Bio


Dr. Nishiga is an Instructor at the Stanford Cardiovascular Institute. As a physician-scientist, he focuses on unraveling the mechanisms of cardiovascular diseases and developing innovative treatments. His research path was sparked by firsthand experiences with heart failure patients during his clinical practice, where he recognized the urgent need for more effective treatments. After completing his cardiology fellowship in Japan, he pursued a Ph.D. at Kyoto University, focusing on the role of microRNAs and non-coding RNAs in heart failure, cardiac fibrosis, and atherosclerosis. Currently at Stanford and under the guidance of Dr. Joseph Wu, Dr. Nishiga's postdoctoral research leverages iPSCs and CRISPR technology. His primary research areas include the cardiac impacts of cancer therapies, the cardiovascular effects of COVID-19, and the influence of marijuana use on vascular inflammation.

Academic Appointments


Honors & Awards


  • NIH K99/R00 Pathway to Independence Award, National Heart, Lung, and Blood Institute (NHLBI) (2023 - 2028)
  • TRDRP Postdoctoral Fellowship, Tobacco-Related Disease Research Program (TRDRP), University of California, Office of the President (2020 - 2022)
  • JSPS Oversees Research Fellowship, Japan Society for the Promotion of Science (JSPS) (2018 - 2020)
  • Astellas Oversees Research Fellowship, Astellas Foundation for Research on Metabolic Disorders (2017 - 2018)

Professional Education


  • PhD, Kyoto University, Cardiovascular Medicine (2017)
  • Cardiology Fellowship, Tenri Hospital (Nara, Japan), Cardiology (2012)
  • Residency, Tenri Hospital (Nara, Japan), Internal Medicine (2009)
  • MD, Kyoto University, Medicine (2007)

All Publications


  • Cryo-electron tomography reveals the structural diversity of cardiac proteins in their cellular context. bioRxiv : the preprint server for biology Woldeyes, R. A., Nishiga, M., Vander Roest, A. S., Engel, L., Giri, P., Montenegro, G. C., Wu, A. C., Dunn, A. R., Spudich, J. A., Bernstein, D., Schmid, M. F., Wu, J. C., Chiu, W. 2023

    Abstract

    Cardiovascular diseases are a leading cause of death worldwide, but our understanding of the underlying mechanisms is limited, in part because of the complexity of the cellular machinery that controls the heart muscle contraction cycle. Cryogenic electron tomography (cryo-ET) provides a way to visualize diverse cellular machinery while preserving contextual information like subcellular localization and transient complex formation, but this approach has not been widely applied to the study of heart muscle cells (cardiomyocytes). Here, we deploy a platform for studying cardiovascular disease by combining cryo-ET with human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs). After developing a cryo-ET workflow for visualizing macromolecules in hiPSC-CMs, we reconstructed sub-nanometer resolution structures of the human thin filament, a central component of the contractile machinery. We also visualized a previously unobserved organization of a regulatory complex that connects muscle contraction to calcium signaling (the troponin complex), highlighting the value of our approach for interrogating the structures of cardiac proteins in their cellular context.

    View details for DOI 10.1101/2023.10.26.564098

    View details for PubMedID 37961228

    View details for PubMedCentralID PMC10634850

  • Adverse Impact of Cannabis on Human Health. Annual review of medicine Chandy, M., Nishiga, M., Wei, T. T., Hamburg, N. M., Nadeau, K., Wu, J. C. 2023

    Abstract

    Cannabis, the most commonly used recreational drug, is illicit in many areas of the world. With increasing decriminalization and legalization, cannabis use is increasing in the United States and other countries. The adverse effects of cannabis are unclear because its status as a Schedule 1 drug in the United States restricts research. Despite a paucity of data, cannabis is commonly perceived as a benign or even beneficial drug. However, recent studies show that cannabis has adverse cardiovascular and pulmonary effects and is linked with malignancy. Moreover, case reports have shown an association between cannabis use and neuropsychiatric disorders. With growing availability, cannabis misuse by minors has led to increasing incidences of overdose and toxicity. Though difficult to detect, cannabis intoxication may be linked to impaired driving and motor vehicle accidents. Overall, cannabis use is on the rise, and adverse effects are becoming apparent in clinical data sets. Expected final online publication date for the Annual Review of Medicine, Volume 75 is January 2024. Please see http://www.annualreviews.org/page/journal/pubdates for revised estimates.

    View details for DOI 10.1146/annurev-med-052422-020627

    View details for PubMedID 37582489

  • Cannabinoid receptor 1 antagonist genistein attenuates marijuana-induced vascular inflammation. Cell Wei, T. T., Chandy, M., Nishiga, M., Zhang, A., Kumar, K. K., Thomas, D., Manhas, A., Rhee, S., Justesen, J. M., Chen, I. Y., Wo, H. T., Khanamiri, S., Yang, J. Y., Seidl, F. J., Burns, N. Z., Liu, C., Sayed, N., Shie, J. J., Yeh, C. F., Yang, K. C., Lau, E., Lynch, K. L., Rivas, M., Kobilka, B. K., Wu, J. C. 2022

    Abstract

    Epidemiological studies reveal that marijuana increases the risk of cardiovascular disease (CVD); however, little is known about the mechanism. Δ9-tetrahydrocannabinol (Δ9-THC), the psychoactive component of marijuana, binds to cannabinoid receptor 1 (CB1/CNR1) in the vasculature and is implicated in CVD. A UK Biobank analysis found that cannabis was an risk factor for CVD. We found that marijuana smoking activated inflammatory cytokines implicated in CVD. In silico virtual screening identified genistein, a soybean isoflavone, as a putative CB1 antagonist. Human-induced pluripotent stem cell-derived endothelial cells were used to model Δ9-THC-induced inflammation and oxidative stress via NF-κB signaling. Knockdown of the CB1 receptor with siRNA, CRISPR interference, and genistein attenuated the effects of Δ9-THC. In mice, genistein blocked Δ9-THC-induced endothelial dysfunction in wire myograph, reduced atherosclerotic plaque, and had minimal penetration of the central nervous system. Genistein is a CB1 antagonist that attenuates Δ9-THC-induced atherosclerosis.

    View details for DOI 10.1016/j.cell.2022.04.005

    View details for PubMedID 35489334

  • Ferroptosis of Pacemaker Cells in COVID-19. Circulation research Nishiga, M., Jahng, J. W., Wu, J. C. 2022; 130 (7): 978-980

    View details for DOI 10.1161/CIRCRESAHA.122.320951

    View details for PubMedID 35357897

  • The use of new CRISPR tools in cardiovascular research and medicine. Nature reviews. Cardiology Nishiga, M., Liu, C., Qi, L. S., Wu, J. C. 2022

    Abstract

    Many novel CRISPR-based genome-editing tools, with a wide variety of applications, have been developed in the past few years. The original CRISPR-Cas9 system was developed as a tool to alter genomic sequences in living organisms in a simple way. However, the functions of new CRISPR tools are not limited to conventional genome editing mediated by non-homologous end-joining or homology-directed repair but expand into gene-expression control, epigenome editing, single-nucleotide editing, RNA editing and live-cell imaging. Furthermore, genetic perturbation screening by multiplexing guide RNAs is gaining popularity as a method to identify causative genes and pathways in an unbiased manner. New CRISPR tools can also be applied to ex vivo or in vivo therapeutic genome editing for the treatment of conditions such as hyperlipidaemia. In this Review, we first provide an overview of the diverse new CRISPR tools that have been developed to date. Second, we summarize how these new CRISPR tools are being used to study biological processes and disease mechanisms in cardiovascular research and medicine. Finally, we discuss the prospect of therapeutic genome editing by CRISPR tools to cure genetic cardiovascular diseases.

    View details for DOI 10.1038/s41569-021-00669-3

    View details for PubMedID 35145236

  • Deciphering pathogenicity of variants of uncertain significance with CRISPR-edited iPSCs. Trends in genetics : TIG Guo, H., Liu, L., Nishiga, M., Cong, L., Wu, J. C. 2021

    Abstract

    Genetic variants play an important role in conferring risk for cardiovascular diseases (CVDs). With the rapid development of next-generation sequencing (NGS), thousands of genetic variants associated with CVDs have been identified by genome-wide association studies (GWAS), but the function of more than 40% of genetic variants is still unknown. This gap of knowledge is a barrier to the clinical application of the genetic information. However, determining the pathogenicity of a variant of uncertain significance (VUS) is challenging due to the lack of suitable model systems and accessible technologies. By combining clustered regularly interspaced short palindromic repeats (CRISPR) and human induced pluripotent stem cells (iPSCs), unprecedented advances are now possible in determining the pathogenicity of VUS in CVDs. Here, we summarize recent progress and new strategies in deciphering pathogenic variants for CVDs using CRISPR-edited human iPSCs.

    View details for DOI 10.1016/j.tig.2021.08.009

    View details for PubMedID 34509299

  • CRISPRi/a Screening with Human iPSCs. Methods in molecular biology (Clifton, N.J.) Nishiga, M., Qi, L. S., Wu, J. C. 2021; 2320: 261-281

    Abstract

    Identifying causative genes in a given phenotype or disease model is important for biological discovery and drug development. The recent development of the CRISPR/Cas9 system has enabled unbiased and large-scale genetic perturbation screens to identify causative genes by knocking out many genes in parallel and selecting cells with desired phenotype of interest. However, compared to cancer cell lines, human somatic cells including cardiomyocytes (CMs), neuron cells, and endothelial cells are not easy targets of CRISPR screens because CRISPR screens require a large number of isogenic cells to be cultured and thus primary cells from patients are not ideal. The combination of CRISPR screens with induced pluripotent stem cell (iPSC) technology would be a powerful tool to identify causative genes and pathways because iPSCs can be expanded easily and differentiated to any cell type in principle. Here we describe a robust protocol for CRISPR screening using human iPSCs. Because each screening is different and needs to be customized depending on the cell types and phenotypes of interest, we show an example of CRISPR knockdown screening using CRISPRi system to identify essential genes to differentiate iPSCs to CMs.

    View details for DOI 10.1007/978-1-0716-1484-6_23

    View details for PubMedID 34302664

  • microRNA-33 maintains adaptive thermogenesis via enhanced sympathetic nerve activity. Nature communications Horie, T., Nakao, T., Miyasaka, Y., Nishino, T., Matsumura, S., Nakazeki, F., Ide, Y., Kimura, M., Tsuji, S., Rodriguez, R. R., Watanabe, T., Yamasaki, T., Xu, S., Otani, C., Miyagawa, S., Matsushita, K., Sowa, N., Omori, A., Tanaka, J., Nishimura, C., Nishiga, M., Kuwabara, Y., Baba, O., Watanabe, S., Nishi, H., Nakashima, Y., Picciotto, M. R., Inoue, H., Watanabe, D., Nakamura, K., Sasaki, T., Kimura, T., Ono, K. 2021; 12 (1): 843

    Abstract

    Adaptive thermogenesis is essential for survival, and therefore is tightly regulated by a central neural circuit. Here, we show that microRNA (miR)-33 in the brain is indispensable for adaptive thermogenesis. Cold stress increases miR-33 levels in the hypothalamus and miR-33-/- mice are unable to maintain body temperature in cold environments due to reduced sympathetic nerve activity and impaired brown adipose tissue (BAT) thermogenesis. Analysis of miR-33f/f dopamine-beta-hydroxylase (DBH)-Cre mice indicates the importance of miR-33 in Dbh-positive cells. Mechanistically, miR-33 deficiency upregulates gamma-aminobutyric acid (GABA)A receptor subunit genes such as Gabrb2 and Gabra4. Knock-down of these genes in Dbh-positive neurons rescues the impaired cold-induced thermogenesis in miR-33f/f DBH-Cre mice. Conversely, increased gene dosage of miR-33 in mice enhances thermogenesis. Thus, miR-33 in the brain contributes to maintenance of BAT thermogenesis and whole-body metabolism via enhanced sympathetic nerve tone through suppressing GABAergic inhibitory neurotransmission. This miR-33-mediated neural mechanism may serve as a physiological adaptive defense mechanism for several stresses including cold stress.

    View details for DOI 10.1038/s41467-021-21107-5

    View details for PubMedID 33594062

  • Macrophages: Potential Therapeutic Target of Myocardial Injury in COVID-19. Circulation research Nishiga, M., Wu, J. C. 2021; 129 (1): 47-49

    View details for DOI 10.1161/CIRCRESAHA.121.319446

    View details for PubMedID 34166080

  • Lionheart LincRNA alleviates cardiac systolic dysfunction under pressure overload. Communications biology Kuwabara, Y., Tsuji, S., Nishiga, M., Izuhara, M., Ito, S., Nagao, K., Horie, T., Watanabe, S., Koyama, S., Kiryu, H., Nakashima, Y., Baba, O., Nakao, T., Nishino, T., Sowa, N., Miyasaka, Y., Hatani, T., Ide, Y., Nakazeki, F., Kimura, M., Yoshida, Y., Inada, T., Kimura, T., Ono, K. 2020; 3 (1): 434

    Abstract

    Recent high-throughput approaches have revealed a vast number of transcripts with unknown functions. Many of these transcripts are long noncoding RNAs (lncRNAs), and intergenic region-derived lncRNAs are classified as long intergenic noncoding RNAs (lincRNAs). Although Myosin heavy chain 6 (Myh6) encoding primary contractile protein is down-regulated in stressed hearts, the underlying mechanisms are not fully clarified especially in terms of lincRNAs. Here, we screen upregulated lincRNAs in pressure overloaded hearts and identify a muscle-abundant lincRNA termed Lionheart. Compared with controls, deletion of the Lionheart in mice leads to decreased systolic function and a reduction in MYH6 protein levels following pressure overload. We reveal decreased MYH6 results from an interaction between Lionheart and Purine-rich element-binding protein A after pressure overload. Furthermore, human LIONHEART levels in left ventricular biopsy specimens positively correlate with cardiac systolic function. Our results demonstrate Lionheart plays a pivotal role in cardiac remodeling via regulation of MYH6.

    View details for DOI 10.1038/s42003-020-01164-0

    View details for PubMedID 32792557

  • COVID-19 and cardiovascular disease: from basic mechanisms to clinical perspectives. Nature reviews. Cardiology Nishiga, M. n., Wang, D. W., Han, Y. n., Lewis, D. B., Wu, J. C. 2020

    Abstract

    Coronavirus disease 2019 (COVID-19), caused by a strain of coronavirus known as severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), has become a global pandemic that has affected the lives of billions of individuals. Extensive studies have revealed that SARS-CoV-2 shares many biological features with SARS-CoV, the zoonotic virus that caused the 2002 outbreak of severe acute respiratory syndrome, including the system of cell entry, which is triggered by binding of the viral spike protein to angiotensin-converting enzyme 2. Clinical studies have also reported an association between COVID-19 and cardiovascular disease. Pre-existing cardiovascular disease seems to be linked with worse outcomes and increased risk of death in patients with COVID-19, whereas COVID-19 itself can also induce myocardial injury, arrhythmia, acute coronary syndrome and venous thromboembolism. Potential drug-disease interactions affecting patients with COVID-19 and comorbid cardiovascular diseases are also becoming a serious concern. In this Review, we summarize the current understanding of COVID-19 from basic mechanisms to clinical perspectives, focusing on the interaction between COVID-19 and the cardiovascular system. By combining our knowledge of the biological features of the virus with clinical findings, we can improve our understanding of the potential mechanisms underlying COVID-19, paving the way towards the development of preventative and therapeutic solutions.

    View details for DOI 10.1038/s41569-020-0413-9

    View details for PubMedID 32690910

  • Utility of collagen-derived peptides as markers of organ injury in patients with acute heart failure. Open heart Nagao, K. n., Tamura, A. n., Sato, Y. n., Hata, R. n., Kawase, Y. n., Kadota, K. n., Horie, T. n., Sowa, N. n., Nishiga, M. n., Ono, K. n., Inada, T. n., Tanaka, M. n. 2020; 7 (1): e001041

    Abstract

    This study aims to investigate the time-dependent prognostic utility of two fibrosis markers representing organ fibrogenesis (N-terminal propeptide of procollagen III (PIIINP) and type IV collagen 7S (P4NP 7S)) in patients with acute heart failure (HF).390 patients with acute HF were dichotomised based on the median value of fibrosis markers at discharge. The primary outcome measure was a composite of cardiac death and HF hospitalisation.P4NP 7S significantly declined during hospitalisation, whereas PIIINP did not. The cumulative 90-day and 365-day incidence of the primary outcome measure was 16.6% vs 16.0% (p=0.42) and 33.3% vs 28.4% (p=0.34) in the patients with high versus low PIIINP; 19.9% vs 13.0% (p=0.04) and 32.3% vs 29.0% (p=0.34) in the patients with high and low P4NP 7S, respectively. After adjusting for confounders, high P4NP 7S correlated with significant excess risk relative to low P4NP 7S for both 90-day and 365-day primary outcome measure (adjusted HR, 1.50; 95% CI, 1.02 to 2.21; p=0.04 and adjusted HR, 1.89; 95% CI, 1.11 to 3.26; p=0.02, respectively), which was driven by significant association of high P4NP 7S with higher incidence of HF hospitalisation. Furthermore, P4NP 7S exhibited an additive value to conventional prognostic factors for predicting 90-day outcome (p=0.038 for net reclassification improvement; p=0.0068 for integrated discrimination improvement). High PIIINP did not correlate with significant excess risk for both 90-day and 365-day outcome.This study suggests a possible role of P4NP 7S in the risk stratification of patients with acute HF.

    View details for DOI 10.1136/openhrt-2019-001041

    View details for PubMedID 32341786

    View details for PubMedCentralID PMC7174059

  • Therapeutic genome editing in cardiovascular diseases. Advanced drug delivery reviews Nishiga, M. n., Qi, L. S., Wu, J. C. 2020

    Abstract

    During the past decade, developments in genome editing technology have fundamentally transformed biomedical research. In particular, the CRISPR/Cas9 system has been extensively applied because of its simplicity and ability to alter genomic sequences within living organisms, and an ever increasing number of CRISPR/Cas9-based molecular tools are being developed for a wide variety of applications. While genome editing tools have been used for many aspects of biological research, they also have enormous potential to be used for genome editing therapy to treat a broad range of diseases. For some hematopoietic diseases, clinical trials of therapeutic genome editing with CRISPR/Cas9 are already starting phase I. In the cardiovascular field, genome editing tools have been utilized to understand the mechanisms of diseases such as cardiomyopathy, arrythmia, and lipid metabolism, which now open the door to therapeutic genome editing. Currently, therapeutic genome editing in the cardiovascular field is centered on liver-targeting strategies to reduce cardiovascular risks. Targeting the heart is more challenging. In this review, we discuss the potential applications, recent advances, and current limitations of therapeutic genome editing in the cardiovascular field.

    View details for DOI 10.1016/j.addr.2020.02.003

    View details for PubMedID 32092381

  • Cardioprotective Effects of VCPModulator KUS121 in Murine and Porcine Models of Myocardial Infarction. JACC. Basic to translational science Ide, Y., Horie, T., Saito, N., Watanabe, S., Otani, C., Miyasaka, Y., Kuwabara, Y., Nishino, T., Nakao, T., Nishiga, M., Nishi, H., Nakashima, Y., Nakazeki, F., Koyama, S., Kimura, M., Tsuji, S., Rodriguez, R. R., Xu, S., Yamasaki, T., Watanabe, T., Yamamoto, M., Yanagita, M., Kimura, T., Kakizuka, A., Ono, K. 2019; 4 (6): 701–14

    Abstract

    No effective treatment is yet available to reduce infarct size and improve clinical outcomes after acute myocardial infarction by enhancing early reperfusion therapy using primary percutaneous coronary intervention. The study showed that Kyoto University Substance 121 (KUS121) reduced endoplasmic reticulum stress, maintained adenosine triphosphate levels, and ameliorated the infarct size in a murine cardiac ischemia and reperfusion injury model. The study confirmed the cardioprotective effect of KUS121 in a porcine ischemia and reperfusion injury model. These findings confirmed that KUS121 is a promising novel therapeutic agent for myocardial infarction in conjunction with primary percutaneous coronary intervention.

    View details for DOI 10.1016/j.jacbts.2019.06.001

    View details for PubMedID 31709319

  • Identification of Differential Roles of MicroRNA-33a and -33b During Atherosclerosis Progression With Genetically Modified Mice. Journal of the American Heart Association Koyama, S., Horie, T., Nishino, T., Baba, O., Sowa, N., Miyasaka, Y., Kuwabara, Y., Nakao, T., Nishiga, M., Nishi, H., Nakashima, Y., Nakazeki, F., Ide, Y., Kimura, M., Tsuji, S., Ruiz Rodriguez, R., Xu, S., Yamasaki, T., Otani, C., Watanabe, T., Nakamura, T., Hasegawa, K., Kimura, T., Ono, K. 2019; 8 (13): e012609

    Abstract

    Background Micro RNA (miR)-33 targets cholesterol transporter ATP -binding cassette protein A1 and other antiatherogenic targets and contributes to atherogenic progression. Its inhibition or deletion is known to result in the amelioration of atherosclerosis in mice. However, mice lack the other member of the miR-33 family, miR-33b, which exists in humans and other large mammals. Thus, precise evaluation and comparison of the responsibilities of these 2 miRs during the progression of atherosclerosis has not been reported, although they are essential. Methods and Results In this study, we performed a comprehensive analysis of the difference between the function of miR-33a and miR-33b using genetically modified mice. We generated 4 strains with or without miR-33a and miR-33b. Comparison between mice with only miR-33a (wild-type mice) and mice with only miR-33b (miR-33a-/-/miR-33b+/+) revealed the dominant expression of miR-33b in the liver. To evaluate the whole body atherogenic potency of miR-33a and miR-33b, we developed apolipoprotein E-deficient/miR-33a+/+/miR-33b-/- mice and apolipoprotein E-deficient/miR-33a-/-/miR-33b+/+ mice. With a high-fat and high-cholesterol diet, the apolipoprotein E-deficient/miR-33a-/-/miR-33b+/+ mice developed increased atherosclerotic plaque versus apolipoprotein E-deficient/miR-33a+/+/miR-33b-/- mice, in line with the predominant expression of miR-33b in the liver and worsened serum cholesterol profile. By contrast, a bone marrow transplantation study showed no significant difference, which was consistent with the relevant expression levels of miR-33a and miR-33b in bone marrow cells. Conclusions The miR-33 family exhibits differences in distribution and regulation and particularly in the progression of atherosclerosis; miR-33b would be more potent than miR-33a.

    View details for DOI 10.1161/JAHA.119.012609

    View details for PubMedID 31242815

  • MiR-33a is a therapeutic target in SPG4-related hereditary spastic paraplegia human neurons. Clinical science (London, England : 1979) Nakazeki, F., Tsuge, I., Horie, T., Imamura, K., Tsukita, K., Hotta, A., Baba, O., Kuwabara, Y., Nishino, T., Nakao, T., Nishiga, M., Nishi, H., Nakashima, Y., Ide, Y., Koyama, S., Kimura, M., Tsuji, S., Naitoh, M., Suzuki, S., Izumi, Y., Kawarai, T., Kaji, R., Kimura, T., Inoue, H., Ono, K. 2019

    Abstract

    Recent reports, including ours, have indicated that microRNA (miR)-33 located within the intron of sterol regulatory element binding protein (SREBP) 2 controls cholesterol homeostasis and can be a potential therapeutic target for the treatment of atherosclerosis. Here we show that SPAST , which encodes a microtubule-severing protein called spastin, was a novel target gene of miR-33 in human. Actually, the miR-33 binding site in the SPAST 3'-UTR is conserved not in mice but in mid to large mammals, and it is impossible to clarify the role of miR-33 on SPAST in mice. We demonstrated that inhibition of miR-33a, a major form of miR-33 in human neurons, via locked nucleic acid-anti-miR (LNA) ameliorated the pathological phenotype in HSP-SPG4 patient iPSC-derived cortical neurons. Thus, miR-33a can be a potential therapeutic target for the treatment of HSP-SPG4.

    View details for PubMedID 30777884

  • An in Vivo miRNA Delivery System for Restoring Infarcted Myocardium. ACS nano Yang, H. n., Qin, X. n., Wang, H. n., Zhao, X. n., Liu, Y. n., Wo, H. T., Liu, C. n., Nishiga, M. n., Chen, H. n., Ge, J. n., Sayed, N. n., Abilez, O. J., Ding, D. n., Heilshorn, S. C., Li, K. n. 2019

    Abstract

    A major challenge in myocardial infarction (MI)-related heart failure treatment using microRNA is the efficient and sustainable delivery of miRNAs into myocardium to achieve functional improvement through stimulation of intrinsic myocardial restoration. In this study, we established an in vivo delivery system using polymeric nanoparticles to carry miRNA (miNPs) for localized delivery within a shear-thinning injectable hydrogel. The miNPs triggered proliferation of human embryonic stem cell-derived cardiomyocytes and endothelial cells (hESC-CMs and hESC-ECs) and promoted angiogenesis in hypoxic conditions, showing significantly lower cytotoxicity than Lipofectamine. Furthermore, one injected dose of hydrogel/miNP in MI rats demonstrated significantly improved cardiac functions: increased ejection fraction from 45% to 64%, reduced scar size from 20% to 10%, and doubled capillary density in the border zone compared to the control group at 4 weeks. As such, our results indicate that this injectable hydrogel/miNP composite can deliver miRNA to restore injured myocardium efficiently and safely.

    View details for DOI 10.1021/acsnano.9b03343

    View details for PubMedID 31149806

  • Hepatokine alpha1-Microglobulin Signaling Exacerbates Inflammation and Disturbs Fibrotic Repair in Mouse Myocardial Infarction. Scientific reports Hakuno, D., Kimura, M., Ito, S., Satoh, J., Nakashima, Y., Horie, T., Kuwabara, Y., Nishiga, M., Ide, Y., Baba, O., Nishi, H., Nakao, T., Nishino, T., Nakazeki, F., Koyama, S., Hanada, R., Randolph, R. R., Endo, J., Kimura, T., Ono, K. 2018; 8 (1): 16749

    Abstract

    Acute cardiac rupture and adverse left ventricular (LV) remodeling causing heart failure are serious complications of acute myocardial infarction (MI). While cardio-hepatic interactions have been recognized, their role in MI remains unknown. We treated cultured cardiomyocytes with conditioned media from various cell types and analyzed the media by mass spectrometry to identify alpha1-microglobulin (AM) as an Akt-activating hepatokine. In mouse MI model, AM protein transiently distributed in the infarct and border zones during the acute phase, reflecting infiltration of AM-bound macrophages. AM stimulation activated Akt, NFkappaB, and ERK signaling and enhanced inflammation as well as macrophage migration and polarization, while inhibited fibrogenesis-related mRNA expression in cultured macrophages and cardiac fibroblasts. Intramyocardial AM administration exacerbated macrophage infiltration, inflammation, and matrix metalloproteinase 9 mRNA expression in the infarct and border zones, whereas disturbed fibrotic repair, then provoked acute cardiac rupture in MI. Shotgun proteomics and lipid pull-down analysis found that AM partly binds to phosphatidic acid (PA) for its signaling and function. Furthermore, systemic delivery of a selective inhibitor of diacylglycerol kinase alpha-mediated PA synthesis notably reduced macrophage infiltration, inflammation, matrix metalloproteinase activity, and adverse LV remodeling in MI. Therefore, targeting AM signaling could be a novel pharmacological option to mitigate adverse LV remodeling in MI.

    View details for PubMedID 30425314

  • SREBF1/MicroRNA-33b Axis Exhibits Potent Effect on Unstable Atherosclerotic Plaque Formation In Vivo. Arteriosclerosis, thrombosis, and vascular biology Nishino, T., Horie, T., Baba, O., Sowa, N., Hanada, R., Kuwabara, Y., Nakao, T., Nishiga, M., Nishi, H., Nakashima, Y., Nakazeki, F., Ide, Y., Koyama, S., Kimura, M., Nagata, M., Yoshida, K., Takagi, Y., Nakamura, T., Hasegawa, K., Miyamoto, S., Kimura, T., Ono, K. 2018; 38 (10): 2460-2473

    Abstract

    Objective- Atherosclerosis is a common disease caused by a variety of metabolic and inflammatory disturbances. MicroRNA (miR)-33a within SREBF2 (sterol regulatory element-binding factor 2) is a potent target for treatment of atherosclerosis through regulating both aspects; however, the involvement of miR-33b within SREBF1 remains largely unknown. Although their host genes difference could lead to functional divergence of miR-33a/b, we cannot dissect the roles of miR-33a/b in vivo because of lack of miR-33b sequences in mice, unlike human. Approach and Results- Here, we analyzed the development of atherosclerosis using miR-33b knock-in humanized mice under apolipoprotein E-deficient background. MiR-33b is prominent both in human and mice on atheroprone condition. MiR-33b reduced serum high-density lipoprotein cholesterol levels and systemic reverse cholesterol transport. MiR-33b knock-in macrophages showed less cholesterol efflux capacity and higher inflammatory state via regulating lipid rafts. Thus, miR-33b promotes vulnerable atherosclerotic plaque formation. Furthermore, bone marrow transplantation experiments strengthen proatherogenic roles of macrophage miR-33b. Conclusions- Our data demonstrated critical roles of SREBF1-miR-33b axis on both lipid profiles and macrophage phenotype remodeling and indicate that miR-33b is a promising target for treating atherosclerosis.

    View details for DOI 10.1161/ATVBAHA.118.311409

    View details for PubMedID 30354203

  • Circulating markers of collagen types I, III, and IV in patients with dilated cardiomyopathy: relationships with myocardial collagen expression. ESC heart failure Nagao, K., Inada, T., Tamura, A., Kajitani, K., Shimamura, K., Yukawa, H., Aida, K., Sowa, N., Nishiga, M., Horie, T., Makita, T., Ono, K., Tanaka, M. 2018

    Abstract

    AIMS: Collagen-derived peptides such as collagen I C-terminal telopeptide (CITP) and procollagen III N-terminal propeptide (PIIINP) have been conventionally used as markers of cardiac fibrosis. Collagen IV 7S domain (P4NP 7S) has been recently reported to be correlated with haemodynamics in patients with acute heart failure. We investigated whether these markers reflect cardiac remodelling and myocardial collagen expression.METHODS AND RESULTS: In 80 patients with dilated cardiomyopathy, relationships of CITP, PIIINP, and P4NP 7S to clinical and echocardiographic variables were analysed. CITP and PIIINP were inversely correlated with estimated glomerular filtration rate (r=-0.41, P<0.001 and r=-0.32, P=0.004, respectively); P4NP 7S was positively correlated with B-type natriuretic peptide (r=0.32, P=0.003) and gamma-glutamyltransferase (r=0.38, P<0.001). These correlations were significant even after adjustment by potential confounders, whereas all three collagen markers were not independently correlated with ejection fraction nor with left ventricular (LV) diastolic diameter. In 33 patients undergoing endomyocardial biopsy, myocardial collagen I and III mRNA expressions were correlated with LV end-diastolic volume index (r=0.42, P=0.02 and r=0.54, P=0.002, respectively), whereas myocardial collagen IV mRNA expression was not correlated with LV end-diastolic volume index nor with ejection fraction. Each collagen-derived peptide was not significantly correlated with the myocardial expression of their corresponding collagen mRNA.CONCLUSIONS: Our study shows that CITP, PIIINP, and P4NP 7S do not reflect myocardial collagen mRNA expression but presumably reflect extra-cardiac organ injury in heart failure.

    View details for PubMedID 30273997

  • SREBF1/MicroRNA-33b Axis Exhibits Potent Effect on Unstable Atherosclerotic Plaque Formation In Vivo ARTERIOSCLEROSIS THROMBOSIS AND VASCULAR BIOLOGY Nishino, T., Horie, T., Baba, O., Sowa, N., Hanada, R., Kuwabara, Y., Nakao, T., Nishiga, M., Nishi, H., Nakashima, Y., Nakazeki, F., Ide, Y., Koyama, S., Kimura, M., Nagata, M., Yoshida, K., Takagi, Y., Nakamura, T., Hasegawa, K., Miyamoto, S., Kimura, T., Ono, K. 2018; 38 (10): 2460–73
  • Loss of periostin ameliorates adipose tissue inflammation and fibrosis in vivo. Scientific reports Nakazeki, F., Nishiga, M., Horie, T., Nishi, H., Nakashima, Y., Baba, O., Kuwabara, Y., Nishino, T., Nakao, T., Ide, Y., Koyama, S., Kimura, M., Tsuji, S., Sowa, N., Yoshida, S., Conway, S. J., Yanagita, M., Kimura, T., Ono, K. 2018; 8 (1): 8553

    Abstract

    Recent evidence suggests that the accumulation of macrophages as a result of obesity-induced adipose tissue hypoxia is crucial for the regulation of tissue fibrosis, but the molecular mechanisms underlying adipose tissue fibrosis are still unknown. In this study, we revealed that periostin (Postn) is produced at extraordinary levels by adipose tissue after feeding with a high-fat diet (HFD). Postn was secreted at least from macrophages in visceral adipose tissue during the development of obesity, possibly due to hypoxia. Postn-/- mice had lower levels of crown-like structure formation and fibrosis in adipose tissue and were protected from liver steatosis. These mice also showed amelioration in systemic insulin resistance compared with HFD-fed WT littermates. Mice deficient in Postn in their hematopoietic compartment also had lower levels of inflammation in adipose tissue, in parallel with a reduction in ectopic lipid accumulation compared with the controls. Our data indicated that the regulation of Postn in visceral fat could be beneficial for the maintenance of healthy adipose tissue in obesity.

    View details for PubMedID 29867212

  • MicroRNA-33 regulates the population of peripheral inflammatory Ly6Chigh monocytes through dual pathways. Molecular and cellular biology Baba, O., Horie, T., Nakao, T., Hakuno, D., Nakashima, Y., Nishi, H., Kuwabara, Y., Nishiga, M., Nishino, T., Ide, Y., Nakazeki, F., Koyama, S., Kimura, M., Hanada, R., Kawahara, M., Kimura, T., Ono, K. 2018

    Abstract

    MicroRNA (miR)-33 targets ATP-binding cassette transporter A1 (ABCA1), and its deficiency increases serum HDL-cholesterol (HDL-C) and ameliorates atherosclerosis. Although we previously reported that miR-33 deficiency increased peripheral Ly6Chigh monocytes on ApoE-deficient background, the effect of miR-33 on monocyte population is not fully elucidated especially on wild-type (WT) background.We found that Ly6Chigh monocytes in miR-33-/- mice were decreased in peripheral blood and increased in bone marrow (BM). Expansion of myeloid progenitors and decreased apoptosis in LSK (Lin-Sca1+c-Kit+) cells were observed in miR-33-/- mice. BM transplantation study and competitive repopulation assay revealed that hematopoietic miR-33 deficiency caused myeloid expansion and increased peripheral Ly6Chigh monocytes, and nonhematopoietic miR-33 deficiency caused reduced peripheral Ly6Chigh monocytes. Expression of High mobility group AT-hook 2 (HMGA2) targeted by miR-33 increased in miR-33 deficient LSK cells, and its knockdown abolished the reduction of apoptosis. Transduction of human apolipoprotein A1 and ABCA1 in WT mouse liver increased HDL-C and reduced peripheral Ly6Chigh monocyte.These data indicate that miR-33 deficiency affects distribution of inflammatory monocytes through dual pathways. One is caused by enhancement of Hmga2 expression in hematopoietic stem cells to increase Ly6Chigh monocytes and the other is due to the elevation of HDL-C to decrease peripheral Ly6Chigh monocytes.

    View details for PubMedID 29712758

  • Induced pluripotent stem cells as a biopharmaceutical factory for extracellular vesicles. European heart journal Nishiga, M. n., Guo, H. n., Wu, J. C. 2018

    View details for PubMedID 29547885

  • Dynamic changes of serum microRNA-122-5p through therapeutic courses indicates amelioration of acute liver injury accompanied by acute cardiac decompensation. ESC heart failure Koyama, S., Kuragaichi, T., Sato, Y., Kuwabara, Y., Usami, S., Horie, T., Baba, O., Hakuno, D., Nakashima, Y., Nishino, T., Nishiga, M., Nakao, T., Arai, H., Kimura, T., Ono, K. 2017; 4 (2): 112-121

    Abstract

    Recent studies have shown that serum microRNA (miR) abundance is informative for the diagnosis or prognosis of heart failure. However, the dynamics and kinetics of miRs in acute heart failure are largely unknown. Serial measurement and analysis of serum miRs changes in individuals along their therapeutic course could reduce inter-individual variation and should detect potentially important serum miRs related to disease mechanisms. Based on this concept, we profiled serum miR signatures of blood samples that were obtained sequentially on the day of admission and on hospital Day 7.This prospective, observational study included 42 consecutive acute heart failure patients (74 ± 1 years old, 24 male). From admission to Day 7, most of the patients showed clinical improvement. In such a cohort, we detected several fluctuations of serum miRs by two distinct screening methods (quantitative PCR and high-throughput sequencing). One of these fluctuating serum miRs, miR-122-5p, decreased significantly from Day 1 to Day 7 [median arbitrary unit (1st:3rd quantile value); 4.62 [2.39:12.3] to 3.07 [1.67:5.39], P = 0.007]. This fluctuation was significantly correlated with changes in serum liver function markers (estimated coefficient and 95% confidence interval; vs change in aspartate aminotransferase 1.69, 0.890-2.484, P < 0.001 and r = 0.560, vs change in alanine aminotransferase 1.09, 0.406-1.771, P = 0.007 and r = 0.428).The serum miR signature of patients with acute heart failure might indicate the severity of the disease or patients' response to therapeutic intervention. Notably, serum miR-122-5p levels reflect liver damage in this condition.

    View details for DOI 10.1002/ehf2.12123

    View details for PubMedID 28451447

    View details for PubMedCentralID PMC5396046

  • MicroRNA-33 Controls Adaptive Fibrotic Response in the Remodeling Heart by Preserving Lipid Raft Cholesterol CIRCULATION RESEARCH Nishiga, M., Horie, T., Kuwabara, Y., Nagao, K., Baba, O., Nakao, T., Nishino, T., Hakuno, D., Nakashima, Y., Nishi, H., Nakazeki, F., Ide, Y., Koyama, S., Kimura, M., Hanada, R., Nakamura, T., Inada, T., Hasegawa, K., Conway, S. J., Kita, T., Kimura, T., Ono, K. 2017; 120 (5): 835-847

    Abstract

    Heart failure and atherosclerosis share the underlying mechanisms of chronic inflammation followed by fibrosis. A highly conserved microRNA (miR), miR-33, is considered as a potential therapeutic target for atherosclerosis because it regulates lipid metabolism and inflammation. However, the role of miR-33 in heart failure remains to be elucidated.To clarify the role of miR-33 involved in heart failure.We first investigated the expression levels of miR-33a/b in human cardiac tissue samples with dilated cardiomyopathy. Increased expression of miR-33a was associated with improving hemodynamic parameters. To clarify the role of miR-33 in remodeling hearts, we investigated the responses to pressure overload by transverse aortic constriction in miR-33-deficient (knockout [KO]) mice. When mice were subjected to transverse aortic constriction, miR-33 expression levels were significantly upregulated in wild-type left ventricles. There was no difference in hypertrophic responses between wild-type and miR-33KO hearts, whereas cardiac fibrosis was ameliorated in miR-33KO hearts compared with wild-type hearts. Despite the ameliorated cardiac fibrosis, miR-33KO mice showed impaired systolic function after transverse aortic constriction. We also found that cardiac fibroblasts were mainly responsible for miR-33 expression in the heart. Deficiency of miR-33 impaired cardiac fibroblast proliferation, which was considered to be caused by altered lipid raft cholesterol content. Moreover, cardiac fibroblast-specific miR-33-deficient mice also showed decreased cardiac fibrosis induced by transverse aortic constriction as systemic miR-33KO mice.Our results demonstrate that miR-33 is involved in cardiac remodeling, and it preserves lipid raft cholesterol content in fibroblasts and maintains adaptive fibrotic responses in the remodeling heart.

    View details for DOI 10.1161/CIRCRESAHA.116.309528

    View details for Web of Science ID 000395592500022

    View details for PubMedID 27920122

  • Prevention of neointimal formation using miRNA-126-containing nanoparticle-conjugated stents in a rabbit model PLOS ONE Izuhara, M., Kuwabara, Y., Saito, N., Yamamoto, E., Hakuno, D., Nakashima, Y., Horie, T., Baba, O., Nishiga, M., Nakao, T., Nishino, T., Nakazeki, F., Ide, Y., Kimura, M., Kimura, T., Ono, K. 2017; 12 (3)

    Abstract

    Despite recent progress with drug-eluting stents, restenosis and thrombosis after endovascular intervention are still major limitations in the treatment of cardiovascular diseases. These problems are possibly caused by inappropriate inhibition of neointimal formation and retardation of re-endothelialization on the surface of the stents. miR-126 has been shown to have the potential to enhance vascular endothelial cell proliferation.We designed and constructed a 27-nt double strand RNA (dsRNA) conjugated to cholesterol, which has high membrane permeability, and formed mature miR-126 after transfection. For site-specific induction of miR-126, we utilized poly (DL-lactide-co-glycolide) nanoparticles (NPs). miR-126-dsRNA-containing NPs (miR-126 NPs) significantly reduced the protein expression of a previously identified miR-126 target, SPRED1, in human umbilical vascular endothelial cells (HUVECs), and miR-126 NPs enhanced the proliferation and migration of HUVECs. On the other hand, miR-126 NPs reduced the proliferation and migration of vascular smooth muscle cells, via the suppression of IRS-1. Finally, we developed a stent system that eluted miR-126. This delivery system exhibited significant inhibition of neointimal formation in a rabbit model of restenosis.miR-126 NP-conjugated stents significantly inhibited the development of neointimal hyperplasia in rabbits. The present study may indicate the possibility of a novel therapeutic option to prevent restenosis after angioplasty.

    View details for DOI 10.1371/journal.pone.0172798

    View details for Web of Science ID 000396011300033

    View details for PubMedID 28253326

    View details for PubMedCentralID PMC5333844

  • Genetic Ablation of MicroRNA-33 Attenuates Inflammation and Abdominal Aortic Aneurysm Formation via Several Anti-Inflammatory Pathways. Arteriosclerosis, thrombosis, and vascular biology Nakao, T. n., Horie, T. n., Baba, O. n., Nishiga, M. n., Nishino, T. n., Izuhara, M. n., Kuwabara, Y. n., Nishi, H. n., Usami, S. n., Nakazeki, F. n., Ide, Y. n., Koyama, S. n., Kimura, M. n., Sowa, N. n., Ohno, S. n., Aoki, H. n., Hasegawa, K. n., Sakamoto, K. n., Minatoya, K. n., Kimura, T. n., Ono, K. n. 2017; 37 (11): 2161–70

    Abstract

    Abdominal aortic aneurysm (AAA) is an increasingly prevalent and ultimately fatal disease with no effective pharmacological treatment. Because matrix degradation induced by vascular inflammation is the major pathophysiology of AAA, attenuation of this inflammation may improve its outcome. Previous studies suggested that miR-33 (microRNA-33) inhibition and genetic ablation of miR-33 increased serum high-density lipoprotein cholesterol and attenuated atherosclerosis.MiR-33a-5p expression in central zone of human AAA was higher than marginal zone. MiR-33 deletion attenuated AAA formation in both mouse models of angiotensin II- and calcium chloride-induced AAA. Reduced macrophage accumulation and monocyte chemotactic protein-1 expression were observed in calcium chloride-induced AAA walls in miR-33-/- mice. In vitro experiments revealed that peritoneal macrophages from miR-33-/- mice showed reduced matrix metalloproteinase 9 expression levels via c-Jun N-terminal kinase inactivation. Primary aortic vascular smooth muscle cells from miR-33-/- mice showed reduced monocyte chemotactic protein-1 expression by p38 mitogen-activated protein kinase attenuation. Both of the inactivation of c-Jun N-terminal kinase and p38 mitogen-activated protein kinase were possibly because of the increase of ATP-binding cassette transporter A1 that is a well-known target of miR-33. Moreover, high-density lipoprotein cholesterol derived from miR-33-/- mice reduced expression of matrix metalloproteinase 9 in macrophages and monocyte chemotactic protein-1 in vascular smooth muscle cells. Bone marrow transplantation experiments indicated that miR-33-deficient bone marrow cells ameliorated AAA formation in wild-type recipients. MiR-33 deficiency in recipient mice was also shown to contribute the inhibition of AAA formation.These data strongly suggest that inhibition of miR-33 will be effective as a novel strategy for treating AAA.

    View details for PubMedID 28882868

  • Predictors of Rapid Progression and Clinical Outcome of Asymptomatic Severe Aortic Stenosis CIRCULATION JOURNAL Nishimura, S., Izumi, C., Nishiga, M., Amano, M., Imamura, S., Onishi, N., Tamaki, Y., Enomoto, S., Miyake, M., Tamura, T., Kondo, H., Kaitani, K., Nakagawa, Y. 2016; 80 (8): 1863-1869

    Abstract

    The optimal timing of aortic valve replacement (AVR) is controversial in patients with asymptomatic severe aortic stenosis (AS) except when very severe. Prediction of progression of severe AS is helpful in deciding on the timing of AVR. The purpose of this study was to clarify the predictors of progression rate and clinical outcomes of severe AS.We retrospectively investigated 140 consecutive patients with asymptomatic severe AS (aortic valve area [AVA], 0.75-1.0 cm(2)). First-year progression rate and annual progression rate of AVA and of aortic jet velocity (AV-Vel) were calculated. Cardiac events were examined and the predictors of rapid progression and cardiac events were analyzed. The median follow-up period was 36 months. The median annual progression rate was -0.05 cm(2)/year for AVA and 0.22 m/s/year for AV-Vel. Dyslipidemia, moderate-severe calcification, and first-year AV-Vel progression ≥0.22 m/s/year were independent predictors of cardiac events. Cardiac event-free rate was lower in patients with AV-Vel first-year progression rate ≥0.22 m/s/year than in those with a lower rate. Diabetes and moderate-severe calcification were related to first-year rapid progression.The annual progression rate of severe AS was -0.05 cm(2)/year for AVA and 0.22 m/s/year for AV-Vel. Patients with first-year rapid progression or severely calcified aortic valve should be carefully observed while considering an early operation. (Circ J 2016; 80: 1863-1869).

    View details for DOI 10.1253/circj.CJ-16-0333

    View details for Web of Science ID 000381437500034

    View details for PubMedID 27334030

  • Prognostic Significance of ST-Segment Elevation in Leads V1-2 in Patients With Severe Aortic Stenosis CIRCULATION JOURNAL Taniguchi, T., Shiomi, H., Kosuge, M., Morimoto, T., Nakatsuma, K., Nishiga, M., Sasa, T., Saito, N., Kimura, T. 2016; 80 (2): 526-?

    Abstract

    ST-segment elevation (STE) in leads V1-2 is often observed in patients with severe aortic stenosis (AS), but its significance remains unknown.We retrospectively evaluated baseline ECGs and 5-year clinical outcomes in 211 consecutive patients with severe AS, defined as peak aortic jet velocity (Aortic Vmax) >4.0 m/s, or mean aortic pressure gradient >40 mmHg, or aortic valve area (AVA) <1.0 cm(2). The primary outcome measure was a composite of death or surgical aortic valve replacement (AVR). Patients with STE in leads V1-2(≥0.15 mV) had greater Aortic Vmax and smaller AVA than patients without. With a median follow-up of 4.9 years, the cumulative 5-year incidence of death or AVR was significantly higher in patients with STE in leads V1-2 than in patients without (91.4% vs. 77.1%; P=0.003). After adjusting for confounders, STE in leads V1-2 was independently associated with higher risk for death or AVR (hazard ratio, 1.53; 95% confidence interval, 1.06-2.22; P=0.02). In 64 asymptomatic patients without any indication for AVR at initial diagnosis of severe AS, the cumulative incidence of AVR was significantly higher in patients with STE in leads V1-2 than in patients without (57.6% vs. 30.5%; P<0.001).STE in leads V1-2 independently predicted poorer prognosis and more frequent need for AVR in patients with severe AS.

    View details for DOI 10.1253/circj.CJ-15-0641

    View details for Web of Science ID 000369376900037

    View details for PubMedID 26742587

  • Expression Patterns of miRNA-423-5p in the Serum and Pericardial Fluid in Patients Undergoing Cardiac Surgery PLOS ONE Miyamoto, S., Usami, S., Kuwabara, Y., Horie, T., Baba, O., Hakuno, D., Nakashima, Y., Nishiga, M., Izuhara, M., Nakao, T., Nishino, T., Ide, Y., Nakazeki, F., Wang, J., Ueyama, K., Kimura, T., Ono, K. 2015; 10 (11)

    Abstract

    Recently, it has been reported that specific microRNA (miRNA) levels are elevated in serum and can be used as biomarkers in patients with cardiovascular diseases. However, miRNAs expression profiles and their sources in pericardial fluid (PF) are unclear.The purpose of this study was to identify the levels of miRNAs in PF in relation to those in the serum in patients undergoing cardiac surgery. Serum (S) and PF from patients undergoing coronary artery bypass graft (CABG) due to stable angina pectoris (sAP) and unstable AP (uAP) and aortic valve replacement due to aortic stenosis (AS) were analyzed for the detection of miRNAs. We named these samples S-sAP, S-uAP, S-AS, PF-sAP, PF-uAP, and PF-AS, respectively. We first measured the levels of miR-423-5p, which was recognized previously as a biomarker for heart failure. miR-423-5p levels were significantly higher in PF than serum. Although there was no difference in miR-423-5p levels among the PF-AS, PF-sAP, and PF-uAP, its levels were significantly elevated in S-uAP compared with those in S-AS and S-sAP. In order to clarify the source of miR-423-5p in PF, we measured the levels of muscle-enriched miR-133a and vascular-enriched miR-126 and miR-92a in the same samples. miR-133a levels were significantly higher in serum than in PF, and it was elevated in S-uAP compared with S-AS. miR-126 level was significantly increased in serum compared with PF, and the level of miR-92a the similar tendency. miR-423-5p is located in the first intron of NSRP1. There is another miRNA, miR-3184, encoded in the opposite direction in the same region. In vitro experiments indicated that the duplex of miR-423-5p and miR-3184-3p was more resistant to RNase than the duplex of miR-423-5p and miR-133-3p, which may help to stabilize miR-423-5p in the PF.Our results suggested that miR-423-5p is enriched in PF, and serum miR-423-5p may be associate with uAP. Its expression pattern was different to that of muscle- and vascular-enriched miRNAs, miR-133a, miR-126, and miR-92a.

    View details for DOI 10.1371/journal.pone.0142904

    View details for Web of Science ID 000364480900080

    View details for PubMedID 26562412

    View details for PubMedCentralID PMC4642962

  • Acute myocardial infarction and 30-year coronary aneurysm follow-up by serial angiography in a young adult with Kawasaki disease. Cardiovascular intervention and therapeutics Matsushita, K., Tamura, T., Nishiga, M., Kaitani, K., Izumi, C., Nakagawa, Y. 2015; 30 (2): 142-146

    Abstract

    The number of adult patients with coronary artery disease caused by Kawasaki disease (KD) is gradually increasing, but some patients drop out of clinical follow-up. However, careful, long-term follow-up and establishment of an optimal treatment strategy are needed in these patients, as well as cooperation between pediatric and adult cardiologists. We report a case of acute myocardial infarction in a young adult patient with KD whose serial coronary angiography was followed up for over 30 years. Successful reperfusion therapy was performed with thrombus aspiration, rotational atherectomy, and implantation of a drug-eluting stent.

    View details for DOI 10.1007/s12928-014-0262-8

    View details for PubMedID 24729026

  • MicroRNA-451 Exacerbates Lipotoxicity in Cardiac Myocytes and High-Fat Diet-Induced Cardiac Hypertrophy in Mice Through Suppression of the LKB1/AMPK Pathway CIRCULATION RESEARCH Kuwabara, Y., Horie, T., Baba, O., Watanabe, S., Nishiga, M., Usami, S., Izuhara, M., Nakao, T., Nishino, T., Otsu, K., Kita, T., Kimura, T., Ono, K. 2015; 116 (2): 279-U217

    Abstract

    In some patients with type 2 diabetes mellitus (DM) without hypertension, cardiac hypertrophy and attenuated cardiac function are observed, and this insult is termed diabetic cardiomyopathy. To date, microRNA (miRNAs or miR) functions in diabetic cardiomyopathy remain to be elucidated.To clarify the functions of miRNAs involved in diabetic cardiomyopathy caused by type 2 DM.C57BL/6 mice were fed a high-fat diet (HFD) for 20 weeks, which induced obesity and type 2 DM. miRNA microarray analyses and real-time polymerase chain reaction revealed that miR-451 levels were significantly increased in the type 2 DM mouse hearts. Because excess supply of saturated fatty acids is a cause of diabetic cardiomyopathy, we stimulated neonatal rat cardiac myocytes with palmitic acid and confirmed that miR-451 expression was increased in a dose- and time-dependent manner. Loss of miR-451 function ameliorated palmitate-induced lipotoxicity in neonatal rat cardiac myocytes. Calcium-binding protein 39 (Cab39) is a scaffold protein of liver kinase B1 (LKB1), an upstream kinase of AMP-activated protein kinase (AMPK). Cab39 was a direct target of miR-451 in neonatal rat cardiac myocytes and Cab39 overexpression rescued the lipotoxicity. To clarify miR-451 functions in vivo, we generated cardiomyocyte-specific miR-451 knockout mice. HFD-induced cardiac hypertrophy and contractile reserves were ameliorated in cardiomyocyte-specific miR-451 knockout mice compared with control mice. Protein levels of Cab39 and phosphorylated AMPK were increased and phosphorylated mammalian target of rapamycin (mTOR) was reduced in cardiomyocyte-specific miR-451 knockout mouse hearts compared with control mouse hearts.Our results demonstrate that miR-451 is involved in diabetic cardiomyopathy through suppression of the LKB1/AMPK pathway.

    View details for DOI 10.1161/CIRCRESAHA.116.304707

    View details for Web of Science ID 000347939000013

    View details for PubMedID 25362209

  • MicroRNA-33b knock-in mice for an intron of sterol regulatory element-binding factor 1 (Srebf1) exhibit reduced HDL-C in vivo SCIENTIFIC REPORTS Horie, T., Nishino, T., Baba, O., Kuwabara, Y., Nakao, T., Nishiga, M., Usami, S., Izuhara, M., Nakazeki, F., Ide, Y., Koyama, S., Sowa, N., Yahagi, N., Shimano, H., Nakamura, T., Hasegawa, K., Kume, N., Yokode, M., Kita, T., Kimura, T., Ono, K. 2014; 4

    Abstract

    MicroRNAs (miRs) are small non-protein-coding RNAs that bind to specific mRNAs and inhibit translation or promote mRNA degradation. Recent reports, including ours, indicated that miR-33a located within the intron of sterol regulatory element-binding protein (SREBP) 2 controls cholesterol homeostasis and can be a possible therapeutic target for treating atherosclerosis. Primates, but not rodents, express miR-33b from an intron of SREBF1. Therefore, humanized mice, in which a miR-33b transgene is inserted within a Srebf1 intron, are required to address its function in vivo. We successfully established miR-33b knock-in (KI) mice and found that protein levels of known miR-33a target genes, such as ABCA1, ABCG1, and SREBP-1, were reduced compared with those in wild-type mice. As a consequence, macrophages from the miR-33b KI mice had a reduced cholesterol efflux capacity via apoA-I and HDL-C. Moreover, HDL-C levels were reduced by almost 35% even in miR-33b KI hetero mice compared with the control mice. These results indicate that miR-33b may account for lower HDL-C levels in humans than those in mice and that miR-33b is possibly utilized for a feedback mechanism to regulate its host gene SREBF1. Our mice will also aid in elucidating the roles of miR-33a/b in different genetic disease models.

    View details for DOI 10.1038/srep05312

    View details for Web of Science ID 000337341400009

    View details for PubMedID 24931346

    View details for PubMedCentralID PMC4058878

  • MicroRNA-33 regulates sterol regulatory element-binding protein 1 expression in mice NATURE COMMUNICATIONS Horie, T., Nishino, T., Baba, O., Kuwabara, Y., Nakao, T., Nishiga, M., Usami, S., Izuhara, M., Sowa, N., Yahagi, N., Shimano, H., Matsumura, S., Inoue, K., Marusawa, H., Nakamura, T., Hasegawa, K., Kume, N., Yokode, M., Kita, T., Kimura, T., Ono, K. 2013; 4

    Abstract

    MicroRNAs (miRs) are small non-protein-coding RNAs that bind to specific mRNAs and inhibit translation or promote mRNA degradation. Recent reports have indicated that miR-33, which is located within the intron of sterol regulatory element-binding protein (SREBP) 2, controls cholesterol homoeostasis and may be a potential therapeutic target for the treatment of atherosclerosis. Here we show that deletion of miR-33 results in marked worsening of high-fat diet-induced obesity and liver steatosis. Using miR-33(-/-)Srebf1(+/-) mice, we demonstrate that SREBP-1 is a target of miR-33 and that the mechanisms leading to obesity and liver steatosis in miR-33(-/-) mice involve enhanced expression of SREBP-1. These results elucidate a novel interaction between SREBP-1 and SREBP-2 mediated by miR-33 in vivo.

    View details for DOI 10.1038/ncomms3883

    View details for Web of Science ID 000329396200001

    View details for PubMedID 24300912

    View details for PubMedCentralID PMC3863899

  • Effects of Medical Treatment on the Prognosis and Risk of Embolic Events in Patients with Severe Aortic Plaque JOURNAL OF ATHEROSCLEROSIS AND THROMBOSIS Nishiga, M., Izumi, C., Matsutani, H., Hashiwada, S., Takahashi, S., Hayama, Y., Nakajima, S., Sakamoto, J., Hanazawa, K., Miyake, M., Tamura, T., Kondo, H., Motooka, M., Kaitani, K., Nakagawa, Y. 2013; 20 (11): 821-829

    Abstract

    The optimal treatment strategy for patients with aortic atheroma is not well established because data regarding medical treatment for such patients are lacking, especially with respect to the Japanese population. The purpose of this study was to clarify the effects of medical treatment on the risk of embolic events and mortality in patients with severe aortic plaque.We retrospectively investigated 75 consecutive patients with severe aortic plaque detected on transesophageal echocardiography (TEE) between 1995 and 2005. The occurrence of embolic events and all-cause death in the period after TEE was assessed. The cumulative incidence of subsequent embolic events and death was evaluated in relation to specific medical treatments, including statins, antiplatelet drugs and warfarin.Embolic events occurred in 27 patients (36%) and death occurred in 37 patients (49%) during follow-up (5.6±3.0 years). The patients who experienced embolic events had a significantly higher prevalence of previous embolic events, atrial fibrillation and hemodialysis than the patients who did not experience embolic events. Univariate and multivariate analyses showed that the use of statins and/or antiplatelet drugs was significantly associated with a low incidence of death but not with a low incidence of embolic events. On the other hand, warfarin exhibited neither beneficial nor harmful effects on the incidence of embolic events or death.Statin and antiplatelet drugs have beneficial effects on the prognosis of patients with severe aortic plaque diagnosed on TEE.

    View details for Web of Science ID 000327863900005

    View details for PubMedID 23955519

  • A case of significantly increased mitral regurgitation early after atrial septal defect closure. Journal of echocardiography Nishiga, M., Izumi, C., Matsutani, H., Hashiwada, S., Takahashi, S., Hayama, Y., Nakajima, S., Sakamoto, J., Hanazawa, K., Miyake, M., Tamura, T., Kondo, H., Motooka, M., Kaitani, K., Nakagawa, Y. 2012; 10 (2): 69-71

    Abstract

    We report a rare case in which mitral regurgitation (MR) was exacerbated to a severe level early after atrial septal defect (ASD) closure, even though the female patient had preoperatively mild MR and mild changes in mitral valve (MV) and sinus rhythm. The mechanism of increased MR was considered as poor coaptation and tethering of the MV due to the restricted motion of the posterior leaflet in addition to geometric changes of the left ventricle (LV) after ASD closure.

    View details for DOI 10.1007/s12574-012-0123-3

    View details for PubMedID 27278048

  • Progression of Isolated Tricuspid Regurgitation Late After Left-Sided Valve Surgery - Clinical Features and Mechanisms CIRCULATION JOURNAL Izumi, C., Miyake, M., Takahashi, S., Matsutani, H., Hashiwada, S., Kuwano, K., Hayashi, H., Nakajima, S., Nishiga, M., Hanazawa, K., Sakamoto, J., Kondo, H., Tamura, T., Kaitani, K., Yamanaka, K., Nakagawa, Y. 2011; 75 (12): 2902-2907

    Abstract

    Severe tricuspid regurgitation (TR) sometimes develops late after left-sided valve surgery without left heart failure, pulmonary hypertension or rheumatic tricuspid valve. The purpose of the present study was to investigate clinical characteristics and mechanisms of severe isolated TR late after left-sided valve surgery.A total of 372 consecutive patients who underwent left-sided valve surgery between 1990 and 2003 and who were followed up with echocardiography for at least 5 years, were retrospectively investigated. The mean follow-up period was 9.4 years. Clinical background, preoperative and postoperative echocardiographic parameters were evaluated. Among the 372 patients, severe isolated TR was detected in 23 patients, which developed at a mean of 8.6 years after surgery. Twenty-two of 23 patients had undergone mitral valve surgery. Multivariate logistic regression analysis identified the presence of preoperative atrial fibrillation and preoperative ejection fraction as independent determinants for the development of severe isolated TR. In patients with severe isolated TR, the tricuspid annular diameter and the right atrial area were already enlarged early after surgery and both of these increased prior to TR progression.Severe isolated TR developing late after mitral valve surgery is not uncommon, thus it is important to recognize this disease entity. Annular dilatation was the main cause of isolated TR and serial echocardiographic data are important to detect progression of isolated TR and to assess its mechanisms.

    View details for DOI 10.1253/circj.CJ-11-0718

    View details for Web of Science ID 000297567600032

    View details for PubMedID 21946358

  • Cardiac tamponade during transesophageal echocardiography in a patient with infective endocarditis. Journal of echocardiography Miyake, M., Izumi, C., Kuwano, K., Honjo, G., Matsutani, H., Hashiwada, S., Takahashi, S., Nishiga, M., Nakajima, S., Yamao, K., Hanazawa, K., Sakamoto, J., Yoshitani, K., Motooka, M., Kaitani, K., Izumi, T., Kobashi, Y., Nakagawa, Y. 2010; 8 (1): 25-27

    Abstract

    An 81-year-old man with a history of diabetes mellitus and end-stage renal disease was admitted because of infective endocarditis. During transesophageal echocardiography (TEE), pericardial effusion rapidly increased and led to cardiac tamponade. Despite intensive therapy, the patient did not recover. Autopsy showed hemopericardium, ruptured sinus of Valsalva, and vegetation on the aortic valve. Our case suggests that cardiac tamponade due to the rupture of a sinus of Valsalva can occur in patients with aortic valve endocarditis complicated by perivalvular abscess. Therefore, we must be aware of this devastating complication and take preventive measures when performing TEE in such patients.

    View details for DOI 10.1007/s12574-009-0023-3

    View details for PubMedID 27278541