Professional Education


  • Doctor of Philosophy, University Of Helsinki (2019)
  • Doctor of Medicine, Ludwig Maximilian Universitat Munchen (2016)
  • Doctor of Philosophy (PhD), University of Helsinki, Medicine (2019)
  • Doctor of Medicine (MD), Ludwig-Maximilians-Universität München, Medicine (2016)

Stanford Advisors


All Publications


  • CYP2C19 loss-of-function alleles and use of omeprazole or esomeprazole increase the risk of cardiovascular outcomes in patients using clopidogrel. Clinical and translational science Ramste, M., Ritvos, M., Hayrynen, S., Kiiski, J. I., Niemi, M., Sinisalo, J. 2023

    Abstract

    Our aim was to investigate in a real-life prospective patient cohort how CYP2C19 loss-of-function (LOF) variants and CYP2C19 inhibitor omeprazole or esomeprazole influence the incidence of cardiovascular events in patients using clopidogrel. Data based simultaneously on these factors are conflicting and sparse. A cohort of prospective patients (n=1972) with acute coronary syndrome (ACS) (n=1302) or symptomatic chronic coronary disease (n=656) was followed for 365days after hospitalization with information on purchased prescription drugs, hospital discharge, death and genotype for CYP2C19*2, CYP2C19*3, and CYP2C19*8 LOF variants. The primary study outcome measurement was cardiovascular death or recurring myocardial infarction or stroke. Altogether, 608 patients (30.8%) carried CYP2C19 LOF alleles. During the 365-day follow-up 252 patients (12.8%) had an ischemic vascular event. Cardiovascular events were significantly more frequent in carriers of CYP2C19 LOF alleles [14.8% (95% CI, 11.7-17.8)] than in non-carriers [10.8% (95% CI, 9.0-12.6)] (P = .0159). Omeprazole or esomeprazole use was similar among LOF allele carriers (n=131, 21.5%) and non-carriers (n=250, 18.3%) (P = .185). Cardiovascular events were significantly more common in a composite group consisting of all CYP2C19 LOF carriers regardless of PPI use status and non-carriers using omeprazole or esomeprazole than in non-carriers not using omeprazole or esomeprazole: 14.8% (95% CI, 12.2-17.3) vs. 9.9% (95% CI, 8.0-11.9) (P = .00173). We observed significantly more cardiovascular events in carriers of CYP2C19 LOF variants and in non-carriers using omeprazole or esomeprazole. For optimal patient care, both genetics and concomitant medication should be considered.

    View details for DOI 10.1111/cts.13608

    View details for PubMedID 37551775

  • Genetic predictors of lifelong medication-use patterns in cardiometabolic diseases. Nature medicine Kiiskinen, T., Helkkula, P., Krebs, K., Karjalainen, J., Saarentaus, E., Mars, N., Lehisto, A., Zhou, W., Cordioli, M., Jukarainen, S., Ramo, J. T., Mehtonen, J., Veerapen, K., Rasanen, M., Ruotsalainen, S., Maasha, M., FinnGen, Niiranen, T., Tuomi, T., Salomaa, V., Kurki, M., Pirinen, M., Palotie, A., Daly, M., Ganna, A., Havulinna, A. S., Milani, L., Ripatti, S. 2023

    Abstract

    Little is known about the genetic determinants of medication use in preventing cardiometabolic diseases. Using the Finnish nationwide drug purchase registry with follow-up since 1995, we performed genome-wide association analyses of longitudinal patterns of medication use in hyperlipidemia, hypertension and type 2 diabetes in up to 193,933 individuals (55% women) in the FinnGen study. In meta-analyses of up to 567,671 individuals combining FinnGen with the Estonian Biobank and the UK Biobank, we discovered 333 independent loci (P<5*10-9) associated with medication use. Fine-mapping revealed 494 95% credible sets associated with the total number of medication purchases, changes in medication combinations or treatment discontinuation, including 46 credible sets in 40 loci not associated with the underlying treatment targets. The polygenic risk scores (PRS) for cardiometabolic risk factors were strongly associated with the medication-use behavior. A medication-use enhanced multitrait PRS for coronary artery disease matched the performance of a risk factor-based multitrait coronary artery disease PRS in an independent sample (UK Biobank, n=343,676). In summary, we demonstrate medication-based strategies for identifying cardiometabolic risk loci and provide genome-wide tools for preventing cardiovascular diseases.

    View details for DOI 10.1038/s41591-022-02122-5

    View details for PubMedID 36653479

  • The SARS-CoV-2 receptor ACE2 is expressed in mouse pericytes but not endothelial cells: Implications for COVID-19 vascular research. Stem cell reports Muhl, L., He, L., Sun, Y., Andaloussi Mae, M., Pietila, R., Liu, J., Genove, G., Zhang, L., Xie, Y., Leptidis, S., Mocci, G., Stritt, S., Osman, A., Anisimov, A., Hemanthakumar, K. A., Rasanen, M., Hansson, E. M., Bjorkegren, J., Vanlandewijck, M., Blomgren, K., Makinen, T., Peng, X., Hu, Y., Ernfors, P., Arnold, T. D., Alitalo, K., Lendahl, U., Betsholtz, C. 2022

    Abstract

    Humanized mouse models and mouse-adapted SARS-CoV-2 virus are increasingly used to study COVID-19 pathogenesis, so it is important to learn where the SARS-CoV-2 receptor ACE2 is expressed. Here we mapped ACE2 expression during mouse postnatal development and in adulthood. Pericytes in the CNS, heart, and pancreas express ACE2 strongly, as do perineurial and adrenal fibroblasts, whereas endothelial cells do not at any location analyzed. In a number of other organs, pericytes do not express ACE2, including in the lung where ACE2 instead is expressed in bronchial epithelium and alveolar type II cells. The onset of ACE2 expression is organ specific: in bronchial epithelium already at birth, in brain pericytes before, and in heart pericytes after postnatal day 10.5. Establishing the vascular localization of ACE2 expression is central to correctly interpret data from modeling COVID-19 in the mouse and may shed light on the cause of vascular COVID-19 complications.

    View details for DOI 10.1016/j.stemcr.2022.03.016

    View details for PubMedID 35452595

  • Cardiac-specific VEGFB overexpression reduces lipoprotein lipase activity and improves insulin action in rat heart AMERICAN JOURNAL OF PHYSIOLOGY-ENDOCRINOLOGY AND METABOLISM Shang, R., Lal, N., Lee, C., Zhai, Y., Puri, K., Seira, O., Boushel, R. C., Sultan, I., Rasanen, M., Alitalo, K., Hussein, B., Rodrigues, B. 2021; 321 (6): E753-E765

    Abstract

    Cardiac muscle uses multiple sources of energy including glucose and fatty acid (FA). The heart cannot synthesize FA and relies on obtaining it from other sources, with lipoprotein lipase (LPL) breakdown of lipoproteins suggested to be a key source of FA for cardiac use. Recent work has indicated that cardiac vascular endothelial growth factor B (VEGFB) overexpression expands the coronary vasculature and facilitates metabolic reprogramming that favors glucose utilization. We wanted to explore whether this influence of VEGFB on cardiac metabolism involves regulation of LPL activity with consequent effects on lipotoxicity and insulin signaling. The transcriptomes of rats with and without cardiomyocyte-specific overexpression of human VEGFB were compared by using RNA sequencing. Isolated perfused hearts or cardiomyocytes incubated with heparin were used to enable measurement of LPL activity. Untargeted metabolomic analysis was performed for quantification of cardiac lipid metabolites. Cardiac insulin sensitivity was evaluated using fast-acting insulin. Isolated heart and cardiomyocytes were used to determine transgene-encoded VEGFB isoform secretion patterns and mitochondrial oxidative capacity using high-resolution respirometry and extracellular flux analysis. In vitro, transgenic cardiomyocytes incubated overnight and thus exposed to abundantly secreted VEGFB isoforms, in the absence of any in vivo confounding regulators of cardiac metabolism, demonstrated higher basal oxygen consumption. In the whole heart, VEGFB overexpression induced an angiogenic response that was accompanied by limited cardiac LPL activity through multiple mechanisms. This was associated with a lowered accumulation of lipid intermediates, diacylglycerols and lysophosphatidylcholine, that are known to influence insulin action. In response to exogenous insulin, transgenic hearts demonstrated increased insulin sensitivity. In conclusion, the interrogation of VEGFB function on cardiac metabolism uncovered an intriguing and previously unappreciated effect to lower LPL activity and prevent lipid metabolite accumulation to improve insulin action. VEGFB could be a potential cardioprotective therapy to treat metabolic disorders, for example, diabetes.NEW & NOTEWORTHY In hearts overexpressing vascular endothelial growth factor B (VEGFB), besides its known angiogenic response, multiple regulatory mechanisms lowered coronary LPL. This was accompanied by limited cardiac lipid metabolite accumulation with an augmentation of cardiac insulin action. Our data for the first time links VEGFB to coronary LPL in regulation of cardiac metabolism. VEGFB may be cardioprotective in metabolic disorders like diabetes.

    View details for DOI 10.1152/ajpendo.00219.2021

    View details for Web of Science ID 000733736100002

    View details for PubMedID 34747201

  • VEGF-B Promotes Endocardium-Derived Coronary Vessel Development and Cardiac Regeneration CIRCULATION Rasanen, M., Sultan, I., Paech, J., Hemanthakumar, K., Yu, W., He, L., Tang, J., Sun, Y., Hlushchuk, R., Huan, X., Armstrong, E., Khoma, O., Mervaala, E., Djonov, V., Betsholtz, C., Zhou, B., Kivela, R., Alitalo, K. 2021; 143 (1): 65-77

    Abstract

    Recent discoveries have indicated that, in the developing heart, sinus venosus and endocardium provide major sources of endothelium for coronary vessel growth that supports the expanding myocardium. Here we set out to study the origin of the coronary vessels that develop in response to vascular endothelial growth factor B (VEGF-B) in the heart and the effect of VEGF-B on recovery from myocardial infarction.We used mice and rats expressing a VEGF-B transgene, VEGF-B-gene-deleted mice and rats, apelin-CreERT, and natriuretic peptide receptor 3-CreERT recombinase-mediated genetic cell lineage tracing and viral vector-mediated VEGF-B gene transfer in adult mice. Left anterior descending coronary vessel ligation was performed, and 5-ethynyl-2'-deoxyuridine-mediated proliferating cell cycle labeling; flow cytometry; histological, immunohistochemical, and biochemical methods; single-cell RNA sequencing and subsequent bioinformatic analysis; microcomputed tomography; and fluorescent- and tracer-mediated vascular perfusion imaging analyses were used to study the development and function of the VEGF-B-induced vessels in the heart.We show that cardiomyocyte overexpression of VEGF-B in mice and rats during development promotes the growth of novel vessels that originate directly from the cardiac ventricles and maintain connection with the coronary vessels in subendocardial myocardium. In adult mice, endothelial proliferation induced by VEGF-B gene transfer was located predominantly in the subendocardial coronary vessels. Furthermore, VEGF-B gene transduction before or concomitantly with ligation of the left anterior descending coronary artery promoted endocardium-derived vessel development into the myocardium and improved cardiac tissue remodeling and cardiac function.The myocardial VEGF-B transgene promotes the formation of endocardium-derived coronary vessels during development, endothelial proliferation in subendocardial myocardium in adult mice, and structural and functional rescue of cardiac tissue after myocardial infarction. VEGF-B could provide a new therapeutic strategy for cardiac neovascularization after coronary occlusion to rescue the most vulnerable myocardial tissue.

    View details for DOI 10.1161/CIRCULATIONAHA.120.050635

    View details for Web of Science ID 000639303400011

    View details for PubMedID 33203221

  • Susceptibility to Cardiac Arrhythmias and Sympathetic Nerve Growth in VEGF-B Overexpressing Myocardium MOLECULAR THERAPY Lahteenvuo, J., Hatinen, O., Kuivanen, A., Huusko, J., Paananen, J., Lahteenvuo, M., Nurro, J., Hedman, M., Hartikainen, J., Laham-Karam, N., Makinen, P., Rasanen, M., Alitalo, K., Rosenzweig, A., Yla-Herttuala, S. 2020; 28 (7): 1731-1740

    Abstract

    VEGF-B gene therapy is a promising proangiogenic treatment for ischemic heart disease, but, unexpectedly, we found that high doses of VEGF-B promote ventricular arrhythmias (VAs). VEGF-B knockout, alpha myosin heavy-chain promoter (αMHC)-VEGF-B transgenic mice, and pigs transduced intramyocardially with adenoviral (Ad)VEGF- B186 were studied. Immunostaining showed a 2-fold increase in the number of nerves per field (76 vs. 39 in controls, p < 0.001) and an abnormal nerve distribution in the hypertrophic hearts of 11- to 20-month-old αMHC-VEGF-B mice. AdVEGF-B186 gene transfer (GT) led to local sprouting of nerve endings in pig myocardium (141 vs. 78 nerves per field in controls, p < 0.05). During dobutamine stress, 60% of the αMHC-VEGF-B hypertrophic mice had arrhythmias as compared to 7% in controls, and 20% of the AdVEGF-B186-transduced pigs and 100% of the combination of AdVEGF-B186- and AdsVEGFR-1-transduced pigs displayed VAs and even ventricular fibrillation. AdVEGF-B186 GT significantly increased the risk of sudden cardiac death in pigs when compared to any other GT with different VEGFs (hazard ratio, 500.5; 95% confidence interval [CI] 46.4-5,396.7; p < 0.0001). In gene expression analysis, VEGF-B induced the upregulation of Nr4a2, ATF6, and MANF in cardiomyocytes, molecules previously linked to nerve growth and differentiation. Thus, high AdVEGF-B186 overexpression induced nerve growth in the adult heart via a VEGFR-1 signaling-independent mechanism, leading to an increased risk of VA and sudden cardiac death.

    View details for DOI 10.1016/j.ymthe.2020.03.011

    View details for Web of Science ID 000548374800021

    View details for PubMedID 32243833

    View details for PubMedCentralID PMC7335717

  • Systemic Blockade of ACVR2B Ligands Protects Myocardium from Acute Ischemia-Reperfusion Injury MOLECULAR THERAPY Magga, J., Vainio, L., Kilpio, T., Hulmi, J. J., Taponen, S., Lin, R., Rasanen, M., Szabo, Z., Gao, E., Rahtu-Korpela, L., Alakoski, T., Ulvila, J., Laitinen, M., Pasternack, A., Koch, W. J., Alitalo, K., Kivela, R., Ritvos, O., Kerkela, R. 2019; 27 (3): 600-610

    Abstract

    Activin A and myostatin, members of the transforming growth factor (TGF)-β superfamily of secreted factors, are potent negative regulators of muscle growth, but their contribution to myocardial ischemia-reperfusion (IR) injury is not known. The aim of this study was to investigate if activin 2B (ACVR2B) receptor ligands contribute to myocardial IR injury. Mice were treated with soluble ACVR2B decoy receptor (ACVR2B-Fc) and subjected to myocardial ischemia followed by reperfusion for 6 or 24 h. Systemic blockade of ACVR2B ligands by ACVR2B-Fc was protective against cardiac IR injury, as evidenced by reduced infarcted area, apoptosis, and autophagy and better preserved LV systolic function following IR. ACVR2B-Fc modified cardiac metabolism, LV mitochondrial respiration, as well as cardiac phenotype toward physiological hypertrophy. Similar to its protective role in IR injury in vivo, ACVR2B-Fc antagonized SMAD2 signaling and cell death in cardiomyocytes that were subjected to hypoxic stress. ACVR2B ligand myostatin was found to exacerbate hypoxic stress. In addition to acute cardioprotection in ischemia, ACVR2B-Fc provided beneficial effects on cardiac function in prolonged cardiac stress in cardiotoxicity model. By blocking myostatin, ACVR2B-Fc potentially reduces cardiomyocyte death and modifies cardiomyocyte metabolism for hypoxic conditions to protect the heart from IR injury.

    View details for DOI 10.1016/j.ymthe.2019.01.013

    View details for Web of Science ID 000460402000013

    View details for PubMedID 30765322

    View details for PubMedCentralID PMC6404100

  • Single-cell RNA sequencing of mouse brain and lung vascular and vessel-associated cell types SCIENTIFIC DATA He, L., Vanlandewijck, M., Mae, M., Andrae, J., Ando, K., Del Gaudio, F., Nahar, K., Lebouvier, T., Lavina, B., Gouveia, L., Sun, Y., Raschperger, E., Segerstolpe, A., Liu, J., Gustafsson, S., Rasanen, M., Zarb, Y., Mochizuki, N., Keller, A., Lendahl, U., Betsholtz, C. 2018; 5: 180160

    Abstract

    Vascular diseases are major causes of death, yet our understanding of the cellular constituents of blood vessels, including how differences in their gene expression profiles create diversity in vascular structure and function, is limited. In this paper, we describe a single-cell RNA sequencing (scRNA-seq) dataset that defines vascular and vessel-associated cell types and subtypes in mouse brain and lung. The dataset contains 3,436 single cell transcriptomes from mouse brain, which formed 15 distinct clusters corresponding to cell (sub)types, and another 1,504 single cell transcriptomes from mouse lung, which formed 17 cell clusters. In order to allow user-friendly access to our data, we constructed a searchable database (http://betsholtzlab.org/VascularSingleCells/database.html). Our dataset constitutes a comprehensive molecular atlas of vascular and vessel-associated cell types in the mouse brain and lung, and as such provides a strong foundation for future studies of vascular development and diseases.

    View details for DOI 10.1038/sdata.2018.160

    View details for Web of Science ID 000442285500001

    View details for PubMedID 30129931

    View details for PubMedCentralID PMC6103262

  • Frequency, Reasons, and Impact of Premature Ticagrelor Discontinuation in Patients Undergoing Coronary Revascularization in Routine Clinical Practice Results From the Bern Percutaneous Coronary Intervention Registry CIRCULATION-CARDIOVASCULAR INTERVENTIONS Zanchin, T., Temperli, F., Karagiannis, A., Zanchin, C., Rasanen, M., Koskinas, K. C., Stortecky, S., Hunziker, L., Praz, F., Blochlinger, S., Moro, C., Moschovitis, A., Seiler, C., Billinger, M., Heg, D., Pilgrim, T., Valgimigli, M., Windecker, S., Raber, L. 2018; 11 (5): e006132

    Abstract

    Although ticagrelor has improved clinical outcomes among patients with acute coronary syndrome compared with clopidogrel, adherence to this new antiplatelet agent in real-world practice has not been fully investigated.Between November 2011 and June 2014, 1278 of 4831 consecutive patients (26.5%) undergoing percutaneous coronary intervention at a tertiary care center were treated with ticagrelor. Premature ticagrelor cessation was categorized into (1) change, when ticagrelor was replaced by prasugrel; (2) de-escalation, when ticagrelor was replaced by clopidogrel; and (3) premature discontinuation, when ticagrelor was discontinued without P2Y12 inhibitor replacement. Of 1278 patients treated with ticagrelor, premature treatment cessation occurred in 212 patients (17%). De-escalation to clopidogrel was the most frequent scenario (57%; n=120), followed by premature discontinuation (28%; n=60) and change to prasugrel (15%; n=32). Reasons for ticagrelor cessation included adverse effects (49%), initiation of oral anticoagulation (19%), and unspecified general practitioner preference (10%). Most frequent adverse effects leading to premature ticagrelor cessation were bleeding (41%), dyspnea (29%), and gastrointestinal symptoms (18%). Premature ticagrelor cessation was not associated with an increased risk of cardiac death, myocardial infarction, or stroke (hazard ratio, 0.73; 95% confidence interval: 0.40-1.32; P=0.29).Premature ticagrelor cessation in routine clinical practice occurred in 1 of 6 patients and was primarily related to adverse effects among which bleeding and dyspnea were the most frequent. Although premature ticagrelor cessation was not associated with adverse cardiovascular outcomes, this finding requires careful interpretation in view of the modest sample size.URL: https://www.clinicaltrials.gov. Unique identifier: NCT02241291.

    View details for DOI 10.1161/CIRCINTERVENTIONS.117.006132

    View details for Web of Science ID 000435567100002

    View details for PubMedID 29748219

  • Prevention of chemotherapy-induced cachexia by ACVR2B ligand blocking has different effects on heart and skeletal muscle JOURNAL OF CACHEXIA SARCOPENIA AND MUSCLE Hulmi, J. J., Nissinen, T. A., Rasanen, M., Degerman, J., Lautaoja, J. H., Hemanthakumar, K., Backman, J. T., Ritvos, O., Silvennoinen, M., Kivela, R. 2018; 9 (2): 417-432

    Abstract

    Toxicity of chemotherapy on skeletal muscles and the heart may significantly contribute to cancer cachexia, mortality, and decreased quality of life. Doxorubicin (DOX) is an effective cytostatic agent, which unfortunately has toxic effects on many healthy tissues. Blocking of activin receptor type IIB (ACVR2B) ligands is an often used strategy to prevent skeletal muscle loss, but its effects on the heart are relatively unknown.The effects of DOX treatment with or without pre-treatment with soluble ACVR2B-Fc (sACVR2B-Fc) were investigated. The mice were randomly assigned into one of the three groups: (1) vehicle (PBS)-treated controls, (2) DOX-treated mice (DOX), and (3) DOX-treated mice administered with sACVR2B-Fc during the experiment (DOX + sACVR2B-Fc). DOX was administered with a cumulative dose of 24 mg/kg during 2 weeks to investigate cachexia outcome in the heart and skeletal muscle. To understand similarities and differences between skeletal and cardiac muscles in their responses to chemotherapy, the tissues were collected 20 h after a single DOX (15 mg/kg) injection and analysed with genome-wide transcriptomics and mRNA and protein analyses. The combination group was pre-treated with sACVR2B-Fc 48 h before DOX administration. Major findings were also studied in mice receiving only sACVR2B-Fc.The DOX treatment induced similar (~10%) wasting in skeletal muscle and the heart. However, transcriptional changes in response to DOX were much greater in skeletal muscle. Pathway analysis and unbiased transcription factor analysis showed that p53-p21-REDD1 is the main common pathway activated by DOX in both skeletal and cardiac muscles. These changes were attenuated by blocking ACVR2B ligands especially in skeletal muscle. Tceal7 (3-fold to 5-fold increase), transferrin receptor (1.5-fold increase), and Ccl21 (0.6-fold to 0.9-fold decrease) were identified as novel genes responsive to blocking ACVR2B ligands. Overall, at the transcriptome level, ACVR2B ligand blocking had only minor influence in the heart while it had marked effects in skeletal muscle. The same was also true for the effects on tissue wasting. This may be explained in part by about 18-fold higher gene expression of myostatin in skeletal muscle compared with the heart.Cardiac and skeletal muscles display similar atrophy after DOX treatment, but the mechanisms for this may differ between the tissues. The present results suggest that p53-p21-REDD1 signalling is the main common DOX-activated pathway in these tissues and that blocking activin receptor ligands attenuates this response, especially in skeletal muscle supporting the overall stronger effects of this treatment in skeletal muscles.

    View details for DOI 10.1002/jcsm.12265

    View details for Web of Science ID 000428885800018

    View details for PubMedID 29230965

    View details for PubMedCentralID PMC5879968

  • A molecular atlas of cell types and zonation in the brain vasculature NATURE Vanlandewijck, M., He, L., Mae, M., Andrae, J., Ando, K., Del Gaudio, F., Nahar, K., Lebouvier, T., Lavina, B., Gouveia, L., Sun, Y., Raschperger, E., Rasanen, M., Zarb, Y., Mochizuki, N., Keller, A., Lendahl, U., Betsholtz, C. 2018; 554 (7693): 475-+

    Abstract

    Cerebrovascular disease is the third most common cause of death in developed countries, but our understanding of the cells that compose the cerebral vasculature is limited. Here, using vascular single-cell transcriptomics, we provide molecular definitions for the principal types of blood vascular and vessel-associated cells in the adult mouse brain. We uncover the transcriptional basis of the gradual phenotypic change (zonation) along the arteriovenous axis and reveal unexpected cell type differences: a seamless continuum for endothelial cells versus a punctuated continuum for mural cells. We also provide insight into pericyte organotypicity and define a population of perivascular fibroblast-like cells that are present on all vessel types except capillaries. Our work illustrates the power of single-cell transcriptomics to decode the higher organizational principles of a tissue and may provide the initial chapter in a molecular encyclopaedia of the mammalian vasculature.

    View details for DOI 10.1038/nature25739

    View details for Web of Science ID 000425597400036

    View details for PubMedID 29443965

  • VEGF-B gene therapy inhibits doxorubicin-induced cardiotoxicity by endothelial protection PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCES OF THE UNITED STATES OF AMERICA Rasanen, M., Degerman, J., Nissinen, T. A., Miinalainen, I., Kerkela, R., Siltanen, A., Backman, J. T., Mervaala, E., Hulmi, J. J., Kivela, R., Alitalo, K. 2016; 113 (46): 13144-13149

    Abstract

    Congestive heart failure is one of the leading causes of disability in long-term survivors of cancer. The anthracycline antibiotic doxorubicin (DOX) is used to treat a variety of cancers, but its utility is limited by its cumulative cardiotoxicity. As advances in cancer treatment have decreased cancer mortality, DOX-induced cardiomyopathy has become an increasing problem. However, the current means to alleviate the cardiotoxicity of DOX are limited. We considered that vascular endothelial growth factor-B (VEGF-B), which promotes coronary arteriogenesis, physiological cardiac hypertrophy, and ischemia resistance, could be an interesting candidate for prevention of DOX-induced cardiotoxicity and congestive heart failure. To study this, we administered an adeno-associated viral vector expressing VEGF-B or control vector to normal and tumor-bearing mice 1 wk before DOX treatment, using doses mimicking the concentrations used in the clinics. VEGF-B treatment completely inhibited the DOX-induced cardiac atrophy and whole-body wasting. VEGF-B also prevented capillary rarefaction in the heart and improved endothelial function in DOX-treated mice. VEGF-B also protected cultured endothelial cells from apoptosis and restored their tube formation. VEGF-B increased left ventricular volume without compromising cardiac function, reduced the expression of genes associated with pathological remodeling, and improved cardiac mitochondrial respiration. Importantly, VEGF-B did not affect serum or tissue concentrations of DOX or augment tumor growth. By inhibiting DOX-induced endothelial damage, VEGF-B could provide a novel therapeutic possibility for the prevention of chemotherapy-associated cardiotoxicity in cancer patients.

    View details for DOI 10.1073/pnas.1616168113

    View details for Web of Science ID 000388970100074

    View details for PubMedID 27799559

    View details for PubMedCentralID PMC5135329

  • Systemic blockade of ACVR2B ligands prevents chemotherapy-induced muscle wasting by restoring muscle protein synthesis without affecting oxidative capacity or atrogenes SCIENTIFIC REPORTS Nissinen, T. A., Degerman, J., Rasanen, M., Poikonen, A. R., Koskinen, S., Mervaala, E., Pasternack, A., Ritvos, O., Kivela, R., Hulmi, J. J. 2016; 6: 32695

    Abstract

    Doxorubicin is a widely used and effective chemotherapy drug. However, cardiac and skeletal muscle toxicity of doxorubicin limits its use. Inhibiting myostatin/activin signalling can prevent muscle atrophy, but its effects in chemotherapy-induced muscle wasting are unknown. In the present study we investigated the effects of doxorubicin administration alone or combined with activin receptor ligand pathway blockade by soluble activin receptor IIB (sACVR2B-Fc). Doxorubicin administration decreased body mass, muscle size and bone mineral density/content in mice. However, these effects were prevented by sACVR2B-Fc administration. Unlike in many other wasting situations, doxorubicin induced muscle atrophy without markedly increasing typical atrogenes or protein degradation pathways. Instead, doxorubicin decreased muscle protein synthesis which was completely restored by sACVR2B-Fc. Doxorubicin administration also resulted in impaired running performance without effects on skeletal muscle mitochondrial capacity/function or capillary density. Running performance and mitochondrial function were unaltered by sACVR2B-Fc administration. Tumour experiment using Lewis lung carcinoma cells demonstrated that sACVR2B-Fc decreased the cachectic effects of chemotherapy without affecting tumour growth. These results demonstrate that blocking ACVR2B signalling may be a promising strategy to counteract chemotherapy-induced muscle wasting without damage to skeletal muscle oxidative capacity or cancer treatment.

    View details for DOI 10.1038/srep32695

    View details for Web of Science ID 000384257600001

    View details for PubMedID 27666826

    View details for PubMedCentralID PMC5036092

  • Endothelial Bmx tyrosine kinase activity is essential for myocardial hypertrophy and remodeling PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCES OF THE UNITED STATES OF AMERICA Holopainen, T., Rasanen, M., Anisimov, A., Tuomainen, T., Zheng, W., Tvorogov, D., Hulmi, J. J., Andersson, L. C., Cenni, B., Tavi, P., Mervaala, E., Kivela, R., Alitalo, K. 2015; 112 (42): 13063-13068

    Abstract

    Cardiac hypertrophy accompanies many forms of heart disease, including ischemic disease, hypertension, heart failure, and valvular disease, and it is a strong predictor of increased cardiovascular morbidity and mortality. Deletion of bone marrow kinase in chromosome X (Bmx), an arterial nonreceptor tyrosine kinase, has been shown to inhibit cardiac hypertrophy in mice. This finding raised the possibility of therapeutic use of Bmx tyrosine kinase inhibitors, which we have addressed here by analyzing cardiac hypertrophy in gene-targeted mice deficient in Bmx tyrosine kinase activity. We found that angiotensin II (Ang II)-induced cardiac hypertrophy is significantly reduced in mice deficient in Bmx and in mice with inactivated Bmx tyrosine kinase compared with WT mice. Genome-wide transcriptomic profiling showed that Bmx inactivation suppresses myocardial expression of genes related to Ang II-induced inflammatory and extracellular matrix responses whereas expression of RNAs encoding mitochondrial proteins after Ang II administration was maintained in Bmx-inactivated hearts. Very little or no Bmx mRNA was expressed in human cardiomyocytes whereas human cardiac endothelial cells expressed abundant amounts. Ang II stimulation of endothelial cells increased Bmx phosphorylation, and Bmx gene silencing inhibited downstream STAT3 signaling, which has been implicated in cardiac hypertrophy. Furthermore, activation of the mechanistic target of rapamycin complex 1 pathway by Ang II treatment was decreased in the Bmx-deficient hearts. Our results demonstrate that inhibition of the cross-talk between endothelial cells and cardiomyocytes by Bmx inactivation suppresses Ang II-induced signals for cardiac hypertrophy. These results suggest that the endothelial Bmx tyrosine kinase could provide a target to attenuate the development of cardiac hypertrophy.

    View details for DOI 10.1073/pnas.1517810112

    View details for Web of Science ID 000363138600049

    View details for PubMedID 26430242

    View details for PubMedCentralID PMC4620883

  • VEGF-B-induced vascular growth leads to metabolic reprogramming and ischemia resistance in the heart EMBO MOLECULAR MEDICINE Kivela, R., Bry, M., Robciuc, M. R., Rasanen, M., Taavitsainen, M., Silvola, J. U., Saraste, A., Hulmi, J. J., Anisimov, A., Mayranpaa, M. I., Lindeman, J. H., Eklund, L., Hellberg, S., Hlushchuk, R., Zhuang, Z. W., Simons, M., Djonov, V., Knuuti, J., Mervaala, E., Alitalo, K. 2014; 6 (3): 307-321

    Abstract

    Angiogenic growth factors have recently been linked to tissue metabolism. We have used genetic gain- and loss-of function models to elucidate the effects and mechanisms of action of vascular endothelial growth factor-B (VEGF-B) in the heart. A cardiomyocyte-specific VEGF-B transgene induced an expanded coronary arterial tree and reprogramming of cardiomyocyte metabolism. This was associated with protection against myocardial infarction and preservation of mitochondrial complex I function upon ischemia-reperfusion. VEGF-B increased VEGF signals via VEGF receptor-2 to activate Erk1/2, which resulted in vascular growth. Akt and mTORC1 pathways were upregulated and AMPK downregulated, readjusting cardiomyocyte metabolic pathways to favor glucose oxidation and macromolecular biosynthesis. However, contrasting with a previous theory, there was no difference in fatty acid uptake by the heart between the VEGF-B transgenic, gene-targeted or wildtype rats. Importantly, we also show that VEGF-B expression is reduced in human heart disease. Our data indicate that VEGF-B could be used to increase the coronary vasculature and to reprogram myocardial metabolism to improve cardiac function in ischemic heart disease.

    View details for DOI 10.1002/emmm.201303147

    View details for Web of Science ID 000332389500003

    View details for PubMedID 24448490

    View details for PubMedCentralID PMC3958306