1
|
Zhu Y, He H, Qiu H, Zhang X, Wang L, Li W. Prognostic Nutritional Index Combined with Triglyceride-Glucose Index to Contrast a Nomogram for Predicting Contrast-Induced Kidney Injury in Type 2 Diabetes Mellitus Patients with Acute Coronary Syndrome After Percutaneous Coronary Intervention. Clin Interv Aging 2023; 18:1663-1673. [PMID: 37810953 PMCID: PMC10559899 DOI: 10.2147/cia.s429957] [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: 08/03/2023] [Accepted: 09/27/2023] [Indexed: 10/10/2023] Open
Abstract
Objective Our objective was to develop and validate a nomogram model aiming at predicting the risk of contrast-induced acute kidney injury (CI-AKI) following percutaneous coronary intervention (PCI) in patients suffering from type 2 diabetes mellitus (T2DM) and also diagnosed with acute coronary syndrome (ACS). Methods The study gathered data from 722 T2DM patients with ACS who received PCI treatment at the Affiliated Hospital of Xuzhou Medical University between February 2019 and December 2022, serving as the training set. Considering the validation set, the study included 217 patients who received PCI at the East Affiliated Hospital of Xuzhou Medical University. The patients were classified into CI-AKI and non-CI-AKI groups. The study employed univariate and multivariate logistic analysis for identifying independent risk factors for CI-AKI, followed by developing a predictive nomogram model for CI-AKI risk using R software. The predictive performance and clinical utility of the nomogram were assessed through internal and external validation, utilizing the areas under the receiver operating characteristic curve (AUC-ROC), the Hosmer-Lemeshow test and calibration correction curve, and decision curve analysis (DCA). Results The nomogram comprised four variables: age, estimated glomerular filtration rate (eGFR), triglyceride-glucose (TyG) index, and prognostic nutritional index (PNI). The AUC-ROC were 0.785 (95% confidence interval (CI) 0.729-0.841) and 0.802 (95% CI 0.699-0.905) for the training and validation cohorts, respectively, indicating a high discriminative ability of the nomogram. The calibration assessment and decision curve analysis have substantiated the strong concordance and clinical usefulness of the aforementioned. Conclusion The nomogram exhibits favorable discrimination and accuracy, enabling it to visually and individually identify pre-procedure high-risk patients, and possesses a predictive capacity regarding CI-AKI incidence after PCI in patients diagnosed with both T2DM and ACS.
Collapse
Affiliation(s)
- Yinghua Zhu
- Institute of Cardiovascular Diseases, Xuzhou Medical University, Xuzhou, People’s Republic of China
| | - Haiyan He
- Department of Cardiology, Xuzhou Municipal Hospital Affiliated to Xuzhou Medical University, Xuzhou, People’s Republic of China
| | - Hang Qiu
- Institute of Cardiovascular Diseases, Xuzhou Medical University, Xuzhou, People’s Republic of China
| | - Xudong Zhang
- Institute of Cardiovascular Diseases, Xuzhou Medical University, Xuzhou, People’s Republic of China
| | - Linsheng Wang
- Institute of Cardiovascular Diseases, Xuzhou Medical University, Xuzhou, People’s Republic of China
| | - Wenhua Li
- Institute of Cardiovascular Diseases, Xuzhou Medical University, Xuzhou, People’s Republic of China
- Department of Cardiology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, People’s Republic of China
| |
Collapse
|
2
|
Patschan D, Patschan S, Matyukhin I, Hoffmeister M, Lauxmann M, Ritter O, Dammermann W. Metabolomics in Acute Kidney Injury: The Experimental Perspective. J Clin Med Res 2023; 15:283-291. [PMID: 37434774 PMCID: PMC10332883 DOI: 10.14740/jocmr4913] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Accepted: 05/08/2023] [Indexed: 07/13/2023] Open
Abstract
Acute kidney injury (AKI) affects increasing numbers of in-hospital patients in Central Europe and the USA, the prognosis remains poor. Although substantial progress has been achieved in the identification of molecular/cellular processes that induce and perpetuate AKI, more integrated pathophysiological perspectives are missing. Metabolomics enables the identification of low-molecular-weight (< 1.5 kD) substances from biological specimens such as certain types of fluid or tissue. The aim of the article was to review the literature on metabolic profiling in experimental AKI and to answer the question if metabolomics allows the integration of distinct pathophysiological events such as tubulopathy and microvasculopathy in ischemic and toxic AKI. The following databases were searched for references: PubMed, Web of Science, Cochrane Library, Scopus. The period lasted from 1940 until 2022. The following terms were utilized: "acute kidney injury" OR "acute renal failure" OR "AKI" AND "metabolomics" OR "metabolic profiling" OR "omics" AND "ischemic" OR "toxic" OR "drug-induced" OR "sepsis" OR "LPS" OR "cisplatin" OR "cardiorenal" OR "CRS" AND "mouse" OR "mice" OR "murine" OR "rats" OR "rat". Additional search terms were "cardiac surgery", "cardiopulmonary bypass", "pig", "dog", and "swine". In total, 13 studies were identified. Five studies were related to ischemic, seven studies to toxic (lipopolysaccharide (LPS), cisplatin), and one study to heat shock-associated AKI. Only one study, related to cisplatin-induced AKI, was performed as a targeted analysis. The majority of the studies identified multiple metabolic deteriorations upon ischemia/the administration of LPS or cisplatin (e.g., amino acid, glucose, lipid metabolism). Particularly, abnormalities in the lipid homeostasis were shown under almost all experimental conditions. LPS-induced AKI most likely depends on the alterations in the tryptophan metabolism. Metabolomics studies provide a deeper understanding of pathophysiological links between distinct processes that are responsible for functional impairment/structural damage in ischemic or toxic or other types of AKI.
Collapse
Affiliation(s)
- Daniel Patschan
- Department of Medicine 1, Cardiology, Angiology, Nephrology, University Hospital Brandenburg of the Brandenburg Medical School Theodor Fontane, Brandenburg, Germany
- Faculty of Health Sciences Brandenburg, Brandenburg Medical School Theodor Fontane, Germany
| | - Susann Patschan
- Department of Medicine 1, Cardiology, Angiology, Nephrology, University Hospital Brandenburg of the Brandenburg Medical School Theodor Fontane, Brandenburg, Germany
| | - Igor Matyukhin
- Department of Medicine 1, Cardiology, Angiology, Nephrology, University Hospital Brandenburg of the Brandenburg Medical School Theodor Fontane, Brandenburg, Germany
| | - Meike Hoffmeister
- Faculty of Health Sciences Brandenburg, Brandenburg Medical School Theodor Fontane, Germany
- Institute of Biochemistry, Brandenburg Medical School Theodor Fontane, Brandenburg, Germany
| | - Martin Lauxmann
- Institute of Biochemistry, Brandenburg Medical School Theodor Fontane, Brandenburg, Germany
| | - Oliver Ritter
- Department of Medicine 1, Cardiology, Angiology, Nephrology, University Hospital Brandenburg of the Brandenburg Medical School Theodor Fontane, Brandenburg, Germany
- Faculty of Health Sciences Brandenburg, Brandenburg Medical School Theodor Fontane, Germany
| | - Werner Dammermann
- Faculty of Health Sciences Brandenburg, Brandenburg Medical School Theodor Fontane, Germany
- Department of Medicine 2, Gastroenterology, Diabetes, Endocrinology, University Hospital Brandenburg of the Brandenburg Medical School Theodor Fontane, Brandenburg, Germany
| |
Collapse
|
3
|
Wu D, Liu J, Zhou C, Ma W, Zhou L, Ge Y, Jia R. Immunomagnetic Delivery of Adipose-Derived Endothelial Progenitor Cells for the Repair of Renal Ischemia-Reperfusion Injury in a Rat Model. Bioengineering (Basel) 2023; 10:bioengineering10050509. [PMID: 37237579 DOI: 10.3390/bioengineering10050509] [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: 02/04/2023] [Revised: 04/19/2023] [Accepted: 04/21/2023] [Indexed: 05/28/2023] Open
Abstract
Renal ischemia-reperfusion injury (IRI) is a significant cause of acute kidney injury (AKI) and usually brings severe public health consequences. Adipose-derived endothelial progenitor cell (AdEPCs) transplantation is beneficial for AKI but suffers from low delivery efficiency. This study was conducted to explore the protective effects of magnetically delivered AdEPCs on the repair of renal IRI. Two types of magnetic delivery methods, namely the endocytosis magnetization (EM) method and the immunomagnetic (IM) method were fabricated using PEG@Fe3O4 and CD133@Fe3O4, and their cytotoxicities in AdEPCs were assessed. In the renal IRI rat model, magnetic AdEPCs were injected via the tail vein and a magnet was placed beside the injured kidney for magnetic guidance. The distribution of transplanted AdEPCs, renal function, and tubular damage were evaluated. Our results suggested that CD133@Fe3O4 had the minimum negative effects on the proliferation, apoptosis, angiogenesis, and migration of AdEPCs compared with PEG@Fe3O4. Renal magnetic guidance could significantly enhance the transplantation efficiency and the therapeutic outcomes of AdEPCs-PEG@Fe3O4 and AdEPCs-CD133@Fe3O4 in the injured kidneys. However, under renal magnetic guidance, AdEPCs-CD133@Fe3O4 had stronger therapeutic effects than PEG@Fe3O4 after renal IRI. The immunomagnetic delivery of AdEPCs with CD133@Fe3O4 could be a promising therapeutic strategy for renal IRI.
Collapse
Affiliation(s)
- Di Wu
- Department of Urology, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, China
| | - Jingyu Liu
- Department of Urology, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, China
| | - Changcheng Zhou
- Department of Urology, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, China
| | - Wenjie Ma
- Department of Urology, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, China
| | - Liuhua Zhou
- Department of Urology, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, China
| | - Yuzheng Ge
- Department of Urology, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, China
| | - Ruipeng Jia
- Department of Urology, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, China
| |
Collapse
|
4
|
Lemmer D, Schmidt J, Kummer K, Lemmer B, Wrede A, Seitz C, Balcarek P, Schwarze K, Müller GA, Patschan D, Patschan S. Impairment of muscular endothelial cell regeneration in dermatomyositis. Front Neurol 2022; 13:952699. [PMID: 36330424 PMCID: PMC9623165 DOI: 10.3389/fneur.2022.952699] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Accepted: 09/01/2022] [Indexed: 11/22/2022] Open
Abstract
Background and aim Inflammatory myopathies are heterogeneous in terms of etiology, (immuno)pathology, and clinical findings. Endothelial cell injury, as it occurs in DM, is a common feature of numerous inflammatory and non-inflammatory vascular diseases. Vascular regeneration is mediated by both local and blood-derived mechanisms, such as the mobilization and activation of so-called proangiogenic cells (PACs) or early endothelial progenitor cells (eEPCs). The current study aimed to evaluate parameters of eEPC integrity in dermatomyositis (DM), compared to necrotizing myopathy (NM) and to non-myopathic controls. Methods Blood samples from DM and NM patients were compared to non-myositis controls and analyzed for the following parameters: circulating CD133+/VEGFR-2+ cells, number of colony-forming unit endothelial cells (CFU-ECs), concentrations of angiopoietin 1, vascular endothelial growth factor (VEGF), and CXCL-16. Muscle biopsies from DM and NM subjects underwent immunofluorescence analysis for CXCR6, nestin, and CD31 (PECAM-1). Finally, myotubes, derived from healthy donors, were stimulated with serum samples from DM and NM patients, subsequently followed by RT-PCR for the following candidates: IL-1β, IL-6, nestin, and CD31. Results Seventeen (17) DM patients, 7 NM patients, and 40 non-myositis controls were included. CD133+/VEGFR-2+ cells did not differ between the groups. Both DM and NM patients showed lower CFU-ECs than controls. In DM, intramuscular CD31 abundances were significantly reduced, which indicated vascular rarefaction. Muscular CXCR6 was elevated in both diseases. Circulating CXCL-16 was higher in DM and NM in contrast, compared to controls. Serum from patients with DM but not NM induced a profound upregulation of mRNS expression of CD31 and IL-6 in cultured myotubes. Conclusion Our study demonstrates the loss of intramuscular microvessels in DM, accompanied by endothelial activation in DM and NM. Vascular regeneration was impaired in DM and NM. The findings suggest a role for inflammation-associated vascular damage in the pathogenesis of DM.
Collapse
Affiliation(s)
- D. Lemmer
- Department of Nephrology and Rheumatology, University Medical Center Göttingen, Göttingen, Germany
- Immanuel Krankenhaus Berlin, Medical Center of Rheumatology Berlin-Buch, Berlin, Germany
| | - J. Schmidt
- Department of Neurology and Pain Treatment, Immanuel Klinik Rüdersdorf, University Hospital of the Brandenburg Medical School Theodor Fontane, Rüdersdorf bei Berlin, Germany
- Faculty of Health Sciences Brandenburg, Brandenburg Medical School Theodor Fontane, Rüdersdorf bei Berlin, Germany
- Department of Neurology, Neuromuscular Center, University Medical Center Göttingen, Göttingen, Germany
| | - K. Kummer
- Department of Neurology, Neuromuscular Center, University Medical Center Göttingen, Göttingen, Germany
| | - B. Lemmer
- Department of Physics, Georg-August-University Göttingen, Göttingen, Germany
| | - A. Wrede
- Department of Neuropathology, University Medical Center Göttingen, Göttingen, Germany
| | - C. Seitz
- Department of Dermatology, Allergology and Venereology, University Medical Center Göttingen, Göttingen, Germany
| | - P. Balcarek
- Department of Trauma Surgery, Orthopedics and Plastic Surgery, University Medical Center Göttingen, Göttingen, Germany
- Arcus Klinik, Pforzheim, Germany
| | - K. Schwarze
- Department of Nephrology and Rheumatology, University Medical Center Göttingen, Göttingen, Germany
| | - G. A. Müller
- Department of Nephrology and Rheumatology, University Medical Center Göttingen, Göttingen, Germany
| | - D. Patschan
- Faculty of Health Sciences Brandenburg, Brandenburg Medical School Theodor Fontane, Rüdersdorf bei Berlin, Germany
- Department of Medicine 1, Cardiology, Angiology, and Nephrology, University Hospital Brandenburg of the Brandenburg Medical School Theodor Fontane, Branderburg, Germany
| | - S. Patschan
- Faculty of Health Sciences Brandenburg, Brandenburg Medical School Theodor Fontane, Rüdersdorf bei Berlin, Germany
- Department of Medicine 1, Cardiology, Angiology, and Nephrology, University Hospital Brandenburg of the Brandenburg Medical School Theodor Fontane, Branderburg, Germany
- *Correspondence: S. Patschan
| |
Collapse
|
5
|
Chen K, Li Y, Xu L, Qian Y, Liu N, Zhou C, Liu J, Zhou L, Xu Z, Jia R, Ge YZ. Comprehensive insight into endothelial progenitor cell-derived extracellular vesicles as a promising candidate for disease treatment. Stem Cell Res Ther 2022; 13:238. [PMID: 35672766 PMCID: PMC9172199 DOI: 10.1186/s13287-022-02921-0] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2022] [Accepted: 05/29/2022] [Indexed: 12/21/2022] Open
Abstract
Endothelial progenitor cells (EPCs), which are a type of stem cell, have been found to have strong angiogenic and tissue repair capabilities. Extracellular vesicles (EVs) contain many effective components, such as cellular proteins, microRNAs, messenger RNAs, and long noncoding RNAs, and can be secreted by different cell types. The functions of EVs depend mainly on their parent cells. Many researchers have conducted functional studies of EPC-derived EVs (EPC-EVs) and showed that they exhibit therapeutic effects on many diseases, such as cardiovascular disease, acute kidney injury, acute lung injury, and sepsis. In this review article, we comprehensively summarized the biogenesis and functions of EPCs and EVs and the potent role of EPC-EVs in the treatment of various diseases. Furthermore, the current problems and future prospects have been discussed, and further studies are needed to compare the therapeutic effects of EVs derived from various stem cells, which will contribute to the accelerated translation of these applications in a clinical setting.
Collapse
Affiliation(s)
- Ke Chen
- Department of Urology, Nanjing First Hospital, Nanjing Medical University, No. 68 Changle Road, Nanjing, 210006, Jiangsu, People's Republic of China
| | - Yang Li
- Department of Urology, Nanjing First Hospital, Nanjing Medical University, No. 68 Changle Road, Nanjing, 210006, Jiangsu, People's Republic of China
| | - Luwei Xu
- Department of Urology, Nanjing First Hospital, Nanjing Medical University, No. 68 Changle Road, Nanjing, 210006, Jiangsu, People's Republic of China
| | - Yiguan Qian
- Department of Urology, Nanjing First Hospital, Nanjing Medical University, No. 68 Changle Road, Nanjing, 210006, Jiangsu, People's Republic of China
| | - Ning Liu
- Department of Urology, Nanjing First Hospital, Nanjing Medical University, No. 68 Changle Road, Nanjing, 210006, Jiangsu, People's Republic of China
| | - Changcheng Zhou
- Department of Urology, Nanjing First Hospital, Nanjing Medical University, No. 68 Changle Road, Nanjing, 210006, Jiangsu, People's Republic of China
| | - Jingyu Liu
- Department of Urology, Nanjing First Hospital, Nanjing Medical University, No. 68 Changle Road, Nanjing, 210006, Jiangsu, People's Republic of China
| | - Liuhua Zhou
- Department of Urology, Nanjing First Hospital, Nanjing Medical University, No. 68 Changle Road, Nanjing, 210006, Jiangsu, People's Republic of China
| | - Zheng Xu
- Department of Urology, Nanjing First Hospital, Nanjing Medical University, No. 68 Changle Road, Nanjing, 210006, Jiangsu, People's Republic of China
| | - Ruipeng Jia
- Department of Urology, Nanjing First Hospital, Nanjing Medical University, No. 68 Changle Road, Nanjing, 210006, Jiangsu, People's Republic of China.
| | - Yu-Zheng Ge
- Department of Urology, Nanjing First Hospital, Nanjing Medical University, No. 68 Changle Road, Nanjing, 210006, Jiangsu, People's Republic of China.
| |
Collapse
|
6
|
NOX1 and NOX2: two enzymes that promote endothelial-to-mesenchymal transition induced by melanoma conditioned media. Pharmacol Res 2022; 177:106097. [DOI: 10.1016/j.phrs.2022.106097] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Revised: 01/20/2022] [Accepted: 01/21/2022] [Indexed: 12/11/2022]
|
7
|
Gaut JP, Liapis H. Acute kidney injury pathology and pathophysiology: a retrospective review. Clin Kidney J 2020; 14:526-536. [PMID: 33623675 PMCID: PMC7886540 DOI: 10.1093/ckj/sfaa142] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Indexed: 12/12/2022] Open
Abstract
Acute kidney injury (AKI) is the clinical term used for decline or loss of renal function. It is associated with chronic kidney disease (CKD) and high morbidity and mortality. However, not all causes of AKI lead to severe consequences and some are reversible. The underlying pathology can be a guide for treatment and assessment of prognosis. The Kidney Disease: Improving Global Outcomes guidelines recommend that the cause of AKI should be identified if possible. Renal biopsy can distinguish specific AKI entities and assist in patient management. This review aims to show the pathology of AKI, including glomerular and tubular diseases.
Collapse
Affiliation(s)
- Joseph P Gaut
- Department of Pathology and Immunology and Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Helen Liapis
- Department of Pathology and Immunology and Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| |
Collapse
|
8
|
Winter RL, Tian Y, Caldwell FJ, Seeto WJ, Koehler JW, Pascoe DA, Fan S, Gaillard P, Lipke EA, Wooldridge AA. Cell engraftment, vascularization, and inflammation after treatment of equine distal limb wounds with endothelial colony forming cells encapsulated within hydrogel microspheres. BMC Vet Res 2020; 16:43. [PMID: 32019556 PMCID: PMC7001230 DOI: 10.1186/s12917-020-2269-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Accepted: 01/27/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Endothelial colony forming cells (ECFCs) may be useful therapeutically in conditions with poor blood supply, such as distal limb wounds in the horse. Encapsulation of ECFCs into injectable hydrogel microspheres may ensure cell survival and cell localization to improve neovascularization and healing. Autologous ECFCs were isolated from 6 horses, labeled with quantum nanodots (QD), and a subset were encapsulated in poly(ethylene) glycol fibrinogen microspheres (PEG-Fb MS). Full-thickness dermal wounds were created on each distal limb and injected with empty PEG-Fb MS, serum, ECFCs, or ECFCs encapsulated into PEG- Fb MS (ECFC/MS). Analysis included wound surface area (WSA), granulation tissue scoring (GS), thermography, collagen density staining, and immunohistochemical staining for endothelial and inflammatory cells. The purpose of this study was to track cell location and evaluate wound vascularization and inflammatory response after injection of ECFC/MS or naked ECFCs in equine distal limb wounds. RESULTS ECFCs were found near and within newly formed blood vessels up to 3 weeks after injection. ECFC and ECFC/MS groups had the greatest blood vessel quantity at week 1 in the wound periphery. Wounds treated with ECFCs and ECFC/MS had the lowest density of neutrophils and macrophages at week 4. There were no significant effects of ECFC or ECFC/MS treatment on other measured parameters. CONCLUSIONS Injection of microsphere encapsulated ECFCs was practical for clinical use and well-tolerated. The positive ECFC treatment effects on blood vessel density and wound inflammation warrant further investigation.
Collapse
Affiliation(s)
- Randolph L. Winter
- Department of Clinical Sciences, Auburn University, Auburn, AL USA
- Department of Clinical Sciences, Ohio State University, Columbus, OH USA
| | - Yuan Tian
- Department of Chemical Engineering, Auburn University, Auburn, AL USA
| | - Fred J. Caldwell
- Department of Clinical Sciences, Auburn University, Auburn, AL USA
| | - Wen J. Seeto
- Department of Chemical Engineering, Auburn University, Auburn, AL USA
| | - Jey W. Koehler
- Department of Pathobiology, Auburn University, Auburn, AL USA
| | | | - Shirley Fan
- Department of Mathematics, Auburn University, Auburn, AL USA
| | | | | | | |
Collapse
|
9
|
Patschan D, Schwarze K, Henze E, Hoffmann JC, Patschan S, Ritter O, Muller GA. Acute Kidney Injury-Associated Systemic Inflammation Is Aggravated in Insulin-Dependent Diabetes Mellitus. J Clin Med Res 2019; 11:720-723. [PMID: 31636787 PMCID: PMC6785280 DOI: 10.14740/jocmr3852] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2019] [Accepted: 08/12/2019] [Indexed: 12/11/2022] Open
Abstract
Background Acute kidney injury (AKI) significantly worsens the prognosis of hospitalized patients. Diabetes mellitus (DM) affects a growing number of individuals in the western world. DM subjects are at a higher risk for acquiring AKI during the stay at the hospital. The current study intended to quantify serum levels of specific immunomodulatory cytokines in diabetic mice suffering from AKI. Methods DM was induced in male C57/Bl6N mice by systemic injections of beta cell-toxic streptozotocin. Animals underwent bilateral renal ischemia (45 min) 6 weeks later. Results Post-ischemic diabetic mice showed significantly differing serum concentrations of the majority of all analytes as compared to untreated controls and non-diabetic (post-ischemic) animals. Conclusions Together, our data suggest DM-associated immune activation in AKI. One may suppose that inadequate stimulation of the humoral/cellular immune response potentially contributes to the higher ischemia susceptibility of the organ in DM.
Collapse
Affiliation(s)
- Daniel Patschan
- Klinik fur Kardiologie, Angiologie und Nephrologie, Innere Medizin I, Klinikum Brandenburg, Medizinische Hochschule Brandenburg, Brandenburg, Germany
| | - Katrin Schwarze
- Klinik fur Nephrologie und Rheumatologie, Universitatsmedizin Gottingen, Gottingen, Germany
| | - Elvira Henze
- Klinik fur Nephrologie und Rheumatologie, Universitatsmedizin Gottingen, Gottingen, Germany
| | | | - Susann Patschan
- Klinik fur Kardiologie, Angiologie und Nephrologie, Innere Medizin I, Klinikum Brandenburg, Medizinische Hochschule Brandenburg, Brandenburg, Germany
| | - Oliver Ritter
- Klinik fur Kardiologie, Angiologie und Nephrologie, Innere Medizin I, Klinikum Brandenburg, Medizinische Hochschule Brandenburg, Brandenburg, Germany
| | - Gerhard Anton Muller
- Klinik fur Nephrologie und Rheumatologie, Universitatsmedizin Gottingen, Gottingen, Germany
| |
Collapse
|
10
|
Molecular Mechanisms of the Acute Kidney Injury to Chronic Kidney Disease Transition: An Updated View. Int J Mol Sci 2019; 20:ijms20194941. [PMID: 31590461 PMCID: PMC6801733 DOI: 10.3390/ijms20194941] [Citation(s) in RCA: 85] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Revised: 10/03/2019] [Accepted: 10/04/2019] [Indexed: 02/08/2023] Open
Abstract
Increasing evidence has demonstrated the bidirectional link between acute kidney injury (AKI) and chronic kidney disease (CKD) such that, in the clinical setting, the new concept of a unified syndrome has been proposed. The pathophysiological reasons, along with the cellular and molecular mechanisms, behind the ability of a single, acute, apparently self-limiting event to drive chronic kidney disease progression are yet to be explained. This acute injury could promote progression to chronic disease through different pathways involving the endothelium, the inflammatory response and the development of fibrosis. The interplay among endothelial cells, macrophages and other immune cells, pericytes and fibroblasts often converge in the tubular epithelial cells that play a central role. Recent evidence has strengthened this concept by demonstrating that injured tubules respond to acute tubular necrosis through two main mechanisms: The polyploidization of tubular cells and the proliferation of a small population of self-renewing renal progenitors. This alternative pathophysiological interpretation could better characterize functional recovery after AKI.
Collapse
|
11
|
Patschan S, Vogt M, Bakhtiari D, Bramlage CP, Henze E, Muller GA, Krause A, Patschan D. Humoral and Cellular Patterns of Early Endothelial Progenitor Cells in Relation to the Cardiovascular Risk in Axial Spondylarthritis. J Clin Med Res 2019; 11:391-400. [PMID: 31143305 PMCID: PMC6522236 DOI: 10.14740/jocmr3441w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2018] [Accepted: 04/30/2018] [Indexed: 11/21/2022] Open
Abstract
Background Spondylarthritis (SpA) significantly affects sacroiliac, intervertebral and peripheral joints. Patients with SpA suffer from increased cardiovascular risk (CVR). The endothelial progenitor cell (EPC) system critically perpetuates vascular repair. The aim of the study was to evaluate circulating EPCs in axial (ax)SpA with special attention on parameters of disease activity and CVR. Methods Disease activity and functional impairment were quantified in 50 axSpA patients by using standardized parameters (Bath ankylosing spondylitis disease activity index (BASDAI), C-reactive protein (CRP), finger-floor distance (FFD) and Ott’ sign). Circulating EPCs and EPC regeneration were analyzed (fluorescence-activated cell sorting (FACS) and colony-forming unit (CFU) assay). Serum vasomodulatory mediators were quantified by enzyme-linked immunosorbent assay (ELISA). Results EPC colony numbers were lower in axSpA as compared to controls. Females displayed more colonies than males. In addition, fewer colonies were observed in smokers, in patients with a BASDAI of below 4 and in hypertension. Circulating CD133+/KDR+ cells did not differ between the groups. Follow-up analysis (33 months later) did not show any differences in gender, colony formation, CD133+/KDR+ cells or serum levels of vasomodulatory mediators if related to the categories of BASDAI, Ott’ sign or FFD. Conclusions EPC colony formation is significantly affected in axSpA with particularly low levels in males. EPC-related parameters do not allow predicting disease activity-related or functional parameters nor are they useful for CVR assessment in SpA.
Collapse
Affiliation(s)
- Susann Patschan
- Clinic of Nephrology and Rheumatology, University Hospital of Gottingen, Gottingen, Germany.,Department of Cardiology, Pulmonology, Angiology and Nephrology, Brandenburg Medical School, University Hospital Brandenburg, Brandenburg, Germany
| | - Maria Vogt
- Clinic of Nephrology and Rheumatology, University Hospital of Gottingen, Gottingen, Germany
| | - Donia Bakhtiari
- Clinic of Nephrology and Rheumatology, University Hospital of Gottingen, Gottingen, Germany
| | - Carsten Peter Bramlage
- Clinic of Nephrology and Rheumatology, University Hospital of Gottingen, Gottingen, Germany
| | - Elvira Henze
- Clinic of Nephrology and Rheumatology, University Hospital of Gottingen, Gottingen, Germany
| | - Gerhard Anton Muller
- Clinic of Nephrology and Rheumatology, University Hospital of Gottingen, Gottingen, Germany
| | - Andreas Krause
- Rheumatology and Clinical Immunology, Immanuel-Krankenhaus Berlin, Berlin, Germany
| | - Daniel Patschan
- Department of Cardiology, Pulmonology, Angiology and Nephrology, Brandenburg Medical School, University Hospital Brandenburg, Brandenburg, Germany
| |
Collapse
|
12
|
Imaging VEGF Receptors and α vβ 3 Integrins in a Mouse Hindlimb Ischemia Model of Peripheral Arterial Disease. Mol Imaging Biol 2019; 20:963-972. [PMID: 29687324 DOI: 10.1007/s11307-018-1191-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
PURPOSE To compare targeted imaging of vascular endothelial growth factor (VEGF) receptors vs. αvβ3 integrins in a mouse hindlimb ischemia model of peripheral artery disease. PROCEDURES Male wild-type (WT) C57BL/6 mice (8- to 10-week old) (n = 24) underwent left femoral artery ligation. The right leg served as control. Five days later, mice were injected with either VEGF receptor targeting [99mTc]DOTA-PEG-scVEGF ([99mTc]scV) (n = 8) or with αvβ3-targeting tracer [99mTc]HYNIC-cycloRGD ([99mTc]RGD) (n = 8) and underwent single photon emission computed tomography (SPECT) x-ray computed tomography imaging. To assess non-specific [99mTc]scV uptake, six additional mice received a mixture of [99mTc]scV and 30-fold excess of targeting protein, scVEGF. Tracer uptake as %ID was measured using volumetric regions encompassing the hindlimb muscles and as %ID/g from harvested limb muscles. Double and triple immunofluorescent analysis on tissue sections established localization of αvβ3, VEGFR-1, VEGFR-2, as well as certain cell lineage markers. RESULTS Tracer uptake, as %ID/g, was higher in ligated limbs of mice injected with [99mTc]scV compared to ligated hindlimbs in mice injected with [99mTc]RGD (p = 0.02). The ratio of tracer uptake for ligated/control hindlimb was borderline higher for [99mTc]scV than for [99mTc]RGD (p = 0.06). Immunofluorescent analysis showed higher prevalence of VEGFR-1, VEGFR-2, and αvβ3, in damaged vs. undamaged hindlimb tissue, but with little co-localization of these markers. Double immunofluorescent staining showed partial co-localization of VEGFR-1, VEGFR-2, and αvβ3, with endothelial cell marker FVIII, but not with CD31. Immunostaining for VEGFR-1 and VEGFR-2 additionally co-localized with lineage markers for endothelial progenitor cell and monocytes/macrophages, with a more diverse pattern of co-localization for VEGFR-2. CONCLUSION In a mouse hindlimb ischemia model of peripheral artery disease, [99mTc]scV SPECT tracer-targeting VEGF receptors showed a more robust signal than [99mTc]RGD tracer-targeting αvβ3. Immunofluorescent analysis suggests that uptake of [99mTc]scV and [99mTc]RGD in damaged tissue is due to non-overlapping cell populations and reflects different dynamic processes and that enhanced uptake of [99mTc]scV may be due to the presence of VEGF receptors on additional cell types.
Collapse
|
13
|
Zhang Z, Gao Y, Qiao X. WITHDRAWN: Spleen tyrosine kinase (SYK) protects renal tubular epithelial cell against hypoxia injury in children with acute kidney injury. Gene 2018:S0378-1119(18)31156-9. [PMID: 30408549 DOI: 10.1016/j.gene.2018.11.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2017] [Revised: 03/13/2018] [Accepted: 11/03/2018] [Indexed: 11/21/2022]
Abstract
This article has been withdrawn at the request of the author(s) and/or editor. The Publisher apologizes for any inconvenience this may cause. The full Elsevier Policy on Article Withdrawal can be found at https://www.elsevier.com/about/our-business/policies/article-withdrawal.
Collapse
Affiliation(s)
- Zhiyong Zhang
- Medicine School of Xi'an Jiaotong University, Xi'an 710061, Shaanxi, China
| | - Ya Gao
- Department of Pediatric Surgery, the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004, Shaanxi, China.
| | - Ximin Qiao
- Central Hospital of Xianyang, Xianyang 712000, Shaanxi, China
| |
Collapse
|
14
|
Ozkok A, Yildiz A. Endothelial Progenitor Cells and Kidney Diseases. Kidney Blood Press Res 2018; 43:701-718. [PMID: 29763891 DOI: 10.1159/000489745] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2018] [Accepted: 05/03/2018] [Indexed: 01/12/2023] Open
Abstract
Endothelial progenitor cells (EPC) are bone marrow derived or tissue-resident cells that play major roles in the maintenance of vascular integrity and repair of endothelial damage. Although EPCs may be capable of directly engrafting and regenerating the endothelium, the most important effects of EPCs seem to be depended on paracrine effects. In recent studies, specific microvesicles and mRNAs have been found to mediate the pro-angiogenic and regenerative effects of EPCs on endothelium. EPC counts have important prognostic implications in cardiovascular diseases (CVD). Uremia and inflammation are associated with lower EPC counts which probably contribute to increased CVD risks in patients with chronic kidney disease. Beneficial effects of the EPC therapies have been shown in studies performed on different models of CVD and kidney diseases such as acute and chronic kidney diseases and glomerulonephritis. However, lack of a clear definition and specific marker of EPCs is the most important problem causing difficulties in interpretation of the results of the studies investigating EPCs.
Collapse
Affiliation(s)
- Abdullah Ozkok
- University of Health Sciences, Umraniye Training and Research Hospital, Department of Nephrology, Istanbul, Turkey,
| | - Alaattin Yildiz
- Istanbul University, Istanbul Faculty of Medicine, Department of Nephrology, Istanbul, Turkey
| |
Collapse
|
15
|
Patschan D, Buschmann I, Ritter O, Kribben A. Cell-Based Therapies in Acute Kidney Injury (AKI). Kidney Blood Press Res 2018; 43:673-681. [PMID: 29734169 DOI: 10.1159/000489624] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Accepted: 04/26/2018] [Indexed: 11/19/2022] Open
Abstract
Acute kidney injury frequently occurs in hospitalized patients all over the world. The prognosis remains poor since specific therapies for promoting kidney regeneration/repair are still missing. In recent years cell-based strategies have improved AKI outcomes under experimental circumstances. Four groups of cells, each of them displaying certain biological and functional characteristics have been evaluated in AKI, induced Pluripotent Stem Cells (iPSCs), Spermatagonial Stem Cells (SSCs), Proangiogenic Cells (PACs) and Endothelial Colony Forming Cells (ECFCs), and Mesenchymal Stem Cells (MSCs). All of these have been documented to stabilize either parameters of kidney excretory dysfunction and/or certain morphological parameters. The mechanisms responsible for AKI protection include direct (cell incorporation) and indirect processes, the latter being mediated by humoral factors and particularly by the production of so-called extracellular vesicles. Cell-derived vesicular organelles have been shown to carry pro-regenerative micro-RNA molecules which stabilize the vascular and tubular function. The first trials in humans have been initiated, the majority of such trials employs MSCs. However, any transfer of cell-based strategies in the clinical practice is potentially associated with significant difficulties. These include cell availability, tolerance and competence. The article intends to summarize essential informations about all of the four populations mentioned above and to discuss implications for the management of human AKI.
Collapse
Affiliation(s)
- Daniel Patschan
- Innere Medizin I, Kardiologie, Angiologie, Nephrologie, Klinikum Brandenburg, Medizinische Hoch-schule Brandenburg, Brandenburg, Germany,
| | - Ivo Buschmann
- Innere Medizin I, Kardiologie, Angiologie, Nephrologie, Klinikum Brandenburg, Medizinische Hoch-schule Brandenburg, Brandenburg, Germany
| | - Oliver Ritter
- Innere Medizin I, Kardiologie, Angiologie, Nephrologie, Klinikum Brandenburg, Medizinische Hoch-schule Brandenburg, Brandenburg, Germany
| | - Andreas Kribben
- Klinik für Nephrologie, Universitätsklinikum Essen, Essen, Germany
| |
Collapse
|
16
|
Felice F, Piras AM, Rocchiccioli S, Barsotti MC, Santoni T, Pucci A, Burchielli S, Chiellini F, Ucciferri N, Solaro R, Altomare A, Cecchettini A, Di Stefano R. Endothelial progenitor cell secretome delivered by novel polymeric nanoparticles in ischemic hindlimb. Int J Pharm 2018. [PMID: 29526620 DOI: 10.1016/j.ijpharm.2018.03.015] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Endothelial progenitor cells (EPCs) contribute to ischemic tissue repair by paracrine secretion up-regulated by hypoxia. In this study we use novel nanoparticles (NPs) as carriers for a controlled release of EPC secretome (CM) to improve their angiogenic properties. The in vivo effect in ischemic hindlimb rat model was evaluated, comparing hypoxic EPC-CM-NPs with hypoxic EPC-CM alone. A proteomic characterization of hypoxic CM and the in vitro effect on endothelial cells (HUVECs) were also performed. Up to 647 protein, 17 of which with angiogenic properties, were upregulated by hypoxia. Moreover, hypoxic EPC-CM significantly promoted capillary-like structures on Matrigel. A significant increase of blood perfusion in ischemic limbs at 2 weeks with EPC-CM-loaded NPs as compared to both EPC-CM and control and a significant increase of capillary formation were observed. The use of EPC-CM-NPs significantly improved neoangiogenesis in vivo, underlining the advantages of controlled release in regenerative medicine.
Collapse
Affiliation(s)
- Francesca Felice
- Laboratory of Cardiovascular Research, Department of Surgical, Medical and Molecular Pathology and Critical Care Medicine, University of Pisa, Pisa, Italy
| | - Anna Maria Piras
- Laboratory of Bioactive Polymeric Materials for Biomedical and Environmental Applications (BIOLab), Department of Chemistry and Industrial Chemistry, University of Pisa, Pisa, Italy
| | | | - Maria Chiara Barsotti
- Laboratory of Cardiovascular Research, Department of Surgical, Medical and Molecular Pathology and Critical Care Medicine, University of Pisa, Pisa, Italy
| | - Tatiana Santoni
- Laboratory of Cardiovascular Research, Department of Surgical, Medical and Molecular Pathology and Critical Care Medicine, University of Pisa, Pisa, Italy
| | - Angela Pucci
- Histopathology Department, University Hospital, Pisa, Italy
| | - Silvia Burchielli
- Tuscany Gabriele Monasterio Foundation and Center of Experimental Biomedicine, CNR-National Research Council, Pisa, Italy
| | - Federica Chiellini
- Laboratory of Bioactive Polymeric Materials for Biomedical and Environmental Applications (BIOLab), Department of Chemistry and Industrial Chemistry, University of Pisa, Pisa, Italy
| | | | - Roberto Solaro
- Laboratory of Bioactive Polymeric Materials for Biomedical and Environmental Applications (BIOLab), Department of Chemistry and Industrial Chemistry, University of Pisa, Pisa, Italy
| | - Angelina Altomare
- Laboratory of Bioactive Polymeric Materials for Biomedical and Environmental Applications (BIOLab), Department of Chemistry and Industrial Chemistry, University of Pisa, Pisa, Italy
| | - Antonella Cecchettini
- Institute of Clinical Physiology, CNR, Pisa, Italy; Department of Clinical and Experimental Medicine, University of Pisa, Italy
| | - Rossella Di Stefano
- Laboratory of Cardiovascular Research, Department of Surgical, Medical and Molecular Pathology and Critical Care Medicine, University of Pisa, Pisa, Italy.
| |
Collapse
|
17
|
Zhang FQ, Lu W, Yuan WX, Li X. Regulation of c-Jun N-Terminal Protein Kinase (JNK) Pathway in Apoptosis of Endothelial Outgrowth Cells Induced by Asymmetric Dimethylarginine. Med Sci Monit 2017; 23:2535-2542. [PMID: 28546532 PMCID: PMC5455802 DOI: 10.12659/msm.904718] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2017] [Accepted: 04/18/2017] [Indexed: 01/03/2023] Open
Abstract
BACKGROUND Endothelial outgrowth cells (EOCs) are terminal endothelial progenitor cells (EPCs). Asymmetric dimethylarginine (ADMA) has been identified as a novel risk factor for cardiovascular diseases. Our aim in the present study was to investigate the effect of regulation of asymmetric dimethylarginine (ADMA) on EOCs apoptosis and to explore the underlining mechanisms of c-Jun N-terminal protein kinase (JNK) pathway in the process. MATERIAL AND METHODS EOCs were harvested from umbilical cord blood and obtained by using density gradient centrifugation and adhesive culture methods. Endothelial characteristics were identified by immunohistochemistry and fluorescence staining. EOCs were treated with different concentrations of ADMA and detected by flow cytometry. After JNK specific inhibitor (SP600125) was added, EOCs apoptosis protein expressions were measured by Western blot analysis. Proliferation, migration, and vascularization were detected by CCK-8 assay, wound healing assay, and tube-like formation assay, respectively. RESULTS EOCs were successfully extracted from umbilical cord blood and different concentrations of ADMA aggravated EOCs apoptosis. ADMA distinctly activates the phosphorylation activity of JNK. Supplementation of JNK-specific inhibitor (SP600125) decreased expression of Bax and cleaved caspase 3/9, and alleviated ADMA-induced apoptosis. SP600125 also promoted angiogenesis viability. CONCLUSIONS The JNK pathway participates in the apoptosis-promoting process of EOCs, and targeted inhibition of the JNK pathway can alleviate ADMA-induced injury, which I s the potential underlying mechanism of vascular endothelium injury in ischemic stroke.
Collapse
Affiliation(s)
| | | | | | - Xin Li
- Corresponding Author: Xin Li, e-mail:
| |
Collapse
|