1
|
Amioka N, Franklin MK, Kukida M, Zhu L, Moorleghen JJ, Howatt DA, Katsumata Y, Mullick AE, Yanagita M, Martinez-Irizarry MM, Sandoval RM, Dunn KW, Sawada H, Daugherty A, Lu HS. Renal Proximal Tubule Cell-Specific Megalin Deletion Does Not Affect Atherosclerosis But Induces Tubulointerstitial Nephritis in Mice Fed a Western Diet. Arterioscler Thromb Vasc Biol 2025; 45:74-89. [PMID: 39569521 DOI: 10.1161/atvbaha.124.321366] [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: 06/08/2024] [Accepted: 10/31/2024] [Indexed: 11/22/2024]
Abstract
BACKGROUND Pharmacological inhibition of megalin (also known as LRP2 [low-density lipoprotein receptor-related protein-2]) attenuates atherosclerosis in hypercholesterolemic mice. Since megalin is abundant in renal proximal tubule cells (PTCs), the purpose of this study was to determine whether PTC-specific deletion of megalin reduces hypercholesterolemia-induced atherosclerosis in mice. METHODS Female Lrp2 f/f mice were bred with male Ndrg1-Cre ERT2 +/0 mice to develop PTC-LRP2 +/+ and PTC-LRP2 -/- littermates. To study atherosclerosis, all mice were bred to an LDL (low-density lipoprotein) receptor -/- background and fed a Western diet to induce atherosclerosis. RESULTS PTC-specific megalin deletion did not attenuate atherosclerosis in LDL receptor -/- mice in either sex. Serendipitously, we discovered that PTC-specific megalin deletion led to interstitial infiltration of CD68+ cells and tubular atrophy. The pathology was only evident in male PTC-LRP2 -/- mice fed a Western diet but not in mice fed a normal laboratory diet. Renal pathologies were also observed in male PTC-LRP2 -/- mice in an LDL receptor +/+ background fed the same Western diet, demonstrating that the renal pathologies were dependent on diet and not on hypercholesterolemia. In contrast, female PTC-LRP2 -/- mice had no apparent renal pathologies. In vivo multiphoton microscopy demonstrated that PTC-specific megalin deletion dramatically diminished ALB (albumin) accumulation in PTCs within 10 days of Western diet feeding. RNA-sequencing analyses demonstrated the upregulation of inflammation-related pathways in the kidney. CONCLUSIONS PTC-specific megalin deletion does not affect atherosclerosis but leads to tubulointerstitial nephritis in mice fed a Western diet, with severe pathologies in male mice.
Collapse
MESH Headings
- Animals
- Low Density Lipoprotein Receptor-Related Protein-2/genetics
- Low Density Lipoprotein Receptor-Related Protein-2/metabolism
- Low Density Lipoprotein Receptor-Related Protein-2/deficiency
- Diet, Western/adverse effects
- Kidney Tubules, Proximal/metabolism
- Kidney Tubules, Proximal/pathology
- Female
- Male
- Atherosclerosis/genetics
- Atherosclerosis/pathology
- Atherosclerosis/metabolism
- Atherosclerosis/prevention & control
- Mice, Knockout
- Disease Models, Animal
- Nephritis, Interstitial/genetics
- Nephritis, Interstitial/pathology
- Nephritis, Interstitial/metabolism
- Mice
- Mice, Inbred C57BL
- Hypercholesterolemia/genetics
- Hypercholesterolemia/complications
- Hypercholesterolemia/metabolism
- Receptors, LDL/genetics
- Receptors, LDL/deficiency
- Antigens, Differentiation, Myelomonocytic/metabolism
- Antigens, Differentiation, Myelomonocytic/genetics
- Plaque, Atherosclerotic
- CD68 Molecule
Collapse
Affiliation(s)
- Naofumi Amioka
- Saha Cardiovascular Research Center and Saha Aortic Center (N.A., M.K.F., M.K., L.Z., J.J.M., D.A.H., H.S., A.D., H.S.L.)
| | - Michael K Franklin
- Saha Cardiovascular Research Center and Saha Aortic Center (N.A., M.K.F., M.K., L.Z., J.J.M., D.A.H., H.S., A.D., H.S.L.)
| | | | - Liyuan Zhu
- Saha Cardiovascular Research Center and Saha Aortic Center (N.A., M.K.F., M.K., L.Z., J.J.M., D.A.H., H.S., A.D., H.S.L.)
| | - Jessica J Moorleghen
- Saha Cardiovascular Research Center and Saha Aortic Center (N.A., M.K.F., M.K., L.Z., J.J.M., D.A.H., H.S., A.D., H.S.L.)
| | - Deborah A Howatt
- Saha Cardiovascular Research Center and Saha Aortic Center (N.A., M.K.F., M.K., L.Z., J.J.M., D.A.H., H.S., A.D., H.S.L.)
| | - Yuriko Katsumata
- Sanders-Brown Center on Aging (Y.K.)
- Department of Biostatistics (Y.K.)
| | | | - Motoko Yanagita
- Department of Nephrology, Kyoto University Graduate School of Medicine (M.Y.)
- WPI Institute for the Advanced Study of Human Biology, Kyoto University, Japan (M.Y.)
| | | | - Ruben M Sandoval
- Department of Medicine, Indiana University, Indianapolis (M.M.M.-I., R.M.S., K.W.D.)
| | - Kenneth W Dunn
- Department of Medicine, Indiana University, Indianapolis (M.M.M.-I., R.M.S., K.W.D.)
| | - Hisashi Sawada
- Saha Cardiovascular Research Center and Saha Aortic Center (N.A., M.K.F., M.K., L.Z., J.J.M., D.A.H., H.S., A.D., H.S.L.)
- Department of Physiology, University of Kentucky, Lexington (H.S., A.D., H.S.L.)
| | - Alan Daugherty
- Saha Cardiovascular Research Center and Saha Aortic Center (N.A., M.K.F., M.K., L.Z., J.J.M., D.A.H., H.S., A.D., H.S.L.)
- Department of Physiology, University of Kentucky, Lexington (H.S., A.D., H.S.L.)
| | - Hong S Lu
- Saha Cardiovascular Research Center and Saha Aortic Center (N.A., M.K.F., M.K., L.Z., J.J.M., D.A.H., H.S., A.D., H.S.L.)
- Department of Physiology, University of Kentucky, Lexington (H.S., A.D., H.S.L.)
| |
Collapse
|
2
|
Sopori S, Kavinay K, Bhan S, Saxena S, Medha M, Kumar R, Dhar A, Bhat A. CLOCK gene 3'UTR and exon 9 polymorphisms show a strong association with essential hypertension in a North Indian population. BMC Med Genomics 2024; 17:289. [PMID: 39696277 DOI: 10.1186/s12920-024-02056-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Accepted: 11/26/2024] [Indexed: 12/20/2024] Open
Abstract
BACKGROUND Hypertension (HTN) is a medical condition characterized by persistent systolic and diastolic blood pressures of ≥ 140 mmHg and ≥ 90 mmHg, respectively. With more than 1200 million adult patients aged 30-79 years worldwide according to the latest WHO data, HTN is a major health risk factor; more importantly, 46% of patients are unaware of this condition. Essential hypertension (EH), also known as primary hypertension, is the predominant subtype and has a complex etiology that involves both genetic and non-genetic factors. Majority of living organisms are influenced by the light and dark cycle of a day and respond to these changes through an intricate clock referred to as the "biological clock" or "circadian rhythm". The connection between circadian rhythm and blood pressure is well established, with many studies supporting the role of circadian rhythm gene mutation(s)/polymorphism(s) in EH. To date, no such data are available from any Indian population. METHODS This case‒control study was conducted on 405 EH patients and 505 healthy controls belonging to the Jammu region of North India after an informed consent was obtained from the participants. A total of three single nucleotide variants, two in the CLOCK gene (rs1801260 and rs34789226) and one in the BMAL1/ARNTL gene (rs6486121), were selected for genotyping. Genotyping was performed via the RFLP technique, and the applicable statistical analyses were performed via the SPSS and SNPStats programs. RESULTS Logistic regression analysis revealed a statistically significant association of both CLOCK gene variants rs1801260 (T > C 3'UTR) and rs34789226 (C > T Exon 9) and a nonsignificant association of the BMAL1/ARNTL intronic variant rs6486121 (C > T) with EH. The 3'UTR variant showed a statistically significant association under the codominant (p < 0.0001), dominant (p < 0.0001), and recessive (p = 0.0004) models. In contrast, the exon 9 variant showed a statistically significant negative association under the codominant (p = 0.003) and dominant (p = 0.015) models only. The rs6486121/rs1801260 and rs1801260/rs34789226/rs6486121 haplotypes showed significant differences in their distribution between cases and controls (p < 0.0001). Certain genotypes and haplotypes were found more common in hypertensive males than females. CONCLUSION This is a first report linking circadian rhythm gene polymorphisms with EH in any Indian population. The statistically significant association of the CLOCK gene 3'UTR and exon 9 polymorphisms with EH, highlight the potential role of this gene and probably other genes of the circadian pathway in the etiology of EH in the study population. Additionally, our study also revealed that certain genotypes are making males more susceptible to EH.
Collapse
Affiliation(s)
- Shreya Sopori
- Centre for Molecular Biology, Central University of Jammu, Jammu, UT Jammu and Kashmir, 181143, India
| | - Kavinay Kavinay
- Centre for Molecular Biology, Central University of Jammu, Jammu, UT Jammu and Kashmir, 181143, India
| | - Sonali Bhan
- Centre for Molecular Biology, Central University of Jammu, Jammu, UT Jammu and Kashmir, 181143, India
| | - Shreya Saxena
- Centre for Molecular Biology, Central University of Jammu, Jammu, UT Jammu and Kashmir, 181143, India
| | - Medha Medha
- Centre for Molecular Biology, Central University of Jammu, Jammu, UT Jammu and Kashmir, 181143, India
| | - Rakesh Kumar
- Department of Biotechnology, Shri Mata Vaishnav Devi University, Katra, 182320, India
| | - Arti Dhar
- Department of Pharmacy, Birla Institute of Technology and Sciences Pilani, Hyderabad Campus, Telangana, 500078, India
| | - Audesh Bhat
- Centre for Molecular Biology, Central University of Jammu, Jammu, UT Jammu and Kashmir, 181143, India.
| |
Collapse
|
3
|
Vernail VL, Lucas L, Miller AJ, Arnold AC. Angiotensin-(1-7) and Central Control of Cardiometabolic Outcomes: Implications for Obesity Hypertension. Int J Mol Sci 2024; 25:13320. [PMID: 39769086 PMCID: PMC11677932 DOI: 10.3390/ijms252413320] [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: 11/18/2024] [Revised: 12/06/2024] [Accepted: 12/10/2024] [Indexed: 01/11/2025] Open
Abstract
Hypertension is a leading independent risk factor for the development of cardiovascular disease, the leading cause of death globally. Importantly, the prevalence of hypertension is positively correlated with obesity, with obesity-related hypertension being difficult to treat due to a lack of current guidelines in this population as well as limited efficacy and adverse off-target effects of currently available antihypertensive therapeutics. This highlights the need to better understand the mechanisms linking hypertension with obesity to develop optimal therapeutic approaches. In this regard, the renin-angiotensin system, which is dysregulated in both hypertension and obesity, is a prime therapeutic target. While research and therapies have typically focused on the deleterious angiotensin II axis of the renin-angiotensin system, emerging evidence shows that targeting the protective angiotensin-(1-7) axis also improves cardiovascular and metabolic functions in animal models of obesity hypertension. While the precise mechanisms involved remain under investigation, in addition to peripheral actions, evidence exists to support a role for the central nervous system in the beneficial cardiometabolic effects of angiotensin-(1-7). This review will highlight emerging translational studies exploring the cardiovascular and metabolic regulatory actions of angiotensin-(1-7), with an emphasis on its central actions in brain regions including the brainstem and hypothalamus. An improved understanding of the central mechanisms engaged by angiotensin-(1-7) to regulate cardiovascular and metabolic functions may provide insight into the potential of targeting this hormone as a novel therapeutic approach for obesity-related hypertension.
Collapse
Affiliation(s)
- Victoria L. Vernail
- Department of Neural and Behavioral Sciences, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA; (V.L.V.); or (A.J.M.)
| | - Lillia Lucas
- Department of Neural and Behavioral Sciences, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA; (V.L.V.); or (A.J.M.)
| | - Amanda J. Miller
- Department of Neural and Behavioral Sciences, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA; (V.L.V.); or (A.J.M.)
- Department of Physical Therapy, Lebanon Valley College, Annville, PA 17003, USA
| | - Amy C. Arnold
- Department of Neural and Behavioral Sciences, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA; (V.L.V.); or (A.J.M.)
| |
Collapse
|
4
|
Rajpoot A, Aggarwal T, Sharma V. Unraveling the enigma of cardiac damage caused by lead: Understanding the intricate relationship between oxidative stress and other multifactorial mechanisms. Toxicology 2024; 509:153984. [PMID: 39481524 DOI: 10.1016/j.tox.2024.153984] [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: 09/23/2024] [Revised: 10/26/2024] [Accepted: 10/28/2024] [Indexed: 11/02/2024]
Abstract
Lead (Pb) exposure remains a pressing concern in the realm of public health, with a mounting body of evidence underscoring its adverse impact on cardiovascular well-being. The exposure to Lead instigates the production of reactive oxygen species (ROS), leading to consequential cellular and physiological damage and a perturbation in redox equilibrium. The resultant oxidative stress, induced by ROS, disrupts endothelial functionality, propagates inflammatory processes, and initiates vascular remodeling, collectively contributing to the advancement of cardiovascular diseases (CVDs). The objective of this current review is to comprehensively expound upon the intricate mechanisms through which Lead induced toxicity affects cardiac cells. Additionally, it briefly addresses the ramifications of Lead exposure on the development of three interconnected cardiovascular conditions: atherosclerosis, hypertension, and myocardial infarction. Furthermore, the discourse delves into the specific repercussions of Lead exposure on lipid metabolism, blood pressure regulation, and cardiac performance, culminating in the initiation and progression of atherosclerotic plaque formation, elevated blood pressure, and an augmented risk of myocardial infarction. By understanding these intricate mechanisms, targeted interventions may be devised to counteract the deleterious effects of Lead on cardiovascular health. Thus, this review offers novel avenues for preventive and therapeutic strategies, ultimately serving to alleviate the burden of cardiovascular diseases associated with Lead toxicity.
Collapse
Affiliation(s)
- Anjali Rajpoot
- Department of Bioscience and Biotechnology, Banasthali Vidyapith, Rajasthan 304022, India.
| | - Tanya Aggarwal
- Department of Bioscience and Biotechnology, Banasthali Vidyapith, Rajasthan 304022, India.
| | - Veena Sharma
- Department of Bioscience and Biotechnology, Banasthali Vidyapith, Rajasthan 304022, India.
| |
Collapse
|
5
|
Singh KD, Karnik SS. Implications of β-Arrestin biased signaling by angiotensin II type 1 receptor for cardiovascular drug discovery and therapeutics. Cell Signal 2024; 124:111410. [PMID: 39270918 DOI: 10.1016/j.cellsig.2024.111410] [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: 07/31/2024] [Revised: 09/06/2024] [Accepted: 09/10/2024] [Indexed: 09/15/2024]
Abstract
Angiotensin II receptors, Type 1 (AT1R) and Type 2 (AT2R) are 7TM receptors that play critical roles in both the physiological and pathophysiological regulation of the cardiovascular system. While AT1R blockers (ARBs) have proven beneficial in managing cardiac, vascular and renal maladies they cannot completely halt and reverse the progression of pathologies. Numerous experimental and animal studies have demonstrated that β-arrestin biased AT1R-ligands (such as SII-AngII, S1I8, TRV023, and TRV027) offer cardiovascular benefits by blocking the G protein signaling while retaining the β-arrestin signaling. However, these ligands failed to show improvement in heart-failure outcome over the placebo in a phase IIb clinical trial. One major limitation of current β-arrestin biased AT1R-ligands is that they are peptides with short half-lives, limiting their long-term efficacy in patients. Additionally, β-arrestin biased AT1R-ligand peptides, may inadvertently block AT2R, a promiscuous receptor, potentially negating its beneficial effects in post-myocardial infarction (MI) patients. Therefore, developing a small molecule β-arrestin biased AT1R-ligand with a longer half-life and specificity to AT1R could be more effective in treating heart failure. This approach has the potential to revolutionize the treatment of cardiovascular diseases by offering more sustained and targeted therapeutic effects.
Collapse
Affiliation(s)
- Khuraijam Dhanachandra Singh
- Department of Cardiovascular and Metabolic Sciences, Cleveland Clinic Lerner College of Medicine at Case Western Reserve University, Cleveland Clinic, USA.
| | - Sadashiva S Karnik
- Department of Cardiovascular and Metabolic Sciences, Cleveland Clinic Lerner College of Medicine at Case Western Reserve University, Cleveland Clinic, USA.
| |
Collapse
|
6
|
Lassiter K, Aloui L, Greene ES, Maqaeda M, Tabler T, Dridi S, Wideman RF, Orlowski S, Bottje WG. Water homeostasis gene expression in the kidney of broilers divergently selected for water conversion ratio. Poult Sci 2024; 104:104560. [PMID: 39615323 PMCID: PMC11647239 DOI: 10.1016/j.psj.2024.104560] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Revised: 11/08/2024] [Accepted: 11/21/2024] [Indexed: 01/05/2025] Open
Abstract
Divergent selection of broilers for water conversion ratio has established and high-(HWE) and low- water efficient (LWE) broiler lines. Two 2 × 2 factorial experiments were conducted to assess the gene expression profile of systems involved in renal water homeostasis. In Exp. 1, male and female HWE and LWE broilers were individually phenotyped between 4 and 6 wks of age to determine growth performance and water conversion ratio (g water intake/g body weight gain). Kidney samples were obtained from 5 males and 5 females from each line. In Exp. 2, HWE and modern random bred (MRB) broilers were placed in 12 controlled-environmental chambers (2 floor pens/chamber, 6 chambers/line, 11 birds per pen, 132 birds/line) on day of hatch. The broilers were brooded at thermoneutral temperatures from 0 to 4 wks. From 4 to 7 wks, broilers were maintained at thermoneutral (TN, 25 °C) or exposed to cyclic heat stress (HS, 35 °C, 8h/day) conditions. Body weight, feed intake, and water intake were recorded. Kidney samples were collected, flash frozen in liquid nitrogen, and kept at -80 °C for gene expression analysis. Data were analyzed by Two-way ANOVA and means compared by Tukey's HSD multiple comparison test. Molecular analyses from Exp. 1 showed that the renal expression of arginine vasopressin (AVP), angiotensinogen (AGT), angiotensin II receptor type 1 and 2 (AT1/2), sodium-potassium ATPase subunit B1 (ATP1B1), and aquaporin 3 (AQP3) were upregulated in HWE compared to the LWE line. In contrast, mRNA expression of mesotocin receptor (MTR), AT1/2, AQP1/2, and occludin were significantly higher in females than in males. In Exp. 2, target genes were regulated in environment and/or line-dependent manner. The renal expression of heat shock proteins 70 and 90, AVP receptor 2 (AVPR2), AGT, renin, AT1/2, and AQP1was significantly upregulated in HS compared to TN birds, however AVPR2 expression was significantly higher in HWE compared to MRB birds. Together, the up-regulation of AVP, the renin-angiotensin system (RAS), and AQP in HWE, female, or under HS conditions suggests a better renal water reabsorption to support water use efficiency.
Collapse
Affiliation(s)
- Kentu Lassiter
- Center of Excellence for Poultry Science, Division of Agriculture, University of Arkansas, Fayetteville, Arkansas, USA.
| | - Loujain Aloui
- Higher School of Agriculture of Mograne, University of Carthage, Zaghouan, Tunisia
| | - Elizabeth S Greene
- Center of Excellence for Poultry Science, Division of Agriculture, University of Arkansas, Fayetteville, Arkansas, USA
| | - Marciela Maqaeda
- Center of Excellence for Poultry Science, Division of Agriculture, University of Arkansas, Fayetteville, Arkansas, USA
| | - Travis Tabler
- Center of Excellence for Poultry Science, Division of Agriculture, University of Arkansas, Fayetteville, Arkansas, USA
| | - Sami Dridi
- Center of Excellence for Poultry Science, Division of Agriculture, University of Arkansas, Fayetteville, Arkansas, USA
| | - Robert F Wideman
- Center of Excellence for Poultry Science, Division of Agriculture, University of Arkansas, Fayetteville, Arkansas, USA
| | - Sara Orlowski
- Center of Excellence for Poultry Science, Division of Agriculture, University of Arkansas, Fayetteville, Arkansas, USA
| | - Walter G Bottje
- Center of Excellence for Poultry Science, Division of Agriculture, University of Arkansas, Fayetteville, Arkansas, USA.
| |
Collapse
|
7
|
Song L, Qiu Q, Ju F, Zheng C. Mechanisms of doxorubicin-induced cardiac inflammation and fibrosis; therapeutic targets and approaches. Arch Biochem Biophys 2024; 761:110140. [PMID: 39243924 DOI: 10.1016/j.abb.2024.110140] [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: 07/14/2024] [Revised: 08/28/2024] [Accepted: 09/04/2024] [Indexed: 09/09/2024]
Abstract
Doxorubicin plays a pivotal role in the treatment of various malignancies. Despite its efficacy, the cardiotoxicity associated with doxorubicin limits its clinical utility. The cardiotoxic nature of doxorubicin is attributed to several mechanisms, including its interference with mitochondrial function, the generation of reactive oxygen species (ROS), and the subsequent damage to cardiomyocyte DNA, proteins, and lipids. Furthermore, doxorubicin disrupts the homeostasis of cardiac-specific transcription factors and signaling pathways, exacerbating cardiac dysfunction. Oxidative stress, cell death, and other severe changes, such as mitochondrial dysfunction, activation of pro-oxidant enzymes, the renin-angiotensin system (RAS), endoplasmic reticulum (ER) stress, and infiltration of immune cells in the heart after treatment with doxorubicin, may cause inflammatory and fibrotic responses. Fibrosis and inflammation can lead to a range of disorders in the heart, resulting in potential cardiac dysfunction and disease. Various adjuvants have shown potential in preclinical studies to mitigate these challenges associated with cardiac inflammation and fibrosis. Antioxidants, plant-based products, specific inhibitors, and cardioprotective drugs may be recommended to alleviate cardiotoxicity. This review explores the complex mechanisms of doxorubicin-induced heart inflammation and fibrosis, identifies possible cellular and molecular targets, and investigates potential substances that could help reduce these harmful effects.
Collapse
Affiliation(s)
- Linghua Song
- Department of Pharmacy, Yantai Mountain Hospital, Yantai City, Shandong Province, 264001, China
| | - Qingzhuo Qiu
- Medical Imaging Department of Qingdao Women and Children's Hospital, 266000, China
| | - Fei Ju
- Department of Critical Care, Medicine East Hospital of Qingdao Municipal Hospital, 266000, China
| | - Chunyan Zheng
- Cadre Health Office of Zibo Central Hospital in Shandong Province, 255000, China.
| |
Collapse
|
8
|
Zhang S, Li S, Xie S, Cui L, Gao Y, Wang Y. The Role of Ca 2+/PI3K/Akt/eNOS/NO Pathway in Astragaloside IV-Induced Inhibition of Endothelial Inflammation Triggered by Angiotensin II. Mediators Inflamm 2024; 2024:3193950. [PMID: 39512364 PMCID: PMC11540887 DOI: 10.1155/2024/3193950] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Revised: 09/29/2024] [Accepted: 10/09/2024] [Indexed: 11/15/2024] Open
Abstract
Inflammation induced by angiotensin II (Ang II) is a key event in the progression of numerous cardiovascular diseases. Astragaloside IV (AS-IV), a glycoside extracted from Astragalus membranaceus Bunge, has been shown to inhibit Ang II-induced inflammatory responses in vivo. However, the mechanisms underlying the beneficial effects are still unclear. This study investigated whether AS-IV attenuates endothelial inflammation induced by Ang II via the activation of endothelial nitric oxide synthase (eNOS)/nitric oxide (NO) pathway. Human umbilical vein endothelial cells (HUVECs) were cultured in the presence of AS-IV with or without the specific inhibitor of NOS or Ca2+- and phosphatidylinositol 3-kinase (PI3K)/Akt-dependent cascade prior to Ang II exposure. Incubation of HUVECs with AS-IV enhanced NO production and eNOSser1177 phosphorylation. These responses were abrogated by the inhibition of NOS or Ca2+- and PI3K/Akt-dependent pathway. In addition, preincubation of HUVECs with AS-IV inhibited Ang II-induced cytokine and chemokine production, adhesion molecule expression, monocyte adhesion, and nuclear factor kappa B (NF-κB) activation as evidenced by the attenuation of inhibitor of kappa B alpha phosphorylation and subsequent NF-κB DNA binding. These effects of AS-IV were abolished by the suppression of NOS or Ca2+- and PI3K/Akt-dependent cascade. Our findings indicate that AS-IV attenuates inflammatory responses triggered by Ang II possibly via the activation of Ca2+/PI3K/Akt/eNOS/NO pathway in endothelial cells.
Collapse
Affiliation(s)
- Shiyu Zhang
- Division of Cardiology and Central Laboratory, First Affiliated Hospital, Henan University of Traditional Chinese Medicine, Zhengzhou 450000, China
| | - Shijie Li
- Division of Cardiology and Central Laboratory, First Affiliated Hospital, Henan University of Traditional Chinese Medicine, Zhengzhou 450000, China
| | - Shiyang Xie
- Division of Cardiology and Central Laboratory, First Affiliated Hospital, Henan University of Traditional Chinese Medicine, Zhengzhou 450000, China
| | - Lin Cui
- Division of Cardiology and Central Laboratory, First Affiliated Hospital, Henan University of Traditional Chinese Medicine, Zhengzhou 450000, China
| | - Yuan Gao
- Division of Cardiology and Central Laboratory, First Affiliated Hospital, Henan University of Traditional Chinese Medicine, Zhengzhou 450000, China
| | - Youping Wang
- Division of Cardiology and Central Laboratory, First Affiliated Hospital, Henan University of Traditional Chinese Medicine, Zhengzhou 450000, China
| |
Collapse
|
9
|
Tripathi A, Pandey VK, Sharma G, Sharma AR, Taufeeq A, Jha AK, Kim JC. Genomic Insights into Dementia: Precision Medicine and the Impact of Gene-Environment Interaction. Aging Dis 2024; 15:2113-2135. [PMID: 38607741 PMCID: PMC11346410 DOI: 10.14336/ad.2024.0322] [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: 01/15/2024] [Accepted: 03/22/2024] [Indexed: 04/14/2024] Open
Abstract
The diagnosis, treatment, and management of dementia provide significant challenges due to its chronic cognitive impairment. The complexity of this condition is further highlighted by the impact of gene-environment interactions. A recent strategy combines advanced genomics and precision medicine methods to explore the complex genetic foundations of dementia. Utilizing the most recent research in the field of neurogenetics, the importance of precise genetic data in explaining the variation seen in dementia patients can be investigated. Gene-environment interactions are important because they influence genetic susceptibilities and aid in the development and progression of dementia. Modified to each patient's genetic profile, precision medicine has the potential to detect groups at risk and make previously unheard-of predictions about the course of diseases. Precision medicine techniques have the potential to completely transform treatment and diagnosis methods. Targeted medications that target genetic abnormalities will probably appear, providing the possibility for more efficient and customized medical interventions. Investigating the relationship between genes and the environment may lead to preventive measures that would enable people to change their surroundings and minimize the risk of dementia, leading to the improved lifestyle of affected people. This paper provides a comprehensive overview of the genomic insights into dementia, emphasizing the pivotal role of precision medicine, and gene-environment interactions.
Collapse
Affiliation(s)
- Anjali Tripathi
- Department of Biotechnology, Sharda School of Engineering and Technology, Sharda University, Greater Noida, Uttar Pradesh, India
| | - Vinay Kumar Pandey
- Division of Research & Innovation (DRI), School of Applied & Life Sciences, Uttaranchal University, Dehradun, Uttarakhand, India
| | - Garima Sharma
- Department of Biomedical Science & Institute of Bioscience and Biotechnology, Kangwon National University, Chuncheon 24341, Republic of Korea
| | - Ashish Ranjan Sharma
- Institute for Skeletal Aging & Orthopedic Surgery, Hallym University-Chuncheon Sacred Heart Hospital, Chuncheon-si, 24252, Gangwon-do, Republic of Korea
| | - Anam Taufeeq
- Department of Biotechnology, Faculty of Engineering and Technology, Rama University, Kanpur, Uttar Pradesh, India
| | - Abhimanyu Kumar Jha
- Department of Biotechnology, Sharda School of Engineering and Technology, Sharda University, Greater Noida, Uttar Pradesh, India
| | - Jin-Chul Kim
- Department of Biomedical Science & Institute of Bioscience and Biotechnology, Kangwon National University, Chuncheon 24341, Republic of Korea
| |
Collapse
|
10
|
Amioka N, Franklin MK, Kukida M, Zhu L, Moorleghen JJ, Howatt DA, Katsumata Y, Mullick AE, Yanagita M, Martinez-Irizarry MM, Sandoval RM, Dunn KW, Sawada H, Daugherty A, Lu HS. Renal Proximal Tubule Cell-specific Megalin Deletion Does Not Affect Atherosclerosis But Induces Tubulointerstitial Nephritis in Mice Fed Western Diet. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.11.592234. [PMID: 38798535 PMCID: PMC11118422 DOI: 10.1101/2024.05.11.592234] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
Background Pharmacological inhibition of megalin (also known as low-density lipoprotein receptor-related protein 2: LRP2) attenuates atherosclerosis in hypercholesterolemic mice. Since megalin is abundant in renal proximal tubule cells (PTCs), the purpose of this study was to determine whether PTC-specific deletion of megalin reduces hypercholesterolemia-induced atherosclerosis in mice. Methods Female Lrp2 f/f mice were bred with male Ndrg1-Cre ERT2 +/0 mice to develop PTC-LRP2 +/+ and -/- littermates. To study atherosclerosis, all mice were bred to an LDL receptor -/- background and fed a Western diet to induce atherosclerosis. Results PTC-specific megalin deletion did not attenuate atherosclerosis in LDL receptor -/- mice in either sex. Serendipitously, we discovered that PTC-specific megalin deletion led to interstitial infiltration of CD68+ cells and tubular atrophy. The pathology was only evident in male PTC-LRP2 -/- mice fed the Western diet, but not in mice fed a normal laboratory diet. Renal pathologies were also observed in male PTC-LRP2 -/- mice in an LDL receptor +/+ background fed the same Western diet, demonstrating that the renal pathologies were dependent on diet and not hypercholesterolemia. In contrast, female PTC-LRP2 -/- mice had no apparent renal pathologies. In vivo multiphoton microscopy demonstrated that PTC-specific megalin deletion dramatically diminished albumin accumulation in PTCs within 10 days of Western diet feeding. RNA sequencing analyses demonstrated the upregulation of inflammation-related pathways in kidney. Conclusions PTC-specific megalin deletion does not affect atherosclerosis, but leads to tubulointerstitial nephritis in mice fed Western diet, with severe pathologies in male mice.
Collapse
Affiliation(s)
- Naofumi Amioka
- Saha Cardiovascular Research Center and Saha Aortic Center
| | | | | | - Liyuan Zhu
- Saha Cardiovascular Research Center and Saha Aortic Center
| | | | | | | | | | - Motoko Yanagita
- Department of Nephrology, Kyoto University Graduate School of Medicine
- Institute for the Advanced Study of Human Biology (WPI-ASHBi), Kyoto University, Kyoto, Japan
| | | | - Ruben M. Sandoval
- Department of Medicine, Indiana University, Indianapolis, Indiana, USA
| | - Kenneth W. Dunn
- Department of Medicine, Indiana University, Indianapolis, Indiana, USA
| | - Hisashi Sawada
- Saha Cardiovascular Research Center and Saha Aortic Center
- Department of Physiology, University of Kentucky, Lexington, Kentucky, USA
| | - Alan Daugherty
- Saha Cardiovascular Research Center and Saha Aortic Center
- Department of Physiology, University of Kentucky, Lexington, Kentucky, USA
| | - Hong S. Lu
- Saha Cardiovascular Research Center and Saha Aortic Center
- Department of Physiology, University of Kentucky, Lexington, Kentucky, USA
| |
Collapse
|
11
|
Șoșdean R, Dănilă MD, Ionică LN, Pescariu AS, Mircea M, Ionac A, Mornoș C, Luca CT, Feier HB, Muntean DM, Sturza A. Monoamine Oxidase Contributes to Valvular Oxidative Stress: A Prospective Observational Pilot Study in Patients with Severe Mitral Regurgitation. Int J Mol Sci 2024; 25:10307. [PMID: 39408637 PMCID: PMC11477003 DOI: 10.3390/ijms251910307] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Revised: 09/09/2024] [Accepted: 09/20/2024] [Indexed: 10/20/2024] Open
Abstract
Monoamine oxidases (MAOs), mitochondrial enzymes that constantly produce hydrogen peroxide (H2O2) as a byproduct of their activity, have been recently acknowledged as contributors to oxidative stress in cardiometabolic pathologies. The present study aimed to assess whether MAOs are mediators of valvular oxidative stress and interact in vitro with angiotensin 2 (ANG2) to mimic the activation of the renin-angiotensin system. To this aim, valvular tissue samples were harvested from 30 patients diagnosed with severe primary mitral regurgitation and indication for surgical repair. Their reactive oxygen species (ROS) levels were assessed by means of a ferrous oxidation xylenol orange (FOX) assay, while MAO expression was assessed by immune fluorescence (protein) and qRT-PCR (mRNA). The experiments were performed using native valvular tissue acutely incubated or not with angiotensin 2 (ANG2), MAO inhibitors (MAOI) and the angiotensin receptor blocker, irbesartan (Irb). Correlations between oxidative stress and echocardiographic parameters were also analyzed. Ex vivo incubation with ANG2 increased MAO-A and -B expression and ROS generation. The level of valvular oxidative stress was negatively correlated with the left ventricular ejection fraction. MAOI and Irb reduced valvular H2O2. production. In conclusion, both MAO isoforms are expressed in pathological human mitral valves and contribute to local oxidative stress and ventricular functional impairment and can be modulated by the local renin-angiotensin system.
Collapse
Affiliation(s)
- Raluca Șoșdean
- Department VI—Cardiology, “Victor Babeș” University of Medicine and Pharmacy of Timișoara, E. Murgu Sq. no 2, 300041 Timișoara, Romania; (R.Ș.); (A.S.P.); (A.I.); (C.M.); (C.T.L.)
- Research Centre of the Institute of Cardiovascular Diseases, G. Adam Str. no 13A, 300310 Timișoara, Romania; (M.M.); (H.B.F.)
| | - Maria D. Dănilă
- Department III—Pathophysiology, “Victor Babeș” University of Medicine and Pharmacy of Timișoara, E. Murgu Sq. no 2, 300041 Timișoara, Romania; (D.M.M.); (A.S.)
- Centre for Translational Research and Systems Medicine, “Victor Babeș” University of Medicine and Pharmacy of Timișoara, E. Murgu Sq. no 2, 300041 Timișoara, Romania;
| | - Loredana N. Ionică
- Centre for Translational Research and Systems Medicine, “Victor Babeș” University of Medicine and Pharmacy of Timișoara, E. Murgu Sq. no 2, 300041 Timișoara, Romania;
- Department X—Medical Semiotics I, “Victor Babeș” University of Medicine and Pharmacy of Timișoara, E. Murgu Sq. no 2, 300041 Timișoara, Romania
| | - Alexandru S. Pescariu
- Department VI—Cardiology, “Victor Babeș” University of Medicine and Pharmacy of Timișoara, E. Murgu Sq. no 2, 300041 Timișoara, Romania; (R.Ș.); (A.S.P.); (A.I.); (C.M.); (C.T.L.)
- Research Centre of the Institute of Cardiovascular Diseases, G. Adam Str. no 13A, 300310 Timișoara, Romania; (M.M.); (H.B.F.)
| | - Monica Mircea
- Research Centre of the Institute of Cardiovascular Diseases, G. Adam Str. no 13A, 300310 Timișoara, Romania; (M.M.); (H.B.F.)
| | - Adina Ionac
- Department VI—Cardiology, “Victor Babeș” University of Medicine and Pharmacy of Timișoara, E. Murgu Sq. no 2, 300041 Timișoara, Romania; (R.Ș.); (A.S.P.); (A.I.); (C.M.); (C.T.L.)
- Research Centre of the Institute of Cardiovascular Diseases, G. Adam Str. no 13A, 300310 Timișoara, Romania; (M.M.); (H.B.F.)
| | - Cristian Mornoș
- Department VI—Cardiology, “Victor Babeș” University of Medicine and Pharmacy of Timișoara, E. Murgu Sq. no 2, 300041 Timișoara, Romania; (R.Ș.); (A.S.P.); (A.I.); (C.M.); (C.T.L.)
- Research Centre of the Institute of Cardiovascular Diseases, G. Adam Str. no 13A, 300310 Timișoara, Romania; (M.M.); (H.B.F.)
| | - Constantin T. Luca
- Department VI—Cardiology, “Victor Babeș” University of Medicine and Pharmacy of Timișoara, E. Murgu Sq. no 2, 300041 Timișoara, Romania; (R.Ș.); (A.S.P.); (A.I.); (C.M.); (C.T.L.)
- Research Centre of the Institute of Cardiovascular Diseases, G. Adam Str. no 13A, 300310 Timișoara, Romania; (M.M.); (H.B.F.)
| | - Horea B. Feier
- Research Centre of the Institute of Cardiovascular Diseases, G. Adam Str. no 13A, 300310 Timișoara, Romania; (M.M.); (H.B.F.)
- Department VI—Cardiovascular Surgery, “Victor Babeș” University of Medicine and Pharmacy of Timișoara, E. Murgu Square no 2, 300041 Timișoara, Romania
| | - Danina M. Muntean
- Department III—Pathophysiology, “Victor Babeș” University of Medicine and Pharmacy of Timișoara, E. Murgu Sq. no 2, 300041 Timișoara, Romania; (D.M.M.); (A.S.)
- Centre for Translational Research and Systems Medicine, “Victor Babeș” University of Medicine and Pharmacy of Timișoara, E. Murgu Sq. no 2, 300041 Timișoara, Romania;
| | - Adrian Sturza
- Department III—Pathophysiology, “Victor Babeș” University of Medicine and Pharmacy of Timișoara, E. Murgu Sq. no 2, 300041 Timișoara, Romania; (D.M.M.); (A.S.)
- Centre for Translational Research and Systems Medicine, “Victor Babeș” University of Medicine and Pharmacy of Timișoara, E. Murgu Sq. no 2, 300041 Timișoara, Romania;
- “Pius Brinzeu” Emergency County Hospital, 300723 Timisoara, Romania
| |
Collapse
|
12
|
Apostolopoulos V, Georgiou N, Tzeli D, Mavromoustakos T, Moore GJ, Kelaidonis K, Matsoukas MT, Tsiodras S, Swiderski J, Kate Gadanec L, Zulli A, Chasapis CT, Matsoukas JM. Density functional theory and enzyme studies support interactions between angiotensin receptor blockers and angiotensin converting enzyme-2: Relevance to coronavirus 2019. Bioorg Chem 2024; 150:107602. [PMID: 38959647 DOI: 10.1016/j.bioorg.2024.107602] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2024] [Revised: 06/14/2024] [Accepted: 06/26/2024] [Indexed: 07/05/2024]
Abstract
The binding affinities and interactions between eight drug candidates, both commercially available (candesartan; losartan; losartan carboxylic acid; nirmatrelvir; telmisartan) and newly synthesized benzimidazole-N-biphenyltetrazole (ACC519T), benzimidazole bis-N,N'-biphenyltetrazole (ACC519T(2) and 4-butyl-N,N-bis([2-(2H-tetrazol-5-yl)biphenyl-4-yl]) methyl (BV6), and the active site of angiotensin-converting enzyme-2 (ACE2) were evaluated for their potential as inhibitors against SARS-CoV-2 and regulators of ACE2 function through Density Functional Theory methodology and enzyme activity assays, respectively. Notably, telmisartan and ACC519T(2) exhibited pronounced binding affinities, forming strong interactions with ACE2's active center, favorably accepting proton from the guanidinium group of arginine273. The ordering of candidates by binding affinity and reactivity descriptors, emerged as telmisartan > ACC519T(2) > candesartan > ACC519T > losartan carboxylic acid > BV6 > losartan > nirmatrelvir. Proton transfers among the active center amino acids revealed their interconnectedness, highlighting a chain-like proton transfer involving tyrosine, phenylalanine, and histidine. Furthermore, these candidates revealed their potential antiviral abilities by influencing proton transfer within the ACE2 active site. Furthermore, through an in vitro pharmacological assays we determined that candesartan and the BV6 derivative, 4-butyl-N,N0-bis[20-2Htetrazol-5-yl)bipheyl-4-yl]methyl)imidazolium bromide (BV6(K+)2) also contain the capacity to increase ACE2 functional activity. This comprehensive analysis collectively underscores the promise of these compounds as potential therapeutic agents against SARS-CoV-2 by targeting crucial protein interactions.
Collapse
Affiliation(s)
- Vasso Apostolopoulos
- Institute for Health and Sport, Victoria University, Melbourne, Victoria 3030, Australia; Immunology Program, Australian Institute for Musculoskeletal Science (AIMSS), Melbourne, Victoria 3021, Australia.
| | - Nikitas Georgiou
- Laboratory of Organic Chemistry, Department of Chemistry, National and Kapodistrian University of Athens, Panepistimioupolis Zografou, 11571 Athens, Greece.
| | - Demeter Tzeli
- Laboratory of Physical Chemistry, Department of Chemistry, National and Kapodistrian University of Athens, Panepistimioupolis Zografou, 11571 Athens, Greece; Theoretical and Physical Chemistry Institute, National Hellenic Research Foundation, 48 Vassileos Constantinou Ave., 11635 Athens, Greece.
| | - Thomas Mavromoustakos
- Laboratory of Organic Chemistry, Department of Chemistry, National and Kapodistrian University of Athens, Panepistimioupolis Zografou, 11571 Athens, Greece.
| | - Graham J Moore
- Pepmetics Inc., 772 Murphy Place, Victoria, BC V8Y 3H4, Canada; Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 1N4, Canada.
| | | | | | - Sotirios Tsiodras
- 4th Department of Internal Medicine, School of Medicine, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Jordan Swiderski
- Institute for Health and Sport, Victoria University, Melbourne, Victoria 3030, Australia.
| | - Laura Kate Gadanec
- Institute for Health and Sport, Victoria University, Melbourne, Victoria 3030, Australia.
| | - Anthony Zulli
- Institute for Health and Sport, Victoria University, Melbourne, Victoria 3030, Australia.
| | - Christos T Chasapis
- Institute of Chemical Biology, National Hellenic Research Foundation, 11635 Athens, Greece.
| | - John M Matsoukas
- Institute for Health and Sport, Victoria University, Melbourne, Victoria 3030, Australia; Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 1N4, Canada; NewDrug PC, Patras Science Park, Patras, 26504, Greece; Department of Chemistry, University of Patras, Patras, Greece.
| |
Collapse
|
13
|
Summart R, Imsoonthornruksa S, Yongsawatdigul J, Ketudat-Cairns M, Udomsil N. Characterization and molecular docking of tetrapeptides with cellular antioxidant and ACE inhibitory properties from cricket ( Acheta domesticus) protein hydrolysate. Heliyon 2024; 10:e35156. [PMID: 39166016 PMCID: PMC11333913 DOI: 10.1016/j.heliyon.2024.e35156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Revised: 07/23/2024] [Accepted: 07/23/2024] [Indexed: 08/22/2024] Open
Abstract
Wide-ranging bioactivities of enzymatically digested insect protein to produce peptides have been targeted for functional food development. In this study, fractionated peptides obtained from cricket (Acheta domesticus) protein hydrolysate by alcalase digestion were identified and evaluated for their bioactivities. Peptide fractions F44, F45, and F46, isolated through size exclusion chromatography, demonstrated strong cytoprotective effects on SH-SY5Y and HepG2 cells exposed to H2O2. This was evidenced by a 2-fold decrease in reactive oxygen species (ROS) accumulation in the cells and a 3-fold upregulation of genes encoding antioxidant enzymes. The F45 peptide fractions also showed chemical antioxidant activities ranging from approximately 290 to 393 mg trolox/g peptide, measured by DPPH, ABTS, and FRAP assays. Furthermore, F45 demonstrated the highest angiotensin-converting enzyme I (ACE) inhibitory activity, 57.93 %. F45 induced higher levels of Nrf2, SOD1, SOD2, CAT, GSR, and GPx4 gene expression in SH-SY5Y and HepG2 cells compared to cells treated with H2O2 and no peptides (p < 0.05). Cells treated with H2O2 and F45 exhibited significantly increased antioxidant enzyme activity, including SOD, CAT, GSR, and GPx (p < 0.05). The F45B fraction from F45 was sequenced to obtain FVEG and FYDQ tetrapeptides. Molecular docking analysis revealed their high binding affinity to cellular antioxidant enzymes (SOD, CAT, GSR, GPx1, and GPx4), an antioxidant-related protein (Keap1), and ACE. These results suggest that the novel tetrapeptides from Acheta domesticus demonstrate important biological activities, establishing them as significant cellular antioxidant activities and a potential source of antihypertensive peptides.
Collapse
Affiliation(s)
- Ratasark Summart
- Division of Food Technology, Mahidol University Kanchanaburi Campus, Kanchanaburi, 71150, Thailand
| | - Sumeth Imsoonthornruksa
- Center for Biomolecular Structure Function and Application, School of Biotechnology, Suranaree University of Technology, Nakhon Ratchasima, 30000, Thailand
| | - Jirawat Yongsawatdigul
- School of Food Technology, Institute of Agricultural Technology, Suranaree University of Technology, Nakhon Ratchasima, 30000, Thailand
| | - Mariena Ketudat-Cairns
- Center for Biomolecular Structure Function and Application, School of Biotechnology, Suranaree University of Technology, Nakhon Ratchasima, 30000, Thailand
| | - Natteewan Udomsil
- Division of Food Technology, Mahidol University Kanchanaburi Campus, Kanchanaburi, 71150, Thailand
| |
Collapse
|
14
|
Wu W, Liu R, Guo S, Song W, Hua Y, Hong M, Zheng J, Zhu Y, Cao P, Duan JA. Mechanism and functional substances of Saiga antelope horn in treating hypertension with liver-yang hyperactivity syndrome explored using network pharmacology and metabolomics. JOURNAL OF ETHNOPHARMACOLOGY 2024; 330:118193. [PMID: 38636578 DOI: 10.1016/j.jep.2024.118193] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Revised: 04/07/2024] [Accepted: 04/11/2024] [Indexed: 04/20/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Saiga antelope horn (SAH) is a traditional Chinese medicine for treating hypertension with liver-yang hyperactivity syndrome (Gan-Yang-Shang-Kang, GYSK), that has a long history of clinical application and precise efficacy, but its mechanism and functional substances are still unknown. Based on the demand for alternative research on the rare and endangered SAH, the group designed and carried out the following studies. AIM OF THE STUDY The purpose of this research was to demonstrate the functional substances and mechanisms of SAH in the treatment of GYSK hypertension. MATERIALS AND METHODS The GYSK-SHR model was constructed by administering a decoction of aconite to spontaneously hypertensive rats (SHRs). Blood pressure (BP), behavioural tests related to GYSK, and pathological changes in the kidneys, heart and aorta were measured to investigate the effects of SAH on GYSK-SHRs. Proteomic analysis was used to identify the keratins and peptides of SAH. Moreover, network pharmacology and plasma metabolomics studies were carried out to reveal the mechanisms by which functional peptides in SAH regulate GYSK-hypertension. RESULTS SAH has a significant antihypertensive effect on GYSK hypertensive animals. It has also been proven to be effective in protecting the function and structural integrity of the kidneys, heart and aorta. Moreover, SAH improved the abnormalities of 31 plasma biomarkers in rats. By constructing a "biomarker-target-peptide" network, 10 functional peptides and two key targets were screened for antihypertensive effects of SAH. The results indicated that SAH may exert a therapeutic effect by re-establishing the imbalance of renin-angiotensin (RAS) system. CONCLUSIONS Functional peptides from keratin contained in SAH are the main material basis for the treatment of GYSK-hypertension and exhibited the protective effect on the GYSK-SHR model through the RAS system.
Collapse
Affiliation(s)
- Wenxing Wu
- National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, and Jiangsu Key Laboratory for High Technology Research of Traditional Chinese Medicine Formulae, Nanjing University of Chinese Medicine, Nanjing, 210023, China; School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China; Animal-Derived Chinese Medicine and Functional Peptides International Collaboration Joint Laboratory, Nanjing, 210023, China
| | - Rui Liu
- National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, and Jiangsu Key Laboratory for High Technology Research of Traditional Chinese Medicine Formulae, Nanjing University of Chinese Medicine, Nanjing, 210023, China; School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China; Animal-Derived Chinese Medicine and Functional Peptides International Collaboration Joint Laboratory, Nanjing, 210023, China.
| | - Sheng Guo
- National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, and Jiangsu Key Laboratory for High Technology Research of Traditional Chinese Medicine Formulae, Nanjing University of Chinese Medicine, Nanjing, 210023, China; School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Wencong Song
- National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, and Jiangsu Key Laboratory for High Technology Research of Traditional Chinese Medicine Formulae, Nanjing University of Chinese Medicine, Nanjing, 210023, China; School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Yongqing Hua
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Min Hong
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Jie Zheng
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Yue Zhu
- National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, and Jiangsu Key Laboratory for High Technology Research of Traditional Chinese Medicine Formulae, Nanjing University of Chinese Medicine, Nanjing, 210023, China; School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Peng Cao
- National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, and Jiangsu Key Laboratory for High Technology Research of Traditional Chinese Medicine Formulae, Nanjing University of Chinese Medicine, Nanjing, 210023, China; Animal-Derived Chinese Medicine and Functional Peptides International Collaboration Joint Laboratory, Nanjing, 210023, China
| | - Jin-Ao Duan
- National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, and Jiangsu Key Laboratory for High Technology Research of Traditional Chinese Medicine Formulae, Nanjing University of Chinese Medicine, Nanjing, 210023, China; School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China; Animal-Derived Chinese Medicine and Functional Peptides International Collaboration Joint Laboratory, Nanjing, 210023, China.
| |
Collapse
|
15
|
Wang F, Liu L, Ruan H, Chen X, Zhang Y, Yu Z, Li Y, Guan Y, Wang J, Huang K, Yu S, Cao Y, Ding C, Chang L, Huang Y, Chen X, Lv Q, Ma C. A randomized, double-blind, placebo-controlled, phase IIa, clinical study on investigating the efficacy and safety of SPH3127 tablet in patients with essential hypertension. Hypertens Res 2024; 47:1925-1933. [PMID: 38632457 PMCID: PMC11224011 DOI: 10.1038/s41440-024-01657-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Revised: 01/29/2024] [Accepted: 03/01/2024] [Indexed: 04/19/2024]
Abstract
Around 70% of patients diagnosed with hypertension exhibit increased levels of renin. SPH3127, an inventive renin inhibitor, has shown favorable tolerability and sustained pharmacodynamic inhibitory impact on plasma renin activity (PRA) during previous phase I trials. This phase II study was conducted to investigate the efficacy and safety of SPH3127 in patients with essential hypertension. This study was conducted in patients with mild to moderate essential hypertension, utilizing a randomized, double-blind, placebo-controlled design. The patients were administered either tablet of SPH3127 at doses of 50 mg, 100 mg, or 200 mg, or a placebo. A total of 122 patients were included in the study, with 121 patients included in the full analysis set. Among these patients, there were 30 individuals in each subgroup receiving different dosage regimens of SPH3127, and 31 patients in the placebo group. The reductions in mean sitting diastolic blood pressure (msDBP) after 8 weeks compared to baseline were 5.7 ± 9.5, 8.6 ± 8.8, and 3.8 ± 10.6 mmHg in the SPH3127 50-, 100-, and 200 mg groups, respectively. In the placebo group, the reduction was 3.1 ± 8.4 mmHg. The corresponding reductions in mean sitting systolic blood pressure (msSBP) were 11.8 ± 13.0, 13.8 ± 11.2, 11.1 ± 13.1, and 7.7 ± 9.7 mmHg in each respective group. SPH3127 is a promising drug for the treatment of patients with essential hypertension. The recommended dosage is 100 mg daily.Clinical trial registration: This study was registered in ClinicalTrials.gov (NCT03756103).
Collapse
Affiliation(s)
- Fang Wang
- Department of Cardiology, Beijing Hospital, No. 1 Dahua Road, Dongcheng District, Beijing, China
| | - Ling Liu
- Department of Cardiovascular Medicine, The Second Xiangya Hospital of Central South University, No. 139, Renmin Middle Road, Changsha, Hunan Province, China
| | - Hongyun Ruan
- Department of Cardiology, Xuzhou Central Hospital, No. 199 Jiefang South Road, Quanshan District, Xuzhou, Jiangsu Province, China
| | - Xiaoping Chen
- Department of Cardiology, West China Hospital of Sichuan University, No. 37 Guoxue Lane, Wuhou District, Chengdu, Sichuan Province, China
| | - Yue Zhang
- Department of Cardiology, The affiliated Hospital of inner Mongolia Medical University, No.1, Tongtong North Street, Huimin District, Hohhot, Inner Mongolia Autonomous Region, China
| | - Zaixin Yu
- Department of Cardiology, Xiangya Hospital Central South University, No. 87, Xiangya Road, Kaifu District, Changsha, Hunan Province, China
| | - Yuhui Li
- Department of Cardiology 1, Guangdong Second Provincial Central Hospital, No. 466, Xingang Middle Road, Haizhu District, Guangzhou, Guangdong Province, China
| | - Yang Guan
- Department of Cardiology 2, Beijing Anzhen Hospital, Capital Medical University, No. 2 Anzhen Road, Chaoyang District, Beijing, China
| | - Jiguang Wang
- Department of Hypertension, Ruijin Hospital Affiliated to The Shanghai Jiao Tong University Medical School, No. 197, Ruijin 2nd Road, Huangpu District, Shanghai, China
| | - Kai Huang
- Department of Cardiology, Union Hospital Tongji Medial College Huazhong University of Science and Technology, No. 1277 Jiefang Avenue, Wuhan City, Hubei Province, China
| | - Shunjiang Yu
- Department of New Drug Registration and Clinical R&D, Shanghai Pharmaceuticals Holding Co. Ltd, No. 200 Taicang Road, Huangpu District, Shanghai, China
| | - Yuanyuan Cao
- Department of New Drug Registration and Clinical R&D, Shanghai Pharmaceuticals Holding Co. Ltd, No. 200 Taicang Road, Huangpu District, Shanghai, China
| | - Cungang Ding
- Department of New Drug Registration and Clinical R&D, Shanghai Pharmaceuticals Holding Co. Ltd, No. 200 Taicang Road, Huangpu District, Shanghai, China
| | - Lin Chang
- Department of New Drug Registration and Clinical R&D, Shanghai Pharmaceuticals Holding Co. Ltd, No. 200 Taicang Road, Huangpu District, Shanghai, China
| | - Yaohua Huang
- Department of New Drug Registration and Clinical R&D, Shanghai Pharmaceuticals Holding Co. Ltd, No. 200 Taicang Road, Huangpu District, Shanghai, China
| | - Xiangjuan Chen
- Department of New Drug Registration and Clinical R&D, Shanghai Pharmaceuticals Holding Co. Ltd, No. 200 Taicang Road, Huangpu District, Shanghai, China
| | - Qiang Lv
- Department of Cardiology 2, Beijing Anzhen Hospital, Capital Medical University, No. 2 Anzhen Road, Chaoyang District, Beijing, China.
| | - Changsheng Ma
- Department of Cardiology 2, Beijing Anzhen Hospital, Capital Medical University, No. 2 Anzhen Road, Chaoyang District, Beijing, China.
| |
Collapse
|
16
|
Tu C, Yang S, Yang M, Liu L, Tao J, Zhang L, Huang X, Tian Y, Li N, Lin L, Qin Z. Mechanisms of persistent hemolysis-induced middle kidney injury in grass carp (Ctenopharyngodon idella). FISH & SHELLFISH IMMUNOLOGY 2024; 150:109603. [PMID: 38704112 DOI: 10.1016/j.fsi.2024.109603] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 04/20/2024] [Accepted: 05/02/2024] [Indexed: 05/06/2024]
Abstract
Infection-induced hemolysis results in intravascular hemolysis, which releases hemoglobin (Hb) into the tissues. Free Hb exhibits cytotoxic, oxidative, and pro-inflammatory effects, leading to systemic inflammation, vascular constriction dysfunction, thrombosis, and proliferative vascular lesions. Currently, the impact of intravascular hemolysis on the middle kidney in fish is unclear. Here, the injection of phenylhydrazine (PHZ) was used to establish a persistent hemolysis model in grass carp. The determination results revealed that the PHZ-induced hemolysis caused conspicuous tissue damage in the kidneys of grass carp, increased the levels of Cr in the serum and the expression indicators of kidney injury-related genes in the middle kidney. Prussian blue staining indicated that PHZ-induced hemolysis significantly increased the deposition of iron ions in the kidneys of grass carp, and activated the expression levels of iron metabolism-related genes. The results of oxidative damage-related experiments indicate that under PHZ treatment, the activity of middle kidney cells decreases, and the production of oxidative damage markers malondialdehyde (MDA) and 4-hydroxynonenal (4-HNE) increases, simultaneously inhibiting the activity of antioxidant enzymes and upregulating the transcription levels of antioxidant enzyme-related genes. Additionally, the analysis of inflammatory factors revealed a significant upregulation of genes associated with inflammation induced by PHZ-induced hemolysis. The transcriptome analysis was performed to further explore the molecular regulatory effects of hemolysis on tissues, the analysis revealed the treatment of PHZ activated various of programmed cell death (PCD) pathways, including ferroptosis, apoptosis, and autophagy. In summary, this study found that sustained hemolysis in fish results in Hb and iron ion deposition in middle kidney, promoting oxidative damage, ultimately inducing various forms of PCD.
Collapse
Affiliation(s)
- Chengming Tu
- Guangdong Provincial Water Environment and Aquatic Products Security Engineering Technology Research Center, Guangzhou Key Laboratory of Aquatic Animal Diseases and Waterfowl Breeding, College of Animal Sciences and Technology, Zhongkai University of Agriculture and Engineering, Guangzhou, Guangdong Province, 510222, China
| | - Shiyi Yang
- Guangdong Provincial Water Environment and Aquatic Products Security Engineering Technology Research Center, Guangzhou Key Laboratory of Aquatic Animal Diseases and Waterfowl Breeding, College of Animal Sciences and Technology, Zhongkai University of Agriculture and Engineering, Guangzhou, Guangdong Province, 510222, China
| | - Minxuan Yang
- Guangdong Provincial Water Environment and Aquatic Products Security Engineering Technology Research Center, Guangzhou Key Laboratory of Aquatic Animal Diseases and Waterfowl Breeding, College of Animal Sciences and Technology, Zhongkai University of Agriculture and Engineering, Guangzhou, Guangdong Province, 510222, China
| | - Lihan Liu
- Guangdong Provincial Water Environment and Aquatic Products Security Engineering Technology Research Center, Guangzhou Key Laboratory of Aquatic Animal Diseases and Waterfowl Breeding, College of Animal Sciences and Technology, Zhongkai University of Agriculture and Engineering, Guangzhou, Guangdong Province, 510222, China
| | - Junjie Tao
- Guangdong Provincial Water Environment and Aquatic Products Security Engineering Technology Research Center, Guangzhou Key Laboratory of Aquatic Animal Diseases and Waterfowl Breeding, College of Animal Sciences and Technology, Zhongkai University of Agriculture and Engineering, Guangzhou, Guangdong Province, 510222, China
| | - Linpeng Zhang
- Guangdong Provincial Water Environment and Aquatic Products Security Engineering Technology Research Center, Guangzhou Key Laboratory of Aquatic Animal Diseases and Waterfowl Breeding, College of Animal Sciences and Technology, Zhongkai University of Agriculture and Engineering, Guangzhou, Guangdong Province, 510222, China
| | - Xiaoman Huang
- Guangdong Provincial Water Environment and Aquatic Products Security Engineering Technology Research Center, Guangzhou Key Laboratory of Aquatic Animal Diseases and Waterfowl Breeding, College of Animal Sciences and Technology, Zhongkai University of Agriculture and Engineering, Guangzhou, Guangdong Province, 510222, China
| | - Ye Tian
- Guangdong Provincial Water Environment and Aquatic Products Security Engineering Technology Research Center, Guangzhou Key Laboratory of Aquatic Animal Diseases and Waterfowl Breeding, College of Animal Sciences and Technology, Zhongkai University of Agriculture and Engineering, Guangzhou, Guangdong Province, 510222, China
| | - Ningjing Li
- Guangdong Provincial Water Environment and Aquatic Products Security Engineering Technology Research Center, Guangzhou Key Laboratory of Aquatic Animal Diseases and Waterfowl Breeding, College of Animal Sciences and Technology, Zhongkai University of Agriculture and Engineering, Guangzhou, Guangdong Province, 510222, China
| | - Li Lin
- Guangdong Provincial Water Environment and Aquatic Products Security Engineering Technology Research Center, Guangzhou Key Laboratory of Aquatic Animal Diseases and Waterfowl Breeding, College of Animal Sciences and Technology, Zhongkai University of Agriculture and Engineering, Guangzhou, Guangdong Province, 510222, China.
| | - Zhendong Qin
- Guangdong Provincial Water Environment and Aquatic Products Security Engineering Technology Research Center, Guangzhou Key Laboratory of Aquatic Animal Diseases and Waterfowl Breeding, College of Animal Sciences and Technology, Zhongkai University of Agriculture and Engineering, Guangzhou, Guangdong Province, 510222, China.
| |
Collapse
|
17
|
Bian J, Zhu Y, Tian P, Yang Q, Li Z. Adaptor protein HIP-55 promotes macrophage M1 polarization through promoting AP-1 complex activation. Cell Signal 2024; 117:111124. [PMID: 38417633 DOI: 10.1016/j.cellsig.2024.111124] [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: 12/15/2023] [Revised: 02/23/2024] [Accepted: 02/25/2024] [Indexed: 03/01/2024]
Abstract
Overwhelming macrophage M1 polarization induced by malfunction of the renin-angiotensin-aldosterone system (RAAS) initiates inflammatory responses, which play a crucial role in various cardiovascular diseases. However, the underlying regulatory mechanism remains elusive. Here, we identified adaptor protein HIP-55 as a critical regulator of macrophage M1 polarization. The expression of HIP-55 was upregulated in M1 macrophage induced by Ang II. Overexpression of HIP-55 significantly promoted Ang II-induced macrophage M1 polarization, whereas genetic deletion of HIP-55 inhibited the Ang II-induced macrophage M1 polarization. Mechanistically, HIP-55 facilitated activator protein-1 (AP-1) complex activation induced by Ang II via promoting ERK1/2 and JNK phosphorylation. Moreover, blocking AP-1 complex activation can attenuate the function of HIP-55 in macrophage polarization. Collectively, our results reveal the role of HIP-55 in macrophage polarization and provide potential therapeutic insights for cardiovascular diseases associated with RAAS dysfunction.
Collapse
Affiliation(s)
- Jingwei Bian
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital; Beijing Key Laboratory of Cardiovascular Receptors Research; Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Ministry of Health; State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing 100191, China
| | - Yuzhong Zhu
- Wuxi School of Medicine, Jiangnan University, Wuxi 214122, China
| | - Panhui Tian
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital; Beijing Key Laboratory of Cardiovascular Receptors Research; Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Ministry of Health; State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing 100191, China; Department of Medical Genetics, Center for Medical Genetics, Peking University Health Science Center, Beijing 100191, China
| | - Qiqi Yang
- Wuxi School of Medicine, Jiangnan University, Wuxi 214122, China
| | - Zijian Li
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital; Beijing Key Laboratory of Cardiovascular Receptors Research; Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Ministry of Health; State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing 100191, China; Department of Pharmacy, Peking University Third Hospital, Beijing 100191, China.
| |
Collapse
|
18
|
Khan RN, Saporito AF, Zenon J, Goodman L, Zelikoff JT. Traffic-related air pollution in marginalized neighborhoods: a community perspective. Inhal Toxicol 2024; 36:343-354. [PMID: 38618680 DOI: 10.1080/08958378.2024.2331259] [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: 11/15/2023] [Accepted: 03/10/2024] [Indexed: 04/16/2024]
Abstract
OBJECTIVES Marginalized communities are exposed to higher levels of traffic-related air pollution (TRAP) than the general population. TRAP exposure is linked to pulmonary toxicity, neurotoxicity, and cardiovascular toxicity often through mechanisms of inflammation and oxidative stress. Early life exposure to TRAP is also implicated in higher rates of asthma in these same communities. There is a critical need for additional epidemiological, in vivo, and in vitro studies to define the health risks of TRAP exposure affecting the most vulnerable groups to set strict, protective air pollution standards in these communities. MATERIALS AND METHODS A literature review was conducted to summarize recent findings (2010-2024) concerning TRAP exposure and toxic mechanisms that are relevant to the most affected underserved communities. CONCLUSIONS Guided by the perspectives of NYC community scientists, this contemporary review of toxicological and epidemiological studies considers how the exposome could lead to disproportionate exposures and health effects in underserved populations.
Collapse
Affiliation(s)
- Rahanna N Khan
- Division of Environmental Medicine, NYU Grossman School of Medicine, New York, NY, USA
| | - Antonio F Saporito
- Division of Environmental Medicine, NYU Grossman School of Medicine, New York, NY, USA
| | - Jania Zenon
- Division of Environmental Medicine, NYU Grossman School of Medicine, New York, NY, USA
| | | | - Judith T Zelikoff
- Division of Environmental Medicine, NYU Grossman School of Medicine, New York, NY, USA
| |
Collapse
|
19
|
Coste B, Delmas P. PIEZO Ion Channels in Cardiovascular Functions and Diseases. Circ Res 2024; 134:572-591. [PMID: 38422173 DOI: 10.1161/circresaha.123.322798] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/02/2024]
Abstract
The cardiovascular system provides blood supply throughout the body and as such is perpetually applying mechanical forces to cells and tissues. Thus, this system is primed with mechanosensory structures that respond and adapt to changes in mechanical stimuli. Since their discovery in 2010, PIEZO ion channels have dominated the field of mechanobiology. These have been proposed as the long-sought-after mechanosensitive excitatory channels involved in touch and proprioception in mammals. However, more and more pieces of evidence point to the importance of PIEZO channels in cardiovascular activities and disease development. PIEZO channel-related cardiac functions include transducing hemodynamic forces in endothelial and vascular cells, red blood cell homeostasis, platelet aggregation, and arterial blood pressure regulation, among others. PIEZO channels contribute to pathological conditions including cardiac hypertrophy and pulmonary hypertension and congenital syndromes such as generalized lymphatic dysplasia and xerocytosis. In this review, we highlight recent advances in understanding the role of PIEZO channels in cardiovascular functions and diseases. Achievements in this quickly expanding field should open a new road for efficient control of PIEZO-related diseases in cardiovascular functions.
Collapse
Affiliation(s)
- Bertrand Coste
- Centre de Recherche en CardioVasculaire et Nutrition, Aix-Marseille Université - INSERM 1263 - INRAE 1260, Marseille, France
| | - Patrick Delmas
- Centre de Recherche en CardioVasculaire et Nutrition, Aix-Marseille Université - INSERM 1263 - INRAE 1260, Marseille, France
| |
Collapse
|
20
|
Choudhary R, Kumari S, Ali M, Thinlas T, Rabyang S, Mishra A. Respiratory tract infection: an unfamiliar risk factor in high-altitude pulmonary edema. Brief Funct Genomics 2024; 23:38-45. [PMID: 36528814 DOI: 10.1093/bfgp/elac048] [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: 09/05/2022] [Revised: 10/26/2022] [Accepted: 11/04/2022] [Indexed: 01/21/2024] Open
Abstract
The dramatic changes in physiology at high altitude (HA) as a result of the characteristic hypobaric hypoxia condition can modify innate and adaptive defense mechanisms of the body. As a consequence, few sojourners visiting HA with mild or asymptomatic infection may have an enhanced susceptibility to high-altitude pulmonary edema (HAPE), an acute but severe altitude sickness. It develops upon rapid ascent to altitudes above 2500 m, in otherwise healthy individuals. Though HAPE has been studied extensively, an elaborate exploration of the HA disease burden and the potential risk factors associated with its manifestation are poorly described. The present review discusses respiratory tract infection (RTI) as an unfamiliar but important risk factor in enhancing HAPE susceptibility in sojourners for two primary reasons. First, the symptoms of RTI s resemble those of HAPE. Secondly, the imbalanced pathways contributing to vascular dysfunction in HAPE also participate in the pathogenesis of the infectious processes. These pathways have a crucial role in shaping host response against viral and bacterial infections and may further worsen the clinical outcomes at HA. Respiratory tract pathogenic agents, if screened in HAPE patients, can help in ascertaining their role in disease risk and also point toward their association with the disease severity. The microbial screenings and identifications of pathogens with diseases are the foundation for describing potential molecular mechanisms underlying host response to the microbial challenge. The prior knowledge of such infections may predict the manifestation of disease etiology and provide better therapeutic options.
Collapse
Affiliation(s)
- Raushni Choudhary
- Cardio Respiratory Disease Unit, CSIR-Institute of Genomics and Integrative Biology, Delhi 110007, India
| | - Swati Kumari
- Cardio Respiratory Disease Unit, CSIR-Institute of Genomics and Integrative Biology, Delhi 110007, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Manzoor Ali
- Cardio Respiratory Disease Unit, CSIR-Institute of Genomics and Integrative Biology, Delhi 110007, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Tashi Thinlas
- Department of Medicine, Sonam Norboo Memorial Hospital, Leh 194101, India
| | - Stanzen Rabyang
- Department of Medicine, Sonam Norboo Memorial Hospital, Leh 194101, India
| | - Aastha Mishra
- Cardio Respiratory Disease Unit, CSIR-Institute of Genomics and Integrative Biology, Delhi 110007, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| |
Collapse
|
21
|
Bakhashab S, O’Neill J, Barber R, Arden C, Weaver JU. Upregulation of Anti-Angiogenic miR-106b-3p Correlates Negatively with IGF-1 and Vascular Health Parameters in a Model of Subclinical Cardiovascular Disease: Study with Metformin Therapy. Biomedicines 2024; 12:171. [PMID: 38255276 PMCID: PMC10813602 DOI: 10.3390/biomedicines12010171] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2023] [Revised: 01/08/2024] [Accepted: 01/09/2024] [Indexed: 01/24/2024] Open
Abstract
Well-controlled type 1 diabetes mellitus (T1DM) is regarded as a model of subclinical cardiovascular disease (CVD), characterized by inflammation and adverse vascular health. However, the underlying mechanisms are not fully understood. We investigated insulin-like growth factor-1 (IGF-1) and IGF-binding protein-3 (IGFBP-3) levels, their correlation to miR-106b-3p expression in a subclinical CVD model, and the cardioprotective effect of metformin. A total of 20 controls and 29 well-controlled T1DM subjects were studied. Plasma IGF-1, IGFBP-3 levels, and miR-106b-3p expression in colony-forming unit-Hills were analyzed and compared with vascular markers. miR-106b-3p was upregulated in T1DM (p < 0.05) and negatively correlated with pro-angiogenic markers CD34+/100-lymphocytes (p < 0.05) and IGF-1 (p < 0.05). IGF-1 was downregulated in T1DM (p < 0.01), which was associated with increased inflammatory markers TNF-α, CRP, and IL-10 and reduced CD34+/100-lymphocytes. IGFBP-3 had no significant results. Metformin had no effect on IGF-1 but significantly reduced miR-106b-3p (p < 0.0001). An Ingenuity Pathway analysis predicted miR-106b-3p to inhibit PDGFA, PIK3CG, GDNF, and ADAMTS13, which activated CVD. Metformin was predicted to be cardioprotective by inhibiting miR-106b-3p. In conclusion: Subclinical CVD is characterized by a cardio-adverse profile of low IGF-1 and upregulated miR-106b-3p. We demonstrated that the cardioprotective effect of metformin may be via downregulation of upregulated miR-106b-3p and its effect on downstream targets.
Collapse
Affiliation(s)
- Sherin Bakhashab
- Biochemistry Department, King Abdulaziz University, P.O. Box 80218, Jeddah 21589, Saudi Arabia;
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne NE2 4HH, UK; (J.O.); (R.B.)
- Center of Excellence in Genomic Medicine Research, King Abdulaziz University, P.O. Box 80216, Jeddah 21589, Saudi Arabia
| | - Josie O’Neill
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne NE2 4HH, UK; (J.O.); (R.B.)
- Biosciences Institute, Newcastle University, Newcastle upon Tyne NE2 4HH, UK;
| | - Rosie Barber
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne NE2 4HH, UK; (J.O.); (R.B.)
- Biosciences Institute, Newcastle University, Newcastle upon Tyne NE2 4HH, UK;
| | - Catherine Arden
- Biosciences Institute, Newcastle University, Newcastle upon Tyne NE2 4HH, UK;
| | - Jolanta U. Weaver
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne NE2 4HH, UK; (J.O.); (R.B.)
- Department of Diabetes, Queen Elizabeth Hospital, Newcastle upon Tyne NE9 6SH, UK
- Vascular Biology and Medicine Theme, Newcastle University, Newcastle upon Tyne NE1 7RU, UK
| |
Collapse
|
22
|
Omidkhah N, Hadizadeh F, Ghodsi R, Kesharwani P, Sahebkar A. In silico Evaluation of NO-Sartans against SARS-CoV-2. Curr Drug Discov Technol 2024; 21:e050324227669. [PMID: 38445698 DOI: 10.2174/0115701638279362240223070810] [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: 10/20/2023] [Revised: 01/16/2024] [Accepted: 01/29/2024] [Indexed: 03/07/2024]
Abstract
INTRODUCTION Numerous clinical trials are currently investigating the potential of nitric oxide (NO) as an antiviral agent against coronaviruses, including SARS-CoV-2. Additionally, some researchers have reported positive effects of certain Sartans against SARS-CoV-2. METHOD Considering the impact of NO-Sartans on the cardiovascular system, we have compiled information on the general structure, synthesis methods, and biological studies of synthesized NOSartans. In silico evaluation of all NO-Sartans and approved sartans against three key SARS-CoV- -2 targets, namely Mpro (PDB ID: 6LU7), NSP16 (PDB ID: 6WKQ), and ACE-2 (PDB ID: 1R4L), was performed using MOE. RESULTS Almost all NO-Sartans and approved sartans demonstrated promising results in inhibiting these SARS-CoV-2 targets. Compound 36 (CLC-1280) showed the best docking scores against the three evaluated targets and was further evaluated using molecular dynamics (MD) simulations. CONCLUSION Based on our in silico studies, CLC-1280 (a Valsartan dinitrate) has the potential to be considered as an inhibitor of the SARS-CoV-2 virus. However, further in vitro and in vivo evaluations are necessary for the drug development process.
Collapse
Affiliation(s)
- Negar Omidkhah
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Medicinal Chemistry, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Farzin Hadizadeh
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Medicinal Chemistry, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Razieh Ghodsi
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Medicinal Chemistry, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Prashant Kesharwani
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard University, New Delhi, 110062, India
| | - Amirhossein Sahebkar
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
23
|
Wee CL, Azemi AK, Mokhtar SS, Yahaya S, Yaacob NS, Rasool AHG. Vitamin D deficiency enhances vascular oxidative stress, inflammation, and angiotensin II levels in the microcirculation of diabetic patients. Microvasc Res 2023; 150:104574. [PMID: 37390963 DOI: 10.1016/j.mvr.2023.104574] [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: 03/27/2023] [Revised: 06/08/2023] [Accepted: 06/27/2023] [Indexed: 07/02/2023]
Abstract
Low vitamin D (vitD) levels have been reported to be a risk factor for diabetes-related cardiovascular complications. This study examined the effects of vitD deficiency on oxidative stress (OS), inflammation, and levels of the vasoconstrictor angiotensin II (Ang II) in the microvascular tissue of type 2 diabetic patients. Patients were categorized into (i) vitD non-deficient diabetics (DNP, n = 10) and (ii) vitD-deficient diabetics (DDP, n = 10), based on their serum 25(OH)D levels. Subcutaneous fat tissues with intact blood vessels were collected during lower limb surgical procedures. The blood vessel were isolated; measurements of the antioxidant enzyme superoxide dismutase (SOD) activity, OS marker malondialdehyde (MDA), Ang II, and the inflammatory marker, TNF-α of the microvascular tissues were determined. Elevated MDA levels and reduced SOD activity, with higher levels of TNF-α and Ang II were observed in the microvascular tissues of DDP compared to DNP. VitD deficiency did not associate with glycemic parameters (fasting blood glucose and glycated hemoglobin) levels. In conclusion, vitD deficiency was correlated with higher microvascular tissue OS, inflammation, and Ang II levels in type 2 diabetic patients. This may contribute to early vasculopathy that occurs in diabetic patients, thus, may contribute to the planning of therapeutic strategies to delay or prevent cardiovascular complications.
Collapse
Affiliation(s)
- Chee Lee Wee
- Department of Pharmacology, School of Medical Sciences, Universiti Sains Malaysia (Health Campus), 16150 Kota Bharu, Kelantan, Malaysia; Department of Chemical Pathology, School of Medical Sciences, Universiti Sains Malaysia (Health Campus), 16150 Kota Bharu, Kelantan, Malaysia.
| | - Ahmad Khusairi Azemi
- Department of Pharmacology, School of Medical Sciences, Universiti Sains Malaysia (Health Campus), 16150 Kota Bharu, Kelantan, Malaysia; Institute of Marine Biotechnology, Universiti Malaysia Terengganu, 21030 Kuala Terengganu, Terengganu, Malaysia.
| | - Siti Safiah Mokhtar
- Department of Pharmacology, School of Medical Sciences, Universiti Sains Malaysia (Health Campus), 16150 Kota Bharu, Kelantan, Malaysia.
| | - Sahran Yahaya
- Department of Orthopedics, School of Medical Sciences, Universiti Sains Malaysia (Health Campus), 16150 Kota Bharu, Kelantan, Malaysia; Hospital Universiti Sains Malaysia, Kubang Kerian, 16150 Kota Bharu, Kelantan, Malaysia.
| | - Nik Soriani Yaacob
- Department of Chemical Pathology, School of Medical Sciences, Universiti Sains Malaysia (Health Campus), 16150 Kota Bharu, Kelantan, Malaysia.
| | - Aida Hanum Ghulam Rasool
- Department of Pharmacology, School of Medical Sciences, Universiti Sains Malaysia (Health Campus), 16150 Kota Bharu, Kelantan, Malaysia; Hospital Universiti Sains Malaysia, Kubang Kerian, 16150 Kota Bharu, Kelantan, Malaysia.
| |
Collapse
|
24
|
Kukida M, Amioka N, Ye D, Chen H, Moorleghen JJ, Liang CL, Howatt DA, Katsumata Y, Yanagita M, Sawada H, Daugherty A, Lu HS. Manipulation of components of the renin angiotensin system in renal proximal tubules fails to alter atherosclerosis in hypercholesterolemic mice. Front Cardiovasc Med 2023; 10:1250234. [PMID: 37655218 PMCID: PMC10466789 DOI: 10.3389/fcvm.2023.1250234] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Accepted: 08/03/2023] [Indexed: 09/02/2023] Open
Abstract
Background and objective Whole body manipulation of the renin-angiotensin system (RAS) consistently exerts profound effects on experimental atherosclerosis development. A deficit in the literature has been a lack of attention to the effects of sex. Also, based on data with gene-deleted mice, the site of RAS activity that influences lesion formation is at an unknown distant location. Since angiotensin (AngII) concentrations are high in kidney and the major components of the RAS are present in renal proximal tubule cells (PTCs), this study evaluated the role of the RAS in PTCs in atherosclerosis development. Methods and results Mice with an LDL receptor -/- background were fed Western diet to induce hypercholesterolemia and atherosclerosis. We first demonstrated the role of AT1 receptor antagonism on atherosclerosis in both sexes. Losartan, an AngII type 1 (AT1) receptor blocker, had greater blood pressure-lowering effects in females than males, but equivalent effects between sexes in reducing atherosclerotic lesion size. To determine the roles of renal AT1a receptor and angiotensin-converting enzyme (ACE), either component was deleted in PTCs after weaning using a tamoxifen-inducible Cre expressed under the control of an Ndrg1 promoter. Despite profound deletion of AT1a receptor or ACE in PTCs, the absence of either protein did not influence development of atherosclerosis in either sex. Conversely, mice expressing human angiotensinogen and renin in PTCs or expressing human angiotensinogen in liver but human renin in PTCs did not change atherosclerotic lesion size in male mice. Conclusion Whole-body AT1R inhibition reduced atherosclerosis equivalently in both male and female mice; however, PTC-specific manipulation of the RAS components had no effects on hypercholesterolemia-induced atherosclerosis.
Collapse
Affiliation(s)
- Masayoshi Kukida
- Saha Cardiovascular Research Center, University of Kentucky, Lexington, KY, United States
| | - Naofumi Amioka
- Saha Cardiovascular Research Center, University of Kentucky, Lexington, KY, United States
| | - Dien Ye
- Saha Cardiovascular Research Center, University of Kentucky, Lexington, KY, United States
| | - Hui Chen
- Saha Cardiovascular Research Center, University of Kentucky, Lexington, KY, United States
| | - Jessica J. Moorleghen
- Saha Cardiovascular Research Center, University of Kentucky, Lexington, KY, United States
| | - Ching-Ling Liang
- Saha Cardiovascular Research Center, University of Kentucky, Lexington, KY, United States
| | - Deborah A. Howatt
- Saha Cardiovascular Research Center, University of Kentucky, Lexington, KY, United States
| | - Yuriko Katsumata
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, United States
- Department of Biostatistics, University of Kentucky, Lexington, KY, United States
| | - Motoko Yanagita
- Department of Nephrology, Kyoto University Graduate School of Medicine, Kyoto, Japan
- Institute for the Advanced Study of Human Biology (WPI-ASHBi), Kyoto University, Kyoto, Japan
| | - Hisashi Sawada
- Saha Cardiovascular Research Center, University of Kentucky, Lexington, KY, United States
- Saha Aortic Center, University of Kentucky, Lexington, KY, United States
- Department of Physiology, University of Kentucky, Lexington, KY, United States
| | - Alan Daugherty
- Saha Cardiovascular Research Center, University of Kentucky, Lexington, KY, United States
- Saha Aortic Center, University of Kentucky, Lexington, KY, United States
- Department of Physiology, University of Kentucky, Lexington, KY, United States
| | - Hong S. Lu
- Saha Cardiovascular Research Center, University of Kentucky, Lexington, KY, United States
- Saha Aortic Center, University of Kentucky, Lexington, KY, United States
- Department of Physiology, University of Kentucky, Lexington, KY, United States
| |
Collapse
|
25
|
Shoemaker R, Poglitsch M, Huang H, Vignes K, Srinivasan A, Cockerham C, Schadler A, Bauer JA, O’Brien JM. Activation of the Renin-Angiotensin-Aldosterone System Is Attenuated in Hypertensive Compared with Normotensive Pregnancy. Int J Mol Sci 2023; 24:12728. [PMID: 37628909 PMCID: PMC10454898 DOI: 10.3390/ijms241612728] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 08/04/2023] [Accepted: 08/07/2023] [Indexed: 08/27/2023] Open
Abstract
Hypertension during pregnancy increases the risk of adverse maternal and fetal outcomes, but the mechanisms of pregnancy hypertension are not precisely understood. Elevated plasma renin activity and aldosterone concentrations play an important role in the normal physiologic adaptation to pregnancy. These effectors are reduced in patients with pregnancy hypertension, creating an opportunity to define the features of the renin-angiotensin-aldosterone system (RAAS) that are characteristic of this disorder. In the current study, we used a novel LC-MS/MS-based methodology to develop comprehensive profiles of RAAS peptides and effectors over gestation in a cohort of 74 pregnant women followed prospectively for the development of gestational hypertension and pre-eclampsia (HYP, 27 patients) versus those remaining normotensive (NT, 47 patients). In NT pregnancy, the plasma renin activity surrogate, (PRA-S, calculated from the sum of Angiotensin I + Angiotensin II) and aldosterone concentrations significantly increased from the first to the third trimester, accompanied by a modest increase in the concentrations of angiotensin peptide metabolites. In contrast, in HYP pregnancies, PRA-S and angiotensin peptides were largely unchanged over gestation, and third-trimester aldosterone concentrations were significantly lower compared with those in NT pregnancies. The results indicated that the predominant features of pregnancies that develop HYP are stalled or waning activation of the RAAS in the second half of pregnancy (accompanied by unchanging levels of angiotensin peptides) and the attenuated secretion of aldosterone.
Collapse
Affiliation(s)
- Robin Shoemaker
- Department of Dietetics and Human Nutrition, University of Kentucky, Lexington, KY 40506, USA
| | | | - Hong Huang
- Department of Pediatrics, University of Kentucky, Lexington, KY 40536, USA
| | - Katherine Vignes
- Division of Maternal Fetal Medicine, Department of Obstetrics and Gynecology, University of Kentucky, Lexington, KY 40506, USA
| | - Aarthi Srinivasan
- Division of Maternal Fetal Medicine, Department of Obstetrics and Gynecology, University of Kentucky, Lexington, KY 40506, USA
| | - Cynthia Cockerham
- Division of Maternal Fetal Medicine, Department of Obstetrics and Gynecology, University of Kentucky, Lexington, KY 40506, USA
| | - Aric Schadler
- Department of Pediatrics, University of Kentucky, Lexington, KY 40536, USA
| | - John A. Bauer
- Department of Pediatrics, University of Kentucky, Lexington, KY 40536, USA
| | - John M. O’Brien
- Division of Maternal Fetal Medicine, Department of Obstetrics and Gynecology, University of Kentucky, Lexington, KY 40506, USA
| |
Collapse
|
26
|
Amioka N, Wu CH, Sawada H, Ito S, Pettey AC, Wu C, Moorleghen JJ, Howatt DA, Graf GA, Vander Kooi CW, Daugherty A, Lu HS. Functional Exploration of Conserved Sequences in the Distal Face of Angiotensinogen-Brief Report. Arterioscler Thromb Vasc Biol 2023; 43:1524-1532. [PMID: 37345525 PMCID: PMC10527926 DOI: 10.1161/atvbaha.122.318930] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Accepted: 06/12/2023] [Indexed: 06/23/2023]
Abstract
BACKGROUND Angiotensinogen (AGT) is an essential component in the renin-angiotensin system. AGT has highly conserved sequences in the loop and β-sheet regions among species; however, their functions have not been studied. METHODS Adeno-associated viral vector (AAV) serotype 2/8 encoding mouse AGT with mutations of conserved sequences in the loop (AAV.loop-Mut), β-sheet (AAV.βsheet-Mut), or both regions (AAV.loop/βsheet-Mut) was injected into male hepatocyte-specific AGT-deficient (hepAGT-/-) mice in an LDL (low-density lipoprotein) receptor-deficient background. AAV containing mouse wild-type AGT (AAV.mAGT) or a null vector (AAV.null) were used as controls. Two weeks after AAV administration, all mice were fed a western diet for 12 weeks. To determine how AGT secretion is regulated in hepatocytes, AAVs containing the above mutations were transducted into HepG2 cells. RESULTS In hepAGT-/- mice infected with AAV.loop-Mut or βsheet-Mut, plasma AGT concentrations, systolic blood pressure, and atherosclerosis were comparable to those in AAV.mAGT-infected mice. Interestingly, plasma AGT concentrations, systolic blood pressure, and atherosclerotic lesion size in hepAGT-/- mice infected with AAV.loop/βsheet-Mut were not different from mice infected with AAV.null. In contrast, hepatic Agt mRNA abundance was elevated to a comparable magnitude as AAV.mAGT-infected mice. Immunostaining showed that AGT protein was accumulated in hepatocytes of mice infected with AAV.loop/βsheet-Mut or HepG2 cells transducted with AAV.loop/βsheet-Mut. Accumulated AGT was not located in the endoplasmic reticulum. CONCLUSIONS The conserved sequences in either the loop or β-sheet region individually have no effect on AGT regulation, but the conserved sequences in both regions synergistically contribute to the secretion of AGT from hepatocytes.
Collapse
Affiliation(s)
- Naofumi Amioka
- Saha Cardiovascular Research Center, University of Kentucky, Lexington, KY
- Saha Aortic Center, University of Kentucky, Lexington, KY
| | - Chia-Hua Wu
- Saha Cardiovascular Research Center, University of Kentucky, Lexington, KY
- Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY
| | - Hisashi Sawada
- Saha Cardiovascular Research Center, University of Kentucky, Lexington, KY
- Saha Aortic Center, University of Kentucky, Lexington, KY
- Department of Physiology, University of Kentucky, Lexington, KY
| | - Sohei Ito
- Saha Cardiovascular Research Center, University of Kentucky, Lexington, KY
- Saha Aortic Center, University of Kentucky, Lexington, KY
| | - Alex C. Pettey
- Saha Cardiovascular Research Center, University of Kentucky, Lexington, KY
- Saha Aortic Center, University of Kentucky, Lexington, KY
- Department of Physiology, University of Kentucky, Lexington, KY
| | - Congqing Wu
- Saha Cardiovascular Research Center, University of Kentucky, Lexington, KY
- Saha Aortic Center, University of Kentucky, Lexington, KY
- Department of Surgery, University of Kentucky, Lexington, KY
- Department of Microbiology, Immunology, and Molecular Genetics University of Kentucky, Lexington, KY
| | - Jessica J. Moorleghen
- Saha Cardiovascular Research Center, University of Kentucky, Lexington, KY
- Saha Aortic Center, University of Kentucky, Lexington, KY
| | - Deborah A. Howatt
- Saha Cardiovascular Research Center, University of Kentucky, Lexington, KY
- Saha Aortic Center, University of Kentucky, Lexington, KY
| | - Gregory A. Graf
- Saha Cardiovascular Research Center, University of Kentucky, Lexington, KY
- Department of Physiology, University of Kentucky, Lexington, KY
| | - Craig W. Vander Kooi
- Department of Molecular and Cellular Biochemistry University of Kentucky, Lexington, KY
| | - Alan Daugherty
- Saha Cardiovascular Research Center, University of Kentucky, Lexington, KY
- Saha Aortic Center, University of Kentucky, Lexington, KY
- Department of Physiology, University of Kentucky, Lexington, KY
| | - Hong S. Lu
- Saha Cardiovascular Research Center, University of Kentucky, Lexington, KY
- Saha Aortic Center, University of Kentucky, Lexington, KY
- Department of Physiology, University of Kentucky, Lexington, KY
| |
Collapse
|
27
|
Ji N, Li Y, Wei J, Huang L, Lin WH, Li G. The Changes of Cardiovascular Neurotransmitter Levels under Low-Intensity Focused Ultrasound Stimulation of the Vagus Nerve. ANNUAL INTERNATIONAL CONFERENCE OF THE IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. ANNUAL INTERNATIONAL CONFERENCE 2023; 2023:1-4. [PMID: 38083122 DOI: 10.1109/embc40787.2023.10340334] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2023]
Abstract
BACKGROUND Our previous study has shown that stimulation of the vagus nerve with low-intensity focused ultrasound could modulate blood pressure (BP), but the underlying mechanisms remain unclear. This study investigated the changes of cardiovascular neurotransmitter levels to indirectly evaluate the responses of the autonomic nervous system and renin-angiotensin system under low-intensity focused ultrasound stimulation (FUS) of the vagus nerve. METHODS Cardiovascular neurotransmitter levels of epinephrine (EPI), norepinephrine (NE), and angiotensin II (ANGII) were measured and compared before and after the FUS in seven spontaneously hypertensive rats; and were also measured and compared between a target stimulation group (FUS, n = 6) and non-target stimulation group (Control, n = 5) after stimulation to exclude the influence of potential confounding factors. RESULTS The t-test results showed that the levels of EPI, NE, and ANGII were significantly decreased (P < 0.05) after stimulation compared to before stimulation. Additionally, the levels of NE and EPI were significantly lower (P < 0.05) in the FUS group than in the Control group after stimulation, indicating that the activities of the sympathetic nervous system and renin-angiotensin system of the vagus nerve might be inhibited by FUS of the vagus nerve. CONCLUSION These findings reveal the mechanism of BP lowing in response to FUS of the vagus nerve.Clinical Relevance-This study revealed the mechanism of BP lowering in response to focused ultrasound stimulation of the vagus nerve through analyzing the changes of cardiovascular neurotransmitter levels.
Collapse
|
28
|
Liu Y, Liu S, Zhao J, Wu K, Xu B, Wang W. Increased plasma renin by vasodilators promotes the progression of abdominal aortic aneurysm. Front Pharmacol 2023; 14:1174278. [PMID: 37383707 PMCID: PMC10299739 DOI: 10.3389/fphar.2023.1174278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2023] [Accepted: 05/22/2023] [Indexed: 06/30/2023] Open
Abstract
Background: It is well-accepted that antihypertensive therapy is the cornerstone of treatment for abdominal aortic aneurysm (AAA) patients with hypertension. Direct-acting vasodilators were used in the treatment of hypertension by directly relaxing vascular smooth muscle but may have destructive effects on the aortic wall by activating the renin-angiotensin system axis. Their roles in AAA disease remain to be elucidated. In this study, we used hydralazine and minoxidil, two classical direct-acting vasodilators, to investigate their influence and potential mechanisms on AAA disease. Methods and results: In this study, we investigated the plasma renin level and plasma renin activity in AAA patients. Simultaneously, age and gender ratio-matched patients diagnosed with peripheral artery disease and varicose veins were selected as the control group using a ratio of 1:1:1. Our regression analysis suggested both the plasma renin level and plasma renin activity are positively associated with AAA development. In view of the well-established relationship between direct-acting vasodilators and increased plasma renin concentration, we established a porcine pancreatic elastase-infused AAA mouse model, followed by oral administration of hydralazine (250 mg/L) and minoxidil (120 mg/L) to investigate effects of direct-acting vasodilators on AAA disease. Our results suggested both hydralazine and minoxidil promoted the progression of AAA with increased aortic degeneration. Mechanistically, the vasodilators aggravated aortic inflammation by increased leukocyte infiltration and inflammatory cytokine secretion. Conclusion and relevance: The plasma renin level and plasma renin activity are positively associated with AAA development. Direct vasodilators aggravated experimental AAA progression, which raised cautionary concerns about their applications in AAA disease.
Collapse
Affiliation(s)
- Yu Liu
- Department of General and Vascular Surgery, Xiangya Hospital, Central South University, Changsha, China
| | - Shuai Liu
- Department of General and Vascular Surgery, Xiangya Hospital, Central South University, Changsha, China
| | - Jiani Zhao
- Department of General and Vascular Surgery, Xiangya Hospital, Central South University, Changsha, China
| | - Kemin Wu
- Department of General and Vascular Surgery, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Baohui Xu
- Department of Surgery, Stanford University School of Medicine, Stanford, CA, United States
| | - Wei Wang
- Department of General and Vascular Surgery, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
29
|
Kanugula AK, Kaur J, Batra J, Ankireddypalli AR, Velagapudi R. Renin-Angiotensin System: Updated Understanding and Role in Physiological and Pathophysiological States. Cureus 2023; 15:e40725. [PMID: 37350982 PMCID: PMC10283427 DOI: 10.7759/cureus.40725] [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] [Accepted: 06/20/2023] [Indexed: 06/24/2023] Open
Abstract
The classical view of the renin-angiotensin system (RAS) is that of the circulating hormone pathway involved in salt and water homeostasis and blood pressure regulation. It is also involved in the pathogenesis of cardiac and renal disorders. This led to the creation of drugs blocking the actions of this classical pathway, which improved cardiac and renal outcomes. Our understanding of the RAS has significantly expanded with the discovery of new peptides involved in this complex pathway. Over the last two decades, a counter-regulatory or protective pathway has been discovered that opposes the effects of the classical pathway. Components of RAS are also implicated in the pathogenesis of obesity and its metabolic diseases. The continued discovery of newer molecules also provides novel therapeutic targets to improve disease outcomes. This article aims to provide an overview of an updated understanding of the RAS, its role in physiological and pathological processes, and potential novel therapeutic options from RAS for managing cardiorenal disorders, obesity, and related metabolic disorders.
Collapse
Affiliation(s)
- Ashok Kumar Kanugula
- Department of Internal Medicine, Wellstar Health System - Spalding Regional Hospital, Griffin, USA
| | - Jasleen Kaur
- Department of Endocrinology, Diabetes, and Metabolism, HealthPartners, Minneapolis, USA
| | - Jaskaran Batra
- Department of Internal Medicine, Univerity of Pittsburg Medical Center (UPMC) McKeesport, McKeesport, USA
| | | | - Ravikanth Velagapudi
- Department of Pulmonary and Critical Care Medicine, Spectrum Health/Michigan State University, Grand Rapids, USA
| |
Collapse
|
30
|
Swiderski J, Gadanec LK, Apostolopoulos V, Moore GJ, Kelaidonis K, Matsoukas JM, Zulli A. Role of Angiotensin II in Cardiovascular Diseases: Introducing Bisartans as a Novel Therapy for Coronavirus 2019. Biomolecules 2023; 13:787. [PMID: 37238657 PMCID: PMC10216788 DOI: 10.3390/biom13050787] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Revised: 04/26/2023] [Accepted: 04/27/2023] [Indexed: 05/28/2023] Open
Abstract
Cardiovascular diseases (CVDs) are the main contributors to global morbidity and mortality. Major pathogenic phenotypes of CVDs include the development of endothelial dysfunction, oxidative stress, and hyper-inflammatory responses. These phenotypes have been found to overlap with the pathophysiological complications of coronavirus disease 2019 (COVID-19). CVDs have been identified as major risk factors for severe and fatal COVID-19 states. The renin-angiotensin system (RAS) is an important regulatory system in cardiovascular homeostasis. However, its dysregulation is observed in CVDs, where upregulation of angiotensin type 1 receptor (AT1R) signaling via angiotensin II (AngII) leads to the AngII-dependent pathogenic development of CVDs. Additionally, the interaction between the spike protein of severe acute respiratory syndrome coronavirus 2 with angiotensin-converting enzyme 2 leads to the downregulation of the latter, resulting in the dysregulation of the RAS. This dysregulation favors AngII/AT1R toxic signaling pathways, providing a mechanical link between cardiovascular pathology and COVID-19. Therefore, inhibiting AngII/AT1R signaling through angiotensin receptor blockers (ARBs) has been indicated as a promising therapeutic approach to the treatment of COVID-19. Herein, we review the role of AngII in CVDs and its upregulation in COVID-19. We also provide a future direction for the potential implication of a novel class of ARBs called bisartans, which are speculated to contain multifunctional targeting towards COVID-19.
Collapse
Affiliation(s)
- Jordan Swiderski
- Institute for Health and Sport, Victoria University, Melbourne, VIC 3030, Australia; (J.S.); (L.K.G.); (V.A.)
| | - Laura Kate Gadanec
- Institute for Health and Sport, Victoria University, Melbourne, VIC 3030, Australia; (J.S.); (L.K.G.); (V.A.)
| | - Vasso Apostolopoulos
- Institute for Health and Sport, Victoria University, Melbourne, VIC 3030, Australia; (J.S.); (L.K.G.); (V.A.)
- Immunology Program, Australian Institute for Musculoskeletal Science, Melbourne, VIC 3021, Australia
| | - Graham J. Moore
- Pepmetics Incorporated, 772 Murphy Place, Victoria, BC V8Y 3H4, Canada;
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada
| | | | - John M. Matsoukas
- Institute for Health and Sport, Victoria University, Melbourne, VIC 3030, Australia; (J.S.); (L.K.G.); (V.A.)
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada
- NewDrug PC, Patras Science Park, 26500 Patras, Greece;
- Department of Chemistry, University of Patras, 26504 Patras, Greece
| | - Anthony Zulli
- Institute for Health and Sport, Victoria University, Melbourne, VIC 3030, Australia; (J.S.); (L.K.G.); (V.A.)
| |
Collapse
|
31
|
Yu J, Tang Y, Wang Y, Zhou M, Li Y, Hong J, Li C, Xu B, Guo X, Mao J. Serum exosomes derived from spontaneously hypertensive rats induce cardiac hypertrophy in vitro and in vivo by increasing autocrine release of angiotensin II in cardiomyocytes. Biochem Pharmacol 2023; 210:115462. [PMID: 36849061 DOI: 10.1016/j.bcp.2023.115462] [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: 11/27/2022] [Revised: 02/17/2023] [Accepted: 02/21/2023] [Indexed: 02/27/2023]
Abstract
Identifying the key factors mediating the progression from hypertension to cardiac hypertrophy is critically important for developing a strategy to protect against heart failure. Serum exosomes have been revealed to be involved in the development of cardiovascular disease. In the current study, we found that either serum or serum exosomes derived from SHR induced hypertrophy in H9c2 cardiomyocytes. SHR Exo injection through the tail vein for 8 weeks induced left ventricular wall thickening and decreased cardiac function in C57BL/6 mice. SHR Exo carried the renin-angiotensin system (RAS) proteins AGT, renin, and ACE into cardiomyocytes, which increased the autocrine secretion of Ang II. Moreover, the AT1-type receptor antagonist telmisartan prevented hypertrophy of H9c2 cells induced by SHR Exo.These results identified a novel role of exosomes derived from SHR serum in cardiac hypertrophy and revealed that SHR Exo induced cardiac hypertrophy by carrying AGT, renin, and ACE proteins into cardiomyocytes to increase their autocrine secretion of Ang II. The emergence of this new mechanism will help us better understand how hypertension progresses to cardiac hypertrophy.
Collapse
Affiliation(s)
- Jingwei Yu
- Guangdong Provincial Key Laboratory of Pharmaceutical Bioactive Substances and School of Life Sciences and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou 510006, China; These authors contributed equally to this work
| | - Yuan Tang
- Guangdong Provincial Key Laboratory of Pharmaceutical Bioactive Substances and School of Life Sciences and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou 510006, China; These authors contributed equally to this work
| | - Yu Wang
- Guangdong Provincial Key Laboratory of Pharmaceutical Bioactive Substances and School of Life Sciences and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou 510006, China; These authors contributed equally to this work
| | - Mi Zhou
- Guangdong Provincial Key Laboratory of Pharmaceutical Bioactive Substances and School of Life Sciences and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou 510006, China; These authors contributed equally to this work
| | - Yanwen Li
- Guangdong Provincial Key Laboratory of Pharmaceutical Bioactive Substances and School of Life Sciences and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Jiahuan Hong
- Guangdong Provincial Key Laboratory of Pharmaceutical Bioactive Substances and School of Life Sciences and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Chunmei Li
- Guangdong Provincial Key Laboratory of Pharmaceutical Bioactive Substances and School of Life Sciences and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Bin Xu
- Guangdong Provincial Key Laboratory of Pharmaceutical Bioactive Substances and School of Life Sciences and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Xinmin Guo
- Department of Ultrasound, Guangzhou Red Cross Hospital, Medical College, Jinan University, Guangzhou, Guangdong 510220, China.
| | - Jianwen Mao
- Guangdong Provincial Key Laboratory of Pharmaceutical Bioactive Substances and School of Life Sciences and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou 510006, China.
| |
Collapse
|
32
|
Katayama IA, Huang Y, Garza AE, Brooks DL, Williams JS, Nascimento MM, Heimann JC, Pojoga LH. Longitudinal changes in blood pressure are preceded by changes in albuminuria and accelerated by increasing dietary sodium intake. Exp Gerontol 2023; 173:112114. [PMID: 36738979 PMCID: PMC10965150 DOI: 10.1016/j.exger.2023.112114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Revised: 01/31/2023] [Accepted: 02/01/2023] [Indexed: 02/06/2023]
Abstract
BACKGROUND Dietary sodium is a well-known risk factor for cardiovascular and renal disease; however, direct evidence of the longitudinal changes that occur with aging, and the influence of dietary sodium on the age-associated alterations are scarce. METHODS C57BL/6 mice were maintained for 13 months on a low (LS, 0.02 % Na+), normal (NS, 0.3 % Na+) or high (HS, 1.6 % Na+) salt diet. We assessed 1) the longitudinal trajectories for two markers of cardiovascular and renal dysfunction (blood pressure (BP) and albuminuria), as well as hormonal changes, and 2) end-of-study cardiac and renal parameters. RESULTS The effect of aging on BP and kidney damage did not reach significance levels in the LS group; however, relative to baseline, there were significant increases in these parameters for animals maintained on NS and HS diets, starting as early as month 7 and month 5, respectively. Furthermore, changes in albuminuria preceded the changes in BP relative to baseline, irrespective of the diet. Circulating aldosterone and plasma renin activity displayed the expected decreasing trends with age and dietary sodium loading. As compared to LS - higher dietary sodium consumption associated with increasing trends in left ventricular mass and volume indices, consistent with an eccentric dilated phenotype. Functional and molecular markers of kidney dysfunction displayed similar trends with increasing long-term sodium levels: higher renovascular resistance, increased glomerular volumes, as well as higher levels of renal angiotensin II type 1 and mineralocorticoid receptors, and lower renal Klotho levels. CONCLUSION Our study provides a timeline for the development of cardiorenal dysfunction with aging, and documents that increasing dietary salt accelerates the age-induced phenotypes. In addition, we propose albuminuria as a prognostic biomarker for the future development of hypertension. Last, we identified functional and molecular markers of renal dysfunction that associate with long-term dietary salt loading.
Collapse
Affiliation(s)
- Isis Akemi Katayama
- Division of Endocrinology, Diabetes and Hypertension, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA; Laboratory of Experimental Hypertension, Department of Internal Medicine, University of São Paulo School of Medicine, São Paulo, Brazil
| | - Yuefei Huang
- Division of Endocrinology, Diabetes and Hypertension, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Amanda E Garza
- Division of Endocrinology, Diabetes and Hypertension, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Danielle L Brooks
- Division of Endocrinology, Diabetes and Hypertension, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Jonathan S Williams
- Division of Endocrinology, Diabetes and Hypertension, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Mariana M Nascimento
- Laboratory of Experimental Hypertension, Department of Internal Medicine, University of São Paulo School of Medicine, São Paulo, Brazil
| | - Joel C Heimann
- Laboratory of Experimental Hypertension, Department of Internal Medicine, University of São Paulo School of Medicine, São Paulo, Brazil
| | - Luminita H Pojoga
- Division of Endocrinology, Diabetes and Hypertension, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
33
|
Okuno K, Torimoto K, Cicalese SM, Preston K, Rizzo V, Hashimoto T, Coffman TM, Sparks MA, Eguchi S. Angiotensin II Type 1A Receptor Expressed in Smooth Muscle Cells is Required for Hypertensive Vascular Remodeling in Mice Infused With Angiotensin II. Hypertension 2023; 80:668-677. [PMID: 36628961 PMCID: PMC9931681 DOI: 10.1161/hypertensionaha.122.20601] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Accepted: 12/29/2022] [Indexed: 01/12/2023]
Abstract
BACKGROUND Ang II (angiotensin II) type 1 (AT1) receptors play a critical role in cardiovascular diseases such as hypertension. Rodents have 2 types of AT1 receptor (AT1A and AT1B) of which knock-in Tagln-mediated smooth muscle AT1A silencing attenuated Ang II-induced hypertension. Although vascular remodeling, a significant contributor to organ damage, occurs concurrently with hypertension in Ang II-infused mice, the contribution of smooth muscle AT1A in this process remains unexplored. Accordingly, it is hypothesized that smooth muscle AT1A receptors exclusively contribute to both medial thickening and adventitial fibrosis regardless of the presence of hypertension. METHODS About 1 µg/kg per minute Ang II was infused for 2 weeks in 2 distinct AT1A receptor silenced mice, knock-in Tagln-mediated constitutive smooth muscle AT1A receptor silenced mice, and Myh11-mediated inducible smooth muscle AT1A together with global AT1B silenced mice for evaluation of hypertensive cardiovascular remodeling. RESULTS Medial thickness, adventitial collagen deposition, and immune cell infiltration in aorta were increased in control mice but not in both smooth muscle AT1A receptor silenced mice. Coronary arterial perivascular fibrosis in response to Ang II infusion was also attenuated in both AT1A receptor silenced mice. Ang II-induced cardiac hypertrophy was attenuated in constitutive smooth muscle AT1A receptor silenced mice. However, Ang II-induced cardiac hypertrophy and hypertension were not altered in inducible smooth muscle AT1A receptor silenced mice. CONCLUSIONS Smooth muscle AT1A receptors mediate Ang II-induced vascular remodeling including medial hypertrophy and inflammatory perivascular fibrosis regardless of the presence of hypertension. Our data suggest an independent etiology of blood pressure elevation and hypertensive vascular remodeling in response to Ang II.
Collapse
Affiliation(s)
- Keisuke Okuno
- Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University, Philadelphia, PA (K.O., K.T., S.M.C., K.P., V.R., S.E.)
| | - Keiichi Torimoto
- Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University, Philadelphia, PA (K.O., K.T., S.M.C., K.P., V.R., S.E.)
| | - Stephanie M Cicalese
- Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University, Philadelphia, PA (K.O., K.T., S.M.C., K.P., V.R., S.E.)
| | - Kyle Preston
- Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University, Philadelphia, PA (K.O., K.T., S.M.C., K.P., V.R., S.E.)
| | - Victor Rizzo
- Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University, Philadelphia, PA (K.O., K.T., S.M.C., K.P., V.R., S.E.)
| | - Tomoki Hashimoto
- Barrow Aneurysm and AVM Research Center, Departments of Neurosurgery and Neurobiology, Barrow Neurological Institute, Phoenix, AZ (T.H.)
| | - Thomas M Coffman
- Division of Nephrology, Department of Medicine, Duke University Medical Center, Durham, NC (T.M.C., M.A.S.)
- Program in Cardiovascular and Metabolic Disorders, Duke-NUS Medical School, Singapore (T.M.C.)
| | - Matthew A Sparks
- Division of Nephrology, Department of Medicine, Duke University Medical Center, Durham, NC (T.M.C., M.A.S.)
| | - Satoru Eguchi
- Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University, Philadelphia, PA (K.O., K.T., S.M.C., K.P., V.R., S.E.)
| |
Collapse
|
34
|
He D, Peng X, Xie H, Peng R, Li Q, Guo Y, Wang W, He H, Chen Y. Genetic Variations in Angiotensinogen Gene and Risk of Preeclampsia: A Pilot Study. J Clin Med 2023; 12:jcm12041509. [PMID: 36836041 PMCID: PMC9966751 DOI: 10.3390/jcm12041509] [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: 12/18/2022] [Revised: 01/27/2023] [Accepted: 01/30/2023] [Indexed: 02/17/2023] Open
Abstract
Preeclampsia (PE) is a typical hypertensive disorders of pregnancy (HDP) which can cause substantial morbidity and mortality in both pregnant women and fetuses. The renin-angiotensin system (RAS) genes are the main HDP-causing genes, and Angiotensinogen (AGT) as the initial substrate can directly reflect the activity of the entire RAS. However, the association between AGT SNPs and PE risk has rarely been confirmed. This study was carried out to determine whether AGT SNPs could affect the risk of PE in 228 cases and 358 controls. The genotyping result revealed that the AGT rs7079 TT carrier was related to increased PE risk. Further stratified analysis illustrated that the rs7079 TT genotype significantly increased the PE risk in subgroups of Age < 35, BMI < 25, Albumin (ALB) ≥ 30 and Aspartate aminotransferase (AST) < 30. These findings demonstrated that the rs7079 might be a promising candidate SNP strongly associated with PE susceptibility.
Collapse
Affiliation(s)
- Dong He
- Guangdong Provincial Key Laboratory of Pathogenesis of Heart and Spleen and Prescription Drugs Research, Department of Pharmacology, School of Chinese Pharmaceutical Science, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Xianglan Peng
- Department of Obstetrics and Gynecology, Department of Gynecologic Oncology Research Office, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou 510150, China
| | - Hongkai Xie
- Guangdong Provincial Key Laboratory of Pathogenesis of Heart and Spleen and Prescription Drugs Research, Department of Pharmacology, School of Chinese Pharmaceutical Science, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Rui Peng
- Department of Obstetrics and Gynecology, Department of Gynecologic Oncology Research Office, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou 510150, China
| | - Qixuan Li
- Department of Obstetrics and Gynecology, Department of Gynecologic Oncology Research Office, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou 510150, China
| | - Yitong Guo
- Guangdong Provincial Key Laboratory of Pathogenesis of Heart and Spleen and Prescription Drugs Research, Department of Pharmacology, School of Chinese Pharmaceutical Science, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Wei Wang
- Guangdong Provincial Key Laboratory of Pathogenesis of Heart and Spleen and Prescription Drugs Research, Department of Pharmacology, School of Chinese Pharmaceutical Science, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Hong He
- Department of Obstetrics and Gynecology, Department of Gynecologic Oncology Research Office, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou 510150, China
- Correspondence: (H.H.); (Y.C.)
| | - Yang Chen
- Guangdong Provincial Key Laboratory of Pathogenesis of Heart and Spleen and Prescription Drugs Research, Department of Pharmacology, School of Chinese Pharmaceutical Science, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
- Correspondence: (H.H.); (Y.C.)
| |
Collapse
|
35
|
Dabidi Roshan V, Ahmadian M, Nasiri K, Akbari A, Ghasemi M, Nasrollahi Borujeni N, Zahedmanesh F, Nabavi Chashmi SM, Imani F. Exercise-induced expression of SARS-CoV-2 entry receptors: impact of mask modality, sex, and exercise intensity. J Sports Med Phys Fitness 2023; 63:319-328. [PMID: 35686871 DOI: 10.23736/s0022-4707.22.14093-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
BACKGROUND Wearing a facemask affects physiological responses to exercise. We explored how exercising with a facemask affects the expression of SARS-CoV-2 entry receptor (angiotensin-converting enzyme 2 [ACE2]) and some associated genes (angiotensin type-1 receptors [AT<inf>1</inf>R]; Mas receptor [MasR]; hypoxia-inducible factor 1α [HIF-1α]; endothelial nitric oxide synthase [eNOS]) among healthy males and females. METHODS One hundred forty-four apparently healthy individuals (72 females; age: 30±6) were allocated to three mask groups of 48 (N95, Surgical, No Mask) with two exercise subgroups for each mask for both sexes. Participants in each experimental group performed either a submaximal (walking with no grade) or maximal (a modified Bruce Protocol) treadmill exercise test. Blood samples were collected before and after each exercise test and used to analyze the mRNA expression of the genes studied. RESULTS The post-exercise expression of genes examined were comparable between Surgical, N95, and No Mask (P>0.05). ACE2 was significantly greater in Surgical and N95 against No Mask at baseline and following moderate-intensity exercise (P<0.05). Whilst similar expressions were noted for MasR and eNOS (P>0.05), AT<inf>1</inf>R was greater in N95 than Surgical following high-intensity exercise (P<0.05). HIF-1α following either exercise intensity was significantly lower in N95 than Surgical (P<0.05). AT<inf>1</inf>R and HIF-1α were similar between Surgical and N95 against No Mask (P>0.05). ACE2 and AT<inf>1</inf>R were significantly higher in either mask modality than No Mask in males at baseline and postexercise (P<0.05). HIF-1α, MasR, and eNOS expressions were comparable between all mask groups in either sex (P<0.05). CONCLUSIONS Our findings suggest that wearing a facemask does not differentiate the gene expression of SARS-CoV-2 entry receptor following exercise among both sexes.
Collapse
Affiliation(s)
- Valiollah Dabidi Roshan
- Department of Exercise Physiology, Faculty of Sport Science, University of Mazandaran, Babolsar, Iran - .,Athletic Performance and Health Research Center, Faculty of Sport Science, University of Mazandaran, Babolsar, Iran -
| | - Mehdi Ahmadian
- School of Kinesiology, University of British Columbia, Vancouver, BC, Canada
| | - Khadijeh Nasiri
- Department of Exercise Physiology, Faculty of Sport Science, University of Mazandaran, Babolsar, Iran
| | - Abolfazl Akbari
- School of Veterinary Medicine, Department of Physiology, Shiraz University, Shiraz, Iran
| | - Mohammad Ghasemi
- Department of Exercise Physiology, Faculty of Sport Science, University of Mazandaran, Babolsar, Iran
| | | | - Foruzan Zahedmanesh
- Department of Exercise Physiology, Faculty of Sport Science, University of Mazandaran, Babolsar, Iran
| | - Seyedeh M Nabavi Chashmi
- Department of Exercise Physiology, Faculty of Sport Science, University of Mazandaran, Babolsar, Iran
| | - Fattaneh Imani
- Department of Exercise Physiology, Faculty of Sport Science, University of Mazandaran, Babolsar, Iran
| |
Collapse
|
36
|
Ghalayini J, Boulianne GL. Deciphering mechanisms of action of ACE inhibitors in neurodegeneration using Drosophila models of Alzheimer's disease. Front Neurosci 2023; 17:1166973. [PMID: 37113150 PMCID: PMC10126366 DOI: 10.3389/fnins.2023.1166973] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Accepted: 03/17/2023] [Indexed: 04/29/2023] Open
Abstract
Alzheimer's disease (AD) is a devastating neurodegenerative disorder for which there is no cure. Recently, several studies have reported a significant reduction in the incidence and progression of dementia among some patients receiving antihypertensive medications such as angiotensin-converting enzyme inhibitors (ACE-Is) and angiotensin receptor blockers (ARBs). Why these drugs are beneficial in some AD patients and not others is unclear although it has been shown to be independent of their role in regulating blood pressure. Given the enormous and immediate potential of ACE-Is and ARBs for AD therapeutics it is imperative that we understand how they function. Recently, studies have shown that ACE-Is and ARBs, which target the renin angiotensin system in mammals, are also effective in suppressing neuronal cell death and memory defects in Drosophila models of AD despite the fact that this pathway is not conserved in flies. This suggests that the beneficial effects of these drugs may be mediated by distinct and as yet, identified mechanisms. Here, we discuss how the short lifespan and ease of genetic manipulations available in Drosophila provide us with a unique and unparalleled opportunity to rapidly identify the targets of ACE-Is and ARBs and evaluate their therapeutic effectiveness in robust models of AD.
Collapse
Affiliation(s)
- Judy Ghalayini
- Program in Developmental and Stem Cell Biology, Peter Gilgin Center for Research and Learning, The Hospital for Sick Children, Toronto, ON, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
| | - Gabrielle L. Boulianne
- Program in Developmental and Stem Cell Biology, Peter Gilgin Center for Research and Learning, The Hospital for Sick Children, Toronto, ON, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
- *Correspondence: Gabrielle L. Boulianne,
| |
Collapse
|
37
|
Gupta P, Rani V. The Surging Mechanistic Role of Angiotensin Converting Enzyme 2 in Human Pathologies: A Potential Approach for Herbal Therapeutics. Curr Drug Targets 2023; 24:1046-1054. [PMID: 37861036 DOI: 10.2174/0113894501247616231009065415] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Revised: 07/27/2023] [Accepted: 09/15/2023] [Indexed: 10/21/2023]
Abstract
Advancements in biological sciences revealed the significant role of angiotensin-converting enzyme 2 (ACE2), a key cell surface receptor in various human pathologies. ACE2 is a metalloproteinase that not only functions in the regulation of Angiotensin II but also possesses some non-catalytic roles in the human body. There is considerable uncertainty regarding its protein expression, despite its presence in virtually all organs. The level of ACE2 expression and its subcellular localisation in humans may be a key determinant of susceptibility to various infections, symptoms, and outcomes of numerous diseases. Therefore, we summarize the distribution and expression pattern of ACE2 in different cell types related to all major human tissues and organs. Moreover, this review constitutes accumulated evidences of the important resources for further studies on ACE2 Inhibitory capacity via different natural compounds in order to understand its mechanism as the potential drug target in disease pathophysiology and to aid in the development of an effective therapeutic approach towards the various diseases.
Collapse
Affiliation(s)
- Priyadarshini Gupta
- Transcriptome laboratory, Centre of Emerging Diseases, Department of Biotechnology, Jaypee Institute of Information Technology, Sector-62, Noida, Uttar Pradesh, India
| | - Vibha Rani
- Transcriptome laboratory, Centre of Emerging Diseases, Department of Biotechnology, Jaypee Institute of Information Technology, Sector-62, Noida, Uttar Pradesh, India
| |
Collapse
|
38
|
MacLachlan R, Evans CE, Chai SY, Good MA, Kehoe PG, Miners JS. Age-related reduction in brain ACE-2 is not exacerbated by Alzheimer's disease pathology in mouse models of Alzheimer's disease. AGING BRAIN 2023; 3:100062. [PMID: 36911263 PMCID: PMC9997187 DOI: 10.1016/j.nbas.2022.100062] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Revised: 10/26/2022] [Accepted: 12/20/2022] [Indexed: 12/30/2022] Open
Abstract
An imbalance in the circulatory and organ-specific renin-angiotensin system (RAS) pathways is associated with age-related dysfunction and disease including cardiovascular burden and more recently Alzheimer's disease (AD). It is currently unclear whether an age-associated imbalance in components of the RAS within the brain precedes the onset of AD or whether a RAS imbalance is associated with the onset of disease pathology and cognitive decline. Angiotensin-converting enzyme-1 (ACE-1) and -2 (ACE-2) protein (ELISA) and enzyme activity (FRET assay), markers of the classical and counter-regulatory RAS axis respectively, and Ang-II and Ang-(1-7) peptide levels (ELISA), were measured in the left cortex across four transgenic AD mouse models of amyloid pathology (5xFAD - 2, 6, and 12 months of age; Apd9 - 3-4, 12, and 18 months of age; Tg2576 - 3-4 and 24 months of age; and PDAPP - 3-4, 7, 11, 15, and 18 months of age) and littermate wild-type (WT) controls. ACE-1 level, and enzyme activity, was unaltered in relation to age in WT mice and across all four models. In contrast, ACE-2 level and enzyme activity, was reduced and Ang-II increased with ageing in both WT animals and disease models. The changes in ACE-2 and Ang-II in AD models mirrored WT mice, except for the 5xFAD model, when the reduction in ACE-2 (and elevated Ang-II) was observed at a younger age. These data indicate an age-related dysregulation of brain RAS is likely to be driven by a reduction in ACE-2. The reduction in ACE-2 occurs at a young age, coinciding with early pathological changes and the initial deposition of Aβ, and preceding neuronal loss and cognitive decline, in the transgenic AD models. However, the age-related loss was mirrored in WT mice suggesting that the change was independent of pathological Aβ deposition.
Collapse
Affiliation(s)
- Robert MacLachlan
- Dementia Research Group, Clinical Neurosciences, Bristol Medical School, University of Bristol, Learning and Research Building, Southmead Hospital, BS10 5NB, United Kingdom
| | - Charles E Evans
- School of Psychology, Cardiff University, Cardiff CF10 3AT, United Kingdom
| | - Siew Yeen Chai
- Monash Biomedicine Discovery Institute, Department of Physiology, Monash University, Clayton, Victoria 3800, Australia
| | - Mark A Good
- School of Psychology, Cardiff University, Cardiff CF10 3AT, United Kingdom
| | - Patrick Gavin Kehoe
- Dementia Research Group, Clinical Neurosciences, Bristol Medical School, University of Bristol, Learning and Research Building, Southmead Hospital, BS10 5NB, United Kingdom
| | - J Scott Miners
- Dementia Research Group, Clinical Neurosciences, Bristol Medical School, University of Bristol, Learning and Research Building, Southmead Hospital, BS10 5NB, United Kingdom
| |
Collapse
|
39
|
Dini I, De Biasi MG, Mancusi A. An Overview of the Potentialities of Antimicrobial Peptides Derived from Natural Sources. Antibiotics (Basel) 2022; 11:1483. [PMID: 36358138 PMCID: PMC9686932 DOI: 10.3390/antibiotics11111483] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Revised: 10/20/2022] [Accepted: 10/21/2022] [Indexed: 07/21/2023] Open
Abstract
Antimicrobial peptides (AMPs) are constituents of the innate immune system in every kind of living organism. They can act by disrupting the microbial membrane or without affecting membrane stability. Interest in these small peptides stems from the fear of antibiotics and the emergence of microorganisms resistant to antibiotics. Through membrane or metabolic disruption, they defend an organism against invading bacteria, viruses, protozoa, and fungi. High efficacy and specificity, low drug interaction and toxicity, thermostability, solubility in water, and biological diversity suggest their applications in food, medicine, agriculture, animal husbandry, and aquaculture. Nanocarriers can be used to protect, deliver, and improve their bioavailability effectiveness. High cost of production could limit their use. This review summarizes the natural sources, structures, modes of action, and applications of microbial peptides in the food and pharmaceutical industries. Any restrictions on AMPs' large-scale production are also taken into consideration.
Collapse
Affiliation(s)
- Irene Dini
- Department of Pharmacy, University of Naples Federico II, Via Domenico Montesano 49, 80131 Napoli, Italy
| | | | - Andrea Mancusi
- Department of Food Microbiology, Istituto Zooprofilattico Sperimentale del Mezzogiorno, Via Salute 2, 80055 Portici, Italy
| |
Collapse
|
40
|
Pelle MC, Zaffina I, Lucà S, Forte V, Trapanese V, Melina M, Giofrè F, Arturi F. Endothelial Dysfunction in COVID-19: Potential Mechanisms and Possible Therapeutic Options. Life (Basel) 2022; 12:1605. [PMID: 36295042 PMCID: PMC9604693 DOI: 10.3390/life12101605] [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: 09/05/2022] [Revised: 09/29/2022] [Accepted: 10/12/2022] [Indexed: 11/06/2022] Open
Abstract
SARS-CoV-2, a novel coronavirus found in Wuhan (China) at the end of 2019, is the etiological agent of the current pandemic that is a heterogeneous disease, named coronavirus disease 2019 (COVID-19). SARS-CoV-2 affects primarily the lungs, but it can induce multi-organ involvement such as acute myocardial injury, myocarditis, thromboembolic eventsandrenal failure. Hypertension, chronic kidney disease, diabetes mellitus and obesity increase the risk of severe complications of COVID-19. There is no certain explanation for this systemic COVID-19 involvement, but it could be related to endothelial dysfunction, due to direct (endothelial cells are infected by the virus) and indirect damage (systemic inflammation) factors. Angiotensin-converting enzyme 2 (ACE2), expressed in human endothelium, has a fundamental role in severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection. In fact, ACE2 is used as a receptor by SARS-CoV-2, leading to the downregulation of these receptors on endothelial cells; once inside, this virus reduces the integrity of endothelial tissue, with exposure of prothrombotic molecules, platelet adhesion, activation of coagulation cascades and, consequently, vascular damage. Systemic microangiopathy and thromboembolism can lead to multi-organ failure with an elevated risk of death. Considering the crucial role of the immunological response and endothelial damage in developing the severe form of COVID-19, in this review, we will attempt to clarify the underlying pathophysiological mechanisms.
Collapse
Affiliation(s)
- Maria Chiara Pelle
- Unit of Internal Medicine, Department of Medical and Surgical Sciences, University “Magna Graecia” of Catanzaro, 88100 Catanzaro, Italy
| | - Isabella Zaffina
- Unit of Internal Medicine, Department of Medical and Surgical Sciences, University “Magna Graecia” of Catanzaro, 88100 Catanzaro, Italy
| | - Stefania Lucà
- Unit of Internal Medicine, Department of Medical and Surgical Sciences, University “Magna Graecia” of Catanzaro, 88100 Catanzaro, Italy
| | - Valentina Forte
- Unit of Internal Medicine, Department of Medical and Surgical Sciences, University “Magna Graecia” of Catanzaro, 88100 Catanzaro, Italy
| | - Vincenzo Trapanese
- Unit of Internal Medicine, Department of Medical and Surgical Sciences, University “Magna Graecia” of Catanzaro, 88100 Catanzaro, Italy
| | - Melania Melina
- Unit of Internal Medicine, Department of Medical and Surgical Sciences, University “Magna Graecia” of Catanzaro, 88100 Catanzaro, Italy
| | - Federica Giofrè
- Unit of Internal Medicine, Department of Medical and Surgical Sciences, University “Magna Graecia” of Catanzaro, 88100 Catanzaro, Italy
| | - Franco Arturi
- Unit of Internal Medicine, Department of Medical and Surgical Sciences, University “Magna Graecia” of Catanzaro, 88100 Catanzaro, Italy
- Research Centre for the Prevention and Treatment of Metabolic Diseases (CR METDIS), University “Magna Graecia” of Catanzaro, 88100 Catanzaro, Italy
| |
Collapse
|
41
|
Cruz-López EO, Ye D, Wu C, Lu HS, Uijl E, Mirabito Colafella KM, Danser AHJ. Angiotensinogen Suppression: A New Tool to Treat Cardiovascular and Renal Disease. Hypertension 2022; 79:2115-2126. [PMID: 35904033 PMCID: PMC9444253 DOI: 10.1161/hypertensionaha.122.18731] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Multiple types of renin-angiotensin system (RAS) blockers exist, allowing interference with the system at the level of renin, angiotensin-converting enzyme, or the angiotensin II receptor. Yet, in particular, for the treatment of hypertension, the number of patients with uncontrolled hypertension continues to rise, either due to patient noncompliance or because of the significant renin rises that may, at least partially, overcome the effect of RAS blockade (RAS escape). New approaches to target the RAS are either direct antisense oligonucleotides that inhibit angiotensinogen RNA translation, or small interfering RNA (siRNA) that function via the RNA interference pathway. Since all angiotensins stem from angiotensinogen, lowering angiotensinogen has the potential to circumvent the RAS escape phenomenon. Moreover, antisense oligonucleotides and small interfering RNA require injections only every few weeks to months, which might reduce noncompliance. Of course, angiotensinogen suppression also poses a threat in situations where the RAS is acutely needed, for instance in women becoming pregnant during treatment, or in cases of emergency, when severe hypotension occurs. This review discusses all preclinical data on angiotensinogen suppression, as well as the limited clinical data that are currently available. It concludes that it is an exciting new tool to target the RAS with high specificity and a low side effect profile. Its long-term action might revolutionize pharmacotherapy, as it could overcome compliance problems. Preclinical and clinical programs are now carefully investigating its efficacy and safety profile, allowing an optimal introduction as a novel drug to treat cardiovascular and renal diseases in due time.
Collapse
Affiliation(s)
- Edwyn O Cruz-López
- Division of Pharmacology and Vascular Medicine, Department of Internal Medicine, Erasmus MC, University Medical Center Rotterdam, the Netherlands (E.O.C.L., D.Y., E.U., A.H.J.D.)
| | - Dien Ye
- Division of Pharmacology and Vascular Medicine, Department of Internal Medicine, Erasmus MC, University Medical Center Rotterdam, the Netherlands (E.O.C.L., D.Y., E.U., A.H.J.D.)
| | - Congqing Wu
- Saha Cardiovascular Research Center (C.W., H.S.L.), University of Kentucky.,Department of Surgery (C.W.), University of Kentucky
| | - Hong S Lu
- Saha Cardiovascular Research Center (C.W., H.S.L.), University of Kentucky.,Department of Physiology (H.S.L.), University of Kentucky
| | - Estrellita Uijl
- Division of Pharmacology and Vascular Medicine, Department of Internal Medicine, Erasmus MC, University Medical Center Rotterdam, the Netherlands (E.O.C.L., D.Y., E.U., A.H.J.D.)
| | | | - A H Jan Danser
- Division of Pharmacology and Vascular Medicine, Department of Internal Medicine, Erasmus MC, University Medical Center Rotterdam, the Netherlands (E.O.C.L., D.Y., E.U., A.H.J.D.)
| |
Collapse
|
42
|
Plasma Angiotensin Converting Enzyme 2 (ACE2) Activity in Healthy Controls and Patients with Cardiovascular Risk Factors and/or Disease. J Pers Med 2022; 12:jpm12091495. [PMID: 36143280 PMCID: PMC9501250 DOI: 10.3390/jpm12091495] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 09/05/2022] [Accepted: 09/08/2022] [Indexed: 11/18/2022] Open
Abstract
Angiotensin converting enzyme 2 (ACE2) is an endogenous negative regulator of the renin-angiotensin system, a key factor in the development of cardiovascular disease (CVD). ACE2 is also used by SARS-CoV-2 for host cell entry. Given that COVID-19 is associated with hypercoagulability, it is timely to explore the potential relationship between plasma ACE2 activity and the coagulation profile. In this cross-sectional study, ACE2 activity and global coagulation assays (GCA) including thromboelastography, thrombin, and fibrin generation were measured in adult healthy controls (n = 123; mean age 41 ± 17 years; 35% male) and in patients with cardiovascular risk factors and/or disease (n = 258; mean age 65 ± 14 years; 55% male). ACE2 activity was significantly lower in controls compared to patients with cardiovascular risk factors and/or disease (median 0.10 (0.02, 3.33) vs. 5.99 (1.95, 10.37) pmol/mL/min, p < 0.001). Of the healthy controls, 48% had undetectable ACE2 activity. Controls with detectable ACE2 had lower maximum amplitude (p < 0.001). In patients with cardiovascular risk factors and/or disease, those in the 3rd tertile were older and male (p = 0.002), with a higher Framingham grade and increased number of cardiovascular risk factors (p < 0.001). In conclusion, plasma ACE2 activity is undetectable to very low in young healthy controls with minimal clinically relevant associations to GCA. Patients with cardiovascular risk factors and/or disease have increased plasma ACE2 activity, suggesting that it may be an important biomarker of endothelial dysfunction and atherosclerosis.
Collapse
|
43
|
Interactions between the intrarenal dopaminergic and the renin-angiotensin systems in the control of systemic arterial pressure. Clin Sci (Lond) 2022; 136:1205-1227. [PMID: 35979889 DOI: 10.1042/cs20220338] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Revised: 07/31/2022] [Accepted: 08/03/2022] [Indexed: 11/17/2022]
Abstract
Systemic arterial hypertension is one of the leading causes of morbidity and mortality in the general population, being a risk factor for many cardiovascular diseases. Although its pathogenesis is complex and still poorly understood, some systems appear to play major roles in its development. This review aims to update the current knowledge on the interaction of the intrarenal renin-angiotensin system (RAS) and dopaminergic system in the development of hypertension, focusing on recent scientific hallmarks in the field. The intrarenal RAS, composed of several peptides and receptors, has a critical role in the regulation of blood pressure (BP) and, consequently, the development of hypertension. The RAS is divided into two main intercommunicating axes: the classical axis, composed of angiotensin-converting enzyme, angiotensin II, and angiotensin type 1 receptor, and the ACE2/angiotensin-(1-7)/Mas axis, which appears to modulate the effects of the classical axis. Dopamine and its receptors are also increasingly showing an important role in the pathogenesis of hypertension, as abnormalities in the intrarenal dopaminergic system impair the regulation of renal sodium transport, regardless of the affected dopamine receptor subtype. There are five dopamine receptors, which are divided into two major subtypes: the D1-like (D1R and D5R) and D2-like (D2R, D3R, and D4R) receptors. Mice deficient in any of the five dopamine receptor subtypes have increased BP. Intrarenal RAS and the dopaminergic system have complex interactions. The balance between both systems is essential to regulate the BP homeostasis, as alterations in the control of both can lead to hypertension.
Collapse
|
44
|
Potue P, Chiangsaen P, Maneesai P, Khamseekaew J, Pakdeechote P, Chankitisakul V, Boonkum W, Duanghaklang N, Duangjinda M. Effects of Thai native chicken breast meat consumption on serum uric acid level, biochemical parameters, and antioxidant activities in rats. Sci Rep 2022; 12:14056. [PMID: 35982129 PMCID: PMC9388516 DOI: 10.1038/s41598-022-18484-2] [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: 03/13/2022] [Accepted: 08/12/2022] [Indexed: 11/18/2022] Open
Abstract
This study aimed to evaluate the effect of a high protein diet comprising breast meat from commercial broiler (BR), Thai native (PD), and commercial broiler × Thai native crossbred (KKU-ONE) chicken on serum uric acid, biochemical parameters, and antioxidant activities in rats. Male Sprague–Dawley rats were divided into four groups. The control group received a standard chow diet, and the other three groups were fed a high protein diet (70% standard diet + 30% BR, PD, or KKU-ONE chicken breast) for five weeks. The PD- and KKU-ONE-fed rats had lower plasma total cholesterol and triglyceride levels than the control rats. A decrease in HDL-c was also observed in rats fed a diet containing BR. Liver weight, liver enzyme, plasma ALP, xanthine oxidase activity, serum uric acid, creatinine, superoxide production, and plasma malondialdehyde levels increased in BR-fed rats. The findings of this study might provide evidence to support the use of Thai native and Thai native crossbred chicken breast meat as functional foods.
Collapse
Affiliation(s)
- Prapassorn Potue
- Department of Physiology, Faculty of Medicine, Khon Kaen University, Khon Kaen, 40002, Thailand
| | | | - Putcharawipa Maneesai
- Department of Physiology, Faculty of Medicine, Khon Kaen University, Khon Kaen, 40002, Thailand
| | - Juthamas Khamseekaew
- Department of Physiology, Faculty of Medicine, Khon Kaen University, Khon Kaen, 40002, Thailand
| | - Poungrat Pakdeechote
- Department of Physiology, Faculty of Medicine, Khon Kaen University, Khon Kaen, 40002, Thailand
| | - Vibuntita Chankitisakul
- Department of Animal Science, Faculty of Agriculture, Khon Kaen University, Khon Kaen, 40002, Thailand.,Network Center for Animal Breeding and Omics Research, Khon Kaen University, Khon Kaen, 40002, Thailand
| | - Wuttigrai Boonkum
- Department of Animal Science, Faculty of Agriculture, Khon Kaen University, Khon Kaen, 40002, Thailand.,Network Center for Animal Breeding and Omics Research, Khon Kaen University, Khon Kaen, 40002, Thailand
| | - Natthaya Duanghaklang
- Network Center for Animal Breeding and Omics Research, Khon Kaen University, Khon Kaen, 40002, Thailand
| | - Monchai Duangjinda
- Department of Animal Science, Faculty of Agriculture, Khon Kaen University, Khon Kaen, 40002, Thailand. .,Network Center for Animal Breeding and Omics Research, Khon Kaen University, Khon Kaen, 40002, Thailand.
| |
Collapse
|
45
|
Lee WC, Wu PY, Huang JC, Tsai YC, Chiu YW, Chen SC, Chang JM, Chen HC. Sex Difference in the Associations among Obesity-Related Indices with Incident Hypertension in a Large Taiwanese Population Follow-Up Study. J Pers Med 2022; 12:jpm12060972. [PMID: 35743756 PMCID: PMC9225143 DOI: 10.3390/jpm12060972] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Revised: 06/08/2022] [Accepted: 06/14/2022] [Indexed: 12/01/2022] Open
Abstract
Hypertension is a major risk factor for stroke, atherosclerosis, and other cardiovascular diseases, and obesity is a major risk factor for hypertension. The aim of this longitudinal study was to investigate sex differences in the correlations among obesity-related indices and incident hypertension in a large Taiwanese cohort. We included 21,466 enrollees in the Taiwan Biobank and followed them for 4 years. Of the 21,466 patients enrolled in this study, 6899 (mean age, 49.6 ± 10.9 years) were male and 14,567 (mean age, 49.7 ± 10.0 years) were female. Data on visceral adiposity index (VAI), waist-to-height ratio (WHtR), waist-to-hip ratio (WHR), lipid accumulation product (LAP), conicity index (CI), body roundness index (BRI), body mass index (BMI), body adiposity index (BAI), and abdominal volume index (AVI) were collected and analyzed. The results showed that all of the studied obesity-related indices were significantly associated with incident hypertension. Among them, WHtR was the strongest predictor of hypertension in both sexes. In addition, interactions between VAI, LAP, CI, BMI, and AVI with sex on incident hypertension were also statistically significant. CI and AVI were more strongly associated with hypertension in the men than in the women, while VAI, LAP, and BMI were more strongly associated with hypertension in the women. In conclusion, the studied obesity-related indices were found to be predictors of incident hypertension, and there were differences in the associations between the male and female participants. Our findings may imply that reducing body weight may be associated with a lower risk of developing hypertension.
Collapse
Affiliation(s)
- Wen-Chi Lee
- Department of General Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807, Taiwan;
| | - Pei-Yu Wu
- Division of Nephrology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807, Taiwan; (P.-Y.W.); (J.-C.H.); (Y.-C.T.); (Y.-W.C.); (J.-M.C.); (H.-C.C.)
- Department of Internal Medicine, Kaohsiung Municipal Siaogang Hospital, Kaohsiung Medical University, Kaohsiung 812, Taiwan
| | - Jiun-Chi Huang
- Division of Nephrology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807, Taiwan; (P.-Y.W.); (J.-C.H.); (Y.-C.T.); (Y.-W.C.); (J.-M.C.); (H.-C.C.)
- Department of Internal Medicine, Kaohsiung Municipal Siaogang Hospital, Kaohsiung Medical University, Kaohsiung 812, Taiwan
- Faculty of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Yi-Chun Tsai
- Division of Nephrology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807, Taiwan; (P.-Y.W.); (J.-C.H.); (Y.-C.T.); (Y.-W.C.); (J.-M.C.); (H.-C.C.)
- Faculty of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Yi-Wen Chiu
- Division of Nephrology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807, Taiwan; (P.-Y.W.); (J.-C.H.); (Y.-C.T.); (Y.-W.C.); (J.-M.C.); (H.-C.C.)
- Faculty of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Szu-Chia Chen
- Division of Nephrology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807, Taiwan; (P.-Y.W.); (J.-C.H.); (Y.-C.T.); (Y.-W.C.); (J.-M.C.); (H.-C.C.)
- Department of Internal Medicine, Kaohsiung Municipal Siaogang Hospital, Kaohsiung Medical University, Kaohsiung 812, Taiwan
- Faculty of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Research Center for Environmental Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Correspondence: ; Tel.: +886-7-8036783 (ext. 3440); Fax: +886-7-8063346
| | - Jer-Ming Chang
- Division of Nephrology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807, Taiwan; (P.-Y.W.); (J.-C.H.); (Y.-C.T.); (Y.-W.C.); (J.-M.C.); (H.-C.C.)
- Faculty of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Hung-Chun Chen
- Division of Nephrology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807, Taiwan; (P.-Y.W.); (J.-C.H.); (Y.-C.T.); (Y.-W.C.); (J.-M.C.); (H.-C.C.)
- Faculty of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| |
Collapse
|
46
|
Ye D, Wu C, Chen H, Liang CL, Howatt DA, Franklin MK, Moorleghen JJ, Tyagi SC, Uijl E, Danser AHJ, Sawada H, Daugherty A, Lu HS. Fludrocortisone Induces Aortic Pathologies in Mice. Biomolecules 2022; 12:825. [PMID: 35740952 PMCID: PMC9220881 DOI: 10.3390/biom12060825] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Revised: 06/07/2022] [Accepted: 06/08/2022] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND AND OBJECTIVE In an experiment designed to explore the mechanisms of fludrocortisone-induced high blood pressure, we serendipitously observed aortic aneurysms in mice infused with fludrocortisone. The purpose of this study was to investigate whether fludrocortisone induces aortic pathologies in both normocholesterolemic and hypercholesterolemic mice. METHODS AND RESULTS Male adult C57BL/6J mice were infused with either vehicle (85% polyethylene glycol 400 (PEG-400) and 15% dimethyl sulfoxide (DMSO); n = 5) or fludrocortisone (12 mg/kg/day dissolved in 85% PEG-400 and 15% DMSO; n = 15) for 28 days. Fludrocortisone-infused mice had higher systolic blood pressure, compared to mice infused with vehicle. Fludrocortisone induced aortic pathologies in 4 of 15 mice with 3 having pathologies in the ascending and aortic arch regions and 1 having pathology in both the ascending and descending thoracic aorta. No pathologies were noted in abdominal aortas. Subsequently, we infused either vehicle (n = 5/group) or fludrocortisone (n = 15/group) into male ApoE -/- mice fed a normal laboratory diet or LDL receptor -/- mice fed either normal or Western diet. Fludrocortisone increased systolic blood pressure, irrespective of mouse strain or diet. In ApoE -/- mice infused with fludrocortisone, 2 of 15 mice had ascending aortic pathologies, but no mice had abdominal aortic pathologies. In LDL receptor -/- mice fed normal diet, 5 had ascending/arch pathologies and 1 had pathologies in the ascending, arch, and suprarenal aortic regions. In LDL receptor -/- mice fed Western diet, 2 died of aortic rupture in either the descending thoracic or abdominal region, and 2 of the 13 survived mice had ascending/arch aortic pathologies. Aortic pathologies included hemorrhage, wall thickening or thinning, or dilation. Only ascending aortic diameter in LDLR -/- mice fed Western diet reached statistical significance, compared to their vehicle. CONCLUSION Fludrocortisone induces aortic pathologies independent of hypercholesterolemia. As indicated by the findings in mouse studies, people who are taking or have taken fludrocortisone might have an increased risk of aortic pathologies.
Collapse
Affiliation(s)
- Dien Ye
- Saha Cardiovascular Research Center, Lexington, KY 40536, USA; (D.Y.); (C.W.); (H.C.); (C.-L.L.); (D.A.H.); (M.K.F.); (J.J.M.); (S.C.T.); (H.S.); (A.D.)
- Division of Pharmacology and Vascular Medicine, Department of Internal Medicine, Erasmus University Medical Center, 3015 CN Rotterdam, The Netherlands; (E.U.); (A.H.J.D.)
| | - Congqing Wu
- Saha Cardiovascular Research Center, Lexington, KY 40536, USA; (D.Y.); (C.W.); (H.C.); (C.-L.L.); (D.A.H.); (M.K.F.); (J.J.M.); (S.C.T.); (H.S.); (A.D.)
- Saha Cardiovascular Research Center, Department of Surgery, College of Medicine, University of Kentucky, Lexington, KY 40536, USA
| | - Hui Chen
- Saha Cardiovascular Research Center, Lexington, KY 40536, USA; (D.Y.); (C.W.); (H.C.); (C.-L.L.); (D.A.H.); (M.K.F.); (J.J.M.); (S.C.T.); (H.S.); (A.D.)
| | - Ching-Ling Liang
- Saha Cardiovascular Research Center, Lexington, KY 40536, USA; (D.Y.); (C.W.); (H.C.); (C.-L.L.); (D.A.H.); (M.K.F.); (J.J.M.); (S.C.T.); (H.S.); (A.D.)
| | - Deborah A. Howatt
- Saha Cardiovascular Research Center, Lexington, KY 40536, USA; (D.Y.); (C.W.); (H.C.); (C.-L.L.); (D.A.H.); (M.K.F.); (J.J.M.); (S.C.T.); (H.S.); (A.D.)
| | - Michael K. Franklin
- Saha Cardiovascular Research Center, Lexington, KY 40536, USA; (D.Y.); (C.W.); (H.C.); (C.-L.L.); (D.A.H.); (M.K.F.); (J.J.M.); (S.C.T.); (H.S.); (A.D.)
| | - Jessica J. Moorleghen
- Saha Cardiovascular Research Center, Lexington, KY 40536, USA; (D.Y.); (C.W.); (H.C.); (C.-L.L.); (D.A.H.); (M.K.F.); (J.J.M.); (S.C.T.); (H.S.); (A.D.)
| | - Samuel C. Tyagi
- Saha Cardiovascular Research Center, Lexington, KY 40536, USA; (D.Y.); (C.W.); (H.C.); (C.-L.L.); (D.A.H.); (M.K.F.); (J.J.M.); (S.C.T.); (H.S.); (A.D.)
- Saha Cardiovascular Research Center, Department of Surgery, College of Medicine, University of Kentucky, Lexington, KY 40536, USA
- Department of Physiology, University of Kentucky, Lexington, KY 40536, USA
- Saha Aortic Center, University of Kentucky, Lexington, KY 40536, USA
| | - Estrellita Uijl
- Division of Pharmacology and Vascular Medicine, Department of Internal Medicine, Erasmus University Medical Center, 3015 CN Rotterdam, The Netherlands; (E.U.); (A.H.J.D.)
| | - A. H. Jan Danser
- Division of Pharmacology and Vascular Medicine, Department of Internal Medicine, Erasmus University Medical Center, 3015 CN Rotterdam, The Netherlands; (E.U.); (A.H.J.D.)
| | - Hisashi Sawada
- Saha Cardiovascular Research Center, Lexington, KY 40536, USA; (D.Y.); (C.W.); (H.C.); (C.-L.L.); (D.A.H.); (M.K.F.); (J.J.M.); (S.C.T.); (H.S.); (A.D.)
- Department of Physiology, University of Kentucky, Lexington, KY 40536, USA
- Saha Aortic Center, University of Kentucky, Lexington, KY 40536, USA
| | - Alan Daugherty
- Saha Cardiovascular Research Center, Lexington, KY 40536, USA; (D.Y.); (C.W.); (H.C.); (C.-L.L.); (D.A.H.); (M.K.F.); (J.J.M.); (S.C.T.); (H.S.); (A.D.)
- Department of Physiology, University of Kentucky, Lexington, KY 40536, USA
- Saha Aortic Center, University of Kentucky, Lexington, KY 40536, USA
| | - Hong S. Lu
- Saha Cardiovascular Research Center, Lexington, KY 40536, USA; (D.Y.); (C.W.); (H.C.); (C.-L.L.); (D.A.H.); (M.K.F.); (J.J.M.); (S.C.T.); (H.S.); (A.D.)
- Department of Physiology, University of Kentucky, Lexington, KY 40536, USA
- Saha Aortic Center, University of Kentucky, Lexington, KY 40536, USA
| |
Collapse
|
47
|
Zhou A, Selvanayagam JB, Hyppönen E. Non-linear Mendelian randomization analyses support a role for vitamin D deficiency in cardiovascular disease risk. Eur Heart J 2022; 43:1731-1739. [PMID: 34891159 DOI: 10.1093/eurheartj/ehab809] [Citation(s) in RCA: 107] [Impact Index Per Article: 35.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Revised: 09/28/2021] [Accepted: 11/12/2021] [Indexed: 11/14/2022] Open
Abstract
AIMS Low vitamin D status is associated with a higher risk for cardiovascular diseases (CVDs). Although most existing linear Mendelian randomization (MR) studies reported a null effect of vitamin D on CVD risk, a non-linear effect cannot be excluded. Our aim was to apply the non-linear MR design to investigate the association of serum 25-hydroxyvitamin D [25(OH)D] concentration with CVD risk. METHODS AND RESULTS The non-linear MR analysis was conducted in the UK Biobank with 44 519 CVD cases and 251 269 controls. Blood pressure (BP) and cardiac-imaging-derived phenotypes were included as secondary outcomes. Serum 25(OH)D concentration was instrumented using 35 confirmed genome-wide significant variants.We also estimated the potential reduction in CVD incidence attributable to correction of low vitamin D status. There was a L-shaped association between genetically predicted serum 25(OH)D and CVD risk (Pnon-linear = 0.007), where CVD risk initially decreased steeply with increasing concentrations and levelled off at around 50 nmol/L. A similar association was seen for systolic (Pnon-linear = 0.03) and diastolic (Pnon-linear = 0.07) BP. No evidence of association was seen for cardiac-imaging phenotypes (P = 0.05 for all). Correction of serum 25(OH)D level below 50 nmol/L was predicted to result in a 4.4% reduction in CVD incidence (95% confidence interval: 1.8- 7.3%). CONCLUSION Vitamin D deficiency can increase the risk of CVD. Burden of CVD could be reduced by population-wide correction of low vitamin D status.
Collapse
Affiliation(s)
- Ang Zhou
- Australian Center for Precision Health, University of South Australia Cancer Research Institute, GPO Box 2471, Adelaide, SA 5001, Australia
- South Australian Health and Medical Research Institute, North Terrace, Adelaide, SA 5000, Australia
| | - Joseph B Selvanayagam
- South Australian Health and Medical Research Institute, North Terrace, Adelaide, SA 5000, Australia
- Department of Cardiovascular Medicine, Flinders Medical Centre, Bedford Park, Adelaide, SA 5042, Australia
| | - Elina Hyppönen
- Australian Center for Precision Health, University of South Australia Cancer Research Institute, GPO Box 2471, Adelaide, SA 5001, Australia
- South Australian Health and Medical Research Institute, North Terrace, Adelaide, SA 5000, Australia
- Population, Policy and Practice, UCL Great Ormond Street Institute of Child Health, 30 Guilford Street, London, UK
| |
Collapse
|
48
|
Lertampaiporn S, Hongsthong A, Wattanapornprom W, Thammarongtham C. Ensemble-AHTPpred: A Robust Ensemble Machine Learning Model Integrated With a New Composite Feature for Identifying Antihypertensive Peptides. Front Genet 2022; 13:883766. [PMID: 35571042 PMCID: PMC9096110 DOI: 10.3389/fgene.2022.883766] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Accepted: 04/04/2022] [Indexed: 11/13/2022] Open
Abstract
Hypertension or elevated blood pressure is a serious medical condition that significantly increases the risks of cardiovascular disease, heart disease, diabetes, stroke, kidney disease, and other health problems, that affect people worldwide. Thus, hypertension is one of the major global causes of premature death. Regarding the prevention and treatment of hypertension with no or few side effects, antihypertensive peptides (AHTPs) obtained from natural sources might be useful as nutraceuticals. Therefore, the search for alternative/novel AHTPs in food or natural sources has received much attention, as AHTPs may be functional agents for human health. AHTPs have been observed in diverse organisms, although many of them remain underinvestigated. The identification of peptides with antihypertensive activity in the laboratory is time- and resource-consuming. Alternatively, computational methods based on robust machine learning can identify or screen potential AHTP candidates prior to experimental verification. In this paper, we propose Ensemble-AHTPpred, an ensemble machine learning algorithm composed of a random forest (RF), a support vector machine (SVM), and extreme gradient boosting (XGB), with the aim of integrating diverse heterogeneous algorithms to enhance the robustness of the final predictive model. The selected feature set includes various computed features, such as various physicochemical properties, amino acid compositions (AACs), transitions, n-grams, and secondary structure-related information; these features are able to learn more information in terms of analyzing or explaining the characteristics of the predicted peptide. In addition, the tool is integrated with a newly proposed composite feature (generated based on a logistic regression function) that combines various feature aspects to enable improved AHTP characterization. Our tool, Ensemble-AHTPpred, achieved an overall accuracy above 90% on independent test data. Additionally, the approach was applied to novel experimentally validated AHTPs, obtained from recent studies, which did not overlap with the training and test datasets, and the tool could precisely predict these AHTPs.
Collapse
Affiliation(s)
- Supatcha Lertampaiporn
- Biochemical Engineering and Systems Biology Research Group, National Center for Genetic Engineering and Biotechnology, National Science and Technology Development Agency at King Mongkut’s University of Technology Thonburi, Bangkok, Thailand
| | - Apiradee Hongsthong
- Biochemical Engineering and Systems Biology Research Group, National Center for Genetic Engineering and Biotechnology, National Science and Technology Development Agency at King Mongkut’s University of Technology Thonburi, Bangkok, Thailand
| | - Warin Wattanapornprom
- Applied Computer Science Program, Department of Mathematics, Faculty of Science, King Mongkut’s University of Technology Thonburi, Bangkok, Thailand
| | - Chinae Thammarongtham
- Biochemical Engineering and Systems Biology Research Group, National Center for Genetic Engineering and Biotechnology, National Science and Technology Development Agency at King Mongkut’s University of Technology Thonburi, Bangkok, Thailand
- *Correspondence: Chinae Thammarongtham,
| |
Collapse
|
49
|
Kuai Y, Huang H, Dai X, Zhang Z, Bai Z, Chen J, Fang F, Pan J, Li X, Wang J, Li Y. In PICU acute kidney injury stage 3 or mortality is associated with early excretion of urinary renin. Pediatr Res 2022; 91:1149-1155. [PMID: 34083760 DOI: 10.1038/s41390-021-01592-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Revised: 05/12/2021] [Accepted: 05/17/2021] [Indexed: 11/09/2022]
Abstract
BACKGROUND Urinary renin is proposed to be a novel prognostic biomarker of acute kidney injury (AKI) in adults. The intention of our study was to evaluate the early predictive value of urinary renin for AKI and pediatric intensive care unit (PICU) mortality in critically ill children. METHODS The first available urine sample during the first 24 h after admission was collected upon PICU admission for the measurement of renin using ELISA. Urinary renin concentrations were corrected for urinary creatinine (urinary renin-to-creatinine ratio, uRenCR). AKI was defined based on KDIGO criteria. RESULTS Of the 207 children, 22 developed AKI, including 6 with stage 1, 6 with stage 2, and 10 with stage 3, and 14 died during PICU stay. There was a significant difference in uRenCR between non-AKI children and those with AKI stage 3 (P = 0.001), but not with AKI stage 1 or 2. The uRenCR remained associated with AKI stage 3 and PICU mortality after adjustment for potential confounders. The area under the receiver operating characteristic curve of uRenCR for discrimination of AKI stage 3 was 0.805, and PICU mortality was 0.801. CONCLUSIONS Urinary renin was associated with the increased risk for AKI stage 3 and PICU mortality in critically ill children. IMPACT Urinary renin is proposed to be a novel prognostic biomarker of AKI in adult patients. There are some differences between children and adults in physiological and pathophysiological characteristics. This study demonstrated that urinary renin was associated with the increased risk for AKI stage 3 and PICU mortality in critically ill children. Accurate identification of patients with severe renal injury or at high risk for mortality early in the disease course could augment the efficacy of available interventions and improve patient outcomes.
Collapse
Affiliation(s)
- Yuxian Kuai
- Department of Nephrology and Immunology, Children's Hospital of Soochow University, Suzhou, Jiangsu province, China
| | - Hui Huang
- Department of Nephrology and Immunology, Children's Hospital of Soochow University, Suzhou, Jiangsu province, China
| | - Xiaomei Dai
- Department of Nephrology and Immunology, Children's Hospital of Soochow University, Suzhou, Jiangsu province, China
| | - Zhongyue Zhang
- Department of Nephrology and Immunology, Children's Hospital of Soochow University, Suzhou, Jiangsu province, China
| | - Zhenjiang Bai
- Pediatric Intensive Care Unit, Children's Hospital of Soochow University, Suzhou, Jiangsu province, China
| | - Jiao Chen
- Pediatric Intensive Care Unit, Children's Hospital of Soochow University, Suzhou, Jiangsu province, China
| | - Fang Fang
- Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou, Jiangsu province, China
| | - Jian Pan
- Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou, Jiangsu province, China
| | - Xiaozhong Li
- Department of Nephrology and Immunology, Children's Hospital of Soochow University, Suzhou, Jiangsu province, China
| | - Jian Wang
- Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou, Jiangsu province, China
| | - Yanhong Li
- Department of Nephrology and Immunology, Children's Hospital of Soochow University, Suzhou, Jiangsu province, China. .,Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou, Jiangsu province, China.
| |
Collapse
|
50
|
Saed Aldien A, Ganesan GS, Wahbeh F, Al-Nassr N, Altarawneh H, Al Theyab L, Saed Aldien S, Tomerak S, Naveed H, Elshazly MB, Zakaria D. Systemic Inflammation May Induce Cardiac Injury in COVID-19 Patients Including Children and Adolescents Without Underlying Cardiovascular Diseases: A Systematic Review. CARDIOVASCULAR REVASCULARIZATION MEDICINE 2022; 35:169-178. [PMID: 33952432 PMCID: PMC8046745 DOI: 10.1016/j.carrev.2021.04.007] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Revised: 04/07/2021] [Accepted: 04/07/2021] [Indexed: 12/15/2022]
Abstract
Coronavirus disease 2019(COVID-19) is an ongoing global pandemic with a daily increasing number of affected individuals and a relatively high mortality rate. COVID-19 patients that develop cardiac injury are at increased risk of a worse clinical course with higher rates of mortality. Increasing amounts of evidence suggest that a system-wide inflammatory response and a cytokine storm mediated type syndrome plays a crucial role in disease progression. This systematic review investigates the possible role of hyperinflammation in inducing cardiac injury as one of the severe complications of COVID-19. A systematic literature search was performed using PubMed, Embase and Scopus databases to identify relevant clinical studies that investigated cardiovascular injury manifestations and reported inflammatory and cardiac biomarkers in COVID-19 patients. Only 29 studies met our inclusion criteria and the majority of these studies demonstrated significantly elevated inflammatory and cardiac blood markers. It was evident that underlying cardiovascular diseases may increase the risk of developing cardiac injury. However, many COVID-19 patients included in this review, developed different types of cardiac injury without having any underlying cardiovascular diseases. Furthermore, many of these patients were either children or adolescents. Therefore, age and comorbidities may not always be the two main risk factors that dictate the severity and outcome of COVID-19. Further investigations are required to understand the underlying mechanisms of pathogenicity as an urgent requirement to develop the appropriate treatment and prevention strategies. These strategies may specifically target hyperinflammation as a suspected driving factor for some of the severe complications of COVID-19.
Collapse
Affiliation(s)
- Arwa Saed Aldien
- Weill Cornell Medicine Qatar, Qatar Foundation, Education City, Al, Luqta St. Ar-Rayyan, P.O. Box 24144, Doha, Qatar
| | - Gowrii S Ganesan
- Weill Cornell Medicine Qatar, Qatar Foundation, Education City, Al, Luqta St. Ar-Rayyan, P.O. Box 24144, Doha, Qatar
| | - Farah Wahbeh
- Weill Cornell Medicine Qatar, Qatar Foundation, Education City, Al, Luqta St. Ar-Rayyan, P.O. Box 24144, Doha, Qatar
| | - Noor Al-Nassr
- Weill Cornell Medicine Qatar, Qatar Foundation, Education City, Al, Luqta St. Ar-Rayyan, P.O. Box 24144, Doha, Qatar
| | - Heba Altarawneh
- Weill Cornell Medicine Qatar, Qatar Foundation, Education City, Al, Luqta St. Ar-Rayyan, P.O. Box 24144, Doha, Qatar
| | - Lolwa Al Theyab
- Weill Cornell Medicine Qatar, Qatar Foundation, Education City, Al, Luqta St. Ar-Rayyan, P.O. Box 24144, Doha, Qatar
| | | | - Sara Tomerak
- Weill Cornell Medicine Qatar, Qatar Foundation, Education City, Al, Luqta St. Ar-Rayyan, P.O. Box 24144, Doha, Qatar
| | - Hiba Naveed
- Weill Cornell Medicine Qatar, Qatar Foundation, Education City, Al, Luqta St. Ar-Rayyan, P.O. Box 24144, Doha, Qatar
| | - Mohamed B Elshazly
- Weill Cornell Medicine Qatar, Qatar Foundation, Education City, Al, Luqta St. Ar-Rayyan, P.O. Box 24144, Doha, Qatar
| | - Dalia Zakaria
- Weill Cornell Medicine Qatar, Qatar Foundation, Education City, Al, Luqta St. Ar-Rayyan, P.O. Box 24144, Doha, Qatar.
| |
Collapse
|