1
|
Liu W, Wang H, Mu Q, Gong T. Taste receptor T1R3 regulates testosterone synthesis via the cAMP-PKA-SP1 pathway in testicular Leydig cells. Theriogenology 2025; 231:210-221. [PMID: 39476553 DOI: 10.1016/j.theriogenology.2024.10.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Revised: 10/18/2024] [Accepted: 10/21/2024] [Indexed: 11/15/2024]
Abstract
Taste receptor type 1 subunit 3 (T1R3) is a G protein-coupled receptor encoded by the TAS1R3 gene that can be specifically activated by certain sweeteners or umami agents for sweet/umami recognition. T1R3 is a potential target for regulating male reproduction. However, studies on the impact of non-nutritive sweeteners on reproduction are limited. In the present study, we evaluated the impact of the non-nutritive sweeteners (saccharin sodium, sucralose and acesulfame-K) on testosterone synthesis in testicular Leydig cells of Xiang pigs by comparing the relative abundance of mRNA transcripts and protein expression of T1R3, steroidogenic related factors, and intracellular cyclic adenosine monophosphate (cAMP), protein kinase A (PKA), as well as testosterone levels using Western blotting, reverse transcription-quantitative polymerase chain reaction (RT-qPCR) and enzyme-linked immunosorbent assay (ELISA). To clarify the specific mechanism, a dual luciferase assay was used to uncover the relationship between the transcription factors and steroidogenic enzyme. The acute intratesticular injection of a typical non-nutritive sweeteners was conducted to verify this impact in mouse. The results showed that saccharin sodium not only enhanced T1R3 expression in Leydig cells of Xiang pigs, but also caused significant increases in testosterone, cAMP, PKA, phosphorylation of specificity protein 1 (p-SP1), total protein of specificity protein 1 (SP1), steroidogenic acute regulatory protein (StAR), and 3β-hydroxysteroid dehydrogenase type 1 (3β-HSD1) (P < 0.05). Similarly, treatment of Leydig cells with sucralose and acesulfame-K also increased testosterone level, protein expression of T1R3, 17-α-hydroxylase/17, 20-lyase (CYP17A1), and 3β-HSD1 (P < 0.05). Treatment with SQ22536 (an adenylate cyclas inhibitor) or H89 (a PKA inhibitor) significantly reduced saccharin sodium-induced protein levels of p-SP1, StAR, CYP17A1, and 3β-HSD1 (P < 0.05). In addition, a dual luciferase assay further demonstrated that SP1 significantly increased the promoter activity of CYP17A1 (P < 0.05). When mouse testes were injected with saccharin sodium, T1R3, p-SP1, CYP17A1, and 3β-HSD1 were upregulated, leading to a significant testicular increase in testosterone and cAMP levels (P < 0.05). These results suggest a mechanism by which the taste receptor T1R3 regulates testosterone production, and this mechanism may be linked to the cAMP-PKA pathway. Understanding the interrelationship between T1R3 and the cAMP-PKA-SP1 pathway contributes to clarify the regulatory mechanisms of male reproduction.
Collapse
Affiliation(s)
- Wenjiao Liu
- Key Laboratory of Animal Genetics, Breeding and Reproduction in The Plateau Mountainous Region, Ministry of Education, Guizhou University, Guiyang, 550025, China; Key Laboratory of Animal Genetics, Breeding and Reproduction, Guiyang, 550025, China; College of Animal Science, Guizhou University, Guiyang, 550025, China
| | - Han Wang
- Key Laboratory of Animal Genetics, Breeding and Reproduction in The Plateau Mountainous Region, Ministry of Education, Guizhou University, Guiyang, 550025, China; Key Laboratory of Animal Genetics, Breeding and Reproduction, Guiyang, 550025, China; College of Animal Science, Guizhou University, Guiyang, 550025, China
| | - Qi Mu
- Key Laboratory of Animal Genetics, Breeding and Reproduction in The Plateau Mountainous Region, Ministry of Education, Guizhou University, Guiyang, 550025, China; Key Laboratory of Animal Genetics, Breeding and Reproduction, Guiyang, 550025, China; College of Animal Science, Guizhou University, Guiyang, 550025, China
| | - Ting Gong
- Key Laboratory of Animal Genetics, Breeding and Reproduction in The Plateau Mountainous Region, Ministry of Education, Guizhou University, Guiyang, 550025, China; Key Laboratory of Animal Genetics, Breeding and Reproduction, Guiyang, 550025, China; College of Animal Science, Guizhou University, Guiyang, 550025, China.
| |
Collapse
|
2
|
Mińczuk K, Schlicker E, Krzyżewska A, Malinowska B. Angiotensin 1-7 injected into the rat paraventricular nucleus of hypothalamus increases blood pressure and heart rate via various receptors. Neuropharmacology 2024:110279. [PMID: 39732324 DOI: 10.1016/j.neuropharm.2024.110279] [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: 11/11/2024] [Revised: 12/06/2024] [Accepted: 12/22/2024] [Indexed: 12/30/2024]
Abstract
Although angiotensin 1-7 (Ang 1-7) and its role as a part of the "protective" axis of the renin-angiotensin system are well described in the literature, the mechanisms of its angiotensin II-like pressor and tachycardic effects following its acute central administration are not fully understood. It was the aim of the present study to examine which receptors contribute to the aforementioned cardiovascular effects. Ang 1-7 and antagonists for glutamate, GABA, vasopressin, thromboxane A2 (TP), α1-adrenergic, and P2X purinoceptors or modulators of oxidative stress were injected into the paraventricular nucleus of the hypothalamus (PVN) of urethane-anesthetized male Wistar rats. Acute injection of Ang 1-7 into the PVN increased blood pressure (BP) by about 15 mmHg and heart rate (HR) by about 14 beats/min. After preinjection with bicuculline (GABAA receptor antagonist), CNQX + D-AP5 (AMPA/kainate and NMDA receptor antagonists) and SQ29548 (TP receptor antagonist) the BP and HR reactions to Ang 1-7 were attenuated or abolished. The vasopressin V1A and V1B receptor antagonists conivaptan and nelivaptan, and the NADPH oxidase inhibitor apocynin even reversed the pressor and tachycardic effects of Ang 1-7. Antagonists of P2X (PPADS) and α1-adrenergic receptors (prazosin), the free radical scavenger tempol and the superoxide dismutase inhibitor DETC did not modify the cardiovascular effects of Ang 1-7. The (Mas receptor-related) rise in BP and HR evoked by Ang 1-7 administered to the rat PVN is linked to glutamate, vasopressin, GABAA and thromboxane receptors, and to oxidative stress, but does not seem to involve α1-adrenergic or P2X receptors.
Collapse
Affiliation(s)
- K Mińczuk
- Department of Experimental Physiology and Pathophysiology, Medical University of Białystok, ul. Mickiewicza 2A, 15-222 Białystok, Poland.
| | - E Schlicker
- Department of Pharmacology and Toxicology, University of Bonn, Venusberg-Campus 1, 53127 Bonn, Germany
| | - A Krzyżewska
- Department of Experimental Physiology and Pathophysiology, Medical University of Białystok, ul. Mickiewicza 2A, 15-222 Białystok, Poland
| | - B Malinowska
- Department of Experimental Physiology and Pathophysiology, Medical University of Białystok, ul. Mickiewicza 2A, 15-222 Białystok, Poland
| |
Collapse
|
3
|
Xue Q, Liu C, Zhang D, Li M, Li Y. α-Mangostin Attenuates Blood Pressure and Reverses Vascular Remodeling by Balancing ACE/AT1R and ACE2/Ang-(1-7)/MasR Axes in Ang II-Infused Hypertensive Mice. Phytother Res 2024; 38:5918-5929. [PMID: 39410864 PMCID: PMC11634819 DOI: 10.1002/ptr.8353] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Revised: 08/20/2024] [Accepted: 09/21/2024] [Indexed: 12/13/2024]
Abstract
Hyperuricemia is a common comorbidity of hypertension and probably has a causal relationship with hypertension. Alpha-mangostin (α-MG) has been reported to have uric acid lowering effect. This study aimed to investigate the dual effects of α-MG on blood pressure (BP) and uric acid levels in angiotensin II (Ang II)-infused hypertensive mice. Male C57BL/6 mice were randomized into five groups: control, Ang II infusion (500 ng/kg/min for 2 weeks), Ang II infusion with gavage administration of α-MG 4.0 and 8.0 mg/kg and benzbromarone (25 mg/kg) respectively. BP, uric acid levels, vascular structure and function, and renin-Ang II system expressions in the aorta were assessed. Treatment with α-MG reduced BP, improved endothelial relaxation, and reversed aortic wall thickening and collagen deposition in Ang II-induced hypertensive mice. It also downregulated Ang II receptor 1 (AT1R) and angiotensin converting enzyme (ACE) expression, while upregulating ACE2, Mas receptor (MasR), and angiotensin (1-7) in the aorta. Moreover, α-MG demonstrated a significant enhancement in uric acid clearance and reduction in serum uric acid levels. Conversely, benzbromarone did not result in a decrease in BP, indicating that the hypotensive effect of α-MG may not be necessarily dependent on its urate-lowering properties. α-MG can attenuate Ang II-induced hypertension and reverse vascular remodeling, potentially by balancing the ACE/Ang II/AT1R axis and the ACE2/Ang-(1-7)/MasR axis. Our findings provide insights into α-MG as a novel anti-hypertensive drug especially in patients with hyperuricemia.
Collapse
Affiliation(s)
- Qi‐Qi Xue
- Department of Cardiovascular Medicine, Shanghai Institute of Hypertension, Shanghai Key Laboratory of Hypertension, National Research Centre for Translational Medicine, Ruijin HospitalShanghai Jiatong University School of MedicineShanghaiChina
| | - Chu‐Hao Liu
- Department of Cardiovascular Medicine, Shanghai Institute of Hypertension, Shanghai Key Laboratory of Hypertension, National Research Centre for Translational Medicine, Ruijin HospitalShanghai Jiatong University School of MedicineShanghaiChina
| | - Dong‐Yan Zhang
- Department of Cardiovascular Medicine, Shanghai Institute of Hypertension, Shanghai Key Laboratory of Hypertension, National Research Centre for Translational Medicine, Ruijin HospitalShanghai Jiatong University School of MedicineShanghaiChina
| | - Ming‐Xuan Li
- Department of Cardiovascular Medicine, Shanghai Institute of Hypertension, Shanghai Key Laboratory of Hypertension, National Research Centre for Translational Medicine, Ruijin HospitalShanghai Jiatong University School of MedicineShanghaiChina
| | - Yan Li
- Department of Cardiovascular Medicine, Shanghai Institute of Hypertension, Shanghai Key Laboratory of Hypertension, National Research Centre for Translational Medicine, Ruijin HospitalShanghai Jiatong University School of MedicineShanghaiChina
| |
Collapse
|
4
|
Frame AA, Nist KM, Kim K, Puleo F, Moreira JD, Swaldi H, McKenna J, Wainford RD. Integrated renal and sympathetic mechanisms underlying the development of sex- and age-dependent hypertension and the salt sensitivity of blood pressure. GeroScience 2024; 46:6435-6458. [PMID: 38976131 PMCID: PMC11494650 DOI: 10.1007/s11357-024-01266-1] [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: 05/13/2024] [Accepted: 06/25/2024] [Indexed: 07/09/2024] Open
Abstract
Aging is a non-modifiable understudied risk factor for hypertension. We hypothesized that sympathetically mediated activation of renal sodium reabsorption drives age-dependent hypertension and the salt sensitivity of blood pressure (BP). Using 3-, 8-, and 16-month-old male and female Sprague-Dawley rats as a model of normal aging, we assessed BP, indices of sympathetic tone, and the physiological responses to acute and chronic sodium challenge including sodium chloride cotransporter (NCC) regulation. The effects of renal nerve ablation and NCC antagonism were assessed in hypertensive male rats. We observed sex-dependent impaired renal sodium handling (24 h sodium balance (meq), male 3-month 0.36 ± 0.1 vs. 16-month 0.84 ± 0.2; sodium load excreted during 5% bodyweight isotonic saline volume expansion (%) male 3-month 77 ± 5 vs. 16-month 22 ± 8), hypertension (MAP (mmHg) male 3-month 123 ± 4 vs. 16-month 148 ± 6), and the salt sensitivity of BP in aged male, but not female, rats. Attenuated sympathoinhibitory afferent renal nerve (ARN) responses contributed to increased sympathetic tone and hypertension in male rats. Increased sympathetic tone contributes to renal sodium retention, in part through increased NCC activity via a dysfunctional with-no-lysine kinase-(WNK) STE20/SPS1-related proline/alanine-rich kinase signaling pathway, to drive hypertension and the salt sensitivity of BP in aged male rats. NCC antagonism and renal nerve ablation, which reduced WNK dysfunction and decreased NCC activity, attenuated age-dependent hypertension in male Sprague-Dawley rats. The contribution of an impaired sympathoinhibitory ARN reflex to sex- and age-dependent hypertension in an NCC-dependent manner, via an impaired WNK1/WNK4 dynamic, suggests this pathway as a mechanism-based target for the treatment of age-dependent hypertension.
Collapse
Affiliation(s)
- Alissa A Frame
- Department of Pharmacology & Experimental Therapeutics and the Whitaker Cardiovascular Institute, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Kayla M Nist
- Department of Anatomy & Neurobiology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Kiyoung Kim
- Department of Pharmacology & Experimental Therapeutics and the Whitaker Cardiovascular Institute, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Franco Puleo
- Department of Pharmacology & Experimental Therapeutics and the Whitaker Cardiovascular Institute, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Jesse D Moreira
- Department of Health Sciences, Sargent College, Boston University, Boston, MA, USA
| | - Hailey Swaldi
- Division of Cardiology, Emory University School of Medicine, 1750 Haygood Drive, Atlanta, GA, N22030322, USA
| | - James McKenna
- Division of Cardiology, Emory University School of Medicine, 1750 Haygood Drive, Atlanta, GA, N22030322, USA
| | - Richard D Wainford
- Department of Pharmacology & Experimental Therapeutics and the Whitaker Cardiovascular Institute, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA.
- Division of Cardiology, Emory University School of Medicine, 1750 Haygood Drive, Atlanta, GA, N22030322, USA.
| |
Collapse
|
5
|
Irigoyen MC, Fetter C, De Angelis K. Advances on the Experimental Research in Resistant Hypertension. Curr Hypertens Rep 2024; 26:475-482. [PMID: 39023702 DOI: 10.1007/s11906-024-01315-2] [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] [Accepted: 06/24/2024] [Indexed: 07/20/2024]
Abstract
PURPOSE OF REVIEW Resistant Hypertension (RH) poses a significant public health challenge, contributing to increased mortality, cardiovascular events and organ damage. Both clinical and experimental research are striving for higher standards in a translational manner to integrate new findings and confirm hypotheses. Considering that many are the aspects of RH that are still under investigation, this review aims to shed light on the advances made in experimental research concerning RH. It seeks to underscore the pivotal role of experimental studies in shaping clinical practices and also explore future perspectives. RECENT FINDINGS It is important to emphasize the significance of experimental models, primarily for advancing our understanding: experimental models have greatly contributed to our comprehension of the underlying mechanisms in RH, including factors like sympathetic activation, endothelial dysfunction and structural vessel abnormalities. Secondly, for assessing treatment approaches: animal models have also played a crucial role in evaluating the potential effectiveness of diverse treatment approaches for RH. These encompass both pharmacological options, involving combinations of established drugs or novel pharmaceuticals, and non-pharmacological alternatives, which include surgical procedures like renal denervation, medical devices like baroreceptor stimulators, and lifestyle modifications. The most lacking component in translational research is the fact that there is no well-established animal model that perfectly replicates RH. Consequently, alternative strategies, including the combination of models, must be considered. What remains clear is that the development of animal models closely mimicking RH holds the promise of providing valuable insights into the essential mechanisms and responses necessary to combat or slow the global progression of RH.
Collapse
Affiliation(s)
- Maria Claudia Irigoyen
- Hypertension Unit, Heart Institute (InCor), Sao Paulo University Medical School, Sao Paulo, Brazil.
| | - Claudia Fetter
- Laboratory of Clinical Investigation (LIC), Institute of Cardiology of Rio Grande do Sul/ Cardiology University Foundation (IC- FUC), Porto Alegre, Brazil
| | - Kátia De Angelis
- Department of Physiology, Federal University of São Paulo (UNIFESP), Universidade Nove de Julho (UNINOVE), Sao Paulo, Brazil
| |
Collapse
|
6
|
Xue QQ, Liu CH, Li Y. Decoding the anti-hypertensive mechanism of α-mangostin based on network pharmacology, molecular docking and experimental validation. Mol Med 2024; 30:234. [PMID: 39592923 PMCID: PMC11600633 DOI: 10.1186/s10020-024-01001-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Accepted: 11/14/2024] [Indexed: 11/28/2024] Open
Abstract
BACKGROUND Hypertension is a leading risk factor for disability and deaths worldwide. Evidence indicates that alpha-mangostin(α-MG) can reduce blood pressure and improve target organ damage. Nonetheless, its pharmacological targets and potential mechanisms of action remain inadequately elucidated. METHOD We used SwissTargetPrediction to identify α-MG's drug targets and DisGeNET, GeneCards, CTD, and GEO databases for hypertension-related targets, and then determined antihypertensive therapeutic targets of α-MG by intersecting these targets. GO functional enrichment analysis, KEGG pathway analysis, and disease association analysis were conducted using the DAVID database and R package "clusterprofile", visualized with Cytoscape software. The binding affinity of α-MG to identified targets was confirmed through molecular docking using Autodock Vina v.1.2.2 software. The impact of α-MG on target genes was validated using an Angiotensin II-induced hypertensive mouse model and RT-qPCR. RESULTS A total of 51 potential antihypertensive therapeutic targets for α-MG were identified by intersecting 109 drug targets with 821 disease targets. Furthermore, 10 cellular component terms, 10 disease terms, and the top 20 enriched biological processes, molecular functions, and KEGG pathways related to α-MG's antihypertensive effects were documented. Molecular docking studies indicated a strong binding affinity of α-MG with the HSP90AA1 domain. In Ang II-induced hypertensive mice aorta, treatment with α-MG effectively reversed the aberrant mRNA expression of TNF, HSP90AA1, NFKB1, PPARG, SIRT1, PTGS2, and RELA. CONCLUSION Our analyses showed that TNF, HSP90AA1, NFKB1, PPARG, SIRT1, PTGS2, and RELA might be α-MG's potential therapeutic targets for hypertension, laying groundwork for further investigation into its pharmacological mechanisms and clinical uses.
Collapse
Affiliation(s)
- Qi-Qi Xue
- Department of Cardiovascular Medicine, Shanghai Key Laboratory of Hypertension, Shanghai Institute of Hypertension, National Research Centre for Translational Medicine, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Ruijin 2nd Rd 197, Shanghai, 200025, China
| | - Chu-Hao Liu
- Department of Cardiovascular Medicine, Shanghai Key Laboratory of Hypertension, Shanghai Institute of Hypertension, National Research Centre for Translational Medicine, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Ruijin 2nd Rd 197, Shanghai, 200025, China
| | - Yan Li
- Department of Cardiovascular Medicine, Shanghai Key Laboratory of Hypertension, Shanghai Institute of Hypertension, National Research Centre for Translational Medicine, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Ruijin 2nd Rd 197, Shanghai, 200025, China.
| |
Collapse
|
7
|
Tiwari A, De A, Sinha A. Malaria and Hypertension: What Is the Direction of Association? FUNCTION 2024; 5:zqae037. [PMID: 39215464 DOI: 10.1093/function/zqae037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Revised: 08/07/2024] [Accepted: 08/21/2024] [Indexed: 09/04/2024] Open
Affiliation(s)
- Aparna Tiwari
- ICMR-National Institute of Malaria Research, New Delhi 110077, India
| | - Auley De
- ICMR-National Institute of Malaria Research, New Delhi 110077, India
| | - Abhinav Sinha
- ICMR-National Institute of Malaria Research, New Delhi 110077, India
- Academy of Scientific and Innovative Research, Ghaziabad 201002, India
| |
Collapse
|
8
|
Daghbouche-Rubio N, Álvarez-Miguel I, Flores VA, Rojo-Mencía J, Navedo M, Nieves-Citrón M, Cidad P, Pérez-García MT, López-López JR. The P2Y6 Receptor as a Potential Keystone in Essential Hypertension. FUNCTION 2024; 5:zqae045. [PMID: 39322240 DOI: 10.1093/function/zqae045] [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: 07/25/2024] [Revised: 09/23/2024] [Accepted: 09/23/2024] [Indexed: 09/27/2024] Open
Abstract
Essential hypertension (HT) is a highly prevalent cardiovascular disease of unclear physiopathology. Pharmacological studies suggest that purinergic P2Y6 receptors (P2ry6) play important roles in cardiovascular function and may contribute to angiotensin II (AgtII) pathophysiological effects. Here, we tested the hypothesis that functional coupling between P2ry6 and AgtII receptors mediates altered vascular reactivity in HT. For this, a multipronged approach was implemented using mesenteric vascular smooth muscle cells (VSMCs) and arteries from Blood Pressure Normal (BPN) and Blood Pressure High (BPH) mice. Differential transcriptome profiling of mesenteric artery VSMCs identified P2ry6 purinergic receptor mRNA as one of the top upregulated transcripts in BPH. P2Y receptor activation elicited distinct vascular responses in mesenteric arteries from BPN and BPH mice. Accordingly, 10 µm UTP produced a contraction close to half-maximal activation in BPH arteries but no response in BPN vessels. AgtII-induced contraction was also higher in BPH mice despite having lower AgtII receptor type-1 (Agtr1) expression and was sensitive to P2ry6 modulators. Proximity ligation assay and super-resolution microscopy showed closer localization of Agtr1 and P2ry6 at/near the membrane of BPH mice. This proximal association was reduced in BPN mice, suggesting a functional role for Agtr1-P2ry6 complexes in the hypertensive phenotype. Intriguingly, BPN mice were resistant to AgtII-induced HT and showed reduced P2ry6 expression in VSMCs. Altogether, results suggest that increased functional coupling between P2ry6 and Agtr1 may contribute to enhanced vascular reactivity during HT. In this regard, blocking P2ry6 could be a potential pharmacological strategy to treat HT.
Collapse
MESH Headings
- Animals
- Essential Hypertension/metabolism
- Essential Hypertension/genetics
- Mice
- Receptors, Purinergic P2/metabolism
- Receptors, Purinergic P2/genetics
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Muscle, Smooth, Vascular/drug effects
- Male
- Mesenteric Arteries/metabolism
- Mesenteric Arteries/drug effects
- Mesenteric Arteries/physiopathology
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/drug effects
- Mice, Inbred C57BL
- Angiotensin II/pharmacology
- Blood Pressure/genetics
Collapse
Affiliation(s)
- Nuria Daghbouche-Rubio
- Departamento de Bioquímica y Biología Molecular y Fisiología e Instituto de Biología y Genética Molecular (IBGM), Universidad de Valladolid y Consejo Superior de Investigaciones Científicas (CSIC), Valladolid, 47003, Spain
| | - Inés Álvarez-Miguel
- Departamento de Bioquímica y Biología Molecular y Fisiología e Instituto de Biología y Genética Molecular (IBGM), Universidad de Valladolid y Consejo Superior de Investigaciones Científicas (CSIC), Valladolid, 47003, Spain
| | | | - Jorge Rojo-Mencía
- Departamento de Bioquímica y Biología Molecular y Fisiología e Instituto de Biología y Genética Molecular (IBGM), Universidad de Valladolid y Consejo Superior de Investigaciones Científicas (CSIC), Valladolid, 47003, Spain
| | - Manuel Navedo
- Department of Pharmacology, University of California Davis, Davis, CA 95616, USA
| | | | - Pilar Cidad
- Departamento de Bioquímica y Biología Molecular y Fisiología e Instituto de Biología y Genética Molecular (IBGM), Universidad de Valladolid y Consejo Superior de Investigaciones Científicas (CSIC), Valladolid, 47003, Spain
| | - M Teresa Pérez-García
- Departamento de Bioquímica y Biología Molecular y Fisiología e Instituto de Biología y Genética Molecular (IBGM), Universidad de Valladolid y Consejo Superior de Investigaciones Científicas (CSIC), Valladolid, 47003, Spain
| | - José R López-López
- Departamento de Bioquímica y Biología Molecular y Fisiología e Instituto de Biología y Genética Molecular (IBGM), Universidad de Valladolid y Consejo Superior de Investigaciones Científicas (CSIC), Valladolid, 47003, Spain
| |
Collapse
|
9
|
Zhang Y, Arzaghi H, Ma Z, Roye Y, Musah S. Epigenetics of Hypertensive Nephropathy. Biomedicines 2024; 12:2622. [PMID: 39595187 PMCID: PMC11591919 DOI: 10.3390/biomedicines12112622] [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: 10/08/2024] [Revised: 11/05/2024] [Accepted: 11/13/2024] [Indexed: 11/28/2024] Open
Abstract
Hypertensive nephropathy (HN) is a leading cause of chronic kidney disease (CKD) and end-stage renal disease (ESRD), contributing to significant morbidity, mortality, and rising healthcare costs. In this review article, we explore the role of epigenetic mechanisms in HN progression and their potential therapeutic implications. We begin by examining key epigenetic modifications-DNA methylation, histone modifications, and non-coding RNAs-observed in kidney disease. Next, we discuss the underlying pathophysiology of HN and highlight current in vitro and in vivo models used to study the condition. Finally, we compare various types of HN-induced renal injury and their associated epigenetic mechanisms with those observed in other kidney injury models, drawing inferences on potential epigenetic therapies for HN. The information gathered in this work indicate that epigenetic mechanisms can drive the progression of HN by regulating key molecular signaling pathways involved in renal damage and fibrosis. The limitations of Renin-Angiotensin-Aldosterone System (RAAS) inhibitors underscore the need for alternative treatments targeting epigenetic pathways. This review emphasizes the importance of further research into the epigenetic regulation of HN to develop more effective therapies and preventive strategies. Identifying novel epigenetic markers could provide new therapeutic opportunities for managing CKD and reducing the burden of ESRD.
Collapse
Affiliation(s)
- Yize Zhang
- Department of Biomedical Engineering, Pratt School of Engineering, Duke University, Durham, NC 27708, USA
| | - Hamidreza Arzaghi
- Department of Biomedical Engineering, Pratt School of Engineering, Duke University, Durham, NC 27708, USA
| | - Zhehan Ma
- Department of Biomedical Engineering, Pratt School of Engineering, Duke University, Durham, NC 27708, USA
| | - Yasmin Roye
- Department of Biomedical Engineering, Pratt School of Engineering, Duke University, Durham, NC 27708, USA
| | - Samira Musah
- Department of Biomedical Engineering, Pratt School of Engineering, Duke University, Durham, NC 27708, USA
- Center for Biomolecular and Tissue Engineering, Duke University, Durham, NC 27708, USA
- Division of Nephrology, Department of Medicine, Duke University School of Medicine, Durham, NC 27710, USA
- Department of Cell Biology, Duke University, Durham, NC 27710, USA
- Affiliate Faculty of the Developmental and Stem Cell Biology Program, Duke Regeneration Center, and Duke MEDx Initiative, Duke University, Durham, NC 27710, USA
| |
Collapse
|
10
|
Anilkumar S A, Dutta S, Aboo S, Ismail A. Vitamin D as a modulator of molecular pathways involved in CVDs: Evidence from preclinical studies. Life Sci 2024; 357:123062. [PMID: 39288869 DOI: 10.1016/j.lfs.2024.123062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Revised: 08/27/2024] [Accepted: 09/12/2024] [Indexed: 09/19/2024]
Abstract
Vitamin D deficiency (VDD) is a widespread global health issue, affecting nearly a billion individuals worldwide, and mounting evidence links it to an increased risk of cardiovascular diseases like hypertension, atherosclerosis, and heart failure. The discovery of vitamin D receptors and metabolizing enzymes in cardiac and vascular cells, coupled with experimental studies, underscores the complex relationship between vitamin D and cardiovascular health. This review aims to synthesize and critically evaluate the preclinical evidence elucidating the role of vitamin D in cardiovascular health. We examined diverse preclinical in vitro (cardiomyocyte cell line) models and in vivo models, including knockout mice, diet-induced deficiency, and disease-specific animal models (hypertension, hypertrophy and myocardial infarction). These studies reveal that vitamin D modulates vascular tone, and prevents fibrosis and hypertrophy through effects on major signal transduction pathways (NF-kB, Nrf2, PI3K/AKT/mTOR, Calcineurin/NFAT, TGF-β/Smad, AMPK) and influences epigenetic mechanisms governing inflammation, oxidative stress, and pathological remodeling. In vitro studies elucidate vitamin D's capacity to promote cardiomyocyte differentiation and inhibit pathological remodeling. In vivo studies further uncovered detrimental cardiac effects of VDD, while supplementation with vitamin D in cardiovascular disease (CVD) models demonstrated its protective effects by decreasing inflammation, attenuating hypertrophy, reduction in plaque formation, and improving cardiac function. Hence, this comprehensive review emphasizes the critical role of vitamin D in cardiovascular health and its potential as a preventive/therapeutic strategy in CVDs. However, further research is needed to translate these findings into clinical applications as there are discrepancies between preclinical and clinical studies.
Collapse
Affiliation(s)
- Athira Anilkumar S
- Department of Endocrinology, ICMR-National Institute of Nutrition, Hyderabad, India
| | - Soumam Dutta
- Department of Endocrinology, ICMR-National Institute of Nutrition, Hyderabad, India
| | - Shabna Aboo
- Department of Endocrinology, ICMR-National Institute of Nutrition, Hyderabad, India.
| | - Ayesha Ismail
- Department of Endocrinology, ICMR-National Institute of Nutrition, Hyderabad, India.
| |
Collapse
|
11
|
Castagna A, Mango G, Martinelli N, Marzano L, Moruzzi S, Friso S, Pizzolo F. Sodium Chloride Cotransporter in Hypertension. Biomedicines 2024; 12:2580. [PMID: 39595146 PMCID: PMC11591633 DOI: 10.3390/biomedicines12112580] [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: 10/07/2024] [Revised: 10/29/2024] [Accepted: 11/08/2024] [Indexed: 11/28/2024] Open
Abstract
The sodium chloride cotransporter (NCC) is essential for electrolyte balance, blood pressure regulation, and pathophysiology of hypertension as it mediates the reabsorption of ultrafiltered sodium in the renal distal convoluted tubule. Given its pivotal role in the maintenance of extracellular fluid volume, the NCC is regulated by a complex network of cellular pathways, which eventually results in either its phosphorylation, enhancing sodium and chloride ion absorption from urines, or dephosphorylation and ubiquitination, which conversely decrease NCC activity. Several factors could influence NCC function, including genetic alterations, hormonal stimuli, and pharmacological treatments. The NCC's central role is also highlighted by several abnormalities resulting from genetic mutations in its gene and consequently in its structure, leading to dysregulation of blood pressure control. In the last decade, among other improvements, the acquisition of knowledge on the NCC and other renal ion channels has been favored by studies on extracellular vesicles (EVs). Dietary sodium and potassium intake are also implicated in the tuning of NCC activity. In this narrative review, we present the main cornerstones and recent evidence related to NCC control, focusing on the context of blood pressure pathophysiology, and promising new therapeutical approaches.
Collapse
Affiliation(s)
- Annalisa Castagna
- Department of Medicine, University of Verona, 37134 Verona, Italy; (A.C.); (G.M.); (N.M.); (S.F.)
| | - Gabriele Mango
- Department of Medicine, University of Verona, 37134 Verona, Italy; (A.C.); (G.M.); (N.M.); (S.F.)
| | - Nicola Martinelli
- Department of Medicine, University of Verona, 37134 Verona, Italy; (A.C.); (G.M.); (N.M.); (S.F.)
| | - Luigi Marzano
- Unit of Internal Medicine B, Department of Medicine, University of Verona School of Medicine, Azienda Ospedaliera Universitaria Integrata Verona, Policlinico “G.B. Rossi”, 37134 Verona, Italy; (L.M.); (S.M.)
| | - Sara Moruzzi
- Unit of Internal Medicine B, Department of Medicine, University of Verona School of Medicine, Azienda Ospedaliera Universitaria Integrata Verona, Policlinico “G.B. Rossi”, 37134 Verona, Italy; (L.M.); (S.M.)
| | - Simonetta Friso
- Department of Medicine, University of Verona, 37134 Verona, Italy; (A.C.); (G.M.); (N.M.); (S.F.)
| | - Francesca Pizzolo
- Department of Medicine, University of Verona, 37134 Verona, Italy; (A.C.); (G.M.); (N.M.); (S.F.)
| |
Collapse
|
12
|
Cseh D, Middlemiss JE, Mäki-Petäjä KM, Hubsch A, Wilkinson IB, McEniery CM. Cardiovascular responses to experimental weight gain in humans: a feasibility study. J Hypertens 2024; 42:2011-2017. [PMID: 39119814 PMCID: PMC11451930 DOI: 10.1097/hjh.0000000000003830] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Revised: 07/01/2024] [Accepted: 07/21/2024] [Indexed: 08/10/2024]
Abstract
OBJECTIVE Obesity and hypertension share a well known association. However, the mechanisms underlying their relationship are not well understood. Our goal was to assess the feasibility of a longitudinal, interventional weight gain study with detailed cardiovascular measurements in humans. METHODS Sixteen healthy, normotensive, young, male volunteers (28 ± 7 years) were enrolled. Body composition, biochemical and cardiovascular data were obtained at baseline, and after an 8-week period of overfeeding (800-1000 kcal/day). Blood pressure (BP), cardiac output (CO) and peripheral vascular resistance (PVR) were determined, as were the minimum forearm vascular resistance (MFVR), forearm blood flow (FBF) response to mental stress and heart rate variability (HRV) parameters. RESULTS Overfeeding resulted in a median weight gain of 5.6 kg [interquartile range (IQR) 4.6-6.4 kg; P < 0.001]. Seated systolic and diastolic BP were significantly increased by 10 ± 9 and 4 ± 6 mmHg, respectively, after weight gain ( P < 0.001 and P = 0.011, respectively). CO also increased and PVR decreased significantly as a result of weight gain ( P = 0.032 and P = 0.044, respectively). MFVR was also significantly decreased after weight gain ( P = 0.023). The FBF response to mental stress was blunted significantly ( P = 0.002), and sympathovagal balance and responsiveness to orthostatic challenge altered moderately after weight gain. CONCLUSION Our overfeeding regimen resulted in moderate weight gain and significant increases in BP. An increase in CO is likely to be the dominant mechanism underlying the observed BP changes, with decreases in PVR partially compensating for these effects. Experimental weight gain, coupled with detailed cardiovascular phenotyping, is a feasible model to examine potential mechanisms underlying obesity-associated hypertension in young adults.
Collapse
|
13
|
Souza ATP, Freitas GP, Lopes HB, Weffort D, Adolpho LF, Gomes MPO, Oliveira FS, Almeida ALG, Beloti MM, Rosa AL. Efficacy of mesenchymal stem cell-based therapy on the bone repair of hypertensive rats. Oral Dis 2024; 30:5118-5128. [PMID: 38764359 DOI: 10.1111/odi.15004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Revised: 04/14/2024] [Accepted: 05/07/2024] [Indexed: 05/21/2024]
Abstract
OBJECTIVE Hypertension disrupts the bone integrity and its repair ability. This study explores the efficiency of a therapy based on the application of mesenchymal stem cells (MSCs) to repair bone defects of spontaneously hypertensive rats (SHR). METHODS First, we evaluated SHR in terms of bone morphometry and differentiation of MSCs into osteoblasts. Then, the effects of the interactions between MSCs from normotensive rats (NTR-MSCs) cocultured with SHR (SHR-MSCs) on the osteoblast differentiation of both cell populations were evaluated. Also, bone formation into calvarial defects of SHR treated with NTR-MSCs was analyzed. RESULTS Hypertension induced bone loss evidenced by reduced bone morphometric parameters of femurs of SHR compared with NTR as well as decreased osteoblast differentiation of SHR-MSCs compared with NTR-MSCs. NTR-MSCs partially restored the capacity of SHR-MSCs to differentiate into osteoblasts, while SHR-MSCs exhibited a slight negative effect on NTR-MSCs. An enhanced bone repair was observed in defects treated with NTR-MSCs compared with control, stressing this cell therapy efficacy even in bones damaged by hypertension. CONCLUSION The use of MSCs derived from a heathy environment can be in the near future a smart approach to treat bone loss in the context of regenerative dentistry for oral rehabilitation of hypertensive patients.
Collapse
Affiliation(s)
- Alann Thaffarell Portilho Souza
- Bone Research Lab, Ribeirão Preto School of Dentistry, University of São Paulo, Ribeirão Preto, Brazil
- School of Dentistry, Metropolitan University Center of the Amazon (UNIFAMAZ), Belém, Brazil
| | - Gileade Pereira Freitas
- Bone Research Lab, Ribeirão Preto School of Dentistry, University of São Paulo, Ribeirão Preto, Brazil
- School of Dentistry, Federal University of Goiás, Goiânia, Brazil
| | - Helena Bacha Lopes
- Bone Research Lab, Ribeirão Preto School of Dentistry, University of São Paulo, Ribeirão Preto, Brazil
| | - Denise Weffort
- Bone Research Lab, Ribeirão Preto School of Dentistry, University of São Paulo, Ribeirão Preto, Brazil
| | - Leticia Faustino Adolpho
- Bone Research Lab, Ribeirão Preto School of Dentistry, University of São Paulo, Ribeirão Preto, Brazil
| | | | | | | | - Marcio Mateus Beloti
- Bone Research Lab, Ribeirão Preto School of Dentistry, University of São Paulo, Ribeirão Preto, Brazil
| | - Adalberto Luiz Rosa
- Bone Research Lab, Ribeirão Preto School of Dentistry, University of São Paulo, Ribeirão Preto, Brazil
| |
Collapse
|
14
|
Byun JW, Paeng JC, Kim YJ, Lee SP, Lee YS, Choi H, Kang KW, Cheon GJ. Evaluation of Fibroblast Activation Protein Expression Using 68Ga-FAPI46 PET in Hypertension-Induced Tissue Changes. J Nucl Med 2024; 65:1776-1781. [PMID: 39327013 DOI: 10.2967/jnumed.124.267489] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Accepted: 09/05/2024] [Indexed: 09/28/2024] Open
Abstract
Chronic hypertension leads to injury and fibrosis in major organs. Fibroblast activation protein (FAP) is one of key molecules in tissue fibrosis, and 68Ga-labeled FAP inhibitor-46 (FAPI46) PET is a recently developed method for evaluating FAP. The aim of this study was to evaluate FAP expression and fibrosis in a hypertension model and to test the feasibility of 68Ga-FAPI46 PET in hypertension. Methods: Hypertension was induced in mice by angiotensin II infusion for 4 wk. 68Ga-FAPI46 biodistribution studies and PET scanning were conducted at 1, 2, and 4 wk after hypertension modeling, and uptake in the major organs was measured. The FAP expression and fibrosis formation of the heart and kidney tissues were analyzed and compared with 68Ga-FAPI46 uptake. Subgroups of the hypertension model underwent angiotensin receptor blocker administration and high-dose FAPI46 blocking, for comparison. As a preliminary human study, 68Ga-FAPI46 PET images of lung cancer patients were analyzed and compared between hypertension and control groups. Results: Uptake of 68Ga-FAPI46 in the heart and kidneys was significantly higher in the hypertension group than in the sham group as early as week 1 and decreased after week 2. The uptake was specifically blocked in the high-dose blocking study. Immunohistochemistry also revealed FAP expression in both heart and kidney tissues. However, overt fibrosis was observed in the heart, whereas it was absent from the kidneys. The angiotensin receptor blocker-treated group showed lower uptake in the heart and kidneys than did the hypertension group. In the pilot human study, renal uptake of 68Ga-FAPI46 significantly differed between the hypertension and control groups. Conclusion: In hypertension, FAP expression is increased in the heart and kidneys from the early phases and decreases over time. FAP expression appears to represent fibrosis activity preceding or underlying fibrotic tissue formation. 68Ga-FAPI46 PET has potential as an effective imaging method for evaluating FAP expression in progressive fibrosis by hypertension.
Collapse
Affiliation(s)
- Jung Woo Byun
- Department of Nuclear Medicine, Seoul National University College of Medicine, Seoul, Republic of Korea
- Department of Nuclear Medicine, Seoul National University Hospital, Seoul, Republic of Korea
- Department of Biomedical Sciences, Seoul National University Graduate School, Seoul, Republic of Korea
- Cancer Research Institute, Seoul National University, Seoul, Republic of Korea
| | - Jin Chul Paeng
- Department of Nuclear Medicine, Seoul National University College of Medicine, Seoul, Republic of Korea;
- Department of Nuclear Medicine, Seoul National University Hospital, Seoul, Republic of Korea
- Institute of Radiation Medicine, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Young Ju Kim
- Department of Nuclear Medicine, Seoul National University College of Medicine, Seoul, Republic of Korea
- Cancer Research Institute, Seoul National University, Seoul, Republic of Korea
| | - Seung-Pyo Lee
- Division of Cardiology, Department of Internal Medicine, Seoul National University Hospital, Seoul, Republic of Korea; and
| | - Yun-Sang Lee
- Department of Nuclear Medicine, Seoul National University College of Medicine, Seoul, Republic of Korea
- Department of Nuclear Medicine, Seoul National University Hospital, Seoul, Republic of Korea
- Institute of Radiation Medicine, Seoul National University College of Medicine, Seoul, Republic of Korea
- Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, Seoul National University, Seoul, Republic of Korea
| | - Hongyoon Choi
- Department of Nuclear Medicine, Seoul National University College of Medicine, Seoul, Republic of Korea
- Department of Nuclear Medicine, Seoul National University Hospital, Seoul, Republic of Korea
- Institute of Radiation Medicine, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Keon Wook Kang
- Department of Nuclear Medicine, Seoul National University College of Medicine, Seoul, Republic of Korea
- Department of Nuclear Medicine, Seoul National University Hospital, Seoul, Republic of Korea
- Department of Biomedical Sciences, Seoul National University Graduate School, Seoul, Republic of Korea
- Cancer Research Institute, Seoul National University, Seoul, Republic of Korea
| | - Gi Jeong Cheon
- Department of Nuclear Medicine, Seoul National University College of Medicine, Seoul, Republic of Korea
- Department of Nuclear Medicine, Seoul National University Hospital, Seoul, Republic of Korea
- Institute of Radiation Medicine, Seoul National University College of Medicine, Seoul, Republic of Korea
- Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, Seoul National University, Seoul, Republic of Korea
| |
Collapse
|
15
|
Wojtacha P, Bogdańska-Chomczyk E, Majewski MK, Obremski K, Majewski MS, Kozłowska A. Renal Inflammation, Oxidative Stress, and Metabolic Abnormalities During the Initial Stages of Hypertension in Spontaneously Hypertensive Rats. Cells 2024; 13:1771. [PMID: 39513878 PMCID: PMC11545559 DOI: 10.3390/cells13211771] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Revised: 10/03/2024] [Accepted: 10/17/2024] [Indexed: 11/16/2024] Open
Abstract
Background: Hypertension is a major cause of mortality worldwide. The kidneys play a crucial role in regulating blood pressure and fluid volume. The relationship between the kidneys and hypertension is complex, involving factors such as the renin-angiotensin system, oxidative stress, and inflammation. This study aims to assess the levels of inflammatory markers, oxidative stress, and metabolic factors in the kidneys, focusing on their potential role in early renal damage and their association with the development of hypertension. Methods: This study was designed to compare the levels of selected inflammatory markers, e.g., interleukins, tumor necrosis factor-α (TNF-α), transforming growth factor, and serine/threonine-protein (mTOR); oxidative stress markers such as malondialdehyde, sulfhydryl group, and glucose (GLC); and metabolic markers among other enzymes, such as alanine transaminase (ALT), aspartate transaminase (AST), hexokinase II (HK-II), and hypoxia-inducible factor-1α (HIF-1α), as well as creatinine in the kidneys of spontaneously hypertensive rats (SHR/NCrl, n = 12) and Wistar Kyoto rats (WKY/NCrl, n = 12). Both juvenile (5 weeks old) and maturing (10 weeks old) specimens were examined using spectrophotometric methods, e.g., ELISA. Results: Juvenile SHRs exhibited reduced renal levels of all studied cytokines and chemokines, with lower oxidative stress and deficits in the mTOR and HK-II levels compared to the age-matched WKYs. Maturing SHRs showed increased renal levels of interleukin-1β (IL-1β), IL-6, IL-18, and TNF-α, alongside elevated carbonyl stress and increased HIF-1α as opposed to their control peers. The levels of all other studied markers were normalized in these animals, except for ALT (increased), ALP, and GLC (both reduced). Conclusions: This study underscores the significant impact of inflammatory, oxidative stress, and metabolic marker changes on renal function. Juvenile SHRs display lower marker levels, indicating an immature immune response and potential subclinical kidney damage that may contribute to hypertension development. In contrast, mature SHRs exhibit chronic inflammation, oxidative dysregulation, and metabolic disturbances, suggesting cellular damage. These changes create a feedback loop that worsens kidney function and accelerates hypertension progression, highlighting the kidneys' crucial role in both initiating and exacerbating this condition.
Collapse
Affiliation(s)
- Paweł Wojtacha
- Department of Psychology and Sociology of Health and Public Health, University of Warmia and Mazury, Warszawska Av, 10-082 Olsztyn, Poland
| | - Ewelina Bogdańska-Chomczyk
- Department of Human Physiology and Pathophysiology, Collegium Medicum, University of Warmia and Mazury, Warszawska Av, 30, 10-082 Olsztyn, Poland; (E.B.-C.); (M.K.M.)
| | - Mariusz Krzysztof Majewski
- Department of Human Physiology and Pathophysiology, Collegium Medicum, University of Warmia and Mazury, Warszawska Av, 30, 10-082 Olsztyn, Poland; (E.B.-C.); (M.K.M.)
| | - Kazimierz Obremski
- Department of Veterinary Prevention and Feed Hygiene, Faculty of Veterinary Medicine, University of Warmia and Mazury, Oczapowskiego 13/29, 10-718 Olsztyn, Poland;
| | - Michał Stanisław Majewski
- Department of Pharmacology and Toxicology, Faculty of Medicine, University of Warmia and Mazury, Warszawska Av, 30, 10-082 Olsztyn, Poland;
| | - Anna Kozłowska
- Department of Human Physiology and Pathophysiology, Collegium Medicum, University of Warmia and Mazury, Warszawska Av, 30, 10-082 Olsztyn, Poland; (E.B.-C.); (M.K.M.)
| |
Collapse
|
16
|
Xu B, Dissanayake LV, Levchenko V, Zietara A, Kravtsova O, Staruschenko A. Deletion of Kcnj16 altered transcriptomic and metabolomic profiles of Dahl salt-sensitive rats. iScience 2024; 27:110901. [PMID: 39328933 PMCID: PMC11424968 DOI: 10.1016/j.isci.2024.110901] [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: 11/07/2023] [Revised: 04/06/2024] [Accepted: 09/04/2024] [Indexed: 09/28/2024] Open
Abstract
The inwardly rectifying K+ channel Kir5.1 (Kcnj16) is essential in renal salt handling and blood pressure control. However, the underlying mechanisms are not fully understood. Here, we integrated transcriptomics and metabolomics to comprehensively profile the changes in genes and metabolites in the Dahl salt-sensitive (SS) rat lacking Kcnj16 to identify potential mechanisms. Consistent with the phenotype of knockout (KO) rats, the transcriptomic profile predicted reduced blood pressure, kidney damage, and increased ion transport. Canonical pathway analysis suggested activation of metabolic-related pathways while suppression of immune response-related pathways in KO rats. Untargeted metabolomic analysis revealed different metabolic profiles between wild-type (WT) and KO rats. Integration of transcriptomic and metabolomic profiles suggested altered tricarboxylic acid (TCA) cycle, amino acid metabolism, and reactive oxygen species (ROS) metabolism that are related to SS hypertension. In conclusion, besides increased ion transport, our data suggest suppressed immune response-related and altered metabolic-related pathways of SS rats lacking Kir5.1.
Collapse
Affiliation(s)
- Biyang Xu
- Department of Molecular Pharmacology & Physiology, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | - Lashodya V Dissanayake
- Department of Molecular Pharmacology & Physiology, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | - Vladislav Levchenko
- Department of Molecular Pharmacology & Physiology, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | - Adrian Zietara
- Department of Molecular Pharmacology & Physiology, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | - Olha Kravtsova
- Department of Molecular Pharmacology & Physiology, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | - Alexander Staruschenko
- Department of Molecular Pharmacology & Physiology, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
- Hypertension and Kidney Research Center, University of South Florida, Tampa, FL, USA
- James A. Haley Veteran's Hospital, Tampa, FL, USA
| |
Collapse
|
17
|
Tukhovskaya EA, Ismailova AM, Perepechenova NA, Slashcheva GA, Palikov VA, Palikova YA, Rzhevsky DI, Rykov VA, Novikova NI, Dyachenko IA, Murashev AN. Development and Worsening of Hypertension with Age in Male Wistar Rats as a Physiological Model of Age-Related Hypertension: Correction of Hypertension with Taxifolin. Int J Mol Sci 2024; 25:11216. [PMID: 39456996 PMCID: PMC11509042 DOI: 10.3390/ijms252011216] [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: 09/30/2024] [Revised: 10/11/2024] [Accepted: 10/16/2024] [Indexed: 10/28/2024] Open
Abstract
To preclinically study the effectiveness of new antihypertensive drugs, various animal hypertension models are used. However, most of them do not correspond to primary hypertension, which develops in people with age. We used male Wistar rats of 4, 10, 12 and 18 months old. The animals were divided according to systolic blood pressure (SBP) into normotensive (SBP ≤ 114 mmHg) or hypertensive (SBP ≥ 115 mmHg). Within hypertensive animals, two cohorts were distinguished-with SBP below and above 125 mmHg. The animals received 100 µg/kg of taxifolin intraperitoneally for 7 days. A significant difference was shown between animals with SBP above and below 115 mmHg, as well as between cohorts of hypertensive animals with SBP above and below 125 mmHg within each age. The number of animals with elevated SBP increased with age both for clusters with an SBP above 115 mmHg and for cohorts with an SBP above 125 mmHg. Administration of taxifolin led to a significant decrease in the SBP only in hypertensive animals. A physiological model of age-related hypertension was obtained in male Wistar rats. It has been shown that hypertension develops and worsens with age. In preclinical studies, it should be taken into account that drugs may have different effects depending on the initial SBP of the animals.
Collapse
Affiliation(s)
- Elena A. Tukhovskaya
- Biological Testing Laboratory, Shemyakin-Ovchinnicov Institute of Bioorganic Chemistry (Branch), Russian Academy of Sciences, Prospekt Nauki, 6, Pushchino 142290, Russia
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Sommer G, Rodríguez López C, Hirschkorn A, Calimano G, Marques-Lopes J, Milner TA, Glass MJ. Estrogen Receptor Beta Agonist Influences Presynaptic NMDA Receptor Distribution in the Paraventricular Hypothalamic Nucleus Following Hypertension in a Mouse Model of Perimenopause. BIOLOGY 2024; 13:819. [PMID: 39452127 PMCID: PMC11505520 DOI: 10.3390/biology13100819] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Revised: 10/01/2024] [Accepted: 10/05/2024] [Indexed: 10/26/2024]
Abstract
Women become susceptible to hypertension as they transition to menopause (i.e., perimenopause); however, the underlying mechanisms are unclear. Animal studies using an accelerated ovarian failure (AOF) model of peri-menopause (peri-AOF) demonstrate that peri-AOF hypertension is associated with increased postsynaptic NMDA receptor plasticity in the paraventricular hypothalamic nucleus (PVN), a brain area critical for blood pressure regulation. However, recent evidence indicates that presynaptic NMDA receptors also play a role in neural plasticity. Here, using immuno-electron microscopy, we examine the influence of peri-AOF hypertension on the subcellular distribution of the essential NMDA GluN1 receptor subunit in PVN axon terminals in peri-AOF and in male mice. Hypertension was produced by 14-day slow-pressor angiotensin II (AngII) infusion. The involvement of estrogen signaling was investigated by co-administering an estrogen receptor beta (ERß) agonist. Although AngII induced hypertension in both peri-AOF and male mice, peri-AOF females showed higher cytoplasmic GluN1 levels. In peri-AOF females, activation of ERß blocked hypertension and increased plasmalemmal GluN1 in axon terminals. In contrast, stimulation of ERß did not inhibit hypertension or influence presynaptic GluN1 localization in males. These results indicate that sex-dependent recruitment of presynaptic NMDA receptors in the PVN is influenced by ERß signaling in mice during early ovarian failure.
Collapse
Affiliation(s)
- Garrett Sommer
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, 407 East 61st Street, New York, NY 10065, USA (C.R.L.); (J.M.-L.)
| | - Claudia Rodríguez López
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, 407 East 61st Street, New York, NY 10065, USA (C.R.L.); (J.M.-L.)
| | - Adi Hirschkorn
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, 407 East 61st Street, New York, NY 10065, USA (C.R.L.); (J.M.-L.)
| | - Gianna Calimano
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, 407 East 61st Street, New York, NY 10065, USA (C.R.L.); (J.M.-L.)
| | - Jose Marques-Lopes
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, 407 East 61st Street, New York, NY 10065, USA (C.R.L.); (J.M.-L.)
- Center for Translational Health and Medical Biotechnology Research (TBIO)/Health Research Network (RISE-HEALTH), ESS, Polytechnic of Porto, R. Dr. António Bernardino de Almeida, 400, 4200-072 Porto, Portugal
| | - Teresa A. Milner
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, 407 East 61st Street, New York, NY 10065, USA (C.R.L.); (J.M.-L.)
| | - Michael J. Glass
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, 407 East 61st Street, New York, NY 10065, USA (C.R.L.); (J.M.-L.)
| |
Collapse
|
19
|
Munteanu C, Popescu C, Vlădulescu-Trandafir AI, Onose G. Signaling Paradigms of H 2S-Induced Vasodilation: A Comprehensive Review. Antioxidants (Basel) 2024; 13:1158. [PMID: 39456412 PMCID: PMC11505308 DOI: 10.3390/antiox13101158] [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: 08/28/2024] [Revised: 09/21/2024] [Accepted: 09/23/2024] [Indexed: 10/28/2024] Open
Abstract
Hydrogen sulfide (H2S), a gas traditionally considered toxic, is now recognized as a vital endogenous signaling molecule with a complex physiology. This comprehensive study encompasses a systematic literature review that explores the intricate mechanisms underlying H2S-induced vasodilation. The vasodilatory effects of H2S are primarily mediated by activating ATP-sensitive potassium (K_ATP) channels, leading to membrane hyperpolarization and subsequent relaxation of vascular smooth muscle cells (VSMCs). Additionally, H2S inhibits L-type calcium channels, reducing calcium influx and diminishing VSMC contraction. Beyond ion channel modulation, H2S profoundly impacts cyclic nucleotide signaling pathways. It stimulates soluble guanylyl cyclase (sGC), increasing the production of cyclic guanosine monophosphate (cGMP). Elevated cGMP levels activate protein kinase G (PKG), which phosphorylates downstream targets like vasodilator-stimulated phosphoprotein (VASP) and promotes smooth muscle relaxation. The synergy between H2S and nitric oxide (NO) signaling further amplifies vasodilation. H2S enhances NO bioavailability by inhibiting its degradation and stimulating endothelial nitric oxide synthase (eNOS) activity, increasing cGMP levels and potent vasodilatory responses. Protein sulfhydration, a post-translational modification, plays a crucial role in cell signaling. H2S S-sulfurates oxidized cysteine residues, while polysulfides (H2Sn) are responsible for S-sulfurating reduced cysteine residues. Sulfhydration of key proteins like K_ATP channels and sGC enhances their activity, contributing to the overall vasodilatory effect. Furthermore, H2S interaction with endothelium-derived hyperpolarizing factor (EDHF) pathways adds another layer to its vasodilatory mechanism. By enhancing EDHF activity, H2S facilitates the hyperpolarization and relaxation of VSMCs through gap junctions between endothelial cells and VSMCs. Recent findings suggest that H2S can also modulate transient receptor potential (TRP) channels, particularly TRPV4 channels, in endothelial cells. Activating these channels by H2S promotes calcium entry, stimulating the production of vasodilatory agents like NO and prostacyclin, thereby regulating vascular tone. The comprehensive understanding of H2S-induced vasodilation mechanisms highlights its therapeutic potential. The multifaceted approach of H2S in modulating vascular tone presents a promising strategy for developing novel treatments for hypertension, ischemic conditions, and other vascular disorders. The interaction of H2S with ion channels, cyclic nucleotide signaling, NO pathways, ROS (Reactive Oxygen Species) scavenging, protein sulfhydration, and EDHF underscores its complexity and therapeutic relevance. In conclusion, the intricate signaling paradigms of H2S-induced vasodilation offer valuable insights into its physiological role and therapeutic potential, promising innovative approaches for managing various vascular diseases through the modulation of vascular tone.
Collapse
Affiliation(s)
- Constantin Munteanu
- Department of Biomedical Sciences, Faculty of Medical Bioengineering, University of Medicine and Pharmacy “Grigore T. Popa” Iași, 700454 Iași, Romania
- Neuromuscular Rehabilitation Clinic Division, Clinical Emergency Hospital “Bagdasar-Arseni”, 041915 Bucharest, Romania; (A.-I.V.-T.); (G.O.)
| | - Cristina Popescu
- Neuromuscular Rehabilitation Clinic Division, Clinical Emergency Hospital “Bagdasar-Arseni”, 041915 Bucharest, Romania; (A.-I.V.-T.); (G.O.)
- Faculty of Medicine, University of Medicine and Pharmacy “Carol Davila”, 020022 Bucharest, Romania
| | - Andreea-Iulia Vlădulescu-Trandafir
- Neuromuscular Rehabilitation Clinic Division, Clinical Emergency Hospital “Bagdasar-Arseni”, 041915 Bucharest, Romania; (A.-I.V.-T.); (G.O.)
- Faculty of Medicine, University of Medicine and Pharmacy “Carol Davila”, 020022 Bucharest, Romania
| | - Gelu Onose
- Neuromuscular Rehabilitation Clinic Division, Clinical Emergency Hospital “Bagdasar-Arseni”, 041915 Bucharest, Romania; (A.-I.V.-T.); (G.O.)
- Faculty of Medicine, University of Medicine and Pharmacy “Carol Davila”, 020022 Bucharest, Romania
| |
Collapse
|
20
|
Jovanović M, Stevanović B, Pajović V, Tasić T, Lozić M, Đukić L, Kosić M, Murphy D, Japundžić-Žigon N. Vasopressin and cardiovascular autonomic adjustment in chronic hypertensive pregnancy. Hypertens Res 2024; 47:2393-2404. [PMID: 39039283 PMCID: PMC11374678 DOI: 10.1038/s41440-024-01769-6] [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: 02/27/2024] [Revised: 05/24/2024] [Accepted: 06/06/2024] [Indexed: 07/24/2024]
Abstract
Chronic hypertensive pregnancy (CHP) is a growing health issue with unknown etiology. Vasopressin (VP), a nonapeptide synthesized in paraventricular (PVN) and supraoptic nucleus (SON), is a well-known neuroendocrine and autonomic modulator of the cardiovascular system, related to hypertension development. We quantified gene expression of VP and its receptors, V1aR and V1bR, within the PVN and SON in CHP and normal pregnancy, and assessed levels of secreted plasma VP. Also, we evaluated autonomic cardiovascular adaptations to CHP using spectral indices of blood pressure (BPV) and heart rate (HRV) short-term variability, and spontaneous baroreflex sensitivity (BRS). Experiments were performed in female spontaneously hypertensive rats (SHRs) and in normotensive Wistar rats (WRs). Animals were equipped with a radiotelemetry probe for continuous hemodynamic recordings before and during pregnancy. BPV, HRV and BRS were assessed using spectral analysis and the sequence method, respectively. Plasma VP was determined by ELISA whilst VP, V1aR, and V1bR gene expression was analyzed by real-time-quantitative PCR (RT-qPCR). The results show that non-pregnant SHRs exhibit greater VP, V1aR, and V1bR gene expression in both PVN and SON respectively, compared to Wistar dams. Pregnancy decreased VP gene expression in the SON of SHRs but increased it in the PVN and SON of WRs. Pregnant SHRs exhibited a marked drop in plasma VP concentration associated with BP normalization. This triggered marked tachycardia, heart rate variability increase, and BRS increase in pregnant SHRs. It follows that regardless of BP normalization in late pregnancy, SHRs exhibit cardiovascular vulnerability and compensate by recruiting vagal mechanisms. Pregnant SHR dams have reduced expression of VP in SON associated with increased V1bR expression, lower plasma VP, normal BP during late pregnancy and marked signs of enhanced sympathetic cardiac stimulation (increased HR and LFHR variability) and recruitment of vagal mechanisms (enhancement of BRS and HFHR variability).
Collapse
Affiliation(s)
- Mirjana Jovanović
- Department of Pathophysiology, University of Belgrade Faculty of Medicine, Belgrade, RS, Serbia
| | | | - Vladislav Pajović
- Department of Pharmacology, University of Belgrade Faculty of Medicine, Belgrade, RS, Serbia
| | - Tatjana Tasić
- University of Belgrade Faculty of Dentistry, Belgrade, RS, Serbia
| | - Maja Lozić
- Department of Pharmacology, University of Belgrade Faculty of Medicine, Belgrade, RS, Serbia
| | - Ljiljana Đukić
- University of Belgrade Faculty of Dentistry, Belgrade, RS, Serbia
| | - Marija Kosić
- Department of Pharmacology, University of Belgrade Faculty of Medicine, Belgrade, RS, Serbia
| | - David Murphy
- Bristol Medical School: Translational Health Sciences, University of Bristol, Bristol, UK
| | - Nina Japundžić-Žigon
- Department of Pharmacology, University of Belgrade Faculty of Medicine, Belgrade, RS, Serbia.
| |
Collapse
|
21
|
Fayyaz AU, Eltony M, Prokop LJ, Koepp KE, Borlaug BA, Dasari S, Bois MC, Margulies KB, Maleszewski JJ, Wang Y, Redfield MM. Pathophysiological insights into HFpEF from studies of human cardiac tissue. Nat Rev Cardiol 2024:10.1038/s41569-024-01067-1. [PMID: 39198624 DOI: 10.1038/s41569-024-01067-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/18/2024] [Indexed: 09/01/2024]
Abstract
Heart failure with preserved ejection fraction (HFpEF) is a major, worldwide health-care problem. Few therapies for HFpEF exist because the pathophysiology of this condition is poorly defined and, increasingly, postulated to be diverse. Although perturbations in other organs contribute to the clinical profile in HFpEF, altered cardiac structure, function or both are the primary causes of this heart failure syndrome. Therefore, studying myocardial tissue is fundamental to improve pathophysiological insights and therapeutic discovery in HFpEF. Most studies of myocardial changes in HFpEF have relied on cardiac tissue from animal models without (or with limited) confirmatory studies in human cardiac tissue. Animal models of HFpEF have evolved based on theoretical HFpEF aetiologies, but these models might not reflect the complex pathophysiology of human HFpEF. The focus of this Review is the pathophysiological insights gained from studies of human HFpEF myocardium. We outline the rationale for these studies, the challenges and opportunities in obtaining myocardial tissue from patients with HFpEF and relevant comparator groups, the analytical approaches, the pathophysiological insights gained to date and the remaining knowledge gaps. Our objective is to provide a roadmap for future studies of cardiac tissue from diverse cohorts of patients with HFpEF, coupling discovery biology with measures to account for pathophysiological diversity.
Collapse
Affiliation(s)
- Ahmed U Fayyaz
- Department of Cardiovascular Disease, Division of Circulatory Failure, Mayo Clinic, Rochester, MN, USA
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
| | - Muhammad Eltony
- Department of Cardiovascular Disease, Division of Circulatory Failure, Mayo Clinic, Rochester, MN, USA
| | - Larry J Prokop
- Mayo Clinic College of Medicine and Science, Library Reference Service, Rochester, MN, USA
| | - Katlyn E Koepp
- Department of Cardiovascular Disease, Division of Circulatory Failure, Mayo Clinic, Rochester, MN, USA
| | - Barry A Borlaug
- Department of Cardiovascular Disease, Division of Circulatory Failure, Mayo Clinic, Rochester, MN, USA
| | - Surendra Dasari
- Mayo Clinic College of Medicine and Science, Computational Biology, Rochester, MN, USA
| | - Melanie C Bois
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
| | - Kenneth B Margulies
- Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Joesph J Maleszewski
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
| | - Ying Wang
- Department of Cardiovascular Disease, Division of Circulatory Failure, Mayo Clinic, Rochester, MN, USA
| | - Margaret M Redfield
- Department of Cardiovascular Disease, Division of Circulatory Failure, Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|
22
|
Faienza S, Citterio L, Messaggio E, Zagato L, Lanzani C, Simonini M, Canciani B, Sanvito F, Rampoldi L, Pavlovic D, Manunta P. A novel mouse model recapitulates the effects of rs2254524 variant in the lanosterol synthase gene on salt sensitivity and organ damage. J Hypertens 2024:00004872-990000000-00523. [PMID: 39248148 DOI: 10.1097/hjh.0000000000003843] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Accepted: 07/29/2024] [Indexed: 09/10/2024]
Abstract
OBJECTIVE The blood pressure (BP) response to salt intake (salt sensitivity) shows great variability among individuals and is more frequent in hypertensive patients. Elevated levels of the steroid hormone Endogenous Ouabain (EO) are associated with hypertension (HT) and salt sensitivity. The lanosterol synthase gene ( LSS ) plays a key role in the biosynthesis of steroids and its rs2254524 variant (Val642Leu) is linked to salt sensitivity in humans. This study aims to investigate the pathophysiological significance of the Lss missense variation in a new knock-in mouse model of salt-sensitive HT onset. METHODS We generated a mouse model carrying the murine homolog (Val643Leu) of the human LSS variant. C57BL/6N LssV643L/V643L mice were fed different NaCl diets (low-salt, LSD; normal-salt, NSD; high-salt, HSD) and were characterized at functional, histological, and molecular levels. RESULTS At baseline, mutant mice showed an enlarged kidney compared to the wild-type (WT) counterpart, but the Lss V643L variant did not affect EO biosynthesis nor systolic BP at 3 and 12 months. In HSD, we observed an increased systolic BP only in 12-month-old LssV643L/V643L mice, compared to NSD. Moreover, only the HSD LssV643L/V643L mice showed cardiac hypertrophy and a higher incidence of cardiac fibrosis compared to WT at 12 months. Finally, the Lss mRNA level was differentially regulated by HSD in the adrenal gland, liver, and heart of LssV643L/V643L mice compared to WT. CONCLUSIONS The novel Lss mouse model resembles the salt-sensitive HT phenotype observed in hypertensive patients and provides a good model of salt-sensitive HT and HT-mediated organ damage.
Collapse
Affiliation(s)
- Sipontina Faienza
- Genomics of Renal Diseases and Hypertension Unit, IRCCS Ospedale San Raffaele
- Università Vita-Salute San Raffaele
| | - Lorena Citterio
- Genomics of Renal Diseases and Hypertension Unit, IRCCS Ospedale San Raffaele
| | | | - Laura Zagato
- Genomics of Renal Diseases and Hypertension Unit, IRCCS Ospedale San Raffaele
| | - Chiara Lanzani
- Genomics of Renal Diseases and Hypertension Unit, IRCCS Ospedale San Raffaele
- Università Vita-Salute San Raffaele
| | - Marco Simonini
- Genomics of Renal Diseases and Hypertension Unit, IRCCS Ospedale San Raffaele
| | | | - Francesca Sanvito
- Pathology Unit, Department of Experimental Oncology, IRCCS Ospedale San Raffaele
| | - Luca Rampoldi
- Università Vita-Salute San Raffaele
- Molecular Genetics of Renal Disorders Unit, Division of Genetics and Cell Biology, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Davor Pavlovic
- Institute of Cardiovascular Sciences, University of Birmingham, Edgbaston, Wolfson Drive, Birmingham, UK
| | - Paolo Manunta
- Genomics of Renal Diseases and Hypertension Unit, IRCCS Ospedale San Raffaele
- Università Vita-Salute San Raffaele
| |
Collapse
|
23
|
Shen D, Zhu Y, Mao J, Lin R, Jiang X, Liang L, Peng J, Cao Y, Dong S, He K, Wang N. Highly sensitive and accurate measurement of underivatized phosphoenolpyruvate in plasma and serum via EDTA-facilitated hydrophilic interaction liquid chromatography-tandem mass spectrometry. Talanta 2024; 275:126134. [PMID: 38692044 DOI: 10.1016/j.talanta.2024.126134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Revised: 04/08/2024] [Accepted: 04/18/2024] [Indexed: 05/03/2024]
Abstract
Phosphoenolpyruvate (PEP) is an essential intermediate metabolite that is involved in various vital biochemical reactions. However, achieving the direct and accurate quantification of PEP in plasma or serum poses a significant challenge owing to its strong polarity and metal affinity. In this study, a sensitive method for the direct determination of PEP in plasma and serum based on ethylenediaminetetraacetic acid (EDTA)-facilitated hydrophilic interaction liquid chromatography-tandem mass spectrometry was developed. Superior chromatographic retention and peak shapes were achieved using a zwitterionic stationary-phase HILIC column with a metal-inert inner surface. Efficient dechelation of PEP-metal complexes in serum/plasma samples was achieved through the introduction of EDTA, resulting in a significant enhancement of the PEP signal. A PEP isotopically labelled standard was employed as a surrogate analyte for the determination of endogenous PEP, and validation assessments proved the sensitivity, selectivity, and reproducibility of this method. The method was applied to the comparative quantification of PEP in plasma and serum samples from mice and rats, as well as in HepG2 cells, HEK293T cells, and erythrocytes; the results confirmed its applicability in PEP-related biomedical research. The developed method can quantify PEP in diverse biological matrices, providing a feasible opportunity to investigate the role of PEP in relevant biomedical research.
Collapse
Affiliation(s)
- Danning Shen
- National Center of Biomedical Analysis, Beijing, 100850, China
| | - Yingjie Zhu
- National Center of Biomedical Analysis, Beijing, 100850, China
| | - Jie Mao
- National Center of Biomedical Analysis, Beijing, 100850, China
| | - Runfeng Lin
- National Center of Biomedical Analysis, Beijing, 100850, China
| | - Xin Jiang
- National Center of Biomedical Analysis, Beijing, 100850, China
| | - Longhui Liang
- National Center of Biomedical Analysis, Beijing, 100850, China
| | - Jing Peng
- National Center of Biomedical Analysis, Beijing, 100850, China
| | - Yanqing Cao
- National Center of Biomedical Analysis, Beijing, 100850, China
| | - Suhe Dong
- National Center of Biomedical Analysis, Beijing, 100850, China
| | - Kun He
- National Center of Biomedical Analysis, Beijing, 100850, China.
| | - Na Wang
- National Center of Biomedical Analysis, Beijing, 100850, China.
| |
Collapse
|
24
|
Mao X, Wu S, Huang D, Li C. Complications and comorbidities associated with antineoplastic chemotherapy: Rethinking drug design and delivery for anticancer therapy. Acta Pharm Sin B 2024; 14:2901-2926. [PMID: 39027258 PMCID: PMC11252465 DOI: 10.1016/j.apsb.2024.03.006] [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: 11/26/2023] [Revised: 01/29/2024] [Accepted: 02/10/2024] [Indexed: 07/20/2024] Open
Abstract
Despite the considerable advancements in chemotherapy as a cornerstone modality in cancer treatment, the prevalence of complications and pre-existing diseases is on the rise among cancer patients along with prolonged survival and aging population. The relationships between these disorders and cancer are intricate, bearing significant influence on the survival and quality of life of individuals with cancer and presenting challenges for the prognosis and outcomes of malignancies. Herein, we review the prevailing complications and comorbidities that often accompany chemotherapy and summarize the lessons to learn from inadequate research and management of this scenario, with an emphasis on possible strategies for reducing potential complications and alleviating comorbidities, as well as an overview of current preclinical cancer models and practical advice for establishing bio-faithful preclinical models in such complex context.
Collapse
Affiliation(s)
- Xiaoman Mao
- College of Pharmaceutical Sciences, Southwest University, Chongqing 400715, China
| | - Shuang Wu
- Medical Research Institute, Southwest University, Chongqing 400715, China
| | - Dandan Huang
- College of Pharmaceutical Sciences, Southwest University, Chongqing 400715, China
| | - Chong Li
- College of Pharmaceutical Sciences, Southwest University, Chongqing 400715, China
- Medical Research Institute, Southwest University, Chongqing 400715, China
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| |
Collapse
|
25
|
Grano de Oro A, Kumariya S, Mell B, Zubcevic J, Joe B, Osman I. Spontaneous vascular dysfunction in Dahl salt-sensitive male rats raised without a high-salt diet. Physiol Rep 2024; 12:e16165. [PMID: 39048525 PMCID: PMC11268988 DOI: 10.14814/phy2.16165] [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: 04/11/2024] [Revised: 07/09/2024] [Accepted: 07/17/2024] [Indexed: 07/27/2024] Open
Abstract
Dahl salt-sensitive (SS) rats fed a high-salt diet, but not low-salt, exhibit vascular dysfunction. Several substrains of SS rats exist that differ in their blood pressure phenotypes and salt sensitivity. The goal of this study was to investigate whether the John-Rapp-derived SS rat (SS/Jr), which exhibits spontaneous hypertension on a low-salt diet, presents with hallmarks of vascular dysfunction observed in another experimental model of hypertension independent of dietary salt, the spontaneously hypertensive rat (SHR). Endothelium-intact aortic rings and mesenteric resistance arteries were isolated from low-salt fed adult male SS/Jr rats and SHRs, or their respective controls, for isometric wire myography. Vessels were challenged with cumulative concentrations of various vasoactive substances, in the absence or presence of nitric oxide synthase or cyclooxygenase inhibitors. Despite showing some differences in their responses to various vasoactive substances, both SS/Jr rats and SHRs exhibited key features of vascular dysfunction, including endothelial dysfunction and hyperresponsiveness to vasocontractile agonists. In conclusion, this study provides evidence to support the utility of the SS/Jr rat strain maintained on a low-salt diet as a valid experimental model for vascular dysfunction, a key feature of human hypertension.
Collapse
Affiliation(s)
- Arturo Grano de Oro
- Department of Physiology and Pharmacology, Center for Hypertension and Personalized MedicineUniversity of Toledo, College of Medicine and Life SciencesToledoOhioUSA
| | - Sanjana Kumariya
- Department of Physiology and Pharmacology, Center for Hypertension and Personalized MedicineUniversity of Toledo, College of Medicine and Life SciencesToledoOhioUSA
| | - Blair Mell
- Department of Physiology and Pharmacology, Center for Hypertension and Personalized MedicineUniversity of Toledo, College of Medicine and Life SciencesToledoOhioUSA
| | - Jasenka Zubcevic
- Department of Physiology and Pharmacology, Center for Hypertension and Personalized MedicineUniversity of Toledo, College of Medicine and Life SciencesToledoOhioUSA
| | - Bina Joe
- Department of Physiology and Pharmacology, Center for Hypertension and Personalized MedicineUniversity of Toledo, College of Medicine and Life SciencesToledoOhioUSA
| | - Islam Osman
- Department of Physiology and Pharmacology, Center for Hypertension and Personalized MedicineUniversity of Toledo, College of Medicine and Life SciencesToledoOhioUSA
| |
Collapse
|
26
|
Peter JK, Umene R, Wu CH, Nakamura Y, Washimine N, Yamamoto R, Ngugi C, Linge K, Kweri JK, Inoue T. Renal macrophages induce hypertension and kidney fibrosis in Angiotensin II salt mice model. Biochem Biophys Res Commun 2024; 715:149997. [PMID: 38678782 DOI: 10.1016/j.bbrc.2024.149997] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Revised: 04/12/2024] [Accepted: 04/23/2024] [Indexed: 05/01/2024]
Abstract
The immune system is involved in hypertension development with different immune cells reported to have either pro or anti-hypertensive effects. In hypertension, immune cells have been thought to infiltrate blood pressure-regulating organs, resulting in either elevation or reduction of blood pressure. There is controversy over whether macrophages play a detrimental or beneficial role in the development of hypertension, and the few existing studies have yielded conflicting results. This study aimed to determine the effects of angiotensin II (Ang II) salt-induced hypertension on renal immune cells and to determine whether renal macrophages are involved in the induction of hypertension. Hypertension was induced by administration of Ang II and saline for two weeks. The effects of hypertension on kidney immune cells were assessed using flow cytometry. Macrophage infiltration in the kidney was assessed by immunohistochemistry and kidney fibrosis was assessed using trichrome stain and kidney real time-qPCR. Liposome encapsulated clodronate was used to deplete macrophages in C57BL/6J mice and investigate the direct role of macrophages in hypertension induction. Ang II saline mice group developed hypertension, had increased renal macrophages, and had increased expression of Acta2 and Col1a1 and kidney fibrotic areas. Macrophage depletion blunted hypertension development and reduced the expression of Acta2 and Col1a1 in the kidney and kidney fibrotic areas in Ang II saline group. The results of this study demonstrate that macrophages infiltrate the kidneys and increase kidney fibrosis in Ang II salt-induced hypertension, and depletion of macrophages suppresses the development of hypertension and decreases kidney fibrosis. This indicates that macrophages play a direct role in hypertension development. Hence macrophages have a potential to be considered as therapeutic target in hypertension management.
Collapse
Affiliation(s)
- Joseph Kasyoki Peter
- Department of Physiology of Visceral Function and Body Fluid, Graduate School of Biomedical Sciences, Nagasaki University, Japan; Department of Medical Physiology, School of Medicine, Jomo Kenyatta University of Agriculture and Technology, Kenya; Department of Clinical Medicine, School of Medicine, Jomo Kenyatta University of Agriculture and Technology, Kenya
| | - Ryusuke Umene
- Department of Physiology of Visceral Function and Body Fluid, Graduate School of Biomedical Sciences, Nagasaki University, Japan; Department of Nephrology, Graduate School of Biomedical Sciences, Nagasaki University, Japan.
| | - Chia-Hsien Wu
- Department of Physiology of Visceral Function and Body Fluid, Graduate School of Biomedical Sciences, Nagasaki University, Japan
| | - Yasuna Nakamura
- Department of Physiology of Visceral Function and Body Fluid, Graduate School of Biomedical Sciences, Nagasaki University, Japan
| | - Norito Washimine
- Department of Physiology of Visceral Function and Body Fluid, Graduate School of Biomedical Sciences, Nagasaki University, Japan; Department of Nephrology, Graduate School of Biomedical Sciences, Nagasaki University, Japan
| | - Ryoko Yamamoto
- Department of Physiology of Visceral Function and Body Fluid, Graduate School of Biomedical Sciences, Nagasaki University, Japan
| | - Caroline Ngugi
- Department of Medical Microbiology, School of Biomedical Sciences, Jomo Kenyatta University of Agriculture and Technology, Kenya
| | - Kavoo Linge
- Department of Medical Physiology, School of Medicine, Jomo Kenyatta University of Agriculture and Technology, Kenya
| | - Joseph K Kweri
- Department of Human Anatomy, School of Medicine, Jomo Kenyatta University of Agriculture and Technology, Kenya
| | - Tsuyoshi Inoue
- Department of Physiology of Visceral Function and Body Fluid, Graduate School of Biomedical Sciences, Nagasaki University, Japan.
| |
Collapse
|
27
|
Sinclair D, Canty AJ, Ziebell JM, Woodhouse A, Collins JM, Perry S, Roccati E, Kuruvilla M, Leung J, Atkinson R, Vickers JC, Cook AL, King AE. Experimental laboratory models as tools for understanding modifiable dementia risk. Alzheimers Dement 2024; 20:4260-4289. [PMID: 38687209 PMCID: PMC11180874 DOI: 10.1002/alz.13834] [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: 12/13/2023] [Revised: 02/29/2024] [Accepted: 03/18/2024] [Indexed: 05/02/2024]
Abstract
Experimental laboratory research has an important role to play in dementia prevention. Mechanisms underlying modifiable risk factors for dementia are promising targets for dementia prevention but are difficult to investigate in human populations due to technological constraints and confounds. Therefore, controlled laboratory experiments in models such as transgenic rodents, invertebrates and in vitro cultured cells are increasingly used to investigate dementia risk factors and test strategies which target them to prevent dementia. This review provides an overview of experimental research into 15 established and putative modifiable dementia risk factors: less early-life education, hearing loss, depression, social isolation, life stress, hypertension, obesity, diabetes, physical inactivity, heavy alcohol use, smoking, air pollution, anesthetic exposure, traumatic brain injury, and disordered sleep. It explores how experimental models have been, and can be, used to address questions about modifiable dementia risk and prevention that cannot readily be addressed in human studies. HIGHLIGHTS: Modifiable dementia risk factors are promising targets for dementia prevention. Interrogation of mechanisms underlying dementia risk is difficult in human populations. Studies using diverse experimental models are revealing modifiable dementia risk mechanisms. We review experimental research into 15 modifiable dementia risk factors. Laboratory science can contribute uniquely to dementia prevention.
Collapse
Affiliation(s)
- Duncan Sinclair
- Wicking Dementia Research and Education Centre, University of TasmaniaHobartTasmaniaAustralia
| | - Alison J. Canty
- Wicking Dementia Research and Education Centre, University of TasmaniaHobartTasmaniaAustralia
- Global Brain Health Institute, Trinity CollegeDublinIreland
| | - Jenna M. Ziebell
- Wicking Dementia Research and Education Centre, University of TasmaniaHobartTasmaniaAustralia
| | - Adele Woodhouse
- Wicking Dementia Research and Education Centre, University of TasmaniaHobartTasmaniaAustralia
| | - Jessica M. Collins
- Wicking Dementia Research and Education Centre, University of TasmaniaHobartTasmaniaAustralia
| | - Sharn Perry
- Wicking Dementia Research and Education Centre, University of TasmaniaHobartTasmaniaAustralia
| | - Eddy Roccati
- Wicking Dementia Research and Education Centre, University of TasmaniaHobartTasmaniaAustralia
| | - Maneesh Kuruvilla
- Wicking Dementia Research and Education Centre, University of TasmaniaHobartTasmaniaAustralia
| | - Jacqueline Leung
- Wicking Dementia Research and Education Centre, University of TasmaniaHobartTasmaniaAustralia
| | - Rachel Atkinson
- Wicking Dementia Research and Education Centre, University of TasmaniaHobartTasmaniaAustralia
| | - James C. Vickers
- Wicking Dementia Research and Education Centre, University of TasmaniaHobartTasmaniaAustralia
| | - Anthony L. Cook
- Wicking Dementia Research and Education Centre, University of TasmaniaHobartTasmaniaAustralia
| | - Anna E. King
- Wicking Dementia Research and Education Centre, University of TasmaniaHobartTasmaniaAustralia
| |
Collapse
|
28
|
Pagani E, Ropke CD, Soares CM, Perez SAC, Benevides PJC, Barbosa BS, Carvalho ACB, Behrens MD. Technology Readiness Level Roadmap for Developing Innovative Herbal Medicinal Products. Pharmaceuticals (Basel) 2024; 17:703. [PMID: 38931370 PMCID: PMC11206302 DOI: 10.3390/ph17060703] [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: 04/19/2024] [Revised: 05/15/2024] [Accepted: 05/17/2024] [Indexed: 06/28/2024] Open
Abstract
Despite the vast global botanical diversity, the pharmaceutical development of herbal medicinal products (HMPs) remains underexploited. Of over 370,000 described plant species, only a few hundred are utilized in HMPs. Most of these have originated from traditional use, and only a minority come from megadiverse countries. Exploiting the pharmacological synergies of the hundreds of compounds found in poorly studied plant species may unlock new therapeutic possibilities, enhance megadiverse countries' scientific and socio-economic development, and help conserve biodiversity. However, extensive constraints in the development process of HMPs pose significant barriers to transforming this unsatisfactory socio-economic landscape. This paper proposes a roadmap to overcome these challenges, based on the technology readiness levels (TRLs) introduced by NASA to assess the maturity of technologies. It aims to assist research entities, manufacturers, and funding agencies from megadiverse countries in the discovery, development, and global market authorization of innovative HMPs that comply with regulatory standards from ANVISA, EMA, and FDA, as well as WHO and ICH guidelines.
Collapse
Affiliation(s)
- Eduardo Pagani
- Medical Department, Azidus Brasil, Valinhos 13271-130, SP, Brazil
- Centroflora Group, Innovation Department, Campinas 06460-040, SP, Brazil
| | | | - Cristiane Mota Soares
- Project Management Office, Vice Direction of Education, Research and Innovation, Institute of Drug Technology Farmanguinhos, Oswaldo Cruz Foundation, Rio de Janeiro 21041-250, RJ, Brazil
| | - Sandra Aurora Chavez Perez
- Project Management Office, Vice Direction of Education, Research and Innovation, Institute of Drug Technology Farmanguinhos, Oswaldo Cruz Foundation, Rio de Janeiro 21041-250, RJ, Brazil
| | | | | | - Ana Cecilia Bezerra Carvalho
- GMESP, Brazilian Health Regulatory Agency, Agência Nacional de Vigilância Sanitária (ANVISA), Brasília 71205-050, DF, Brazil
| | - Maria Dutra Behrens
- Natural Products Department, Vice Direction of Education, Research and Innovation, Institute of Drug Technology Farmanguinhos, Oswaldo Cruz Foundation, Rio de Janeiro 21041-250, RJ, Brazil
| |
Collapse
|
29
|
Choi Y, Lee ES, Woo SK, Lee KC, Chung HK, Kang JH. Feasibility Study of Single-Photon Emission Computed Tomography with Iodine-123 Labeled Metaiodobenzylguanidine for Preclinical Evaluation of Labetalol as a β-Adrenergic Receptor Blocker. Mol Pharm 2024; 21:2435-2440. [PMID: 38626389 PMCID: PMC11080995 DOI: 10.1021/acs.molpharmaceut.3c01240] [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: 01/05/2024] [Revised: 04/04/2024] [Accepted: 04/04/2024] [Indexed: 04/18/2024]
Abstract
Among clinically used radiopharmaceuticals, iodine-123 labeled metaiodobenzylguanidine ([123I]mIBG) serves for diagnosing neuroendocrine tumors and obtaining images of myocardial sympathetic innervation. mIBG, a structural analogue of norepinephrine (NE), a neurotransmitter acting in peripheral and central nerves, follows a pathway similar to NE, transmitting signals through the NE transporter (NET) located at synaptic terminals. It moves through the body without decomposing, enabling noninvasive image evaluation. In this study, we aimed to quantify [123I]mIBG uptake in the adrenal glands using small animal single-photon emission computed tomography/computed tomography (SPECT/CT) images post [123I]mIBG administration. We investigated the possibility of assessing the effectiveness of β-adrenergic receptor blockers by quantifying SPECT/CT images and biodistribution results to determine the degree of [123I]mIBG uptake in the adrenal glands treated with labetalol, a known β-adrenergic receptor blocker. Upon intravenous administration of [123I]mIBG to mice, SPECT/CT images were acquired over time to confirm the in vivo distribution pattern, revealing a clear uptake in the adrenal glands. Labetalol inhibited the uptake of [123I]mIBG in cell lines expressing NET. A decrease in [123I]mIBG uptake in the adrenal glands was observed in the labetalol-treated group compared with the normal group through SPECT/CT imaging and biodistribution studies. These results demonstrate that SPECT/CT imaging with [123I]mIBG could be applicable for evaluating the preclinical efficacy of new antihypertensive drug candidates such as labetalol, a β-adrenergic receptor blocker.
Collapse
Affiliation(s)
- Yiseul Choi
- Korea
Radioisotope Center for Pharmaceuticals, Korea Institute of Radiological and Medical Sciences (KIRAMS), Seoul 01812, Korea
| | - Eun Sang Lee
- Korea
Radioisotope Center for Pharmaceuticals, Korea Institute of Radiological and Medical Sciences (KIRAMS), Seoul 01812, Korea
| | - Sang-Keun Woo
- Division
of Applied RI, Korea Institute of Radiological
and Medical Sciences (KIRAMS), Seoul 01812, Korea
| | - Kyo Chul Lee
- Division
of Applied RI, Korea Institute of Radiological
and Medical Sciences (KIRAMS), Seoul 01812, Korea
| | - Hye Kyung Chung
- Korea
Radioisotope Center for Pharmaceuticals, Korea Institute of Radiological and Medical Sciences (KIRAMS), Seoul 01812, Korea
| | - Joo Hyun Kang
- Korea
Radioisotope Center for Pharmaceuticals, Korea Institute of Radiological and Medical Sciences (KIRAMS), Seoul 01812, Korea
| |
Collapse
|
30
|
Guo R, Spyropoulos F, Michel T. FRBM Mini REVIEW: Chemogenetic approaches to probe redox dysregulation in heart failure. Free Radic Biol Med 2024; 217:173-178. [PMID: 38565399 PMCID: PMC11221410 DOI: 10.1016/j.freeradbiomed.2024.03.027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Revised: 03/28/2024] [Accepted: 03/30/2024] [Indexed: 04/04/2024]
Abstract
Chemogenetics refers to experimental methods that use novel recombinant proteins that can be dynamically and uniquely regulated by specific biochemicals. Chemogenetic approaches allow the precise manipulation of cellular signaling to delineate the molecular pathways involved in both physiological and pathological disease states. Approaches utilizing yeast d-amino acid oxidase (DAAO) enable manipulation of intracellular redox metabolism through generation of hydrogen peroxide in the presence of d-amino acids and have led to the development of new and informative animal models to characterize the impact of oxidative stress in heart failure and neurodegeneration. These chemogenetic models, in which DAAO expression is regulated by different tissue-specific promoters, have led to a range of cardiac phenotypes. This review discusses chemogenetic approaches to manipulate oxidative stress in models of heart failure. These approaches provide new insights into the relationships between redox metabolism and normal and pathologic states in the heart, as well as in other diseases characterized by oxidative stress.
Collapse
Affiliation(s)
- Ruby Guo
- Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, 02115, USA
| | - Fotios Spyropoulos
- Newborn Medicine Division, Department of Pediatrics, Brigham and Women's Hospital, Harvard Medical School, Boston, 02115, USA
| | - Thomas Michel
- Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, 02115, USA.
| |
Collapse
|
31
|
Zheng K, Layton AT. Predicting sex differences in the effects of diuretics in renal epithelial transport during angiotensin II-induced hypertension. Am J Physiol Renal Physiol 2024; 326:F737-F750. [PMID: 38482554 DOI: 10.1152/ajprenal.00398.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Revised: 03/05/2024] [Accepted: 03/06/2024] [Indexed: 04/26/2024] Open
Abstract
Chronic angiotensin II (ANG II) infusion is an experimental model that induces hypertension in rodents. The natriuresis, diuresis, and blood pressure responses differ between males and females. This is perhaps not unexpected, given the rodent kidney, which plays a key role in blood pressure regulation, exhibits marked sex differences. Under normotensive conditions, compared with males, the female rat nephron exhibits lower Na+/H+ exchanger 3 (NHE3) activity along the proximal tubule but higher Na+ transporter activities along the distal segments. ANG II infusion-induced hypertension induces a pressure natriuretic response that reduces NHE3 activity and shifts Na+ transport capacity downstream. The goals of this study were to apply a computational model of epithelial transport along a rat nephron 1) to understand how a 14-day ANG II infusion impacts segmental electrolyte transport in male and female rat nephrons and 2) to identify and explain any sex differences in the effects of loop diuretics, thiazide diuretics, and K+-sparing diuretics. Model simulations suggest that the NHE3 downregulation in the proximal tubule is a major contributor to natriuresis and diuresis in hypertension, with the effects stronger in males. All three diuretics are predicted to induce stronger natriuretic and diuretic effects under hypertension compared with normotension, with relative increases in sodium excretion higher in hypertensive females than in males. The stronger natriuretic responses can be explained by the downstream shift of Na+ transport load in hypertension and by the larger distal transport load in females, both of which limit the ability of the distal segments to further elevate their Na+ transport.NEW & NOTEWORTHY Sex differences in the prevalence of hypertension are found in human and animal models. The kidney, which regulates blood pressure, exhibits sex differences in morphology, hemodynamics, and membrane transporter distributions. This computational modeling study provides insights into how the sexually dimorphic responses to a 14-day angiotensin II infusion differentially impact segmental electrolyte transport in rats. Simulations of diuretic administration explain how the natriuretic and diuretic effects differ between normotension and hypertension and between the sexes.
Collapse
Affiliation(s)
- Kaixin Zheng
- Department of Applied Mathematics, University of Waterloo, Waterloo, Ontario, Canada
| | - Anita T Layton
- Department of Applied Mathematics, University of Waterloo, Waterloo, Ontario, Canada
- Department of Biology, Cheriton School of Computer Science, and School of Pharmacology, University of Waterloo, Waterloo, Ontario, Canada
| |
Collapse
|
32
|
Pacholko A, Iadecola C. Hypertension, Neurodegeneration, and Cognitive Decline. Hypertension 2024; 81:991-1007. [PMID: 38426329 PMCID: PMC11023809 DOI: 10.1161/hypertensionaha.123.21356] [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] [Indexed: 03/02/2024]
Abstract
Elevated blood pressure is a well-established risk factor for age-related cognitive decline. Long linked to cognitive impairment on vascular bases, increasing evidence suggests a potential association of hypertension with the neurodegenerative pathology underlying Alzheimer disease. Hypertension is well known to disrupt the structural and functional integrity of the cerebral vasculature. However, the mechanisms by which these alterations lead to brain damage, enhance Alzheimer pathology, and promote cognitive impairment remain to be established. Furthermore, critical questions concerning whether lowering blood pressure by antihypertensive medications prevents cognitive impairment have not been answered. Recent developments in neurovascular biology, brain imaging, and epidemiology, as well as new clinical trials, have provided insights into these critical issues. In particular, clinical and basic findings on the link between neurovascular dysfunction and the pathobiology of neurodegeneration have shed new light on the overlap between vascular and Alzheimer pathology. In this review, we will examine the progress made in the relationship between hypertension and cognitive impairment and, after a critical evaluation of the evidence, attempt to identify remaining knowledge gaps and future research directions that may advance our understanding of one of the leading health challenges of our time.
Collapse
Affiliation(s)
- Anthony Pacholko
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY
| | - Costantino Iadecola
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY
| |
Collapse
|
33
|
Fang G, Tian Y, Huang S, Zhang X, Liu Y, Li Y, Du J, Gao S. KLF15 maintains contractile phenotype of vascular smooth muscle cells and prevents thoracic aortic dissection by interacting with MRTFB. J Biol Chem 2024; 300:107260. [PMID: 38582447 PMCID: PMC11061230 DOI: 10.1016/j.jbc.2024.107260] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Revised: 03/26/2024] [Accepted: 03/28/2024] [Indexed: 04/08/2024] Open
Abstract
Thoracic aortic dissection (TAD) is a highly dangerous cardiovascular disorder caused by weakening of the aortic wall, resulting in a sudden tear of the internal face. Progressive loss of the contractile apparatus in vascular smooth muscle cells (VSMCs) is a major event in TAD. Exploring the endogenous regulators essential for the contractile phenotype of VSMCs may aid the development of strategies to prevent TAD. Krüppel-like factor 15 (KLF15) overexpression was reported to inhibit TAD formation; however, the mechanisms by which KLF15 prevents TAD formation and whether KLF15 regulates the contractile phenotype of VSMCs in TAD are not well understood. Therefore, we investigated these unknown aspects of KLF15 function. We found that KLF15 expression was reduced in human TAD samples and β-aminopropionitrile monofumarate-induced TAD mouse model. Klf15KO mice are susceptible to both β-aminopropionitrile monofumarate- and angiotensin II-induced TAD. KLF15 deficiency results in reduced VSMC contractility and exacerbated vascular inflammation and extracellular matrix degradation. Mechanistically, KLF15 interacts with myocardin-related transcription factor B (MRTFB), a potent serum response factor coactivator that drives contractile gene expression. KLF15 silencing represses the MRTFB-induced activation of contractile genes in VSMCs. Thus, KLF15 cooperates with MRTFB to promote the expression of contractile genes in VSMCs, and its dysfunction may exacerbate TAD. These findings indicate that KLF15 may be a novel therapeutic target for the treatment of TAD.
Collapse
MESH Headings
- Animals
- Humans
- Male
- Mice
- Angiotensin II/metabolism
- Angiotensin II/pharmacology
- Aortic Aneurysm, Thoracic/metabolism
- Aortic Aneurysm, Thoracic/genetics
- Aortic Aneurysm, Thoracic/pathology
- Dissection, Thoracic Aorta
- Kruppel-Like Transcription Factors/metabolism
- Kruppel-Like Transcription Factors/genetics
- Mice, Inbred C57BL
- Mice, Knockout
- Muscle Contraction/genetics
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Muscle, Smooth, Vascular/cytology
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Phenotype
- Transcription Factors/metabolism
- Transcription Factors/genetics
Collapse
Affiliation(s)
- Guangming Fang
- Collaborative Innovation Centre for Cardiovascular Disorders, The Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education, Beijing Anzhen Hospital, Beijing Institute of Heart, Lung and Blood Vessel Diseases, Capital Medical University, Beijing, China
| | - Yexuan Tian
- Collaborative Innovation Centre for Cardiovascular Disorders, The Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education, Beijing Anzhen Hospital, Beijing Institute of Heart, Lung and Blood Vessel Diseases, Capital Medical University, Beijing, China
| | - Shan Huang
- Collaborative Innovation Centre for Cardiovascular Disorders, The Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education, Beijing Anzhen Hospital, Beijing Institute of Heart, Lung and Blood Vessel Diseases, Capital Medical University, Beijing, China
| | - Xiaoping Zhang
- Collaborative Innovation Centre for Cardiovascular Disorders, The Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education, Beijing Anzhen Hospital, Beijing Institute of Heart, Lung and Blood Vessel Diseases, Capital Medical University, Beijing, China
| | - Yan Liu
- Collaborative Innovation Centre for Cardiovascular Disorders, The Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education, Beijing Anzhen Hospital, Beijing Institute of Heart, Lung and Blood Vessel Diseases, Capital Medical University, Beijing, China
| | - Yulin Li
- Collaborative Innovation Centre for Cardiovascular Disorders, The Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education, Beijing Anzhen Hospital, Beijing Institute of Heart, Lung and Blood Vessel Diseases, Capital Medical University, Beijing, China
| | - Jie Du
- Collaborative Innovation Centre for Cardiovascular Disorders, The Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education, Beijing Anzhen Hospital, Beijing Institute of Heart, Lung and Blood Vessel Diseases, Capital Medical University, Beijing, China.
| | - Shijuan Gao
- Collaborative Innovation Centre for Cardiovascular Disorders, The Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education, Beijing Anzhen Hospital, Beijing Institute of Heart, Lung and Blood Vessel Diseases, Capital Medical University, Beijing, China.
| |
Collapse
|
34
|
Hopper SE, Weiss D, Mikush N, Jiang B, Spronck B, Cavinato C, Humphrey JD, Figueroa CA. Central Artery Hemodynamics in Angiotensin II-Induced Hypertension and Effects of Anesthesia. Ann Biomed Eng 2024; 52:1051-1066. [PMID: 38383871 PMCID: PMC11418744 DOI: 10.1007/s10439-024-03440-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Accepted: 12/30/2023] [Indexed: 02/23/2024]
Abstract
Systemic hypertension is a strong risk factor for cardiovascular, neurovascular, and renovascular diseases. Central artery stiffness is both an initiator and indicator of hypertension, thus revealing a critical relationship between the wall mechanics and hemodynamics. Mice have emerged as a critical animal model for studying effects of hypertension and much has been learned. Regardless of the specific mouse model, data on changes in cardiac function and hemodynamics are necessarily measured under anesthesia. Here, we present a new experimental-computational workflow to estimate awake cardiovascular conditions from anesthetized data, which was then used to quantify effects of chronic angiotensin II-induced hypertension relative to normotension in wild-type mice. We found that isoflurane anesthesia had a greater impact on depressing hemodynamics in angiotensin II-infused mice than in controls, which led to unexpected results when comparing anesthetized results between the two groups of mice. Through comparison of the awake simulations, however, in vivo relevant effects of angiotensin II-infusion on global and regional vascular structure, properties, and hemodynamics were found to be qualitatively consistent with expectations. Specifically, we found an increased in vivo vascular stiffness in the descending thoracic aorta and suprarenal abdominal aorta, leading to increases in pulse pressure in the distal aorta. These insights allow characterization of the impact of regionally varying vascular remodeling on hemodynamics and mouse-to-mouse variations due to induced hypertension.
Collapse
Affiliation(s)
- S E Hopper
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - D Weiss
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA
| | - N Mikush
- Translational Research Imaging Center, Yale School of Medicine, New Haven, CT, USA
| | - B Jiang
- Department of Thyroid and Vascular Surgery, 1st Hospital of China Medical University, Shen Yang, China
| | - B Spronck
- Department of Biomedical Engineering, Maastricht University, Maastricht, The Netherlands
| | - C Cavinato
- LMGC, Universite' Montpellier, CNRS, Montpellier, France
| | - J D Humphrey
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA.
| | - C A Figueroa
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
- Department of Surgery, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
35
|
Fu Y, Xiang Y, Wei Q, Ilatovskaya D, Dong Z. Rodent models of AKI and AKI-CKD transition: an update in 2024. Am J Physiol Renal Physiol 2024; 326:F563-F583. [PMID: 38299215 PMCID: PMC11208034 DOI: 10.1152/ajprenal.00402.2023] [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: 12/13/2023] [Revised: 01/29/2024] [Accepted: 01/29/2024] [Indexed: 02/02/2024] Open
Abstract
Despite known drawbacks, rodent models are essential tools in the research of renal development, physiology, and pathogenesis. In the past decade, rodent models have been developed and used to mimic different etiologies of acute kidney injury (AKI), AKI to chronic kidney disease (CKD) transition or progression, and AKI with comorbidities. These models have been applied for both mechanistic research and preclinical drug development. However, current rodent models have their limitations, especially since they often do not fully recapitulate the pathophysiology of AKI in human patients, and thus need further refinement. Here, we discuss the present status of these rodent models, including the pathophysiologic compatibility, clinical translational significance, key factors affecting model consistency, and their main limitations. Future efforts should focus on establishing robust models that simulate the major clinical and molecular phenotypes of human AKI and its progression.
Collapse
Affiliation(s)
- Ying Fu
- Department of Nephrology, Institute of Nephrology, The Second Xiangya Hospital at Central South University, Changsha, People's Republic of China
| | - Yu Xiang
- Department of Nephrology, Institute of Nephrology, The Second Xiangya Hospital at Central South University, Changsha, People's Republic of China
| | - Qingqing Wei
- Department of Cellular Biology and Anatomy, Medical College of Georgia at Augusta University and Charlie Norwood Veterans Affairs Medical Center, Augusta, Georgia, United States
| | - Daria Ilatovskaya
- Department of Physiology, Medical College of Georgia at Augusta University, Augusta, Georgia, United States
| | - Zheng Dong
- Department of Nephrology, Institute of Nephrology, The Second Xiangya Hospital at Central South University, Changsha, People's Republic of China
- Department of Cellular Biology and Anatomy, Medical College of Georgia at Augusta University and Charlie Norwood Veterans Affairs Medical Center, Augusta, Georgia, United States
- Research Department, Charlie Norwood Veterans Affairs Medical Center, Augusta, Georgia, United States
| |
Collapse
|
36
|
Faraci FM, Scheer FA. Hypertension: Causes and Consequences of Circadian Rhythms in Blood Pressure. Circ Res 2024; 134:810-832. [PMID: 38484034 PMCID: PMC10947115 DOI: 10.1161/circresaha.124.323515] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Accepted: 02/16/2024] [Indexed: 03/19/2024]
Abstract
Hypertension is extremely common, affecting approximately 1 in every 2 adults globally. Chronic hypertension is the leading modifiable risk factor for cardiovascular disease and premature mortality worldwide. Despite considerable efforts to define mechanisms that underlie hypertension, a potentially major component of the disease, the role of circadian biology has been relatively overlooked in both preclinical models and humans. Although the presence of daily and circadian patterns has been observed from the level of the genome to the whole organism, the functional and structural impact of biological rhythms, including mechanisms such as circadian misalignment, remains relatively poorly defined. Here, we review the impact of daily rhythms and circadian systems in regulating blood pressure and the onset, progression, and consequences of hypertension. There is an emphasis on the impact of circadian biology in relation to vascular disease and end-organ effects that, individually or in combination, contribute to complex phenotypes such as cognitive decline and the loss of cardiac and brain health. Despite effective treatment options for some individuals, control of blood pressure remains inadequate in a substantial portion of the hypertensive population. Greater insight into circadian biology may form a foundation for novel and more widely effective molecular therapies or interventions to help in the prevention, treatment, and management of hypertension and its related pathophysiology.
Collapse
Affiliation(s)
- Frank M. Faraci
- Department of Internal Medicine, Francois M. Abboud Cardiovascular Center, Carver College of Medicine, University of Iowa, Iowa City, IA 52242-1081
- Department of Neuroscience and Pharmacology, Francois M. Abboud Cardiovascular Center, Carver College of Medicine, University of Iowa, Iowa City, IA 52242-1081
| | - Frank A.J.L. Scheer
- Division of Sleep Medicine, Harvard Medical School, Brigham and Women's Hospital, Boston, Massachusetts, 02115
- Medical Chronobiology Program, Division of Sleep and Circadian Disorders, Departments of Medicine and Neurology, Brigham and Women's Hospital, Boston, Massachusetts, 02115
| |
Collapse
|
37
|
Carnevale L, Perrotta M, Mastroiacovo F, Perrotta S, Migliaccio A, Fardella V, Pacella J, Fardella S, Pallante F, Carnevale R, Carnevale D, Lembo G. Advanced Magnetic Resonance Imaging to Define the Microvascular Injury Driven by Neuroinflammation in the Brain of a Mouse Model of Hypertension. Hypertension 2024; 81:636-647. [PMID: 38174566 DOI: 10.1161/hypertensionaha.123.21940] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Accepted: 12/20/2023] [Indexed: 01/05/2024]
Abstract
BACKGROUND Hypertension is one of the main risk factors for dementia and cognitive impairment. METHODS We used the model of transverse aortic constriction to induce chronic pressure overload in mice. We characterized brain injury by advanced translational applications of magnetic resonance imaging. In parallel, we analyzed peripheral target organ damage induced by chronic pressure overload by ultrasonography. Microscopical characterization of brain vasculature was performed as well, together with the analysis of immune and inflammatory markers. RESULTS We identified a specific structural, microstructural, and functional brain injury. In particular, we highlighted a regional enlargement of the hypothalamus, microstructural damage in the white matter of the fimbria, and a reduction of the cerebral blood flow. A parallel analysis performed by confocal microscopy revealed a correspondent tissue damage evidenced by a reduction of cerebral capillary density, paired with loss of pericyte coverage. We assessed cognitive impairment and cardiac damage induced by hypertension to perform correlation analyses with the brain injury severity. At the mechanistic level, we found that CD8+T cells, producing interferon-γ, infiltrated the brain of hypertensive mice. By neutralizing this proinflammatory cytokine, we obtained a rescue of the phenotype, demonstrating their crucial role in establishing the microvascular damage. CONCLUSIONS Overall, we have used translational tools to comprehensively characterize brain injury in a mouse model of hypertension induced by chronic pressure overload. We have identified early cerebrovascular damage in hypertensive mice, sustained by CD8+IFN-γ+T lymphocytes, which fuel neuroinflammation to establish the injury of brain capillaries.
Collapse
Affiliation(s)
- Lorenzo Carnevale
- Department of Angiocardioneurology and Translational Medicine, IRCCS INM Neuromed, Pozzilli, Italy (L.C., M.P., F.M., S.P., A.M., V.F., J.P., S.F., F.P., R.C., D.C., G.L.)
| | - Marialuisa Perrotta
- Department of Angiocardioneurology and Translational Medicine, IRCCS INM Neuromed, Pozzilli, Italy (L.C., M.P., F.M., S.P., A.M., V.F., J.P., S.F., F.P., R.C., D.C., G.L.)
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy (M.P., D.C., G.L.)
| | - Francesco Mastroiacovo
- Department of Angiocardioneurology and Translational Medicine, IRCCS INM Neuromed, Pozzilli, Italy (L.C., M.P., F.M., S.P., A.M., V.F., J.P., S.F., F.P., R.C., D.C., G.L.)
| | - Sara Perrotta
- Department of Angiocardioneurology and Translational Medicine, IRCCS INM Neuromed, Pozzilli, Italy (L.C., M.P., F.M., S.P., A.M., V.F., J.P., S.F., F.P., R.C., D.C., G.L.)
| | - Agnese Migliaccio
- Department of Angiocardioneurology and Translational Medicine, IRCCS INM Neuromed, Pozzilli, Italy (L.C., M.P., F.M., S.P., A.M., V.F., J.P., S.F., F.P., R.C., D.C., G.L.)
| | - Valentina Fardella
- Department of Angiocardioneurology and Translational Medicine, IRCCS INM Neuromed, Pozzilli, Italy (L.C., M.P., F.M., S.P., A.M., V.F., J.P., S.F., F.P., R.C., D.C., G.L.)
| | - Jacopo Pacella
- Department of Angiocardioneurology and Translational Medicine, IRCCS INM Neuromed, Pozzilli, Italy (L.C., M.P., F.M., S.P., A.M., V.F., J.P., S.F., F.P., R.C., D.C., G.L.)
| | - Stefania Fardella
- Department of Angiocardioneurology and Translational Medicine, IRCCS INM Neuromed, Pozzilli, Italy (L.C., M.P., F.M., S.P., A.M., V.F., J.P., S.F., F.P., R.C., D.C., G.L.)
| | - Fabio Pallante
- Department of Angiocardioneurology and Translational Medicine, IRCCS INM Neuromed, Pozzilli, Italy (L.C., M.P., F.M., S.P., A.M., V.F., J.P., S.F., F.P., R.C., D.C., G.L.)
| | - Raimondo Carnevale
- Department of Angiocardioneurology and Translational Medicine, IRCCS INM Neuromed, Pozzilli, Italy (L.C., M.P., F.M., S.P., A.M., V.F., J.P., S.F., F.P., R.C., D.C., G.L.)
| | - Daniela Carnevale
- Department of Angiocardioneurology and Translational Medicine, IRCCS INM Neuromed, Pozzilli, Italy (L.C., M.P., F.M., S.P., A.M., V.F., J.P., S.F., F.P., R.C., D.C., G.L.)
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy (M.P., D.C., G.L.)
| | - Giuseppe Lembo
- Department of Angiocardioneurology and Translational Medicine, IRCCS INM Neuromed, Pozzilli, Italy (L.C., M.P., F.M., S.P., A.M., V.F., J.P., S.F., F.P., R.C., D.C., G.L.)
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy (M.P., D.C., G.L.)
| |
Collapse
|
38
|
Pal S, Sharma S, Porwal K, Tiwari MC, Khan YA, Kumar S, Kumar N, Chattopadhyay N. The Role of Osteogenic Effect and Vascular Function in Bone Health in Hypertensive Rats: A Study of Anti-hypertensive and Hemorheologic Drugs. Calcif Tissue Int 2024; 114:295-309. [PMID: 38102510 DOI: 10.1007/s00223-023-01170-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2023] [Accepted: 11/22/2023] [Indexed: 12/17/2023]
Abstract
Vascular dysfunction contributes to the development of osteopenia in hypertensive patients, as decreased blood supply to bones results in tissue damage and dysfunction. The effect of anti-hypertensive medicines on bone mass in hypertensive individuals is inconclusive because of the varied mechanism of their action, and suggests that reducing blood pressure (BP) alone is insufficient to enhance bone mass in hypertension. Pentoxifylline (PTX), a hemorheological drug, improves blood flow by reducing blood viscosity and angiogenesis, also has an osteogenic effect. We hypothesized that improving vascular function is critical to increasing bone mass in hypertension. To test this, we screened various anti-hypertensive drugs for their in vitro osteogenic effect, from which timolol and hydralazine were selected. In adult female spontaneously hypertensive rats (SHRs), timolol and hydralazine did not improve vascular function and bone mass, but PTX improved both. In female SHR animals, PTX restored bone mass, strength and mineralization, up to the level of normotensive control rats. In addition, we observed lower blood vasculature in the femur of adult SHR animals, and PTX restored them. PTX also restored the bone vascular and angiogenesis parameters that had been impaired in OVX SHR compared to sham SHR. This study demonstrates the importance of vascular function in addition to increased bone mass for improving bone health as achieved by PTX without affecting BP, and suggests a promising treatment option for osteoporosis in hypertensive patients, particularly at-risk postmenopausal women.
Collapse
Affiliation(s)
- Subhashis Pal
- Division of Endocrinology, CSIR-Central Drug Research Institute, Council of Scientific and Industrial Research, Lucknow, 226031, India
| | - Shivani Sharma
- Division of Endocrinology, CSIR-Central Drug Research Institute, Council of Scientific and Industrial Research, Lucknow, 226031, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Konica Porwal
- Division of Endocrinology, CSIR-Central Drug Research Institute, Council of Scientific and Industrial Research, Lucknow, 226031, India
| | - Mahesh C Tiwari
- Division of Endocrinology, CSIR-Central Drug Research Institute, Council of Scientific and Industrial Research, Lucknow, 226031, India
| | - Yasir A Khan
- Division of Endocrinology, CSIR-Central Drug Research Institute, Council of Scientific and Industrial Research, Lucknow, 226031, India
| | - Saroj Kumar
- Department of Mechanical Engineering, Indian Institute of Technology Ropar, Rupnagar, 140001, Punjab, India
| | - Navin Kumar
- Department of Mechanical Engineering, Indian Institute of Technology Ropar, Rupnagar, 140001, Punjab, India
| | - Naibedya Chattopadhyay
- Division of Endocrinology, CSIR-Central Drug Research Institute, Council of Scientific and Industrial Research, Lucknow, 226031, India.
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India.
- Division of Endocrinology, CSIR-Central Drug Research Institute, Sector 10, Jankipuram Extension, Sitapur Road, Lucknow, 226 031, India.
| |
Collapse
|
39
|
Norambuena-Soto I, Deng Y, Brenner C, Lavandero S, Wang ZV. NAD in pathological cardiac remodeling: Metabolic regulation and beyond. Biochim Biophys Acta Mol Basis Dis 2024; 1870:167038. [PMID: 38281710 PMCID: PMC10922927 DOI: 10.1016/j.bbadis.2024.167038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2023] [Revised: 01/05/2024] [Accepted: 01/19/2024] [Indexed: 01/30/2024]
Abstract
Nicotinamide adenine dinucleotide (NAD) coenzymes are carriers of high energy electrons in metabolism and also play critical roles in numerous signaling pathways. NAD metabolism is decreased in various cardiovascular diseases. Importantly, stimulation of NAD biosynthesis protects against heart disease under different pathological conditions. In this review, we describe pathways for both generation and catabolism of NAD coenzymes and the respective changes of these pathways in the heart under cardiac diseases, including pressure overload, myocardial infarction, cardiometabolic disease, cancer treatment cardiotoxicity, and heart failure. We next provide an update on the strategies and treatments to increase NAD levels, such as supplementation of NAD precursors, in the heart that prevent or reverse cardiomyopathy. We also introduce the approaches to manipulate NAD consumption enzymes to ameliorate cardiac disease. Finally, we discuss the mechanisms associated with improvements in cardiac function by NAD coenzymes, differentiating between mitochondria-dependent effects and those independent of mitochondrial metabolism.
Collapse
Affiliation(s)
- Ignacio Norambuena-Soto
- Department of Diabetes and Cancer Metabolism, Beckman Research Institute, City of Hope National Medical Center, Duarte, CA 91010, USA; Advanced Center for Chronic Diseases (ACCDiS), Facultad Ciencias Químicas y Farmacéuticas & Facultad Medicina, Universidad de Chile, Santiago 8380494, Chile
| | - Yingfeng Deng
- Department of Diabetes and Cancer Metabolism, Beckman Research Institute, City of Hope National Medical Center, Duarte, CA 91010, USA
| | - Charles Brenner
- Department of Diabetes and Cancer Metabolism, Beckman Research Institute, City of Hope National Medical Center, Duarte, CA 91010, USA
| | - Sergio Lavandero
- Advanced Center for Chronic Diseases (ACCDiS), Facultad Ciencias Químicas y Farmacéuticas & Facultad Medicina, Universidad de Chile, Santiago 8380494, Chile; Cardiology Division, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390-8573, USA.
| | - Zhao V Wang
- Department of Diabetes and Cancer Metabolism, Beckman Research Institute, City of Hope National Medical Center, Duarte, CA 91010, USA.
| |
Collapse
|
40
|
Gong T, Mu Q, Xu Y, Wang W, Meng L, Feng X, Liu W, Ao Z, Zhang Y, Chen X, Xu H. Expression of the umami taste receptor T1R1/T1R3 in porcine testis of: Function in regulating testosterone synthesis and autophagy in Leydig cells. J Steroid Biochem Mol Biol 2024; 236:106429. [PMID: 38035949 DOI: 10.1016/j.jsbmb.2023.106429] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Revised: 10/31/2023] [Accepted: 11/17/2023] [Indexed: 12/02/2023]
Abstract
Testosterone is a vital male hormone responsible for male sexual characteristics. The taste receptor family 1 subunit 3 (T1R3) regulates testosterone synthesis and autophagy in non-taste cells, and the links with the taste receptor family 1 subunit 1 (T1R1) for umami perception. However, little is known about these mechanisms. Thus, we aimed to determine the relationship between the umami taste receptor (T1R1/T1R3) and testosterone synthesis or autophagy in testicular Leydig cells of the Xiang pig. There was a certain proportion of spermatogenic tubular dysplasia in the Xiang pig at puberty, in which autophagy was enhanced, and the testosterone level was increased with a weak expression of T1R3. Silenced T1R3 decreased testosterone level and intracellular cyclic adenosine monophosphate (cAMP) content and inhibited the messenger RNA (mRNA) expression levels of testosterone synthesis enzyme genes [steroidogenic acute regulatory protein (StAR), hydroxy-delta-5-steroid dehydrogenase, 3 beta- and steroid delta-isomerase 1 (3β-HSD1), cytochrome P450 family 17 subfamily A member 1 (CYP17A1) and hydroxysteroid 17-beta dehydrogenase 3 (17β-HSD3)]. In addition, T1R3 increased the number of acidic autophagy bubbles and upregulated the expression levels of autophagy markers [Microtubule-associated protein 1 A/1B-light chain 3 (LC3) and Beclin-1] in testicular Leydig cells of the Xiang pig. Using an umami tasting agonist (10 mM L-glutamate for 6 h), the activation of T1R1/T1R3 enhanced the testosterone synthesis ability by increasing the intracellular cAMP level and upregulated the expression levels of StAR, 3β-HSD1, CYP17A1 and 17β-HSD3 in Leydig cells. Furthermore, the number of acidic autophagy bubbles decreased in the T1R1/T1R3-activated group with the downregulation of the expression levels of the autophagy markers, including LC3 and Beclin-1. These data suggest that the function of T1R1/T1R3 expressed in testicular Leydig cells of the Xiang pig is related to testosterone synthesis and autophagy.
Collapse
Affiliation(s)
- Ting Gong
- Key Laboratory of Animal Genetics, Breeding and Reproduction in The Plateau Mountainous Region, Ministry of Education, Guizhou University, Guiyang 550025, Guizhou, PR China; Guizhou Provincial Key Laboratory of Animal Genetics, Breeding and Reproduction, Guizhou University, Guiyang 550025, Guizhou, PR China; College of Animal Science, Guizhou University, Guiyang 550025, Guizhou, PR China.
| | - Qi Mu
- Key Laboratory of Animal Genetics, Breeding and Reproduction in The Plateau Mountainous Region, Ministry of Education, Guizhou University, Guiyang 550025, Guizhou, PR China; Guizhou Provincial Key Laboratory of Animal Genetics, Breeding and Reproduction, Guizhou University, Guiyang 550025, Guizhou, PR China; College of Animal Science, Guizhou University, Guiyang 550025, Guizhou, PR China
| | - Yongjian Xu
- Key Laboratory of Animal Genetics, Breeding and Reproduction in The Plateau Mountainous Region, Ministry of Education, Guizhou University, Guiyang 550025, Guizhou, PR China; Qiannan Buyi and Miao Autonomous Prefecture Bureau of Agriculture and Rural Affairs, PR China
| | - Weiyong Wang
- Key Laboratory of Animal Genetics, Breeding and Reproduction in The Plateau Mountainous Region, Ministry of Education, Guizhou University, Guiyang 550025, Guizhou, PR China; Guizhou Provincial Key Laboratory of Animal Genetics, Breeding and Reproduction, Guizhou University, Guiyang 550025, Guizhou, PR China; College of Animal Science, Guizhou University, Guiyang 550025, Guizhou, PR China
| | - Lijie Meng
- Key Laboratory of Animal Genetics, Breeding and Reproduction in The Plateau Mountainous Region, Ministry of Education, Guizhou University, Guiyang 550025, Guizhou, PR China; Guizhou Provincial Key Laboratory of Animal Genetics, Breeding and Reproduction, Guizhou University, Guiyang 550025, Guizhou, PR China; College of Animal Science, Guizhou University, Guiyang 550025, Guizhou, PR China
| | - Xianzhou Feng
- Key Laboratory of Animal Genetics, Breeding and Reproduction in The Plateau Mountainous Region, Ministry of Education, Guizhou University, Guiyang 550025, Guizhou, PR China; Guizhou Provincial Key Laboratory of Animal Genetics, Breeding and Reproduction, Guizhou University, Guiyang 550025, Guizhou, PR China; College of Animal Science, Guizhou University, Guiyang 550025, Guizhou, PR China
| | - Wenjiao Liu
- Key Laboratory of Animal Genetics, Breeding and Reproduction in The Plateau Mountainous Region, Ministry of Education, Guizhou University, Guiyang 550025, Guizhou, PR China; Guizhou Provincial Key Laboratory of Animal Genetics, Breeding and Reproduction, Guizhou University, Guiyang 550025, Guizhou, PR China; College of Animal Science, Guizhou University, Guiyang 550025, Guizhou, PR China
| | - Zheng Ao
- Key Laboratory of Animal Genetics, Breeding and Reproduction in The Plateau Mountainous Region, Ministry of Education, Guizhou University, Guiyang 550025, Guizhou, PR China; Guizhou Provincial Key Laboratory of Animal Genetics, Breeding and Reproduction, Guizhou University, Guiyang 550025, Guizhou, PR China; College of Animal Science, Guizhou University, Guiyang 550025, Guizhou, PR China
| | - Yiyu Zhang
- Key Laboratory of Animal Genetics, Breeding and Reproduction in The Plateau Mountainous Region, Ministry of Education, Guizhou University, Guiyang 550025, Guizhou, PR China; Guizhou Provincial Key Laboratory of Animal Genetics, Breeding and Reproduction, Guizhou University, Guiyang 550025, Guizhou, PR China; College of Animal Science, Guizhou University, Guiyang 550025, Guizhou, PR China
| | - Xiang Chen
- Key Laboratory of Animal Genetics, Breeding and Reproduction in The Plateau Mountainous Region, Ministry of Education, Guizhou University, Guiyang 550025, Guizhou, PR China; Guizhou Provincial Key Laboratory of Animal Genetics, Breeding and Reproduction, Guizhou University, Guiyang 550025, Guizhou, PR China; College of Animal Science, Guizhou University, Guiyang 550025, Guizhou, PR China
| | - Houqiang Xu
- Key Laboratory of Animal Genetics, Breeding and Reproduction in The Plateau Mountainous Region, Ministry of Education, Guizhou University, Guiyang 550025, Guizhou, PR China; Guizhou Provincial Key Laboratory of Animal Genetics, Breeding and Reproduction, Guizhou University, Guiyang 550025, Guizhou, PR China; College of Animal Science, Guizhou University, Guiyang 550025, Guizhou, PR China
| |
Collapse
|
41
|
Ray A, Stelloh C, Liu Y, Meyer A, Geurts AM, Cowley AW, Greene AS, Liang M, Rao S. Histone Modifications and Their Contributions to Hypertension. Hypertension 2024; 81:229-239. [PMID: 38031837 PMCID: PMC11229175 DOI: 10.1161/hypertensionaha.123.21755] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2023]
Abstract
Essential hypertension, a multifaceted disorder, is a worldwide health problem. A complex network of genetic, epigenetic, physiological, and environmental components regulates blood pressure (BP), and any dysregulation of this network may result in hypertension. Growing evidence suggests a role for epigenetic factors in BP regulation. Any alterations in the expression or functions of these epigenetic regulators may dysregulate various determinants of BP, thereby promoting the development of hypertension. Histone posttranslational modifications are critical epigenetic regulators that have been implicated in hypertension. Several studies have demonstrated a clear association between the increased expression of some histone-modifying enzymes, especially HDACs (histone deacetylases), and hypertension. In addition, treatment with HDAC inhibitors lowers BP in hypertensive animal models, providing an excellent opportunity to design new drugs to treat hypertension. In this review, we discuss the potential contribution of different histone modifications to the regulation of BP.
Collapse
Affiliation(s)
- Atrayee Ray
- Versiti Blood Research Institute, Milwaukee, WI (A.R., C.S., A.M., S.R.)
- Department of Physiology, Center of Systems Molecular Medicine (A.R., A.M.G., A.C.), Medical College of Wisconsin, Milwaukee
| | - Cary Stelloh
- Versiti Blood Research Institute, Milwaukee, WI (A.R., C.S., A.M., S.R.)
| | - Yong Liu
- Department of Physiology, The University of Arizona, Tucson (Y.L., M.L.)
| | - Alison Meyer
- Versiti Blood Research Institute, Milwaukee, WI (A.R., C.S., A.M., S.R.)
| | - Aron M Geurts
- Department of Physiology, Center of Systems Molecular Medicine (A.R., A.M.G., A.C.), Medical College of Wisconsin, Milwaukee
| | - Allen W Cowley
- Department of Physiology, Center of Systems Molecular Medicine (A.R., A.M.G., A.C.), Medical College of Wisconsin, Milwaukee
| | | | - Mingyu Liang
- Department of Physiology, The University of Arizona, Tucson (Y.L., M.L.)
| | - Sridhar Rao
- Versiti Blood Research Institute, Milwaukee, WI (A.R., C.S., A.M., S.R.)
- Department of Pediatrics, Section of Hematology/Oncology/Transplantation (S.R.), Medical College of Wisconsin, Milwaukee
| |
Collapse
|
42
|
Toczek M, Schlicker E, Remiszewski P, Malinowska B. Function of Presynaptic Inhibitory Cannabinoid CB 1 Receptors in Spontaneously Hypertensive Rats and Its Modification by Enhanced Endocannabinoid Tone. Int J Mol Sci 2024; 25:858. [PMID: 38255931 PMCID: PMC10815615 DOI: 10.3390/ijms25020858] [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: 12/07/2023] [Revised: 01/04/2024] [Accepted: 01/08/2024] [Indexed: 01/24/2024] Open
Abstract
We studied whether the function of presynaptic inhibitory cannabinoid CB1 receptors on the sympathetic nerve fibres innervating resistance vessels is increased in spontaneously hypertensive rats (SHR) like in deoxycorticosterone (DOCA)-salt hypertension. An increase in diastolic blood pressure (DBP) was induced by electrical stimulation of the preganglionic sympathetic neurons or by phenylephrine injection in pithed SHR and normotensive Wistar-Kyoto rats (WKY). The electrically (but not the phenylephrine) induced increase in DBP was inhibited by the cannabinoid receptor agonist CP55940, similarly in both groups, and by the endocannabinoid reuptake inhibitor AM404 in SHR only. The effect of CP55940 was abolished/reduced by the CB1 receptor antagonist AM251 (in both groups) and in WKY by endocannabinoid degradation blockade, i.e., the monoacylglycerol lipase (MAGL) inhibitor MJN110 and the dual fatty acid amide hydrolase (FAAH)/MAGL inhibitor JZL195 but not the FAAH inhibitor URB597. MJN110 and JZL195 tended to enhance the effect of CP55940 in SHR. In conclusion, the function of presynaptic inhibitory CB1 receptors depends on the hypertension model. Although no differences occurred between SHR and WKY under basal experimental conditions, the CB1 receptor function was better preserved in SHR when the endocannabinoid tone was increased by the inhibition of MAGL or the endocannabinoid transporter.
Collapse
Affiliation(s)
- Marek Toczek
- Department of Experimental Physiology and Pathophysiology, Medical University of Białystok, Mickiewicza Str. 2A, 15-222 Białystok, Poland; (P.R.); (B.M.)
| | - Eberhard Schlicker
- Department of Pharmacology and Toxicology, University of Bonn, Venusberg-Campus 1, 53127 Bonn, Germany;
| | - Patryk Remiszewski
- Department of Experimental Physiology and Pathophysiology, Medical University of Białystok, Mickiewicza Str. 2A, 15-222 Białystok, Poland; (P.R.); (B.M.)
| | - Barbara Malinowska
- Department of Experimental Physiology and Pathophysiology, Medical University of Białystok, Mickiewicza Str. 2A, 15-222 Białystok, Poland; (P.R.); (B.M.)
| |
Collapse
|
43
|
Drury ER, Wu J, Gigliotti JC, Le TH. Sex differences in blood pressure regulation and hypertension: renal, hemodynamic, and hormonal mechanisms. Physiol Rev 2024; 104:199-251. [PMID: 37477622 PMCID: PMC11281816 DOI: 10.1152/physrev.00041.2022] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Revised: 06/06/2023] [Accepted: 07/16/2023] [Indexed: 07/22/2023] Open
Abstract
The teleology of sex differences has been argued since at least as early as Aristotle's controversial Generation of Animals more than 300 years BC, which reflects the sex bias of the time to contemporary readers. Although the question "why are the sexes different" remains a topic of debate in the present day in metaphysics, the recent emphasis on sex comparison in research studies has led to the question "how are the sexes different" being addressed in health science through numerous observational studies in both health and disease susceptibility, including blood pressure regulation and hypertension. These efforts have resulted in better understanding of differences in males and females at the molecular level that partially explain their differences in vascular function and renal sodium handling and hence blood pressure and the consequential cardiovascular and kidney disease risks in hypertension. This review focuses on clinical studies comparing differences between men and women in blood pressure over the life span and response to dietary sodium and highlights experimental models investigating sexual dimorphism in the renin-angiotensin-aldosterone, vascular, sympathetic nervous, and immune systems, endothelin, the major renal sodium transporters/exchangers/channels, and the impact of sex hormones on these systems in blood pressure homeostasis. Understanding the mechanisms governing sex differences in blood pressure regulation could guide novel therapeutic approaches in a sex-specific manner to lower cardiovascular risks in hypertension and advance personalized medicine.
Collapse
Affiliation(s)
- Erika R Drury
- Division of Nephrology, Department of Medicine, University of Rochester Medical Center, Rochester, New York, United States
| | - Jing Wu
- Division of Nephrology, Department of Medicine, University of Rochester Medical Center, Rochester, New York, United States
- Department of Pharmacology and Physiology, University of Rochester Medical Center, Rochester, New York, United States
| | - Joseph C Gigliotti
- Department of Integrative Physiology and Pharmacology, Liberty University College of Osteopathic Medicine, Lynchburg, Virginia, United States
| | - Thu H Le
- Division of Nephrology, Department of Medicine, University of Rochester Medical Center, Rochester, New York, United States
| |
Collapse
|
44
|
Baggeroer CE, Cambronero FE, Savan NA, Jefferson AL, Santisteban MM. Basic Mechanisms of Brain Injury and Cognitive Decline in Hypertension. Hypertension 2024; 81:34-44. [PMID: 37732479 PMCID: PMC10840624 DOI: 10.1161/hypertensionaha.123.19939] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/22/2023]
Abstract
Dementia affects almost 50 million adults worldwide, and remains a major cause of death and disability. Hypertension is a leading risk factor for dementia, including Alzheimer disease and Alzheimer disease-related dementias. Although this association is well-established, the mechanisms underlying hypertension-induced cognitive decline remain poorly understood. By exploring the mechanisms mediating the detrimental effects of hypertension on the brain, studies have aimed to provide therapeutic insights and strategies on how to protect the brain from the effects of blood pressure elevation. In this review, we focus on the basic mechanisms contributing to the cerebrovascular adaptions to elevated blood pressure and hypertension-induced microvascular injury. We also assess the cellular mechanisms of neurovascular unit dysfunction, focusing on the premise that cognitive impairment ensues when the dynamic metabolic demands of neurons are not met due to neurovascular uncoupling, and summarize cognitive deficits across various rodent models of hypertension as a resource for investigators. Despite significant advances in antihypertensive therapy, hypertension remains a critical risk factor for cognitive decline, and several questions remain about the development and progression of hypertension-induced cognitive impairment.
Collapse
Affiliation(s)
- Caroline E. Baggeroer
- Vanderbilt Memory and Alzheimer’s Center, Vanderbilt University Medical Center, Nashville, TN
| | - Francis E. Cambronero
- Vanderbilt Memory and Alzheimer’s Center, Vanderbilt University Medical Center, Nashville, TN
| | - N. Anna Savan
- Medical Scientist Training Program, Yale University, New Haven, CT
| | - Angela L. Jefferson
- Vanderbilt Memory and Alzheimer’s Center, Vanderbilt University Medical Center, Nashville, TN
- Department of Neurology, Vanderbilt University Medical Center, Nashville, TN
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN
| | - Monica M. Santisteban
- Vanderbilt Memory and Alzheimer’s Center, Vanderbilt University Medical Center, Nashville, TN
- Department of Neurology, Vanderbilt University Medical Center, Nashville, TN
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN
| |
Collapse
|
45
|
Wilkinson CM, Kalisvaart AC, Kung TF, Abrahart AH, Khiabani E, Colbourne F. Tissue Compliance and Intracranial Pressure Responses to Large Intracerebral Hemorrhage in Young and Aged Spontaneously Hypertensive Rats. Hypertension 2024; 81:151-161. [PMID: 37909235 PMCID: PMC10734784 DOI: 10.1161/hypertensionaha.123.21628] [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: 06/08/2023] [Accepted: 10/12/2023] [Indexed: 11/02/2023]
Abstract
BACKGROUND After a large intracerebral hemorrhage (ICH), the hematoma and swelling cause intracranial pressure (ICP) to increase, sometimes causing brain herniation and death. This is partly countered by widespread tissue compliance, an acute decrease in tissue volume distal to the stroke, at least in young healthy animals. Intracranial compensation dynamics seem to vary with age, but there is no data on old animals or those with hypertension, major factors influencing ICH risk and outcome. METHODS We assessed hematoma volume, edema, ICP, and functional deficits in young and aged spontaneously hypertensive rats (SHRs) and young normotensive control strains after collagenase-induced ICH. Macroscopic and microscopic brain volume fractions, such as contralateral hemisphere volume, cortical thickness, and neuronal morphology, were assessed via histological and stereological techniques. RESULTS Hematoma volume was 52% larger in young versus aged SHRs; surprisingly, aged SHRs still experienced proportionally worse outcomes following ICH, with 2× greater elevations in edema and ICP relative to bleed volume and 3× the degree of tissue compliance. Aged SHRs also experienced equivalent neurological deficits following ICH compared with their younger counterparts, despite the lack of significant age-related behavioral effects. Importantly, tissue compliance occurred across strains and age groups and was not impaired by hypertension or old age. CONCLUSIONS Aged SHRs show considerable capacity for tissue compliance following ICH and seem to rely on such mechanisms more heavily in settings of elevated ICP. Therefore, the ICP compensation response to ICH mass effect varies across the lifespan according to risk factors such as chronic hypertension.
Collapse
Affiliation(s)
- Cassandra M. Wilkinson
- Department of Psychology (C.M.W., A.C.J.K., T.F.C.K., A.H.A., E.K., F.C.), University of Alberta, Edmonton, Canada
| | - Anna C.J. Kalisvaart
- Department of Psychology (C.M.W., A.C.J.K., T.F.C.K., A.H.A., E.K., F.C.), University of Alberta, Edmonton, Canada
| | - Tiffany F.C. Kung
- Department of Psychology (C.M.W., A.C.J.K., T.F.C.K., A.H.A., E.K., F.C.), University of Alberta, Edmonton, Canada
| | - Ashley H. Abrahart
- Department of Psychology (C.M.W., A.C.J.K., T.F.C.K., A.H.A., E.K., F.C.), University of Alberta, Edmonton, Canada
| | - Elmira Khiabani
- Department of Psychology (C.M.W., A.C.J.K., T.F.C.K., A.H.A., E.K., F.C.), University of Alberta, Edmonton, Canada
| | - Frederick Colbourne
- Department of Psychology (C.M.W., A.C.J.K., T.F.C.K., A.H.A., E.K., F.C.), University of Alberta, Edmonton, Canada
- Neuroscience and Mental Health Institute (F.C.), University of Alberta, Edmonton, Canada
| |
Collapse
|
46
|
Seropian IM, Cassaglia P, Miksztowicz V, González GE. Unraveling the role of galectin-3 in cardiac pathology and physiology. Front Physiol 2023; 14:1304735. [PMID: 38170009 PMCID: PMC10759241 DOI: 10.3389/fphys.2023.1304735] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Accepted: 12/05/2023] [Indexed: 01/05/2024] Open
Abstract
Galectin-3 (Gal-3) is a carbohydrate-binding protein with multiple functions. Gal-3 regulates cell growth, proliferation, and apoptosis by orchestrating cell-cell and cell-matrix interactions. It is implicated in the development and progression of cardiovascular disease, and its expression is increased in patients with heart failure. In atherosclerosis, Gal-3 promotes monocyte recruitment to the arterial wall boosting inflammation and atheroma. In acute myocardial infarction (AMI), the expression of Gal-3 increases in infarcted and remote zones from the beginning of AMI, and plays a critical role in macrophage infiltration, differentiation to M1 phenotype, inflammation and interstitial fibrosis through collagen synthesis. Genetic deficiency of Gal-3 delays wound healing, impairs cardiac remodeling and function after AMI. On the contrary, Gal-3 deficiency shows opposite results with improved remodeling and function in other cardiomyopathies and in hypertension. Pharmacologic inhibition with non-selective inhibitors is also protective in cardiac disease. Finally, we recently showed that Gal-3 participates in normal aging. However, genetic absence of Gal-3 in aged mice exacerbates pathological hypertrophy and increases fibrosis, as opposed to reduced fibrosis shown in cardiac disease. Despite some gaps in understanding its precise mechanisms of action, Gal-3 represents a potential therapeutic target for the treatment of cardiovascular diseases and the management of cardiac aging. In this review, we summarize the current knowledge regarding the role of Gal-3 in the pathophysiology of heart failure, atherosclerosis, hypertension, myocarditis, and ischemic heart disease. Furthermore, we describe the physiological role of Gal-3 in cardiac aging.
Collapse
Affiliation(s)
- Ignacio M. Seropian
- Laboratorio de Patología Cardiovascular Experimental e Hipertensión Arterial, Instituto de Investigaciones Biomédicas (UCA-CONICET), Facultad de Ciencias Médicas Universidad Católica Argentina, Buenos Aires, Argentina
- Servicio de Hemodinamia, Hospital Italiano de Buenos Aires, Buenos Aires, Argentina
| | - Pablo Cassaglia
- Departamento de Patología, Instituto de Salud Comunitaria, Universidad Nacional de Hurlingham, Buenos Aires, Argentina
| | - Verónica Miksztowicz
- Laboratorio de Patología Cardiovascular Experimental e Hipertensión Arterial, Instituto de Investigaciones Biomédicas (UCA-CONICET), Facultad de Ciencias Médicas Universidad Católica Argentina, Buenos Aires, Argentina
| | - Germán E. González
- Laboratorio de Patología Cardiovascular Experimental e Hipertensión Arterial, Instituto de Investigaciones Biomédicas (UCA-CONICET), Facultad de Ciencias Médicas Universidad Católica Argentina, Buenos Aires, Argentina
- Departamento de Patología, Instituto de Salud Comunitaria, Universidad Nacional de Hurlingham, Buenos Aires, Argentina
| |
Collapse
|
47
|
Sams A, Haanes KA, Holm A, Kazantzi S, Mikkelsen LF, Edvinsson L, Brain S, Sheykhzade M. Heterogeneous vasomotor responses in segments from Göttingen Minipigs coronary, cerebral, and mesenteric artery: A comparative study. Vascul Pharmacol 2023; 153:107231. [PMID: 37730143 DOI: 10.1016/j.vph.2023.107231] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Revised: 09/13/2023] [Accepted: 09/17/2023] [Indexed: 09/22/2023]
Abstract
Göttingen Minipigs (GM) are used as an important preclinical model for cardiovascular safety pharmacology and for evaluation of cardiovascular drug targets. To improve the translational value of the GM model, the current study represents a basic characterization of vascular responses to endothelial regulators and sympathetic, parasympathetic, and sensory neurotransmitters in different anatomical origins. The aim of the current comparative and descriptive study is to use myography to characterize the vasomotor responses of coronary artery isolated from GM and compare the responses to those obtained from parallel studies using cerebral and mesenteric arteries. The selected agonists for sympathetic (norepinephrine), parasympathetic (carbachol), sensory (calcitonin gene-related peptide, CGRP), and endothelial pathways (endothelin-1, ET-1, and bradykinin) were used for comparison. Further, the robust nature of the vasomotor responses was evaluated after 24 h of cold storage of vascular tissue mimicking the situation under which human biopsies are often kept before experiments or grafting is feasible. Results show that bradykinin and CGRP consistently dilated, and endothelin consistently contracted artery segments from coronary, cerebral, and mesenteric origin. By comparison, norepinephrine and carbachol, had responses that varied with the anatomical source of the tissues. To support the basic characterization of GM vasomotor responses, we demonstrated the presence of mRNA encoding selected vascular receptors (CGRP- and ETA-receptors) in fresh artery segments. In conclusion, the vasomotor responses of isolated coronary, cerebral, and mesenteric arteries to selected agonists of endothelial, sympathetic, parasympathetic, and sensory pathways are different and the phenotypes are similar to sporadic human findings.
Collapse
Affiliation(s)
- Anette Sams
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark; Department of Clinical Experimental Research, Rigshospitalet, Glostrup, Denmark; Epoqe Pharma, Ole Maaloes Vej 3, 2200 Copenhagen N, Denmark.
| | | | - Anja Holm
- Department of Clinical Experimental Research, Rigshospitalet, Glostrup, Denmark; Center for RNA Medicine, Department of Clinical Medicine, Aalborg University, Copenhagen, Denmark
| | - Spyridoula Kazantzi
- Department of Clinical Experimental Research, Rigshospitalet, Glostrup, Denmark
| | | | - Lars Edvinsson
- Department of Clinical Experimental Research, Rigshospitalet, Glostrup, Denmark
| | - Susan Brain
- Section of Vascular Biology & Inflammation, School of Cardiovascular Medicine & Research, BHF Centre of Excellence, King's College London, London, United Kingdom
| | - Majid Sheykhzade
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark
| |
Collapse
|
48
|
Finkel S, Sweet S, Locke T, Smith S, Wang Z, Sandini C, Imredy J, He Y, Durante M, Lagrutta A, Feinberg A, Lee A. FRESH™ 3D bioprinted cardiac tissue, a bioengineered platform for in vitro pharmacology. APL Bioeng 2023; 7:046113. [PMID: 38046544 PMCID: PMC10693443 DOI: 10.1063/5.0163363] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Accepted: 10/30/2023] [Indexed: 12/05/2023] Open
Abstract
There is critical need for a predictive model of human cardiac physiology in drug development to assess compound effects on human tissues. In vitro two-dimensional monolayer cultures of cardiomyocytes provide biochemical and cellular readouts, and in vivo animal models provide information on systemic cardiovascular response. However, there remains a significant gap in these models due to their incomplete recapitulation of adult human cardiovascular physiology. Recent efforts in developing in vitro models from engineered heart tissues have demonstrated potential for bridging this gap using human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) in three-dimensional tissue structure. Here, we advance this paradigm by implementing FRESH™ 3D bioprinting to build human cardiac tissues in a medium throughput, well-plate format with controlled tissue architecture, tailored cellular composition, and native-like physiological function, specifically in its drug response. We combined hiPSC-CMs, endothelial cells, and fibroblasts in a cellular bioink and FRESH™ 3D bioprinted this mixture in the format of a thin tissue strip stabilized on a tissue fixture. We show that cardiac tissues could be fabricated directly in a 24-well plate format were composed of dense and highly aligned hiPSC-CMs at >600 million cells/mL and, within 14 days, demonstrated reproducible calcium transients and a fast conduction velocity of ∼16 cm/s. Interrogation of these cardiac tissues with the β-adrenergic receptor agonist isoproterenol showed responses consistent with positive chronotropy and inotropy. Treatment with calcium channel blocker verapamil demonstrated responses expected of hiPSC-CM derived cardiac tissues. These results confirm that FRESH™ 3D bioprinted cardiac tissues represent an in vitro platform that provides data on human physiological response.
Collapse
Affiliation(s)
| | | | - Tyler Locke
- FluidForm, Inc., Waltham, Massachusetts 02451, USA
| | - Sydney Smith
- FluidForm, Inc., Waltham, Massachusetts 02451, USA
| | - Zhefan Wang
- FluidForm, Inc., Waltham, Massachusetts 02451, USA
| | | | - John Imredy
- In Vitro Safety Pharmacology, Genetic and Cellular Toxicology, Merck & Co. Inc., Rahway, New Jersey 07065, USA
| | - Yufang He
- Division of Technology, Infrastructure, Operations and Experience, Merck & Co. Inc., Rahway, New Jersey 07065, USA
| | - Marc Durante
- Division of Technology, Infrastructure, Operations and Experience, Merck & Co. Inc., Rahway, New Jersey 07065, USA
| | - Armando Lagrutta
- In Vitro Safety Pharmacology, Genetic and Cellular Toxicology, Merck & Co. Inc., Rahway, New Jersey 07065, USA
| | | | - Andrew Lee
- FluidForm, Inc., Waltham, Massachusetts 02451, USA
| |
Collapse
|
49
|
West-Livingston L, Lim JW, Lee SJ. Translational tissue-engineered vascular grafts: From bench to bedside. Biomaterials 2023; 302:122322. [PMID: 37713761 DOI: 10.1016/j.biomaterials.2023.122322] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Revised: 09/01/2023] [Accepted: 09/09/2023] [Indexed: 09/17/2023]
Abstract
Cardiovascular disease is a primary cause of mortality worldwide, and patients often require bypass surgery that utilizes autologous vessels as conduits. However, the limited availability of suitable vessels and the risk of failure and complications have driven the need for alternative solutions. Tissue-engineered vascular grafts (TEVGs) offer a promising solution to these challenges. TEVGs are artificial vascular grafts made of biomaterials and/or vascular cells that can mimic the structure and function of natural blood vessels. The ideal TEVG should possess biocompatibility, biomechanical mechanical properties, and durability for long-term success in vivo. Achieving these characteristics requires a multi-disciplinary approach involving material science, engineering, biology, and clinical translation. Recent advancements in scaffold fabrication have led to the development of TEVGs with improved functional and biomechanical properties. Innovative techniques such as electrospinning, 3D bioprinting, and multi-part microfluidic channel systems have allowed the creation of intricate and customized tubular scaffolds. Nevertheless, multiple obstacles must be overcome to apply these innovations effectively in clinical practice, including the need for standardized preclinical models and cost-effective and scalable manufacturing methods. This review highlights the fundamental approaches required to successfully fabricate functional vascular grafts and the necessary translational methodologies to advance their use in clinical practice.
Collapse
Affiliation(s)
- Lauren West-Livingston
- Wake Forest Institute for Regenerative Medicine, Wake Forest University School of Medicine, Winston-Salem, NC, 27157, USA; Department of Vascular and Endovascular Surgery, Duke University, Durham, NC, 27712, USA
| | - Jae Woong Lim
- Wake Forest Institute for Regenerative Medicine, Wake Forest University School of Medicine, Winston-Salem, NC, 27157, USA; Department of Thoracic and Cardiovascular Surgery, Soonchunhyang University Hospital, Bucheon-Si, Gyeonggi-do, 420-767, Republic of Korea
| | - Sang Jin Lee
- Wake Forest Institute for Regenerative Medicine, Wake Forest University School of Medicine, Winston-Salem, NC, 27157, USA.
| |
Collapse
|
50
|
Guo ZL, Tjen-A-Looi SC, Nguyen AT, Fu LW, Su HF, Gong YD, Malik S. Adenosine A 2A receptors in the rostral ventrolateral medulla participate in blood pressure decrease with electroacupuncture in hypertensive rats. Front Cardiovasc Med 2023; 10:1275952. [PMID: 37928764 PMCID: PMC10620741 DOI: 10.3389/fcvm.2023.1275952] [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: 08/15/2023] [Accepted: 10/06/2023] [Indexed: 11/07/2023] Open
Abstract
Acupuncture is increasingly used to manage high blood pressure (BP) as a complementary therapy. However, the mechanisms underlying its hypotensive effects remain unclear. Our previous studies have shown that electroacupuncture (EA) at the ST36-37 acupoints, overlying the deep peroneal nerve, attenuates pressor responses through adenosine A2A receptors (A2AR) in the rostral ventrolateral medulla (rVLM). However, it is uncertain whether rVLM A2AR contributes to EA's BP-lowering effect in sustained hypertension. We hypothesized that a course of EA treatment lowers BP, in part, through the activation of adenosine A2AR in the rVLM in hypertensive rats. To mimic essential hypertension in the clinic, we performed EA in conscious Dahl salt-sensitive hypertensive rats (DSHRs). EA (0.1-0.4 mA, 2 Hz) was applied at ST36-37 for 30 min twice weekly for four weeks, while sham-EA was conducted in a similar manner but without electrical input. In hypertensive rats, BP was reduced by EA (n = 14) but neither by sham-EA (n = 14) nor in the absence of needling (n = 8). Following four weeks of eight treatments and then under anesthesia, EA's modulatory effect on elevated BP was reversed by unilateral rVLM microinjection of SCH 58261 (1 mM in 50 nl; an A2AR antagonist; n = 7; P < 0.05) but not the vehicle (n = 5) in EA-treated DSHRs. Activation of rVLM A2AR in DSHRs treated with sham-EA by an A2AR agonist, CGS-21680 (0.4 mM in 50 nl; n = 8), decreased BP. Unilateral administration of SCH 58261 or CGS-21680 into the rVLM did not alter basal BP in Dahl salt-sensitive rats fed a regular diet with normal BP. The A2AR level in the rVLM after EA was increased compared to the sham-EA and untreated DSHRs (n = 5 in each group; all P < 0.05). These data suggest that a 4-week twice weekly EA treatment reduced BP in salt-sensitive hypertensive rats likely through adenosine-mediated A2AR in the rVLM.
Collapse
Affiliation(s)
- Zhi-Ling Guo
- Susan-Samueli Integrative Health Institute and Department of Medicine, College of Health Sciences, University of California, Irvine, CA, United States
| | | | | | | | | | | | | |
Collapse
|