1
|
Kim HD, Kim EN, Lim JH, Kim Y, Ban TH, Lee H, Kim YS, Park CW, Choi BS. Phosphodiesterase inhibitor ameliorates senescent changes of renal interstitial pericytes in aging kidney. Aging Cell 2024; 23:e14075. [PMID: 38155524 PMCID: PMC10928568 DOI: 10.1111/acel.14075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Revised: 12/12/2023] [Accepted: 12/14/2023] [Indexed: 12/30/2023] Open
Abstract
Pericytes are mesenchymal cells that surround endothelial cells, playing a crucial role in angiogenesis and vessel maturation. Additionally, they are associated with interstitial fibrosis as a major contributor to renal myofibroblasts. In this study, we aim to investigate whether the phosphodiesterase inhibitor, pentoxifylline (PTX), can ameliorate aging-related functional and histological deterioration in the kidney. We subjected aging C57BL/6 mice, dividing into young, aging, and PTX-treated aging groups. Renal function, albuminuria, and histological changes were assessed. Interstitial pericytes were assessed by immunohistochemistry analysis. We examined changes in pericytes in elderly patients using human kidney tissue obtained from healthy kidney donors for kidney transplantation. In vitro experiments with human pericytes and endothelial cells were performed. Aging mice exhibited declined renal function, increased albuminuria, and aging-related histological changes including mesangial expansion and tubulointerstitial fibrosis. Notably, number of pericytes declined in aging kidneys, and myofibroblasts increased. PTX treatment ameliorated albuminuria, histological alterations, and microvascular rarefaction, as well as modulated angiopoietin expression. In vitro experiments showed PTX reduced cellular senescence and inflammation. Human kidney analysis confirmed similar pericyte changes in aging kidneys. The phosphodiesterase inhibitor, PTX preserved microvascular density and improved renal interstitial fibrosis and inflammation in aging mice kidneys. These protective effects were suggested to be associated with the amelioration of pericytes reduction and the transition to myofibroblasts. Additionally, the upregulation of angiopoietin-1 expression may exert potential impacts. To the best of our knowledge, this is the first report on the changes in renal interstitial pericytes in aging human kidneys.
Collapse
Affiliation(s)
- Hyung Duk Kim
- Department of Internal Medicine, Eunpyeong St. Mary's Hospital, College of MedicineThe Catholic University of KoreaSeoulRepublic of Korea
| | - Eun Nim Kim
- Department of Internal Medicine, College of MedicineThe Catholic University of KoreaSeoulRepublic of Korea
| | - Ji Hee Lim
- Department of Internal Medicine, College of MedicineThe Catholic University of KoreaSeoulRepublic of Korea
| | - Yaeni Kim
- Department of Internal Medicine, Seoul St. Mary's Hospital, College of MedicineThe Catholic University of KoreaSeoulRepublic of Korea
| | - Tae Hyun Ban
- Department of Internal Medicine, Eunpyeong St. Mary's Hospital, College of MedicineThe Catholic University of KoreaSeoulRepublic of Korea
| | - Hajeong Lee
- Department of Internal MedicineSeoul National University HospitalSeoulRepublic of Korea
| | - Yon Su Kim
- Department of Internal MedicineSeoul National University HospitalSeoulRepublic of Korea
| | - Cheol Whee Park
- Department of Internal Medicine, Seoul St. Mary's Hospital, College of MedicineThe Catholic University of KoreaSeoulRepublic of Korea
| | - Bum Soon Choi
- Department of Internal Medicine, Eunpyeong St. Mary's Hospital, College of MedicineThe Catholic University of KoreaSeoulRepublic of Korea
| |
Collapse
|
2
|
Chou YH, Pan SY, Shih HM, Lin SL. Update of pericytes function and their roles in kidney diseases. J Formos Med Assoc 2024; 123:307-317. [PMID: 37586973 DOI: 10.1016/j.jfma.2023.08.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Accepted: 08/01/2023] [Indexed: 08/18/2023] Open
Abstract
Studies have highlighted the significant involvement of kidney pericytes in renal fibrosis. Kidney pericytes, classified as interstitial mesenchymal cells, are extensively branched, collagen-producing cells that closely interact with endothelial cells. This article aims to provide an overview of the recent advancements in understanding the physiological functions of pericytes and their roles in kidney diseases. In a healthy kidney, pericytes have essential physiological function in angiogenesis, erythropoietin (EPO) production, and the regulation of renal blood flow. Nevertheless, pericyte-myofibroblast transition has been identified as the primary cause of disease progression in acute kidney injury (AKI)-to-chronic kidney disease (CKD) continuum. Our recent research has demonstrated that hypoxia-inducible factor-2α (HIF-2α) regulates erythropoietin production in pericytes. However, this production is repressed by EPO gene hypermethylation and HIF-2α downregulation which were induced by transforming growth factor-β1-activated DNA methyltransferase and activin receptor-like kinase-5 signaling pathway during renal fibrosis, respectively. Additionally, AKI induces epigenetic modifications in pericytes, rendering them more prone to extracellular matrix production, cell migration and proliferation, thereby contributing to subsequent capillary rarefaction and renal fibrosis. Further investigation into the specific functions and roles of different subpopulations of pericytes may contribute for the development of targeted therapies aimed at attenuating kidney disease and mitigating their adverse effects.
Collapse
Affiliation(s)
- Yu-Hsiang Chou
- Renal Division, Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Szu-Yu Pan
- Renal Division, Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan; Department of Integrated Diagnostics and Therapeutics, National Taiwan University Hospital, Taipei, Taiwan
| | - Hong-Mou Shih
- Division of Nephrology, Department of Internal Medicine, MacKay Memorial Hospital, Taipei, Taiwan
| | - Shuei-Liong Lin
- Renal Division, Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan; Graduate Institute of Physiology, College of Medicine, National Taiwan University, Taipei, Taiwan; Research Center for Developmental Biology and Regenerative Medicine, National Taiwan University, Taipei, Taiwan.
| |
Collapse
|
3
|
Zhang M, Ye Z, Yuan E, Lv X, Zhang Y, Tan Y, Xia C, Tang J, Huang J, Li Z. Imaging-based deep learning in kidney diseases: recent progress and future prospects. Insights Imaging 2024; 15:50. [PMID: 38360904 PMCID: PMC10869329 DOI: 10.1186/s13244-024-01636-5] [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: 11/19/2023] [Accepted: 01/27/2024] [Indexed: 02/17/2024] Open
Abstract
Kidney diseases result from various causes, which can generally be divided into neoplastic and non-neoplastic diseases. Deep learning based on medical imaging is an established methodology for further data mining and an evolving field of expertise, which provides the possibility for precise management of kidney diseases. Recently, imaging-based deep learning has been widely applied to many clinical scenarios of kidney diseases including organ segmentation, lesion detection, differential diagnosis, surgical planning, and prognosis prediction, which can provide support for disease diagnosis and management. In this review, we will introduce the basic methodology of imaging-based deep learning and its recent clinical applications in neoplastic and non-neoplastic kidney diseases. Additionally, we further discuss its current challenges and future prospects and conclude that achieving data balance, addressing heterogeneity, and managing data size remain challenges for imaging-based deep learning. Meanwhile, the interpretability of algorithms, ethical risks, and barriers of bias assessment are also issues that require consideration in future development. We hope to provide urologists, nephrologists, and radiologists with clear ideas about imaging-based deep learning and reveal its great potential in clinical practice.Critical relevance statement The wide clinical applications of imaging-based deep learning in kidney diseases can help doctors to diagnose, treat, and manage patients with neoplastic or non-neoplastic renal diseases.Key points• Imaging-based deep learning is widely applied to neoplastic and non-neoplastic renal diseases.• Imaging-based deep learning improves the accuracy of the delineation, diagnosis, and evaluation of kidney diseases.• The small dataset, various lesion sizes, and so on are still challenges for deep learning.
Collapse
Affiliation(s)
- Meng Zhang
- Department of Radiology, West China Hospital, Sichuan University, No. 37 Guoxue Alley, Chengdu, 610041, China
- Medical Equipment Innovation Research Center, West China Hospital, Sichuan University, No. 37 Guoxue Alley, Chengdu, 610041, China
- Med+X Center for Manufacturing, West China Hospital, Sichuan University, No. 37 Guoxue Alley, Chengdu, 610041, China
| | - Zheng Ye
- Department of Radiology, West China Hospital, Sichuan University, No. 37 Guoxue Alley, Chengdu, 610041, China
| | - Enyu Yuan
- Department of Radiology, West China Hospital, Sichuan University, No. 37 Guoxue Alley, Chengdu, 610041, China
| | - Xinyang Lv
- Department of Radiology, West China Hospital, Sichuan University, No. 37 Guoxue Alley, Chengdu, 610041, China
| | - Yiteng Zhang
- Department of Radiology, West China Hospital, Sichuan University, No. 37 Guoxue Alley, Chengdu, 610041, China
| | - Yuqi Tan
- Department of Radiology, West China Hospital, Sichuan University, No. 37 Guoxue Alley, Chengdu, 610041, China
| | - Chunchao Xia
- Department of Radiology, West China Hospital, Sichuan University, No. 37 Guoxue Alley, Chengdu, 610041, China
| | - Jing Tang
- Department of Radiology, West China Hospital, Sichuan University, No. 37 Guoxue Alley, Chengdu, 610041, China.
| | - Jin Huang
- Medical Equipment Innovation Research Center, West China Hospital, Sichuan University, No. 37 Guoxue Alley, Chengdu, 610041, China.
- Med+X Center for Manufacturing, West China Hospital, Sichuan University, No. 37 Guoxue Alley, Chengdu, 610041, China.
| | - Zhenlin Li
- Department of Radiology, West China Hospital, Sichuan University, No. 37 Guoxue Alley, Chengdu, 610041, China.
| |
Collapse
|
4
|
Udzik J, Pacholewicz J, Biskupski A, Safranow K, Wojciechowska-Koszko I, Kwiatkowski P, Roszkowska P, Rogulska K, Dziedziejko V, Marcinowska Z, Kwiatkowski S, Kwiatkowska E. Higher perfusion pressure and pump flow during cardiopulmonary bypass are beneficial for kidney function-a single-centre prospective study. Front Physiol 2024; 15:1257631. [PMID: 38420620 PMCID: PMC10899324 DOI: 10.3389/fphys.2024.1257631] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Accepted: 01/29/2024] [Indexed: 03/02/2024] Open
Abstract
Background: Kidneys play an essential role in the circulatory system, regulating blood pressure and intravascular volume. They are also set on maintaining an adequate filtration pressure in the glomerulus. During the CPB, a decrease in systemic blood pressure and hemoglobin concentration may lead to renal ischemia and subsequent acute kidney injury. Methods: One hundred nine adult patients were prospectively enrolled in this study. The intervention in this study was increasing the flow of the CPB pump to reach the target MAP of > 90 mmHg during the procedure. The control group had a standard pump flow of 2.4 L/min/m2. Results: Standard pump flow of 2.4 L/min/m2 resulted in mean MAP < 90 mmHg during the CPB in most patients in the control group. Maintaining a higher MAP during CPB in this study population did not affect CSA-AKI incidence. However, it increased the intraoperative and postoperative diuresis and decreased renin release associated with CPB. Higher MAP during the CPB did not increase the incidence of cerebrovascular complications after the operation; patients in the highest MAP group had the lowest incidence of postoperative delirium, but the result did not obtain statistical significance. Conclusion: Maintaining MAP > 90 mmHg during the CPB positively impacts intraoperative and postoperative kidney function. It significantly reduces renal hypoperfusion during the procedure compared to MAP < 70 mmHg. MAP > 90 mmHg is safe for the central nervous system, and preliminary results suggest that it may have a beneficial impact on the incidence of postoperative delirium.
Collapse
Affiliation(s)
- Jakub Udzik
- Cardiac Surgery Department, Pomeranian Medical University, Szczecin, Poland
| | - Jerzy Pacholewicz
- Cardiac Surgery Department, Pomeranian Medical University, Szczecin, Poland
| | - Andrzej Biskupski
- Cardiac Surgery Department, Pomeranian Medical University, Szczecin, Poland
| | - Krzysztof Safranow
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University, Szczecin, Poland
| | | | - Paweł Kwiatkowski
- Department of Diagnostic Immunology, Pomeranian Medical University, Szczecin, Poland
| | - Paulina Roszkowska
- Department of Diagnostic Immunology, Pomeranian Medical University, Szczecin, Poland
| | - Karolina Rogulska
- Department of Diagnostic Immunology, Pomeranian Medical University, Szczecin, Poland
| | - Violetta Dziedziejko
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University, Szczecin, Poland
| | - Zuzanna Marcinowska
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University, Szczecin, Poland
| | - Sebastian Kwiatkowski
- Department of Obstetrics and Gynecology, Pomeranian Medical University, Szczecin, Poland
| | - Ewa Kwiatkowska
- Department of Nephrology, Transplantology and Internal Medicine, Pomeranian Medical University, Szczecin, Poland
| |
Collapse
|
5
|
Endo A, Hirose T, Sato S, Ito H, Takahashi C, Ishikawa R, Kamada A, Oba-Yabana I, Kimura T, Takahashi K, Mori T. Sodium glucose cotransporter 2 inhibitor suppresses renal injury in rats with renal congestion. Hypertens Res 2024; 47:33-45. [PMID: 37749334 PMCID: PMC10766540 DOI: 10.1038/s41440-023-01437-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 09/10/2023] [Accepted: 09/10/2023] [Indexed: 09/27/2023]
Abstract
Renal congestion is an issue of cardiorenal syndrome in patients with heart failure. Recent clinical and basic studies suggest a renoprotective potential of sodium-glucose cotransporter (SGLT) 2 inhibitors. However, the effect on renal congestion and its mechanism is not fully understood. Thus, we aimed to clarify the effect of SGLT inhibition in a renal congestion model. Renal congestion was induced in the left kidney of male Sprague-Dawley rats by ligation of the inferior vena cava between the renal veins. The SGLT2 inhibitor tofogliflozin or vehicle was orally administered daily from the day before IVC ligation until two days after surgery. On the third postoperative day, both the right control kidney and the left congested kidney were harvested and analyzed. Kidney weight and water content was increased, and renal injury and fibrosis were observed in the left congested kidney. Kidney weight gain and hydration were improved with tofogliflozin treatment. Additionally, this treatment effectively reduced renal injury and fibrosis, particularly in the renal cortex. SGLT2 expression was observed in the congested kidney, but suppressed in the damaged tubular cells. Molecules associated with inflammation were increased in the congested kidney and reversed by tofogliflozin treatment. Mitochondrial dysfunction provoked by renal congestion was also improved by tofogliflozin treatment. Tofogliflozin protects against renal damage induced by renal congestion. SGLT2 inhibitors could be a candidate strategy for renal impairment associated with heart failure.
Collapse
Affiliation(s)
- Akari Endo
- Division of Nephrology and Endocrinology, Faculty of Medicine, Tohoku Medical and Pharmaceutical University, Sendai, Japan
- Department of Endocrinology and Applied Medical Science, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Takuo Hirose
- Division of Nephrology and Endocrinology, Faculty of Medicine, Tohoku Medical and Pharmaceutical University, Sendai, Japan.
- Department of Endocrinology and Applied Medical Science, Tohoku University Graduate School of Medicine, Sendai, Japan.
- Division of Integrative Renal Replacement Therapy, Faculty of Medicine, Tohoku Medical and Pharmaceutical University, Sendai, Japan.
| | - Shigemitsu Sato
- Division of Integrative Renal Replacement Therapy, Faculty of Medicine, Tohoku Medical and Pharmaceutical University, Sendai, Japan
| | - Hiroki Ito
- Division of Nephrology and Endocrinology, Faculty of Medicine, Tohoku Medical and Pharmaceutical University, Sendai, Japan
- Department of Endocrinology and Applied Medical Science, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Chika Takahashi
- Division of Integrative Renal Replacement Therapy, Faculty of Medicine, Tohoku Medical and Pharmaceutical University, Sendai, Japan
| | - Risa Ishikawa
- Division of Nephrology and Endocrinology, Faculty of Medicine, Tohoku Medical and Pharmaceutical University, Sendai, Japan
| | - Ayaka Kamada
- Division of Nephrology and Endocrinology, Faculty of Medicine, Tohoku Medical and Pharmaceutical University, Sendai, Japan
| | - Ikuko Oba-Yabana
- Division of Nephrology and Endocrinology, Faculty of Medicine, Tohoku Medical and Pharmaceutical University, Sendai, Japan
| | - Tomoyoshi Kimura
- Division of Nephrology and Endocrinology, Faculty of Medicine, Tohoku Medical and Pharmaceutical University, Sendai, Japan
| | - Kazuhiro Takahashi
- Department of Endocrinology and Applied Medical Science, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Takefumi Mori
- Division of Nephrology and Endocrinology, Faculty of Medicine, Tohoku Medical and Pharmaceutical University, Sendai, Japan.
- Division of Integrative Renal Replacement Therapy, Faculty of Medicine, Tohoku Medical and Pharmaceutical University, Sendai, Japan.
| |
Collapse
|
6
|
McLarnon SR. Pathophysiology of Red Blood Cell Trapping in Ischemic Acute Kidney Injury. Compr Physiol 2023; 14:5325-5343. [PMID: 38158367 DOI: 10.1002/cphy.c230010] [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: 01/03/2024]
Abstract
Red blood cell (RBC) trapping describes the accumulation of RBCs in the microvasculature of the kidney outer medulla that occurs following ischemic acute kidney injury (AKI). Despite its prominence in human kidneys following AKI, as well as evidence from experimental models demonstrating that the severity of RBC trapping is directly correlated with renal recovery, to date, RBC trapping has not been a primary focus in understanding the pathogenesis of ischemic kidney injury. New evidence from rodent models suggests that RBC trapping is responsible for much of the tubular injury occurring in the initial hours after kidney reperfusion from ischemia. This early injury appears to result from RBC cytotoxicity and closely reflects the injury profile observed in human kidneys, including sloughing of the medullary tubules and the formation of heme casts in the distal tubules. In this review, we discuss what is currently known about RBC trapping. We conclude that RBC trapping is likely avoidable. The primary causes of RBC trapping are thought to include rheologic alterations, blood coagulation, tubular cell swelling, and increased vascular permeability; however, new data indicate that a mismatch in blood flow between the cortex and medulla where medullary perfusion is maintained during cortical ischemia is also likely critical. The mechanism(s) by which RBC trapping contributes to renal functional decline require more investigation. We propose a renewed focus on the mechanisms mediating RBC trapping, and RBC trapping-associated injury is likely to provide important knowledge for improving AKI outcomes. © 2024 American Physiological Society. Compr Physiol 14:5325-5343, 2024.
Collapse
Affiliation(s)
- Sarah R McLarnon
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| |
Collapse
|
7
|
Udzik J, Pacholewicz J, Biskupski A, Walerowicz P, Januszkiewicz K, Kwiatkowska E. Alterations to Kidney Physiology during Cardiopulmonary Bypass-A Narrative Review of the Literature and Practical Remarks. J Clin Med 2023; 12:6894. [PMID: 37959359 PMCID: PMC10647422 DOI: 10.3390/jcm12216894] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2023] [Accepted: 10/31/2023] [Indexed: 11/15/2023] Open
Abstract
INTRODUCTION According to different authors, cardiac surgery-associated acute kidney injury (CSA-AKI) incidence can be as high as 20-50%. This complication increases postoperative morbidity and mortality and impairs long-term kidney function in some patients. This review aims to summarize current knowledge regarding alterations to renal physiology during cardiopulmonary bypass (CPB) and to discuss possible nephroprotective strategies for cardiac surgeries. Relevant sections: Systemic and renal circulation, Vasoactive drugs, Fluid balance and Osmotic regulation and Inflammatory response. CONCLUSIONS Considering the available scientific evidence, it is concluded that adequate kidney perfusion and fluid balance are the most critical factors determining postoperative kidney function. By adequate perfusion, one should understand perfusion with proper oxygen delivery and sufficient perfusion pressure. Maintaining the fluid balance is imperative for a normal kidney filtration process, which is essential for preserving the intra- and postoperative kidney function. FUTURE DIRECTIONS The review of the available literature regarding kidney function during cardiac surgery revealed a need for a more holistic approach to this subject.
Collapse
Affiliation(s)
- Jakub Udzik
- Department of Cardiac Surgery, Pomeranian Medical University, Powstancow Wielkopolskich 72, 70-111 Szczecin, Poland; (J.P.); (A.B.); (P.W.)
| | - Jerzy Pacholewicz
- Department of Cardiac Surgery, Pomeranian Medical University, Powstancow Wielkopolskich 72, 70-111 Szczecin, Poland; (J.P.); (A.B.); (P.W.)
| | - Andrzej Biskupski
- Department of Cardiac Surgery, Pomeranian Medical University, Powstancow Wielkopolskich 72, 70-111 Szczecin, Poland; (J.P.); (A.B.); (P.W.)
| | - Paweł Walerowicz
- Department of Cardiac Surgery, Pomeranian Medical University, Powstancow Wielkopolskich 72, 70-111 Szczecin, Poland; (J.P.); (A.B.); (P.W.)
| | - Kornelia Januszkiewicz
- Department of Anesthesiology, Intensive Care and Acute Intoxications, Pomeranian Medical University, Powstancow Wielkopolskich 72, 70-111 Szczecin, Poland;
| | - Ewa Kwiatkowska
- Clinical Department of Nephrology, Transplantology and Internal Medicine, Pomeranian Medical University, 70-111 Szczecin, Poland;
| |
Collapse
|
8
|
Tanaka S, Portilla D, Okusa MD. Role of perivascular cells in kidney homeostasis, inflammation, repair and fibrosis. Nat Rev Nephrol 2023; 19:721-732. [PMID: 37608184 DOI: 10.1038/s41581-023-00752-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/19/2023] [Indexed: 08/24/2023]
Abstract
Perivascular niches in the kidney comprise heterogeneous cell populations, including pericytes and fibroblasts, with distinct functions. These perivascular cells have crucial roles in preserving kidney homeostasis as they maintain microvascular networks by stabilizing the vasculature and regulating capillary constriction. A subset of kidney perivascular cells can also produce and secrete erythropoietin; this ability can be enhanced with hypoxia-inducible factor-prolyl hydroxylase inhibitors, which are used to treat anaemia in chronic kidney disease. In the pathophysiological state, kidney perivascular cells contribute to the progression of kidney fibrosis, partly via transdifferentiation into myofibroblasts. Moreover, perivascular cells are now recognized as major innate immune sentinels in the kidney that produce pro-inflammatory cytokines and chemokines following injury. These mediators promote immune cell infiltration, leading to persistent inflammation and progression of kidney fibrosis. The crosstalk between perivascular cells and tubular epithelial, immune and endothelial cells is therefore a key process in physiological and pathophysiological states. Here, we examine the multiple roles of kidney perivascular cells in health and disease, focusing on the latest advances in this field of research.
Collapse
Affiliation(s)
- Shinji Tanaka
- Division of Nephrology and Endocrinology, The University of Tokyo Graduate School of Medicine, Tokyo, Japan.
| | - Didier Portilla
- Division of Nephrology and Center for Immunity, Inflammation, and Regenerative Medicine, University of Virginia, Charlottesville, VA, USA
| | - Mark D Okusa
- Division of Nephrology and Center for Immunity, Inflammation, and Regenerative Medicine, University of Virginia, Charlottesville, VA, USA.
| |
Collapse
|
9
|
Wildman SS, Dunn K, Van Beusecum JP, Inscho EW, Kelley S, Lilley RJ, Cook AK, Taylor KD, Peppiatt-Wildman CM. A novel functional role for the classic CNS neurotransmitters, GABA, glycine, and glutamate, in the kidney: potent and opposing regulators of the renal vasculature. Am J Physiol Renal Physiol 2023; 325:F38-F49. [PMID: 37102686 PMCID: PMC10511176 DOI: 10.1152/ajprenal.00425.2021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 03/29/2023] [Accepted: 04/22/2023] [Indexed: 04/28/2023] Open
Abstract
The presence of a renal GABA/glutamate system has previously been described; however, its functional significance in the kidney remains undefined. We hypothesized, given its extensive presence in the kidney, that activation of this GABA/glutamate system would elicit a vasoactive response from the renal microvessels. The functional data here demonstrate, for the first time, that activation of endogenous GABA and glutamate receptors in the kidney significantly alters microvessel diameter with important implications for influencing renal blood flow. Renal blood flow is regulated in both the renal cortical and medullary microcirculatory beds via diverse signaling pathways. GABA- and glutamate-mediated effects on renal capillaries are strikingly similar to those central to the regulation of central nervous system capillaries, that is, exposing renal tissue to physiological concentrations of GABA, glutamate, and glycine led to alterations in the way that contractile cells, pericytes, and smooth muscle cells, regulate microvessel diameter in the kidney. Since dysregulated renal blood flow is linked to chronic renal disease, alterations in the renal GABA/glutamate system, possibly through prescription drugs, could significantly impact long-term kidney function.NEW & NOTEWORTHY Functional data here offer novel insight into the vasoactive activity of the renal GABA/glutamate system. These data show that activation of endogenous GABA and glutamate receptors in the kidney significantly alters microvessel diameter. Furthermore, the results show that these antiepileptic drugs are as potentially challenging to the kidney as nonsteroidal anti-inflammatory drugs.
Collapse
Affiliation(s)
| | - Kadeshia Dunn
- Division of Natural Sciences, University of Kent, Kent, United Kingdom
| | - Justin P Van Beusecum
- Ralph H. Johnson Veterans Affairs Medical Center, Charleston, South Carolina, United States
- Medical University of South Carolina, Charleston, South Carolina, United States
| | - Edward W Inscho
- Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, United States
| | - Stephen Kelley
- Division of Natural Sciences, University of Kent, Kent, United Kingdom
| | - Rebecca J Lilley
- Division of Natural Sciences, University of Kent, Kent, United Kingdom
| | - Anthony K Cook
- Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, United States
| | - Kirsti D Taylor
- Division of Natural Sciences, University of Kent, Kent, United Kingdom
| | | |
Collapse
|
10
|
Campos Pamplona C, Moers C, Leuvenink HGD, van Leeuwen LL. Expanding the Horizons of Pre-Transplant Renal Vascular Assessment Using Ex Vivo Perfusion. Curr Issues Mol Biol 2023; 45:5437-5459. [PMID: 37504261 PMCID: PMC10378498 DOI: 10.3390/cimb45070345] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Revised: 06/21/2023] [Accepted: 06/27/2023] [Indexed: 07/29/2023] Open
Abstract
Recently, immense efforts have focused on improving the preservation of (sub)optimal donor organs by means of ex vivo perfusion, which enables the opportunity for organ reconditioning and viability assessment. However, there is still no biomarker that correlates with renal viability. Therefore, it is essential to explore new techniques for pre-transplant assessment of organ quality to guarantee successful long-term transplantation outcomes. The renal vascular compartment has received little attention in machine perfusion studies. In vivo, proper renal vascular and endothelial function is essential for maintaining homeostasis and long-term graft survival. In an ex vivo setting, little is known about vascular viability and its implications for an organ's suitability for transplant. Seeing that endothelial damage is the first step in a cascade of disruptions and maintaining homeostasis is crucial for positive post-transplant outcomes, further research is key to clarifying the (patho)physiology of the renal vasculature during machine perfusion. In this review, we aim to summarize key aspects of renal vascular physiology, describe the role of the renal vasculature in pathophysiological settings, and explain how ex vivo perfusion plays a role in either unveiling or targeting such processes. Additionally, we discuss potentially new vascular assessment tools during ex vivo renal perfusion.
Collapse
Affiliation(s)
- Carolina Campos Pamplona
- Department of Surgery-Organ Donation and Transplantation, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands
| | - Cyril Moers
- Department of Surgery-Organ Donation and Transplantation, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands
| | - Henri G D Leuvenink
- Department of Surgery-Organ Donation and Transplantation, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands
| | - L Leonie van Leeuwen
- Department of Surgery-Organ Donation and Transplantation, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands
| |
Collapse
|
11
|
Lilley RJ, Taylor KD, Wildman SSP, Peppiatt-Wildman CM. Inflammatory mediators act at renal pericytes to elicit contraction of vasa recta and reduce pericyte density along the kidney medullary vascular network. Front Physiol 2023; 14:1194803. [PMID: 37362447 PMCID: PMC10288992 DOI: 10.3389/fphys.2023.1194803] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Accepted: 06/01/2023] [Indexed: 06/28/2023] Open
Abstract
Introduction: Regardless of initiating cause, renal injury promotes a potent pro-inflammatory environment in the outer medulla and a concomitant sustained decrease in medullary blood flow (MBF). This decline in MBF is believed to be one of the critical events in the pathogenesis of acute kidney injury (AKI), yet the precise cellular mechanism underlying this are still to be fully elucidated. MBF is regulated by contractile pericyte cells that reside on the descending vasa recta (DVR) capillaries, which are the primary source of blood flow to the medulla. Methods: Using the rat and murine live kidney slice models, we investigated the acute effects of key medullary inflammatory mediators TNF-α, IL-1β, IL-33, IL-18, C3a and C5a on vasa recta pericytes, the effect of AT1-R blocker Losartan on pro-inflammatory mediator activity at vasa recta pericytes, and the effect of 4-hour sustained exposure on immunolabelled NG2+ pericytes. Results and discussion: Exposure of rat and mouse kidney slices to TNF-α, IL-18, IL-33, and C5a demonstrated a real-time pericyte-mediated constriction of DVR. When pro-inflammatory mediators were applied in the presence of Losartan the inflammatory mediator-mediated constriction that had previously been observed was significantly attenuated. When live kidney slices were exposed to inflammatory mediators for 4-h, we noted a significant reduction in the number of NG2+ positive pericytes along vasa recta capillaries in both rat and murine kidney slices. Data collected in this study demonstrate that inflammatory mediators can dysregulate pericytes to constrict DVR diameter and reduce the density of pericytes along vasa recta vessels, further diminishing the regulatory capacity of the capillary network. We postulate that preliminary findings here suggest pericytes play a role in AKI.
Collapse
Affiliation(s)
- Rebecca J. Lilley
- Division of Natural Sciences, University of Kent, Kent, United Kingdom
| | - Kirsti D. Taylor
- Division of Natural Sciences, University of Kent, Kent, United Kingdom
| | | | | |
Collapse
|
12
|
Xia M, Jiao L, Wang XH, Tong M, Yao MD, Li XM, Yao J, Li D, Zhao PQ, Yan B. Single-cell RNA sequencing reveals a unique pericyte type associated with capillary dysfunction. Theranostics 2023; 13:2515-2530. [PMID: 37215579 PMCID: PMC10196835 DOI: 10.7150/thno.83532] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Accepted: 04/10/2023] [Indexed: 05/24/2023] Open
Abstract
Background: Capillary dysfunction has been implicated in a series of life- threatening vascular diseases characterized by pericyte and endothelial cell (EC) degeneration. However, the molecular profiles that govern the heterogeneity of pericytes have not been fully elucidated. Methods: Single-cell RNA sequencing was conducted on oxygen-induced proliferative retinopathy (OIR) model. Bioinformatics analysis was conducted to identify specific pericytes involved in capillary dysfunction. qRT-PCRs and western blots were conducted to detect Col1a1 expression pattern during capillary dysfunction. Matrigel co-culture assays, PI staining, and JC-1 staining was conducted to determine the role of Col1a1 in pericyte biology. IB4 and NG2 staining was conducted to determine the role of Col1a1 in capillary dysfunction. Results: We constructed an atlas of > 76,000 single-cell transcriptomes from 4 mouse retinas, which could be annotated to 10 distinct retinal cell types. Using the sub-clustering analysis, we further characterized retinal pericytes into 3 different subpopulations. Notably, GO and KEGG pathway analysis demonstrated that pericyte sub-population 2 was identified to be vulnerable to retinal capillary dysfunction. Based on the single-cell sequencing results, Col1a1 was identified as a marker gene of pericyte sub-population 2 and a promising therapeutic target for capillary dysfunction. Col1a1 was abundantly expressed in pericytes and its expression was obviously upregulated in OIR retinas. Col1a1 silencing could retard the recruitment of pericytes toward endothelial cells and aggravated hypoxia-induced pericyte apoptosis in vitro. Col1a1 silencing could reduce the size of neovascular area and avascular area in OIR retinas and suppressed pericyte-myofibroblast transition and endothelial-mesenchymal transition. Moreover, Col1a1 expression was up-regulated in the aqueous humor of the patients with proliferative diabetic retinopathy (PDR) or retinopathy of prematurity (ROP) and up-regulated in the proliferative membranes of PDR patients. Conclusions: These findings enhance the understanding of the complexity and heterogeneity of retinal cells and have important implications for future treatment of capillary dysfunction.
Collapse
Affiliation(s)
- Min Xia
- The Fourth School of Clinical Medicine, Nanjing Medical University, Nanjing 210000, China
- The Affiliated Eye Hospital, Nanjing Medical University, Nanjing 210000, China
| | - Lyu Jiao
- Department of Ophthalmology, Xinhua Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai 200092, China
| | - Xiao-Han Wang
- Department of Ophthalmology, Xinhua Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai 200092, China
| | - Min Tong
- Eye Institute, Eye & ENT Hospital, Shanghai Medical College, Fudan University, Shanghai 200030, China
| | - Mu-Di Yao
- Eye Institute, Eye & ENT Hospital, Shanghai Medical College, Fudan University, Shanghai 200030, China
| | - Xiu-Miao Li
- The Affiliated Eye Hospital, Nanjing Medical University, Nanjing 210000, China
| | - Jin Yao
- The Fourth School of Clinical Medicine, Nanjing Medical University, Nanjing 210000, China
- The Affiliated Eye Hospital, Nanjing Medical University, Nanjing 210000, China
| | - Dan Li
- Eye Institute, Eye & ENT Hospital, Shanghai Medical College, Fudan University, Shanghai 200030, China
| | - Pei-Quan Zhao
- Department of Ophthalmology, Xinhua Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai 200092, China
| | - Biao Yan
- Eye Institute, Eye & ENT Hospital, Shanghai Medical College, Fudan University, Shanghai 200030, China
- NHC Key Laboratory of Myopia (Fudan University), Key Laboratory of Myopia, Chinese Academy of Medical Sciences, and Shanghai Key Laboratory of Visual Impairment and Restoration (Fudan University), Shanghai 200030, China
| |
Collapse
|
13
|
Huang R, Fu P, Ma L. Kidney fibrosis: from mechanisms to therapeutic medicines. Signal Transduct Target Ther 2023; 8:129. [PMID: 36932062 PMCID: PMC10023808 DOI: 10.1038/s41392-023-01379-7] [Citation(s) in RCA: 225] [Impact Index Per Article: 112.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2022] [Revised: 02/12/2023] [Accepted: 02/20/2023] [Indexed: 03/19/2023] Open
Abstract
Chronic kidney disease (CKD) is estimated to affect 10-14% of global population. Kidney fibrosis, characterized by excessive extracellular matrix deposition leading to scarring, is a hallmark manifestation in different progressive CKD; However, at present no antifibrotic therapies against CKD exist. Kidney fibrosis is identified by tubule atrophy, interstitial chronic inflammation and fibrogenesis, glomerulosclerosis, and vascular rarefaction. Fibrotic niche, where organ fibrosis initiates, is a complex interplay between injured parenchyma (like tubular cells) and multiple non-parenchymal cell lineages (immune and mesenchymal cells) located spatially within scarring areas. Although the mechanisms of kidney fibrosis are complicated due to the kinds of cells involved, with the help of single-cell technology, many key questions have been explored, such as what kind of renal tubules are profibrotic, where myofibroblasts originate, which immune cells are involved, and how cells communicate with each other. In addition, genetics and epigenetics are deeper mechanisms that regulate kidney fibrosis. And the reversible nature of epigenetic changes including DNA methylation, RNA interference, and chromatin remodeling, gives an opportunity to stop or reverse kidney fibrosis by therapeutic strategies. More marketed (e.g., RAS blockage, SGLT2 inhibitors) have been developed to delay CKD progression in recent years. Furthermore, a better understanding of renal fibrosis is also favored to discover biomarkers of fibrotic injury. In the review, we update recent advances in the mechanism of renal fibrosis and summarize novel biomarkers and antifibrotic treatment for CKD.
Collapse
Affiliation(s)
- Rongshuang Huang
- Kidney Research Institute, Division of Nephrology, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Ping Fu
- Kidney Research Institute, Division of Nephrology, West China Hospital, Sichuan University, Chengdu, 610041, China.
| | - Liang Ma
- Kidney Research Institute, Division of Nephrology, West China Hospital, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
14
|
Heinl ES, Broeker KAE, Lehrmann C, Heydn R, Krieger K, Ortmaier K, Tauber P, Schweda F. Localization of natriuretic peptide receptors A, B, and C in healthy and diseased mouse kidneys. Pflugers Arch 2023; 475:343-360. [PMID: 36480070 PMCID: PMC9908653 DOI: 10.1007/s00424-022-02774-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Revised: 11/16/2022] [Accepted: 11/18/2022] [Indexed: 12/13/2022]
Abstract
The natriuretic peptides (NPs) ANP (atrial natriuretic peptide) and BNP (B-type natriuretic peptide) mediate their widespread effects by activating the natriuretic peptide receptor-A (NPR-A), while C-type natriuretic peptide (CNP) acts via natriuretic peptide receptor-B (NPR-B). NPs are removed from the circulation by internalization via the natriuretic peptide clearance receptor natriuretic peptide receptor-C (NPR-C). In addition to their well-known functions, for instance on blood pressure, all three NPs confer significant cardioprotection and renoprotection. Since neither the NP-mediated renal functions nor the renal target cells of renoprotection are completely understood, we performed systematic localization studies of NP receptors using in situ hybridization (RNAscope) in mouse kidneys. NPR-A mRNA is highly expressed in glomeruli (mainly podocytes), renal arterioles, endothelial cells of peritubular capillaries, and PDGFR-receptor β positive (PDGFR-β) interstitial cells. No NPR-A mRNA was detected by RNAscope in the tubular system. In contrast, NPR-B expression is highest in proximal tubules. NPR-C is located in glomeruli (mainly podocytes), in endothelial cells and PDGFR-β positive cells. To test for a possible regulation of NPRs in kidney diseases, their distribution was studied in adenine nephropathy. Signal intensity of NPR-A and NPR-B mRNA was reduced while their spatial distribution was unaltered compared with healthy kidneys. In contrast, NPR-C mRNA signal was markedly enhanced in cell clusters of myofibroblasts in fibrotic areas of adenine kidneys. In conclusion, the primary renal targets of ANP and BNP are glomerular, vascular, and interstitial cells but not the tubular compartment, while the CNP receptor NPR-B is highly expressed in proximal tubules. Further studies are needed to clarify the function and interplay of this specific receptor expression pattern.
Collapse
Affiliation(s)
- Elena-Sofia Heinl
- Institute for Physiology, University Regensburg, Regensburg, Germany.
| | | | - Claudia Lehrmann
- grid.7727.50000 0001 2190 5763Institute for Physiology, University Regensburg, Regensburg, Germany
| | - Rosmarie Heydn
- grid.7727.50000 0001 2190 5763Institute for Physiology, University Regensburg, Regensburg, Germany
| | - Katharina Krieger
- grid.7727.50000 0001 2190 5763Institute for Physiology, University Regensburg, Regensburg, Germany
| | - Katharina Ortmaier
- grid.7727.50000 0001 2190 5763Institute for Physiology, University Regensburg, Regensburg, Germany
| | - Philipp Tauber
- grid.7727.50000 0001 2190 5763Institute for Physiology, University Regensburg, Regensburg, Germany
| | - Frank Schweda
- Institute for Physiology, University Regensburg, Regensburg, Germany.
| |
Collapse
|
15
|
Inhibition of platelet-derived growth factor pathway suppresses tubulointerstitial injury in renal congestion. J Hypertens 2022; 40:1935-1949. [PMID: 35983805 PMCID: PMC9451920 DOI: 10.1097/hjh.0000000000003191] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
OBJECTIVE Increased central venous pressure in congestive heart failure is responsible for renal dysfunction, which is mediated by renal venous congestion. Pericyte detachment from capillaries after renal congestion might trigger renal fibrogenesis via pericyte-myofibroblast transition (PMT). Platelet-derived growth factor receptors (PDGFRs), which are PMT indicators, were upregulated in our recently established renal congestion model. This study was designed to determine whether inhibition of the PDGFR pathway could suppress tubulointerstitial injury after renal congestion. METHODS The inferior vena cava between the renal veins was ligated in male Sprague-Dawley rats, inducing congestion only in the left kidney. Imatinib mesylate or vehicle were injected intraperitoneally daily from 1 day before the operation. Three days after the surgery, the effect of imatinib was assessed by physiological, morphological and molecular methods. The inhibition of PDGFRs against transforming growth factor-β1 (TGFB1)-induced fibrosis was also tested in human pericyte cell culture. RESULTS Increased kidney weight and renal fibrosis were observed in the congested kidneys. Upstream inferior vena cava (IVC) pressure immediately increased to around 20 mmHg after IVC ligation in both the imatinib and saline groups. Although vasa recta dilatation and pericyte detachment under renal congestion were maintained, imatinib ameliorated the increased kidney weight and suppressed renal fibrosis around the vasa recta. TGFB1-induced elevation of fibrosis markers in human pericytes was suppressed by PDGFR inhibitors at the transcriptional level. CONCLUSION The activation of the PDGFR pathway after renal congestion was responsible for renal congestion-induced fibrosis. This mechanism could be a candidate therapeutic target for renoprotection against renal congestion-induced tubulointerstitial injury.
Collapse
|
16
|
Edwards A, Kurtcuoglu V. Renal blood flow and oxygenation. Pflugers Arch 2022; 474:759-770. [PMID: 35438336 PMCID: PMC9338895 DOI: 10.1007/s00424-022-02690-y] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Revised: 03/19/2022] [Accepted: 03/21/2022] [Indexed: 02/07/2023]
Abstract
Our kidneys receive about one-fifth of the cardiac output at rest and have a low oxygen extraction ratio, but may sustain, under some conditions, hypoxic injuries that might lead to chronic kidney disease. This is due to large regional variations in renal blood flow and oxygenation, which are the prerequisite for some and the consequence of other kidney functions. The concurrent operation of these functions is reliant on a multitude of neuro-hormonal signaling cascades and feedback loops that also include the regulation of renal blood flow and tissue oxygenation. Starting with open questions on regulatory processes and disease mechanisms, we review herein the literature on renal blood flow and oxygenation. We assess the current understanding of renal blood flow regulation, reasons for disparities in oxygen delivery and consumption, and the consequences of disbalance between O2 delivery, consumption, and removal. We further consider methods for measuring and computing blood velocity, flow rate, oxygen partial pressure, and related parameters and point out how limitations of these methods constitute important hurdles in this area of research. We conclude that to obtain an integrated understanding of the relation between renal function and renal blood flow and oxygenation, combined experimental and computational modeling studies will be needed.
Collapse
Affiliation(s)
- Aurelie Edwards
- Department of Biomedical Engineering, Boston University, 44 Cummington Mall, Boston, MA, 02215, USA
| | - Vartan Kurtcuoglu
- Institute of Physiology, University of Zurich, Winterthurerstrasse 190, 8057, Zurich, Switzerland.
- National Center of Competence in Research, Kidney.CH, University of Zurich, Zurich, Switzerland.
- Zurich Center for Integrative Human Physiology, University of Zurich, Zurich, Switzerland.
| |
Collapse
|
17
|
Grgurevic L, Novak R, Salai G, Hrkac S, Mocibob M, Vojtusek IK, Laganovic M. Stage II of Chronic Kidney Disease-A Tipping Point in Disease Progression? Biomedicines 2022; 10:1522. [PMID: 35884827 PMCID: PMC9313233 DOI: 10.3390/biomedicines10071522] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Revised: 06/20/2022] [Accepted: 06/25/2022] [Indexed: 11/17/2022] Open
Abstract
Chronic kidney disease (CKD) is the progressive loss of renal function. Although advances have been made in understanding the progression of CKD, key molecular events in complex pathophysiological mechanisms that mark each stage of renal failure remain largely unknown. Changes in plasma protein profiles in different disease stages are important for identification of early diagnostic markers and potential therapeutic targets. The goal of this study was to determine the molecular profile of each CKD stage (from 1 to 5), aiming to specifically point out markedly expressed or downregulated proteins. We performed a cross-sectional shotgun-proteomic study of pooled plasma across CKD stages and compared them to healthy controls. After sample pooling and heparin-column purification we analysed proteomes from healthy to CKD stage 1 through 5 participants' plasma by liquid-chromatography/mass-spectrometry. We identified 453 proteins across all study groups. Our results indicate that key events, which may later affect the course of disease progression and the overall pathophysiological background, are most pronounced in CKD stage 2, with an emphasis on inflammation, lipoprotein metabolism, angiogenesis and tissue regeneration. We hypothesize that CKD stage 2 is the tipping point in disease progression and a suitable point in disease course for the development of therapeutic solutions.
Collapse
Affiliation(s)
- Lovorka Grgurevic
- Department of Anatomy, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia
- Center for Translational and Clinical Research, Department of Proteomics, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia; (R.N.); (G.S.); (S.H.)
| | - Rudjer Novak
- Center for Translational and Clinical Research, Department of Proteomics, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia; (R.N.); (G.S.); (S.H.)
| | - Grgur Salai
- Center for Translational and Clinical Research, Department of Proteomics, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia; (R.N.); (G.S.); (S.H.)
- Department of Pulmonology, University Hospital Dubrava, 10000 Zagreb, Croatia
| | - Stela Hrkac
- Center for Translational and Clinical Research, Department of Proteomics, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia; (R.N.); (G.S.); (S.H.)
- Department of Emergency Medicine, University Hospital Centre Zagreb, 10000 Zagreb, Croatia
| | - Marko Mocibob
- Department of Chemistry, Faculty of Science, University of Zagreb, 10000 Zagreb, Croatia;
| | - Ivana Kovacevic Vojtusek
- Department of Nephrology, Arterial Hypertension, Dialysis and Transplantation, University Hospital Center Zagreb, 10000 Zagreb, Croatia;
| | - Mario Laganovic
- Department of Nephrology, University Hospital Merkur, 10000 Zagreb, Croatia;
| |
Collapse
|
18
|
Xu M, Lichtenberger FB, Erdoǧan C, Lai E, Persson PB, Patzak A, Khedkar PH. Nitric Oxide Signalling in Descending Vasa Recta after Hypoxia/Re-Oxygenation. Int J Mol Sci 2022; 23:7016. [PMID: 35806018 PMCID: PMC9266395 DOI: 10.3390/ijms23137016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 06/20/2022] [Accepted: 06/21/2022] [Indexed: 01/27/2023] Open
Abstract
Reduced renal medullary oxygen supply is a key factor in the pathogenesis of acute kidney injury (AKI). As the medulla exclusively receives blood through descending vasa recta (DVR), dilating these microvessels after AKI may help in renoprotection by restoring renal medullary blood flow. We stimulated the NO-sGC-cGMP signalling pathway in DVR at three different levels before and after hypoxia/re-oxygenation (H/R). Rat DVR were isolated and perfused under isobaric conditions. The phosphodiesterase 5 (PDE5) inhibitor sildenafil (10-6 mol/L) impaired cGMP degradation and dilated DVR pre-constricted with angiotensin II (Ang II, 10-6 mol/L). Dilations by the soluble guanylyl cyclase (sGC) activator BAY 60-2770 as well as the nitric oxide donor sodium nitroprusside (SNP, 10-3 mol/L) were equally effective. Hypoxia (0.1% O2) augmented DVR constriction by Ang II, thus potentially aggravating tissue hypoxia. H/R left DVR unresponsive to sildenafil, yet sGC activation by BAY 60-2770 effectively dilated DVR. Dilation to SNP under H/R is delayed. In conclusion, H/R renders PDE5 inhibition ineffective in dilating the crucial vessels supplying the area at risk for hypoxic damage. Stimulating sGC appears to be the most effective in restoring renal medullary blood flow after H/R and may prove to be the best target for maintaining oxygenation to this vulnerable area of the kidney.
Collapse
Affiliation(s)
- Minze Xu
- Institute of Translational Physiology, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Charitéplatz 1, 10117 Berlin, Germany; (M.X.); (F.-B.L.); (C.E.); (P.B.P.); (P.H.K.)
| | - Falk-Bach Lichtenberger
- Institute of Translational Physiology, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Charitéplatz 1, 10117 Berlin, Germany; (M.X.); (F.-B.L.); (C.E.); (P.B.P.); (P.H.K.)
| | - Cem Erdoǧan
- Institute of Translational Physiology, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Charitéplatz 1, 10117 Berlin, Germany; (M.X.); (F.-B.L.); (C.E.); (P.B.P.); (P.H.K.)
| | - Enyin Lai
- Department of Physiology, Zhejiang University School of Medicine, Hangzhou 310058, China;
| | - Pontus B. Persson
- Institute of Translational Physiology, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Charitéplatz 1, 10117 Berlin, Germany; (M.X.); (F.-B.L.); (C.E.); (P.B.P.); (P.H.K.)
| | - Andreas Patzak
- Institute of Translational Physiology, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Charitéplatz 1, 10117 Berlin, Germany; (M.X.); (F.-B.L.); (C.E.); (P.B.P.); (P.H.K.)
| | - Pratik H. Khedkar
- Institute of Translational Physiology, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Charitéplatz 1, 10117 Berlin, Germany; (M.X.); (F.-B.L.); (C.E.); (P.B.P.); (P.H.K.)
| |
Collapse
|
19
|
Mediators of Regional Kidney Perfusion during Surgical Pneumo-Peritoneum Creation and the Risk of Acute Kidney Injury—A Review of Basic Physiology. J Clin Med 2022; 11:jcm11102728. [PMID: 35628855 PMCID: PMC9142947 DOI: 10.3390/jcm11102728] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Revised: 05/05/2022] [Accepted: 05/05/2022] [Indexed: 02/04/2023] Open
Abstract
Acute kidney injury (AKI), especially if recurring, represents a risk factor for future chronic kidney disease. In intensive care units, increased intra-abdominal pressure is well-recognized as a significant contributor to AKI. However, the importance of transiently increased intra-abdominal pressures procedures is less commonly appreciated during laparoscopic surgery, the use of which has rapidly increased over the last few decades. Unlike the well-known autoregulation of the renal cortical circulation, medulla perfusion is modulated via partially independent regulatory mechanisms and strongly impacted by changes in venous and lymphatic pressures. In our review paper, we will provide a comprehensive overview of this evolving topic, covering a broad range from basic pathophysiology up to and including current clinical relevance and examples. Key regulators of oxidative stress such as ischemia-reperfusion injury, the activation of inflammatory response and humoral changes interacting with procedural pneumo-peritoneum formation and AKI risk will be recounted. Moreover, we present an in-depth review of the interaction of pneumo-peritoneum formation with general anesthetic agents and animal models of congestive heart failure. A better understanding of the relationship between pneumo-peritoneum formation and renal perfusion will support basic and clinical research, leading to improved clinical care and collaboration among specialists.
Collapse
|
20
|
Andersen SB, Taghavi I, Søgaard SB, Hoyos CAV, Nielsen MB, Jensen JA, Sørensen CM. Super-Resolution Ultrasound Imaging Can Quantify Alterations in Microbubble Velocities in the Renal Vasculature of Rats. Diagnostics (Basel) 2022; 12:1111. [PMID: 35626267 PMCID: PMC9140053 DOI: 10.3390/diagnostics12051111] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Revised: 04/04/2022] [Accepted: 04/27/2022] [Indexed: 11/17/2022] Open
Abstract
Super-resolution ultrasound imaging, based on the localization and tracking of single intravascular microbubbles, makes it possible to map vessels below 100 µm. Microbubble velocities can be estimated as a surrogate for blood velocity, but their clinical potential is unclear. We investigated if a decrease in microbubble velocity in the arterial and venous beds of the renal cortex, outer medulla, and inner medulla was detectable after intravenous administration of the α1-adrenoceptor antagonist prazosin. The left kidneys of seven rats were scanned with super-resolution ultrasound for 10 min before, during, and after prazosin administration using a bk5000 ultrasound scanner and hockey-stick probe. The super-resolution images were manually segmented, separating cortex, outer medulla, and inner medulla. Microbubble tracks from arteries/arterioles were separated from vein/venule tracks using the arterial blood flow direction. The mean microbubble velocities from each scan were compared. This showed a significant prazosin-induced velocity decrease only in the cortical arteries/arterioles (from 1.59 ± 0.38 to 1.14 ± 0.31 to 1.18 ± 0.33 mm/s, p = 0.013) and outer medulla descending vasa recta (from 0.70 ± 0.05 to 0.66 ± 0.04 to 0.69 ± 0.06 mm/s, p = 0.026). Conclusively, super-resolution ultrasound imaging makes it possible to detect and differentiate microbubble velocity responses to prazosin simultaneously in the renal cortical and medullary vascular beds.
Collapse
Affiliation(s)
- Sofie Bech Andersen
- Department of Biomedical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark; (S.B.S.); (C.M.S.)
- Department of Diagnostic Radiology, University Hospital Rigshospitalet, 2100 Copenhagen, Denmark;
| | - Iman Taghavi
- Center for Fast Ultrasound Imaging, Department of Health Technology, Technical University of Denmark, 2800 Lyngby, Denmark; (I.T.); (J.A.J.)
| | - Stinne Byrholdt Søgaard
- Department of Biomedical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark; (S.B.S.); (C.M.S.)
- Department of Diagnostic Radiology, University Hospital Rigshospitalet, 2100 Copenhagen, Denmark;
| | | | - Michael Bachmann Nielsen
- Department of Diagnostic Radiology, University Hospital Rigshospitalet, 2100 Copenhagen, Denmark;
- Department of Clinical Medicine, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Jørgen Arendt Jensen
- Center for Fast Ultrasound Imaging, Department of Health Technology, Technical University of Denmark, 2800 Lyngby, Denmark; (I.T.); (J.A.J.)
| | - Charlotte Mehlin Sørensen
- Department of Biomedical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark; (S.B.S.); (C.M.S.)
| |
Collapse
|
21
|
Lim JH, Yook JM, Oh SH, Jeon SJ, Noh HW, Jung HY, Choi JY, Cho JH, Kim CD, Kim YL, Park SH. Paricalcitol Improves Hypoxia-Induced and TGF-β1-Induced Injury in Kidney Pericytes. Int J Mol Sci 2021; 22:ijms22189751. [PMID: 34575914 PMCID: PMC8472327 DOI: 10.3390/ijms22189751] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Revised: 09/05/2021] [Accepted: 09/06/2021] [Indexed: 11/16/2022] Open
Abstract
Recently, the role of kidney pericytes in kidney fibrosis has been investigated. This study aims to evaluate the effect of paricalcitol on hypoxia-induced and TGF-β1-induced injury in kidney pericytes. The primary cultured pericytes were pretreated with paricalcitol (20 ng/mL) for 90 min before inducing injury, and then they were exposed to TGF-β1 (5 ng/mL) or hypoxia (1% O2 and 5% CO2). TGF-β1 increased α-SMA and other fibrosis markers but reduced PDGFRβ expression in pericytes, whereas paricalcitol reversed the changes. Paricalcitol inhibited the TGF-β1-induced cell migration of pericytes. Hypoxia increased TGF-β1, α-SMA and other fibrosis markers but reduced PDGFRβ expression in pericyte, whereas paricalcitol reversed them. Hypoxia activated the HIF-1α and downstream molecules including prolyl hydroxylase 3 and glucose transporter-1, whereas paricalcitol attenuated the activation of the HIF-1α-dependent molecules and TGF-β1/Smad signaling pathways in hypoxic pericytes. The gene silencing of HIF-1α vanished the hypoxia-induced TGF-β1, α-SMA upregulation, and PDGFRβ downregulation. The effect of paricalcitol on the HIF-1α-dependent changes of fibrosis markers was not significant after the gene silencing of HIF-1α. In addition, hypoxia aggravated the oxidative stress in pericytes, whereas paricalcitol reversed the oxidative stress by increasing the antioxidant enzymes in an HIF-1α-independent manner. In conclusion, paricalcitol improved the phenotype changes of pericyte to myofibroblast in TGF-β1-stimulated pericytes. In addition, paricalcitol improved the expression of fibrosis markers in hypoxia-exposed pericytes both in an HIF-1α-dependent and independent manner.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - Sun-Hee Park
- Correspondence: ; Tel.: +82-53-200-5547; Fax: +82-53-426-9464
| |
Collapse
|
22
|
Zehra T, Cupples WA, Braam B. Tubuloglomerular Feedback Synchronization in Nephrovascular Networks. J Am Soc Nephrol 2021; 32:1293-1304. [PMID: 33833078 PMCID: PMC8259654 DOI: 10.1681/asn.2020040423] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
To perform their functions, the kidneys maintain stable blood perfusion in the face of fluctuations in systemic BP. This is done through autoregulation of blood flow by the generic myogenic response and the kidney-specific tubuloglomerular feedback (TGF) mechanism. The central theme of this paper is that, to achieve autoregulation, nephrons do not work as single units to manage their individual blood flows, but rather communicate electrically over long distances to other nephrons via the vascular tree. Accordingly, we define the nephrovascular unit (NVU) to be a structure consisting of the nephron, glomerulus, afferent arteriole, and efferent arteriole. We discuss features that require and enable distributed autoregulation mediated by TGF across the kidney. These features include the highly variable topology of the renal vasculature which creates variability in circulation and the potential for mismatch between tubular oxygen demand and delivery; the self-sustained oscillations in each NVU arising from the autoregulatory mechanisms; and the presence of extensive gap junctions formed by connexins and their properties that enable long-distance transmission of TGF signals. The existence of TGF synchronization across the renal microvascular network enables an understanding of how NVUs optimize oxygenation-perfusion matching while preventing transmission of high systemic pressure to the glomeruli, which could lead to progressive glomerular and vascular injury.
Collapse
Affiliation(s)
- Tayyaba Zehra
- Department of Medicine, University of Alberta, Edmonton, Alberta, Canada
| | - William A. Cupples
- Department of Physiology and Kinesiology, Simon Fraser University, Burnaby, British Columbia, Canada
| | - Branko Braam
- Department of Medicine, University of Alberta, Edmonton, Alberta, Canada,Department of Physiology, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
23
|
Schrankl J, Fuchs M, Broeker K, Daniel C, Kurtz A, Wagner C. Localization of angiotensin II type 1 receptor gene expression in rodent and human kidneys. Am J Physiol Renal Physiol 2021; 320:F644-F653. [PMID: 33615887 DOI: 10.1152/ajprenal.00550.2020] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
The kidneys are an important target for angiotensin II (ANG II). In adult kidneys, the effects of ANG II are mediated mainly by ANG II type 1 (AT1) receptors. AT1 receptor expression has been reported for a variety of different cell types within the kidneys, suggesting a broad spectrum of actions for ANG II. Since there have been heterogeneous results in the literature regarding the intrarenal distribution of AT1 receptors, this study aimed to obtain a comprehensive overview about the localization of AT1 receptor expression in mouse, rat, and human kidneys. Using the cell-specific and high-resolution RNAscope technique, we performed colocalization experiments with various cell markers to specifically discriminate between different segments of the tubular and vascular system. Overall, we found a similar pattern of AT1 mRNA expression in mouse, rat, and human kidneys. AT1 receptors were detected in mesangial cells and renin-producing cells. In addition, AT1 mRNA was found in interstitial cells of the cortex and outer medulla. In rodents, late afferent and early efferent arterioles expressed AT1 receptor mRNA, but larger vessels of the investigated species showed no AT1 expression. Tubular expression of AT1 mRNA was species dependent with a strong expression in proximal tubules of mice, whereas expression was undetectable in human tubular cells. These findings suggest that the (juxta)glomerular area and tubulointerstitium are conserved expression sites for AT1 receptors across species and might present the main target sites for ANG II in adult human and rodent kidneys.NEW & NOTEWORTHY Angiotensin II (ANG II) type 1 (AT1) receptors are essential for mediating the effects of ANG II in the kidneys. This study aimed to obtain a comprehensive overview about the cell-specific localization of AT1 receptor expression in rodent and human kidneys using the novel RNAscope technique. We found that the conserved AT1 receptor mRNA expression sites across species are the (juxta)glomerular areas and tubulointerstitium, which might present main target sites for ANG II in adult human and rodent kidneys.
Collapse
Affiliation(s)
- Julia Schrankl
- Institute of Physiology, University of Regensburg, Regensburg, Germany
| | - Michaela Fuchs
- Institute of Physiology, University of Regensburg, Regensburg, Germany
| | - Katharina Broeker
- Institute of Physiology, University of Regensburg, Regensburg, Germany
| | - Christoph Daniel
- Department of Nephropathology, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, Germany
| | - Armin Kurtz
- Institute of Physiology, University of Regensburg, Regensburg, Germany
| | - Charlotte Wagner
- Institute of Physiology, University of Regensburg, Regensburg, Germany
| |
Collapse
|
24
|
Meijer EM, van Dijk CGM, Kramann R, Verhaar MC, Cheng C. Implementation of Pericytes in Vascular Regeneration Strategies. TISSUE ENGINEERING PART B-REVIEWS 2021; 28:1-21. [PMID: 33231500 DOI: 10.1089/ten.teb.2020.0229] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
For the survival and integration of complex large-sized tissue-engineered (TE) organ constructs that exceed the maximal nutrients and oxygen diffusion distance required for cell survival, graft (pre)vascularization to ensure medium or blood supply is crucial. To achieve this, the morphology and functionality of the microcapillary bed should be mimicked by incorporating vascular cell populations, including endothelium and mural cells. Pericytes play a crucial role in microvascular function, blood vessel stability, angiogenesis, and blood pressure regulation. In addition, tissue-specific pericytes are important in maintaining specific functions in different organs, including vitamin A storage in the liver, renin production in the kidneys and maintenance of the blood-brain-barrier. Together with their multipotential differentiation capacity, this makes pericytes the preferred cell type for application in TE grafts. The use of a tissue-specific pericyte cell population that matches the TE organ may benefit organ function. In this review, we provide an overview of the literature for graft (pre)-vascularization strategies and highlight the possible advantages of using tissue-specific pericytes for specific TE organ grafts. Impact statement The use of a tissue-specific pericyte cell population that matches the tissue-engineered (TE) organ may benefit organ function. In this review, we provide an overview of the literature for graft (pre)vascularization strategies and highlight the possible advantages of using tissue-specific pericytes for specific TE organ grafts.
Collapse
Affiliation(s)
- Elana M Meijer
- Department of Nephrology and Hypertension, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Christian G M van Dijk
- Department of Nephrology and Hypertension, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Rafael Kramann
- Division of Nephrology and Institute of Experimental Medicine and Systems Biology, University Hospital RWTH Aachen, Aachen, Germany.,Department of Internal Medicine, Nephrology and Transplantation, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Marianne C Verhaar
- Department of Nephrology and Hypertension, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Caroline Cheng
- Department of Nephrology and Hypertension, University Medical Center Utrecht, Utrecht, The Netherlands.,Experimental Cardiology, Department of Cardiology, Thorax Center Erasmus University Medical Center, Rotterdam, The Netherlands
| |
Collapse
|
25
|
Stabinska J, Ljimani A, Zöllner HJ, Wilken E, Benkert T, Limberg J, Esposito I, Antoch G, Wittsack HJ. Spectral diffusion analysis of kidney intravoxel incoherent motion MRI in healthy volunteers and patients with renal pathologies. Magn Reson Med 2021; 85:3085-3095. [PMID: 33462838 DOI: 10.1002/mrm.28631] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Revised: 10/22/2020] [Accepted: 11/12/2020] [Indexed: 12/12/2022]
Abstract
PURPOSE To assess the feasibility of measuring tubular and vascular signal fractions in the human kidney using nonnegative least-square (NNLS) analysis of intravoxel incoherent motion data collected in healthy volunteers and patients with renal pathologies. METHODS MR imaging was performed at 3 Tesla in 12 healthy subjects and 3 patients with various kidney pathologies (fibrotic kidney disease, failed renal graft, and renal masses). Relative signal fractions f and mean diffusivities of the diffusion components in the cortex, medulla, and renal lesions were obtained using the regularized NNLS fitting of the intravoxel incoherent motion data. Test-retest repeatability of the NNLS approach was tested in 5 volunteers scanned twice. RESULTS In the healthy kidneys, the NNLS method yielded diffusion spectra with 3 distinguishable components that may be linked to the slow tissue water diffusion, intermediate tubular and vascular flow, and fast blood flow in larger vessels with the relative signal fractions, fslow , finterm and ffast , respectively. In the pathological kidneys, the diffusion spectra varied substantially from those acquired in the healthy kidneys. Overall, the renal cyst showed substantially higher finterm and lower fslow , whereas the fibrotic kidney, failed renal graft, and renal cell carcinoma demonstrated the opposite trend. CONCLUSION NNLS-based intravoxel incoherent motion could potentially become a valuable tool in assessing changes in tubular and vascular volume fractions under pathophysiological conditions.
Collapse
Affiliation(s)
- Julia Stabinska
- Department of Diagnostic and Interventional Radiology, Medical Faculty, Heinrich Heine University Dusseldorf, Düsseldorf, Germany
| | - Alexandra Ljimani
- Department of Diagnostic and Interventional Radiology, Medical Faculty, Heinrich Heine University Dusseldorf, Düsseldorf, Germany
| | - Helge Jörn Zöllner
- Department of Diagnostic and Interventional Radiology, Medical Faculty, Heinrich Heine University Dusseldorf, Düsseldorf, Germany.,Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD, USA.,F. M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, MD, USA
| | - Enrica Wilken
- Department of Diagnostic and Interventional Radiology, Medical Faculty, Heinrich Heine University Dusseldorf, Düsseldorf, Germany
| | - Thomas Benkert
- MR Application Development, Siemens Healthcare GmbH, Erlangen, Germany
| | - Juliane Limberg
- Institute of Pathology, Medical Faculty, Heinrich Heine University Dusseldorf, Düsseldorf, Germany
| | - Irene Esposito
- Institute of Pathology, Medical Faculty, Heinrich Heine University Dusseldorf, Düsseldorf, Germany
| | - Gerald Antoch
- Department of Diagnostic and Interventional Radiology, Medical Faculty, Heinrich Heine University Dusseldorf, Düsseldorf, Germany
| | - Hans-Jörg Wittsack
- Department of Diagnostic and Interventional Radiology, Medical Faculty, Heinrich Heine University Dusseldorf, Düsseldorf, Germany
| |
Collapse
|
26
|
Ku MC, Fernández-Seara MA, Kober F, Niendorf T. Noninvasive Renal Perfusion Measurement Using Arterial Spin Labeling (ASL) MRI: Basic Concept. Methods Mol Biol 2021; 2216:229-239. [PMID: 33476003 PMCID: PMC9703206 DOI: 10.1007/978-1-0716-0978-1_13] [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] [Indexed: 06/12/2023]
Abstract
The kidney is a complex organ involved in the excretion of metabolic products as well as the regulation of body fluids, osmolarity, and homeostatic status. These functions are influenced in large part by alterations in the regional distribution of blood flow between the renal cortex and medulla. Renal perfusion is therefore a key determinant of glomerular filtration. Therefore the quantification of regional renal perfusion could provide important insights into renal function and renal (patho)physiology. Arterial spin labeling (ASL) based perfusion MRI techniques, can offer a noninvasive and reproducible way of measuring renal perfusion in animal models. This chapter addresses the basic concept of ASL-MRI.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 two separate chapters describing the experimental procedure and data analysis.
Collapse
Affiliation(s)
- Min-Chi Ku
- Berlin Ultrahigh Field Facility (B.U.F.F.), Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | | | - Frank Kober
- Aix-Marseille Université, CNRS, Centre de Résonance Magnétique Biologique et Médicale (CRMBM), Marseille, France
| | - Thoralf Niendorf
- Berlin Ultrahigh Field Facility (B.U.F.F.), Max Delbrück Center for Molecular Medicine (MDC) in the Helmholtz Association, Berlin, Germany
| |
Collapse
|
27
|
Kim HJ, Kim SH, Kim M, Baik H, Park SJ, Kang MS, Kim DH, Kim BW, Markowitz SD, Bae KB. Inhibition of 15-PGDH prevents ischemic renal injury by the PGE 2/EP 4 signaling pathway mediating vasodilation, increased renal blood flow, and increased adenosine/A 2A receptors. Am J Physiol Renal Physiol 2020; 319:F1054-F1066. [PMID: 33135478 DOI: 10.1152/ajprenal.00103.2020] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
In the present study, we demonstrated the marked activity of SW033291, an inhibitor of 15-hydoxyprostaglandin dehydrogenase (15-PGDH), in preventing acute kidney injury (AKI) in a murine model of ischemia-reperfusion injury. AKI due to ischemic injury represents a significant clinical problem. PGE2 is vasodilatory in the kidney, but it is rapidly degraded in vivo due to catabolism by 15-PGDH. We investigated the potential of SW033291, a potent and specific 15-PGDH inhibitor, as prophylactic treatment for ischemic AKI. Prophylactic administration of SW033291 significantly increased renal tissue PGE2 levels and increased post-AKI renal blood flow and renal arteriole area. In parallel, prophylactic SW033291 decreased post-AKI renal morphology injury scores and tubular apoptosis and markedly reduced biomarkers of renal injury that included blood urea nitrogen, creatinine, neutrophil gelatinase-associated lipocalin, and kidney injury molecule-1. Prophylactic SW033291 also reduced post-AKI induction of proinflammatory cytokines, high-mobility group box 1, and malondialdehyde. Protective effects of SW033291 were mediated by PGE2 signaling, as they could be blocked by pharmacological inhibition of PGE2 synthesis. Consistent with activation of PGE2 signaling, SW033291 induced renal levels of both EP4 receptors and cAMP, along with other vasodilatory effectors, including AMP, adenosine, and the adenosine A2A receptor. The protective effects of SW0333291 could largely be achieved with a single prophylactic dose of the drug. Inhibition of 15-PGDH may thus represent a novel strategy for prophylaxis of ischemic AKI in multiple clinical settings, including renal transplantation and cardiovascular surgery.
Collapse
Affiliation(s)
- Hye Jung Kim
- Department of Surgery, Busan Paik Hospital, Inje University College of Medicine, Busan, Republic of Korea.,Paik Institute for Clinical Research, Inje University College of Medicine, Busan, Republic of Korea
| | - Sun-Hee Kim
- Department of Surgery, Busan Paik Hospital, Inje University College of Medicine, Busan, Republic of Korea.,In-Dang Bio Medical Research Institute, Busan Paik Hospital, Inje University College of Medicine, Busan, Republic of Korea
| | - Minjung Kim
- Department of Surgery, Busan Paik Hospital, Inje University College of Medicine, Busan, Republic of Korea.,Paik Institute for Clinical Research, Inje University College of Medicine, Busan, Republic of Korea
| | - HyungJoo Baik
- Department of Surgery, Busan Paik Hospital, Inje University College of Medicine, Busan, Republic of Korea
| | - Seok Ju Park
- Paik Institute for Clinical Research, Inje University College of Medicine, Busan, Republic of Korea.,Department of Nephrology, Busan Paik Hospital, Inje University College of Medicine, Busan, Republic of Korea
| | - Mi Seon Kang
- Department of Pathology, Inje University College of Medicine, Busan, Republic of Korea
| | - Dong-Hyun Kim
- Department of Pharmacology, Inje University College of Medicine, Busan, Republic of Korea
| | - Byeong Woo Kim
- Department of Nephrology, Haeundae Bumin Hospital, Busan, Republic of Korea
| | - Sanford D Markowitz
- Department of Medicine, Case Western Reserve University, and Seidman Cancer Center, University Hospitals, Cleveland, Ohio
| | - Ki Beom Bae
- Department of Surgery, Busan Paik Hospital, Inje University College of Medicine, Busan, Republic of Korea.,Paik Institute for Clinical Research, Inje University College of Medicine, Busan, Republic of Korea
| |
Collapse
|
28
|
Vlachopanos G, Schizas D, Hasemaki N, Georgalis A. Pathophysiology of Contrast-Induced Acute Kidney Injury (CIAKI). Curr Pharm Des 2020; 25:4642-4647. [PMID: 31820694 DOI: 10.2174/1381612825666191210152944] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Accepted: 11/06/2020] [Indexed: 12/24/2022]
Abstract
Contrast-induced acute kidney injury (CIAKI) is a severe complication associated with the use of iodinated contrast media (CM); a sudden but potentially reversible fall in glomerular filtration rate (GFR) typically occurring 48-72 hours after CM administration. Principal risk factors related with the presentation of CIAKI are preexisting chronic kidney disease and diabetes mellitus. Studies on CIAKI present considerable complexity because of differences in CM type and dose, controversies in definition and baseline comorbidities. Despite that, it should be noted that CIAKI poses a serious health problem because it is a very common cause of hospitalacquired AKI, linked to increased morbidity and mortality and utilizing growing healthcare resources. The pathogenesis of CIAKI is heterogeneous and, thus, is incompletely understood. Three basic mechanisms appear to simultaneously occur for CIAKI development: Renal vasoconstriction and medullary hypoxia, tubular cell toxicity and reactive oxygen species formation. The relative contribution of each one of these mechanisms is unknown but they ultimately lead to epithelial and endothelial cell apoptosis and GFR reduction. Further research is needed in order to better clarify CIAKI pathophysiology and accordingly introduce effective preventive and therapeutic strategies.
Collapse
Affiliation(s)
- Georgios Vlachopanos
- Department of Nephrology, Rethymno General Hospital, 19-21 Triantallidou Str., 74100 Rethymn, Greece
| | - Dimitrios Schizas
- First Department of Surgery, National and Kapodistrian University of Athens, Laikon General Hospital, 17 Agiou Thoma Str., 11527 Athens, Greece
| | - Natasha Hasemaki
- First Department of Surgery, National and Kapodistrian University of Athens, Laikon General Hospital, 17 Agiou Thoma Str., 11527 Athens, Greece
| | - Argyrios Georgalis
- Clinic for Transplantation Immunology and Nephrology, University Hospital Basel, Petersgraben 4, 4031 Basel, Switzerland
| |
Collapse
|
29
|
Vallon V, Unwin R, Inscho EW, Leipziger J, Kishore BK. Extracellular Nucleotides and P2 Receptors in Renal Function. Physiol Rev 2019; 100:211-269. [PMID: 31437091 DOI: 10.1152/physrev.00038.2018] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The understanding of the nucleotide/P2 receptor system in the regulation of renal hemodynamics and transport function has grown exponentially over the last 20 yr. This review attempts to integrate the available data while also identifying areas of missing information. First, the determinants of nucleotide concentrations in the interstitial and tubular fluids of the kidney are described, including mechanisms of cellular release of nucleotides and their extracellular breakdown. Then the renal cell membrane expression of P2X and P2Y receptors is discussed in the context of their effects on renal vascular and tubular functions. Attention is paid to effects on the cortical vasculature and intraglomerular structures, autoregulation of renal blood flow, tubuloglomerular feedback, and the control of medullary blood flow. The role of the nucleotide/P2 receptor system in the autocrine/paracrine regulation of sodium and fluid transport in the tubular and collecting duct system is outlined together with its role in integrative sodium and fluid homeostasis and blood pressure control. The final section summarizes the rapidly growing evidence indicating a prominent role of the extracellular nucleotide/P2 receptor system in the pathophysiology of the kidney and aims to identify potential therapeutic opportunities, including hypertension, lithium-induced nephropathy, polycystic kidney disease, and kidney inflammation. We are only beginning to unravel the distinct physiological and pathophysiological influences of the extracellular nucleotide/P2 receptor system and the associated therapeutic perspectives.
Collapse
Affiliation(s)
- Volker Vallon
- Departments of Medicine and Pharmacology, University of California San Diego & VA San Diego Healthcare System, San Diego, California; Centre for Nephrology, Division of Medicine, University College London, London, United Kingdom; IMED ECD CVRM R&D, AstraZeneca, Gothenburg, Sweden; Department of Medicine, Division of Nephrology, The University of Alabama at Birmingham, Birmingham, Alabama; Department of Biomedicine/Physiology, Aarhus University, Aarhus, Denmark; Departments of Internal Medicine and Nutrition and Integrative Physiology, and Center on Aging, University of Utah Health & Nephrology Research, VA Salt Lake City Healthcare System, Salt Lake City, Utah
| | - Robert Unwin
- Departments of Medicine and Pharmacology, University of California San Diego & VA San Diego Healthcare System, San Diego, California; Centre for Nephrology, Division of Medicine, University College London, London, United Kingdom; IMED ECD CVRM R&D, AstraZeneca, Gothenburg, Sweden; Department of Medicine, Division of Nephrology, The University of Alabama at Birmingham, Birmingham, Alabama; Department of Biomedicine/Physiology, Aarhus University, Aarhus, Denmark; Departments of Internal Medicine and Nutrition and Integrative Physiology, and Center on Aging, University of Utah Health & Nephrology Research, VA Salt Lake City Healthcare System, Salt Lake City, Utah
| | - Edward W Inscho
- Departments of Medicine and Pharmacology, University of California San Diego & VA San Diego Healthcare System, San Diego, California; Centre for Nephrology, Division of Medicine, University College London, London, United Kingdom; IMED ECD CVRM R&D, AstraZeneca, Gothenburg, Sweden; Department of Medicine, Division of Nephrology, The University of Alabama at Birmingham, Birmingham, Alabama; Department of Biomedicine/Physiology, Aarhus University, Aarhus, Denmark; Departments of Internal Medicine and Nutrition and Integrative Physiology, and Center on Aging, University of Utah Health & Nephrology Research, VA Salt Lake City Healthcare System, Salt Lake City, Utah
| | - Jens Leipziger
- Departments of Medicine and Pharmacology, University of California San Diego & VA San Diego Healthcare System, San Diego, California; Centre for Nephrology, Division of Medicine, University College London, London, United Kingdom; IMED ECD CVRM R&D, AstraZeneca, Gothenburg, Sweden; Department of Medicine, Division of Nephrology, The University of Alabama at Birmingham, Birmingham, Alabama; Department of Biomedicine/Physiology, Aarhus University, Aarhus, Denmark; Departments of Internal Medicine and Nutrition and Integrative Physiology, and Center on Aging, University of Utah Health & Nephrology Research, VA Salt Lake City Healthcare System, Salt Lake City, Utah
| | - Bellamkonda K Kishore
- Departments of Medicine and Pharmacology, University of California San Diego & VA San Diego Healthcare System, San Diego, California; Centre for Nephrology, Division of Medicine, University College London, London, United Kingdom; IMED ECD CVRM R&D, AstraZeneca, Gothenburg, Sweden; Department of Medicine, Division of Nephrology, The University of Alabama at Birmingham, Birmingham, Alabama; Department of Biomedicine/Physiology, Aarhus University, Aarhus, Denmark; Departments of Internal Medicine and Nutrition and Integrative Physiology, and Center on Aging, University of Utah Health & Nephrology Research, VA Salt Lake City Healthcare System, Salt Lake City, Utah
| |
Collapse
|
30
|
Castellano G, Stasi A, Franzin R, Sallustio F, Divella C, Spinelli A, Netti GS, Fiaccadori E, Cantaluppi V, Crovace A, Staffieri F, Lacitignola L, Grandaliano G, Simone S, Pertosa GB, Gesualdo L. LPS-Binding Protein Modulates Acute Renal Fibrosis by Inducing Pericyte-to-Myofibroblast Trans-Differentiation through TLR-4 Signaling. Int J Mol Sci 2019; 20:ijms20153682. [PMID: 31357597 PMCID: PMC6696277 DOI: 10.3390/ijms20153682] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Revised: 07/23/2019] [Accepted: 07/24/2019] [Indexed: 01/10/2023] Open
Abstract
During sepsis, the increased synthesis of circulating lipopolysaccharide (LPS)-binding protein (LBP) activates LPS/TLR4 signaling in renal resident cells, leading to acute kidney injury (AKI). Pericytes are the major source of myofibroblasts during chronic kidney disease (CKD), but their involvement in AKI is poorly understood. Here, we investigate the occurrence of pericyte-to-myofibroblast trans-differentiation (PMT) in sepsis-induced AKI. In a swine model of sepsis-induced AKI, PMT was detected within 9 h from LPS injection, as evaluated by the reduction of physiologic PDGFRβ expression and the dysfunctional α-SMA increase in peritubular pericytes. The therapeutic intervention by citrate-based coupled plasma filtration adsorption (CPFA) significantly reduced LBP, TGF-β, and endothelin-1 (ET-1) serum levels, and furthermore preserved PDGFRβ and decreased α-SMA expression in renal biopsies. In vitro, both LPS and septic sera led to PMT with a significant increase in Collagen I synthesis and α-SMA reorganization in contractile fibers by both SMAD2/3-dependent and -independent TGF-β signaling. Interestingly, the removal of LBP from septic plasma inhibited PMT. Finally, LPS-stimulated pericytes secreted LBP and TGF-β and underwent PMT also upon TGF-β receptor-blocking, indicating the crucial pro-fibrotic role of TLR4 signaling. Our data demonstrate that the selective removal of LBP may represent a therapeutic option to prevent PMT and the development of acute renal fibrosis in sepsis-induced AKI.
Collapse
Affiliation(s)
- Giuseppe Castellano
- Nephrology, Dialysis and Transplantation Unit, Department of Emergency and Organ Transplantation, University of Bari, 70124 Bari, Italy.
| | - Alessandra Stasi
- Nephrology, Dialysis and Transplantation Unit, Department of Emergency and Organ Transplantation, University of Bari, 70124 Bari, Italy
| | - Rossana Franzin
- Nephrology, Dialysis and Transplantation Unit, Department of Emergency and Organ Transplantation, University of Bari, 70124 Bari, Italy
| | - Fabio Sallustio
- Nephrology, Dialysis and Transplantation Unit, Department of Emergency and Organ Transplantation, University of Bari, 70124 Bari, Italy
- Department of Basic Medical Sciences, Neuroscience and Sense Organs, University of Bari, 70124 Bari, Italy
| | - Chiara Divella
- Nephrology, Dialysis and Transplantation Unit, Department of Emergency and Organ Transplantation, University of Bari, 70124 Bari, Italy
| | - Alessandra Spinelli
- Nephrology, Dialysis and Transplantation Unit, Department of Emergency and Organ Transplantation, University of Bari, 70124 Bari, Italy
| | - Giuseppe Stefano Netti
- Nephrology, Dialysis and Transplantation Unit, Department of Medical and Surgical Sciences, University of Foggia, 71122 Foggia, Italy
| | - Enrico Fiaccadori
- Nephrology Unit, Department of Medicine and Surgery, University of Parma, 43121 Parma, Italy
| | - Vincenzo Cantaluppi
- Department of Translational Medicine, University of Piemonte Orientale, 28100 Novara, Italy
| | - Antonio Crovace
- Veterinary Surgery Unit, Department of Emergency and Organ Transplantation, University of Bari, 70010 Bari, Italy
| | - Francesco Staffieri
- Veterinary Surgery Unit, Department of Emergency and Organ Transplantation, University of Bari, 70010 Bari, Italy
| | - Luca Lacitignola
- Veterinary Surgery Unit, Department of Emergency and Organ Transplantation, University of Bari, 70010 Bari, Italy
| | - Giuseppe Grandaliano
- Nephrology, Dialysis and Transplantation Unit, Department of Medical and Surgical Sciences, University of Foggia, 71122 Foggia, Italy
| | - Simona Simone
- Nephrology, Dialysis and Transplantation Unit, Department of Emergency and Organ Transplantation, University of Bari, 70124 Bari, Italy
| | - Giovanni Battista Pertosa
- Nephrology, Dialysis and Transplantation Unit, Department of Emergency and Organ Transplantation, University of Bari, 70124 Bari, Italy
| | - Loreto Gesualdo
- Nephrology, Dialysis and Transplantation Unit, Department of Emergency and Organ Transplantation, University of Bari, 70124 Bari, Italy
| |
Collapse
|
31
|
Wang Y, Zhang X, Hua Z, Xie L, Jiang X, Wang R, Gao P, Ren K. Blood Oxygen Level-Dependent Imaging and Intravoxel Incoherent Motion MRI of Early Contrast-Induced Acute Kidney Injury in a Rabbit Model. Kidney Blood Press Res 2019; 44:496-512. [PMID: 31256149 DOI: 10.1159/000500052] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Accepted: 04/01/2019] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND To evaluate the application of blood oxygenation level-dependent (BOLD)imaging and intravoxel incoherent motion (IVIM) magnetic resonance imaging (MRI) on assessing early contrast-induced acute kidney injury (CIAKI). MATERIALS Sixty rabbits were randomly chosen to undergo iohexol (1.0, 2.5, and 5.0 [gI/kg], respectively; n = 15 for each group) or saline injection (n = 15). In each group, 6 rabbits underwent MRI at 24 h before injection and after injection of iohexol or saline (1 h and 1, 2, 3, and 4 days); meanwhile, out of the remaining 9 rabbits, 3 were chosen for MRI acquisition, and then they were killed at specific time points (1 h, 1 day, and 3 days, respectively). RESULTS The strong attenuation of pure molecular diffusion (D), apparent diffusion coefficient (ADC), and perfusion fraction (f) was observed at 1 day, while pseudodiffusion coefficient (D*) showed a significant decrease at 1 h after iohexol injection. A distinct elevation of apparent transverse relaxation rate (R2*) reached the maximum levels on day 1, which was consistent with the expression of hypoxia-inducible factor-1α and vascular endothelial growth factor. ADC, D, and R2* correlated well with histopathological parameters and biochemical parameters. CONCLUSION BOLD combined with IVIM is effective to monitor renal pathophysiology associated with CIAKI.
Collapse
Affiliation(s)
- Yongfang Wang
- Department of Radiology, First Hospital of China Medical University, Shenyang, China
| | - Xin Zhang
- Department of Radiology, First Hospital of China Medical University, Shenyang, China
| | - Zhengxu Hua
- Department of Radiology, First Hospital of China Medical University, Shenyang, China
| | - Lizhi Xie
- GE Healthcare, MR Research China, Beijing, China
| | - Xuan Jiang
- Cardiac Surgery, First Hospital of China Medical University, Shenyang, China
| | - Rongjia Wang
- Department of Radiology, First Hospital of China Medical University, Shenyang, China
| | - Peirong Gao
- Department of Ultrasound, First Hospital of China Medical University, Shenyang, China
| | - Ke Ren
- Department of Radiology, Xiang'an Hospital of Xiamen University, Xiamen, China,
| |
Collapse
|
32
|
|
33
|
Emans TW, Janssen BJ, Joles JA, Krediet CP. Nitric Oxide Synthase Inhibition Induces Renal Medullary Hypoxia in Conscious Rats. J Am Heart Assoc 2018; 7:e009501. [PMID: 30371226 PMCID: PMC6201463 DOI: 10.1161/jaha.118.009501] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Accepted: 06/18/2018] [Indexed: 12/14/2022]
Abstract
Background Renal hypoxia, implicated as crucial factor in onset and progression of chronic kidney disease, may be attributed to reduced nitric oxide because nitric oxide dilates vasculature and inhibits mitochondrial oxygen consumption. We hypothesized that chronic nitric oxide synthase inhibition would induce renal hypoxia. Methods and Results Oxygen-sensitive electrodes, attached to telemeters, were implanted in either renal cortex (n=6) or medulla (n=7) in rats. After recovery and stabilization, baseline oxygenation ( pO 2) was recorded for 1 week. To inhibit nitric oxide synthase, N-ω-nitro-l-arginine (L-NNA; 40 mg/kg/day) was administered via drinking water for 2 weeks. A separate group (n=8), instrumented with blood pressure telemeters, followed the same protocol. L-NNA rapidly induced hypertension (165±6 versus 108±3 mm Hg; P<0.001) and proteinuria (79±12 versus 17±2 mg/day; P<0.001). Cortical pO 2, after initially dipping, returned to baseline and then increased. Medullary pO 2 decreased progressively (up to -19±6% versus baseline; P<0.05). After 14 days of L-NNA, amplitude of diurnal medullary pO 2 was decreased (3.7 [2.2-5.3] versus 7.9 [7.5-8.4]; P<0.01), whereas amplitudes of blood pressure and cortical pO 2 were unaltered. Terminal glomerular filtration rate (1374±74 versus 2098±122 μL/min), renal blood flow (5014±336 versus 9966±905 μL/min), and sodium reabsorption efficiency (13.0±0.8 versus 22.8±1.7 μmol/μmol) decreased (all P<0.001). Conclusions For the first time, we show temporal development of renal cortical and medullary oxygenation during chronic nitric oxide synthase inhibition in unrestrained conscious rats. Whereas cortical pO 2 shows transient changes, medullary pO 2 decreased progressively. Chronic L-NNA leads to decreased renal perfusion and sodium reabsorption efficiency, resulting in progressive medullary hypoxia, suggesting that juxtamedullary nephrons are potentially vulnerable to prolonged nitric oxide depletion.
Collapse
Affiliation(s)
- Tonja W. Emans
- Internal Medicine‐NephrologyAmsterdam UMC / Academic Medical Centre at the University of AmsterdamThe Netherlands
- Nephrology and HypertensionUniversity Medical Centre UtrechtThe Netherlands
| | - Ben J. Janssen
- Pharmacology and ToxicologyMaastricht UniversityThe Netherlands
| | - Jaap A. Joles
- Nephrology and HypertensionUniversity Medical Centre UtrechtThe Netherlands
| | - C.T. Paul Krediet
- Internal Medicine‐NephrologyAmsterdam UMC / Academic Medical Centre at the University of AmsterdamThe Netherlands
| |
Collapse
|
34
|
Shaw I, Rider S, Mullins J, Hughes J, Péault B. Pericytes in the renal vasculature: roles in health and disease. Nat Rev Nephrol 2018; 14:521-534. [DOI: 10.1038/s41581-018-0032-4] [Citation(s) in RCA: 68] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
|
35
|
Castellano G, Franzin R, Stasi A, Divella C, Sallustio F, Pontrelli P, Lucarelli G, Battaglia M, Staffieri F, Crovace A, Stallone G, Seelen M, Daha MR, Grandaliano G, Gesualdo L. Complement Activation During Ischemia/Reperfusion Injury Induces Pericyte-to-Myofibroblast Transdifferentiation Regulating Peritubular Capillary Lumen Reduction Through pERK Signaling. Front Immunol 2018; 9:1002. [PMID: 29875766 PMCID: PMC5974049 DOI: 10.3389/fimmu.2018.01002] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2017] [Accepted: 04/23/2018] [Indexed: 12/21/2022] Open
Abstract
Pericytes are one of the principal sources of scar-forming myofibroblasts in chronic kidneys disease. However, the modulation of pericyte-to-myofibroblast transdifferentiation (PMT) in the early phases of acute kidney injury is poorly understood. Here, we investigated the role of complement in inducing PMT after transplantation. Using a swine model of renal ischemia/reperfusion (I/R) injury, we found the occurrence of PMT after 24 h of I/R injury as demonstrated by reduction of PDGFRβ+/NG2+ cells with increase in myofibroblasts marker αSMA. In addition, PMT was associated with significant reduction in peritubular capillary luminal diameter. Treatment by C1-inhibitor (C1-INH) significantly preserved the phenotype of pericytes maintaining microvascular density and capillary lumen area at tubulointerstitial level. In vitro, C5a transdifferentiated human pericytes in myofibroblasts, with increased αSMA expression in stress fibers, collagen I production, and decreased antifibrotic protein Id2. The C5a-induced PMT was driven by extracellular signal-regulated kinases phosphorylation leading to increase in collagen I release that required both non-canonical and canonical TGFβ pathways. These results showed that pericytes are a pivotal target of complement activation leading to a profibrotic maladaptive cellular response. Our studies suggest that C1-INH may be a potential therapeutic strategy to counteract the development of PMT and capillary lumen reduction in I/R injury.
Collapse
Affiliation(s)
- Giuseppe Castellano
- Nephrology, Dialysis and Transplantation Unit, Department of Emergency and Organ Transplantation, University of Bari Aldo Moro, Bari, Italy
| | - Rossana Franzin
- Nephrology, Dialysis and Transplantation Unit, Department of Emergency and Organ Transplantation, University of Bari Aldo Moro, Bari, Italy
| | - Alessandra Stasi
- Nephrology, Dialysis and Transplantation Unit, Department of Emergency and Organ Transplantation, University of Bari Aldo Moro, Bari, Italy
| | - Chiara Divella
- Nephrology, Dialysis and Transplantation Unit, Department of Emergency and Organ Transplantation, University of Bari Aldo Moro, Bari, Italy
| | - Fabio Sallustio
- Nephrology, Dialysis and Transplantation Unit, Department of Emergency and Organ Transplantation, University of Bari Aldo Moro, Bari, Italy.,Department of Basic Medical Sciences, Neuroscience and Sense Organs, University of Bari Aldo Moro, Bari, Italy
| | - Paola Pontrelli
- Nephrology, Dialysis and Transplantation Unit, Department of Emergency and Organ Transplantation, University of Bari Aldo Moro, Bari, Italy
| | - Giuseppe Lucarelli
- Urology, Andrology and Renal Transplantation Unit, Department of Emergency and Organ Transplantation, University of Bari Aldo Moro, Bari, Italy
| | - Michele Battaglia
- Urology, Andrology and Renal Transplantation Unit, Department of Emergency and Organ Transplantation, University of Bari Aldo Moro, Bari, Italy
| | - Francesco Staffieri
- Veterinary Surgery Unit, Department of Emergency and Organ Transplantation, University of Bari Aldo Moro, Bari, Italy
| | - Antonio Crovace
- Veterinary Surgery Unit, Department of Emergency and Organ Transplantation, University of Bari Aldo Moro, Bari, Italy
| | - Giovanni Stallone
- Nephrology, Dialysis and Transplantation Unit, Department of Medical and Surgical Sciences, University of Foggia, Foggia, Italy
| | - Marc Seelen
- Division of Nephrology, Department of Internal Medicine, University of Groningen, University Medical Centre Groningen, Groningen, Netherlands
| | - Mohamed R Daha
- Division of Nephrology, Department of Internal Medicine, University of Groningen, University Medical Centre Groningen, Groningen, Netherlands.,Department of Nephrology, Leiden University Medical Centre, Leiden, Netherlands
| | - Giuseppe Grandaliano
- Nephrology, Dialysis and Transplantation Unit, Department of Medical and Surgical Sciences, University of Foggia, Foggia, Italy
| | - Loreto Gesualdo
- Nephrology, Dialysis and Transplantation Unit, Department of Emergency and Organ Transplantation, University of Bari Aldo Moro, Bari, Italy
| |
Collapse
|
36
|
Zhang Z, Payne K, Pallone TL. Adaptive responses of rat descending vasa recta to ischemia. Am J Physiol Renal Physiol 2018; 314:F373-F380. [PMID: 28814437 DOI: 10.1152/ajprenal.00062.2017] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
tested whether rat descending vasa recta (DVR) undergo regulatory adaptations after the kidney is exposed to ischemia. Left kidneys (LK) were subjected to 30-min renal artery cross clamp. After 48 h, the postischemic LK and contralateral right kidney (RK) were harvested for study. When compared with DVR isolated from either sham-operated LK or the contralateral RK, postischemic LK DVR markedly increased their NO generation. The selective inducible NOS (iNOS) inhibitor 1400W blocked the NO response. Immunoblots from outer medullary homogenates showed a parallel 2.6-fold increase in iNOS expression ( P = 0.01). Microperfused postischemic LK DVR exposed to angiotensin II (ANG II, 10 nM), constricted less than those from the contralateral RK, and constricted more when exposed to 1400W (10 µM). Resting membrane potentials of pericytes from postischemic LK DVR pericytes were hyperpolarized relative to contralateral RK pericytes (62.0 ± 1.6 vs. 51.8 ± 2.2 mV, respectively, P < 0.05) or those from sham-operated LK (54.9 ± 2.1 mV, P < 0.05). Blockade of NO generation with 1400W did not repolarize postischemic pericytes (62.5 ± 1.4 vs. 61.1 ± 3.4 mV); however, control pericytes were hyperpolarized by exposure to NO donation from S-nitroso- N-acetyl- dl-penicillamine (51.5 ± 2.9 to 62.1 ± 1.4 mV, P < 0.05). We conclude that postischemic adaptations intrinsic to the DVR wall occur after ischemia. A rise in 1400W sensitive NO generation and iNOS expression occurs that is associated with diminished contractile responses to ANG II. Pericyte hyperpolarization occurs that is not explained by the rise in ambient NO generation within the DVR wall.
Collapse
Affiliation(s)
- Zhong Zhang
- Division of Nephrology, Department of Medicine, University of Maryland School of Medicine , Baltimore, Maryland
| | - Kristie Payne
- Division of Nephrology, Department of Medicine, University of Maryland School of Medicine , Baltimore, Maryland
| | - Thomas L Pallone
- Division of Nephrology, Department of Medicine, University of Maryland School of Medicine , Baltimore, Maryland.,Baltimore Veterans Administration Medical Center , Baltimore, Maryland
| |
Collapse
|
37
|
O’Carroll AM, Salih S, Griffiths PR, Bijabhai A, Knepper MA, Lolait SJ. Expression and functional implications of the renal apelinergic system in rodents. PLoS One 2017; 12:e0183094. [PMID: 28817612 PMCID: PMC5560558 DOI: 10.1371/journal.pone.0183094] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2017] [Accepted: 07/28/2017] [Indexed: 02/06/2023] Open
Abstract
Apelin binds to the G protein-coupled apelin receptor (APJ; gene name aplnr) to modulate diverse physiological systems including cardiovascular function, and hydromineral and metabolic balance. Recently a second endogenous ligand for APJ, named apela, has been discovered. We confirm that apela activates signal transduction pathways (ERK activation) in cells expressing the cloned rat APJ. Previous studies suggest that exogenous apela is diuretic, attributable wholly or in part to an action on renal APJ. Thus far the cellular distribution of apela in the kidney has not been reported. We have utilized in situ hybridization histochemistry to reveal strong apela labelling in the inner medulla (IM), with lower levels observed in the inner stripe of the outer medulla (ISOM), of rat and mouse kidneys. This contrasts with renal aplnr expression where the converse is apparent, with intense labelling in the ISOM (consistent with vasa recta labelling) and low-moderate hybridization in the IM, in addition to labelling of glomeruli. Apelin is found in sparsely distributed cells amongst more prevalent aplnr-labelled cells in extra-tubular regions of the medulla. This expression profile is supported by RNA-Seq data that shows that apela, but not apelin or aplnr, is highly expressed in microdissected rat kidney tubules. If endogenous tubular apela promotes diuresis in the kidney it could conceivably do this by interacting with APJ in vasculature, or via an unknown receptor in the tubules. The comparative distribution of apela, apelin and aplnr in the rodent kidney lays the foundation for future work on how the renal apelinergic system interacts.
Collapse
Affiliation(s)
- Anne-Marie O’Carroll
- Bristol Medical School, HW-LINE, University of Bristol, Bristol, United Kingdom
- * E-mail:
| | - Sabrine Salih
- Bristol Medical School, HW-LINE, University of Bristol, Bristol, United Kingdom
| | - Philip R. Griffiths
- Bristol Medical School, HW-LINE, University of Bristol, Bristol, United Kingdom
| | - Aarifah Bijabhai
- Bristol Medical School, HW-LINE, University of Bristol, Bristol, United Kingdom
| | - Mark A. Knepper
- Epithelial Systems Biology Laboratory, Systems Biology Center, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Stephen J. Lolait
- Bristol Medical School, HW-LINE, University of Bristol, Bristol, United Kingdom
| |
Collapse
|
38
|
|
39
|
Kennedy-Lydon T. Immune Functions and Properties of Resident Cells in the Heart and Cardiovascular System: Pericytes. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 1003:93-103. [PMID: 28667555 DOI: 10.1007/978-3-319-57613-8_5] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
This chapter provides an introduction to pericyte physiology. Pericytes are smooth muscle-like cells that wrap around vessels and arterioles. Here, we discuss their structure, function, contractility and interaction with other cells including immune cells and finally their role in pathological processes. Additionally, we discuss recent studies describing pericyte populations in the heart and their potential as targets for future cardiac therapeutic interventions.
Collapse
|
40
|
Wnuk M, Anderegg MA, Graber WA, Buergy R, Fuster DG, Djonov V. Neuropilin1 regulates glomerular function and basement membrane composition through pericytes in the mouse kidney. Kidney Int 2016; 91:868-879. [PMID: 27988210 DOI: 10.1016/j.kint.2016.10.010] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2016] [Revised: 10/04/2016] [Accepted: 10/06/2016] [Indexed: 12/28/2022]
Abstract
Neuropilin1 (Nrp1) is a co-receptor best known to regulate the development of endothelial cells and is a target of anticancer therapies. However, its role in other vascular cells including pericytes is emergent. The kidney is an organ with high pericyte density and cancer patients develop severe proteinuria following administration of NRP1B-neutralizing antibody combined with bevacizumab. Therefore, we investigated whether Nrp1 regulates glomerular capillary integrity after completion of renal development using two mouse models; tamoxifen-inducible NG2Cre to delete Nrp1 specifically in pericytes and administration of Nrp1-neutralizing antibodies. Specific Nrp1 deletion in pericytes did not affect pericyte number but mutant mice developed hematuria with glomerular basement membrane defects. Despite foot process effacement, albuminuria was absent and expression of podocyte proteins remained unchanged upon Nrp1 deletion. Additionally, these mice displayed dilation of the afferent arteriole and glomerular capillaries leading to glomerular hyperfiltration. Nidogen-1 mRNA was downregulated and collagen4α3 mRNA was upregulated with no significant effect on the expression of other basement membrane genes in the mutant mice. These features were phenocopied by treating wild-type mice with Nrp1-neutralizing antibodies. Thus, our results reveal a postdevelopmental role of Nrp1 in renal pericytes as an important regulator of glomerular basement membrane integrity. Furthermore, our study offers novel mechanistic insights into renal side effects of Nrp1 targeting cancer therapies.
Collapse
Affiliation(s)
- Monika Wnuk
- Institute of Anatomy, University of Bern, Bern, Switzerland
| | - Manuel A Anderegg
- Institute of Biochemistry and Molecular Medicine, University of Bern, Bern, Switzerland
| | | | - Regula Buergy
- Institute of Anatomy, University of Bern, Bern, Switzerland
| | - Daniel G Fuster
- Institute of Biochemistry and Molecular Medicine, University of Bern, Bern, Switzerland; Division of Nephrology, Hypertension, and Clinical Pharmacology, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Valentin Djonov
- Institute of Anatomy, University of Bern, Bern, Switzerland.
| |
Collapse
|
41
|
Warmke N, Griffin KJ, Cubbon RM. Pericytes in diabetes-associated vascular disease. J Diabetes Complications 2016; 30:1643-1650. [PMID: 27592245 DOI: 10.1016/j.jdiacomp.2016.08.005] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2016] [Revised: 08/01/2016] [Accepted: 08/08/2016] [Indexed: 12/21/2022]
Abstract
Pericytes are mural cells that support and stabilise the microvasculature, and are present in all vascular beds. Pericyte-endothelial cell crosstalk is essential in both remodelling and quiescent vasculature, and this complex interaction is often disrupted in disease states. Pericyte loss is believed to be an early hallmark of diabetes-associated microvascular disease, including retinopathy and nephropathy. Here we review the current literature defining pericyte biology in the context of diabetes-associated vascular disease, with a particular focus on whether pericytes contribute actively to disease progression. We also speculate regarding the role of pericytes in the recovery from macrovascular complications, such as critical limb ischaemia. It becomes clear that dysfunctional pericytes are likely to actively induce disease progression by causing vasoconstriction and basement membrane thickening, resulting in tissue ischaemia. Moreover, their altered interactions with endothelial cells are likely to cause abnormal and inadequate neovascularisation in diverse vascular beds. Further research is needed to identify mechanisms by which pericyte function is altered by diabetes, with a view to developing therapeutic approaches that normalise vascular function and remodelling.
Collapse
Affiliation(s)
- Nele Warmke
- Leeds Institute of Cardiovascular and Metabolic Medicine, LIGHT laboratories, The University of Leeds, Clarendon Way, Leeds, LS2 9JT, United Kingdom
| | - Kathryn J Griffin
- Leeds Institute of Cardiovascular and Metabolic Medicine, LIGHT laboratories, The University of Leeds, Clarendon Way, Leeds, LS2 9JT, United Kingdom
| | - Richard M Cubbon
- Leeds Institute of Cardiovascular and Metabolic Medicine, LIGHT laboratories, The University of Leeds, Clarendon Way, Leeds, LS2 9JT, United Kingdom.
| |
Collapse
|
42
|
Gomez IG, Roach AM, Nakagawa N, Amatucci A, Johnson BG, Dunn K, Kelly MC, Karaca G, Zheng TS, Szak S, Peppiatt-Wildman CM, Burkly LC, Duffield JS. TWEAK-Fn14 Signaling Activates Myofibroblasts to Drive Progression of Fibrotic Kidney Disease. J Am Soc Nephrol 2016; 27:3639-3652. [PMID: 27026366 DOI: 10.1681/asn.2015111227] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2015] [Accepted: 02/16/2016] [Indexed: 01/15/2023] Open
Abstract
The identification of the cellular origins of myofibroblasts has led to the discovery of novel pathways that potentially drive myofibroblast perpetuation in disease. Here, we further investigated the role of innate immune signaling pathways in this process. In mice, renal injury-induced activation of pericytes, which are myofibroblast precursors attached to endothelial cells, led to upregulated expression of TNF receptor superfamily member 12a, also known as fibroblast growth factor-inducible 14 (Fn14), by these cells. In live rat kidney slices, administration of the Fn14 ligand, TNF-related weak inducer of apoptosis (TWEAK), promoted pericyte-dependent vasoconstriction followed by pericyte detachment from capillaries. In vitro, administration of TWEAK activated and differentiated pericytes into cytokine-producing myofibroblasts, and further activated established myofibroblasts in a manner requiring canonical and noncanonical NF-κB signaling pathways. Deficiency of Fn14 protected mouse kidneys from fibrogenesis, inflammation, and associated vascular instability after in vivo injury, and was associated with loss of NF-κB signaling. In a genetic model of spontaneous CKD, therapeutic delivery of anti-TWEAK blocking antibodies attenuated disease progression, preserved organ function, and increased survival. These results identify the TWEAK-Fn14 signaling pathway as an important factor in myofibroblast perpetuation, fibrogenesis, and chronic disease progression.
Collapse
Affiliation(s)
- Ivan G Gomez
- Research & Development, Biogen, Cambridge, Massachusetts.,Division of Nephrology, Departments of Medicine & Pathology, and Institute of Stem Cell & Regenerative Medicine, University of Washington, Seattle, Washington; and
| | - Allie M Roach
- Research & Development, Biogen, Cambridge, Massachusetts.,Division of Nephrology, Departments of Medicine & Pathology, and Institute of Stem Cell & Regenerative Medicine, University of Washington, Seattle, Washington; and
| | - Naoki Nakagawa
- Division of Nephrology, Departments of Medicine & Pathology, and Institute of Stem Cell & Regenerative Medicine, University of Washington, Seattle, Washington; and
| | - Aldo Amatucci
- Research & Development, Biogen, Cambridge, Massachusetts
| | - Bryce G Johnson
- Research & Development, Biogen, Cambridge, Massachusetts.,Division of Nephrology, Departments of Medicine & Pathology, and Institute of Stem Cell & Regenerative Medicine, University of Washington, Seattle, Washington; and
| | - Kadeshia Dunn
- Medway School of Pharmacy, University of Kent, Chatham, Kent, United Kingdom
| | - Mark C Kelly
- Medway School of Pharmacy, University of Kent, Chatham, Kent, United Kingdom
| | - Gamze Karaca
- Research & Development, Biogen, Cambridge, Massachusetts
| | | | - Suzanne Szak
- Research & Development, Biogen, Cambridge, Massachusetts
| | | | - Linda C Burkly
- Research & Development, Biogen, Cambridge, Massachusetts;
| | - Jeremy S Duffield
- Research & Development, Biogen, Cambridge, Massachusetts; .,Division of Nephrology, Departments of Medicine & Pathology, and Institute of Stem Cell & Regenerative Medicine, University of Washington, Seattle, Washington; and
| |
Collapse
|
43
|
Briguori C, Visconti G, Donahue M, De Micco F, Focaccio A, Golia B, Signoriello G, Ciardiello C, Donnarumma E, Condorelli G. RenalGuard system in high-risk patients for contrast-induced acute kidney injury. Am Heart J 2016; 173:67-76. [PMID: 26920598 DOI: 10.1016/j.ahj.2015.12.005] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2015] [Accepted: 12/04/2015] [Indexed: 11/17/2022]
Abstract
BACKGROUND High urine flow rate (UFR) has been suggested as a target for effective prevention of contrast-induced acute kidney injury (CI-AKI). The RenalGuard therapy (saline infusion plus furosemide controlled by the RenalGuard system) facilitates the achievement of this target. METHODS Four hundred consecutive patients with an estimated glomerular filtration rate ≤30 mL/min per 1.73 m(2) and/or a high predicted risk (according to the Mehran score ≥11 and/or the Gurm score >7%) treated by the RenalGuard therapy were analyzed. The primary end points were (1) the relationship between CI-AKI and UFR during preprocedural, intraprocedural, and postprocedural phases of the RenalGuard therapy and (2) the rate of acute pulmonary edema and impairment in electrolytes balance. RESULTS Urine flow rate was significantly lower in the patients with CI-AKI in the preprocedural phase (208 ± 117 vs 283 ± 160 mL/h, P < .001) and in the intraprocedural phase (389 ± 198 vs 483 ± 225 mL/h, P = .009). The best threshold for CI-AKI prevention was a mean intraprocedural phase UFR ≥450 mL/h (area under curve 0.62, P = .009, sensitivity 80%, specificity 46%). Performance of percutaneous coronary intervention (hazard ratio [HR] 4.13, 95% CI 1.81-9.10, P < .001), the intraprocedural phase UFR <450 mL/h (HR 2.27, 95% CI 1.05-2.01, P = .012), and total furosemide dose >0.32 mg/kg (HR 5.03, 95% CI 2.33-10.87, P < .001) were independent predictors of CI-AKI. Pulmonary edema occurred in 4 patients (1%). Potassium replacement was required in 16 patients (4%). No patients developed severe hypomagnesemia, hyponatremia, or hypernatremia. CONCLUSIONS RenalGuard therapy is safe and effective in reaching high UFR. Mean intraprocedural UFR ≥450 mL/h should be the target for optimal CI-AKI prevention.
Collapse
Affiliation(s)
- Carlo Briguori
- Laboratory of Interventional Cardiology and Department of Cardiology, Clinica Mediterranea, Naples, Italy.
| | - Gabriella Visconti
- Laboratory of Interventional Cardiology and Department of Cardiology, Clinica Mediterranea, Naples, Italy
| | - Michael Donahue
- Laboratory of Interventional Cardiology and Department of Cardiology, Clinica Mediterranea, Naples, Italy
| | - Francesca De Micco
- Laboratory of Interventional Cardiology and Department of Cardiology, Clinica Mediterranea, Naples, Italy
| | - Amelia Focaccio
- Laboratory of Interventional Cardiology and Department of Cardiology, Clinica Mediterranea, Naples, Italy
| | - Bruno Golia
- Laboratory of Interventional Cardiology and Department of Cardiology, Clinica Mediterranea, Naples, Italy
| | - Giuseppe Signoriello
- Department of Mental Health and Preventive Medicine, Second University of Naples, Naples, Italy
| | | | | | - Gerolama Condorelli
- Department of Cellular and Molecular Biology and Pathology, "Federico II" University of Naples, Naples, Italy
| |
Collapse
|
44
|
Lactate Transport and Receptor Actions in Retina: Potential Roles in Retinal Function and Disease. Neurochem Res 2015; 41:1229-36. [PMID: 26677077 DOI: 10.1007/s11064-015-1792-x] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2015] [Revised: 11/06/2015] [Accepted: 11/08/2015] [Indexed: 01/01/2023]
Abstract
In retina, like in brain, lactate equilibrates across cell membranes via monocarboxylate transporters and in the extracellular space by diffusion, forming a basis for the action of lactate as a transmitter of metabolic signals. In the present paper, we argue that the lactate receptor GPR81, also known as HCAR1, may contribute importantly to the control of retinal cell functions in health and disease. GPR81, a G-protein coupled receptor, is known to downregulate cAMP both in adipose and nervous tissue. The receptor also acts through other down-stream mechanisms to control functions, such as excitability, metabolism and inflammation. Recent publications predict effects of the lactate receptor on neurodegeneration. Neurodegenerative diseases in retina, where the retinal ganglion cells die, notably glaucoma and diabetic retinopathy, may be linked to disturbed lactate homeostasis. Pilot studies reveal high GPR81 mRNA in retina and indicate GPR81 localization in Müller cells and retinal ganglion cells. Moreover, monocarboxylate transporters are expressed in retinal cells. We envision that lactate receptors and transporters could be useful future targets of novel therapeutic strategies to protect neurons and prevent or counteract glaucoma as well as other retinal diseases.
Collapse
|
45
|
Verma SK, Molitoris BA. Renal endothelial injury and microvascular dysfunction in acute kidney injury. Semin Nephrol 2015; 35:96-107. [PMID: 25795503 DOI: 10.1016/j.semnephrol.2015.01.010] [Citation(s) in RCA: 158] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The kidney is comprised of heterogeneous cell populations that function together to perform a number of tightly controlled, complex and interdependent processes. Renal endothelial cells contribute to vascular tone, regulation of blood flow to local tissue beds, modulation of coagulation and inflammation, and vascular permeability. Both ischemia and sepsis have profound effects on the renal endothelium, resulting in microvascular dysregulation resulting in continued ischemia and further injury. In recent years, the concept of the vascular endothelium as an organ that is both the source of and target for inflammatory injury has become widely appreciated. Here we revisit the renal endothelium in the light of ever evolving molecular advances.
Collapse
Affiliation(s)
- Sudhanshu Kumar Verma
- Nephrology Division, Department of Medicine, Indiana University School of Medicine, The Roudebush VA Medical Center, Indiana Center for Biological Microscopy, Indianapolis, IN
| | - Bruce A Molitoris
- Nephrology Division, Department of Medicine, Indiana University School of Medicine, The Roudebush VA Medical Center, Indiana Center for Biological Microscopy, Indianapolis, IN.
| |
Collapse
|
46
|
Moreno C, Llinás MT, Rodriguez F, Moreno JM, Salazar FJ. Nitric oxide, prostaglandins and angiotensin II in the regulation of renal medullary blood flow during volume expansion. J Physiol Biochem 2015; 72:1-8. [PMID: 26611113 DOI: 10.1007/s13105-015-0450-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2015] [Accepted: 11/17/2015] [Indexed: 11/29/2022]
Abstract
Regulation of medullary blood flow (MBF) is essential in maintaining renal function and blood pressure. However, it is unknown whether outer MBF (OMBF) and papillary blood flow (PBF) are regulated independently when extracellular volume (ECV) is enhanced. The aim of this study was to determine whether OMBF and PBF are differently regulated and whether there is an interaction between nitric oxide (NO), prostaglandins (PGs) and angiotensin II (Ang II) in regulating OMBF and PBF when ECV is enhanced. To achieve these goals, OMBF and PBF were measured by laser-Doppler in volume-expanded rats treated with a cyclooxygenase inhibitor (meclofenamate, 3 mg/kg) and/or a NO synthesis inhibitor (L-nitro-arginine methyl ester (L-NAME), 3 μg/kg/min) and/or Ang II (10 ng/kg/min). OMBF was unchanged by NO or PGs synthesis inhibition but decreased by 36 % (P < 0.05) when L-NAME and meclofenamate were infused simultaneously. PBF was similarly reduced by L-NAME (12 %), meclofenamate (17 %) or L-NAME + meclofenamate (19 %). Ang II did not modify OMBF, but it led to a similar decrease (P < 0.05) in OMBF when it was administered to rats with reduced NO (32 %), PGs (36 %) or NO and PGs (37 %) synthesis. In contrast, the fall in PBF induced by Ang II (12 %) was enhanced (P < 0.05) by the simultaneous PGs (30 %) or PGs and NO (31 %) synthesis inhibition but not in L-NAME-treated rats (20 %). This study presents novel findings suggesting that blood flows to the outer medulla and renal papilla are differently regulated and showing that there is a complex interaction between NO, PGs and Ang II in regulating OMBF and PBF when ECV is enhanced.
Collapse
Affiliation(s)
- Carol Moreno
- Cardiovascular and Metabolic Diseases, MedImmune, Cambridge, UK
| | - María T Llinás
- Department of Physiology, School of Medicine, University of Murcia, Murcia, 30100, Spain.,Regional Campus of International Excellence "Mare Nostrum", Murcia, Spain.,Instituto Murciano de Investigación Biomédica, Murcia, Spain
| | - Francisca Rodriguez
- Department of Physiology, School of Medicine, University of Murcia, Murcia, 30100, Spain.,Regional Campus of International Excellence "Mare Nostrum", Murcia, Spain.,Instituto Murciano de Investigación Biomédica, Murcia, Spain
| | - Juan M Moreno
- Department of Physiology, School of Medicine, University of Murcia, Murcia, 30100, Spain.,Regional Campus of International Excellence "Mare Nostrum", Murcia, Spain.,Instituto Murciano de Investigación Biomédica, Murcia, Spain
| | - F Javier Salazar
- Department of Physiology, School of Medicine, University of Murcia, Murcia, 30100, Spain. .,Regional Campus of International Excellence "Mare Nostrum", Murcia, Spain. .,Instituto Murciano de Investigación Biomédica, Murcia, Spain.
| |
Collapse
|
47
|
Affiliation(s)
- S. Reuter
- Klinik für Innere Medizin III; AG Experimentelle Nephrologie; Universitätsklinikum Jena; Jena Germany
| | - R. Mrowka
- Klinik für Innere Medizin III; AG Experimentelle Nephrologie; Universitätsklinikum Jena; Jena Germany
| |
Collapse
|
48
|
Kennedy-Lydon T, Crawford C, Wildman SS, Peppiatt-Wildman CM. Nonsteroidal anti-inflammatory drugs alter vasa recta diameter via pericytes. Am J Physiol Renal Physiol 2015. [PMID: 26202223 DOI: 10.1152/ajprenal.00199.2015] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
We have previously shown that vasa recta pericytes are known to dilate vasa recta capillaries in the presence of PGE2 and contract vasa recta capillaries when endogenous production of PGE2 is inhibited by the nonselective nonsteroidal anti-inflammatory drug (NSAID) indomethacin. In the present study, we used a live rat kidney slice model to build on these initial observations and provide novel data that demonstrate that nonselective, cyclooxygenase-1-selective, and cyclooxygenase -2-selective NSAIDs act via medullary pericytes to elicit a reduction of vasa recta diameter. Real-time images of in situ vasa recta were recorded, and vasa recta diameters at pericyte and nonpericyte sites were measured offline. PGE2 and epoprostenol (a prostacyclin analog) evoked dilation of vasa recta specifically at pericyte sites, and PGE2 significantly attenuated pericyte-mediated constriction of vasa recta evoked by both endothelin-1 and ANG II. NSAIDs (indomethacin > SC-560 > celecoxib > meloxicam) evoked significantly greater constriction of vasa recta capillaries at pericyte sites than at nonpericyte sites, and indomethacin significantly attenuated the pericyte-mediated vasodilation of vasa recta evoked by PGE2, epoprostenol, bradykinin, and S-nitroso-N-acetyl-l-penicillamine. Moreover, a reduction in PGE2 was measured using an enzyme immune assay after superfusion of kidney slices with indomethacin. In addition, immunohistochemical techniques were used to demonstrate the population of EP receptors in the medulla. Collectively, these data demonstrate that pericytes are sensitive to changes in PGE2 concentration and may serve as the primary mechanism underlying NSAID-associated renal injury and/or further compound-associated tubular damage.
Collapse
Affiliation(s)
- Teresa Kennedy-Lydon
- Urinary System Physiology Unit, Medway School of Pharmacy, The Universities of Kent and Greenwich at Medway, Kent, United Kingdom
| | - Carol Crawford
- Urinary System Physiology Unit, Medway School of Pharmacy, The Universities of Kent and Greenwich at Medway, Kent, United Kingdom
| | - Scott S Wildman
- Urinary System Physiology Unit, Medway School of Pharmacy, The Universities of Kent and Greenwich at Medway, Kent, United Kingdom
| | - Claire M Peppiatt-Wildman
- Urinary System Physiology Unit, Medway School of Pharmacy, The Universities of Kent and Greenwich at Medway, Kent, United Kingdom
| |
Collapse
|
49
|
Evans KK, Nawata CM, Pannabecker TL. Isolation and perfusion of rat inner medullary vasa recta. Am J Physiol Renal Physiol 2015; 309:F300-4. [PMID: 26062876 DOI: 10.1152/ajprenal.00214.2015] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2015] [Accepted: 06/04/2015] [Indexed: 11/22/2022] Open
Abstract
Outer medullary isolated descending vasa recta have proven to be experimentally tractable, and consequently much has been learned about outer medullary vasa recta endothelial transport, pericyte contractile mechanisms, and tubulovascular interactions. In contrast, inner medullary vasa recta have never been isolated from any species, and therefore isolated vasa recta function has never been subjected to in vitro quantitative evaluation. As we teased out inner medullary thin limbs of Henle's loops from the Munich-Wistar rat, we found that vasa recta could be isolated using similar protocols. We isolated ∼30 inner medullary vasa recta from 23 adult male Munich-Wistar rats and prepared them for brightfield or electron microscopy, gene expression analysis by RT-PCR, or isolated tubule microperfusion. Morphological characteristics include branching and nonbranching segments exhibiting a thin endothelium, axial surface filaments radiating outward giving vessels a hairy appearance, and attached interstitial cells. Electron microscopy shows multiple cells, tight junctions, and either continuous or fenestrated endothelia. Isolated vasa recta express genes encoding the urea transporter UT-B and/or the fenestral protein PV-1, genes expressed in descending or ascending vasa recta, respectively. The transepithelial NaCl permeability (383.3 ± 60.0 × 10(-5) cm/s, mean ± SE, n = 4) was determined in isolated perfused vasa recta. Future quantitative analyses of isolated inner medullary vasa recta should provide structural and functional details important for more fully understanding fluid and solute flows through the inner medulla and their associated regulatory pathways.
Collapse
Affiliation(s)
- Kristen K Evans
- Department of Physiology, University of Arizona Health Sciences Center, Tucson, Arizona
| | - C Michele Nawata
- Department of Physiology, University of Arizona Health Sciences Center, Tucson, Arizona
| | - Thomas L Pannabecker
- Department of Physiology, University of Arizona Health Sciences Center, Tucson, Arizona
| |
Collapse
|
50
|
Stefanska A, Eng D, Kaverina N, Duffield JS, Pippin JW, Rabinovitch P, Shankland SJ. Interstitial pericytes decrease in aged mouse kidneys. Aging (Albany NY) 2015; 7:370-82. [PMID: 26081073 PMCID: PMC4505164 DOI: 10.18632/aging.100756] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/15/2023]
Abstract
With increasing age, the kidney undergoes characteristic changes in the glomerular and tubulo-interstitial compartments, which are ultimately accompanied by reduced kidney function. Studies have shown age-related loss of peritubular vessels. Normal peritubular vessel tone, function and survival depend on neighboring pericytes. Pericyte detachment leads to vascular damage, which can be accompanied by their differentiation to fibroblasts and myofibroblasts, a state that favors matrix production. To better understand the fate of pericytes in the aged kidney, 27 month-old mice were studied. Compared to 3 month-old young adult mice, aged kidneys showed a substantial decrease in capillaries, identified by CD31 staining, in both cortex and medulla. This was accompanied by a marked decrease in surrounding NG2+ / PDGFRβ+ pericytes. This decrease was more pronounced in the medulla. Capillaries devoid of pericytes were typically dilated in aged mice. Aged kidneys were also characterized by interstitial fibrosis due to increased collagen-I and -III staining. This was accompanied by an increase in the number of pericytes that acquired a pro-fibrotic phenotype, identified by increased PDGFRβ+ / αSMA+ staining. These findings are consistent with the decline in kidney interstitial pericytes as a critical step in the development of changes to the peritubular vasculature with aging, and accompanying fibrosis.
Collapse
Affiliation(s)
- Ania Stefanska
- Division of Nephrology, Department of Medicine, University of Washington, Seattle, WA 98104, USA
| | - Diana Eng
- Division of Nephrology, Department of Medicine, University of Washington, Seattle, WA 98104, USA
| | - Natalya Kaverina
- Division of Nephrology, Department of Medicine, University of Washington, Seattle, WA 98104, USA
| | - Jeremy S. Duffield
- Division of Nephrology, Department of Medicine, University of Washington, Seattle, WA 98104, USA
- Biogen Idec, Cambridge, MA 02142, USA
| | - Jeffrey W. Pippin
- Division of Nephrology, Department of Medicine, University of Washington, Seattle, WA 98104, USA
| | - Peter Rabinovitch
- Department of Pathology, University of Washington, Seattle, WA 98195, USA
| | - Stuart J. Shankland
- Division of Nephrology, Department of Medicine, University of Washington, Seattle, WA 98104, USA
| |
Collapse
|