1
|
Sato Y, Yoshihisa A, Ohashi N, Takeishi R, Sekine T, Nishiura K, Ogawara R, Ichimura S, Kimishima Y, Yokokawa T, Miura S, Misaka T, Sato T, Oikawa M, Kobayashi A, Yamaki T, Nakazato K, Takeishi Y. Association of nighttime very short-term blood pressure variability determined by pulse transit time with adverse prognosis in patients with heart failure. Hypertens Res 2025; 48:1305-1314. [PMID: 39833554 DOI: 10.1038/s41440-025-02102-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Revised: 12/27/2024] [Accepted: 12/29/2024] [Indexed: 01/22/2025]
Abstract
Long-term blood pressure (BP) variability (BPV) is associated with adverse prognosis in patients with heart failure. However, the clinical significance of very short-term (beat-to-beat) BPV is unclear. We collected data on nighttime pulse transit time-based continuous beat-to-beat BP measurement in patients with heart failure (n = 366, median age 72.0, male sex 53.3%). Coefficient of variation (CoV) of pulse transit time-based BP was considered as very short-term BPV. The primary outcome was a composite of heart failure hospitalization or cardiac death. Median values (25th and 75th percentiles) of systolic and diastolic BP CoV were 3.6% (2.8%, 4.5%) and 5.1% (3.8%, 6.5%), respectively. During a median follow-up period of 1084 days after BPV evaluation, 71 patients experienced the primary outcome. When the patients were divided into tertiles based on the systolic and diastolic BPV, the primary outcome occurred most frequently in the highest tertile of BPV. Multivariable Cox proportional hazard analysis revealed that systolic and diastolic BPV, as continuous variables, were independently associated with the primary outcome (hazard ratio 1.199 and 1.101, respectively). In conclusion, high nighttime very short-term BPV was associated with adverse prognosis in patients with heart failure.
Collapse
Affiliation(s)
- Yu Sato
- Department of Cardiovascular Medicine, Fukushima Medical University, Fukushima, Japan
| | - Akiomi Yoshihisa
- Department of Cardiovascular Medicine, Fukushima Medical University, Fukushima, Japan.
- Department of Clinical Laboratory Sciences, Fukushima Medical University, Fukushima, Japan.
| | - Naoto Ohashi
- Department of Cardiovascular Medicine, Fukushima Medical University, Fukushima, Japan
| | - Ryohei Takeishi
- Department of Cardiovascular Medicine, Fukushima Medical University, Fukushima, Japan
| | - Toranosuke Sekine
- Department of Cardiovascular Medicine, Fukushima Medical University, Fukushima, Japan
| | - Kazuto Nishiura
- Department of Cardiovascular Medicine, Fukushima Medical University, Fukushima, Japan
| | - Ryo Ogawara
- Department of Cardiovascular Medicine, Fukushima Medical University, Fukushima, Japan
| | - Shohei Ichimura
- Department of Cardiovascular Medicine, Fukushima Medical University, Fukushima, Japan
| | - Yusuke Kimishima
- Department of Cardiovascular Medicine, Fukushima Medical University, Fukushima, Japan
| | - Tetsuro Yokokawa
- Department of Cardiovascular Medicine, Fukushima Medical University, Fukushima, Japan
| | - Shunsuke Miura
- Department of Cardiovascular Medicine, Fukushima Medical University, Fukushima, Japan
| | - Tomofumi Misaka
- Department of Cardiovascular Medicine, Fukushima Medical University, Fukushima, Japan
- Department of Community Cardiovascular Medicine, Fukushima Medical University, Fukushima, Japan
| | - Takamasa Sato
- Department of Cardiovascular Medicine, Fukushima Medical University, Fukushima, Japan
- Department of Community Cardiovascular Medicine, Fukushima Medical University, Fukushima, Japan
| | - Masayoshi Oikawa
- Department of Cardiovascular Medicine, Fukushima Medical University, Fukushima, Japan
| | - Atsushi Kobayashi
- Department of Cardiovascular Medicine, Fukushima Medical University, Fukushima, Japan
| | - Takayoshi Yamaki
- Department of Cardiovascular Medicine, Fukushima Medical University, Fukushima, Japan
| | - Kazuhiko Nakazato
- Department of Cardiovascular Medicine, Fukushima Medical University, Fukushima, Japan
| | - Yasuchika Takeishi
- Department of Cardiovascular Medicine, Fukushima Medical University, Fukushima, Japan
| |
Collapse
|
2
|
Li J, Sun Y, Ren J, Wu Y, He Z. Machine Learning for In-hospital Mortality Prediction in Critically Ill Patients With Acute Heart Failure: A Retrospective Analysis Based on the MIMIC-IV Database. J Cardiothorac Vasc Anesth 2025; 39:666-674. [PMID: 39779429 DOI: 10.1053/j.jvca.2024.12.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Revised: 11/03/2024] [Accepted: 12/09/2024] [Indexed: 01/11/2025]
Abstract
BACKGROUND The incidence, mortality, and readmission rates for acute heart failure (AHF) are high, and the in-hospital mortality for AHF patients in the intensive care unit (ICU) is higher. However, there is currently no method to accurately predict the mortality of AHF patients. METHODS The Medical Information Mart for Intensive Care Ⅳ (MIMIC-Ⅳ) database was used to perform a retrospective study. Patients meeting the inclusion criteria were identified from the MIMIC-Ⅳ database and randomly divided into a training set (n = 3,580, 70%) and a validation set (n = 1,534, 30%). The variates collected include demographic data, vital signs, comorbidities, laboratory test results, and treatment information within 24 hours of ICU admission. By using the least absolute shrinkage and selection operator (LASSO) regression model in the training set, variates that affect the in-hospital mortality of AHF patients were screened. Subsequently, in the training set, five common machine learning (ML) algorithms were applied to construct models using variates selected by LASSO to predict the in-hospital mortality of AHF patients. The predictive ability of the models was evaluated for sensitivity, specificity, accuracy, the area under the curve of receiver operating characteristics, and clinical net benefit in the validation set. To obtain a model with the best predictive ability, the predictive ability of common scoring systems was compared with the best ML model. RESULTS Among the 5,114 patients, in-hospital mortality was 12.5%. Comparing the area under the curve, the XGBoost model had the best predictive ability among all ML models, and the XGBoost model was chosen as the final model for its higher net benefit. Its predictive ability was superior to common scoring systems. CONCLUSIONS The XGBoost model can effectively predict the in-hospital mortality of AHF patients admitted to the ICU, which may assist clinicians in precise management and early intervention for patients with AHF to reduce mortality.
Collapse
Affiliation(s)
- Jun Li
- Department of Anesthesiology, Dazhou Central Hospital, Dazhou, Sichuan, China
| | - Yiwu Sun
- Department of Anesthesiology, Dazhou Central Hospital, Dazhou, Sichuan, China
| | - Jie Ren
- Department of Anesthesiology, Guizhou Provincial People's Hospital, Guiyang, Guizhou, China
| | - Yifan Wu
- Department of Anesthesiology, Shanghai Sixth People's Hospital, Shanghai, China
| | - Zhaoyi He
- Surgical Anesthesia Center, The First People's Hospital of Longquanyi District, Chengdu, China.
| |
Collapse
|
3
|
Zhang S, Huang Y, Han C, Chen M, Yang Z, Wang C. Circulating mitochondria carrying cGAS promote endothelial Secreted group IIA phospholipase A2-mediated neuroinflammation through activating astroglial/microglial Integrin-alphavbeta3 in subfornical organ to augment central sympathetic overdrive in heart failure rats. Int Immunopharmacol 2025; 144:113649. [PMID: 39586230 DOI: 10.1016/j.intimp.2024.113649] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Revised: 11/13/2024] [Accepted: 11/14/2024] [Indexed: 11/27/2024]
Abstract
BACKGROUND Sympathoexcitation, a manifestation of heart-brain axis dysregulation, contributes to the progression of heart failure (HF). Our recent study revealed that circulating mitochondria (C-Mito), a newly identified mediator of multi-organ communication, promote sympathoexcitation in HF by aggravating endothelial cell (EC)-derived neuroinflammation in the subfornical organ (SFO), the cardiovascular autonomic neural center. The precise molecular mechanism by which C-Mito promotes SFO-induced endothelial neuroinflammation has not been fully elucidated. OBJECTIVE C-Mito carrying cGAS promote sympathoexcitation by targeting PLA2G2A in ECs of the SFO in HF rats. METHODS Male Sprague-Dawley (SD) rats received a subcutaneous injection of isoprenaline (ISO) at a dosage of 5 mg/kg/day for seven consecutive days to establish a HF model. C-Mito were isolated from HF rats and evaluated. The level of cGAS, a dsDNA sensor recently discovered to be directly localized on the outer membrane of mitochondria, was detected in C-Mito. C-Mito from HF rats (C-MitoHF) or control rats (C-MitoCtrl) were intravenously infused into HF rats. The accumulation of C-Mito in the ECs in the SFO was detected via double immunofluorescence staining. The SFO was processed for RNA sequencing (RNA-Seq) analysis. Secreted group IIA phospholipase A2 (PLA2G2A), the key gene involved in C-MitoHF-associated SFO dysfunction, was identified via bioinformatics analysis. Upregulation of PLA2G2A in the SFO ECs was assessed via immunofluorescence staining and immunoblotting, and PLA2G2A activity was evaluated. The interaction between cGAS and PLA2G2A was detected via co-immunoprecipitation. The dowstream molecular mechanisms of which PLA2G2A induced astroglial/microglial activation were also investigated. AAV9-TIE-shRNA (PLA2G2A) was introduced into the SFO to specifically knockdown endothelial PLA2G2A. Neuronal activation and glial proinflammatory polarization in the SFO were also evaluated. Renal sympathetic nerve activity (RSNA) was measured to evaluate central sympathetic output. Cardiac sympathetic hyperinnervation, myocardial remodeling, and left ventricular systolic function were assessed in C-Mito-treated HF rats. RESULTS Respiratory functional incompetence and oxidative damage were observed in C-MitoHF compared with C-MitoCtrl. Surprisingly, cGAS protein levels in C-MitoHF were significantly higher than those in C-MitoCtrl, while blocking cGAS with its specific inhibitor, RU.521, mitigated respiratory dysfunction and oxidative injury in C-MitoHF. C-Mito entered the ECs of the SFO in HF rats. RNA sequencing revealed that PLA2G2A is a key molecule for the induction of SFO dysfunction by C-MitoHF. The immunoblotting and immunofluorescence results confirmed that, compared with C-MitoCtrl, C-MitoHF increased endothelial PLA2G2A expression in the SFO of HF rats, which could be alleviated by attenuating C-MitoHF-localized cGAS. Furthermore, we found that cGAS directly interacts with PLA2G2A, increased the activity of PLA2AG2, which produced arachidonic acid, and also promoted PLA2G2A secretion in brain ECs. In addition, the inhibition of PLA2G2A in brain ECs significantly mitigated the proinflammatory effect of conditioned cell culture medium from C-MitoHF-treated ECs on astroglia and microglia. Also, we found that PLA2G2A secreted from ECs insulted by C-Mito induced neuroinflammation through activating astriglial/microglial Integrin-alphavbeta3 in the SFO, which further promote central sympathetic overdrive in HF rats. Specific knockdown of endothelial PLA2G2A in the SFO mitigated C-MitoHF-induced presympathetic neuronal sensitization, cardiac sympathetic hyperinnervation, RSNA activation, myocardial remodeling, and systolic dysfunction in HF rats. CONCLUSION C-Mito carrying cGAS promoted cardiac sympathoexcitation by directly targeting PLA2G2A in the ECs of the SFO in HF rats. Secreted PLA2G2A derived from ECs insulted by C-Mito induced neuroinflammation through activating astriglial/microglial Integrin-alphavbeta3 in the SFO, which further promote central sympathetic overdrive in HF rats. Our study indicated that inhibiting cGAS in C-Mito might be a potential treatment for central sympathetic overdrive in HF.
Collapse
Affiliation(s)
- Shutian Zhang
- Department of Cardiac Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, China; Shanghai Institute of Cardiovascular Diseases, Shanghai 200032, China.
| | - Yijun Huang
- Department of Cardiac Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, China; Shanghai Institute of Cardiovascular Diseases, Shanghai 200032, China
| | - Chengzhi Han
- Department of Cardiac Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, China; Shanghai Institute of Cardiovascular Diseases, Shanghai 200032, China
| | - Maoxiang Chen
- Department of Cardiac Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, China; Shanghai Institute of Cardiovascular Diseases, Shanghai 200032, China
| | - Zhaohua Yang
- Department of Cardiac Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, China; Shanghai Institute of Cardiovascular Diseases, Shanghai 200032, China.
| | - Chunsheng Wang
- Department of Cardiac Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, China; Shanghai Institute of Cardiovascular Diseases, Shanghai 200032, China.
| |
Collapse
|
4
|
Mi Y, Yu H, Wang P, Miao Y, Teng X, Jin S, Xiao L, Xue H, Tian D, Guo Q, Wu Y. Tyrosine hydroxylase-positive neurons in the rostral ventrolateral medulla mediate sympathetic activation in sepsis. Life Sci 2024; 358:123118. [PMID: 39384147 DOI: 10.1016/j.lfs.2024.123118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Revised: 09/26/2024] [Accepted: 10/05/2024] [Indexed: 10/11/2024]
Abstract
AIM Sepsis results in high mortality and is associated with organ dysfunction caused by infection. The present study aimed to elucidate whether early-stage sympathetic activation is associated with the prognosis of sepsis and its possible mechanisms. METHODS Patients with sepsis and healthy controls were included. Sepsis in rats was induced by lipopolysaccharide. Dexmedetomidine, a α2-adrenergic receptor agonist, was used in patients and rats with sepsis to evaluate the role of the sympathetic nervous system in sepsis. Holter monitoring was used to detect heart rate variability, while plasma samples were obtained to measure levels of norepinephrine and inflammatory markers. Mean arterial pressure, heart rate, and renal sympathetic nerve activity were recorded. Immunofluorescence was used to detect the activation of neurons in the rostral ventrolateral medulla (RVLM). RESULTS In patients with sepsis, plasma levels of norepinephrine and interleukin-1β were higher compared with those in controls and positively correlated with acute physiology and chronic health evaluation (APACHE) II. SDNN and SDANN were significantly reduced as well as negatively correlated with APACHE II. Meanwhile, rats with sepsis showed increased of sympathetic outflow and plasma levels of norepinephrine, with increased c-fos levels in the RVLM. Treatment with dexmedetomidine could improve prognosis. Lesion of tyrosine hydroxylase-positive neurons in the RVLM attenuated sympathetic activation and target organs damage in septic rats as well as improved survival. CONCLUSION The results suggest that tyrosine hydroxylase-positive neurons in the RVLM might contribute to the prognosis of sepsis via activation of the sympathetic nervous system, while dexmedetomidine could ameliorate sepsis via inhibiting sympathetic activation.
Collapse
Affiliation(s)
- Yuan Mi
- Department of Physiology, Institute of Basic Medicine, Hebei Medical University, 361 Zhongshan East Road, Shijiazhuang 050017, China; Department of Emergency, The Fourth Hospital of Hebei Medical University, Shijiazhuang 050011, China
| | - Hao Yu
- Department of Physiology, Institute of Basic Medicine, Hebei Medical University, 361 Zhongshan East Road, Shijiazhuang 050017, China
| | - Ping Wang
- Department of Physiology, Institute of Basic Medicine, Hebei Medical University, 361 Zhongshan East Road, Shijiazhuang 050017, China
| | - Yuxin Miao
- Department of Physiology, Institute of Basic Medicine, Hebei Medical University, 361 Zhongshan East Road, Shijiazhuang 050017, China
| | - Xu Teng
- Department of Physiology, Institute of Basic Medicine, Hebei Medical University, 361 Zhongshan East Road, Shijiazhuang 050017, China
| | - Sheng Jin
- Department of Physiology, Institute of Basic Medicine, Hebei Medical University, 361 Zhongshan East Road, Shijiazhuang 050017, China
| | - Lin Xiao
- Department of Physiology, Institute of Basic Medicine, Hebei Medical University, 361 Zhongshan East Road, Shijiazhuang 050017, China
| | - Hongmei Xue
- Department of Physiology, Institute of Basic Medicine, Hebei Medical University, 361 Zhongshan East Road, Shijiazhuang 050017, China
| | - Danyang Tian
- Department of Physiology, Institute of Basic Medicine, Hebei Medical University, 361 Zhongshan East Road, Shijiazhuang 050017, China
| | - Qi Guo
- Department of Physiology, Institute of Basic Medicine, Hebei Medical University, 361 Zhongshan East Road, Shijiazhuang 050017, China; Experimental Center for Teaching, Hebei Medical University, Shijiazhuang 050017, China; Hebei Key Laboratory of Cardiovascular Homeostasis and Aging, 050017 Hebei, China.
| | - Yuming Wu
- Department of Physiology, Institute of Basic Medicine, Hebei Medical University, 361 Zhongshan East Road, Shijiazhuang 050017, China; Hebei Collaborative Innovation Center for Cardio-Cerebrovascular Disease, Shijiazhuang 050017, China; The Key Laboratory of Neural and Vascular Biology, Ministry of Education, China; Hebei Key Laboratory of Cardiovascular Homeostasis and Aging, 050017 Hebei, China.
| |
Collapse
|
5
|
Li X, Zhou Y, Wang F, Wang L. Sex-Dimorphic Kidney-Brain Connectivity Map of Mice. Neurosci Bull 2024; 40:1445-1457. [PMID: 38896358 PMCID: PMC11422536 DOI: 10.1007/s12264-024-01240-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Accepted: 12/09/2023] [Indexed: 06/21/2024] Open
Abstract
The kidneys are essential organs that help maintain homeostasis, and their function is regulated by the neural system. Despite the anatomical multi-synaptic connection between the central autonomic nuclei and the kidneys, it remains unclear whether there are any variations in neural connections between the nervous systems and the renal cortex and medulla in male and female mice. Here, we used the pseudorabies virus to map the central innervation network of the renal cortex and medulla in both sexes. The data revealed that specific brain regions displayed either a contralateral-bias or ipsilateral-bias pattern while kidney-innervating neurons distributed symmetrically in the midbrain and hindbrain. Sex differences were observed in the distribution of neurons connected to the left kidney, as well as those connected to the renal cortex and medulla. Our findings provide a comprehensive understanding of the brain-kidney network in both males and females and may help shed light on gender differences in kidney function and disease susceptibility in humans.
Collapse
Affiliation(s)
- Xulin Li
- Shenzhen Key Laboratory of Neuropsychiatric Modulation, Shenzhen-Hong Kong Institute of Brain Science, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
- CAS Key Laboratory of Brain Connectome and Manipulation, the Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
- Guangdong Provincial Key Laboratory of Brain Connectome and Behavior, the Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
- University of Chinese Academy of Sciences, Beijing, 101408, China
| | - Yuan Zhou
- Shenzhen Key Laboratory of Neuropsychiatric Modulation, Shenzhen-Hong Kong Institute of Brain Science, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
- CAS Key Laboratory of Brain Connectome and Manipulation, the Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
- Guangdong Provincial Key Laboratory of Brain Connectome and Behavior, the Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
- University of Chinese Academy of Sciences, Beijing, 101408, China
| | - Feng Wang
- Shenzhen Key Laboratory of Neuropsychiatric Modulation, Shenzhen-Hong Kong Institute of Brain Science, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China.
- CAS Key Laboratory of Brain Connectome and Manipulation, the Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China.
- Guangdong Provincial Key Laboratory of Brain Connectome and Behavior, the Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China.
| | - Liping Wang
- Shenzhen Key Laboratory of Neuropsychiatric Modulation, Shenzhen-Hong Kong Institute of Brain Science, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China.
- CAS Key Laboratory of Brain Connectome and Manipulation, the Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China.
- Guangdong Provincial Key Laboratory of Brain Connectome and Behavior, the Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China.
| |
Collapse
|
6
|
Dhyani N, Tian C, Gao L, Rudebush TL, Zucker IH. Nrf2-Keap1 in Cardiovascular Disease: Which Is the Cart and Which the Horse? Physiology (Bethesda) 2024; 39:0. [PMID: 38687468 PMCID: PMC11460534 DOI: 10.1152/physiol.00015.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 04/23/2024] [Accepted: 04/24/2024] [Indexed: 05/02/2024] Open
Abstract
High levels of oxidant stress in the form of reactive oxidant species are prevalent in the circulation and tissues in various types of cardiovascular disease including heart failure, hypertension, peripheral arterial disease, and stroke. Here we review the role of nuclear factor erythroid 2-related factor 2 (Nrf2), an important and widespread antioxidant and anti-inflammatory transcription factor that may contribute to the pathogenesis and maintenance of cardiovascular diseases. We review studies showing that downregulation of Nrf2 exacerbates heart failure, hypertension, and autonomic function. Finally, we discuss the potential for using Nrf2 modulation as a therapeutic strategy for cardiovascular diseases and autonomic dysfunction.
Collapse
Affiliation(s)
- Neha Dhyani
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, Nebraska, United States
| | - Changhai Tian
- Department of Toxicology and Cancer Biology, University of Kentucky College of Medicine, Lexington, Kentucky, United States
| | - Lie Gao
- Department of Anesthesiology, University of Nebraska Medical Center, Omaha, Nebraska, United States
| | - Tara L Rudebush
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, Nebraska, United States
| | - Irving H Zucker
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, Nebraska, United States
| |
Collapse
|
7
|
Fujiu K. Brain-Heart Dialogue - Decoding Its Role in Homeostasis and Cardiovascular Disease. Circ J 2024; 88:1354-1359. [PMID: 37967922 DOI: 10.1253/circj.cj-23-0579] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/17/2023]
Abstract
Despite advancements in treatments for heart failure and lethal arrhythmias, achieving satisfactory life prognoses remains a challenge. A fresh perspective on the pathogenesis of heart disease is imperative to improve these prognoses. Our research has highlighted the role of cardiac macrophages in inhibiting the onset of heart failure and sudden cardiac death. We have recently unveiled a collaborative mechanism involving immune cells, brain neural networks, and the kidneys, which work in concert to combat cardiovascular diseases. This intricate organ network, orchestrated by the brain neural network and immune system, is pivotal in maintaining whole-body homeostasis. Disruptions in this harmonious interplay can precipitate various conditions, including heart failure and multiple organ failure, underscoring the significance of technological advancements in analytical methods and the advent of artificial intelligence. Recent strides in circulatory organ research have facilitated concurrent high-level analysis of the neural network and cardiovascular system. This review encapsulates these cutting-edge reports, evaluates the progress of research anchored in the fundamental concept that system failure of the cardiovascular organ precipitates cardiovascular disease, and offers valuable insights to guide future research.
Collapse
Affiliation(s)
- Katsuhito Fujiu
- Department of Cardiovascular Medicine, the University of Tokyo
| |
Collapse
|
8
|
Stutsman N, Habecker B, Pavlovic N, Jurgens CY, Woodward WR, Lee CS, Denfeld QE. Sympathetic dysfunction is associated with worse fatigue and early and subtle symptoms in heart failure: an exploratory sex-stratified analysis. Eur J Cardiovasc Nurs 2024; 23:532-539. [PMID: 38196102 PMCID: PMC11257751 DOI: 10.1093/eurjcn/zvad121] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Revised: 11/20/2023] [Accepted: 11/21/2023] [Indexed: 01/11/2024]
Abstract
AIMS Physical symptoms impact patients with heart failure (HF) despite treatment advancements; however, our understanding of the pathogenic mechanisms underlying HF symptoms remains limited, including sex differences therein. The objective of this study was to quantify associations between sympathetic markers [norepinephrine (NE) and 3,4-dihydroxyphenylglycol (DHPG)] and physical symptoms in patients with HF and to explore sex differences in these associations. METHODS AND RESULTS We performed a secondary analysis of combined data from two studies: outpatients with HF (n = 111), and patients prior to left ventricular assist device implantation (n = 38). Physical symptoms were measured with the Heart Failure Somatic Perception Scale (HFSPS) dyspnoea and early/subtle symptom subscales and the Functional Assessment in Chronic Illness Therapy Fatigue Scale (FACIT-F) to capture dyspnoea, early symptoms of decompensation, and fatigue. Norepinephrine and DHPG were measured with high-performance liquid chromatography with electrochemical detection. Multivariate linear regression was used to quantify associations between symptoms and sympathetic markers. The sample (n = 149) was 60.8 ± 15.7 years, 41% women, and 71% non-ischaemic aetiology. Increased plasma NE and NE:DHPG ratio were associated with worse FACIT-F scores (P = 0.043 and P = 0.013, respectively). Increased plasma NE:DHPG ratio was associated with worse HFSPS early/subtle symptoms (P = 0.025). In sex-stratified analyses, increased NE:DHPG ratio was associated with worse FACIT-F scores (P = 0.011) and HFSPS early/subtle scores (P = 0.022) among women but not men. CONCLUSION In patients with HF, sympathetic dysfunction is associated with worse fatigue and early/subtle physical symptoms with associations stronger in women than men.
Collapse
Affiliation(s)
- Nina Stutsman
- Oregon Health & Science University, School of Nursing, 3455 SW U.S. Veteran’s Hospital Road, Portland, OR 97239, USA
| | - Beth Habecker
- Oregon Health & Science University, Knight Cardiovascular Institute, 3181 SW Sam Jackson Park Road, Portland, OR 97239, USA
- Department of Chemical Physiology and Biochemistry, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Portland, OR 97239, USA
| | - Noelle Pavlovic
- Johns Hopkins School of Public Health, 615 N Wolfe Street, Baltimore, MD 21205, USA
| | - Corrine Y Jurgens
- Boston College, William F. Connell School of Nursing, 140 Commonwealth Avenue, Chestnut Hill, MA 02467, USA
| | - William R Woodward
- Department of Chemical Physiology and Biochemistry, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Portland, OR 97239, USA
| | - Christopher S Lee
- Boston College, William F. Connell School of Nursing, 140 Commonwealth Avenue, Chestnut Hill, MA 02467, USA
- Australian Catholic University, 115 Victoria Parade, Fitzroy, VIC 3065, Australia
| | - Quin E Denfeld
- Oregon Health & Science University, School of Nursing, 3455 SW U.S. Veteran’s Hospital Road, Portland, OR 97239, USA
- Oregon Health & Science University, Knight Cardiovascular Institute, 3181 SW Sam Jackson Park Road, Portland, OR 97239, USA
| |
Collapse
|
9
|
Durante A. Marathon not sprint: fatigue and early symptoms detected with sympathetic dysfunction in patients with heart failure. Eur J Cardiovasc Nurs 2024; 23:e75-e76. [PMID: 38227551 DOI: 10.1093/eurjcn/zvae003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Accepted: 01/02/2024] [Indexed: 01/18/2024]
Affiliation(s)
- Angela Durante
- Department of Translational Medicine, University of Eastern Piedmont Amedeo Avogadro, Via Solaroli 17, Palazzo Bellini, Room P89b, 28100 Novara, Italy
| |
Collapse
|
10
|
Peng R, Shi J, Jiang M, Qian D, Yan Y, Bai H, Yu M, Cao X, Fu S, Lu S. Electroacupuncture Improves Cardiac Function via Inhibiting Sympathetic Remodeling Mediated by Promoting Macrophage M2 Polarization in Myocardial Infarction Mice. Mediators Inflamm 2024; 2024:8237681. [PMID: 38974599 PMCID: PMC11227948 DOI: 10.1155/2024/8237681] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 03/24/2024] [Accepted: 06/07/2024] [Indexed: 07/09/2024] Open
Abstract
Electroacupuncture (EA) at the Neiguan acupoint (PC6) has shown significant cardioprotective effects. Sympathetic nerves play an important role in maintaining cardiac function after myocardial infarction (MI). Previous studies have found that EA treatment may improve cardiac function by modulating sympathetic remodeling after MI. However, the mechanism in how EA affects sympathetic remodeling and improves cardiac function remains unclear. The aim of this study is to investigate the cardioprotective mechanism of EA after myocardial ischemic injury by improving sympathetic remodeling and promoting macrophage M2 polarization. We established a mouse model of MI by occluding coronary arteries in male C57/BL6 mice. EA treatment was performed at the PC6 with current intensity (1 mA) and frequency (2/15 Hz). Cardiac function was evaluated using echocardiography. Heart rate variability in mice was assessed via standard electrocardiography. Myocardial fibrosis was evaluated by Sirius red staining. Levels of inflammatory factors were assessed using RT-qPCR. Sympathetic nerve remodeling was assessed through ELISA, western blotting, immunohistochemistry, and immunofluorescence staining. Macrophage polarization was evaluated using flow cytometry. Our results indicated that cardiac systolic function improved significantly after EA treatment, with an increase in fractional shortening and ejection fraction. Myocardial fibrosis was significantly mitigated in the EA group. The sympathetic nerve marker tyrosine hydroxylase and the nerve sprouting marker growth-associated Protein 43 were significantly reduced in the EA group, indicating that sympathetic remodeling was significantly reduced. EA treatment also promoted macrophage M2 polarization, reduced levels of inflammatory factors TNF-α, IL-1β, and IL-6, and decreased macrophage-associated nerve growth factor in myocardial tissue. To sum up, our results suggest that EA at PC6 attenuates sympathetic remodeling after MI to promote macrophage M2 polarization and improve cardiac function.
Collapse
Affiliation(s)
- Rou Peng
- Key Laboratory of Acupuncture and Medicine Research of Ministry of EducationNanjing University of Chinese Medicine, Nanjing 210023, China
| | - Junjing Shi
- The Second People's Hospital of Qidong, South Ring Road No. 229, Lvsigang Town, Qidong, Jiangsu Province 226200, China
| | - Minjiao Jiang
- Key Laboratory of Acupuncture and Medicine Research of Ministry of EducationNanjing University of Chinese Medicine, Nanjing 210023, China
| | - Danying Qian
- Key Laboratory of Acupuncture and Medicine Research of Ministry of EducationNanjing University of Chinese Medicine, Nanjing 210023, China
| | - Yuhang Yan
- Key Laboratory of Acupuncture and Medicine Research of Ministry of EducationNanjing University of Chinese Medicine, Nanjing 210023, China
| | - Hua Bai
- Key Laboratory of Acupuncture and Medicine Research of Ministry of EducationNanjing University of Chinese Medicine, Nanjing 210023, China
| | - Meiling Yu
- Key Laboratory of Acupuncture and Medicine Research of Ministry of EducationNanjing University of Chinese Medicine, Nanjing 210023, China
| | - Xin Cao
- Acupuncture and Chronobiology Key Laboratory of Sichuan ProvinceAcupuncture and Tuina School/Third Teaching HospitalChengdu University of Traditional Chinese Medicine, Chengdu 610075, China
| | - Shuping Fu
- Key Laboratory of Acupuncture and Medicine Research of Ministry of EducationNanjing University of Chinese Medicine, Nanjing 210023, China
| | - Shengfeng Lu
- Key Laboratory of Acupuncture and Medicine Research of Ministry of EducationNanjing University of Chinese Medicine, Nanjing 210023, China
- School of Elderly Care Services and ManagementNanjing University of Chinese Medicine, Nanjing 210023, China
| |
Collapse
|
11
|
Yatsu S, Kasai T, Naito R, Matsumoto H, Murata A, Shitara J, Shiroshita N, Kato M, Kawana F, Sato A, Ishiwata S, Shimizu M, Kato T, Suda S, Hiki M, Minamino T. Impact of sleep-disordered breathing on overnight changes in arterial stiffness in patients with acute heart failure. Hypertens Res 2024; 47:342-351. [PMID: 37783770 DOI: 10.1038/s41440-023-01448-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 08/25/2023] [Accepted: 08/31/2023] [Indexed: 10/04/2023]
Abstract
Overnight increases in arterial stiffness associated with sleep-disordered breathing may adversely affect patients with acute heart failure. Thus, we investigated overnight changes in arterial stiffness and their association with sleep-disordered breathing in patients hospitalized for acute heart failure. Consecutive patients with acute heart failure were enrolled. All participants underwent overnight full polysomnography following the initial improvement of acute signs and symptoms of acute heart failure. The arterial stiffness parameter, cardio-ankle vascular index (CAVI), was assessed before and after polysomnography. Overall, 60 patients (86.7% men) were analyzed. CAVI significantly increased overnight (from 8.4 ± 1.6 at night to 9.1 ± 1.7 in the morning, P < 0.001) in addition to systolic and diastolic blood pressure (from 114.1 mmHg to 121.6 mmHg, P < 0.001; and from 70.1 mmHg to 78.2 mmHg, P < 0.001, respectively). Overnight increase in CAVI (ΔCAVI ≥ 0) was observed in 42 patients (70%). The ΔCAVI ≥ 0 group was likely to have moderate-to-severe sleep-disordered breathing (i.e., apnea-hypopnea index ≥15, 55.6% vs 80.9%, P = 0.047) and greater obstructive respiratory events (29.4% vs 58.5%, P = 0.041). In multivariable analysis, moderate-to-severe sleep-disordered breathing and greater obstructive respiratory events were independently correlated with an overnight increase in CAVI (P = 0.033 and P = 0.042, respectively). In patients hospitalized for acute heart failure, arterial stiffness, as assessed by CAVI, significantly increased overnight. Moderate-to-severe sleep-disordered breathing and obstructive respiratory events may play an important role in the overnight increase in cardio-ankle vascular index.
Collapse
Affiliation(s)
- Shoichiro Yatsu
- Department of Cardiovascular Biology and Medicine, Juntendo University School of Medicine, Tokyo, Japan
- Department of Cardiovascular Medicine, Juntendo University Shizuoka Hospital, Shizuoka, Japan
| | - Takatoshi Kasai
- Department of Cardiovascular Biology and Medicine, Juntendo University School of Medicine, Tokyo, Japan.
- Cardiovascular Respiratory Sleep Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan.
- Department of Cardiovascular Management and Remote Monitoring, Juntendo University Graduate School of Medicine, Tokyo, Japan.
- Sleep and Sleep-Disordered Breathing Center, Juntendo University Hospital, Tokyo, Japan.
| | - Ryo Naito
- Department of Cardiovascular Biology and Medicine, Juntendo University School of Medicine, Tokyo, Japan
- Cardiovascular Respiratory Sleep Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Hiroki Matsumoto
- Department of Cardiovascular Biology and Medicine, Juntendo University School of Medicine, Tokyo, Japan
| | - Azusa Murata
- Department of Cardiovascular Biology and Medicine, Juntendo University School of Medicine, Tokyo, Japan
| | - Jun Shitara
- Department of Cardiovascular Biology and Medicine, Juntendo University School of Medicine, Tokyo, Japan
- Department of Cardiovascular Medicine, Juntendo University Shizuoka Hospital, Shizuoka, Japan
| | - Nanako Shiroshita
- Department of Cardiovascular Management and Remote Monitoring, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Mitsue Kato
- Cardiovascular Respiratory Sleep Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Fusae Kawana
- Cardiovascular Respiratory Sleep Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Akihiro Sato
- Department of Cardiovascular Biology and Medicine, Juntendo University School of Medicine, Tokyo, Japan
| | - Sayaki Ishiwata
- Department of Cardiovascular Biology and Medicine, Juntendo University School of Medicine, Tokyo, Japan
| | - Megumi Shimizu
- Department of Cardiovascular Biology and Medicine, Juntendo University School of Medicine, Tokyo, Japan
| | - Takao Kato
- Department of Cardiovascular Biology and Medicine, Juntendo University School of Medicine, Tokyo, Japan
| | - Shoko Suda
- Department of Cardiovascular Biology and Medicine, Juntendo University School of Medicine, Tokyo, Japan
| | - Masaru Hiki
- Department of Cardiovascular Biology and Medicine, Juntendo University School of Medicine, Tokyo, Japan
| | - Tohru Minamino
- Department of Cardiovascular Biology and Medicine, Juntendo University School of Medicine, Tokyo, Japan
- Japan Agency for Medical Research and Development-Core Research for Evolutionary Medical Science and Technology (AMED-CREST), Japan Agency for Medical Research and Development, Tokyo, Japan
| |
Collapse
|
12
|
Zhang S, Zhao D, Yang Z, Wang F, Yang S, Wang C. Circulating mitochondria promoted endothelial cGAS-derived neuroinflammation in subfornical organ to aggravate sympathetic overdrive in heart failure mice. J Neuroinflammation 2024; 21:27. [PMID: 38243316 PMCID: PMC10799549 DOI: 10.1186/s12974-024-03013-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Accepted: 01/03/2024] [Indexed: 01/21/2024] Open
Abstract
BACKGROUND Sympathoexcitation contributes to myocardial remodeling in heart failure (HF). Increased circulating pro-inflammatory mediators directly act on the Subfornical organ (SFO), the cardiovascular autonomic center, to increase sympathetic outflow. Circulating mitochondria (C-Mito) are the novel discovered mediators for inter-organ communication. Cyclic GMP-AMP synthase (cGAS) is the pro-inflammatory sensor of damaged mitochondria. OBJECTIVES This study aimed to assess the sympathoexcitation effect of C-Mito in HF mice via promoting endothelial cGAS-derived neuroinflammation in the SFO. METHODS C-Mito were isolated from HF mice established by isoprenaline (0.0125 mg/kg) infusion via osmotic mini-pumps for 2 weeks. Structural and functional analyses of C-Mito were conducted. Pre-stained C-Mito were intravenously injected every day for 2 weeks. Specific cGAS knockdown (cGAS KD) in the SFO endothelial cells (ECs) was achieved via the administration of AAV9-TIE-shRNA (cGAS) into the SFO. The activation of cGAS in the SFO ECs was assessed. The expression of the mitochondrial redox regulator Dihydroorotate dehydrogenase (DHODH) and its interaction with cGAS were also explored. Neuroinflammation and neuronal activation in the SFO were evaluated. Sympathetic activity, myocardial remodeling, and cardiac systolic dysfunction were measured. RESULTS C-Mito were successfully isolated, which showed typical structural characteristics of mitochondria with double-membrane and inner crista. Further analysis showed impaired respiratory complexes activities of C-Mito from HF mice (C-MitoHF) accompanied by oxidative damage. C-Mito entered ECs, instead of glial cells and neurons in the SFO of HF mice. C-MitoHF increased the level of ROS and cytosolic free double-strand DNA (dsDNA), and activated cGAS in cultured brain endothelial cells. Furthermore, C-MitoHF highly expressed DHODH, which interacted with cGAS to facilitate endothelial cGAS activation. C-MitoHF aggravated endothelial inflammation, microglial/astroglial activation, and neuronal sensitization in the SFO of HF mice, which could be ameliorated by cGAS KD in the ECs of the SFO. Further analysis showed C-MitoHF failed to exacerbate sympathoexcitation and myocardial sympathetic hyperinnervation in cGAS KD HF mice. C-MitoHF promoted myocardial fibrosis and hypertrophy, and cardiac systolic dysfunction in HF mice, which could be ameliorated by cGAS KD. CONCLUSION Collectively, we demonstrated that damaged C-MitoHF highly expressed DHODH, which promoted endothelial cGAS activation in the SFO, hence aggravating the sympathoexcitation and myocardial injury in HF mice, suggesting that C-Mito might be the novel therapeutic target for sympathoexcitation in HF.
Collapse
Affiliation(s)
- Shutian Zhang
- Department of Cardiac Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
- Shanghai Institute of Cardiovascular Diseases, Shanghai, 200032, China
| | - Dajun Zhao
- Department of Cardiac Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
- Shanghai Institute of Cardiovascular Diseases, Shanghai, 200032, China
| | - Zhaohua Yang
- Department of Cardiac Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
- Shanghai Institute of Cardiovascular Diseases, Shanghai, 200032, China
| | - Fanshun Wang
- Department of Cardiac Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
- Shanghai Institute of Cardiovascular Diseases, Shanghai, 200032, China
| | - Shouguo Yang
- Department of Cardiac Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, China.
- Shanghai Institute of Cardiovascular Diseases, Shanghai, 200032, China.
| | - Chunsheng Wang
- Department of Cardiac Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, China.
- Shanghai Institute of Cardiovascular Diseases, Shanghai, 200032, China.
| |
Collapse
|
13
|
Takeshita Y, Nomura C, Murai H, Mukai Y, Hirai T, Hamaoka T, Tokuno S, Tanaka T, Goto H, Nakano Y, Usui S, Nakajima K, Takamura M, Takamura T. Study Protocol for the Pleiotropic Effects of Sodium-Glucose Cotransporter 2 Inhibitor on Organ-Specific Sympathetic Nerve Activity and Insulin Sensitivity in Participants with Type 2 Diabetes. Diabetes Ther 2024; 15:269-280. [PMID: 37883004 PMCID: PMC10786788 DOI: 10.1007/s13300-023-01497-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Accepted: 10/12/2023] [Indexed: 10/27/2023] Open
Abstract
INTRODUCTION Hyperinsulinemia and hyperglycemia are associated with exaggerated systemic sympathetic nerve activity (SNA) in patients with type 2 diabetes. Sodium-glucose cotransporter 2 (SGLT2) inhibitors lower insulin levels, whereas sulfonylureas increase insulin levels. We will test whether these two classes of antidiabetic agents have different effects on SNA. METHODS The present study is an ongoing, 24-week, one-center (only Kanazawa University Hospital), open-label, randomized, parallel trial (jRCTs 041200035). Participants with type 2 diabetes with multiple atherosclerosis risk factors are randomly assigned in a 1:1 manner to receive 2.5 mg luseogliflozin or 0.5 mg glimepiride once daily. The sample size was calculated to be 14 in each group, with a significance level of 0.05 and a power of 0.80. The design required 40 evaluable study participants. Our primary endpoint will be the change in muscle SNA (MSNA). The secondary endpoints included organ-specific insulin sensitivity measured by a hyperinsulinemic-euglycemic clamp study using an artificial pancreas combined with a stable isotope-labeled glucose infusion, bioelectrical impedance analysis, and organ-specific (cardiac, renal, and hepatic) 123I-meta-iodobenzylguanidine (MIBG) innervation imaging. PLANNED OUTCOMES Study recruitment started in April 2020 and will end in June 2024, with 40 participants randomized into the two groups. The treatment follow-up of the participants is currently ongoing and is due to finish by March 2025. TRIAL REGISTRATION The study protocol has been approved by the Certified Review Board, Kanazawa University, Ishikawa, Japan, in accordance with the guidelines stipulated in the Declaration of Helsinki (CRB4180005, 2019-001). This trial is registered with the Japan Registry of Clinical Trials, jRCTs 041200035.
Collapse
Affiliation(s)
- Yumie Takeshita
- Department of Endocrinology and Metabolism, Kanazawa University Graduate School of Medical Sciences, 13-1 Takara-machi, Kanazawa, Ishikawa, 920-8640, Japan
| | - Chiaki Nomura
- Department of Endocrinology and Metabolism, Kanazawa University Graduate School of Medical Sciences, 13-1 Takara-machi, Kanazawa, Ishikawa, 920-8640, Japan
| | - Hisayoshi Murai
- Department of Cardiovascular Medicine, Kanazawa University Graduate School of Medical Sciences, 13-1 Takara-machi, Kanazawa, Ishikawa, 920-8640, Japan
| | - Yusuke Mukai
- Department of Cardiovascular Medicine, Kanazawa University Graduate School of Medical Sciences, 13-1 Takara-machi, Kanazawa, Ishikawa, 920-8640, Japan
| | - Tadayuki Hirai
- Department of Cardiovascular Medicine, Kanazawa University Graduate School of Medical Sciences, 13-1 Takara-machi, Kanazawa, Ishikawa, 920-8640, Japan
| | - Takuto Hamaoka
- Department of Cardiovascular Medicine, Kanazawa University Graduate School of Medical Sciences, 13-1 Takara-machi, Kanazawa, Ishikawa, 920-8640, Japan
| | - Shota Tokuno
- Department of Endocrinology and Metabolism, Kanazawa University Graduate School of Medical Sciences, 13-1 Takara-machi, Kanazawa, Ishikawa, 920-8640, Japan
| | - Takeo Tanaka
- Department of Endocrinology and Metabolism, Kanazawa University Graduate School of Medical Sciences, 13-1 Takara-machi, Kanazawa, Ishikawa, 920-8640, Japan
| | - Hisanori Goto
- Department of Endocrinology and Metabolism, Kanazawa University Graduate School of Medical Sciences, 13-1 Takara-machi, Kanazawa, Ishikawa, 920-8640, Japan
| | - Yujiro Nakano
- Department of Endocrinology and Metabolism, Kanazawa University Graduate School of Medical Sciences, 13-1 Takara-machi, Kanazawa, Ishikawa, 920-8640, Japan
| | - Soichiro Usui
- Department of Cardiovascular Medicine, Kanazawa University Graduate School of Medical Sciences, 13-1 Takara-machi, Kanazawa, Ishikawa, 920-8640, Japan
| | - Kenichi Nakajima
- Department of Functional Imaging and Artificial Intelligence, Kanazawa University, 13-1 Takara-machi, Kanazawa, Ishikawa, 920-8640, Japan
| | - Masayuki Takamura
- Department of Cardiovascular Medicine, Kanazawa University Graduate School of Medical Sciences, 13-1 Takara-machi, Kanazawa, Ishikawa, 920-8640, Japan
| | - Toshinari Takamura
- Department of Endocrinology and Metabolism, Kanazawa University Graduate School of Medical Sciences, 13-1 Takara-machi, Kanazawa, Ishikawa, 920-8640, Japan.
- Department of Comprehensive Metabology, Kanazawa University Graduate School of Medical Sciences, 13-1 Takara-machi, Kanazawa, Ishikawa, 920-8640, Japan.
| |
Collapse
|
14
|
Yang L, Yin L, Hu M, Zhao W, Wang C, Chen Y, Li Z, Wang L. Preliminary Evaluation of 18F-Labeled Benzylguanidine Analogs as NET Tracers for Myocardial Infarction Diagnosis. Mol Imaging Biol 2023; 25:1125-1134. [PMID: 37580463 DOI: 10.1007/s11307-023-01844-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 07/27/2023] [Accepted: 07/28/2023] [Indexed: 08/16/2023]
Abstract
PURPOSE Heart failure (HF) remains a major cause of late morbidity and mortality after myocardial infarction (MI). To date, no clinically established 18F-labeled sympathetic nerve PET tracers for monitoring myocardial infarction are available. Therefore, in this study, we synthesized a series of 18F-labeled benzyl guanidine analogs and evaluated their efficacy as cardiac neuronal norepinephrine transporter (NET) tracers for myocardial imaging. We also investigated the preliminary diagnostic capabilities of these tracers in myocardial infarction animal models, as well as the structure-activity relationship of these tracers. PROCEDURES Three benzyl guanidine-NET tracers, including [18F]1, [18F]2, and [18F]3, were synthesized and evaluated in vivo as PET tracers in a myocardial infarction mouse model. [18F]LMI1195 was used as a positive control for the tracers. H&E staining of the isolated myocardial infarction heart tissue sections was performed to verify the efficacy of the selected PET tracer. RESULTS Our data show that [18F]3 had a moderate decay corrected labeling yield (~10%) and high radiochemical purity (>95%) compared to other tracers. The uptake of [18F]3 in normal mouse hearts was 1.7±0.1%ID/cc at 1 h post-injection (p. i.), while it was 2.4±0.1, 2.6±0.9, and 2.1±0.4%ID/cc in the MI mouse hearts at 1, 2, and 3 days after surgery, respectively. Compared with [18F]LMI1195, [18F]3 had a better myocardial imaging effect in terms of the contrast between normal and MI hearts. The area of myocardial infarction shown by PET imaging corresponded well with the infarcted tissue demonstrated by H&E staining. CONCLUSIONS With an obvious cardiac uptake contrast between normal mice and the myocardial infarction mouse model, [18F]3 appears to be a potential tool in the diagnosis of myocardial infarction. Therefore, it is necessary to conduct further structural modification studies on the chemical structure of [18F]3 to improve its in vivo stability and diagnostic detection ability to achieve reliable and practical imaging effects.
Collapse
Affiliation(s)
- Liping Yang
- Department of Nuclear Medicine, Nuclear Medicine and Molecular Imaging Key Laboratory of Sichuan Province, The Affiliated Hospital of Southwest Medical University, No. 25 Taiping St, Jiangyang District, Luzhou, Sichuan, China
- School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
- Academician (Expert) Workstation of Sichuan Province, Luzhou, Sichuan, China
| | - Liping Yin
- Department of Nuclear Medicine, Nuclear Medicine and Molecular Imaging Key Laboratory of Sichuan Province, The Affiliated Hospital of Southwest Medical University, No. 25 Taiping St, Jiangyang District, Luzhou, Sichuan, China
- School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
- Academician (Expert) Workstation of Sichuan Province, Luzhou, Sichuan, China
| | - Mei Hu
- Department of Nuclear Medicine, Nuclear Medicine and Molecular Imaging Key Laboratory of Sichuan Province, The Affiliated Hospital of Southwest Medical University, No. 25 Taiping St, Jiangyang District, Luzhou, Sichuan, China
- School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
- Academician (Expert) Workstation of Sichuan Province, Luzhou, Sichuan, China
| | - Weiling Zhao
- Department of Radiology, Lineberger Comprehensive Cancer Center, and Biomedical Research Imaging Center, University of North Carolina, Chapel Hill, NC, USA
| | - Changjiang Wang
- Department of Nuclear Medicine, Nuclear Medicine and Molecular Imaging Key Laboratory of Sichuan Province, The Affiliated Hospital of Southwest Medical University, No. 25 Taiping St, Jiangyang District, Luzhou, Sichuan, China
- School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
- Academician (Expert) Workstation of Sichuan Province, Luzhou, Sichuan, China
| | - Yue Chen
- Department of Nuclear Medicine, Nuclear Medicine and Molecular Imaging Key Laboratory of Sichuan Province, The Affiliated Hospital of Southwest Medical University, No. 25 Taiping St, Jiangyang District, Luzhou, Sichuan, China.
- Academician (Expert) Workstation of Sichuan Province, Luzhou, Sichuan, China.
| | - Zibo Li
- Department of Radiology, Lineberger Comprehensive Cancer Center, and Biomedical Research Imaging Center, University of North Carolina, Chapel Hill, NC, USA.
| | - Li Wang
- Department of Nuclear Medicine, Nuclear Medicine and Molecular Imaging Key Laboratory of Sichuan Province, The Affiliated Hospital of Southwest Medical University, No. 25 Taiping St, Jiangyang District, Luzhou, Sichuan, China.
- Academician (Expert) Workstation of Sichuan Province, Luzhou, Sichuan, China.
| |
Collapse
|
15
|
Manolis AA, Manolis TA, Manolis AS. Neurohumoral Activation in Heart Failure. Int J Mol Sci 2023; 24:15472. [PMID: 37895150 PMCID: PMC10607846 DOI: 10.3390/ijms242015472] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Revised: 10/16/2023] [Accepted: 10/21/2023] [Indexed: 10/29/2023] Open
Abstract
In patients with heart failure (HF), the neuroendocrine systems of the sympathetic nervous system (SNS), the renin-angiotensin-aldosterone system (RAAS) and the arginine vasopressin (AVP) system, are activated to various degrees producing often-observed tachycardia and concomitant increased systemic vascular resistance. Furthermore, sustained neurohormonal activation plays a key role in the progression of HF and may be responsible for the pathogenetic mechanisms leading to the perpetuation of the pathophysiology and worsening of the HF signs and symptoms. There are biomarkers of activation of these neurohormonal pathways, such as the natriuretic peptides, catecholamine levels and neprilysin and various newer ones, which may be employed to better understand the mechanisms of HF drugs and also aid in defining the subgroups of patients who might benefit from specific therapies, irrespective of the degree of left ventricular dysfunction. These therapies are directed against these neurohumoral systems (neurohumoral antagonists) and classically comprise beta blockers, angiotensin-converting enzyme (ACE) inhibitors/angiotensin receptor blockers and vaptans. Recently, the RAAS blockade has been refined by the introduction of the angiotensin receptor-neprilysin inhibitor (ARNI) sacubitril/valsartan, which combines the RAAS inhibition and neprilysin blocking, enhancing the actions of natriuretic peptides. All these issues relating to the neurohumoral activation in HF are herein reviewed, and the underlying mechanisms are pictorially illustrated.
Collapse
Affiliation(s)
- Antonis A. Manolis
- First Department of Cardiology, Evagelismos Hospital, 106 76 Athens, Greece;
| | - Theodora A. Manolis
- Department of Psychiatry, Aiginiteio University Hospital, 115 28 Athens, Greece;
| | - Antonis S. Manolis
- First Department of Cardiology, Ippokrateio University Hospital, 115 27 Athens, Greece
| |
Collapse
|
16
|
Odnoshivkina JG, Sibgatullina GV, Petrov AM. Lipid-dependent regulation of neurotransmitter release from sympathetic nerve endings in mice atria. BIOCHIMICA ET BIOPHYSICA ACTA. BIOMEMBRANES 2023; 1865:184197. [PMID: 37394027 DOI: 10.1016/j.bbamem.2023.184197] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 05/25/2023] [Accepted: 06/26/2023] [Indexed: 07/04/2023]
Abstract
Neurotransmitter release from sympathetic terminals is a key avenue for heart regulation. Herein, presynaptic exocytotic activity was monitored in mice atrial tissue using a false fluorescent neurotransmitter FFN511, a substrate for monoamine transporters. FFN511 labeling had similarity with tyrosine hydroxylase immunostaining. High [K+]o depolarization caused FFN511 release, which was augmented by reserpine, an inhibitor of neurotransmitter uptake. However, reserpine lost the ability to increase depolarization-induced FFN511 unloading after depletion of ready releasable pool with hyperosmotic sucrose. Cholesterol oxidase and sphingomyelinase modified atrial membranes, changing in opposite manner fluorescence of lipid ordering-sensitive probe. Plasmalemmal cholesterol oxidation increased FFN511 release upon K+-depolarization and more markedly potentiated FFN511 unloading in the presence of reserpine. Hydrolysis of plasmalemmal sphingomyelin profoundly enhanced the rate of FFN511 loss due to K+-depolarization, but completely prevented potentiating action of reserpine on FFN511 unloading. If cholesterol oxidase or sphingomyelinase got access to membranes of recycling synaptic vesicles, then the enzyme effects were suppressed. Hence, a fast neurotransmitter reuptake dependent on exocytosis of vesicles from ready releasable pool occurs during presynaptic activity. This reuptake can be enhanced or inhibited by plasmalemmal cholesterol oxidation or sphingomyelin hydrolysis, respectively. These modifications of plasmalemmal (but not vesicular) lipids increase the evoked neurotransmitter release.
Collapse
Affiliation(s)
- Julia G Odnoshivkina
- Laboratory of Biophysics of Synaptic Processes, Kazan Institute of Biochemistry and Biophysics, Federal Research Center "Kazan Scientific Center of RAS", 2/31 Lobachevsky Street, Box 30, Kazan 420111, Russia; Kazan State Medical University, 49 Butlerova Street, Kazan 420012, Russia
| | - Guzel V Sibgatullina
- Laboratory of Biophysics of Synaptic Processes, Kazan Institute of Biochemistry and Biophysics, Federal Research Center "Kazan Scientific Center of RAS", 2/31 Lobachevsky Street, Box 30, Kazan 420111, Russia
| | - Alexey M Petrov
- Laboratory of Biophysics of Synaptic Processes, Kazan Institute of Biochemistry and Biophysics, Federal Research Center "Kazan Scientific Center of RAS", 2/31 Lobachevsky Street, Box 30, Kazan 420111, Russia; Kazan Federal University, 18 Kremlyovskaya Street, Kazan 420008, Russia.
| |
Collapse
|
17
|
Tang H, Xu C, Zhang P, Luo T, Huang Y, Yang X. A profile of SGLT-2 inhibitors in hyponatremia: The evidence to date. Eur J Pharm Sci 2023; 184:106415. [PMID: 36870579 DOI: 10.1016/j.ejps.2023.106415] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2022] [Revised: 02/14/2023] [Accepted: 02/27/2023] [Indexed: 03/06/2023]
Abstract
Hyponatremia is the most common electrolyte disorder in clinical practice, which may lead to life-threatening complications. Several lines of evidence suggest that hyponatremia is associated not only with significant increases in length of stay, cost, and financial burden, but also with increased morbidity and mortality. Hyponatremia is also considered to be a negative prognostic factor in patients with heart failure and cancer. Although multiple therapeutic methods are available for treating hyponatremia, most have some limitations, such as poor compliance, rapid correction of serum Na+, other negative side effects and high cost. Given these limitations, identifying novel therapies for hyponatremia is essential. Recent clinical studies have shown that SGLT-2 inhibitors (SGLT 2i) significantly increased serum Na+ levels and were well tolerated by patients who underwent this treatment. Therefore, oral administration of SGLT 2i appears to be an effective treatment for hyponatremia. This article will briefly review the etiology of hyponatremia and integrated control of sodium within the kidney, current therapies for hyponatremia, potential mechanisms and efficacy of SGLT 2i for hyponatremia, and the benefits in cardiovascular, cancer, and kidney disease by regulating sodium and water balance.
Collapse
Affiliation(s)
- Hui Tang
- Department of Pharmacy, The Affiliated Hospital of Southwest Medical University, Luzhou 646000, China; School of Pharmacy, Southwest Medical University, Luzhou 646000, China
| | - Changjing Xu
- Department of Pharmacy, The Affiliated Hospital of Southwest Medical University, Luzhou 646000, China
| | - Piao Zhang
- Department of Pharmacy, Ya 'an People's Hospital, Ya 'an, Sichuan 646000, China
| | - Taimin Luo
- Department of pharmacy, Chengdu Seventh People's Hospital, Chengdu, Sichuan 610000, China
| | - Yilan Huang
- Department of Pharmacy, The Affiliated Hospital of Southwest Medical University, Luzhou 646000, China; School of Pharmacy, Southwest Medical University, Luzhou 646000, China.
| | - Xuping Yang
- Department of Pharmacy, The Affiliated Hospital of Southwest Medical University, Luzhou 646000, China; School of Pharmacy, Southwest Medical University, Luzhou 646000, China.
| |
Collapse
|
18
|
Persistent Hypochloremia Is Associated with Adverse Prognosis in Patients Repeatedly Hospitalized for Heart Failure. J Clin Med 2023; 12:jcm12041257. [PMID: 36835793 PMCID: PMC9962161 DOI: 10.3390/jcm12041257] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Revised: 01/30/2023] [Accepted: 02/03/2023] [Indexed: 02/09/2023] Open
Abstract
BACKGROUND Hypochloremia reflects neuro-hormonal activation in patients with heart failure (HF). However, the prognostic impact of persistent hypochloremia in those patients remains unclear. METHODS We collected the data of patients who were hospitalized for HF at least twice between 2010 and 2021 (n = 348). Dialysis patients (n = 26) were excluded. The patients were divided into four groups based on the absence/presence of hypochloremia (<98 mmol/L) at discharge from their first and second hospitalizations: Group A (patients without hypochloremia at their first and second hospitalizations, n = 243); Group B (those with hypochloremia at their first hospitalization and without hypochloremia at their second hospitalization, n = 29); Group C (those without hypochloremia at their first hospitalization and with hypochloremia at their second hospitalization, n = 34); and Group D (those with hypochloremia at their first and second hospitalizations, n = 16). RESULTS a Kaplan-Meier analysis revealed that all-cause mortality and cardiac mortality were the highest in Group D compared to the other groups. A multivariable Cox proportional hazard analysis revealed that persistent hypochloremia was independently associated with both all-cause death (hazard ratio 3.490, p < 0.001) and cardiac death (hazard ratio 3.919, p < 0.001). CONCLUSIONS In patients with HF, prolonged hypochloremia over two hospitalizations is associated with an adverse prognosis.
Collapse
|
19
|
Alieva AM, Teplova NV, Reznik EV, Ettinger OA, Faradzhov RA, Khachirova EA, Kovtiukh IV, Kotikova IA, Sysoeva DA, Bigushev IR, Nikitin IG. Catestanin – a promising biological marker for heart failure: A review. CONSILIUM MEDICUM 2022. [DOI: 10.26442/20751753.2022.10.201873] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
The epidemic of heart failure (HF) is one of the problems that the global health system has been facing for decades. HF is a multicomponent clinical syndrome caused by dysfunction of the heart and its pathological remodeling. In addition to the well-known natriuretic peptides, a number of cardiovascular biological markers have now been identified that provide clinicians with additional opportunities in diagnosing, classifying, predicting, and monitoring the effectiveness of treating patients with HF. From the position of establishing the sympathetic load in patients with HF, it seems very promising to assess the concentrations of catestatin. The presented data of our literature review suggest that catestatin is probably a reliable biological marker of the activity of the sympathetic division of the autonomic nervous system, and its elevated concentrations in patients with HF reflect the severity of the pathological process. However, despite the reliable results of studies, the clinical significance of assessing the values of this marker both separately and in the framework of a multimarker model requires further study in larger prospective clinical studies.
Collapse
|
20
|
Li YL. Stellate Ganglia and Cardiac Sympathetic Overactivation in Heart Failure. Int J Mol Sci 2022; 23:ijms232113311. [PMID: 36362099 PMCID: PMC9653702 DOI: 10.3390/ijms232113311] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2022] [Revised: 10/28/2022] [Accepted: 10/29/2022] [Indexed: 11/06/2022] Open
Abstract
Heart failure (HF) is a major public health problem worldwide, especially coronary heart disease (myocardial infarction)-induced HF with reduced ejection fraction (HFrEF), which accounts for over 50% of all HF cases. An estimated 6 million American adults have HF. As a major feature of HF, cardiac sympathetic overactivation triggers arrhythmias and sudden cardiac death, which accounts for nearly 50–60% of mortality in HF patients. Regulation of cardiac sympathetic activation is highly integrated by the regulatory circuitry at multiple levels, including afferent, central, and efferent components of the sympathetic nervous system. Much evidence, from other investigators and us, has confirmed the afferent and central neural mechanisms causing sympathoexcitation in HF. The stellate ganglion is a peripheral sympathetic ganglion formed by the fusion of the 7th cervical and 1st thoracic sympathetic ganglion. As the efferent component of the sympathetic nervous system, cardiac postganglionic sympathetic neurons located in stellate ganglia provide local neural coordination independent of higher brain centers. Structural and functional impairments of cardiac postganglionic sympathetic neurons can be involved in cardiac sympathetic overactivation in HF because normally, many effects of the cardiac sympathetic nervous system on cardiac function are mediated via neurotransmitters (e.g., norepinephrine) released from cardiac postganglionic sympathetic neurons innervating the heart. This review provides an overview of cardiac sympathetic remodeling in stellate ganglia and potential mechanisms and the role of cardiac sympathetic remodeling in cardiac sympathetic overactivation and arrhythmias in HF. Targeting cardiac sympathetic remodeling in stellate ganglia could be a therapeutic strategy against malignant cardiac arrhythmias in HF.
Collapse
Affiliation(s)
- Yu-Long Li
- Department of Emergency Medicine, University of Nebraska Medical Center, Omaha, NE 68198, USA; ; Tel.: +1-402-559-3016; Fax: +1-402-559-9659
- Department of Cellular & Integrative Physiology, University of Nebraska Medical Center, Omaha, NE 68198, USA
| |
Collapse
|