scholarly journals Antiplatelet agents maintain arteriovenous fistula and graft function in patients receiving hemodialysis: A nationwide case–control study

PLoS ONE ◽  
2018 ◽  
Vol 13 (10) ◽  
pp. e0206011 ◽  
Author(s):  
Yung-Ho Hsu ◽  
Yu-Chun Yen ◽  
Yi-Chun Lin ◽  
Li-Chin Sung
2006 ◽  
Vol 25 (5) ◽  
pp. 579-584 ◽  
Author(s):  
N. HUSSAIN ◽  
R. ALSULAIMAN ◽  
P. BURTIN ◽  
Y. TOUBOUTI ◽  
E. RAHME ◽  
...  

Blood ◽  
2019 ◽  
Vol 134 (Supplement_1) ◽  
pp. 4425-4425
Author(s):  
Yuan Kong ◽  
Xie-Na Cao ◽  
Zhong-Shi Lyu ◽  
Xiao-Dong Mo ◽  
Qi Wen ◽  
...  

Background: Endothelial cells(ECs) serve as an instructive platform to support hematopoietic stem cells(HSCs) for homeostasis, however the underlying mechanism by which ECs regulate HSCs remains unclear. In humans, poor graft function(PGF) is characterized by pancytopenia post allogeneic hematopoietic stem cell transplantation(allo-HSCT) and serves as an appropriate model to study hematopoiesis in real world. Our series of studies reported that the impaired BM ECs are responsible for defective hematopoiesis post allo-HSCT(2013 BBMT, 2014 BBMT, 2015 BMT, 2016 Blood, 2018 AJH, 2019 Blood Advances), whereas prophylactic strategies to improve BM ECs promote hematopoietic reconstitution post allo-HSCT(2019 Blood Advances), further indicating the critical role of BM ECs in regulating HSCs. Autophagy, an essential homeostatic process responsible for nutrient deprivation, can be activated as cytoprotective response. Conversely, over-activated autophagy contributes to cancer progression. Beclin-1, an autophagy-related protein, plays a critical role in the formation of autophagosomes. Accumulating evidence has demonstrated the essential roles of ECs in supporting HSCs and of autophagy in benefiting ECs, raising the question of whether EC autophagy plays a critical role in supporting HSCs. Moreover, the effect of EC autophagy on HSC support needs to be elucidated and validated in a human pancytopenia disease model. Aims: The current study was performed to investigate whether the autophagy status in ECs regulates their ability to support hematopoiesis. Moreover, we evaluated the effect of EC autophagy on HSC support in PGF patients, and the results may provide a promising therapeutic target for PGF patients post allo-HSCT. Methods: Human Umbilical Vein Endothelial Cells(HUVECs) were genetically knockdown or overexpressed of Beclin-1. RNA-seq analyses were performed to elucidate the regulatory mechanism underlying hematopoietic supporting ability of HUVECs, which were further confirmed by qRT-PCR. In vitro pharmacologic regulations of autophagy(Rapamycin, an autophagy activator, and Hydroxychloroquine, an autophagy inhibitor) were administrated to HUVECs. Subsequently, a prospective case-control study was conducted. The levels of autophagy-related markers(LC3, Beclin1, and p62), and intracellular autophagosomes in BM ECs from PGF patients and their matched good graft function(GGF) patients were evaluated by monodansylcadaverine(MDC) staining, flow cytometry, western blot and transmission electron microscopy. HUVECs or BM ECs from PGF patients were cocultured with normal CD34+ cells. The quantity and function, especially the HSCs supporting ability of HUVECs or BM ECs from PGF patients were evaluated by colony-forming unit assay after Rapamycin treatment. Results: Inhibiting autophagy by Beclin-1 knockdown significantly reduced the hematopoiesis-supporting ability of HUVECs, which could be restored by activating autophagy through up-regulating Beclin-1. During the above process, autophagy positively regulated hematopoiesis-regulating genes in HUVECs. In addition, in vitro pharmacologic regulations of autophagy by Rapamycin or Hydroxychloroquine could modulate the hematopoietic supporting ability of HUVECs through regulating autophagy. Moreover, genetic and pharmacologic down-regulating autophagy in HUVECs impaired their quantity and function, which could be restored by up-regulating autophagy in HUVECs. Subsequently, the prospective case-control study demonstrated that defective autophagy, reduced Beclin-1 expression and deficient CFU plating efficiency in BM ECs from PGF patients when compared to their matched GGF patients. Importantly, Rapamycin quantitatively and functionally improved the impaired BM ECs from PGF patients in vitro, and especially enhanced their ability to support HSCs by activating the defective autophagy. Summary / Conclusions: Our results suggest that the autophagy status of ECs modulates their ability to support hematopoiesis by regulating the Beclin-1 pathway. Defective autophagy in BM ECs may be involved in the pathogenesis of PGF post allo-HSCT. Although further validation is required, our data suggest that Rapamycin could improve the impaired HSC-supporting ability of ECs by activating autophagy, thus providing a promising therapeutic approach for PGF patients. Disclosures No relevant conflicts of interest to declare.


2019 ◽  
Author(s):  
Maria Vargas ◽  
Pasquale Buonanno ◽  
Andrea Sica ◽  
Emanuele Sabatella ◽  
Francesco P. D’Alessio ◽  
...  

Abstract Background Rocuronium can be used in patients with severe renal failure (creatinine clearance <30 ml/min) but the duration of muscle relaxation is longer and results in an increased risk of postoperative residual curarization. Rocuronium can be antagonized by sugammadex but the elimination of the complex they made (rocuronium-sugammadex complex) varies according to the renal function. Two case reports/series reported the use of rocuronium – sugammadex complex during renal transplantation. This retrospective case-control study aimed to investigate the effects of rocuronium – sugammadex, used during the renal transplantation, on transplanted kidney function.Methods We analyzed 113 medical records of patients undergoing kidney transplantation from January 2015 to December 2018. 47 medical records were excluded because they did not report the administration of rocuronium + sugammadex or cisatracurium + neostigmine during the transplantation. For each medical records the following data were recorded the characteristics of patients and of kidney donor. Blood creatinine, blood urea, blood sodium, blood potassium, blood calcium levels were collected at the day before the surgery, the day of surgery, after 6 hours (hrs), 12 hrs, 24 hrs, 48 hrs and 72 hrs. Diuresis, urinary sodium and urinary potassium levels were collected at the day of surgery, after 6 hrs, 12 hrs, 24 hrs, 48 hrs and 72 hrs. Results We collected data from 66 medical reports. Blood creatinine levels at 6 hrs, 12 hrs and 24 hrs were significantly lower in roc + sug group than cis + neo group (crea 6 hrs =0.05, crea 12 hrs p=0.038, crea 24 hrs p= 0.049). Blood urea levels for 24 hrs after the transplantation were significantly lower in in roc + sug group than cis + neo group (urea 0 hrs p=0.025, urea 6 hrs p= 0.011, urea 12 hrs p=0.03, urea 24 hrs p=0.011). We found no statistically significant differences in blood sodium, blood potassium, blood calcium, diuresis, urinary sodium, urinary potassium levels before and after the transplantation.Conclusions In this retrospective case-control study, the use of rocuronium and sugammadex during the renal transplant surgery did not affect the recovery of the graft function during the first week after the transplantation.


Blood ◽  
2014 ◽  
Vol 124 (21) ◽  
pp. 3883-3883
Author(s):  
Yuan Kong ◽  
Yue Hu ◽  
Yuan-yuan Zhang ◽  
Yu-Hong Chen ◽  
Wei Han ◽  
...  

Abstract Background: Poor graft function (PGF), defined as the presence of 2 or 3 cytopenic counts (ANC ≤0.5×109/L and PLT ≤20×109/L or Hb ≤70 g/L) for at least 3 consecutive days beyond day +28 post-transplantation with transfusion requirement, in the presence of complete donor chimerism, and in the absence of severe graft-versus-host disease (GVHD) or hematological relapse, remains a serious complication after allogeneic hematopoietic stem cell transplantation (allo-HSCT). Emerging evidence from mouse studies suggested that effective hematopoiesis depends on a particular bone marrow (BM) microenvironment in which hematopoietic stem cells (HSCs) reside. We recently found that the impaired BM microenvironment may contribute to the occurrence of PGF post-HSCT using a prospective nested case-control study (Kong Y, et al. Biol Blood Marrow Transplant.2013;19:1465-1473). However, it remains unclear whether the quantitative and functional abnormalities of HSCs are involved in the pathogenesis of PGF. Aims: To evaluate whether the quantitative and functional abnormalities of CD34+cells in BM are involved in the pathogenesis of PGF after allo-HSCT. Methods: The normal multilineage differentiation capacity of the CD34+ cells sorted from the donors BM of PGF and good graft function (GGF) patients underwent allo-HSCT were investigated in vivo using sublethally irradiated NOD-PrkdcscidIL2rgnull mice by intra-BM injection. Subsequently, a prospective nested case-control study was performed enrolling 15 patients with PGF, 30 matched patients with GGF after allo-HSCT and their healthy donors (HDs). To minimize the potential influence of the length of time after allo-HSCT, the PGF and GGF patients had their HSCs tested at a matched median time point after allo-HSCT. Quantification of CD34+ cells was performed by flow cytometry. Owning to the limited numbers of CD34+cells in PGF patients, colony-forming cell (CFC) assay, instead of xenograft assay, was performed to investigate the multi-pluripotency of post-HSCT HSCs in vitro. Additionally, the quiescent cell cycle status and reactive oxygen species (ROS) production of HSCs in the post-HSCT BM were further analyzed by flow cytometry in the aforementioned three groups of subjects. The study was approved by the Ethics Committee of Peking University People’s Hospital and written informed consent was obtained from all subjects. Results: CD34+ BM cells from donors of PGF and GGF patients demonstrated no significant differences of normal multilineage differentiation capacity in NOD-PrkdcscidIL2rgnull mice. In the prospective nested case-control study, the demographic and clinical characteristics showed no significant difference between allo-HSCT patients with PGF and those with GGF. The percentage of CD34+ cells significantly decreased in the BM of PGF patients post-HSCT, although similar amounts of CD34+ cells were transplanted. We observed a remarkable decrease in stem cell/progenitor frequency in post-HSCT CD34+ BM cells of PGF patients as determined by CFC assay. Additionally, post-HSCT BM of PGF patients contained a significantly lower number of quiescent cells in the CD34+CD38low fraction compared to the BM of GGF patients and HDs. Moreover, significantly higher levels of ROS were observed in BM CD34+, CD34+CD38low, CD34+CD38high fractions of PGF patients following allo-HSCT compared to that in GGF patients and HDs. Summary/Conclusion: Although the frequency and function of CD34+ BM cells were revealed normal pre-HSCT, CD34+ BM cells were quantitatively and functionally impaired in patients with PGF following allo-HSCT. Our data indicate that the diminished regenerative capacity of BM of PGF patients following allo-HSCT is possibly related to a loss of quiescence and a reduced tolerability of CD34+ cells to oxidative stress. Acknowledgment: Supported by the National Natural Science Foundation of China (grant nos. 81370638&81230013), the Beijing Municipal Science and Technology Program (grant no. Z141100000214011), and Peking University People’s Hospital Research and Development Funds (grant no. RDB2012-23). Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2015 ◽  
Vol 126 (23) ◽  
pp. 4337-4337
Author(s):  
Yuan Kong ◽  
Yue Hu ◽  
Yang Song ◽  
Yu-Tong Wang ◽  
Zheng-Fan Jiang ◽  
...  

Abstract Background: Poor graft function (PGF) is a serious complication after allogeneic hematopoietic stem cell transplantation (allo-HSCT). However, the mechanisms underlying PGF remain to be elucidated, which increases the difficulty of managing PGF. In murine study, effective cross-talk between hematopoietic stem cells (HSCs) and bone marrow (BM) micro-environment plays an important role in hematopoiesis. HSCs occupy a hypoxic BM micro-environment to protect them from oxidative stress, whereas excessive reactive oxygen species (ROS) could inhibit HSCs self-renewal and induce HSCs exhaustion resulting in hematopoietic dysfunction. We recently reported that the impaired BM micro-environment may contribute to the occurrence of PGF post-HSCT using a prospective nested case-control study (Kong Y, et al. Biol Blood Marrow Transplant. 2013;19:1465-1473). Nevertheless, it is largely unknown whether the quantitatively and functionally impaired HSCs pre- and post-HSCT operate in the occurrence of PGF in allotransplants patients. Aims: To investigate whether the quantitative and functional abnormalities of the donor BM CD34+ cells pre- and post-HSCT are involved in the pathogenesis of PGF. Methods: The hematopoietic reconstitution activities of the CD34+ cells, sorted from the donors' BM of PGF and good graft function (GGF) patients, were evaluated in xenografted NOD-Prkdcscid IL2rgnull mice as an indicator of donors' HSCs function. To further investigate the effect of oxidative stress on normal hematopoiesis post-HSCT, the BM CD34+ cells of GGF allotransplant patients were treated of hydrogen peroxide (H2O2) with or without antioxidant N-acetyl-L-cysteine (NAC) in vitro. Subsequently, a prospective nested case-control study was performed enrolling 15 patients with PGF, 30 matched patients with GGF after allo-HSCT and their healthy donors. Quantification of the frequency, intracellular ROS levels, and cell cycle status of the BM CD34+ cells were analyzed by flow cytometry pre- and post-HSCT. Colony-forming capacity was investigated in CD34+ cells post-HSCT in vitro. The study was approved by the Ethics Committee of Peking University People's Hospital and written informed consent was obtained from all subjects. Results: The hematopoietic reconstitution activity of the BM CD34+ cells in NOD-Prkdcscid IL2rgnull mice demonstrated no significant differences between the donors of PGF and GGF patients. In the subsequent in vitro study, increased ROS were found to play an important role in the exhaustion of the quiescent BM CD34+ cells of GGF patients, whereas treatment of ROS-abrogated CD34+ cells with the antioxidant NAC could partially, but significantly restore the exhaustion and colony-forming capacity of CD34+ cells. In the prospective nested case-control study, all patient- and therapy-related variables were similar between patients with PGF and GGF. Polymerase chain reaction DNA fingerprinting of the STRs confirmed 100% donor chimerism in these patients. The frequency, intracellular ROS levels and cell cycle status of the transplanted donor BM CD34+ cells showed no remarkable differences pre-HSCT. Nevertheless, the percentage of CD34+ cells post-HSCT and their colony-forming capacity, especially the quiescent CD34+ CD38low fraction, decreased remarkably in PGF patients when compared to that in GGF patients. Notably, significantly increased ROS levels were observed in CD34+ and CD34+ CD38low fractions of PGF patients post-HSCT. Summary/Conclusion: Although the frequency and function of the transplanted donor BM CD34+ cells of PGF were demonstrated normal pre-HSCT, the increased levels of ROS and exhaustion of the quiescent CD34+ cells may operate in PGF post-HSCT. Our preliminary data indicate that an impaired BM micro-environment which may hamper the hematopoietic reconstitution of the donor HSCs in the recipients, rather than the defective donor HSCs, was involved in the occurrence of PGF. Therefore, novel therapeutic approaches, such as antioxidative therapy to maintain hypoxia BM micro-environment, promise to facilitate hematopoietic recovery in PGF. Acknowledgment: Supported by the National Natural Science Foundation of China (grant nos. 81370638&81230013), and the Beijing Municipal Science and Technology Program (grant nos. Z141100000214011& Z151100004015164& Z151100001615020). Disclosures No relevant conflicts of interest to declare.


2016 ◽  
Vol 25 (8) ◽  
pp. 889-897 ◽  
Author(s):  
Chao-Feng Lin ◽  
Hung-Yi Chiou ◽  
Ya-Hui Chang ◽  
Ju-Chi Liu ◽  
Yen-Ni Hung ◽  
...  

Sign in / Sign up

Export Citation Format

Share Document