, measured by the stringent criterion of skin grafting, can be achieved using a noncytoablative treatment regimen.


Reproductive Biomedicine Online | 2006

Human embryonic stem cells are immunogenic in allogeneic and xenogeneic settings

Karl-Henrik Grinnemo; Makiko Kumagai-Braesch; Agneta Mânsson-Broberg; Heli Skottman; Xiaojin Hao; Anwar J. Siddiqui; Agneta Andersson; Anne-Marie Strömberg; Riita Lahesmaa; Outi Hovatta; Christer Sylvén; Matthias Corbascio; Göran Dellgren

Recent studies have suggested that human embryonic stem cells (HESC) are immune-privileged and may thereby circumvent rejection. The expression of immunologically active molecules was studied by DNA microarray analysis and by flow cytometry. HESC were transplanted into immunologically competent mice and traced by fluorescence in-situ hybridization (FISH) and immunohistochemistry. The ability of HESC to directly and indirectly induce immune responses in CD4+ T-cells from naive and transplanted mice was studied. Their ability to induce immune responses of human CD4+ T-cells, when cultured in the presence of dendritic cells (DC) syngeneic to responder T-cells, was also analysed. HESC demonstrated expression of HLA class I and HLA class II genes, but the cell surface expression of HLA class II molecules was low even after incubation with IFNgamma. In wild-type mice, HESC could be demonstrated by FISH until 3 days after transplantation and were surrounded by heavy infiltrates of T-cells and macrophages. HESC induced a similar immune response as human fibroblast cells (HFib) on naive and immunized T-cells, both directly and in the presence of syngeneic DC. A similar response was observed in the allogeneic setting. It is concluded that HESC are immunologically inert and do not inhibit immune responses during direct or indirect antigen presentation, and they were acutely rejected in a xenogeneic setting.


Transplantation | 1997

Analysis of the B7 costimulatory pathway in allograft rejection.

Thomas C. Pearson; Diane Z. Alexander; Matthias Corbascio; Rose Hendrix; Shannon C. Ritchie; Peter S. Linsley; Denise Faherty; Christian P. Larsen

BACKGROUND Blockade of the B7/CD28 costimulation pathway with the fusion protein, CTLA4-Ig, has been shown to prolong allograft survival in numerous rodent models, suggesting that this pathway is functionally important in the allograft rejection response. This pathway is complex and consists of at least the B7-1, B7-1a, B7-1cyt II, and B7-2 molecules on the antigen-presenting cell and CD28 and CTLA4 molecules on the T cell. METHODS The intragraft transcript expression of the B7 molecules and their counterreceptors was defined using reverse transcriptase-polymerase chain reaction in the vascularized mouse cardiac allograft model. In addition, the functional significance of these molecules was investigated both in vitro in the mixed leukocyte response (MLR) and in vivo in the vascularized mouse cardiac allograft model. RESULTS Intragraft expression of B7-1, B7-1a, B7-1cyt II, B7-2, CD28, and CTLA4 transcripts is up-regulated in allografts when compared with both normal untransplanted hearts and syngeneic transplants at between 5 and 12 days after transplant. Both anti-B7-1 and anti-B7-2 monoclonal antibodies alone inhibited T-cell proliferation in the MLR, however, equivalent maximal inhibition was obtained by a combination of these agents or by CTLA4-Ig. Likewise, in the mouse cardiac allograft model, both anti-B7-1 and anti-B7-2 modestly prolonged graft survival. However, an increased survival was obtained with either a combination of anti-B7-1 and anti-B7-2 or CTLA4-Ig. Blockade of the B7/CD28 pathway in the MLR using T cells from CD28 knockout mice had no effect on the proliferative response. Likewise, blockade of the B7/CD28 pathway did not effect the rate of rejection of cardiac allografts by CD28 knockout recipients. CONCLUSIONS These data suggest that both B7-1 and B7-2 have an important role in allograft rejection in the mouse vascularized cardiac allograft model.


Cell and Tissue Research | 2008

Immunogenicity of human embryonic stem cells

Karl-Henrik Grinnemo; Christer Sylvén; Outi Hovatta; Göran Dellgren; Matthias Corbascio

Human embryonic stem cells (HESC) are pluripotent stem cells isolated from the inner cell mass of human blastocysts. With the first successful culturing of HESC, a new era of regenerative medicine was born. HESC can differentiate into almost any cell type and, in the future, might replace solid organ transplantation and even be used to treat progressive degenerative diseases such as Parkinson’s disease. Although this sounds promising, certain obstacles remain with regard to their clinical use, such as culturing HESC under well-defined conditions without exposure to animal proteins, the risk of teratoma development and finally the avoidance of immune rejection. In this review, we discuss the immunological properties of HESC and various strategic solutions to circumvent immune rejection, such as stem cell banking, somatic cell nuclear transfer and the induction of tolerance by co-stimulation blockade and mixed chimerism.


Annals of Medicine | 2006

Human mesenchymal stem cells do not differentiate into cardiomyocytes in a cardiac ischemic xenomodel

Karl-Henrik Grinnemo; Agneta Månsson-Broberg; Katarina LeBlanc; Matthias Corbascio; Eva Wärdell; Anwar J. Siddiqui; Xiaojin Hao; Christer Sylvén; Göran Dellgren

AIM. As the capability of human mesenchymal stem cells (hMSC) to engraft, differentiate and improve myocardial function cannot be studied in humans, exploration was performed in a xenomodel. METHODS. The rats were divided into three groups depending on the type of rats used (Rowett nude (RNU) or Fischer rats +/− immunosuppression). Different groups were treated with intramyocardial injection of hMSC (1–2 million) either directly or three days after ligation of the left anterior descending artery (LAD). Myocardial function was investigated by echocardiography. The hMSC were identified with fluorescence in situ hybridization and myocardial differentiation was assessed by immunohistochemistry. RESULTS. When hMSC were injected directly after LAD ligation they could be identified in half (8/16) of the RNU rats (without immunosuppression) at 4 weeks. When injected 3 days after LAD ligation in immunosuppressed RNU rats they were identified in all (6/6) rats at 6 weeks. The surviving hMSC showed signs of differentiation into fibroblasts. No cardiomyocyte differentiation was observed. There was no difference in myocardial function in treated animals compared to controls. CONCLUSIONS. The hMSC survived in this xenomodel up to 6 weeks. However, hMSC required implantation into immunoincompetent animals as well as immunosuppression to survive, indicating that these cells are otherwise rejected. Furthermore, these cells did not differentiate into cardiomyocytes nor did they improve heart function in this xenomodel.


International Journal of Cancer | 2006

Microvascular endothelial cells increase proliferation and inhibit apoptosis of native human acute myelogenous leukemia blasts

Kimberley Joanne Hatfield; Anita Ryningen; Matthias Corbascio; Øystein Bruserud

Interactions between acute myelogenous leukemia (AML) blasts and neighbouring endothelial cells in the bone marrow seem important both for disease development and susceptibility to chemotherapy. We investigated the effects of soluble mediators released by microvascular endothelial cells on native human AML cells. AML cells derived from 33 patients were cocultured with microvascular endothelial cells, separated by a semipermeable membrane. We investigated the effect of coculture on AML cell proliferation, viability/apoptosis and cytokine release. Coculture increased AML cell proliferation, and this growth enhancement included the clonogenic leukemia cell subset. Increased release of several soluble mediators was also detected (interleukin 3, interleukin 6, granulocyte‐macrophage and granulocyte colony‐stimulating factors) in cocultures. Our cytokine neutralization experiments suggest that an intercellular crosstalk involving several soluble mediators contribute to the increased leukemia cell proliferation. The presence of endothelial cells had an additional antiapoptotic effect on the AML cells. The endothelial cells did not have any growth‐enhancing effect on native human acute lymphoblastic leukemia cells. Our in vitro results suggest that the release of soluble mediators by microvascular endothelial cells supports leukemic hematopoiesis through paracrine mechanisms by direct enhancement of AML blast proliferation and by inhibition of leukemic cell apoptosis.


Stem Cells and Development | 2010

Islet-1 cells are cardiac progenitors present during the entire lifespan: from the embryonic stage to adulthood.

Rami Genead; Christian Danielsson; Agneta Andersson; Matthias Corbascio; Anders Franco-Cereceda; Christer Sylvén; Karl-Henrik Grinnemo

The aim of this study was to longitudinally characterize the distribution of cells actively expressing the progenitor transcription factor islet-1 (Isl1+) during the embryonic life, the postnatal period, and adulthood. In this study, we have used direct immunohistochemical staining toward the protein Isl1 in a longitudinal rat model. Cells actively expressing Isl1 were traced in embryos from gestational day (GD) 11 until adulthood. In early cardiac development (GD 11), the Isl1+ progenitors were located in a greater abundance in the paracardiac regions, areas suggested to be the second heart field. To a lesser extent, Isl1+ cells were present within the bulbotruncal region and the truncus arteriosus. During the following days until GD 15, the Isl1+ cells were mainly observed at the proximal outflow tract (OFT) and at the inflow area of the right atrium. No Isl1+ cells were detected in the left ventricle. Compared with GD 11, more Isl1+ cells seemed to co-express cardiomyocyte markers and a minority of the Isl1+ cells was undifferentiated. Unexpectedly, only few undifferentiated Isl1+ cells were Ki67+ while a lot of TnT+ cardiomyocytes were proliferating in the ventricles. After birth, immature Isl1+ cells were still present in the OFT where they resided until adulthood. Our data suggest that during embryogenesis, Isl1+ cells migrate from extracardiac regions into the proximal part of the heart, proliferating and giving rise to cardioblasts. Unexpectedly, only a minority of the Isl1+ cells while a majority of ventricular cardiomyocytes were proliferating. The Isl1+ cell pool persists into adulthood, which might open up new strategies to repair damaged myocardium.


Stem Cells | 2008

Costimulation Blockade Induces Tolerance to HESC Transplanted to the Testis and Induces Regulatory T‐Cells to HESC Transplanted into the Heart

Karl-Henrik Grinnemo; Rami Genead; Makiko Kumagai-Braesch; Agneta Andersson; Christian Danielsson; Agneta Månsson-Broberg; Göran Dellgren; Anne-Marie Strömberg; Henrik Ekberg; Outi Hovatta; Christer Sylvén; Matthias Corbascio

In order to study the ability of costimulation blockade to induce tolerance to human embryonic stem cells (HESC), severe combined immunodeficient (SCID), and immunocompetent C57BL/6 mice treated with costimulation blockade received intratesticular and intramyocardial HESC transplants. All SCID mice with intratesticular HESC transplants developed teratoma. When SCID mice were transplanted intramyocardially, only two of five mice developed teratoma‐like tumors. C57BL/6 mice transplanted intratesticularly and treated with costimulation blockade all developed teratoma and were surrounded by CD4+CD25+Foxp3+ T‐cells, while isotype control treated recipients rejected their grafts. Most C57BL/6 mice transplanted intramyocardially and treated with costimulation blockade demonstrated lymphocytic infiltrates 1 month after transplantation, whereas one maintained its graft. Isolation of regulatory T‐cells from intramyocardial transplanted recipients treated with costimulation blockade demonstrated specificity toward undifferentiated HESC and down‐regulated naive T‐cell activation toward HESC. These results demonstrate that costimulation blockade is sufficiently robust to induce tolerance to HESC in the immune‐privileged environment of the testis. HESC specific regulatory T‐cells developed to HESC transplanted to the heart and the success of transplantation was similar to that seen in SCID mice.

Collaboration


Dive into the Matthias Corbascio's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Karl-Henrik Grinnemo

Karolinska University Hospital

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Christer Sylvén

Karolinska University Hospital

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Agneta Andersson

Karolinska University Hospital

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge