51
|
Malik S, Ali ZS, Al-Rawi R, Lavercombe W, Gupta S, Zhou Z, Farina JM, Marcotte L, Baranchuk A. Emotions & Heart:Exploring the Impact of Negative Emotions on Cardiovascular Health. Curr Probl Cardiol 2025; 50:102989. [PMID: 39848354 DOI: 10.1016/j.cpcardiol.2025.102989] [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: 01/11/2025] [Accepted: 01/16/2025] [Indexed: 01/25/2025]
Abstract
Negative emotions can have a significant impact on individuals, which then influences their cardiovascular system. However, the underlying pathophysiological mechanisms and clinical implications of this association remain inadequately defined. A narrative review of pertinent literature was conducted to examine the pathophysiology, clinical manifestations, and treatment related to the interplay between emotions and conditions such as takotsubo cardiomyopathy, atherosclerosis, acute plaque rupture, and cardiac arrhythmias. Negative emotions can instigate a chronic stress response, which in turn heightens sympathetic nervous system activity and increases vulnerability to cardiovascular diseases. This intricate relationship between emotional states and cardiovascular health underscores the necessity for targeted lifestyle interventions and clinical strategies aimed at mitigating the adverse effects of negative emotions.
Collapse
Affiliation(s)
- Shaun Malik
- Faculty of Health Sciences, School of Medicine, Queen's University, Kingston, Ontario, Canada
| | - Zain S Ali
- Faculty of Health Sciences, School of Medicine, Queen's University, Kingston, Ontario, Canada; Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada
| | - Reem Al-Rawi
- Faculty of Health Sciences, School of Medicine, Queen's University, Kingston, Ontario, Canada
| | | | - Shyla Gupta
- Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Zier Zhou
- Faculty of Health Sciences, School of Medicine, Queen's University, Kingston, Ontario, Canada
| | - Juan M Farina
- Department of Cardiovascular and Thoracic Surgery, Mayo Clinic, Phoenix, AZ, USA
| | - Laura Marcotte
- Faculty of Health Sciences, School of Medicine, Queen's University, Kingston, Ontario, Canada; Division of General Internal Medicine, Queen's University, Kingston, Ontario, Canada
| | - Adrian Baranchuk
- Faculty of Health Sciences, School of Medicine, Queen's University, Kingston, Ontario, Canada; Division of Cardiology, Queen's University, Kingston, Ontario, Canada.
| |
Collapse
|
52
|
Kaplan A, El‐Samadi L, Zahreddine R, Amin G, Booz GW, Zouein FA. Canonical or non-canonical, all aspects of G protein-coupled receptor kinase 2 in heart failure. Acta Physiol (Oxf) 2025; 241:e70010. [PMID: 39960030 PMCID: PMC11831727 DOI: 10.1111/apha.70010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 01/12/2025] [Accepted: 01/20/2025] [Indexed: 02/20/2025]
Abstract
G protein-coupled receptor kinase 2 (GRK2) with its multidomain structure performs various crucial cellular functions under both normal and pathological conditions. Overexpression of GRK2 is linked to cardiovascular diseases, and its inhibition or deletion has been shown to be protective. The functions of GRK2 extend beyond G protein-coupled receptor (GPCR) signaling, influencing non-GPCR substrates as well. Increased GRK2 in heart failure (HF) initially may be protective but ultimately leads to maladaptive effects such as GPCR desensitization, insulin resistance, and apoptosis. The multifunctional nature of GRK2, including its action in hypertrophic gene expression, insulin signaling, and cardiac fibrosis, highlights its complex role in HF pathogenesis. Additionally, GRK2 is involved in mitochondrial biogenesis and lipid metabolism. GRK2 also regulates epinephrine secretion from the adrenal gland and its increase in circulating lymphocytes can be used to monitor HF status. Overall, GRK2 is a multifaceted protein with significant implications for HF and the regulation of GRK2 is crucial for understanding and treating cardiovascular diseases.
Collapse
Affiliation(s)
- Abdullah Kaplan
- Department of Pharmacology and ToxicologyAmerican University of Beirut Faculty of MedicineBeirutLebanon
- The Cardiovascular, Renal, and Metabolic Diseases Research Center of ExcellenceAmerican University of Beirut Medical CenterBeirutLebanon
- Cardiology ClinicKemer Public HospitalAntalyaTurkey
| | - Lana El‐Samadi
- Department of Pharmacology and ToxicologyAmerican University of Beirut Faculty of MedicineBeirutLebanon
- The Cardiovascular, Renal, and Metabolic Diseases Research Center of ExcellenceAmerican University of Beirut Medical CenterBeirutLebanon
| | - Rana Zahreddine
- Department of Pharmacology and ToxicologyAmerican University of Beirut Faculty of MedicineBeirutLebanon
- The Cardiovascular, Renal, and Metabolic Diseases Research Center of ExcellenceAmerican University of Beirut Medical CenterBeirutLebanon
| | - Ghadir Amin
- Department of Pharmacology and ToxicologyAmerican University of Beirut Faculty of MedicineBeirutLebanon
- The Cardiovascular, Renal, and Metabolic Diseases Research Center of ExcellenceAmerican University of Beirut Medical CenterBeirutLebanon
- Department of Pharmacology and Toxicology, School of MedicineUniversity of Mississippi Medical CenterJacksonMississippiUSA
| | - George W. Booz
- Department of Pharmacology and Toxicology, School of MedicineUniversity of Mississippi Medical CenterJacksonMississippiUSA
| | - Fouad A. Zouein
- Department of Pharmacology and ToxicologyAmerican University of Beirut Faculty of MedicineBeirutLebanon
- The Cardiovascular, Renal, and Metabolic Diseases Research Center of ExcellenceAmerican University of Beirut Medical CenterBeirutLebanon
- Department of Pharmacology and Toxicology, School of MedicineUniversity of Mississippi Medical CenterJacksonMississippiUSA
| |
Collapse
|
53
|
Bussmann B, Ayagama T, Liu K, Li D, Herring N. Bayliss Starling Prize Lecture 2023: Neuropeptide-Y being 'unsympathetic' to the broken hearted. J Physiol 2025; 603:1841-1864. [PMID: 38847435 PMCID: PMC11955873 DOI: 10.1113/jp285370] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Accepted: 05/01/2024] [Indexed: 04/01/2025] Open
Abstract
William Bayliss and Ernest Starling are not only famous as pioneers in cardiovascular physiology, but also responsible for the discovery of the first hormone (from the Greek 'excite or arouse'), the intestinal signalling molecule and neuropeptide secretin in 1902. Our research group focuses on neuropeptides and neuromodulators that influence cardiovascular autonomic control as potential biomarkers in disease and tractable targets for therapeutic intervention. Acute myocardial infarction (AMI) and chronic heart failure (CHF) result in high levels of cardiac sympathetic stimulation, which is a poor prognostic indicator. Although beta-blockers improve mortality in these conditions by preventing the action of the neurotransmitter noradrenaline, a substantial residual risk remains. Recently, we have identified the sympathetic co-transmitter neuropeptide-Y (NPY) as being released during AMI, leading to larger infarcts and life-threatening arrhythmia in both animal models and patients. Here, we discuss recently published data demonstrating that peripheral venous NPY levels are associated with heart failure hospitalisation and mortality after AMI, and all cause cardiovascular mortality in CHF, even when adjusting for known risk factors (including brain natriuretic peptide). We have investigated the mechanistic basis for these observations in human and rat stellate ganglia and cardiac tissue, manipulating NPY neurochemistry at the same time as using state-of-the-art imaging techniques, to establish the receptor pathways responsible for NPY signalling. We propose NPY as a new mechanistic biomarker in AMI and CHF patients and aim to determine whether specific NPY receptor blockers can prevent arrhythmia and attenuate the development of heart failure.
Collapse
Affiliation(s)
- Benjamin Bussmann
- Burdon Sanderson Cardiac Science Centre, Department of Physiology, Anatomy and GeneticsUniversity of OxfordOxfordUK
| | - Thamali Ayagama
- Burdon Sanderson Cardiac Science Centre, Department of Physiology, Anatomy and GeneticsUniversity of OxfordOxfordUK
| | - Kun Liu
- Burdon Sanderson Cardiac Science Centre, Department of Physiology, Anatomy and GeneticsUniversity of OxfordOxfordUK
| | - Dan Li
- Burdon Sanderson Cardiac Science Centre, Department of Physiology, Anatomy and GeneticsUniversity of OxfordOxfordUK
| | - Neil Herring
- Burdon Sanderson Cardiac Science Centre, Department of Physiology, Anatomy and GeneticsUniversity of OxfordOxfordUK
| |
Collapse
|
54
|
Bastos JM, Colaço B, Baptista R, Gavina C, Vitorino R. Innovations in heart failure management: The role of cutting-edge biomarkers and multi-omics integration. JOURNAL OF MOLECULAR AND CELLULAR CARDIOLOGY PLUS 2025; 11:100290. [PMID: 40129519 PMCID: PMC11930597 DOI: 10.1016/j.jmccpl.2025.100290] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/30/2024] [Revised: 02/11/2025] [Accepted: 02/27/2025] [Indexed: 03/26/2025]
Abstract
Heart failure (HF) remains a major cause of morbidity and mortality worldwide and represents a major challenge for diagnosis, prognosis and treatment due to its heterogeneity. Traditional biomarkers such as BNP and NT-proBNP are valuable but insufficient to capture the complexity of HF, especially phenotypes such as HF with preserved ejection fraction (HFpEF). Recent advances in multi-omics technology and novel biomarkers such as cell-free DNA (cfDNA), microRNAs (miRNAs), ST2 and galectin-3 offer transformative potential for HF management. This review explores the integration of these innovative biomarkers into clinical practice and highlights their benefits, such as improved diagnostic accuracy, enhanced risk stratification and non-invasive monitoring capabilities. By leveraging multi-omics approaches, including lipidomics and metabolomics, clinicians can uncover new pathways, refine the classification of HF phenotypes, and develop personalized therapeutic strategies tailored to individual patient profiles. Remarkable advances in proteomics and metabolomics have identified biomarkers associated with key HF mechanisms such as mitochondrial dysfunction, inflammation and fibrosis, paving the way for targeted therapies and early interventions. Despite the promising results, significant challenges remain in translating these findings into routine care, including high costs, technical limitations and the need for large-scale validation studies. This report argues for an integrative, multi-omics-based model to overcome these obstacles and emphasizes the importance of collaboration between researchers, clinicians and policy makers. By linking innovative science with practical applications, multi-omics approaches have the potential to redefine HF management and lead to better patient outcomes and more sustainable healthcare systems.
Collapse
Affiliation(s)
- Jose Mesquita Bastos
- Department of Medical Sciences, Institute of Biomedicine iBiMED, University of Aveiro, 3810-193 Aveiro, Portugal
- Cardiology Department, Hospital Infante D. Pedro, Centro Hospitalar do Baixo Vouga, Aveiro, Portugal
| | - Beatriz Colaço
- Department of Medical Sciences, Institute of Biomedicine iBiMED, University of Aveiro, 3810-193 Aveiro, Portugal
- Cardiology Department, Hospital Infante D. Pedro, Centro Hospitalar do Baixo Vouga, Aveiro, Portugal
| | - Rui Baptista
- Department of Cardiology, Centro Hospitalar de Entre o Douro e Vouga, Santa Maria da Feira, Portugal
- Faculty of Medicine, University of Coimbra, Coimbra, Portugal
- University of Coimbra, Center for Innovative Biomedicine and Biotechnology (CIBB), Coimbra, Portugal
- Clinical Academic Center of Coimbra (CACC), Coimbra, Portugal
| | - Cristina Gavina
- Pedro Hispano Hospital - ULS Matosinhos, Matosinhos, Portugal
- Cardiology Department, Faculty of Medicine, University of Porto, Oporto, Portugal
- RISE- Health Research Network, Faculty of Medicine, University of Porto, Oporto, Portugal
| | - Rui Vitorino
- Department of Medical Sciences, Institute of Biomedicine iBiMED, University of Aveiro, 3810-193 Aveiro, Portugal
- Cardiovascular R&D Centre - UnIC@RISE, Department of Surgery and Physiology, Faculty of Medicine of the University of Porto, 4200-319 Porto, Portugal
| |
Collapse
|
55
|
Newman AAC, Dalman JM, Moore KJ. Cardiovascular Disease and Cancer: A Dangerous Liaison. Arterioscler Thromb Vasc Biol 2025; 45:359-371. [PMID: 39781742 PMCID: PMC11864891 DOI: 10.1161/atvbaha.124.319863] [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: 11/08/2024] [Revised: 12/10/2024] [Accepted: 12/20/2024] [Indexed: 01/12/2025]
Abstract
The field of cardio-oncology has traditionally focused on the impact of cancer and its therapies on cardiovascular health. Mounting clinical and preclinical evidence, however, indicates that the reverse may also be true: cardiovascular disease can itself influence tumor growth and metastasis. Numerous epidemiological studies have reported that individuals with prevalent cardiovascular disease have an increased incidence of cancer. In parallel, studies using preclinical mouse models of myocardial infarction, heart failure, and cardiac remodeling support the notion that cardiovascular disorders accelerate the growth of solid tumors and metastases. These findings have ushered in a new and burgeoning field termed reverse cardio-oncology that investigates the impact of cardiovascular disease pathophysiology on cancer emergence and progression. Recent studies have begun to illuminate the mechanisms driving this relationship, including shared risk factors, reprogramming of immune responses, changes in gene expression, and the release of cardiac factors that result in selective advantages for tumor cells or their local milieu, thus exacerbating cancer pathology. Here, we review the evidence supporting the relationship between cardiovascular disease and cancer, the mechanistic pathways enabling this connection, and the implications of these findings for patient care.
Collapse
Affiliation(s)
- Alexandra A C Newman
- Cardiovascular Research Center, New York University Langone Health, New York, NY 10016, USA
- Leon H. Charney Division of Cardiology, Department of Medicine, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Jessie M Dalman
- Cardiovascular Research Center, New York University Langone Health, New York, NY 10016, USA
- Leon H. Charney Division of Cardiology, Department of Medicine, New York University Grossman School of Medicine, New York, NY 10016, USA
- University of Michigan Medical School, Ann Arbor, MI 48104, USA
| | - Kathryn J Moore
- Cardiovascular Research Center, New York University Langone Health, New York, NY 10016, USA
- Leon H. Charney Division of Cardiology, Department of Medicine, New York University Grossman School of Medicine, New York, NY 10016, USA
- Department of Cell Biology, New York University Grossman School of Medicine, New York, NY 10016, USA
| |
Collapse
|
56
|
Kharrat A, Nissimov S, Zhu F, Deshpande P, Jain A. Cardiopulmonary Physiology of Hypoxemic Respiratory Failure Among Preterm Infants with Septic Shock. J Pediatr 2025; 278:114384. [PMID: 39510164 DOI: 10.1016/j.jpeds.2024.114384] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Revised: 10/28/2024] [Accepted: 10/28/2024] [Indexed: 11/15/2024]
Abstract
OBJECTIVE To examine cardiopulmonary physiological alterations associated with hypoxemic respiratory failure (HRF; fraction of inspired oxygen ≥0.60) among preterm neonates requiring vasopressors/inotropes during sepsis (septic shock). STUDY DESIGN We conducted a retrospective cohort study from 2015 through 2022 at a tertiary neonatal intensive care unit. Neonates <34 weeks gestational age who had septic shock and underwent a comprehensive targeted neonatal echocardiography (TNE) ≤72 hours of sepsis onset were included. TNE findings of patients with shock and HRF were compared with those with shock without HRF. Indices of pulmonary vascular resistance (PVR), right ventricular (RV) and left ventricular (LV) systolic and diastolic function, measured using conventional, tissue Doppler imaging and speckle-tracking echocardiography, were examined. RESULTS Of 52 included infants with septic shock, 19 (37%) also had HRF. Baseline characteristics were similar. On TNE, although the HRF group more frequently had bidirectional/right-to-left flow across the patent ductus arteriosus (67% vs 33%; P = .08), all indices of PVR and RV function were similar. However, the HRF group demonstrated reduced LV systolic function (ejection fraction, 51.8% ± 12.3% vs 62.6% ± 13.0%; global peak systolic longitudinal strain -15.2% ± 4.5% vs -18.6% ± 4.5%), diastolic function (early [2.3 ± 1.0/s vs 3.6 ± 1.2/s]) and late (2.4/s [IQR, 1.9-2.6/s] vs 2.8/s [2.3-3.5/s] diastolic strain rate), and higher frequency of LV output <150 mL/min/kg (44% vs 12%) (all P < .05). CONCLUSIONS Acute HRF occurring in preterm neonates with septic shock is associated with alterations in TNE measures of LV function, and not PVR or RV function. Future studies should evaluate the impact of supporting LV function in these patients.
Collapse
Affiliation(s)
- Ashraf Kharrat
- Department of Pediatrics, Mount Sinai Hospital, Toronto, Ontario, Canada; Department of Pediatrics, University of Toronto, Toronto, Ontario, Canada.
| | - Sagee Nissimov
- Department of Pediatrics, Mount Sinai Hospital, Toronto, Ontario, Canada
| | - Faith Zhu
- Department of Pediatrics, Mount Sinai Hospital, Toronto, Ontario, Canada; Department of Pediatrics, University of Toronto, Toronto, Ontario, Canada
| | - Poorva Deshpande
- Department of Pediatrics, Mount Sinai Hospital, Toronto, Ontario, Canada; Department of Pediatrics, University of Toronto, Toronto, Ontario, Canada
| | - Amish Jain
- Department of Pediatrics, Mount Sinai Hospital, Toronto, Ontario, Canada; Department of Pediatrics, University of Toronto, Toronto, Ontario, Canada; Lunenfeld-Tanenbaum Research Institute, Toronto, Ontario, Canada
| |
Collapse
|
57
|
Huang W, Lou A, Wang J, Wang Y, Zhang W, Li J, Wang S, Geng S, Wang G, Li X. TMBIM1 ameliorates sepsis-induced cardiac dysfunction by promoting Parkin-mediated mitophagy. FASEB J 2025; 39:e70397. [PMID: 39937566 DOI: 10.1096/fj.202402599rr] [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: 10/28/2024] [Revised: 02/02/2025] [Accepted: 02/05/2025] [Indexed: 02/13/2025]
Abstract
Myocardial dysfunction is a significant complication of sepsis that is associated with elevated mortality rates. Transmembrane BAX inhibitor motif containing 1 (TMBIM1), a stress-responsive protein, has garnered interest in the field of cardiovascular disease for its cardioprotective properties. Nevertheless, the role of TMBIM1 on sepsis-induced cardiac dysfunction (SICD) remains unknown. Here, our findings revealed a significant elevation in TMBIM1 expression within the myocardium following endotoxin challenge and further demonstrate the cardioprotective effects of TMBIM1 through adenovirus-mediated gene manipulation. Notably, lipopolysaccharide exposure markedly induced mitochondrial dysfunction in cardiomyocytes, which was effectively alleviated by TMBIM1 overexpression, while TMBIM1 knockdown exacerbated this dysfunction. Moreover, in cardiomyocytes subjected to endotoxin challenge, TMBIM1 was observed to interact with Parkin, facilitating its translocation from the cytosol to damaged mitochondria. This interaction enhanced the activation of mitophagy, thereby promoting the clearance of dysfunctional mitochondria and subsequently mitigating cellular injury. Hence, targeting TMBIM1 could be a novel therapeutic strategy for treating SICD.
Collapse
Affiliation(s)
- Weichang Huang
- Department of Emergency Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
- State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Department of Critical Care Medicine, The 10th Affiliated Hospital of Southern Medical University (Dongguan People's Hospital), Southern Medical University, Dongguan, China
| | - Anni Lou
- Department of Emergency Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Jun Wang
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yuegang Wang
- Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Wenyong Zhang
- Department of Emergency Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Jierui Li
- Department of Emergency Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Shuanghu Wang
- The Laboratory of Clinical Pharmacy, The People's Hospital of Lishui, Lishui, Zhejiang, China
| | - Shiyu Geng
- Department of Emergency Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
- State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Guozhen Wang
- Department of Emergency Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
- State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Xu Li
- Department of Emergency Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
- State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
58
|
Voelker TL, Westhoff M, del Villar SG, Thai PN, Chiamvimonvat N, Nieves-Cintrón M, Dickson EJ, Dixon RE. Phosphoinositide Depletion and Compensatory β-adrenergic Signaling in Angiotensin II-Induced Heart Disease: Protection Through PTEN Inhibition. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.23.639781. [PMID: 40060428 PMCID: PMC11888262 DOI: 10.1101/2025.02.23.639781] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 03/16/2025]
Abstract
Contractile dysfunction, hypertrophy, and cell death during heart failure are linked to altered Ca2+ handling, and elevated levels of the hormone angiotensin II (AngII), which signals through Gq-coupled AT1 receptors, initiating hydrolysis of PIP2. Chronic elevation of AngII contributes to cardiac pathology, but the mechanisms linking sustained AngII signaling to heart dysfunction remain incompletely understood. Here, we demonstrate that chronic AngII exposure profoundly disrupts cardiac phosphoinositide homeostasis, triggering a cascade of cellular adaptations that ultimately impair cardiac function. Using in vivo AngII infusion combined with phospholipid mass spectrometry, super-resolution microscopy, and functional analyses, we show that sustained AngII signaling reduces PI(4,5)P2 levels and triggers extensive redistribution of CaV1.2 channels from t-tubules to various endosomal compartments. Despite this t-tubular channel loss, enhanced sympathetic drive maintains calcium currents and transients through increased channel phosphorylation via PKA and CaMKII pathways. However, this compensation proves insufficient as cardiac function progressively declines, marked by pathological hypertrophy, t-tubule disruption, and diastolic dysfunction. Notably, we identify depletion of PI(3,4,5)P3 as a critical mediator of AngII-induced cardiac pathology. While preservation of PI(3,4,5)P3 levels through PTEN inhibition did not prevent cellular remodeling or calcium handling changes, it protected against cardiac dysfunction, suggesting effects primarily through reduction of fibrosis. These findings reveal a complex interplay between phosphoinositide signaling, ion channel trafficking, and sympathetic activation in AngII-induced cardiac pathology. Moreover, they establish maintenance of PI(3,4,5)P3 as a promising therapeutic strategy for hypertensive heart disease and as a potential protective adjunct therapy during clinical AngII administration.
Collapse
Affiliation(s)
- Taylor L. Voelker
- Dept. of Physiology and Membrane Biology, University of California, Davis, CA, 95616, USA; present address: Department of Neuroscience, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Maartje Westhoff
- Dept. of Physiology and Membrane Biology, University of California, Davis, CA, 95616, USA
| | - Silvia G. del Villar
- Dept. of Physiology and Membrane Biology, University of California, Davis, CA, 95616, USA
| | - Phung N. Thai
- Department of Internal Medicine, Division of Cardiovascular Medicine, University of California, Davis, Davis, CA 95616, USA; present address: David Geffen School of Medicine at University of California, Los Angeles, CA 90095, USA
| | - Nipavan Chiamvimonvat
- Department of Internal Medicine, Division of Cardiovascular Medicine, University of California, Davis, Davis, CA 95616, USA; Dept. of Pharmacology, University of California, Davis, CA, 95616, USA; Department of Veterans Affairs, Northern California Health Care System, Mather, CA 95655, USA; present address: Department of Basic Medical Sciences and Translational Cardiovascular Research Center, University of Arizona College of Medicine, Phoenix, AZ 85004, USA
| | | | - Eamonn J. Dickson
- Dept. of Physiology and Membrane Biology, University of California, Davis, CA, 95616, USA
| | - Rose E. Dixon
- Dept. of Physiology and Membrane Biology, University of California, Davis, CA, 95616, USA
| |
Collapse
|
59
|
Han Y, Dawson JR, DeMarco KR, Rouen KC, Ngo K, Bekker S, Yarov-Yarovoy V, Clancy CE, Xiang YK, Ahn SH, Vorobyov I. Molecular simulations reveal intricate coupling between agonist-bound β-adrenergic receptors and G protein. iScience 2025; 28:111741. [PMID: 39898043 PMCID: PMC11787599 DOI: 10.1016/j.isci.2024.111741] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Revised: 10/24/2024] [Accepted: 12/18/2024] [Indexed: 02/04/2025] Open
Abstract
G protein-coupled receptors (GPCRs) and G proteins transmit signals from hormones and neurotransmitters across cell membranes, initiating downstream signaling and modulating cellular behavior. Using advanced computer modeling and simulation, we identified atomistic-level structural, dynamic, and energetic mechanisms of norepinephrine (NE) and stimulatory G protein (Gs) interactions with β-adrenergic receptors (βARs), crucial GPCRs for heart function regulation and major drug targets. Our analysis revealed distinct binding behaviors of NE within β1AR and β2AR despite identical orthosteric binding pockets. β2AR had an additional binding site, explaining variations in NE binding affinities. Simulations showed significant differences in NE dissociation pathways and receptor interactions with the Gs. β1AR binds Gs more strongly, while β2AR induces greater conformational changes in the α subunit of Gs. Furthermore, GTP and GDP binding to Gs may disrupt coupling between NE and βAR, as well as between βAR and Gs. These findings may aid in designing precise βAR-targeted drugs.
Collapse
Affiliation(s)
- Yanxiao Han
- Department of Physiology and Membrane Biology, University of California, Davis, Davis, CA 95616, USA
| | - John R.D. Dawson
- Department of Physiology and Membrane Biology, University of California, Davis, Davis, CA 95616, USA
- Biophysics Graduate Group, University of California, Davis, Davis, CA 95616, USA
| | - Kevin R. DeMarco
- Department of Physiology and Membrane Biology, University of California, Davis, Davis, CA 95616, USA
| | - Kyle C. Rouen
- Department of Physiology and Membrane Biology, University of California, Davis, Davis, CA 95616, USA
- Biophysics Graduate Group, University of California, Davis, Davis, CA 95616, USA
| | - Khoa Ngo
- Department of Physiology and Membrane Biology, University of California, Davis, Davis, CA 95616, USA
- Biophysics Graduate Group, University of California, Davis, Davis, CA 95616, USA
| | - Slava Bekker
- American River College, Sacramento, CA 95841, USA
| | - Vladimir Yarov-Yarovoy
- Department of Physiology and Membrane Biology, University of California, Davis, Davis, CA 95616, USA
- Department of Anesthesiology and Pain Medicine, University of California, Davis, Davis, CA 95616, USA
- Center for Precision Medicine and Data Science, University of California, Davis, Davis, CA 95616, USA
| | - Colleen E. Clancy
- Department of Physiology and Membrane Biology, University of California, Davis, Davis, CA 95616, USA
- Center for Precision Medicine and Data Science, University of California, Davis, Davis, CA 95616, USA
- Department of Pharmacology, University of California, Davis, Davis, CA 95616, USA
| | - Yang K. Xiang
- Department of Pharmacology, University of California, Davis, Davis, CA 95616, USA
- VA Northern California Health Care System, Mather, CA 95655, USA
| | - Surl-Hee Ahn
- Department of Chemical Engineering, University of California, Davis, Davis, CA 95616, USA
| | - Igor Vorobyov
- Department of Physiology and Membrane Biology, University of California, Davis, Davis, CA 95616, USA
- Department of Pharmacology, University of California, Davis, Davis, CA 95616, USA
| |
Collapse
|
60
|
Yan H, Li X, Liu B, Wu B, Chen C, He L, Wu G. Stellate Ganglionectomy Attenuates Pressure Overload-Induced Cardiac Hypertrophy and Dysfunction. FRONT BIOSCI-LANDMRK 2025; 30:26268. [PMID: 40018929 DOI: 10.31083/fbl26268] [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: 08/25/2024] [Revised: 10/28/2024] [Accepted: 12/10/2024] [Indexed: 03/01/2025]
Abstract
BACKGROUND Enhanced cardiac sympathetic activity contributes to chronic heart failure (CHF). Interventions targeting the stellate ganglion (SG) can reduce this activity, potentially slowing the progression of cardiovascular diseases. This study examined the effects and mechanisms of stellate ganglionectomy on myocardial hypertrophy and cardiac dysfunction caused by pressure overload. METHODS A rat model of pressure overload was created using abdominal aortic constriction. Four groups were studied: the sham surgery, abdominal aortic coarctation (AB), aortic constriction plus left stellate ganglionectomy (LSG), and aortic constriction plus right stellate ganglionectomy (RSG) groups. Cardiac function was assessed via echocardiography, and myocardial hypertrophy and fibrosis were evaluated using hematoxylin-eosin staining (H&E) and Masson staining. Serum atrial natriuretic peptides (ANP) and norepinephrine (NE) levels were measured using enzyme linked immunosorbent assay (ELISA), and the levels of the molecular markers tyrosine hydroxylase (TH) and growth-associated protein-43 (GAP43) were analyzed using Western blotting and PCR. Calcium calmodulin dependent protein kinase II (CaMKII) and phosphorylated Ryanodine Receptor 2 (p-RyR2) expression were also investigated. RESULTS Stellate ganglionectomy significantly reduced myocardial hypertrophy and improved cardiac function, as indicated by decreased left ventricular posterior wall thickness (LVPWD) (p < 0.01), left ventricular end-diastolic diameter (LVEDD) and volume (p < 0.001), left ventricular end-diastolic volume (LVEDV) (p < 0.001), increased left ventricular ejection fraction (LVEF) (p < 0.001) and left ventricular fractional shortening (LVFS) (p < 0.001). Histological analysis confirmed reduced myocardial dilation. Molecular analysis revealed decreased CaMKII/RyR2 signaling (p < 0.001) and lower NE levels (p < 0.01), suggesting reduced neurohormonal stress. CONCLUSIONS Stellate ganglionectomy alleviates hypertrophy and cardiac dysfunction caused by pressure overload, likely through inhibition of the CaMKII/RyR2 pathway, underscoring its potential as a therapeutic approach.
Collapse
Affiliation(s)
- Hui Yan
- Department of Pharmacy, Wuhan No.1 Hospital, 430022 Wuhan, Hubei, China
| | - Xiujun Li
- Department of Cardiology, Hubei Provincial Hospital of Traditional Chinese Medicine, Affiliated Hospital of Hubei University of Chinese Medicine, 431799 Wuhan, Hubei, China
| | - Beilei Liu
- Department of Cardiology, Hubei Provincial Hospital of Traditional Chinese Medicine, Affiliated Hospital of Hubei University of Chinese Medicine, 431799 Wuhan, Hubei, China
| | - Bin Wu
- Department of Cardiology, Hubei Provincial Hospital of Traditional Chinese Medicine, Affiliated Hospital of Hubei University of Chinese Medicine, 431799 Wuhan, Hubei, China
| | - Changgui Chen
- Department of Pharmacy, Wuhan No.1 Hospital, 430022 Wuhan, Hubei, China
| | - Liqun He
- Department of Pharmacy, Wuhan No.1 Hospital, 430022 Wuhan, Hubei, China
| | - Gang Wu
- Department of Cardiology, Hubei Key Laboratory of Cardiology, Renmin Hospital of Wuhan University, Cardiovascular Research Institute, Wuhan University, 430060 Wuhan, Hubei, China
| |
Collapse
|
61
|
Zhu Y, Zhang Q, Wang Y, Liu W, Zeng S, Yuan Q, Zhang K. Identification of Necroptosis and Immune Infiltration in Heart Failure Through Bioinformatics Analysis. J Inflamm Res 2025; 18:2465-2481. [PMID: 39991658 PMCID: PMC11847454 DOI: 10.2147/jir.s502203] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2024] [Accepted: 02/10/2025] [Indexed: 02/25/2025] Open
Abstract
Purpose Heart failure (HF) remains a leading cause of mortality and morbidity in cardiovascular disease. Research has shown that necroptosis contributes to HF, and immune infiltration has been reported to be implicated in HF. However, the specific mechanisms by which necroptosis and immune infiltration promote HF remain poorly understood. This study aims to elucidate these mechanisms, thereby providing new insights for future therapeutic strategies. Methods and Results In the GSE21610 dataset, there were 1848 differentially expressed genes (DEGs), 14 of which related to necroptosis (NRDEGs) in HF. Gene Set Enrichment Analysis (GSEA) indicated that Th1 and Th2 cell differentiation, TGF-beta signaling, Renin secretion, and Wnt signaling pathways may be closely associated with HF. The NRDEGs may play a role in responding to mechanical stimuli, membrane rafts, cytokine receptor binding, or the necroptosis signaling pathway. The protein-protein interaction (PPI) network identified EGFR, TXN, FASLG, MAPK14, and CASP8 as hub NRDEGs. Furthermore, immune infiltration analysis of CIBERSORT algorithm suggested that M2 macrophages, memory B cells, monocytes, regulatory T cells (Tregs), follicular helper T cells, and gamma delta T cells may participate in the development of HF. The hub NRDEGs, including EGFR, FASLG, and TXN, exhibited significant correlations with various immune cell types. Finally, animal models confirmed that in the HF group, EGFR and FASLG were up-regulated, while TXN was down-regulated. Conclusion The present findings demonstrate that necroptosis and immune infiltration are associated with the development of HF. This study provides valuable insights and recommendations for the clinical management of HF.
Collapse
Affiliation(s)
- Yuanting Zhu
- Department of Cardiology, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, People’s Republic of China
| | - Qiang Zhang
- Department of Emergency Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, People’s Republic of China
| | - Yanbo Wang
- Department of Cardiology, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, People’s Republic of China
| | - Wenqiang Liu
- Digestive Diseases Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, People’s Republic of China
| | - Shichen Zeng
- Clinical Medicine, Changsha Medical University, Changsha, People’s Republic of China
| | - Qinghua Yuan
- Department of Cardiology, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, People’s Republic of China
| | - Kun Zhang
- Department of Cardiology, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, People’s Republic of China
| |
Collapse
|
62
|
Liu Z, Li F, Li N, Chen Y, Chen Z. MicroRNAs as regulators of cardiac dysfunction in sepsis: pathogenesis and diagnostic potential. Front Cardiovasc Med 2025; 12:1517323. [PMID: 40041174 PMCID: PMC11876399 DOI: 10.3389/fcvm.2025.1517323] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Accepted: 01/29/2025] [Indexed: 03/06/2025] Open
Abstract
Introduction Sepsis, a life-threatening condition arising from an uncontrolled immune response to infection, can lead to organ dysfunction, with severe inflammation potentially causing multiple organ failures. Sepsis-induced cardiac dysfunction (SIMD) is a common and severe complication of sepsis, significantly increasing patient mortality. Understanding the pathogenesis of SIMD is crucial for improving treatment, and microRNAs (miRNAs) have emerged as important regulators in this process. Methods A comprehensive literature search was conducted in PubMed, Science Direct, and Embase databases up to September 2024. The search terms included ["miRNA" or "microRNA"] and ["Cardiac" or "Heart"] and ["Sepsis" or "Septic"], with the language limited to English. After initial filtering by the database search engine, Excel software was used to further screen references. Duplicate articles, those without abstracts or full texts, and review/meta-analyses or non-English articles were excluded. Finally, 106 relevant research articles were included for data extraction and analysis. Results The pathogenesis of SIMD is complex and involves mitochondrial dysfunction, oxidative stress, cardiomyocyte apoptosis and pyroptosis, dysregulation of myocardial calcium homeostasis, myocardial inhibitory factors, autonomic nervous regulation disorders, hemodynamic changes, and myocardial structural alterations. miRNAs play diverse roles in SIMD. They are involved in regulating the above-mentioned pathological processes. Discussion Although significant progress has been made in understanding the role of miRNAs in SIMD, there are still challenges. Some studies on the pathogenesis of SIMD have limitations such as small sample sizes and failure to account for confounding factors. Research on miRNAs also faces issues like inconsistent measurement techniques and unclear miRNA-target gene relationships. Moreover, the translation of miRNA-based research into clinical applications is hindered by problems related to miRNA stability, delivery mechanisms, off-target effects, and long-term safety. In conclusion, miRNAs play a significant role in the pathogenesis of SIMD and have potential as diagnostic biomarkers. Further research is needed to overcome existing challenges and fully exploit the potential of miRNAs in the diagnosis and treatment of SIMD.
Collapse
Affiliation(s)
- Zhen Liu
- Research Center of Experimental Acupuncture Science, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- School of Acupuncture-Moxibustion and Tuina, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Feiyang Li
- Research Center of Experimental Acupuncture Science, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- School of Acupuncture-Moxibustion and Tuina, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Ningcen Li
- Research Center of Experimental Acupuncture Science, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- School of Acupuncture-Moxibustion and Tuina, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yong Chen
- Department of Critical Care Medicine, Tianjin Hospital of ITCWM Nankai Hospital, Tianjin, China
| | - Zelin Chen
- Research Center of Experimental Acupuncture Science, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- School of Acupuncture-Moxibustion and Tuina, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| |
Collapse
|
63
|
Zadorozny L, Du J, Supanekar N, Annamalai K, Yu Q, Wang M. Caveolin and oxidative stress in cardiac pathology. Front Physiol 2025; 16:1550647. [PMID: 40041164 PMCID: PMC11876135 DOI: 10.3389/fphys.2025.1550647] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Accepted: 01/27/2025] [Indexed: 03/06/2025] Open
Abstract
Caveolins interact with signaling molecules within caveolae and subcellular membranes. Dysregulation of caveolin function and protein abundance contributes to cardiac pathophysiological processes, driving the development and progression of heart disease. Reactive oxygen species (ROS) play a critical role in maintaining cellular homeostasis and are key contributors to the pathophysiological mechanisms of cardiovascular disorders. Caveolins have been shown to modulate oxidative stress and regulate redox homeostasis. However, the specific roles of caveolins, particularly caveolin-1 and caveolin-3, in regulating ROS production during cardiac pathology remain unclear. This mini-review article highlights the correlation between caveolins and oxidative stress in maintaining cardiovascular health and modulating cardiac diseases, specifically in myocardial ischemia, heart failure, diabetes-induced metabolic cardiomyopathy, and septic cardiomyopathy. A deeper understanding of caveolin-mediated mechanisms may pave the way for innovative therapeutic approaches to treat cardiovascular diseases.
Collapse
Affiliation(s)
- Lauren Zadorozny
- Center for Surgical Sciences, Department of Surgery, Indiana University School of Medicine, Indianapolis, IN, United States
- Division of Cardiothoracic Surgery, Department of Surgery, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Jiayue Du
- Center for Surgical Sciences, Department of Surgery, Indiana University School of Medicine, Indianapolis, IN, United States
- Division of Cardiothoracic Surgery, Department of Surgery, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Neil Supanekar
- Center for Surgical Sciences, Department of Surgery, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Karthik Annamalai
- Center for Surgical Sciences, Department of Surgery, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Qing Yu
- Center for Surgical Sciences, Department of Surgery, Indiana University School of Medicine, Indianapolis, IN, United States
- Division of Cardiothoracic Surgery, Department of Surgery, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Meijing Wang
- Center for Surgical Sciences, Department of Surgery, Indiana University School of Medicine, Indianapolis, IN, United States
- Division of Cardiothoracic Surgery, Department of Surgery, Indiana University School of Medicine, Indianapolis, IN, United States
| |
Collapse
|
64
|
Qiu X, Zheng Q, Luo D, Ming Y, Zhang T, Pu W, Ai M, He J, Peng Y. Rational Design, Synthesis, and Biological Evaluation of Novel c-Met Degraders for Lung Cancer Therapy. J Med Chem 2025; 68:2815-2839. [PMID: 39882685 DOI: 10.1021/acs.jmedchem.4c02129] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2025]
Abstract
Cellular-mesenchymal epithelial transition factor (c-Met) is an attractive target for treating multiple cancers. Despite plentiful c-Met inhibitors have been developed, some issues, including the acquired drug resistance to c-Met inhibitors, have emerged to hamper their application in clinical treatment. Degradation of c-Met offers an opportunity to solve these issues. In this study, we developed a series of c-Met degraders, and the optimal compound 22b can efficiently degrade c-Met with a DC50 value of 0.59 nM in EBC-1 cells. Mechanistic studies revealed that compound 22b induced c-Met degradation via proteasome-mediated pathway. In addition, compound 22b suppressed the proliferation and also induced apoptosis of EBC-1 cells, outperforming the corresponding inhibitor tepotinib. Importantly, compound 22b showed favorable pharmacokinetic properties and significantly induced tumor regression in a xenograft model without obvious toxicity. In brief, this study provided compound 22b as a novel c-Met degrader for lung cancer therapy.
Collapse
Affiliation(s)
- Xingyang Qiu
- Center for Molecular Oncology, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy and Cancer Center, State Key Laboratory of Respiratory Health and Multimorbidity, West China Hospital, Sichuan University, 610064 Chengdu, China
| | - Qingquan Zheng
- Center for Molecular Oncology, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy and Cancer Center, State Key Laboratory of Respiratory Health and Multimorbidity, West China Hospital, Sichuan University, 610064 Chengdu, China
| | - Dongdong Luo
- Center for Molecular Oncology, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy and Cancer Center, State Key Laboratory of Respiratory Health and Multimorbidity, West China Hospital, Sichuan University, 610064 Chengdu, China
| | - Yue Ming
- Center for Molecular Oncology, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy and Cancer Center, State Key Laboratory of Respiratory Health and Multimorbidity, West China Hospital, Sichuan University, 610064 Chengdu, China
| | - Tingting Zhang
- Center for Molecular Oncology, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy and Cancer Center, State Key Laboratory of Respiratory Health and Multimorbidity, West China Hospital, Sichuan University, 610064 Chengdu, China
| | - Wenchen Pu
- Center for Molecular Oncology, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy and Cancer Center, State Key Laboratory of Respiratory Health and Multimorbidity, West China Hospital, Sichuan University, 610064 Chengdu, China
| | - Min Ai
- Center for Molecular Oncology, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy and Cancer Center, State Key Laboratory of Respiratory Health and Multimorbidity, West China Hospital, Sichuan University, 610064 Chengdu, China
| | - Jianhua He
- Center for Molecular Oncology, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy and Cancer Center, State Key Laboratory of Respiratory Health and Multimorbidity, West China Hospital, Sichuan University, 610064 Chengdu, China
| | - Yong Peng
- Center for Molecular Oncology, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy and Cancer Center, State Key Laboratory of Respiratory Health and Multimorbidity, West China Hospital, Sichuan University, 610064 Chengdu, China
- Frontiers Medical Center, Tianfu Jincheng Laboratory, 610212 Chengdu, China
| |
Collapse
|
65
|
Rode F, Pavlović N, Jordan A, Radić M, Lisičić A, Tomić SS, Kursar J, Manola Š, Jurin I. The use of beta-blockers for heart failure with reduced ejection fraction in the era of SGLT2 inhibitors - are we still afraid to up-titrate? Heart Vessels 2025:10.1007/s00380-025-02525-7. [PMID: 39934336 DOI: 10.1007/s00380-025-02525-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Accepted: 01/29/2025] [Indexed: 02/13/2025]
Abstract
Beta-blockers are one of the four major pillars of guideline-directed medical therapy (GDMT) for heart failure with reduced ejection fraction (HFrEF). The therapy has presented the best effects when up-titrated to evidence-based target doses. Despite their proven benefits, physicians have traditionally shown reluctance to up-titrate beta-blockers because of their negative inotropic and chronotropic effects. The effects of newly introduced sodium-glucose cotransporter 2 inhibitors (SGLT2I) in treating HFrEF might open more room for adequate beta-blockers up-titration. The goal of this study was to evaluate the up-titration practice, and impact of target doses of beta-blockers in patients with HFrEF receiving SGLT2I. This is a prospective cohort study involving patients with HFrEF receiving SGLT2I therapy. Baseline use and dosing to the evidence-based targets were examined. We compared the groups of patients receiving maximally titrated beta-blockers versus incompletely titrated. Primary outcome was composite of (1) rehospitalization or revisit to emergency unit due to the heart failure; (2) all-cause death and major adverse cardiac events (MACE). Secondary outcomes were heart rate at rest, left ventricular ejection fraction, NT-proBNP, and NYHA status at 6 and 12 months of follow-up. Study endpoints were documented via telephone interviews, regular outpatient follow-up, or by electronic hospital records. This study included a total of 458 patients with median follow-up time of 365 (186-502) days. A total of 122 (26.6%) patients had beta-blockers maximally up-titrated. The results show that adherence to maximal target doses of β-blocker therapy significantly reduces hazard of death or MACE comparing to not using maximal doses of β-blocker (factor 0.43). Hazard reduction was not statistically significant for composite of rehospitalization or revisit to emergency unit due to HF. Maximal doses of beta-blockers did not result in a significant decrease in resting heart rate. Our real-world data have highlighted the prevalence of incomplete titration of beta-blockers. Although it has been shown that evidence-based target dosing of beta-blockers reduces death and MACE, there is still room for improvement with up-titrating beta-blockers in eligible patients.
Collapse
Affiliation(s)
- Fran Rode
- University Hospital Dubrava, Gojka Šuška 6, 10000, Zagreb, Croatia.
| | - Nikola Pavlović
- University Hospital Dubrava, Gojka Šuška 6, 10000, Zagreb, Croatia
| | - Ana Jordan
- University Hospital Dubrava, Gojka Šuška 6, 10000, Zagreb, Croatia
| | - Marija Radić
- University Hospital Dubrava, Gojka Šuška 6, 10000, Zagreb, Croatia
| | - Ante Lisičić
- University Hospital Dubrava, Gojka Šuška 6, 10000, Zagreb, Croatia
| | | | - Jelena Kursar
- University Hospital Dubrava, Gojka Šuška 6, 10000, Zagreb, Croatia
| | - Šime Manola
- University Hospital Dubrava, Gojka Šuška 6, 10000, Zagreb, Croatia
| | - Ivana Jurin
- University Hospital Dubrava, Gojka Šuška 6, 10000, Zagreb, Croatia
| |
Collapse
|
66
|
Niu L, Gao M, Li Y, Wang C, Zhang C, Duan H, Li H, Wang F, Ge J. Effects of the stress hormone norepinephrine on the probiotic properties of Levilactobacillus: antibacterial colonization, anti-inflammation, and antioxidation. Front Microbiol 2025; 16:1526362. [PMID: 39996081 PMCID: PMC11849050 DOI: 10.3389/fmicb.2025.1526362] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Accepted: 01/16/2025] [Indexed: 02/26/2025] Open
Abstract
Probiotics as antibiotic alternatives are unstable for use under stress in clinical applications. To explore the influence of catecholamine hormones on probiotic bacterial inhibition and antimicrobial activity, we tested the effects of norepinephrine (NE) on Levilactobacillus in vitro and in a mouse model. The in vitro results showed that in the presence of NE, 80% of Levilactobacillus strains showed increased growth rate and more than 80% of the strains indicated lower antimicrobial activity at 22 h. Furthermore, in the mouse model, NE weakens the protective effect of L. brevis 23,017 on Escherichia coli infection, which is shown by the decreased ability of antibacterial colonization, antioxidation, and anti-inflammation, and downregulating the expression of antioxidant genes and intestinal mucosal barrier-related genes. At the same time, the addition of NE modulates the bacterial microbiota richness and diversity in the intestine, disrupting the balance of intestinal probiotics. These findings provide evidence that NE reduces the probiotic ability of Levilactobacillus and illustrates the plasticity of the probiotics in response to the intestinal microenvironment under stress.
Collapse
Affiliation(s)
- Lingdi Niu
- Heilongjiang Provincial Key Laboratory of Zoonosis, College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Mingchun Gao
- Heilongjiang Provincial Key Laboratory of Zoonosis, College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Yifan Li
- Heilongjiang Provincial Key Laboratory of Zoonosis, College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Chaonan Wang
- Heilongjiang Provincial Key Laboratory of Zoonosis, College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China
| | - Chuankun Zhang
- Heilongjiang Provincial Key Laboratory of Zoonosis, College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Haoyuan Duan
- Heilongjiang Provincial Key Laboratory of Zoonosis, College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Hai Li
- Heilongjiang Provincial Key Laboratory of Zoonosis, College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Fang Wang
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China
| | - Junwei Ge
- Heilongjiang Provincial Key Laboratory of Zoonosis, College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| |
Collapse
|
67
|
Lian Y, Zhang H, Xing W, Li S, Lai X, Jia S, Shang J, Liu H. Global Research Trends and Focus on the Link Between Heart Failure and NLRP3 Inflammasome: A Bibliometric Analysis From 2010 to 2024. J Multidiscip Healthc 2025; 18:697-710. [PMID: 39949850 PMCID: PMC11822290 DOI: 10.2147/jmdh.s505356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Accepted: 01/21/2025] [Indexed: 02/16/2025] Open
Abstract
Background Heart failure (HF) is characterized by elevated morbidity, mortality, and rehospitalization frequencies. This condition imposes a considerable medical burden and fiscal strain on society. Inflammation plays a crucial role in the inception, advancement, and outcome of HF. Despite mounting evidence demonstrating the pivotal function of the NLRP3 inflammasome in HF, a thorough bibliometric examination of research focal points and trajectories in this domain has yet to be undertaken. Methods Publications related to the NLRP3 inflammasome in HF were retrieved from the Web of Science database spanning 2010-2024. The acquired data were subsequently analyzed utilizing various visualization instruments, including Citespace, VOSviewer, Scimago Graphica, and Microsoft Office Excel 2021. Results A total of 282 papers were included in the analysis, authored by 2,130 researchers from 500 institutions across 34 nations/regions. China emerged as a significant contributor to this field, producing the highest number of outputs. Antonio Abbate was identified as the most prolific author. Virginia Commonwealth University and Wuhan University were the institutions with the highest publication output. INTERNATIONAL IMMUNOPHARMACOLOGY was the periodical with the most numerous publications in this sphere. CIRCULATION, however, received the highest number of citations, indicating its substantial influence on investigations in this field. Contemporary research focal points primarily concentrate on the activation and inhibition pathways of the NLRP3 inflammasome, the exploration of novel HF targets, and the association between HF and mitochondrial function. Future research trajectories are likely to encompass investigations into the relationship between HF and pyroptosis, as well as clinical studies on pharmaceuticals targeting the NLRP3 inflammasome as a therapeutic approach for HF. Conclusion This investigation provides a comprehensive bibliometric analysis and synopsis of NLRP3 inflammable-related studies in HF. The findings offer a conceptual foundation for further research on the NLRP3 inflammasome in HF and provide valuable guidance for future research directions in this domain.
Collapse
Affiliation(s)
- YanJie Lian
- Department of Cardiovascular, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, 100010, People’s Republic of China
- Graduate School of Capital Medical University, Beijing, 100069, People’s Republic of China
| | - Heyi Zhang
- Department of Cardiovascular, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, 100010, People’s Republic of China
- Graduate School of Capital Medical University, Beijing, 100069, People’s Republic of China
| | - Wenlong Xing
- Department of Cardiovascular, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, 100010, People’s Republic of China
| | - Sinai Li
- Beijing Institute of Traditional Chinese Medicine, Beijing, 100010, People’s Republic of China
| | - Xiaolei Lai
- Department of Cardiovascular, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, 100010, People’s Republic of China
| | - Sihan Jia
- Department of Cardiovascular, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, 100010, People’s Republic of China
| | - JuJu Shang
- Department of Cardiovascular, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, 100010, People’s Republic of China
| | - Hongxu Liu
- Department of Cardiovascular, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, 100010, People’s Republic of China
| |
Collapse
|
68
|
Kamel R, Bourcier A, Margaria JP, Jin V, Varin A, Hivonnait A, Mercier‐Nomé F, Mika D, Ghigo A, Charpentier F, Algalarrondo V, Hirsch E, Fischmeister R, Vandecasteele G, Leroy J. Cardiac Gene Therapy With Phosphodiesterase 2A Limits Remodeling and Arrhythmias in Mouse Models of Heart Failure. J Am Heart Assoc 2025; 14:e037343. [PMID: 39895533 PMCID: PMC12074716 DOI: 10.1161/jaha.124.037343] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Accepted: 12/23/2024] [Indexed: 02/04/2025]
Abstract
BACKGROUND PDE2 (phosphodiesterase 2) is upregulated in human heart failure. Cardiac PDE2-transgenic mice are protected against contractile dysfunction and arrhythmias in heart failure but whether an acute elevation of PDE2 could be of therapeutic value remains elusive. This hypothesis was tested using cardiac PDE2 gene transfer in preclinical models of heart failure. METHODS AND RESULTS C57BL/6 male mice were injected with serotype 9 adeno-associated viruses encoding for PDE2A. This led to a ≈10-fold rise of PDE2A protein levels that affected neither cardiac structure nor function in healthy mice. Two weeks after inoculation with serotype 9 adeno-associated viruses, mice were implanted with minipumps delivering either NaCl, isoproterenol (60 mg/kg per day), or isoproterenol and phenylephrine (30 mg/kg per day each) for 2 weeks. In mice injected with serotype 9 adeno-associated viruses encoding for LUC (luciferase), isoproterenol or isoproterenol+phenylephrine infusion induced left ventricular hypertrophy, decreased ejection fraction unveiled by echocardiography, and promoted fibrosis and apoptosis assessed by Masson's trichrome and Tunel, respectively. Furthermore, inotropic responses to isoproterenol of ventricular cardiomyocytes isolated from isoproterenol+phenylephrine-LUC mice loaded with 1 μmol/L Fura-2AM and stimulated at 1 Hz to record calcium transients and sarcomere shortening were dampened. Spontaneous calcium waves at the cellular level were promoted as well as ventricular arrhythmias evoked in vivo by catheter-mediated ventricular pacing after isoproterenol (1.5 mg/kg) and atropine (1 mg/kg) injection. However, increased PDE2A blunted these adverse outcomes evoked by sympathomimetic amines. CONCLUSIONS Cardiac gene therapy with PDE2A limits left ventricle remodeling, dysfunction, and arrhythmias evoked by catecholamines, providing evidence that increasing PDE2A activity acutely could prevent progression toward heart failure.
Collapse
MESH Headings
- Animals
- Heart Failure/therapy
- Heart Failure/physiopathology
- Heart Failure/genetics
- Heart Failure/enzymology
- Genetic Therapy/methods
- Disease Models, Animal
- Mice, Inbred C57BL
- Male
- Ventricular Remodeling
- Arrhythmias, Cardiac/genetics
- Arrhythmias, Cardiac/physiopathology
- Arrhythmias, Cardiac/enzymology
- Arrhythmias, Cardiac/prevention & control
- Arrhythmias, Cardiac/therapy
- Cyclic Nucleotide Phosphodiesterases, Type 2/genetics
- Cyclic Nucleotide Phosphodiesterases, Type 2/metabolism
- Cyclic Nucleotide Phosphodiesterases, Type 2/biosynthesis
- Myocytes, Cardiac/enzymology
- Myocytes, Cardiac/drug effects
- Myocytes, Cardiac/pathology
- Dependovirus/genetics
- Mice
- Hypertrophy, Left Ventricular/physiopathology
- Hypertrophy, Left Ventricular/genetics
- Hypertrophy, Left Ventricular/enzymology
- Isoproterenol
- Apoptosis
- Ventricular Function, Left
- Fibrosis
Collapse
Affiliation(s)
- Rima Kamel
- Université Paris‐Saclay, Inserm, Signaling and Cardiovascular Pathophysiology, UMR‐S 1180OrsayFrance
| | - Aurélia Bourcier
- Université Paris‐Saclay, Inserm, Signaling and Cardiovascular Pathophysiology, UMR‐S 1180OrsayFrance
| | - Jean Piero Margaria
- Université Paris‐Saclay, Inserm, Signaling and Cardiovascular Pathophysiology, UMR‐S 1180OrsayFrance
- Molecular Biotechnology Center “Guido Tarone”, Department of Molecular Biotechnology and Health SciencesUniversity of TorinoItaly
- Cystic Kidney Disorders Unit, Division of Genetics and Cell BiologyUniversità Vita‐Salute San RaffaeleMilanItaly
| | - Valentin Jin
- Université Paris‐Saclay, Inserm, Signaling and Cardiovascular Pathophysiology, UMR‐S 1180OrsayFrance
| | - Audrey Varin
- Université Paris‐Saclay, Inserm, Signaling and Cardiovascular Pathophysiology, UMR‐S 1180OrsayFrance
| | - Agnès Hivonnait
- Nantes Université, CNRS, INSERM, l’institut du thoraxNantesFrance
| | | | - Delphine Mika
- Université Paris‐Saclay, Inserm, Signaling and Cardiovascular Pathophysiology, UMR‐S 1180OrsayFrance
| | - Alessandra Ghigo
- Molecular Biotechnology Center “Guido Tarone”, Department of Molecular Biotechnology and Health SciencesUniversity of TorinoItaly
| | | | - Vincent Algalarrondo
- Université Paris‐Saclay, Inserm, Signaling and Cardiovascular Pathophysiology, UMR‐S 1180OrsayFrance
| | - Emilio Hirsch
- Molecular Biotechnology Center “Guido Tarone”, Department of Molecular Biotechnology and Health SciencesUniversity of TorinoItaly
| | - Rodolphe Fischmeister
- Université Paris‐Saclay, Inserm, Signaling and Cardiovascular Pathophysiology, UMR‐S 1180OrsayFrance
| | - Grégoire Vandecasteele
- Université Paris‐Saclay, Inserm, Signaling and Cardiovascular Pathophysiology, UMR‐S 1180OrsayFrance
| | - Jérôme Leroy
- Université Paris‐Saclay, Inserm, Signaling and Cardiovascular Pathophysiology, UMR‐S 1180OrsayFrance
| |
Collapse
|
69
|
Gao C, Song N, Wang Y. A New GRK2 Inhibitor for Heart Failure. JACC Basic Transl Sci 2025; 10:218-220. [PMID: 40131156 PMCID: PMC11897434 DOI: 10.1016/j.jacbts.2024.12.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Accepted: 12/04/2024] [Indexed: 03/26/2025]
Affiliation(s)
- Chen Gao
- Department of Pharmacology, Physiology and Neurobiology, University of Cincinnati, School of Medicine, Cincinnati, Ohio, USA
| | - Ningjing Song
- Department of Pharmacology, Physiology and Neurobiology, University of Cincinnati, School of Medicine, Cincinnati, Ohio, USA
| | - Yibin Wang
- Signature Research Program in Cardiovascular and Metabolic Diseases, DukeNUS Medical School, Singapore; National Heart Center of Singapore and National Heart Research Institute of Singapore, Singapore; Department of Medicine, Duke University School of Medicine, North Carolina, USA.
| |
Collapse
|
70
|
Ge Y, Ma E, Guo X, Wang Q, Zhu W, Ren D, Wo D. Baicalin Prevents Chronic β-AR Agonist-Induced Heart Failure via Preventing Oxidative Stress and Overactivation of the NADPH Oxidase NOX2. J Cell Mol Med 2025; 29:e70388. [PMID: 39988987 PMCID: PMC11847971 DOI: 10.1111/jcmm.70388] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2024] [Accepted: 01/17/2025] [Indexed: 02/25/2025] Open
Abstract
Heart failure (HF) remains the leading cause of mortality worldwide. Although various drugs are currently used in the treatment of HF, including angiotensin receptor blockers, angiotensin-converting enzyme inhibitors and beta blockers, none of these drugs can reverse the physiological remodelling of the heart associated with HF. Therefore, discovering novel drugs that can limit the extent of HF or prevent the structural dysfunction of the heart during HF progression is urgently needed. Baicalin is a natural flavonoid widely used in Traditional Chinese Medicine for its anti-inflammatory and anti-oxidative effects; however, the role of baicalin in chronic HF, in particular its underlying mechanisms of action, remains largely unelucidated. Murine models of beta-adrenergic receptor agonist (β-AR)-induced HF were induced via chronic induction with isoproterenol (ISO) for 4 weeks. Furthermore, we examined the effects and mechanisms of baicalin in protecting against ISO-induced cardiac impairment and HF. Daily administrations of baicalin robustly protected against chronic ISO-induced pathophysiological changes of the heart, including cardiac hypertrophy, reduced ejection fraction, fibrosis and remodelling. Baicalin also strongly inhibited the production of reactive oxygen and nitrogen species in the heart by preventing overactivation of the NADPH oxidase NOX2. Hence, the cardioprotective effects of baicalin in preventing chronic β-AR-induced HF were due to preventing the overactivation of NOX2 and generation of excessive oxidative stress. Our findings provide new mechanistic insight and suggest the therapeutic potential of baicalin as a novel drug in the treatment of chronic HF.
Collapse
Affiliation(s)
- Yixuan Ge
- Academy of Integrative Medicine, College of Integrative Medicine, Fujian Key Laboratory of Integrative Medicine on GeriatricFujian University of Traditional Chinese Medicine, Fujian Key Laboratory of Integrative Medicine on GeriatricFuzhouFujianChina
| | - En Ma
- Academy of Integrative Medicine, College of Integrative Medicine, Fujian Key Laboratory of Integrative Medicine on GeriatricFujian University of Traditional Chinese Medicine, Fujian Key Laboratory of Integrative Medicine on GeriatricFuzhouFujianChina
| | - Xiaowei Guo
- Academy of Integrative Medicine, College of Integrative Medicine, Fujian Key Laboratory of Integrative Medicine on GeriatricFujian University of Traditional Chinese Medicine, Fujian Key Laboratory of Integrative Medicine on GeriatricFuzhouFujianChina
| | - Qing Wang
- Academy of Integrative Medicine, College of Integrative Medicine, Fujian Key Laboratory of Integrative Medicine on GeriatricFujian University of Traditional Chinese Medicine, Fujian Key Laboratory of Integrative Medicine on GeriatricFuzhouFujianChina
| | - Weidong Zhu
- Academy of Integrative Medicine, College of Integrative Medicine, Fujian Key Laboratory of Integrative Medicine on GeriatricFujian University of Traditional Chinese Medicine, Fujian Key Laboratory of Integrative Medicine on GeriatricFuzhouFujianChina
| | - Dan‐ni Ren
- Academy of Integrative Medicine, College of Integrative Medicine, Fujian Key Laboratory of Integrative Medicine on GeriatricFujian University of Traditional Chinese Medicine, Fujian Key Laboratory of Integrative Medicine on GeriatricFuzhouFujianChina
| | - Da Wo
- Academy of Integrative Medicine, College of Integrative Medicine, Fujian Key Laboratory of Integrative Medicine on GeriatricFujian University of Traditional Chinese Medicine, Fujian Key Laboratory of Integrative Medicine on GeriatricFuzhouFujianChina
| |
Collapse
|
71
|
Ni Y, Tong Q, Xu M, Gu J, Ye H. Gut Microbiota-Induced Modulation of the Central Nervous System Function in Parkinson's Disease Through the Gut-Brain Axis and Short-Chain Fatty Acids. Mol Neurobiol 2025; 62:2480-2492. [PMID: 39134825 DOI: 10.1007/s12035-024-04370-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Accepted: 07/12/2024] [Indexed: 01/28/2025]
Abstract
Recent insights into Parkinson's disease (PD), a progressive neurodegenerative disorder, suggest a significant influence of the gut microbiome on its pathogenesis and progression through the gut-brain axis. This study integrates 16S rRNA sequencing, high-throughput transcriptomic sequencing, and animal model experiments to explore the molecular mechanisms underpinning the role of gut-brain axis in PD, with a focus on short-chain fatty acids (SCFAs) mediated by the SCFA receptors FFAR2 and FFAR3. Our findings highlighted prominent differences in the gut microbiota composition between PD patients and healthy individuals, particularly in taxa such as Escherichia_Shigella and Bacteroidetes, which potentially impact SCFA levels through secondary metabolite biosynthesis. Notably, fecal microbiota transplantation (FMT) from healthy to 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced PD mouse models significantly improved motor function, enhanced dopamine and serotonin levels in the striatum, and increased the number of dopaminergic neurons in the substantia nigra while reducing glial cell activation. This therapeutic effect was associated with increased levels of SCFAs such as acetate, propionate, and butyrate in the gut of MPTP-lesioned mice. Moreover, transcriptomic analyses revealed upregulated expression of FFAR2 and FFAR3 in MPTP-lesioned mice, indicating their crucial role in mediating the benefits of FMT on the central nervous system. These results provide compelling evidence that gut microbiota and SCFAs play a critical role in modulating the gut-brain axis, offering new insights into PD's etiology and potential targets for therapeutic intervention.
Collapse
Affiliation(s)
- Yiting Ni
- Department of Neurology, The Third Affiliated Hospital of Shanghai University (Wenzhou People's Hospital), Wenzhou, 325000, China
| | - Qiaowen Tong
- Department of Neurology, The Third Affiliated Hospital of Shanghai University (Wenzhou People's Hospital), Wenzhou, 325000, China
| | - Mengying Xu
- Department of Neurology, The Third Affiliated Hospital of Shanghai University (Wenzhou People's Hospital), Wenzhou, 325000, China
| | - Jiayi Gu
- Department of Neurology, The Third Affiliated Hospital of Shanghai University (Wenzhou People's Hospital), Wenzhou, 325000, China
| | - Hua Ye
- Department of Neurology, The Third Affiliated Hospital of Shanghai University (Wenzhou People's Hospital), Wenzhou, 325000, China.
| |
Collapse
|
72
|
Roy R, Schumacher SM, Murphy HC, Grondolsky J, Rosales TO, Chuprun JK, Gao E, Zhao H, Berretta RM, Hobby ARH, Houser SR, Avramova LV, Tesmer JJG, Koch WJ. Therapeutic Efficacy of a Novel Pharmacologic GRK2 Inhibitor in Multiple Animal Models of Heart Failure. JACC Basic Transl Sci 2025; 10:202-217. [PMID: 40131155 PMCID: PMC11897459 DOI: 10.1016/j.jacbts.2024.10.008] [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: 06/19/2023] [Revised: 10/02/2024] [Accepted: 10/02/2024] [Indexed: 03/26/2025]
Abstract
GRK2 is the most prominent G protein-coupled receptor kinase that is upregulated in heart failure (HF), and inhibiting GRK2 has improved cardiac function in mice. CCG258208, generated from the paroxetine scaffold, which has GRK2 inhibitory properties, has a 50-fold higher selectivity for GRK2 at 100-fold lower doses. We evaluated CCG258208 in 2 mice HF models and found that CCG258208 has robust therapeutic effects. In a chronic mini-swine HF model, acute administration of CCG258208 enhanced dobutamine inotropic responses. Our results indicate that CCG258208 has robust cardioprotective and HF-reversing effects in different HF models and it stands as a promising lead for HF therapy.
Collapse
Affiliation(s)
- Rajika Roy
- Center for Translational Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania, USA; Department of Surgery (Division of Cardiovascular and Thoracic Surgery), Duke Cardiovascular Research Center, Duke University School of Medicine, Durham, North Carolina, USA
| | - Sarah M Schumacher
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Haley Christine Murphy
- Center for Translational Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania, USA
| | - Jessica Grondolsky
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Thiele Osvaldt Rosales
- Center for Translational Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania, USA; Department of Surgery (Division of Cardiovascular and Thoracic Surgery), Duke Cardiovascular Research Center, Duke University School of Medicine, Durham, North Carolina, USA
| | - J Kurt Chuprun
- Center for Translational Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania, USA; Department of Surgery (Division of Cardiovascular and Thoracic Surgery), Duke Cardiovascular Research Center, Duke University School of Medicine, Durham, North Carolina, USA
| | - Erhe Gao
- Center for Translational Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania, USA
| | - Huaqing Zhao
- Center for Biostatistics and Epidemiology, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania, USA
| | - Remus M Berretta
- Center for Cardiovascular Research, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania, USA
| | - Alexander R H Hobby
- Center for Cardiovascular Research, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania, USA
| | - Steven R Houser
- Center for Cardiovascular Research, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania, USA
| | - Larisa V Avramova
- Department of Biological Sciences, Purdue University, West Lafayette, Indiana, USA; Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, Indiana, USA
| | - John J G Tesmer
- Department of Biological Sciences, Purdue University, West Lafayette, Indiana, USA; Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, Indiana, USA
| | - Walter J Koch
- Center for Translational Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania, USA; Department of Surgery (Division of Cardiovascular and Thoracic Surgery), Duke Cardiovascular Research Center, Duke University School of Medicine, Durham, North Carolina, USA.
| |
Collapse
|
73
|
Yi L, Han N, Li Z, Jiang H, Cao Z. Relaxin-2 promotes osteoblastic differentiation mediated by epidermal growth factor and epidermal growth factor receptor signaling. Biotechnol Appl Biochem 2025; 72:260-267. [PMID: 39219221 DOI: 10.1002/bab.2661] [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: 12/11/2023] [Accepted: 08/16/2024] [Indexed: 09/04/2024]
Abstract
Loss of osteogenic differentiation potential of osteoblasts has been associated with the pathogenesis of osteoporosis. Thus, stimulation of osteoblastic differentiation is a therapeutic strategy for osteoporosis. Relaxin-2 is a peptide hormone with potent biological functions. However, the effects of Relaxin-2 in osteoblastic differentiation and osteoporosis have not been reported before. Here, we report a novel physiological role of Relaxin-2 in promoting osteoblastic differentiation and mineralization of MC3T3-E1 cells. Our results indicate that exposure to Relaxin-2 upregulated the expression, and elevated the activity of alkaline phosphatase (ALP) when MC3T3-E1 cells were cultured in osteogenic differentiation medium (OM). Additionally, Relaxin-2 upregulated the mRNA levels of osteocalcin (ocn), osteopontin (opn), and collagen type I alpha 1 (Col1a1). The alizarin red S staining assay revealed that Relaxin-2 promoted the mineralization of MC3T3-E1 cells. We also found that Relaxin-2 increased the expression of Runx-2 as well as the epidermal growth factor (EGF) and epidermal growth factor receptor (EGFR). Importantly, silencing of EGF abolished the effects of Relaxin-2 in osteoblastic differentiation and related gene expression. These findings suggest that Relaxin-2 stimulates osteogenic differentiation through activating EGF/EGFR signaling.
Collapse
Affiliation(s)
- Lankai Yi
- Department of Hand, Foot, and Orthopedics Surgery, Weifang People's Hospital, Weifang, Shandong Province, China
| | - Ning Han
- Department of Hand, Foot, and Orthopedics Surgery, Weifang People's Hospital, Weifang, Shandong Province, China
| | - Zhong Li
- Department of Hand, Foot, and Orthopedics Surgery, Weifang People's Hospital, Weifang, Shandong Province, China
| | - Housen Jiang
- Department of Hand, Foot, and Orthopedics Surgery, Weifang People's Hospital, Weifang, Shandong Province, China
| | - Zhenhao Cao
- Department of Hand, Foot, and Orthopedics Surgery, Weifang People's Hospital, Weifang, Shandong Province, China
| |
Collapse
|
74
|
Al Khafaji AT, Barakat AM, Shayyal AJ, Taan AA, Aboqader Al-Aouadi RF. Managing Doxorubicin Cardiotoxicity: Insights Into Molecular Mechanisms and Protective Strategies. J Biochem Mol Toxicol 2025; 39:e70155. [PMID: 39887483 DOI: 10.1002/jbt.70155] [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: 12/04/2024] [Revised: 12/30/2024] [Accepted: 01/16/2025] [Indexed: 02/01/2025]
Abstract
Cancer ranks as the second leading cause of death in the United States and poses a significant health challenge globally. Numerous therapeutic options exist for treating cancer, with chemotherapy being one of the most prominent. Chemotherapy involves the use of antineoplastic drugs, either alone or in combination with other medications, to target and kill cancer cells. However, these drugs can also adversely affect healthy cells, leading to various side effects. Among the most commonly used chemotherapy agents are anthracyclines, which include doxorubicin, daunorubicin, and epirubicin. Doxorubicin is particularly notable for its effectiveness but is also associated with significant cardiotoxicity, a common concern for patients undergoing chemotherapy. Unfortunately, there is currently no definitive treatment to prevent or reverse this cardiotoxicity. The cardiac effects of doxorubicin can manifest in several ways, including changes in electrocardiograms, arrhythmias, myocarditis, pericarditis, myocardial infarction, cardiomyopathy, heart failure, and congestive heart failure. These complications may arise during treatment, shortly after it concludes, or even weeks later. Various mechanisms have been proposed to explain doxorubicin-induced cardiotoxicity. Key factors include the inhibition of topoisomerase IIβ, mitochondrial damage, reactive oxygen species (ROS) production due to iron metabolism, increased oxidative stress, heightened inflammatory responses, and elevated rates of apoptosis and necrosis within cardiac tissue. This review article will provide a comprehensive overview of the current state of knowledge regarding doxorubicin-induced cardiomyopathy. We will explore the underlying molecular mechanisms contributing to this condition and discuss emerging therapeutic strategies aimed at mitigating its impact on cancer survivors.
Collapse
Affiliation(s)
| | | | | | - Ali Adnan Taan
- Nasr City Hospital for Health Insurance, Ministry of Health, Cairo, Egypt
| | | |
Collapse
|
75
|
Gao H, Wang X, Yang Q. Septic cardiomyopathy or myocardial infarction?: A case report of septic shock with ST-segment elevation on ECG. Medicine (Baltimore) 2025; 104:e41454. [PMID: 39889150 PMCID: PMC11789888 DOI: 10.1097/md.0000000000041454] [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: 09/18/2024] [Revised: 01/12/2025] [Accepted: 01/17/2025] [Indexed: 02/02/2025] Open
Abstract
RATIONALE Sepsis is one of the most prevalent and deadly diseases today. Sepsis involving the heart can progress to septic cardiomyopathy; however, there is a lack of uniform diagnostic criteria. A review of the literature reveals a paucity of literature on sepsis combined with acute myocardial infarction (AMI) and no reports on emergency surgical treatment. PATIENTS CONCERNS A 52-year-old patient with trauma-induced sepsis leading to acute heart failure with elevated ST-segment on electrocardiogram and postoperative coronary angiography suggestive of AMI. DIAGNOSES Small bowel rupture, infectious shock, AMI, hypertensive disease, old cerebral infarction. INTERVENTIONS The patient was admitted to the hospital and immediately underwent emergency surgery to remove the infected focus, with treatment with meropenem for anti-infection, ambroxol for sputum, parenteral nutritional support, sedation and analgesia, esmolol to control the ventricular rate, uradil to control blood pressure, and transfusion of red blood cells and plasma for correction of anemia and coagulation functions. Coronary angiography was performed 6 months later. OUTCOMES The patient was discharged after showing signs of improvement and was subsequently monitored in an outpatient clinic setting. At the time of writing, the patient is still alive and well. LESSONS In cases of acute heart failure resulting from trauma-induced sepsis, it is crucial to consider myocardial ischemia as a potential factor. Early surgical removal of infected foci may prove beneficial in improving the patient's prognosis. However, differentiating between septic cardiomyopathy and sepsis-combined myocardial infarction can be challenging, and the appropriateness of the diagnostic criteria for sepsis at this stage is debatable.
Collapse
Affiliation(s)
- Haolei Gao
- Department of Anesthesiology, Weihai Municipal Hospital, Cheeloo College of Medicine, Shandong University, Weihai City, Shandong Province, China
| | - Xiaodong Wang
- Department of Anesthesiology, Weihai Municipal Hospital, Cheeloo College of Medicine, Shandong University, Weihai City, Shandong Province, China
| | - Qingyue Yang
- Department of Critical Care Medicine, Laizhou People’s Hospital, Yantai City, Shandong Province, China
| |
Collapse
|
76
|
Zheng E, Warchoł I, Mejza M, Możdżan M, Strzemińska M, Bajer A, Madura P, Żak J, Plewka M. Exploring Anti-Inflammatory Treatment as Upstream Therapy in the Management of Atrial Fibrillation. J Clin Med 2025; 14:882. [PMID: 39941553 PMCID: PMC11818443 DOI: 10.3390/jcm14030882] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Revised: 01/05/2025] [Accepted: 01/14/2025] [Indexed: 02/16/2025] Open
Abstract
Inflammation has been widely recognized as one of the major pathophysiological drivers of the development of atrial fibrillation (AF), which works in tandem with other risk factors of AF including obesity, diabetes, hypertension, and heart failure (HF). Our current understanding of the role of inflammation in the natural history of AF remains elusive; however, several key players, including the NLRP3 (NLR family pyrin domain containing 3) inflammasome, have been acknowledged to be heavily influential on chronic inflammation in the atrial myocardium, which leads to fibrosis and eventual degradation of its electrical function. Nevertheless, our current methods of pharmacological modalities with reported immunomodulatory properties, including well-established classes of drugs e.g., drugs targeting the renin-angiotensin-aldosterone system (RAAS), statins, and vitamin D, have proven effective in reducing the overall risk of developing AF, the onset of postoperative atrial fibrillation (POAF), and reducing overall mortality among patients with AF. This might bring hope for further progress in developing new treatment modalities targeting cellular checkpoints of the NLRP3 inflammasome pathway, or revisiting other well-known anti-inflammatory drugs e.g., colchicine, vitamin C, nonsteroidal anti-inflammatory drugs (NSAIDs), glucocorticosteroids, and antimalarial drugs. In our review, we aim to find relevant upstream anti-inflammatory treatment methods for the management of AF and present the most current real-world evidence of their clinical utility.
Collapse
|
77
|
Sumneang N, Kobroob A, Phungphong S, Boonhoh W, Punsawad C, Kangwan N. Fermented Houttuynia cordata Juice Exerts Cardioprotective Effects by Alleviating Cardiac Inflammation and Apoptosis in Rats with Lipopolysaccharide-Induced Sepsis. Nutrients 2025; 17:501. [PMID: 39940359 PMCID: PMC11820264 DOI: 10.3390/nu17030501] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2025] [Revised: 01/27/2025] [Accepted: 01/28/2025] [Indexed: 02/16/2025] Open
Abstract
BACKGROUND/OBJECTIVES Sepsis-induced cardiac dysfunction is a major problem that often leads to severe complications and a poor prognosis. Despite the growing awareness of its impact, effective treatment options for sepsis-induced cardiac dysfunction remain limited. To date, fermented products of Houttuynia cordata (HC), known for its rich bioactive properties, have shown potential in modulating inflammatory and oxidative stress pathways. However, treatment with fermented HC juice (FHJ) in lipopolysaccharide (LPS)-induced sepsis in rats has not been investigated. METHODS Rats were pretreated with FHJ at doses of 200 mg/kg and 400 mg/kg for 2 weeks. After that, the rats were injected with a single dose of LPS (10 mg/kg), and 12 h after injection, they developed sepsis-induced cardiac dysfunction. Then, cardiac function, oxidative stress, inflammation, apoptosis, and cardiac injury markers were determined. RESULTS Pretreatment with FHJ at doses of 200 mg/kg and 400 mg/kg prevented LPS-induced cardiac dysfunction in rats by attenuating cardiac inflammation (IL-1β, TLR-4, and NF-κB levels), oxidative stress (MDA levels), and apoptosis (cleaved-caspase 3 and Bax/Bcl-2 expression) and reducing markers of cardiac injury (LDH and CK-MB levels). CONCLUSIONS These results suggest that FHJ could be a potential therapeutic agent for sepsis-induced heart disease.
Collapse
Affiliation(s)
- Natticha Sumneang
- Department of Medical Science, School of Medicine, Walailak University, Nakhon Si Thammarat 80160, Thailand; (N.S.); (S.P.); (C.P.)
- Research Center in Tropical Pathobiology, Walailak University, Nakhon Si Thammarat 80160, Thailand
| | - Anongporn Kobroob
- Division of Physiology, School of Medical Sciences, University of Phayao, Phayao 56000, Thailand;
| | - Sukanya Phungphong
- Department of Medical Science, School of Medicine, Walailak University, Nakhon Si Thammarat 80160, Thailand; (N.S.); (S.P.); (C.P.)
- Research Center in Tropical Pathobiology, Walailak University, Nakhon Si Thammarat 80160, Thailand
| | - Worakan Boonhoh
- Akkhraratchakumari Veterinary College, Walailak University, Nakhon Si Thammarat 80160, Thailand;
| | - Chuchard Punsawad
- Department of Medical Science, School of Medicine, Walailak University, Nakhon Si Thammarat 80160, Thailand; (N.S.); (S.P.); (C.P.)
- Research Center in Tropical Pathobiology, Walailak University, Nakhon Si Thammarat 80160, Thailand
| | - Napapan Kangwan
- Division of Physiology, School of Medical Sciences, University of Phayao, Phayao 56000, Thailand;
| |
Collapse
|
78
|
Wang L, Dai X, Yu L, Li H, Zhang X, Yu Q, Lv X, Wang Y, Zhang S, Hao G, Wang H, Wang Z. Dexmedetomidine therapy promotes cardiac dysfunction and increases mortality in sepsis: A translational study. Int Immunopharmacol 2025; 146:113924. [PMID: 39732103 DOI: 10.1016/j.intimp.2024.113924] [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: 10/22/2024] [Revised: 12/18/2024] [Accepted: 12/19/2024] [Indexed: 12/30/2024]
Abstract
Previous studies demonstrated that dexmedetomidine (Dex) posttreatment aggravated myocardial dysfunction and reduced survival in septic mice. Yet, whether Dex elicits similar effects in septic patients as defined by Sepsis-3 remains unknown. This study sought to assess the effects of Dex-based sedation on mortality and cardiac dysfunction in septic patients defined by Sepsis-3 and to further reveal the mechanisms in septic rats. In the retrospective cohort study, patients were categorised into sepsis with Dex, other sedatives (propofol or midazolam) or without sedatives, mortality at 28 days were compared, and patients with measurements of cardiovascular biomarkers and echocardiography were used to examine the effect of Dex on cardiac dysfunction. Septic rats and Langendorff-perfused isolated rat hearts were used, cardiac function, mortality and pro-inflammatory mediators were analyzed. The all-cause mortality of septic patients receiving Dex reached to 35.2 % on Day 28, significantly higher than that of patients with other sedatives (16.1 %), while no difference with group of no sedatives (27.3 %). Patients in Dex group showed lower left ventricular EF and lateral mitral annular early diastolic peak velocities, but higher interventricular septum diastolic dimension compared to those with other sedatives. The plasma levels of H-FABP, NT-proBNP and HMGB1 in Dex and other sedative groups showed no difference, while both were significantly lower than the group of no sedative. Notably, Dex posttreatment deteriorated cardiac dysfunction, increasing mortality in septic rats with enhanced systemic and myocardial proinflammatory mediators, including TNF-α, IL-1β, IL-6 and VCAM-1. Mechanistical study by Langendorff-perfusion revealed that Dex directly acted on the heart, aggravating LPS-induced myocardial inflammation and dysfunction. These results suggest that Dex increases mortality and deteriorates myocardial dysfunction compared with other sedatives in septic patients defined by Sepsis 3.0, maybe partly through promoting proinflammatory response via directly acting on the heart.
Collapse
Affiliation(s)
- Liaoyuan Wang
- Medical Imaging Center, the First Affiliated Hospital of Jinan University, Guangzhou 510632, Guangdong, China
| | - Xiaomeng Dai
- Department of Pathology, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Lei Yu
- Department of Pathophysiology, Key Laboratory of State Administration of Traditional Chinese Medicine of the People's Republic of China, School of Medicine, Jinan University, Guangzhou 510632, Guangdong, China
| | - Hongmei Li
- Department of Pathophysiology, Key Laboratory of State Administration of Traditional Chinese Medicine of the People's Republic of China, School of Medicine, Jinan University, Guangzhou 510632, Guangdong, China
| | - Xue Zhang
- Department of Pathophysiology, Key Laboratory of State Administration of Traditional Chinese Medicine of the People's Republic of China, School of Medicine, Jinan University, Guangzhou 510632, Guangdong, China
| | - Qing Yu
- Department of Pathophysiology, Key Laboratory of State Administration of Traditional Chinese Medicine of the People's Republic of China, School of Medicine, Jinan University, Guangzhou 510632, Guangdong, China
| | - Xiuxiu Lv
- Department of Pathophysiology, Key Laboratory of State Administration of Traditional Chinese Medicine of the People's Republic of China, School of Medicine, Jinan University, Guangzhou 510632, Guangdong, China
| | - Yiyang Wang
- Department of Pathophysiology, Key Laboratory of State Administration of Traditional Chinese Medicine of the People's Republic of China, School of Medicine, Jinan University, Guangzhou 510632, Guangdong, China
| | - Shuixing Zhang
- Medical Imaging Center, the First Affiliated Hospital of Jinan University, Guangzhou 510632, Guangdong, China
| | - Guang Hao
- Department of Epidemiology and Biostatistics, School of Public Health, Guangdong Pharmaceutical University, Guangzhou, Guangdong, China.
| | - Huadong Wang
- Department of Pathophysiology, Key Laboratory of State Administration of Traditional Chinese Medicine of the People's Republic of China, School of Medicine, Jinan University, Guangzhou 510632, Guangdong, China.
| | - Zhigang Wang
- Department of Critical Care Medicine, the First Affiliated Hospital of Jinan University, Guangzhou 510632, Guangdong, China.
| |
Collapse
|
79
|
Biswal N, Harish R, Roshan M, Samudrala S, Jiao X, Pestell RG, Ashton AW. Role of GPCR Signaling in Anthracycline-Induced Cardiotoxicity. Cells 2025; 14:169. [PMID: 39936961 PMCID: PMC11817789 DOI: 10.3390/cells14030169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Revised: 11/27/2024] [Accepted: 11/27/2024] [Indexed: 02/13/2025] Open
Abstract
Anthracyclines are a class of chemotherapeutics commonly used to treat a range of cancers. Despite success in improving cancer survival rates, anthracyclines have dose-limiting cardiotoxicity that prevents more widespread clinical utility. Currently, the therapeutic options for these patients are limited to the iron-chelating agent dexrazoxane, the only FDA-approved drug for anthracycline cardiotoxicity. However, the clinical use of dexrazoxane has failed to replicate expectations from preclinical studies. A limited list of GPCRs have been identified as pathogenic in anthracycline-induced cardiotoxicity, including receptors (frizzled, adrenoreceptors, angiotensin II receptors) previously implicated in cardiac remodeling in other pathologies. The RNA sequencing of iPSC-derived cardiac myocytes from patients has increased our understanding of the pathogenic mechanisms driving cardiotoxicity. These data identified changes in the expression of novel GPCRs, heterotrimeric G proteins, and the regulatory pathways that govern downstream signaling. This review will capitalize on insights from these experiments to explain aspects of disease pathogenesis and cardiac remodeling. These data provide a cornucopia of possible unexplored potential pathways by which we can reduce the cardiotoxic side effects, without compromising the anti-cancer effects, of doxorubicin and provide new therapeutic options to improve the recovery and quality of life for patients undergoing chemotherapy.
Collapse
Affiliation(s)
- Nimish Biswal
- School of Medicine, Xavier University at Aruba, Oranjestad, Aruba (X.J.); (R.G.P.)
| | - Ritika Harish
- Pennsylvania Cancer and Regenerative Medicine Research Center, Baruch S. Blumberg Institute, Wynnewood, PA 19096, USA;
| | - Minahil Roshan
- School of Medicine, Xavier University at Aruba, Oranjestad, Aruba (X.J.); (R.G.P.)
| | - Sathvik Samudrala
- School of Medicine, Xavier University at Aruba, Oranjestad, Aruba (X.J.); (R.G.P.)
| | - Xuanmao Jiao
- School of Medicine, Xavier University at Aruba, Oranjestad, Aruba (X.J.); (R.G.P.)
- Pennsylvania Cancer and Regenerative Medicine Research Center, Baruch S. Blumberg Institute, Wynnewood, PA 19096, USA;
| | - Richard G. Pestell
- School of Medicine, Xavier University at Aruba, Oranjestad, Aruba (X.J.); (R.G.P.)
- Pennsylvania Cancer and Regenerative Medicine Research Center, Baruch S. Blumberg Institute, Wynnewood, PA 19096, USA;
- The Wistar Institute, Philadelphia, PA 19104, USA
| | - Anthony W. Ashton
- School of Medicine, Xavier University at Aruba, Oranjestad, Aruba (X.J.); (R.G.P.)
- Pennsylvania Cancer and Regenerative Medicine Research Center, Baruch S. Blumberg Institute, Wynnewood, PA 19096, USA;
- Division of Perinatal Research, Kolling Institute of Medical Research, University of Sydney, St Leonards, NSW 2065, Australia
- Division of Cardiovascular Medicine, Lankenau Institute for Medical Research, Wynnewood, PA 19096, USA
| |
Collapse
|
80
|
Rampanelli E, Romp N, Troise AD, Ananthasabesan J, Wu H, Gül IS, De Pascale S, Scaloni A, Bäckhed F, Fogliano V, Nieuwdorp M, Bui TPN. Gut bacterium Intestinimonas butyriciproducens improves host metabolic health: evidence from cohort and animal intervention studies. MICROBIOME 2025; 13:15. [PMID: 39833973 PMCID: PMC11744835 DOI: 10.1186/s40168-024-02002-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Accepted: 12/10/2024] [Indexed: 01/22/2025]
Abstract
BACKGROUND The human gut microbiome strongly influences host metabolism by fermenting dietary components into metabolites that signal to the host. Our previous work has shown that Intestinimonas butyriciproducens is a prevalent commensal bacterium with the unique ability to convert dietary fructoselysine to butyrate, a well-known signaling molecule with proven health benefits. Dietary fructoselysine is an abundant Amadori product formed in foods during thermal treatment and is part of foods rich in dietary advanced glycation end products which have been associated with cardiometabolic disease. It is therefore of interest to investigate the causal role of this bacterium and fructoselysine metabolism in metabolic disorders. RESULTS We assessed associations of I. butyriciproducens with metabolic risk biomarkers at both strain and functional levels using a human cohort characterized by fecal metagenomic analysis. We observed that the level of the bacterial strain as well as fructoselysine fermentation genes were negatively associated with BMI, triglycerides, HbA1c, and fasting insulin levels. We also investigated the fructoselysine degradation capacity within the Intestinimonas genus using a culture-dependent approach and found that I. butyriciproducens is a key player in the butyrogenic fructoselysine metabolism in the gut. To investigate the function of I. butyriciproducens in host metabolism, we used the diet-induced obesity mouse model to mimic the human metabolic syndrome. Oral supplementation with I. butyriciproducens counteracted body weight gain, hyperglycemia, and adiposity. In addition, within the inguinal white adipose tissue, bacterial administration reduced inflammation and promoted pathways involved in browning and insulin signaling. The observed effects may be partly attributable to the formation of the short-chain fatty acids butyrate from dietary fructoselysine, as butyrate plasma and cecal levels were significantly increased by the bacterial strain, thereby contributing to the systemic effects of the bacterial treatment. CONCLUSIONS I. butyriciproducens ameliorates host metabolism in the context of obesity and may therefore be a good candidate for new microbiota-therapeutic approaches to prevent or treat metabolic diseases. Video Abstract.
Collapse
Affiliation(s)
- Elena Rampanelli
- Department of Experimental Vascular Medicine, Amsterdam UMC, Amsterdam, the Netherlands
- Amsterdam Institute for Immunology and Infectious Diseases, Amsterdam, the Netherlands
| | - Nadia Romp
- Department of Experimental Vascular Medicine, Amsterdam UMC, Amsterdam, the Netherlands
- Amsterdam Institute for Immunology and Infectious Diseases, Amsterdam, the Netherlands
| | - Antonio Dario Troise
- Proteomics, Metabolomics & Mass Spectrometry Laboratory, Institute for the Animal Production System in the Mediterranean Environment, National Research Council, 80055, Portici (Naples), Italy
| | | | - Hao Wu
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan Microbiome Center, and Human Phenome Institute, Fudan University, Shanghai, 200438, China
| | | | - Sabrina De Pascale
- Proteomics, Metabolomics & Mass Spectrometry Laboratory, Institute for the Animal Production System in the Mediterranean Environment, National Research Council, 80055, Portici (Naples), Italy
| | - Andrea Scaloni
- Proteomics, Metabolomics & Mass Spectrometry Laboratory, Institute for the Animal Production System in the Mediterranean Environment, National Research Council, 80055, Portici (Naples), Italy
| | - Fredrik Bäckhed
- The Wallenberg Laboratory, Department of Molecular and Clinical Medicine, Sahlgrenska Academy, University of Gothenburg, Bruna Stråket 16, 41345, Gothenburg, Sweden
| | - Vincenzo Fogliano
- Department of Food Quality and Design, Wageningen University, Wageningen, the Netherlands
| | - Max Nieuwdorp
- Department of Experimental Vascular Medicine, Amsterdam UMC, Amsterdam, the Netherlands
| | - Thi Phuong Nam Bui
- Department of Experimental Vascular Medicine, Amsterdam UMC, Amsterdam, the Netherlands.
- Laboratory of Microbiology, Wageningen University, Wageningen, the Netherlands.
| |
Collapse
|
81
|
Zhou W, Fang J, Jia Q, Meng H, Liu F, Mao J. Transcription factor specificity protein (SP) family in renal physiology and diseases. PeerJ 2025; 13:e18820. [PMID: 39850832 PMCID: PMC11756367 DOI: 10.7717/peerj.18820] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Accepted: 12/15/2024] [Indexed: 01/25/2025] Open
Abstract
Dysregulated specificity proteins (SPs), members of the C2H2 zinc-finger family, are crucial transcription factors (TFs) with implications for renal physiology and diseases. This comprehensive review focuses on the role of SP family members, particularly SP1 and SP3, in renal physiology and pathology. A detailed analysis of their expression and cellular localization in the healthy human kidney is presented, highlighting their involvement in fatty acid metabolism, electrolyte regulation, and the synthesis of important molecules. The review also delves into the diverse roles of SPs in various renal diseases, including renal ischemia/reperfusion injury, diabetic nephropathy, renal interstitial fibrosis, and lupus nephritis, elucidating their molecular mechanisms and potential as therapeutic targets. The review further discusses pharmacological modulation of SPs and its implications for treatment. Our findings provide a comprehensive understanding of SPs in renal health and disease, offering new avenues for targeted therapeutic interventions and precision medicine in nephrology.
Collapse
Affiliation(s)
- Wei Zhou
- Department of Nephrology, Children’s Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, Zhejiang, China
| | - Jiaxi Fang
- Department of Ultrasound, Taizhou Central Hospital, Taizhou, Zhejiang, China
| | - Qingqing Jia
- Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, Shanghai, China
| | - Hanyan Meng
- Department of Nephrology, Children’s Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, Zhejiang, China
| | - Fei Liu
- Department of Nephrology, Children’s Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, Zhejiang, China
| | - Jianhua Mao
- Department of Nephrology, Children’s Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, Zhejiang, China
| |
Collapse
|
82
|
Jacobs B, Derese I, Derde S, Vander Perre S, Pauwels L, Van den Berghe G, Gunst J, Langouche L. A murine model of acute and prolonged abdominal sepsis, supported by intensive care, reveals time-dependent metabolic alterations in the heart. Intensive Care Med Exp 2025; 13:6. [PMID: 39821755 PMCID: PMC11748666 DOI: 10.1186/s40635-025-00715-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Accepted: 01/09/2025] [Indexed: 01/19/2025] Open
Abstract
BACKGROUND Sepsis-induced cardiomyopathy (SICM) often occurs in the acute phase of sepsis and is associated with increased mortality due to cardiac dysfunction. The pathogenesis remains poorly understood, and no specific treatments are available. Although SICM is considered reversible, emerging evidence suggests potential long-term sequelae. We hypothesized that metabolic and inflammatory cardiac changes, previously observed in acute sepsis as potential drivers of SICM, partially persist in prolonged sepsis. METHODS In 24-week-old C57BL/6J mice, sepsis was induced by cecal ligation and puncture, followed by intravenous fluid resuscitation, subcutaneous analgesics and antibiotics, and, in the prolonged phase, by parenteral nutrition. Mice were killed after 5 days of sepsis (prolonged sepsis, n = 15). For comparison, we included acutely septic mice killed at 30 h (acute sepsis, n = 15) and healthy controls animals (HC, n = 15). Cardiac tissue was collected for assessment of inflammatory and metabolic markers through gene expression, metabolomic analysis and histological assessment. RESULTS In prolonged sepsis, cardiac expression of IL-1β and IL-6 and macrophage infiltration remained upregulated (p ≤ 0.05). In contrast, tissue levels of Krebs cycle intermediates and adenosine phosphates were normal, whereas NADPH levels were low in prolonged sepsis (p ≤ 0.05). Gene expression of fatty acid transporters and of the glucose transporter Slc2a1 was upregulated in prolonged sepsis (p ≤ 0.01). Lipid staining and glycogen content were elevated in prolonged sepsis together with increased gene expression of enzymes responsible for lipogenesis and glycogen synthesis (p ≤ 0.05). Intermediate glycolytic metabolites (hexose-phosphates, GADP, DHAP) were elevated (p ≤ 0.05), but gene expression of several enzymes for glycolysis and mitochondrial oxidation of pyruvate, fatty-acyl-CoA and ketone bodies to acetyl-CoA were suppressed in prolonged sepsis (p ≤ 0.05). Key metabolic transcription factors PPARα and PGC-1α were downregulated in acute, but upregulated in prolonged, sepsis (p ≤ 0.05 for both). Ketone body concentrations were normal but ketolytic enzymes remained suppressed (p ≤ 0.05). Amino acid metabolism showed mild, mixed changes. CONCLUSIONS Our results suggest myocardial lipid and glycogen accumulation and suppressed mitochondrial oxidation, with a functionally intact Krebs cycle, in the prolonged phase of sepsis, together with ongoing myocardial inflammation. Whether these alterations have functional consequences and predispose to long-term sequelae of SICM needs further research.
Collapse
Affiliation(s)
- Bart Jacobs
- Clinical Division and Laboratory of Intensive Care Medicine, Department of Cellular and Molecular Medicine, KU Leuven, Herestraat 49, O&N1 Box 503, 3000, Louvain, Belgium
| | - Inge Derese
- Clinical Division and Laboratory of Intensive Care Medicine, Department of Cellular and Molecular Medicine, KU Leuven, Herestraat 49, O&N1 Box 503, 3000, Louvain, Belgium
| | - Sarah Derde
- Clinical Division and Laboratory of Intensive Care Medicine, Department of Cellular and Molecular Medicine, KU Leuven, Herestraat 49, O&N1 Box 503, 3000, Louvain, Belgium
| | - Sarah Vander Perre
- Clinical Division and Laboratory of Intensive Care Medicine, Department of Cellular and Molecular Medicine, KU Leuven, Herestraat 49, O&N1 Box 503, 3000, Louvain, Belgium
| | - Lies Pauwels
- Clinical Division and Laboratory of Intensive Care Medicine, Department of Cellular and Molecular Medicine, KU Leuven, Herestraat 49, O&N1 Box 503, 3000, Louvain, Belgium
| | - Greet Van den Berghe
- Clinical Division and Laboratory of Intensive Care Medicine, Department of Cellular and Molecular Medicine, KU Leuven, Herestraat 49, O&N1 Box 503, 3000, Louvain, Belgium
| | - Jan Gunst
- Clinical Division and Laboratory of Intensive Care Medicine, Department of Cellular and Molecular Medicine, KU Leuven, Herestraat 49, O&N1 Box 503, 3000, Louvain, Belgium
| | - Lies Langouche
- Clinical Division and Laboratory of Intensive Care Medicine, Department of Cellular and Molecular Medicine, KU Leuven, Herestraat 49, O&N1 Box 503, 3000, Louvain, Belgium.
| |
Collapse
|
83
|
Zhao M, Hou Y, Yuan M, Ma S, Yue Y. Clinical efficacy and hemodynamic effects of levosimendan in cardiac surgery patients after surgery. J Cardiothorac Surg 2025; 20:43. [PMID: 39773754 PMCID: PMC11705681 DOI: 10.1186/s13019-024-03316-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2024] [Accepted: 12/26/2024] [Indexed: 01/11/2025] Open
Abstract
OBJECTIVE To investigate the therapeutic effect of levosimendan on hemodynamics in patients undergoing major cardiac surgery and presenting with acute postoperative heart failure. METHODS The subjects of the study were 160 patients with severe cardiac conditions who underwent surgery and had acute heart failure. Eighty cases each were assigned to the research and control groups using a random number table. Document the general patient data for each of the two groups; compare the clinical outcomes of the two groups. The hemodynamic states of the two groups were compared both before and after therapy. 48 h after surgery, echocardiography was performed in both groups to determine cardiac function. 48 h after surgery, N-terminal pro-brain B-type natriuretic peptide (NT-Pro-BNP) levels were compared between the two groups. RESULTS The overall effective rate was significantly higher in the research group (92.5%) compared to the control group (76.25%, P < 0.05). Post-treatment, the research group demonstrated a significant reduction in CVP (9.25 ± 2.11 cmH2O vs. 11.36 ± 3.08 cmH2O, P < 0.001), heart rate (100.30 ± 8.69 bpm vs. 105.74 ± 7.69 bpm, P < 0.001), and lactic acid levels (1.68 ± 0.59 mmol/L vs. 2.69 ± 0.55 mmol/L, P < 0.001). The research group also showed improvements in SBP (117.23 ± 8.74 mmHg vs. 113.25 ± 7.55 mmHg, P = 0.002) and urine output (4.21 ± 1.76 mL/kg/h vs. 3.65 ± 1.23 mL/kg/h, P = 0.021). Cardiac function indicators 48 h after surgery indicated a higher LVEF (55.21 ± 8.04% vs. 47.18 ± 6.60%, P < 0.001) and lower LVEDVi and LVESVi in the research group (P < 0.001 for both). NT-Pro-BNP levels were significantly lower in the research group (6010.19 ± 1208.52 pg/mL vs. 9663.21 ± 2391.34 pg/mL, P < 0.001). The incidence of complications was lower in the research group (5% vs. 22.5%, P = 0.001). CONCLUSION Cardiac surgery patients are prone to complications with acute heart failure after surgery. Treatment with levosimendan can significantly improve clinical efficacy and reduce complications. It can also effectively improve patients' cardiac function and promote hemodynamic stability.
Collapse
Affiliation(s)
- Meiling Zhao
- Department of Critical Care Medicine, Zibo Central Hospital, Zibo, 255000, China
| | - Yunfeng Hou
- Department of Critical Care Medicine, The First Affiliated Hospital of Shandong First Medical University, JiNan, 250014, China
| | - Meng Yuan
- Department of Critical Care Medicine, Zibo Central Hospital, Zibo, 255000, China
| | - Shuang Ma
- Department of Critical Care Medicine, Zibo Central Hospital, Zibo, 255000, China
| | - Yifeng Yue
- Department of Anesthesiology, Zibo Central Hospital, No.10 Shanghai Road, Zhangdian District, Zibo City, 255000, Shandong Province, China.
| |
Collapse
|
84
|
Laradhi AO, Shan Y, Allawy ME. Psychological wellbeing and treatment adherence among cardio-renal syndrome patients in Yemen: a cross section study. Front Med (Lausanne) 2025; 11:1439704. [PMID: 39839638 PMCID: PMC11747712 DOI: 10.3389/fmed.2024.1439704] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Accepted: 12/19/2024] [Indexed: 01/23/2025] Open
Abstract
Background Anxiety and depression are associated with adverse outcomes in cardiorenal syndrome patients undergoing hemodialysis, including decreased quality of life, poorer clinical parameters, and lower treatment adherence. Objective This study aimed to examine the level of psychological wellbeing and its relationship with treatment adherence among dialysis patients with cardiorenal syndrome. Methods This cross-sectional descriptive study was conducted between February and May 2021 on convenience sampling of 100 patients in two dialysis centers in Hadhramout, Yemen. Patients' depression and anxiety levels were assessed using the Hospital Anxiety and Depression Scale (HADS)-Arabic version, and patient treatment adherence was assessed using the Treatment Adherence Questionnaire (TAQ). Descriptive statistics, Pearson's correlation analysis, and multiple linear regression analyzes were performed to analyze data with a significance level set at p < 0.05. Results The mean age ± standard deviation of participants was 53.46 ± 14.24 years. Most (90%) of patients had moderate to high levels of anxiety and depression. Most of the patients (87%) had a low level of treatment adherence. The findings revealed that psychological wellbeing is significantly association with treatment adherence t = 2.577 (95% CI 0.029, 0.225), p = 0.011. Conclusion Anxiety and depression symptoms occurred more frequently among dialysis patients with cardiorenal syndrome, and there was a significant association between psychological wellbeing and treatment adherence. Our findings suggest that nurse managers should take into account that adding psychotherapies into the present cardiorenal syndrome treatment programs would improve patients' clinical and psychological parameters and, consequently, their clinical outcomes while taking patient heterogeneity and resource limitations into consideration.
Collapse
Affiliation(s)
- Adel Omar Laradhi
- School of Nursing and Health, Zhengzhou University, Zhengzhou, China
- College of Nursing, University of Hail, Hail, Saudi Arabia
| | - Yan Shan
- School of Nursing and Health, Zhengzhou University, Zhengzhou, China
| | - Mohamed Elsayed Allawy
- Department of Nursing Sciences, College of Applied Medical Sciences, Prince Sattam bin Abdulaziz University, Wadi Alddawasir, Saudi Arabia
- Medical- Surgical Nursing Department, Faculty of Nursing Suez Canal University, Ismailia, Egypt
| |
Collapse
|
85
|
Bibi F, Aslam A, Naseem H, Khurshid H, Haider A, Ashraf H, Rafaqat S. Adipokines Pathogenesis in Heart Failure. CARDIOVASCULAR INNOVATIONS AND APPLICATIONS 2025; 10. [DOI: 10.15212/cvia.2024.0065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/02/2025] Open
Abstract
Many studies have reported that obesity causes heart failure (HF) pathogenesis. The elevated circulating levels of angiopoietin-like protein 2 (ANGPTL2) observed in patients with HF suggest potential links among elevated ANGPTL2 levels, metabolic disturbances, and inflammation. C1q/TNF-related protein 3 and C1q/TNF-related protein 9 are diminished in patients with HF with reduced ejection fraction, in proportion to disease severity, and are associated with elevated morbidity and mortality. In addition, fibroblast growth factor 21 (FGF21) has been suggested to be involved in the pathophysiology of diastolic HF. Further studies are necessary to determine whether FGF21 plays a causal role in HF, and whether measuring circulating FGF21 might effectively improve HF prediction, diagnosis, and prognosis. Osteopontin has also been reported to be upregulated in patients with HF and therefore might potentially serve as a novel prognostic biomarker in patients with chronic HF. A considerable number of patients with community-acquired HF have elevated tumor necrosis factor-alpha, which is associated with significantly diminished survival and significantly elevated risk of HF.
Collapse
|
86
|
Theofilis P, Oikonomou E, Vlachakis PK, Karakasis P, Dimitriadis K, Sagris M, Pamporis K, Drakopoulou M, Siasos G, Tsioufis K, Tousoulis D. Sodium-Glucose Cotransporter 2 Inhibitors and Changes in Epicardial Adipose Tissue: A Systematic Literature Review And Meta-Analysis. Curr Vasc Pharmacol 2025; 23:204-212. [PMID: 39819408 DOI: 10.2174/0115701611330060241204062248] [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: 05/30/2024] [Revised: 11/02/2024] [Accepted: 11/08/2024] [Indexed: 01/19/2025]
Abstract
INTRODUCTION Sodium-glucose cotransporter 2 (SGLT2) inhibitors have emerged as a groundbreaking class of antidiabetic medications renowned for their glucose-lowering effects and cardiovascular benefits. Recent studies have suggested that SGLT2 inhibitors may extend their influence beyond glycemic control to impact adipose tissue physiology, particularly within the epicardial adipose depot. Epicardial adipose tissue (EAT), an actively secretory organ surrounding the heart, has been implicated in the modulation of cardiovascular risk. AIMS This systematic review and meta-analysis aims to systematically review and synthesize existing literature on the effects of SGLT2 inhibitors on EAT. METHODS We performed a literature search for studies assessing the changes in epicardial adipose tissue volume/thickness before and after treatment with an SGLT2 inhibitor. We excluded reviews, editorials, case reports/case series, experimental studies, and studies that did not use SGLT2 inhibitors as the intervention. The main outcome of interest was the change in EAT volume/thickness at follow-up. RESULTS The literature search yielded 72 results. After the application of the exclusion criteria, a total of 11 studies were selected for data extraction and inclusion in the meta-analysis. A mean of 6.57ml decreased EAT volume, and EAT thickness was reduced by a mean of 1.55mm. We detected that treatment with an SGLT2 inhibitor was associated with decreased EAT volume/thickness compared to the control group (SMD -1.79, 95% CI -2.91 to -0.66, p<0.01). There was substantial betweenstudy heterogeneity (I2: 94%, p<0.001). Results remained robust even after the exclusion of any single study. Subgroup analysis revealed a significantly greater effect size in randomized studies. Funnel plot inspection and Egger's regression test did not indicate the presence of publication bias. CONCLUSION This meta-analysis suggests that SGLT2 inhibitors use is associated with a reduction in EAT volume/thickness, posing as a potential mechanism of their beneficial effects in heart failure (HF) outcomes.
Collapse
Affiliation(s)
- Panagiotis Theofilis
- 1st Department of Cardiology, "Hippokration" General Hospital, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Evangelos Oikonomou
- 3rd Department of Cardiology, Sotiria General Hospital, Athens, 11527, Greece
| | - Panayotis K Vlachakis
- 1st Department of Cardiology, "Hippokration" General Hospital, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Paschalis Karakasis
- 2nd Department of Cardiology, Ippokrateio General Hospital of Thessaloniki, Thessaloniki, 54642, Greece
| | - Kyriakos Dimitriadis
- 1st Department of Cardiology, "Hippokration" General Hospital, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Marios Sagris
- 1st Department of Cardiology, "Hippokration" General Hospital, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Konstantinos Pamporis
- 1st Department of Cardiology, "Hippokration" General Hospital, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Maria Drakopoulou
- 1st Department of Cardiology, "Hippokration" General Hospital, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Gerasimos Siasos
- 3rd Department of Cardiology, Sotiria General Hospital, Athens, 11527, Greece
| | - Konstantinos Tsioufis
- 1st Department of Cardiology, "Hippokration" General Hospital, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Dimitris Tousoulis
- 1st Department of Cardiology, "Hippokration" General Hospital, National and Kapodistrian University of Athens, 11527 Athens, Greece
| |
Collapse
|
87
|
Dimitriadis K, Pitsiori D, Alexiou P, Pyrpyris N, Sakalidis A, Beneki E, Iliakis P, Tatakis F, Theofilis P, Tsioufis P, Konstantinidis D, Aggeli K, Tsioufis K. Modulating Sympathetic Nervous System With the Use of SGLT2 Inhibitors: Where There Is Smoke, There Is Fire? J Cardiovasc Pharmacol 2025; 85:12-20. [PMID: 39436317 DOI: 10.1097/fjc.0000000000001644] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Accepted: 09/24/2024] [Indexed: 10/23/2024]
Abstract
Heart failure (HF) has become even more prevalent in recent years, because of improved diagnostics and an increase in the risk factors predisposing to its pathology. Sodium-glucose cotransporter 2 inhibitors (SGLT2i) emerged as one of the key pharmacotherapy options for both reduced and preserved ejection fraction, providing cardio- and renoprotection and improving mortality and cardiovascular (CV) outcomes. The pleiotropism of SGLT2i has led to multiple efforts to understand their distinct pathophysiologic interactions with various pathways, including microcirculation, endothelial dysfunction, and inflammation. More recently, the role of SGLT2i on the sympathetic nervous system (SNS) is starting to be recognized, especially because observations of retained or reduced heart rate despite volume contraction have been noted by investigators in the large clinical trials testing the safety and efficacy of these agents. Both preclinical and clinical studies have been performed, with conflicting results. Interestingly, in both settings, although there are indications of SNS modulation by SGLT2i, other studies contradict such findings, without showing, however, worsening of the autonomic homeostasis. Given the importance of neuromodulation in HF, in both pharmacologic and interventional therapies, in this review, we aim to describe the role of SNS in CV disease, focusing on HF, analyze preclinical and clinical data regarding the efficacy of SGLT2i in modulating autonomic dysfunction by examining various markers of SNS activation, and provide the most plausible theoretical backgrounds on the mechanism of benefit of SNS from the inhibition of SGLT2 receptors.
Collapse
Affiliation(s)
- Kyriakos Dimitriadis
- First Department of Cardiology, School of Medicine, National and Kapodistrian University of Athens, Hippokration General Hospital, Athens, Greece
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
88
|
Mosa AK, Majeed S, Ghafil FA, Hadi NR. Potential cardioprotective effect of trimetazidine in mice model of endotoxemia: role of AMPK-Nrf2. WIADOMOSCI LEKARSKIE (WARSAW, POLAND : 1960) 2025; 78:35-44. [PMID: 40023854 DOI: 10.36740/wlek/190820] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/04/2025]
Abstract
OBJECTIVE Aim: To clarify the potential cardioprotective effect of Trimetazidine against experimentally sepsis-caused endotoxic cardiac injury damage in mice. PATIENTS AND METHODS Materials and Methods: 24 Mice were divided into four groups (n=6): Sham group, CLP group DMSO group, trimetazidine-treated group 50 mg/kg IP, 1hr before CLP, then the animals were sacrificed 24 hr after CLP and tissue sample was taken for measurement of TNF-α, TNF-αr1, IL-1β, HO-1, MPO, caspase-11, F2-isoprostane and serum troponin by ELISA and gene expression of AMPK-Nrf2 by qpcr and histopathological study. RESULTS Results: trimetazidine treated group showed significant changes as compared with clp group regarding TNF-α, TNF-αr1, IL-1β, HO-1, MPO, CASPASE-11, F2-ISOPROSTANE as well as affect tissue mRNA expression of AMPK-Nrf2 genes p<0.05. CONCLUSION Conclusions: We evaluate that Trimetazidine has cardio protective effects due to its anti-inflammatory and anti-oxidative action. Also, trimetazidine showed a cardio-protective effect as they affect tissue mRNA expression of AMPK-Nrf2 genes.
Collapse
Affiliation(s)
- Alaa Kadhum Mosa
- PHARMACOLOGY AND THERAPEUTIC DEPARTMENT, FACULTY OF MEDICINE, UNIVERSITY OF KUFA, KUFA, IRAQ
| | - Sahar Majeed
- PHARMACOLOGY AND THERAPEUTIC DEPARTMENT, FACULTY OF MEDICINE, UNIVERSITY OF KUFA, KUFA, IRAQ
| | | | - Najah Rayish Hadi
- PHARMACOLOGY AND THERAPEUTIC DEPARTMENT, FACULTY OF MEDICINE, UNIVERSITY OF KUFA, KUFA, IRAQ
| |
Collapse
|
89
|
Saxena J, Agarwal G, Das S, Kumar A, Thakkar K, Kaushik S, Srivatsava VK, Siddiqui AJ, Jyoti A. Immunopharmacological Insights into Cordyceps spp.: Harnessing Therapeutic Potential for Sepsis. Curr Pharm Des 2025; 31:823-842. [PMID: 39694962 DOI: 10.2174/0113816128326301240920040036] [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: 04/26/2024] [Revised: 07/29/2024] [Accepted: 08/06/2024] [Indexed: 12/20/2024]
Abstract
Cordyceps spp. (CS), a well-known medicinal mushroom that belongs to Tibetan medicine and is predominantly found in the high altitudes in the Himalayas. CS is a rich reservoir of various bioactive substances including nucleosides, sterols flavonoids, peptides, and phenolic compounds. The bioactive compounds and CS extract have antibacterial, antioxidant, immunomodulatory, and inflammatory properties in addition to organ protection properties across a range of disease states. The study aimed to review the potential of CS, a medicinal mushroom, as a treatment for sepsis. While current sepsis drugs have side effects, CS shows promise due to its anti-inflammatory, antioxidant, and antibacterial properties. We have performed an extensive literature search based on published original and review articles in Scopus and PubMed. The keywords used were Cordyceps, sepsis, and inflammation. Studies indicate that CS extract and bioactive compounds target free radicals including oxidative as well as nitrosative stress, lower inflammation, and modulate the immune system, all of which are critical components in sepsis. The brain, liver, kidneys, lungs, and heart are among the organs that CS extracts may be able to shield against harm during sepsis. Traditional remedies with anti-inflammatory and protective qualities, such as Cordyceps mushrooms, are promising in sepsis. However, more research including clinical trials is required to validate the usefulness of CS metabolites in terms of organ protection and fight infections in sepsis.
Collapse
Affiliation(s)
- Juhi Saxena
- Department of Biotechnology, Parul Institute of Technology, Parul University, Vadodara, Gujarat, India
| | - Gaurang Agarwal
- Department of Life Science, Parul Institute of Applied Science, Parul University, Vadodara, Gujarat, India
| | - Sarvjeet Das
- Department of Life Science, Parul Institute of Applied Science, Parul University, Vadodara, Gujarat, India
| | - Anshu Kumar
- Department of Life Science, Parul Institute of Applied Science, Parul University, Vadodara, Gujarat, India
| | - Krish Thakkar
- Department of Biotechnology, Parul Institute of Technology, Parul University, Vadodara, Gujarat, India
| | - Sanket Kaushik
- Amity Institute of Biotechnology, Amity University, Jaipur, Rajasthan, India
| | | | - Arif Jamal Siddiqui
- Department of Biology, College of Science, University of Ha'il, Ha'il, P.O. Box 2440, Saudi Arabia
| | - Anupam Jyoti
- Department of Life Science, Parul Institute of Applied Science, Parul University, Vadodara, Gujarat, India
| |
Collapse
|
90
|
Zhang ZB, Cheng YW, Xu L, Li JQ, Pan X, Zhu M, Chen XH, Sun AJ, Lin JR, Gao PJ. Activation of β3-adrenergic receptor by mirabegron prevents aortic dissection/aneurysm by promoting lymphangiogenesis in perivascular adipose tissue. Cardiovasc Res 2024; 120:2307-2319. [PMID: 39288197 DOI: 10.1093/cvr/cvae213] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Revised: 05/06/2024] [Accepted: 07/23/2024] [Indexed: 09/19/2024] Open
Abstract
AIMS β3-Adrenergic receptor (β3-AR) is essential for cardiovascular homoeostasis through regulating adipose tissue function. Perivascular adipose tissue (PVAT) has been implicated in the pathogenesis of aortic dissection and aneurysm (AD/AA). Here, we aim to investigate β3-AR activation-mediated PVAT function in AD/AA. METHODS AND RESULTS Aortas from patients with thoracic aortic dissection (TAD) were collected to detect β3-AR expression in PVAT. ApoE-/- and β-aminopropionitrile monofumarate (BAPN)-treated C57BL/6 mice were induced with Angiotensin II (AngII) to simulate AD/AA and subsequently received either placebo or mirabegron, a β3-AR agonist. The results demonstrated an up-regulation of β3-AR in PVAT of TAD patients and AD/AA mice. Moreover, activation of β3-AR by mirabegron significantly prevented AngII-induced AD/AA formation in mice. RNA-sequencing analysis of adipocytes from PVAT revealed a notable increase of the lymphangiogenic factor, vascular endothelial growth factor C (VEGF-C), in mirabegron-treated mice. Consistently, enhanced lymphangiogenesis was found in PVAT with mirabegron treatment. Mechanistically, the number of CD4+/CD8+ T cells and CD11c+ cells was reduced in PVAT but increased in adjacent draining lymph nodes of mirabegron-treated mice, indicating the improved draining and clearance of inflammatory cells in PVAT by lymphangiogenesis. Importantly, adipocyte-specific VEGF-C knockdown by the adeno-associated virus system restrained lymphangiogenesis and exacerbated inflammatory cell infiltration in PVAT, which ultimately abolished the protection of mirabegron on AD/AA. In addition, the conditional medium derived from mirabegron-treated adipocytes activated the proliferation and tube formation of LECs, which was abrogated by the silencing of VEGF-C in adipocytes. CONCLUSION Our findings illustrated the therapeutic potential of β3-AR activation by mirabegron on AD/AA, which promoted lymphangiogenesis by increasing adipocyte-derived VEGF-C and, therefore, ameliorated PVAT inflammation.
Collapse
MESH Headings
- Animals
- Aortic Dissection/metabolism
- Aortic Dissection/pathology
- Aortic Dissection/prevention & control
- Aortic Dissection/chemically induced
- Aortic Dissection/physiopathology
- Aortic Dissection/genetics
- Lymphangiogenesis/drug effects
- Adrenergic beta-3 Receptor Agonists/pharmacology
- Mice, Inbred C57BL
- Receptors, Adrenergic, beta-3/metabolism
- Receptors, Adrenergic, beta-3/genetics
- Acetanilides/pharmacology
- Vascular Endothelial Growth Factor C/metabolism
- Vascular Endothelial Growth Factor C/genetics
- Humans
- Male
- Disease Models, Animal
- Adipose Tissue/metabolism
- Adipose Tissue/drug effects
- Adipose Tissue/pathology
- Aortic Aneurysm, Thoracic/metabolism
- Aortic Aneurysm, Thoracic/pathology
- Aortic Aneurysm, Thoracic/prevention & control
- Aortic Aneurysm, Thoracic/physiopathology
- Aortic Aneurysm, Thoracic/genetics
- Aortic Aneurysm, Thoracic/chemically induced
- Aortic Aneurysm, Thoracic/drug therapy
- Mice, Knockout, ApoE
- Thiazoles/pharmacology
- Signal Transduction
- Aorta, Thoracic/metabolism
- Aorta, Thoracic/pathology
- Aorta, Thoracic/physiopathology
- Aorta, Thoracic/drug effects
- Angiotensin II
- Adipocytes/metabolism
- Adipocytes/pathology
- Adipocytes/drug effects
- Cells, Cultured
- Mice
- Lymphatic Vessels/metabolism
- Lymphatic Vessels/drug effects
- Lymphatic Vessels/pathology
- Lymphatic Vessels/physiopathology
- Female
- Middle Aged
Collapse
Affiliation(s)
- Ze-Bei Zhang
- Department of Cardiovascular Medicine, State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, Shanghai Institute of Hypertension, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin, 2 Road, Shanghai 200025, China
| | - Yu-Wen Cheng
- Department of Cardiovascular Medicine, State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, Shanghai Institute of Hypertension, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin, 2 Road, Shanghai 200025, China
- Department of Cardiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lian Xu
- Department of Cardiovascular Medicine, State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, Shanghai Institute of Hypertension, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin, 2 Road, Shanghai 200025, China
| | - Jia-Qi Li
- Department of Cardiovascular Medicine, State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, Shanghai Institute of Hypertension, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin, 2 Road, Shanghai 200025, China
| | - Xin Pan
- Department of Gerontology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Min Zhu
- Department of Cardiovascular Medicine, State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, Shanghai Institute of Hypertension, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin, 2 Road, Shanghai 200025, China
| | - Xiao-Hui Chen
- Department of Cardiovascular Medicine, State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, Shanghai Institute of Hypertension, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin, 2 Road, Shanghai 200025, China
| | - Ai-Jun Sun
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai, China
| | - Jing-Rong Lin
- Department of Cardiovascular Medicine, State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, Shanghai Institute of Hypertension, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin, 2 Road, Shanghai 200025, China
| | - Ping-Jin Gao
- Department of Cardiovascular Medicine, State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, Shanghai Institute of Hypertension, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin, 2 Road, Shanghai 200025, China
| |
Collapse
|
91
|
Li J, Lu G, Deng H, Gao X, Zhai Y. Irbesartan May Ameliorate Ventricular Remodeling by Inhibiting CREB-Mediated Cardiac Aldosterone Synthesis in Rats with Myocardial Infarction. Int J Mol Sci 2024; 26:198. [PMID: 39796054 PMCID: PMC11719508 DOI: 10.3390/ijms26010198] [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: 11/04/2024] [Revised: 12/25/2024] [Accepted: 12/27/2024] [Indexed: 01/13/2025] Open
Abstract
Irbesartan improves ventricular remodeling (VR) following myocardial infarction (MI). This study investigates whether irbesartan attenuates VR by reducing aldosterone production in the heart and its underlying mechanisms. MI was induced in male Sprague-Dawley rats through coronary artery ligation. The MI rats were randomly assigned to two groups: one received a vehicle, and the other received 100 mg/kg/day of irbesartan for 5 weeks. Cardiac function and myocardial fibrosis were assessed using echocardiography and Masson's trichrome staining, respectively. The impact of angiotensin II (Ang II) stimulation on cardiac microvascular endothelial cells (CMECs) from commercial sources was determined using ELISA, real-time PCR, and Western blotting. Irbesartan reduced left ventricular mass index, collagen composition, and aldosterone levels while enhancing cardiac function in MI rats. In vitro, Ang II time-dependently stimulated aldosterone secretion and CYP11B2 mRNA expression in CMECs (p < 0.05). Additionally, Ang II significantly upregulated p-CREB protein levels. However, these effects were abrogated by irbesartan and partially attenuated by CaMK inhibitor KN93 (p < 0.05). In conclusion, our study demonstrated that improvement in VR by irbesartan coincided with reduced CREB phosphorylation in CMECs and reduced aldosterone synthesis in the non-infarcted tissue. These effects may be mediated by blocking the AT1 receptor.
Collapse
Affiliation(s)
- Jie Li
- Department of Cardiology, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510800, China; (J.L.); (G.L.); (H.D.)
- Key Laboratory on Assisted Circulation, Ministry of Health, Guangzhou 510800, China
| | - Guihua Lu
- Department of Cardiology, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510800, China; (J.L.); (G.L.); (H.D.)
- Key Laboratory on Assisted Circulation, Ministry of Health, Guangzhou 510800, China
| | - Haiwei Deng
- Department of Cardiology, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510800, China; (J.L.); (G.L.); (H.D.)
- Key Laboratory on Assisted Circulation, Ministry of Health, Guangzhou 510800, China
| | - Xiuren Gao
- Department of Cardiology, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510800, China; (J.L.); (G.L.); (H.D.)
- Key Laboratory on Assisted Circulation, Ministry of Health, Guangzhou 510800, China
| | - Yuansheng Zhai
- Department of Cardiology, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510800, China; (J.L.); (G.L.); (H.D.)
- Key Laboratory on Assisted Circulation, Ministry of Health, Guangzhou 510800, China
| |
Collapse
|
92
|
Song C, Yan Q, Ma Y, Li P, Yang Y, Wang Y, Li W, Wan X, Li Y, Zhu R, Liu H, Zhang Z. Modified Zuo Gui Wan Ameliorates Ovariectomy-Induced Osteoporosis in Rats by Regulating the SCFA-GPR41-p38MAPK Signaling Pathway. Drug Des Devel Ther 2024; 18:6359-6377. [PMID: 39741920 PMCID: PMC11687096 DOI: 10.2147/dddt.s482965] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Accepted: 12/14/2024] [Indexed: 01/03/2025] Open
Abstract
Objective Modified Zuo Gui Wan (MZGW) was a combination of Zuo Gui Wan and red yeast rice used for treating osteoporosis (OP), but its mechanism remains unclear. We aimed to validate the anti-OP effect of MZGW and explore its underlying mechanism. Methods An ovariectomy (OVX) rat model in vivo and a RANKL-induced osteoclasts (OCs) model in vitro were established. Key active ingredients in MZGW high dose (MZGW-H) group were detected by UPLC-MS/MS. Micro-CT scans and histomorphology analysis were performed in OVX rats. 16S rRNA gene sequencing was performed to investigate the relationship between the anti-OP effect of MZGW-H and intestinal flora. CCK-8 assay was applied to examine the optimal concentration of Modified Zuo Gui Wan drug serum (MZGW-DS) on osteoclasts. The qRT-PCR and Western blotting were utilized to explore the potential anti-OP pathway of MZGW, namely the SCFA-GPR41-p38MAPK signaling pathway. GPR41 was knocked down to further reverse to verify whether the pathway was the key pathway for MZGW-DS to exert its inhibitory effect on osteoclasts. Results The three main blood components, Ferulic acid, L-Ascorbic acid and Riboflavin, were examined mainly by UPLC-MS/MS. 16S rRNA gene sequencing showed that MZGW-H changed the metabolism of SCFAs. In vivo studies verified that MZGW-H ameliorated microstructure damage, improved histological changes and reduced TRAP, BALP, and BGP in OVX rats by regulating the SCFA-GPR41-p38MAPK signaling pathway. CCK-8 revealed that 5% MZGW-DS group was the most optimal concentration of MZGW-DS to inhibit osteoclast differentiation. In vitro, MZGW-DS was better than peripheral blood concentration of SCFAs in inhibiting osteoclasts. After the knockout of GPR41, MZGW-DS could not inhibit the expression of osteoclast-related protein (CTSK and NFATc1) via SCFA-GPR41-p38MAPK signaling pathway. Conclusion MZGW-H effectively ameliorates OVX-induced osteoporosis partially achieved by increasing SCFAs metabolism and modulating the SCFA-GPR41-p38MAPK signaling pathway.
Collapse
Affiliation(s)
- Changheng Song
- Institute of Basic Theory for Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing, People’s Republic of China
- Department of Endocrinology, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, People’s Republic of China
| | - Qiqi Yan
- Institute of Basic Theory for Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing, People’s Republic of China
| | - Yujie Ma
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, People’s Republic of China
| | - Pei Li
- Institute of Basic Theory for Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing, People’s Republic of China
| | - Ying Yang
- Institute of Basic Theory for Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing, People’s Republic of China
| | - Yuhan Wang
- Institute of Basic Theory for Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing, People’s Republic of China
| | - Wenjie Li
- Institute of Basic Theory for Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing, People’s Republic of China
| | - Xinyu Wan
- Institute of Basic Theory for Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing, People’s Republic of China
| | - Yubo Li
- Institute of Basic Theory for Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing, People’s Republic of China
| | - Ruyuan Zhu
- Institute of Basic Theory for Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing, People’s Republic of China
| | - Haixia Liu
- Institute of Basic Theory for Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing, People’s Republic of China
| | - Zhiguo Zhang
- Institute of Basic Theory for Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing, People’s Republic of China
| |
Collapse
|
93
|
Nádasy GL, Balla A, Dörnyei G, Hunyady L, Szekeres M. Direct Vascular Effects of Angiotensin II (A Systematic Short Review). Int J Mol Sci 2024; 26:113. [PMID: 39795971 PMCID: PMC11719566 DOI: 10.3390/ijms26010113] [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: 11/14/2024] [Revised: 12/19/2024] [Accepted: 12/22/2024] [Indexed: 01/13/2025] Open
Abstract
The octapeptide angiotensin II (Ang II) is a circulating hormone as well as a locally formed agonist synthesized by the angiotensin-converting enzyme (ACE) of endothelial cells. It forms a powerful mechanism to control the amount and pressure of body fluids. All main effects are directed to save body salt and water and ensure blood pressure under basic conditions and in emergencies. All blood vessels respond to stimulation by Ang II; the immediate response is smooth muscle contraction, increasing vascular resistance, and elevating blood pressure. Such effects are conveyed by type 1 angiotensin receptors (AT1Rs) located in the plasma membrane of both endothelial and vascular smooth muscle cells. AT1Rs are heterotrimeric G protein-coupled receptors (GPCRs), but their signal pathways are much more complicated than other GPCRs. In addition to Gq/11, the G12/13, JAK/STAT, Jnk, MAPK, and ERK 1/2, and arrestin-dependent and -independent pathways are activated because of the promiscuous attachment of different signal proteins to the intracellular G protein binding site and to the intracellular C terminal loop. Substantial changes in protein expression follow, including the intracellular inflammation signal protein NF-κB, endothelial contact proteins, cytokines, matrix metalloproteinases (MMPs), and type I protocollagen, eliciting the inflammatory transformation of endothelial and vascular smooth muscle cells and fibrosis. Ang II is an important contributor to vascular pathologies in hypertensive, atherosclerotic, and aneurysmal vascular wall remodeling. Such direct vascular effects are reviewed. In addition to reducing blood pressure, AT1R antagonists and ACE inhibitors have a beneficial effect on the vascular wall by inhibiting pathological wall remodeling.
Collapse
Affiliation(s)
- György L. Nádasy
- Department of Physiology, Faculty of Medicine, Semmelweis University, 37-47 Tűzoltó Street, 1094 Budapest, Hungary; (G.L.N.); (A.B.); (L.H.)
| | - András Balla
- Department of Physiology, Faculty of Medicine, Semmelweis University, 37-47 Tűzoltó Street, 1094 Budapest, Hungary; (G.L.N.); (A.B.); (L.H.)
- HUN-REN-SU Molecular Physiology Research Group, Hungarian Research Network, Semmelweis University, 1094 Budapest, Hungary
| | - Gabriella Dörnyei
- Department of Morphology and Physiology, Faculty of Health Sciences, Semmelweis University, 17 Vas Street, 1088 Budapest, Hungary;
| | - László Hunyady
- Department of Physiology, Faculty of Medicine, Semmelweis University, 37-47 Tűzoltó Street, 1094 Budapest, Hungary; (G.L.N.); (A.B.); (L.H.)
- Institute of Molecular Life Sciences, HUN-REN Research Centre for Natural Sciences, 2 Magyar Tudósok Körútja, 1117 Budapest, Hungary
| | - Mária Szekeres
- Department of Physiology, Faculty of Medicine, Semmelweis University, 37-47 Tűzoltó Street, 1094 Budapest, Hungary; (G.L.N.); (A.B.); (L.H.)
- Department of Morphology and Physiology, Faculty of Health Sciences, Semmelweis University, 17 Vas Street, 1088 Budapest, Hungary;
| |
Collapse
|
94
|
Goorani S, Zangene S, Imig JD. Hypertension: A Continuing Public Healthcare Issue. Int J Mol Sci 2024; 26:123. [PMID: 39795981 PMCID: PMC11720251 DOI: 10.3390/ijms26010123] [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: 11/06/2024] [Revised: 12/21/2024] [Accepted: 12/24/2024] [Indexed: 01/13/2025] Open
Abstract
Hypertension is a cardiovascular disease defined by an elevated systemic blood pressure. This devastating disease afflicts 30-40% of the adult population worldwide. The disease burden for hypertension is great, and it greatly increases the risk of cardiovascular morbidity and mortality. Unfortunately, there are a myriad of factors that result in an elevated blood pressure. These include genetic factors, a sedentary lifestyle, obesity, salt intake, aging, and stress. Although lifestyle modifications have had limited success, anti-hypertensive drugs have been moderately effective in lowering blood pressure. New approaches to control and treat hypertension include digital health tools and compounds that activate the angiotensin receptor type 2 (AT2), which can promote cardiovascular health. Nonetheless, research on hypertension and its management is vital for lessening the significant health and economic burden of this condition.
Collapse
Affiliation(s)
- Samaneh Goorani
- Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Somaye Zangene
- Faculty of Medicine, University of Tehran, Tehran 1416634793, Iran;
| | - John D. Imig
- Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| |
Collapse
|
95
|
Qin X, Zhang M, Chen S, Tang Y, Cui J, Ding G. Short-chain fatty acids in fetal development and metabolism. Trends Mol Med 2024:S1471-4914(24)00329-0. [PMID: 39694776 DOI: 10.1016/j.molmed.2024.11.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2024] [Revised: 11/18/2024] [Accepted: 11/25/2024] [Indexed: 12/20/2024]
Abstract
Short-chain fatty acids (SCFAs), primarily derived from gut microbiota, play a role in regulating fetal development; however, the mechanism remains unclear. Fetal SCFAs levels depends on maternal SCFAs transported via the placenta. Metabolic stress, particularly from diabetes and obesity, can disrupt maternal SCFAs levels, impairing fetal metabolic reprogramming. Dysregulated SCFAs may negatively impact the development of the fetal cardiovascular, nervous, and immune systems, potentially contributing to adverse outcomes in adulthood. This review focuses on recent advances regarding the role of maternal SCFAs in shaping the metabolic profile of offspring, especially in the context of various maternal metabolic disorders. Given that SCFAs may influence fetal development through the placenta-embryo axis, targeted SCFAs supplementation could be a promising strategy against developmental diseases associated with intrauterine risk factors.
Collapse
Affiliation(s)
- Xueyun Qin
- Obstetrics and Gynecology Hospital, Institute of Reproduction and Development, Fudan University, Shanghai, China; Shanghai Key Laboratory of Reproduction and Development, Shanghai, China
| | - Mo Zhang
- Obstetrics and Gynecology Hospital, Institute of Reproduction and Development, Fudan University, Shanghai, China; Shanghai Key Laboratory of Reproduction and Development, Shanghai, China
| | - Shiting Chen
- Obstetrics and Gynecology Hospital, Institute of Reproduction and Development, Fudan University, Shanghai, China; Shanghai Key Laboratory of Reproduction and Development, Shanghai, China
| | - Yunhui Tang
- Obstetrics and Gynecology Hospital, Institute of Reproduction and Development, Fudan University, Shanghai, China; Shanghai Key Laboratory of Reproduction and Development, Shanghai, China
| | - Jiajun Cui
- Obstetrics and Gynecology Hospital, Institute of Reproduction and Development, Fudan University, Shanghai, China; Shanghai Key Laboratory of Reproduction and Development, Shanghai, China
| | - Guolian Ding
- Obstetrics and Gynecology Hospital, Institute of Reproduction and Development, Fudan University, Shanghai, China; Shanghai Key Laboratory of Reproduction and Development, Shanghai, China; Research Units of Embryo Original Diseases, Chinese Academy of Medical Sciences, Shanghai, China.
| |
Collapse
|
96
|
Zhen WJ, Zhang Y, Fu WD, Fu XL, Yan X. Role of immune-related endoplasmic reticulum stress genes in sepsis-induced cardiomyopathy: Novel insights from bioinformatics analysis. PLoS One 2024; 19:e0315582. [PMID: 39671358 PMCID: PMC11642931 DOI: 10.1371/journal.pone.0315582] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2024] [Accepted: 11/27/2024] [Indexed: 12/15/2024] Open
Abstract
BACKGROUND The current study aims to elucidate the key molecular mechanisms linked to endoplasmic reticulum stress (ERS) in the pathogenesis of sepsis-induced cardiomyopathy (SIC) and offer innovative therapeutic targets for SIC. METHODS The study downloaded dataset GSE79962 from the Gene Expression Omnibus database and acquired the ERS-related gene set from GeneCards. It utilized weighted gene co-expression network analysis (WGCNA) and conducted differential expression analysis to identify key modules and genes associated with SIC. The SIC hub genes were determined by the intersection of WGCNA-based hubs, DEGs, and ERS-related genes, followed by protein-protein interaction (PPI) network construction. Enrichment analyses, encompassing GO, KEGG, GSEA, and GSVA, were performed to elucidate potential biological pathways. The CIBERSORT algorithm was employed to analyze immune infiltration patterns. Diagnostic and prognostic models were developed to assess the clinical significance of hub genes in SIC. Additionally, in vivo experiments were conducted to validate the expression of hub genes. RESULTS Differential analysis revealed 1031 differentially expressed genes (DEGs), while WGCNA identified a hub module with 1327 key genes. Subsequently, 13 hub genes were pinpointed by intersecting with ERS-related genes. NOX4, PDHB, SCP2, ACTC1, DLAT, EDN1, and NSDHL emerged as hub ERS-related genes through the protein-protein interaction network, with their diagnostic values confirmed via ROC curves. Diagnostic models incorporating five genes (NOX4, PDHB, ACTC1, DLAT, NSDHL) were validated using the LASSO algorithm, highlighting only the prognostic significance of serum PDHB levels in predicting the survival of septic patients. Additionally, decreased PDHB mRNA and protein expression levels were observed in the cardiac tissue of septic mice compared to control mice. CONCLUSIONS This study elucidated the interplay between metabolism and the immune microenvironment in SIC, providing fresh perspectives on the investigation of potential SIC pathogenesis. PDHB emerged as a significant biomarker of SIC, with implications on its progression through the regulation of ERS and metabolism.
Collapse
Affiliation(s)
- Wan-Jing Zhen
- Department of Anesthesiology, the Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian Province, China
| | - Yan Zhang
- Department of Anesthesiology, Zhuzhou Central Hospital (Zhuzhou Hospital Affiliated to Xiangya School of Medicine), Zhuzhou, Hunan Province, China
| | - Wei-Dong Fu
- Department of Anesthesiology, Zhuzhou Central Hospital (Zhuzhou Hospital Affiliated to Xiangya School of Medicine), Zhuzhou, Hunan Province, China
| | - Xiao-Lei Fu
- Department of Cardiovascular Medicine, Zhuzhou Central Hospital (Zhuzhou Hospital Affiliated to Xiangya School of Medicine), Zhuzhou, Hunan Province, China
| | - Xin Yan
- Department of Cardiovascular Medicine, Zhuzhou Central Hospital (Zhuzhou Hospital Affiliated to Xiangya School of Medicine), Zhuzhou, Hunan Province, China
| |
Collapse
|
97
|
Bauset C, Carda-Diéguez M, Cejudo-Garcés A, Buetas E, Seco-Cervera M, Macias-Ceja DC, Navarro-Vicente F, Esplugues JV, Calatayud S, Mira Á, Ortiz-Masiá D, Barrachina MD, Cosín-Roger J. A disturbed metabolite-GPCR axis is associated with microbial dysbiosis in IBD patients: Potential role of GPR109A in macrophages. Biochim Biophys Acta Mol Basis Dis 2024; 1870:167489. [PMID: 39233260 DOI: 10.1016/j.bbadis.2024.167489] [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: 01/31/2024] [Revised: 06/20/2024] [Accepted: 08/26/2024] [Indexed: 09/06/2024]
Abstract
Inflammatory Bowel Disease (IBD) is a chronic inflammatory disorder of the gastrointestinal tract characterized by disrupted immune function. Indeed, gut microbiota dysbiosis and metabolomic profile alterations, are hallmarks of IBD. In this scenario, metabolite-sensing G-protein coupled receptors (GPCRs), involved in several biological processes, have emerged as pivotal players in the pathophysiology of IBD. The aim of this study was to characterize the axis microbiota-metabolite-GPCR in intestinal surgical resections from IBD patients. Results showed that UC patients had a lower microbiota richness and bacterial load, with a higher proportion of the genus Cellulosimicrobium and a reduced proportion of Escherichia, whereas CD patients showed a decreased abundance of Enterococcus. Furthermore, metabolomic analysis revealed alterations in carboxylic acids, fatty acids, and amino acids in UC and CD samples. These patients also exhibited upregulated expression of most metabolite-sensing GPCRs analysed, which positively correlated with pro-inflammatory and pro-fibrotic markers. The role of GPR109A was studied in depth and increased expression of this receptor was detected in epithelial cells and cells from lamina propria, including CD68+ macrophages, in IBD patients. The treatment with β-hydroxybutyrate increased gene expression of GPR109A, CD86, IL1B and NOS2 in U937-derived macrophages. Besides, when GPR109A was transiently silenced, the mRNA expression and secretion of IL-1β, IL-6 and TNF-α were impaired in M1 macrophages. Finally, the secretome from siGPR109A M1 macrophages reduced the gene and protein expression of COL1A1 and COL3A1 in intestinal fibroblasts. A better understanding of metabolite-sensing GPCRs, such as GPR109A, could establish their potential as therapeutic targets for managing IBD.
Collapse
Affiliation(s)
- Cristina Bauset
- Departamento de Farmacología, Facultad de Medicina, Universidad de Valencia, Valencia, Spain
| | | | - Andrea Cejudo-Garcés
- Departamento de Farmacología, Facultad de Medicina, Universidad de Valencia, Valencia, Spain
| | - Elena Buetas
- Genomics & Health Department, FISABIO Foundation, Valencia, Spain
| | | | | | | | - Juan Vicente Esplugues
- Departamento de Farmacología, Facultad de Medicina, Universidad de Valencia, Valencia, Spain; CIBERehd (Centro de Investigaciones en Red Enfermedad Hepática y Digestiva), Madrid, Spain
| | - Sara Calatayud
- Departamento de Farmacología, Facultad de Medicina, Universidad de Valencia, Valencia, Spain; CIBERehd (Centro de Investigaciones en Red Enfermedad Hepática y Digestiva), Madrid, Spain
| | - Álex Mira
- Genomics & Health Department, FISABIO Foundation, Valencia, Spain; CIBER Center for Epidemiology and Public Health, Madrid, Spain
| | - Dolores Ortiz-Masiá
- CIBERehd (Centro de Investigaciones en Red Enfermedad Hepática y Digestiva), Madrid, Spain; Departamento de Medicina, Facultad de Medicina, Universidad de Valencia, Valencia, Spain.
| | - María Dolores Barrachina
- Departamento de Farmacología, Facultad de Medicina, Universidad de Valencia, Valencia, Spain; CIBERehd (Centro de Investigaciones en Red Enfermedad Hepática y Digestiva), Madrid, Spain.
| | - Jesús Cosín-Roger
- Departamento de Farmacología, Facultad de Medicina, Universidad de Valencia, Valencia, Spain; CIBERehd (Centro de Investigaciones en Red Enfermedad Hepática y Digestiva), Madrid, Spain
| |
Collapse
|
98
|
Lymperopoulos A, Koch WJ. Editorial Overview: Adrenergic signaling in cellular physiology and disease. CURRENT OPINION IN PHYSIOLOGY 2024; 42:100778. [DOI: 10.1016/j.cophys.2024.100778] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2025]
|
99
|
Dong ZK, Wang YF, Li WP, Jin WL. Neurobiology of cancer: Adrenergic signaling and drug repurposing. Pharmacol Ther 2024; 264:108750. [PMID: 39527999 DOI: 10.1016/j.pharmthera.2024.108750] [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: 07/04/2024] [Revised: 10/04/2024] [Accepted: 11/05/2024] [Indexed: 11/16/2024]
Abstract
Cancer neuroscience, as an emerging converging discipline, provides us with new perspectives on the interactions between the nervous system and cancer progression. As the sympathetic nervous system, in particular adrenergic signaling, plays an important role in the regulation of tumor activity at every hierarchical level of life, from the tumor cell to the tumor microenvironment, and to the tumor macroenvironment, it is highly desirable to dissect its effects. Considering the far-reaching implications of drug repurposing for antitumor drug development, such a large number of adrenergic receptor antagonists on the market has great potential as one of the means of antitumor therapy, either as primary or adjuvant therapy. Therefore, this review aims to summarize the impact of adrenergic signaling on cancer development and to assess the status and prospects of intervening in adrenergic signaling as a therapeutic tool against tumors.
Collapse
Affiliation(s)
- Zi-Kai Dong
- The First Clinical Medical College of Lanzhou University, Lanzhou 730000, PR China; Institute of Cancer Neuroscience, Medical Frontier Innovation Research Center, The First Hospital of Lanzhou University, The First Clinical Medical College of Lanzhou University, Lanzhou 730000, PR China
| | - Yong-Fei Wang
- The First Clinical Medical College of Lanzhou University, Lanzhou 730000, PR China; Institute of Cancer Neuroscience, Medical Frontier Innovation Research Center, The First Hospital of Lanzhou University, The First Clinical Medical College of Lanzhou University, Lanzhou 730000, PR China
| | - Wei-Ping Li
- The First Clinical Medical College of Lanzhou University, Lanzhou 730000, PR China; Department of Urology, The First Hospital of Lanzhou University, Lanzhou, Gansu 730000, PR China
| | - Wei-Lin Jin
- The First Clinical Medical College of Lanzhou University, Lanzhou 730000, PR China; Institute of Cancer Neuroscience, Medical Frontier Innovation Research Center, The First Hospital of Lanzhou University, The First Clinical Medical College of Lanzhou University, Lanzhou 730000, PR China.
| |
Collapse
|
100
|
Zhou HM, Yang XY, Yue SJ, Wang WX, Zhang Q, Xu DQ, Li JJ, Tang YP. The identification of metabolites from gut microbiota in coronary heart disease via network pharmacology. ARTIFICIAL CELLS, NANOMEDICINE, AND BIOTECHNOLOGY 2024; 52:145-155. [PMID: 38412071 DOI: 10.1080/21691401.2024.2319827] [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: 08/30/2023] [Accepted: 02/12/2024] [Indexed: 02/29/2024]
Abstract
Although the gut microbial metabolites exhibit potential effects on coronary heart disease (CHD), the underlying mechanism remains unclear. In this study, the active gut microbial metabolites acting on CHD and their potential mechanisms of action were explored through a network pharmacological approach. We collected a total of 208 metabolites from the gutMgene database and 726 overlapping targets from the similarity ensemble approach (SEA) and SwissTargetPrediction (STP) database, and ultimately identified 610 targets relevant to CHD. In conjunction with the gutMGene database, we identified 12 key targets. The targets of exogenous substances were removed, and 10 core targets involved in CHD were eventually retained. The microbiota-metabolites-targets-signalling pathways network analysis revealed that C-type lectin receptor signalling pathway, Lachnospiraceae, Escherichia, mitogen-activated protein kinase 1, prostaglandin-endoperoxidase synthase 2, phenylacetylglutamine and alcoholic acid are notable components of CHD and play important roles in the development of CHD. The results of molecular docking experiments demonstrated that AKT1-glycocholic acid and PTGS2-phenylacetylglutamine complexes may act on C-type lectin receptor signalling pathways. In this study, the key substances and potential mechanisms of gut microbial metabolites were analysed via network pharmacological methods, and a scientific basis and comprehensive idea were provided for the effects of gut microbial metabolites on CHD.
Collapse
Affiliation(s)
- Hao-Ming Zhou
- Key Laboratory of Shaanxi Administration of Traditional Chinese Medicine for TCM Compatibility, State Key Laboratory of Research & Development of Characteristic Qin Medicine Resources (Cultivation), Shaanxi Key Laboratory of Chinese Medicine Fundamentals and New Drugs Research, Shaanxi Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Shaanxi University of Chinese Medicine, Xi'an, China
| | - Xin-Yu Yang
- Department of Pharmacy, Beijing Key Laboratory of Bio-characteristic Profiling for Evaluation of Rational Drug Use, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Shi-Jun Yue
- Key Laboratory of Shaanxi Administration of Traditional Chinese Medicine for TCM Compatibility, State Key Laboratory of Research & Development of Characteristic Qin Medicine Resources (Cultivation), Shaanxi Key Laboratory of Chinese Medicine Fundamentals and New Drugs Research, Shaanxi Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Shaanxi University of Chinese Medicine, Xi'an, China
- International Joint Research Center on Resource Utilization and Quality Evaluation of Traditional Chinese Medicine of Hebei Province, Hebei University of Chinese Medicine, Shijiazhuang, China
| | - Wen-Xiao Wang
- International Joint Research Center on Resource Utilization and Quality Evaluation of Traditional Chinese Medicine of Hebei Province, Hebei University of Chinese Medicine, Shijiazhuang, China
| | - Qiao Zhang
- Key Laboratory of Shaanxi Administration of Traditional Chinese Medicine for TCM Compatibility, State Key Laboratory of Research & Development of Characteristic Qin Medicine Resources (Cultivation), Shaanxi Key Laboratory of Chinese Medicine Fundamentals and New Drugs Research, Shaanxi Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Shaanxi University of Chinese Medicine, Xi'an, China
| | - Ding-Qiao Xu
- Key Laboratory of Shaanxi Administration of Traditional Chinese Medicine for TCM Compatibility, State Key Laboratory of Research & Development of Characteristic Qin Medicine Resources (Cultivation), Shaanxi Key Laboratory of Chinese Medicine Fundamentals and New Drugs Research, Shaanxi Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Shaanxi University of Chinese Medicine, Xi'an, China
| | - Jia-Jia Li
- Key Laboratory of Shaanxi Administration of Traditional Chinese Medicine for TCM Compatibility, State Key Laboratory of Research & Development of Characteristic Qin Medicine Resources (Cultivation), Shaanxi Key Laboratory of Chinese Medicine Fundamentals and New Drugs Research, Shaanxi Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Shaanxi University of Chinese Medicine, Xi'an, China
| | - Yu-Ping Tang
- Key Laboratory of Shaanxi Administration of Traditional Chinese Medicine for TCM Compatibility, State Key Laboratory of Research & Development of Characteristic Qin Medicine Resources (Cultivation), Shaanxi Key Laboratory of Chinese Medicine Fundamentals and New Drugs Research, Shaanxi Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Shaanxi University of Chinese Medicine, Xi'an, China
| |
Collapse
|