Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 81
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Nature ; 572(7769): 335-340, 2019 08.
Article in English | MEDLINE | ID: mdl-31316208

ABSTRACT

Lamin A/C (LMNA) is one of the most frequently mutated genes associated with dilated cardiomyopathy (DCM). DCM related to mutations in LMNA is a common inherited cardiomyopathy that is associated with systolic dysfunction and cardiac arrhythmias. Here we modelled the LMNA-related DCM in vitro using patient-specific induced pluripotent stem cell-derived cardiomyocytes (iPSC-CMs). Electrophysiological studies showed that the mutant iPSC-CMs displayed aberrant calcium homeostasis that led to arrhythmias at the single-cell level. Mechanistically, we show that the platelet-derived growth factor (PDGF) signalling pathway is activated in mutant iPSC-CMs compared to isogenic control iPSC-CMs. Conversely, pharmacological and molecular inhibition of the PDGF signalling pathway ameliorated the arrhythmic phenotypes of mutant iPSC-CMs in vitro. Taken together, our findings suggest that the activation of the PDGF pathway contributes to the pathogenesis of LMNA-related DCM and point to PDGF receptor-ß (PDGFRB) as a potential therapeutic target.


Subject(s)
Cardiomyopathy, Dilated/genetics , Lamin Type A/genetics , Mutation , Platelet-Derived Growth Factor/metabolism , Receptor, Platelet-Derived Growth Factor beta/metabolism , Signal Transduction , Arrhythmias, Cardiac/metabolism , Arrhythmias, Cardiac/pathology , Calcium/metabolism , Cells, Cultured , Chromatin/chemistry , Chromatin/genetics , Chromatin/metabolism , Chromatin Assembly and Disassembly/genetics , Haploinsufficiency/genetics , Homeostasis , Humans , In Vitro Techniques , Induced Pluripotent Stem Cells/pathology , Models, Biological , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Nonsense Mediated mRNA Decay , RNA, Messenger/analysis , RNA, Messenger/genetics , Single-Cell Analysis
2.
Cancer ; 130(12): 2205-2214, 2024 Jun 15.
Article in English | MEDLINE | ID: mdl-38358333

ABSTRACT

BACKGROUND: Patients undergoing autologous hematopoietic cell transplantation (HCT) have a >2-fold risk of developing cardiovascular disease (CVD; heart failure, myocardial infarction, and stroke), compared to the general population. Coronary artery calcium (CAC) is predictive of CVD in nononcology patients but is not as well studied in patients who underwent HCT and survivors of HCT.The objective of this study was to examine the association between CAC and CVD risk and outcomes after HCT in patients with lymphoma. METHODS: This was a retrospective cohort study of 243 consecutive patients who underwent a first autologous HCT for lymphoma between 2009 and 2014. CAC (Agatston score) was determined from chest computed tomography obtained <60 days from HCT. Multivariable Cox regression analysis was used to calculate hazard ratio (HR) estimates and 95% confidence intervals (CIs), adjusted for covariates (age, conventional risk factors [e.g., hypertension and dyslipidemia], and cancer treatment). RESULTS: The median age at HCT was 55.7 years (range, 18.5-75.1 years), 59% were male, and 60% were non-Hispanic White. The prevalence of CAC was 37%. The 5-year CVD incidence for the cohort was 12%, and there was an incremental increase in the incidence according to CAC score: 0 (6%), 1-100 (20%), and >100 (32%) (p = .001). CAC was significantly associated with CVD risk (HR, 3.0; 95% CI, 1.2-7.5) and worse 5-year survival (77% vs. 50%; p < .001; HR, 2.0; 95% CI, 1.1-3.4), compared to those without CAC. CONCLUSIONS: CAC is independently associated with CVD and survival after HCT. This highlights the importance of integrating readily available imaging information in risk stratification and decision-making in patients undergoing HCT, which sets the stage for strategies to optimize outcomes after HCT.


Subject(s)
Cardiovascular Diseases , Hematopoietic Stem Cell Transplantation , Lymphoma , Transplantation, Autologous , Humans , Male , Female , Middle Aged , Hematopoietic Stem Cell Transplantation/adverse effects , Adult , Retrospective Studies , Aged , Lymphoma/therapy , Young Adult , Adolescent , Cardiovascular Diseases/etiology , Cardiovascular Diseases/epidemiology , Coronary Vessels/diagnostic imaging , Coronary Vessels/pathology , Coronary Vessels/metabolism , Risk Factors , Calcium/metabolism , Coronary Artery Disease/epidemiology , Incidence
3.
Circ Res ; 130(4): 632-651, 2022 02 18.
Article in English | MEDLINE | ID: mdl-35175846

ABSTRACT

In both cardiovascular disease and cancer, there are established sex-based differences in prevalence and outcomes. Males and females may also differ in terms of risk of cardiotoxicity following cancer therapy, including heart failure, cardiomyopathy, atherosclerosis, thromboembolism, arrhythmias, and myocarditis. Here, we describe sex-based differences in the epidemiology and pathophysiology of cardiotoxicity associated with anthracyclines, hematopoietic stem cell transplant (HCT), hormone therapy and immune therapy. Relative to males, the risk of anthracycline-induced cardiotoxicity is higher in prepubertal females, lower in premenopausal females, and similar in postmenopausal females. For autologous hematopoietic cell transplant, several studies suggest an increased risk of late heart failure in female lymphoma patients, but sex-based differences have not been shown for allogeneic hematopoietic cell transplant. Hormone therapies including GnRH (gonadotropin-releasing hormone) modulators, androgen receptor antagonists, selective estrogen receptor modulators, and aromatase inhibitors are associated with cardiotoxicity, including arrhythmia and venous thromboembolism. However, sex-based differences have not yet been elucidated. Evaluation of sex differences in cardiotoxicity related to immune therapy is limited, in part, due to low participation of females in relevant clinical trials. However, some studies suggest that females are at increased risk of immune checkpoint inhibitor myocarditis, although this has not been consistently demonstrated. For each of the aforementioned cancer therapies, we consider sex-based differences according to cardiotoxicity management. We identify knowledge gaps to guide future mechanistic and prospective clinical studies. Furthering our understanding of sex-based differences in cancer therapy cardiotoxicity can advance the development of targeted preventive and therapeutic cardioprotective strategies.


Subject(s)
Antineoplastic Agents/adverse effects , Cardiovascular Diseases/chemically induced , Cardiovascular Diseases/epidemiology , Heart Disease Risk Factors , Neoplasms/epidemiology , Sex Characteristics , Anthracyclines/adverse effects , Cardiotoxicity/diagnosis , Cardiotoxicity/epidemiology , Cardiovascular Diseases/diagnosis , Female , Hematopoietic Stem Cell Transplantation/adverse effects , Hormone Replacement Therapy/adverse effects , Humans , Immunotherapy/adverse effects , Male , Neoplasms/drug therapy
4.
Phys Chem Chem Phys ; 26(10): 8390-8396, 2024 Mar 06.
Article in English | MEDLINE | ID: mdl-38406868

ABSTRACT

The realization of quantum advantage with noisy-intermediate-scale quantum (NISQ) machines has become one of the major challenges in computational sciences. Maintaining coherence of a physical system with more than ten qubits is a critical challenge that motivates research on compact system representations to reduce algorithm complexity. Toward this end, the variational quantum eigensolver (VQE) used to perform quantum simulations is considered to be one of the most promising algorithms for quantum chemistry in the NISQ era. We investigate reduced mapping of one spatial orbital to a single qubit to analyze the ground state energy in a way that the Pauli operators of qubits are mapped to the creation/annihilation of singlet pairs of electrons. To include the effect of non-bosonic (or non-paired) excitations, we introduce a simple correction scheme in the electron correlation model approximated by the geometrical mean of the bosonic (or paired) terms. Employing it in a VQE algorithm, we assess ground state energies of H2O, N2, and Li2O in good agreement with full configuration interaction (FCI) models respectively, using only 6, 8, and 12 qubits with quantum gate depths proportional to the squares of the qubit counts. With the adopted seniority-zero approximation that uses only one half of the qubit counts of a conventional VQE algorithm, we find that our non-bosonic correction method reaches reliable quantum chemistry simulations at least for the tested systems.

5.
Opt Express ; 31(19): 30669-30681, 2023 Sep 11.
Article in English | MEDLINE | ID: mdl-37710606

ABSTRACT

We propose a novel heterodyne detection scheme for continuous-variable quantum key distribution (CVQKD), which measures both quadrature components of a quantum signal encoded in optical phase space. The proposed method uses time division to achieve identical performance to conventional heterodyne detection with only a single homodyne detection system. Our method also uses a Faraday-Michelson interferometer to make it independent of polarization drift and eliminate the need for dynamic polarization control. Our method is experimentally demonstrated using the Gaussian-modulated coherent-states (GMCS) protocol over a 20.06 km optical fiber channel, achieving an expected secret key rate of up to 0.187 Mbps.

6.
Curr Oncol Rep ; 25(9): 965-977, 2023 09.
Article in English | MEDLINE | ID: mdl-37273124

ABSTRACT

PURPOSE OF THE REVIEW: There have been increasing reports of cardiovascular complications of androgen deprivation therapy (ADT) leading to worse outcomes among patients with prostate cancer. While this may result from the direct effects of androgen suppression in the cardiovascular systems, there are ADT-type-specific distinct cardiovascular complications suggestive of mechanisms beyond androgen-mediated. Thus, it is critical to understand the biological and clinical impact of ADT on the cardiovascular system. RECENT FINDINGS: Gonadotropin-releasing hormone (GnRH) agonists cause increased cardiovascular events compared to GnRH antagonists. Androgen receptor antagonists are linked to an increased risk of long QT syndrome, torsades de pointes, and sudden cardiac death. Androgen synthesis inhibitors are associated with increased rates of hypertension, atrial tachyarrhythmia, and, in rare incidences, heart failure. ADT increases the risk of cardiovascular disease. The risk among ADT drugs differs and must be evaluated to develop a medically optimal plan for prostate cancer patients.


Subject(s)
Cardiovascular System , Prostatic Neoplasms , Male , Humans , Prostatic Neoplasms/drug therapy , Androgens/therapeutic use , Androgen Antagonists/adverse effects , Gonadotropin-Releasing Hormone/agonists , Gonadotropin-Releasing Hormone/therapeutic use , Biology
7.
Heart Fail Clin ; 18(3): 335-347, 2022 Jul.
Article in English | MEDLINE | ID: mdl-35718410

ABSTRACT

The development of human-induced pluripotent stem cell-derived cardiac cell types has created a new paradigm in assessing drug-induced cardiotoxicity. Advances in genomics and epigenomics have also implicated several genomic loci and biological pathways that may contribute to susceptibility to cancer therapies. In this review, we first provide a brief overview of the cardiotoxicity associated with chemotherapy. We then provide a detailed summary of systems biology approaches being applied to elucidate potential molecular mechanisms involved in cardiotoxicity. Finally, we discuss combining systems biology approaches with iPSC technology to help discover molecular mechanisms associated with cardiotoxicity.


Subject(s)
Induced Pluripotent Stem Cells , Neoplasms , Cardiotoxicity/etiology , Humans , Induced Pluripotent Stem Cells/metabolism , Myocytes, Cardiac/metabolism , Neoplasms/drug therapy , Neoplasms/metabolism , Systems Biology
8.
Circ Res ; 125(2): 212-222, 2019 07 05.
Article in English | MEDLINE | ID: mdl-31079550

ABSTRACT

RATIONALE: Calcium channel blockers (CCBs) are an important class of drugs in managing cardiovascular diseases. Patients usually rely on these medications for the remainder of their lives after diagnosis. Although the acute pharmacological actions of CCBs in the hearts are well-defined, little is known about the drug-specific effects on human cardiomyocyte transcriptomes and physiological alterations after long-term exposure. OBJECTIVE: This study aimed to simulate chronic CCB treatment and to examine both the functional and transcriptomic changes in human cardiomyocytes. METHODS AND RESULTS: We differentiated cardiomyocytes and generated engineered heart tissues from 3 human induced pluripotent stem cell lines and exposed them to 4 different CCBs-nifedipine, amlodipine, diltiazem, and verapamil-at their physiological serum concentrations for 2 weeks. Without inducing cell death and damage to myofilament structure, CCBs elicited line-specific inhibition on calcium kinetics and contractility. While all 4 CCBs exerted similar inhibition on calcium kinetics, verapamil applied the strongest inhibition on cardiomyocyte contractile function. By profiling cardiomyocyte transcriptome after CCB treatment, we identified little overlap in their transcriptome signatures. Verapamil is the only inhibitor that reduced the expression of contraction-related genes, such as MYH (myosin heavy chain) and troponin I, consistent with its depressive effects on contractile function. The reduction of these contraction-related genes may also explain the responsiveness of patients with hypertrophic cardiomyopathy to verapamil in managing left ventricular outflow tract obstruction. CONCLUSIONS: This is the first study to identify the transcriptome signatures of different CCBs in human cardiomyocytes. The distinct gene expression patterns suggest that although the 4 inhibitors act on the same target, they may have distinct effects on normal cardiac cell physiology.


Subject(s)
Calcium Channel Blockers/pharmacology , Induced Pluripotent Stem Cells/drug effects , Myocytes, Cardiac/metabolism , Transcriptome , Amlodipine/pharmacology , Cell Differentiation , Cells, Cultured , Diltiazem/pharmacology , Humans , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Myocytes, Cardiac/cytology , Myocytes, Cardiac/drug effects , Myosin Heavy Chains/genetics , Myosin Heavy Chains/metabolism , Nifedipine/pharmacology , Verapamil/pharmacology
9.
Circulation ; 139(21): 2451-2465, 2019 05 21.
Article in English | MEDLINE | ID: mdl-30866650

ABSTRACT

BACKGROUND: Molecular targeted chemotherapies have been shown to significantly improve the outcomes of patients who have cancer, but they often cause cardiovascular side effects that limit their use and impair patients' quality of life. Cardiac dysfunction induced by these therapies, especially trastuzumab, shows a distinct cardiotoxic clinical phenotype in comparison to the cardiotoxicity induced by conventional chemotherapies. METHODS: We used the human induced pluripotent stem cell-derived cardiomyocyte (iPSC-CM) platform to determine the underlying cellular mechanisms in trastuzumab-induced cardiac dysfunction. We assessed the effects of trastuzumab on structural and functional properties in iPSC-CMs from healthy individuals and performed RNA-sequencing to further examine the effect of trastuzumab on iPSC-CMs. We also generated human induced pluripotent stem cells from patients receiving trastuzumab and examined whether patients' phenotype could be recapitulated in vitro by using patient-specific iPSC-CMs. RESULTS: We found that clinically relevant doses of trastuzumab significantly impaired the contractile and calcium-handling properties of iPSC-CMs without inducing cardiomyocyte death or sarcomeric disorganization. RNA-sequencing and subsequent functional analysis revealed mitochondrial dysfunction and altered the cardiac energy metabolism pathway as primary causes of trastuzumab-induced cardiotoxic phenotype. Human iPSC-CMs generated from patients who received trastuzumab and experienced severe cardiac dysfunction were more vulnerable to trastuzumab treatment than iPSC-CMs generated from patients who did not experience cardiac dysfunction following trastuzumab therapy. It is important to note that metabolic modulation with AMP-activated protein kinase activators could avert the adverse effects induced by trastuzumab. CONCLUSIONS: Our results indicate that alterations in cellular metabolic pathways in cardiomyocytes could be a key mechanism underlying the development of cardiac dysfunction following trastuzumab therapy; therefore, targeting the altered metabolism may be a promising therapeutic approach for trastuzumab-induced cardiac dysfunction.


Subject(s)
Antineoplastic Agents, Immunological/toxicity , Breast Neoplasms/drug therapy , Heart Diseases/chemically induced , Induced Pluripotent Stem Cells/drug effects , Trastuzumab/toxicity , AMP-Activated Protein Kinases/metabolism , Calcium Signaling/drug effects , Cardiotoxicity , Case-Control Studies , Cell Line , Energy Metabolism/drug effects , Female , Heart Diseases/metabolism , Heart Diseases/pathology , Heart Diseases/physiopathology , Humans , Induced Pluripotent Stem Cells/metabolism , Induced Pluripotent Stem Cells/pathology , Myocardial Contraction/drug effects , Phenotype , Risk Factors , Transcriptome/drug effects
10.
Circ Res ; 123(4): 443-450, 2018 08 03.
Article in English | MEDLINE | ID: mdl-29986945

ABSTRACT

RATIONALE: Human-induced pluripotent stem cell-derived endothelial cells (iPSC-ECs) have risen as a useful tool in cardiovascular research, offering a wide gamut of translational and clinical applications. However, inefficiency of the currently available iPSC-EC differentiation protocol and underlying heterogeneity of derived iPSC-ECs remain as major limitations of iPSC-EC technology. OBJECTIVE: Here, we performed droplet-based single-cell RNA sequencing (scRNA-seq) of the human iPSCs after iPSC-EC differentiation. Droplet-based scRNA-seq enables analysis of thousands of cells in parallel, allowing comprehensive analysis of transcriptional heterogeneity. METHODS AND RESULTS: Bona fide iPSC-EC cluster was identified by scRNA-seq, which expressed high levels of endothelial-specific genes. iPSC-ECs, sorted by CD144 antibody-conjugated magnetic sorting, exhibited standard endothelial morphology and function including tube formation, response to inflammatory signals, and production of NO. Nonendothelial cell populations resulting from the differentiation protocol were identified, which included immature cardiomyocytes, hepatic-like cells, and vascular smooth muscle cells. Furthermore, scRNA-seq analysis of purified iPSC-ECs revealed transcriptional heterogeneity with 4 major subpopulations, marked by robust enrichment of CLDN5, APLNR, GJA5, and ESM1 genes, respectively. CONCLUSIONS: Massively parallel, droplet-based scRNA-seq allowed meticulous analysis of thousands of human iPSCs subjected to iPSC-EC differentiation. Results showed inefficiency of the differentiation technique, which can be improved with further studies based on identification of molecular signatures that inhibit expansion of nonendothelial cell types. Subtypes of bona fide human iPSC-ECs were also identified, allowing us to sort for iPSC-ECs with specific biological function and identity.


Subject(s)
Endothelial Cells/metabolism , Induced Pluripotent Stem Cells/metabolism , Transcriptome , Apelin Receptors/genetics , Apelin Receptors/metabolism , Cell Differentiation , Cells, Cultured , Claudin-5/genetics , Claudin-5/metabolism , Connexins/genetics , Connexins/metabolism , Endothelial Cells/cytology , Humans , Induced Pluripotent Stem Cells/cytology , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Proteoglycans/genetics , Proteoglycans/metabolism , Single-Cell Analysis , Gap Junction alpha-5 Protein
11.
Curr Cardiol Rep ; 22(5): 34, 2020 04 29.
Article in English | MEDLINE | ID: mdl-32350632

ABSTRACT

PURPOSE OF REVIEW: COronaVirus Disease 2019 (COVID-19) has spread at unprecedented speed and scale into a global pandemic with cardiovascular risk factors and complications emerging as important disease modifiers. We aim to review available clinical and biomedical literature on cardiovascular risks of COVID-19. RECENT FINDINGS: SARS-CoV2, the virus responsible for COVID-19, enters the cell via ACE2 expressed in select organs. Emerging epidemiological evidence suggest cardiovascular risk factors are associated with increased disease severity and mortality in COVID-19 patients. Patients with a more severe form of COVID-19 are also more likely to develop cardiac complications such as myocardial injury and arrhythmia. The true incidence of and mechanism underlying these events remain elusive. Cardiovascular diseases appear intricately linked with COVID-19, with cardiac complications contributing to the elevated morbidity/mortality of COVID-19. Robust epidemiologic and biologic studies are urgently needed to better understand the mechanism underlying these associations to develop better therapies.


Subject(s)
Cardiovascular Diseases/mortality , Cardiovascular Diseases/physiopathology , Coronavirus Infections/mortality , Coronavirus Infections/physiopathology , Pneumonia, Viral/mortality , Pneumonia, Viral/physiopathology , Angiotensin-Converting Enzyme 2 , Betacoronavirus/metabolism , Betacoronavirus/pathogenicity , Betacoronavirus/ultrastructure , COVID-19 , Cardiovascular Diseases/epidemiology , Cardiovascular Diseases/virology , Comorbidity , Coronavirus/metabolism , Coronavirus/pathogenicity , Coronavirus/ultrastructure , Coronavirus Infections/epidemiology , Coronavirus Infections/virology , Humans , Pandemics , Peptidyl-Dipeptidase A/metabolism , Pneumonia, Viral/epidemiology , Pneumonia, Viral/virology , Risk Factors , SARS-CoV-2 , Treatment Outcome
12.
Curr Cardiol Rep ; 22(5): 32, 2020 04 21.
Article in English | MEDLINE | ID: mdl-32318865

ABSTRACT

PURPOSE OF REVIEW: Coronavirus disease of 2019 (COVID-19) is a cause of significant morbidity and mortality worldwide. While cardiac injury has been demonstrated in critically ill COVID-19 patients, the mechanism of injury remains unclear. Here, we review our current knowledge of the biology of SARS-CoV-2 and the potential mechanisms of myocardial injury due to viral toxicities and host immune responses. RECENT FINDINGS: A number of studies have reported an epidemiological association between history of cardiac disease and worsened outcome during COVID infection. Development of new onset myocardial injury during COVID-19 also increases mortality. While limited data exist, potential mechanisms of cardiac injury include direct viral entry through the angiotensin-converting enzyme 2 (ACE2) receptor and toxicity in host cells, hypoxia-related myocyte injury, and immune-mediated cytokine release syndrome. Potential treatments for reducing viral infection and excessive immune responses are also discussed. COVID patients with cardiac disease history or acquire new cardiac injury are at an increased risk for in-hospital morbidity and mortality. More studies are needed to address the mechanism of cardiotoxicity and the treatments that can minimize permanent damage to the cardiovascular system.


Subject(s)
Coronavirus Infections/complications , Coronavirus Infections/immunology , Heart Diseases/complications , Heart Diseases/immunology , Heart Diseases/virology , Pneumonia, Viral/complications , Pneumonia, Viral/immunology , Angiotensin-Converting Enzyme 2 , Animals , Betacoronavirus , COVID-19 , Coronavirus Infections/therapy , Cytokines/immunology , Humans , Hypoxia/pathology , Myocytes, Cardiac/pathology , Pandemics , Peptidyl-Dipeptidase A/metabolism , Pneumonia, Viral/therapy , SARS-CoV-2
13.
Curr Cardiol Rep ; 22(5): 36, 2020 05 13.
Article in English | MEDLINE | ID: mdl-32405913

ABSTRACT

It has been pointed out that the second paragraph of the section "Treatments for SARS-CoV-2 Infection" contains an error. The original article has been corrected.

14.
Eur Heart J ; 40(45): 3685-3695, 2019 12 01.
Article in English | MEDLINE | ID: mdl-31219556

ABSTRACT

AIMS: Diastolic dysfunction (DD) is common among hypertrophic cardiomyopathy (HCM) patients, causing major morbidity and mortality. However, its cellular mechanisms are not fully understood, and presently there is no effective treatment. Patient-specific induced pluripotent stem cell-derived cardiomyocytes (iPSC-CMs) hold great potential for investigating the mechanisms underlying DD in HCM and as a platform for drug discovery. METHODS AND RESULTS: In the present study, beating iPSC-CMs were generated from healthy controls and HCM patients with DD. Micropatterned iPSC-CMs from HCM patients showed impaired diastolic function, as evidenced by prolonged relaxation time, decreased relaxation rate, and shortened diastolic sarcomere length. Ratiometric Ca2+ imaging indicated elevated diastolic [Ca2+]i and abnormal Ca2+ handling in HCM iPSC-CMs, which were exacerbated by ß-adrenergic challenge. Combining Ca2+ imaging and traction force microscopy, we observed enhanced myofilament Ca2+ sensitivity (measured as dF/Δ[Ca2+]i) in HCM iPSC-CMs. These results were confirmed with genome-edited isogenic iPSC lines that carry HCM mutations, indicating that cytosolic diastolic Ca2+ overload, slowed [Ca2+]i recycling, and increased myofilament Ca2+ sensitivity, collectively impairing the relaxation of HCM iPSC-CMs. Treatment with partial blockade of Ca2+ or late Na+ current reset diastolic Ca2+ homeostasis, restored diastolic function, and improved long-term survival, suggesting that disturbed Ca2+ signalling is an important cellular pathological mechanism of DD. Further investigation showed increased expression of L-type Ca2+channel (LTCC) and transient receptor potential cation channels (TRPC) in HCM iPSC-CMs compared with control iPSC-CMs, which likely contributed to diastolic [Ca2+]i overload. CONCLUSION: In summary, this study recapitulated DD in HCM at the single-cell level, and revealed novel cellular mechanisms and potential therapeutic targets of DD using iPSC-CMs.


Subject(s)
Cardiomyopathy, Hypertrophic/genetics , Heart Failure, Diastolic/physiopathology , Induced Pluripotent Stem Cells/metabolism , Myocytes, Cardiac/metabolism , Calcium/metabolism , Cardiac Myosins/genetics , Cardiomyopathy, Hypertrophic/drug therapy , Cardiomyopathy, Hypertrophic/physiopathology , Carrier Proteins/genetics , Case-Control Studies , Cell Differentiation , Heart Failure, Diastolic/drug therapy , Heart Failure, Diastolic/mortality , Humans , Mutation , Myosin Heavy Chains/genetics , Phenotype , Sarcomeres/physiology , Troponin T/genetics
15.
J Vasc Res ; 56(1): 11-15, 2019.
Article in English | MEDLINE | ID: mdl-30763932

ABSTRACT

Peripartum cardiomyopathy (PPCM) is a rare form of congestive heart failure characterized by left ventricular dysfunction that develops towards the end of pregnancy or during the early postpartum phase. Even though the majority of PPCM patients show partial or complete recovery of their heart functions, the mortality rate of PPCM remains high. Previous research has suggested that vascular dysfunction triggered by late-gestational hormones and potent anti-angiogenic factors play key roles in the pathogenesis of PPCM; however, the exact mechanisms remain elusive due to limited patient tissues for characterization. Here, we report a case of PPCM where the coronary vessels from the patient's explanted heart showed marked vascular dysfunction with impaired nitric oxide response. Importantly, these vessels exhibited deficient adenosine-mediated vasorelaxation when subjected to myograph studies, suggesting impaired Kv7 ion channels. Results from this work may lead to new therapeutic strategies for improving Kv7 function in PPCM patients.


Subject(s)
Cardiomyopathies/etiology , Coronary Artery Disease/etiology , Coronary Vessels/physiopathology , Peripartum Period , Vasodilation , Ventricular Dysfunction, Left/etiology , Ventricular Function, Left , Cardiomyopathies/diagnostic imaging , Cardiomyopathies/physiopathology , Cardiomyopathies/surgery , Coronary Artery Disease/diagnostic imaging , Coronary Artery Disease/physiopathology , Coronary Artery Disease/surgery , Coronary Vessels/diagnostic imaging , Coronary Vessels/metabolism , Disease Progression , Female , Heart Failure/etiology , Heart Failure/physiopathology , Heart Failure/surgery , Heart Transplantation , Humans , KCNQ Potassium Channels/metabolism , Middle Aged , Pregnancy , Ventricular Dysfunction, Left/diagnostic imaging , Ventricular Dysfunction, Left/physiopathology , Ventricular Dysfunction, Left/surgery
17.
Opt Express ; 26(13): 17662-17668, 2018 Jun 25.
Article in English | MEDLINE | ID: mdl-30119576

ABSTRACT

Orthogonal frequency division multiple access (OFDMA) uplink in a passive optical network (PON) requires the delay alignment for OFDMA symbols from remotely distributed optical network units (ONUs). In this paper, we experimentally demonstrate and analyze the performance of a Zadoff-Chu (ZC) sequence-based upstream ranging scheme in an intensity modulation/direct detection (IM/DD)-based OFDMA-PON. The experimental results show that the proposed scheme can achieve upstream synchronization with only marginal inter-carrier interference (ICI) and requires no additional bandwidths in a typical OFDMA transmission with cyclic prefix (CP).

18.
Opt Express ; 25(10): 11894-11909, 2017 May 15.
Article in English | MEDLINE | ID: mdl-28788747

ABSTRACT

Needs for providing security to end users have brought installation of quantum key distribution (QKD) in one-to-many access networks such as passive optical networks. In the networks, a presence of optical power splitters makes issues for secure key rate more important. However, researches for QKD in access networks have mainly focused on implementation issues rather than protocol development for key rate enhancement. Since secure key rate is theoretically limited by a protocol, researches without protocol development cannot overcome the limit of secure key rate given by a protocol. This brings need of researches for protocol development. In this paper, we provide a new approach which provides secure key rate enhancement over the conventional protocol. Specifically, we propose the secure key rate formula in a passive optical network by extending the secure key rate formula based on the decoy-state BB84 protocol. For a passive optical network, we provide a way that incorporates cooperation across end users. Then, we show that the way can mitigate a photon number splitting (PNS) attack which is crucial in an well known decoy BB84 protocol. Especially, the proposed scheme enables multi-photon states to serve as secure keys unlike the conventional decoy BB84 protocol. Numerical simulations demonstrate that our proposed scheme outperforms the decoy BB84 protocol in secure key rate.

SELECTION OF CITATIONS
SEARCH DETAIL