1
|
Mansoor T, Farrukh F, Khalid SN, Abramov D, Michos ED, Mehta A, Paul TK, Dani SS, Al Rifai M, Misra A, Nambi V, Virani SS, Minhas AMK. The future of hypertension pharmacotherapy: Ongoing and future clinical trials for hypertension. Curr Probl Cardiol 2025; 50:102922. [PMID: 39522662 DOI: 10.1016/j.cpcardiol.2024.102922] [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: 11/02/2024] [Accepted: 11/04/2024] [Indexed: 11/16/2024]
Abstract
Hypertension is among the most prevalent diagnoses across the world and increases the risk of many serious health problems, such as stroke, heart disease, and kidney disease. Pharmacological approaches to treat hypertension are often required and reduce blood pressure through mechanisms such as vasodilation, inhibition of the renin-angiotensin-aldosterone pathway, and increased urine output to reduce blood volume, among other mechanisms. Further research is ongoing to find novel pathways and mechanisms to treat hypertension, which we summarize in this review. We used clinicaltrials.gov to gather information about ongoing clinical trials of pharmacological hypertension therapy as of March 2024 and found 103 clinical trials that met our criteria. The interventions of these 103 clinical trials include novel and previously approved pharmacological and dietary supplement therapies for hypertension. We aim to use these clinical trials to provide insight into the future therapies and practices of hypertension treatment.
Collapse
Affiliation(s)
- Taha Mansoor
- Department of Internal Medicine, Western Michigan University Homer Stryker M.D. School of Medicine, Kalamazoo, MI, USA.
| | - Fatima Farrukh
- Department of Internal Medicine, University of Cincinnati, Cincinnati, OH, USA
| | - Subaina N Khalid
- Department of Internal Medicine, SUNY Upstate Medical University, Syracruse, NY, USA
| | - Dmitry Abramov
- Division of Cardiology, Department of Medicine, Loma Linda University Health, Loma Linda, CA, USA
| | - Erin D Michos
- Division of Cardiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Anurag Mehta
- Division of Cardiology, Virginia Commonwealth University, Richmond, VA, USA
| | - Timir K Paul
- Department of Cardiovascular Sciences, University of Tennessee Health Science Center- Nashville, TN, USA
| | - Sourbha S Dani
- Division of Cardiology, Lahey Hospital and Medical Center, Beth Israel Lahey Health, Burlington, MA, USA
| | | | - Arunima Misra
- Section of Cardiology, Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Vijay Nambi
- Section of Cardiology, Department of Medicine, Baylor College of Medicine, Houston, TX, USA; Section of Cardiovascular Research, Baylor College of Medicine, Houston, TX, USA; Michael E. DeBakey Veterans Affair Medical Center, Houston, TX, USA
| | - Salim S Virani
- Department of Medicine, Aga Khan University, Karachi, Pakistan
| | - Abdul Mannan Khan Minhas
- Section of Cardiology, Department of Medicine, Baylor College of Medicine, Houston, TX, USA; Section of Cardiovascular Research, Baylor College of Medicine, Houston, TX, USA
| |
Collapse
|
2
|
Buchan E, Harbi MH, Rickard JJS, Thomas M, Goldberg Oppenheimer P. Advanced biomolecular spectroscopic profiling of cardiovascular disease macromolecular markers: SIL-6, IL-9, LpA, ApoB, PCSK9 and NT-ProBNP for rapid in-situ detection and monitoring. Int J Biol Macromol 2025; 284:138115. [PMID: 39608533 DOI: 10.1016/j.ijbiomac.2024.138115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2024] [Revised: 11/23/2024] [Accepted: 11/25/2024] [Indexed: 11/30/2024]
Abstract
Cardiovascular disease (CVD) remains a major global health concern and a leading cause of morbidity and mortality worldwide. Early-diagnosis and prompt medical attention are crucial in managing and reducing overall impact on health-and-wellbeing, necessitating the development of innovative diagnostics, which transcend traditional methodologies. Raman spectroscopy uniquely provides molecular fingerprinting and structural information, offering insights into biochemical composition. Integration of Raman spectroscopy with advanced machine learning is established as a powerful clinical adjunct for point-of-care detection of CVDs. A non-invasive, label-free spectroscopic platform coupled with neural network algorithm, 'SKiNET' has been developed to accurately detect the biomolecular changes within plasma of CVD versus healthy cohorts, enabling rapid diagnosis and longer-term monitoring, where the real-time capabilities provide dynamic assessment of progression, aligning treatment strategies with evolving states. CVD has been detected and classified via SKiNET with 88.6 %-accuracy, 92.9 %-specificity and 85.1 %-sensitivity and with 83.8 %-accuracy. The hybrid RS-SKiNET bio-molecularly specific detection signposted a comprehensive panel of CVD-indicative biomarkers, including SIL-6, IL-9, LpA, ApoB, PCSK9 and NT-ProBNP, offering important insights into disease mechanisms and risk-stratification. This multidimensional technique holds potential for improved patient-and-healthcare management for CVDs, laying the platform toward high-throughput biomolecular profiling of CVD-indicative macromolecular biomarkers, particularly vital for widespread point-of-care diagnostics and monitoring.
Collapse
Affiliation(s)
- Emma Buchan
- School of Chemical Engineering, College of Engineering and Physical Science, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK
| | - Maan H Harbi
- Pharmacology and Toxicology Department, College of Pharmacy, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Jonathan J S Rickard
- Department of Physics, Cavendish Laboratory, University of Cambridge, JJ Thomson Avenue, Cambridge CB3 0HE, UK
| | - Mark Thomas
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK
| | - Pola Goldberg Oppenheimer
- School of Chemical Engineering, College of Engineering and Physical Science, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK; Healthcare Technologies Institute, Institute of Translational Medicine, Mindelsohn Way, Birmingham B15 2TH, UK.
| |
Collapse
|
3
|
Gowayed MA, Zakaraya ZZ, Abu-Samra N, Elhamammy RH, Abdel Moneim LM, Hafez HA, Moneam IA, Oriquat GA, Kamel MA. Crosstalk between mitochondrial homeostasis and AMPK pathway mediate the receptor-mediated cardioprotective effects of estradiol in ovariectomized female rats. PLoS One 2024; 19:e0312397. [PMID: 39693325 DOI: 10.1371/journal.pone.0312397] [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: 07/24/2024] [Accepted: 10/04/2024] [Indexed: 12/20/2024] Open
Abstract
Estrogen (E2) deficiency is a risk factor for cardiovascular disease (CVD), however, the exact mechanism for the E2 protective effect on CVD remains unclear. This study aimed to investigate the estrogen receptor (ER) and non-receptor mediated effects of E2 treatment on the cardiac expression of adenosine monophosphate-dependent protein kinase (AMPK), autophagic, mitophagy and mitochondrial homeostasis-regulating genes in ovariectomized (OVX) rats. Female rats were divided into two main groups; sham and bilaterally OVX rats, then each group was subdivided into four subgroups according to treatment; untreated, subcutaneously treated with E2 (30 μg/kg), or Fulvestrant (F) (5 mg/Kg), or a combination of both drugs for 28 days. The OVX rats or F-treated sham rats showed dyslipidemia, and marked disturbances in parameters of AMPK signaling, autophagy, mitophagy, mitochondrial fission, fusion and biogenesis. E2 administration to OVX or F-treated sham rats has corrected the disturbed lipid and cardiac profiles, increased AMPK, and restored the balance of cardiac autophagy, mitophagy, and mitochondrial dynamics and homeostasis. Most of these effects in OVX rats were blocked by the ER antagonist (F). Estrogen treatment has cardioprotective effects in OVX females through modulating cardiac mitochondrial homeostasis, mitophagy and autophagy and restoring the AMPK signaling pathway. As witnessed by Fulvestrant, these effects suggest the main role of ER-mediated signaling in regulating mitophagy and plasma and cardiac lipids along with the existence of a post-translational control mechanism or the involvement of estrogenic non-receptor pathway controlling the postmenopausal cardiac mitochondrial energy production machinery that needs further investigation.
Collapse
Affiliation(s)
- Mennatallah A Gowayed
- Department of Pharmacology &Therapeutics, Faculty of Pharmacy and Drug Manufacturing, Pharos University in Alexandria, Alexandria, Egypt
| | - Zainab Zaki Zakaraya
- Biopharmaceutics and Clinical Pharmacy Department, Faculty of Pharmacy, Al-Ahliyya Amman University, Amman, Jordan
| | - Nehal Abu-Samra
- Department of Basic Sciences, Faculty of Physical Therapy, Pharos University in Alexandria, Alexandria, Egypt
| | - Reem H Elhamammy
- Department of Biochemistry, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt
| | - Lobna M Abdel Moneim
- Department of Pharmacology &Therapeutics, Faculty of Pharmacy and Drug Manufacturing, Pharos University in Alexandria, Alexandria, Egypt
| | - Hala A Hafez
- Department of Biochemistry, Medical Research Institute, Alexandria University, Alexandria, Egypt
| | - Ihab A Moneam
- Clinical Laboratory Sciences Department, College of Pharmacy, Almaaqal University, Basra, Iraq
- Supplementary General Sciences Department, Faculty of Dentistry, Future University, New Cairo, Egypt
| | - Ghaleb A Oriquat
- Department of Medical Laboratory Sciences, Faculty of Allied Medical Sciences, Al-Ahliyya Amman University, Amman, Jordan
| | - Maher A Kamel
- Department of Biochemistry, Medical Research Institute, Alexandria University, Alexandria, Egypt
| |
Collapse
|
4
|
Jiang D, Wang J, Wang R, Wu Y. Comprehensive Insights into Mechanisms for Ventricular Remodeling in Right Heart Failure. Rev Cardiovasc Med 2024; 25:426. [PMID: 39742244 PMCID: PMC11683703 DOI: 10.31083/j.rcm2512426] [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: 03/31/2024] [Revised: 07/12/2024] [Accepted: 07/23/2024] [Indexed: 01/03/2025] Open
Abstract
Ventricular remodeling in right heart failure is a complex pathological process involving interactions between multiple mechanisms. Overactivation of the neuro-hormonal pathways, activation of the oxidative stress response, expression of cytokines, apoptosis of cardiomyocytes, and alterations of the extracellular matrix (ECM) are among the major mechanisms involved in the development of ventricular remodeling in right heart failure. These mechanisms are involved in ventricular remodeling, such as myocardial hypertrophy and fibrosis, leading to the deterioration of myocardial systolic and diastolic function. A deeper understanding of these mechanisms can help develop more effective therapeutic strategies in patients with right heart failure (RHF) to improve patient survival and quality of life. Despite the importance of ventricular remodeling in RHF, there are a limited number of studies in this field. This article explores in-depth historical and current information about the specific mechanisms in ventricular remodeling in RHF, providing a theoretical rationale for recognizing its importance in health and disease.
Collapse
Affiliation(s)
- Dongmei Jiang
- Department of General Medicine, First Affiliated Hospital of Xinjiang Medical University, 830011 Urumchi, Xinjiang, China
| | - Jie Wang
- Department of Pharmacy, First Affiliated Hospital of Xinjiang Medical University, 830011 Urumchi, Xinjiang, China
| | - Rui Wang
- Department of General Medicine, First Affiliated Hospital of Xinjiang Medical University, 830011 Urumchi, Xinjiang, China
| | - Yun Wu
- Department of General Medicine, First Affiliated Hospital of Xinjiang Medical University, 830011 Urumchi, Xinjiang, China
| |
Collapse
|
5
|
Kosyakovsky LB, de Boer RA, Ho JE. Screening for Heart Failure: Biomarkers to Detect Heightened Risk in the General Population. Curr Heart Fail Rep 2024; 21:591-603. [PMID: 39287754 DOI: 10.1007/s11897-024-00686-6] [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] [Accepted: 09/04/2024] [Indexed: 09/19/2024]
Abstract
PURPOSE OF REVIEW Heart failure (HF) represents a growing global burden of morbidity and mortality. Identifying individuals at risk for HF development is increasingly important, particularly given the advent of disease-modifying therapies for HF as well as its major risk factors such as obesity actalnd diabetes. We aim to review the key circulating biomarkers associated with future HF which may contribute to HF risk prediction. RECENT FINDINGS While current guidelines recommend the use of natriuretic peptides and cardiac troponins in HF risk stratification, there are a diverse array of other emerging protein, metabolic, transcriptomic, and genomic biomarkers of future HF development. These biomarkers not only lend insight into the underlying pathophysiology of HF, which spans inflammation to cardiac fibrosis, but also offer an opportunity to further refine HF risk in addition to established biomarkers. As evolving techniques in molecular biology enable an increased understanding of the complex biologic contributions to HF pathophysiology, there is an important opportunity to construct integrated clinical and multi-omic models to best capture HF risk. Moving forward, future studies should seek to understand the contributions of sex differences, underlying comorbidity burden, and HF subtypes to an individual's HF risk. Further studies are necessary to fully define the clinical utility of biomarker screening approaches to refine HF risk assessment, as well as to link risk assessment directly to preventive strategies for HF.
Collapse
Affiliation(s)
- Leah B Kosyakovsky
- Division of Cardiology, E/CLS 945, Beth Israel Deaconess Medical Center, 330 Brookline Avenue, Boston, MA, 02215-5491, USA
| | - Rudolf A de Boer
- Department of Cardiology, Cardiovascular Institute, Thorax Center, Erasmus MC, Rotterdam, the Netherlands
| | - Jennifer E Ho
- Division of Cardiology, E/CLS 945, Beth Israel Deaconess Medical Center, 330 Brookline Avenue, Boston, MA, 02215-5491, USA.
| |
Collapse
|
6
|
Anilkumar S A, Dutta S, Aboo S, Ismail A. Vitamin D as a modulator of molecular pathways involved in CVDs: Evidence from preclinical studies. Life Sci 2024; 357:123062. [PMID: 39288869 DOI: 10.1016/j.lfs.2024.123062] [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: 06/18/2024] [Revised: 08/27/2024] [Accepted: 09/12/2024] [Indexed: 09/19/2024]
Abstract
Vitamin D deficiency (VDD) is a widespread global health issue, affecting nearly a billion individuals worldwide, and mounting evidence links it to an increased risk of cardiovascular diseases like hypertension, atherosclerosis, and heart failure. The discovery of vitamin D receptors and metabolizing enzymes in cardiac and vascular cells, coupled with experimental studies, underscores the complex relationship between vitamin D and cardiovascular health. This review aims to synthesize and critically evaluate the preclinical evidence elucidating the role of vitamin D in cardiovascular health. We examined diverse preclinical in vitro (cardiomyocyte cell line) models and in vivo models, including knockout mice, diet-induced deficiency, and disease-specific animal models (hypertension, hypertrophy and myocardial infarction). These studies reveal that vitamin D modulates vascular tone, and prevents fibrosis and hypertrophy through effects on major signal transduction pathways (NF-kB, Nrf2, PI3K/AKT/mTOR, Calcineurin/NFAT, TGF-β/Smad, AMPK) and influences epigenetic mechanisms governing inflammation, oxidative stress, and pathological remodeling. In vitro studies elucidate vitamin D's capacity to promote cardiomyocyte differentiation and inhibit pathological remodeling. In vivo studies further uncovered detrimental cardiac effects of VDD, while supplementation with vitamin D in cardiovascular disease (CVD) models demonstrated its protective effects by decreasing inflammation, attenuating hypertrophy, reduction in plaque formation, and improving cardiac function. Hence, this comprehensive review emphasizes the critical role of vitamin D in cardiovascular health and its potential as a preventive/therapeutic strategy in CVDs. However, further research is needed to translate these findings into clinical applications as there are discrepancies between preclinical and clinical studies.
Collapse
Affiliation(s)
- Athira Anilkumar S
- Department of Endocrinology, ICMR-National Institute of Nutrition, Hyderabad, India
| | - Soumam Dutta
- Department of Endocrinology, ICMR-National Institute of Nutrition, Hyderabad, India
| | - Shabna Aboo
- Department of Endocrinology, ICMR-National Institute of Nutrition, Hyderabad, India.
| | - Ayesha Ismail
- Department of Endocrinology, ICMR-National Institute of Nutrition, Hyderabad, India.
| |
Collapse
|
7
|
Nagaraj G, Vellaichamy E. Triiodo-L-thyronine (T3) downregulates Npr1 gene (coding for natriuretic peptide receptor-A) transcription in H9c2 cells: involvement of β-AR-ROS signaling. Endocrine 2024; 85:1075-1090. [PMID: 38713329 DOI: 10.1007/s12020-024-03849-6] [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: 01/13/2024] [Accepted: 04/20/2024] [Indexed: 05/08/2024]
Abstract
INTRODUCTION Natriuretic peptide receptor-A (NPR-A) signaling system is considered as an intrinsic productive mechanism of the heart that opposes abnormal cardiac remodeling and hypertrophic growth. NPR-A is coded by Npr1 gene, and its expression is downregulated in the hypertrophied heart. AIM We sought to examine the levels of Npr1 gene transcription in triiodo-L-thyronine (T3) treated hypertrophied cardiomyocyte (H9c2) cells, in vitro, and also the involvement of β-adrenergic receptor (β-AR) - Reactive oxygen species (ROS) signaling system in the down-regulation of Npr1 transcription also studied. MAIN METHODS Anti-hypertrophic Npr1 gene transcription was monitored in control and T3-treated (dose and time dependent) H9c2 cells, using a real time PCR method. Further, cell size, intracellular cGMP, ROS, hypertrophy markers (ANP, BNP, α-sk, α-MHC and β-MHC), β-AR, and protein kinase cGMP-dependent 1 (PKG-I) genes expression were also determined. The intracellular cGMP and ROS levels were determined by ELISA and DCF dye method, respectively. In addition, to neutralize T3 mediated ROS generation, H9c2 cells were treated with T3 in the presence and absence of antioxidants [curcumin (CU) or N-acetyl-L-cysteine (NAC)]. RESULTS A dose dependent (10 pM, 100 pM, 1 nM and 10 nM) and time dependent (12 h, 24 h and 48 h) down-regulation of Npr1 gene transcription (20, 39, 60, and 74% respectively; 18, 55, and 85%, respectively) were observed in T3-treated H9c2 cells as compared with control cells. Immunofluorescence analysis also revealed that a marked down regulation of NPR- A protein in T3-treated cells as compared with control cells. Further, a parallel downregulation of cGMP and PKG-I (2.4 fold) were noticed in the T3-treated cells. In contrast, a time dependent increased expression of β-AR (60, 72, and 80% respectively) and ROS (26, 48, and 74%, respectively) levels were noticed in T3-treated H9c2 cells as compared with control cells. Interestingly, antioxidants, CU or NAC co-treated T3 cells displayed a significant reduction in ROS (69 and 81%, respectively) generation and to increased Npr1 gene transcription (81 and 88%, respectively) as compared with T3 alone treated cells. CONCLUSION Our result suggest that down regulation of Npr1 gene transcription is critically involved in T3- induced hypertrophic growth in H9c2 cells, and identifies the cross-talk between T3-β-AR-ROS and NPR-A signaling.
Collapse
Affiliation(s)
- Gopinath Nagaraj
- Peptide Research and Molecular Cardiology Unit, Department of Biochemistry, University of Madras, Guindy campus, Chennai, Tamil Nadu, 600025, India
| | - Elangovan Vellaichamy
- Peptide Research and Molecular Cardiology Unit, Department of Biochemistry, University of Madras, Guindy campus, Chennai, Tamil Nadu, 600025, India.
| |
Collapse
|
8
|
Abassi Z, Hamo‐Giladi DB, Kinaneh S, Heyman SN. The endocrine basis of the cardio-renal axis: New perspectives regarding corin. Physiol Rep 2024; 12:e16105. [PMID: 38942727 PMCID: PMC11213627 DOI: 10.14814/phy2.16105] [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: 02/15/2024] [Revised: 05/10/2024] [Accepted: 05/27/2024] [Indexed: 06/30/2024] Open
Abstract
The central role of natriuretic peptides (NPs) in the complex cardio-renal integrated physiology and organ failure has been revealed over the last four decades. Atrial natriuretic peptide (ANP), the oldest representative of the NPs family, is produced through conversion of proANP to the mature peptide by corin, a trans-membrane protease localized to the cardiac myocyte membrane. Similarly, brain natriuretic peptide (BNP) is generated by furin, which cleaves proBNP to BNP in myocytes. Though the components of NPs system, their synthesis and target organs are well established, understanding their role in the interplay between the heart and the kidney is steadily evolving. In this context, Feldman et al. (New England Journal of Medicine, 389, 1685) recently described patients with hypertension, cardiomyopathy, atrial arrhythmia and left atrial fibrosis, associated with a homozygous loss-of-function variant of the gene encoding corin (Cor-/-). Notably, reduced baseline urinary electrolyte and creatinine excretion have been observed in one of the studied patients. This renal excretory functional impairment could be attributed to the lack of cardiac-derived ANP in these patients, as implied by Feldman et al. Yet, in this mini-review we suggest that this aberrant renal manifestation may principally stem from lack of local ANP production at renal tissue, as corin is normally expressed in proximal tubules, Henle's loop and collecting ducts, with locally produced ANP provoking Na+ and water exertion. Collectively, it seems that beside the classic well-established cardio-renal axis, the renal NPs system functions as local endocrine machinery in the regulation of sodium excretion.
Collapse
Affiliation(s)
- Zaid Abassi
- Ruth & Bruce Rappaport Faculty of Medicine, Technion‐IITHaifaIsrael
- Department of Laboratory MedicineRambam Health Care CampusHaifaIsrael
| | | | - Safa Kinaneh
- Ruth & Bruce Rappaport Faculty of Medicine, Technion‐IITHaifaIsrael
| | - Samuel N. Heyman
- Department of MedicineHadassah Hebrew University Hospital, Mt. Scopus and Herzog HospitalJerusalemIsrael
| |
Collapse
|
9
|
Abassi Z, Azzam ZS, Heyman SN. Corin and Left Atrial Cardiomyopathy, Hypertension, Arrhythmia, and Fibrosis. N Engl J Med 2024; 390:1539. [PMID: 38657261 DOI: 10.1056/nejmc2313870] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/26/2024]
Affiliation(s)
- Zaid Abassi
- Technion-Israel Institute of Technology, Haifa, Israel
| | | | - Samuel N Heyman
- Hadassah Hebrew University Hospital, Mt. Scopus, Jerusalem, Israel
| |
Collapse
|
10
|
Mah CK, Ahmed N, Lopez NA, Lam DC, Pong A, Monell A, Kern C, Han Y, Prasad G, Cesnik AJ, Lundberg E, Zhu Q, Carter H, Yeo GW. Bento: a toolkit for subcellular analysis of spatial transcriptomics data. Genome Biol 2024; 25:82. [PMID: 38566187 PMCID: PMC11289963 DOI: 10.1186/s13059-024-03217-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Accepted: 03/14/2024] [Indexed: 04/04/2024] Open
Abstract
The spatial organization of molecules in a cell is essential for their functions. While current methods focus on discerning tissue architecture, cell-cell interactions, and spatial expression patterns, they are limited to the multicellular scale. We present Bento, a Python toolkit that takes advantage of single-molecule information to enable spatial analysis at the subcellular scale. Bento ingests molecular coordinates and segmentation boundaries to perform three analyses: defining subcellular domains, annotating localization patterns, and quantifying gene-gene colocalization. We demonstrate MERFISH, seqFISH + , Molecular Cartography, and Xenium datasets. Bento is part of the open-source Scverse ecosystem, enabling integration with other single-cell analysis tools.
Collapse
Affiliation(s)
- Clarence K Mah
- Division of Medical Genetics, Department of Medicine, University of California San Diego, La Jolla, CA, USA
- Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, CA, USA
- Sanford Stem Cell Institute Innovation Center, La Jolla, CA, USA
| | - Noorsher Ahmed
- Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, CA, USA
- Sanford Stem Cell Institute Innovation Center, La Jolla, CA, USA
| | - Nicole A Lopez
- Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, CA, USA
| | - Dylan C Lam
- Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, CA, USA
- Sanford Stem Cell Institute Innovation Center, La Jolla, CA, USA
- Division of Biological Sciences, University of California San Diego, La Jolla, CA, USA
| | - Avery Pong
- Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, CA, USA
| | - Alexander Monell
- Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, CA, USA
- Department of Bioengineering, University of California San Diego, La Jolla, CA, USA
| | - Colin Kern
- Center for Epigenomics, University of California San Diego, La Jolla, CA, USA
| | - Yuanyuan Han
- Center for Epigenomics, University of California San Diego, La Jolla, CA, USA
| | - Gino Prasad
- Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, CA, USA
- Department of Bioengineering, University of California San Diego, La Jolla, CA, USA
| | - Anthony J Cesnik
- Department of Bioengineering, Stanford University, Stanford, CA, USA
| | - Emma Lundberg
- Department of Bioengineering, Stanford University, Stanford, CA, USA
- Department of Pathology, Stanford University, Stanford, CA, USA
- Chan-Zuckerberg Biohub, San Francisco, CA, USA
| | - Quan Zhu
- Center for Epigenomics, University of California San Diego, La Jolla, CA, USA
| | - Hannah Carter
- Division of Medical Genetics, Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Gene W Yeo
- Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, CA, USA.
- Sanford Stem Cell Institute Innovation Center, La Jolla, CA, USA.
- Stem Cell Program, University of California San Diego, La Jolla, CA, USA.
- Institute for Genomic Medicine, University of California San Diego, La Jolla, CA, USA.
| |
Collapse
|
11
|
Romero-Becera R, Santamans AM, Arcones AC, Sabio G. From Beats to Metabolism: the Heart at the Core of Interorgan Metabolic Cross Talk. Physiology (Bethesda) 2024; 39:98-125. [PMID: 38051123 DOI: 10.1152/physiol.00018.2023] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Revised: 10/26/2023] [Accepted: 12/01/2023] [Indexed: 12/07/2023] Open
Abstract
The heart, once considered a mere blood pump, is now recognized as a multifunctional metabolic and endocrine organ. Its function is tightly regulated by various metabolic processes, at the same time it serves as an endocrine organ, secreting bioactive molecules that impact systemic metabolism. In recent years, research has shed light on the intricate interplay between the heart and other metabolic organs, such as adipose tissue, liver, and skeletal muscle. The metabolic flexibility of the heart and its ability to switch between different energy substrates play a crucial role in maintaining cardiac function and overall metabolic homeostasis. Gaining a comprehensive understanding of how metabolic disorders disrupt cardiac metabolism is crucial, as it plays a pivotal role in the development and progression of cardiac diseases. The emerging understanding of the heart as a metabolic and endocrine organ highlights its essential contribution to whole body metabolic regulation and offers new insights into the pathogenesis of metabolic diseases, such as obesity, diabetes, and cardiovascular disorders. In this review, we provide an in-depth exploration of the heart's metabolic and endocrine functions, emphasizing its role in systemic metabolism and the interplay between the heart and other metabolic organs. Furthermore, emerging evidence suggests a correlation between heart disease and other conditions such as aging and cancer, indicating that the metabolic dysfunction observed in these conditions may share common underlying mechanisms. By unraveling the complex mechanisms underlying cardiac metabolism, we aim to contribute to the development of novel therapeutic strategies for metabolic diseases and improve overall cardiovascular health.
Collapse
Affiliation(s)
| | | | - Alba C Arcones
- Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain
- Centro Nacional de Investigaciones Oncológicas, Madrid, Spain
| | - Guadalupe Sabio
- Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain
- Centro Nacional de Investigaciones Oncológicas, Madrid, Spain
| |
Collapse
|
12
|
Gu X, Liu M, Wang M, Wang K, Zhou T, Wu Q, Dong N. Corin deficiency alleviates mucosal lesions in a mouse model of colitis induced by dextran sulfate sodium. Life Sci 2024; 339:122446. [PMID: 38246520 DOI: 10.1016/j.lfs.2024.122446] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Revised: 01/10/2024] [Accepted: 01/16/2024] [Indexed: 01/23/2024]
Abstract
AIMS High dietary salt consumption is a risk factor for inflammatory bowel disease (IBD). Corin is a protease that activates atrial natriuretic peptide (ANP), thereby regulating sodium homeostasis. Corin acts in multiple tissues, including the intestine. In mice, corin deficiency impairs intestinal sodium excretion. This study aims to examine if reduced intestinal sodium excretion alters the pathophysiology of IBD. MAIN METHODS Wild-type (WT), Corin knockout (KO), and Corin kidney conditional KO (kcKO) mice were tested in a colitis model induced by dextran sulfide sodium (DSS). Effects of ANP on DSS-induced colitis were tested in WT and Corin KO mice. Body weight changes in the mice were monitored. Necropsy, histological analysis, and immunostaining studies were conducted to examine colon length and mucosal lesions. Fecal sodium levels were measured. RT-PCR was done to analyze proinflammatory genes in colon samples. KEY FINDINGS DSS-treated Corin KO mice had an ameliorated colitis phenotype with less body weight loss, longer colon lengths, smaller mucosal lesions, lower disease scores, more preserved goblet cells, and suppressed proinflammatory genes in the colon. In longitudinal studies, the DSS-treated Corin KO mice had delayed onset of colon mucosal lesions. ANP administration lessened the colitis in WT, but not Corin KO, mice. Analyses of WT, Corin KO, and Corin kcKO mice indicated that fecal sodium excretion, controlled by intestinal corin, may regulate inflammatory responses in DSS-induced colitis in mice. SIGNIFICANCE Our findings indicate a role of corin in intestinal pathophysiology, suggesting that reduced intestinal sodium level may offer protective benefits against IBD.
Collapse
Affiliation(s)
- Xiabing Gu
- Jiangsu Institute of Hematology, The First Affiliated Hospital and Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Protection, Suzhou Medical College, Soochow University, Suzhou, China; Cyrus Tang Hematology Center, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Prevention, Soochow University, Suzhou, China
| | - Meng Liu
- Cyrus Tang Hematology Center, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Prevention, Soochow University, Suzhou, China
| | - Mengting Wang
- Jiangsu Institute of Hematology, The First Affiliated Hospital and Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Protection, Suzhou Medical College, Soochow University, Suzhou, China; Cyrus Tang Hematology Center, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Prevention, Soochow University, Suzhou, China
| | - Kun Wang
- Jiangsu Institute of Hematology, The First Affiliated Hospital and Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Protection, Suzhou Medical College, Soochow University, Suzhou, China; Cyrus Tang Hematology Center, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Prevention, Soochow University, Suzhou, China
| | - Tiantian Zhou
- Cyrus Tang Hematology Center, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Prevention, Soochow University, Suzhou, China
| | - Qingyu Wu
- Cyrus Tang Hematology Center, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Prevention, Soochow University, Suzhou, China.
| | - Ningzheng Dong
- Jiangsu Institute of Hematology, The First Affiliated Hospital and Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Protection, Suzhou Medical College, Soochow University, Suzhou, China; Cyrus Tang Hematology Center, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Prevention, Soochow University, Suzhou, China.
| |
Collapse
|
13
|
Ahmad S, Kumar R. An update of new/potential cardiovascular markers: a narrative review. Mol Biol Rep 2024; 51:179. [PMID: 38252393 DOI: 10.1007/s11033-023-08978-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Accepted: 11/07/2023] [Indexed: 01/23/2024]
Abstract
BACKGROUND Cardiovascular and their associated disease (CVD) is a leading cause of death worldwide, in developed and developing countries, and its prevalence has increased over the past few decades, due to changes in the lifestyle of people. Biomarkers are important tools for diagnosing, analyzing, and providing evidence of pathological conditions of CVD and their associated diseases. METHODS This study reviews historical cardiovascular biomarkers used to diagnose various diseases, their uses, and limitations, as well as the importance of new and emerging biomarkers. CONCLUSION sST2, GDF-15, CD-40, IL-6, and Micro-RNA. Initial studies of the future of cardiac biomarkers are promising, but more research is needed to demonstrate that they are more effective biomarkers of risk factors for CVD development. They also lack the analytical foundation needed for adoption in the medical industry. It is also necessary to determine whether these biomarkers can be used for diagnosis.
Collapse
Affiliation(s)
- Sharique Ahmad
- Department of Pathology, Era's Lucknow Medical College & Hospital, Era University, Lucknow, India
| | - Raushan Kumar
- Department of Pathology, Era's Lucknow Medical College & Hospital, Era University, Lucknow, India.
| |
Collapse
|
14
|
Sofia-Gonçalves A, Guedes-Martins L. Nuchal Translucency and Congenital Heart Defects. Curr Cardiol Rev 2024; 20:1-13. [PMID: 38275068 PMCID: PMC11107467 DOI: 10.2174/011573403x264963231128045500] [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: 06/26/2023] [Revised: 08/18/2023] [Accepted: 10/17/2023] [Indexed: 01/27/2024] Open
Abstract
Nuchal translucency comprises a temporary accumulation of fluid in the subcutaneous tissue on the back of a fetus's neck, which accompanies the crown-rump length and is observed through an ultrasound performed between 11 and 13 weeks + 6 days gestation. Nuchal translucency is considered to be above normal when values are higher than the 95th/99th percentile or equal to or higher than 2.5/3.5 mm. The first connection between increased nuchal translucency and the presence of congenital heart defects is described in the study of Hyett et al., who observed that they are directly proportional. Since that time, several studies have been conducted to understand if nuchal translucency measurements can be used for congenital heart defect screening in euploid fetuses. However, there is great variability in the estimated nuchal translucency cutoff values for congenital heart defect detection. The purpose of this review was to understand how increased nuchal translucency values and congenital heart defects are related and to identify which of these defects are more frequently associated with an increase in these values.
Collapse
Affiliation(s)
- A. Sofia-Gonçalves
- Instituto de Ciências Biomédicas Abel Salazar, University of Porto, 4050-313, Porto, Portugal
- Centro de Medicina Fetal, Medicina Fetal Porto - Centro Materno Infantil do Norte, 4099-001, Porto, Portugal
| | - L. Guedes-Martins
- Instituto de Ciências Biomédicas Abel Salazar, University of Porto, 4050-313, Porto, Portugal
- Centro de Medicina Fetal, Medicina Fetal Porto - Centro Materno Infantil do Norte, 4099-001, Porto, Portugal
- Departamento da Mulher e da Medicina Reprodutiva, Centro Hospitalar Universitário do Porto EPE, Centro Materno Infantil do Norte, Largo Prof. Abel Salazar, 4099-001, Porto, Portugal
- Unidade de Investigação e Formação – Centro Materno Infantil do Norte, 4099-001, Porto, Portugal
- Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-319, Porto, Portugal
| |
Collapse
|
15
|
Sousa MP, Bettencourt P, Brás-Silva C, Pereira C. Biosensors for natriuretic peptides in cardiovascular diseases. A review. Curr Probl Cardiol 2024; 49:102180. [PMID: 37907188 DOI: 10.1016/j.cpcardiol.2023.102180] [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: 10/25/2023] [Accepted: 10/28/2023] [Indexed: 11/02/2023]
Abstract
Heart failure (HF) is a complex clinical syndrome associated with high rates of morbidity and mortality. Over the years, it has been crucial to find accurate biomarkers capable of doing a precise monitor of HF and provide an early diagnosis. Of these, it has been established an important role of natriuretic peptides in HF assessment. Moreover, the development of biosensors has been garnering interest as new diagnostic medical tools. In this review we first provide a general overview of HF, its pathogenesis, and diagnostic features. We then discuss the role of natriuretic peptides in heart failure by characterizing them and point out their potential as biomarkers. Finally, we adress the evolution of biosensors development and the available natriuretic peptides biosensors for disease monitoring.
Collapse
Affiliation(s)
- Mariana P Sousa
- Instituto de Investigação e Inovação em Saúde - i3S, Universidade do Porto, Porto 4200-135, Portugal
| | - Paulo Bettencourt
- Cardiovascular R&D Centre-UnIC@RISE, Department of Surgery and Physiology, Faculty of Medicine, University of Porto, 4200-319, Porto, Portugal
| | - Carmen Brás-Silva
- Cardiovascular R&D Centre-UnIC@RISE, Department of Surgery and Physiology, Faculty of Medicine, University of Porto, 4200-319, Porto, Portugal
| | - Claudia Pereira
- FP-I3ID, Instituto de Investigação, Inovação e Desenvolvimento, FP-BHS, Biomedical and Health Sciences, Universidade Fernando Pessoa, Porto 4249-004, Portugal; HE-FP-Hospital Fernando Pessoa, CECLIN, Center of Clinical Studies, 4420-096 Gondomar, Portugal; FCS-Faculty of Health Sciences, Fernando Pessoa University, 4249-004 Porto, Portugal.
| |
Collapse
|
16
|
Liu Y, Lu X, Chen M, Wei Z, Peng G, Yang J, Tang C, Yu P. Advances in screening, synthesis, modification, and biomedical applications of peptides and peptide aptamers. Biofactors 2024; 50:33-57. [PMID: 37646383 DOI: 10.1002/biof.2001] [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: 05/29/2023] [Accepted: 08/04/2023] [Indexed: 09/01/2023]
Abstract
Peptides and peptide aptamers have emerged as promising molecules for a wide range of biomedical applications due to their unique properties and versatile functionalities. The screening strategies for identifying peptides and peptide aptamers with desired properties are discussed, including high-throughput screening, display screening technology, and in silico design approaches. The synthesis methods for the efficient production of peptides and peptide aptamers, such as solid-phase peptide synthesis and biosynthesis technology, are described, along with their advantages and limitations. Moreover, various modification techniques are explored to enhance the stability, specificity, and pharmacokinetic properties of peptides and peptide aptamers. This includes chemical modifications, enzymatic modifications, biomodifications, genetic engineering modifications, and physical modifications. Furthermore, the review highlights the diverse biomedical applications of peptides and peptide aptamers, including targeted drug delivery, diagnostics, and therapeutic. This review provides valuable insights into the advancements in screening, synthesis, modification, and biomedical applications of peptides and peptide aptamers. A comprehensive understanding of these aspects will aid researchers in the development of novel peptide-based therapeutics and diagnostic tools for various biomedical challenges.
Collapse
Affiliation(s)
- Yijie Liu
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, China
| | - Xiaoling Lu
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, China
| | - Meilun Chen
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, China
| | - Zheng Wei
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, China
| | - Guangnan Peng
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, China
| | - Jie Yang
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, China
| | - Chunhua Tang
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, China
| | - Peng Yu
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, China
| |
Collapse
|
17
|
Yamasaki S, Tokunou T, Maeda T, Horiuchi T. Night-Time Hot Spring Bathing Is Associated with a Lower Systolic Blood Pressure among Japanese Older Adults: A Single-Institution Retrospective Cohort Study. Geriatrics (Basel) 2023; 9:2. [PMID: 38392101 PMCID: PMC10888263 DOI: 10.3390/geriatrics9010002] [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: 08/30/2023] [Revised: 11/28/2023] [Accepted: 12/19/2023] [Indexed: 02/24/2024] Open
Abstract
Hot spring bathing is practiced to help manage hypertension. We retrospectively investigated the effects of hot spring bathing on hypertension with the aim of identifying a novel approach to prevent and manage hypertension. The study cohort comprised 99 patients aged ≥65 years admitted to Kyushu University Beppu Hospital between 1 December 2021 and 30 November 2022 who could walk by themselves and who used hot springs for ≥3 days during their hospital stay. The changes in both systolic and diastolic blood pressure were significantly decreased in the night-time bathing group (n = 21) compared with the noontime (n = 26) and afternoon (n = 52) groups. Night-time hot spring bathing was significantly associated with reduced systolic blood pressure the next morning in older adults. Although prospective randomized controlled trials on night-time hot spring bathing as a hypertension treatment are warranted to investigate whether the practice can prevent hypertension among adults aged ≥65 years, we have initiated a single-center, phase II study on the relationship between sleep quality and quality of life in hypertensive patients after night-time hot spring bathing.
Collapse
Affiliation(s)
- Satoshi Yamasaki
- Department of Internal Medicine, Kyushu University Beppu Hospital, Beppu 874-0838, Japan
- Department of Hematology and Clinical Research Institute, National Hospital Organization Kyushu Medical Center, Fukuoka 810-0065, Japan
| | - Tomotake Tokunou
- Department of Internal Medicine, Kyushu University Beppu Hospital, Beppu 874-0838, Japan
- Division of Basic Medical Science and Fundamental Nursing, Department of Nursing, Fukuoka Nursing College, Fukuoka 814-0193, Japan
| | - Toyoki Maeda
- Department of Internal Medicine, Kyushu University Beppu Hospital, Beppu 874-0838, Japan
| | - Takahiko Horiuchi
- Department of Internal Medicine, Kyushu University Beppu Hospital, Beppu 874-0838, Japan
| |
Collapse
|
18
|
Kaesler N, Cheng M, Nagai J, O’Sullivan J, Peisker F, Bindels EM, Babler A, Moellmann J, Droste P, Franciosa G, Dugourd A, Saez-Rodriguez J, Neuss S, Lehrke M, Boor P, Goettsch C, Olsen JV, Speer T, Lu TS, Lim K, Floege J, Denby L, Costa I, Kramann R. Mapping cardiac remodeling in chronic kidney disease. SCIENCE ADVANCES 2023; 9:eadj4846. [PMID: 38000021 PMCID: PMC10672229 DOI: 10.1126/sciadv.adj4846] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Accepted: 10/24/2023] [Indexed: 11/26/2023]
Abstract
Patients with advanced chronic kidney disease (CKD) mostly die from sudden cardiac death and recurrent heart failure. The mechanisms of cardiac remodeling are largely unclear. To dissect molecular and cellular mechanisms of cardiac remodeling in CKD in an unbiased fashion, we performed left ventricular single-nuclear RNA sequencing in two mouse models of CKD. Our data showed a hypertrophic response trajectory of cardiomyocytes with stress signaling and metabolic changes driven by soluble uremia-related factors. We mapped fibroblast to myofibroblast differentiation in this process and identified notable changes in the cardiac vasculature, suggesting inflammation and dysfunction. An integrated analysis of cardiac cellular responses to uremic toxins pointed toward endothelin-1 and methylglyoxal being involved in capillary dysfunction and TNFα driving cardiomyocyte hypertrophy in CKD, which was validated in vitro and in vivo. TNFα inhibition in vivo ameliorated the cardiac phenotype in CKD. Thus, interventional approaches directed against uremic toxins, such as TNFα, hold promise to ameliorate cardiac remodeling in CKD.
Collapse
Affiliation(s)
- Nadine Kaesler
- Clinic for Renal and Hypertensive Disorders, Rheumatological and Immunological Disease, University Hospital of the RWTH Aachen, Aachen, Germany
- Institute of Experimental Medicine and Systems Biology, University Hospital of the RWTH Aachen, Aachen, Germany
| | - Mingbo Cheng
- Institute for Computational Genomics, University Hospital of the RWTH Aachen, Aachen, Germany
| | - James Nagai
- Institute for Computational Genomics, University Hospital of the RWTH Aachen, Aachen, Germany
| | - James O’Sullivan
- Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, UK
| | - Fabian Peisker
- Institute of Experimental Medicine and Systems Biology, University Hospital of the RWTH Aachen, Aachen, Germany
| | - Eric M. J. Bindels
- Department of Hematology, Erasmus Medical Center, Rotterdam, Netherlands
| | - Anne Babler
- Institute of Experimental Medicine and Systems Biology, University Hospital of the RWTH Aachen, Aachen, Germany
| | - Julia Moellmann
- Department of Internal Medicine I, University Hospital of the RWTH Aachen, Aachen, Germany
| | - Patrick Droste
- Clinic for Renal and Hypertensive Disorders, Rheumatological and Immunological Disease, University Hospital of the RWTH Aachen, Aachen, Germany
- Institute of Pathology, University Hospital of the RWTH Aachen, Aachen, Germany
| | - Giulia Franciosa
- Novo Nordisk Foundation Center for Protein Research, University of Copenhagen, Copenhagen, Denmark
| | - Aurelien Dugourd
- Heidelberg University, Faculty of Medicine, and Heidelberg University Hospital, Institute for Computational Biomedicine, Bioquant, Heidelberg, Germany
| | - Julio Saez-Rodriguez
- Heidelberg University, Faculty of Medicine, and Heidelberg University Hospital, Institute for Computational Biomedicine, Bioquant, Heidelberg, Germany
| | - Sabine Neuss
- Institute of Pathology, University Hospital of the RWTH Aachen, Aachen, Germany
- Helmholtz Institute for Biomedical Engineering, Biointerface Laboratory, RWTH Aachen University, Aachen, Germany
| | - Michael Lehrke
- Department of Internal Medicine I, University Hospital of the RWTH Aachen, Aachen, Germany
| | - Peter Boor
- Clinic for Renal and Hypertensive Disorders, Rheumatological and Immunological Disease, University Hospital of the RWTH Aachen, Aachen, Germany
- Institute of Pathology, University Hospital of the RWTH Aachen, Aachen, Germany
| | - Claudia Goettsch
- Department of Internal Medicine I, University Hospital of the RWTH Aachen, Aachen, Germany
| | - Jesper V. Olsen
- Novo Nordisk Foundation Center for Protein Research, University of Copenhagen, Copenhagen, Denmark
| | - Thimoteus Speer
- Department of Medicine (Nephrology), Goethe University Frankfurt, Frankfurt, Germany
| | - Tzong-Shi Lu
- Brigham and Women’s Hospital, Renal Division, Boston, MA, USA
| | - Kenneth Lim
- Division of Nephrology and Hypertension, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Jürgen Floege
- Clinic for Renal and Hypertensive Disorders, Rheumatological and Immunological Disease, University Hospital of the RWTH Aachen, Aachen, Germany
| | - Laura Denby
- Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, UK
| | - Ivan Costa
- Institute for Computational Genomics, University Hospital of the RWTH Aachen, Aachen, Germany
| | - Rafael Kramann
- Clinic for Renal and Hypertensive Disorders, Rheumatological and Immunological Disease, University Hospital of the RWTH Aachen, Aachen, Germany
- Institute of Experimental Medicine and Systems Biology, University Hospital of the RWTH Aachen, Aachen, Germany
- Department of Internal Medicine, Nephrology and Transplantation, Erasmus Medical Center, Rotterdam, Netherlands
| |
Collapse
|
19
|
Baris Feldman H, Chai Gadot C, Zahler D, Mory A, Aviram G, Elhanan E, Shefer G, Goldiner I, Amir Y, Kurolap A, Ablin JN. Corin and Left Atrial Cardiomyopathy, Hypertension, Arrhythmia, and Fibrosis. N Engl J Med 2023; 389:1685-1692. [PMID: 37913506 DOI: 10.1056/nejmoa2301908] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/03/2023]
Abstract
Two siblings presented with cardiomyopathy, hypertension, arrhythmia, and fibrosis of the left atrium. Each had a homozygous null variant in CORIN, the gene encoding atrial natriuretic peptide (ANP)-converting enzyme. A plasma sample obtained from one of the siblings had no detectable levels of corin or N-terminal pro-ANP but had elevated levels of B-type natriuretic peptide (BNP) and one of the two protein markers of fibrosis that we tested. These and other findings support the hypothesis that BNP cannot fully compensate for a lack of activation of the ANP pathway and that corin is critical to normal ANP activity, left atrial function, and cardiovascular homeostasis.
Collapse
Affiliation(s)
- Hagit Baris Feldman
- From the Genetics Institute and Genomics Center (H.B.F., C.C.G., A.M., E.E., Y.A., A.K.), the Departments of Cardiology (D.Z.), Radiology (G.A.), Nephrology (E.E.), Clinical Laboratories (G.S., I.G.), and Internal Medicine H (J.N.A.) and the Institute of Rheumatology (J.N.A.), Tel Aviv Sourasky Medical Center, and the Faculty of Medicine, Tel Aviv University (H.B.F., D.Z., G.A., I.G., Y.A., J.N.A.) - all in Tel Aviv, Israel
| | - Chofit Chai Gadot
- From the Genetics Institute and Genomics Center (H.B.F., C.C.G., A.M., E.E., Y.A., A.K.), the Departments of Cardiology (D.Z.), Radiology (G.A.), Nephrology (E.E.), Clinical Laboratories (G.S., I.G.), and Internal Medicine H (J.N.A.) and the Institute of Rheumatology (J.N.A.), Tel Aviv Sourasky Medical Center, and the Faculty of Medicine, Tel Aviv University (H.B.F., D.Z., G.A., I.G., Y.A., J.N.A.) - all in Tel Aviv, Israel
| | - David Zahler
- From the Genetics Institute and Genomics Center (H.B.F., C.C.G., A.M., E.E., Y.A., A.K.), the Departments of Cardiology (D.Z.), Radiology (G.A.), Nephrology (E.E.), Clinical Laboratories (G.S., I.G.), and Internal Medicine H (J.N.A.) and the Institute of Rheumatology (J.N.A.), Tel Aviv Sourasky Medical Center, and the Faculty of Medicine, Tel Aviv University (H.B.F., D.Z., G.A., I.G., Y.A., J.N.A.) - all in Tel Aviv, Israel
| | - Adi Mory
- From the Genetics Institute and Genomics Center (H.B.F., C.C.G., A.M., E.E., Y.A., A.K.), the Departments of Cardiology (D.Z.), Radiology (G.A.), Nephrology (E.E.), Clinical Laboratories (G.S., I.G.), and Internal Medicine H (J.N.A.) and the Institute of Rheumatology (J.N.A.), Tel Aviv Sourasky Medical Center, and the Faculty of Medicine, Tel Aviv University (H.B.F., D.Z., G.A., I.G., Y.A., J.N.A.) - all in Tel Aviv, Israel
| | - Galit Aviram
- From the Genetics Institute and Genomics Center (H.B.F., C.C.G., A.M., E.E., Y.A., A.K.), the Departments of Cardiology (D.Z.), Radiology (G.A.), Nephrology (E.E.), Clinical Laboratories (G.S., I.G.), and Internal Medicine H (J.N.A.) and the Institute of Rheumatology (J.N.A.), Tel Aviv Sourasky Medical Center, and the Faculty of Medicine, Tel Aviv University (H.B.F., D.Z., G.A., I.G., Y.A., J.N.A.) - all in Tel Aviv, Israel
| | - Emil Elhanan
- From the Genetics Institute and Genomics Center (H.B.F., C.C.G., A.M., E.E., Y.A., A.K.), the Departments of Cardiology (D.Z.), Radiology (G.A.), Nephrology (E.E.), Clinical Laboratories (G.S., I.G.), and Internal Medicine H (J.N.A.) and the Institute of Rheumatology (J.N.A.), Tel Aviv Sourasky Medical Center, and the Faculty of Medicine, Tel Aviv University (H.B.F., D.Z., G.A., I.G., Y.A., J.N.A.) - all in Tel Aviv, Israel
| | - Gabi Shefer
- From the Genetics Institute and Genomics Center (H.B.F., C.C.G., A.M., E.E., Y.A., A.K.), the Departments of Cardiology (D.Z.), Radiology (G.A.), Nephrology (E.E.), Clinical Laboratories (G.S., I.G.), and Internal Medicine H (J.N.A.) and the Institute of Rheumatology (J.N.A.), Tel Aviv Sourasky Medical Center, and the Faculty of Medicine, Tel Aviv University (H.B.F., D.Z., G.A., I.G., Y.A., J.N.A.) - all in Tel Aviv, Israel
| | - Ilana Goldiner
- From the Genetics Institute and Genomics Center (H.B.F., C.C.G., A.M., E.E., Y.A., A.K.), the Departments of Cardiology (D.Z.), Radiology (G.A.), Nephrology (E.E.), Clinical Laboratories (G.S., I.G.), and Internal Medicine H (J.N.A.) and the Institute of Rheumatology (J.N.A.), Tel Aviv Sourasky Medical Center, and the Faculty of Medicine, Tel Aviv University (H.B.F., D.Z., G.A., I.G., Y.A., J.N.A.) - all in Tel Aviv, Israel
| | - Yam Amir
- From the Genetics Institute and Genomics Center (H.B.F., C.C.G., A.M., E.E., Y.A., A.K.), the Departments of Cardiology (D.Z.), Radiology (G.A.), Nephrology (E.E.), Clinical Laboratories (G.S., I.G.), and Internal Medicine H (J.N.A.) and the Institute of Rheumatology (J.N.A.), Tel Aviv Sourasky Medical Center, and the Faculty of Medicine, Tel Aviv University (H.B.F., D.Z., G.A., I.G., Y.A., J.N.A.) - all in Tel Aviv, Israel
| | - Alina Kurolap
- From the Genetics Institute and Genomics Center (H.B.F., C.C.G., A.M., E.E., Y.A., A.K.), the Departments of Cardiology (D.Z.), Radiology (G.A.), Nephrology (E.E.), Clinical Laboratories (G.S., I.G.), and Internal Medicine H (J.N.A.) and the Institute of Rheumatology (J.N.A.), Tel Aviv Sourasky Medical Center, and the Faculty of Medicine, Tel Aviv University (H.B.F., D.Z., G.A., I.G., Y.A., J.N.A.) - all in Tel Aviv, Israel
| | - Jacob N Ablin
- From the Genetics Institute and Genomics Center (H.B.F., C.C.G., A.M., E.E., Y.A., A.K.), the Departments of Cardiology (D.Z.), Radiology (G.A.), Nephrology (E.E.), Clinical Laboratories (G.S., I.G.), and Internal Medicine H (J.N.A.) and the Institute of Rheumatology (J.N.A.), Tel Aviv Sourasky Medical Center, and the Faculty of Medicine, Tel Aviv University (H.B.F., D.Z., G.A., I.G., Y.A., J.N.A.) - all in Tel Aviv, Israel
| |
Collapse
|
20
|
Cortés M, Lorenzo O, Lumpuy-Castillo J, Martínez-Albaladejo S, Taibo-Urquía M, Pello AM, Bollas AJ, Orejas M, Navas MÁ, Macia E, Martínez ME, Rueda A, Tuñón J. Dapagliflozin Improved Cardiac Function and Structure in Diabetic Patients with Preserved Ejection Fraction: Results of a Single Centre, Observational Prospective Study. J Clin Med 2023; 12:6698. [PMID: 37892836 PMCID: PMC10607224 DOI: 10.3390/jcm12206698] [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: 09/20/2023] [Revised: 10/10/2023] [Accepted: 10/20/2023] [Indexed: 10/29/2023] Open
Abstract
Sodium-glucose cotransporter inhibitors (SGLT2i) have demonstrated a reduction in cardiovascular events in diabetes and heart failure (HF). The mechanisms underlying this benefit are not well known and data are contradictory. The purpose of this study is to analyse the effect of dapagliflozin on cardiac structure and function in patients with normal ejection fraction. Between October 2020 and October 2021, we consecutively included 31 diabetic patients without prior history of SGLT2i use. In all of them, dapagliflozin treatment was started. At inclusion and during six months of follow-up, different clinical, ECG, analytical, and echocardiographic (standard, 3D, and speckle tracking) variables were recorded. After a follow-up period of 6.6 months, an average reduction of 18 g (p = 0.028) in 3D-estimated left ventricle mass was observed. An increase in absolute left ventricle global longitudinal strain (LV-GLS) of 0.3 (p = 0.036) was observed, as well as an increase in isovolumetric relaxation time (IVRT) of 10.5 ms (p = 0.05). Moreover, dapagliflozin decreased the levels of plasma creatin-kinase (CK-MB) and atrial natriuretic peptide (ANP). In conclusion, our data show that the use of SGLT2i is associated with both structural (myocardial mass) and functional (IVRT, LV-GLS) cardiac improvements in a population of diabetic patients with normal ejection fraction.
Collapse
Affiliation(s)
- Marcelino Cortés
- Cardiology Department, Fundación Jiménez Díaz Hospital, 28040 Madrid, Spain; (M.T.-U.); (A.M.P.); (A.J.B.); (M.O.); (M.Á.N.); (E.M.); (M.E.M.); (A.R.); (J.T.)
| | - Oscar Lorenzo
- Laboratory of Diabetes and Vascular Pathology, IIS-Fundación Jiménez Díaz, Universidad Autónoma, 28040 Madrid, Spain; (O.L.); (J.L.-C.); (S.M.-A.)
- Biomedical Research Network on Diabetes and Associated Metabolic Disorders (CIBERDEM), Carlos III National Health Institute, 28029 Madrid, Spain
| | - Jairo Lumpuy-Castillo
- Laboratory of Diabetes and Vascular Pathology, IIS-Fundación Jiménez Díaz, Universidad Autónoma, 28040 Madrid, Spain; (O.L.); (J.L.-C.); (S.M.-A.)
- Biomedical Research Network on Diabetes and Associated Metabolic Disorders (CIBERDEM), Carlos III National Health Institute, 28029 Madrid, Spain
| | - Sacramento Martínez-Albaladejo
- Laboratory of Diabetes and Vascular Pathology, IIS-Fundación Jiménez Díaz, Universidad Autónoma, 28040 Madrid, Spain; (O.L.); (J.L.-C.); (S.M.-A.)
- Biomedical Research Network on Diabetes and Associated Metabolic Disorders (CIBERDEM), Carlos III National Health Institute, 28029 Madrid, Spain
| | - Mikel Taibo-Urquía
- Cardiology Department, Fundación Jiménez Díaz Hospital, 28040 Madrid, Spain; (M.T.-U.); (A.M.P.); (A.J.B.); (M.O.); (M.Á.N.); (E.M.); (M.E.M.); (A.R.); (J.T.)
| | - Ana María Pello
- Cardiology Department, Fundación Jiménez Díaz Hospital, 28040 Madrid, Spain; (M.T.-U.); (A.M.P.); (A.J.B.); (M.O.); (M.Á.N.); (E.M.); (M.E.M.); (A.R.); (J.T.)
| | - Antonio José Bollas
- Cardiology Department, Fundación Jiménez Díaz Hospital, 28040 Madrid, Spain; (M.T.-U.); (A.M.P.); (A.J.B.); (M.O.); (M.Á.N.); (E.M.); (M.E.M.); (A.R.); (J.T.)
| | - Miguel Orejas
- Cardiology Department, Fundación Jiménez Díaz Hospital, 28040 Madrid, Spain; (M.T.-U.); (A.M.P.); (A.J.B.); (M.O.); (M.Á.N.); (E.M.); (M.E.M.); (A.R.); (J.T.)
| | - Miguel Ángel Navas
- Cardiology Department, Fundación Jiménez Díaz Hospital, 28040 Madrid, Spain; (M.T.-U.); (A.M.P.); (A.J.B.); (M.O.); (M.Á.N.); (E.M.); (M.E.M.); (A.R.); (J.T.)
| | - Ester Macia
- Cardiology Department, Fundación Jiménez Díaz Hospital, 28040 Madrid, Spain; (M.T.-U.); (A.M.P.); (A.J.B.); (M.O.); (M.Á.N.); (E.M.); (M.E.M.); (A.R.); (J.T.)
| | - María Esther Martínez
- Cardiology Department, Fundación Jiménez Díaz Hospital, 28040 Madrid, Spain; (M.T.-U.); (A.M.P.); (A.J.B.); (M.O.); (M.Á.N.); (E.M.); (M.E.M.); (A.R.); (J.T.)
| | - Andrea Rueda
- Cardiology Department, Fundación Jiménez Díaz Hospital, 28040 Madrid, Spain; (M.T.-U.); (A.M.P.); (A.J.B.); (M.O.); (M.Á.N.); (E.M.); (M.E.M.); (A.R.); (J.T.)
| | - Jose Tuñón
- Cardiology Department, Fundación Jiménez Díaz Hospital, 28040 Madrid, Spain; (M.T.-U.); (A.M.P.); (A.J.B.); (M.O.); (M.Á.N.); (E.M.); (M.E.M.); (A.R.); (J.T.)
| |
Collapse
|
21
|
Tan K, Foo R, Loh M. Cardiomyopathy in Asian Cohorts: Genetic and Epigenetic Insights. CIRCULATION. GENOMIC AND PRECISION MEDICINE 2023; 16:496-506. [PMID: 37589150 DOI: 10.1161/circgen.123.004079] [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: 08/18/2023]
Abstract
Previous studies on cardiomyopathies have been particularly valuable for clarifying pathological mechanisms in heart failure, an etiologically heterogeneous disease. In this review, we specifically focus on cardiomyopathies in Asia, where heart failure is particularly pertinent. There has been an increase in prevalence of cardiomyopathies in Asia, in sharp contrast with the decline observed in Western countries. Indeed, important disparities in cardiomyopathy incidence, clinical characteristics, and prognosis have been reported in Asian versus White cohorts. These have been accompanied by emerging descriptions of a distinct rare and common genetic basis for disease among Asian cardiomyopathy patients marked by an increased burden of variants with uncertain significance, reclassification of variants deemed pathogenic based on evidence from predominantly White cohorts, and the discovery of Asian-specific cardiomyopathy-associated loci with underappreciated pathogenicity under conventional classification criteria. Findings from epigenetic studies of heart failure, particularly DNA methylation studies, have complemented genetic findings in accounting for the phenotypic variability in cardiomyopathy. Though extremely limited, findings from Asian ancestry-focused DNA methylation studies of cardiomyopathy have shown potential to contribute to general understanding of cardiomyopathy pathophysiology by proposing disease and cause-relevant pathophysiological mechanisms. We discuss the value of multiomics study designs incorporating genetic, methylation, and transcriptomic information for future DNA methylation studies in Asian cardiomyopathy cohorts to yield Asian ancestry-specific insights that will improve risk stratification in the Asian population.
Collapse
Affiliation(s)
- Konstanze Tan
- Lee Kong Chian School of Medicine, Nanyang Technological University, Clinical Sciences Building, Singapore (K.T., M.L.)
| | - Roger Foo
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore (R.F.)
- Department of Cardiology, National University Heart Centre, National University Health System, Singapore (R.F.)
| | - Marie Loh
- Lee Kong Chian School of Medicine, Nanyang Technological University, Clinical Sciences Building, Singapore (K.T., M.L.)
- Genome Institute of Singapore, Singapore (GIS), Agency for Science, Technology and Research (A*STAR) (M.L.)
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, United Kingdom (M.L.)
- National Skin Centre, Singapore (M.L.)
| |
Collapse
|
22
|
Niu Y, Zhou T, Zhang S, Li W, Wang K, Dong N, Wu Q. Corin deficiency impairs cardiac function in mouse models of heart failure. Front Cardiovasc Med 2023; 10:1164524. [PMID: 37636304 PMCID: PMC10450958 DOI: 10.3389/fcvm.2023.1164524] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2023] [Accepted: 08/01/2023] [Indexed: 08/29/2023] Open
Abstract
Introduction Corin is a protease in the natriuretic peptide system. Deleterious CORIN variants are associated with hypertension and heart disease. It remains unclear if and to what extent corin deficiency may contribute to heart failure (HF). Methods Corin knockout (KO) mice were used as a model. Cardiac function was assessed by echocardiography and tissue analysis in Corin KO mice at different ages or subjected to transverse aortic constriction (TAC), which increased pressure overload. Heart and lung tissues were analyzed for cardiac hypertrophy and lung edema using wheat germ agglutinin, Sirius red, Masson's trichrome, and Prussian blue staining. Recombinant corin was tested for its effect on cardiac function in the TAC-operated Corin KO mice. Selected gene expression in the heart was examined by RT-PCR. ELISA was used to analyze factors in plasma. Results Corin KO mice had progressive cardiac dysfunction with cardiac hypertrophy and fibrosis after 9 months of age, likely due to chronic hypertension. When Corin KO mice were subjected to TAC at 10-12 weeks of age, cardiac function decreased more rapidly than in similarly treated wild-type mice. When the TAC-operated Corin KO mice were treated with recombinant corin protein, cardiac dysfunction, hypertrophy, and fibrosis were ameliorated. The corin treatment also decreased the gene expression associated with cardiac hypertrophy and fibrosis, increased plasma cGMP levels, lowered plasma levels of N-terminal pro-atrial natriuretic peptide, angiotensin II, and aldosterone, and lessened lung edema in the Corin KO mice subjected to TAC. Conclusion Corin deficiency impairs cardiac function and exacerbates HF development in mice. Corin protein may be used to reduce cardiac hypertrophy and fibrosis, suppress the renin-angiotensin-aldosterone system, and improve cardiac function in HF.
Collapse
Affiliation(s)
- Yayan Niu
- Cyrus Tang Hematology Center, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Prevention, Medical School, Soochow University, Suzhou, China
| | - Tiantian Zhou
- Cyrus Tang Hematology Center, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Prevention, Medical School, Soochow University, Suzhou, China
| | - Shengnan Zhang
- Cyrus Tang Hematology Center, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Prevention, Medical School, Soochow University, Suzhou, China
- NHC Key Laboratory of Thrombosis and Hemostasis, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Wenguo Li
- Cyrus Tang Hematology Center, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Prevention, Medical School, Soochow University, Suzhou, China
| | - Kun Wang
- Cyrus Tang Hematology Center, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Prevention, Medical School, Soochow University, Suzhou, China
| | - Ningzheng Dong
- Cyrus Tang Hematology Center, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Prevention, Medical School, Soochow University, Suzhou, China
- NHC Key Laboratory of Thrombosis and Hemostasis, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Qingyu Wu
- Cyrus Tang Hematology Center, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Prevention, Medical School, Soochow University, Suzhou, China
| |
Collapse
|
23
|
Wu Q. Natriuretic Peptide Signaling in Uterine Biology and Preeclampsia. Int J Mol Sci 2023; 24:12309. [PMID: 37569683 PMCID: PMC10418983 DOI: 10.3390/ijms241512309] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Revised: 07/30/2023] [Accepted: 07/31/2023] [Indexed: 08/13/2023] Open
Abstract
Endometrial decidualization is a uterine process essential for spiral artery remodeling, embryo implantation, and trophoblast invasion. Defects in endometrial decidualization and spiral artery remodeling are important contributing factors in preeclampsia, a major disorder in pregnancy. Atrial natriuretic peptide (ANP) is a cardiac hormone that regulates blood volume and pressure. ANP is also generated in non-cardiac tissues, such as the uterus and placenta. In recent human genome-wide association studies, multiple loci with genes involved in natriuretic peptide signaling are associated with gestational hypertension and preeclampsia. In cellular experiments and mouse models, uterine ANP has been shown to stimulate endometrial decidualization, increase TNF-related apoptosis-inducing ligand expression and secretion, and enhance apoptosis in arterial smooth muscle cells and endothelial cells. In placental trophoblasts, ANP stimulates adenosine 5'-monophosphate-activated protein kinase and the mammalian target of rapamycin complex 1 signaling, leading to autophagy inhibition and protein kinase N3 upregulation, thereby increasing trophoblast invasiveness. ANP deficiency impairs endometrial decidualization and spiral artery remodeling, causing a preeclampsia-like phenotype in mice. These findings indicate the importance of natriuretic peptide signaling in pregnancy. This review discusses the role of ANP in uterine biology and potential implications of impaired ANP signaling in preeclampsia.
Collapse
Affiliation(s)
- Qingyu Wu
- Cyrus Tang Hematology Center, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Prevention, Soochow University, Suzhou 215123, China
| |
Collapse
|
24
|
Guarino BD, Dado CD, Kumar A, Braza J, Harrington EO, Klinger JR. Deletion of the Npr3 gene increases severity of acute lung injury in obese mice. Pulm Circ 2023; 13:e12270. [PMID: 37528869 PMCID: PMC10387407 DOI: 10.1002/pul2.12270] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Revised: 07/05/2023] [Accepted: 07/09/2023] [Indexed: 08/03/2023] Open
Abstract
Previous studies have shown that atrial natriuretic peptide (ANP) attenuates agonist-induced pulmonary edema and that this effect may be mediated in part by the ANP clearance receptor, natriuretic peptide receptor-C (NPR-C). Obesity has been associated with lower plasma ANP levels due to increased expression of NPR-C, and with decreased severity of acute lung injury (ALI). Therefore, we hypothesized that increased expression of NPR-C may attenuate ALI severity in obese populations. To test this, we examined ALI in Npr3 wild-type (WT) and knockout (KO) mice fed normal chow (NC) or high-fat diets (HFD). After 12 weeks, ALI was induced with intra-tracheal administration of Pseudomonas aeruginosa strain 103 (PA103) or saline. ALI severity was determined by lung wet-to-dry ratio (W/D) along with measurement of cell count, protein levels from bronchoalveolar lavage fluid (BALF), and quantitative polymerase chain reaction was performed on whole lung to measure cytokine/chemokine and Npr3 mRNA expression. ANP levels were measured from plasma. PA103 caused ALI as determined by significant increases in W/D, BALF protein concentration, and whole lung cytokine/chemokine expression. PA103 increased Npr3 expression in the lungs of wild-type (WT) mice regardless of diet. There was a nonsignificant trend toward increased Npr3 expression in the lungs of WT mice fed HFD versus NC. No differences in ALI were seen between Npr3 knockout (KO) mice and WT-fed NC, but Npr3 KO mice fed HFD had a significantly greater W/D and BALF protein concentration than WT mice fed HFD. These findings support the hypothesis that Npr3 may help protect against ALI in obesity.
Collapse
Affiliation(s)
- Brianna D. Guarino
- Vascular Research LabProvidence Veterans Affairs Medical CenterProvidenceRhode IslandUSA
- Department of Medicine, Sleep and Critical Care MedicineRhode Island HospitalProvidenceRhode IslandUSA
- Warren Alpert Medical School of Brown UniversityProvidenceRhode IslandUSA
| | - Christopher D. Dado
- Vascular Research LabProvidence Veterans Affairs Medical CenterProvidenceRhode IslandUSA
- Department of Medicine, Sleep and Critical Care MedicineRhode Island HospitalProvidenceRhode IslandUSA
- Warren Alpert Medical School of Brown UniversityProvidenceRhode IslandUSA
| | - Ashok Kumar
- Vascular Research LabProvidence Veterans Affairs Medical CenterProvidenceRhode IslandUSA
- Warren Alpert Medical School of Brown UniversityProvidenceRhode IslandUSA
| | - Julie Braza
- Vascular Research LabProvidence Veterans Affairs Medical CenterProvidenceRhode IslandUSA
- Warren Alpert Medical School of Brown UniversityProvidenceRhode IslandUSA
| | - Elizabeth O. Harrington
- Vascular Research LabProvidence Veterans Affairs Medical CenterProvidenceRhode IslandUSA
- Department of Medicine, Sleep and Critical Care MedicineRhode Island HospitalProvidenceRhode IslandUSA
- Warren Alpert Medical School of Brown UniversityProvidenceRhode IslandUSA
| | - James R. Klinger
- Vascular Research LabProvidence Veterans Affairs Medical CenterProvidenceRhode IslandUSA
- Department of Medicine, Sleep and Critical Care MedicineRhode Island HospitalProvidenceRhode IslandUSA
- Warren Alpert Medical School of Brown UniversityProvidenceRhode IslandUSA
| |
Collapse
|
25
|
Thimm C, Erichsen L, Wruck W, Adjaye J. Unveiling Angiotensin II and Losartan-Induced Gene Regulatory Networks Using Human Urine-Derived Podocytes. Int J Mol Sci 2023; 24:10551. [PMID: 37445727 DOI: 10.3390/ijms241310551] [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: 06/07/2023] [Revised: 06/20/2023] [Accepted: 06/21/2023] [Indexed: 07/15/2023] Open
Abstract
Podocytes are highly specialized cells that play a pivotal role in the blood filtration process in the glomeruli of the kidney, and their dysfunction leads to renal diseases. For this reason, the study and application of this cell type is of great importance in the field of regenerative medicine. Hypertension is mainly regulated by the renin-angiotensin-aldosterone system (RAAS), with its main mediator being angiotensin II (ANG II). Elevated ANG II levels lead to a pro-fibrotic, inflammatory, and hypertrophic milieu that induces apoptosis in podocytes. The activation of RAAS is critical for the pathogenesis of podocyte injury; as such, to prevent podocyte damage, patients with hypertension are administered drugs that modulate RAAS signaling. A prime example is the orally active, non-peptide, selective angiotensin-II-type I receptor (AGTR1) blocker losartan. Here, we demonstrate that SIX2-positive urine-derived renal progenitor cells (UdRPCs) and their immortalized counterpart (UM51-hTERT) can be directly differentiated into mature podocytes. These podocytes show activation of RAAS after stimulation with ANG II, resulting in ANG II-dependent upregulation of the expression of the angiotensin-II-type I receptor, AGTR1, and the downregulated expression of the angiotensin-II-type II receptor 2 (AGTR2). The stimulation of podocytes with losartan counteracts ANG II-dependent changes, resulting in a dependent favoring of the specific receptor from AGTR1 to AGTR2. Transcriptome analysis revealed 94 losartan-induced genes associated with diverse biological processes and pathways such as vascular smooth muscle contraction, the oxytocin signaling pathway, renin secretion, and ECM-receptor interaction. Co-stimulation with losartan and ANG II induced the exclusive expression of 106 genes associated with DNA methylation or demethylation, cell differentiation, the developmental process, response to muscle stretch, and calcium ion transmembrane transport. These findings highlight the usefulness of UdRPC-derived podocytes in studying the RAAS pathway and nephrotoxicity in various kidney diseases.
Collapse
Affiliation(s)
- Chantelle Thimm
- Institute for Stem Cell Research and Regenerative Medicine, Medical Faculty, Heinrich-Heine University Düsseldorf, 40225 Düsseldorf, Germany
| | - Lars Erichsen
- Institute for Stem Cell Research and Regenerative Medicine, Medical Faculty, Heinrich-Heine University Düsseldorf, 40225 Düsseldorf, Germany
| | - Wasco Wruck
- Institute for Stem Cell Research and Regenerative Medicine, Medical Faculty, Heinrich-Heine University Düsseldorf, 40225 Düsseldorf, Germany
| | - James Adjaye
- Institute for Stem Cell Research and Regenerative Medicine, Medical Faculty, Heinrich-Heine University Düsseldorf, 40225 Düsseldorf, Germany
- EGA Institute for Women's Health, Zayed Centre for Research into Rare Diseases in Children (ZCR), University College London (UCL), 20 Guilford Street, London WC1N 1DZ, UK
| |
Collapse
|
26
|
Silva Cunha P, Antunes DO, Laranjo S, Coutinho A, Abecasis J, Oliveira MM. Case report: Mutation in NPPA gene as a cause of fibrotic atrial myopathy. Front Cardiovasc Med 2023; 10:1149717. [PMID: 37363091 PMCID: PMC10285104 DOI: 10.3389/fcvm.2023.1149717] [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: 01/22/2023] [Accepted: 05/19/2023] [Indexed: 06/28/2023] Open
Abstract
Early-onset atrial fibrillation (AF) can be the manifestation of a genetic atrial myopathy. However, specific genetic identification of a mutation causing atrial fibrosis is rare. We report a case of a young patient with an asymptomatic AF, diagnosed during a routine examination. The cardiac MRI revealed extensive atrial fibrosis and the electrophysiology study showed extensive areas of low voltage. The genetic investigation identified a homozygous pathogenic variant in the NPPA gene in the index case and the presence of the variant in heterozygosity in both parents.
Collapse
Affiliation(s)
- Pedro Silva Cunha
- Arrhythmology, Pacing and Electrophysiology Unit, Cardiology Service, Santa Marta Hospital, Centro Hospitalar Universitário Lisboa Central, Lisbon, Portugal
- Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
- Cardiovascular Department, Hospital Lusíadas Lisboa, Lisbon, Portugal
| | - Diana Oliveira Antunes
- Genetics Department, Hospital Dona Estefânia, Centro Hospitalar Universitário Lisboa Central, Lisbon, Portugal
- GenoMed Diagnóstico Medicina Molecular, Instituto de Medicina Molecular, Lisbon, Portugal
| | - Sérgio Laranjo
- Arrhythmology, Pacing and Electrophysiology Unit, Cardiology Service, Santa Marta Hospital, Centro Hospitalar Universitário Lisboa Central, Lisbon, Portugal
- NOVA Medical School, Universidade NOVA de Lisboa, Lisbon, Portugal
| | - Ana Coutinho
- GenoMed Diagnóstico Medicina Molecular, Instituto de Medicina Molecular, Lisbon, Portugal
| | - João Abecasis
- Cardiovascular Department, Hospital Lusíadas Lisboa, Lisbon, Portugal
- NOVA Medical School, Universidade NOVA de Lisboa, Lisbon, Portugal
| | - Mário Martins Oliveira
- Arrhythmology, Pacing and Electrophysiology Unit, Cardiology Service, Santa Marta Hospital, Centro Hospitalar Universitário Lisboa Central, Lisbon, Portugal
- Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| |
Collapse
|
27
|
MacDonald BT, Elowe NH, Garvie CW, Kaushik VK, Ellinor PT. Identification of a new Corin atrial natriuretic peptide-converting enzyme substrate: Agouti-signaling protein (ASIP). BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.04.26.538495. [PMID: 37162877 PMCID: PMC10168342 DOI: 10.1101/2023.04.26.538495] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/11/2023]
Abstract
Corin is a transmembrane tethered enzyme best known for processing the hormone atrial natriuretic peptide (ANP) in cardiomyocytes to control electrolyte balance and blood pressure. Loss of function mutations in Corin prevent ANP processing and lead to hypertension. Curiously, Corin loss of function variants also result in lighter coat color pigmentation in multiple species. Corin pigmentation effects are dependent on a functional Agouti locus encoding the agouti-signaling protein (ASIP) based on a genetic interaction. However, the nature of this conserved role of Corin has not been defined. Here we report that ASIP is a direct proteolytic substrate of the Corin enzyme.
Collapse
Affiliation(s)
- Bryan T. MacDonald
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA
| | - Nadine H. Elowe
- Center for the Development of Therapeutics, Broad Institute of MIT and Harvard, Cambridge, MA
| | - Colin W. Garvie
- Center for the Development of Therapeutics, Broad Institute of MIT and Harvard, Cambridge, MA
| | - Virendar K. Kaushik
- Center for the Development of Therapeutics, Broad Institute of MIT and Harvard, Cambridge, MA
| | - Patrick T. Ellinor
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA
| |
Collapse
|
28
|
Postmus AC, Kruit JK, Eilers RE, Havinga R, Koster MH, Johmura Y, Nakanishi M, van de Sluis B, Jonker JW. The chemotherapeutic drug doxorubicin does not exacerbate p16 Ink4a-positive senescent cell accumulation and cardiometabolic disease development in young adult female LDLR-deficient mice. Toxicol Appl Pharmacol 2023; 468:116531. [PMID: 37088304 DOI: 10.1016/j.taap.2023.116531] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Revised: 04/05/2023] [Accepted: 04/19/2023] [Indexed: 04/25/2023]
Abstract
Cancer survivors who received chemotherapy, such as the anthracycline doxorubicin (DOX), have an increased risk of developing complications later in life, including the development of chronic metabolic diseases. Although the etiology of this increased risk for late metabolic complications in cancer survivors is poorly understood, a causal role of therapy-induced senescent cells has been suggested. To study the role of cellular senescence in chemotherapy-induced metabolic complications, young adult female low-density lipoprotein receptor-deficient (Ldlr-/-)-p16-3MR mice, in which p16Ink4a-positive (p16Ink4a+) senescent cells can be genetically eliminated, were treated with four weekly injections of DOX (2.5 mg/kg) followed by a high-fat high-cholesterol diet for 12 weeks. While DOX treatment induced known short-term effects, such as reduction in body weight, gonadal fat mass, and adipose tissue inflammation, it was not associated with significant long-term effects on glucose homeostasis, hepatic steatosis, or atherosclerosis. We further found no evidence of DOX-induced accumulation of p16Ink4a+-senescent cells at 1 or 12 weeks after DOX treatment. Neither did we observe an effect of elimination of p16Ink4a+-senescent cells on the development of diet-induced cardiometabolic complications in DOX-treated mice. Other markers for senescence were generally also not affected except for an increase in p21 and Cxcl10 in gonadal white adipose tissue long-term after DOX treatment. Together, our study does not support a significant role for p16Ink4a+-senescent cells in the development of diet-induced cardiometabolic disease in young adult DOX-treated female Ldlr-/- mice. These findings illustrate the need of further studies to understand the link between cancer therapy and cardiometabolic disease development in cancer survivors.
Collapse
Affiliation(s)
- Andrea C Postmus
- Department of Pediatrics, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9713 GZ Groningen, the Netherlands
| | - Janine K Kruit
- Department of Pediatrics, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9713 GZ Groningen, the Netherlands
| | - Roos E Eilers
- Department of Pediatrics, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9713 GZ Groningen, the Netherlands
| | - Rick Havinga
- Department of Pediatrics, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9713 GZ Groningen, the Netherlands
| | - Mirjam H Koster
- Department of Pediatrics, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9713 GZ Groningen, the Netherlands
| | - Yoshikazu Johmura
- Division of Cancer and Senescence Biology, Cancer Research Institute, Institute for Frontier Science Initiative, Kanazawa University, Kakuma, Kanazawa, Japan
| | - Makoto Nakanishi
- Division of Cancer Cell Biology, Institute of Medical Science, University of Tokyo, Tokyo, Japan
| | - Bart van de Sluis
- Department of Pediatrics, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9713 GZ Groningen, the Netherlands
| | - Johan W Jonker
- Department of Pediatrics, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9713 GZ Groningen, the Netherlands.
| |
Collapse
|
29
|
Custodia A, Aramburu-Núñez M, Rodríguez-Arrizabalaga M, Pías-Peleteiro JM, Vázquez-Vázquez L, Camino-Castiñeiras J, Aldrey JM, Castillo J, Ouro A, Sobrino T, Romaus-Sanjurjo D. Biomarkers Assessing Endothelial Dysfunction in Alzheimer's Disease. Cells 2023; 12:cells12060962. [PMID: 36980302 PMCID: PMC10047803 DOI: 10.3390/cells12060962] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 03/07/2023] [Accepted: 03/10/2023] [Indexed: 03/30/2023] Open
Abstract
Alzheimer's disease (AD) is the most common degenerative disorder in the elderly in developed countries. Currently, growing evidence is pointing at endothelial dysfunction as a key player in the cognitive decline course of AD. As a main component of the blood-brain barrier (BBB), the dysfunction of endothelial cells driven by vascular risk factors associated with AD allows the passage of toxic substances to the cerebral parenchyma, producing chronic hypoperfusion that eventually causes an inflammatory and neurotoxic response. In this process, the levels of several biomarkers are disrupted, such as an increase in adhesion molecules that allow the passage of leukocytes to the cerebral parenchyma, increasing the permeability of the BBB; moreover, other vascular players, including endothelin-1, also mediate artery inflammation. As a consequence of the disruption of the BBB, a progressive neuroinflammatory response is produced that, added to the astrogliosis, eventually triggers neuronal degeneration (possibly responsible for cognitive deterioration). Recently, new molecules have been proposed as early biomarkers for endothelial dysfunction that can constitute new therapeutic targets as well as early diagnostic and prognostic markers for AD.
Collapse
Affiliation(s)
- Antía Custodia
- NeuroAging Group (NEURAL), Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), 15706 Santiago de Compostela, Spain
- Centro de Investigación Biomédica en Red en Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Marta Aramburu-Núñez
- NeuroAging Group (NEURAL), Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), 15706 Santiago de Compostela, Spain
- Centro de Investigación Biomédica en Red en Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Mariña Rodríguez-Arrizabalaga
- NeuroAging Group (NEURAL), Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), 15706 Santiago de Compostela, Spain
| | - Juan Manuel Pías-Peleteiro
- NeuroAging Group (NEURAL), Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), 15706 Santiago de Compostela, Spain
- Centro de Investigación Biomédica en Red en Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Laura Vázquez-Vázquez
- NeuroAging Group (NEURAL), Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), 15706 Santiago de Compostela, Spain
- Centro de Investigación Biomédica en Red en Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Javier Camino-Castiñeiras
- NeuroAging Group (NEURAL), Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), 15706 Santiago de Compostela, Spain
- Centro de Investigación Biomédica en Red en Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - José Manuel Aldrey
- NeuroAging Group (NEURAL), Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), 15706 Santiago de Compostela, Spain
- Centro de Investigación Biomédica en Red en Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - José Castillo
- Neuroimaging and Biotechnology Laboratory (NOBEL), Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), 15706 Santiago de Compostela, Spain
| | - Alberto Ouro
- NeuroAging Group (NEURAL), Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), 15706 Santiago de Compostela, Spain
- Centro de Investigación Biomédica en Red en Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Tomás Sobrino
- NeuroAging Group (NEURAL), Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), 15706 Santiago de Compostela, Spain
- Centro de Investigación Biomédica en Red en Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Daniel Romaus-Sanjurjo
- NeuroAging Group (NEURAL), Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), 15706 Santiago de Compostela, Spain
- Centro de Investigación Biomédica en Red en Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, 28029 Madrid, Spain
| |
Collapse
|
30
|
Hatthakone T, Oundavong S, Soejima Y, Sawabe M. Development of a new histological identification method of human sinoatrial node suitable for immunohistochemical study. Anat Sci Int 2023; 98:293-305. [PMID: 36422826 DOI: 10.1007/s12565-022-00697-0] [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: 07/12/2022] [Accepted: 11/07/2022] [Indexed: 11/27/2022]
Abstract
Histological identification of the human sinoatrial node (SAN) remains a challenge. Conventional identification methods, such as Lev's method, have certain limitations. The aim of our study was to develop a new histological identification method that could properly identify the sinoatrial node, applicable to the immunohistochemical study of intra-nodal structures. Thirty-nine human autopsied hearts were included in this study. The cases included 23 men and 16 women ranging in age from 20 to 99 years. The sinoatrial area from eight control samples was cut in the vertical section using the conventional Lev's method. In our new method, called the "En face one-block method," the sinoatrial node was cut in "En face" at the junction of the right border of the right appendage and superior vena cava, placed in one long cassette, and serially cut using a microtome. Immunostaining was performed using primary antibodies against CD31, podoplanin (D2-40), S-100, and other proteins. The average area of the SAN on the slide glass in our new method was 32.2 mm2, which was significantly larger than that (3.59 mm2) of the control samples by Lev's method. The SAN area was positively correlated with age (r = 0.357; p = 0.026), especially in women (r = 0.626; p = 0.0095). The SAN group had significantly lower percentage of CD31-positive blood capillaries, higher percentage of podoplanin-positive lymphatic channels, and S-100-positive peripheral nerves. We successfully developed a novel cutting method applicable to immunohistochemical studies, with which we could provide a bird's-eye view of the sinoatrial nodes.
Collapse
Affiliation(s)
- Thavisouk Hatthakone
- Department of Molecular Pathology, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo, Tokyo, 113-8510, Japan
| | - Sunti Oundavong
- Department of Molecular Pathology, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo, Tokyo, 113-8510, Japan
| | - Yurie Soejima
- Department of Molecular Pathology, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo, Tokyo, 113-8510, Japan
| | - Motoji Sawabe
- Department of Molecular Pathology, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo, Tokyo, 113-8510, Japan.
- Department of Molecular Pathology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo, Tokyo, 113-8519, Japan.
- Department of Diagnostic Pathology, Hitachi General Hospital, 2-1-1 Jonancho, Hitachi, Ibaraki, 317-0077, Japan.
| |
Collapse
|
31
|
Hajializadeh Z, Khaksari M, Dabiri S, Darvishzadeh Mahani F, Raji-Amirhasani A, Bejeshk MA. Protective effects of calorie restriction and 17-β estradiol on cardiac hypertrophy in ovariectomized obese rats. PLoS One 2023; 18:e0282089. [PMID: 37098007 PMCID: PMC10128952 DOI: 10.1371/journal.pone.0282089] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Accepted: 02/06/2023] [Indexed: 04/26/2023] Open
Abstract
Obesity and menopause lead to cardiovascular diseases. Calorie restriction (CR) can modulate estrogen deficiency and obesity-related cardiovascular diseases. The protective effects of CR and estradiol on cardiac hypertrophy in ovariectomized obese rats were explored in this study. The adult female Wistar rats were divided into sham and ovariectomized (OVX) groups that received a high-fat diet (60% HFD) or standard diet (SD) or 30% CR for 16 weeks, and then, 1mg/kg E2 (17-β estradiol) was injected intraperitoneally every 4 days for four weeks in OVX-rats. Hemodynamic parameters were evaluated before and after each diet. Heart tissues were collected for biochemical, histological, and molecular analysis. HFD consumption led to weight gain in sham and OVX rats. In contrast, CR and E2 led to body weight loss in these animals. Also, heart weight (HW), heart weight/body weight (HW/BW) ratio, and left ventricular weight (LVW) were enhanced in OVX rats that received SD and HFD. E2 reduced these indexes in both diet conditions but reduction effects of CR were seen only in HFD groups. HFD and SD feeding increased hemodynamic parameters, ANP (atrial natriuretic peptide) mRNA expression, and TGF-β1(transforming growth factor-beta 1) protein level in the OVX animals, while CR and E2 reduced these factors. Cardiomyocyte diameter and hydroxyproline content were increased in the OVX-HFD groups. Nevertheless, CR and E2 decreased these indicators. The results showed that CR and E2 treatment reduced obesity-induced-cardiac hypertrophy in ovariectomized groups (20% and 24% respectively). CR appears to have almost as reducing effects as estrogen therapy on cardiac hypertrophy. The findings suggest that CR can be considered a therapeutic candidate for postmenopausal cardiovascular disease.
Collapse
Affiliation(s)
- Zahra Hajializadeh
- Physiology Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
| | - Mohammad Khaksari
- Endocrinology and Metabolism Research Center, Institute of Basic and Clinical Physiology Sciences, Kerman University of Medical Sciences Kerman, Kerman, Iran
| | - Shahriar Dabiri
- Pathology and Stem Cell Research Center, Kerman University of Medical Sciences, Kerman, Iran
| | - Fatemeh Darvishzadeh Mahani
- Endocrinology and Metabolism Research Center, Institute of Basic and Clinical Physiology Sciences, Kerman University of Medical Sciences Kerman, Kerman, Iran
| | - Alireza Raji-Amirhasani
- Endocrinology and Metabolism Research Center, Institute of Basic and Clinical Physiology Sciences, Kerman University of Medical Sciences Kerman, Kerman, Iran
| | - Mohammad Abbas Bejeshk
- Department of Physiology and Pharmacology, Afzalipour Faculty of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| |
Collapse
|
32
|
Choudhary MI, Römling U, Nadeem F, Bilal HM, Zafar M, Jahan H, ur-Rahman A. Innovative Strategies to Overcome Antimicrobial Resistance and Tolerance. Microorganisms 2022; 11:microorganisms11010016. [PMID: 36677308 PMCID: PMC9863313 DOI: 10.3390/microorganisms11010016] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Revised: 12/15/2022] [Accepted: 12/16/2022] [Indexed: 12/24/2022] Open
Abstract
Antimicrobial resistance and tolerance are natural phenomena that arose due to evolutionary adaptation of microorganisms against various xenobiotic agents. These adaptation mechanisms make the current treatment options challenging as it is increasingly difficult to treat a broad range of infections, associated biofilm formation, intracellular and host adapted microbes, as well as persister cells and microbes in protected niches. Therefore, novel strategies are needed to identify the most promising drug targets to overcome the existing hurdles in the treatment of infectious diseases. Furthermore, discovery of novel drug candidates is also much needed, as few novel antimicrobial drugs have been introduced in the last two decades. In this review, we focus on the strategies that may help in the development of innovative small molecules which can interfere with microbial resistance mechanisms. We also highlight the recent advances in optimization of growth media which mimic host conditions and genome scale molecular analyses of microbial response against antimicrobial agents. Furthermore, we discuss the identification of antibiofilm molecules and their mechanisms of action in the light of the distinct physiology and metabolism of biofilm cells. This review thus provides the most recent advances in host mimicking growth media for effective drug discovery and development of antimicrobial and antibiofilm agents.
Collapse
Affiliation(s)
- M. Iqbal Choudhary
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Pakistan
- H. E. J. Research Institute of Chemistry, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Pakistan
| | - Ute Römling
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, 171 65 Stockholm, Sweden
- Correspondence: (U.R.); (H.J.); Tel.: +46-8-5248-7319 (U.R.); +92-21-111-232-292 (ext. 301) (H.J.)
| | - Faiza Nadeem
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Pakistan
| | - Hafiz Muhammad Bilal
- H. E. J. Research Institute of Chemistry, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Pakistan
| | - Munirah Zafar
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Pakistan
| | - Humera Jahan
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Pakistan
- Correspondence: (U.R.); (H.J.); Tel.: +46-8-5248-7319 (U.R.); +92-21-111-232-292 (ext. 301) (H.J.)
| | - Atta ur-Rahman
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Pakistan
- H. E. J. Research Institute of Chemistry, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Pakistan
| |
Collapse
|
33
|
Long C, Liu H, Zhan W, Chen L, Wu A, Yang L, Chen S. Null Function of Npr1 Disturbs Immune Response in Colonic Inflammation During Early Postnatal Stage. Inflammation 2022; 45:2419-2432. [PMID: 35794311 PMCID: PMC9646613 DOI: 10.1007/s10753-022-01702-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Revised: 06/01/2022] [Accepted: 06/06/2022] [Indexed: 12/24/2022]
Abstract
Natriuretic peptide receptor 1 (NPR1) is conventionally known as a regulator of vascular homeostasis. Here, we generated an Npr1 knockout mouse model with CRISPR/Cas9 technology and found that homozygous mice (Npr1-/-) exhibited weight loss and poor survival rate during early postnatal stage. Careful examination revealed unexpectedly that Npr1-/- mice developed colitis characterized by shortened colon, evident colonic mucosal damage, increased histopathological score, and higher colonic expression of proinflammatory cytokines interleukin-1B (IL1B) and -6 (IL6). RNA-sequencing analysis revealed that differentially expressed genes were prominently enriched in the biological pathways related to immune response in both spleen and colon of Npr1-/- mice. Cytofluorimetric analysis demonstrated that leukocytes in the spleen were significantly increased, particularly, the populations of neutrophil and CD3+ T cell were elevated but CD4+ T cells were decreased in Npr1-/- mice. Administration of 8-Br-cGMP, a downstream activator of NPR1, restored these immune-cell populations disturbed in Npr1-/- mice and lessened the colitis-related phenotypes. To validate the involvement of Npr1 in colitis, we examined another mouse model induced by dextran sodium sulfate (DSS) and found a decreased Npr1 expression and shifted immune-cell populations as well. Importantly, 8-Br-cGMP treatment exhibited a similar effect in the restoration of immune-cell populations and attenuation of colonic inflammation in DSS mice. Our data indicate that loss of Npr1 possibly interrupts immune response, which is critical to the pathogenesis of colitis in the early life.
Collapse
Affiliation(s)
- Changkun Long
- Vascular Function Laboratory, Human Aging Research Institute and School of Life Science, and Jiangxi Key Laboratory of Human Aging, Nanchang University, Nanchang, 330031, China
| | - Hongfei Liu
- Vascular Function Laboratory, Human Aging Research Institute and School of Life Science, and Jiangxi Key Laboratory of Human Aging, Nanchang University, Nanchang, 330031, China
| | - Wenxing Zhan
- Vascular Function Laboratory, Human Aging Research Institute and School of Life Science, and Jiangxi Key Laboratory of Human Aging, Nanchang University, Nanchang, 330031, China
| | - Liping Chen
- Vascular Function Laboratory, Human Aging Research Institute and School of Life Science, and Jiangxi Key Laboratory of Human Aging, Nanchang University, Nanchang, 330031, China
| | - Andong Wu
- Aging and Vascular DiseasesSchool of Life Scienceand Jiangxi Key Laboratory of Human Aging, Human Aging Research Institute, Nanchang University, Nanchang, 330031, China
| | - Lin Yang
- Department of Nephrology, Jiangxi Provincial People's Hospital, Affiliated to Nanchang University, Nanchang, 330006, China
| | - Shenghan Chen
- Vascular Function Laboratory, Human Aging Research Institute and School of Life Science, and Jiangxi Key Laboratory of Human Aging, Nanchang University, Nanchang, 330031, China.
| |
Collapse
|
34
|
Hajibabaie F, Abedpoor N, Safavi K, Taghian F. Natural remedies medicine derived from flaxseed (secoisolariciresinol diglucoside, lignans, and α-linolenic acid) improve network targeting efficiency of diabetic heart conditions based on computational chemistry techniques and pharmacophore modeling. J Food Biochem 2022; 46:e14480. [PMID: 36239429 DOI: 10.1111/jfbc.14480] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Revised: 08/31/2022] [Accepted: 09/29/2022] [Indexed: 01/14/2023]
Abstract
Cytokine storms lead to cardiovascular diseases (CVDs). Natural herbal compounds are considered the primary source of active agents with the potential to prevent or treat inflammatory-related pathologies such as CVD and diabetes. Flaxseed contains phytochemicals, including secoisolariciresinol diglucoside (SDG), α-linolenic acid (ALA), and lignans, termed "SAL." Hence, we evaluated the effect of the SAL on the H9c2 cardiac cells in hyperlipidemic and hyperglycemic conditions. Here, candidate hub genes, TNF-α, IL6, SIRT1, NRF1, NPPA, and FGF7, were selected as effective genes in diabetic cardiovascular pathogenesis based on in-silico analysis and chemoinformatic. Myocardial infarction (MI) was induced using H9c2 cardiac cells in hyperlipidemic and hyperglycemic conditions. Real-time qPCR was conducted to assess the expression level of hub genes. This study indicated that SAL compounds bound to the Il-6, SIRT1, and TNF-α active sites as druggable candidate proteins based on the chemoinformatics analysis. This study displayed that the TNF-α, IL6, SIRT1, NRF1, NPPA, and FGF7 network dysfunction in MI models were ameliorated by SAL consumption. Furthermore, SAL compounds improved the function and myogenesis of H9c2 cells in hyperlipidemic and hyperglycemic conditions. Our data suggested that phytochemicals obtained from flaxseed might have proposed potential complementary treatment or preventive strategies for MI. PRACTICAL APPLICATIONS: Phytochemicals obtained from flaxseed (SAL) could reverse diabetic heart dysfunction hallmarks and provide new potential treatment approaches in cardiovascular therapy. SAL could be considered complementary and alternative medicines for treating various disorders/diseases singly or synchronizing with prescription drugs.
Collapse
Affiliation(s)
- Fatemeh Hajibabaie
- Department of Physiology, Medicinal Plants Research Center, Isfahan (Khorasgan) Branch, Islamic Azad University, Isfahan, Iran
| | - Navid Abedpoor
- Department of Physiology, Medicinal Plants Research Center, Isfahan (Khorasgan) Branch, Islamic Azad University, Isfahan, Iran
| | - Kamran Safavi
- Department of Plant Biotechnology, Medicinal Plants Research Centre, Isfahan (Khorasgan) Branch, Islamic Azad University, Isfahan, Iran
| | - Farzaneh Taghian
- Department of Sports Physiology, Faculty of Sports Sciences, Isfahan (Khorasgan) Branch, Islamic Azad University, Isfahan, Iran
| |
Collapse
|
35
|
Proprotein Convertase Subtilisin/Kexin 6 in Cardiovascular Biology and Disease. Int J Mol Sci 2022; 23:ijms232113429. [DOI: 10.3390/ijms232113429] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Revised: 11/01/2022] [Accepted: 11/02/2022] [Indexed: 11/06/2022] Open
Abstract
Proprotein convertase subtilisin/kexin 6 (PCSK6) is a secreted serine protease expressed in most major organs, where it cleaves a wide range of growth factors, signaling molecules, peptide hormones, proteolytic enzymes, and adhesion proteins. Studies in Pcsk6-deficient mice have demonstrated the importance of Pcsk6 in embryonic development, body axis specification, ovarian function, and extracellular matrix remodeling in articular cartilage. In the cardiovascular system, PCSK6 acts as a key modulator in heart formation, lipoprotein metabolism, body fluid homeostasis, cardiac repair, and vascular remodeling. To date, dysregulated PCSK6 expression or function has been implicated in major cardiovascular diseases, including atrial septal defects, hypertension, atherosclerosis, myocardial infarction, and cardiac aging. In this review, we describe biochemical characteristics and posttranslational modifications of PCSK6. Moreover, we discuss the role of PCSK6 and related molecular mechanisms in cardiovascular biology and disease.
Collapse
|
36
|
Fatty Acid Amide Hydrolase Deficiency Is Associated with Deleterious Cardiac Effects after Myocardial Ischemia and Reperfusion in Mice. Int J Mol Sci 2022; 23:ijms232012690. [PMID: 36293543 PMCID: PMC9604059 DOI: 10.3390/ijms232012690] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 10/11/2022] [Accepted: 10/19/2022] [Indexed: 11/06/2022] Open
Abstract
Ischemic cardiomyopathy leads to inflammation and left ventricular (LV) dysfunction. Animal studies provided evidence for cardioprotective effects of the endocannabinoid system, including cardiomyocyte adaptation, inflammation, and remodeling. Cannabinoid type-2 receptor (CB2) deficiency led to increased apoptosis and infarctions with worsened LV function in ischemic cardiomyopathy. The aim of our study was to investigate a possible cardioprotective effect of endocannabinoid anandamide (AEA) after ischemia and reperfusion (I/R). Therefore, fatty acid amide hydrolase deficient (FAAH)−/− mice were subjected to repetitive, daily, 15 min, left anterior descending artery (LAD) occlusion over 3 and 7 consecutive days. Interestingly, FAAH−/− mice showed stigmata such as enhanced inflammation, cardiomyocyte loss, stronger remodeling, and persistent scar with deteriorated LV function compared to wild-type (WT) littermates. As endocannabinoids also activate PPAR-α (peroxisome proliferator-activated receptor), PPAR-α mediated effects of AEA were eliminated with PPAR-α antagonist GW6471 i.v. in FAAH−/− mice. LV function was assessed using M-mode echocardiography. Immunohistochemical analysis revealed apoptosis, macrophage accumulation, collagen deposition, and remodeling. Hypertrophy was determined by cardiomyocyte area and heart weight/tibia length. Molecular analyses involved Taqman® RT-qPCR and immune cells were analyzed with fluorescence-activated cell sorting (FACS). Most importantly, collagen deposition was reduced to WT levels when FAAH−/− mice were treated with GW6471. Chemokine ligand-2 (CCL2) expression was significantly higher in FAAH−/− mice compared to WT, followed by higher macrophage infiltration in infarcted areas, both being reversed by GW6471 treatment. Besides restoring antioxidative properties and contractile elements, PPAR-α antagonism also reversed hypertrophy and remodeling in FAAH−/− mice. Finally, FAAH−/−-mice showed more substantial downregulation of PPAR-α compared to WT, suggesting a compensatory mechanism as endocannabinoids are also ligands for PPAR-α, and its activation causes lipotoxicity leading to cardiomyocyte apoptosis. Our study gives novel insights into the role of endocannabinoids acting via PPAR-α. We hypothesize that the increase in endocannabinoids may have partially detrimental effects on cardiomyocyte survival due to PPAR-α activation.
Collapse
|
37
|
Long C, Liu H, Zhan W, Chen L, Yu Z, Tian S, Xiang Y, Chen S, Tian X. Chronological attenuation of NPRA/PKG/AMPK signaling promotes vascular aging and elevates blood pressure. Aging Cell 2022; 21:e13699. [PMID: 36016499 PMCID: PMC9470896 DOI: 10.1111/acel.13699] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Revised: 07/09/2022] [Accepted: 08/02/2022] [Indexed: 01/25/2023] Open
Abstract
Hypertension is common in elderly population. We designed to search comprehensively for genes that are chronologically shifted in their expressions and to define their contributions to vascular aging and hypertension. RNA sequencing was conducted to search for senescence-shifted transcripts in human umbilical vein endothelial cells (HUVECs). Small interfering RNA (siRNA), small-molecule drugs, CRISPR/Cas9 techniques, and imaging were used to determine genes' function and contributions to age-related phenotypes of the endothelial cell and blood vessel. Of 25 genes enriched in the term of "regulation of blood pressure," NPRA was changed most significantly. The decreased NPRA expression was replicated in aortas of aged mice. The knockdown of NPRA promoted HUVEC senescence and it decreased expressions of protein kinase cGMP-dependent 1 (PKG), sirtuin 1 (SIRT1), and endothelial nitric oxide synthase (eNOS). Suppression of NPRA also decreased the phosphorylation of AMP-activated protein kinase (AMPK) as well as the ratio of oxidized nicotinamide adenine dinucleotide (NAD+ )/reduced nicotinamide adenine dinucleotide (NADH) but increased the production of reactive oxygen species (ROS). 8-Br-cGMP (analog of cGMP), or AICAR (AMPK activator), counteracted the observed changes in HUVECs. The Npr1+/- mice presented an elevated systolic blood pressure and their vessels became insensitive to endothelial-dependent vasodilators. Further, vessels from Npr1+/- mice increased Cdkn1a but decreased eNos expressions. These phenotypes were rescued by intravenously administrated 8-Br-cGMP and viral overexpression of human PKG, respectively. In conclusion, we demonstrate NPRA/PKG/AMPK as a novel and critical signaling axis in the modulation of endothelial cell senescence, vascular aging, and hypertension.
Collapse
Affiliation(s)
- Changkun Long
- Vascular Function LaboratoryHuman Aging Research Institute and School of Life Science, Nanchang university, and Jiangxi Key Laboratory of Human AgingNanchangChina
| | - Hongfei Liu
- Vascular Function LaboratoryHuman Aging Research Institute and School of Life Science, Nanchang university, and Jiangxi Key Laboratory of Human AgingNanchangChina
| | - Wenxing Zhan
- Vascular Function LaboratoryHuman Aging Research Institute and School of Life Science, Nanchang university, and Jiangxi Key Laboratory of Human AgingNanchangChina
| | - Liping Chen
- Vascular Function LaboratoryHuman Aging Research Institute and School of Life Science, Nanchang university, and Jiangxi Key Laboratory of Human AgingNanchangChina
| | - Zhenping Yu
- Institute of Translational MedicineNanchang UniversityNanchangChina,School of Life Science, Nanchang UniversityNanchangChina
| | - Shane Tian
- Department of Biochemistry/ChemistryOhio State UniversityColumbusOhioUSA
| | - Yang Xiang
- Metabolic Control and AgingHuman Aging Research Institute and School of Life Science, Nanchang university, and Jiangxi Key Laboratory of Human AgingNanchangChina
| | - Shenghan Chen
- Vascular Function LaboratoryHuman Aging Research Institute and School of Life Science, Nanchang university, and Jiangxi Key Laboratory of Human AgingNanchangChina
| | - Xiao‐Li Tian
- Aging and Vascular DiseasesHuman Aging Research Institute and School of Life Science, Nanchang university, and Jiangxi Key Laboratory of Human AgingNanchangChina
| |
Collapse
|
38
|
Liu X, Chen Q, Ji X, Yu W, Wang T, Han J, Li S, Liu J, Zeng F, Zhao Y, Zhang Y, Luo Q, Wang S, Wang F. Astragaloside IV promotes pharmacological effect of Descurainia sophia seeds on isoproterenol-induced cardiomyopathy in rats by synergistically modulating the myosin motor. Front Pharmacol 2022; 13:939483. [PMID: 36034815 PMCID: PMC9403516 DOI: 10.3389/fphar.2022.939483] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Accepted: 07/12/2022] [Indexed: 11/23/2022] Open
Abstract
Descurainia sophia seeds (DS), Astragalus mongholicus (AM), and their formulas are widely used to treat heart failure caused by various cardiac diseases in traditional Chinese medicine practice. However, the molecular mechanism of action of DS and AM has not been completely understood. Herein, we first used mass spectrometry coupled to UPLC to characterize the chemical components of DS and AM decoctions, then applied MS-based quantitative proteomic analysis to profile protein expression in the heart of rats with isoproterenol-induced cardiomyopathy (ISO-iCM) before and after treated with DS alone or combined with AM, astragaloside IV (AS4), calycosin-7-glucoside (C7G), and Astragalus polysaccharides (APS) from AM. We demonstrated for the first time that DS decoction alone could reverse the most of differentially expressed proteins in the heart of the rats with ISO-iCM, including the commonly recognized biomarkers natriuretic peptides (NPPA) of cardiomyopathy and sarcomeric myosin light chain 4 (MYL4), relieving ISO-iCM in rats, but AM did not pronouncedly improve the pharmacological efficiency of DS. Significantly, we revealed that AS4 remarkably promoted the pharmacological potency of DS by complementarily reversing myosin motor MYH6/7, and further downregulating NPPA and MYL4. In contrast, APS reduced the efficiency of DS due to upregulating NPPA and MYL4. These findings not only provide novel insights to better understanding in the combination principle of traditional Chinese medicine but also highlight the power of mass spectrometric proteomics strategy combined with conventional pathological approaches for the traditional medicine research.
Collapse
|
39
|
Atrial Natriuretic Peptide (ANP) Suppress λ-Carrageenan-Induced Acute Paw Edema by Modulating Pro-inflammatory Genes in Swiss Albino Mice. Int J Pept Res Ther 2022. [DOI: 10.1007/s10989-022-10444-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/16/2022]
|
40
|
Klemens CA, Dissanayake LV, Levchenko V, Zietara A, Palygin O, Staruschenko A. Modulation of blood pressure regulatory genes in the Agtrap-Plod1 locus associated with a deletion in Clcn6. Physiol Rep 2022; 10:e15417. [PMID: 35927940 PMCID: PMC9353118 DOI: 10.14814/phy2.15417] [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: 06/10/2022] [Revised: 07/15/2022] [Accepted: 07/18/2022] [Indexed: 06/15/2023] Open
Abstract
The AGTRAP-PLOD1 locus is a conserved gene cluster containing several blood pressure regulatory genes, including CLCN6, MTHFR, NPPA, and NPPB. Previous work revealed that knockout of Clcn6 on the Dahl Salt-Sensitive (SS) rat background (SS-Clcn6) resulted in lower diastolic blood pressure compared to SS-WT rats. Additionally, a recent study found sickle cell anemia patients with mutations in CLCN6 had improved survival and reduced stroke risk. We investigated whether loss of Clcn6 would delay the mortality of Dahl SS rats on an 8% NaCl (HS) diet. No significant difference in survival was found. The ability of Clcn6 to affect mRNA expression of nearby Mthfr, Nppa, and Nppb genes was also tested. On normal salt (0.4% NaCl, NS) diets, renal Mthfr mRNA and protein expression were significantly increased in the SS-Clcn6 rats. MTHFR reduces homocysteine to methionine, but no differences in circulating homocysteine levels were detected. Nppa mRNA levels in cardiac tissue from SS-Clcn6 rat in both normotensive and hypertensive conditions were significantly reduced compared to SS-WT. Nppb mRNA expression in SS-Clcn6 rats on a NS diet was also substantially decreased. Heightened Mthfr expression would be predicted to be protective; however, diminished Nppa and Nppb expression could be deleterious and by preventing or blunting vasodilation, natriuresis, and diuresis that ought to normally occur to offset blood pressure increases. The conserved nature of this genetic locus in humans and rats suggests more studies are warranted to understand how mutations in and around these genes may be influencing the expression of their neighbors.
Collapse
Affiliation(s)
- Christine A. Klemens
- Department of Molecular Pharmacology and PhysiologyUniversity of South FloridaTampaFloridaUSA
- Hypertension and Kidney Research CenterUniversity of South FloridaTampaFloridaUSA
| | - Lashodya V. Dissanayake
- Department of Molecular Pharmacology and PhysiologyUniversity of South FloridaTampaFloridaUSA
- Department of PhysiologyMedical College of WisconsinMilwaukeeWisconsinUSA
| | - Vladislav Levchenko
- Department of Molecular Pharmacology and PhysiologyUniversity of South FloridaTampaFloridaUSA
| | - Adrian Zietara
- Department of Molecular Pharmacology and PhysiologyUniversity of South FloridaTampaFloridaUSA
- Department of PhysiologyMedical College of WisconsinMilwaukeeWisconsinUSA
| | - Oleg Palygin
- Department of MedicineMedical University of South CarolinaCharlestonSouth CarolinaUSA
| | - Alexander Staruschenko
- Department of Molecular Pharmacology and PhysiologyUniversity of South FloridaTampaFloridaUSA
- Hypertension and Kidney Research CenterUniversity of South FloridaTampaFloridaUSA
- James A. Haley Veterans' HospitalTampaFloridaUSA
| |
Collapse
|
41
|
Zhang X, Li W, Zhou T, Liu M, Wu Q, Dong N. Corin Deficiency Alters Adipose Tissue Phenotype and Impairs Thermogenesis in Mice. BIOLOGY 2022; 11:biology11081101. [PMID: 35892957 PMCID: PMC9329919 DOI: 10.3390/biology11081101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 07/18/2022] [Accepted: 07/20/2022] [Indexed: 11/16/2022]
Abstract
Atrial natriuretic peptide (ANP) is a key regulator in body fluid balance and cardiovascular biology. In addition to its role in enhancing natriuresis and vasodilation, ANP increases lipolysis and thermogenesis in adipose tissue. Corin is a protease responsible for ANP activation. It remains unknown if corin has a role in regulating adipose tissue function. Here, we examined adipose tissue morphology and function in corin knockout (KO) mice. We observed increased weights and cell sizes in white adipose tissue (WAT), decreased levels of uncoupling protein 1 (Ucp1), a brown adipocyte marker in WAT and brown adipose tissue (BAT), and suppressed thermogenic gene expression in BAT from corin KO mice. At regular room temperature, corin KO and wild-type mice had similar metabolic rates. Upon cold exposure at 4 °C, corin KO mice exhibited impaired thermogenic responses and developed hypothermia. In BAT from corin KO mice, the signaling pathway of p38 mitogen-activated protein kinase, peroxisome proliferator-activated receptor c coactivator 1a, and Ucp1 was impaired. In cell culture, ANP treatment increased Ucp1 expression in BAT-derived adipocytes from corin KO mice. These data indicate that corin mediated-ANP activation is an important hormonal mechanism in regulating adipose tissue function and body temperature upon cold exposure in mice.
Collapse
Affiliation(s)
- Xianrui Zhang
- Cyrus Tang Hematology Center, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Prevention, Soochow University, Suzhou 215123, China; (X.Z.); (W.L.); (T.Z.); (M.L.)
- MOH Key Laboratory of Thrombosis and Hemostasis, The First Affiliated Hospital of Soochow University, Suzhou 215006, China
| | - Wenguo Li
- Cyrus Tang Hematology Center, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Prevention, Soochow University, Suzhou 215123, China; (X.Z.); (W.L.); (T.Z.); (M.L.)
- MOH Key Laboratory of Thrombosis and Hemostasis, The First Affiliated Hospital of Soochow University, Suzhou 215006, China
| | - Tiantian Zhou
- Cyrus Tang Hematology Center, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Prevention, Soochow University, Suzhou 215123, China; (X.Z.); (W.L.); (T.Z.); (M.L.)
| | - Meng Liu
- Cyrus Tang Hematology Center, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Prevention, Soochow University, Suzhou 215123, China; (X.Z.); (W.L.); (T.Z.); (M.L.)
| | - Qingyu Wu
- Cyrus Tang Hematology Center, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Prevention, Soochow University, Suzhou 215123, China; (X.Z.); (W.L.); (T.Z.); (M.L.)
- Correspondence: (Q.W.); (N.D.)
| | - Ningzheng Dong
- Cyrus Tang Hematology Center, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Prevention, Soochow University, Suzhou 215123, China; (X.Z.); (W.L.); (T.Z.); (M.L.)
- MOH Key Laboratory of Thrombosis and Hemostasis, The First Affiliated Hospital of Soochow University, Suzhou 215006, China
- Correspondence: (Q.W.); (N.D.)
| |
Collapse
|
42
|
Robberechts R, Poffe C, Hespel P. Exogenous ketosis suppresses diuresis and atrial natriuretic peptide during exercise. J Appl Physiol (1985) 2022; 133:449-460. [PMID: 35771216 DOI: 10.1152/japplphysiol.00061.2022] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
We have previously demonstrated that exogenous ketosis reduces urine production during exercise. However, the underlying physiological mechanism of this anti-diuretic effect remained unclear. Therefore, we investigated whether acute exogenous ketosis by oral ingestion of ketone ester (KE) during a simulated cycling race (RACE) affects the hormonal pathways implicated in fluid balance regulation during exercise. In a double-blind crossover design, 11 well-trained male cyclists participated in RACE consisting of a 3-h submaximal intermittent cycling (IMT180') bout followed by a 15-minute time trial (TT15') in an environmental chamber set at 28 °C and 60 % relative humidity. Fluid intake was adjusted to maintain euhydration. Before and during RACE, the subjects received either a control drink (CON) or the ketone ester (R)-3-hydroxybutyl (R)-3-hydroxybutyrate (KE), which elevated blood β-hydroxybutyrate to ~2-4 mM. Urine output during IMT180' was ~20% lower in KE (1172 ± 557 ml) than in CON (1431 ± 548 ml, p < 0.05). Compared with CON, N-terminal pro-atrial natriuretic peptide (NT-pro ANP) concentration during RACE was ~20% lower in KE (p < 0.05). KE also raised plasma noradrenaline concentrations during RACE. Performance in TT15' was similar between CON and KE. In conclusion, exogenous ketosis suppresses diuresis and downregulates α-natriuretic peptide activity during exercise.
Collapse
Affiliation(s)
- Ruben Robberechts
- Exercise Physiology Research Group, Department of Movement Sciences, KU Leuven,, Leuven, Belgium
| | - Chiel Poffe
- Exercise Physiology Research Group, Department of Movement Sciences, KU Leuven,, Leuven, Belgium
| | - Peter Hespel
- Exercise Physiology Research Group, Department of Movement Sciences, KU Leuven,, Leuven, Belgium.,DBakala Academy-Athletic Performance Center, KU Leuven, Leuven, Belgium
| |
Collapse
|
43
|
Boteanu RM, Suica VI, Uyy E, Ivan L, Cerveanu-Hogas A, Mares RG, Simionescu M, Schiopu A, Antohe F. Short-Term Blockade of Pro-Inflammatory Alarmin S100A9 Favorably Modulates Left Ventricle Proteome and Related Signaling Pathways Involved in Post-Myocardial Infarction Recovery. Int J Mol Sci 2022; 23:ijms23095289. [PMID: 35563680 PMCID: PMC9103348 DOI: 10.3390/ijms23095289] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Revised: 05/05/2022] [Accepted: 05/08/2022] [Indexed: 02/01/2023] Open
Abstract
Prognosis after myocardial infarction (MI) varies greatly depending on the extent of damaged area and the management of biological processes during recovery. Reportedly, the inhibition of the pro-inflammatory S100A9 reduces myocardial damage after MI. We hypothesize that a S100A9 blockade induces changes of major signaling pathways implicated in post-MI healing. Mass spectrometry-based proteomics and gene analyses of infarcted mice left ventricle were performed. The S100A9 blocker (ABR-23890) was given for 3 days after coronary ligation. At 3 and 7 days post-MI, ventricle samples were analyzed versus control and Sham-operated mice. Blockade of S100A9 modulated the expressed proteins involved in five biological processes: leukocyte cell–cell adhesion, regulation of the muscle cell apoptotic process, regulation of the intrinsic apoptotic signaling pathway, sarcomere organization and cardiac muscle hypertrophy. The blocker induced regulation of 36 proteins interacting with or targeted by the cellular tumor antigen p53, prevented myocardial compensatory hypertrophy, and reduced cardiac markers of post-ischemic stress. The blockade effect was prominent at day 7 post-MI when the quantitative features of the ventricle proteome were closer to controls. Blockade of S100A9 restores key biological processes altered post-MI. These processes could be valuable new pharmacological targets for the treatment of ischemic heart. Mass spectrometry data are available via ProteomeXchange with identifier PXD033683.
Collapse
Affiliation(s)
- Raluca Maria Boteanu
- Department of Proteomics, Institute of Cellular Biology and Pathology “N. Simionescu” of the Romanian Academy, 050568 Bucharest, Romania; (R.M.B.); (V.-I.S.); (E.U.); (L.I.); (A.C.-H.); (M.S.)
| | - Viorel-Iulian Suica
- Department of Proteomics, Institute of Cellular Biology and Pathology “N. Simionescu” of the Romanian Academy, 050568 Bucharest, Romania; (R.M.B.); (V.-I.S.); (E.U.); (L.I.); (A.C.-H.); (M.S.)
| | - Elena Uyy
- Department of Proteomics, Institute of Cellular Biology and Pathology “N. Simionescu” of the Romanian Academy, 050568 Bucharest, Romania; (R.M.B.); (V.-I.S.); (E.U.); (L.I.); (A.C.-H.); (M.S.)
| | - Luminita Ivan
- Department of Proteomics, Institute of Cellular Biology and Pathology “N. Simionescu” of the Romanian Academy, 050568 Bucharest, Romania; (R.M.B.); (V.-I.S.); (E.U.); (L.I.); (A.C.-H.); (M.S.)
| | - Aurel Cerveanu-Hogas
- Department of Proteomics, Institute of Cellular Biology and Pathology “N. Simionescu” of the Romanian Academy, 050568 Bucharest, Romania; (R.M.B.); (V.-I.S.); (E.U.); (L.I.); (A.C.-H.); (M.S.)
| | - Razvan Gheorghita Mares
- Department of Pathophysiology, University of Medicine, Pharmacy, Sciences and Technology of Targu Mures, 540142 Targu Mures, Romania; (R.G.M.); (A.S.)
| | - Maya Simionescu
- Department of Proteomics, Institute of Cellular Biology and Pathology “N. Simionescu” of the Romanian Academy, 050568 Bucharest, Romania; (R.M.B.); (V.-I.S.); (E.U.); (L.I.); (A.C.-H.); (M.S.)
| | - Alexandru Schiopu
- Department of Pathophysiology, University of Medicine, Pharmacy, Sciences and Technology of Targu Mures, 540142 Targu Mures, Romania; (R.G.M.); (A.S.)
- Department of Clinical Sciences Malmö, Lund University, 21428 Malmö, Sweden
| | - Felicia Antohe
- Department of Proteomics, Institute of Cellular Biology and Pathology “N. Simionescu” of the Romanian Academy, 050568 Bucharest, Romania; (R.M.B.); (V.-I.S.); (E.U.); (L.I.); (A.C.-H.); (M.S.)
- Correspondence: ; Tel.: +40-213-192-737
| |
Collapse
|
44
|
Corin: A Key Mediator in Sodium Homeostasis, Vascular Remodeling, and Heart Failure. BIOLOGY 2022; 11:biology11050717. [PMID: 35625445 PMCID: PMC9138375 DOI: 10.3390/biology11050717] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Revised: 05/01/2022] [Accepted: 05/05/2022] [Indexed: 12/11/2022]
Abstract
Simple Summary Atrial natriuretic peptide (ANP) is an important hormone that regulates many physiological and pathological processes, including electrolyte and body fluid balance, blood volume and pressure, cardiac channel activity and function, inflammatory response, lipid metabolism, and vascular remodeling. Corin is a transmembrane serine protease that activates ANP. Variants in the CORIN gene are associated with cardiovascular disease, including hypertension, cardiac hypertrophy, atrial fibrillation, heart failure, and preeclampsia. The current data indicate a key role of corin-mediated ANP production and signaling in the maintenance of cardiovascular homeostasis. In this review, we discuss the latest findings regarding the molecular and cellular mechanisms underlying the role of corin in sodium homeostasis, uterine spiral artery remodeling, and heart failure. Abstract Atrial natriuretic peptide (ANP) is a crucial element of the cardiac endocrine function that promotes natriuresis, diuresis, and vasodilation, thereby protecting normal blood pressure and cardiac function. Corin is a type II transmembrane serine protease that is highly expressed in the heart, where it converts the ANP precursor to mature ANP. Corin deficiency prevents ANP activation and causes hypertension and heart disease. In addition to the heart, corin is expressed in other tissues, including those of the kidney, skin, and uterus, where corin-mediated ANP production and signaling act locally to promote sodium excretion and vascular remodeling. These results indicate that corin and ANP function in many tissues via endocrine and autocrine mechanisms. In heart failure patients, impaired natriuretic peptide processing is a common pathological mechanism that contributes to sodium and body fluid retention. In this review, we discuss most recent findings regarding the role of corin in non-cardiac tissues, including the kidney and skin, in regulating sodium homeostasis and body fluid excretion. Moreover, we describe the molecular mechanisms underlying corin and ANP function in supporting orderly cellular events in uterine spiral artery remodeling. Finally, we assess the potential of corin-based approaches to enhance natriuretic peptide production and activity as a treatment of heart failure.
Collapse
|
45
|
Olaniyi KS, Atuma CL, Mahmud H, Saidi AO, Sabinari IW, Akintayo CO, Ajadi IO, Olatunji LA. Restoration of cardiac metabolic flexibility by acetate in high fat diet-induced obesity is independent of ANP/BNP modulation. Can J Physiol Pharmacol 2022; 100:509-520. [PMID: 35395159 DOI: 10.1139/cjpp-2021-0531] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The present study hypothesized that cardiac metabolic inflexibility is dependent on cardiac ANP/BNP alteration and HDAC activity. We further sought to investigate the therapeutic potential of SCFA, acetate in high fat diet (HFD)-induced obese rat model. Adult male Wistar rats were assigned into groups (n = 6/group): Control, Obese, Sodium acetate (NaAc)-treated and Obese+ NaAc-treated groups received distilled water once daily (oral gavage), 40% HFD ad libitum, 200 mg/kg NaAc once daily (oral gavage) and 40% HFD+NaAc respectively. The treatments lasted for 12 weeks. HFD resulted in increased food intake, body weight and cardiac mass. It also caused insulin resistance and enhanced β-cell function, increased fasting insulin, lactate, plasma and cardiac triglyceride, total cholesterol, lipid peroxidation, TNF-α, IL-6, HDAC and cardiac troponin T and γ-Glutamyl transferase and decreased plasma and cardiac GSH with unaltered cardiac ANP and BNP. However, these alterations were averted when treated with acetate. Taken together, these results indicate that obesity induces defective cardiac metabolic flexibility, which is accompanied by elevated level of HDAC and not ANP/BNP alteration. The results also suggest that acetate ameliorates obesity-induced cardiac metabolic inflexibility by suppression of HDAC and independent of ANP/BNP modulation.
Collapse
Affiliation(s)
- Kehinde Samuel Olaniyi
- Afe Babalola University, 470822, Department of Physiology, Ado Ekiti, Nigeria.,College of Health Sciences University of Ilorin P, Department of Physiology, Ilorin, Nigeria;
| | - Chukwubueze L Atuma
- Afe Babalola University, 470822, Department of Physiology, Ado Ekiti, Nigeria;
| | - Hadiza Mahmud
- Afe Babalola University, 470822, Department of Physiology, Ado Ekiti, Nigeria;
| | - Azeezat O Saidi
- Afe Babalola University, 470822, Department of Physiology, Ado Ekiti, Nigeria;
| | | | - Christopher O Akintayo
- Afe Babalola University College of Medicine and Health Sciences, 473846, Cardio/Repro-metabolic and Microbiome Research Unit, Department of Physiology, Ado Ekiti, Ekiti, Nigeria;
| | - Isaac O Ajadi
- Ladoke Akintola University of Technology College of Health Sciences, 215747, Department of Physiology, Osogbo, Osun, Nigeria;
| | - Lawrence A Olatunji
- College of Health Sciences University of Ilorin P, Department of Physiology, Ilorin, Nigeria;
| |
Collapse
|
46
|
Amini P, Amrovani M, Nassaj ZS, Ajorlou P, Pezeshgi A, Ghahrodizadehabyaneh B. Hypertension: Potential Player in Cardiovascular Disease Incidence in Preeclampsia. Cardiovasc Toxicol 2022; 22:391-403. [PMID: 35347585 DOI: 10.1007/s12012-022-09734-w] [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: 10/23/2021] [Accepted: 02/17/2022] [Indexed: 11/28/2022]
Abstract
Preeclampsia (PE) is one of the complications, that threatens pregnant mothers during pregnancy. According to studies, it accounts for 3-7% of all pregnancies, and also is effective in preterm delivery. PE is the third leading cause of death in pregnant women. High blood pressure in PE can increase the risk of developing cardiovascular disease (CVD) in cited individuals, and is one of the leading causes of death in PE individuals. Atrial natriuretic peptide (ANP), Renin-Angiotensin system and nitric oxide (NO) are some of involved factors in regulating blood pressure. Therefore, by identifying the signaling pathways, that are used by these molecules to regulate and modulate blood pressure, appropriate treatment strategies can be provided to reduce blood pressure through target therapy in PE individuals; consequently, it can reduce CVD risk and mortality.
Collapse
Affiliation(s)
- Parya Amini
- Atherosclerosis Research Center, Ahvaz Jundishapour University of Medical Sciences, Ahvaz, Iran
| | - Mehran Amrovani
- High Institute for Education and Research in Transfusion Medicine, Tehran, Iran
| | - Zohre Saleh Nassaj
- Center for Health Related Social and Behavioral Sciences Research, Shahroud University of Medical Sciences, Shahroud, Iran
| | - Parisa Ajorlou
- Department of Medical Genetics, Faculty of Medicine, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
| | - Aiyoub Pezeshgi
- Internal Medicine Department, Zanjan University of Medical Sciences, Zanjan, Iran.
| | | |
Collapse
|
47
|
Apostolidis SA, Sarkar A, Giannini HM, Goel RR, Mathew D, Suzuki A, Baxter AE, Greenplate AR, Alanio C, Abdel-Hakeem M, Oldridge DA, Giles JR, Wu JE, Chen Z, Huang YJ, Belman J, Pattekar A, Manne S, Kuthuru O, Dougherty J, Weiderhold B, Weisman AR, Ittner CAG, Gouma S, Dunbar D, Frank I, Huang AC, Vella LA, Reilly JP, Hensley SE, Rauova L, Zhao L, Meyer NJ, Poncz M, Abrams CS, Wherry EJ. Signaling Through FcγRIIA and the C5a-C5aR Pathway Mediate Platelet Hyperactivation in COVID-19. Front Immunol 2022; 13:834988. [PMID: 35309299 PMCID: PMC8928747 DOI: 10.3389/fimmu.2022.834988] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Accepted: 02/07/2022] [Indexed: 12/12/2022] Open
Abstract
Patients with COVID-19 present with a wide variety of clinical manifestations. Thromboembolic events constitute a significant cause of morbidity and mortality in patients infected with SARS-CoV-2. Severe COVID-19 has been associated with hyperinflammation and pre-existing cardiovascular disease. Platelets are important mediators and sensors of inflammation and are directly affected by cardiovascular stressors. In this report, we found that platelets from severely ill, hospitalized COVID-19 patients exhibited higher basal levels of activation measured by P-selectin surface expression and had poor functional reserve upon in vitro stimulation. To investigate this question in more detail, we developed an assay to assess the capacity of plasma from COVID-19 patients to activate platelets from healthy donors. Platelet activation was a common feature of plasma from COVID-19 patients and correlated with key measures of clinical outcome including kidney and liver injury, and APACHEIII scores. Further, we identified ferritin as a pivotal clinical marker associated with platelet hyperactivation. The COVID-19 plasma-mediated effect on control platelets was highest for patients that subsequently developed inpatient thrombotic events. Proteomic analysis of plasma from COVID-19 patients identified key mediators of inflammation and cardiovascular disease that positively correlated with in vitro platelet activation. Mechanistically, blocking the signaling of the FcγRIIa-Syk and C5a-C5aR pathways on platelets, using antibody-mediated neutralization, IgG depletion or the Syk inhibitor fostamatinib, reversed this hyperactivity driven by COVID-19 plasma and prevented platelet aggregation in endothelial microfluidic chamber conditions. These data identified these potentially actionable pathways as central for platelet activation and/or vascular complications and clinical outcomes in COVID-19 patients. In conclusion, we reveal a key role of platelet-mediated immunothrombosis in COVID-19 and identify distinct, clinically relevant, targetable signaling pathways that mediate this effect.
Collapse
Affiliation(s)
- Sokratis A. Apostolidis
- Institute for Immunology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, United States
- Division of Rheumatology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, United States
| | - Amrita Sarkar
- Division of Hematology, The Children’s Hospital of Philadelphia, Philadelphia, PA, United States
| | - Heather M. Giannini
- Division of Pulmonary, Allergy and Critical Care Medicine, Center for Translational Lung Biology, Lung Biology Institute, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, United States
| | - Rishi R. Goel
- Institute for Immunology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, United States
| | - Divij Mathew
- Institute for Immunology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, United States
| | - Aae Suzuki
- Department of Medicine, University of Pennsylvania School of Medicine, Philadelphia, PA, United States
| | - Amy E. Baxter
- Institute for Immunology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, United States
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, United States
| | - Allison R. Greenplate
- Institute for Immunology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, United States
- Immune Health™, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, United States
| | - Cécile Alanio
- Institute for Immunology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, United States
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, United States
- Parker Institute for Cancer Immunotherapy, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, United States
| | - Mohamed Abdel-Hakeem
- Institute for Immunology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, United States
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, United States
| | - Derek A. Oldridge
- Institute for Immunology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, United States
- Department of Pathology and Laboratory Medicine, Children’s Hospital of Philadelphia, Philadelphia, PA, United States
- Department of Pathology and Laboratory Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, United States
| | - Josephine R. Giles
- Institute for Immunology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, United States
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, United States
- Parker Institute for Cancer Immunotherapy, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, United States
| | - Jennifer E. Wu
- Institute for Immunology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, United States
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, United States
- Parker Institute for Cancer Immunotherapy, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, United States
| | - Zeyu Chen
- Institute for Immunology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, United States
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, United States
| | - Yinghui Jane Huang
- Institute for Immunology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, United States
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, United States
| | - Jonathan Belman
- Institute for Immunology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, United States
- Department of Pathology and Laboratory Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, United States
| | - Ajinkya Pattekar
- Institute for Immunology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, United States
- Immune Health™, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, United States
| | - Sasikanth Manne
- Institute for Immunology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, United States
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, United States
| | - Oliva Kuthuru
- Institute for Immunology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, United States
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, United States
| | - Jeanette Dougherty
- Institute for Immunology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, United States
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, United States
| | - Brittany Weiderhold
- Department of Medicine, University of Pennsylvania School of Medicine, Philadelphia, PA, United States
| | - Ariel R. Weisman
- Division of Pulmonary, Allergy and Critical Care Medicine, Center for Translational Lung Biology, Lung Biology Institute, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, United States
| | - Caroline A. G. Ittner
- Division of Pulmonary, Allergy and Critical Care Medicine, Center for Translational Lung Biology, Lung Biology Institute, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, United States
| | - Sigrid Gouma
- Department of Microbiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, United States
| | - Debora Dunbar
- Division of Infectious Diseases, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Ian Frank
- Division of Infectious Diseases, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Alexander C. Huang
- Institute for Immunology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, United States
- Parker Institute for Cancer Immunotherapy, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, United States
- Division of Hematology and Oncology, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, United States
| | - Laura A. Vella
- Institute for Immunology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, United States
- Division of Infectious Diseases, Department of Pediatrics, Children’s Hospital of Philadelphia, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, United States
| | - John P. Reilly
- Division of Pulmonary, Allergy and Critical Care Medicine, Center for Translational Lung Biology, Lung Biology Institute, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, United States
| | - Scott E. Hensley
- Institute for Immunology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, United States
- Department of Microbiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, United States
| | - Lubica Rauova
- Division of Hematology, The Children’s Hospital of Philadelphia, Philadelphia, PA, United States
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Liang Zhao
- Department of Medicine, University of Pennsylvania School of Medicine, Philadelphia, PA, United States
| | - Nuala J. Meyer
- Division of Pulmonary, Allergy and Critical Care Medicine, Center for Translational Lung Biology, Lung Biology Institute, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, United States
| | - Mortimer Poncz
- Division of Hematology, The Children’s Hospital of Philadelphia, Philadelphia, PA, United States
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Charles S. Abrams
- Department of Medicine, University of Pennsylvania School of Medicine, Philadelphia, PA, United States
| | - E. John Wherry
- Institute for Immunology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, United States
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, United States
- Immune Health™, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, United States
- Parker Institute for Cancer Immunotherapy, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, United States
| |
Collapse
|
48
|
Zhang W, Zhou Y, Dong Y, Liu W, Li H, Song W. Correlation between N-terminal pro-atrial natriuretic peptide, corin, and target organ damage in hypertensive disorders of pregnancy. J Clin Hypertens (Greenwich) 2022; 24:644-651. [PMID: 35199942 PMCID: PMC9106090 DOI: 10.1111/jch.14450] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Revised: 02/10/2022] [Accepted: 02/13/2022] [Indexed: 12/11/2022]
Abstract
The objective was to evaluate the correlation between N‐terminal pro‐atrial natriuretic peptide (NT‐proANP), corin and the severity of target organ injury in hypertensive disorders of pregnancy. A total of 78 women with hypertensive disorders of pregnancy and 49 normotensive pregnancies were enrolled. The clinical characteristics, laboratory index and echocardiogram results were collected. NT‐proANP, corin, sFlt‐1 and PlGF levels were measured. A receiver's operating characteristics (ROC) curve was performed to evaluate the efficacy of predicting target organ injury in the HDP group. The NT‐proANP, corin, and sFlt‐1/PlGF ratio were increased in the HDP group (p < .05). The area under the curve (AUC) predicted by NT‐proANP and corin were larger than sFlt‐1/PlGF ratio (0.779, 0.867, and 0.766, respectively). The creatinine and urine protein were significantly increased, while the estimated glomerular filtration rate (eGFR) was dramatically decreased in the HDP group (p < .05 each). The left atrial diameter (LAD), left atrial volume index (LAVI), left ventricular posterior wall thickness (LVPWT), and left ventricular septal thickness (LVST) were larger in the HDP group (p < .001 each). The NT‐proANP/corin levels were positively correlated with LAD, creatinine, and urine protein, and negatively correlated with eGFR in HDP group (p < .05 each). Multiple regressions demonstrated that NT‐proANP was an independent risk factor of LAD and urine protein, and corin was an independent risk factor of creatinine and eGFR in HDP group. NT‐proANP and corin may be reliable biomarkers for evaluating the severity of target organ damage in the hypertensive disorders of pregnant patients.
Collapse
Affiliation(s)
- Wei Zhang
- Department of Hypertension, The First Affiliated Hospital of Dalian Medical University, DaLian Liaoning, China
| | - Ying Zhou
- Department of Hypertension, The First Affiliated Hospital of Dalian Medical University, DaLian Liaoning, China
| | - Yubing Dong
- Department of Hypertension, The First Affiliated Hospital of Dalian Medical University, DaLian Liaoning, China
| | - Wanyu Liu
- Department of Hypertension, The First Affiliated Hospital of Dalian Medical University, DaLian Liaoning, China
| | - Haiying Li
- The Dalian Obstetrics and Gynecology Hospital, DaLian Liaoning, China
| | - Wei Song
- Department of Hypertension, The First Affiliated Hospital of Dalian Medical University, DaLian Liaoning, China
| |
Collapse
|
49
|
Rajanathan R, Pedersen TM, Thomsen MB, Botker HE, Matchkov VV. Phenylephrine-Induced Cardiovascular Changes in the Anesthetized Mouse: An Integrated Assessment of in vivo Hemodynamics Under Conditions of Controlled Heart Rate. Front Physiol 2022; 13:831724. [PMID: 35250634 PMCID: PMC8891648 DOI: 10.3389/fphys.2022.831724] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Accepted: 01/27/2022] [Indexed: 11/13/2022] Open
Abstract
Objective Investigating the cardiovascular system is challenging due to its complex regulation by humoral and neuronal factors. Despite this complexity, many existing research methods are limited to the assessment of a few parameters leading to an incomplete characterization of cardiovascular function. Thus, we aim to establish a murine in vivo model for integrated assessment of the cardiovascular system under conditions of controlled heart rate. Utilizing this model, we assessed blood pressure, cardiac output, stroke volume, total peripheral resistance, and electrocardiogram (ECG). Hypothesis We hypothesize that (i) our in vivo model can be utilized to investigate cardiac and vascular responses to pharmacological intervention with the α1-agonist phenylephrine, and (ii) we can study cardiovascular function during artificial pacing of the heart, modulating cardiac function without a direct vascular effect. Methods We included 12 mice that were randomly assigned to either vehicle or phenylephrine intervention through intraperitoneal administration. Mice were anesthetized with isoflurane and intubated endotracheally for mechanical ventilation. We measured blood pressure via a solid-state catheter in the aortic arch, blood flow via a probe on the ascending aorta, and ECG from needle electrodes on the extremities. Right atrium was electrically paced at a frequency ranging from 10 to 11.3 Hz before and after either vehicle or phenylephrine administration. Results Phenylephrine significantly increased blood pressure, stroke volume, and total peripheral resistance compared to the vehicle group. Moreover, heart rate was significantly decreased following phenylephrine administration. Pacing significantly decreased stroke volume and cardiac output both prior to and after drug administration. However, phenylephrine-induced changes in blood pressure and total peripheral resistance were maintained with increasing pacing frequencies compared to the vehicle group. Total peripheral resistance was not significantly altered with increasing pacing frequencies suggesting that the effect of phenylephrine is primarily of vascular origin. Conclusion In conclusion, this in vivo murine model is capable of distinguishing between changes in peripheral vascular and cardiac functions. This study underlines the primary effect of phenylephrine on vascular function with secondary changes to cardiac function. Hence, this in vivo model is useful for the integrated assessment of the cardiovascular system.
Collapse
Affiliation(s)
- Rajkumar Rajanathan
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
- *Correspondence: Rajkumar Rajanathan,
| | | | - Morten B. Thomsen
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Hans Erik Botker
- Department of Cardiology, Aarhus University Hospital, Aarhus, Denmark
| | | |
Collapse
|
50
|
Predicting Heart Cell Types by Using Transcriptome Profiles and a Machine Learning Method. Life (Basel) 2022; 12:life12020228. [PMID: 35207515 PMCID: PMC8877019 DOI: 10.3390/life12020228] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Revised: 01/29/2022] [Accepted: 01/29/2022] [Indexed: 11/17/2022] Open
Abstract
The heart is an essential organ in the human body. It contains various types of cells, such as cardiomyocytes, mesothelial cells, endothelial cells, and fibroblasts. The interactions between these cells determine the vital functions of the heart. Therefore, identifying the different cell types and revealing the expression rules in these cell types are crucial. In this study, multiple machine learning methods were used to analyze the heart single-cell profiles with 11 different heart cell types. The single-cell profiles were first analyzed via light gradient boosting machine method to evaluate the importance of gene features on the profiling dataset, and a ranking feature list was produced. This feature list was then brought into the incremental feature selection method to identify the best features and build the optimal classifiers. The results suggested that the best decision tree (DT) and random forest classification models achieved the highest weighted F1 scores of 0.957 and 0.981, respectively. The selected features, such as NPPA, LAMA2, DLC1, and the classification rules extracted from the optimal DT classifier played a crucial role in cardiac structure and function in recent research and enrichment analysis. In particular, some lncRNAs (LINC02019, NEAT1) were found to be quite important for the recognition of different cardiac cell types. In summary, these findings provide a solid academic foundation for the development of molecular diagnostics and biomarker discovery for cardiac diseases.
Collapse
|