1
|
Ou YC, Peng XY, Yang JX, Chen BY, Chen PF, Liu M. Efficacy of catheter-based ultrasound renal denervation in the treatment of hypertension. World J Clin Cases 2025; 13:102853. [DOI: 10.12998/wjcc.v13.i16.102853] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Revised: 12/25/2024] [Accepted: 01/18/2025] [Indexed: 02/10/2025] Open
Abstract
BACKGROUND Hypertension (HTN) is a prevalent chronic health condition that significantly increases the risk of cardiovascular diseases-associated mortalities. Despite the use of antihypertensive medications, numerous patients fail to achieve guideline-recommended blood pressure (BP) targets.
AIM To evaluates the efficacy of catheter-based ultrasound renal denervation (uRDN) for the treatment of HTN.
METHODS Relevant studies were identified through searches in PubMed, Embase, the Cochrane Library, Web of Science, and China National Knowledge Infrastructure, with a cut-off date at April 1, 2024. A random-effects model was employed in this study to mitigate potential biases. The risk of bias for included studies was assessed using the Cochrane Risk of Bias assessment tool. Statistical analyses were conducted using Review Manager version 5.3. This meta-analysis incorporated four studies encompassing a total of 627 patients. The reporting bias of this study was deemed acceptable.
RESULTS Compared to the Sham group, the uRDN group demonstrated a significant reduction in daytime ambulatory systolic BP (SBP) [mean difference (MD) -3.87 mmHg, 95% confidence interval (CI): -7.02 to -0.73, P = 0.02], office SBP (MD -4.13 mmHg, 95%CI: -7.15 to -1.12, P = 0.007), and home SBP (MD -5.51 mmHg, 95%CI: -8.47 to -2.55, P < 0.001). However, there was no statistically significant reduction observed in either 24-hour or nighttime ambulatory SBP levels. Subgroup analysis shows that uRDN can significantly reduce the SBP in patients with non-resistant HTN (MD -6.19 mmHg, MD -6.00 mmHg, MD -7.72 mmHg, MD -5.02 mmHg, MD -3.61 mmHg).
CONCLUSION The current evidence suggests that uRDN may effectively reduce home, office, and daytime SBP in patients with HTN, particularly in those with non-resistant HTN.
Collapse
Affiliation(s)
- Yi-Chao Ou
- Department of Cardiology, Cardiovascular Research Center, Affiliated Hospital of North Sichuan Medical College, Nanchong 637000, Sichuan Province, China
- Department of Clinical Medicine, North Sichuan Medical College, Nanchong 637000, Sichuan Province, China
| | - Xin-Yuan Peng
- Department of Cardiology, Cardiovascular Research Center, Affiliated Hospital of North Sichuan Medical College, Nanchong 637000, Sichuan Province, China
- Department of Clinical Medicine, North Sichuan Medical College, Nanchong 637000, Sichuan Province, China
| | - Jing-Xi Yang
- Department of Cardiology, Cardiovascular Research Center, Affiliated Hospital of North Sichuan Medical College, Nanchong 637000, Sichuan Province, China
| | - Bo-Yu Chen
- Department of Cardiology, Cardiovascular Research Center, Affiliated Hospital of North Sichuan Medical College, Nanchong 637000, Sichuan Province, China
- Department of Clinical Medicine, North Sichuan Medical College, Nanchong 637000, Sichuan Province, China
| | - Peng-Fei Chen
- Department of Cardiology, Cardiovascular Research Center, Affiliated Hospital of North Sichuan Medical College, Nanchong 637000, Sichuan Province, China
| | - Mao Liu
- Department of Cardiology, Cardiovascular Research Center, Affiliated Hospital of North Sichuan Medical College, Nanchong 637000, Sichuan Province, China
| |
Collapse
|
2
|
Callender K, Ogieuhi IJ, Ajekiigbe VO, Adeyemi BA, Agbo CE, Falayi TA, Oladejo A, Ajayi JO, Ajewole S, Adetayo FT, Fakojo OS, Akingbola A, Adeshina GA. Renal Denervation as a Novel Therapeutic Approach for Resistant Hypertension: Mechanisms, Efficacy and Future Directions. High Blood Press Cardiovasc Prev 2025; 32:227-254. [PMID: 39580579 DOI: 10.1007/s40292-024-00696-4] [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: 09/16/2024] [Accepted: 11/02/2024] [Indexed: 11/25/2024] Open
Abstract
Resistant hypertension is a state characterized by sustained hypertension despite adherence to the standard pharmacological treatment with beta-blockers, calcium channel blockers, diuretics, and ACE inhibitors or ARBs. Resistant hypertension is a problem now in cardiovascular medicine because of its association with increased stroke, heart failure, kidney disease, and vision loss. Renal denervation (RDN) is an invasive treatment strategy for patients with hypertension who are unresponsive to pharmacological therapy. Therefore, this procedure has become a feasible alternative, and this review explores and compares with other possible novel options. RDN's mechanisms, efficacy, safety, and future directions are also discussed. No serious side effects have been reported in the short-term use of RDN, but some of the complications include renal stenosis and hypertensive urgencies in the long term. Despite this, RDN can benefit patients who are non-compliant with medications or are intolerant. However, it should also be pointed out that some clinical studies have not given consistent results. RDN may be employed as secondary therapy as opposed to the primary line of treatment in resistant hypertension. Subsequent studies should assess the technique's durability and establish customized approaches to deliver RDN safely while determining specific biomarkers that can predict patients' outcomes.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Samuel Ajewole
- Babcock University Teaching Hospital (BUTH), Ilishan-Remo, Ogun State, Nigeria
| | | | | | - Adewunmi Akingbola
- Department of Public Health, University of Cambridge, Cambridgeshire Old Trinity Schools, CB2 1TN, Cambridge, England, UK
| | - Ganiyat Adekemi Adeshina
- Department of Pharmacology, Therapeutics and Toxicology, Faculty of Basic Medical Sciences, University of Lagos, Lagos, Nigeria
| |
Collapse
|
3
|
Rahman AA, Hildreth CM, Milliken P, Hassan S, Sridhar A, Phillips JK. Impaired inhibitory reno-renal reflex responses in chronic kidney disease. Front Physiol 2025; 16:1544592. [PMID: 40271214 PMCID: PMC12014541 DOI: 10.3389/fphys.2025.1544592] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Accepted: 03/27/2025] [Indexed: 04/25/2025] Open
Abstract
The renal afferent nerves serve as physiologic regulators of efferent renal sympathetic nerve activity (rSNA) as part of the inhibitory reno-renal reflex. Dysregulation of this reflex response may promote sympathoexcitation and subsequent hypertension under pathologic conditions such as chronic kidney disease (CKD). We have undertaken an in-depth characterization of reno-renal reflex function in CKD using an anesthetized rodent model with concurrent physiological outflows assessed. Using anesthetized male Lewis Polycystic Kidney (LPK) rats and normotensive Lewis controls, we investigated the cardiovascular [blood pressure (BP), heart rate (HR) and sympathetic responses (recorded from renal and splanchnic nerves (r/sSNA)] to renal capsaicin (50 µM) and direct electrical stimulation of the whole renal nerve. In Lewis rats, intra-pelvic renal capsaicin injection resulted in a depressor, bradycardic, and sympathoinhibitory response in sSNA with no significant change in rSNA. In contrast, the same stimulus led to a pressor and sympathoexcitatory response in the LPK group. In Lewis rats, low-intensity electrical stimulation (0.2 ms pulses, 15 μA, 2-40 Hz) of the renal nerve elicited a depressor response and bradycardia with concurrent sympathoexcitation (sSNA), whereas high-intensity (150 µA) stimulation induced a biphasic depressor/pressor response and tachycardia. In LPK rats, low-intensity renal nerve electrical stimulation triggered a biphasic depressor/pressor BP response, tachycardia, and sympathoexcitation. High-intensity stimulation similarly caused a biphasic depressor/pressor BP response and tachycardia. The magnitude of the sSNA response and both phases of the blood pressure response was higher in LPK compared to Lewis. All responses showed some degree of frequency dependency. Our results suggest the inhibitory reno-renal reflex is impaired in CKD, with dominance of excitatory reflex response. However, a depressor component remained that could be targeted using implantable neurotechnologies to lower blood pressure in CKD patients safely and effectively.
Collapse
Affiliation(s)
- Ahmed A. Rahman
- Sensory and Autonomic Neuroscience Laboratory, Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, NSW, Australia
- Department of Pediatric Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
| | - Cara M. Hildreth
- Sensory and Autonomic Neuroscience Laboratory, Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, NSW, Australia
| | | | - Sarah Hassan
- Galvani Bioelectronics, Stevenage, United Kingdom
| | - Arun Sridhar
- Galvani Bioelectronics, Stevenage, United Kingdom
| | - Jacqueline K. Phillips
- Sensory and Autonomic Neuroscience Laboratory, Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, NSW, Australia
| |
Collapse
|
4
|
Hu W, Tu H, Wadman MC, Li YL, Zhang D. Renal denervation achieves its antiarrhythmic effect through attenuating macrophage activation and neuroinflammation in stellate ganglia in chronic heart failure. Cardiovasc Res 2025; 120:2420-2433. [PMID: 39321201 PMCID: PMC11976725 DOI: 10.1093/cvr/cvae196] [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: 02/09/2023] [Revised: 05/22/2024] [Accepted: 06/19/2024] [Indexed: 09/27/2024] Open
Abstract
AIMS Renal denervation (RDN) is widely investigated in multiple studies of sympathetically driven cardiovascular diseases. While the therapeutic potential of RDN for ventricular arrhythmia has been reported, the mechanisms responsible for its antiarrhythmic effect are poorly understood. Our recent study showed that macrophage expansion-induced neuroinflammation in the stellate ganglion (SG) was a critical factor for cardiac sympathetic overactivation and ventricular arrhythmogenesis in chronic heart failure (CHF). This study investigates if and how RDN decreases ventricular arrhythmias by attenuating neuroinflammation in cardiac sympathetic post-ganglionic (CSP) neurons in CHF. METHODS AND RESULTS Rat CHF was induced by surgical ligation of the left anterior descending (LAD) coronary artery. At 12 weeks after LAD ligation, completed bilateral RDN was achieved by surgically cutting all the visible renal nerves around the renal artery and vein, followed by applying 70% ethanol around the vessels. Immunofluorescence staining and western blot data showed that expression of granulocyte-macrophage colony-stimulating factor (GM-CSF) and its receptor-α subunit in SGs was increased in CHF rats. RDN not only reduced CHF-elevated GM-CSF levels in kidney, serum, and SGs but also attenuated macrophage expansion and neuroinflammation in SGs from CHF rats. Using flow cytometry, we confirmed that RDN reduced the percentage of macrophages in SGs, which is pathologically increased in CHF. RDN also decreased CHF-enhanced N-type Ca2+ currents in CSP neurons and attenuated CHF-elevated cardiac sympathetic nerve activity. Electrocardiogram data from 24-h continuous telemetry recording in conscious rats revealed that RDN improved CHF-induced heterogeneity of ventricular electrical activities and reduced the duration of spontaneous ventricular tachyarrhythmias in CHF rats. CONCLUSION RDN alleviates cardiac sympathetic overactivation and ventricular arrhythmogenesis through attenuating GM-CSF-induced macrophage activation and neuroinflammation within SGs in CHF. This suggests that manipulation of the GM-CSF signalling pathway could be a novel strategy for achieving the antiarrhythmic effect of RDN in CHF.
Collapse
Affiliation(s)
- Wenfeng Hu
- Department of Emergency Medicine, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Huiyin Tu
- Department of Emergency Medicine, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Michael C Wadman
- Department of Emergency Medicine, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Yu-Long Li
- Department of Emergency Medicine, University of Nebraska Medical Center, Omaha, NE 68198, USA
- Department of Cellular & Integrative Physiology, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Dongze Zhang
- Department of Emergency Medicine, University of Nebraska Medical Center, Omaha, NE 68198, USA
| |
Collapse
|
5
|
Si F, Liu Q, Ma X, Yu J. Effects of Renal Denervation on Cardiac Remodeling, Cardiac Function, and Cardiovascular Neurohormones in Heart Failure with Reduced Ejection Fraction Patients: A Meta-Analysis and Systematic Review. Cardiorenal Med 2025; 15:261-280. [PMID: 40132560 DOI: 10.1159/000545078] [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: 10/23/2024] [Accepted: 02/23/2025] [Indexed: 03/27/2025] Open
Abstract
INTRODUCTION The objective of this study was to evaluate the effects of renal denervation (RDN) on cardiac remodeling, cardiac function, and cardiovascular (CV) neurohormones in heart failure patients with reduced ejection fraction (HFrEF). METHODS We searched PubMed, Embase, Web of Science, and China National Knowledge Infrastructure (CNKI), identifying 6 randomized controlled trials (RCTs) and 9 single-arm studies, totaling 352 participants. Meta-analyses for RCTs and single-arm studies were conducted using STATA 17 software and the metafor package in R, respectively. RESULTS In RCTs, RDN significantly reduced left ventricular end-diastolic diameter (LVEDD) (weighted mean difference [WMD] = -3.55 mm, 95% CI [-5.51, -1.59], p < 0.01), left ventricular end-systolic diameter (LVESD) (WMD = -4.13 mm, 95% CI [-6.08, -2.18], p < 0.01), and significantly increased left ventricular ejection fraction (LVEF) (WMD = 6.30%, 95% CI [4.64, 7.96], p < 0.01) and 6-min walk test (6MWT) distance (WMD = 51.25 m, 95% CI [8.30, 94.20], p < 0.05). Brain natriuretic peptide (BNP) or N-terminal pro-B-type natriuretic peptide (NT-proBNP) levels were significantly reduced (standardized mean difference = -1.24, 95% CI [-1.57, -0.90], p < 0.01). In single-arm studies, RDN significantly reduced LVEDD (MC = -2.41 mm, 95% CI [-3.74, -1.09], p < 0.01), LVESD (MC = -1.72 mm, 95% CI [-2.77, -0.67], p < 0.01), left atrial diameter (MC = -1.62 mm, 95% CI [-3.16, -0.08], p < 0.01), and interventricular septal thickness (IVST) (MC = -0.76 mm, 95% CI [-1.05, -0.47], p < 0.01). RDN significantly increased LVEF (MC = 29.52%, 95% CI [12.74, 46.31], p < 0.01) and 6MWT distance (MC = 100.49 m, 95% CI [49.12, 151.86], p < 0.05). RDN significantly reduced BNP or NT-proBNP levels (SMC = -0.57, 95% CI [-0.83, -0.31], p < 0.01). Our study also found that RDN had varying degrees of reduction on renin, angiotensin II, aldosterone, and norepinephrine in HFrEF patients. Additionally, we found that RDN had no significant effect on SBP/DBP in HFrEF patients but reduced heart rate (WMD = -7.22 bpm, 95% CI [-9.84, -4.60], p < 0.01). CONCLUSION Our meta-analysis demonstrates that RDN can improve cardiac remodeling, enhance cardiac function, reduce CV neurohormones and has no significant effect on blood pressure in patients with HFrEF.
Collapse
Affiliation(s)
- Fei Si
- Department of Cardiology, The Second Hospital and Clinical Medical School, Lanzhou University, Lanzhou, China
| | - Qian Liu
- Department of Cardiology, The Second Hospital and Clinical Medical School, Lanzhou University, Lanzhou, China
| | - Xin Ma
- Department of Cardiology, The Second Hospital and Clinical Medical School, Lanzhou University, Lanzhou, China
| | - Jing Yu
- Department of Cardiology, The Second Hospital and Clinical Medical School, Lanzhou University, Lanzhou, China
| |
Collapse
|
6
|
Salman IM, Ameer OZ, Hassan SF, Sridhar A, Hsieh YH, Lewis SJ. Low-level neurostimulation of the renal nerves as a potential therapeutic strategy for hypertension treatment. Front Pharmacol 2025; 16:1529844. [PMID: 40078273 PMCID: PMC11897555 DOI: 10.3389/fphar.2025.1529844] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2024] [Accepted: 01/30/2025] [Indexed: 03/14/2025] Open
Abstract
Background Neurostimulation is an emerging treatment for conditions like hypertension. The renal nerves, comprising sensory afferent and sympathetic efferent fibers, are crucial for blood pressure (BP) regulation. The inhibitory reno-renal reflex, where central integration of renal sensory input reduces sympathetic outflow and systemic BP, presents a promising target for neurostimulation interventions. We therefore investigated renal nerve stimulation (RNS) as a potential hypertension therapy. Methods Anesthetized male spontaneously hypertensive rats (SHRs) were subjected to low-level RNS at 0.5 mA pulse amplitude and 0.5 ms pulse width for 30 s delivered to the left intact renal nerve at 2.5 and 5.0 Hz. Mean arterial pressure (MAP), heart rate (HR), hindquarter blood flow (HQF), and ipsilateral renal cortical blood flow (RCF) were recorded. Hindquarter resistance (HQR) and renal cortical resistance (RCR) were derived from MAP and flow values. Results RNS significantly reduced MAP, with similar depressor responses at 2.5 (27 ± 3 mmHg) and 5.0 Hz (37 ± 8 mmHg). RNS substantially increased HQF and reduced HQR, with comparable effects at both frequencies. A 5-Hz stimulus markedly reduced RCF and increased RCR of the ipsilateral kidney. When the stimulation frequency was lowered to 2.5 Hz, the changes in RCF and RCR were nearly indistinguishable from baseline. Conclusion Low-level RNS effectively lowers BP in the SHR model of hypertension and may offer a promising therapeutic alternative for hypertension treatment. Physiologically, the observed clinically relevant reductions in BP were primarily due to reductions in vascular resistance. Adjusting stimulus levels can achieve desired hypotensive responses without compromising ipsilateral renal blood supply, typically affected by direct renal sympathetic fiber stimulation.
Collapse
Affiliation(s)
- Ibrahim M. Salman
- Department of Pharmaceutical Sciences, College of Pharmacy, Alfaisal University, Riyadh, Saudi Arabia
| | - Omar Z. Ameer
- Department of Pharmaceutical Sciences, College of Pharmacy, Alfaisal University, Riyadh, Saudi Arabia
| | - Sarah F. Hassan
- Department of Disease Biology, Galvani Bioelectronics, Stevenage, Hertfordshire, United Kingdom
| | - Arun Sridhar
- Department of Disease Biology, Galvani Bioelectronics, Stevenage, Hertfordshire, United Kingdom
| | - Yee-Hsee Hsieh
- Division of Pulmonary, Critical Care, and Sleep Medicine, School of Medicine, Case Western Reserve University, Cleveland, OH, United States
| | - Stephen J. Lewis
- Division of Pulmonology, Allergy and Immunology, Department of Pediatrics, School of Medicine, Case Western Reserve University, Cleveland, OH, United States
- Electrical Stimulation Center, Case Western Reserve University, Cleveland, OH, United States
| |
Collapse
|
7
|
Song Z, Yan M, Zhang S, Hu B, Qing X, Shao Z, Chen S, Lv X, Liu H. Implications of circadian disruption on intervertebral disc degeneration: The mediating role of sympathetic nervous system. Ageing Res Rev 2025; 104:102633. [PMID: 39701186 DOI: 10.1016/j.arr.2024.102633] [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: 09/28/2024] [Revised: 12/05/2024] [Accepted: 12/05/2024] [Indexed: 12/21/2024]
Abstract
The circadian clock orchestrates a broad spectrum of physiological processes, crucially modulating human biology across an approximate 24-hour cycle. The circadian disturbances precipitated by modern lifestyle contribute to the occurrence of low back pain (LBP), mainly ascribed to intervertebral disc degeneration (IVDD). The intervertebral disc (IVD) exhibits rhythmic physiological behaviors, with fluctuations in osmotic pressure and hydration levels that synchronized with the diurnal cycle of activity and rest. Over recent decades, advanced molecular biology techniques have shed light on the association between circadian molecules and IVD homeostasis. The complex interplay between circadian rhythm disruption and IVDD is becoming increasingly evident, with the sympathetic nervous system (SNS) emerging as a potential mediator. Synchronized with circadian rhythm through suprachiasmatic nucleus, the SNS regulates diverse physiological functions and metabolic processes, profoundly influences the structural and functional integrity of the IVD. This review synthesizes the current understanding of circadian regulation and sympathetic innervation of the IVD, highlighting advancements in the comprehension of their interactions. We elucidate the impact of circadian system on the physiological functions of IVD through the SNS, advocating for the adoption of chronotherapy as a brand-new and effective strategy to ameliorate IVDD and alleviate LBP.
Collapse
Affiliation(s)
- Zongmian Song
- Department of Orthopedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Miaoheng Yan
- Department of Orthopedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Shuo Zhang
- School of Medicine, Nankai University, Tianjin 300071, China
| | - Binwu Hu
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Xiangcheng Qing
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Zengwu Shao
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Songfeng Chen
- Department of Orthopedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China.
| | - Xiao Lv
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
| | - Hongjian Liu
- Department of Orthopedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China.
| |
Collapse
|
8
|
Johnson NA, Dickenson JA, MacKenzie BW, Isakovich R, Kalker A, Bouten J, Strzalkowski ND, Harman TS, Holmström P, Kunwar AJ, Thakur N, Dhungel S, Sherpa N, Bigham AW, Brutsaert TD, Day TA. Comparing integrative ventilatory and renal acid-base acclimatization in lowlanders and Tibetan highlanders during ascent to 4,300 m. Proc Natl Acad Sci U S A 2025; 122:e2412561121. [PMID: 39793031 PMCID: PMC11725942 DOI: 10.1073/pnas.2412561121] [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: 08/01/2024] [Accepted: 11/02/2024] [Indexed: 01/12/2025] Open
Abstract
With over 14 million people living above 3,500 m, the study of acclimatization and adaptation to high altitude in human populations is of increasing importance, where exposure to high altitude (HA) imposes a blood oxygenation and acid-base challenge. A sustained and augmented hypoxic ventilatory response protects oxygenation through ventilatory acclimatization, but elicits hypocapnia and respiratory alkalosis. A subsequent renally mediated compensatory metabolic acidosis corrects pH toward baseline values, with a high degree of interindividual variability. Differential renal compensation between acclimatizing lowlanders (LL) and Tibetan highlanders (TH; Sherpa) with ascent was previously unknown. We assessed ventilatory and renal acclimatization between unacclimatized LL and TH during incremental ascent from 1,400 m to 4,300 m in age- and sex-matched groups of 15-LL (8F) and 14-TH (7F) of confirmed Tibetan ancestry. We compared respiratory and renally mediated blood acid-base acclimatization (PCO2, [HCO3-], pH) in both groups before (1,400 m) and following day 8 to 9 of incremental ascent to 4,300 m. We found that following ascent to 4,300 m, LL had significantly lower PCO2 (P <0.0001) and [HCO3-] (P <0.0001), and higher pH (P = 0.0037) than 1,400 m, suggesting respiratory alkalosis and only partial renal compensation. Conversely, TH had significantly lower PCO2 (P < 0.0001) and [HCO3-] (P < 0.0001), but unchanged pH (P = 0.1), suggesting full renal compensation, with significantly lower PCO2 (P = 0.01), [HCO3-] (P < 0.0001) and pH (P = 0.005) than LL at 4,300 m. This demonstration of differential integrative respiratory-renal responses between acclimatizing LL and TH may indicate selective pressure on TH, and highlights the important role of the kidneys in acclimatization.
Collapse
Affiliation(s)
- Nicole A. Johnson
- Department of Biology, Faculty of Science and Technology, Mount Royal University, Calgary, ABT3E 6K6, Canada
| | - Jessica A. Dickenson
- Department of Biology, Faculty of Science and Technology, Mount Royal University, Calgary, ABT3E 6K6, Canada
| | - Benjamin W.L. MacKenzie
- Department of Biology, Faculty of Science and Technology, Mount Royal University, Calgary, ABT3E 6K6, Canada
| | - Rodion Isakovich
- Department of Biology, Faculty of Science and Technology, Mount Royal University, Calgary, ABT3E 6K6, Canada
| | - Anne Kalker
- Radboud University Medical Center, Nijmegen6525 XZ, Netherlands
| | - Janne Bouten
- Department of Movement and Sports Sciences, Ghent University, Ghent29000, Belgium
- Laboratory of Sport, Expertise and Performance, French Institute of Sport (INSEP), Paris75012, France
| | - Nicholas D.J. Strzalkowski
- Department of Biology, Faculty of Science and Technology, Mount Royal University, Calgary, ABT3E 6K6, Canada
| | - Taylor S. Harman
- Department of Exercise Science, Syracuse University, Syracuse, NY13210
| | - Pontus Holmström
- Department of Health Sciences, Mid Sweden University, Östersund831 25, Sweden
| | - Ajaya J. Kunwar
- Kathmandu Center for Genomics and Research Laboratory, Global Hospital, Lalitpur44700, Nepal
| | - Nilam Thakur
- Kathmandu Center for Genomics and Research Laboratory, Global Hospital, Lalitpur44700, Nepal
| | - Sunil Dhungel
- College of Medicine, Nepalese Army Institute of Health Sciences, Kathmandu44600, Nepal
- Medical University of the Americas, Charlestown, Nevis, Saint Kitts and Nevis, West Indies, KN0802
| | - Nima Sherpa
- Glory of Nepal Travels and Tours, Kathmandu44600, Nepal
| | - Abigail W. Bigham
- Department of Anthropology, University of Los Angeles, Los Angeles, CA90095
| | - Tom D. Brutsaert
- Department of Exercise Science, Syracuse University, Syracuse, NY13210
| | - Trevor A. Day
- Department of Biology, Faculty of Science and Technology, Mount Royal University, Calgary, ABT3E 6K6, Canada
| |
Collapse
|
9
|
Evans LC, Dayton A, Osborn JW. Renal nerves in physiology, pathophysiology and interoception. Nat Rev Nephrol 2025; 21:57-69. [PMID: 39363020 DOI: 10.1038/s41581-024-00893-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/27/2024] [Indexed: 10/05/2024]
Abstract
Sympathetic efferent renal nerves have key roles in the regulation of kidney function and blood pressure. Increased renal sympathetic nerve activity is thought to contribute to hypertension by promoting renal sodium retention, renin release and renal vasoconstriction. This hypothesis led to the development of catheter-based renal denervation (RDN) for the treatment of hypertension. Two RDN devices that ablate both efferent and afferent renal nerves received FDA approval for this indication in 2023. However, in animal models, selective ablation of afferent renal nerves resulted in comparable anti-hypertensive effects to ablation of efferent and afferent renal nerves and was associated with a reduction in sympathetic nerve activity. Selective afferent RDN also improved kidney function in a chronic kidney disease model. Notably, the beneficial effects of RDN extend beyond hypertension and chronic kidney disease to other clinical conditions that are associated with elevated sympathetic nerve activity, including heart failure and arrhythmia. These findings suggest that the kidney is an interoceptive organ, as increased renal sensory nerve activity modulates sympathetic activity to other organs. Future studies are needed to translate this knowledge into novel therapies for the treatment of hypertension and other cardiorenal diseases.
Collapse
Affiliation(s)
- Louise C Evans
- Department of Surgery, University of Minnesota, Minneapolis, MN, USA
| | - Alex Dayton
- Division of Nephrology and Hypertension, University of Minnesota, Minneapolis, MN, USA
| | - John W Osborn
- Department of Surgery, University of Minnesota, Minneapolis, MN, USA.
| |
Collapse
|
10
|
Lee JD, Kuo YW, Lee CP, Huang YC, Lee M, Lee TH. Impact of heart rate on eGFR decline in ischemic stroke patients. Clin Kidney J 2025; 18:sfae387. [PMID: 39834622 PMCID: PMC11744309 DOI: 10.1093/ckj/sfae387] [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: 07/09/2024] [Indexed: 01/22/2025] Open
Abstract
Background Resting heart rate is a potent predictor of various renal outcomes. However, the decline rate of renal function in ischemic stroke patients is not well defined. We tested the association of heart rate with estimated eGFR decline and the composite renal outcomes in patients with recent ischemic stroke. Methods The data of 9366 patients with ischemic stroke with an eGFR of ≥30 mL/min/1.73 m2 were retrieved from the Chang Gung Research Database. Mean initial in-hospital heart rates were averaged and categorized into 10-beats-per-minute (bpm) increments. The outcomes were the annualized change in eGFR across the heart rate subgroups and composite renal outcomes, namely a ≥40% sustained decline in eGFR, end-stage renal disease, or renal death. Generalized estimating equation models were used to determine the annualized change in eGFR and Cox proportional hazards regression models were used to estimate the relative hazard of composite renal outcomes by referencing the subgroup with a heart rate of <60 bpm. Results The annual eGFR decline in the patients with a mean heart rate of <60, 60-69, 70-79, 80-89, and ≥90 bpm was 2.12, 2.49, 2.83, 3.35, and 3.90 mL/min/1.73 m2, respectively. Compared with the reference group, the adjusted hazard ratios for composite renal outcomes were 1.17 [95% confidence interval (CI), 0.89-1.53), 1.54 (95% CI, 1.19-2.00), 1.72 (95% CI, 1.30-2.28), and 1.84 (95% CI, 1.29-2.54] for the patients with a heart rate of 60-69, 70-79, 80-89, and ≥90 bpm, respectively. In the subgroup analysis, the associations between higher heart rate and both eGFR decline and composite renal outcomes were more evident and statistically significant in patients without atrial fibrillation. Conclusions A higher heart rate is associated with a faster rate of eGFR decline and an increased risk of composite renal outcomes after ischemic stroke, particularly in patients without atrial fibrillation. These results underscore the importance of heart rate monitoring and management in ischemic stroke patients in sinus rhythm to potentially mitigate renal function decline. Further studies are needed to explore this relationship in patients with atrial fibrillation and across different ethnic groups.
Collapse
Affiliation(s)
- Jiann-Der Lee
- Department of Neurology, Chiayi Chang Gung Memorial Hospital, Chiayi, Taiwan
- College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Ya-Wen Kuo
- Department of Neurology, Chiayi Chang Gung Memorial Hospital, Chiayi, Taiwan
- Department of Nursing, Chang Gung University of Science and Technology, Chiayi Campus, Chiayi, Taiwan
| | - Chuan-Pin Lee
- Health Information and Epidemiology Laboratory, Chiayi Chang Gung Memorial Hospital, Chiayi, Taiwan
| | - Yen-Chu Huang
- Department of Neurology, Chiayi Chang Gung Memorial Hospital, Chiayi, Taiwan
- College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Meng Lee
- Department of Neurology, Chiayi Chang Gung Memorial Hospital, Chiayi, Taiwan
- College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Tsong-Hai Lee
- College of Medicine, Chang Gung University, Taoyuan, Taiwan
- Department of Neurology, Linkou Chang Gung Memorial Hospital, Taoyuan, Taiwan
| |
Collapse
|
11
|
Lin WY, Cheng YH, Liu PY, Hsu SP, Lin SC, Chien CT. Carvedilol through ß1-Adrenoceptor blockade ameliorates glomerulonephritis via inhibition of oxidative stress, apoptosis, autophagy, ferroptosis, endoplasmic reticulum stress and inflammation. Biochem Pharmacol 2024; 230:116570. [PMID: 39401703 DOI: 10.1016/j.bcp.2024.116570] [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: 05/20/2024] [Revised: 08/28/2024] [Accepted: 10/08/2024] [Indexed: 10/28/2024]
Abstract
Glomerulonephritis (GN) is one of the main causes of end stage renal disease and requires an effective treatment for inhibiting GN. Renal nerves through efferent (RENA) and afferent (RANA) innervation to glomeruli regulate the glomerular function. We delineated the role of RENA and RANA on anti-Thy1.1-induced GN. Female Wistar rats were divided into Control, Thy1.1 plus anti-Thy1.1, bilaterally renal nerve denervation (DNX) plus anti-Thy1.1, and topical capsaicin to bilateral renal nerves for selective ablation of RANA (DNAX) plus anti-Thy1.1. We examined RANA and RENA response to anti-Thy1.1 and compared the effect of DNX or DNAX on urinary oxidative stress, renal gp91, tyrosine hydroxylase (TH), calcitonin gene-related peptide (CGRP), apoptosis, autophagy, ferroptosis, antioxidant enzymes, endoplasmic reticulum (ER) stress and inflammation by western blot. Anti-Thy1.1 significantly enhanced RENA, but did not affect RANA. DNX significantly decreased TH and CGRP expression, whereas DNAX only reduced CGRP expression. Anti-Thy1.1 significantly increased glomerulosclerosis injury, urinary protein, electron paramagnetic resonance signals of alpha-(4-pyridyl-N-oxide)-N-tert-butylnitrone adducts, 8-isoprostane and nitrotyrosine levels, NADPH oxidase gp91phox (gp91), macrophage/monocyte (ED-1), GRP-78, Beclin-1/LC3-II, Bax/caspase-3/poly(ADP-ribose) polymerase expression, inflammatory cytokines levels and decreased renal Copper/Zinc superoxide dismutase, Cystine/glutamate transporter (xCT) and Glutathione peroxidase 4 (GPX4) expression vs. Control. The enhanced oxidative parameters or reduced antioxidant defense by anti-Thy1.1 were significantly attenuated by DNX but not DNAX. Additionally, oral ß1-adrenoceptor antagonist-Carvedilol at an early stage reduced anti-Thy1.1 increased proteinuria level and oxidative parameters. Our data suggest that DNX and ß1-adrenoceptor antagonist-Carvedilol efficiently attenuate oxidative stress, inflammation, ER stress, autophagy, ferroptosis and apoptosis in GN.
Collapse
Affiliation(s)
- Wei-Yu Lin
- Department of Life Science, School of Life Science, College of Science, National Taiwan Normal University, Taipei 11677, Taiwan; Department of Urology, Taipei Hospital, Ministry of Health and Welfare, New Taipei City 24213, Taiwan
| | - Yu-Hsuan Cheng
- Department of Life Science, School of Life Science, College of Science, National Taiwan Normal University, Taipei 11677, Taiwan
| | - Pei-Yu Liu
- Department of Life Science, School of Life Science, College of Science, National Taiwan Normal University, Taipei 11677, Taiwan
| | - Shih-Ping Hsu
- Department of Life Science, School of Life Science, College of Science, National Taiwan Normal University, Taipei 11677, Taiwan; Department of Internal Medicine, Far Eastern Memorial Hospital, New Taipei City 220, Taiwan; Department of Industrial Management, Oriental Institute of Technology, New Taipei City 220, Taiwan; General Education Center, Lunghwa University of Science and Technology, Taoyuan, Taiwan.
| | - San-Chi Lin
- Division of Renal Section, Department of Internal Medicine, Keelung Hospital, Ministry of Health and Welfare, Keelung City 201, Taiwan.
| | - Chiang-Ting Chien
- Department of Life Science, School of Life Science, College of Science, National Taiwan Normal University, Taipei 11677, Taiwan.
| |
Collapse
|
12
|
Goto H, Yamamoto Y, Tsujiguchi H, Sato T, Yamamoto R, Takeshita Y, Nakano Y, Kannon T, Hosomichi K, Suzuki K, Nakamura M, Kambayashi Y, Zhao J, Asai A, Katano K, Ogawa A, Fukushima S, Shibata A, Suzuki F, Tsuboi H, Hara A, Kometani M, Karashima S, Yoneda T, Tajima A, Nakamura H, Takamura T. Oxytocin Receptor Polymorphism Is Associated With Sleep Apnea Symptoms. J Endocr Soc 2024; 9:bvae198. [PMID: 39606181 PMCID: PMC11590662 DOI: 10.1210/jendso/bvae198] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Indexed: 11/29/2024] Open
Abstract
Context Oxytocin supplementation improves obstructive sleep apnea (OSA), and animal studies suggest involvement of oxytocin in respiratory control. However, the relationship between endogenous oxytocin signaling and human sleep status remains undetermined. Objective In this study, we approached the contribution of the intrinsic oxytocin-oxytocin receptor (OXTR) system to OSA by genetic association analysis. Methods We analyzed the relationship between OXTR gene polymorphisms and sleep parameters using questionnaire data and sleep measurements in 305 Japanese participants. OSA symptoms were assessed in 225 of these individuals. Results The OXTR rs2254298 A allele was more frequent in those with OSA symptoms than in those without (P = .0087). Although total scores on the Pittsburgh Sleep Quality Index questionnaire did not differ between the genotypes, breathlessness and snoring symptoms associated with OSA were significantly more frequent in individuals with rs2254298 A genotype (P = .00045 and P = .0089 for recessive models, respectively) than the G genotype. A multivariable analysis confirmed these genotype-phenotype associations even after adjusting for age, sex, and body mass index in a sensitivity analysis. Furthermore, objective sleep efficiency measured by actigraph was not significantly different between genotypes; however, subjective sleep efficiency was significantly lower in the rs2254298 A genotype (P = .013) compared with the G genotype. The frequency of the A allele is higher in East Asians, which may contribute to their lean OSA phenotype. Conclusion The OXTR gene may contribute to OSA symptoms via the respiratory control system, although it could be in linkage disequilibrium with a true causal gene.
Collapse
Affiliation(s)
- Hisanori Goto
- Department of Endocrinology and Metabolism, Kanazawa
University Graduate School of Medical Sciences,
Kanazawa, Ishikawa 920-8640, Japan
- Department of Biochemistry and Molecular Vascular Biology,
Kanazawa University Graduate School of Medical Sciences,
Kanazawa, Ishikawa 920-8640, Japan
| | - Yasuhiko Yamamoto
- Department of Biochemistry and Molecular Vascular Biology,
Kanazawa University Graduate School of Medical Sciences,
Kanazawa, Ishikawa 920-8640, Japan
| | - Hiromasa Tsujiguchi
- Department of Hygiene and Public Health, Faculty of Medicine,
Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa
University, Kanazawa City 920-8640,
Japan
- Advanced Preventive Medical Sciences Research Center, Kanazawa
University, Kanazawa, Ishikawa
920-8640, Japan
| | - Takehiro Sato
- Department of Human Biology and Anatomy, Graduate School of
Medicine, University of the Ryukyus, Nishihara,
Okinawa 903-0215, Japan
| | - Reina Yamamoto
- Department of Endocrinology and Metabolism, Kanazawa
University Graduate School of Medical Sciences,
Kanazawa, Ishikawa 920-8640, Japan
| | - Yumie Takeshita
- Department of Endocrinology and Metabolism, Kanazawa
University Graduate School of Medical Sciences,
Kanazawa, Ishikawa 920-8640, Japan
| | - Yujiro Nakano
- Department of Endocrinology and Metabolism, Kanazawa
University Graduate School of Medical Sciences,
Kanazawa, Ishikawa 920-8640, Japan
| | - Takayuki Kannon
- Department of Biomedical Data Science, School of Medicine,
Fujita Health University, Toyoake, Aichi
470-1192, Japan
| | - Kazuyoshi Hosomichi
- Laboratory of Computational Genomics, School of Life Science,
Tokyo University of Pharmacy and Life Sciences,
Hachioji, Tokyo 192-0392, Japan
| | - Keita Suzuki
- Advanced Preventive Medical Sciences Research Center, Kanazawa
University, Kanazawa, Ishikawa
920-8640, Japan
| | - Masaharu Nakamura
- Department of Hygiene and Public Health, Faculty of Medicine,
Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa
University, Kanazawa City 920-8640,
Japan
| | - Yasuhiro Kambayashi
- Department of Hygiene and Public Health, Faculty of Medicine,
Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa
University, Kanazawa City 920-8640,
Japan
- Department of Public Health, Faculty of Veterinary Medicine,
Okayama University of Science, Imabari, Ehime
794-8555, Japan
| | - Jiaye Zhao
- Department of Hygiene and Public Health, Faculty of Medicine,
Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa
University, Kanazawa City 920-8640,
Japan
| | - Atsushi Asai
- Advanced Preventive Medical Sciences Research Center, Kanazawa
University, Kanazawa, Ishikawa
920-8640, Japan
| | - Koji Katano
- Advanced Preventive Medical Sciences Research Center, Kanazawa
University, Kanazawa, Ishikawa
920-8640, Japan
| | - Aya Ogawa
- Department of Hygiene and Public Health, Faculty of Medicine,
Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa
University, Kanazawa City 920-8640,
Japan
| | - Shinobu Fukushima
- Department of Hygiene and Public Health, Faculty of Medicine,
Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa
University, Kanazawa City 920-8640,
Japan
| | - Aki Shibata
- Department of Hygiene and Public Health, Faculty of Medicine,
Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa
University, Kanazawa City 920-8640,
Japan
| | - Fumihiko Suzuki
- Department of Hygiene and Public Health, Faculty of Medicine,
Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa
University, Kanazawa City 920-8640,
Japan
- Department of Geriatric Dentistry, Ohu University School of
Dentistry, Koriyama, Fukushima
963-8611, Japan
| | - Hirohito Tsuboi
- Department of Hygiene and Public Health, Faculty of Medicine,
Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa
University, Kanazawa City 920-8640,
Japan
- Graduate School of Human Sciences, The University of Shiga
Prefecture, Hikone, Shiga 522-8533,
Japan
| | - Akinori Hara
- Department of Hygiene and Public Health, Faculty of Medicine,
Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa
University, Kanazawa City 920-8640,
Japan
- Advanced Preventive Medical Sciences Research Center, Kanazawa
University, Kanazawa, Ishikawa
920-8640, Japan
| | - Mitsuhiro Kometani
- Department of Health Promotion and Medicine of the Future,
Kanazawa University Graduate School of Medical Sciences,
Kanazawa 920-8640, Japan
| | - Shigehiro Karashima
- Institute of Liberal Arts and Science, Kanazawa
University, Kanazawa 920-1192,
Japan
| | - Takashi Yoneda
- Department of Health Promotion and Medicine of the Future,
Kanazawa University Graduate School of Medical Sciences,
Kanazawa 920-8640, Japan
| | - Atsushi Tajima
- Department of Bioinformatics and Genomics, Graduate School of
Advanced Preventive Medical Sciences, Kanazawa University,
Kanazawa, Ishikawa 920-8640, Japan
| | - Hiroyuki Nakamura
- Department of Hygiene and Public Health, Faculty of Medicine,
Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa
University, Kanazawa City 920-8640,
Japan
- Advanced Preventive Medical Sciences Research Center, Kanazawa
University, Kanazawa, Ishikawa
920-8640, Japan
| | - Toshinari Takamura
- Department of Endocrinology and Metabolism, Kanazawa
University Graduate School of Medical Sciences,
Kanazawa, Ishikawa 920-8640, Japan
| |
Collapse
|
13
|
Morones J, Pérez M, Muñoz M, Sánchez E, Ávila M, Topete J, Ventura J, Martínez S. Evaluation of the Effect of an α-Adrenergic Blocker, a PPAR-γ Receptor Agonist, and a Glycemic Regulator on Chronic Kidney Disease in Diabetic Rats. Int J Mol Sci 2024; 25:11372. [PMID: 39518925 PMCID: PMC11545748 DOI: 10.3390/ijms252111372] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Revised: 10/03/2024] [Accepted: 10/16/2024] [Indexed: 11/16/2024] Open
Abstract
Diabetic nephropathy (DN) is a globally widespread complication of diabetes mellitus (DM). Research indicates that pioglitazone and linagliptin mitigate the risk of DN by reducing inflammation, oxidative stress, and fibrosis. The role of tamsulosin in DN is less studied, but it may contribute to reducing oxidative stress and inflammatory responses. The protective effects of combining pioglitazone, linagliptin, and tamsulosin on the kidneys have scarcely been investigated. This study examines the individual and combined effects of these drugs on DN in Wistar rats. Diabetic rats were treated with tamsulosin, pioglitazone, and linagliptin for six weeks. We assessed food and water intake, estimated glomerular filtration rate (eGFR), histological markers, urea, creatinine, glucose, NF-κB, IL-1, IL-10, TGF-β, and Col-IV using immunofluorescence and qPCR. The DN group exhibited hyperglycaemia, reduced eGFR, and tissue damage. Tamsulosin and linagliptin improved eGFR, decreased urinary glucose, and repaired tissue damage. Pioglitazone and its combinations restored serum and urinary markers and reduced tissue damage. Linagliptin lowered serum creatinine and tissue injury. In conclusion, tamsulosin, linagliptin, and pioglitazone demonstrated renoprotective effects in DN.
Collapse
Affiliation(s)
- Jorge Morones
- Department of Morphology, Basic Sciences Center, Universidad Autónoma de Aguascalientes, Aguascalientes 20100, Mexico; (J.M.); (M.P.); (E.S.); (M.Á.)
| | - Mariana Pérez
- Department of Morphology, Basic Sciences Center, Universidad Autónoma de Aguascalientes, Aguascalientes 20100, Mexico; (J.M.); (M.P.); (E.S.); (M.Á.)
| | - Martín Muñoz
- Department of Chemistry, Basic Sciences Center, Universidad Autónoma de Aguascalientes, Aguascalientes 20100, Mexico;
| | - Esperanza Sánchez
- Department of Morphology, Basic Sciences Center, Universidad Autónoma de Aguascalientes, Aguascalientes 20100, Mexico; (J.M.); (M.P.); (E.S.); (M.Á.)
- Family Medicine Unit 8, Instituto Mexicano del Seguro Social (IMSS), Aguascalientes 20180, Mexico
| | - Manuel Ávila
- Department of Morphology, Basic Sciences Center, Universidad Autónoma de Aguascalientes, Aguascalientes 20100, Mexico; (J.M.); (M.P.); (E.S.); (M.Á.)
| | - Jorge Topete
- Department of Nephrology, Regional General Hospital No. 46, Instituto Mexicano del Seguro Social (IMSS), Guadalajara 44910, Mexico;
| | - Javier Ventura
- Department of Morphology, Basic Sciences Center, Universidad Autónoma de Aguascalientes, Aguascalientes 20100, Mexico; (J.M.); (M.P.); (E.S.); (M.Á.)
| | - Sandra Martínez
- Department of Microbiology, Basic Sciences Center, Universidad Autónoma de Aguascalientes, Aguascalientes 20100, Mexico
| |
Collapse
|
14
|
Kusayama T, Nagamori Y, Takeuchi K, Nakagawa Y, Takamura M. Renal autonomic dynamics in hypertension: how can we evaluate sympathetic activity for renal denervation? Hypertens Res 2024; 47:2685-2692. [PMID: 39095482 DOI: 10.1038/s41440-024-01816-2] [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] [Received: 02/19/2024] [Revised: 06/10/2024] [Accepted: 06/29/2024] [Indexed: 08/04/2024]
Abstract
This review explores the various pathophysiological factors influencing antihypertensive effects, involving the regulation of vascular resistance, plasma volume, cardiac function, and the autonomic nervous system, emphasizing the interconnected processes regulating blood pressure (BP). The kidney's pivotal role in BP control and its potential contribution to hypertension is complicated but important to understand the effective mechanisms of renal denervation (RDN), which may be a promising treatment for resistant hypertension. Excessive stimulation of the sympathetic nervous system or the renin-angiotensin-aldosterone system (RAAS) can elevate BP through various physiological changes, contributing to chronic hypertension. Renal sympathetic efferent nerve activation leads to elevated norepinephrine levels and subsequent cascading effects on vasoconstriction, renin release, and sodium reabsorption. RDN reduces BP in resistant hypertension by potentially disrupting sensory afferent nerves, decreasing feedback activation to the central nervous system, and reducing efferent sympathetic nerve activity in the heart and other structures. RDN may also modulate central sympathetic outflow and inhibit renal renin-angiotensin system overactivation. While evidence for RDN efficacy in hypertension is increasing, accurate patient selection becomes crucial, considering complex interactions that vary among patients. This review also discusses methods to evaluate autonomic nerve activity from the golden standard to new potential examination for finding out optimization in stimulation parameters or rigorous patient selection based on appropriate biomarkers.
Collapse
Affiliation(s)
- Takashi Kusayama
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kanazawa University, Ishikawa, Japan.
| | - Yuta Nagamori
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kanazawa University, Ishikawa, Japan
| | - Kazutaka Takeuchi
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kanazawa University, Ishikawa, Japan
| | - Yoichiro Nakagawa
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kanazawa University, Ishikawa, Japan
| | - Masayuki Takamura
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kanazawa University, Ishikawa, Japan
| |
Collapse
|
15
|
Evans LC, Dailey-Krempel B, Lauar MR, Dayton A, Vulchanova L, Osborn JW. Renal interoception in health and disease. Auton Neurosci 2024; 255:103208. [PMID: 39128142 DOI: 10.1016/j.autneu.2024.103208] [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: 04/17/2024] [Revised: 07/19/2024] [Accepted: 07/26/2024] [Indexed: 08/13/2024]
Abstract
Catheter based renal denervation has recently been FDA approved for the treatment of hypertension. Traditionally, the anti-hypertensive effects of renal denervation have been attributed to the ablation of the efferent sympathetic renal nerves. In recent years the role of the afferent sensory renal nerves in the regulation of blood pressure has received increased attention. In addition, afferent renal denervation is associated with reductions in sympathetic nervous system activity. This suggests that reductions in sympathetic drive to organs other than the kidney may contribute to the non-renal beneficial effects observed in clinical trials of catheter based renal denervation. In this review we will provide an overview of the role of the afferent renal nerves in the regulation of renal function and the development of pathophysiologies, both renal and non-renal. We will also describe the central projections of the afferent renal nerves, to give context to the responses seen following their ablation and activation. Finally, we will discuss the emerging role of the kidney as an interoceptive organ. We will describe the potential role of the kidney in the regulation of interoceptive sensitivity and in this context, speculate on the possible pathological consequences of altered renal function.
Collapse
Affiliation(s)
- Louise C Evans
- Department of Surgery, University of Minnesota Medical School, Minneapolis 55455, United States of America
| | - Brianna Dailey-Krempel
- Department of Neuroscience, University of Minnesota, Minneapolis 55455, United States of America
| | - Mariana R Lauar
- Department of Surgery, University of Minnesota Medical School, Minneapolis 55455, United States of America
| | - Alex Dayton
- Division of Nephrology and Hypertension, University of Minnesota Medical School, Minneapolis 55455, United States of America
| | - Lucy Vulchanova
- Department of Neuroscience, University of Minnesota, Minneapolis 55455, United States of America
| | - John W Osborn
- Department of Surgery, University of Minnesota Medical School, Minneapolis 55455, United States of America.
| |
Collapse
|
16
|
Al Ghorani H, Kulenthiran S, Lauder L, Recktenwald MJM, Dederer J, Kunz M, Götzinger F, Ewen S, Ukena C, Böhm M, Mahfoud F. Ultra-long-term efficacy and safety of catheter-based renal denervation in resistant hypertension: 10-year follow-up outcomes. Clin Res Cardiol 2024; 113:1384-1392. [PMID: 38451261 PMCID: PMC11420252 DOI: 10.1007/s00392-024-02417-2] [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: 12/21/2023] [Accepted: 02/16/2024] [Indexed: 03/08/2024]
Abstract
BACKGROUND Randomized sham-controlled trials have confirmed the efficacy and safety of catheter-based renal denervation in hypertension. Data on the very long-term effects of renal denervation are scarce. AIMS This study evaluates the 10-year safety and efficacy of renal denervation in resistant hypertension. METHODS This prospective single-center study included patients with resistant hypertension undergoing radio-frequency renal denervation between 2010 and 2012. Office blood pressure, 24-h ambulatory blood pressure, antihypertensive medication, color duplex sonography, and renal function were assessed after 1-, 2- and 10-years. RESULTS Thirty-nine patients completed the 10-year follow-up (mean follow-up duration 9.4 ± 0.7 years). Baseline office and 24-h ambulatory systolic blood pressure were 164 ± 23 mmHg and 153 ± 16 mmHg, respectively. After 10 years, 24-h ambulatory and office systolic blood pressure were reduced by 16 ± 17 mmHg (P < 0.001) and 14 ± 23 mmHg (P = 0.001), respectively. The number of antihypertensive drugs remained unchanged from 4.9 ± 1.4 to 4.5 ± 1.2 drugs (P = 0.087). The estimated glomerular filtration rate declined within the expected range from 69 (95% CI 63 to 74) to 60 mL/min/1.73m2 (95% CI 53 to 68; P < 0.001) through 10-year follow-up. Three renal artery interventions were documented for progression of pre-existing renal artery stenosis in two patients and one patient with new-onset renal artery stenosis. No other adverse events were observed during the follow-up. CONCLUSION Renal denervation was safe and sustainedly reduced ambulatory and office blood pressure out to 10 years in patients with resistant hypertension.
Collapse
Affiliation(s)
- Hussam Al Ghorani
- Klinik für Innere Medizin III, Kardiologie, Angiologie und Internistische Intensivmedizin, Universitätsklinikum des Saarlandes, Saarland University, Kirrberger Str. 100, Gebäude 41, 66421, Homburg/Saar, Germany.
| | - Saarraaken Kulenthiran
- Klinik für Innere Medizin III, Kardiologie, Angiologie und Internistische Intensivmedizin, Universitätsklinikum des Saarlandes, Saarland University, Kirrberger Str. 100, Gebäude 41, 66421, Homburg/Saar, Germany
| | - Lucas Lauder
- Klinik für Innere Medizin III, Kardiologie, Angiologie und Internistische Intensivmedizin, Universitätsklinikum des Saarlandes, Saarland University, Kirrberger Str. 100, Gebäude 41, 66421, Homburg/Saar, Germany
| | - Michael Johannes Maria Recktenwald
- Klinik für Innere Medizin III, Kardiologie, Angiologie und Internistische Intensivmedizin, Universitätsklinikum des Saarlandes, Saarland University, Kirrberger Str. 100, Gebäude 41, 66421, Homburg/Saar, Germany
| | - Juliane Dederer
- Klinik für Innere Medizin III, Kardiologie, Angiologie und Internistische Intensivmedizin, Universitätsklinikum des Saarlandes, Saarland University, Kirrberger Str. 100, Gebäude 41, 66421, Homburg/Saar, Germany
| | - Michael Kunz
- Klinik für Innere Medizin III, Kardiologie, Angiologie und Internistische Intensivmedizin, Universitätsklinikum des Saarlandes, Saarland University, Kirrberger Str. 100, Gebäude 41, 66421, Homburg/Saar, Germany
| | - Felix Götzinger
- Klinik für Innere Medizin III, Kardiologie, Angiologie und Internistische Intensivmedizin, Universitätsklinikum des Saarlandes, Saarland University, Kirrberger Str. 100, Gebäude 41, 66421, Homburg/Saar, Germany
| | - Sebastian Ewen
- Klinik für Innere Medizin III, Kardiologie, Angiologie und Internistische Intensivmedizin, Universitätsklinikum des Saarlandes, Saarland University, Kirrberger Str. 100, Gebäude 41, 66421, Homburg/Saar, Germany
| | - Christian Ukena
- Klinik für Innere Medizin III, Kardiologie, Angiologie und Internistische Intensivmedizin, Universitätsklinikum des Saarlandes, Saarland University, Kirrberger Str. 100, Gebäude 41, 66421, Homburg/Saar, Germany
- Medizinische Klinik II - Kardiologie/Angiologie, Marien Hospital Herne - Universitätsklinikum der Ruhr Universität Bochum, Hölkeskampring 40, 44625, Herne, Germany
| | - Michael Böhm
- Klinik für Innere Medizin III, Kardiologie, Angiologie und Internistische Intensivmedizin, Universitätsklinikum des Saarlandes, Saarland University, Kirrberger Str. 100, Gebäude 41, 66421, Homburg/Saar, Germany
| | - Felix Mahfoud
- Klinik für Innere Medizin III, Kardiologie, Angiologie und Internistische Intensivmedizin, Universitätsklinikum des Saarlandes, Saarland University, Kirrberger Str. 100, Gebäude 41, 66421, Homburg/Saar, Germany
| |
Collapse
|
17
|
Jiang Q, Yang Q, Zhang C, Hou C, Hong W, Du M, Shan X, Li X, Zhou D, Wen D, Xiong Y, Yang K, Lin Z, Song J, Mo Z, Feng H, Xing Y, Fu X, Liu C, Peng F, Wu L, Li B, Lu W, Yuan JXJ, Wang J, Chen Y. Nephrectomy and high-salt diet inducing pulmonary hypertension and kidney damage by increasing Ang II concentration in rats. Respir Res 2024; 25:288. [PMID: 39080603 PMCID: PMC11290206 DOI: 10.1186/s12931-024-02916-w] [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: 02/07/2024] [Accepted: 07/14/2024] [Indexed: 08/02/2024] Open
Abstract
BACKGROUND Chronic kidney disease (CKD) is a significant risk factor for pulmonary hypertension (PH), a complication that adversely affects patient prognosis. However, the mechanisms underlying this association remain poorly understood. A major obstacle to progress in this field is the lack of a reliable animal model replicating CKD-PH. METHODS This study aimed to establish a stable rat model of CKD-PH. We employed a combined approach, inducing CKD through a 5/6 nephrectomy and concurrently exposing the rats to a high-salt diet. The model's hemodynamics were evaluated dynamically, alongside a comprehensive assessment of pathological changes in multiple organs. Lung tissues and serum samples were collected from the CKD-PH rats to analyze the expression of angiotensin-converting enzyme 2 (ACE2), evaluate the activity of key vascular components within the renin-angiotensin-aldosterone system (RAAS), and characterize alterations in the serum metabolic profile. RESULTS At 14 weeks post-surgery, the CKD-PH rats displayed significant changes in hemodynamic parameters indicative of pulmonary arterial hypertension. Additionally, right ventricular hypertrophy was observed. Notably, no evidence of pulmonary vascular remodeling was found. Further analysis revealed RAAS dysregulation and downregulated ACE2 expression within the pulmonary vascular endothelium of CKD-PH rats. Moreover, the serum metabolic profile of these animals differed markedly from the sham surgery group. CONCLUSIONS Our findings suggest that the development of pulmonary arterial hypertension in CKD-PH rats is likely a consequence of a combined effect: RAAS dysregulation, decreased ACE2 expression in pulmonary vascular endothelial cells, and metabolic disturbances.
Collapse
Grants
- 82370063, 82170069, 82241012, 82120108001, 81970057, 82170065, 82000045, 82270052 National Natural Science Foundation of China
- 82370063, 82170069, 82241012, 82120108001, 81970057, 82170065, 82000045, 82270052 National Natural Science Foundation of China
- National Key Research and Development Program of China
Collapse
Affiliation(s)
- Qian Jiang
- State Key Laboratory of Respiratory Diseases, National Center for Respiratory Medicine, Guangdong Key Laboratory of Vascular Diseases, National Clinical Research Center for Respiratory Diseases, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, 151 Yanjiang Road, Guangzhou, 510120, Guangdong, China
| | - Qifeng Yang
- State Key Laboratory of Respiratory Diseases, National Center for Respiratory Medicine, Guangdong Key Laboratory of Vascular Diseases, National Clinical Research Center for Respiratory Diseases, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, 151 Yanjiang Road, Guangzhou, 510120, Guangdong, China
| | - Chenting Zhang
- State Key Laboratory of Respiratory Diseases, National Center for Respiratory Medicine, Guangdong Key Laboratory of Vascular Diseases, National Clinical Research Center for Respiratory Diseases, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, 151 Yanjiang Road, Guangzhou, 510120, Guangdong, China
| | - Chi Hou
- State Key Laboratory of Respiratory Diseases, National Center for Respiratory Medicine, Guangdong Key Laboratory of Vascular Diseases, National Clinical Research Center for Respiratory Diseases, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, 151 Yanjiang Road, Guangzhou, 510120, Guangdong, China
- Department of Neurology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, Guangdong, China
| | - Wei Hong
- State Key Laboratory of Respiratory Diseases, National Center for Respiratory Medicine, Guangdong Key Laboratory of Vascular Diseases, National Clinical Research Center for Respiratory Diseases, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, 151 Yanjiang Road, Guangzhou, 510120, Guangdong, China
- GMU-GIBH Joint School of Life Sciences, Guangzhou Medical University, Guangzhou, 511436, Guangdong, China
| | - Min Du
- State Key Laboratory of Respiratory Diseases, National Center for Respiratory Medicine, Guangdong Key Laboratory of Vascular Diseases, National Clinical Research Center for Respiratory Diseases, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, 151 Yanjiang Road, Guangzhou, 510120, Guangdong, China
| | - Xiaoqian Shan
- State Key Laboratory of Respiratory Diseases, National Center for Respiratory Medicine, Guangdong Key Laboratory of Vascular Diseases, National Clinical Research Center for Respiratory Diseases, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, 151 Yanjiang Road, Guangzhou, 510120, Guangdong, China
| | - Xuanyi Li
- State Key Laboratory of Respiratory Diseases, National Center for Respiratory Medicine, Guangdong Key Laboratory of Vascular Diseases, National Clinical Research Center for Respiratory Diseases, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, 151 Yanjiang Road, Guangzhou, 510120, Guangdong, China
| | - Dansha Zhou
- State Key Laboratory of Respiratory Diseases, National Center for Respiratory Medicine, Guangdong Key Laboratory of Vascular Diseases, National Clinical Research Center for Respiratory Diseases, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, 151 Yanjiang Road, Guangzhou, 510120, Guangdong, China
| | - Dongmei Wen
- State Key Laboratory of Respiratory Diseases, National Center for Respiratory Medicine, Guangdong Key Laboratory of Vascular Diseases, National Clinical Research Center for Respiratory Diseases, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, 151 Yanjiang Road, Guangzhou, 510120, Guangdong, China
| | - Yuanhui Xiong
- State Key Laboratory of Respiratory Diseases, National Center for Respiratory Medicine, Guangdong Key Laboratory of Vascular Diseases, National Clinical Research Center for Respiratory Diseases, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, 151 Yanjiang Road, Guangzhou, 510120, Guangdong, China
| | - Kai Yang
- State Key Laboratory of Respiratory Diseases, National Center for Respiratory Medicine, Guangdong Key Laboratory of Vascular Diseases, National Clinical Research Center for Respiratory Diseases, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, 151 Yanjiang Road, Guangzhou, 510120, Guangdong, China
| | - Ziying Lin
- State Key Laboratory of Respiratory Diseases, National Center for Respiratory Medicine, Guangdong Key Laboratory of Vascular Diseases, National Clinical Research Center for Respiratory Diseases, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, 151 Yanjiang Road, Guangzhou, 510120, Guangdong, China
| | - Jingjing Song
- Department of Stomatology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, 510120, Guangdong, China
| | - Zhanjie Mo
- State Key Laboratory of Respiratory Diseases, National Center for Respiratory Medicine, Guangdong Key Laboratory of Vascular Diseases, National Clinical Research Center for Respiratory Diseases, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, 151 Yanjiang Road, Guangzhou, 510120, Guangdong, China
| | - Huazhuo Feng
- State Key Laboratory of Respiratory Diseases, National Center for Respiratory Medicine, Guangdong Key Laboratory of Vascular Diseases, National Clinical Research Center for Respiratory Diseases, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, 151 Yanjiang Road, Guangzhou, 510120, Guangdong, China
| | - Yue Xing
- State Key Laboratory of Respiratory Diseases, National Center for Respiratory Medicine, Guangdong Key Laboratory of Vascular Diseases, National Clinical Research Center for Respiratory Diseases, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, 151 Yanjiang Road, Guangzhou, 510120, Guangdong, China
| | - Xin Fu
- State Key Laboratory of Respiratory Diseases, National Center for Respiratory Medicine, Guangdong Key Laboratory of Vascular Diseases, National Clinical Research Center for Respiratory Diseases, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, 151 Yanjiang Road, Guangzhou, 510120, Guangdong, China
| | - Chunli Liu
- State Key Laboratory of Respiratory Diseases, National Center for Respiratory Medicine, Guangdong Key Laboratory of Vascular Diseases, National Clinical Research Center for Respiratory Diseases, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, 151 Yanjiang Road, Guangzhou, 510120, Guangdong, China
| | - Fang Peng
- Department of Critical Medicine, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, Guangdong, China
| | - Liling Wu
- Department of Nephrology, Shenzhen Second People's Hospital, Shenzhen, 518000, Guangdong, China
| | - Bing Li
- State Key Laboratory of Respiratory Diseases, National Center for Respiratory Medicine, Guangdong Key Laboratory of Vascular Diseases, National Clinical Research Center for Respiratory Diseases, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, 151 Yanjiang Road, Guangzhou, 510120, Guangdong, China
| | - Wenju Lu
- State Key Laboratory of Respiratory Diseases, National Center for Respiratory Medicine, Guangdong Key Laboratory of Vascular Diseases, National Clinical Research Center for Respiratory Diseases, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, 151 Yanjiang Road, Guangzhou, 510120, Guangdong, China
| | - Jason X-J Yuan
- Section of Physiology, Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of California San Diego, La Jolla, San Diego, CA, 92093, USA
| | - Jian Wang
- State Key Laboratory of Respiratory Diseases, National Center for Respiratory Medicine, Guangdong Key Laboratory of Vascular Diseases, National Clinical Research Center for Respiratory Diseases, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, 151 Yanjiang Road, Guangzhou, 510120, Guangdong, China.
- Section of Physiology, Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of California San Diego, La Jolla, San Diego, CA, 92093, USA.
- Guangzhou Laboratory, Guangzhou International Bio Island, Guangzhou, 510320, Guangdong, China.
| | - Yuqin Chen
- State Key Laboratory of Respiratory Diseases, National Center for Respiratory Medicine, Guangdong Key Laboratory of Vascular Diseases, National Clinical Research Center for Respiratory Diseases, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, 151 Yanjiang Road, Guangzhou, 510120, Guangdong, China.
- Section of Physiology, Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of California San Diego, La Jolla, San Diego, CA, 92093, USA.
| |
Collapse
|
18
|
Agnesi F, Carlucci L, Burjanadze G, Bernini F, Gabisonia K, Osborn JW, Micera S, Recchia FA. Complex Hemodynamic Responses to Trans-Vascular Electrical Stimulation of the Renal Nerve in Anesthetized Pigs. IEEE OPEN JOURNAL OF ENGINEERING IN MEDICINE AND BIOLOGY 2024; 5:750-758. [PMID: 39691472 PMCID: PMC11651661 DOI: 10.1109/ojemb.2024.3429294] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 07/02/2024] [Accepted: 07/09/2024] [Indexed: 12/19/2024] Open
Abstract
The objective of this study was to characterize hemodynamic changes during trans-vascular stimulation of the renal nerve and their dependence on stimulation parameters. We employed a stimulation catheter inserted in the right renal artery under fluoroscopic guidance, in pigs. Systolic, diastolic and pulse blood pressure and heart rate were recorded during stimulations delivered at different intravascular sites along the renal artery or while varying stimulation parameters (amplitude, frequency, and pulse width). Blood pressure changes during stimulation displayed a pattern more complex than previously described in literature, with a series of negative and positive peaks over the first two minutes, followed by a steady state elevation during the remainder of the stimulation. Pulse pressure and heart rate only showed transient responses, then they returned to baseline values despite constant stimulation. The amplitude of the evoked hemodynamic response was roughly linearly correlated with stimulation amplitude, frequency, and pulse width.
Collapse
Affiliation(s)
- Filippo Agnesi
- Interdisciplinary Research Center “Health Science”Scuola Superiore Sant'AnnaPisa56127Italy
| | - Lucia Carlucci
- Interdisciplinary Research Center “Health Science”Scuola Superiore Sant'AnnaPisa56127Italy
| | - Gia Burjanadze
- Interdisciplinary Research Center “Health Science”Scuola Superiore Sant'AnnaPisa56127Italy
| | - Fabio Bernini
- Interdisciplinary Research Center “Health Science”Scuola Superiore Sant'AnnaPisa56127Italy
| | - Khatia Gabisonia
- Interdisciplinary Research Center “Health Science”Scuola Superiore Sant'AnnaPisa56127Italy
| | - John W Osborn
- Department of Integrative Biology and Physiology, Medical SchoolUniversity of MinnesotaMinneapolisMN55455USA
| | - Silvestro Micera
- The BioRobotics Institute, Department of Excellence in Robotics & AIScuola Superiore Sant'AnnaPisa56127Italy
- Bertarelli Foundation Chair in Translational NeuroEngineering, Centre for Neuroprosthetics and Institute of BioengineeringÉcole Polytechnique Fédérale de Lausanne (EPFL)Lausanne1015Switzerland
| | - Fabio A. Recchia
- Scuola Superiore Sant'AnnaPisa56127Italy
- Lewis Katz School of Medicine, Cardiovascular Research CenterTemple UniversityPhiladelphiaPA19140USA
- Institute of Clinical PhysiologyNational Research CouncilRome00185Italy
| |
Collapse
|
19
|
Katsurada K, Patel KP. The role of afferent renal nerves in regulating sympathetic outflow via central nervous system mechanisms. Hypertens Res 2024; 47:1978-1980. [PMID: 38632458 DOI: 10.1038/s41440-024-01677-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Accepted: 03/20/2024] [Indexed: 04/19/2024]
Affiliation(s)
- Kenichi Katsurada
- Division of Cardiovascular Medicine, Department of Internal Medicine, Jichi Medical University School of Medicine, Shimotsuke, Tochigi, Japan.
- Division of Clinical Pharmacology, Department of Pharmacology, Jichi Medical University School of Medicine, Shimotsuke, Tochigi, Japan.
| | - Kaushik P Patel
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, NE, USA
| |
Collapse
|
20
|
Dulam V, Katta S, Nakka VP. Stroke and Distal Organ Damage: Exploring Brain-Kidney Crosstalk. Neurochem Res 2024; 49:1617-1627. [PMID: 38376748 DOI: 10.1007/s11064-024-04126-8] [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: 12/17/2023] [Revised: 02/11/2024] [Accepted: 02/13/2024] [Indexed: 02/21/2024]
Abstract
Stroke and kidney dysfunction represent significant public health challenges, yet the precise mechanisms connecting these conditions and their severe consequences remain unclear. Individuals experiencing chronic kidney disease (CKD) and acute kidney injury (AKI) are at heightened susceptibility to experiencing repeated strokes. Similarly, a reduced glomerular filtration rate is associated with an elevated risk of suffering a stroke. Prior strokes independently contribute to mortality, end-stage kidney disease, and cardiovascular complications, underscoring the pathological connection between the brain and the kidneys. In cases of AKI, various mechanisms, such as cytokine signaling, leukocyte infiltration, and oxidative stress, establish communication between the brain and the kidneys. The bidirectional relationship between stroke and kidney pathologies involves key factors such as uremic toxins, proteinuria, inflammatory responses, decreased glomerular filtration, impairment of the blood-brain barrier (BBB), oxidative stress, and metabolites produced by the gut microbiota. This review examines potential mechanisms of brain-kidney crosstalk underlying stroke and kidney diseases. It holds significance for comprehending multi-organ dysfunction associated with stroke and for formulating therapeutic strategies to address stroke-induced kidney dysfunction and the bidirectional pathological connection between the kidney and stroke.
Collapse
Affiliation(s)
- Vandana Dulam
- Department of Biochemistry, Acharya Nagarjuna University, Andhra Pradesh, 522510, India
| | - Sireesha Katta
- Department of Biochemistry, Acharya Nagarjuna University, Andhra Pradesh, 522510, India
| | - Venkata Prasuja Nakka
- Department of Biochemistry, Acharya Nagarjuna University, Andhra Pradesh, 522510, India.
| |
Collapse
|
21
|
Wang Z, Li W, Jiang C, Wang J, Hua C, Tang Y, Zhang H, Liu X, Wang Y, Gao M, Lv Q, Dong J, Ma C, Du X. Association between blood pressure variability and risk of kidney function decline in hypertensive patients without chronic kidney disease: a post hoc analysis of Systolic Blood Pressure Intervention Trial study. J Hypertens 2024; 42:1203-1211. [PMID: 38690929 DOI: 10.1097/hjh.0000000000003715] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/03/2024]
Abstract
BACKGROUND Blood pressure variability (BPV) is a risk factor for poor kidney function independent of blood pressure (BP) in chronic kidney disease (CKD). Little is known about the association between kidney function decline and BPV in hypertensive patients without CKD. METHODS A post-hoc analysis of the Systolic Blood Pressure Intervention Trial (SPRINT) was performed. BPV was measured as standard deviation (SD) and average real variability (ARV). Cox proportional hazard models were employed to explore the relationship between BPV and incident CKD and albuminuria. RESULTS A total of 5700 patients were included, with a mean age of 66.4 years old. During a median of 3.29 years follow-up, 150 (2.6%) patients developed CKD and 222 (7.2%) patients developed albuminuria. Patients were divided into four groups according to the quartiles of BPV. Compared with SBPV Q1, the incidence of CKD was higher in SBPV Q2-Q4; hazard ratios and 95% confidence interval were 1.81 (1.07-3.04), 1.85 (1.10-3.12) and 1.90 (1.13-3.19), respectively. The association between incident CKD and albuminuria with DBPV was less significant than SBPV. Similar results were found when measuring BPV as ARV and SD. No interaction was detected in BP-lowering strategy and SBPV on incident CKD and albuminuria ( P > 0.05). CONCLUSION This study found that BPV was a risk factor for incident CKD and albuminuria in patients without CKD, especially SBPV. Although intensive BP control increased the risk of CKD, the association between SBPV and kidney function decline did not differ between the two treatment groups. REGISTRATION URL: https://clinicaltrials.gov/ , Unique identifier: NCT01206062.
Collapse
Affiliation(s)
| | - Wenjie Li
- Department of Cardiology, Anzhen Hospital
| | - Chao Jiang
- Department of Cardiology, Anzhen Hospital
| | - Jue Wang
- Department of Cardiology, Anzhen Hospital
| | - Chang Hua
- Department of Cardiology, Anzhen Hospital
| | | | - Hao Zhang
- Department of Cardiology, Anzhen Hospital
| | - Xinru Liu
- Department of Cardiology, Anzhen Hospital
| | | | | | - Qiang Lv
- Department of Cardiology, Anzhen Hospital
| | - Jianzeng Dong
- Department of Cardiology, Anzhen Hospital
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
| | | | - Xin Du
- Department of Cardiology, Anzhen Hospital
- Heart Health Research Center (HHRC), Beijing, China
- The George Institute for Global Health (Australia), The University of New South Wales, Sydney, Australia
| |
Collapse
|
22
|
Johns EJ. Functional insights into the cardiorenal syndrome. Hypertens Res 2024; 47:1747-1749. [PMID: 38580847 DOI: 10.1038/s41440-024-01665-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2024] [Revised: 03/02/2024] [Accepted: 03/13/2024] [Indexed: 04/07/2024]
Affiliation(s)
- Edward J Johns
- Department of Physiology, College of Medicine and Health, University College Cork, Cork, T12 K8AF, Republic of Ireland.
| |
Collapse
|
23
|
Abdulla MH, AlMarabeh S, Bolger T, Lucking EF, O'Halloran KD, Johns EJ. Effects of intrarenal pelvic infusion of tumour necrosis factor-α and interleukin 1-β on reno-renal reflexes in anaesthetised rats. J Hypertens 2024; 42:1027-1038. [PMID: 38690904 DOI: 10.1097/hjh.0000000000003689] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/03/2024]
Abstract
OBJECTIVE Reno-renal reflexes are disturbed in cardiovascular and hypertensive conditions when elevated levels of pro-inflammatory mediators/cytokines are present within the kidney. We hypothesised that exogenously administered inflammatory cytokines tumour necrosis factor alpha (TNF-α) and interleukin (IL)-1β modulate the renal sympatho-excitatory response to chemical stimulation of renal pelvic sensory nerves. METHODS In anaesthetised rats, intrarenal pelvic infusions of vehicle [0.9% sodium chloride (NaCl)], TNF-α (500 and 1000 ng/kg) and IL-1β (1000 ng/kg) were maintained for 30 min before chemical activation of renal pelvic sensory receptors was performed using randomized intrarenal pelvic infusions of hypertonic NaCl, potassium chloride (KCl), bradykinin, adenosine and capsaicin. RESULTS The increase in renal sympathetic nerve activity (RSNA) in response to intrarenal pelvic hypertonic NaCl was enhanced during intrapelvic TNF-α (1000 ng/kg) and IL-1β infusions by almost 800% above vehicle with minimal changes in mean arterial pressure (MAP) and heart rate (HR). Similarly, the RSNA response to intrarenal pelvic adenosine in the presence of TNF-α (500 ng/kg), but not IL-1β, was almost 200% above vehicle but neither MAP nor HR were changed. There was a blunted sympatho-excitatory response to intrapelvic bradykinin in the presence of TNF-α (1000 ng/kg), but not IL-1β, by almost 80% below vehicle, again without effect on either MAP or HR. CONCLUSION The renal sympatho-excitatory response to renal pelvic chemoreceptor stimulation is modulated by exogenous TNF-α and IL-1β. This suggests that inflammatory mediators within the kidney can play a significant role in modulating the renal afferent nerve-mediated sympatho-excitatory response.
Collapse
Affiliation(s)
- Mohammed H Abdulla
- Department of Physiology, School of Medicine, College of Medicine and Health, University College Cork, Cork, Ireland
| | - Sara AlMarabeh
- Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
- Department of Biopharmaceutics and Clinical Pharmacy, School of Pharmacy, The University of Jordan, Amman, Jordan
| | - Tom Bolger
- Department of Physiology, School of Medicine, College of Medicine and Health, University College Cork, Cork, Ireland
| | - Eric F Lucking
- Department of Physiology, School of Medicine, College of Medicine and Health, University College Cork, Cork, Ireland
| | - Ken D O'Halloran
- Department of Physiology, School of Medicine, College of Medicine and Health, University College Cork, Cork, Ireland
| | - Edward J Johns
- Department of Physiology, School of Medicine, College of Medicine and Health, University College Cork, Cork, Ireland
| |
Collapse
|
24
|
Rahmouni K. Neural Circuits Underlying Reciprocal Cardiometabolic Crosstalk: 2023 Arthur C. Corcoran Memorial Lecture. Hypertension 2024; 81:1233-1243. [PMID: 38533662 PMCID: PMC11096079 DOI: 10.1161/hypertensionaha.124.22066] [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: 03/28/2024]
Abstract
The interplay of various body systems, encompassing those that govern cardiovascular and metabolic functions, has evolved alongside the development of multicellular organisms. This evolutionary process is essential for the coordination and maintenance of homeostasis and overall health by facilitating the adaptation of the organism to internal and external cues. Disruption of these complex interactions contributes to the development and progression of pathologies that involve multiple organs. Obesity-associated cardiovascular risks, such as hypertension, highlight the significant influence that metabolic processes exert on the cardiovascular system. This cardiometabolic communication is reciprocal, as indicated by substantial evidence pointing to the ability of the cardiovascular system to affect metabolic processes, with pathophysiological implications in disease conditions. In this review, I outline the bidirectional nature of the cardiometabolic interaction, with special emphasis on the impact that metabolic organs have on the cardiovascular system. I also discuss the contribution of the neural circuits and autonomic nervous system in mediating the crosstalk between cardiovascular and metabolic functions in health and disease, along with the molecular mechanisms involved.
Collapse
Affiliation(s)
- Kamal Rahmouni
- Department of Neuroscience and Pharmacology, University of Iowa Carver College of Medicine, Iowa City, Iowa
- Veterans Affairs Health Care System, Iowa City, Iowa
- Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City, Iowa
- Fraternal Order of Eagles Diabetes Research Center, University of Iowa Carver College of Medicine, Iowa City, Iowa
- Obesity Research and Education Initiative, University of Iowa Carver College of Medicine, Iowa City, Iowa
- Iowa Neuroscience Institute, University of Iowa Carver College of Medicine, Iowa City, Iowa
| |
Collapse
|
25
|
Pieters TT, Besseling PJ, Bovée DM, Rookmaaker MB, Verhaar MC, Yard B, Hoorn EJ, Joles JA. Discrepancies between transcutaneous and estimated glomerular filtration rates in rats with chronic kidney disease. Kidney Int 2024; 105:1212-1220. [PMID: 38514000 DOI: 10.1016/j.kint.2024.02.020] [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: 06/05/2023] [Revised: 02/03/2024] [Accepted: 02/12/2024] [Indexed: 03/23/2024]
Abstract
Accurate assessment of the glomerular filtration rate (GFR) is crucial for researching kidney disease in rats. Although validation of methods that assess GFR is crucial, large-scale comparisons between different methods are lacking. Both transcutaneous GFR (tGFR) and a newly developed estimated GFR (eGFR) equation by our group provide a low-invasive approach enabling repeated measurements. The tGFR is a single bolus method using FITC-labeled sinistrin to measure GFR based on half-life of the transcutaneous signal, whilst the eGFR is based on urinary sinistrin clearance. Here, we retrospectively compared tGFR, using both 1- and 3- compartment models (tGFR_1c and tGFR_3c, respectively) to the eGFR in a historic cohort of 43 healthy male rats and 84 male rats with various models of chronic kidney disease. The eGFR was on average considerably lower than tGFR-1c and tGFR-3c (mean differences 855 and 216 μL/min, respectively) and only 20 and 47% of measurements were within 30% of each other, respectively. The relative difference between eGFR and tGFR was highest in rats with the lowest GFR. Possible explanations for the divergence are problems inherent to tGFR, such as technical issues with signal measurement, description of the signal kinetics, and translation of half-life to tGFR, which depends on distribution volume. The unknown impact of isoflurane anesthesia used in determining mGFR remains a limiting factor. Thus, our study shows that there is a severe disagreement between GFR measured by tGFR and eGFR, stressing the need for more rigorous validation of the tGFR and possible adjustments to the underlying technique.
Collapse
Affiliation(s)
- Tobias T Pieters
- Department of Nephrology and Hypertension, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Paul J Besseling
- Department of Nephrology and Hypertension, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Dominique M Bovée
- Division of Nephrology and Transplantation, Department of Internal Medicine, Erasmus Medical Center, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Maarten B Rookmaaker
- Department of Nephrology and Hypertension, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Marianne C Verhaar
- Department of Nephrology and Hypertension, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Benito Yard
- Department of Medicine, University Hospital Mannheim, University of Heidelberg, Mannheim, Germany
| | - Ewout J Hoorn
- Division of Nephrology and Transplantation, Department of Internal Medicine, Erasmus Medical Center, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Jaap A Joles
- Department of Nephrology and Hypertension, University Medical Center Utrecht, Utrecht, The Netherlands.
| |
Collapse
|
26
|
Bricher Choque PN, Porter MH, Teixeira MS, Dellê H, Elias RM, Durante B, Dutra MRH, Metz CN, Pavlov VA, Consolim Colombo FM. Cholinergic Stimulation Exerts Cardioprotective Effects and Alleviates Renal Inflammatory Responses after Acute Myocardial Infarction in Spontaneous Hypertensive Rats (SHRs). Pharmaceuticals (Basel) 2024; 17:547. [PMID: 38794117 PMCID: PMC11124479 DOI: 10.3390/ph17050547] [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: 03/07/2024] [Revised: 04/17/2024] [Accepted: 04/18/2024] [Indexed: 05/26/2024] Open
Abstract
BACKGROUND In this investigation, we explored the effects of pharmacological cholinergic stimulation on cardiac function and renal inflammation following acute myocardial infarction (AMI) in spontaneously hypertensive rats (SHRs). METHODS Adult male SHRs were randomized into three experimental groups: sham-operated; AMI + Veh (infarcted, treated with vehicle); and AMI + PY (infarcted, treated with the cholinesterase inhibitor, pyridostigmine bromide (PY)-40 mg/kg, once daily for seven days). Rats were euthanized 7 or 30 days post-surgery. The clinical parameters were assessed on the day before euthanasia. Subsequent to euthanasia, blood samples were collected and renal tissues were harvested for histological and gene expression analyses aimed to evaluate inflammation and injury. RESULTS Seven days post-surgery, the AMI + PY group demonstrated improvements in left ventricular diastolic function and autonomic regulation, and a reduction in renal macrophage infiltration compared to the AMI + Veh group. Furthermore, there was a notable downregulation in pro-inflammatory gene expression and an upregulation in anti-inflammatory gene expression. Analysis 30 days post-surgery showed that PY treatment had a sustained positive effect on renal gene expression, correlated with a decrease in biomarkers, indicative of subclinical kidney injury. CONCLUSIONS Short-term cholinergic stimulation with PY provides both cardiac and renal protection by mitigating the inflammatory response after AMI.
Collapse
Affiliation(s)
- Pamela Nithzi Bricher Choque
- Department of Medicine, Universidade Nove de Julho (Uninove), São Paulo 01504-001, SP, Brazil; (P.N.B.C.); (M.H.P.); (H.D.); (R.M.E.); (M.R.H.D.)
| | - Maria Helena Porter
- Department of Medicine, Universidade Nove de Julho (Uninove), São Paulo 01504-001, SP, Brazil; (P.N.B.C.); (M.H.P.); (H.D.); (R.M.E.); (M.R.H.D.)
| | - Manuella S. Teixeira
- Hypertension Unit, Heart Institute, Medical School, University of São Paulo, São Paulo 05403-900, SP, Brazil; (M.S.T.); (B.D.)
| | - Humberto Dellê
- Department of Medicine, Universidade Nove de Julho (Uninove), São Paulo 01504-001, SP, Brazil; (P.N.B.C.); (M.H.P.); (H.D.); (R.M.E.); (M.R.H.D.)
| | - Rosilene Motta Elias
- Department of Medicine, Universidade Nove de Julho (Uninove), São Paulo 01504-001, SP, Brazil; (P.N.B.C.); (M.H.P.); (H.D.); (R.M.E.); (M.R.H.D.)
| | - Bruno Durante
- Hypertension Unit, Heart Institute, Medical School, University of São Paulo, São Paulo 05403-900, SP, Brazil; (M.S.T.); (B.D.)
| | - Marina Rascio Henriques Dutra
- Department of Medicine, Universidade Nove de Julho (Uninove), São Paulo 01504-001, SP, Brazil; (P.N.B.C.); (M.H.P.); (H.D.); (R.M.E.); (M.R.H.D.)
| | - Christine N. Metz
- The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY 11030, USA; (C.N.M.); (V.A.P.)
- Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY 11550, USA
| | - Valentin A. Pavlov
- The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY 11030, USA; (C.N.M.); (V.A.P.)
- Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY 11550, USA
| | - Fernanda M. Consolim Colombo
- Department of Medicine, Universidade Nove de Julho (Uninove), São Paulo 01504-001, SP, Brazil; (P.N.B.C.); (M.H.P.); (H.D.); (R.M.E.); (M.R.H.D.)
- Hypertension Unit, Heart Institute, Medical School, University of São Paulo, São Paulo 05403-900, SP, Brazil; (M.S.T.); (B.D.)
| |
Collapse
|
27
|
Honetschlägerová Z, Husková Z, Kikerlová S, Sadowski J, Kompanowska-Jezierska E, Táborský M, Vaňourková Z, Kujal P, Červenka L. Renal sympathetic denervation improves pressure-natriuresis relationship in cardiorenal syndrome: insight from studies with Ren-2 transgenic hypertensive rats with volume overload induced using aorto-caval fistula. Hypertens Res 2024; 47:998-1016. [PMID: 38302775 PMCID: PMC10994851 DOI: 10.1038/s41440-024-01583-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 12/20/2023] [Accepted: 12/24/2023] [Indexed: 02/03/2024]
Abstract
The aim was to evaluate the effects of renal denervation (RDN) on autoregulation of renal hemodynamics and the pressure-natriuresis relationship in Ren-2 transgenic rats (TGR) with aorto-caval fistula (ACF)-induced heart failure (HF). RDN was performed one week after creation of ACF or sham-operation. Animals were prepared for evaluation of autoregulatory capacity of renal blood flow (RBF) and glomerular filtration rate (GFR), and of the pressure-natriuresis characteristics after stepwise changes in renal arterial pressure (RAP) induced by aortic clamping. Their basal values of blood pressure and renal function were significantly lower than with innervated sham-operated TGR (p < 0.05 in all cases): mean arterial pressure (MAP) (115 ± 2 vs. 160 ± 3 mmHg), RBF (6.91 ± 0.33 vs. 10.87 ± 0.38 ml.min-1.g-1), urine flow (UF) (11.3 ± 1.79 vs. 43.17 ± 3.24 µl.min-1.g-1) and absolute sodium excretion (UNaV) (1.08 ± 0.27 vs, 6.38 ± 0.76 µmol.min-1.g-1). After denervation ACF TGR showed improved autoregulation of RBF: at lowest RAP level (80 mmHg) the value was higher than in innervated ACF TGR (6.92 ± 0.26 vs. 4.54 ± 0.22 ml.min-1.g-1, p < 0.05). Also, the pressure-natriuresis relationship was markedly improved after RDN: at the RAP of 80 mmHg UF equaled 4.31 ± 0.99 vs. 0.26 ± 0.09 µl.min-1.g-1 recorded in innervated ACF TGR, UNaV was 0.31 ± 0.05 vs. 0.04 ± 0.01 µmol min-1.g-1 (p < 0.05 in all cases). In conclusion, in our model of hypertensive rat with ACF-induced HF, RDN improved autoregulatory capacity of RBF and the pressure-natriuresis relationship when measured at the stage of HF decompensation.
Collapse
Affiliation(s)
- Zuzana Honetschlägerová
- Center for Experimental Medicine, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | - Zuzana Husková
- Center for Experimental Medicine, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | - Soňa Kikerlová
- Center for Experimental Medicine, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | - Janusz Sadowski
- Department of Renal and Body Fluid Physiology, Mossakowski Medical Research Institute, Polish Academy of Sciences, Warsaw, Poland
| | - Elzbieta Kompanowska-Jezierska
- Department of Renal and Body Fluid Physiology, Mossakowski Medical Research Institute, Polish Academy of Sciences, Warsaw, Poland
| | - Miloš Táborský
- Department of Internal Medicine I, Cardiology, University Hospital Olomouc and Palacký University, Olomouc, Czech Republic
| | - Zdenka Vaňourková
- Center for Experimental Medicine, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | - Petr Kujal
- Department of Pathology, 3rd Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Luděk Červenka
- Center for Experimental Medicine, Institute for Clinical and Experimental Medicine, Prague, Czech Republic.
- Department of Internal Medicine I, Cardiology, University Hospital Olomouc and Palacký University, Olomouc, Czech Republic.
| |
Collapse
|
28
|
Milano S, Saponara I, Gerbino A, Lapi D, Lela L, Carmosino M, Dal Monte M, Bagnoli P, Svelto M, Procino G. β3-Adrenoceptor as a new player in the sympathetic regulation of the renal acid-base homeostasis. Front Physiol 2024; 15:1304375. [PMID: 38455846 PMCID: PMC10917900 DOI: 10.3389/fphys.2024.1304375] [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: 09/29/2023] [Accepted: 01/29/2024] [Indexed: 03/09/2024] Open
Abstract
Efferent sympathetic nerve fibers regulate several renal functions activating norepinephrine receptors on tubular epithelial cells. Of the beta-adrenoceptors (β-ARs), we previously demonstrated the renal expression of β3-AR in the thick ascending limb (TAL), the distal convoluted tubule (DCT), and the collecting duct (CD), where it participates in salt and water reabsorption. Here, for the first time, we reported β3-AR expression in the CD intercalated cells (ICCs), where it regulates acid-base homeostasis. Co-localization of β3-AR with either proton pump H+-ATPase or Cl-/HCO3 - exchanger pendrin revealed β3-AR expression in type A, type B, non-A, and non-B ICCs in the mouse kidney. We aimed to unveil the possible regulatory role of β3-AR in renal acid-base homeostasis, in particular in modulating the expression, subcellular localization, and activity of the renal H+-ATPase, a key player in this process. The abundance of H+-ATPase was significantly decreased in the kidneys of β3-AR-/- compared with those of β3-AR+/+ mice. In particular, H+-ATPase reduction was observed not only in the CD but also in the TAL and DCT, which contribute to acid-base transport in the kidney. Interestingly, we found that in in vivo, the absence of β3-AR reduced the kidneys' ability to excrete excess proton in the urine during an acid challenge. Using ex vivo stimulation of mouse kidney slices, we proved that the β3-AR activation promoted H+-ATPase apical expression in the epithelial cells of β3-AR-expressing nephron segments, and this was prevented by β3-AR antagonism or PKA inhibition. Moreover, we assessed the effect of β3-AR stimulation on H+-ATPase activity by measuring the intracellular pH recovery after an acid load in β3-AR-expressing mouse renal cells. Importantly, β3-AR agonism induced a 2.5-fold increase in H+-ATPase activity, and this effect was effectively prevented by β3-AR antagonism or by inhibiting either H+-ATPase or PKA. Of note, in urine samples from patients treated with a β3-AR agonist, we found that β3-AR stimulation increased the urinary excretion of H+-ATPase, likely indicating its apical accumulation in tubular cells. These findings demonstrate that β3-AR activity positively regulates the expression, plasma membrane localization, and activity of H+-ATPase, elucidating a novel physiological role of β3-AR in the sympathetic control of renal acid-base homeostasis.
Collapse
Affiliation(s)
- Serena Milano
- Department of Biosciences, Biotechnologies and Environment, University of Bari, Bari, Italy
| | - Ilenia Saponara
- Department of Biosciences, Biotechnologies and Environment, University of Bari, Bari, Italy
| | - Andrea Gerbino
- Department of Biosciences, Biotechnologies and Environment, University of Bari, Bari, Italy
| | - Dominga Lapi
- Department of Biology, University of Pisa, Pisa, Italy
| | - Ludovica Lela
- Department of Sciences, University of Basilicata, Potenza, Italy
| | - Monica Carmosino
- Department of Sciences, University of Basilicata, Potenza, Italy
| | | | - Paola Bagnoli
- Department of Biology, University of Pisa, Pisa, Italy
| | - Maria Svelto
- Department of Biosciences, Biotechnologies and Environment, University of Bari, Bari, Italy
| | - Giuseppe Procino
- Department of Biosciences, Biotechnologies and Environment, University of Bari, Bari, Italy
| |
Collapse
|
29
|
Azzam O, Nejad SH, Carnagarin R, Nolde JM, Galindo-Kiuchi M, Schlaich MP. Taming resistant hypertension: The promise of novel pharmacologic approaches and renal denervation. Br J Pharmacol 2024; 181:319-339. [PMID: 37715452 DOI: 10.1111/bph.16247] [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: 04/08/2023] [Revised: 08/11/2023] [Accepted: 09/02/2023] [Indexed: 09/17/2023] Open
Abstract
Resistant hypertension is associated with an exceedingly high cardiovascular risk and there remains an unmet therapeutic need driven by pathophysiologic pathways unaddressed by guideline-recommended therapy. While spironolactone is widely considered as the preferable fourth-line drug, its broad application is limited by its side effect profile, especially off-target steroid receptor-mediated effects and hyperkalaemia in at-risk subpopulations. Recent landmark trials have reported promising safety and efficacy results for a number of novel compounds targeting relevant pathophysiologic pathways that remain unopposed by contemporary drugs. These include the dual endothelin receptor antagonist, aprocitentan, the aldosterone synthase inhibitor, baxdrostat and the nonsteroidal mineralocorticoid receptor antagonist finerenone. Furthermore, the evidence base for consideration of catheter-based renal denervation as a safe and effective adjunct therapeutic approach across the clinical spectrum of hypertension has been further substantiated. This review will summarise the recently published evidence on novel antihypertensive drugs and renal denervation in the context of resistant hypertension.
Collapse
Affiliation(s)
- Omar Azzam
- Dobney Hypertension Centre, Medical School-Royal Perth Hospital Unit, Royal Perth Hospital Medical Research Foundation, The University of Western Australia, Perth, Western Australia, Australia
- Department of Nephrology, Royal Perth Hospital, Perth, Western Australia, Australia
| | - Sayeh Heidari Nejad
- Dobney Hypertension Centre, Medical School-Royal Perth Hospital Unit, Royal Perth Hospital Medical Research Foundation, The University of Western Australia, Perth, Western Australia, Australia
| | - Revathy Carnagarin
- Dobney Hypertension Centre, Medical School-Royal Perth Hospital Unit, Royal Perth Hospital Medical Research Foundation, The University of Western Australia, Perth, Western Australia, Australia
| | - Janis M Nolde
- Dobney Hypertension Centre, Medical School-Royal Perth Hospital Unit, Royal Perth Hospital Medical Research Foundation, The University of Western Australia, Perth, Western Australia, Australia
| | - Marcio Galindo-Kiuchi
- Dobney Hypertension Centre, Medical School-Royal Perth Hospital Unit, Royal Perth Hospital Medical Research Foundation, The University of Western Australia, Perth, Western Australia, Australia
| | - Markus P Schlaich
- Dobney Hypertension Centre, Medical School-Royal Perth Hospital Unit, Royal Perth Hospital Medical Research Foundation, The University of Western Australia, Perth, Western Australia, Australia
- Department of Nephrology, Royal Perth Hospital, Perth, Western Australia, Australia
- Department of Cardiology, Royal Perth Hospital, Perth, Western Australia, Australia
| |
Collapse
|
30
|
Seifi B, Kadkhodaei M, Bakhshi E, Sajedizadeh A, Ranjbaran M, Hajiaqaei M. PAG Masked Protective Physical Exercise-Induced High H 2S Levels in 5/6 Nephrectomized Rats. IRANIAN JOURNAL OF PHARMACEUTICAL RESEARCH : IJPR 2024; 23:e145620. [PMID: 39895674 PMCID: PMC11786116 DOI: 10.5812/ijpr-145620] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Revised: 03/30/2024] [Accepted: 04/03/2024] [Indexed: 02/04/2025]
Abstract
BACKGROUND To investigate the mechanisms of exercise therapeutics in preclinical animal models of chronic kidney disease (CKD), PAG (D, L-propargylglycine), an inhibitor of hydrogen sulfide production, was used to examine the protective effects of physical activity on oxidative stress and inflammation levels during CKD. METHODS Male Wistar rats with CKD, induced by the 5/6 nephrectomy procedure and subjected to 8 weeks of exercise training, received injections of PAG, a cystathionine γ-lyase (CSE) inhibitor, at a dose of 19 mg/kg, i.p., twice a week during those 8 weeks. The systolic blood pressure (BP) and renal sympathetic nerve activity (RSNA) were assessed. Additionally, plasma creatinine, BUN, renal hydrogen sulfide (H2S) levels, oxidative stress, and inflammatory markers were evaluated. RESULTS In the PAG group, inhibition of H2S production significantly reversed the improvements in plasma creatinine, BUN, renal malondialdehyde (MDA) level, superoxide dismutase (SOD) activity, TNF-α, and IL-6 that were achieved by exercise. Additionally, high RSNA and high BP, which were also reversed in the PAG group, compared to the CKD group subjected to exercise training. CONCLUSIONS The results suggest that the improvement in BP, oxidative stress, and inflammation status by exercise in CKD may be at least partially due to CSE/H2S signaling.
Collapse
Affiliation(s)
- Behjat Seifi
- Department of Physiology, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Mehri Kadkhodaei
- Department of Physiology, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Enayatollah Bakhshi
- Department of Statistics and Computer, University of Social Welfare and Rehabilitation Sciences, Tehran, Iran
| | - Abdollah Sajedizadeh
- Department of Physiology, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Mina Ranjbaran
- Department of Physiology, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Mahdi Hajiaqaei
- Department of Physiology, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
31
|
Zhao S, Tang C, Weinberger J, Gao D, Hou S. Sprouting of afferent and efferent inputs to pelvic organs after spinal cord injury. J Neuropathol Exp Neurol 2023; 83:20-29. [PMID: 38102789 PMCID: PMC10746698 DOI: 10.1093/jnen/nlad108] [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: 12/17/2023] Open
Abstract
Neural plasticity occurs within the central and peripheral nervous systems after spinal cord injury (SCI). Although central alterations have extensively been studied, it is largely unknown whether afferent and efferent fibers in pelvic viscera undergo similar morphological changes. Using a rat spinal cord transection model, we conducted immunohistochemistry to investigate afferent and efferent innervations to the kidney, colon, and bladder. Approximately 3-4 weeks after injury, immunostaining demonstrated that tyrosine hydroxylase (TH)-labeled postganglionic sympathetic fibers and calcitonin gene-related peptide (CGRP)-immunoreactive sensory terminals sprout in the renal pelvis and colon. Morphologically, sprouted afferent or efferent projections showed a disorganized structure. In the bladder, however, denser CGRP-positive primary sensory fibers emerged in rats with SCI, whereas TH-positive sympathetic efferent fibers did not change. Numerous CGRP-positive afferents were observed in the muscle layer and the lamina propria of the bladder following SCI. TH-positive efferent inputs displayed hypertrophy with large diameters, but their innervation patterns were sustained. Collectively, afferent or efferent inputs sprout widely in the pelvic organs after SCI, which may be one of the morphological bases underlying functional adaptation or maladaptation.
Collapse
Affiliation(s)
- Shunyi Zhao
- Department of Neurobiology and Anatomy, Marion Murray Spinal Cord Research Center, Drexel University College of Medicine, USA
- Department of Pharmacology and Physiology, Drexel University College of Medicine, USA
| | - Chuanxi Tang
- Department of Neurobiology and Cell Biology, Key Laboratory of Neurobiology, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Jeremy Weinberger
- Department of Neurobiology and Anatomy, Marion Murray Spinal Cord Research Center, Drexel University College of Medicine, USA
| | - Dianshuai Gao
- Department of Neurobiology and Cell Biology, Key Laboratory of Neurobiology, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Shaoping Hou
- Department of Neurobiology and Anatomy, Marion Murray Spinal Cord Research Center, Drexel University College of Medicine, USA
| |
Collapse
|
32
|
Xia L, Ye Y, Luo Y, Miao B, Na N. Case report: Application of nonsurgical method in saving transplant renal vein thrombosis caused by acute diarrhea. Front Med (Lausanne) 2023; 10:1275188. [PMID: 38173940 PMCID: PMC10762308 DOI: 10.3389/fmed.2023.1275188] [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: 08/09/2023] [Accepted: 11/27/2023] [Indexed: 01/05/2024] Open
Abstract
Transplant renal vein thrombosis is a rare complication after kidney transplantation, which can seriously threaten graft survival. Though the measures like thrombolytic therapy or operative intervention could be taken to deal with this complication, allograft loss is the most common outcome. Thus, early finding as well as decisive intervention is crucial to saving the graft. Here we present a 46-year-old male patient who underwent kidney transplantation from a cadaveric donor who developed a transplant renal venous thrombosis induced by acute diarrhea more than 1 year after renal transplantation with an initial symptom of sudden anuria and pain in the graft area. Subsequently, serum creatinine levels increased to 810.0 μmol/L. Pelvic CT showed increased vascular density of the transplanted kidney, and contrast-enhanced ultrasound confirmed venous thrombosis. The patient was treated with heparin sodium alone and diuresis gradually resumed. After more than 1 year of follow-up, serum creatinine returned to the baseline level prior to thrombosis. Our case indicates that quick ancillary examination and treatment without hesitation would be indispensable in rescuing allografts with renal vein thrombus. Unfractionated heparin can be recommended as an effective treatment for mid-long-term renal transplantation patients with renal vein thrombosis.
Collapse
Affiliation(s)
| | | | | | | | - Ning Na
- Department of Kidney Transplantation, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| |
Collapse
|
33
|
Petersen JCG, Jonassen TEN, Holstein-Rathlou NH, Petersen LG, Sorensen CM. Dynamic changes in renal sodium handling during sympathetic stimulation in healthy human males. Auton Neurosci 2023; 250:103131. [PMID: 37984257 DOI: 10.1016/j.autneu.2023.103131] [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: 08/10/2023] [Revised: 10/27/2023] [Accepted: 11/13/2023] [Indexed: 11/22/2023]
Abstract
The temporal response of changes in renal sodium reabsorption during increased renal sympathetic nerve activity has not been investigated. Central hypovolemia by application of lower-body negative-pressure (LBNP) elicits baroreceptor mediated sympathetic reflexes to maintain arterial blood pressure. We hypothesized, that during 90 min LBNP, the renal sodium retention would increase rapidly, remain increased during intervention, and return to baseline immediately after end of intervention. METHODS 30 young, healthy, sodium loaded, non-obese males were exposed to -15 mmHg LBNP, -30 mmHg LBNP, -15 mmHg LBNP + renin blockade or time-control (0 mmHg LBNP) for 90 min. Urine was collected every 15 min during 90 min of intervention and 60 min of recovery to identify a possible relation between time of intervention and renal response. RESULTS All intervention groups exhibited a comparable reduction in distal sodium excretion at the end of the intervention (P = 0.46 between groups; -15 mmHg: -3.1 ± 0.9 %, -30 mmHg: -2.9 ± 0.6 %, -15 mmHg + aslikiren: -1.8 ± 0.6 %). -15 mmHg+Aliskiren resulted in a slower onset, but all groups exhibited a continued reduction in sodium excretion after 1 h of recovery despite return to baseline of renin, aldosterone, diuresis and cardiovascular parameters. CONCLUSION Sympathetic stimulation for 90 min via LBNP at -30 mmHg LBNP compared to -15 mmHg did not result in a greater response in fractional Na+ excretion, suggesting that additional baroreceptor unloading did not cause further increases in renal sodium reabsorption. Changes in distal Na+ excretion were linear with respect to time (dose) of intervention, but seem to exhibit a saturation-like effect at a level around 4 %. The lack of recovery after 1 h is also a new finding that warrants further investigation.
Collapse
Affiliation(s)
- J C G Petersen
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark.
| | - T E N Jonassen
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - N-H Holstein-Rathlou
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - L G Petersen
- Department of Aeronautics and Astronautics, Massachusetts Institute of Technology, Cambridge, USA
| | - C M Sorensen
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
34
|
Verdecchia P, Cavallini C, Sclafani R, Santucci A, Notaristefano F, Zingarini G, Colombo GA, Angeli F. Catheter-based renal artery denervation: facts and expectations. Eur J Intern Med 2023; 117:66-77. [PMID: 37544846 DOI: 10.1016/j.ejim.2023.07.041] [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: 07/20/2023] [Revised: 07/27/2023] [Accepted: 07/31/2023] [Indexed: 08/08/2023]
Abstract
Catheter-based renal artery denervation (RAD) is entering a new era. After the disappointing results of SYMPLICITY-HTN 3 trial in year 2014, several technical and methodological advancements led to execution of important SHAM-controlled randomized trials with promising results. Now, the 2023 ESH Guidelines give RAD a class of recommendation II with a Level of Evidence B. Currently, catheter-based RAD has two main areas of application: (a) Hypertensive patients who are still untreated, in whom RAD is a sort of a first-line treatment; (b) Difficult-to-control or true resistant hypertensive patients. Notably, randomized SHAM-controlled trials met their primary end-point in both these conditions. So far, we do not dispose of established predictors of the antihypertensive response to RAD. Some data suggest that younger patients with systo-diastolic hypertension, absence of diffuse atherosclerosis and evidence of sympathetic nervous system overactivity experience a better BP response to the procedure. We reviewed the available data on catheter-based RAD and included an updated meta-analysis of the results of the available SHAM-controlled trials. Overall, the reduction in 24-h systolic blood pressure (BP) after RAD exceeded that after SHAM by 4.58 mmHg (95% CI 3.07-6.10) in untreated patients, and by 3.82 mmHg (95% CI 2.46-5.18) in treated patients, without significant heterogeneity across trials, patient phenotype (untreated versus treated patients) and technique (radiofrequency versus ultrasound). There were no important safety signals related to the procedure. Notably, some data suggest that RAD could be an effective additional approach in patients with atrial fibrillation and other conditions characterized by sympathetic nervous system overactivity.
Collapse
Affiliation(s)
- Paolo Verdecchia
- Division of Cardiology, Hospital S. Maria della Misericordia, Perugia, Italy.
| | - Claudio Cavallini
- Division of Cardiology, Hospital S. Maria della Misericordia, Perugia, Italy
| | - Rocco Sclafani
- Division of Cardiology, Hospital S. Maria della Misericordia, Perugia, Italy
| | - Andrea Santucci
- Division of Cardiology, Hospital S. Maria della Misericordia, Perugia, Italy
| | | | - Gianluca Zingarini
- Division of Cardiology, Hospital S. Maria della Misericordia, Perugia, Italy
| | - Giovanni Andrea Colombo
- Department of Medicine and Technological Innovation (DiMIT), University of Insubria, Varese and Department of Medicine and Cardiopulmonary Rehabilitation, Maugeri Care and Research Institute, IRCCS, Tradate, Italy
| | - Fabio Angeli
- Department of Medicine and Technological Innovation (DiMIT), University of Insubria, Varese and Department of Medicine and Cardiopulmonary Rehabilitation, Maugeri Care and Research Institute, IRCCS, Tradate, Italy
| |
Collapse
|
35
|
Mazza A, Dell'Avvocata F, Torin G, Bulighin F, Battaglia Y, Fiorini F. Does Renal Denervation a Reasonable Treatment Option in Hemodialysis-Dependent Patient with Resistant Hypertension? A Narrative Review. Curr Hypertens Rep 2023; 25:353-363. [PMID: 37672130 PMCID: PMC10598141 DOI: 10.1007/s11906-023-01264-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/22/2023] [Indexed: 09/07/2023]
Abstract
PURPOSE OF REVIEW This narrative review aims to assess the pathophysiology, diagnosis, and treatment of resistant hypertension (RH) in end-stage kidney disease (ESKD) patients on dialysis, with a specific focus on the effect of renal denervation (RDN) on short-term and long-term blood pressure (BP) control. Additionally, we share our experience with the use of RDN in an amyloidotic patient undergoing hemodialysis with RH. RECENT FINDINGS High BP, an important modifiable cardiovascular risk factor, is often observed in patients in ESKD, despite the administration of multiple antihypertensive medications. However, in clinical practice, it remains challenging to identify RH patients on dialysis treatment because of the absence of specific definition for RH in this context. Moreover, the use of invasive approaches, such as RDN, to treat RH is limited by the exclusion of patients with reduced renal function (eGFR < 45 mL/min/1.73 m3) in the clinical trials. Nevertheless, recent studies have reported encouraging results regarding the effectiveness of RDN in stage 3 and 4 chronic kidney disease (CKD) and ESKD patients on dialysis, with reductions in BP of nearly up to 10 mmhg. Although multiple underlying pathophysiological mechanisms contribute to RH, the overactivation of the sympathetic nervous system in ESKD patients on dialysis plays a crucial role. The diagnosis of RH requires both confirmation of adherence to antihypertensive therapy and the presence of uncontrolled BP values by ambulatory BP monitoring or home BP monitoring. Treatment involves a combination of nonpharmacological approaches (such as dry weight reduction, sodium restriction, dialysate sodium concentration reduction, and exercise) and pharmacological treatments. A promising approach for managing of RH is based on catheter-based RDN, through radiofrequency, ultrasound, or alcohol infusion, directly targeting on sympathetic overactivity.
Collapse
Affiliation(s)
- Alberto Mazza
- ESH Excellence Hpertension Centre and Dept. of Internal Medicine, Santa Maria della Misericordia General Hospital, AULSS 5 Polesana, Rovigo, Italy
| | - Fabio Dell'Avvocata
- Cardiovascular Diagnosis and Endoluminal Interventions Unit, Santa Maria della Misericordia General Hospital, AULSS 5 Polesana, Rovigo, Italy
| | - Gioia Torin
- ESH Excellence Hpertension Centre and Dept. of Internal Medicine, Santa Maria della Misericordia General Hospital, AULSS 5 Polesana, Rovigo, Italy
| | - Francesca Bulighin
- Department of Medicine, University of Verona, 37129, Verona, Italy
- Nephrology and Dialysis Unit, Pederzoli Hospital, Via Monte Baldo, 24, 37019, Peschiera del Garda, Italy
| | - Yuri Battaglia
- Department of Medicine, University of Verona, 37129, Verona, Italy.
- Nephrology and Dialysis Unit, Pederzoli Hospital, Via Monte Baldo, 24, 37019, Peschiera del Garda, Italy.
| | - Fulvio Fiorini
- Nephrology, Dialysis and Dietology Unit, Santa Maria della Misericordia General Hospital, AULSS 5 Polesana, Rovigo, Italy
| |
Collapse
|
36
|
Zhou H, Liu H, Li D, Xu Y, Xiao P, Yin Y. Optogenetics supports to identify the function of renal autonomic system. Hypertens Res 2023; 46:2439-2441. [PMID: 37550358 DOI: 10.1038/s41440-023-01379-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Revised: 06/14/2023] [Accepted: 06/29/2023] [Indexed: 08/09/2023]
Affiliation(s)
- Hao Zhou
- Department of Cardiology, the Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Hang Liu
- Department of Cardiology, the Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Dan Li
- Department of Cardiology, the Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yanping Xu
- Department of Cardiology, the Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Peilin Xiao
- Department of Cardiology, the Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yuehui Yin
- Department of Cardiology, the Second Affiliated Hospital of Chongqing Medical University, Chongqing, China.
| |
Collapse
|
37
|
Lauar MR, Evans LC, Van Helden D, Fink GD, Banek CT, Menani JV, Osborn JW. Renal and hypothalamic inflammation in renovascular hypertension: role of afferent renal nerves. Am J Physiol Regul Integr Comp Physiol 2023; 325:R411-R422. [PMID: 37519252 PMCID: PMC10639016 DOI: 10.1152/ajpregu.00072.2023] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Revised: 06/30/2023] [Accepted: 07/25/2023] [Indexed: 08/01/2023]
Abstract
Renal denervation (RDN) is a potential therapy for drug-resistant hypertension. However, whether its effects are mediated by ablation of efferent or afferent renal nerves is not clear. Previous studies have implicated that renal inflammation and the sympathetic nervous system are driven by the activation of afferent and efferent renal nerves. RDN attenuated the renal inflammation and sympathetic activity in some animal models of hypertension. In the 2 kidney,1 clip (2K1C) model of renovascular hypertension, RDN also decreased sympathetic activity; however, mechanisms underlying renal and central inflammation are still unclear. We tested the hypothesis that the mechanisms by which total RDN (TRDN; efferent + afferent) and afferent-specific RDN (ARDN) reduce arterial pressure in 2K1C rats are the same. Male Sprague-Dawley rats were instrumented with telemeters to measure mean arterial pressure (MAP), and after 7 days, a clip was placed on the left renal artery. Rats underwent TRDN, ARDN, or sham surgery of the clipped kidney and MAP was measured for 6 wk. Weekly measurements of water intake (WI), urine output (UO), and urinary copeptin were conducted, and urine was analyzed for cytokines/chemokines. Neurogenic pressor activity (NPA) was assessed at the end of the protocol calculated by the depressor response after intraperitoneal injection of hexamethonium. Rats were euthanized and the hypothalamus and kidneys removed for measurement of cytokine content. MAP, NPA, WI, and urinary copeptin were significantly increased in 2K1C-sham rats, and these responses were abolished by both TRDN and ARDN. 2K1C-sham rats presented with renal and hypothalamic inflammation and these responses were largely mitigated by TRDN and ARDN. We conclude that RDN attenuates 2K1C hypertension primarily by ablation of afferent renal nerves which disrupts bidirectional renal neural-immune pathways.NEW & NOTEWORTHY Hypertension resulting from reduced perfusion of the kidney is dependent on renal sensory nerves, which are linked to inflammation in the kidney and hypothalamus. Afferent renal nerves are required for chronic increases in both water intake and vasopressin release observed following renal artery stenosis. Findings from this study suggest an important role of renal sensory nerves that has previously been underestimated in the pathogenesis of 2K1C hypertension.
Collapse
Affiliation(s)
- Mariana R Lauar
- Department of Surgery, Medical School, University of Minnesota, Minneapolis, Minnesota, United States
- Department of Physiology and Pathology, Dentistry School, São Paulo State University-UNESP, Araraquara, São Paulo, Brazil
| | - Louise C Evans
- Department of Surgery, Medical School, University of Minnesota, Minneapolis, Minnesota, United States
| | - Dusty Van Helden
- Department of Surgery, Medical School, University of Minnesota, Minneapolis, Minnesota, United States
| | - Gregory D Fink
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, Michigan, United States
| | - Christopher T Banek
- Department of Physiology, University of Arizona Health Sciences, Tucson, Arizona, United States
| | - José V Menani
- Department of Physiology and Pathology, Dentistry School, São Paulo State University-UNESP, Araraquara, São Paulo, Brazil
| | - John W Osborn
- Department of Surgery, Medical School, University of Minnesota, Minneapolis, Minnesota, United States
| |
Collapse
|
38
|
Collard D, Velde LVD, Stegehuis VE, Delewi R, Beijk MAM, Zijlstra IJAJ, de Winter RJ, Vogt L, van den Born BJH. Assessment of renal sympathetic control using invasive pressure and flow velocity measurements in humans. Am J Physiol Renal Physiol 2023; 325:F263-F270. [DOI: https:/doi.org/10.1152/ajprenal.00031.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Revised: 06/21/2023] [Accepted: 06/22/2023] [Indexed: 11/30/2023] Open
Abstract
Renal sympathetic innervation is important in the homeostasis of systemic and renal hemodynamics. We showed that renal arterial pressure significantly increased and that flow decreased during static handgrip exercise using direct renal arterial pressure and flow measurements in humans, but with a large difference between individuals. These findings may be useful for future studies aimed to assess the effect of interventions that influence renal sympathetic control.
Collapse
Affiliation(s)
- Didier Collard
- Department of Internal Medicine, Section Vascular Medicine, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Lennart van de Velde
- Department of Internal Medicine, Section Vascular Medicine, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Faculty of Science and Technology, Technical Medical Centre, Multi-Modality Medical Imaging Group, University of Twente, Enschede, The Netherlands
| | - Valerie E. Stegehuis
- Heart Center, Department of Cardiology, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Ronak Delewi
- Heart Center, Department of Cardiology, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Marcel A. M. Beijk
- Heart Center, Department of Cardiology, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | | | - Robbert J. de Winter
- Heart Center, Department of Cardiology, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Liffert Vogt
- Department of Internal Medicine, Section Nephrology, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Bert-Jan H. van den Born
- Department of Internal Medicine, Section Vascular Medicine, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Department of Public and Occupational Health, Amsterdam Public Health Research Institute, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
39
|
Collard D, Velde LVD, Stegehuis VE, Delewi R, Beijk MAM, Zijlstra IJAJ, de Winter RJ, Vogt L, van den Born BJH. Assessment of renal sympathetic control using invasive pressure and flow velocity measurements in humans. Am J Physiol Renal Physiol 2023; 325:F263-F270. [PMID: 37382495 DOI: 10.1152/ajprenal.00031.2023] [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: 02/16/2023] [Revised: 06/21/2023] [Accepted: 06/22/2023] [Indexed: 06/30/2023] Open
Abstract
Renal sympathetic innervation is important in the control of renal and systemic hemodynamics and is a target for pharmacological and catheter-based therapies. The effect of a physiological sympathetic stimulus using static handgrip exercise on renal hemodynamics and intraglomerular pressure in humans is unknown. We recorded renal arterial pressure and flow velocity in patients with a clinical indication for coronary or peripheral angiography using a sensor-equipped guidewire during baseline, handgrip, rest, and hyperemia following intrarenal dopamine (30 μg/kg). Changes in perfusion pressure were expressed as the change in mean arterial pressure, and changes in flow were expressed as a percentage with respect to baseline. Intraglomerular pressure was estimated using a Windkessel model. A total of 18 patients (61% male and 39% female) with a median age of 57 yr (range: 27-85 yr) with successful measurements were included. During static handgrip, renal arterial pressure increased by 15.2 mmHg (range: 4.2-53.0 mmHg), whereas flow decreased by 11.2%, but with a large variation between individuals (range: -13.4 to 49.8). Intraglomerular pressure increased by 4.2 mmHg (range: -3.9 to 22.1 mmHg). Flow velocity under resting conditions remained stable, with a median of 100.6% (range: 82.3%-114.6%) compared with baseline. During hyperemia, maximal flow was 180% (range: 111%-281%), whereas intraglomerular pressure decreased by 9.6 mmHg (interquartile range: 4.8 to 13.9 mmHg). Changes in renal pressure and flow during handgrip exercise were significantly correlated (ρ = -0.68, P = 0.002). Measurement of renal arterial pressure and flow velocity during handgrip exercise allows the identification of patients with higher and lower sympathetic control of renal perfusion. This suggests that hemodynamic measurements may be useful to assess the response to therapeutic interventions aimed at altering renal sympathetic control.NEW & NOTEWORTHY Renal sympathetic innervation is important in the homeostasis of systemic and renal hemodynamics. We showed that renal arterial pressure significantly increased and that flow decreased during static handgrip exercise using direct renal arterial pressure and flow measurements in humans, but with a large difference between individuals. These findings may be useful for future studies aimed to assess the effect of interventions that influence renal sympathetic control.
Collapse
Affiliation(s)
- Didier Collard
- Department of Internal Medicine, Section Vascular Medicine, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Lennart van de Velde
- Department of Internal Medicine, Section Vascular Medicine, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Faculty of Science and Technology, Technical Medical Centre, Multi-Modality Medical Imaging Group, University of Twente, Enschede, The Netherlands
| | - Valerie E Stegehuis
- Heart Center, Department of Cardiology, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Ronak Delewi
- Heart Center, Department of Cardiology, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Marcel A M Beijk
- Heart Center, Department of Cardiology, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - IJsbrand A J Zijlstra
- Department of Radiology, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Robbert J de Winter
- Heart Center, Department of Cardiology, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Liffert Vogt
- Department of Internal Medicine, Section Nephrology, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Bert-Jan H van den Born
- Department of Internal Medicine, Section Vascular Medicine, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Department of Public and Occupational Health, Amsterdam Public Health Research Institute, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
40
|
Wagener M, Dolan E, Arnous S, Galvin J, Murphy AW, Casserly I, Eustace J, O’Connor S, McCreery C, Shand J, Wall C, Matiullah S, Sharif F. Renal Denervation as a Complementary Treatment Option for Uncontrolled Arterial Hypertension: A Situation Assessment. J Clin Med 2023; 12:5634. [PMID: 37685701 PMCID: PMC10488551 DOI: 10.3390/jcm12175634] [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: 07/23/2023] [Revised: 08/20/2023] [Accepted: 08/25/2023] [Indexed: 09/10/2023] Open
Abstract
Uncontrolled arterial hypertension is a major global health issue. Catheter-based renal denervation has shown to lower blood pressure in sham-controlled trials and represents a device-based, complementary treatment option for hypertension. In this situation assessment, the authors, who are practicing experts in hypertension, nephrology, general practice and cardiology in the Republic of Ireland, discuss the current evidence base for the BP-lowering efficacy and safety of catheter-based renal denervation with different modalities. Although important questions remain regarding the identification of responders, and long-term efficacy and safety of the intervention, renal denervation has the potential to provide much-needed help to address hypertension and its adverse consequences. The therapeutic approach needs to be multidisciplinary and personalised to take into account the perspective of patients and healthcare professionals in a shared decision-making process.
Collapse
Affiliation(s)
- Max Wagener
- University Hospital Galway, University of Galway, H91 TK33 Galway, Ireland
| | - Eamon Dolan
- Stroke and Hypertension Unit, Connolly Hospital, D15 X40D Dublin, Ireland
| | - Samer Arnous
- University Hospital Limerick, V94 F858 Limerick, Ireland
| | - Joseph Galvin
- The Mater Misericordiae University Hospital, D07 R2WY Dublin, Ireland
| | - Andrew W. Murphy
- Turloughmore Medical Centre, University of Galway, H91 TK33 Galway, Ireland
| | - Ivan Casserly
- The Mater Misericordiae University Hospital, D07 R2WY Dublin, Ireland
| | | | | | | | - James Shand
- St. Vincent’s University Hospital, D04 T6F4 Dublin, Ireland
| | | | | | - Faisal Sharif
- University Hospital Galway, University of Galway, H91 TK33 Galway, Ireland
| |
Collapse
|
41
|
Hahn RG, Weinberg L, Li Y, Bahlmann H, Bellomo R, Wuethrich PY. Concentrated urine, low urine flow, and postoperative elevation of plasma creatinine: A retrospective analysis of pooled data. PLoS One 2023; 18:e0290071. [PMID: 37590224 PMCID: PMC10434918 DOI: 10.1371/journal.pone.0290071] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Accepted: 08/01/2023] [Indexed: 08/19/2023] Open
Abstract
Elevations of plasma creatinine are common after major surgery, but their pathophysiology is poorly understood. To identify possible contributing mechanisms, we pooled data from eight prospective studies performed in four different countries to study circumstances during which elevation of plasma creatinine occurs. We included 642 patients undergoing mixed major surgeries, mostly open gastrointestinal. Plasma and urinary creatinine and a composite index for renal fluid conservation (Fluid Retention Index, FRI) were measured just before surgery and on the first postoperative morning. Urine flow was measured during the surgery. The results show that patients with a postoperative increase in plasma creatinine by >25% had a high urinary creatinine concentration (11.0±5.9 vs. 8.3±5.6 mmol/L; P< 0001) and higher FRI value (3.2±1.0 vs. 2.9±1.1; P< 0.04) already before surgery was initiated. Progressive increase of plasma creatinine was associated with a gradually lower urine flow and larger blood loss during the surgery (Kruskal-Wallis test, P< 0.001). The patients with an elevation > 25% also showed higher creatinine and a higher FRI value on the first postoperative morning (P< 0.001). Elevations to > 50% of baseline were associated with slightly lower mean arterial pressure (73 ± 10 vs. 80 ± 12 mmHg; P< 0.005). We conclude that elevation of plasma creatinine in the perioperative period was associated with low urine flow and greater blood loss during surgery and with concentrated urine both before and after the surgery. Renal water conservation-related mechanisms seem to contribute to the development of increased plasma creatinine after surgery.
Collapse
Affiliation(s)
- Robert G. Hahn
- Karolinska Institutet at Danderyds Hospital (KIDS), Stockholm, Sweden
| | - Laurence Weinberg
- Department of Anesthesia, Austin Hospital; Melbourne, Australia
- Department of Critical Care, The University of Melbourne, Melbourne, Australia
| | - Yuhong Li
- Department of Anesthesiology, Shulan International Hospital, Shuren University, Hangzhou, 3100004, Zhejiang Province, PR of China
| | - Hans Bahlmann
- Department of Anesthesiology and Intensive Care in Linköping, Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Rinaldo Bellomo
- Department of Intensive Care, Austin Hospital, Melbourne, Australia
- Department of Critical Care, The University of Melbourne; Melbourne, Australia
| | - Patrick Y. Wuethrich
- Department of Anesthesiology and Pain Medicine, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| |
Collapse
|
42
|
Becker BK, Grady CM, Markl AE, Torres Rodriguez AA, Pollock DM. Elevated renal afferent nerve activity in a rat model of endothelin B receptor deficiency. Am J Physiol Renal Physiol 2023; 325:F235-F247. [PMID: 37348026 PMCID: PMC10396274 DOI: 10.1152/ajprenal.00064.2023] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Revised: 06/13/2023] [Accepted: 06/21/2023] [Indexed: 06/24/2023] Open
Abstract
Renal nerves have been an attractive target for interventions aimed at lowering blood pressure; however, the specific roles of renal afferent (sensory) versus efferent sympathetic nerves in mediating hypertension are poorly characterized. A number of studies have suggested that a sympathoexcitatory signal conveyed by renal afferents elicits increases in blood pressure, whereas other studies identified sympathoinhibitory afferent pathways. These sympathoinhibitory pathways have been identified as protective against salt-sensitive increases in blood pressure through endothelin B (ETB) receptor activation. We hypothesized that ETB-deficient (ETB-def) rats, which are devoid of functional ETB receptors except in adrenergic tissues, lack appropriate sympathoinhibition and have lower renal afferent nerve activity following a high-salt diet compared with transgenic controls. We found that isolated renal pelvises from high salt-fed ETB-def animals lack a response to a physiological stimulus, prostaglandin E2, compared with transgenic controls but respond equally to a noxious stimulus, capsaicin. Surprisingly, we observed elevated renal afferent nerve activity in intact ETB-def rats compared with transgenic controls under both normal- and high-salt diets. ETB-def rats have been previously shown to have heightened global sympathetic tone, and we also observed higher total renal sympathetic nerve activity in ETB-def rats compared with transgenic controls under both normal- and high-salt diets. These data indicate that ETB receptors are integral mediators of the sympathoinhibitory renal afferent reflex (renorenal reflex), and, in a genetic rat model of ETB deficiency, the preponderance of sympathoexcitatory renal afferent nerve activity prevails and may contribute to hypertension.NEW & NOTEWORTHY Here, we found that endothelin B receptors are an important contributor to renal afferent nerve responsiveness to a high-salt diet. Rats lacking endothelin B receptors have increased afferent nerve activity that is not responsive to a high-salt diet.
Collapse
Affiliation(s)
- Bryan K Becker
- Cardio-Renal Physiology and Medicine, Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, United States
| | - Caroline M Grady
- Cardio-Renal Physiology and Medicine, Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, United States
| | - Alexa E Markl
- Cardio-Renal Physiology and Medicine, Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, United States
| | - Alfredo A Torres Rodriguez
- Cardio-Renal Physiology and Medicine, Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, United States
| | - David M Pollock
- Cardio-Renal Physiology and Medicine, Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, United States
| |
Collapse
|
43
|
Parvin I, Gauthier MM, Dennis MR, Encinas NM, Nangia EL, Schwartz KL, Banek CT. Sequential afferent and sympathetic renal denervation impact on cardiovascular and renal homeostasis in the male Sprague-Dawley rat. Life Sci 2023; 325:121768. [PMID: 37169146 PMCID: PMC10225348 DOI: 10.1016/j.lfs.2023.121768] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 05/05/2023] [Accepted: 05/05/2023] [Indexed: 05/13/2023]
Abstract
Renal denervation (RDNx) is emerging as a promising treatment for cardiovascular disease, yet the underlying mechanisms and contributions of afferent (sensory) and efferent (sympathetic) renal nerves in healthy conditions remains limited. We hypothesize that sympathetic renal nerves contribute to long-term MAP and renal function, whereas afferent renal nerves do not contribute to the maintenance of cardiovascular and renal function. To test this hypothesis, we performed two experiments. In experiment one, we performed total renal denervation (T-RDNx), ablating afferent and sympathetic renal nerves, in normotensive adult SD rats to determine effects on MAP and renal function. Experiment 2 employed a sequential surgical ablation using: (1) afferent targeted renal denervation (A-RDNx), then (2) sympathetic (T-RDNx) denervation to determine the individual contributions to cardiovascular and renal homeostasis. In experiment 1, MAP decreased following T-RDNx and GFR increased. In experiment 2, A-RDNx led to an increase in MAP but did not change renal function. In contrast, T-RDNx decreased MAP and improved renal filtration. Together, these data partially support our hypothesis that renal sympathetic nerves contribute to the chronic regulation of arterial pressure and renal function. Contrary to the hypothesis, A-RDNx produced an increase in MAP without a detected change in renal function. We concluded that renal sympathetic nerves influence MAP and renal function regulation through a well-defined tonic contribution to renal vascular resistance and sodium reabsorption, whereas afferent renal nerves likely contribute to the maintenance of MAP through a tonic sympatho-inhibitory, negative feedback regulation in the normotensive, healthy rat.
Collapse
Affiliation(s)
- Irin Parvin
- Department of Physiology, University of Arizona College of Medicine, Tucson, AZ, USA
| | - Madeline M Gauthier
- Department of Physiology, University of Arizona College of Medicine, Tucson, AZ, USA
| | - Melissa R Dennis
- Department of Physiology, University of Arizona College of Medicine, Tucson, AZ, USA
| | - Noah M Encinas
- Department of Physiology, University of Arizona College of Medicine, Tucson, AZ, USA
| | - Ellen L Nangia
- Department of Physiology, University of Arizona College of Medicine, Tucson, AZ, USA
| | - Kyle L Schwartz
- Department of Physiology, University of Arizona College of Medicine, Tucson, AZ, USA
| | - Christopher T Banek
- Department of Physiology, University of Arizona College of Medicine, Tucson, AZ, USA.
| |
Collapse
|
44
|
Schwäble Santamaria A, Grassi M, Meeusen JW, Lieske JC, Scott R, Robertson A, Schiffer E. Performance of Nuclear Magnetic Resonance-Based Estimated Glomerular Filtration Rate in a Real-World Setting. Bioengineering (Basel) 2023; 10:717. [PMID: 37370648 PMCID: PMC10295228 DOI: 10.3390/bioengineering10060717] [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: 04/20/2023] [Revised: 05/25/2023] [Accepted: 06/06/2023] [Indexed: 06/29/2023] Open
Abstract
An accurate estimate of glomerular filtration rate (eGFR) is essential for proper clinical management, especially in patients with kidney dysfunction. This prospective observational study evaluated the real-world performance of the nuclear magnetic resonance (NMR)-based GFRNMR equation, which combines creatinine, cystatin C, valine, and myo-inositol with age and sex. We compared GFRNMR performance to that of the 2021 CKD-EPI creatinine and creatinine-cystatin C equations (CKD-EPI2021Cr and CKD-EPI2021CrCys), using 115 fresh routine samples of patients scheduled for urinary iothalamate clearance measurement (mGFR). Median bias to mGFR of the three eGFR equations was comparably low, ranging from 0.4 to 2.0 mL/min/1.73 m2. GFRNMR outperformed the 2021 CKD-EPI equations in terms of precision (interquartile range to mGFR of 10.5 vs. 17.9 mL/min/1.73 m2 for GFRNMR vs. CKD-EPI2021CrCys; p = 0.01) and accuracy (P15, P20, and P30 of 66.1% vs. 48.7% [p = 0.007], 80.0% vs. 60.0% [p < 0.001] and 95.7% vs. 86.1% [p = 0.006], respectively, for GFRNMR vs. CKD-EPI2021CrCys). Clinical parameters such as etiology, comorbidities, or medications did not significantly alter the performance of the three eGFR equations. Altogether, this study confirmed the utility of GFRNMR for accurate GFR estimation, and its potential value in routine clinical practice for improved medical care.
Collapse
Affiliation(s)
| | - Marcello Grassi
- Department of Research and Development, Numares AG, 93053 Regensburg, Germany
| | - Jeffrey W. Meeusen
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN 55905, USA
| | - John C. Lieske
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN 55905, USA
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN 55905, USA
| | - Renee Scott
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN 55905, USA
| | - Andrew Robertson
- Department of Research and Development, Numares AG, 93053 Regensburg, Germany
| | - Eric Schiffer
- Department of Research and Development, Numares AG, 93053 Regensburg, Germany
| |
Collapse
|
45
|
Zhu Q, Xiao L, Cheng G, He J, Yin C, Wang L, Wang Q, Li L, Wei B, Weng Y, Geng F, Shen XZ, Shi P. Self-maintaining macrophages within the kidney contribute to salt and water balance by modulating kidney sympathetic nerve activity. Kidney Int 2023:S0085-2538(23)00343-5. [PMID: 37224917 DOI: 10.1016/j.kint.2023.04.023] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Revised: 04/18/2023] [Accepted: 04/20/2023] [Indexed: 05/26/2023]
Abstract
The kidney is critical in controlling salt and water balance, with the interstitium involved with a variety of components including immune cells in steady state. However, the roles of resident immune cells in kidney physiology are largely unknown. To help unravel some of these unknowns, we employed cell fate mapping, and identified a population of embryo-derived self-maintaining macrophages (SM-MØ) that were independent of the bone marrow in adult mouse kidneys. This kidney-specific SM-MØ population was distinctive from the kidney monocyte-derived macrophages in transcriptome and in their distribution. Specifically, the SM-MØ highly expressed nerve-associated genes; high-resolution confocal microscopy revealed that the SM-MØ in the cortex were in close association with sympathetic nerves and there was a dynamical interaction between macrophages and sympathetic nerves when live kidney sections were monitored. Kidney-specific depletion of the SM-MØ resulted in reduced sympathetic distribution and tone, leading to reduced renin secretion, increased glomerular filtration rate and solute diuresis, which caused salt decompensation and significant weight loss under a low-salt diet challenge. Supplementation of L-3,4-dihydroxyphenylserine which is converted to norepinephrine in vivo rescued the phenotype of SM-MØ-depleted mice. Thus, our findings provide insights in kidney macrophage heterogeneity and address a non-canonical role of macrophages in kidney physiology. In contrast to the well-appreciated way of central regulation, local regulation of sympathetic nerve distribution and activities in the kidney was uncovered.
Collapse
Affiliation(s)
- Qian Zhu
- Department of Physiology, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China;; Department of Cardiology of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China;; Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province, Department of Cardiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Liang Xiao
- Kidney Disease Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Guo Cheng
- Department of Cardiology of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China;; Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Jian He
- Department of Physiology, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China;; Department of Cardiology of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Chunyou Yin
- Department of Physiology, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China;; Department of Cardiology of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Liang Wang
- Department of Neurology of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Qiang Wang
- Department of Laboratory Medicine, Affiliated Zhejiang Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Li Li
- Department of Pharmacy, Affiliated Zhejiang Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Bo Wei
- Department of Cardiology of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China;; Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Yuancheng Weng
- Department of Physiology, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China;; Department of Cardiology of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Fei Geng
- Department of Blood of Transfusion, Affiliated Zhejiang Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Xiao Z Shen
- Department of Physiology, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China;; Department of Cardiology of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China;.
| | - Peng Shi
- Department of Cardiology of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China;; Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China;.
| |
Collapse
|
46
|
Monteiro LM, Barbosa CF, Lichtenecker DCK, Argeri R, Gomes GN. Sex modifies the renal consequences of high fructose consumption introduced after weaning. Front Physiol 2023; 14:1090090. [PMID: 37008005 PMCID: PMC10050681 DOI: 10.3389/fphys.2023.1090090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Accepted: 03/03/2023] [Indexed: 03/17/2023] Open
Abstract
After lactation, many children consume fructose-rich processed foods. However, overconsumption of these foods can predispose individuals to non-communicable chronic diseases, which can have different repercussions depending on the sex. Thus, we evaluated the effects of fructose overload introduced after weaning on the renal function of young rats of both sexes.Methods: After weaning, male and female offspring of Wistar rats were assigned to drink water (the male/water and female/water groups) or 20% D-fructose solution (male/fructose and female/fructose groups). Food and water or fructose solution was offered ad libitum. Rats were evaluated at 4 months. Parameters analyzed: blood pressure, body weight, triglyceride levels, glomerular filtration rate, sodium, potassium, calcium, and magnesium excretion, macrophage infiltration, and eNOS and 8OHdG expression in renal tissue. CEUA-UNIFESP: 2757270117.Results: Fructose intake affected the blood pressure, body weight, and plasma triglyceride in all rats. Glomerular filtration rate was significantly reduced in males that received fructose when compared to that of the control group. Sodium and potassium excretion decreased in all fructose-treated rats; however, the excreted load of these ions was significantly higher in females than in males. In the female control group, calcium excretion was higher than that of the male control group. Fructose overload increased magnesium excretion in females, and also increased macrophage infiltration and reduced eNOS expression in both males and females.Conclusion: Fructose overload introduced after weaning caused metabolic and renal changes in rats. Renal function was more affected in males; however, several significant alterations were also observed in the female-fructose group.
Collapse
Affiliation(s)
- Letícia Maria Monteiro
- Department of Physiology, Escola Paulista de Medicina, Federal University of São Paulo, São Paulo, Brazil
- Postgraduate Program in Translational Medicine, Department of Medicine, Escola Paulista de Medicina, Federal University of São Paulo, São Paulo, Brazil
| | - Celine Farias Barbosa
- Department of Physiology, Escola Paulista de Medicina, Federal University of São Paulo, São Paulo, Brazil
| | | | - Rogério Argeri
- Department of Physiology, Escola Paulista de Medicina, Federal University of São Paulo, São Paulo, Brazil
- Postgraduate Program in Translational Medicine, Department of Medicine, Escola Paulista de Medicina, Federal University of São Paulo, São Paulo, Brazil
| | - Guiomar Nascimento Gomes
- Department of Physiology, Escola Paulista de Medicina, Federal University of São Paulo, São Paulo, Brazil
- *Correspondence: Guiomar Nascimento Gomes,
| |
Collapse
|
47
|
Janas A, Wojakowski W. Peregrine system infusion catheter for neurolytic renal denervation in hypertension: an overview of its safety and efficacy. Expert Rev Med Devices 2023; 20:179-186. [PMID: 36846949 DOI: 10.1080/17434440.2023.2183838] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/01/2023]
Abstract
INTRODUCTION Resistant hypertension (HTN), despite the tremendous advances in pharmacotherapy, is a major global problem. Transcatheter renal denervation (RDN) could be a pertinent strategy for resistant HTN and patients with poor pharmacotherapy adherence. Nonetheless, the adoption of energy-based RDN in clinical practice is slow and alternative approaches are needed. AREAS COVERED The review focuses on the assessment of the Peregrine System Infusion Catheters. The system is designed for chemically mediated transcatheter RDN by the infusion publications on the Peregrine system. The theoretical assumptions for chemically mediated RDN, design of the system, data from preclinical and clinical studies, and further perspectives are discussed. EXPERT OPINION Peregrine System Infusion Catheters are the only catheter on the market designed for chemically mediated RDN by the infusion of the neurolytic agent. Chemical neurolysis more efficiently destroys nerves around the renal artery in comparison to energy-based catheters, due to deeper tissue penetration and circumferential distribution resulting in a wider range of effective nerve injury. Chemically mediated RDN by the infusion of the neurolytic agent (alcohol) has an excellent safety profile as confirmed in initial clinical trials which also suggested high efficacy. Currently, there is an ongoing phase III sham-control study. Other possible applications of this technology include clinical settings like heart failure or atrial fibrillation.
Collapse
Affiliation(s)
- Adam Janas
- Faculty of Medicine and Health Sciences, Andrzej Frycz Modrzewski Cracow University, Kraków, Poland.,American Heart of Poland, Center of Cardiovascular Research and Development, Poland
| | - Wojciech Wojakowski
- Division of Cardiology and Structural Heart Diseases, Medical University of Silesia, Katowice, Poland
| |
Collapse
|
48
|
Renal sympathetic activity: A key modulator of pressure natriuresis in hypertension. Biochem Pharmacol 2023; 208:115386. [PMID: 36535529 DOI: 10.1016/j.bcp.2022.115386] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Revised: 12/11/2022] [Accepted: 12/13/2022] [Indexed: 12/23/2022]
Abstract
Hypertension is a complex disorder ensuing necessarily from alterations in the pressure-natriuresis relationship, the main determinant of long-term control of blood pressure. This mechanism sets natriuresis to the level of blood pressure, so that increasing pressure translates into higher osmotically driven diuresis to reduce volemia and control blood pressure. External factors affecting the renal handling of sodium regulate the pressure-natriuresis relationship so that more or less natriuresis is attained for each level of blood pressure. Hypertension can thus only develop following primary alterations in the pressure to natriuresis balance, or by abnormal activity of the regulation network. On the other hand, increased sympathetic tone is a very frequent finding in most forms of hypertension, long regarded as a key element in the pathophysiological scenario. In this article, we critically analyze the interplay of the renal component of the sympathetic nervous system and the pressure-natriuresis mechanism in the development of hypertension. A special focus is placed on discussing recent findings supporting a role of baroreceptors as a component, along with the afference of reno-renal reflex, of the input to the nucleus tractus solitarius, the central structure governing the long-term regulation of renal sympathetic efferent tone.
Collapse
|
49
|
Zucker IH, Xia Z, Wang HJ. Potential Neuromodulation of the Cardio-Renal Syndrome. J Clin Med 2023; 12:803. [PMID: 36769450 PMCID: PMC9917464 DOI: 10.3390/jcm12030803] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2022] [Revised: 01/16/2023] [Accepted: 01/17/2023] [Indexed: 01/20/2023] Open
Abstract
The cardio-renal syndrome (CRS) type 2 is defined as a progressive loss of renal function following a primary insult to the myocardium that may be either acute or chronic but is accompanied by a decline in myocardial pump performance. The treatment of patients with CRS is difficult, and the disease often progresses to end-stage renal disease that is refractory to conventional therapy. While a good deal of information is known concerning renal injury in the CRS, less is understood about how reflex control of renal sympathetic nerve activity affects this syndrome. In this review, we provide insight into the role of the renal nerves, both from the afferent or sensory side and from the efferent side, in mediating renal dysfunction in CRS. We discuss how interventions such as renal denervation and abrogation of systemic reflexes may be used to alleviate renal dysfunction in the setting of chronic heart failure. We specifically focus on a novel cardiac sensory reflex that is sensitized in heart failure and activates the sympathetic nervous system, especially outflow to the kidney. This so-called Cardiac Sympathetic Afferent Reflex (CSAR) can be ablated using the potent neurotoxin resinferitoxin due to the high expression of Transient Receptor Potential Vanilloid 1 (TRPV1) receptors. Following ablation of the CSAR, several markers of renal dysfunction are reversed in the post-myocardial infarction heart failure state. This review puts forth the novel idea of neuromodulation at the cardiac level in the treatment of CRS Type 2.
Collapse
Affiliation(s)
- Irving H. Zucker
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Zhiqiu Xia
- Department of Anesthesiology, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Han-Jun Wang
- Department of Anesthesiology, University of Nebraska Medical Center, Omaha, NE 68198, USA
| |
Collapse
|
50
|
Sodium Homeostasis, a Balance Necessary for Life. Nutrients 2023; 15:nu15020395. [PMID: 36678265 PMCID: PMC9862583 DOI: 10.3390/nu15020395] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 01/09/2023] [Accepted: 01/10/2023] [Indexed: 01/15/2023] Open
Abstract
Body sodium (Na) levels must be maintained within a narrow range for the correct functioning of the organism (Na homeostasis). Na disorders include not only elevated levels of this solute (hypernatremia), as in diabetes insipidus, but also reduced levels (hyponatremia), as in cerebral salt wasting syndrome. The balance in body Na levels therefore requires a delicate equilibrium to be maintained between the ingestion and excretion of Na. Salt (NaCl) intake is processed by receptors in the tongue and digestive system, which transmit the information to the nucleus of the solitary tract via a neural pathway (chorda tympani/vagus nerves) and to circumventricular organs, including the subfornical organ and area postrema, via a humoral pathway (blood/cerebrospinal fluid). Circuits are formed that stimulate or inhibit homeostatic Na intake involving participation of the parabrachial nucleus, pre-locus coeruleus, medial tuberomammillary nuclei, median eminence, paraventricular and supraoptic nuclei, and other structures with reward properties such as the bed nucleus of the stria terminalis, central amygdala, and ventral tegmental area. Finally, the kidney uses neural signals (e.g., renal sympathetic nerves) and vascular (e.g., renal perfusion pressure) and humoral (e.g., renin-angiotensin-aldosterone system, cardiac natriuretic peptides, antidiuretic hormone, and oxytocin) factors to promote Na excretion or retention and thereby maintain extracellular fluid volume. All these intake and excretion processes are modulated by chemical messengers, many of which (e.g., aldosterone, angiotensin II, and oxytocin) have effects that are coordinated at peripheral and central level to ensure Na homeostasis.
Collapse
|