1
|
Masuda T, Nagata D. Glomerular pressure and tubular oxygen supply: a critical dual target for renal protection. Hypertens Res 2024; 47:3330-3337. [PMID: 39397109 DOI: 10.1038/s41440-024-01944-9] [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: 08/13/2024] [Revised: 09/25/2024] [Accepted: 09/27/2024] [Indexed: 10/15/2024]
Abstract
The primary treatment goal of chronic kidney disease (CKD) is preserving renal function and preventing its progression to end-stage renal disease. Glomerular hypertension and tubular hypoxia are critical risk factors in CKD progression. However, the renal hemodynamics make it difficult to avoid both factors due to the existence of peritubular capillaries that supply oxygen to the renal tubules downstream from the glomerulus through the efferent arteriole. In the treatment strategies for balancing glomerular pressure and tubular oxygen supply, afferent and efferent arterioles of the glomerulus determine glomerular filtration rate and blood flow to the peritubular capillaries. Therefore, sodium-glucose cotransporter 2 inhibitors and angiotensin receptor-neprilysin inhibitors as well as classical renin-angiotensin system inhibitors, which can change the diameter of afferent and/or efferent arterioles, are promising options for balancing this dual target and achieving renal protection. This review focuses on the clinical importance of glomerular pressure and tubular oxygen supply and proposes an effective treatment modality for this dual target.
Collapse
Affiliation(s)
- Takahiro Masuda
- Division of Nephrology, Department of Medicine, Jichi Medical University, Shimotsuke, Tochigi, Japan.
| | - Daisuke Nagata
- Division of Nephrology, Department of Medicine, Jichi Medical University, Shimotsuke, Tochigi, Japan
| |
Collapse
|
2
|
Xu Y, Lu F, Wang M, Wang L, Ye C, Yang S, Wang C. Shen Shuai II recipe improves renal hypoxia to attenuate renal injury in 5/6 renal ablation/infarction rats and effect evaluation using blood oxygenation level-dependent functional magnetic resonance imaging. Ren Fail 2024; 46:2338565. [PMID: 38622926 PMCID: PMC11022919 DOI: 10.1080/0886022x.2024.2338565] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Accepted: 03/31/2024] [Indexed: 04/17/2024] Open
Abstract
Background: Renal hypoxia plays a key role in the progression of chronic kidney disease (CKD). Shen Shuai II Recipe (SSR) has shown good results in the treatment of CKD as a common herbal formula. This study aimed to explore the effect of SSR on renal hypoxia and injury in CKD rats. Methods: Twenty-five Wistar rats underwent 5/6 renal ablation/infarction (A/I) surgery were randomly divided into three groups: 5/6 (A/I), 5/6 (A/I) + losartan (LOS), and 5/6 (A/I) + SSR groups. Another eight normal rats were used as the Sham group. After 8-week corresponding interventions, blood oxygenation level-dependent functional magnetic resonance imaging (BOLD-fMRI) was performed to evaluate renal oxygenation in all rats, and biochemical indicators were used to measure kidney and liver function, hemoglobin, and proteinuria. The expression of fibrosis and hypoxia-related proteins was analyzed using immunoblotting examination. Results: Renal oxygenation, evaluated by BOLD-fMRI as cortical and medullary T2* values (COT2* and MET2*), was decreased in 5/6 (A/I) rats, but increased after SSR treatment. SSR also downregulated the expression of hypoxia-inducible factor-1α (HIF-1α) in 5/6 (A/I) kidneys. With the improvement of renal hypoxia, renal function and fibrosis were improved in 5/6 (A/I) rats, accompanied by reduced proteinuria. Furthermore, the COT2* and MET2* were significantly positively correlated with the levels of creatinine clearance rate (Ccr) and hemoglobin, but negatively associated with the levels of serum creatinine (SCr), blood urea nitrogen (BUN), serum cystatin C (CysC), serum uric acid (UA), 24-h urinary protein (24-h Upr), and urinary albumin:creatinine ratio (UACR). Conclusion: The degree of renal oxygenation reduction is correlated with the severity of renal injury in CKD. SSR can improve renal hypoxia to attenuate renal injury in 5/6 (A/I) rats of CKD.
Collapse
Affiliation(s)
- Yizeng Xu
- Department of Nephrology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Key Laboratory of Liver and Kidney Diseases, Ministry of Education, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- TCM Institute of Kidney Disease, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Shanghai Key Laboratory of Traditional Chinese Clinical Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Fang Lu
- Department of Radiology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Meng Wang
- Department of Nephrology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Key Laboratory of Liver and Kidney Diseases, Ministry of Education, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- TCM Institute of Kidney Disease, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Shanghai Key Laboratory of Traditional Chinese Clinical Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Lingchen Wang
- Department of Nephrology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Key Laboratory of Liver and Kidney Diseases, Ministry of Education, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- TCM Institute of Kidney Disease, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Shanghai Key Laboratory of Traditional Chinese Clinical Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Chaoyang Ye
- Department of Nephrology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Key Laboratory of Liver and Kidney Diseases, Ministry of Education, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- TCM Institute of Kidney Disease, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Shanghai Key Laboratory of Traditional Chinese Clinical Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Shuohui Yang
- Department of Radiology, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Chen Wang
- Department of Nephrology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Key Laboratory of Liver and Kidney Diseases, Ministry of Education, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- TCM Institute of Kidney Disease, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Shanghai Key Laboratory of Traditional Chinese Clinical Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
3
|
Rasmussen SB, Boyko Y, Ranucci M, de Somer F, Ravn HB. Cardiac surgery-Associated acute kidney injury - A narrative review. Perfusion 2024; 39:1516-1530. [PMID: 37905794 DOI: 10.1177/02676591231211503] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2023]
Abstract
Cardiac Surgery-Associated Acute Kidney Injury (CSA-AKI) is a serious complication seen in approximately 20-30% of cardiac surgery patients. The underlying pathophysiology is complex, often involving both patient- and procedure related risk factors. In contrast to AKI occurring after other types of major surgery, the use of cardiopulmonary bypass comprises both additional advantages and challenges, including non-pulsatile flow, targeted blood flow and pressure as well as the ability to manipulate central venous pressure (congestion). With an increasing focus on the impact of CSA-AKI on both short and long-term mortality, early identification and management of high-risk patients for CSA-AKI has evolved. The present narrative review gives an up-to-date summary on definition, diagnosis, underlying pathophysiology, monitoring and implications of CSA-AKI, including potential preventive interventions. The review will provide the reader with an in-depth understanding of how to identify, support and provide a more personalized and tailored perioperative management to avoid development of CSA-AKI.
Collapse
Affiliation(s)
- Sebastian Buhl Rasmussen
- Department of Anaesthesiology and Intensive Care, Odense University Hospital, Odense, Denmark
- Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Yuliya Boyko
- Department of Anaesthesiology and Intensive Care, Odense University Hospital, Odense, Denmark
| | - Marco Ranucci
- Department of Cardiovascular Anaesthesiology and Intensive Care, IRCCS Policlinico San Donato, Milan, Italy
| | | | - Hanne Berg Ravn
- Department of Anaesthesiology and Intensive Care, Odense University Hospital, Odense, Denmark
- Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| |
Collapse
|
4
|
Gazzard SE, Cullen-McEwen LA, Nikulina M, Clever AB, Gardner BS, Smith DW, Lee CJ, Nyengaard JR, Evans RG, Bertram JF. Alterations to peritubular capillary structure in a rat model of kidney interstitial fibrosis: Implications for oxygen diffusion. Anat Rec (Hoboken) 2024. [PMID: 39238265 DOI: 10.1002/ar.25576] [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: 07/03/2024] [Revised: 08/19/2024] [Accepted: 08/28/2024] [Indexed: 09/07/2024]
Abstract
Fibrosis and loss of functional capillary surface area may contribute to renal tissue hypoxia in a range of kidney diseases. However, there is limited quantitative information on the impact of kidney disease on the barriers to oxygen diffusion from cortical peritubular capillaries (PTCs) to kidney epithelial tubules. Here, we used stereological methods to quantify changes in total cortical PTC length and surface area, PTC length and surface densities, and diffusion distances between PTCs and kidney tubules in adenine-induced kidney injury. After 7 days of oral gavage of adenine (100 mg), plasma creatinine was 3.5-fold greater than in vehicle-treated rats, while total kidney weight was 83% greater. The total length of PTCs was similar in adenine-treated (1.47 ± 0.23 km (mean ± standard deviation)) to vehicle-treated (1.24 ± 0.24 km) rats, as was the surface density of PTCs (0.025 ± 0.002 vs. 0.024 ± 0.004 μm2/μm3). The total surface area of PTCs was 69% greater in adenine-treated than vehicle-treated rats. However, the length density of PTCs was 28% less in adenine-treated than vehicle-treated rats. Diffusion distances, from PTCs to the basal membrane of the nearest renal tubule (108%), and to the mid-point of the cytoplasmic height of the nearest tubular epithelial cell (57%), were markedly increased. These findings indicate that, in adenine-induced kidney injury, expansion of the renal cortical interstitium increases the distance required for diffusion of oxygen from PTCs to tubules, rendering the kidney cortex susceptible to hypoxia.
Collapse
Affiliation(s)
- Sarah E Gazzard
- Department of Anatomy and Developmental Biology, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
| | - Luise A Cullen-McEwen
- Department of Anatomy and Developmental Biology, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
| | - Marina Nikulina
- Department of Physiology, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
| | - Arnold B Clever
- Department of Physiology, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
| | - Bruce S Gardner
- School of Mathematics, Statistics, Chemistry and Physics, Murdoch University, Perth, Western Australia, Australia
| | - David W Smith
- Faculty of Engineering and Mathematical Sciences, The University of Western Australia, Perth, Western Australia, Australia
| | - Chang-Joon Lee
- Auckland Bioengineering Institute, The University of Auckland, Auckland, New Zealand
| | - Jens R Nyengaard
- Core Center for Molecular Morphology, Section for Stereology and Microscopy, Department of Clinical Medicine, Aarhus University; and Department of Pathology, Aarhus University Hospital, Aarhus, Denmark
| | - Roger G Evans
- Department of Physiology, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
- Pre-Clinical Critical Care Unit, Florey Institute of Neuroscience and Mental Health, University of Melbourne, Melbourne, Victoria, Australia
| | - John F Bertram
- Department of Anatomy and Developmental Biology, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
| |
Collapse
|
5
|
Faivre A, de Seigneux S. The role of hypoxia in chronic kidney disease: a nuanced perspective. Curr Opin Nephrol Hypertens 2024; 33:414-419. [PMID: 38597413 PMCID: PMC11139247 DOI: 10.1097/mnh.0000000000000989] [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] [Indexed: 04/11/2024]
Abstract
PURPOSE OF REVIEW This review critically examines the role of hypoxia in chronic kidney disease (CKD). While traditionally viewed as detrimental, recent insights suggest a more nuanced understanding of hypoxia's role during renal disease. RECENT FINDINGS Emerging evidence challenges the traditional view that hypoxia is universally harmful in CKD context. We review here the recent evidence about hypoxia and HIF activation in CKD. We also discuss the effect of hypoxia on the renal tissue, and the relative inhibition of different HIF isoforms. Recent advancements in therapies, such as HIF prolyl hydroxylase inhibitors (HIF-PHIs) and sodium-glucose cotransporter 2 (SGLT2) inhibitors seem to target the HIF pathway. These drugs impact anemia associated with CKDbut also renoprotection, hinting at a more complex interplay between hypoxia, HIF activation, and renal health. SUMMARY A certain level of hypoxia and specific HIF pathway activation, especially HIF-α, can be beneficial in CKD progression. Therapeutic strategies targeting HIF stabilization, such as with HIF-PHIs and SGLT2 inhibitors, offer promising avenues for enhancing renal protection. Future investigations should aim at better understanding the precise effects on HIF pathway and optimize their clinical application to improve outcomes for CKD patients.
Collapse
Affiliation(s)
- Anna Faivre
- Service de néphrologie, Département des Spécialités de Médecine Interne, Hôpitaux Universitaires de Genève
- Département de Physiologie Cellulaire et Métabolisme, Université de Genève, Genève, Suisse
| | - Sophie de Seigneux
- Service de néphrologie, Département des Spécialités de Médecine Interne, Hôpitaux Universitaires de Genève
- Département de Physiologie Cellulaire et Métabolisme, Université de Genève, Genève, Suisse
| |
Collapse
|
6
|
Ogurlu B, Hamelink TL, Van Tricht IM, Leuvenink HGD, De Borst MH, Moers C, Pool MBF. Utilizing pathophysiological concepts of ischemia-reperfusion injury to design renoprotective strategies and therapeutic interventions for normothermic ex vivo kidney perfusion. Am J Transplant 2024; 24:1110-1126. [PMID: 38184242 DOI: 10.1016/j.ajt.2024.01.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Revised: 12/13/2023] [Accepted: 01/01/2024] [Indexed: 01/08/2024]
Abstract
Normothermic machine perfusion (NMP) has emerged as a promising tool for the preservation, viability assessment, and repair of deceased-donor kidneys prior to transplantation. These kidneys inevitably experience a period of ischemia during donation, which leads to ischemia-reperfusion injury when NMP is subsequently commenced. Ischemia-reperfusion injury has a major impact on the renal vasculature, metabolism, oxygenation, electrolyte balance, and acid-base homeostasis. With an increased understanding of the underlying pathophysiological mechanisms, renoprotective strategies and therapeutic interventions can be devised to minimize additional injury during normothermic reperfusion, ensure the safe implementation of NMP, and improve kidney quality. This review discusses the pathophysiological alterations in the vasculature, metabolism, oxygenation, electrolyte balance, and acid-base homeostasis of deceased-donor kidneys and delineates renoprotective strategies and therapeutic interventions to mitigate renal injury and improve kidney quality during NMP.
Collapse
Affiliation(s)
- Baran Ogurlu
- Department of Surgery - Organ Donation and Transplantation, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands.
| | - Tim L Hamelink
- Department of Surgery - Organ Donation and Transplantation, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Isa M Van Tricht
- Department of Surgery - Organ Donation and Transplantation, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Henri G D Leuvenink
- Department of Surgery - Organ Donation and Transplantation, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Martin H De Borst
- Department of Internal Medicine, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Cyril Moers
- Department of Surgery - Organ Donation and Transplantation, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Merel B F Pool
- Department of Surgery - Organ Donation and Transplantation, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| |
Collapse
|
7
|
Zhao K, Seeliger E, Niendorf T, Liu Z. Noninvasive Assessment of Diabetic Kidney Disease With MRI: Hype or Hope? J Magn Reson Imaging 2024; 59:1494-1513. [PMID: 37675919 DOI: 10.1002/jmri.29000] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Revised: 08/22/2023] [Accepted: 08/23/2023] [Indexed: 09/08/2023] Open
Abstract
Owing to the increasing prevalence of diabetic mellitus, diabetic kidney disease (DKD) is presently the leading cause of chronic kidney disease and end-stage renal disease worldwide. Early identification and disease interception is of paramount clinical importance for DKD management. However, current diagnostic, disease monitoring and prognostic tools are not satisfactory, due to their low sensitivity, low specificity, or invasiveness. Magnetic resonance imaging (MRI) is noninvasive and offers a host of contrast mechanisms that are sensitive to pathophysiological changes and risk factors associated with DKD. MRI tissue characterization involves structural and functional information including renal morphology (kidney volume (TKV) and parenchyma thickness using T1- or T2-weighted MRI), renal microstructure (diffusion weighted imaging, DWI), renal tissue oxygenation (blood oxygenation level dependent MRI, BOLD), renal hemodynamics (arterial spin labeling and phase contrast MRI), fibrosis (DWI) and abdominal or perirenal fat fraction (Dixon MRI). Recent (pre)clinical studies demonstrated the feasibility and potential value of DKD evaluation with MRI. Recognizing this opportunity, this review outlines key concepts and current trends in renal MRI technology for furthering our understanding of the mechanisms underlying DKD and for supplementing clinical decision-making in DKD. Progress in preclinical MRI of DKD is surveyed, and challenges for clinical translation of renal MRI are discussed. Future directions of DKD assessment and renal tissue characterization with (multi)parametric MRI are explored. Opportunities for discovery and clinical break-through are discussed including biological validation of the MRI findings, large-scale population studies, standardization of DKD protocols, the synergistic connection with data science to advance comprehensive texture analysis, and the development of smart and automatic data analysis and data visualization tools to further the concepts of virtual biopsy and personalized DKD precision medicine. We hope that this review will convey this vision and inspire the reader to become pioneers in noninvasive assessment and management of DKD with MRI. LEVEL OF EVIDENCE: 1 TECHNICAL EFFICACY: Stage 2.
Collapse
Affiliation(s)
- Kaixuan Zhao
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
- Department of Radiology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Artificial Intelligence in Medical Image Analysis and Application, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Erdmann Seeliger
- Institute of Translational Physiology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Thoralf Niendorf
- Berlin Ultrahigh Field Facility (B.U.F.F.), Max Delbrueck Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Zaiyi Liu
- Department of Radiology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Artificial Intelligence in Medical Image Analysis and Application, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| |
Collapse
|
8
|
Higashihara E, Harada T, Fukuhara H. Juxtaglomerular apparatus-mediated homeostatic mechanisms: therapeutic implication for chronic kidney disease. Expert Opin Pharmacother 2024; 25:819-832. [PMID: 38773961 DOI: 10.1080/14656566.2024.2357188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Accepted: 05/15/2024] [Indexed: 05/24/2024]
Abstract
INTRODUCTION Juxtaglomerular apparatus (JGA)-mediated homeostatic mechanism links to how sodium-glucose cotransporter 2 inhibitors (SGLT2is) slow progression of chronic kidney disease (CKD) and may link to how tolvaptan slows renal function decline in autosomal dominant polycystic kidney disease (ADPKD). AREA COVERED JGA-mediated homeostatic mechanism has been hypothesized based on investigations of tubuloglomerular feedback and renin-angiotensin system. We reviewed clinical trials of SGLT2is and tolvaptan to assess the relationship between this mechanism and these drugs. EXPERT OPINION When sodium load to macula densa (MD) increases, MD increases adenosine production, constricting afferent arteriole (Af-art) and protecting glomeruli. Concurrently, MD signaling suppresses renin secretion, increases urinary sodium excretion, and counterbalances reduced sodium filtration. However, when there is marked increase in sodium load per-nephron, as in advanced CKD, MD adenosine production increases, relaxing Af-art and maintaining sodium homeostasis at the expense of glomeruli. The beneficial effects of tolvaptan on renal function in ADPKD may also depend on the JGA-mediated homeostatic mechanisms since tolvaptan inhibits sodium reabsorption in the thick ascending limb.The JGA-mediated homeostatic mechanism regulates Af-arts, constricting to relaxing according to homeostatic needs. Understanding this mechanism may contribute to the development of pharmacotherapeutic compounds and better care for patients with CKD.
Collapse
Affiliation(s)
- Eiji Higashihara
- Department of Urology, Kyorin University School of Medicine, Mitaka, Japan
| | - Takeo Harada
- Department of Renal and Cardiovascular Research, Otsuka Pharmaceutical Co. Ltd, Tokushima, Japan
| | - Hiroshi Fukuhara
- Department of Urology, Kyorin University School of Medicine, Mitaka, Japan
| |
Collapse
|
9
|
Abstract
Almost a hundred years have passed since obstruction of the renal artery has been recognized to raise blood pressure. By now chronic renovascular disease (RVD) due to renal artery stenosis is recognized as a major source of renovascular hypertension and renal disease. In some patients, RVD unaccompanied by noteworthy renal dysfunction or blood pressure elevation may be incidentally identified during peripheral angiography. Nevertheless, in others, RVD might present as a progressive disease associated with diffuse atherosclerosis, leading to loss of renal function, renovascular hypertension, hemodynamic compromise, and a magnified risk for cardiovascular morbidity and mortality. Atherosclerotic RVD leads to renal atrophy, inflammation, and hypoxia but represents a potentially treatable cause of chronic renal failure because until severe fibrosis sets in the ischemic kidney, it retains a robust potential for vascular and tubular regeneration. This remarkable recovery capacity of the kidney begs for early diagnosis and treatment. However, accumulating evidence from both animal studies and randomized clinical trials has convincingly established the inadequate efficacy of renal artery revascularization to fully restore renal function or blood pressure control and has illuminated the potential of therapies targeted to the ischemic renal parenchyma to instigate renal regeneration. Some of the injurious mechanisms identified as potential therapeutic targets included oxidative stress, microvascular disease, inflammation, mitochondrial injury, and cellular senescence. This review recapitulates the intrinsic mechanisms that orchestrate renal damage and recovery in RVD and can be harnessed to introduce remedial opportunities.
Collapse
Affiliation(s)
- Alfonso Eirin
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN, USA
| | - Alejandro R. Chade
- Department of Medical Pharmacology and Physiology, University of Missouri-Columbia, MO
| | - Lilach O. Lerman
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
10
|
Robinson S, Nag A, Peticca B, Prudencio T, Di Carlo A, Karhadkar S. Renal Cell Carcinoma in End-Stage Kidney Disease and the Role of Transplantation. Cancers (Basel) 2023; 16:3. [PMID: 38201432 PMCID: PMC10777936 DOI: 10.3390/cancers16010003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Revised: 12/12/2023] [Accepted: 12/13/2023] [Indexed: 01/12/2024] Open
Abstract
Kidney transplant patients have a higher risk of renal cell carcinoma (RCC) compared to non-transplanted end-stage kidney disease (ESKD) patients. This increased risk has largely been associated with the use of immunosuppression; however, recent genetic research highlights the significance of tissue specificity in cancer driver genes. The implication of tissue specificity becomes more obscure when addressing transplant patients, as two distinct metabolic environments are present within one individual. The oncogenic potential of donor renal tissue is largely unknown but assumed to pose minimal risk to the kidney transplant recipient (KTR). Our review challenges this notion by examining how donor and recipient microenvironments impact a transplant recipient's associated risk of renal cell carcinoma. In doing so, we attempt to encapsulate how ESKD-RCC and KTR-RCC differ in their incidence, pathogenesis, outcome, and approach to management.
Collapse
Affiliation(s)
- Samuel Robinson
- Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA; (S.R.); (B.P.); (T.P.); (A.D.C.)
- Department of Surgery, Temple University Hospital, Philadelphia, PA 19140, USA;
| | - Alena Nag
- Department of Surgery, Temple University Hospital, Philadelphia, PA 19140, USA;
| | - Benjamin Peticca
- Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA; (S.R.); (B.P.); (T.P.); (A.D.C.)
- Department of Surgery, Temple University Hospital, Philadelphia, PA 19140, USA;
| | - Tomas Prudencio
- Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA; (S.R.); (B.P.); (T.P.); (A.D.C.)
- Department of Surgery, Temple University Hospital, Philadelphia, PA 19140, USA;
| | - Antonio Di Carlo
- Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA; (S.R.); (B.P.); (T.P.); (A.D.C.)
- Department of Surgery, Temple University Hospital, Philadelphia, PA 19140, USA;
| | - Sunil Karhadkar
- Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA; (S.R.); (B.P.); (T.P.); (A.D.C.)
- Department of Surgery, Temple University Hospital, Philadelphia, PA 19140, USA;
| |
Collapse
|
11
|
André C, Bodeau S, Kamel S, Bennis Y, Caillard P. The AKI-to-CKD Transition: The Role of Uremic Toxins. Int J Mol Sci 2023; 24:16152. [PMID: 38003343 PMCID: PMC10671582 DOI: 10.3390/ijms242216152] [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: 09/14/2023] [Revised: 10/31/2023] [Accepted: 11/06/2023] [Indexed: 11/26/2023] Open
Abstract
After acute kidney injury (AKI), renal function continues to deteriorate in some patients. In a pro-inflammatory and profibrotic environment, the proximal tubules are subject to maladaptive repair. In the AKI-to-CKD transition, impaired recovery from AKI reduces tubular and glomerular filtration and leads to chronic kidney disease (CKD). Reduced kidney secretion capacity is characterized by the plasma accumulation of biologically active molecules, referred to as uremic toxins (UTs). These toxins have a role in the development of neurological, cardiovascular, bone, and renal complications of CKD. However, UTs might also cause CKD as well as be the consequence. Recent studies have shown that these molecules accumulate early in AKI and contribute to the establishment of this pro-inflammatory and profibrotic environment in the kidney. The objective of the present work was to review the mechanisms of UT toxicity that potentially contribute to the AKI-to-CKD transition in each renal compartment.
Collapse
Affiliation(s)
- Camille André
- Department of Clinical Pharmacology, Amiens Medical Center, 80000 Amiens, France; (S.B.); (Y.B.)
- GRAP Laboratory, INSERM UMR 1247, University of Picardy Jules Verne, 80000 Amiens, France
| | - Sandra Bodeau
- Department of Clinical Pharmacology, Amiens Medical Center, 80000 Amiens, France; (S.B.); (Y.B.)
- MP3CV Laboratory, UR UPJV 7517, University of Picardy Jules Verne, 80000 Amiens, France; (S.K.); (P.C.)
| | - Saïd Kamel
- MP3CV Laboratory, UR UPJV 7517, University of Picardy Jules Verne, 80000 Amiens, France; (S.K.); (P.C.)
- Department of Clinical Biochemistry, Amiens Medical Center, 80000 Amiens, France
| | - Youssef Bennis
- Department of Clinical Pharmacology, Amiens Medical Center, 80000 Amiens, France; (S.B.); (Y.B.)
- MP3CV Laboratory, UR UPJV 7517, University of Picardy Jules Verne, 80000 Amiens, France; (S.K.); (P.C.)
| | - Pauline Caillard
- MP3CV Laboratory, UR UPJV 7517, University of Picardy Jules Verne, 80000 Amiens, France; (S.K.); (P.C.)
- Department of Nephrology, Dialysis and Transplantation, Amiens Medical Center, 80000 Amiens, France
| |
Collapse
|
12
|
Kurahara N, Yutsudo A, Furusawa Y, Yamato O, Miyoshi N, Hifumi T, Yabuki A. Immunohistochemical analysis of renal oxidative damage in senior and geriatric cats with chronic kidney disease. J Comp Pathol 2023; 207:14-17. [PMID: 37871535 DOI: 10.1016/j.jcpa.2023.09.001] [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: 04/03/2023] [Revised: 06/03/2023] [Accepted: 09/20/2023] [Indexed: 10/25/2023]
Abstract
Oxidative stress is a well-known cause of chronic kidney disease (CKD). In this study, renal oxidative damage in azotaemic and non-azotaemic aged cats with naturally occurring CKD was investigated using immunohistochemistry for 8-hydroxy-2'-deoxyguanosine (8-OHdG) and 4-hydroxynonenal (4-HNE) as markers of oxidative tissue damage. Kidneys were obtained from aged (>10 years old) azotaemic (n = 13) and non-azotaemic (n = 7) cats. Immunoreactivity for 8-OHdG was found in the nuclei of glomeruli, proximal and distal tubules, loops of Henle and collecting ducts, whereas 4-HNE-positive signals were detected in the cytoplasm of distal nephrons in azotaemic and non-azotaemic cats. Quantitative analysis did not identify any significant differences between the azotaemic and non-azotaemic groups for any of the parameters examined. These results indicate that renal oxidative damage occurs in the kidneys of aged cats with CKD, regardless of whether they are azotaemic or non-azotaemic, emphasizing the importance of oxidative stress during early-stage CKD in senior and geriatric cats.
Collapse
Affiliation(s)
- Natsume Kurahara
- Laboratory of Veterinary Clinical Pathology, Joint Faculty of Veterinary Medicine, Kagoshima University, 1-21-24 Korimoto, Kagoshima 890-0065, Japan
| | - Ayami Yutsudo
- Laboratory of Veterinary Clinical Pathology, Joint Faculty of Veterinary Medicine, Kagoshima University, 1-21-24 Korimoto, Kagoshima 890-0065, Japan
| | - Yu Furusawa
- Kagoshima University Veterinary Teaching Hospital, Joint Faculty of Veterinary Medicine, Kagoshima University, 1-21-24 Korimoto, Kagoshima 890-0065, Japan
| | - Osamu Yamato
- Laboratory of Veterinary Clinical Pathology, Joint Faculty of Veterinary Medicine, Kagoshima University, 1-21-24 Korimoto, Kagoshima 890-0065, Japan
| | - Noriaki Miyoshi
- Laboratory of Veterinary Histopathology, Joint Faculty of Veterinary Medicine, Kagoshima University, 1-21-24 Korimoto, Kagoshima 890-0065, Japan
| | - Tatsuro Hifumi
- Laboratory of Veterinary Histopathology, Joint Faculty of Veterinary Medicine, Kagoshima University, 1-21-24 Korimoto, Kagoshima 890-0065, Japan
| | - Akira Yabuki
- Laboratory of Veterinary Clinical Pathology, Joint Faculty of Veterinary Medicine, Kagoshima University, 1-21-24 Korimoto, Kagoshima 890-0065, Japan; Kagoshima University Veterinary Teaching Hospital, Joint Faculty of Veterinary Medicine, Kagoshima University, 1-21-24 Korimoto, Kagoshima 890-0065, Japan.
| |
Collapse
|
13
|
Chen H, Duan Q, Zhu H, Wan S, Zhao X, Ye D, Fang X. Short-term association of CO and NO 2 with hospital visits for glomerulonephritis in Hefei, China: a time series study. Front Public Health 2023; 11:1239378. [PMID: 37670834 PMCID: PMC10475946 DOI: 10.3389/fpubh.2023.1239378] [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: 06/13/2023] [Accepted: 07/24/2023] [Indexed: 09/07/2023] Open
Abstract
Objective Recent studies suggest air pollution as an underlying factor to kidney disease. However, there is still limited knowledge about the short-term correlation between glomerulonephritis (GN) and air pollution. Thus, we aim to fill this research gap by investigating the short-term correlation between GN clinical visits and air pollution exposure. Methods Between 2015 and 2019, daily GN visit data from two grade A tertiary hospitals in Hefei City were collected, along with corresponding air pollution and meteorological data. A generalized linear model integrated with a distributed lag nonlinear model was employed to analyze the relationship between GN visits and air pollutants. Moreover, we incorporated a dual pollutant model to account for the combined effects of multiple pollutants. Furthermore, subgroup analyses were performed to identify vulnerable populations based on gender, age, and season. Results The association between 23,475 GN visits and air pollutants was assessed, and significant positive associations were found between CO and NO2 exposure and GN visit risk. The single-day lagged effect model for CO showed increased risks for GN visits from lag0 (RR: 1.129, 95% CI: 1.031-1.236) to lag2 (RR: 1.034, 95% CI: 1.011-1.022), with the highest risk at lag0. In contrast, NO2 displayed a more persistent impact (lag1-lag4) on GN visit risk, peaking at lag2 (RR: 1.017, 95% CI: 1.011-1.022). Within the dual-pollutant model, the significance persisted for both CO and NO2 after adjusting for each other. Subgroup analyses showed that the cumulative harm of CO was greater in the cold-season and older adult groups. Meanwhile, the female group was more vulnerable to the harmful effects of cumulative exposure to NO2. Conclusion Our study indicated that CO and NO2 exposure can raise the risk of GN visits, and female and older adult populations exhibited greater susceptibility.
Collapse
Affiliation(s)
- Haifeng Chen
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, Anhui, China
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei, Anhui, China
| | - Qiong Duan
- Department of Health Management Center, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Huahui Zhu
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, Anhui, China
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei, Anhui, China
| | - Shuai Wan
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, Anhui, China
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei, Anhui, China
| | - Xinyi Zhao
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, Anhui, China
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei, Anhui, China
| | - Dongqing Ye
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, Anhui, China
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei, Anhui, China
| | - Xinyu Fang
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, Anhui, China
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei, Anhui, China
| |
Collapse
|
14
|
Päivärinta J, Anastasiou IA, Koivuviita N, Sharma K, Nuutila P, Ferrannini E, Solini A, Rebelos E. Renal Perfusion, Oxygenation and Metabolism: The Role of Imaging. J Clin Med 2023; 12:5141. [PMID: 37568543 PMCID: PMC10420088 DOI: 10.3390/jcm12155141] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2023] [Revised: 08/02/2023] [Accepted: 08/03/2023] [Indexed: 08/13/2023] Open
Abstract
Thanks to technical advances in the field of medical imaging, it is now possible to study key features of renal anatomy and physiology, but so far poorly explored due to the inherent difficulties in studying both the metabolism and vasculature of the human kidney. In this narrative review, we provide an overview of recent research findings on renal perfusion, oxygenation, and substrate uptake. Most studies evaluating renal perfusion with positron emission tomography (PET) have been performed in healthy controls, and specific target populations like obese individuals or patients with renovascular disease and chronic kidney disease (CKD) have rarely been assessed. Functional magnetic resonance (fMRI) has also been used to study renal perfusion in CKD patients, and recent studies have addressed the kidney hemodynamic effects of therapeutic agents such as glucagon-like receptor agonists (GLP-1RA) and sodium-glucose co-transporter 2 inhibitors (SGLT2-i) in an attempt to characterise the mechanisms leading to their nephroprotective effects. The few available studies on renal substrate uptake are discussed. In the near future, these imaging modalities will hopefully become widely available with researchers more acquainted with them, gaining insights into the complex renal pathophysiology in acute and chronic diseases.
Collapse
Affiliation(s)
- Johanna Päivärinta
- Department of Medicine, Division of Nephrology, Turku University Hospital, 20521 Turku, Finland; (J.P.); (N.K.)
| | - Ioanna A. Anastasiou
- 1st Department of Propaedeutic and Internal Medicine, Medical School, National and Kapodistrian University of Athens, Laiko General Hospital, 11527 Athens, Greece;
| | - Niina Koivuviita
- Department of Medicine, Division of Nephrology, Turku University Hospital, 20521 Turku, Finland; (J.P.); (N.K.)
| | - Kanishka Sharma
- Department of Imaging, Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield S10 2TN, UK;
| | - Pirjo Nuutila
- Turku PET Centre, 20521 Turku, Finland;
- Department of Endocrinology, Turku University Hospital, 20521 Turku, Finland
| | - Ele Ferrannini
- CNR, Institute of Clinical Physiology, 56124 Pisa, Italy;
| | - Anna Solini
- Department of Surgical, Medical, Molecular and Critical Area Pathology, University of Pisa, 56124 Pisa, Italy;
| | - Eleni Rebelos
- Turku PET Centre, 20521 Turku, Finland;
- Department of Clinical and Experimental Medicine, University of Pisa, 56124 Pisa, Italy
| |
Collapse
|
15
|
Russo E, Bussalino E, Macciò L, Verzola D, Saio M, Esposito P, Leoncini G, Pontremoli R, Viazzi F. Non-Haemodynamic Mechanisms Underlying Hypertension-Associated Damage in Target Kidney Components. Int J Mol Sci 2023; 24:ijms24119422. [PMID: 37298378 DOI: 10.3390/ijms24119422] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2023] [Revised: 05/23/2023] [Accepted: 05/27/2023] [Indexed: 06/12/2023] Open
Abstract
Arterial hypertension (AH) is a global challenge that greatly impacts cardiovascular morbidity and mortality worldwide. AH is a major risk factor for the development and progression of kidney disease. Several antihypertensive treatment options are already available to counteract the progression of kidney disease. Despite the implementation of the clinical use of renin-angiotensin aldosterone system (RAAS) inhibitors, gliflozins, endothelin receptor antagonists, and their combination, the kidney damage associated with AH is far from being resolved. Fortunately, recent studies on the molecular mechanisms of AH-induced kidney damage have identified novel potential therapeutic targets. Several pathophysiologic pathways have been shown to play a key role in AH-induced kidney damage, including inappropriate tissue activation of the RAAS and immunity system, leading to oxidative stress and inflammation. Moreover, the intracellular effects of increased uric acid and cell phenotype transition showed their link with changes in kidney structure in the early phase of AH. Emerging therapies targeting novel disease mechanisms could provide powerful approaches for hypertensive nephropathy management in the future. In this review, we would like to focus on the interactions of pathways linking the molecular consequences of AH to kidney damage, suggesting how old and new therapies could aim to protect the kidney.
Collapse
Affiliation(s)
- Elisa Russo
- U.O.C. Nefrologia e Dialisi, Ospedale San Luca, 55100 Lucca, Italy
| | - Elisabetta Bussalino
- IRCCS Ospedale Policlinico San Martino, 16132 Genova, Italy
- Department of Internal Medicine, University of Genova, 16132 Genova, Italy
| | - Lucia Macciò
- IRCCS Ospedale Policlinico San Martino, 16132 Genova, Italy
| | | | - Michela Saio
- S.S.D. Nefrologia e Dialisi, Ospedale di Sestri Levante, 16124 Genova, Italy
| | - Pasquale Esposito
- IRCCS Ospedale Policlinico San Martino, 16132 Genova, Italy
- Department of Internal Medicine, University of Genova, 16132 Genova, Italy
| | - Giovanna Leoncini
- IRCCS Ospedale Policlinico San Martino, 16132 Genova, Italy
- Department of Internal Medicine, University of Genova, 16132 Genova, Italy
| | - Roberto Pontremoli
- IRCCS Ospedale Policlinico San Martino, 16132 Genova, Italy
- Department of Internal Medicine, University of Genova, 16132 Genova, Italy
| | - Francesca Viazzi
- IRCCS Ospedale Policlinico San Martino, 16132 Genova, Italy
- Department of Internal Medicine, University of Genova, 16132 Genova, Italy
| |
Collapse
|
16
|
Yu Y, Wu H, Liu C, Zhang C, Song Y, Ma Y, Li H, Lou J, Liu Y, Cao J, Zhang H, Xu Z, Evans RG, Duan C, Mi W. Intraoperative renal desaturation and postoperative acute kidney injury in older patients undergoing liver resection: A prospective cohort study. J Clin Anesth 2023; 87:111084. [PMID: 36905791 DOI: 10.1016/j.jclinane.2023.111084] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2022] [Revised: 02/15/2023] [Accepted: 02/18/2023] [Indexed: 03/13/2023]
Abstract
STUDY OBJECTIVE To determine the association between intraoperative renal tissue desaturation as measured using near-infrared spectroscopy and increased likelihood of developing postoperative acute kidney injury (AKI) in older patients undergoing hepatectomy. DESIGN A multicenter prospective cohort study. SETTING The study was conducted at two tertiary hospitals in China from September 2020 to October 2021. PATIENTS 157 older patients (≥ 60 years) undergoing open hepatectomy surgery. INTERVENTIONS AND MEASUREMENTS Renal tissue oxygen saturation was continuously monitored during operation using near-infrared spectroscopy. The exposure of interest was intraoperative renal desaturation, defined as at least 20% relative decline in renal tissue oxygen saturation from baseline. The primary outcome was postoperative AKI, defined using the Kidney Disease: Improving Global Outcomes criteria according to the serum creatinine criteria. MAIN RESULTS Renal desaturation occurred in 70 of 157 patients. Postoperative AKI was observed in 23% (16/70) and 8% (7/87) of patients with versus without renal desaturation. Patients with renal desaturation were at higher risk of AKI than patients without renal desaturation (adjusted odds ratio 3.41, 95% confidence interval: 1.12-10.36, p = 0.031). Predictive performance was 65.2% sensitivity and 33.6% specificity for hypotension alone, 69.6% sensitivity and 59.7% specificity for renal desaturation alone, and 95.7% sensitivity and 26.9% specificity for combined use of hypotension and renal desaturation. CONCLUSIONS Intraoperative renal desaturation occurred in >40% in our sample of older patients undergoing liver resection and was associated with increased risk of AKI. Intraoperative near-infrared spectroscopy monitoring enhances the detection of AKI.
Collapse
Affiliation(s)
- Yao Yu
- Department of Anesthesiology, The First Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Haotian Wu
- Department of Anesthesiology, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing, China
| | - Chang Liu
- Department of Anesthesiology, The First Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Changsheng Zhang
- Department of Anesthesiology, The First Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Yuxiang Song
- Department of Anesthesiology, The First Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Yulong Ma
- Department of Anesthesiology, The First Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Hao Li
- Department of Anesthesiology, The First Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Jingsheng Lou
- Department of Anesthesiology, The First Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Yanhong Liu
- Department of Anesthesiology, The First Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Jiangbei Cao
- Department of Anesthesiology, The First Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Huan Zhang
- Department of Anesthesiology, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing, China
| | - Zhipeng Xu
- Department of Anesthesiology, the Affiliated Hospital of Medical School, Ningbo University, Ningbo, China
| | - Roger G Evans
- Cardiovascular Disease Program, Biomedicine Discovery Institute and Department of Physiology, Monash University, Melbourne, Australia; Florey Institute of Neurosciences and Mental Health, University of Melbourne, Melbourne, Australia
| | - Chongyang Duan
- Department of Biostatistics, School of Public Health, Southern Medical University, Guangzhou, China.
| | - Weidong Mi
- Department of Anesthesiology, The First Medical Center of Chinese PLA General Hospital, Beijing, China.
| |
Collapse
|
17
|
Verma N, Despa F. The association between renal accumulation of pancreatic amyloid-forming amylin and renal hypoxia. Front Endocrinol (Lausanne) 2023; 14:1104662. [PMID: 36875454 PMCID: PMC9978768 DOI: 10.3389/fendo.2023.1104662] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Accepted: 02/06/2023] [Indexed: 02/18/2023] Open
Abstract
Chronic kidney disease (CKD) is increasing worldwide and is associated with diabetic states (obesity, prediabetes and type-2 diabetes mellitus). The kidney is intrinsically susceptible to low oxygen (hypoxia) and renal hypoxia plays a vital role in the progression of CKD. Recent studies suggest an association between CKD and renal deposition of amyloid-forming amylin secreted from the pancreas. Renal accumulation of amyloid-forming amylin is associated with hypertension, mitochondrial dysfunction, increased production of reactive oxygen species (ROS) and activation of hypoxia signaling in the kidney. In this review we will discuss potential associations between renal amylin amyloid accumulation, hypertension, and mechanism of hypoxia-induced kidney dysfunction, including activation of hypoxia-inducible factors (HIFs) and mitochondrial dysfunction.
Collapse
Affiliation(s)
- Nirmal Verma
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY, United States
| | | |
Collapse
|
18
|
Deng J, Li L, Feng Y, Yang J. Comprehensive Management of Blood Pressure in Patients with Septic AKI. J Clin Med 2023; 12:jcm12031018. [PMID: 36769666 PMCID: PMC9917880 DOI: 10.3390/jcm12031018] [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: 11/27/2022] [Revised: 01/18/2023] [Accepted: 01/19/2023] [Indexed: 01/31/2023] Open
Abstract
Acute kidney injury (AKI) is one of the serious complications of sepsis in clinical practice, and is an important cause of prolonged hospitalization, death, increased medical costs, and a huge medical burden to society. The pathogenesis of AKI associated with sepsis is relatively complex and includes hemodynamic abnormalities due to inflammatory response, oxidative stress, and shock, which subsequently cause a decrease in renal perfusion pressure and eventually lead to ischemia and hypoxia in renal tissue. Active clinical correction of hypotension can effectively improve renal microcirculatory disorders and promote the recovery of renal function. Furthermore, it has been found that in patients with a previous history of hypertension, small changes in blood pressure may be even more deleterious for kidney function. Therefore, the management of blood pressure in patients with sepsis-related AKI will directly affect the short-term and long-term renal function prognosis. This review summarizes the pathophysiological mechanisms of microcirculatory disorders affecting renal function, fluid management, vasopressor, the clinical blood pressure target, and kidney replacement therapy to provide a reference for the clinical management of sepsis-related AKI, thereby promoting the recovery of renal function for the purpose of improving patient prognosis.
Collapse
Affiliation(s)
- Junhui Deng
- Department of Nephrology, The Third Affiliated Hospital of Chongqing Medical University, Chongqing 400120, China
| | - Lina Li
- Department of Nephrology, The Third Affiliated Hospital of Chongqing Medical University, Chongqing 400120, China
| | - Yuanjun Feng
- Department of Renal Rheumatology, Space Hospital Affiliated to Zunyi Medical University, Zunyi 563002, China
| | - Jurong Yang
- Department of Nephrology, The Third Affiliated Hospital of Chongqing Medical University, Chongqing 400120, China
- Correspondence: or
| |
Collapse
|
19
|
Schutter R, van Varsseveld OC, Lantinga VA, Pool MBF, Hamelink TH, Potze JH, Leuvenink HGD, Laustsen C, Borra RJH, Moers C. Magnetic resonance imaging during warm ex vivo kidney perfusion. Artif Organs 2023; 47:105-116. [PMID: 35996889 PMCID: PMC10086841 DOI: 10.1111/aor.14391] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Revised: 06/30/2022] [Accepted: 08/02/2022] [Indexed: 01/04/2023]
Abstract
BACKGROUND The shortage of donor organs for transplantation remains a worldwide problem. The utilization of suboptimal deceased donors enlarges the pool of potential organs, yet consequently, clinicians face the difficult decision of whether these sub-optimal organs are of sufficient quality for transplantation. Novel technologies could play a pivotal role in making pre-transplant organ assessment more objective and reliable. METHODS Ex vivo normothermic machine perfusion (NMP) at temperatures around 35-37°C allows organ quality assessment in a near-physiological environment. Advanced magnetic resonance imaging (MRI) techniques convey unique information about an organ's structural and functional integrity. The concept of applying magnetic resonance imaging during renal normothermic machine perfusion is novel in both renal and radiological research and we have developed the first MRI-compatible NMP setup for human-sized kidneys. RESULTS We were able to obtain a detailed and real-time view of ongoing processes inside renal grafts during ex vivo perfusion. This new technique can visualize structural abnormalities, quantify regional flow distribution, renal metabolism, and local oxygen availability, and track the distribution of ex vivo administered cellular therapy. CONCLUSION This platform allows for advanced pre-transplant organ assessment, provides a new realistic tool for studies into renal physiology and metabolism, and may facilitate therapeutic tracing of pharmacological and cellular interventions to an isolated kidney.
Collapse
Affiliation(s)
- Rianne Schutter
- Department of Surgery - Organ Donation and Transplantation, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Otis C van Varsseveld
- Department of Surgery - Organ Donation and Transplantation, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Veerle A Lantinga
- Department of Surgery - Organ Donation and Transplantation, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Merel B F Pool
- Department of Surgery - Organ Donation and Transplantation, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Tim H Hamelink
- Department of Surgery - Organ Donation and Transplantation, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Jan Hendrik Potze
- Department of Radiology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Henri G D Leuvenink
- Department of Surgery - Organ Donation and Transplantation, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Christoffer Laustsen
- Department of Clinical Medicine, The MR Research Center, Aarhus University, Aarhus, Denmark
| | - Ronald J H Borra
- Department of Radiology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Cyril Moers
- Department of Surgery - Organ Donation and Transplantation, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| |
Collapse
|
20
|
Hu RT, Lankadeva YR, Yanase F, Osawa EA, Evans RG, Bellomo R. Continuous bladder urinary oxygen tension as a new tool to monitor medullary oxygenation in the critically ill. Crit Care 2022; 26:389. [PMID: 36527088 PMCID: PMC9758873 DOI: 10.1186/s13054-022-04230-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Accepted: 11/09/2022] [Indexed: 12/23/2022] Open
Abstract
Acute kidney injury (AKI) is common in the critically ill. Inadequate renal medullary tissue oxygenation has been linked to its pathogenesis. Moreover, renal medullary tissue hypoxia can be detected before biochemical evidence of AKI in large mammalian models of critical illness. This justifies medullary hypoxia as a pathophysiological biomarker for early detection of impending AKI, thereby providing an opportunity to avert its evolution. Evidence from both animal and human studies supports the view that non-invasively measured bladder urinary oxygen tension (PuO2) can provide a reliable estimate of renal medullary tissue oxygen tension (tPO2), which can only be measured invasively. Furthermore, therapies that modify medullary tPO2 produce corresponding changes in bladder PuO2. Clinical studies have shown that bladder PuO2 correlates with cardiac output, and that it increases in response to elevated cardiopulmonary bypass (CPB) flow and mean arterial pressure. Clinical observational studies in patients undergoing cardiac surgery involving CPB have shown that bladder PuO2 has prognostic value for subsequent AKI. Thus, continuous bladder PuO2 holds promise as a new clinical tool for monitoring the adequacy of renal medullary oxygenation, with its implications for the recognition and prevention of medullary hypoxia and thus AKI.
Collapse
Affiliation(s)
- Raymond T. Hu
- grid.410678.c0000 0000 9374 3516Department of Anaesthesia, Austin Health, Heidelberg, VIC Australia ,grid.1008.90000 0001 2179 088XDepartment of Critical Care, Melbourne Medical School, The University of Melbourne, Parkville, VIC Australia
| | - Yugeesh R. Lankadeva
- grid.1008.90000 0001 2179 088XDepartment of Critical Care, Melbourne Medical School, The University of Melbourne, Parkville, VIC Australia ,grid.1008.90000 0001 2179 088XPre-Clinical Critical Care Unit, Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC Australia
| | - Fumitake Yanase
- grid.414094.c0000 0001 0162 7225Department of Intensive Care, Austin Hospital, Heidelberg, Australia
| | - Eduardo A. Osawa
- Cardiology Intensive Care Unit, DF Star Hospital, Brasília, Brazil ,grid.472984.4D’Or Institute for Research and Education (IDOR), DF Star Hospital, Brasília, Brazil
| | - Roger G. Evans
- grid.1008.90000 0001 2179 088XPre-Clinical Critical Care Unit, Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC Australia ,grid.1002.30000 0004 1936 7857Cardiovascular Disease Program, Biomedicine Discovery Institute and Department of Physiology, Monash University, Clayton, VIC Australia
| | - Rinaldo Bellomo
- grid.1008.90000 0001 2179 088XDepartment of Critical Care, Melbourne Medical School, The University of Melbourne, Parkville, VIC Australia ,grid.414094.c0000 0001 0162 7225Department of Intensive Care, Austin Hospital, Heidelberg, Australia ,grid.1002.30000 0004 1936 7857Australian and New Zealand Intensive Care Research Centre, Monash University, Melbourne, Australia ,grid.416153.40000 0004 0624 1200Department of Intensive Care, Royal Melbourne Hospital, Parkville, Australia
| |
Collapse
|
21
|
Nie Y, Wang L, You X, Wang X, Wu J, Zheng Z. Low dimensional nanomaterials for treating acute kidney injury. J Nanobiotechnology 2022; 20:505. [PMID: 36456976 PMCID: PMC9714216 DOI: 10.1186/s12951-022-01712-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2022] [Accepted: 11/15/2022] [Indexed: 12/02/2022] Open
Abstract
Acute kidney injury (AKI) is one of the most common severe complications among hospitalized patients. In the absence of specific drugs to treat AKI, hemodialysis remains the primary clinical treatment for AKI patients. AKI treatment has received significant attention recently due to the excellent drug delivery capabilities of low-dimensional nanomaterials (LDNs) and their unique therapeutic effects. Diverse LDNs have been proposed to treat AKI, with promising results and the potential for future clinical application. This article aims to provide an overview of the pathogenesis of AKI and the recent advances in the treatment of AKI using different types of LDNs. In addition, it is intended to provide theoretical support for the design of LDNs and implications for AKI treatment.
Collapse
Affiliation(s)
- Yuanpeng Nie
- Department of Nephrology, Center of Kidney and Urology, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, 518107, China
| | - Liying Wang
- Department of Hematology, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, 518107, China
| | - Xinru You
- Department of Pediatrics, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, 518107, China
| | - Xiaohua Wang
- Bioscience and Biomedical Engineering Thrust, The Hong Kong University of Science and Technology (Guangzhou), Nansha, Guangzhou, 511400, China
| | - Jun Wu
- Department of Hematology, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, 518107, China.
- Bioscience and Biomedical Engineering Thrust, The Hong Kong University of Science and Technology (Guangzhou), Nansha, Guangzhou, 511400, China.
- Division of Life Science, The Hong Kong University of Science and Technology, Hong Kong SAR, China.
| | - Zhihua Zheng
- Department of Nephrology, Center of Kidney and Urology, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, 518107, China.
| |
Collapse
|
22
|
Xiang L, Calderon AS, Klemcke HG, Hinojosa-Laborde C, Becerra SC, Ryan KL. A novel animal model to study delayed resuscitation following traumatic hemorrhage. J Appl Physiol (1985) 2022; 133:814-821. [PMID: 36007893 PMCID: PMC9512111 DOI: 10.1152/japplphysiol.00335.2022] [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: 06/13/2022] [Revised: 08/16/2022] [Accepted: 08/16/2022] [Indexed: 11/22/2022] Open
Abstract
A focus of combat casualty care research is to develop treatments for when full resuscitation after hemorrhage is delayed. However, few animal models exist to investigate such treatments. Given the kidney's susceptibility to ischemia, we determined how delayed resuscitation affects renal function in a model of traumatic shock. Rats were randomized into three groups: resuscitation after 1 h (ETH-1) or 2 h (ETH-2) of extremity trauma and hemorrhagic shock, and sham control. ETH was induced in anesthetized rats with muscle injury and fibula fracture, followed by pressure-controlled hemorrhage [mean arterial pressure (MAP) = 55 mmHg] for 1 or 2 h. Rats were then resuscitated with whole blood until MAP stabilized between 90 and 100 mmHg for 30 min. MAP, glomerular filtration rate (GFR), creatinine, blood gases, and fractional excretion of sodium (nFENa+) were measured for 3 days. Compared with control, ETH-1 and ETH-2 exhibited decreases in GFR and nFENa+, and increases in circulating lactate, creatinine, and blood urea nitrogen (BUN) before and within 30 min after resuscitation. The increases in creatinine, BUN, and potassium were greater in ETH-2 than in ETH-1, whereas lactate levels were similar between ETH-1 and ETH-2 before and after resuscitation. All measurements were normalized in ETH-1 within 2 days after resuscitation, with 22% mortality. However, ETH-2 exhibited a prolonged impairment of GFR, increased nFENa+, and a 66% mortality. Resuscitation 1 h after injury therefore preserves renal function, whereas further delay of resuscitation irreversibly impairs renal function and increases mortality. This animal model can be used to explore treatments for prolonged prehospital care following traumatic hemorrhage.NEW & NOTEWORTHY A focus of combat casualty care research is to develop treatment where full resuscitation after hemorrhage is delayed. However, animal models of combat-related hemorrhagic shock in which to determine physiological outcomes of such delays and explore potential treatment for golden hour extension are lacking. In this study, we filled this knowledge gap by establishing a traumatic shock model with reproducible development of AKI and shock-related complications determined by the time of resuscitation.
Collapse
Affiliation(s)
- Lusha Xiang
- U.S. Army Institute of Surgical Research, JBSA-Fort Sam Houston, San Antonio, Texas
| | - Alfredo S Calderon
- U.S. Army Institute of Surgical Research, JBSA-Fort Sam Houston, San Antonio, Texas
| | - Harold G Klemcke
- U.S. Army Institute of Surgical Research, JBSA-Fort Sam Houston, San Antonio, Texas
| | | | - Sandra C Becerra
- U.S. Army Institute of Surgical Research, JBSA-Fort Sam Houston, San Antonio, Texas
| | - Kathy L Ryan
- U.S. Army Institute of Surgical Research, JBSA-Fort Sam Houston, San Antonio, Texas
| |
Collapse
|
23
|
Liu X, Du H, Sun Y, Shao L. Role of abnormal energy metabolism in the progression of chronic kidney disease and drug intervention. Ren Fail 2022; 44:790-805. [PMID: 35535500 PMCID: PMC9103584 DOI: 10.1080/0886022x.2022.2072743] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Chronic kidney disease (CKD) is a severe clinical syndrome with significant socioeconomic impact worldwide. Orderly energy metabolism is essential for normal kidney function and energy metabolism disorders are increasingly recognized as an important player in CKD. Energy metabolism disorders are characterized by ATP deficits and reactive oxygen species increase. Oxygen and mitochondria are essential for ATP production, hypoxia and mitochondrial dysfunction both affect the energy production process. Renin-angiotensin and adenine signaling pathway also play important regulatory roles in energy metabolism. In addition, disturbance of energy metabolism is a key factor in the development of hereditary nephropathy such as autosomal dominant polycystic kidney disease. Currently, drugs with clinically clear renal function protection, such as Angiotensin II Type 1 receptor blockers and fenofibrate, have been proven to improve energy metabolism disorders. The sodium-glucose co-transporter inhibitors 2 that can mediate glucose metabolism disorders not only delay the progress of diabetic nephropathy, but also have significant protective effects in non-diabetic nephropathy. Hypoxia-inducible factor enhances ATP production to the kidney by improving renal oxygen supply and increasing glycolysis, and the mitochondria targeted peptides (SS-31) plays a protective role by stabilizing the mitochondrial inner membrane. Moreover, several drugs are being studied and are predicted to have potential renal protective properties. We propose that the regulation of energy metabolism represents a promising strategy to delay the progression of CKD.
Collapse
Affiliation(s)
- Xuyan Liu
- Department of Nephrology, The Affiliated Qingdao Municipal Hospital of Qingdao University, Qingdao, China
| | - Huasheng Du
- Department of Nephrology, The Affiliated Qingdao Municipal Hospital of Qingdao University, Qingdao, China
| | - Yan Sun
- Department of Nephrology, The Affiliated Qingdao Municipal Hospital of Qingdao University, Qingdao, China
| | - Leping Shao
- Department of Nephrology, The Affiliated Qingdao Municipal Hospital of Qingdao University, Qingdao, China
| |
Collapse
|
24
|
Thévenod F, Schreiber T, Lee WK. Renal hypoxia-HIF-PHD-EPO signaling in transition metal nephrotoxicity: friend or foe? Arch Toxicol 2022; 96:1573-1607. [PMID: 35445830 PMCID: PMC9095554 DOI: 10.1007/s00204-022-03285-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Accepted: 03/14/2022] [Indexed: 12/18/2022]
Abstract
The kidney is the main organ that senses changes in systemic oxygen tension, but it is also the key detoxification, transit and excretion site of transition metals (TMs). Pivotal to oxygen sensing are prolyl-hydroxylases (PHDs), which hydroxylate specific residues in hypoxia-inducible factors (HIFs), key transcription factors that orchestrate responses to hypoxia, such as induction of erythropoietin (EPO). The essential TM ion Fe is a key component and regulator of the hypoxia–PHD–HIF–EPO (HPHE) signaling axis, which governs erythropoiesis, angiogenesis, anaerobic metabolism, adaptation, survival and proliferation, and hence cell and body homeostasis. However, inadequate concentrations of essential TMs or entry of non-essential TMs in organisms cause toxicity and disrupt health. Non-essential TMs are toxic because they enter cells and displace essential TMs by ionic and molecular mimicry, e. g. in metalloproteins. Here, we review the molecular mechanisms of HPHE interactions with TMs (Fe, Co, Ni, Cd, Cr, and Pt) as well as their implications in renal physiology, pathophysiology and toxicology. Some TMs, such as Fe and Co, may activate renal HPHE signaling, which may be beneficial under some circumstances, for example, by mitigating renal injuries from other causes, but may also promote pathologies, such as renal cancer development and metastasis. Yet some other TMs appear to disrupt renal HPHE signaling, contributing to the complex picture of TM (nephro-)toxicity. Strikingly, despite a wealth of literature on the topic, current knowledge lacks a deeper molecular understanding of TM interaction with HPHE signaling, in particular in the kidney. This precludes rationale preventive and therapeutic approaches to TM nephrotoxicity, although recently activators of HPHE signaling have become available for therapy.
Collapse
Affiliation(s)
- Frank Thévenod
- Institute for Physiology, Pathophysiology and Toxicology, ZBAF, Witten/Herdecke University, Stockumer Strasse 12, 58453, Witten, Germany.
| | - Timm Schreiber
- Institute for Physiology, Pathophysiology and Toxicology, ZBAF, Witten/Herdecke University, Stockumer Strasse 12, 58453, Witten, Germany
| | - Wing-Kee Lee
- Physiology and Pathophysiology of Cells and Membranes, Medical School EWL, Bielefeld University, R.1 B2-13, Morgenbreede 1, 33615 Bielefeld, Germany
| |
Collapse
|
25
|
Jägers J, Kirsch M, Cantore M, Karaman O, Ferenz KB. Artificial oxygen carriers in organ preservation: Dose dependency in a rat model of ex-vivo normothermic kidney perfusion. Artif Organs 2022; 46:1783-1793. [PMID: 35435266 DOI: 10.1111/aor.14264] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Revised: 02/16/2022] [Accepted: 04/08/2022] [Indexed: 11/28/2022]
Abstract
INTRODUCTION Organ preservation through ex-vivo normothermic perfusion (EVNP) with albumin-derived perfluorocarbon-based artificial oxygen carriers (A-AOCs) consisting of albumin-derived perfluorodecalin-filled nanocapsules prior to transplantation would be a promising approach to avoid hypoxic tissue injury during organ storage. METHODS The kidneys of 16 rats underwent EVNP for 2 h with plasma-like solution (5% bovine serum albumin, Ringer-Saline, inulin) with or without A-AOCs in different volume fractions (0%, 2%, 4%, or 8%). Cell death was determined using TdT-mediated dUTP-biotin nick end labeling (TUNEL). Aspartate transaminase (AST) activity in both perfusate and urine as well as the glomerular filtration rate (GFR) were determined. The hypoxia inducible factors 1α and 2α (HIF-1α und -2α) were quantified in tissue homogenates. RESULTS GFR was substantially decreased in the presence of 0%, 2%, and 8% A-AOC but not of 4%. In accordance, hypoxia-mediated cell death, as indicated by both AST activity and TUNEL-positive cells, was significantly decreased in the 4% group compared to the control group. The stabilization of HIF-1α and 2α decreased with 4% and 8% but not with 2% A-AOCs. CONCLUSION The dosage of 4% A-AOCs in EVNP was most effective in maintaining the physiological renal function.
Collapse
Affiliation(s)
- Johannes Jägers
- Institute of Physiology, Essen University Hospital, University of Duisburg-Essen, Essen, Germany.,Department of Biomedicine, Aarhus University, Aarhus C, Denmark
| | - Michael Kirsch
- Institute of Physiological Chemistry, Essen University Hospital, University of Duisburg-Essen, Essen, Germany
| | - Miriam Cantore
- Institute of Physiology, Essen University Hospital, University of Duisburg-Essen, Essen, Germany
| | - Ozan Karaman
- Institute of Physiology, Essen University Hospital, University of Duisburg-Essen, Essen, Germany.,Institute of Physiological Chemistry, Essen University Hospital, University of Duisburg-Essen, Essen, Germany
| | - Katja Bettina Ferenz
- Institute of Physiology, Essen University Hospital, University of Duisburg-Essen, Essen, Germany.,CeNIDE (Center for Nanointegration Duisburg-Essen) University of Duisburg-Essen, Duisburg, Germany
| |
Collapse
|
26
|
Lal H, Singh P, Ponmalai K, Prasad R, Singh SP, Yadav P, Singh A, Bhadauria D, Kumar S, Agarwal V, Mishra P. Role of blood oxygen level-dependent magnetic resonance imaging in studying renal oxygenation changes in renal artery stenosis. Abdom Radiol (NY) 2022; 47:1112-1123. [PMID: 35059812 DOI: 10.1007/s00261-022-03408-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 12/31/2021] [Accepted: 01/05/2022] [Indexed: 11/29/2022]
Abstract
AIM Primary objective of this study was to compare R2* value of the post-stenotic kidney with contralateral kidney, kidneys of essential hypertensive patients, and healthy subjects using blood oxygen level-dependent magnetic resonance imaging (BOLD MRI) technique. The secondary objective was to study the effect of severity of stenosis and viability of kidneys on R2* value. METHODS We compared 4 groups of kidneys including 92 with renal artery stenosis, 37 normal contralateral kidneys of unilateral renal artery stenosis patients, 62 kidneys of essential hypertensive patients, and 40 kidneys of healthy controls using BOLD MRI. Deoxyhemoglobin level represented by R2* was calculated before and after giving furosemide and was compared among different groups. RESULTS Baseline means cortical R2* value did not differ between groups. Response to furosemide was reduced in stenotic kidneys as compared to essential hypertensive and healthy control groups (p < 0.001). The mean R2* value of the contralateral normal kidney group was not significantly different from the stenotic group. Baseline R2* value and delta R2* values did not differ between different degrees of stenosis. Higher mean cortical R2* was seen in stenotic kidneys which were small (< 7 cm) in size (24.27 ± 5.65 vs 21.7 ± 3.88; p value 0.02) or with poor corticomedullary differentiation (24.64 ± 5.8 vs 20.74 ± 3.34; p value 0.006) as compared to other stenotic kidneys. Similarly, the delta R2* value was also blunted in these small shrunken kidneys (p value < 0.001). CONCLUSION R2* values on BOLD MRI are significantly different between kidneys with and without renal artery stenosis and can potentially also predict the utility of revascularization.
Collapse
Affiliation(s)
- Hira Lal
- Department of Radiology, Sanjay Gandhi Post Graduate Institute of Medical Sciences, Raebareli Road, Lucknow, U.P., 226014, India.
- Department of Radiology, Sanjay Gandhi Post Graduate Institute of Medical Sciences, Raibareli Road, Lucknow, Uttar Pradesh, 226014, India.
| | - Priya Singh
- Department of Radiology, Sanjay Gandhi Post Graduate Institute of Medical Sciences, Raebareli Road, Lucknow, U.P., 226014, India
| | - Kaushik Ponmalai
- Department of Radiology, Sanjay Gandhi Post Graduate Institute of Medical Sciences, Raebareli Road, Lucknow, U.P., 226014, India
| | - Raghunandan Prasad
- Department of Radiology, Sanjay Gandhi Post Graduate Institute of Medical Sciences, Raebareli Road, Lucknow, U.P., 226014, India
| | - Surya Pratap Singh
- Department of Radiology, Sanjay Gandhi Post Graduate Institute of Medical Sciences, Raebareli Road, Lucknow, U.P., 226014, India
| | - Priyank Yadav
- Department of Urology and Renal Transplantation, Sanjay Gandhi Post Graduate Institute of Medical Sciences, Raebareli Road, Lucknow, U.P., 226014, India
| | - Anuradha Singh
- Department of Radiology, Sanjay Gandhi Post Graduate Institute of Medical Sciences, Raebareli Road, Lucknow, U.P., 226014, India
| | - Dharmendra Bhadauria
- Department of Nephrology, Sanjay Gandhi Post Graduate Institute of Medical Sciences, Raebareli Road, Lucknow, U.P., 226014, India
| | - Sudeep Kumar
- Department of Cardiology, Sanjay Gandhi Post Graduate Institute of Medical Sciences, Raebareli Road, Lucknow, U.P., 226014, India
| | - Vikas Agarwal
- Department of Immunology, Sanjay Gandhi Post Graduate Institute of Medical Sciences, Raebareli Road, Lucknow, U.P., 226014, India
| | - Prabhakar Mishra
- Department of Biostatistics and Health Informatics, Sanjay Gandhi Post Graduate Institute of Medical Sciences, Raibareli Road, Lucknow, U.P., 226014, India
| |
Collapse
|
27
|
Wang B, Li ZL, Zhang YL, Wen Y, Gao YM, Liu BC. Hypoxia and chronic kidney disease. EBioMedicine 2022; 77:103942. [PMID: 35290825 PMCID: PMC8921539 DOI: 10.1016/j.ebiom.2022.103942] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Revised: 02/22/2022] [Accepted: 03/01/2022] [Indexed: 12/12/2022] Open
Abstract
Hypoxia is an inherent pathophysiological characteristic of chronic kidney disease (CKD), which is closely associated with the development of renal inflammation and fibrosis, as well as CKD-related complications such as anaemia, cardiovascular events, and sarcopenia. This review outlined the characteristics of oxygen supply in the kidney, changes in oxygen metabolism and factors leading to hypoxia in CKD. Mechanistically, we discussed how hypoxia contributes to renal injury as well as complications associated with CKD. Furthermore, we also discussed the potential therapeutic approaches that target chronic hypoxia, as well as the challenges in the study of oxygen homeostasis imbalance in CKD.
Collapse
Affiliation(s)
- Bin Wang
- Institute of Nephrology, Zhong Da Hospital, Southeast University School of Medicine, Nanjing, Jiangsu, China
| | - Zuo-Lin Li
- Institute of Nephrology, Zhong Da Hospital, Southeast University School of Medicine, Nanjing, Jiangsu, China
| | - Yi-Lin Zhang
- Institute of Nephrology, Zhong Da Hospital, Southeast University School of Medicine, Nanjing, Jiangsu, China
| | - Yi Wen
- Institute of Nephrology, Zhong Da Hospital, Southeast University School of Medicine, Nanjing, Jiangsu, China
| | - Yue-Ming Gao
- Institute of Nephrology, Zhong Da Hospital, Southeast University School of Medicine, Nanjing, Jiangsu, China
| | - Bi-Cheng Liu
- Institute of Nephrology, Zhong Da Hospital, Southeast University School of Medicine, Nanjing, Jiangsu, China.
| |
Collapse
|
28
|
Correia MJ, Pimpão AB, Fernandes DGF, Morello J, Sequeira CO, Calado J, Antunes AMM, Almeida MS, Branco P, Monteiro EC, Vicente JB, Serpa J, Pereira SA. Cysteine as a Multifaceted Player in Kidney, the Cysteine-Related Thiolome and Its Implications for Precision Medicine. Molecules 2022; 27:1416. [PMID: 35209204 PMCID: PMC8874463 DOI: 10.3390/molecules27041416] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Revised: 02/15/2022] [Accepted: 02/17/2022] [Indexed: 11/16/2022] Open
Abstract
In this review encouraged by original data, we first provided in vivo evidence that the kidney, comparative to the liver or brain, is an organ particularly rich in cysteine. In the kidney, the total availability of cysteine was higher in cortex tissue than in the medulla and distributed in free reduced, free oxidized and protein-bound fractions (in descending order). Next, we provided a comprehensive integrated review on the evidence that supports the reliance on cysteine of the kidney beyond cysteine antioxidant properties, highlighting the relevance of cysteine and its renal metabolism in the control of cysteine excess in the body as a pivotal source of metabolites to kidney biomass and bioenergetics and a promoter of adaptive responses to stressors. This view might translate into novel perspectives on the mechanisms of kidney function and blood pressure regulation and on clinical implications of the cysteine-related thiolome as a tool in precision medicine.
Collapse
Affiliation(s)
- Maria João Correia
- CEDOC, NOVA Medical School, Universidade Nova de Lisboa, 1169-056 Lisboa, Portugal; (M.J.C.); (A.B.P.); (J.M.); (C.O.S.); (M.S.A.); (P.B.); (E.C.M.); (J.S.)
| | - António B. Pimpão
- CEDOC, NOVA Medical School, Universidade Nova de Lisboa, 1169-056 Lisboa, Portugal; (M.J.C.); (A.B.P.); (J.M.); (C.O.S.); (M.S.A.); (P.B.); (E.C.M.); (J.S.)
| | - Dalila G. F. Fernandes
- Instituto de Tecnologia Química e Biológica António Xavier (ITQB NOVA), 2780-157 Oeiras, Portugal; (D.G.F.F.); (J.B.V.)
| | - Judit Morello
- CEDOC, NOVA Medical School, Universidade Nova de Lisboa, 1169-056 Lisboa, Portugal; (M.J.C.); (A.B.P.); (J.M.); (C.O.S.); (M.S.A.); (P.B.); (E.C.M.); (J.S.)
| | - Catarina O. Sequeira
- CEDOC, NOVA Medical School, Universidade Nova de Lisboa, 1169-056 Lisboa, Portugal; (M.J.C.); (A.B.P.); (J.M.); (C.O.S.); (M.S.A.); (P.B.); (E.C.M.); (J.S.)
| | - Joaquim Calado
- Centre for Toxicogenomics and Human Health (ToxOmics), Genetics, Oncology and Human Toxicology, Nova Medical School/Faculdade de Ciências Médicas, Universidade Nova de Lisboa, 1169-056 Lisboa, Portugal;
- Nephrology Department, Centro Hospitalar Universitário de Lisboa Central, 1069-166 Lisboa, Portugal
| | - Alexandra M. M. Antunes
- Centro de Química Estrutural, Institute of Molecular Sciences, Instituto Superior Técnico, 1049-001 Lisboa, Portugal;
| | - Manuel S. Almeida
- CEDOC, NOVA Medical School, Universidade Nova de Lisboa, 1169-056 Lisboa, Portugal; (M.J.C.); (A.B.P.); (J.M.); (C.O.S.); (M.S.A.); (P.B.); (E.C.M.); (J.S.)
- Hospital de Santa Cruz, Centro Hospitalar de Lisboa Ocidental, 2790-134 Carnaxide, Portugal
| | - Patrícia Branco
- CEDOC, NOVA Medical School, Universidade Nova de Lisboa, 1169-056 Lisboa, Portugal; (M.J.C.); (A.B.P.); (J.M.); (C.O.S.); (M.S.A.); (P.B.); (E.C.M.); (J.S.)
- Hospital de Santa Cruz, Centro Hospitalar de Lisboa Ocidental, 2790-134 Carnaxide, Portugal
| | - Emília C. Monteiro
- CEDOC, NOVA Medical School, Universidade Nova de Lisboa, 1169-056 Lisboa, Portugal; (M.J.C.); (A.B.P.); (J.M.); (C.O.S.); (M.S.A.); (P.B.); (E.C.M.); (J.S.)
| | - João B. Vicente
- Instituto de Tecnologia Química e Biológica António Xavier (ITQB NOVA), 2780-157 Oeiras, Portugal; (D.G.F.F.); (J.B.V.)
| | - Jacinta Serpa
- CEDOC, NOVA Medical School, Universidade Nova de Lisboa, 1169-056 Lisboa, Portugal; (M.J.C.); (A.B.P.); (J.M.); (C.O.S.); (M.S.A.); (P.B.); (E.C.M.); (J.S.)
- Instituto Português de Oncologia de Lisboa Francisco Gentil (IPOLFG), 1099-023 Lisboa, Portugal
| | - Sofia A. Pereira
- CEDOC, NOVA Medical School, Universidade Nova de Lisboa, 1169-056 Lisboa, Portugal; (M.J.C.); (A.B.P.); (J.M.); (C.O.S.); (M.S.A.); (P.B.); (E.C.M.); (J.S.)
| |
Collapse
|
29
|
Tomar N, Zhang X, Kandel SM, Sadri S, Yang C, Liang M, Audi SH, Cowley AW, Dash RK. Substrate-dependent differential regulation of mitochondrial bioenergetics in the heart and kidney cortex and outer medulla. BIOCHIMICA ET BIOPHYSICA ACTA. BIOENERGETICS 2022; 1863:148518. [PMID: 34864090 PMCID: PMC8957717 DOI: 10.1016/j.bbabio.2021.148518] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Revised: 10/29/2021] [Accepted: 11/20/2021] [Indexed: 05/05/2023]
Abstract
The kinetics and efficiency of mitochondrial oxidative phosphorylation (OxPhos) can depend on the choice of respiratory substrates. Furthermore, potential differences in this substrate dependency among different tissues are not well-understood. Here, we determined the effects of different substrates on the kinetics and efficiency of OxPhos in isolated mitochondria from the heart and kidney cortex and outer medulla (OM) of Sprague-Dawley rats. The substrates were pyruvate+malate, glutamate+malate, palmitoyl-carnitine+malate, alpha-ketoglutarate+malate, and succinate±rotenone at saturating concentrations. The kinetics of OxPhos were interrogated by measuring mitochondrial bioenergetics under different ADP perturbations. Results show that the kinetics and efficiency of OxPhos are highly dependent on the substrates used, and this dependency is distinctly different between heart and kidney. Heart mitochondria showed higher respiratory rates and OxPhos efficiencies for all substrates in comparison to kidney mitochondria. Cortex mitochondria respiratory rates were higher than OM mitochondria, but OM mitochondria OxPhos efficiencies were higher than cortex mitochondria. State 3 respiration was low in heart mitochondria with succinate but increased significantly in the presence of rotenone, unlike kidney mitochondria. Similar differences were observed in mitochondrial membrane potential. Differences in H2O2 emission in the presence of succinate±rotenone were observed in heart mitochondria and to a lesser extent in OM mitochondria, but not in cortex mitochondria. Bioenergetics and H2O2 emission data with succinate±rotenone indicate that oxaloacetate accumulation and reverse electron transfer may play a more prominent regulatory role in heart mitochondria than kidney mitochondria. These studies provide novel quantitative data demonstrating that the choice of respiratory substrates affects mitochondrial responses in a tissue-specific manner.
Collapse
Affiliation(s)
- Namrata Tomar
- Department of Biomedical Engineering, Medical College of Wisconsin, Milwaukee WI-53226, United States of America
| | - Xiao Zhang
- Department of Biomedical Engineering, Medical College of Wisconsin, Milwaukee WI-53226, United States of America
| | - Sunil M Kandel
- Department of Biomedical Engineering, Medical College of Wisconsin, Milwaukee WI-53226, United States of America
| | - Shima Sadri
- Department of Biomedical Engineering, Medical College of Wisconsin, Milwaukee WI-53226, United States of America
| | - Chun Yang
- Department of Physiology, Medical College of Wisconsin, Milwaukee WI-53226, United States of America
| | - Mingyu Liang
- Department of Physiology, Medical College of Wisconsin, Milwaukee WI-53226, United States of America; Center of Systems Molecular Medicine, Medical College of Wisconsin, Milwaukee WI-53226, United States of America
| | - Said H Audi
- Department of Biomedical Engineering, Marquette University, Milwaukee WI-53223, United States of America
| | - Allen W Cowley
- Department of Physiology, Medical College of Wisconsin, Milwaukee WI-53226, United States of America; Center of Systems Molecular Medicine, Medical College of Wisconsin, Milwaukee WI-53226, United States of America.
| | - Ranjan K Dash
- Department of Biomedical Engineering, Medical College of Wisconsin, Milwaukee WI-53226, United States of America; Department of Physiology, Medical College of Wisconsin, Milwaukee WI-53226, United States of America; Center of Systems Molecular Medicine, Medical College of Wisconsin, Milwaukee WI-53226, United States of America.
| |
Collapse
|
30
|
Miura T, Kuno A, Tanaka M. Diabetes modulation of the myocardial infarction- acute kidney injury axis. Am J Physiol Heart Circ Physiol 2022; 322:H394-H405. [PMID: 35089809 DOI: 10.1152/ajpheart.00639.2021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Since there is crosstalk in functions of the heart and kidney, acute or chronic injury in one of the two organs provokes adaptive and/or maladaptive responses in both organs, leading to cardiorenal syndrome (CRS). Acute kidney injury (AKI) induced by acute heart failure is referred to as type 1 CRS, and a frequent cause of this type of CRS is acute myocardial infarction (AMI). Diabetes mellitus increases the risk of AMI and also the risk of AKI of various causes. However, there have been only a few studies in which animal models of diabetes were used to examine how diabetes modulates AMI-induced AKI. In this review, we summarize findings regarding the mechanisms of type 1 CRS and the impact of diabetes on both AMI and renal susceptibility to AKI and we discuss mechanisms by which diabetes modulates AMI-induced AKI. Hemodynamic alterations induced by AMI could be augmented by diabetes via its detrimental effect on infarct size and contractile function of the non-infarcted region in the heart. Diabetes increases susceptibility of renal cells to hypoxia and oxidative stress by modulation of signaling pathways that regulate cell survival and autophagy. Recent studies have shown that diabetes mellitus even at early stage of cardiomyopathy/nephropathy predisposes the kidney to AMI-induced AKI, in which activation of toll-like receptors and reactive oxygen species derived from NADPH oxidases are involved. Further analysis of crosstalk between diabetic cardiomyopathy and diabetic kidney disease is necessary for obtaining a more comprehensive understanding of modulation of the AMI-AKI axis by diabetes.
Collapse
Affiliation(s)
- Tetsuji Miura
- Department of Clinical Pharmacology, Faculty of Pharmaceutical Sciences, Hokkaido University of Science, Sapporo, Japan.,Department of Cardiovascular, Renal and Metabolic Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Atsushi Kuno
- Department of Cardiovascular, Renal and Metabolic Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan.,Department of Pharmacology, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Marenao Tanaka
- Department of Cardiovascular, Renal and Metabolic Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan
| |
Collapse
|
31
|
Tiwari R, Kapitsinou PP. Role of Endothelial Prolyl-4-Hydroxylase Domain Protein/Hypoxia-Inducible Factor Axis in Acute Kidney Injury. Nephron Clin Pract 2022; 146:243-248. [PMID: 34515168 PMCID: PMC8885783 DOI: 10.1159/000518632] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Accepted: 07/21/2021] [Indexed: 01/03/2023] Open
Abstract
Ischemia reperfusion injury (IRI) results from a cessation or restriction of blood supply to an organ followed by reestablishment of perfusion and reoxygenation. In the kidney, IRI due to transplantation, cardiac surgery with cardiopulmonary bypass, and other major vascular surgeries contributes to acute kidney injury (AKI), a clinical condition associated with significant morbidity and mortality in hospitalized patients. In the postischemic kidney, endothelial damage promotes inflammatory responses and leads to persistent hypoxia of the renal tubular epithelium. Like other cell types, endothelial cells respond to low oxygen tension by multiple hypoxic signaling mechanisms. Key mediators of adaptation to hypoxia are hypoxia-inducible factors (HIF)-1 and -2, transcription factors whose activity is negatively regulated by prolyl-hydroxylase domain proteins 1 to 3 (PHD1 to PHD3). The PHD/HIF axis controls several processes determining injury outcome, including ATP generation, cell survival, proliferation, and angiogenesis. Here, we discuss recent advances in our understanding of the endothelial-derived PHD/HIF signaling and its effects on postischemic AKI.
Collapse
Affiliation(s)
- Ratnakar Tiwari
- Department of Medicine and Feinberg Cardiovascular and Renal Research Institute, Northwestern University Feinberg School of Medicine, Chicago, IL
| | - Pinelopi P. Kapitsinou
- Department of Medicine and Feinberg Cardiovascular and Renal Research Institute, Northwestern University Feinberg School of Medicine, Chicago, IL.,Address correspondence and Lead contact: Dr. Pinelopi P. Kapitsinou, Division of Nephrology and Feinberg Cardiovascular and Renal Research Institute, Feinberg School of Medicine, Northwestern University, 303 East Superior Street, SQBRC 8-408, Chicago, IL 60611.
| |
Collapse
|
32
|
[Acute kidney injury in intensive care unit: A review]. Nephrol Ther 2021; 18:7-20. [PMID: 34872863 DOI: 10.1016/j.nephro.2021.07.324] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Revised: 07/20/2021] [Accepted: 07/23/2021] [Indexed: 12/18/2022]
Abstract
Acute kidney injury is a common complication in intensive care unit. Its incidence is variable according to the studies. It is considered to occur in more than 50 % of patients. Acute kidney injury is responsible for an increase in morbidity (length of hospitalization, renal replacement therapy) but also for excess mortality. The commonly accepted definition of acute kidney injury comes from the collaborative workgroup named Kidney Disease: Improving Global Outcomes (KDIGO). It made it possible to standardize practices and raise awareness among practitioners about monitoring plasma creatinine and also diuresis. Acute kidney injury in intensive care unit is a systemic disease including circulatory, endothelial, epithelial and cellular function involvement and an acute kidney injury is not accompanied by ad integrum repair. After prolonged injury, inadequate repair begins with a fibrotic process. Several mechanisms are involved (cell cycle arrest, epithelial-mesenchymal transition, mitochondrial dysfunction) and result in improper repair. A continuum exists between acute kidney disease and chronic kidney disease, characterized by different renal recovery phenotypes. Thus, preventive measures to prevent the occurrence of kidney damage play a major role in management. The nephrologist must be involved at every stage, from the prevention of the first acute kidney injury (upon arrival in intensive care unit) to long-term follow-up and the care of a chronic kidney disease.
Collapse
|
33
|
Dewitte A, Labat A, Duvignaud PA, Bouche G, Joannes-Boyau O, Ripoche J, Hilbert G, Gruson D, Rubin S, Ouattara A, Boyer A, Combe C. High mean arterial pressure target to improve sepsis-associated acute kidney injury in patients with prior hypertension: a feasibility study. Ann Intensive Care 2021; 11:139. [PMID: 34553274 PMCID: PMC8458519 DOI: 10.1186/s13613-021-00925-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Accepted: 09/06/2021] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND The optimal mean arterial pressure (MAP) in cases of septic shock is still a matter of debate in patients with prior hypertension. An MAP between 75 and 85 mmHg can improve glomerular filtration rate (GFR) but its effect on tubular function is unknown. We assessed the effects of high MAP level on glomerular and tubular renal function in two intensive care units of a teaching hospital. Inclusion criteria were patients with a history of chronic hypertension and developing AKI in the first 24 h of septic shock. Data were collected during two 6 h periods of MAP regimen administered consecutively after haemodynamic stabilisation in an order depending on the patient's admission unit: a high-target period (80-85 mmHg) and a low-target period (65-70 mmHg). The primary endpoint was the creatinine clearance (CrCl) calculated from urine and serum samples at the end of each MAP period by the UV/P formula. RESULTS 26 patients were included. Higher urine output (+0.2 (95%:0, 0.4) mL/kg/h; P = 0.04), urine sodium (+6 (95% CI 0.2, 13) mmol/L; P = 0.04) and lower serum creatinine (- 10 (95% CI - 17, - 3) µmol/L; P = 0.03) were observed during the high-MAP period as compared to the low-MAP period, resulting in a higher CrCl (+25 (95% CI 11, 39) mL/mn; P = 0.002). The urine creatinine, urine-plasma creatinine ratio, urine osmolality, fractional excretion of sodium and urea showed no significant variation. The KDIGO stage at inclusion only interacted with serum creatinine variation and low level of sodium excretion at inclusion did not interact with these results. CONCLUSIONS In the early stage of sepsis-associated AKI, a high-MAP target in patients with a history of hypertension was associated with a higher CrCl, but did not affect the kidneys' ability to concentrate urine, which may reflect no effect on tubular function.
Collapse
Affiliation(s)
- Antoine Dewitte
- CHU Bordeaux, Department of Anaesthesia and Critical Care, Magellan Medico-Surgical Centre, F-33000, Bordeaux, France.
- Univ. Bordeaux, CNRS, UMR 5164, ImmunoConcEpT, F-33000, Bordeaux, France.
| | - Aurore Labat
- CHU Bordeaux, Department of Nephrology-Transplantation-Dialysis-Apheresis, Hôpital Pellegrin, F-33000, Bordeaux, France
| | - Pierre-Antoine Duvignaud
- CHU Bordeaux, Department of Anaesthesia and Critical Care, Magellan Medico-Surgical Centre, F-33000, Bordeaux, France
| | | | - Olivier Joannes-Boyau
- CHU Bordeaux, Department of Anaesthesia and Critical Care, Magellan Medico-Surgical Centre, F-33000, Bordeaux, France
| | - Jean Ripoche
- Univ. Bordeaux, INSERM, UMR 1026, F-33000, Bordeaux, France
| | - Gilles Hilbert
- CHU Bordeaux, Medical Intensive Care Unit, Hôpital Pellegrin, F-33000, Bordeaux, France
| | - Didier Gruson
- CHU Bordeaux, Medical Intensive Care Unit, Hôpital Pellegrin, F-33000, Bordeaux, France
| | - Sébastien Rubin
- CHU Bordeaux, Department of Nephrology-Transplantation-Dialysis-Apheresis, Hôpital Pellegrin, F-33000, Bordeaux, France
| | - Alexandre Ouattara
- CHU Bordeaux, Department of Anaesthesia and Critical Care, Magellan Medico-Surgical Centre, F-33000, Bordeaux, France
- Univ. Bordeaux, INSERM, UMR 1034, Biology of Cardiovascular Diseases, F-33600, Pessac, France
| | - Alexandre Boyer
- CHU Bordeaux, Medical Intensive Care Unit, Hôpital Pellegrin, F-33000, Bordeaux, France
| | - Christian Combe
- CHU Bordeaux, Department of Nephrology-Transplantation-Dialysis-Apheresis, Hôpital Pellegrin, F-33000, Bordeaux, France
| |
Collapse
|
34
|
Kidney Microcirculation as a Target for Innovative Therapies in AKI. J Clin Med 2021; 10:jcm10184041. [PMID: 34575154 PMCID: PMC8471583 DOI: 10.3390/jcm10184041] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Revised: 08/29/2021] [Accepted: 09/02/2021] [Indexed: 12/19/2022] Open
Abstract
Acute kidney injury (AKI) is a serious multifactorial conditions accompanied by the loss of function and damage. The renal microcirculation plays a crucial role in maintaining the kidney’s functional and structural integrity for oxygen and nutrient supply and waste product removal. However, alterations in microcirculation and oxygenation due to renal perfusion defects, hypoxia, renal tubular, and endothelial damage can result in AKI and the loss of renal function regardless of systemic hemodynamic changes. The unique structural organization of the renal microvasculature and the presence of autoregulation make it difficult to understand the mechanisms and the occurrence of AKI following disorders such as septic, hemorrhagic, or cardiogenic shock; ischemia/reperfusion; chronic heart failure; cardiorenal syndrome; and hemodilution. In this review, we describe the organization of microcirculation, autoregulation, and pathophysiological alterations leading to AKI. We then suggest innovative therapies focused on the protection of the renal microcirculation and oxygenation to prevent AKI.
Collapse
|
35
|
Ow CPC, Trask-Marino A, Betrie AH, Evans RG, May CN, Lankadeva YR. Targeting Oxidative Stress in Septic Acute Kidney Injury: From Theory to Practice. J Clin Med 2021; 10:jcm10173798. [PMID: 34501245 PMCID: PMC8432047 DOI: 10.3390/jcm10173798] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 08/17/2021] [Accepted: 08/23/2021] [Indexed: 12/17/2022] Open
Abstract
Sepsis is the leading cause of acute kidney injury (AKI) and leads to increased morbidity and mortality in intensive care units. Current treatments for septic AKI are largely supportive and are not targeted towards its pathophysiology. Sepsis is commonly characterized by systemic inflammation and increased production of reactive oxygen species (ROS), particularly superoxide. Concomitantly released nitric oxide (NO) then reacts with superoxide, leading to the formation of reactive nitrogen species (RNS), predominantly peroxynitrite. Sepsis-induced ROS and RNS can reduce the bioavailability of NO, mediating renal microcirculatory abnormalities, localized tissue hypoxia and mitochondrial dysfunction, thereby initiating a propagating cycle of cellular injury culminating in AKI. In this review, we discuss the various sources of ROS during sepsis and their pathophysiological interactions with the immune system, microcirculation and mitochondria that can lead to the development of AKI. We also discuss the therapeutic utility of N-acetylcysteine and potential reasons for its efficacy in animal models of sepsis, and its inefficacy in ameliorating oxidative stress-induced organ dysfunction in human sepsis. Finally, we review the pre-clinical studies examining the antioxidant and pleiotropic actions of vitamin C that may be of benefit for mitigating septic AKI, including future implications for clinical sepsis.
Collapse
Affiliation(s)
- Connie P. C. Ow
- Preclinical Critical Care Unit, Florey Institute of Neuroscience and Mental Health, Melbourne, VIC 3052, Australia; (C.P.C.O.); (A.T.-M.); (A.H.B.); (R.G.E.); (C.N.M.)
- Department of Cardiac Physiology, National Cerebral and Cardiovascular Center Research Institute, Osaka 564-8565, Japan
| | - Anton Trask-Marino
- Preclinical Critical Care Unit, Florey Institute of Neuroscience and Mental Health, Melbourne, VIC 3052, Australia; (C.P.C.O.); (A.T.-M.); (A.H.B.); (R.G.E.); (C.N.M.)
| | - Ashenafi H. Betrie
- Preclinical Critical Care Unit, Florey Institute of Neuroscience and Mental Health, Melbourne, VIC 3052, Australia; (C.P.C.O.); (A.T.-M.); (A.H.B.); (R.G.E.); (C.N.M.)
- Melbourne Dementia Research Centre, Florey Institute of Neuroscience and Mental Health, Melbourne, VIC 3052, Australia
| | - Roger G. Evans
- Preclinical Critical Care Unit, Florey Institute of Neuroscience and Mental Health, Melbourne, VIC 3052, Australia; (C.P.C.O.); (A.T.-M.); (A.H.B.); (R.G.E.); (C.N.M.)
- Cardiovascular Disease Program, Biomedicine Discovery Institute and Department of Physiology, Monash University, Melbourne, VIC 3800, Australia
| | - Clive N. May
- Preclinical Critical Care Unit, Florey Institute of Neuroscience and Mental Health, Melbourne, VIC 3052, Australia; (C.P.C.O.); (A.T.-M.); (A.H.B.); (R.G.E.); (C.N.M.)
- Department of Critical Care, Melbourne Medical School, University of Melbourne, Melbourne, VIC 3052, Australia
| | - Yugeesh R. Lankadeva
- Preclinical Critical Care Unit, Florey Institute of Neuroscience and Mental Health, Melbourne, VIC 3052, Australia; (C.P.C.O.); (A.T.-M.); (A.H.B.); (R.G.E.); (C.N.M.)
- Department of Critical Care, Melbourne Medical School, University of Melbourne, Melbourne, VIC 3052, Australia
- Correspondence: ; Tel.: +61-3-8344-0417; Fax: +61-3-9035-3107
| |
Collapse
|
36
|
Morimoto N, Honda S, Terai A, Tanabe M, Otani M, Shioji S, Hirasawa S, Aki S, Aoyagi M, Tanaka H. PTRA is useful for renal artery angina by atherosclerotic plaque rupture with unilateral functioning kidney. CEN Case Rep 2021; 11:84-89. [PMID: 34379303 DOI: 10.1007/s13730-021-00634-6] [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: 03/03/2021] [Accepted: 08/07/2021] [Indexed: 10/20/2022] Open
Abstract
We report a case of acute ischemic nephropathy in a patient with severe renal artery stenosis and bradycardia due to sick sinus syndrome. An 83-year-old Japanese woman with a history of hypertension was diagnosed with sick sinus syndrome and scheduled for pacemaker implantation. Four days prior to admission for the procedure, she experienced sudden-onset severe right flank pain that persisted for 1 day. On the day of admission, her serum creatinine level increased from 1.35 mg/dL, measured 2 weeks earlier, to 7.04 mg/dL. Laboratory examinations showed elevated C-reactive protein and lactate dehydrogenase levels. A computed tomography scan showed a severely atrophied left kidney, suggesting that it was non-functioning. Doppler ultrasonography of the right renal artery showed an extended acceleration time, suggesting proximal stenosis. Magnetic resonance imaging showed no enhancement in the proximal portions of the right renal artery, consistent with severe stenosis or occlusion. The patient developed severe bradycardia with lightheadedness; as a result, pacemaker implantation was performed on post-admission day 7. On day 10, digital subtraction angiography revealed diffuse severe stenosis of the right renal artery; intravascular ultrasonography suggested plaque rupture. Percutaneous transluminal renal angioplasty (PTRA) was performed and a drug-eluting stent was placed. On day 11, hemodialysis was performed owing to deteriorating renal function. The patient's renal function dramatically improved shortly thereafter. This case highlights the importance of PTRA for select patients, as it can potentially save some patients from chronic dialysis, and outlines the possible implications of bradycardia in the pathogenesis of ischemic nephropathy.
Collapse
Affiliation(s)
- Nobuhisa Morimoto
- Department of Nephrology, Yokosuka Kyosai Hospital, 1-16 Yonegahamadori, Yokosuka, Kanagawa, 238-8558, Japan.
| | - Seiichiro Honda
- Department of Nephrology, Yokosuka Kyosai Hospital, 1-16 Yonegahamadori, Yokosuka, Kanagawa, 238-8558, Japan
| | - Ayumi Terai
- Department of Nephrology, Yokosuka Kyosai Hospital, 1-16 Yonegahamadori, Yokosuka, Kanagawa, 238-8558, Japan
| | - Madoka Tanabe
- Department of Nephrology, Yokosuka Kyosai Hospital, 1-16 Yonegahamadori, Yokosuka, Kanagawa, 238-8558, Japan
| | - Megumi Otani
- Department of Nephrology, Yokosuka Kyosai Hospital, 1-16 Yonegahamadori, Yokosuka, Kanagawa, 238-8558, Japan
| | - Shingo Shioji
- Department of Nephrology, Yokosuka Kyosai Hospital, 1-16 Yonegahamadori, Yokosuka, Kanagawa, 238-8558, Japan
| | - Suguru Hirasawa
- Department of Nephrology, Yokosuka Kyosai Hospital, 1-16 Yonegahamadori, Yokosuka, Kanagawa, 238-8558, Japan
| | - Shota Aki
- Department of Nephrology, Yokosuka Kyosai Hospital, 1-16 Yonegahamadori, Yokosuka, Kanagawa, 238-8558, Japan
| | - Makoto Aoyagi
- Department of Nephrology, Yokosuka Kyosai Hospital, 1-16 Yonegahamadori, Yokosuka, Kanagawa, 238-8558, Japan
| | - Hiroyuki Tanaka
- Department of Nephrology, Yokosuka Kyosai Hospital, 1-16 Yonegahamadori, Yokosuka, Kanagawa, 238-8558, Japan
| |
Collapse
|
37
|
Spencer S, Wheeler-Jones C, Elliott J. Hypoxia and chronic kidney disease: Possible mechanisms, therapeutic targets, and relevance to cats. Vet J 2021; 274:105714. [PMID: 34252550 DOI: 10.1016/j.tvjl.2021.105714] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Revised: 07/01/2021] [Accepted: 07/06/2021] [Indexed: 10/20/2022]
Abstract
There is mounting evidence that kidney ischaemia/hypoxia plays an important role in feline chronic kidney disease (CKD) development and progression, as well as in human disease and laboratory animal models. Ischaemic acute kidney injury is widely accepted as a cause of CKD in people and data from laboratory species has identified some of the pathways underlying this continuum. Experimental kidney ischaemia in cats results in morphological changes, namely chronic tubulointerstitial inflammation, tubulointerstitial fibrosis, and tubular atrophy, akin to those observed in naturally-occurring CKD. Multiple situations are envisaged that could result in acute or chronic episodes of kidney hypoxia in cats, while risk factors identified in epidemiological studies provide further support that kidney hypoxia contributes to spontaneously occurring feline CKD. This review evaluates the evidence for the role of kidney ischaemia/hypoxia in feline CKD and the proposed mechanisms and consequences of kidney hypoxia. As no effective treatments exist that substantially slow or prevent feline CKD progression, there is a need for novel therapeutic strategies. Targeting kidney hypoxia is one such promising approach, with therapies including those that attenuate the hypoxia-inducible factor (HIF) pathway already being utilised in human CKD.
Collapse
Affiliation(s)
- Sarah Spencer
- Comparative Biomedical Sciences, The Royal Veterinary College, Royal College Street, London NW1 0TU, UK.
| | - Caroline Wheeler-Jones
- Comparative Biomedical Sciences, The Royal Veterinary College, Royal College Street, London NW1 0TU, UK
| | - Jonathan Elliott
- Comparative Biomedical Sciences, The Royal Veterinary College, Royal College Street, London NW1 0TU, UK
| |
Collapse
|
38
|
Perspectives on the Role of Magnetic Resonance Imaging (MRI) for Noninvasive Evaluation of Diabetic Kidney Disease. J Clin Med 2021; 10:jcm10112461. [PMID: 34199385 PMCID: PMC8199575 DOI: 10.3390/jcm10112461] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 05/27/2021] [Accepted: 05/28/2021] [Indexed: 01/09/2023] Open
Abstract
Renal magnetic resonance imaging (MRI) techniques are currently in vogue, as they provide in vivo information on renal volume, function, metabolism, perfusion, oxygenation, and microstructural alterations, without the need for exogenous contrast media. New imaging biomarkers can be identified using these tools, which represent a major advance in the understanding and study of the different pathologies affecting the kidney. Diabetic kidney disease (DKD) is one of the most important diseases worldwide due to its high prevalence and impact on public health. However, its multifactorial etiology poses a challenge for both basic and clinical research. Therefore, the use of novel renal MRI techniques is an attractive step forward in the comprehension of DKD, both in its pathogenesis and in its detection and surveillance in the clinical practice. This review article outlines the most promising MRI techniques in the study of DKD, with the purpose of stimulating their clinical translation as possible tools for the diagnosis, follow-up, and monitoring of the clinical impacts of new DKD treatments.
Collapse
|
39
|
Beneficial Effects of Vasopressin Compared With Norepinephrine on Renal Perfusion, Oxygenation, and Function in Experimental Septic Acute Kidney Injury. Crit Care Med 2021; 48:e951-e958. [PMID: 32931198 DOI: 10.1097/ccm.0000000000004511] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
OBJECTIVES To compare the effects of restoring mean arterial pressure with vasopressin or norepinephrine on systemic hemodynamics, renal blood flow, intrarenal perfusion and oxygenation, and renal function in ovine septic acute kidney injury. DESIGN Interventional Study. SETTING Research Institute. SUBJECTS Adult Merino ewes. INTERVENTIONS Flow probes were implanted on the pulmonary and renal arteries (and the mesenteric artery in sheep that received vasopressin). Fiber-optic probes were implanted in the renal cortex and medulla to measure tissue perfusion and oxygen tension (PO2). Conscious sheep were administered Escherichia coli to induce septic acute kidney injury. Vasopressin (0.03 IU/min [0.03-0.05 IU/min]; n = 7) or norepinephrine (0.60 μg/kg/min [0.30-0.70 μg/kg/min]; n = 7) was infused IV and titrated to restore baseline mean arterial pressure during 24-30 hours of sepsis. MEASUREMENTS AND MAIN RESULTS Ovine septic acute kidney injury was characterized by reduced mean arterial pressure (-16% ± 2%) and creatinine clearance (-65% ± 9%) and increased renal blood flow (+34% ± 7%) but reduced renal medullary perfusion (-44% ± 7%) and PO2 (-47% ± 10%). Vasopressin infusion did not significantly affect renal medullary perfusion or PO2 and induced a sustained (6 hr) ~2.5-fold increase in creatinine clearance. Vasopressin reduced sepsis-induced mesenteric hyperemia (+61 ± 13 to +9% ± 6%). Norepinephrine transiently (2 hr) improved creatinine clearance (by ~3.5-fold) but worsened renal medullary ischemia (to -64% ± 7%) and hypoxia (to -71% ± 6%). CONCLUSIONS In ovine septic acute kidney injury, restoration of mean arterial pressure with vasopressin induced a more sustained improvement in renal function than norepinephrine, without exacerbating renal medullary ischemia and hypoxia or reducing mesenteric blood flow below baseline values.
Collapse
|
40
|
Singh AK, Kolligundla LP, Francis J, Pasupulati AK. Detrimental effects of hypoxia on glomerular podocytes. J Physiol Biochem 2021; 77:193-203. [PMID: 33835424 DOI: 10.1007/s13105-021-00788-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Accepted: 01/11/2021] [Indexed: 12/19/2022]
Abstract
Hypoxia-inducible factor1 (HIF1) plays a pivotal role in ensuring cells adapt to low-oxygen conditions. Depletion of oxygen, a co-substrate during hydroxylation of prolyl (P402 and P564) residues of HIF1⍺, evades HIF1⍺ ubiquitination and enables its dimerization with HIF1β to mediate global transcriptional response to hypoxia. Though HIF1 is largely considered eliciting a protective role during physiological or pathological hypoxia or ischemia, elevated HIF1 during chronic hypoxia contributes to glomerular diseases' pathology and proteinuria. The glomerulus is responsible for renal permselectivity and excretion of ultra-filtrated urine. Podocytes are the glomerulus' major cell types and are instrumental for glomerular filtration, permselectivity, and glomerular basement membrane maintenance. Podocyte injury is expected to impair the efficiency of glomerular filtration and manifestation of glomerulosclerosis and proteinuria. Accumulated evidence suggests that podocytes are susceptible to various insults during chronic hypoxia, including podocyte EMT, slit-diaphragm dysfunction, foot process effacement, and cytoskeletal derangement due to accumulation of HIF1. This review discusses how hypoxia/HIF1 signaling regulates various features and function of podocytes during exposure to chronic hypoxia or inducing HIF1 by various chemical modulators.
Collapse
Affiliation(s)
- Ashish K Singh
- Department of Biochemistry, School of Life Sciences, University of Hyderabad, Hyderabad, India
| | - Lakshmi P Kolligundla
- Department of Biochemistry, School of Life Sciences, University of Hyderabad, Hyderabad, India
| | - Justus Francis
- Department of Biochemistry, School of Life Sciences, University of Hyderabad, Hyderabad, India
| | - Anil K Pasupulati
- Department of Biochemistry, School of Life Sciences, University of Hyderabad, Hyderabad, India.
| |
Collapse
|
41
|
Textor SC, Abumoawad A, Saad A, Ferguson C, Dietz A. Stem Cell Therapy for Microvascular Injury Associated with Ischemic Nephropathy. Cells 2021; 10:cells10040765. [PMID: 33807289 PMCID: PMC8066553 DOI: 10.3390/cells10040765] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2021] [Revised: 03/22/2021] [Accepted: 03/26/2021] [Indexed: 12/15/2022] Open
Abstract
Ischemic nephropathy reflects progressive loss of kidney function due to large vessel atherosclerotic occlusive disease. Recent studies indicate that this process is characterized by microvascular rarefaction, increased tissue hypoxia and activation of inflammatory processes of tissue injury. This review summarizes the rationale and application of functional MR imaging to evaluate tissue oxygenation in human subjects that defines the limits of renal adaptation to reduction in blood flow, development of increasingly severe tissue hypoxia and recruitment of inflammatory injury pathways in ischemic nephropathy. Human mesenchymal stromal/stem cells (MSC) are capable of modifying angiogenic pathways and immune responses, but the potency of these effects vary between individuals and various clinical characteristics including age and chronic kidney disease and levels of hypoxia. We summarize recently completed first-in-human studies applying intrarenal infusion of autologous adipose-derived MSC in human subjects with ischemic nephropathy that demonstrate a rise in blood flow and reduction in tissue hypoxia consistent with partial repair of microvascular injury, even without restoring main renal arterial blood flow. Inflammatory biomarkers in the renal vein of post-stenotic kidneys fell after MSC infusion. These changes were associated with modest but significant dose-related increments in kidney function. These data provide support a role for autologous MSC in repair of microvascular injury associated with tissue hypoxia.
Collapse
Affiliation(s)
- Stephen C. Textor
- Mayo Clinic, Division of Nephrology and Hypertension, Rochester, MN 55905, USA;
- Correspondence:
| | - Abdu Abumoawad
- Department of Medicine University of Missouri, Kansas, MO 64108, USA;
| | - Ahmed Saad
- Department of Medicine Creighton University School of Medicine, Omaha, NE 68124, USA;
| | | | - Allan Dietz
- Mayo Clinic, Human Cell Therapy Laboratory, Rochester, MN 55905, USA;
| |
Collapse
|
42
|
Lu F, Yang J, Yang S, Bernd K, Fu C, Yang C, Xu H, Liu M, Zhan S, Wang C, Guo R, Wu Y. Use of Three-Dimensional Arterial Spin Labeling to Evaluate Renal Perfusion in Patients With Chronic Kidney Disease. J Magn Reson Imaging 2021; 54:1152-1163. [PMID: 33769645 DOI: 10.1002/jmri.27609] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 03/06/2021] [Accepted: 03/09/2021] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND A noninvasive method for evaluating renal blood flow (RBF) in patients with chronic kidney disease (CKD) may have clinical value in disease staging, management, and prognostication. PURPOSE To evaluate effectiveness of three-dimensional pseudocontinuous arterial spin labeling (pCASL) and pulsed arterial spin labeling (PASL) in assessment of cortex and outer medulla (cortex/OM) RBF in CKD patients and healthy volunteers (HVs). STUDY TYPE Prospective, in a single institution. SUBJECTS A total of 48 CKD patients (stage 1, 2, 3, and 4-5: N = 11, 12, 13, and 12, respectively) and 18 HVs FIELD STRENGTH/SEQUENCE: 3 T, pCASL, and PASL with a three-dimensional hybrid gradient echo/spin echo sequence. ASSESSMENT Quality of RBF images derived from pCASL and PASL were evaluated and RBF in cortex/OM measured. Clinical and laboratory data were recorded. STATISTICAL TESTS Image quality differences between pCASL and PASL were evaluated with Wilcoxon signed-rank test. For both methods, analysis of variance, followed by Fisher's LSD-t test, was used to determine whether RBF differed between CKD stages and HVs. Pearson correlation coefficients were calculated to assess strength of relationships between cortex/OM RBF and data from clinical and laboratory tests. RESULTS Image quality differences were significantly higher in pCASL than PASL in both patients and HVs (both P < 0.05). For pCASL, cortex/OM RBF of patients were significantly lower than those of HVs (P < 0.05). Cortex/OM RBF were higher in S1 and S2 patients than those in S3 and S4-5 (P < 0.05). For PASL, only RBF in cortex of S1 and S2 patients were significantly higher than those of S4-5 (P < 0.05). Good correlations between pCASL RBF and estimated glomerular filtration (eGFR) were found in cortex/OM of patients (rho = 0.796 and 0.798, respectively, both P < 0.05), higher than those between PASL RBF and eGFR (rho = 0.430 and 0.374, respectively, both P < 0.05). DATA CONCLUSION Three-dimensional pCASL may potentially be a noninvasive technique to assess renal perfusion in CKD patients in different stages. LEVEL OF EVIDENCE 1 TECHNICAL EFFICACY: Stage 2.
Collapse
Affiliation(s)
- Fang Lu
- Department of Radiology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jing Yang
- Department of Nephrology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Shuohui Yang
- Department of Radiology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Kuehn Bernd
- MR Applications Development, Siemens Healthcare, Erlangen, Germany
| | - Caixia Fu
- MR Applications Development, Siemens Shenzhen Magnetic Resonance Ltd, Shenzhen, China
| | - Chenyao Yang
- Department of Radiology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Huihui Xu
- Department of Radiology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Mengxiao Liu
- MR Scientific Marketing, Siemens Healthcare, Shanghai, China
| | - Songhua Zhan
- Department of Radiology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Chen Wang
- Department of Nephrology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Rongfang Guo
- Department of Radiology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yu Wu
- Department of Radiology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
43
|
Mou Z, Guan T, Chen L. Risk Factors of Acute Kidney Injury in ECMO Patients: A Systematic Review and Meta-Analysis. J Intensive Care Med 2021; 37:267-277. [PMID: 33761767 DOI: 10.1177/08850666211003485] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
PURPOSE Acute kidney injury (AKI) is one of the most common complications in patients receiving extracorporeal membrane oxygenation (ECMO), but there is no systematic analysis regarding its risk factors. This meta-analysis aims to determine the risk factors of AKI in adult patients with ECMO treatment. METHODS Two authors independently carried out a systemic literature search using PubMed, Web of Science, and Embase until April 20, 2020 (inclusive) to enroll 12 studies reporting the necessary clinical characteristics. The Gender (male), age, APACHE II score, SOFA score, cancer, diabetes mellitus (DM), intra-aortic balloon pump (IABP), postcardiotomy, and ECMO supporting duration were pooled for further analysis by STATA. RESULTS Adult patients receiving ECMO who develop AKI and severe AKI incidents are usually older or have a higher APACHE II scores; in addition, severe AKI is related to higher SOFA scores, DM, and longer duration of ECMO support. CONCLUSIONS Patients with these clinical characteristics should be paid more attention during ECMO. There remains a need for additional studies to validate these conclusions and to detect additional AKI risk factors for ECMO patients.
Collapse
Affiliation(s)
- Zhixiang Mou
- Department of Nephrology, 66366Zhongshan Hospital Xiamen University, Xiamen, China
| | - Tianjun Guan
- Department of Nephrology, 66366Zhongshan Hospital Xiamen University, Xiamen, China
| | - Lan Chen
- Department of Nephrology, 66366Zhongshan Hospital Xiamen University, Xiamen, China
| |
Collapse
|
44
|
Dinc B, Yilmaz VT, Aycan İO, Kisaoglu A, Dandin O, Aydinli B, Hadimioglu N, Ertug Z. Effect of post-perfusion hyperoxemia on early graft function in renal transplant recipients: a retrospective observational cohort study. Ir J Med Sci 2021; 190:1539-1545. [PMID: 33398714 DOI: 10.1007/s11845-020-02499-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Accepted: 12/25/2020] [Indexed: 11/25/2022]
Abstract
BACKGROUND The effects of hyperoxemia on the transplanted grafts arouse interest nowadays, particularly intraoperative hyperoxemia, on transplant kidney function and survival in the 1-year post-operative period. AIMS We aimed to investigate the effect of post-perfusion (5 min after perfusion) hyperoxemia on early graft function and survival in renal transplant recipients. METHODS Two hundred forty-seven living donor kidney transplant recipients were included in the study. Patients were divided into the three groups according to their partial arterial oxygen pressure in post-perfusion blood gas samples: group 1: normoxia (n = 52, PaO2 pressure: < 120 mmHg, 103 ± 13); group 2: moderate hyperoxemia (n = 121, PaO2: 120-200 mmHg, 169 ± 21); group 3: severe hyperoxemia (n = 74, PaO2: > 200 mmHg, 233 ± 25). Graft functions (serum creatinine levels, estimated-glomerular filtration rate values, spot urine protein/creatinine ratio), survival rates, and groups' clinical outcomes were compared in the first year after transplantation. RESULTS Graft survival rates were similar in the groups and the rate of BK virus viremia was the lowest in the group 3 (groups 1, 2, and 3: 15.4% (n = 8), 6.6% (n = 8), 1.4% (n = 1), respectively, P: 0.009). Serum creatinine and proteinuria levels were lower, and estimated-glomerular filtration rate values were higher in group 3. A negative correlation between partial arterial oxygen pressure and serum creatinine levels and a positive correlation with estimated-glomerular filtration rate value were noted. These results were confirmed by univariate and multivariate analyses. CONCLUSIONS We demonstrated that the kidney transplant recipients with post-perfusion hyperoxemia have better early graft functions and lower BK virus viremia rates. TRIAL REGISTRATION ClinicalTrials.gov Identifier: NCT04420897.
Collapse
Affiliation(s)
- Bora Dinc
- Department of Anaesthesiology and Reanimation, Akdeniz University Medical School, Antalya, Turkey
| | - Vural T Yilmaz
- Division of Nephrology, Department of Internal Medicine, Akdeniz University Medical School, Antalya, Turkey.
| | - İlker O Aycan
- Department of Anaesthesiology and Reanimation, Akdeniz University Medical School, Antalya, Turkey
| | - Abdullah Kisaoglu
- Department of General Surgery, Akdeniz University Medical School, Antalya, Turkey
| | - Ozgur Dandin
- Department of General Surgery, Akdeniz University Medical School, Antalya, Turkey
| | - Bulent Aydinli
- Department of General Surgery, Akdeniz University Medical School, Antalya, Turkey
| | - Necmiye Hadimioglu
- Department of Anaesthesiology and Reanimation, Akdeniz University Medical School, Antalya, Turkey
| | - Zeki Ertug
- Department of Anaesthesiology and Reanimation, Akdeniz University Medical School, Antalya, Turkey
| |
Collapse
|
45
|
Cantow K, Evans RG, Grosenick D, Gladytz T, Niendorf T, Flemming B, Seeliger E. Quantitative Assessment of Renal Perfusion and Oxygenation by Invasive Probes: Basic Concepts. Methods Mol Biol 2021; 2216:89-107. [PMID: 33475996 PMCID: PMC9703258 DOI: 10.1007/978-1-0716-0978-1_6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/21/2023]
Abstract
Renal tissue hypoperfusion and hypoxia are early key elements in the pathophysiology of acute kidney injury of various origins, and may also promote progression from acute injury to chronic kidney disease. Here we describe basic principles of methodology to quantify renal hemodynamics and tissue oxygenation by means of invasive probes in experimental animals. Advantages and disadvantages of the various methods are discussed in the context of the heterogeneity of renal tissue perfusion and oxygenation.This chapter is based upon work from the COST Action PARENCHIMA, a community-driven network funded by the European Cooperation in Science and Technology (COST) program of the European Union, which aims to improve the reproducibility and standardization of renal MRI biomarkers. This introduction chapter is complemented by a separate chapter describing the experimental procedure and data analysis.
Collapse
Affiliation(s)
- Kathleen Cantow
- Working Group Integrative Kidney Physiology, Institute of Physiology, Charité-University Medicine Berlin, Berlin, Germany
| | - Roger G Evans
- Cardiovascular Disease Program, Biomedicine Discovery Institute and Department of Physiology, Monash University, Melbourne, Australia
| | - Dirk Grosenick
- Physikalisch-Technische Bundesanstalt (German Federal Metrologic Institute), Berlin, Germany
| | - Thomas Gladytz
- Physikalisch-Technische Bundesanstalt (German Federal Metrologic Institute), Berlin, Germany
| | - Thoralf Niendorf
- Berlin Ultrahigh Field Facility (B.U.F.F.), Max Delbrück Center for Molecular Medicine (MDC) in the Helmholtz Association, Berlin, Germany
| | - Bert Flemming
- Working Group Integrative Kidney Physiology, Institute of Physiology, Charité-University Medicine Berlin, Berlin, Germany
| | - Erdmann Seeliger
- Working Group Integrative Kidney Physiology, Institute of Physiology, Charité-University Medicine Berlin, Berlin, Germany.
| |
Collapse
|
46
|
Abosamak MF, Lippi G, Benoit SW, Henry BM, Shama AAA. Bladder urine oxygen partial pressure monitoring: Could it be a tool for early detection of acute kidney injury? EGYPTIAN JOURNAL OF ANAESTHESIA 2021. [DOI: 10.1080/11101849.2021.1878686] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022] Open
Affiliation(s)
- Mohammed Fawzi Abosamak
- Department of Anesthesia and Intensive Care, Faculty of Medicine, Tanta University, Tanta, Egypt
| | - Giuseppe Lippi
- Department of Neuroscience, Section of Clinical Biochemistry, Biomedicine and Movement, University of Verona, Verona, Italy
| | - Stefanie W. Benoit
- Division of Nephrology and Hypertension, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
- Department of Pediatrics, University of Cincinnati, College of Medicine, Cincinnati, OH, USA
| | - Brandon Michael Henry
- Cardiac Intensive Care Unit, The Heart Institute, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
| | | |
Collapse
|
47
|
Cantow K, Ladwig-Wiegard M, Flemming B, Fekete A, Hosszu A, Seeliger E. Reversible (Patho)Physiologically Relevant Test Interventions: Rationale and Examples. Methods Mol Biol 2021; 2216:57-73. [PMID: 33475994 DOI: 10.1007/978-1-0716-0978-1_4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Renal tissue hypoperfusion and hypoxia are early key elements in the pathophysiology of acute kidney injury of various origins, and may also promote progression from acute injury to chronic kidney disease. Here we describe test interventions that are used to study the control of renal hemodynamics and oxygenation in experimental animals in the context of kidney-specific control of hemodynamics and oxygenation. The rationale behind the use of the individual tests, the physiological responses of renal hemodynamics and oxygenation, the use in preclinical studies, and the possible application in humans are discussed.This chapter is based upon work from the COST Action PARENCHIMA, a community-driven network funded by the European Cooperation in Science and Technology (COST) program of the European Union, which aims to improve the reproducibility and standardization of renal MRI biomarkers.
Collapse
Affiliation(s)
- Kathleen Cantow
- Working Group Integrative Kidney Physiology, Institute of Physiology, Charité-University Medicine Berlin, Berlin, Germany
| | - Mechthild Ladwig-Wiegard
- Institute of Animal Welfare, Animal Behavior and Laboratory Animal Science, Free University Berlin, Berlin, Germany
| | - Bert Flemming
- Working Group Integrative Kidney Physiology, Institute of Physiology, Charité-University Medicine Berlin, Berlin, Germany
| | - Andrea Fekete
- 1st Department of Pediatrics, Semmelweis University, Budapest, Hungary
| | - Adam Hosszu
- 1st Department of Pediatrics, Semmelweis University, Budapest, Hungary
| | - Erdmann Seeliger
- Working Group Integrative Kidney Physiology, Institute of Physiology, Charité-University Medicine Berlin, Berlin, Germany.
| |
Collapse
|
48
|
La Porta E, Lanino L, Calatroni M, Caramella E, Avella A, Quinn C, Faragli A, Estienne L, Alogna A, Esposito P. Volume Balance in Chronic Kidney Disease: Evaluation Methodologies and Innovation Opportunities. Kidney Blood Press Res 2021; 46:396-410. [PMID: 34233334 DOI: 10.1159/000515172] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Accepted: 02/10/2021] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND Patients affected by chronic kidney disease are at a risk of cardiovascular morbidity and mortality. Body fluids unbalance is one of the main characteristics of this condition, as fluid overload is highly prevalent in patients affected by the cardiorenal syndrome. SUMMARY We describe the state of the art and new insights into body volume evaluation. The mechanisms behind fluid balance are often complex, mainly because of the interplay of multiple regulatory systems. Consequently, its management may be challenging in clinical practice and even more so out-of-hospital. Availability of novel technologies offer new opportunities to improve the quality of care and patients' outcome. Development and validation of new technologies could provide new tools to reduce costs for the healthcare system, promote personalized medicine, and boost home care. Due to the current COVID-19 pandemic, a proper monitoring of chronic patients suffering from fluid unbalances is extremely relevant. Key Message: We discuss the main mechanisms responsible for fluid overload in different clinical contexts, including hemodialysis, peritoneal dialysis, and heart failure, emphasizing the potential impact provided by the implementation of the new technologies.
Collapse
Affiliation(s)
- Edoardo La Porta
- Department of Cardionephrology, Istituto Clinico Di Alta Specialità (ICLAS), Rapallo, Italy
- Department of Internal Medicine (DIMI), University of Genoa, Genoa, Italy
| | - Luca Lanino
- Department of Internal Medicine (DIMI), University of Genoa, Genoa, Italy
| | - Marta Calatroni
- Division of Nephrology, Humanitas Clinical and Research Center, Milan, Italy
| | - Elena Caramella
- Division of Nephrology and Dialysis, Ospedale Sant'Anna, San Fermo della Battaglia, Como, Italy
| | - Alessandro Avella
- Division of Nephrology and Dialysis, Ospedale di Circolo e Fondazione Macchi, Varese, Italy
| | - Caroline Quinn
- Department of Biological Sciences, Rensselaer Polytechnic Institute, Troy, New York, USA
| | - Alessandro Faragli
- Department of Internal Medicine and Cardiology, Deutsches Herzzentrum Berlin, Berlin, Germany
- Department of Internal Medicine and Cardiology, Campus Virchow-Klinikum, Charité - Universitätsmedizin Berlin, Berlin, Germany
- Berlin Institute of Health (BIH), Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
| | - Luca Estienne
- Department of Nephrology and Dialysis, SS. Antonio e Biagio e Cesare Arrigo Hospital, Alessandria, Italy
| | - Alessio Alogna
- Department of Internal Medicine and Cardiology, Campus Virchow-Klinikum, Charité - Universitätsmedizin Berlin, Berlin, Germany
- Berlin Institute of Health (BIH), Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
| | - Pasquale Esposito
- Division of Nephrology, Department of Internal Medicine, Dialysis and Transplantation, University of Genoa and IRCCS Policlinico San Martino, Genoa, Italy
| |
Collapse
|
49
|
Poudel N, Zheng S, Schinderle CM, Sun N, Hu S, Okusa MD. Peritubular Capillary Oxygen Consumption in Sepsis-Induced AKI: Multi-Parametric Photoacoustic Microscopy. Nephron Clin Pract 2020; 144:621-625. [PMID: 33147592 DOI: 10.1159/000511167] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Accepted: 08/24/2020] [Indexed: 12/22/2022] Open
Abstract
Understanding and measuring parameters responsible for the pathogenesis of sepsis-induced AKI (SI-AKI) is critical in developing therapies. Blood flow to the kidney is heterogeneous, partly due to the existence of dynamic networks of capillaries in various regions, responding differentially to oxygen demand in cortex versus medulla. High energy demand regions, especially the outer medulla, are susceptible to hypoxia and subject to damage during SI-AKI. Proximal tubule epithelial cells in the cortex and the outer medulla can also undergo metabolic reprogramming during SI-AKI to maintain basal physiological status and to avoid potential damage. Current data on the assessment of renal hemodynamics and oxygen metabolism during sepsis is limited. Preclinical and clinical studies show changes in renal hemodynamics associated with SI-AKI, and in clinical settings, interventions to manage renal hemodynamics seem to help improve disease outcomes in some cases. Lack of proper tools to assess temporospatial changes in peritubular blood flow and tissue oxygen metabolism is a barrier to our ability to understand microcirculatory dynamics and oxygen consumption and their role in the pathogenesis of SI-AKI. Current tools to assess renal oxygenation are limited in their usability as these cannot perform continuous simultaneous measurement of renal hemodynamics and oxygen metabolism. Multi-parametric photo-acoustic microscopy (PAM) is a new tool that can measure real-time changes in microhemodynamics and oxygen metabolism. Use of multi-parametric PAM in combination with advanced intravital imaging techniques has the potential to understand the contribution of microhemodynamic and tissue oxygenation alterations to SI-AKI.
Collapse
Affiliation(s)
- Nabin Poudel
- Division of Nephrology and Center for Immunity, Inflammation and Regenerative Medicine, University of Virginia, Charlottesville, Virginia, USA
| | - Shuqiu Zheng
- Division of Nephrology and Center for Immunity, Inflammation and Regenerative Medicine, University of Virginia, Charlottesville, Virginia, USA
| | - Colleen M Schinderle
- Division of Nephrology and Center for Immunity, Inflammation and Regenerative Medicine, University of Virginia, Charlottesville, Virginia, USA
| | - Naidi Sun
- Department of Biomedical Engineering, Washington University at St. Louis, St. Louis, Missouri, USA
| | - Song Hu
- Department of Biomedical Engineering, Washington University at St. Louis, St. Louis, Missouri, USA
| | - Mark D Okusa
- Division of Nephrology and Center for Immunity, Inflammation and Regenerative Medicine, University of Virginia, Charlottesville, Virginia, USA,
| |
Collapse
|
50
|
Iguchi N, Kosaka J, Iguchi Y, Evans RG, Bellomo R, May CN, Lankadeva YR. Systemic haemodynamic, renal perfusion and renal oxygenation responses to changes in inspired oxygen fraction during total intravenous or volatile anaesthesia. Br J Anaesth 2020; 125:192-200. [DOI: 10.1016/j.bja.2020.03.033] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Revised: 03/13/2020] [Accepted: 03/18/2020] [Indexed: 02/03/2023] Open
|