Expanded Hemodialysis Therapy Ameliorates Uremia-Induced Systemic Microinflammation and Endothelial Dysfunction by Modulating VEGF, TNF-α and AP-1 Signaling

Rusan Catar, Guido Moll, Julian Kamhieh-Milz, Christian Luecht, Lei Chen, Hongfan Zhao, Lucas Ernst, Kevin Willy, Matthias Girndt, Roman Fiedler, Janusz Witowski, Henning Morawietz, Olle Ringdén, Duska Dragun, Kai-Uwe Eckardt, Ralf Schindler, Daniel Zickler, Rusan Catar, Guido Moll, Julian Kamhieh-Milz, Christian Luecht, Lei Chen, Hongfan Zhao, Lucas Ernst, Kevin Willy, Matthias Girndt, Roman Fiedler, Janusz Witowski, Henning Morawietz, Olle Ringdén, Duska Dragun, Kai-Uwe Eckardt, Ralf Schindler, Daniel Zickler

Abstract

Abstract: Systemic chronic microinflammation and altered cytokine signaling, with adjunct cardiovascular disease (CVD), endothelial maladaptation and dysfunction is common in dialysis patients suffering from end-stage renal disease and associated with increased morbidity and mortality. New hemodialysis filters might offer improvements. We here studied the impact of novel improved molecular cut-off hemodialysis filters on systemic microinflammation, uremia and endothelial dysfunction. Human endothelial cells (ECs) were incubated with uremic serum obtained from patients treated with two different hemodialysis regimens in the Permeability Enhancement to Reduce Chronic Inflammation (PERCI-II) crossover clinical trial, comparing High-Flux (HF) and Medium Cut-Off (MCO) membranes, and then assessed for their vascular endothelial growth factor (VEGF) production and angiogenesis. Compared to HF membranes, dialysis with MCO membranes lead to a reduction in proinflammatory mediators and reduced endothelial VEGF production and angiogenesis. Cytokine multiplex screening identified tumor necrosis factor (TNF) superfamily members as promising targets. The influence of TNF-α and its soluble receptors (sTNF-R1 and sTNF-R2) on endothelial VEGF promoter activation, protein release, and the involved signaling pathways was analyzed, revealing that this detrimental signaling was indeed induced by TNF-α and mediated by AP-1/c-FOS signaling. In conclusion, uremic toxins, in particular TNF-signaling, promote endothelial maladaptation, VEGF expression and aberrant angiogenesis, which can be positively modulated by dialysis with novel MCO membranes.

Translational perspective and graphical abstract: Systemic microinflammation, altered cytokine signaling, cardiovascular disease, and endothelial maladaptation/dysfunction are common clinical complications in dialysis patients suffering from end-stage renal disease. We studied the impact of novel improved medium-cut-off hemodialysis filters on uremia and endothelial dysfunction. We can show that uremic toxins, especially TNF-signaling, promote endothelial maladaptation, VEGF expression and aberrant angiogenesis, which can be positively modulated by dialysis with novel improved medium-cut-off membranes.

Trial registration: ClinicalTrials.gov NCT02084381.

Keywords: cardiovascular disease; chronic kidney disease; end-stage renal disease; endothelial cell (dys)function; expanded hemodialysis therapy; tumor necrosis factor alpha (TNF-alpha); uremic toxins / systemic microinflammation; vascular endothelial growth factor (VEGF).

Conflict of interest statement

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Copyright © 2021 Catar, Moll, Kamhieh-Milz, Luecht, Chen, Zhao, Ernst, Willy, Girndt, Fiedler, Witowski, Morawietz, Ringdén, Dragun, Eckardt, Schindler and Zickler.

Figures

Graphical Abstract
Graphical Abstract
Systemic microinflammation, altered cytokine signaling and cardiovascular diseases are common in hemodialysis patients contributing to the highly increased cardiovascular morbidity and mortality. One of the pathological causes is the endothelial maladaptation and dysfunction associated with uremia and chronic systemic microinflammation. We here elucidate the molecular and biological mechanisms how endothelial maladaptation is induced, and most importantly also how it can be reversed, with in vivo validation in a crossover randomized multi-center clinical study comparing novel improved medium-cut-off (MCO) dialyzers to standard-of-care high-flux (HF) dialyzers.
Figure 1
Figure 1
Study design and hypothesis: Expanded Hemodialysis Therapy ameliorates systemic Inflammation and endothelial maladaptation and dysfunction. (A) The medical need for Expanded Hemodialysis Therapy and clinical study design: The hemodialysis field has been shown a near exponential growth in the past decades, with >167.000 publications on PUBMED containing the search-term “Hemodialysis” in 2020. Recently, particular attention has been placed into lowering chronic treatment-associated adverse cardiovascular diseases (CVD) and new optimized treatment concepts, such as “Expanded Hemodialysis Therapy” with improved molecular cut-off hemodialyzers (8, 27). Within the PERCI-II study n=48 hemodialysis patients underwent crossover randomized multi-center comparison employing novel medium-cut-off (MCO; MCOI-Ci400, Gambro) dialyzers in comparison to standard of care high-flux (HF) hemodialyzers (PERCI-II-MCO; ClinicalTrials.gov: NCT02084381) (28). These novel MCO dialyzers have an improved molecular size cut-off, which positively modulates systemic microinflammation (28). (B) Goal of the follow up study: To Elucidate the Molecular and Biological Mechanisms: In the present study, we explore the molecular signaling mechanisms underlying this positive antiinflammatory shift and evaluate promising leads identified during the first screen in 2017. In particular, we study the modulation of TNF-superfamily members in sera of patients undergoing MCO dialysis and how this impacts on uremia- and TNF-α-induced endothelial maladaptation and dysfunction (left panel) and the molecular mechanisms (right panel), resulting in aberrant VEGF induction and angiogenesis. Our VEGF promoter activation studies and adjunct signaling pathway experiments elucidated that this detrimental uremia- and TNF-α-induced signaling is mediated via AP-1/c-FOS signaling and that alterations in the serum ratio between TNF-α and sTNF-R1, but not sTNF-R2, are potential indicators for endothelial maladaptation. These findings provide new avenues for molecular targets and treatment modalities to reduce chronic microinflammation in the context of hemodialysis.
Figure 2
Figure 2
Hemodialysis with improved MCO dialyzers normalizes endothelial VEGF production and maladaptive angiogenesis upon uremic serum exposure in vitro. (A) Schematics of patient serum collection for analysis of endothelial VEGF expression and angiogenesis/endothelial tube formation after stimulation of ECs with respective sera. Within the PERCI-II study, patients underwent alternating hemodialysis (HD; in 4 weeks intervals) with either high-flux (HF) or medium cut-off (MCO) dialyzers (n=23-25 patients). Upon a 4-weeks wash-in phase on HF dialyzers, patients were allocated for 4 weeks to either HF or MCO dialyzers, followed by a 4-weeks wash-out phase on HF dialyzers, followed by a 12-weeks allocation to HF or MCO dialyzer. The top row shows regimen A (HF, MCO, HF, HF) and the bottom row shows regimen B (HF, HF, MCO, MCO). The sera/time points used for analysis in the second part of the figure are indicated with red stars: end of wash-in, end of phase 1, 2, 3; and (B, C) Endothelial VEGF mRNA expression (AU; arbitrary units, 3-hour stimulation; n=23-25), and (D, E) VEGF protein release (pg/ml) upon 24-hour stimulation with either 10% HF-HD or 10% MCO-HD serum (n=23-25), and (F, G) Endothelial tube formation (TMSL/field; n=23-25) upon stimulated with either 10% HF-HD or 10% MCO-HD serum for 16 hours, as compared to healthy serum (HS) controls. ANOVA, Mean ± SEM, with *P < 0.05, **P < 0.01, and ***P < 0.001. ns, not significant.
Figure 3
Figure 3
MCO dialysis alters the systemic TNF-α/s-TNF-R1-ratio correlating with endothelial protection in vitro and a beneficial shift in serum cytokine levels in vivo. The patients underwent the two different hemodialysis (HD) regimes as indicated in (either HF/MCO/MCO or MCO/HF/HF; n=23-25) and the levels of soluble mediators at the different time points were analyzed with multiplex Milliplex and Luminex technology (28). (A) Unsupervised clustering heat-map analysis of biomarkers (rows) and patients (columns); (B) Quantification of TNF-α, and sTNF-R1 and sTNF-R1-R2 and their corresponding ratios; (C) Correlation of the TNF-α/sTNF-R1- and TNF-α/sTNF-R2-ratios with endothelial VEGF release (pg/ml) or endothelial tube formation (TMSL/field) upon stimulation with either 10% HF or 10% MCO patient serum; and (D) Patient serum profiling for biomarkers of endothelial activation and systemic inflammation. [Legend to (B) and (D)] Central legend (simplified depiction corresponding to the clinical trial scheme shown in ) indicates the underlying color code of the samples/time points in the box plots from regimen and the length of their phases (Phase 1 and 2 four weeks and phase 3 eight weeks, abbreviated as P1, P2, and P3 with duration shown in brackets), which show cytokine analysis of serum samples at the end of HF wash-in phase (Phase 0, the standard proinflammatory baseline before start of Regimen A or B which indicated in red), and the end of Phase 1, 2, and 3 (HF shown in orange, and MCO shown in green in legend and corresponding box plots), and the analyzed samples correspond to the end of the phases (Corresponding to the red stars in Figure 2A). Each box plot is labeled with the corresponding dialysis filter device (HF or MCO) and the trial stage (P0, P1, P2, and P3), as indicate in the central legend. ANOVA, Box plots Tukey with interquartile range, with *P < 0.05, **P < 0.01, and ***P < 0.001. ns, not significant.
Figure 4
Figure 4
Elevated TNF-α and VEGF levels in uremic serum and VEGF induction in ECs by uremic serum, but omission of VEGF induction by TNF-α blockade. (A) Levels of TNF-α (pg/ml), sTNF-R1 (ng/ml) and VEGF (pg/ml), in sera derived from healthy control subjects or uremic hemodialysis patients (n=14), used to generate the healthy and uremic serum pools (HSP and USP, respectively; Mann-Whitney test, Box plots min-max range); (B, C) Kinetics and dose-response of endothelial cell (EC) VEGF mRNA (AU; arbitrary units; n=6) and protein production (pg/ml) in response to incubation with HSP or USP (both 2way-ANOVA); (B) To assess kinetics of VEGF production, the ECs were incubated for different time points (1-24 hours) with 10% serum with the peak of VEGF mRNA expression detected at 3 hours and maximal protein expression at 6-24 hours; (C) To assess the dose-response of VEGF production the ECs were incubated with different concentrations of (1-20% serum) with maximal VEGF mRNA expression and protein secretion being detected in response to 10-20% serum after 3 and 24 hours of incubation respectively; and (D–F) The effect of either: (D) Anti-IL-1 receptor antagonist Anakinra, or (E) Anti-TNF-α blocking antibody Infliximab, on human uremic serum-induced VEGF release in ECs. The cells were pre-treated with or without either Anakinra or Infliximab for 1 hour, followed by stimulation for 24 hours with either 10% USP or 10% HSP (n=7), and ECs were subsequently assessed for VEGF release (both 2way-ANOVA); and (F) Dose-dependent effect of 1-20% USP vs. 1-20% HSP on EC viability, with assessment of EC viability (% viable cells, n = 6) with the WST-8 cell viability assay after 24-hour stimulation (2way-ANOVA). Box plots min-max range with Mann-Whitney-test, other plots 2way-ANOVA-testing with mean ± SEM, with *P <0.05, **P <0.01, and ***P <0.001.
Figure 5
Figure 5
TNF-α concentration-dependent VEGF promoter activation and angiogenesis and VEGF promoter sequences responsive to TNF-α stimulation. (A–C) ECs were stimulated with different concentrations of TNF-α (1 to 1000 pg/ml) to assess: (A) VEGF promoter activation (RLU; relative luciferase activity, 3-hour stimulation with TNF-α, n=4) in cells transfected with the pLuc 2068 full-length luciferase-reporter construct; and (B) VEGF mRNA expression (AU; arbitrary units, 3-hour stimulation with TNF-α, n=4); and (C) Endothelial tube formation (TMSL/field; total master segment length per field averaged of 5 assessed fields per condition, 16-hour stimulation with TNF-α, n=12). (D) To identify VEGF promoter sequences responsive to TNF-α stimulation ECs were transfected with different VEGF promoter constructs subjected to progressive 5’-deletions (full-length -2018 construct and -1286, -267, and -52 deletion) and the relative luciferase activity (RLU, n=4) determined in cells stimulated for 6 hours with TNF-α (1 pg/mL or 1000 pg/ml) compared to unstimulated resting control cells, demonstrating a loss in VEGF promoter activity upon truncation of the promoter region spanning the positions -267 to -52, and simultaneous identification of a potential high affinity AP-1/c-FOS binding site at position -102 with computation analysis. (E) Validation of the AP-1/c-FOS transcription factor-binding site with EMSA (one representative experiment shown) using biotin-labeled double-stranded oligonucleotides targeting to the calculated AP-1/c-FOS positions at -95 to -119 of the corresponding VEGF promoter region. ECs were stimulated for 6 h with TNF-a (1 pg/mL or 1000 pg/ml) and nuclear fractions analyzed for formation of nuclear complexes with the c-FOS probe (labeled as a shift with arrow); and (F) Effect of sTNF-R1 (1, 10, and 100 ng/ml) on the modulation of TNF-α induced c-FOS mRNA-expression (AU; 3-hour stimulation with 1 or 1000 pg/ml TNF-α, n=4) with corresponding TNF-α/sTNF-R1 ratios (0.01, 0.1, 1.0, 10, 100, 1000). 1way-ANOVA with Mean ± SEM, with *P < 0.05, **P < 0.01, and ***P < 0.001. ns, not significant.
Figure 6
Figure 6
Analysis of the molecular signaling pathways underling TNF-α induced endothelial VEGF induction and angiogenesis identifies AP-1/cFOS-signaling. (A, B) Role of the AP-1/c-FOS signaling pathway in low and high TNF-α level-induced endothelial VEGF induction (AU; n=5) shown in (A) and in vitro angiogenesis/endothelial tube formation (TMSL/field; total master segment length per field averaged out of 5 assessed fields per condition, 16-hour stimulation with TNF-α, n=6) shown in (B). Cells were first pre-treated with or without AP-1 blocker SR-11302 (10 nM) for 1 hour, followed by stimulation for 16 hours with or without 1 or 1000 pg/ml TNF-α in the presence or absence TNF-α blocking antibody Infliximab (100 ug/ml). Mann-Whitney-test with mean ± SEM, *P < 0.05 and **P < 0.01.

References

    1. Eckardt KU, Coresh J, Devuyst O, Johnson RJ, Kottgen A, Levey AS, et al. . Evolving Importance of Kidney Disease: From Subspecialty to Global Health Burden. Lancet (2013) 382:158–69. doi: 10.1016/S0140-6736(13)60439-0
    1. Remuzzi G, Benigni A, Finkelstein FO, Grunfeld JP, Joly D, Katz I, et al. . Kidney Failure: Aims for the Next 10 Years and Barriers to Success. Lancet (2013) 382:353–62. doi: 10.1016/S0140-6736(13)60438-9
    1. Bikbov B, Purcell CA, Levey AS, Smith M, Abdoli A, Abebe M, et al. . Global, Regional, and National Burden of Chronic Kidney Disease, 1990–2017: A Systematic Analysis for the Global Burden of Disease Study 2017. Lancet (2020) 395:709–33. doi: 10.1016/S0140-6736(20)30045-3
    1. Nadim MK, Forni LG, Mehta RL, Connor MJ, Liu KD, Ostermann M, et al. . COVID-19-Associated Acute Kidney Injury: Consensus Report of the 25th Acute Disease Quality Initiative (ADQI) Workgroup. Nat Rev Nephrol (2020) 16:747–64. doi: 10.1038/s41581-020-00356-5
    1. Perico L, Benigni A, Casiraghi F, Ng LFP, Renia L, Remuzzi G. Immunity, Endothelial Injury and Complement-Induced Coagulopathy in COVID-19. Nat Rev Nephrol (2021) 17:46–64. doi: 10.1038/s41581-020-00357-4
    1. Ronco C, Reis T. Kidney Involvement in COVID-19 and Rationale for Extracorporeal Therapies. Nat Rev Nephrol (2020) 16:308–10. doi: 10.1038/s41581-020-0284-7
    1. Morawietz H, Julius U, Bornstein SR. Cardiovascular Diseases, Lipid-Lowering Therapies and European Registries in the COVID-19 Pandemic. Cardiovasc Res (2020) 116:e122–5. doi: 10.1093/cvr/cvaa176
    1. Ronco C, Clark WR. Haemodialysis Membranes. Nat Rev Nephrol (2018) 14:394–410. doi: 10.1038/s41581-018-0002-x
    1. Storr M, Ward RA. Membrane Innovation: Closer to Native Kidneys. Nephrol Dialysis Transplant (2018) 33:iii22–7. doi: 10.1093/ndt/gfy228
    1. Moll G, Ankrum JA, Kamhieh-Milz J, Bieback K, Ringden O, Volk HD, et al. . Intravascular Mesenchymal Stromal/Stem Cell Therapy Product Diversification: Time for New Clinical Guidelines. Trends Mol Med (2019) 25:149–63. doi: 10.1016/j.molmed.2018.12.006
    1. Barcia R, Nguyen S, O’Rourke B, Igo P, Tilles A, Miller B, et al. . Immune Reprogramming in Human Subjects After Extracorporeal Mesenchymal Stromal Cell Therapy. Cytotherapy (2020) 22:S11. doi: 10.1016/j.jcyt.2020.03.473
    1. Roemhild A, Otto NM, Moll G, Abou-El-Enein M, Kaiser D, Bold G, et al. . Regulatory T Cells for Minimising Immune Suppression in Kidney Transplantation: Phase I/IIa Clinical Trial. Bmj-Brit Med J (2020) 371. doi: 10.1136/bmj.m3734
    1. Sarnak MJ, Levey AS, Schoolwerth AC, Coresh J, Culleton B, Hamm LL, et al. . Kidney Disease as a Risk Factor for Development of Cardiovascular Disease: A Statement From the American Heart Association Councils on Kidney in Cardiovascular Disease, High Blood Pressure Research, Clinical Cardiology, and Epidemiology and Prevention. Circulation (2003) 108:2154–69. doi: 10.1161/01.CIR.0000095676.90936.80
    1. Go AS, Chertow GM, Fan D, McCulloch CE, Hsu CY. Chronic Kidney Disease and the Risks of Death, Cardiovascular Events, and Hospitalization. N Engl J Med (2004) 351:1296–305. doi: 10.1056/NEJMoa041031
    1. Blacher J, Guerin AP, Pannier B, Marchais SJ, London GM. Arterial Calcifications, Arterial Stiffness, and Cardiovascular Risk in End-Stage Renal Disease. Hypertension (2001) 38:938–42. doi: 10.1161/hy1001.096358
    1. Shroff R, Long DA, Shanahan C. Mechanistic Insights Into Vascular Calcification in CKD. J Am Soc Nephrology: JASN (2013) 24:179–89. doi: 10.1681/ASN.2011121191
    1. Zickler D, Luecht C, Willy K, Chen L, Witowski J, Girndt M, et al. . Tumour Necrosis Factor-Alpha in Uraemic Serum Promotes Osteoblastic Transition and Calcification of Vascular Smooth Muscle Cells via Extracellular Signal-Regulated Kinases and Activator Protein 1/c-FOS-Mediated Induction of Interleukin 6 Expression. Nephrol Dial Transplant (2018) 33:574–85. doi: 10.1093/ndt/gfx316
    1. Henaut L, Massy ZA. New Insights Into the Key Role of Interleukin 6 in Vascular Calcification of Chronic Kidney Disease. Nephrol Dial Transplant (2018) 33:543–8. doi: 10.1093/ndt/gfx379
    1. Henaut L, Mary A, Chillon JM, Kamel S, Massy ZA. The Impact of Uremic Toxins on Vascular Smooth Muscle Cell Function. Toxins (Basel) (2018) 10. doi: 10.3390/toxins10060218
    1. Zoccali C. Endothelial Dysfunction in CKD: A New Player in Town? Nephrol Dial Transplant (2008) 23:783–5. doi: 10.1093/ndt/gfm924
    1. Merino A, Portoles J, Selgas R, Ojeda R, Buendia P, Ocana J, et al. . Effect of Different Dialysis Modalities on Microinflammatory Status and Endothelial Damage. Clin J Am Soc Nephrology: CJASN (2010) 5:227–34. doi: 10.2215/CJN.03260509
    1. Anderson CE, Hamm LL, Batuman G, Kumbala DR, Chen CS, Kallu SG, et al. . The Association of Angiogenic Factors and Chronic Kidney Disease. BMC Nephrol (2018) 19:117. doi: 10.1186/s12882-018-0909-2
    1. Rothuizen TC, Ocak G, Verschuren JJ, Dekker FW, Rabelink TJ, Jukema JW, et al. . Candidate Gene Analysis of Mortality in Dialysis Patients. PloS One (2015) 10:e0143079. doi: 10.1371/journal.pone.0143079
    1. Stenvinkel P, Ketteler M, Johnson RJ, Lindholm B, Pecoits-Filho R, Riella M, et al. . IL-10, IL-6, and TNF-Alpha: Central Factors in the Altered Cytokine Network of Uremia–the Good, the Bad, and the Ugly. Kidney Int (2005) 67:1216–33. doi: 10.1111/j.1523-1755.2005.00200.x
    1. Gimbrone MA, Jr, Garcia-Cardena G. Endothelial Cell Dysfunction and the Pathobiology of Atherosclerosis. Circ Res (2016) 118:620–36. doi: 10.1161/CIRCRESAHA.115.306301
    1. Libby P, Luscher T. COVID-19 is, in the End, an Endothelial Disease. Eur Heart J (2020) 41:3038–44. doi: 10.1093/eurheartj/ehaa623
    1. Ronco C, Marchionna N, Brendolan A, Neri M, Lorenzin A, Martínez Rueda AJ. Expanded Haemodialysis: From Operational Mechanism to Clinical Results. Nephrol Dial Transplant (2018) 33:iii41–7. doi: 10.1093/ndt/gfy202
    1. Zickler D, Schindler R, Willy K, Martus P, Pawlak M, Storr M, et al. . Medium Cut-Off (MCO) Membranes Reduce Inflammation in Chronic Dialysis Patients-A Randomized Controlled Clinical Trial. PloS One (2017) 12:e0169024. doi: 10.1371/journal.pone.0169024
    1. Kirsch AH, Lyko R, Nilsson LG, Beck W, Amdahl M, Lechner P, et al. . Performance of Hemodialysis With Novel Medium Cut-Off Dialyzers. Nephrol Dial Transplant (2017) 32:165–72. doi: 10.1093/ndt/gfw310
    1. Cobo G, Lindholm B, Stenvinkel P. Chronic Inflammation in End-Stage Renal Disease and Dialysis. Nephrol Dial Transplant (2018) 33:iii35–40. doi: 10.1093/ndt/gfy175
    1. Girndt M, Fiedler R, Martus P, Pawlak M, Storr M, Bohler T, et al. . High Cut-Off Dialysis in Chronic Haemodialysis Patients. Eur J Clin Invest (2015) 45:1333–40. doi: 10.1111/eci.12559
    1. Yuan J, Guo Q, Qureshi AR, Anderstam B, Eriksson M, Heimburger O, et al. . Circulating Vascular Endothelial Growth Factor (VEGF) and its Soluble Receptor 1 (sVEGFR-1) are Associated With Inflammation and Mortality in Incident Dialysis Patients. Nephrol Dial Transplant (2013) 28:2356–63. doi: 10.1093/ndt/gft256
    1. Catar R, Witowski J, Wagner P, Annett Schramm I, Kawka E, Philippe A, et al. . The Proto-Oncogene C-Fos Transcriptionally Regulates VEGF Production During Peritoneal Inflammation. Kidney Int (2013) 84:1119–28. doi: 10.1038/ki.2013.217
    1. Catar R, Witowski J, Zhu N, Lucht C, Derrac Soria A, Uceda Fernandez J, et al. . IL-6 Trans-Signaling Links Inflammation With Angiogenesis in the Peritoneal Membrane. J Am Soc Nephrology: JASN (2017) 28:1188–99. doi: 10.1681/ASN.2015101169
    1. Ferrara N, Gerber HP. The Role of Vascular Endothelial Growth Factor in Angiogenesis. Acta Haematol (2001) 106:148–56. doi: 10.1159/000046610
    1. Domigan CK, Warren CM, Antanesian V, Happel K, Ziyad S, Lee S, et al. . Autocrine VEGF Maintains Endothelial Survival Through Regulation of Metabolism and Autophagy. J Cell Sci (2015) 128:2236–48. doi: 10.1242/jcs.163774
    1. Catar R, Moll G, Hosp I, Simon M, Luecht C, Zhao H, et al. . Transcriptional Regulation of Thrombin-Induced Endothelial VEGF Induction and Proangiogenic Response. Cells (2021) 10:910. doi: 10.3390/cells10040910
    1. Descamps-Latscha B, Herbelin A, Nguyen AT, Roux-Lombard P, Zingraff J, Moynot A, et al. . Balance Between IL-1 Beta, TNF-Alpha, and Their Specific Inhibitors in Chronic Renal Failure and Maintenance Dialysis. Relationships With Activation Markers of T Cells, B Cells, and Monocytes. J Immunol (1995) 154:882–92.
    1. van Riemsdijk-van Overbeeke IC, Baan CC, Hesse CJ, Loonen EH, Niesters HG, Zietse R, et al. . TNF-Alpha: mRNA, Plasma Protein Levels and Soluble Receptors in Patients on Chronic Hemodialysis, on CAPD and With End-Stage Renal Failure. Clin Nephrol (2000) 53:115–23.
    1. Speeckaert MM, Speeckaert R, Laute M, Vanholder R, Delanghe JR. Tumor Necrosis Factor Receptors: Biology and Therapeutic Potential in Kidney Diseases. Am J Nephrol (2012) 36:261–70. doi: 10.1159/000342333
    1. Naserian S, Leclerc M, Shamdani S, Uzan G. Current Preventions and Treatments of aGVHD: From Pharmacological Prophylaxis to Innovative Therapies. Front Immunol (2020) 11. doi: 10.3389/fimmu.2020.607030
    1. Maisonpierre PC, Suri C, Jones PF, Bartunkova S, Wiegand SJ, Radziejewski C, et al. . Angiopoietin-2, a Natural Antagonist for Tie2 That Disrupts In Vivo Angiogenesis. Science (1997) 277:55–60. doi: 10.1126/science.277.5322.55
    1. Eroglu E, Gottschalk B, Charoensin S, Blass S, Bischof H, Rost R, et al. . Development of Novel FP-Based Probes for Live-Cell Imaging of Nitric Oxide Dynamics. Nat Commun (2016) 7:10623. doi: 10.1038/ncomms10623
    1. Eroglu E, Saravi SSS, Sorrentino A, Steinhorn B, Michel T. Discordance Between eNOS Phosphorylation and Activation Revealed by Multispectral Imaging and Chemogenetic Methods. Proc Natl Acad Sci USA (2019) 116:20210–7. doi: 10.1073/pnas.1910942116
    1. Andrzejewska A, Catar R, Schoon J, Qazi TH, Sass FA, Jacobi D, et al. . Multi-Parameter Analysis of Biobanked Human Bone Marrow Stromal Cells Shows Little Influence for Donor Age and Mild Comorbidities on Phenotypic and Functional Properties. Front Immunol (2019) 10:2474. doi: 10.3389/fimmu.2019.02474
    1. Moll G, Rasmusson-Duprez I, von Bahr L, Connolly-Andersen AM, Elgue G, Funke L, et al. . Are Therapeutic Human Mesenchymal Stromal Cells Compatible With Human Blood? Stem Cells (2012) 30:1565–74. doi: 10.1002/stem.1111
    1. Moll G, Alm JJ, Davies LC, von Bahr L, Heldring N, Stenbeck-Funke L, et al. . Do Cryopreserved Mesenchymal Stromal Cells Display Impaired Immunomodulatory and Therapeutic Properties? Stem Cells (2014) 32:2430–42. doi: 10.1002/stem.1729
    1. Finkenzeller G, Sparacio A, Technau A, Marmé D, Siemeister G. Sp1 Recognition Sites in the Proximal Promoter of the Human Vascular Endothelial Growth Factor Gene are Essential for Platelet-Derived Growth Factor-Induced Gene Expression. Oncogene (1997) 15:669–76. doi: 10.1038/sj.onc.1201219
    1. Hegner B, Weber M, Dragun D, Schulze-Lohoff E. Differential Regulation of Smooth Muscle Markers in Human Bone Marrow-Derived Mesenchymal Stem Cells. J Hypertens (2005) 23:1191–202. doi: 10.1097/01.hjh.0000170382.31085.5d
    1. Giacca M, Zacchigna S. VEGF Gene Therapy: Therapeutic Angiogenesis in the Clinic and Beyond. Gene Ther (2012) 19:622–9. doi: 10.1038/gt.2012.17
    1. Marsico G, Martin-Saldaña S, Pandit A. Therapeutic Biomaterial Approaches to Alleviate Chronic Limb Threatening Ischemia. Adv Sci 8(7):2003119. doi: 10.1002/advs.202003119
    1. Qadura M, Terenzi DC, Verma S, Al-Omran M, Hess DA. Concise Review: Cell Therapy for Critical Limb Ischemia: An Integrated Review of Preclinical and Clinical Studies. Stem Cells (2018) 36:161–71. doi: 10.1002/stem.2751
    1. Belgore FM, Blann AD, Li-Saw-Hee FL, Beevers DG, Lip GY. Plasma Levels of Vascular Endothelial Growth Factor and its Soluble Receptor (SFlt-1) in Essential Hypertension. Am J Cardiol (2001) 87:805–7. doi: 10.1016/s0002-9149(00)01512-5
    1. Mazidi M, Rezaie P, Kengne AP, Stathopoulou MG, Azimi-Nezhad M, Siest S. VEGF, the Underlying Factor for Metabolic Syndrome; Fact or Fiction? Diabetes Metab Syndr (2017) 11(Suppl 1):S61–4. doi: 10.1016/j.dsx.2016.12.004
    1. Farber A, Eberhardt RT. The Current State of Critical Limb Ischemia: A Systematic Review. JAMA Surg (2016) 151:1070–7. doi: 10.1001/jamasurg.2016.2018
    1. Ernandez T, Mayadas T. Immunoregulatory Role of TNFa in Inflammatory Kidney Diseases. Kidney Int (2009) 76:262–76. doi: 10.1038/ki.2009.142
    1. Connolly-Andersen AM, Moll G, Andersson C, Akerstrom S, Karlberg H, Douagi I, et al. . Crimean-Congo Hemorrhagic Fever Virus Activates Endothelial Cells. J Virol (2011) 85:7766–74. doi: 10.1128/JVI.02469-10
    1. Sierra-Parraga JM, Merino A, Eijken M, Leuvenink H, Ploeg R, Møller BK, et al. . Reparative Effect of Mesenchymal Stromal Cells on Endothelial Cells After Hypoxic and Inflammatory Injury. Stem Cell Res Ther (2020) 11:352. doi: 10.1186/s13287-020-01869-3
    1. Nouri Barkestani M, Shamdani S, Afshar Bakshloo M, Arouche N, Bambai B, Uzan G, et al. . Tnfα Priming Through its Interaction With TNFR2 Enhances Endothelial Progenitor Cell Immunosuppressive Effect: New Hope for Their Widespread Clinical Application. Cell Commun Signal (2021) 19:1. doi: 10.1186/s12964-020-00683-x
    1. Zickler D, Willy K, Girndt M, Fiedler R, Martus P, Storr M, et al. . High Cut-Off Dialysis in Chronic Haemodialysis Patients Reduces Serum Procalcific Activity. Nephrol Dial Transplant (2016) 31:1706–12. doi: 10.1093/ndt/gfw293
    1. Willy K, Hulko M, Storr M, Speidel R, Gauss J, Schindler R, et al. . In Vitro Dialysis of Cytokine-Rich Plasma With High and Medium Cut-Off Membranes Reduces Its Procalcific Activity. Artif Organs (2017) 41:803–9. doi: 10.1111/aor.12884
    1. Moll G, Jitschin R, von Bahr L, Rasmusson-Duprez I, Sundberg B, Lonnies L, et al. . Mesenchymal Stromal Cells Engage Complement and Complement Receptor Bearing Innate Effector Cells to Modulate Immune Responses. PloS One (2011) 6:e21703. doi: 10.1371/journal.pone.0021703
    1. Moll G, Hult A, von Bahr L, Alm JJ, Heldring N, Hamad OA, et al. . Do ABO Blood Group Antigens Hamper the Therapeutic Efficacy of Mesenchymal Stromal Cells? PloS One (2014) 9:e85040. doi: 10.1371/journal.pone.0085040
    1. Moll G, Ignatowicz L, Catar R, Luecht C, Sadeghi B, Hamad O, et al. . Different Procoagulant Activity of Therapeutic Mesenchymal Stromal Cells Derived From Bone Marrow and Placental Decidua. Stem Cells Dev (2015) 24:2269–79. doi: 10.1089/scd.2015.0120
    1. Taye A, Sawamura T, Morawietz H. Aldosterone Augments LOX-1-Mediated Low-Density Lipoprotein Uptake in Human Umbilical Artery Endothelial Cells. Pharmacol Rep: PR (2010) 62:311–8. doi: 10.1016/S1734-1140(10)70271-1

Source: PubMed

3
Sottoscrivi