Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 121
Filter
Add more filters

Publication year range
1.
Kidney Int ; 105(2): 347-363, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38040290

ABSTRACT

Natural killer (NK) cells mediate spontaneous cell-mediated cytotoxicity and antibody-dependent cell-mediated cytotoxicity. This dual functionality could enable their participation in chronic active antibody-mediated rejection (CA-ABMR). Earlier microarray profiling studies have not subcategorized antibody-mediated rejection into CA-ABMR and active-ABMR, and the gene expression pattern of CA-ABMR has not been compared with that of T cell-mediated rejection (TCMR). To fill these gaps, we RNA sequenced human kidney allograft biopsies categorized as CA-ABMR, active-ABMR, TCMR, or No Rejection (NR). Among the 15,910 genes identified in the biopsies, 60, 114, and 231 genes were uniquely overexpressed in CA-ABMR, TCMR, and active-ABMR, respectively; compared to NR, 50 genes were shared between CA-ABMR and active-ABMR, and 164 genes between CA-ABMR and TCMR. The overexpressed genes were annotated to NK cells and T cells in CA-ABMR and TCMR, and to neutrophils and monocytes in active-ABMR. The NK cell cytotoxicity and allograft rejection pathways were enriched in CA-ABMR. Genes encoding perforin, granzymes, and death receptor were overexpressed in CA-ABMR versus active-ABMR but not compared to TCMR. NK cell cytotoxicity pathway gene set variation analysis score was higher in CA-ABMR compared to active-ABMR but not in TCMR. Principal component analysis of the deconvolved immune cellular transcriptomes separated CA-ABMR and TCMR from active-ABMR and NR. Immunohistochemistry of kidney allograft biopsies validated a higher proportion of CD56+ NK cells in CA-ABMR than in active-ABMR. Thus, CA-ABMR was exemplified by the overexpression of the NK cell cytotoxicity pathway gene set and, surprisingly, molecularly more like TCMR than active-ABMR.


Subject(s)
Kidney Transplantation , Humans , Kidney Transplantation/adverse effects , Transcriptome , Graft Rejection , Kidney/pathology , Antibodies , Gene Expression Profiling , Allografts , Sequence Analysis, RNA
2.
Curr Opin Organ Transplant ; 28(2): 117-125, 2023 04 01.
Article in English | MEDLINE | ID: mdl-36757681

ABSTRACT

PURPOSE OF REVIEW: Because all functioning nephrons contribute to urine formation, we reasoned that urine would be a suitable substitute to kidney allograft biopsy to discern human kidney allograft status. In view of compelling data that ribonucleic acid (RNA) sequencing outperforms microarray-based profiling, we performed RNA sequencing of urinary cells and kidney allograft biopsies to define the transcriptional landscape of allograft rejection. RECENT FINDINGS: Whole genome transcriptome profiling identified unique and shared gene signatures of acute T cell mediated rejection (TCMR) and antibody mediated rejection (AMR). We found that biopsy rejection signatures are enriched in urinary cells and that the immune cellular landscape is more diverse and enriched in urine compared to biopsies. Towards a patient friendly protocol for urinary cell messenger RNA (mRNA) profiling, we developed a filtration-based protocol for the initial processing of urine at home and demonstrated excellent performance characteristics of the filter- based protocol. SUMMARY: Acute rejection signatures are enriched in urinary cells. Urinary cell mRNA profiles are diagnostic and prognostic of acute rejection and could serve as yardsticks of in-vivo immune status. RNA sequencing yields insights into mechanisms of rejection and helps prioritize therapeutic targets. The filtration protocol for home processing of urine may optimize immune surveillance.


Subject(s)
Kidney Transplantation , Humans , Kidney Transplantation/adverse effects , RNA, Messenger/genetics , RNA, Messenger/urine , Kidney/pathology , Transplantation, Homologous , Biopsy , Allografts , Graft Rejection/diagnosis , Graft Rejection/genetics
3.
J Physiol ; 600(22): 4827-4848, 2022 11.
Article in English | MEDLINE | ID: mdl-36181482

ABSTRACT

Loss of function mutations in store-operated Ca2+ entry (SOCE) are associated with severe paediatric disorders in humans, including combined immunodeficiency, anaemia, thrombocytopenia, anhidrosis and muscle hypotonia. Given its central role in immune cell activation, SOCE has been a therapeutic target for autoimmune and inflammatory diseases. Treatment for such chronic diseases would require prolonged SOCE inhibition. It is, however, unclear whether chronic SOCE inhibition is viable therapeutically. Here we address this issue using a novel genetic mouse model (SOCE hypomorph) with deficient SOCE, nuclear factor of activated T cells activation, and T cell cytokine production. SOCE hypomorph mice develop and reproduce normally and do not display muscle weakness or overt anhidrosis. They do, however, develop cardiovascular complications, including hypertension and tachycardia, which we show are due to increased sympathetic autonomic nervous system activity and not cardiac or vascular smooth muscle autonomous defects. These results assert that chronic SOCE inhibition is viable therapeutically if the cardiovascular complications can be managed effectively clinically. They further establish the SOCE hypomorph line as a genetic model to define the therapeutic window of SOCE inhibition and dissect toxicities associated with chronic SOCE inhibition in a tissue-specific fashion. KEY POINTS: A floxed stromal interaction molecule 1 (STIM1) hypomorph mouse model was generated with significant reduction in Ca2+ influx through store-operated Ca2+ entry (SOCE), resulting in defective nuclear translocation of nuclear factor of activated T cells, cytokine production and inflammatory response. The hypomorph mice are viable and fertile, with no overt defects. Decreased SOCE in the hypomorph mice is due to poor translocation of the mutant STIM1 to endoplasmic reticulum-plasma membrane contact sites resulting in fewer STIM1 puncta. Hypomorph mice have similar susceptibility to controls to develop diabetes but exhibit tachycardia and hypertension. The hypertension is not due to increased vascular smooth muscle contractility or vascular remodelling. The tachycardia is not due to heart-specific defects but rather seems to be due to increased circulating catecholamines in the hypomorph. Therefore, long term SOCE inhibition is viable if the cardiovascular defects can be managed clinically.


Subject(s)
Hypertension , Hypohidrosis , Animals , Child , Humans , Mice , Calcium/metabolism , Calcium Signaling , Cytokines/metabolism , ORAI1 Protein/genetics , Stromal Interaction Molecule 1/genetics , Stromal Interaction Molecule 1/metabolism , Cardiovascular System/metabolism
4.
Proc Natl Acad Sci U S A ; 116(37): 18738-18744, 2019 09 10.
Article in English | MEDLINE | ID: mdl-31451660

ABSTRACT

High-throughput metagenomic sequencing offers an unbiased approach to identify pathogens in clinical samples. Conventional metagenomic sequencing, however, does not integrate information about the host, which is often critical to distinguish infection from infectious disease, and to assess the severity of disease. Here, we explore the utility of high-throughput sequencing of cell-free DNA (cfDNA) after bisulfite conversion to map the tissue and cell types of origin of host-derived cfDNA, and to profile the bacterial and viral metagenome. We applied this assay to 51 urinary cfDNA isolates collected from a cohort of kidney transplant recipients with and without bacterial and viral infection of the urinary tract. We find that the cell and tissue types of origin of urinary cfDNA can be derived from its genome-wide profile of methylation marks, and strongly depend on infection status. We find evidence of kidney and bladder tissue damage due to viral and bacterial infection, respectively, and of the recruitment of neutrophils to the urinary tract during infection. Through direct comparison to conventional metagenomic sequencing as well as clinical tests of infection, we find this assay accurately captures the bacterial and viral composition of the sample. The assay presented here is straightforward to implement, offers a systems view into bacterial and viral infections of the urinary tract, and can find future use as a tool for the differential diagnosis of infection.


Subject(s)
Cell-Free Nucleic Acids/isolation & purification , Host-Pathogen Interactions/genetics , Metagenome/genetics , Metagenomics/methods , Postoperative Complications/diagnosis , Urinary Tract Infections/diagnosis , Bacterial Infections/diagnosis , Bacterial Infections/microbiology , Bacterial Infections/urine , Biomarkers/urine , Cell-Free Nucleic Acids/genetics , Cell-Free Nucleic Acids/urine , DNA Methylation/genetics , DNA, Bacterial/genetics , DNA, Bacterial/isolation & purification , DNA, Bacterial/urine , DNA, Viral/genetics , DNA, Viral/isolation & purification , DNA, Viral/urine , Diagnosis, Differential , Female , High-Throughput Nucleotide Sequencing , Host-Pathogen Interactions/immunology , Humans , Kidney/cytology , Kidney/immunology , Kidney/microbiology , Kidney/pathology , Kidney Failure, Chronic/surgery , Kidney Transplantation/adverse effects , Male , Neutrophil Infiltration/immunology , Postoperative Complications/immunology , Postoperative Complications/microbiology , Postoperative Complications/urine , Transplant Recipients , Urinary Bladder/cytology , Urinary Bladder/immunology , Urinary Bladder/microbiology , Urinary Bladder/pathology , Urinary Tract Infections/immunology , Urinary Tract Infections/microbiology , Urinary Tract Infections/urine , Virus Diseases/diagnosis , Virus Diseases/immunology , Virus Diseases/urine , Virus Diseases/virology
5.
Clin Chem ; 68(1): 163-171, 2021 12 30.
Article in English | MEDLINE | ID: mdl-34718476

ABSTRACT

BACKGROUND: Metagenomic sequencing of microbial cell-free DNA (cfDNA) in blood and urine is increasingly used as a tool for unbiased infection screening. The sensitivity of metagenomic cfDNA sequencing assays is determined by the efficiency by which the assay recovers microbial cfDNA vs host-specific cfDNA. We hypothesized that the choice of methods used for DNA isolation, DNA sequencing library preparation, and sequencing would affect the sensitivity of metagenomic cfDNA sequencing. METHODS: We characterized the fragment length biases inherent to select DNA isolation and library preparation procedures and developed a model to correct for these biases. We analyzed 305 cfDNA sequencing data sets, including publicly available data sets and 124 newly generated data sets, to evaluate the dependence of the sensitivity of metagenomic cfDNA sequencing on pre-analytical variables. RESULTS: Length bias correction of fragment length distributions measured from different experimental procedures revealed the ultrashort (<100 bp) nature of microbial-, mitochondrial-, and host-specific urinary cfDNA. The sensitivity of metagenomic sequencing assays to detect the clinically reported microorganism differed by more than 5-fold depending on the combination of DNA isolation and library preparation used. CONCLUSIONS: Substantial gains in the sensitivity of microbial and other short fragment recovery can be achieved by easy-to-implement changes in the sample preparation protocol, which highlights the need for standardization in the liquid biopsy field.


Subject(s)
Cell-Free Nucleic Acids , DNA Fragmentation , Sequence Analysis, DNA , Bias , Cell-Free Nucleic Acids/genetics , DNA , Humans , Metagenomics/methods
6.
Clin Transplant ; 35(2): e14200, 2021 02.
Article in English | MEDLINE | ID: mdl-33349997

ABSTRACT

Identifying kidney transplant recipients at risk for graft failure following BK virus nephropathy (BKVN) may allow personalization of therapy. We have reported that a noninvasive composite signature of urinary cell level of plasminogen activator inhibitor-1(PAI-1) mRNA and serum creatinine level, measured at the time of BKVN diagnosis, is prognostic of graft failure. In this investigation, we determined whether the composite signature is prognostic of graft failure in an independent cohort of 25 patients with BKVN. Of the 25 patients, 8 developed graft failure and 17 did not. We measured urinary cell levels of PAI-1 mRNA, 18S rRNA, and BKV VP1 mRNA at the time of BKVN diagnosis and evaluated clinical parameters including Banff pathology scores, acute rejection, and graft function. The area under the receiver operating characteristic curve for the noninvasive composite signature was 0.95 (P < .001) for prognosticating graft failure. The previously reported threshold of -0.858 predicted graft failure with a sensitivity of 75% and a specificity of 94%. Our current study validates the use of composite signature and the threshold of -0.858 to identify those at risk for graft failure following BKVN diagnosis, and supports future studies utilizing the composite signature score to personalize treatment of BKVN.


Subject(s)
BK Virus , Kidney Diseases , Kidney Transplantation , Polyomavirus Infections , Tumor Virus Infections , Allografts , BK Virus/genetics , Graft Rejection/diagnosis , Graft Rejection/etiology , Humans , Kidney Transplantation/adverse effects , Polyomavirus Infections/diagnosis , Polyomavirus Infections/etiology , Prognosis
7.
Nephrol Dial Transplant ; 35(7): 1250-1261, 2020 07 01.
Article in English | MEDLINE | ID: mdl-32678882

ABSTRACT

BACKGROUND: Kidney graft recipients receiving immunosuppressive therapy may be at heightened risk for coronavirus disease 2019 (Covid-19) and adverse outcomes. It is therefore important to characterize the clinical course and outcome of Covid-19 in this population and identify safe therapeutic strategies. METHODS: We performed a retrospective chart review of 73 adult kidney graft recipients evaluated for Covid-19 from 13 March to 20 April 2020. Primary outcomes included recovery from symptoms, acute kidney injury, graft failure and case fatality rate. RESULTS: Of the 73 patients screened, 54 tested positive for severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2)-39 with moderate to severe symptoms requiring hospital admission and 15 with mild symptoms managed in the ambulatory setting. Hospitalized patients were more likely to be male, of Hispanic ethnicity and to have cardiovascular disease. In the hospitalized group, tacrolimus dosage was reduced in 46% of patients and mycophenolate mofetil (MMF) therapy was stopped in 61% of patients. None of the ambulatory patients had tacrolimus reduction or discontinuation of MMF. Azithromycin or doxycycline was prescribed at a similar rate among hospitalized and ambulatory patients (38% versus 40%). Hydroxychloroquine was prescribed in 79% of hospitalized patients. Graft failure requiring hemodialysis occurred in 3 of 39 hospitalized patients (8%) and 7 patients died, resulting in a case fatality rate of 13% among Covid-19-positive patients and 18% among hospitalized Covid-19-positive patients. CONCLUSIONS: Data from our study suggest that a strategy of systematic triage to outpatient or inpatient care, early management of concurrent bacterial infections and judicious adjustment of immunosuppressive drugs rather than cessation is feasible in kidney transplant recipients with Covid-19.


Subject(s)
Betacoronavirus , Coronavirus Infections/complications , Graft Rejection/therapy , Hydroxychloroquine/therapeutic use , Immunosuppression Therapy/methods , Kidney Transplantation , Mycophenolic Acid/therapeutic use , Pneumonia, Viral/complications , Adult , Aged , Aged, 80 and over , Allografts , Antimalarials/therapeutic use , COVID-19 , Coronavirus Infections/drug therapy , Coronavirus Infections/epidemiology , Enzyme Inhibitors/therapeutic use , Female , Graft Rejection/complications , Graft Rejection/epidemiology , Humans , Immunosuppressive Agents/therapeutic use , Incidence , Male , Middle Aged , New York City/epidemiology , Pandemics , Pneumonia, Viral/drug therapy , Pneumonia, Viral/epidemiology , Retrospective Studies , SARS-CoV-2 , Transplant Recipients
8.
Am J Transplant ; 19(2): 488-500, 2019 02.
Article in English | MEDLINE | ID: mdl-29920927

ABSTRACT

Posttransplant diarrhea is associated with kidney allograft failure and death, but its etiology remains unknown in the majority of cases. Because altered gut microbial ecology is a potential basis for diarrhea, we investigated whether posttransplant diarrhea is associated with gut dysbiosis. We enrolled 71 kidney allograft recipients for serial fecal specimen collections in the first 3 months of transplantation and profiled the gut microbiota using 16S ribosomal RNA (rRNA) gene V4-V5 deep sequencing. The Shannon diversity index was significantly lower in 28 diarrheal fecal specimens from 25 recipients with posttransplant diarrhea than in 112 fecal specimens from 46 recipients without posttransplant diarrhea. We found a lower relative abundance of 13 commensal genera (Benjamini-Hochberg adjusted P ≤ .15) in the diarrheal fecal specimens including the same 4 genera identified in our prior study. The 28 diarrheal fecal specimens were also evaluated by a multiplexed polymerase chain reaction (PCR) assay for 22 bacterial, viral, and protozoan gastrointestinal pathogens, and 26 specimens were negative for infectious etiologies. Using PICRUSt (Phylogenetic Investigation of Communities by Reconstruction of Unobserved States) to predict metagenomic functions, we found that diarrheal fecal specimens had a lower abundance of metabolic genes. Our findings suggest that posttransplant diarrhea is not associated with common infectious diarrheal pathogens but with a gut dysbiosis.


Subject(s)
Bacteria/growth & development , Diarrhea/etiology , Dysbiosis/etiology , Gastrointestinal Microbiome , Graft Rejection/etiology , Kidney Failure, Chronic/surgery , Kidney Transplantation/adverse effects , Adult , Bacteria/genetics , Bacteria/isolation & purification , Case-Control Studies , Cohort Studies , Diarrhea/pathology , Dysbiosis/pathology , Feces/microbiology , Female , Follow-Up Studies , Glomerular Filtration Rate , Graft Rejection/pathology , Graft Survival , Humans , Kidney Function Tests , Male , Middle Aged , Postoperative Complications , Prognosis , RNA, Ribosomal, 16S/genetics , Risk Factors
9.
Transpl Infect Dis ; 21(6): e13167, 2019 Dec.
Article in English | MEDLINE | ID: mdl-31502737

ABSTRACT

BACKGROUND: In kidney transplant recipients, gastrointestinal (GI) pathogens in feces are only evaluated during diarrheal episodes. Little is known about the prevalence of GI pathogens in asymptomatic individuals in this population. METHODS: We recruited 142 kidney transplant recipients who provided a non-diarrheal fecal sample within the first 10 days after transplantation. The specimens were evaluated for GI pathogens using the BioFire® FilmArray® GI Panel (BioFire Diagnostics, LLC), which tests for 22 pathogens. The fecal microbiome was also characterized using 16S rRNA gene sequencing of the V4-V5 hypervariable region. We evaluated whether detection of Clostridioides difficile and other GI pathogens was associated with post-transplant diarrhea within the first 3 months after transplantation. RESULTS: Among the 142 subjects, a potential pathogen was detected in 43 (30%) using the GI Panel. The most common organisms detected were C difficile (n = 24, 17%), enteropathogenic Escherichia coli (n = 8, 6%), and norovirus (n = 5, 4%). Detection of a pathogen on the GI panel or detection of C difficile alone was not associated with future post-transplant diarrhea (P > .05). The estimated number of gut bacterial species was significantly lower in subjects colonized with C difficile than those not colonized with a GI pathogen (P = .01). CONCLUSION: Colonization with GI pathogens, particularly C difficile, is common at the time of kidney transplantation but does not predict subsequent diarrhea. Detection of C difficile carriage was associated with decreased microbial diversity and may be a biomarker of gut dysbiosis.


Subject(s)
Asymptomatic Infections/epidemiology , Dysbiosis/epidemiology , Feces/microbiology , Gastrointestinal Microbiome/genetics , Kidney Transplantation/adverse effects , Adult , Aged , Clostridioides difficile/genetics , Clostridioides difficile/isolation & purification , DNA, Bacterial/isolation & purification , Dysbiosis/diagnosis , Dysbiosis/microbiology , Enteropathogenic Escherichia coli/genetics , Enteropathogenic Escherichia coli/isolation & purification , Female , Humans , Intestinal Mucosa/immunology , Intestinal Mucosa/microbiology , Male , Middle Aged , Norovirus/genetics , Norovirus/isolation & purification , RNA, Ribosomal, 16S/genetics , RNA, Viral/isolation & purification , Retrospective Studies
10.
Transpl Infect Dis ; 21(6): e13180, 2019 Dec.
Article in English | MEDLINE | ID: mdl-31544324

ABSTRACT

BACKGROUND: The gut microbiome is being associated increasingly with development of infections besides Clostridium difficile infection. A recent study found an association between butyrate-producing gut (BPG) bacteria and less frequent development of lower respiratory viral infections in allogeneic hematopoietic stem cell transplant recipients (Haak et al, Blood 131(26): 2978, 2018). In this investigation, we examine the relationship between the abundance of BPG bacteria and the development of viral infections in a cohort of kidney transplant recipients. METHODS: We recruited 168 kidney transplant recipients who provided 510 fecal specimens in the first 3 months after transplantation and profiled the gut microbiota using 16S rRNA gene sequencing of the V4-V5 hypervariable region. We classified the kidney transplant recipients into higher BPG Bacteria Group and lower BPG Bacteria Group using the same criteria of 1% relative gut abundance of BPG bacteria as the Haak et al study. RESULTS: Administration of antibiotics against anaerobes was associated with a significant decrease in the relative gut abundance of BPG bacteria. The higher BPG Bacteria Group was associated with less development of respiratory viral infections (Hazard Ratio [HR]: 0.28, P = .01) but not with less development of CMV viremia (HR: 0.38, P = .13) or BK viremia (HR: 1.02, P = .98) at 2 years post transplantation. CONCLUSION: Our pilot investigation supports future validation of the relationship between high relative gut abundance of BPG bacteria and decreased risk for development of respiratory viral infections.


Subject(s)
Bacteria/isolation & purification , Gastrointestinal Microbiome/physiology , Kidney Transplantation/adverse effects , Respiratory Tract Infections/epidemiology , Virus Diseases/epidemiology , Anti-Bacterial Agents/adverse effects , Antibiotic Prophylaxis/adverse effects , Bacteria/drug effects , Bacteria/immunology , Bacteria/metabolism , Bacterial Infections/immunology , Bacterial Infections/prevention & control , Butyrates/metabolism , DNA, Bacterial/isolation & purification , Feces/microbiology , Female , Follow-Up Studies , Gastrointestinal Microbiome/drug effects , Graft Rejection/immunology , Graft Rejection/prevention & control , Humans , Immunosuppressive Agents/adverse effects , Male , Middle Aged , Pilot Projects , RNA, Ribosomal, 16S/genetics , Respiratory Tract Infections/immunology , Respiratory Tract Infections/virology , Virus Diseases/immunology , Virus Diseases/virology
11.
Am J Transplant ; 18(10): 2429-2442, 2018 10.
Article in English | MEDLINE | ID: mdl-29659169

ABSTRACT

Advances in bioinformatics allow identification of single nucleotide polymorphisms (variants) from RNA sequence data. In an allograft biopsy, 2 genomes contribute to the RNA pool, 1 from the donor organ and the other from the infiltrating recipient's cells. We hypothesize that imbalances in genetic variants of RNA sequence data of kidney allograft biopsies provide an objective measure of cellular infiltration of the allograft. We performed mRNA sequencing of 40 kidney allograft biopsies, selected to represent a comprehensive range of diagnostic categories. We analyzed the sequencing reads of these biopsies and of 462 lymphoblastoid cell lines from the 1000 Genomes Project, for RNA variants. The ratio of heterozygous to nonreference genome homozygous variants (Het/Hom ratio) on all autosomes was determined for each sample, and the estimation of stromal and immune cells in malignant tumors using expression data (ESTIMATE) score was computed as a complementary estimate of the degree of cellular infiltration into biopsies. The Het/Hom ratios (P = .02) and the ESTIMATE scores (P < .001) were associated with the biopsy diagnosis. Both measures correlated significantly (r = .67, P < .0001), even though the Het/Hom ratio is based on mRNA sequence variation, while the ESTIMATE score uses mRNA expression. Het/Hom ratio and the ESTIMATE score may offer unbiased and quantitative parameters for characterizing cellular traffic into human kidney allografts.


Subject(s)
Biomarkers/analysis , Graft Rejection/diagnosis , High-Throughput Nucleotide Sequencing/methods , Kidney Transplantation/adverse effects , Polymorphism, Single Nucleotide , Adult , Allografts , Computational Biology , Female , Graft Rejection/etiology , Heterozygote , Homozygote , Humans , Male , Middle Aged
12.
Immunol Rev ; 258(1): 218-40, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24517436

ABSTRACT

Kidney allograft status is currently characterized using the invasive percutaneous needle core biopsy procedure. The procedure has become safer over the years, but challenges and complications still exist including sampling error, interobserver variability, bleeding, arteriovenous fistula, graft loss, and even death. Because the most common type of acute rejection is distinguished by inflammatory cells exiting the intravascular compartment and gaining access to the renal tubular space, we reasoned that a kidney allograft may function as an in vivo flow cytometer and sort cells involved in rejection into urine. To test this idea, we developed quantitative polymerase chain reaction (PCR) assays for absolute quantification of mRNA and pre-amplification protocols to overcome the low RNA yield from urine. Here, we review our single center urinary cell mRNA profiling studies that led to the multicenter Clinical Trials in Organ Transplantation (CTOT-04) study and the discovery and validation of a 3-gene signature of 18S rRNA-normalized measures of CD3ε mRNA and IP-10 mRNA and 18S rRNA that is diagnostic and predictive of acute cellular rejection in the kidney allograft. We also review our development of a 4-gene signature of mRNAs for vimentin, NKCC2, E-cadherin, and 18S rRNA diagnostic of interstitial fibrosis/tubular atrophy (IF/TA).


Subject(s)
Gene Expression Profiling , Genetic Testing , Graft Rejection/genetics , Graft Rejection/urine , Kidney Transplantation , RNA, Messenger/urine , Allografts , Biopsy , Gene Expression Profiling/methods , Genetic Markers , Genetic Testing/methods , Graft Rejection/diagnosis , Graft Rejection/immunology , Humans , Kidney Transplantation/adverse effects , Polymerase Chain Reaction , Predictive Value of Tests , Treatment Outcome
13.
PLoS Comput Biol ; 12(9): e1005088, 2016 Sep.
Article in English | MEDLINE | ID: mdl-27684477

ABSTRACT

Current strategies to improve graft outcome following kidney transplantation consider information at the human leukocyte antigen (HLA) loci. Cell surface antigens, in addition to HLA, may serve as the stimuli as well as the targets for the anti-allograft immune response and influence long-term graft outcomes. We therefore performed exome sequencing of DNA from kidney graft recipients and their living donors and estimated all possible cell surface antigens mismatches for a given donor/recipient pair by computing the number of amino acid mismatches in trans-membrane proteins. We designated this tally as the allogenomics mismatch score (AMS). We examined the association between the AMS and post-transplant estimated glomerular filtration rate (eGFR) using mixed models, considering transplants from three independent cohorts (a total of 53 donor-recipient pairs, 106 exomes, and 239 eGFR measurements). We found that the AMS has a significant effect on eGFR (mixed model, effect size across the entire range of the score: -19.4 [-37.7, -1.1], P = 0.0042, χ2 = 8.1919, d.f. = 1) that is independent of the HLA-A, B, DR matching, donor age, and time post-transplantation. The AMS effect is consistent across the three independent cohorts studied and similar to the strong effect size of donor age. Taken together, these results show that the AMS, a novel tool to quantify amino acid mismatches in trans-membrane proteins in individual donor/recipient pair, is a strong, robust predictor of long-term graft function in kidney transplant recipients.

14.
Clin Transplant ; 31(11)2017 Nov.
Article in English | MEDLINE | ID: mdl-28921709

ABSTRACT

We studied the causes and predictors of death-censored kidney allograft failure among 1670 kidney recipients transplanted at our center in the corticosteroid-free maintenance immunosuppression era. As of January 1, 2012, we identified 137 recipients with allograft failure; 130 of them (cases) were matched 1-1 for recipient age, calendar year of transplant, and donor type with 130 recipients with functioning grafts (controls). Median time to allograft failure was 29 months (interquartile range: 18-51). Physician-validated and biopsy-confirmed categories of allograft failure were as follows: acute rejection (21%), glomerular disease (19%), transplant glomerulopathy (13%), interstitial fibrosis tubular atrophy (10%), and polyomavirus-associated nephropathy (7%). Graft failures were attributed to medical conditions in 21% and remained unresolved in 9%. Donor race, donor age, human leukocyte antigen mismatches, serum creatinine, urinary protein, acute cellular rejection, acute antibody-mediated rejection, BK viremia, and CMV viremia were associated with allograft failure. Independent predictors of allograft failure were acute cellular rejection (odds ratio: 18.31, 95% confidence interval: 5.28-63.45) and urine protein ≥1 g/d within the first year post-transplantation (5.85, 2.37-14.45). Serum creatinine ≤1.5 mg/dL within the first year post-transplantation reduced the odds (0.29, 0.13-0.64) of allograft failure. Our study has identified modifiable risk factors to reduce the burden of allograft failure.


Subject(s)
Adrenal Cortex Hormones , Graft Rejection/etiology , Graft Survival/drug effects , Immunosuppressive Agents/therapeutic use , Kidney Failure, Chronic/surgery , Kidney Transplantation/adverse effects , Adult , Case-Control Studies , Female , Follow-Up Studies , Glomerular Filtration Rate , Graft Rejection/drug therapy , Graft Rejection/pathology , Humans , Kidney Function Tests , Male , Middle Aged , Postoperative Complications , Prognosis , Retrospective Studies , Risk Factors
15.
J Am Soc Nephrol ; 27(2): 626-36, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26047788

ABSTRACT

Noninvasive diagnosis and prognostication of acute cellular rejection in the kidney allograft may help realize the full benefits of kidney transplantation. To investigate whether urine metabolites predict kidney allograft status, we determined levels of 749 metabolites in 1516 urine samples from 241 kidney graft recipients enrolled in the prospective multicenter Clinical Trials in Organ Transplantation-04 study. A metabolite signature of the ratio of 3-sialyllactose to xanthosine in biopsy specimen-matched urine supernatants best discriminated acute cellular rejection biopsy specimens from specimens without rejection. For clinical application, we developed a high-throughput mass spectrometry-based assay that enabled absolute and rapid quantification of the 3-sialyllactose-to-xanthosine ratio in urine samples. A composite signature of ratios of 3-sialyllactose to xanthosine and quinolinate to X-16397 and our previously reported urinary cell mRNA signature of 18S ribosomal RNA, CD3ε mRNA, and interferon-inducible protein-10 mRNA outperformed the metabolite signatures and the mRNA signature. The area under the receiver operating characteristics curve for the composite metabolite-mRNA signature was 0.93, and the signature was diagnostic of acute cellular rejection with a specificity of 84% and a sensitivity of 90%. The composite signature, developed using solely biopsy specimen-matched urine samples, predicted future acute cellular rejection when applied to pristine samples taken days to weeks before biopsy. We conclude that metabolite profiling of urine offers a noninvasive means of diagnosing and prognosticating acute cellular rejection in the human kidney allograft, and that the combined metabolite and mRNA signature is diagnostic and prognostic of acute cellular rejection with very high accuracy.


Subject(s)
Allografts/metabolism , Graft Rejection/urine , Kidney Transplantation , Kidney/metabolism , Acute Disease , Adolescent , Adult , Aged , Child , Child, Preschool , Female , Graft Rejection/metabolism , Humans , Infant , Male , Middle Aged , Predictive Value of Tests , Prospective Studies , Young Adult
16.
N Engl J Med ; 369(1): 20-31, 2013 Jul 04.
Article in English | MEDLINE | ID: mdl-23822777

ABSTRACT

BACKGROUND: The standard test for the diagnosis of acute rejection in kidney transplants is the renal biopsy. Noninvasive tests would be preferable. METHODS: We prospectively collected 4300 urine specimens from 485 kidney-graft recipients from day 3 through month 12 after transplantation. Messenger RNA (mRNA) levels were measured in urinary cells and correlated with allograft-rejection status with the use of logistic regression. RESULTS: A three-gene signature of 18S ribosomal (rRNA)-normalized measures of CD3ε mRNA and interferon-inducible protein 10 (IP-10) mRNA, and 18S rRNA discriminated between biopsy specimens showing acute cellular rejection and those not showing rejection (area under the curve [AUC], 0.85; 95% confidence interval [CI], 0.78 to 0.91; P<0.001 by receiver-operating-characteristic curve analysis). The cross-validation estimate of the AUC was 0.83 by bootstrap resampling, and the Hosmer-Lemeshow test indicated good fit (P=0.77). In an external-validation data set, the AUC was 0.74 (95% CI, 0.61 to 0.86; P<0.001) and did not differ significantly from the AUC in our primary data set (P=0.13). The signature distinguished acute cellular rejection from acute antibody-mediated rejection and borderline rejection (AUC, 0.78; 95% CI, 0.68 to 0.89; P<0.001). It also distinguished patients who received anti-interleukin-2 receptor antibodies from those who received T-cell-depleting antibodies (P<0.001) and was diagnostic of acute cellular rejection in both groups. Urinary tract infection did not affect the signature (P=0.69). The average trajectory of the signature in repeated urine samples remained below the diagnostic threshold for acute cellular rejection in the group of patients with no rejection, but in the group with rejection, there was a sharp rise during the weeks before the biopsy showing rejection (P<0.001). CONCLUSIONS: A molecular signature of CD3ε mRNA, IP-10 mRNA, and 18S rRNA levels in urinary cells appears to be diagnostic and prognostic of acute cellular rejection in kidney allografts. (Funded by the National Institutes of Health and others.).


Subject(s)
Chemokine CXCL10/genetics , Graft Rejection/diagnosis , Intracellular Signaling Peptides and Proteins/genetics , Kidney Transplantation , RNA, Messenger/urine , RNA, Ribosomal/urine , Acute Disease , Adult , Area Under Curve , Chemokine CXCL10/urine , Female , Graft Rejection/genetics , Humans , Intracellular Signaling Peptides and Proteins/urine , Male , Middle Aged , Prospective Studies , RNA Polymerase I , RNA, Ribosomal, 18S/urine , ROC Curve , Real-Time Polymerase Chain Reaction , Sensitivity and Specificity , Transcriptome
17.
J Am Soc Nephrol ; 25(7): 1586-97, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24610929

ABSTRACT

Noninvasive tests to differentiate the basis for acute dysfunction of the kidney allograft are preferable to invasive allograft biopsies. We measured absolute levels of 26 prespecified mRNAs in urine samples collected from kidney graft recipients at the time of for-cause biopsy for acute allograft dysfunction and investigated whether differential diagnosis of acute graft dysfunction is feasible using urinary cell mRNA profiles. We profiled 52 urine samples from 52 patients with biopsy specimens indicating acute rejection (26 acute T cell-mediated rejection and 26 acute antibody-mediated rejection) and 32 urine samples from 32 patients with acute tubular injury without acute rejection. A stepwise quadratic discriminant analysis of mRNA measures identified a linear combination of mRNAs for CD3ε, CD105, TLR4, CD14, complement factor B, and vimentin that distinguishes acute rejection from acute tubular injury; 10-fold cross-validation of the six-gene signature yielded an estimate of the area under the curve of 0.92 (95% confidence interval, 0.86 to 0.98). In a decision analysis, the six-gene signature yielded the highest net benefit across a range of reasonable threshold probabilities for biopsy. Next, among patients diagnosed with acute rejection, a similar statistical approach identified a linear combination of mRNAs for CD3ε, CD105, CD14, CD46, and 18S rRNA that distinguishes T cell-mediated rejection from antibody-mediated rejection, with a cross-validated estimate of the area under the curve of 0.81 (95% confidence interval, 0.68 to 0.93). Incorporation of these urinary cell mRNA signatures in clinical decisions may reduce the number of biopsies in patients with acute dysfunction of the kidney allograft.


Subject(s)
Graft Rejection/diagnosis , Graft Rejection/urine , Kidney Transplantation , Primary Graft Dysfunction/diagnosis , Primary Graft Dysfunction/urine , RNA, Messenger/urine , Acute Disease , Diagnosis, Differential , Female , Humans , Kidney Tubules , Male , Middle Aged , Urine/cytology
18.
BMC Med ; 12: 125, 2014 Jul 29.
Article in English | MEDLINE | ID: mdl-25070128

ABSTRACT

BACKGROUND: The epidemics of HIV and hypertension are converging in sub-Saharan Africa. Due to antiretroviral therapy (ART), more HIV-infected adults are living longer and gaining weight, putting them at greater risk for hypertension and kidney disease. The relationship between hypertension, kidney disease and long-term ART among African adults, though, remains poorly defined. Therefore, we determined the prevalences of hypertension and kidney disease in HIV-infected adults (ART-naive and on ART >2 years) compared to HIV-negative adults. We hypothesized that there would be a higher hypertension prevalence among HIV-infected adults on ART, even after adjusting for age and adiposity. METHODS: In this cross-sectional study conducted between October 2012 and April 2013, consecutive adults (>18 years old) attending an HIV clinic in Tanzania were enrolled in three groups: 1) HIV-negative controls, 2) HIV-infected, ART-naive, and 3) HIV-infected on ART for >2 years. The main study outcomes were hypertension and kidney disease (both defined by international guidelines). We compared hypertension prevalence between each HIV group versus the control group by Fisher's exact test. Logistic regression was used to determine if differences in hypertension prevalence were fully explained by confounding. RESULTS: Among HIV-negative adults, 25/153 (16.3%) had hypertension (similar to recent community survey data). HIV-infected adults on ART had a higher prevalence of hypertension (43/150 (28.7%), P = 0.01) and a higher odds of hypertension even after adjustment (odds ratio (OR) = 2.19 (1.18 to 4.05), P = 0.01 in the best model). HIV-infected, ART-naive adults had a lower prevalence of hypertension (8/151 (5.3%), P = 0.003) and a lower odds of hypertension after adjustment (OR= 0.35 (0.15 to 0.84), P = 0.02 in the best model). Awareness of hypertension was ≤ 25% among hypertensive adults in all three groups. Kidney disease was common in all three groups (25.6% to 41.3%) and strongly associated with hypertension (P <0.001 for trend); among hypertensive participants, 50/76 (65.8%) had microalbuminuria and 20/76 (26.3%) had an estimated glomerular filtration rate (eGFR) <60 versus 33/184 (17.9%) and 16/184 (8.7%) participants with normal blood pressure. CONCLUSIONS: HIV-infected adults on ART >2 years had two-fold greater odds of hypertension than HIV-negative controls. HIV-infected adults with hypertension were rarely aware of their diagnosis but often have evidence of kidney disease. Intensive hypertension screening and education are needed in HIV-clinics in sub-Saharan Africa. Further studies should determine if chronic, dysregulated inflammation may accelerate hypertension in this population.


Subject(s)
HIV Infections/epidemiology , Renal Insufficiency/epidemiology , Adult , Anti-Retroviral Agents/administration & dosage , Comorbidity , Cross-Sectional Studies , Female , HIV Infections/complications , Humans , Hypertension/complications , Hypertension/epidemiology , Logistic Models , Male , Middle Aged , Odds Ratio , Prevalence , Renal Insufficiency/complications , Tanzania/epidemiology
19.
J Am Soc Nephrol ; 24(9): 1376-85, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23787913

ABSTRACT

The ability to achieve immunologic tolerance after transplantation is a therapeutic goal. Here, we report interim results from an ongoing trial of tolerance in HLA-identical sibling renal transplantation. The immunosuppressive regimen included alemtuzumab induction, donor hematopoietic stem cells, tacrolimus/mycophenolate immunosuppression converted to sirolimus, and complete drug withdrawal by 24 months post-transplantation. Recipients were considered tolerant if they had normal biopsies and renal function after an additional 12 months without immunosuppression. Of the 20 recipients enrolled, 10 had at least 36 months of follow-up after transplantation. Five of these 10 recipients had immunosuppression successfully withdrawn for 16-36 months (tolerant), 2 had disease recurrence, and 3 had subclinical rejection in protocol biopsies (nontolerant). Microchimerism disappeared after 1 year, and CD4(+)CD25(high)CD127(-)FOXP3(+) regulatory T cells and CD19(+)IgD/M(+)CD27(-) B cells were increased through 5 years post-transplantation in both tolerant and nontolerant recipients. Immune/inflammatory gene expression pathways in the peripheral blood and urine, however, were differentially downregulated between tolerant and nontolerant recipients. In summary, interim results from this trial of tolerance in HLA-identical renal transplantation suggest that predictive genomic biomarkers, but not immunoregulatory phenotyping, may be able to discriminate tolerant from nontolerant patients.


Subject(s)
DNA/genetics , Genome/genetics , HLA Antigens/genetics , HLA Antigens/immunology , Immune Tolerance/immunology , Kidney Transplantation/immunology , Adult , Biomarkers , Biopsy , Chimerism , Female , Follow-Up Studies , Graft Rejection/immunology , Graft Rejection/prevention & control , Hematopoietic Stem Cell Transplantation , Humans , Immunosuppressive Agents/therapeutic use , Kidney/pathology , Kidney Transplantation/pathology , Male , Middle Aged , Retrospective Studies
20.
J Immunol Methods ; 532: 113714, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38936464

ABSTRACT

INTRODUCTION: Acute rejection (AR) undermines the life-extending benefits of kidney transplantation and is diagnosed using the invasive biopsy procedure. T cell-mediated rejection (TCMR), antibody-mediated rejection (ABMR), or concurrent TCMR + ABMR (Mixed Rejection [MR]) are the three major types of AR. Development of noninvasive biomarkers diagnostic of AR due to any of the three types is a useful addition to the diagnostic armamentarium. METHODS: We developed customized RT-qPCR assays and measured urinary cell mRNA copy numbers in 145 biopsy-matched urine samples from 126 kidney allograft recipients. We determined whether the urinary cell three-gene signature diagnostic of TCMR (Suthanthiran et al., 2013) discriminates patients with no rejection biopsies (NR, n = 50) from those with ABMR (n = 28) or MR (n = 20) biopsies. RESULTS: The urinary cell three-gene signature discriminated all three types of rejection biopsies from NR biopsies (P < 0.0001, One-way ANOVA). Dunnett's multiple comparisons test yielded P < 0.0001 for NR vs. TCMR; P < 0.001 for NR vs. ABMR; and P < 0.0001 for NR vs. MR. By bootstrap resampling, optimism-corrected area under the receiver operating characteristic curve (AUC) was 0.749 (bias-corrected 95% confidence interval [CI], 0.638 to 0.840) for NR vs. TCMR (P < 0.0001); 0.780 (95% CI, 0.656 to 0.878) for NR vs. ABMR (P < 0.0001); and 0.857 (95% CI, 0.727 to 0.947) for NR vs. MR (P < 0.0001). All three rejection categories were distinguished from NR biopsies with similar accuracy (all AUC comparisons P > 0.05). CONCLUSION: The urinary cell three-gene signature score discriminates AR due to TCMR, ABMR or MR from NR biopsies in human kidney allograft recipients.


Subject(s)
Graft Rejection , Kidney Transplantation , Humans , Graft Rejection/urine , Graft Rejection/diagnosis , Graft Rejection/immunology , Graft Rejection/genetics , Graft Rejection/pathology , Kidney Transplantation/adverse effects , Male , Female , Middle Aged , Adult , Biopsy , Biomarkers/urine , Transcriptome , Allografts/immunology , Gene Expression Profiling , Acute Disease , Aged , ROC Curve
SELECTION OF CITATIONS
SEARCH DETAIL