Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
1.
Proc Natl Acad Sci U S A ; 113(40): 11306-11311, 2016 10 04.
Article in English | MEDLINE | ID: mdl-27655895

ABSTRACT

Although multiagent combination chemotherapy is curative in a significant fraction of childhood acute lymphoblastic leukemia (ALL) patients, 20% of cases relapse and most die because of chemorefractory disease. Here we used whole-exome and whole-genome sequencing to analyze the mutational landscape at relapse in pediatric ALL cases. These analyses identified numerous relapse-associated mutated genes intertwined in chemotherapy resistance-related protein complexes. In this context, RAS-MAPK pathway-activating mutations in the neuroblastoma RAS viral oncogene homolog (NRAS), kirsten rat sarcoma viral oncogene homolog (KRAS), and protein tyrosine phosphatase, nonreceptor type 11 (PTPN11) genes were present in 24 of 55 (44%) cases in our series. Interestingly, some leukemias showed retention or emergence of RAS mutant clones at relapse, whereas in others RAS mutant clones present at diagnosis were replaced by RAS wild-type populations, supporting a role for both positive and negative selection evolutionary pressures in clonal evolution of RAS-mutant leukemia. Consistently, functional dissection of mouse and human wild-type and mutant RAS isogenic leukemia cells demonstrated induction of methotrexate resistance but also improved the response to vincristine in mutant RAS-expressing lymphoblasts. These results highlight the central role of chemotherapy-driven selection as a central mechanism of leukemia clonal evolution in relapsed ALL, and demonstrate a previously unrecognized dual role of RAS mutations as drivers of both sensitivity and resistance to chemotherapy.


Subject(s)
Clonal Evolution/genetics , Genes, ras , Mutation/genetics , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology , Base Sequence , Cell Line, Tumor , Drug Resistance, Neoplasm/drug effects , Drug Resistance, Neoplasm/genetics , Humans , Methotrexate/pharmacology , Methotrexate/therapeutic use , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Vincristine/pharmacology , Vincristine/therapeutic use
2.
J Immunol ; 197(2): 644-54, 2016 07 15.
Article in English | MEDLINE | ID: mdl-27271569

ABSTRACT

CD8(+) T cells develop increased sensitivity following Ag experience, and differences in sensitivity exist between T cell memory subsets. How differential TCR signaling between memory subsets contributes to sensitivity differences is unclear. We show in mouse effector memory T cells (TEM) that >50% of lymphocyte-specific protein tyrosine kinase (Lck) exists in a constitutively active conformation, compared with <20% in central memory T cells (TCM). Immediately proximal to Lck signaling, we observed enhanced Zap-70 phosphorylation in TEM following TCR ligation compared with TCM Furthermore, we observed superior cytotoxic effector function in TEM compared with TCM, and we provide evidence that this results from a lower probability of TCM reaching threshold signaling owing to the decreased magnitude of TCR-proximal signaling. We provide evidence that the differences in Lck constitutive activity between CD8(+) TCM and TEM are due to differential regulation by SH2 domain-containing phosphatase-1 (Shp-1) and C-terminal Src kinase, and we use modeling of early TCR signaling to reveal the significance of these differences. We show that inhibition of Shp-1 results in increased constitutive Lck activity in TCM to levels similar to TEM, as well as increased cytotoxic effector function in TCM Collectively, this work demonstrates a role for constitutive Lck activity in controlling Ag sensitivity, and it suggests that differential activities of TCR-proximal signaling components may contribute to establishing the divergent effector properties of TCM and TEM. This work also identifies Shp-1 as a potential target to improve the cytotoxic effector functions of TCM for adoptive cell therapy applications.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Immunologic Memory/physiology , Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/immunology , T-Lymphocyte Subsets/immunology , Animals , Blotting, Western , CD8-Positive T-Lymphocytes/metabolism , Cytotoxicity, Immunologic/immunology , Flow Cytometry , Humans , Lymphocyte Activation/immunology , Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/metabolism , Markov Chains , Mice , Polymerase Chain Reaction , T-Lymphocyte Subsets/metabolism , Time-Lapse Imaging
3.
J Transl Med ; 12: 342, 2014 Dec 10.
Article in English | MEDLINE | ID: mdl-25491880

ABSTRACT

BACKGROUND: Matrix metalloproteinase-23 (MMP-23) can block the voltage-gated potassium channel Kv1.3, whose function is important for sustained Ca(2+) signaling during T cell activation. MMP-23 may also alter T cell activity and phenotype through cleavage of proteins affecting cytokine and chemokine signaling. We therefore tested the hypothesis that MMP-23 can negatively regulate the anti-tumor T cell response in human melanoma. METHODS: We characterized MMP-23 expression in primary melanoma patients who received adjuvant immunotherapy. We examined the association of MMP-23 with the anti-tumor immune response - as assessed by the prevalence of tumor-infiltrating lymphocytes and Foxp3(+) regulatory T cells. Further, we examined the association between MMP-23 expression and response to immunotherapy. Considering also an in trans mechanism, we examined the association of melanoma MMP-23 and melanoma Kv1.3 expression. RESULTS: Our data revealed an inverse association between primary melanoma MMP-23 expression and the anti-tumor T cell response, as demonstrated by decreased tumor-infiltrating lymphocytes (TIL) (P = 0.05), in particular brisk TILs (P = 0.04), and a trend towards an increased proportion of immunosuppressive Foxp3(+) regulatory T cells (P = 0.07). High melanoma MMP-23 expression is also associated with recurrence in patients treated with immune biologics (P = 0.037) but not in those treated with vaccines (P = 0.64). Further, high melanoma MMP-23 expression is associated with shorter periods of progression-free survival for patients receiving immune biologics (P = 0.025). On the other hand, there is no relationship between melanoma MMP-23 and melanoma Kv1.3 expression (P = 0.27). CONCLUSIONS: Our data support a role for MMP-23 as a potential immunosuppressive target in melanoma, as well as a possible biomarker for informing melanoma immunotherapies.


Subject(s)
Immunotherapy , Matrix Metalloproteinases/metabolism , Melanoma/enzymology , Adult , Aged , Aged, 80 and over , Female , Humans , Male , Melanoma/immunology , Melanoma/therapy , Middle Aged , T-Lymphocytes, Regulatory/immunology , Young Adult
4.
Nat Genet ; 46(2): 166-70, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24413734

ABSTRACT

Peripheral T cell lymphomas (PTCLs) are a heterogeneous and poorly understood group of non-Hodgkin lymphomas. Here we combined whole-exome sequencing of 12 tumor-normal DNA pairs, RNA sequencing analysis and targeted deep sequencing to identify new genetic alterations in PTCL transformation. These analyses identified highly recurrent epigenetic factor mutations in TET2, DNMT3A and IDH2 as well as a new highly prevalent RHOA mutation encoding a p.Gly17Val alteration present in 22 of 35 (67%) angioimmunoblastic T cell lymphoma (AITL) samples and in 8 of 44 (18%) PTCL, not otherwise specified (PTCL-NOS) samples. Mechanistically, the RHOA Gly17Val protein interferes with RHOA signaling in biochemical and cellular assays, an effect potentially mediated by the sequestration of activated guanine-exchange factor (GEF) proteins. In addition, we describe new and recurrent, albeit less frequent, genetic defects including mutations in FYN, ATM, B2M and CD58 implicating SRC signaling, impaired DNA damage response and escape from immune surveillance mechanisms in the pathogenesis of PTCL.


Subject(s)
Epigenesis, Genetic/genetics , Lymphoma, T-Cell, Peripheral/genetics , Proto-Oncogene Proteins c-fyn/genetics , rhoA GTP-Binding Protein/genetics , Ataxia Telangiectasia Mutated Proteins/genetics , Base Sequence , CD58 Antigens/genetics , Computational Biology , DNA (Cytosine-5-)-Methyltransferases/genetics , DNA Methyltransferase 3A , DNA-Binding Proteins/genetics , Dioxygenases , Escherichia coli , Exome/genetics , Fluorescent Antibody Technique , HEK293 Cells , High-Throughput Nucleotide Sequencing , Humans , Isocitrate Dehydrogenase/genetics , Molecular Sequence Data , Mutation, Missense/genetics , Proto-Oncogene Proteins/genetics , Sequence Analysis, RNA
5.
Cancer Cell ; 24(6): 766-76, 2013 Dec 09.
Article in English | MEDLINE | ID: mdl-24291004

ABSTRACT

Glucocorticoid resistance is a major driver of therapeutic failure in T cell acute lymphoblastic leukemia (T-ALL). Here, we identify the AKT1 kinase as a major negative regulator of the NR3C1 glucocorticoid receptor protein activity driving glucocorticoid resistance in T-ALL. Mechanistically, AKT1 impairs glucocorticoid-induced gene expression by direct phosphorylation of NR3C1 at position S134 and blocking glucocorticoid-induced NR3C1 translocation to the nucleus. Moreover, we demonstrate that loss of PTEN and consequent AKT1 activation can effectively block glucocorticoid-induced apoptosis and induce resistance to glucocorticoid therapy. Conversely, pharmacologic inhibition of AKT with MK2206 effectively restores glucocorticoid-induced NR3C1 translocation to the nucleus, increases the response of T-ALL cells to glucocorticoid therapy, and effectively reverses glucocorticoid resistance in vitro and in vivo.


Subject(s)
Dexamethasone/therapeutic use , Heterocyclic Compounds, 3-Ring/pharmacology , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Active Transport, Cell Nucleus , Animals , Drug Resistance, Neoplasm , Humans , Mice , PTEN Phosphohydrolase/physiology , Phosphorylation , Proto-Oncogene Proteins c-akt/physiology , Receptors, Glucocorticoid/metabolism
6.
Blood ; 122(14): 2425-32, 2013 Oct 03.
Article in English | MEDLINE | ID: mdl-23908464

ABSTRACT

The SH2B adaptor protein 3 (SH2B3) gene encodes a negative regulator of cytokine signaling with a critical role in the homeostasis of hematopoietic stem cells and lymphoid progenitors. Here, we report the identification of germline homozygous SH2B3 mutations in 2 siblings affected with developmental delay and autoimmunity, one in whom B-precursor acute lymphoblastic leukemia (ALL) developed. Mechanistically, loss of SH2B3 increases Janus kinase-signal transducer and activator of transcription signaling, promotes lymphoid cell proliferation, and accelerates leukemia development in a mouse model of NOTCH1-induced ALL. Moreover, extended mutation analysis showed homozygous somatic mutations in SH2B3 in 2 of 167 ALLs analyzed. Overall, these results demonstrate a Knudson tumor suppressor role for SH2B3 in the pathogenesis of ALL and highlight a possible link between genetic predisposition factors in the pathogenesis of autoimmunity and leukemogenesis.


Subject(s)
Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Proteins/genetics , Adaptor Proteins, Signal Transducing , Animals , Autoimmune Diseases/genetics , Base Sequence , Blotting, Western , Child , Child, Preschool , DNA Mutational Analysis , Developmental Disabilities/genetics , Female , Genotype , Germ-Line Mutation , Humans , Infant , Infant, Newborn , Intracellular Signaling Peptides and Proteins , Male , Mice , Mice, Inbred C57BL , Molecular Sequence Data , Oligonucleotide Array Sequence Analysis , Pedigree , Siblings
7.
Hum Immunol ; 74(9): 1219-24, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23756164

ABSTRACT

Cytotoxic T lymphocyte antigen 4 (CTLA-4) plays a key inhibitory role during T lymphocyte activation. The CTLA4 gene is translated into two proteic isoforms: a full-length protein (flCTLA-4) and a soluble counterpart. We explored the expression of both isoforms on healthy subjects. Whereas in non-stimulated cells the flCTLA-4 isoform is predominant, after stimulation the expression of the soluble form rapidly increases, reaching its maximum 24h after and falling again to the basal levels 72 h after stimulation. In contrast, the flCTLA-4 mRNA levels increase is slower, reaching the maximum level 72 h after stimulation. The presence of the T allele in the promoter positions -1722 and -318 is associated with an increased transcriptional activity and this effect seems to be synergic. We conclude that the kinetics of CTLA-4 isoform expression are sequential, and that the promoter polymorphisms -1722(C/T) and -318(C/T) are involved in the control of the CTLA4 transcription.


Subject(s)
CTLA-4 Antigen/metabolism , Protein Isoforms/metabolism , T-Lymphocytes/immunology , CTLA-4 Antigen/immunology , Cells, Cultured , Cloning, Molecular , Enzyme-Linked Immunosorbent Assay , Gene Expression Regulation/genetics , Genotype , Humans , Lymphocyte Activation , Polymorphism, Genetic , Promoter Regions, Genetic/genetics , Protein Isoforms/immunology , Spain , Transgenes/genetics
8.
Cell ; 153(7): 1552-66, 2013 Jun 20.
Article in English | MEDLINE | ID: mdl-23791182

ABSTRACT

Sequencing efforts led to the identification of somatic mutations that could affect the self-renewal and differentiation of cancer-initiating cells. One such recurrent mutation targets the binding pocket of the ubiquitin ligase Fbxw7. Missense FBXW7 mutations are prevalent in various tumors, including T cell acute lymphoblastic leukemia (T-ALL). To study the effects of such lesions, we generated animals carrying regulatable Fbxw7 mutant alleles. Here, we show that these mutations specifically bolster cancer-initiating cell activity in collaboration with Notch1 oncogenes but spare normal hematopoietic stem cell function. We were also able to show that FBXW7 mutations specifically affect the ubiquitylation and half-life of c-Myc protein, a key T-ALL oncogene. Using animals carrying c-Myc fusion alleles, we connected Fbxw7 function to c-Myc abundance and correlated c-Myc expression to leukemia-initiating activity. Finally, we demonstrated that small-molecule-mediated suppression of MYC activity leads to T-ALL remission, suggesting an effective therapeutic strategy.


Subject(s)
Cell Cycle Proteins/metabolism , F-Box Proteins/metabolism , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Proto-Oncogene Proteins c-myc/metabolism , Ubiquitin-Protein Ligases/metabolism , Animals , Cell Cycle Proteins/genetics , Disease Models, Animal , F-Box Proteins/genetics , F-Box-WD Repeat-Containing Protein 7 , Hematopoietic Stem Cells/metabolism , Humans , Mice , Mice, Knockout , Mutation, Missense , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Proto-Oncogene Proteins c-myc/antagonists & inhibitors , Receptor, Notch1/metabolism , Tumor Suppressor Protein p53/metabolism , Ubiquitin-Protein Ligases/genetics , Ubiquitination
9.
Nat Med ; 19(3): 368-71, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23377281

ABSTRACT

Acute lymphoblastic leukemia (ALL) is an aggressive hematological tumor resulting from the malignant transformation of lymphoid progenitors. Despite intensive chemotherapy, 20% of pediatric patients and over 50% of adult patients with ALL do not achieve a complete remission or relapse after intensified chemotherapy, making disease relapse and resistance to therapy the most substantial challenge in the treatment of this disease. Using whole-exome sequencing, we identify mutations in the cytosolic 5'-nucleotidase II gene (NT5C2), which encodes a 5'-nucleotidase enzyme that is responsible for the inactivation of nucleoside-analog chemotherapy drugs, in 20/103 (19%) relapse T cell ALLs and 1/35 (3%) relapse B-precursor ALLs. NT5C2 mutant proteins show increased nucleotidase activity in vitro and conferred resistance to chemotherapy with 6-mercaptopurine and 6-thioguanine when expressed in ALL lymphoblasts. These results support a prominent role for activating mutations in NT5C2 and increased nucleoside-analog metabolism in disease progression and chemotherapy resistance in ALL.


Subject(s)
5'-Nucleotidase/genetics , Antineoplastic Agents/therapeutic use , Drug Resistance, Neoplasm/genetics , Mercaptopurine/therapeutic use , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , 5'-Nucleotidase/metabolism , Arabinonucleosides/pharmacology , Arabinonucleosides/therapeutic use , Base Sequence , Cell Line , HEK293 Cells , Humans , Mutation , Precursor Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Recurrence , Sequence Analysis, DNA , Thioguanine/therapeutic use
10.
Nat Med ; 18(3): 436-40, 2012 Feb 26.
Article in English | MEDLINE | ID: mdl-22366949

ABSTRACT

The TLX1 and TLX3 transcription factor oncogenes have a key role in the pathogenesis of T cell acute lymphoblastic leukemia (T-ALL). Here we used reverse engineering of global transcriptional networks to decipher the oncogenic regulatory circuit controlled by TLX1 and TLX3. This systems biology analysis defined T cell leukemia homeobox 1 (TLX1) and TLX3 as master regulators of an oncogenic transcriptional circuit governing T-ALL. Notably, a network structure analysis of this hierarchical network identified RUNX1 as a key mediator of the T-ALL induced by TLX1 and TLX3 and predicted a tumor-suppressor role for RUNX1 in T cell transformation. Consistent with these results, we identified recurrent somatic loss-of-function mutations in RUNX1 in human T-ALL. Overall, these results place TLX1 and TLX3 at the top of an oncogenic transcriptional network controlling leukemia development, show the power of network analyses to identify key elements in the regulatory circuits governing human cancer and identify RUNX1 as a tumor-suppressor gene in T-ALL.


Subject(s)
Core Binding Factor Alpha 2 Subunit/genetics , Core Binding Factor Alpha 2 Subunit/metabolism , Gene Regulatory Networks/genetics , Homeodomain Proteins/metabolism , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Proto-Oncogene Proteins/metabolism , Tumor Suppressor Proteins/metabolism , Cell Line, Tumor , Cell Transformation, Neoplastic , Homeodomain Proteins/genetics , Humans , Mutation , Oligonucleotide Array Sequence Analysis , Oncogenes/genetics , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Protein Conformation , Proto-Oncogene Proteins/genetics , Tumor Suppressor Proteins/genetics
11.
Biol Blood Marrow Transplant ; 18(1): 100-5, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21703972

ABSTRACT

CTLA-4 (cytotoxic T-lymphocyte antigen-4) plays a pivotal role in inhibiting T cell activation through competitive interaction with B7 molecules and interruption of costimulatory signals mediated by CD28. Polymorphisms on the CTLA-4 gene have been previously associated with autoimmune diseases, predisposition to leukemic relapse, and with graft-versus-host disease (GVHD) or relapse after allogeneic transplant. As CTLA-4 is expressed on T-lymphocytes, the aim of this study was to determine whether the donor CTLA-4 CT60 genotype also influences clinical outcome even after T cell depletion with CD34-positive selection. We studied 136 patient-donor pairs. Overall survival (OS) was worse for those patients who received grafts from a donor with the CT60 AA genotype rather than from a donor with the AG or GG genotype (35.6% vs 49.4%; P = .043). This association was confirmed through multivariate analysis, which identified the donor CT60 genotype as an independent risk factor for OS (P = .008; hazard ratio [HR]: 2.24, 95% confidence interval [CI]: 1.23-4.08). The donor CT60 AA genotype was also associated with lower disease-free survival, this being related to an increased risk of relapse (P = .001; HR: 3.41, 95% CI: 1.67-6.96) and a trend toward higher transplant-related mortality. These associations were stronger when considering only patients in the early stage of disease. Our results suggest that graft-versus-leukemia (GVL) activity after T cell depletion is conditioned by the donor CTLA-4 genotype.


Subject(s)
CTLA-4 Antigen/genetics , Hematopoietic Stem Cell Transplantation/methods , T-Lymphocytes/immunology , Tissue Donors , Adolescent , Adult , CTLA-4 Antigen/immunology , Disease-Free Survival , Female , Genotype , Hematopoietic Stem Cell Transplantation/adverse effects , Histocompatibility , Humans , Male , Middle Aged , Siblings , Survival Rate , Transplantation, Homologous , Treatment Outcome , Young Adult
12.
J Exp Med ; 208(13): 2571-9, 2011 Dec 19.
Article in English | MEDLINE | ID: mdl-22162831

ABSTRACT

Early immature T cell acute lymphoblastic leukemias (T-ALLs) account for ~5-10% of pediatric T-ALLs and are associated with poor prognosis. However, the genetic defects that drive the biology of these tumors remain largely unknown. In this study, analysis of microarray gene expression signatures in adult T-ALL demonstrated a high prevalence of early immature leukemias and revealed a close relationship between these tumors and myeloid leukemias. Many adult immature T-ALLs harbored mutations in myeloid-specific oncogenes and tumor suppressors including IDH1, IDH2, DNMT3A, FLT3, and NRAS. Moreover, we identified ETV6 mutations as a novel genetic lesion uniquely present in immature adult T-ALL. Our results demonstrate that early immature adult T-ALL represents a heterogeneous category of leukemias characterized by the presence of overlapping myeloid and T-ALL characteristics, and highlight the potential role of ETV6 mutations in these tumors.


Subject(s)
Mutation , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Proto-Oncogene Proteins c-ets/genetics , Proto-Oncogene Proteins c-ets/metabolism , Repressor Proteins/genetics , Repressor Proteins/metabolism , Adult , Cell Line, Tumor , Female , Gene Expression Profiling , Gene Expression Regulation, Leukemic/genetics , Genes, Tumor Suppressor , Humans , Infant , Male , Oligonucleotide Array Sequence Analysis , ETS Translocation Variant 6 Protein
13.
J Vis Exp ; (44)2010 Oct 22.
Article in English | MEDLINE | ID: mdl-21048669

ABSTRACT

T-cell receptors (TCRs) play a central role in the immune system. TCRs on T-cell surfaces can specifically recognize peptide antigens presented by antigen presenting cells (APCs). This recognition leads to the activation of T-cells and a series of functional outcomes (e.g. cytokine production, killing of the target cells). Understanding the functional role of TCRs is critical to harness the power of the immune system to treat a variety of immunology related diseases (e.g. cancer or autoimmunity). It is convenient to study TCRs in mouse models, which can be accomplished in several ways. Making TCR transgenic mouse models is costly and time-consuming and currently there are only a limited number of them available. Alternatively, mice with antigen-specific T-cells can be generated by bone marrow chimera. This method also takes several weeks and requires expertise. Retroviral transduction of TCRs into in vitro activated mouse T-cells is a quick and relatively easy method to obtain T-cells of desired peptide-MHC specificity. Antigen-specific T-cells can be generated in one week and used in any downstream applications. Studying transduced T-cells also has direct application to human immunotherapy, as adoptive transfer of human T-cells transduced with antigen-specific TCRs is an emerging strategy for cancer treatment. Here we present a protocol to retrovirally transduce TCRs into in vitro activated mouse T-cells. Both human and mouse TCR genes can be used. Retroviruses carrying specific TCR genes are generated and used to infect mouse T-cells activated with anti-CD3 and anti-CD28 antibodies. After in vitro expansion, transduced T-cells are analyzed by flow cytometry.


Subject(s)
Receptors, Antigen, T-Cell/genetics , Retroviridae/genetics , T-Lymphocytes/physiology , Transfection/methods , Animals , Epitopes, T-Lymphocyte/genetics , Epitopes, T-Lymphocyte/immunology , Humans , Mice , Models, Animal , Receptors, Antigen, T-Cell/immunology , T-Lymphocytes/immunology , Transduction, Genetic
14.
Transplantation ; 84(9): 1103-11, 2007 Nov 15.
Article in English | MEDLINE | ID: mdl-17998864

ABSTRACT

BACKGROUND: The presence of a few circulating donor cells in recipient's blood was first thought to be only an epiphenomenon of solid organ transplantation, also called microchimerism, but several authors have suggested that these circulating cells may contribute to tolerance induction. This study aims to assess the rate of microchimerism after kidney transplantation and determine its influence on acute rejection in a 4-year follow-up. METHODS: A total of 84 single-kidney recipients were included for microchimerism detection and quantification 2, 6, 12, and 18 months after transplantation by specific detection of non-shared STR, VNTR, human leukocyte antigen-A, -B, -DRB1, and SRY alleles. Kinetic establishment of microchimerism was monitored in a double kidney transplanted recipient for 150 min after declamping and after 7 days. RESULTS: Microchimerism was detected in 56.2% of kidney recipients 2 months after transplantation (M2): this fell to 30.1% at 12 months. In renal calcineurin inhibitor-based immunosuppression cohort (n=73), the microchimerism-negative group (n=32) showed 37.9% biopsy-proven acute rejection (BPAR), whereas in the microchimerism-positive group (n=41), no recipient did (P<0.001). Regardless of immunosuppression, BPAR incidence was 35.6% and 4.9%, respectively (P<0.001). Multivariate study showed microchimerism as a protective factor against BPAR (odds ratio: 8.3; 95% confidence interval: 1.8 to 37.9; P = 0.006), blinding other well-known rejection-risk variables. Microchimerism M2 presence did not correlate with a multifactorial critical outcome such as late graft loss. CONCLUSION: Microchimerism was frequent after kidney transplantation and correlated with a significantly lower incidence of rejection. We propose that early microchimerism monitoring could help early detection of low rejection-risk recipients.


Subject(s)
Hematopoietic Stem Cells/physiology , Kidney Transplantation/immunology , Transplantation Chimera , Follow-Up Studies , HLA Antigens/genetics , Histocompatibility Testing , Humans , Kinetics , Polymerase Chain Reaction , Time Factors , Treatment Failure , Treatment Outcome
15.
Blood ; 110(1): 461-7, 2007 Jul 01.
Article in English | MEDLINE | ID: mdl-17384200

ABSTRACT

CTLA-4 is an inhibitory molecule that down-regulates T-cell activation. Although polymorphisms at CTLA-4 have been correlated with autoimmune diseases their association with clinical outcome after allogeneic hematopoietic stem cell transplantation (allo-HSCT) has yet to be explored. A total of 5 CTLA-4 single-nucleotide polymorphisms were genotyped on 536 HLA-identical sibling donors of allo-HSC transplants. Genotypes were tested for an association with patients' posttransplantation outcomes. The effect of the polymorphisms on cytotoxic T-lymphocyte antigen 4 (CTLA-4) mRNA and protein production were determined in 60 healthy control participants. We observed a reduction in the mRNA expression of the soluble CTLA-4 isoform in the presence of a G allele at CT60 and +49. Patients receiving stem cells from a donor with at least 1 G allele in position CT60 had worse overall survival (56.2% vs 69.8% at 5 years; P = .001; hazard ratio [HR], 3.80; 95% confidence interval [CI], 1.75-8.22), due to a higher risk of relapse (P = .049; HR, 1.71; 95% CI, 1.00-2.93). Acute graft-versus-host disease (aGVHD) was more frequent in patients receiving CT60 AA stem cells (P = .033; HR, 1.54; 95% CI, 1.03-2.29). This is the first study to report an association between polymorphisms at CTLA-4 and clinical outcome after allo-HSCT. The CT60 genotype influences relapse and aGVHD, probably due to its action on CTLA-4 alternative splicing.


Subject(s)
Antigens, CD/genetics , Antigens, Differentiation/genetics , HLA Antigens/immunology , Hematopoietic Stem Cell Transplantation/methods , Histocompatibility/genetics , Polymorphism, Genetic , Adolescent , Adult , CTLA-4 Antigen , Child , Child, Preschool , Female , Genotype , Graft vs Host Disease/etiology , Hematopoietic Stem Cell Transplantation/adverse effects , Hematopoietic Stem Cell Transplantation/mortality , Humans , Infant , Male , Middle Aged , Siblings , Survival Rate , Transplantation, Homologous , Treatment Outcome
SELECTION OF CITATIONS
SEARCH DETAIL
...