Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Exp Hematol Oncol ; 13(1): 59, 2024 Jun 03.
Article in English | MEDLINE | ID: mdl-38831452

ABSTRACT

Type I interferon (IFN) is a potent antitumoral drug, with an important history in the treatment of hematologic malignancies. However, its pleiotropic nature leads to severe dose-limiting toxicities that blunt its therapeutic potential. To achieve selective targeting of specific immune or tumor cells, AcTakines (Activity-on-Target Cytokines), i.e., immunocytokines utilizing attenuated cytokines, and clinically optimized A-Kines™ were developed. In syngeneic murine models, the CD20-targeted murine IFNα2-based AcTaferons (AFNs) have demonstrated clear antitumoral effects, with excellent tolerability. The current study explores the antitumoral potential of the humanized huCD20-Fc-AFN in 5 different humanized patient derived xenograft (PDX) models of huCD20+ aggressive B non-Hodgkin lymphomas (B-NHLs). The huCD20-Fc-AFN consists of a huCD20-specific single-domain antibody (VHH) linked through a heterodimeric 'knob-in-hole' human IgG1 Fc molecule to an attenuated huIFNα2 sequence. An in vitro targeting efficacy of up to 1.000-fold could be obtained, without detectable in vivo toxicities, except for selective (on-target) and reversible B cell depletion. Treatment with huCD20-Fc-AFN significantly increased the median overall survival (mOS) in both non-humanized (mOS 31 to 45 days; HR = 0.26; p = 0.001), and humanized NSG/NOG mice (mOS 34 to 80 days; HR = 0.37; p < 0.0001). In humanized mice, there was a trend for increased survival when compared to equimolar rituximab (mOS 49 to 80 days; HR = 0.73; p = 0.09). The antitumoral effects of huCD20-Fc-AFN were partly due to direct effects of type I IFN on the tumor cells, but additional effects via the human immune system are essential to obtain long-term remissions. To conclude, huCD20-Fc-AFN could provide a novel therapeutic strategy for huCD20-expressing aggressive B-NHLs.

2.
Mol Cancer ; 22(1): 191, 2023 11 29.
Article in English | MEDLINE | ID: mdl-38031106

ABSTRACT

Despite major improvements in immunotherapeutic strategies, the immunosuppressive tumor microenvironment remains a major obstacle for the induction of efficient antitumor responses. In this study, we show that local delivery of a bispecific Clec9A-PD-L1 targeted type I interferon (AcTaferon, AFN) overcomes this hurdle by reshaping the tumor immune landscape.Treatment with the bispecific AFN resulted in the presence of pro-immunogenic tumor-associated macrophages and neutrophils, increased motility and maturation profile of cDC1 and presence of inflammatory cDC2. Moreover, we report empowered diversity in the CD8+ T cell repertoire and induction of a shift from naive, dysfunctional CD8+ T cells towards effector, plastic cytotoxic T lymphocytes together with increased presence of NK and NKT cells as well as decreased regulatory T cell levels. These dynamic changes were associated with potent antitumor activity. Tumor clearance and immunological memory, therapeutic immunity on large established tumors and blunted tumor growth at distant sites were obtained upon co-administration of a non-curative dose of chemotherapy.Overall, this study illuminates further application of type I interferon as a safe and efficient way to reshape the suppressive tumor microenvironment and induce potent antitumor immunity; features which are of major importance in overcoming the development of metastases and tumor cell resistance to immune attack. The strategy described here has potential for application across to a broad range of cancer types.


Subject(s)
Interferon Type I , Neoplasms , Humans , CD8-Positive T-Lymphocytes , Interferon Type I/metabolism , Tumor Microenvironment , B7-H1 Antigen/metabolism , Neoplasms/metabolism , Immunotherapy , Cell Line, Tumor
3.
Sci Rep ; 11(1): 21575, 2021 11 03.
Article in English | MEDLINE | ID: mdl-34732771

ABSTRACT

Type I Interferon (IFN) was the very first drug approved for the treatment of Multiple Sclerosis (MS), and is still frequently used as a first line therapy. However, systemic IFN also causes considerable side effects, affecting therapy adherence and dose escalation. In addition, the mechanism of action of IFN in MS is multifactorial and still not completely understood. Using AcTaferons (Activity-on-Target IFNs, AFNs), optimized IFN-based immunocytokines that allow cell-specific targeting, we have previously demonstrated that specific targeting of IFN activity to dendritic cells (DCs) can protect against experimental autoimmune encephalitis (EAE), inducing in vivo tolerogenic protective effects, evidenced by increased indoleamine-2,3-dioxygenase (IDO) and transforming growth factor ß (TGFß) release by plasmacytoid (p) DCs and improved immunosuppressive capacity of regulatory T and B cells. We here report that targeting type I IFN activity specifically towards B cells also provides strong protection against EAE, and that targeting pDCs using SiglecH-AFN can significantly add to this protective effect. The superior protection achieved by simultaneous targeting of both B lymphocytes and pDCs correlated with improved IL-10 responses in B cells and conventional cDCs, and with a previously unseen very robust IDO response in several cells, including all B and T lymphocytes, cDC1 and cDC2.


Subject(s)
B-Lymphocytes/metabolism , Dendritic Cells/metabolism , Encephalomyelitis, Autoimmune, Experimental/therapy , Interferons/metabolism , Animals , Antibodies/chemistry , Biotechnology , Disease Progression , Immunosuppressive Agents , Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism , Interferon Type I/metabolism , Lymphocyte Count , Lymphocytes/cytology , Male , Mice , Mice, Inbred C57BL , Peptides/chemistry , Signal Transduction , T-Lymphocytes/immunology , Transforming Growth Factor beta/metabolism
4.
Cell Mol Life Sci ; 76(6): 1201-1214, 2019 Mar.
Article in English | MEDLINE | ID: mdl-30659329

ABSTRACT

Leptin links body energy stores to high energy demanding processes like reproduction and immunity. Based on leptin's role in autoimmune diseases and cancer, several leptin and leptin receptor (LR) antagonists have been developed, but these intrinsically lead to unwanted weight gain. Here, we report on the uncoupling of leptin's metabolic and immune functions based on the cross talk with the epidermal growth factor receptor (EGFR). We show that both receptors spontaneously interact and, remarkably, that this complex can partially overrule the lack of LR activation by a leptin antagonistic mutein. Moreover, this leptin mutant induces EGFR phosphorylation comparable to wild-type leptin. Exploiting this non-canonical leptin signalling pathway, we identified a camelid single-domain antibody that selectively inhibits this LR-EGFR cross talk without interfering with homotypic LR signalling. Administration in vivo showed that this single-domain antibody did not interfere with leptin's metabolic functions, but could reverse the leptin-driven protection against starvation-induced thymic and splenic atrophy. These findings offer new opportunities for the design and clinical application of selective leptin and LR antagonists that avoid unwanted metabolic side effects.


Subject(s)
Leptin/immunology , Leptin/metabolism , Receptors, Leptin/antagonists & inhibitors , Receptors, Leptin/metabolism , Single-Domain Antibodies/pharmacology , Animals , Camelids, New World/immunology , ErbB Receptors/genetics , ErbB Receptors/metabolism , Female , HEK293 Cells , Humans , Leptin/genetics , Ligands , Mice, Inbred C57BL , Mutation , Protein Binding/drug effects , Receptor Cross-Talk/drug effects , Receptors, Leptin/genetics , Signal Transduction
5.
J Autoimmun ; 97: 70-76, 2019 02.
Article in English | MEDLINE | ID: mdl-30467068

ABSTRACT

Type I Interferon (IFN) is widely used for multiple sclerosis (MS) treatment, but its side effects are limiting and its mechanism of action still unknown. Furthermore, 30-50% of MS patients are unresponsive, and IFN can even induce relapses. Fundamental understanding of the cellular target(s) of IFN will help to optimize treatments by reducing side effects and separating beneficial from detrimental effects. To improve clinical systemic IFN usage, we are developing AcTaferons (Activity-on-Target IFNs = AFNs), optimized IFN-based immunocytokines that allow cell-specific targeting. In experimental autoimmune encephalitis (EAE) in mice, high dose WT mIFNα could delay disease, but caused mortality and severe hematological deficits. In contrast, AFN targeted to dendritic cells (DC, via Clec9A) protected without mortality or hematological consequences. Conversely, CD8-targeted AFN did not protect and exacerbated weight loss, indicating the presence of both protective and unfavorable IFN effects in EAE. Comparing Clec9A-, XCR1-and SiglecH-targeting, we found that targeting AFN to plasmacytoid (p) and conventional (c) DC is superior and non-toxic compared to WT mIFN. DC-targeted AFN increased pDC numbers and their tolerogenic potential, evidenced by increased TGFß and IDO synthesis and regulatory T cell induction. In addition, both regulatory T and B cells produced significantly more immunosuppressive TGFß and IL-10. In conclusion, specific DC-targeting of IFN activity induces a robust in vivo tolerization, efficiently protecting against EAE, without noticeable side effects. Thus, dissecting positive and negative IFN effects via cell-specific targeting may result in better and safer MS therapy and response rates.


Subject(s)
Dendritic Cells/immunology , Dendritic Cells/metabolism , Encephalomyelitis, Autoimmune, Experimental/etiology , Encephalomyelitis, Autoimmune, Experimental/metabolism , Immune Tolerance , Interferons/metabolism , Animals , B7-H1 Antigen/metabolism , Biomarkers , CTLA-4 Antigen/metabolism , Disease Models, Animal , Disease Susceptibility , Encephalomyelitis, Autoimmune, Experimental/pathology , Male , Mice , Models, Biological
6.
Oncoimmunology ; 7(3): e1398876, 2018.
Article in English | MEDLINE | ID: mdl-29399401

ABSTRACT

Despite approval for the treatment of various malignancies, clinical application of cytokines such as type I interferon (IFN) is severely impeded by their systemic toxicity. AcTakines (Activity-on-Target cytokines) are optimized immunocytokines that, when injected in mice, only reveal their activity upon cell-specific impact. We here show that type I IFN-derived AcTaferon targeted to the tumor displays strong antitumor activity without any associated toxicity, in contrast with wild type IFN. Treatment with CD20-targeted AcTaferon of CD20+ lymphoma tumors or melanoma tumors engineered to be CD20+, drastically reduced tumor growth. This antitumor effect was completely lost in IFNAR- or Batf3-deficient mice, and depended on IFN signaling in conventional dendritic cells. Also the presence of, but not the IFN signaling in, CD8+ T lymphocytes was critical for proficient antitumor effects. When combined with immunogenic chemotherapy, low-dose TNF, or immune checkpoint blockade strategies such as anti-PDL1, anti-CTLA4 or anti-LAG3, complete tumor regressions and subsequent immunity (memory) were observed, still without any concomitant morbidity, again in sharp contrast with wild type IFN. Interestingly, the combination therapy of tumor-targeted AcTaferon with checkpoint inhibiting antibodies indicated its ability to convert nonresponding tumors into responders. Collectively, our findings demonstrate that AcTaferon targeted to tumor-specific surface markers may provide a safe and generic addition to cancer (immuno)therapies.

7.
Cancer Res ; 78(2): 463-474, 2018 01 15.
Article in English | MEDLINE | ID: mdl-29187401

ABSTRACT

An ideal generic cancer immunotherapy should mobilize the immune system to destroy tumor cells without harming healthy cells and remain active in case of recurrence. Furthermore, it should preferably not rely on tumor-specific surface markers, as these are only available in a limited set of malignancies. Despite approval for treatment of various cancers, clinical application of cytokines is still impeded by their multiple toxic side effects. Type I IFN has a long history in the treatment of cancer, but its multifaceted activity pattern and complex side effects prevent its clinical use. Here we develop AcTakines (Activity-on-Target cytokines), optimized (mutated) immunocytokines that are up to 1,000-fold more potent on target cells, allowing specific signaling in selected cell types only. Type I IFN-derived AcTaferon (AFN)-targeting Clec9A+ dendritic cells (DC) displayed strong antitumor activity in murine melanoma, breast carcinoma, and lymphoma models and against human lymphoma in humanized mice without any detectable toxic side effects. Combined with immune checkpoint blockade, chemotherapy, or low-dose TNF, complete tumor regression and long-lasting tumor immunity were observed, still without adverse effects. Our findings indicate that DC-targeted AFNs provide a novel class of highly efficient, safe, and broad-spectrum off-the-shelf cancer immunotherapeutics with no need for a tumor marker.Significance: Targeted type I interferon elicits powerful antitumor efficacy, similar to wild-type IFN, but without any toxic side effects. Cancer Res; 78(2); 463-74. ©2017 AACR.


Subject(s)
Cytokines/chemistry , Dendritic Cells/immunology , Immunotherapy , Interferon Type I/pharmacology , Mammary Neoplasms, Experimental/therapy , Melanoma, Experimental/therapy , Animals , Apoptosis , Cell Proliferation , Combined Modality Therapy , Cytokines/metabolism , Dendritic Cells/metabolism , Dendritic Cells/pathology , Female , Mammary Neoplasms, Experimental/immunology , Mammary Neoplasms, Experimental/metabolism , Mammary Neoplasms, Experimental/pathology , Melanoma, Experimental/immunology , Melanoma, Experimental/metabolism , Melanoma, Experimental/pathology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Tumor Cells, Cultured
8.
Nat Commun ; 6: 8482, 2015 Oct 07.
Article in English | MEDLINE | ID: mdl-26442659

ABSTRACT

Oxidative stress, a central mediator of cardiovascular disease, results in loss of the prosthetic haem group of soluble guanylate cyclase (sGC), preventing its activation by nitric oxide (NO). Here we introduce Apo-sGC mice expressing haem-free sGC. Apo-sGC mice are viable and develop hypertension. The haemodynamic effects of NO are abolished, but those of the sGC activator cinaciguat are enhanced in apo-sGC mice, suggesting that the effects of NO on smooth muscle relaxation, blood pressure regulation and inhibition of platelet aggregation require sGC activation by NO. Tumour necrosis factor (TNF)-induced hypotension and mortality are preserved in apo-sGC mice, indicating that pathways other than sGC signalling mediate the cardiovascular collapse in shock. Apo-sGC mice allow for differentiation between sGC-dependent and -independent NO effects and between haem-dependent and -independent sGC effects. Apo-sGC mice represent a unique experimental platform to study the in vivo consequences of sGC oxidation and the therapeutic potential of sGC activators.


Subject(s)
Cardiovascular System/metabolism , Guanylate Cyclase/genetics , Heme/genetics , Muscle, Smooth, Vascular/metabolism , Nitric Oxide/metabolism , Receptors, Cytoplasmic and Nuclear/genetics , Animals , Benzoates/pharmacology , Blood Pressure/drug effects , Cardiovascular System/drug effects , Gene Knock-In Techniques , Hypertension/genetics , Hypotension/chemically induced , Hypotension/genetics , Mice , Mice, Transgenic , Muscle, Smooth, Vascular/drug effects , Oxidative Stress/drug effects , Platelet Aggregation/drug effects , Soluble Guanylyl Cyclase , Tumor Necrosis Factor-alpha/pharmacology
9.
Crit Care Med ; 42(8): e560-9, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24717467

ABSTRACT

OBJECTIVE: Early detection and start of appropriate treatment are highly correlated with survival of sepsis and septic shock, but the currently available predictive tools are not sensitive enough to identify patients at risk. DESIGN: Linear (time and frequency domain) and nonlinear (unifractal and multiscale complexity dynamics) measures of beat-to-beat interval variability were analyzed in two mouse models of inflammatory shock to determine if they are sensitive enough to predict outcome. SETTING: University research laboratory. SUBJECTS: Blood pressure transmitter-implanted female C57BL/6J mice. INTERVENTIONS: IV administration of tumor necrosis factor (n = 11) or lipopolysaccharide (n = 14). MEASUREMENTS AND MAIN RESULTS: Contrary to linear indices of variability, unifractal dynamics, and absolute heart rate or blood pressure, quantification of complex beat-to-beat dynamics using multiscale entropy was able to predict survival outcome starting as early as 40 minutes after induction of inflammatory shock. Based on these results, a new and clinically relevant index of multiscale entropy was developed that scores the key features of a multiscale entropy profile. Contrary to multiscale entropy, multiscale entropy scoring can be followed as a function of time to monitor disease progression with limited loss of information. CONCLUSIONS: Analysis of multiscale complexity of beat-to-beat dynamics at high temporal resolution has potential as a sensitive prognostic tool with translational power that can predict survival outcome in systemic inflammatory conditions such as sepsis and septic shock.


Subject(s)
Entropy , Linear Models , Nonlinear Dynamics , Severity of Illness Index , Systemic Inflammatory Response Syndrome/diagnosis , Animals , Blood Pressure , Early Diagnosis , Female , Heart Rate , Lipopolysaccharides , Mice , Mice, Inbred C57BL , Prognosis , Survival Rate , Systemic Inflammatory Response Syndrome/chemically induced , Treatment Outcome , Tumor Necrosis Factor-alpha
10.
PLoS One ; 8(8): e72155, 2013.
Article in English | MEDLINE | ID: mdl-24015214

ABSTRACT

Sepsis and septic shock are associated with high mortality rates and the majority of sepsis patients die due to complications of multiple organ failure (MOF). The cyclic GMP (cGMP) producing enzyme soluble guanylate cyclase (sGC) is crucially involved in the regulation of (micro)vascular homeostasis, cardiac function and, consequently, organ function. However, it can become inactivated when exposed to reactive oxygen species (ROS). The resulting heme-free sGC can be reactivated by the heme- and nitric oxide (NO)-independent sGC activator BAY 58-2667 (Cinaciguat). We report that late (+8 h) post-treatment with BAY 58-2667 in a mouse model can protect against lethal endotoxic shock. Protection was associated with reduced hypothermia, circulating IL-6 levels, cardiomyocyte apoptosis, and mortality. In contrast to BAY 58-2667, the sGC stimulator BAY 41-2272 and the phosphodiesterase 5 inhibitor Sildenafil did not have any beneficial effect on survival, emphasizing the importance of the selectivity of BAY 58-2667 for diseased vessels and tissues. Hemodynamic parameters (blood pressure and heart rate) were decreased, and linear and nonlinear indices of blood pressure variability, reflective for (un)coupling of the communication between the autonomic nervous system and the heart, were improved after late protective treatment with BAY 58-2667. In conclusion, our results demonstrate the pivotal role of the NO/sGC axis in endotoxic shock. Stabilization of sGC function with BAY 58-2667 can prevent mortality when given in the correct treatment window, which probably depends on the dynamics of the heme-free sGC pool, in turn influenced by oxidative stress. We speculate that, considering the central role of sGC signaling in many pathways required for maintenance of (micro)circulatory homeostasis, BAY 58-2667 supports organ function by recoupling inter-organ communication pathways.


Subject(s)
Benzoates/pharmacology , Enzyme Activators/pharmacology , Shock, Septic/drug therapy , Animals , Apoptosis/drug effects , Blood Pressure/drug effects , Drug Evaluation, Preclinical , Drug Synergism , Female , Guanylate Cyclase , Heart Rate/drug effects , Lipopolysaccharides/pharmacology , Mice , Mice, Inbred C57BL , Myocardium/immunology , Myocardium/pathology , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/physiology , Phosphodiesterase 5 Inhibitors/pharmacology , Piperazines/pharmacology , Purines/pharmacology , Pyrazoles/pharmacology , Pyridines/pharmacology , Shock, Septic/immunology , Shock, Septic/physiopathology , Sildenafil Citrate , Sulfones/pharmacology
11.
Cardiovasc Res ; 100(1): 28-35, 2013 Oct 01.
Article in English | MEDLINE | ID: mdl-23787001

ABSTRACT

AIMS: Septic shock is the leading cause of death in intensive care units worldwide, resulting from a progressive systemic inflammatory reaction causing cardiovascular and organ failure. Nitric oxide (NO) is a potent vasodilator and inhibition of NO synthases (NOS) can increase blood pressure in septic shock. However, NOS inhibition does not improve outcome, on the contrary, and certain NO donors may even provide protection. In addition, NOS produce superoxide in case of substrate or cofactor deficiency or oxidation. We hypothesized that excessive systemic iNOS-derived NO production is insufficient to trigger cardiovascular failure and shock. METHODS AND RESULTS: We found that the systemic injection with various synthetic Toll-like receptor-2 (TLR2), TLR3, or TLR9 agonists triggered systemic NO production identical to that of lipopolysaccharide (LPS) or tumour necrosis factor. In contrast to the latter, however, these agonists did not cause hypothermia or any other signs of discomfort or morbidity, and inflammatory cytokine production was low. TLR2 stimulation with the triacylated lipopeptide Pam3CSK4 not only caused identical NO levels in circulation, but also identical iNOS expression patterns as LPS. Nevertheless, Pam3CSK4 did not cause hypotension, bradycardia, reduced blood flow, or inadequate tissue perfusion in the kidney or the liver. CONCLUSION: We demonstrate that excessive iNOS-derived NO in circulation is not necessarily linked to concomitant cardiovascular collapse, morbidity, or mortality. As such, our data indicate that the central role of iNOS-derived NO in inflammation-associated cardiovascular failure may be overestimated.


Subject(s)
Nitric Oxide/biosynthesis , Shock, Septic/etiology , Toll-Like Receptor 2/physiology , Animals , Cytokines/physiology , Female , Lipopolysaccharides/pharmacology , Mice , Mice, Inbred C57BL , Morbidity , Nitric Oxide Synthase/physiology , Systemic Inflammatory Response Syndrome/etiology
12.
J Mol Med (Berl) ; 88(9): 921-30, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20496172

ABSTRACT

Septic shock is associated with life-threatening vasodilation and hypotension. To cause vasodilation, vascular endothelium may release nitric oxide (NO), prostacyclin (PGI2), and the elusive endothelium-derived hyperpolarizing factor (EDHF). Although NO is critical in controlling vascular tone, inhibiting NO in septic shock does not improve outcome, on the contrary, precipitating the search for alternative therapeutic targets. Using a hyperacute tumor necrosis factor (TNF)-induced shock model in mice, we found that shock can develop independently of the known vasodilators NO, cGMP, PGI2, or epoxyeicosatrienoic acids. However, the antioxidant tempol efficiently prevented hypotension, bradycardia, hypothermia, and mortality, indicating the decisive involvement of reactive oxygen species (ROS) in these phenomena. Also, in classical TNF or lipopolysaccharide-induced shock models, tempol protected significantly. Experiments with (cell-permeable) superoxide dismutase or catalase, N-acetylcysteine and apocynin suggest that the ROS-dependent shock depends on intracellular (*)OH radicals. Potassium channels activated by ATP (K(ATP)) or calcium (K(Ca)) are important mediators of vascular relaxation. While NO and PGI2-induced vasodilation involves K(ATP) and large-conductance BK(Ca) channels, small-conductance SK(Ca) channels mediate vasodilation induced by EDHF. Interestingly, also SK(Ca) inhibition completely prevented the ROS-dependent shock. Our data thus indicate that intracellular (*)OH and SK(Ca) channels represent interesting new therapeutic targets for inflammatory shock. Moreover, they may also explain why antioxidants other than tempol fail to provide survival benefit during shock.


Subject(s)
Hypotension/etiology , Reactive Oxygen Species/metabolism , Shock, Septic/etiology , Small-Conductance Calcium-Activated Potassium Channels/metabolism , Animals , Biological Factors/metabolism , Female , Hypotension/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Nitric Oxide/metabolism , Shock, Septic/metabolism , Tumor Necrosis Factor-alpha/metabolism
13.
Cardiovasc Res ; 79(1): 179-86, 2008 Jul 01.
Article in English | MEDLINE | ID: mdl-18339647

ABSTRACT

AIM: The effects of nitric oxide (NO) in the cardiovascular system are attributed in part to cGMP synthesis by the alpha1beta1 isoform of soluble guanylate cyclase (sGC). Because available sGC inhibitors are neither enzyme- nor isoform-specific, we generated knockout mice for the alpha1 subunit (sGCalpha1(-/-) mice) in order to investigate the function of sGCalpha1beta1 in the regulation of blood pressure and cardiac function. METHODS AND RESULTS: Blood pressure was evaluated, using both non-invasive and invasive haemodynamic techniques, in intact and gonadectomized male and female sGCalpha1(-/-) and wild-type (WT) mice. Cardiac function was assessed with a conductance catheter inserted in the left ventricle of male and female sGCalpha1(-/-) and WT mice. Male sGCalpha1(-/-) mice developed hypertension (147 +/- 2 mmHg), whereas female sGCalpha1(-/-) mice did not (115 +/- 2 mmHg). Orchidectomy and treatment with an androgen receptor antagonist prevented hypertension, while ovariectomy did not influence the phenotype. Chronic testosterone treatment increased blood pressure in ovariectomized sGCalpha1(-/-) mice but not in WT mice. The NO synthase inhibitor Nomega-nitro-L-arginine methyl ester hydrochloride raised blood pressure similarly in male and female WT and sGCalpha1(-/-) mice. The ability of NO donor compounds to reduce blood pressure was slightly attenuated in sGCalpha1(-/-) male and female mice as compared to WT mice. The direct sGC stimulator BAY 41-2272 reduced blood pressure only in WT mice. Increased cardiac contractility and arterial elastance as well as impaired ventricular relaxation were observed in both male and female sGCalpha1(-/-) mice. CONCLUSION: These findings demonstrate that sGCalpha1beta1-derived cGMP signalling has gender-specific and testosterone-dependent cardiovascular effects and reveal that the effects of NO on systemic blood pressure do not require sGCalpha1beta1.


Subject(s)
Blood Pressure/physiology , Guanylate Cyclase/metabolism , Hypertension/metabolism , Nitric Oxide/physiology , Receptors, Cytoplasmic and Nuclear/metabolism , Sex Characteristics , Animals , Cyclic GMP/metabolism , Female , Guanylate Cyclase/genetics , Hypertension/physiopathology , Isoenzymes , Male , Mice , Myocardial Contraction/physiology , Orchiectomy , Ovariectomy , Pyrazoles/pharmacology , Pyridines/pharmacology , Receptors, Cytoplasmic and Nuclear/genetics , Signal Transduction/physiology , Soluble Guanylyl Cyclase , Testosterone/metabolism , Vascular Resistance/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...