Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
1.
Front Med (Lausanne) ; 8: 708316, 2021.
Article in English | MEDLINE | ID: mdl-34540864

ABSTRACT

Background: Delayed graft function (DGF) is a common complication after kidney transplantation (KT) with a poor clinical outcome. There are no accurate biomarkers for the early prediction of DGF. Macrophage migration inhibitory factor (MIF) release during surgery plays a key role in protecting the kidney, and may be a potential biomarker for predicting post-transplant renal allograft recovery. Methods: Recipients who underwent KT between July 2020 and December 2020 were enrolled in the study. Plasma MIF levels were tested in recipients at different time points, and the correlation between plasma MIF and DGF in recipients was evaluated. This study was registered in the Chinese Clinical Trial Registry (ChiCTR2000035596). Results: Intraoperative MIF levels were different between immediate, slowed, and delayed graft function groups (7.26 vs. 6.49 and 5.59, P < 0.001). Plasma MIF was an independent protective factor of DGF (odds ratio = 0.447, 95% confidence interval [CI] 0.264-0.754, P = 0.003). Combining plasma MIF level and donor terminal serum creatinine provided the best predictive power for DGF (0.872; 95%CI 0.795-0.949). Furthermore, plasma MIF was significantly associated with allograft function at 1-month post-transplant (R 2 = 0.42, P < 0.001). Conclusion: Intraoperative MIF, as an independent protective factor for DGF, has excellent diagnostic performance for predicting DGF and is worthy of further exploration.

2.
Aging (Albany NY) ; 13(6): 8440-8453, 2021 03 10.
Article in English | MEDLINE | ID: mdl-33714205

ABSTRACT

We previously showed that donor plasma mitochondrial DNA (dmtDNA) levels were correlated with renal allograft function. The aim of the current study was to determine whether dmtDNA levels are associated with the occurrence of antibody-mediated rejection (ABMR). This is a retrospective open cohort study comprised of 167 donors and 323 recipients enrolled from January 2015 to December 2017. We quantified the mtDNA level present in donor plasma using quantitative real-time polymerase chain reaction. The average plasma dmtDNA level in the acute rejection (AR) group was higher than that of the control group (0.156 versus 0.075, p<0.001). Multivariate logistic regression analysis showed that dmtDNA levels were also significantly associated with AR (OR=1.588, 95% CI 1.337-4.561, p<0.001). When the dmtDNA level was >0.156, the probability of AR was 62.9%. The plasma dmtDNA level in the ABMR group was significantly higher than that of the T cell-mediated rejection group (0.185 versus 0.099, p=0.032). The area under the receiver operating characteristic curve of dmtDNA for prediction of ABMR was as high as 0.910 (95% CI 0.843-0.977). We demonstrated that plasma dmtDNA was an independent risk factor for ABMR, which is valuable in organ evaluation. dmtDNA level is a possible first predictive marker for ABMR.


Subject(s)
Biomarkers/blood , DNA, Mitochondrial/blood , Graft Rejection/blood , Kidney Transplantation , Adult , Allografts , Cohort Studies , Female , Humans , Male , Middle Aged , Retrospective Studies , Tissue Donors
3.
Chin Med J (Engl) ; 133(5): 561-570, 2020 Mar 05.
Article in English | MEDLINE | ID: mdl-32053570

ABSTRACT

BACKGROUND: Although the use of expanded-criteria donors (ECDs) alleviates the problem of organ shortage, it significantly increases the incidence of delayed graft function (DGF). DGF is a common complication after kidney transplantation; however, the effect of DGF on graft loss is uncertain based on the published literature. Hence, the aim of this study was to determine the relationship between DGF and allograft survival. METHODS: We conducted a retrospective, multicenter, observation cohort study. A total of 284 deceased donors and 541 recipients between February 2012 and March 2017 were included. We used logistic regression analysis to verify the association between clinical parameters and DGF, and Cox proportional hazards models were applied to quantify the hazard ratios of DGF for kidney graft loss. RESULTS: Among the 284 deceased donors, 65 (22.8%) donors were ECD. Of the 541 recipients, 107 (19.8%) recipients developed DGF, and this rate was higher with ECD kidneys than with standard-criteria donor (SCD) kidneys (29.2% vs. 17.1%; P = 0.003). The 5-year graft survival rate was not significantly different between SCD kidney recipients with and without DGF (95.8% vs. 95.4%; P = 0.580). However, there was a significant difference between ECD kidney recipients with and without DGF (71.4% vs. 97.6%; P = 0.001), and the adjusted hazard ratio (HR) for graft loss for recipients with DGF was 1.885 (95% confidence interval [CI] = 1.305-7.630; P = 0.024). Results showed that induction therapy with anti-thymocyte globulin was protective against DGF (odds ratio = 0.359; 95% CI = 0.197-0.652; P = 0.001) with all donor kidneys and a protective factor for graft survival (HR = 0.308; 95% CI = 0.130-0.728; P = 0.007) with ECD kidneys. CONCLUSION: DGF is an independent risk factor for graft survival in recipients with ECD kidneys, but not SCD kidneys.


Subject(s)
Kidney Transplantation/methods , Adult , Cohort Studies , Female , Graft Survival/physiology , Humans , Male , Middle Aged , Multicenter Studies as Topic , Proportional Hazards Models , Retrospective Studies , Tissue Donors
4.
Transplantation ; 103(11): 2347-2358, 2019 11.
Article in English | MEDLINE | ID: mdl-30747854

ABSTRACT

BACKGROUND: The lack of accurate biomarkers makes it difficult to determine whether organs are suitable for transplantation. Mitochondrial DNA (mtDNA) correlates with tissue damage and kidney disease, making it a potential biomarker in organ evaluation. METHODS: Donors who had experienced cardiac death and successfully donated their kidneys between January 2015 and May 2017 were included this study. We detected the level of mtDNA in the plasma of the donor using quantitative real-time polymerase chain reaction and then statistically analyzed the relationship between the level of mtDNA and the delayed graft function (DGF) of the recipient. RESULTS: The incidence of DGF or slowed graft function (SGF) increased by 4 times (68% versus 16%, P < 0.001) when the donor mtDNA (dmtDNA) level was >0.114. When dmtDNA levels were >0.243, DGF and primary nonfunction were approximately 100% and 44%, respectively. Moreover, dmtDNA was an independent risk factor for slowed graft function and DGF. A prediction model for DGF based on dmtDNA achieved an area under the receiver operating characteristic curve for a prediction score as high as 0.930 (95% confidence interval 0.856-1.000), and the validation cohort results showed that the sensitivity and specificity of the model were 100% and 78%, respectively. dmtDNA levels were correlated with 6-month allograft function (R=0.332, P < 0.001) and 1-year graft survival (79% versus 99%, P < 0.001). CONCLUSIONS: We conclusively demonstrated that plasma dmtDNA was an independent risk factor for DGF, which is valuable in organ evaluation. dmtDNA is a possible first predictive marker for primary nonfunction and worth further evaluation.


Subject(s)
DNA, Mitochondrial/blood , Delayed Graft Function/etiology , Kidney Transplantation , Kidney/immunology , Plasma/metabolism , Tissue Donors , Adult , Allografts/immunology , Biomarkers , Delayed Graft Function/genetics , Female , Graft Rejection/epidemiology , Graft Survival , Humans , Immunohistochemistry , Incidence , Kidney Failure, Chronic/complications , Kidney Failure, Chronic/surgery , Male , Middle Aged , Postoperative Complications , ROC Curve , Retrospective Studies , Risk Factors , Transplantation, Homologous/adverse effects
5.
BMC Nephrol ; 19(1): 164, 2018 07 04.
Article in English | MEDLINE | ID: mdl-29973175

ABSTRACT

BACKGROUND: Organ donation after brain death (DBD) is the standard strategy for organ transplantation; however, the concept of brain death is not universally accepted due to cultural beliefs and barriers amongst billions of people worldwide. Hence, a novel donation pattern has been established in China which outlines the concept of donation after brain death followed by circulatory death (DBCD). Differently from any current donation classification, this new concept is formulated based on combination of recognizing brain death and circulatory death. Should approval be gained for this definition and approach, DBCD will pave a novel donation option for billions of people who cannot accept DBD due to their cultural beliefs. METHODS: A multi-center, cohort study was conducted from February 2012 to December 2015. 523 kidney transplant recipients from four kidney transplant institutions were enrolled into the study, of which, 383 received kidneys from DBCD, and 140 from DBD. Graft and recipient survivals following transplantation were retrospectively analyzed. Postoperative complications including delayed graft function,, and acute rejection, were also analyzed for both groups. RESULTS: DBCD could achieve comparable graft and recipient survivals in comparison with DBD (Log-rank P = 0.32 and 0.86,respectively). One-year graft and recipient survivals were equal between DBCD and DBD groups (97.4% versus 97.9%, P = 0.10;98.4% versus 98.6%, P = 1.0, respectively). Furthermore, DBCD did not increase incidences of postoperative complications compared with DBD, including delayed graft function (19.3% versus 22.1%, P = 0.46) and acute rejection (9.1% versus 8.6%, P = 1.0). Additionally, antithymocyte globulin as induction therapy and shorter warm ischemia time decreased incidence of delayed graft function in DBCD group (16.8% on antithymocyte globulin versus 27.2% on basiliximab, P = 0.03; 16.7% on ≤18 min versus 26.7% on > 18 min group, P = 0.03). CONCLUSIONS: Kidney donation through DBCD achieves equally successful outcomes as DBD, and could provide a feasible path to graft availability for billions of people who face barriers to organ donation from DBD.


Subject(s)
Allografts/physiology , Brain Death/diagnosis , Kidney Transplantation/methods , Shock/diagnosis , Tissue and Organ Procurement/methods , Adult , Brain Death/pathology , Cohort Studies , Female , Graft Survival/physiology , Humans , Kidney Transplantation/standards , Male , Middle Aged , Retrospective Studies , Shock/pathology , Tissue and Organ Procurement/standards , Treatment Outcome , Young Adult
6.
Kidney Blood Press Res ; 43(3): 893-903, 2018.
Article in English | MEDLINE | ID: mdl-29871001

ABSTRACT

BACKGROUND/AIMS: Delayed graft function (DGF) is a common complication following kidney transplantation adversely affecting graft outcomes. Donation after brain death followed by circulatory death (DBCD), a novel donation pattern, is expected to correlate with high incidence of DGF. However, little information is available about factors associated with DGF in DBCD. METHODS: A total of 383 kidney transplants from DBCD donation in three institutions were enrolled. Associations of DGF with the clinical characteristics of recipients and donors were quantified. RESULTS: In this retrospective multi-center study, the incidence of DGF was 19.3%. Lower incidence of DGF was found in recipients for whom antithymocyte globulin was used for induction (p < 0.05), which was an independent protective factor against DGF (odds ratio [OR] = 0.48; 95% CI 0.27-0.86). Two novel explicative variables were recognized as independent risk factors, including use of vasoactive drugs (OR = 3.15; 95% CI 1.39-7.14) and cardiopulmonary resuscitation (OR = 2.51; 95% CI 1.05-6.00), which contributed significantly to increased risk of DGF (p < 0.05). Prolonged warm ischemia time (> 18 min; OR = 2.42; 95% CI 1.36-4.32), was also predictive of DGF in DBCD. A prediction model was developed and achieved an area under the curve of 0.89 in predicting DGF when combined with reported parameters. CONCLUSION: The novel factors, confirmed for the first time in our study, will help to improve risk prediction of DGF and to determine optimal interventions to prevent DGF in clinical practice.


Subject(s)
Delayed Graft Function/diagnosis , Kidney Transplantation/methods , Tissue Donors , Adult , Area Under Curve , Brain Death , Cohort Studies , Delayed Graft Function/etiology , Female , Humans , Male , Middle Aged , Prognosis , Retrospective Studies , Risk Factors , Shock/mortality
7.
J Transl Med ; 16(1): 52, 2018 03 07.
Article in English | MEDLINE | ID: mdl-29514693

ABSTRACT

BACKGROUND: Kidneys from deceased donors are being used to meet the growing need for grafts. However, delayed graft function (DGF) and acute rejection incidences are high, leading to adverse effects on graft outcomes. Optimal induction intervention should include both renal structure injury repair and immune response suppression. Mesenchymal stem cells (MSCs) with potent anti-inflammatory, regenerative, and immune-modulatory properties are considered a candidate to prevent DGF and acute rejection in renal transplantation. Thus, this prospective multicenter paired study aimed to assess the clinical value of allogeneic MSCs as induction therapy to prevent both DGF and acute rejection in deceased donor renal transplantation. METHODS: Forty-two renal allograft recipients were recruited and divided into trial and control groups. The trial group (21 cases) received 2 × 106/kg human umbilical-cord-derived MSCs (UC-MSCs) via the peripheral vein before renal transplantation, and 5 × 106 cells via the renal artery during the surgical procedure. All recipients received standard induction therapy. Incidences of DGF and biopsy-proven acute rejection were recorded postoperatively and severe postoperative complications were assessed. Graft and recipient survivals were also evaluated. RESULTS: Treatment with UC-MSCs achieved comparable graft and recipient survivals with non-MSC treatment (P = 0.97 and 0.15, respectively). No increase in postoperative complications, including DGF and acute rejection, were observed (incidence of DGF: 9.5% in the MSC group versus 33.3% in the non-MSC group, P = 0.13; Incidence of acute rejection: 14.3% versus 4.8%, P = 0.61). Equal postoperative estimated glomerular filtration rates were found between the two groups (P = 0.88). All patients tolerated the MSCs infusion without adverse clinical effects. Additionally, a multiprobe fluorescence in situ hybridization assay revealed that UC-MSCs administered via the renal artery were absent from the recipient's biopsy sample. CONCLUSIONS: Umbilical-cord-derived MSCs can be used as clinically feasible and safe induction therapy. Adequate timing and frequency of UC-MSCs administration may have a significant effect on graft and recipient outcomes. Trial registration NCT02490020 . Registered on June 29 2015.


Subject(s)
Kidney Transplantation , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/cytology , Adult , Feasibility Studies , Female , Follow-Up Studies , Glomerular Filtration Rate , Humans , Kaplan-Meier Estimate , Male , Pilot Projects , Postoperative Complications/etiology , Tissue Donors , Transplantation, Homologous , Treatment Outcome , Umbilical Cord/cytology
8.
Int J Mol Med ; 41(1): 25-32, 2018 Jan.
Article in English | MEDLINE | ID: mdl-29115389

ABSTRACT

Ischemia/reperfusion injury (IRI) commonly occurs in renal transplantation. Erythropoietin (EPO) exerts a protective effect in IRI. To investigate the underlying molecular mechanism, rat models of renal IRI were established and treated with EPO and/or lentivirus­mediated EPO-siRNA, the signal transducer and activator of transcription 6 (STAT6) inhibitor AS1517499, the JNK inhibitor SP600125, the p38 mitogen-activated protein kinase (MAPK) inhibitor SB203580, and the nuclear factor (NF)-κB inhibitor lactacystin. Histological examination revealed that EPO protected the kidney from IRI, through decreasing the extent of tissue congestion and inflammatory cell infiltration; however, EPO siRNA did not exert the same protective effect. In addition, the EPO level was inversely associated with renal IRI. EPO downregulated the expression of interferon-γ, interleukin (IL)-4, creatinine and caspase-3, and upregulated the expression of IL-10, thymic stromal lymphopoietin, STAT6, p-JNK and p-p38, while the opposite effects were observed with the administration of EPO-siRNA and the specific respective inhibitors. Further results revealed that MAPK (p-JNK and p-p38) acted upstream of NF-κB, and that NF-κB signaling regulated the expression of caspase-1 and -3, which may be responsible for the cytotoxicity associated with IRI. Taken together, the results of the present study demonstrated that EPO exerted a protective effect in renal IRI via the STAT6/MAPK/NF-κB pathway. This protective effect of EPO may improve reperfusion tolerance in ischemic kidneys and benefit transplant recipients.


Subject(s)
Erythropoietin/administration & dosage , Reperfusion Injury/drug therapy , Reperfusion Injury/genetics , STAT6 Transcription Factor/genetics , p38 Mitogen-Activated Protein Kinases/genetics , Acetylcysteine/administration & dosage , Acetylcysteine/analogs & derivatives , Animals , Anthracenes/administration & dosage , Apoptosis/drug effects , Apoptosis/genetics , Erythropoietin/genetics , Gene Expression Regulation/genetics , Humans , Imidazoles/administration & dosage , Interferon-gamma/genetics , Kidney/metabolism , Kidney/pathology , Kidney Transplantation/adverse effects , Lentivirus/genetics , MAP Kinase Kinase 4/antagonists & inhibitors , MAP Kinase Kinase 4/genetics , NF-kappa B/antagonists & inhibitors , NF-kappa B/genetics , Pyridines/administration & dosage , Pyrimidines/administration & dosage , RNA, Small Interfering/administration & dosage , RNA, Small Interfering/genetics , Rats , Reperfusion Injury/pathology , STAT6 Transcription Factor/antagonists & inhibitors , Signal Transduction/drug effects , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors
9.
Trials ; 18(1): 545, 2017 Nov 16.
Article in English | MEDLINE | ID: mdl-29145879

ABSTRACT

BACKGROUND: Using kidneys from deceased donors is an available strategy to meet the growing need of grafts. However, higher incidences of delayed graft function (DGF) and acute rejection exert adverse effects on graft outcomes. Since ischemia-reperfusion injury (IRI) and ongoing process of immune response to grafts are the major causes of DGF and acute rejection, the optimal induction intervention should possess capacities of both repairing renal structure injury and suppressing immune response simultaneously. Mesenchymal stem cells (MSCs) with potent anti-inflammatory, regenerative and immune-modulatory properties are considered as a candidate to prevent both DGF and acute rejection in renal transplantation. Previous studies just focused on the safety of autologous MSCs on living-related donor renal transplants, and lack of concomitant controls and the sufficient sample size and source of MSCs. Here, we propose a prospective multicenter controlled study to assess the clinical value of allogeneic MSCs in preventing both DGF and acute rejection simultaneously as induction therapy in deceased-donor renal transplantation. METHODS/DESIGN: Renal allograft recipients (n = 100) will be recruited and divided into trial and control groups, and 50 patients in the trial group will be administered with a dose of 2 × 106 per kilogram human umbilical-cord-derived MSCs (UC-MSCs) via peripheral vein injection preoperatively, and a dose of 5 × 106 cells via renal arterial injection during surgery, with standard induction therapy. Incidences of postoperative DGF and biopsy-proved acute rejection (BPAR) will be recorded and analyzed. Additionally, other clinical parameters such as baseline demographics, graft and recipient survival and other severe postoperative complications, including complicated urinary tract infection, severe pneumonia, and severe bleeding, will be also assessed. DISCUSSION: This study will clarify the clinical value of UC-MSCs in preventing DGF and acute rejection simultaneously in deceased-donor renal transplantation, and provide evidence as to whether allogeneic MSCs can be used as clinically feasible and safe induction therapy. TRIAL REGISTRATION: ClinicalTrials.gov, NCT02490020 . Registered on 29 June 2015.


Subject(s)
Clinical Protocols , Delayed Graft Function/prevention & control , Graft Rejection/prevention & control , Kidney Transplantation/adverse effects , Mesenchymal Stem Cell Transplantation , Acute Disease , Adolescent , Adult , Humans , Middle Aged , Prospective Studies , Transplantation, Homologous , Young Adult
10.
Stem Cell Res ; 24: 89-93, 2017 10.
Article in English | MEDLINE | ID: mdl-29034901

ABSTRACT

The ATP-sensitive potassium channel is an octameric complex, and one of its subunits, namely Kir6.2, is encoded by the KCNJ11 gene. Mutations in KCNJ11 result in hyperinsulinism or diabetes mellitus, associated with abnormal insulin secretion. Here, using CRISPR/Cas9 editing, we established a homozygous mutant KCNJ11 cell line, WAe001-A-12, which was generated by a 62-bp deletion in the coding sequence of the human embryonic stem cell line H1. It was confirmed that this deletion in the KCNJ11 gene did not affect the protein expression levels of key pluripotent factors. Additionally, normal karyotype and differentiation potency were observed for the cell line.


Subject(s)
CRISPR-Cas Systems/genetics , Embryonic Stem Cells/metabolism , Human Embryonic Stem Cells/metabolism , Potassium Channels, Inwardly Rectifying/genetics , Cell Differentiation , Cell Line , Humans , Potassium Channels, Inwardly Rectifying/metabolism
11.
Stem Cell Res ; 18: 67-69, 2017 01.
Article in English | MEDLINE | ID: mdl-28395809

ABSTRACT

A patient specific point mutation (c.1288G>T) of Men1 gene was introduced into wide type iPSC line with CRISPR/Cas9 and single-stranded donor oligonucleotides carrying the mutation. The mutated iPSC line has a heterozygous c.1288G>T mutation on exon-9 of Men1 that was confirmed by sequencing analysis. The karyotype of this line was normal and the pluripotency was demonstrated by its ability to differentiate into three germ layers. These artificially created Men1 mutation in wild type iPSC line will help to dissect out the molecular basis of two patients carried the same mutation from one family who were differentially represented hypoglycemia.


Subject(s)
CRISPR-Cas Systems/genetics , Induced Pluripotent Stem Cells/cytology , Oligodeoxyribonucleotides/genetics , Proto-Oncogene Proteins/genetics , Base Sequence , Cell Differentiation , Cell Line , DNA Mutational Analysis , Embryoid Bodies/metabolism , Embryoid Bodies/pathology , Exons , Gene Editing , Genotype , Heterozygote , Humans , Induced Pluripotent Stem Cells/metabolism , Karyotype , Male , Microscopy, Fluorescence , Multiple Endocrine Neoplasia Type 1/genetics , Multiple Endocrine Neoplasia Type 1/metabolism , Multiple Endocrine Neoplasia Type 1/pathology , Oligodeoxyribonucleotides/metabolism , Point Mutation , Transcription Factors/genetics , Transcription Factors/metabolism
12.
Ther Clin Risk Manag ; 13: 279-285, 2017.
Article in English | MEDLINE | ID: mdl-28280347

ABSTRACT

BACKGROUND: Aldolase A (ALDOA) is a glycolytic enzyme that drives the glycolytic metabolic pathway in mammalian cells. The overexpression of ALDOA was observed in a variety of cancers including clear-cell renal cell carcinoma (ccRCC). However, little was known about the clinicopathological significance and prognostic value of ALDOA in ccRCC patients. METHODS: The expression of ALDOA was detected using immunohistochemical staining in 162 formalin-fixed, paraffin-embedded ccRCC sections. Prognostic outcomes correlated with ALDOA were examined using Kaplan-Meier analysis and the Cox proportional hazards model. RESULTS: In patients with ccRCC, increased cytoplasmic ALDOA expression was positively associated with tumor size (P=0.021), TNM stages (P=0.034), lymph node metastasis (P=0.020), and overall survival (OS) (P<0.001). Kaplan-Meier analysis showed that high cytoplasmic expression of ALDOA was associated with a statistically significant lower OS (P<0.001). Multivariate analysis demonstrated that ALDOA expression was an independent and significant prognostic factor (HR =3.561, 95% CI =1.715-7.396, P=0.001). ALDOA expression was not associated with significant prognostic deference in the subgroups of TNM stage I patients or pT1 patients. CONCLUSION: Our results suggest that ALDOA expression is an independent prognostic factor for OS in patients with ccRCC.

13.
Onco Targets Ther ; 9: 7199-7205, 2016.
Article in English | MEDLINE | ID: mdl-27932890

ABSTRACT

High-mobility group AT-hook 2 (HMGA2) is involved in a wide spectrum of biological processes and is upregulated in several tumors, but its role in renal carcinoma remains unclear. The aim of this study was to examine the expression of HMGA2 and its relationship to the overall survival (OS) of patients with non-metastatic clear cell renal cell carcinoma (ccRCC) following surgery. The expression of HMGA2 was evaluated retrospectively by immunohistochemistry (IHC) in 162 patients with ccRCC who underwent nephrectomy in 2003 and 2004. An IHC analysis revealed that HMGA2 was expressed in the nuclei of tumor cells in 146 (90.1%) patients with ccRCC. The level of HMGA2 was positively correlated with tumor size, lymph node metastasis, and Fuhrman Grade. A Kaplan-Meier analysis with log-rank test found that patients with high HMGA2 expression had a poor outcome and that patients with low HMGA2 expression had better survival. Cox regression analysis showed that HMGA2 expression could serve as an independent prognostic factor for ccRCC patients. The efficacy of the following prognostic models was improved when HMGA2 expression was added: tumor node metastasis stage, UCLA Integrated Scoring System, Mayo Clinic stage, size, grade, and necrosis score. In summary, this study showed that HMGA2 expression is an independent prognostic factor for OS in patients with ccRCC. HMGA2 was found to be a valuable biomarker for ccRCC progression.

14.
Mol Immunol ; 79: 22-31, 2016 11.
Article in English | MEDLINE | ID: mdl-27689750

ABSTRACT

Immature dendritic cells (iDCs) are bone marrow-derived professional antigen-presenting cells, exhibit very low levels of the co-stimulatory molecules CD80 (B7-1), CD86 (B7-2), and CD40 and major histocompatibility complex (MHC) class II and play a critical role in triggering antigen-specific immunotolerance. The enzyme indoleamine 2, 3-dioxygenase (IDO) is a cytosolic tryptophan catabolism rate-limiting step enzyme. IDO secreted by DCs shows an association with the suppression of T-cell responses and promotion of tolerance. In this study, BN rat recipients were pre-injected with donor renal alloantigen-treated recipient iDCs before kidney transplantation. The renal allograft exhibited a lighter renal rejection response, prolonged graft survival time, and an increasing content of CD4+CD25+Foxp3+ regulatory T cells (Tregs). Additionally, up-regulated secretion of Th2 cytokines were found in recipient sera post-transplantation. Transfection of si-IDO1 RNA into renal-antigen-treated recipient iDCs reversed these changes, which suggested that IDO channel signaling may be involved in iDC-induced allograft immunotolerance. These results suggested that iDC-induced and IDO-mediated allograft immunotolerance might be a potentially feasible tactic to prolong allograft survival, in addition to immunosuppressive drugs.


Subject(s)
Dendritic Cells/immunology , Immune Tolerance/immunology , Indoleamine-Pyrrole 2,3,-Dioxygenase/immunology , Kidney Transplantation , Transplantation Immunology/immunology , Allografts/immunology , Animals , Blotting, Western , Dendritic Cells/enzymology , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Graft Survival , Hematopoietic Stem Cells/enzymology , Hematopoietic Stem Cells/immunology , Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism , Lymphocyte Culture Test, Mixed , Male , Rats , T-Lymphocytes, Regulatory/immunology
15.
Oncotarget ; 7(28): 44039-44046, 2016 Jul 12.
Article in English | MEDLINE | ID: mdl-27270655

ABSTRACT

The data on the impact of the neutrophil-to-lymphocyte ratio (NLR) in metastatic renal cell carcinoma (mRCC) patients receiving tyrosine kinase inhibitors (TKIs) are inconsistent. We therefore performed a meta-analysis to assess the prognostic value of pretreatment NLR in patients treated with TKIs for mRCC. We searched the Embase, Medline, PubMed, Cochrane and ISI Web of Knowledge to identify clinical studies that had evaluated the association between the pretreatment NLR and prognosis in mRCC patients. Prognostic outcomes included overall survival (OS) and progression-free survival (PFS). Nine studies encompassing a total of 1091 participants were included. We found that a high NLR was an effective prognostic marker of both OS (pooled HR: 1.93, 95% CI: 1.35-2.77; P = 0.0003) and PFS (pooled HR: 2.12, 95% CI: 1.42-3.17; P = 0.0002). Subgroup analysis revealed that studies reporting a NLR ≥ 3 showed a more significant effect of NLR on both OS (pooled HR: 2.50, 95% CI: 1.99-3.14; P = 0.0003) and PFS (pooled HR: 2.17, 95% CI: 1.26-3.75). This meta-analysis suggests that high pretreatment NLR is associated with a poor prognosis in mRCC patients receiving TKI treatment.


Subject(s)
Carcinoma, Renal Cell/drug therapy , Kidney Neoplasms/drug therapy , Lymphocytes/pathology , Neutrophils/pathology , Protein-Tyrosine Kinases/therapeutic use , Carcinoma, Renal Cell/pathology , Disease-Free Survival , Humans , Kidney Neoplasms/pathology , Leukocyte Count , Prognosis , Treatment Outcome
16.
Cell Biochem Funct ; 30(2): 139-44, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22105543

ABSTRACT

Nephrotoxic serum nephritis (NSN) is a well-established animal model of glomerulonephritis, a frequent clinical condition with a high mortality rate owing to the ineffectiveness of current therapies. Mesenchymal stem cells (MSCs) are adult stem cells with potential as novel therapies in regenerative medicine owing to the absence of allogenic rejection. Glial cell-derived neurotrophic factor (GDNF) acts as a morphogen in kidney development. The therapeutic effectiveness of bone marrow MSCs overexpressing GDNF (GDNF-MSCs) was evaluated in an NSN rat model. An adenoviral vector was used to transduce MSCs with GDNF and a green fluorescent protein reporter gene. Then, GDNF-MSCs were injected into NSN rats via the renal artery. The influence of GDNF on renal injury was assessed. The location of GDNF-MSCs in kidneys was detected using fluorescence microscopy, cells were counted, and kidney function was measured. Infusion of GNDF-MSCs enhanced the recovery of renal function in NSN rats. MSCs were detected in the kidney cortex after injection. Compared with control MSCs, GDNF-MSCs led to significantly better renal function and injury recovery in NSN rats. GDNF has a positive effect on MSC differentiation in renal tissue. Owing to their highly renoprotective capacity, GDNF-MSCs represent a possible novel cell-based paradigm for treatment of glomerulonephritis.


Subject(s)
Gene Expression , Glial Cell Line-Derived Neurotrophic Factor/genetics , Glial Cell Line-Derived Neurotrophic Factor/metabolism , Glomerulonephritis/physiopathology , Glomerulonephritis/therapy , Kidney/physiopathology , Mesenchymal Stem Cells/metabolism , Animals , Cell Differentiation , Cell- and Tissue-Based Therapy , Cells, Cultured , Disease Models, Animal , Glomerulonephritis/genetics , Glomerulonephritis/metabolism , Humans , Kidney/metabolism , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/cytology , Rats
17.
Chin Med J (Engl) ; 124(17): 2723-7, 2011 Sep.
Article in English | MEDLINE | ID: mdl-22040431

ABSTRACT

BACKGROUND: Thymokidney has been reported as an approach for a vascularized thymus for transplantation to induce donor specific tolerance. A completely thymectomized model which ensures that the obtained thymus is not injured has not been developed yet and it would be useful for evaluating autologous thymokidney function in rats. METHODS: Adult Sprague-Dawley male rats weighing 150 - 300 g (n = 30) underwent non-invasive intubation with the assistance of an improved self-made wedge-shaped cannula made from a 2-ml plastic syringe and transillumination from the anterior tracheal area by an operation spotlight. The rats then received a thoracotomy while their breathing was supported by a small animal ventilator, and both lobes of the thymus were entirely extirpated under a 10× microscope. The postoperative survival rate of the rats was recorded, and changes in the T-cell reservoir from 9 of 30 rats within 21 days after surgery were monitored using flow cytometry. The complete thymectomy rate was confirmed by autopsy and histological examination on 21 days post-operation. RESULTS: The postoperative survival rate of rats was 100%. The exsected thymus was free of injury and the rate of complete thymectomy was 100%. CONCLUSIONS: This model has a stable survival rate and complete thymectomy is able to be achieved. The obtained thymus tissue is free of injury and can be used for transplantation.


Subject(s)
Intubation, Intratracheal/methods , Thymectomy/methods , Animals , Male , Rats , Rats, Sprague-Dawley , Thoracotomy/methods , Thymus Gland/surgery
18.
Chin Med J (Engl) ; 124(8): 1213-6, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21542999

ABSTRACT

BACKGROUND: For the renal transplant recipients, anemia is one of the common complications and becomes a major medical issue before transplantation. Haemoglobin (Hb) is used as a prognostic indicator, although the optimal pre-transplantation Hb concentration associated with positive prognosis is still controversial. The aim of this study was to detect the optimal Hb concentration on predicting the graft survival and function. METHODS: A retrospective cohort study was conducted by reviewing the medical records of the patients who received renal transplantations at our center from January 2004 to June 2008. Patients were divided into two groups: high Hb group (≥ 100 g/L, n = 79) and low Hb group (< 100 g/L, n = 63). There was no significant difference between the two groups regarding sex, age, blood type and tissue types. Renal function among the two groups was measured and compared. Panel reacting antigens (PRA) of all the recipients were negative. The effect of preoperative hemoglobin concentration on the postoperative renal function recovery in both groups was further analyzed. RESULTS: A total of 14 acute rejection episodes occurred, including 5 patients in the high Hb group (7.9%) and 9 in the low Hb group (11.4%, P > 0.05). The serum creatinine level at one-year post-transplantation of the low Hb group was significantly higher than that of the high Hb group ((117.8 ± 36.3) µmol/L vs. (103.1 ± 35.5) µmol/L, P < 0.05). For one-year actuarial patient and graft survival, incidence of delayed graft function (DGF), serum creatinine concentrations at 1, 3, 6 months post-transplantation, the incidence of cytomegalovirus (CMV) infection, post-transplantation anemia (PTA) and post-transplantation diabetes mellitus (PTDM) of both groups, there were no statistically significant differences. CONCLUSION: Pre-transplantation Hb concentration has significant effect on one-year creatinine concentration, but can not significantly affect acute rejection episodes, DGF, PTA, CMV infection and PTDM.


Subject(s)
Graft Rejection/blood , Hemoglobins/metabolism , Kidney Transplantation/adverse effects , Adult , Creatinine/blood , Female , Graft Survival/physiology , Humans , Immunosuppressive Agents/therapeutic use , Male , Middle Aged , Postoperative Period , Retrospective Studies
19.
J Mater Sci Mater Med ; 22(3): 547-55, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21318627

ABSTRACT

A superior drug controlled release system capable of achieving efficient osteogenesis is in imperative demand because of limited bone substitute tissue for the treatment of bone defect. In the present study, we investigated the potential of using poly(ε-caprolactone)-hydroxyapatite (PCL-HA) composite microspheres as an injectable bone repair vehicle by controlled release of alendronate (AL), a medicine that belongs to the bisphosphonates family. The PCL/HA-AL microspheres were prepared with solid/oil/water emulsion technique, which included two processes: (1) AL was loaded on the hydroxyapatite nanoparticles; (2) the HA-AL complex was built in the PCL matrix. The spherical PCL/HA-AL microspheres were characterized with its significantly improved encapsulation efficiency of hydrophilic AL and better sustained release. Human bone mesenchymal stem cells (hMSCs) were cultured on the surface of these microspheres and exhibited high proliferative profile. Specifically, in osteogenic medium, hMSCs on the surface of PCL/HA-AL microspheres displayed superior osteogenic differentiation which was verified by alkaline phosphatase activity assay. In conclusion, by presenting strong osteogenic commitment of hMSCs in vitro, the PCL/HA-AL microspheres have the potential to be used as an injectable vehicle for local therapy of bone defect.


Subject(s)
Alendronate/chemistry , Bone Substitutes/chemistry , Durapatite/chemistry , Mesenchymal Stem Cells/cytology , Polyesters/chemistry , Alkaline Phosphatase/metabolism , Bone and Bones/chemistry , Bone and Bones/cytology , Bone and Bones/pathology , Cell Proliferation , Cell Survival , Humans , Microscopy, Electron, Scanning/methods , Microspheres , Osteogenesis , Particle Size , Spectroscopy, Fourier Transform Infrared/methods
20.
Chin Med J (Engl) ; 122(3): 344-8, 2009 Feb 05.
Article in English | MEDLINE | ID: mdl-19236816

ABSTRACT

BACKGROUND: The prospects of using immature CD8a(+) dendritic cells (DC2) to establish transplant immunologic tolerance and treatments for autoimmune diseases in the future are promising. However, the methods for inducing DC2 are still being explored. The present study was aimed to investigate the optimal in vitro conditions for preparing large numbers of predominant DC2 from murine bone marrow cells. METHODS: Three groups of bone marrow cells cultured under different conditions were examined, namely a cytokine-induced experimental group (cytokine group), a control group with a low concentration of granulocyte-macrophage colony stimulating factor (GM-CSF, low GM-CSF group) and a control group without endogenous cytokines. The cytokine group was cultured with 5 ng/ml GM-CSF, 25 ng/ml Flt3 ligand (Flt3L), 20 ng/ml interleukin 4 (IL-4) and 100 ng/ml stem cell factor (SCF). The low GM-CSF control group was cultured with 0.4 ng/ml GM-CSF, 25 ng/ml Flt3L and 100 ng/ml SCF, without IL-4. The control group without exogenous cytokines was cultured without additional cytokines. All cells were cultured at 37 degrees C under 5% CO2. On days 3, 7 and 16, 4-color flow cytometry was carried out to analyze the cell phenotypes, and the total cell numbers were counted to analyze the cell yields. Phase-contrast microscopy was used to observe the cell morphologies. RESULTS: The cytokine group exhibited higher proportions of typical immature CD8a(+) DC, especially on day 3, but the total cell number and DC2 proportion decreased during prolonged culture. The low GM-CSF control group showed the same tendencies as the cytokine group on days 16 and 22, but produced higher total cell numbers (P<0.05) with lower DC2 proportions and cell numbers. The control group without exogenous cytokines spontaneously generated a certain proportion of DC2, but with low total cell and DC2 numbers that decreased rapidly, especially during prolonged culture (days 7 and 16, P<0.05). CONCLUSIONS: Culture in the presence of 5 ng/ml GM-CSF, 25 ng/ml Flt3L, 20 ng/ml IL-4 and 100 ng/ml SCF can rapidly induce large quantities of predominant immature CD8a(+) DC from murine bone marrow cells. Therefore, these represent optimal culture conditions for preparing murine immature DC2 in vitro.


Subject(s)
CD8 Antigens/metabolism , CD8-Positive T-Lymphocytes/cytology , Cell Culture Techniques/methods , Animals , Bone Marrow Cells/cytology , Bone Marrow Cells/drug effects , CD8-Positive T-Lymphocytes/drug effects , Cells, Cultured , Flow Cytometry , Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology , Male , Mice , Mice, Inbred BALB C , Microscopy, Phase-Contrast
SELECTION OF CITATIONS
SEARCH DETAIL
...