1
|
Eirin A, Siddiqi S, Hughes AG, Jiang Y, Zhu XY, Kazeminia S, Lu B, Xing L, Lu B, Tang H, Xue A, Lerman A, Textor SC, Lerman LO. Renovascular Disease and Mitochondrial Dysfunction in Human Mesenchymal Stem Cells. J Am Soc Nephrol 2024; 35:1507-1519. [PMID: 39012704 PMCID: PMC11543019 DOI: 10.1681/asn.0000000000000440] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Accepted: 07/09/2024] [Indexed: 07/18/2024] Open
Abstract
Key Points Renovascular disease impairs the capacity of human adipose tissue–derived mesenchymal stem/stromal cells to repair ischemic murine kidneys. miR-378h modulated the capacity of renovascular disease adipose tissue–derived mesenchymal stem/stromal cells to repair ischemic kidneys in vivo . Background Renovascular disease leads to renal ischemia, hypertension, and eventual kidney failure. Autologous transplantation of adipose tissue–derived mesenchymal stem/stromal cells (MSCs) improves perfusion and oxygenation in stenotic human kidneys, but associated atherosclerosis and hypertension might blunt their effectiveness. We hypothesized that renovascular disease alters the human MSC transcriptome and impairs their reparative potency. Methods MSCs were harvested from subcutaneous abdominal fat of patients with renovascular disease and healthy volunteers (n =3 each), characterized and subsequently injected (5×105/200 μ l) into mice 2 weeks after renal artery stenosis or sham surgery (n =6/group). Two weeks later, mice underwent imaging and tissue studies. MSCs from healthy volunteers and in those with renovascular disease were also characterized by mRNA/microRNA (miRNA) sequencing. Based on these, MSC proliferation and mitochondrial damage were assessed in vitro before and after miRNA modulation and in vivo in additional renal artery stenosis mice administered with MSCs from renovascular disease pretreated with miR-378h mimic (n =5) or inhibitor (n =4). Results MSCs engrafted in stenotic mouse kidneys. Healthy volunteer MSCs (but not renovascular disease MSCs) decreased BP, improved serum creatinine levels and stenotic-kidney cortical perfusion and oxygenation, and attenuated peritubular capillary loss, tubular injury, and fibrosis. Genes upregulated in renovascular disease MSCs versus healthy volunteer MSCs were mostly implicated in transcription and cell proliferation, whereas those downregulated encoded mainly mitochondrial proteins. Upregulated miRNAs, including miR-378h, primarily target nuclear-encoded mitochondrial genes, whereas downregulated miRNAs mainly target genes implicated in transcription and cell proliferation. MSC proliferation was similar, but their mitochondrial structure and reparative function both in vivo and in vitro improved after miR-378h inhibition. Conclusions Renovascular disease impaired the reparative capacity of human MSCs, possibly by dysregulating miR-378h that targets mitochondrial genes. Podcast This article contains a podcast at https://dts.podtrac.com/redirect.mp3/www.asn-online.org/media/podcast/JASN/2024_08_21_ASN0000000000000440.mp3
Collapse
Affiliation(s)
- Alfonso Eirin
- Division of Nephrology and Hypertension, Department of Medicine, Mayo Clinic, Rochester, Minnesota
- Department of Cardiovascular Diseases, Mayo Clinic, Rochester, Minnesota
| | - Sarosh Siddiqi
- Division of Nephrology and Hypertension, Department of Medicine, Mayo Clinic, Rochester, Minnesota
| | - Autumn G. Hughes
- Division of Nephrology and Hypertension, Department of Medicine, Mayo Clinic, Rochester, Minnesota
| | - Yamei Jiang
- Division of Nephrology and Hypertension, Department of Medicine, Mayo Clinic, Rochester, Minnesota
| | - Xiang-Yang Zhu
- Division of Nephrology and Hypertension, Department of Medicine, Mayo Clinic, Rochester, Minnesota
| | - Sara Kazeminia
- Division of Nephrology and Hypertension, Department of Medicine, Mayo Clinic, Rochester, Minnesota
| | - Bo Lu
- Division of Nephrology and Hypertension, Department of Medicine, Mayo Clinic, Rochester, Minnesota
| | - Li Xing
- Division of Nephrology and Hypertension, Department of Medicine, Mayo Clinic, Rochester, Minnesota
| | - Brandon Lu
- Division of Nephrology and Hypertension, Department of Medicine, Mayo Clinic, Rochester, Minnesota
| | - Hui Tang
- Division of Nephrology and Hypertension, Department of Medicine, Mayo Clinic, Rochester, Minnesota
| | - Ailing Xue
- Division of Nephrology and Hypertension, Department of Medicine, Mayo Clinic, Rochester, Minnesota
| | - Amir Lerman
- Department of Cardiovascular Diseases, Mayo Clinic, Rochester, Minnesota
| | - Stephen C. Textor
- Division of Nephrology and Hypertension, Department of Medicine, Mayo Clinic, Rochester, Minnesota
| | - Lilach O. Lerman
- Division of Nephrology and Hypertension, Department of Medicine, Mayo Clinic, Rochester, Minnesota
- Department of Cardiovascular Diseases, Mayo Clinic, Rochester, Minnesota
| |
Collapse
|
2
|
Fu J, Lin Z, Zhang B, Song L, Qin N, Qiu J, Yang M, Zou Y. Magnetic Resonance Imaging in Atherosclerotic Renal Artery Stenosis: The Update and Future Directions from Interventional Perspective. KIDNEY DISEASES (BASEL, SWITZERLAND) 2024; 10:23-31. [PMID: 38322626 PMCID: PMC10843188 DOI: 10.1159/000534499] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Accepted: 08/23/2023] [Indexed: 02/08/2024]
Abstract
Background Atherosclerotic renal artery stenosis (ARAS) is a condition where the renal arteries become narrowed due to atherosclerosis, leading to reduced blood flow to the kidneys and various renal complications. The effectiveness of interventional treatments, such as renal artery angioplasty and stenting, remains debated, making patient selection for these procedures challenging. Summary This review focuses on the diagnosis and management of ARAS, with a particular emphasis on the potential role of functional magnetic resonance imaging (MRI) in evaluating renal function and mechanisms. By summarizing current diagnostic approaches and outcomes of interventional treatments, the review highlights the importance of informed clinical decision-making in ARAS management. Functional MRI emerges as a promising noninvasive tool to assess renal function, aiding in patient stratification and treatment planning. Key Messages The efficacy of interventional treatments for ARAS requires further investigation and careful patient selection. Functional MRI holds promise as a noninvasive means to assess renal function and mechanisms, potentially guiding more effective clinical decisions in ARAS management. Advancing research in diagnostic methods, particularly functional MRI, can enhance our understanding and improve the treatment outcomes for ARAS patients.
Collapse
Affiliation(s)
- Jia Fu
- Department of Interventional Radiology and Vascular Surgery, Peking University First Hospital, Beijing, China
| | - Zhiyong Lin
- Department of Radiology, Peking University First Hospital, Beijing, China
| | - Bihui Zhang
- Department of Interventional Radiology and Vascular Surgery, Peking University First Hospital, Beijing, China
| | - Li Song
- Department of Interventional Radiology and Vascular Surgery, Peking University First Hospital, Beijing, China
| | - Naishan Qin
- Department of Radiology, Peking University First Hospital, Beijing, China
| | - Jianxing Qiu
- Department of Radiology, Peking University First Hospital, Beijing, China
| | - Min Yang
- Department of Interventional Radiology and Vascular Surgery, Peking University First Hospital, Beijing, China
| | - Yinghua Zou
- Department of Interventional Radiology and Vascular Surgery, Peking University First Hospital, Beijing, China
| |
Collapse
|
3
|
Shaughnessey EM, Kann SH, Charest JL, Vedula EM. Human Kidney Proximal Tubule-Microvascular Model Facilitates High-Throughput Analyses of Structural and Functional Effects of Ischemia-Reperfusion Injury. Adv Biol (Weinh) 2024; 8:e2300127. [PMID: 37786311 DOI: 10.1002/adbi.202300127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 08/27/2023] [Indexed: 10/04/2023]
Abstract
Kidney ischemia reperfusion injury (IRI) poses a major global healthcare burden, but effective treatments remain elusive. IRI involves a complex interplay of tissue-level structural and functional changes caused by interruptions in blood and filtrate flow and reduced oxygenation. Existing in vitro models poorly replicate the in vivo injury environment and lack means of monitoring tissue function during the injury process. Here, a high-throughput human primary kidney proximal tubule (PT)-microvascular model is described, which facilitates in-depth structural and rapid functional characterization of IRI-induced changes in the tissue barrier. The PREDICT96 (P96) microfluidic platform's user-controlled fluid flow can mimic the conditions of IR to induce pronounced changes in cell structure that resemble clinical and in vivo phenotypes. High-throughput trans-epi/endo-thelial electrical resistance (TEER) sensing is applied to non-invasively track functional changes in the PT-microvascular barrier during the two-stage injury process and over repeated episodes of injury. Notably, ischemia causes an initial increase in tissue TEER followed by a sudden increase in permeability upon reperfusion, and this biphasic response occurs only with the loss of both fluid flow and oxygenation. This study demonstrates the potential of the P96 kidney IRI model to enhance understanding of IRI and fuel therapeutic development.
Collapse
Affiliation(s)
- Erin M Shaughnessey
- Draper Scholar, The Charles Stark Draper Laboratory Inc., 555 Technology Square, Cambridge, MA, 02139, USA
- Department of Biomedical Engineering, Tufts University, 4 Colby Street, Medford, MA, 02155, USA
| | - Samuel H Kann
- Draper Scholar, The Charles Stark Draper Laboratory Inc., 555 Technology Square, Cambridge, MA, 02139, USA
- Department of Mechanical Engineering, Boston University, 110 Cummington Mall, Boston, MA, 02215, USA
| | - Joseph L Charest
- The Charles Stark Draper Laboratory Inc., 555 Technology Square, Cambridge, MA, 02139, USA
| | - Else M Vedula
- The Charles Stark Draper Laboratory Inc., 555 Technology Square, Cambridge, MA, 02139, USA
| |
Collapse
|
4
|
Sabra MS, Hemida FK, Allam EAH. Adenine model of chronic renal failure in rats to determine whether MCC950, an NLRP3 inflammasome inhibitor, is a renopreventive. BMC Nephrol 2023; 24:377. [PMID: 38114914 PMCID: PMC10731818 DOI: 10.1186/s12882-023-03427-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Accepted: 12/06/2023] [Indexed: 12/21/2023] Open
Abstract
BACKGROUND Chronic renal failure (CRF) is defined by a significant decline in renal function that results in decreased salt filtration and inhibition of tubular reabsorption, which ultimately causes volume enlargement. This study evaluated the potential renopreventive effects of the NLRP3 inflammasome inhibitor MCC950 in adenine-induced CRF in rats due to conflicting evidence on the effects of MCC950 on the kidney. METHODS Since the majority of the kidney tubular abnormalities identified in people with chronic renal disease are comparable to those caused by adding 0.75 percent of adenine powder to a rat's diet each day for four weeks, this method has received broad approval as a model for evaluating kidney damage. Throughout the test, blood pressure was checked weekly and at the beginning. Additionally, oxidative stress factors, urine sample examination, histological modifications, and immunohistochemical adjustments of caspase-3 and interleukin-1 beta (IL-1) levels in renal tissues were carried out. RESULTS Results revealed that MCC950, an inhibitor of the NLRP3 inflammasome, had a renopreventive effect, which was demonstrated by a reduction in blood pressure readings and an improvement in urine, serum, and renal tissue indicators that indicate organ damage. This was also demonstrated by the decrease in neutrophil gelatinase-associated lipocalin tubular expression (NGAL). The NLRP3 inflammasome inhibitor MCC950 was found to significantly alleviate the worsening renal cellular alterations evidenced by increased expression of caspase-3 and IL-1, according to immunohistochemical tests. CONCLUSION The NLRP3 inflammasome inhibitor MCC950 demonstrated renopreventive effects in the CRF rat model, suggesting that it might be used as a treatment strategy to stop the progression of CRF.
Collapse
Affiliation(s)
- Mahmoud S Sabra
- Pharmacology Department, Faculty of Veterinary Medicine, Assiut University, Assiut, 71526, Egypt.
| | - Fahmy K Hemida
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Assiut University, Assiut, 71526, Egypt
| | - Essmat A H Allam
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Assiut University, Assiut, 71526, Egypt
| |
Collapse
|
5
|
Rajagopalan KS, Glasstetter LM, Zhu XY, Thaler R, Tang H, Jordan KL, Saadiq IM, Herrmann SM, Chade AR, Irazabal MV, Lerman LO, Eirin A. Renal Ischemia Induces Epigenetic Changes in Apoptotic, Proteolytic, and Mitochondrial Genes in Swine Scattered Tubular-like Cells. Cells 2022; 11:1803. [PMID: 35681498 PMCID: PMC9180447 DOI: 10.3390/cells11111803] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Revised: 05/19/2022] [Accepted: 05/27/2022] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Scattered tubular-like cells (STCs) are dedifferentiated renal tubular cells endowed with progenitor-like characteristics to repair injured parenchymal cells. STCs may be damaged and rendered ineffective by renal artery stenosis (RAS), but the underlying processes remain unclear. We hypothesized that RAS alters the epigenetic landscape on DNA and the ensuing gene transcriptional profile of swine STCs. METHODS CD24+/CD133+ STCs were isolated from pig kidneys after 10 weeks of RAS or sham (n = 3 each) and their whole 5-methylcytosine (5mC) and 5-hydroxymethylcytosine (5hmC) profiles were examined by 5mC and 5hmC immunoprecipitation sequencing (MeDIP-/hMeDIP-seq, respectively). A subsequent integrated (MeDIP/hMeDIP-seq/mRNA-seq) analysis was performed by comparing all online available gene sets using Gene Set Enrichment Analysis. Apoptosis, proteolysis, and mitochondrial structure and function were subsequently evaluated in vitro. RESULTS Differential expression (DE) analysis revealed 239 genes with higher and 236 with lower 5mC levels and 275 genes with higher and 315 with lower 5hmC levels in RAS-STCs compared to Normal-STCs (fold change ≥1.4 or ≤0.7, p ≤ 0.05). Integrated MeDIP-/hMeDIP-seq/mRNA-seq analysis identified several overlapping (DE-5mC/mRNA and DE-5hmC/mRNA levels) genes primarily implicated in apoptosis, proteolysis, and mitochondrial functions. Furthermore, RAS-STCs exhibited decreased apoptosis, mitochondrial matrix density, and ATP production, and increased intracellular amino acid concentration and ubiquitin expression. CONCLUSIONS Renal ischemia induces epigenetic changes in apoptosis-, proteolysis-, and mitochondria-related genes, which correlate with alterations in the transcriptomic profile and corresponding function of swine STCs. These observations may contribute to developing novel targeted interventions to preserve the reparative potency of STCs in renal disease.
Collapse
Affiliation(s)
- Kamalnath S. Rajagopalan
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN 55901, USA; (K.S.R.); (L.M.G.); (X.-Y.Z.); (H.T.); (K.L.J.); (I.M.S.); (S.M.H.); (M.V.I.); (L.O.L.)
| | - Logan M. Glasstetter
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN 55901, USA; (K.S.R.); (L.M.G.); (X.-Y.Z.); (H.T.); (K.L.J.); (I.M.S.); (S.M.H.); (M.V.I.); (L.O.L.)
| | - Xiang-Yang Zhu
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN 55901, USA; (K.S.R.); (L.M.G.); (X.-Y.Z.); (H.T.); (K.L.J.); (I.M.S.); (S.M.H.); (M.V.I.); (L.O.L.)
| | - Roman Thaler
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN 55901, USA;
| | - Hui Tang
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN 55901, USA; (K.S.R.); (L.M.G.); (X.-Y.Z.); (H.T.); (K.L.J.); (I.M.S.); (S.M.H.); (M.V.I.); (L.O.L.)
| | - Kyra L. Jordan
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN 55901, USA; (K.S.R.); (L.M.G.); (X.-Y.Z.); (H.T.); (K.L.J.); (I.M.S.); (S.M.H.); (M.V.I.); (L.O.L.)
| | - Ishran M. Saadiq
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN 55901, USA; (K.S.R.); (L.M.G.); (X.-Y.Z.); (H.T.); (K.L.J.); (I.M.S.); (S.M.H.); (M.V.I.); (L.O.L.)
| | - Sandra M. Herrmann
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN 55901, USA; (K.S.R.); (L.M.G.); (X.-Y.Z.); (H.T.); (K.L.J.); (I.M.S.); (S.M.H.); (M.V.I.); (L.O.L.)
| | - Alejandro R. Chade
- Department of Physiology and Biophysics, Medicine and Radiology, University of Mississippi Medical Center, Jackson, MS 55901, USA;
| | - Maria V. Irazabal
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN 55901, USA; (K.S.R.); (L.M.G.); (X.-Y.Z.); (H.T.); (K.L.J.); (I.M.S.); (S.M.H.); (M.V.I.); (L.O.L.)
| | - Lilach O. Lerman
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN 55901, USA; (K.S.R.); (L.M.G.); (X.-Y.Z.); (H.T.); (K.L.J.); (I.M.S.); (S.M.H.); (M.V.I.); (L.O.L.)
| | - Alfonso Eirin
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN 55901, USA; (K.S.R.); (L.M.G.); (X.-Y.Z.); (H.T.); (K.L.J.); (I.M.S.); (S.M.H.); (M.V.I.); (L.O.L.)
| |
Collapse
|
6
|
Emergent players in renovascular disease. Clin Sci (Lond) 2022; 136:239-256. [PMID: 35129198 DOI: 10.1042/cs20210509] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Revised: 01/25/2022] [Accepted: 01/26/2022] [Indexed: 02/07/2023]
Abstract
Renovascular disease (RVD) remains a common etiology of secondary hypertension. Recent clinical trials revealed unsatisfactory therapeutic outcomes of renal revascularization, leading to extensive investigation to unravel key pathophysiological mechanisms underlying irreversible functional loss and structural damage in the chronically ischemic kidney. Research studies identified complex interactions among various players, including inflammation, fibrosis, mitochondrial injury, cellular senescence, and microvascular remodeling. This interplay resulted in a shift of our understanding of RVD from a mere hemodynamic disorder to a pro-inflammatory and pro-fibrotic pathology strongly influenced by systemic diseases like metabolic syndrome (MetS), hypertension, diabetes mellitus, and hyperlipidemia. Novel diagnostic approaches have been tested for early detection and follow-up of RVD progression, using new imaging techniques and biochemical markers of renal injury and dysfunction. Therapies targeting some of the pathological pathways governing the development of RVD have shown promising results in animal models, and a few have moved from bench to clinical research. This review summarizes evolving understanding in chronic ischemic kidney injury.
Collapse
|
7
|
Nyvad J, Lerman A, Lerman LO. With a Little Help From My Friends: the Role of the Renal Collateral Circulation in Atherosclerotic Renovascular Disease. Hypertension 2022; 79:717-725. [PMID: 35135307 PMCID: PMC8917080 DOI: 10.1161/hypertensionaha.121.17960] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The collateral circulation can adapt to bypass major arteries with limited flow and serves a crucial protective role in coronary, cerebral, and peripheral arterial disease. Emerging evidence indicates that the renal collateral circulation can similarly adapt and thereby limit kidney ischemia in atherosclerotic renovascular disease. These adaptations predominantly include recruitment of preexisting microvessels for arteriogenesis, with de novo vessel formation playing a limited role. Yet, adaptations of the renal collateral circulation in renovascular disease are often insufficient to fully compensate for the limited flow within an obstructed renal artery and may be hampered by the severity of obstruction or patient comorbidities. Experimental strategies have attempted to circumvent limitations of collateral formation and improve the prognosis of patients with various ischemic vascular territories. These have included pharmacological approaches such as endothelial growth factors, renin-angiotensin-aldosterone system blockade, and If-channel-blockers, as well as interventions like preconditioning, exercise, enhanced external counter-pulsation, and low-energy shock-wave therapy. However, few of these strategies have been implemented in atherosclerotic renovascular disease. This review summarizes current understanding regarding the development of renal collateral circulation in atherosclerotic renovascular disease. Studies are needed to apply lessons learned in other vascular beds in the setting of atherosclerotic renovascular disease to develop new treatment regimens for this patient group.
Collapse
Affiliation(s)
- Jakob Nyvad
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN. (J.N., L.O.L.).,Department of Nephrology and Hypertension, Aarhus University Hospital, Aarhus, Denmark (J.N.)
| | - Amir Lerman
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN. (A.L.)
| | - Lilach O Lerman
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN. (J.N., L.O.L.)
| |
Collapse
|
8
|
Walther CP, Civitello AB, Liao KK, Navaneethan SD. Nephrology Considerations in the Management of Durable and Temporary Mechanical Circulatory Support. KIDNEY360 2022; 3:569-579. [PMID: 35582171 PMCID: PMC9034823 DOI: 10.34067/kid.0003382021] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Accepted: 01/04/2022] [Indexed: 01/10/2023]
Abstract
Durable and temporary mechanical circulatory support (MCS) use is growing for a range of cardiovascular indications. Kidney dysfunction is common in people evaluated for or receiving durable or temporary MCS and portends worse outcomes. This kidney dysfunction can be due to preexisting kidney chronic kidney disease (CKD), acute kidney injury (AKI) related to acute cardiovascular disease necessitating MCS, AKI due to cardiac procedures, and acute and chronic MCS effects and complications. Durable MCS, with implantable continuous flow pumps, is used for long-term support in advanced heart failure refractory to guideline-directed medical and device therapy, either permanently or as a bridge to heart transplantation. Temporary MCS-encompassing in this review intra-aortic balloon pumps (IABP), axial flow pumps, centrifugal flow pumps, and venoarterial ECMO-is used for diverse situations: high-risk percutaneous coronary interventions (PCI), acute decompensated heart failure, cardiogenic shock, and resuscitation after cardiac arrest. The wide adoption of MCS makes it imperative to improve understanding of the effects of MCS on kidney health/function and of kidney health/function on MCS outcomes. The complex structure and functions of the kidney, and the complex health states of individuals receiving MCS, makes investigations in this area challenging, and current knowledge is limited. Fortunately, the increasing nephrology toolbox of noninvasive kidney health/function assessments may enable development and testing of individualized management strategies and therapeutics in the future. We review technology, epidemiology, pathophysiology, clinical considerations, and future directions in MCS and nephrology.
Collapse
Affiliation(s)
- Carl P. Walther
- Selzman Institute for Kidney Health, Section of Nephrology, Department of Medicine, Baylor College of Medicine, Houston, Texas
| | - Andrew B. Civitello
- Section of Cardiology, Department of Medicine, Baylor College of Medicine, Houston, Texas,Advanced Heart Failure Center of Excellence, Baylor College of Medicine, Houston, Texas
| | - Kenneth K. Liao
- Division of Cardiothoracic Transplantation and Circulatory Support, Department of Surgery, Baylor College of Medicine, Houston, Texas
| | - Sankar D. Navaneethan
- Selzman Institute for Kidney Health, Section of Nephrology, Department of Medicine, Baylor College of Medicine, Houston, Texas,Section of Nephrology, Michael E. DeBakey Veterans Affairs Medical Center, Houston, Texas,Institute of Clinical and Translational Research, Baylor College of Medicine, Houston, Texas
| |
Collapse
|
9
|
Hickson LJ, Herrmann SM, McNicholas BA, Griffin MD. Progress toward the Clinical Application of Mesenchymal Stromal Cells and Other Disease-Modulating Regenerative Therapies: Examples from the Field of Nephrology. KIDNEY360 2021; 2:542-557. [PMID: 34316720 PMCID: PMC8312727 DOI: 10.34067/kid.0005692020] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Accepted: 01/27/2021] [Indexed: 02/07/2023]
Abstract
Drawing from basic knowledge of stem-cell biology, embryonic development, wound healing, and aging, regenerative medicine seeks to develop therapeutic strategies that complement or replace conventional treatments by actively repairing diseased tissue or generating new organs and tissues. Among the various clinical-translational strategies within the field of regenerative medicine, several can be broadly described as promoting disease resolution indirectly through local or systemic interactions with a patient's cells, without permanently integrating or directly forming new primary tissue. In this review, we focus on such therapies, which we term disease-modulating regenerative therapies (DMRT), and on the extent to which they have been translated into the clinical arena in four distinct areas of nephrology: renovascular disease (RVD), sepsis-associated AKI (SA-AKI), diabetic kidney disease (DKD), and kidney transplantation (KTx). As we describe, the DMRT that has most consistently progressed to human clinical trials for these indications is mesenchymal stem/stromal cells (MSCs), which potently modulate ischemic, inflammatory, profibrotic, and immune-mediated tissue injury through diverse paracrine mechanisms. In KTx, several early-phase clinical trials have also tested the potential for ex vivo-expanded regulatory immune cell therapies to promote donor-specific tolerance and prevent or resolve allograft injury. Other promising DMRT, including adult stem/progenitor cells, stem cell-derived extracellular vesicles, and implantable hydrogels/biomaterials remain at varying preclinical stages of translation for these renal conditions. To date (2021), no DMRT has gained market approval for use in patients with RVD, SA-AKI, DKD, or KTx, and clinical trials demonstrating definitive, cost-effective patient benefits are needed. Nonetheless, exciting progress in understanding the disease-specific mechanisms of action of MSCs and other DMRT, coupled with increasing knowledge of the pathophysiologic basis for renal-tissue injury and the experience gained from pioneering early-phase clinical trials provide optimism that influential, regenerative treatments for diverse kidney diseases will emerge in the years ahead.
Collapse
Affiliation(s)
- LaTonya J. Hickson
- Division of Nephrology and Hypertension, Department of Medicine, Mayo Clinic, Jacksonville, Florida
| | - Sandra M. Herrmann
- Division of Nephrology and Hypertension, Department of Medicine, Mayo Clinic, Rochester, Minnesota
| | - Bairbre A. McNicholas
- Regenerative Medicine Institute (REMEDI) at CÚRAM Centre for Research in Medical Devices, School of Medicine, National University of Ireland Galway, Ireland
- Nephrology Services, Galway University Hospitals, Saolta University Healthcare System, Galway, Ireland
- Critical Care Services, Galway University Hospitals, Saolta University Healthcare System, Galway, Ireland
| | - Matthew D. Griffin
- Regenerative Medicine Institute (REMEDI) at CÚRAM Centre for Research in Medical Devices, School of Medicine, National University of Ireland Galway, Ireland
- Nephrology Services, Galway University Hospitals, Saolta University Healthcare System, Galway, Ireland
| |
Collapse
|
10
|
Signorini L, Zaza G, Gambaro G. The challenge of early glomerular filtration rate decline in response to antihypertensive treatment and chronic kidney disease outcomes. Nephrol Dial Transplant 2020; 37:222-229. [PMID: 33155053 DOI: 10.1093/ndt/gfaa171] [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: 01/18/2020] [Indexed: 11/14/2022] Open
Abstract
Hypertension and chronic kidney disease (CKD) are closely linked pathological processes. Combating high blood pressure (BP) is an essential part of preventing CKD progression and reducing cardiovascular (CV) risk. Data from recent randomized controlled trials on patients at high CV risk showed the beneficial effects of intensive action to meet BP targets on mortality related to CV disease. The impact of meeting such targets on renal function is still unclear, however, particularly for patients with CKD. This issue has been the object of several post hoc analyses because lowering BP definitely has a nephroprotective role, but the early decline in glomerular filtration rate (GFR) associated with antihypertensive therapies and strict BP targets is still a concern in nephrology clinical practice. The present review discusses the results of studies on this topic, focusing specifically on the clinical significance of early GFR decline in response to treatment with angiotensin-converting enzyme inhibitor/angiotensin receptor blocker, or to different BP targets, in terms of renal and CV outcomes, and how this tips the balance towards continuing or discontinuing antihypertensive therapy.
Collapse
Affiliation(s)
| | - Gianluigi Zaza
- Renal Unit, Department of Medicine, University Hospital of Verona, Verona, Italia
| | - Giovanni Gambaro
- Renal Unit, Department of Medicine, University Hospital of Verona, Verona, Italia
| |
Collapse
|
11
|
Improved renal outcomes after revascularization of the stenotic renal artery in pigs by prior treatment with low-energy extracorporeal shockwave therapy. J Hypertens 2020; 37:2074-2082. [PMID: 31246892 DOI: 10.1097/hjh.0000000000002158] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
BACKGROUND Revascularization does not restore renal function in most patients with atherosclerotic renal artery stenosis (RAS), likely because of inflammation and fibrosis within the stenotic kidney. Low-energy shockwave therapy (LE-SWT) stimulates angiogenesis in the stenotic kidney, but its ability to improve renal function and structure after revascularization remains unexplored. We tested the hypothesis that a LE-SWT regimen before percutaneous transluminal renal angioplasty (PTRA) would enable PTRA to restore renal function in hypercholesterolemic pigs with RAS (HC+RAS), and that this would be associated with attenuation of renal inflammation and fibrosis. METHODS AND RESULTS Twenty-six pigs were studied after 16 weeks of HC+RAS, HC+RAS treated with PTRA with or without a preceding LE-SWT regimen (bi-weekly for 3 weeks), and controls. Single-kidney renal blood flow (RBF), glomerular filtration rate (GFR), and oxygenation were assessed in vivo using imaging 4 weeks after PTRA, and then inflammation and fibrosis ex vivo.Four weeks after successful PTRA, blood pressure fell similarly in both revascularized groups. Yet, stenotic-kidney GFR remained lower in HC+RAS and HC+RAS+PTRA (P < 0.01 vs. normal), but was improved in HC+RAS+PTRA+SW (P > 0.05 vs. normal). Furthermore, reduced inflammation, medullary fibrosis, and cortical hypoxia were only shown in swine stenotic kidneys pretreated with LE-SWT before PTRA 4 weeks later. CONCLUSION LE-SWT delivery before revascularization permitted PTRA to improve function and decrease cortical and medullary damage in the stenotic swine kidney. This study, therefore, supports the use of an adjunct SW pretreatment to enhance the success of PTRA in blunting loss of kidney function in experimental HC+RAS.
Collapse
|
12
|
Afarideh M, Zhang X, Ferguson CM, Glockner JF, Lerman A, Textor SC, Lerman LO. Peristenotic Collateral Circulation in Atherosclerotic Renovascular Disease: Association With Kidney Function and Response to Treatment. Hypertension 2020; 76:497-505. [PMID: 32507040 DOI: 10.1161/hypertensionaha.120.15057] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The significance of peristenotic collateral circulation (PCC) development around a stenotic renal artery is unknown. We tested the hypothesis that PCC is linked to loss of kidney function and recovery potential in patients with atherosclerotic renovascular disease (ARVD). Thirty-four patients with ARVD were assigned to medical-therapy with or without revascularization based on clinical indications. The PCC was visualized using multidetector computed tomography and defined relative to segmental arteries in patients with essential hypertension. PCC number before and 3 months after treatment was correlated with various renal parameters. Thirty-four stenotic kidneys from 30 patients were analyzed. PCC number correlated inversely with kidney volume. ARVD-stenotic kidneys with baseline PCC (collateral ARVD [C-ARVD], n=13) associated with elevated 24-hour urine protein and stenotic kidney vein level of tumor necrosis factor-α, lower single-kidney volume and blood flow, and greater hypoxia than in stenotic kidneys with no PCC (no collateral ARVD [NC-ARVD], n=17). Revascularization (but not medical-therapy alone) improved stenotic kidney function and reduced inflammation in both NC-ARVD and C-ARVD. In C-ARVD, revascularization also increased stenotic kidney volume, blood flow, and oxygenation to levels comparable to NC-ARVD, and induced PCC regression. However, revascularization improved systolic blood pressure, plasma renin activity, and filtration fraction only in NC-ARVD. Therefore, patients with C-ARVD have greater kidney dysfunction, atrophy, hypoxia, and inflammation compared with patients with NC-ARVD, suggesting that PCC does not effectively protect the stenotic kidney in ARVD. Renal artery revascularization improved in C-ARVD stenotic kidney function, but not hypertension or renin-angiotensin system activation. These observations may help direct management of patients with ARVD.
Collapse
Affiliation(s)
| | | | | | - James F Glockner
- From the Division of Nephrology and Hypertension and the Departments of Radiology (J.F.G.), Mayo Clinic, Rochester, MN
| | - Amir Lerman
- Cardiovascular Medicine (A.L.), Mayo Clinic, Rochester, MN
| | | | | |
Collapse
|
13
|
Chen XJ, Zhang X, Jiang K, Krier JD, Zhu X, Conley S, Lerman A, Lerman LO. Adjunctive mesenchymal stem/stromal cells augment microvascular function in poststenotic kidneys treated with low-energy shockwave therapy. J Cell Physiol 2020; 235:9806-9818. [PMID: 32430932 DOI: 10.1002/jcp.29794] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Revised: 05/05/2020] [Accepted: 05/05/2020] [Indexed: 12/21/2022]
Abstract
Effective therapeutic strategies are needed to preserve renal function in patients with atherosclerotic renal artery stenosis (ARAS). Low-energy shockwave therapy (SW) and adipose tissue-derived mesenchymal stem/stromal cells (MSCs) both stimulate angiogenesis repair of stenotic kidney injury. This study tested the hypothesis that intrarenal delivery of adipose tissue-derived MSCs would enhance the capability of SW to preserve stenotic kidney function and structure. Twenty-two pigs were studied after 16 weeks of ARAS, ARAS treated with a SW regimen (bi-weekly for 3 weeks) with or without subsequent intrarenal delivery of adipose tissue-derived MSCs and controls. Four weeks after treatment, single-kidney renal blood flow (RBF) before and after infusion of acetylcholine, glomerular filtration rate (GFR), and oxygenation were assessed in vivo and the renal microcirculation, fibrosis, and oxidative stress ex vivo. Mean arterial pressure remained higher in ARAS, ARAS + SW, and ARAS + SW + MSC compared with normal. Both SW and SW + MSC similarly elevated the decreased stenotic kidney GFR and RBF observed in ARAS to normal levels. Yet, SW + MSC significantly improved RBF response to acetylcholine in ARAS, and attenuated capillary loss and oxidative stress more than SW alone. Density of larger microvessels was similarly increased by both interventions. Therefore, although significant changes in functional outcomes were not observed in a short period of time, adjunct MSCs enhanced pro-angiogenic effect of SW to improve renal microvascular outcomes, suggesting this as an effective stratege for long-term management of renovascular disease.
Collapse
Affiliation(s)
- Xiao-Jun Chen
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota.,Department of Nephrology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Xin Zhang
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota
| | - Kai Jiang
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota
| | - James D Krier
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota
| | - Xiangyang Zhu
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota
| | - Sabena Conley
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota
| | - Amir Lerman
- Department of Cardiovascular Diseases, Mayo Clinic, Rochester, Minnesota
| | - Lilach O Lerman
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota.,Department of Cardiovascular Diseases, Mayo Clinic, Rochester, Minnesota
| |
Collapse
|
14
|
Simeoni M, Borrelli S, Garofalo C, Fuiano G, Esposito C, Comi A, Provenzano M. Atherosclerotic-nephropathy: an updated narrative review. J Nephrol 2020; 34:125-136. [PMID: 32270411 DOI: 10.1007/s40620-020-00733-0] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Accepted: 04/03/2020] [Indexed: 12/13/2022]
Abstract
The increased prevalence of chronic kidney disease (CKD) in elderly patients recognizes, as main cause, the long-term exposure to atherosclerosis and hypertension. Chronic ischemic damage due to critical renal arterial stenosis induces oxidative stress and intra-renal inflammation, resulting in fibrosis and microvascular remodelling, that is the histological picture of atherosclerotic renal vascular disease (ARVD). The concomitant presence of a long history of hypertension may generate intimal thickening and luminal narrowing of renal arteries and arterioles, glomerulosclerosis, interstitial fibrosis and tubular atrophy, more typically expression of hypertensive nephropathy. These complex mechanisms contribute to the development of CKD and the progression to End Stage Kidney Disease. In elderly CKD patients, the distinction among these nephropathies may be problematic; therefore, ischemic and hypertensive nephropathies can be joined in a unique clinical syndrome defined as atherosclerotic nephropathy. The availability of novel diagnostic procedures, such as intra-vascular ultrasound and BOLD-MRI, in addition to traditional imaging, have opened new scenarios, because these tools allow to identify ischemic lesions responsive to renal revascularization. Indeed, although trials have deflated the role of renal revascularization on the renal outcomes, it should be still used to avoid dialysis initiation and/or to reduce blood pressure in selected elderly patients at high risk. Nonetheless, lifestyle modifications (smoking cessation, increased physical activity), statins and antiplatelet use, as well as cautious use of renin-angiotensin system inhibitors, remain the main therapeutic approach aimed at slowing the renal damage progression. Mesenchymal stem cells and Micro-RNA are promising target of anti-fibrotic therapy, which might provide potential benefit in ARVD patients, though safety and efficacy profile in humans is unknown too.
Collapse
Affiliation(s)
| | - Silvio Borrelli
- Nephrology and Dialysis Unit, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Carlo Garofalo
- Nephrology and Dialysis Unit, University of Campania "Luigi Vanvitelli", Naples, Italy.
| | - Giorgio Fuiano
- Nephrology Units at University "Magna Graecia", Catanzaro, Italy
| | | | - Alessandro Comi
- Nephrology Units at University "Magna Graecia", Catanzaro, Italy
| | | |
Collapse
|
15
|
Abstract
PURPOSE OF REVIEW Renovascular disease (RVD) remains an important cause of hypertension and renal dysfunction. Given the failure of renal revascularization to provide consistent clinical benefit in the Cardiovascular Outcomes for Renal Artery Lesions trial among others, further research has underscored the need for mechanistically targeted interventions to improve renal outcomes in patients in RVD. This review discusses novel therapeutic approaches for RVD in the post-Cardiovascular Outcomes for Renal Artery Lesions era. RECENT FINDINGS Emerging evidence indicates that renal inflammation, microvascular remodeling, and mitochondrial damage accelerate progression of renal injury and are important determinants of the response to revascularization. Experimental studies have identified interventions capable of ameliorating renal inflammation (e.g., cytokine inhibitors, mesenchymal stem cells), microvascular remodeling (proangiogenic interventions), and mitochondrial injury (mito-protective drugs), alone or combined with renal revascularization, to preserve the structure and function of the poststenotic kidney. Recent prospective pilot studies in patients with atherosclerotic RVD demonstrate the safety and feasibility of some of such interventions to protect the kidney. SUMMARY Experimental studies and pilot clinical trials suggest that therapies targeting renal inflammation, microvascular remodeling, and mitochondrial damage have the potential to preserve the structure and function of the stenotic kidney. Further studies in larger cohorts are needed to confirm their renoprotective effects and clinical role in human RVD.
Collapse
|
16
|
Renal Artery Stenosis Alters Gene Expression in Swine Scattered Tubular-Like Cells. Int J Mol Sci 2019; 20:ijms20205069. [PMID: 31614781 PMCID: PMC6829501 DOI: 10.3390/ijms20205069] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2019] [Revised: 10/07/2019] [Accepted: 10/09/2019] [Indexed: 12/14/2022] Open
Abstract
Background: Scattered tubular-like cells (STCs) proliferate and differentiate to support neighboring injured renal tubular cells during recovery from insults. Renal artery stenosis (RAS) induces renal ischemia and hypertension and leads to loss of kidney function, but whether RAS alters renal endogenous repair mechanisms, such as STCs, remains unknown. We hypothesize that RAS in swine modifies the messenger RNA (mRNA) profile of STCs, blunting their in vitro reparative capacity. Methods: CD24+/CD133+ STCs were isolated from pig kidneys after 10-weeks of RAS or sham (n = 3 each) and their gene cargo analyzed using high-throughput mRNAseq. Expression profiles for upregulated and downregulated mRNAs in RAS-STCs were functionally interpreted by gene ontology analysis. STC activation was assessed by counting the total number of STCs in pig kidney sections using flow cytometry, whereas cell proliferation was assessed in vitro. Results: Of all expressed genes, 1430 genes were upregulated and 315 downregulated in RAS- versus Normal-STCs. Expression of selected candidate genes followed the same fold change directions as the mRNAseq findings. Genes upregulated in RAS-STCs were involved in cell adhesion, extracellular matrix remodeling, and kidney development, whereas those downregulated in RAS-STCs are related to cell cycle and cytoskeleton. The percentage of STCs from dissociated kidney cells was higher in RAS versus Normal pigs, but their proliferation rate was blunted. Conclusions: Renal ischemia and hypertension in swine induce changes in the mRNA profile of STCs, associated with increased STC activation and impaired proliferation. These observations suggest that RAS may alter the reparative capacity of STCs.
Collapse
|
17
|
Reply. J Hypertens 2019; 37:2302-2303. [PMID: 31567761 DOI: 10.1097/hjh.0000000000002242] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
18
|
Dominiczak AF, Kuo D, Bhalla V, Granger JP, Griffin KA. Celebrating 40 Years of Accomplishments. Hypertension 2018; 73:3-6. [PMID: 30571572 DOI: 10.1161/hypertensionaha.118.12252] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Anna F Dominiczak
- From the Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, United Kingdom (A.F.D.)
| | - Denise Kuo
- American Heart Association, Dallas, TX (D.K.)
| | - Vivek Bhalla
- Department of Medicine, Division of Nephrology, Stanford University, Palo Alto, CA (V.B.)
| | - Joey P Granger
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, MS (J.P.G.)
| | - Karen A Griffin
- Loyola University Medical Center, Maywood, IL (K.A.G.).,Edward Hines, Jr. VA, Hines, IL (K.A.G.)
| |
Collapse
|