Network


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

Hotspot


Dive into the research topics where Karl-Otto Larsen is active.

Publication


Featured researches published by Karl-Otto Larsen.


Journal of Applied Physiology | 2009

Elevated levels of activin A in clinical and experimental pulmonary hypertension.

Arne Yndestad; Karl-Otto Larsen; Erik Øie; Thor Ueland; Camilla Smith; Bente Halvorsen; Ivar Sjaastad; Ole Henning Skjønsberg; Turid M. Pedersen; Ole-Gunnar Anfinsen; Jan Kristian Damås; Geir Christensen; Pål Aukrust; Arne K. Andreassen

Activin A, a member of the transforming growth factor (TGF)-beta superfamily, is involved in regulation of tissue remodeling and inflammation. Herein, we wanted to explore a role for activin A in pulmonary hypertension (PH). Circulating levels of activin A and its binding protein follistatin were measured in patients with PH (n = 47) and control subjects (n = 14). To investigate synthesis and localization of pulmonary activin A, we utilized an experimental model of hypoxia-induced PH. In mouse lungs, we also explored signaling pathways that can be activated by activin A, such as phosphorylation of Smads, which are mediators of TGF-beta signaling. Possible pathophysiological mechanisms initiated by activin A were explored by exposing pulmonary arterial smooth muscle cells in culture to this cytokine. Elevated levels of activin A and follistatin were found in patients with PH, and activin A levels were significantly related to mortality. Immunohistochemistry of lung autopsies from PH patients and lungs with experimental PH localized activin A primarily to alveolar macrophages and bronchial epithelial cells. Mice with PH exhibited increased pulmonary levels of mRNA for activin A and follistatin in the lungs, and also elevated pulmonary levels of phosphorylated Smad2. Finally, we found that activin A increased proliferation and induced gene expression of endothelin-1 and plasminogen activator inhibitor-1 in pulmonary artery smooth muscle cells, mediators that could contribute to vascular remodeling. Our findings in both clinical and experimental studies suggest a role for activin A in the development of various types of PH.


Cardiovascular Research | 2008

Diastolic dysfunction in alveolar hypoxia: a role for interleukin-18-mediated increase in protein phosphatase 2A

Karl-Otto Larsen; Birgitte Lygren; Ivar Sjaastad; Kurt A. Krobert; K. Arnkværn; Geir Florholmen; Ann-Kristin Ruud Larsen; Finn Olav Levy; Kjetil Taskén; Ole Henning Skjønsberg; Geir Christensen

AIMS Chronic obstructive pulmonary disease with alveolar hypoxia is associated with diastolic dysfunction in the right and left ventricle (LV). LV diastolic dysfunction is not caused by increased afterload, and we recently showed that reduced phosphorylation of phospholamban at serine (Ser) 16 may explain the reduced relaxation of the myocardium. Here, we study the mechanisms leading to the hypoxia-induced reduction in phosphorylation of phospholamban at Ser16. METHODS AND RESULTS In C57Bl/6j mice exposed to 10% oxygen, signalling molecules were measured in cardiac tissue, sarcoplasmic reticulum (SR)-enriched membrane preparations, and serum. Cardiomyocytes isolated from neonatal mice were exposed to interleukin (IL)-18 for 24 h. The beta-adrenergic pathway in the myocardium was not altered by alveolar hypoxia, as assessed by measurements of beta-adrenergic receptor levels, adenylyl cyclase activity, and subunits of cyclic AMP-dependent protein kinase. However, alveolar hypoxia led to a significantly higher amount (124%) and activity (234%) of protein phosphatase (PP) 2A in SR-enriched membrane preparations from LV compared with control. Serum levels of an array of cytokines were assayed, and a pronounced increase in IL-18 was observed. In isolated cardiomyocytes, treatment with IL-18 increased the amount and activity of PP2A, and reduced phosphorylation of phospholamban at Ser16 to 54% of control. CONCLUSION Our results indicate that the diastolic dysfunction observed in alveolar hypoxia might be caused by increased circulating IL-18, thereby inducing an increase in PP2A and a reduction in phosphorylation of phospholamban at Ser16.


American Journal of Physiology-lung Cellular and Molecular Physiology | 2015

Absence of the inflammasome adaptor ASC reduces hypoxia-induced pulmonary hypertension in mice.

Fadila Telarevic Cero; Vigdis Hillestad; Ivar Sjaastad; Arne Yndestad; Pål Aukrust; Trine Ranheim; Ida G. Lunde; Maria Belland Olsen; Egil Lien; Lili Zhang; Solveig Bjærum Haugstad; Else Marit Løberg; Geir Christensen; Karl-Otto Larsen; Ole Henning Skjønsberg

Pulmonary hypertension is a serious condition that can lead to premature death. The mechanisms involved are incompletely understood although a role for the immune system has been suggested. Inflammasomes are part of the innate immune system and consist of the effector caspase-1 and a receptor, where nucleotide-binding oligomerization domain-like receptor pyrin domain-containing 3 (NLRP3) is the best characterized and interacts with the adaptor protein apoptosis-associated speck-like protein containing a caspase-recruitment domain (ASC). To investigate whether ASC and NLRP3 inflammasome components are involved in hypoxia-induced pulmonary hypertension, we utilized mice deficient in ASC and NLRP3. Active caspase-1, IL-18, and IL-1β, which are regulated by inflammasomes, were measured in lung homogenates in wild-type (WT), ASC(-/-), and NLRP3(-/-) mice, and phenotypical changes related to pulmonary hypertension and right ventricular remodeling were characterized after hypoxic exposure. Right ventricular systolic pressure (RVSP) of ASC(-/-) mice was significantly lower than in WT exposed to hypoxia (40.8 ± 1.5 mmHg vs. 55.8 ± 2.4 mmHg, P < 0.001), indicating a substantially reduced pulmonary hypertension in mice lacking ASC. Magnetic resonance imaging further supported these findings by demonstrating reduced right ventricular remodeling. RVSP of NLRP3(-/-) mice exposed to hypoxia was not significantly altered compared with WT hypoxia. Whereas hypoxia increased protein levels of caspase-1, IL-18, and IL-1β in WT and NLRP3(-/-) mice, this response was absent in ASC(-/-) mice. Moreover, ASC(-/-) mice displayed reduced muscularization and collagen deposition around arteries. In conclusion, hypoxia-induced elevated right ventricular pressure and remodeling were attenuated in mice lacking the inflammasome adaptor protein ASC, suggesting that inflammasomes play an important role in the pathogenesis of pulmonary hypertension.


American Journal of Physiology-lung Cellular and Molecular Physiology | 2011

Lack of CCR7 induces pulmonary hypertension involving perivascular leukocyte infiltration and inflammation

Karl-Otto Larsen; Arne Yndestad; Ivar Sjaastad; Else Marit Løberg; Ingeborg Løstegaard Goverud; Bente Halvorsen; Jing Jia; Arne K. Andreassen; Cathrine Husberg; Sofia Jonasson; Martin Lipp; Geir Christensen; Pål Aukrust; Ole Henning Skjønsberg

The chemokine receptor CCR7 regulates lymphocyte trafficking, and CCR7 deficiency induces infiltration of T and B cells adjacent to vessels in mouse lungs. Perivascular infiltration of T and B cells has also been found in human pulmonary arterial hypertension, and downregulation of the CCR7 receptor in circulating leukocytes of such patients has been observed. To investigate whether changes in the CCR7 system contribute to the pathogenesis of pulmonary hypertension, we utilized mice deficient of the CCR7 receptor. The cardiopulmonary and inflammatory responses of CCR7 depletion were evaluated in CCR7-deficient and wild-type mice. Measurements of cytokines upregulated in the animal model were also performed in patients with pulmonary hypertension and controls and in vascular smooth muscle cells. We found that mice lacking CCR7 had increased right ventricular systolic pressure, reduced pulmonary artery acceleration time, increased right ventricular/tibial length ratio, Rho kinase-mediated pulmonary vasoconstriction, and increased muscularization of distal arteries, indicating pulmonary hypertension. These mice also showed increased perivascular infiltration of leukocytes, consisting mainly of T and B cells, and increased mRNA levels of the inflammatory cytokines interleukin-12 and CX3CL1 within pulmonary tissue. Increased serum levels of interleukin-12 and CX3CL1 were also observed in patients with pulmonary hypertension, particularly in those with pulmonary hypertension associated with connective tissue disorder. In smooth muscle cells, interleukin-12 induced secretion of the angiogenic cytokine interleukin-8. We conclude that these results suggest a role for CCR7 in the development of pulmonary arterial hypertension, at least in some subgroups, possibly via pulmonary infiltration of lymphocytes and secretion of interleukin-12 and CX3CL1.


Experimental Lung Research | 2012

IL-18 and IL-12 synergy induces matrix degrading enzymes in the lung

Fadila Telarevic Cero; Vigdis Hillestad; Else Marit Løberg; Geir Christensen; Karl-Otto Larsen; Ole Henning Skjønsberg

ABSTRACT Interleukin (IL)-18 is a pro-inflammatory cytokine suggested to be involved in the development of pulmonary emphysema and inflammation. Studies involving immunology and cancer have revealed that IL-18 can have synergistic effects with IL-12. We have studied the presence of IL-18 and IL-12 receptors (IL-18R/IL-12R) in the lungs and whether IL-18 and IL-12, alone or in combination, have the ability to initiate the induction of mediators related to the development of emphysema and inflammation. The expression of the IL-18R was abundant in lungs compared to other organs (heart, liver, and spleen), and the IL-12R was also expressed in lung tissue. Mice treated with i.p. injection of recombinant murine IL-18 or IL-12 expressed significantly higher pulmonary mRNA levels of the matrix degrading enzymes metalloproteinase (MMP) 12 and cathepsin S, in addition to interferon-γ, tumor necrosis factor-α, and CXC chemokine ligand 9 (CXCL9) (all P < .05) than controls (received PBS). Treatment with IL-18 and IL-12 in combination showed an even more pronounced induction of these mediators, as well as a significant increase in MMP-9, IL-6, IL-1β, and transforming growth factor-β (P < .05). Furthermore, cellular apoptosis in lung tissue was induced. Immunohistochemical analysis revealed T-cell infiltration in pulmonary vessels following co-stimulation. In summary, IL-18 and IL-12 exert a synergistic effect on the lungs by inducing MMPs, cathepsins S, and pro-inflammatory cytokines, which may promote pulmonary emphysema and inflammation. The synergy between IL-18 and IL-12 involves infiltration of T-cells in the lungs, possibly induced by the T-cell chemoattractant CXCL9.


Acta Physiologica | 2015

IL-18 neutralization during alveolar hypoxia improves left ventricular diastolic function in mice.

Vigdis Hillestad; Emil K.S. Espe; Fadila Telarevic Cero; Karl-Otto Larsen; Ivar Sjaastad; Ståle Nygård; Ole Henning Skjønsberg; Geir Christensen

In patients, an association exists between pulmonary diseases and diastolic dysfunction of the left ventricle (LV). We have previously shown that alveolar hypoxia in mice induces LV diastolic dysfunction and that mice exposed to hypoxia have increased levels of circulating interleukin‐18 (IL‐18), suggesting involvement of IL‐18 in development of diastolic dysfunction. IL‐18 binding protein (IL‐18BP) is a natural inhibitor of IL‐18. In this study, we hypothesized that neutralization of IL‐18 during alveolar hypoxia would improve LV diastolic function.


American Journal of Physiology-heart and Circulatory Physiology | 2006

Alveolar hypoxia induces left ventricular diastolic dysfunction and reduces phosphorylation of phospholamban in mice.

Karl-Otto Larsen; Ivar Sjaastad; Aud Svindland; Kurt A. Krobert; Ole Henning Skjønsberg; Geir Christensen


Journal of Clinical Oncology | 2017

Thromboembolic events after high-intensity training during cisplatin-based chemotherapy for testicular cancer.

Lene Thorsen; Sophie D. Fosså; Hege Sagstuen Haugnes; Torgrim Tandstad; Marianne Brydøy; Torbjørn Wisløff; Elisabeth Edvardsen; Gunhild Maria Gjerset; Karl-Otto Larsen; Per Morten Sandset; Carola E. Henriksson; Helene Fs Negaard


European Respiratory Journal | 2017

Change in pulmonary function and cardiorespiratory fitness following chemotherapy in testicular cancer patients – a pilot study

Elisabeth Edvardsen; Karl-Otto Larsen; Sophie D. Fosså; Tone Ikdahl; Jon Håvard Loge; Lene Thorsen


European Respiratory Journal | 2017

A role for the inflammasome effector caspase-1 in hypoxia-induced pulmonary hypertension

Camilla Udjus; Fadila Telarevic Cero; Bente Halvorsen; Cathrine R. Carlson; Bård Bendiksen; Emil K.S. Espe; Ivar Sjåstad; Else Marit Løberg; Arne Yndestad; Pål Aukrust; Geir Christensen; Ole Henning Skjønsberg; Karl-Otto Larsen

Collaboration


Dive into the Karl-Otto Larsen's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Ivar Sjaastad

Oslo University Hospital

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
Researchain Logo
Decentralizing Knowledge