Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
Add more filters










Publication year range
1.
Nat Commun ; 14(1): 8051, 2023 Dec 05.
Article in English | MEDLINE | ID: mdl-38052872

ABSTRACT

Gene editing strategies for cystic fibrosis are challenged by the complex barrier properties of airway epithelia. We previously reported that the amphiphilic S10 shuttle peptide non-covalently combined with CRISPR-associated (Cas) ribonucleoprotein (RNP) enabled editing of human and mouse airway epithelial cells. Here, we derive the S315 peptide as an improvement over S10 in delivering base editor RNP. Following intratracheal aerosol delivery of Cy5-labeled peptide in rhesus macaques, we confirm delivery throughout the respiratory tract. Subsequently, we target CCR5 with co-administration of ABE8e-Cas9 RNP and S315. We achieve editing efficiencies of up-to 5.3% in rhesus airway epithelia. Moreover, we document persistence of edited epithelia for up to 12 months in mice. Finally, delivery of ABE8e-Cas9 targeting the CFTR R553X mutation restores anion channel function in cultured human airway epithelia. These results demonstrate the therapeutic potential of base editor delivery with S315 to functionally correct the CFTR R553X mutation in respiratory epithelia.


Subject(s)
Cystic Fibrosis Transmembrane Conductance Regulator , Epithelial Cells , Animals , Humans , Mice , Macaca mulatta/metabolism , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Epithelial Cells/metabolism , Respiratory Mucosa/metabolism , Ribonucleoproteins/metabolism , Peptides/genetics , CRISPR-Cas Systems
2.
Front Immunol ; 14: 1231916, 2023.
Article in English | MEDLINE | ID: mdl-37675109

ABSTRACT

Introduction: Natural Killer (NK) cells hold the potential to shift cell therapy from a complex autologous option to a universal off-the-shelf one. Although NK cells have demonstrated efficacy and safety in the treatment of leukemia, the limited efficacy of NK cell-based immunotherapies against solid tumors still represents a major hurdle. In the immunosuppressive tumor microenvironment (TME), inhibitory interactions between cancer and immune cells impair antitumoral immunity. KLRC1 gene encodes the NK cell inhibitory receptor NKG2A, which is a potent NK cell immune checkpoint. NKG2A specifically binds HLA-E, a non-classical HLA class I molecule frequently overexpressed in tumors, leading to the transmission of inhibitory signals that strongly impair NK cell function. Methods: To restore NK cell cytotoxicity against HLA-E+ tumors, we have targeted the NKG2A/HLA-E immune checkpoint by using a CRISPR-mediated KLRC1 gene editing. Results: KLRC1 knockout resulted in a reduction of 81% of NKG2A+ cell frequency in ex vivo expanded human NK cells post-cell sorting. In vitro, the overexpression of HLA-E by tumor cells significantly inhibited wild-type (WT) NK cell cytotoxicity with p-values ranging from 0.0071 to 0.0473 depending on tumor cell lines. In contrast, KLRC1 KO NK cells exhibited significantly higher cytotoxicity when compared to WT NK cells against four different HLA-E+ solid tumor cell lines, with p-values ranging from<0.0001 to 0.0154. Interestingly, a proportion of 43.5% to 60.2% of NKG2A- NK cells within the edited NK cell population was sufficient to reverse at its maximum the HLA-E-mediated inhibition of NK cell cytotoxicity. The expression of the activating receptor NKG2C was increased in KLRC1 KO NK cells and contributed to the improved NK cell cytotoxicity against HLA-E+ tumors. In vivo, the adoptive transfer of human KLRC1 KO NK cells significantly delayed tumor progression and increased survival in a xenogeneic mouse model of HLA-E+ metastatic breast cancer, as compared to WT NK cells (p = 0.0015). Conclusions: Our results demonstrate that KLRC1 knockout is an effective strategy to improve NK cell antitumor activity against HLA-E+ tumors and could be applied in the development of NK cell therapy for solid tumors.


Subject(s)
Killer Cells, Natural , Leukemia , Humans , Animals , Mice , Receptors, Natural Killer Cell , Protein Transport , Tumor Microenvironment , HLA-E Antigens
3.
Mol Oncol ; 17(10): 2109-2125, 2023 10.
Article in English | MEDLINE | ID: mdl-37086156

ABSTRACT

The androgen receptor (AR) is an established orchestrator of cell metabolism in prostate cancer (PCa), notably by inducing an oxidative mitochondrial program. Intriguingly, AR regulates cytoplasmic isocitrate dehydrogenase 1 (IDH1), but not its mitochondrial counterparts IDH2 and IDH3. Here, we aimed to understand the functional role of IDH1 in PCa. Mouse models, in vitro human PCa cell lines, and human patient-derived organoids (PDOs) were used to study the expression and activity of IDH enzymes in the normal prostate and PCa. Genetic and pharmacological inhibition of IDH1 was then combined with extracellular flux analyses and gas chromatography-mass spectrometry for metabolomic analyses and cancer cell proliferation in vitro and in vivo. In PCa cells, more than 90% of the total IDH activity is mediated through IDH1 rather than its mitochondrial counterparts. This profile seems to originate from the specialized prostate metabolic program, as observed using mouse prostate and PDOs. Pharmacological and genetic inhibition of IDH1 impaired mitochondrial respiration, suggesting that this cytoplasmic enzyme contributes to the mitochondrial tricarboxylic acid cycle (TCA) in PCa. Mass spectrometry-based metabolomics confirmed this hypothesis, showing that inhibition of IDH1 impairs carbon flux into the TCA cycle. Consequently, inhibition of IDH1 decreased PCa cell proliferation in vitro and in vivo. These results demonstrate that PCa cells have a hybrid cytoplasmic-mitochondrial TCA cycle that depends on IDH1. This metabolic enzyme represents a metabolic vulnerability of PCa cells and a potential new therapeutic target.


Subject(s)
Citric Acid Cycle , Prostatic Neoplasms , Male , Mice , Animals , Humans , Isocitrate Dehydrogenase/genetics , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/metabolism , Mitochondria/metabolism , Cytosol/metabolism
4.
Res Sq ; 2023 Feb 17.
Article in English | MEDLINE | ID: mdl-36824928

ABSTRACT

Gene editing strategies for cystic fibrosis are challenged by the complex barrier properties of airway epithelia. We previously reported that the amphiphilic S10 shuttle peptide non-covalently combined with CRISPR-associated (Cas) ribonucleoprotein (RNP) enabled editing of human and mouse airway epithelial cells. Here, to improve base editor RNP delivery, we optimized S10 to derive the S315 peptide. Following intratracheal aerosol of Cy5-labeled peptide cargo in rhesus macaques, we confirmed delivery throughout the respiratory tract. Subsequently, we targeted CCR5 with co-administration of ABE8e-Cas9 RNP and S315. We achieved editing efficiencies of up to 5.3% in rhesus airway epithelia. Moreover, we documented persistence of edited epithelia for up to 12 months in mice. Finally, delivery of ABE8e-Cas9 targeting the CFTR R553X mutation restored anion channel function in cultured human airway epithelial cells. These results demonstrate the therapeutic potential of base editor delivery with S315 to functionally correct the CFTR R553X mutation in respiratory epithelia.

5.
Mol Metab ; 62: 101516, 2022 08.
Article in English | MEDLINE | ID: mdl-35598879

ABSTRACT

OBJECTIVE: The prostate is metabolically unique: it produces high levels of citrate for secretion via a truncated tricarboxylic acid (TCA) cycle to maintain male fertility. In prostate cancer (PCa), this phenotype is reprogrammed, making it an interesting therapeutic target. However, how the truncated prostate TCA cycle works is still not completely understood. METHODS: We optimized targeted metabolomics in mouse and human organoid models in ex vivo primary culture. We then used stable isotope tracer analyses to identify the pathways that fuel citrate synthesis. RESULTS: First, mouse and human organoids were shown to recapitulate the unique citrate-secretory program of the prostate, thus representing a novel model that reproduces this unusual metabolic profile. Using stable isotope tracer analysis, several key nutrients were shown to allow the completion of the prostate TCA cycle, revealing a much more complex metabolic profile than originally anticipated. Indeed, along with the known pathway of aspartate replenishing oxaloacetate, glutamine was shown to fuel citrate synthesis through both glutaminolysis and reductive carboxylation in a GLS1-dependent manner. In human organoids, aspartate entered the TCA cycle at the malate entry point, upstream of oxaloacetate. Our results demonstrate that the citrate-secretory phenotype of prostate organoids is supported by the known aspartate-oxaloacetate-citrate pathway, but also by at least three additional pathways: glutaminolysis, reductive carboxylation, and aspartate-malate conversion. CONCLUSIONS: Our results add a significant new dimension to the prostate citrate-secretory phenotype, with at least four distinct pathways being involved in citrate synthesis. Better understanding this distinctive citrate metabolic program will have applications in both male fertility as well as in the development of novel targeted anti-metabolic therapies for PCa.


Subject(s)
Citric Acid Cycle , Malates , Animals , Aspartic Acid/metabolism , Citrates/metabolism , Citric Acid/metabolism , Humans , Malates/metabolism , Male , Metabolic Networks and Pathways , Mice , Oxaloacetates/metabolism , Prostate/metabolism
6.
Cancer Prev Res (Phila) ; 9(2): 189-95, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26645279

ABSTRACT

Cigarette smoking is the most important known risk factor for urinary bladder cancer. Selected arylamines in cigarette smoke are recognized human bladder carcinogens and undergo biotransformation through several detoxification pathways, such as the glutathione S-transferases (GST), and uridine-diphospho-glucuronosyltransferases (UGT) pathways. GSTM1 deletion status and UGT1A1*28 rs8175347 genotypes were assessed in 189 non-muscle-invasive bladder cancers (NMIBC) patients with pTa (77.2%) and pT1 (22.8%) tumors and a mean follow-up of 5.6 years, to investigate whether two common functional polymorphisms in GSTM1 and UGT1A1 genes and smoking history are associated with recurrence-free survival of patients with NMIBC. Most patients were current (48.7%) or previous (35.4%) cigarette smokers and 15.9% never smoked. Tumor recurrence occurred in 65.1% of patients, at a median time of 12.9 months. Upon multivariate analysis, previous and current smokers approximately tripled their risk of recurrences [HR = 2.76; 95% confidence interval (CI), 1.03-7.40 and HR = 2.93; 95% CI, 1.08-7.94, respectively]. When adjusted for age, smoking status, stage, grade, gender, and presence of carcinoma in situ, carriers of GSTM1 (+/- and -/-) and UGT1A1*28/*28 alleles were significantly at risk of NMIBC recurrence (HR = 10.05; 95% CI, 1.35-75.1 and HR = 1.91; 95% CI, 1.01-3.62, respectively). Compared with nonsmokers with UGT1A1*1/*1 and *1/*28 genotypes, previous and current smokers homozygous for the UGT1A1*28 allele demonstrated a risk of recurrence of 4.95 (95% CI, 1.02-24.0) and 5.32 (95% CI, 2.07-13.7), respectively. This study establishes a connection between GSTM1, UGT1A1, and tobacco exposure as prognostic markers of NMIBC recurrence in bladder cancer patients. These findings warrant validation in larger cohorts.


Subject(s)
Glucuronosyltransferase/genetics , Glutathione Transferase/genetics , Neoplasm Recurrence, Local/pathology , Polymorphism, Genetic/genetics , Smoking/genetics , Urinary Bladder Neoplasms/pathology , Aged , Biomarkers, Tumor/genetics , Female , Follow-Up Studies , Gene-Environment Interaction , Genetic Predisposition to Disease , Genotype , Humans , Male , Middle Aged , Neoplasm Grading , Neoplasm Invasiveness , Neoplasm Recurrence, Local/etiology , Neoplasm Recurrence, Local/metabolism , Neoplasm Staging , Prognosis , Risk Factors , Survival Rate , Urinary Bladder Neoplasms/etiology , Urinary Bladder Neoplasms/metabolism
7.
J Pharmacol Exp Ther ; 345(1): 95-101, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23386248

ABSTRACT

Despite the importance of UDP-glucuronosyltransferase (UGT) 1A1*28 in irinotecan pharmacogenetics, our capability to predict drug-induced severe toxicity remains limited. We aimed at identifying novel genetic markers that would improve prediction of irinotecan toxicity and response in advanced colorectal cancer patients treated with folic acid (leucovorin), fluorouracil (5-FU), and irinotecan (camptosar)-based regimens. The relationships between UGT1A candidate markers across the gene (n = 21) and toxicity were prospectively evaluated in 167 patients. We included variants in the 3'untranscribed region (3'UTR) of the UGT1A locus, not studied in this context yet. These genetic markers were further investigated in 250 Italian FOLFIRI-treated patients. Several functional UGT1A variants, including UGT1A1*28, significantly influenced risk of severe hematologic toxicity. As previously reported in the Italian cohort, a 5-marker risk haplotype [haplotype II (HII); UGTs 1A9/1A7/1A1] was associated with severe neutropenia in our cohort [odds ratio (OR) = 2.43; P = 0.004]. The inclusion of a 3'UTR single-nucleotide polymorphism (SNP) permitted refinement of the previously defined HI, in which HIa was associated with the absence of severe neutropenia in combined cohorts (OR = 0.55; P = 0.038). Among all tested UGT1A variations and upon multivariate analyses, no UGT1A1 SNPs remained significant, whereas three SNPs located in the central region of UGT1A were linked to neutropenia grade 3-4. Haplotype analyses of these markers with the 3'UTR SNP allowed the identification of a protective HI (OR = 0.50; P = 0.048) and two risk haplotypes, HII and HIII, characterized by 2 and 3 unfavorable alleles, respectively, revealing a dosage effect (ORs of 2.15 and 5.28; P ≤ 0.030). Our results suggest that specific SNPs in UGT1A, other than UGT1A1*28, may influence irinotecan toxicity and should be considered to refine pharmacogenetic testing.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/toxicity , Colorectal Neoplasms/genetics , Glucuronosyltransferase/genetics , Haplotypes , Neutropenia/chemically induced , Polymorphism, Single Nucleotide , 3' Untranslated Regions , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Camptothecin/administration & dosage , Camptothecin/analogs & derivatives , Camptothecin/therapeutic use , Camptothecin/toxicity , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/pathology , Female , Fluorouracil/administration & dosage , Fluorouracil/therapeutic use , Fluorouracil/toxicity , Genetic Markers , Humans , Irinotecan , Leucovorin/administration & dosage , Leucovorin/therapeutic use , Leucovorin/toxicity , Male , Middle Aged , Neoplasm Metastasis , Neutropenia/genetics , Prospective Studies , Severity of Illness Index
8.
Biochem J ; 450(1): 107-14, 2013 Feb 15.
Article in English | MEDLINE | ID: mdl-23148825

ABSTRACT

The oligomerization of UGTs [UDP (uridine diphosphate)-glucuronosyltransferases] modulates their enzyme activities. Recent findings also indicate that glucuronidation is negatively regulated by the formation of inactive oligomeric complexes between UGT1A enzymes [i1 (isoform 1)] and an enzymatically inactive alternatively spliced i2 (isoform 2). In the present paper, we assessed whether deletion of the UGT-interacting domains previously reported to be critical for enzyme function might be involved in i1-i2 interactions. The bilirubin-conjugating UGT1A1 was used as a prototype. We also explored whether intermolecular disulfide bonds are involved in i1-i2 interactions and the potential role of selected cysteine residues. Co-immunoprecipitation assays showed that UGT1A1 lacking the SP (signal peptide) alone or also lacking the transmembrane domain (absent from i2) did not self-interact, but still interacted with i2. The deletion of other N- or C-terminal domains did not compromise i1-i2 complex formation. Under non-reducing conditions, we also observed formation of HMWCs (high-molecular-mass complexes) for cells overexpressing i1 and i2. The presence of UGTs in these complexes was confirmed by MS. Mutation of individual cysteine residues throughout UGT1A1 did not compromise i1-i1 or i1-i2 complex formation. These findings are compatible with the hypothesis that the interaction between i1 and i2 proteins (either transient or stable) involves binding of more than one domain that probably differs from those involved in i1-i1 interactions.


Subject(s)
Alternative Splicing , Glucuronosyltransferase/chemistry , Glucuronosyltransferase/genetics , Bilirubin/metabolism , Glucuronosyltransferase/metabolism , HEK293 Cells , Humans , Immunoprecipitation , Protein Interaction Domains and Motifs , Protein Isoforms/chemistry , Protein Isoforms/genetics , Protein Isoforms/metabolism
9.
Pharmacogenet Genomics ; 21(10): 631-41, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21881541

ABSTRACT

OBJECTIVE: UGT2B7 is a key member of the UDP-glucuronosyltransferase (UGT) family that participates in glucuronidation of endogenous compounds and pharmaceuticals. Much evidence suggests a large interindividual variability of UGT2B7-mediated glucuronidation, which is still unexplained by polymorphisms. We hypothesized that alternative splicing may be responsible for the variability in the UGT2B7 function. METHODS: We carried out a comprehensive scan for additional exons at this locus and discovered multiple alternative splicing events. We then determined transcript expression profiles across a large variety of human tissues and assessed some of these variants for their glucuronidation activity in human cells. RESULTS: In-depth analysis of the UGT2B7 gene structure led to the discovery of six novel exons. Kidney and liver samples presented the greatest enrichment of tissue-specific splicing, with 21 new UGT2B7 transcripts isolated. Furthermore, transcription from the proximal promoter (exon 1), associated with the active UGT2B7 mRNA isoform 1 (UGT2B7_v1), is most commonly observed in the gastrointestinal tract, whereas a distal promoter (exon 1a) induces the exclusion of the canonical exon 1 and is active in hormone-related tissues. We also showed that novel transcripts with alternative 3' ends could be translated into proteins lacking glucuronosyltransferase activity in human cells but acting as negative regulators on transferase activity when coexpressed with the active UGT2B7 protein. CONCLUSION: Our findings point toward a significant variability in structure, abundance, and tissue-specific UGT2B7 transcriptome, in addition to novel functions for UGT2B7-derived proteins, all of which may ensure the production of tissue-specific proteomes and functions.


Subject(s)
Alternative Splicing/genetics , Glucuronosyltransferase/genetics , RNA, Messenger/genetics , Exons/genetics , Gene Expression Profiling , Humans , Isoenzymes/genetics , Organ Specificity/genetics , Promoter Regions, Genetic , RNA Precursors/genetics , Tissue Distribution/genetics , Transcription, Genetic
10.
Drug Metab Dispos ; 39(7): 1127-30, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21487055

ABSTRACT

Tacrolimus (Tacro) is a potent immunosuppressant and a central agent in the prevention of posttransplantation rejection. Tacro is characterized by a narrow therapeutic index and wide interindividual pharmacokinetic fluctuation. The contribution of human UDP-glucuronosyltransferase (UGT) in its metabolism has not been extensively studied. In vitro metabolism studies support that the liver produced Tacro-glucuronide (Tacro-G) while its formation was minimal or undetectable in the presence of intestine and kidney microsomes. Among 16 human UGTs tested, UGT1A4 was the sole enzyme involved in Tacro-G formation. This conclusion is supported by the finding of inhibition with a specific substrate of UGT1A4 lamotrigine with K(i) values similar for both human liver and UGT1A4 microsomes and the correlation with trifluoperazine-glucuronide formation by liver microsomes (r(s) = 0.551; p = 0.02). Formation of Tacro-G by liver samples varied among individuals (6.4-fold variation; n = 16), and common nonsynonymous variants may contribute to this variability. In the human embryonic kidney 293 cellular model, no significant differences in enzyme kinetics could be revealed for UGT1A4*2 (P(24)T) and *3 (L(48)V), whereas the allozyme *4 (R(11)W) displayed a 2-fold higher velocity (p < 0.01) compared with the UGT1A4*1 enzyme preparation. In human liver samples, carriers of the UGT1A4 variants did not display statistically different efficiency in Tacro-G formation compared with homozygote for the reference genotype UGT1A4*1/*1. We conclude that UGT1A4 is the major isoform involved in Tacro glucuronidation, whereas additional studies are required to assess the contribution of UGT1A4 genetic factors in tacrolimus glucuronidation variability.


Subject(s)
Glucuronides/metabolism , Glucuronosyltransferase/metabolism , Immunosuppressive Agents/metabolism , Tacrolimus/metabolism , Humans , In Vitro Techniques , Mass Spectrometry
11.
J Pathol ; 223(3): 425-35, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21171088

ABSTRACT

Glucuronidation by UDP-glucuronyltransferase (UGT) enzymes is the prevailing conjugative pathway for the metabolism of both xenobiotics and endogenous compounds. Alterations in this pathway, such as those generated by common genetic polymorphisms, have been shown to significantly impact on the health of individuals, influencing cancer susceptibility, responsiveness to drugs and drug-induced toxicity. Alternative usage of terminal exons leads to UGT1A-derived splice variants, namely the classical and enzymatically active isoforms 1 (i1) and the novel enzymatically inactive isoforms 2 (i2). In vitro functional data from heterologous expression and RNA interference experiments indicate that these i2 isoforms act as negative modulators of glucuronidation, likely by forming inactive complexes with active isoform 1. We used specific antibodies against either active i1 or inactive i2 proteins to examine their distribution in major drug-metabolizing tissues. Data revealed that UGT1A_i1 and inactive UGT1A_i2 are co-produced in the same tissue structures, including liver, kidney, stomach, intestine and colon. Examination of the cellular distribution and semi-quantitative level of expression of UGT1As revealed heterogeneous expression of i1 and i2 proteins, with increased expression of i2 in liver tumours and decreased levels of i1 and i2 in colon cancer specimens, compared to normal tissues. These differences in expression may be relevant to human colon and liver cancer tumorigenesis. Our data clearly demonstrate the similar immunolocalization of active and inactive UGT1A isoforms in most UGT1A-expressing cell types of major tissues involved in drug metabolism. These expression patterns are consistent with a dominant-negative function for the i2 encoded by the UGT1A gene.


Subject(s)
Colonic Neoplasms/enzymology , Glucuronosyltransferase/metabolism , Liver Neoplasms/enzymology , Alternative Splicing , Humans , Intestine, Small/enzymology , Isoenzymes/metabolism , Kidney/enzymology , Stomach/enzymology
12.
Drug Metab Dispos ; 38(10): 1785-9, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20610558

ABSTRACT

UDP-glucuronosyltransferases (UGTs) are major mediators in conjugative metabolism. Current data suggest that UGTs, which are anchored in the endoplasmic reticulum membrane, can oligomerize with each other and/or with other metabolic enzymes, a process that may influence their enzymatic activities. We demonstrated previously that the UGT1A locus encodes previously unknown isoforms (denoted "i2"), by alternative usage of the terminal exon 5. Although i2 proteins lack transferase activity, we showed that knockdown of endogenous i2 levels enhanced cellular UGT1A-i1 activity. In this study, we explored the potential of multiple active UGT1A_i1 proteins (UGT1A1, UGT1A3, UGT1A4, UGT1A6, UGT1A7, UGT1A8, UGT1A9, and UGT1A10) to interact with all spliced i2s by coimmunoprecipitation. We further studied the functional consequences of coexpressing various combinations of spliced i1s and i2s from highly similar UGTs, namely UGT1A7, UGT1A8, and UGT1A9, based on expression profiles observed in human tissues. The i1 isoform of each UGT1A coimmunoprecipitated its respective i2 homolog as well as all other i2s, indicating that they can form heteromeric complexes. Functional data further support the fact that i2 splice species alter glucuronidation activity of i1s independently of the identity of the i2, although the degree of inhibition varied, suggesting that this phenomenon may occur in tissues expressing such combinations of splice forms. These results provide biochemical evidence to support the inhibitory effect of i2s on multiple active UGT1As, probably through formation of inactive heteromeric assemblies of i1s and inactive i2s. The relative abundance of active/inactive oligomeric complexes may thus determine transferase activity.


Subject(s)
Alternative Splicing , Glucuronosyltransferase/antagonists & inhibitors , Glucuronosyltransferase/genetics , Multienzyme Complexes/metabolism , Blotting, Western , Cell Line , Colon/enzymology , Esophagus/enzymology , Humans , Immunoprecipitation , Intestine, Small/enzymology , Liver/enzymology , Protein Isoforms , Protein Multimerization , Transfection
13.
BMC Mol Biol ; 11: 9, 2010 Jan 22.
Article in English | MEDLINE | ID: mdl-20096102

ABSTRACT

BACKGROUND: UDP-glucuronosyltransferase 1A1 (UGT1A1) is a pivotal enzyme involved in metabolism of SN-38, the active metabolite of irinotecan commonly used to treat metastatic colorectal cancer. We previously demonstrated aberrant methylation of specific CpG dinucleotides in UGT1A1-negative cells, and revealed that methylation state of the UGT1A1 5'-flanking sequence is negatively correlated with gene transcription. Interestingly, one of these CpG dinucleotides (CpG -4) is found close to a HNF1 response element (HRE), known to be involved in activation of UGT1A1 gene expression, and within an upstream stimulating factor (USF) binding site. RESULTS: Gel retardation assays revealed that methylation of CpG-4 directly affect the interaction of USF1/2 with its cognate sequence without altering the binding for HNF1-alpha. Luciferase assays sustained a role for USF1/2 and HNF1-alpha in UGT1A1 regulation in colon cancer cells. Based on the differential expression profiles of HNF1A gene in colon cell lines, we also assessed whether methylation affects its expression. In agreement with the presence of CpG islands in the HNF1A promoter, treatments of UGT1A1-negative HCT116 colon cancer cells with a DNA methyltransferase inhibitor restore HNF1A gene expression, as observed for UGT1A1. CONCLUSIONS: This study reveals that basal UGT1A1 expression in colon cells is positively regulated by HNF1-alpha and USF, and negatively regulated by DNA methylation. Besides, DNA methylation of HNF1A could also play an important role in regulating additional cellular drug metabolism and transporter pathways. This process may contribute to determine local inactivation of drugs such as the anticancer agent SN-38 by glucuronidation and define tumoral response.


Subject(s)
Colonic Neoplasms/genetics , Gene Expression Regulation, Neoplastic , Glucuronosyltransferase/genetics , Hepatocyte Nuclear Factor 1/metabolism , Azacitidine/analogs & derivatives , Azacitidine/pharmacology , Base Sequence , Colonic Neoplasms/metabolism , CpG Islands , Cytidine Triphosphate/analogs & derivatives , Cytidine Triphosphate/pharmacology , DNA Methylation , Electrophoretic Mobility Shift Assay , Glucuronosyltransferase/metabolism , HCT116 Cells , HT29 Cells , Hepatocyte Nuclear Factor 1/genetics , Humans , Molecular Sequence Data , Promoter Regions, Genetic , Up-Regulation
14.
Drug Metab Rev ; 42(1): 24-44, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19857043

ABSTRACT

The human uridine diphospho (UDP)-glucuronosyltransferase (UGT) superfamily comprises enzymes responsible for a major biotransformation phase II pathway: the glucuronidation process. The UGT enzymes are located in the endoplasmic reticulum of almost all tissues, where they catalyze the inactivation of several endogenous and exogenous molecules, including bilirubin, sex steroids, numerous prescribed drugs, and environmental toxins. This metabolic pathway is particularly variable. The influence of inheritable polymorphisms in human UGT-encoding genes has been extensively documented and was shown to be responsible for a fraction of the observed phenotypic variability. Other key genomic processes are likely underlying this diversity; these include copy-number variations, epigenetic factors, and newly discovered splicing mechanisms. This review will discuss novel molecular aspects that may be determinant to UGT phenotypes.


Subject(s)
Alternative Splicing/genetics , Gene Duplication , Glucuronosyltransferase/genetics , Glucuronosyltransferase/metabolism , Polymorphism, Genetic , Polymorphism, Single Nucleotide/genetics , Animals , Cell Line , Gene Expression Regulation, Enzymologic/genetics , Genetic Variation , Genome, Human , Genotype , Humans , Kinetics , Pharmacogenetics/methods , Phenotype , RNA, Messenger/analysis
15.
J Biol Chem ; 285(6): 3600-3607, 2010 Feb 05.
Article in English | MEDLINE | ID: mdl-19996319

ABSTRACT

This study investigated the molecular mechanisms underlying the regulatory effect of the newly discovered 45-kDa enzymatically inactive UGT1A spliced polypeptides, named isoform i2, upon UGT1A-mediated glucuronidation. Initially, using an inducible system that mimics the relative abundance of isoforms 1 and 2 of UGT1A1 in human tissues, the rates of formation of glucuronides were significantly reduced. We then used a heterologous system constitutively expressing both isoforms i1 and i2 for an in-depth investigation of the presence of spliced i2 on glucuronidation kinetics. UGT1A1, UGT1A7, and UGT1A8 were selected as candidates for these studies. In all cases, co-expression of i1 and i2 in HEK293 cells leads to a significant reduction of the velocity of the glucuronidation reaction without affecting the affinity (K(m) (app)) for all substrates tested and the K(m) for the co-substrate, UDP-glucuronic acid. The data are consistent with a dominant-negative model of inhibition but do not sustain with an UGT1A_i2-mediated inhibition by competitive binding for substrate or the co-substrate. In contrast, the data from the co-immunoprecipitation experiments are indicative of the existence of a mixture homo-oligomeric (i1-i1 or i2-i2) and hetero-oligomeric (i1-i2) complexes in which the i2-i2 and i1-i2 subunits would be inactive. Thus, protein-protein interactions are likely responsible for the inhibition of active UGT1A_i1 by i2 spliced polypeptides. This new regulatory mechanism may alternatively modulate cellular response to endo/xeno stimulus.


Subject(s)
Alternative Splicing , Glucuronides/metabolism , Glucuronosyltransferase/metabolism , Peptides/metabolism , Blotting, Western , Cell Line , Ecdysone/pharmacology , Glucuronosyltransferase/chemistry , Glucuronosyltransferase/genetics , Humans , Immunoprecipitation , Isoenzymes/chemistry , Isoenzymes/genetics , Isoenzymes/metabolism , Kinetics , Peptides/genetics , Protein Binding/drug effects , Protein Multimerization , Signal Transduction , Substrate Specificity , Transfection
16.
Hum Mutat ; 30(9): 1310-9, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19572376

ABSTRACT

UGT2B17 and UGT2B28 are among the most commonly deleted genes in humans and encode members of the uridine diphosphate (UDP)-glucuronosyltransferase 2B (UGT2B) subfamily. They are involved, along with UGT2B15, in the catabolism of sex-steroid hormones. Despite the recent biomedical interest in UGT2B17 and UGT2B28 copy-number variations (CNVs) within human populations, the impact of their gene dosage has been hampered by the lack of precise molecular identification of the common deletion breakpoints within high homology sequence regions on chromosome 4. We have characterized these common deletions and report their coexistence in Caucasians, along with the p.D85Y (rs1902023:G>T) functional polymorphism of UGT2B15. Segmental duplications of 4.9 kb for UGT2B17 and 6.8 kb for UGT2B28 comprise purine-rich recombination sites located 117 kb and 108 kb apart on both ends of the deletions. CNVs of UGT2B17 and UGT2B28 occur in Caucasians at 27% and 13.5%, respectively. While only 43% have two copies of both genes, 57% harbor at least one deletion. Their co-occurrence on 5% of chromosomes creates a 225-kb genomic gap. CNVs of both UGT2B17 and UGT2B28, with the co-occurrence of UGT2B15:p.D85Y, generate seven distinct haplotypes. Restricting the analyses to CNV of the UGT2B17 gene without evaluating UGT2B28 CNV, along with the genotype of UGT2B15, may over- or underestimate the impact of each gene under physiological conditions or disease states.


Subject(s)
Gene Dosage , Glucuronosyltransferase/genetics , Polymorphism, Genetic , Base Sequence , Genetic Variation , Genotype , Gonadal Steroid Hormones/metabolism , Haplotypes , Humans , Minor Histocompatibility Antigens , Molecular Sequence Data , Sequence Deletion , White People
17.
Drug Metab Dispos ; 37(9): 1793-6, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19487252

ABSTRACT

The non-nucleoside reverse transcriptase inhibitor efavirenz (EFV) is directly conjugated by the UDP-glucuronosyltransferase (UGT) pathway to form EFV-N-glucuronide (EFV-G), but the enzyme(s) involved has not yet been identified. The glucuronidation of EFV was screened with UGT1A and UGT2B enzymes expressed in a heterologous system, and UGT2B7 was shown to be the only reactive enzyme. The apparent K(m) value of UGT2B7 (21 microM) is similar to the value observed for human liver microsomes (24 microM), whereas the variant allozyme UGT2B7*2 (Tyr(268)) displayed similar kinetic parameters. Because 3'-azido-3'-deoxythymidine (AZT), one of the most current nucleotide reverse transcriptase inhibitors prescribed in combination with EFV, is also conjugated by UGT2B7, the potential metabolic interaction between EFV and AZT has been studied using human liver microsomes. Glucuronidation of both drugs was inhibited by one another, in a concentration-dependent manner. At K(m) values (25 and 1000 microM for EFV and AZT, respectively), EFV inhibited AZT glucuronidation by 47%, whereas AZT inhibited EFV glucuronidation by 23%. With a K(i) value of 17 microM for AZT-glucuronide formation, EFV appears to be one of the most selective and potent competitive inhibitor of AZT glucuronidation in vitro. Moreover, assuming that concentrations of EFV achieved in plasma (C(max) = 12.9 microM) are in a range similar to its K(i) value, it was estimated that EFV could produce a theoretical 43% inhibition of AZT glucuronidation in vivo. We conclude that UGT2B7 has a major role in EFV glucuronidation and that EFV could potentially interfere with the hepatic glucuronidation of AZT.


Subject(s)
Anti-HIV Agents/metabolism , Anti-HIV Agents/pharmacology , Benzoxazines/metabolism , Benzoxazines/pharmacology , Glucuronides/metabolism , Glucuronosyltransferase/metabolism , Reverse Transcriptase Inhibitors/metabolism , Reverse Transcriptase Inhibitors/pharmacology , Zidovudine/pharmacology , Alkynes , Chromatography, High Pressure Liquid , Cyclopropanes , Drug Interactions , Humans , In Vitro Techniques , Indicators and Reagents , Kinetics , Microsomes, Liver/drug effects , Microsomes, Liver/metabolism , Tandem Mass Spectrometry
18.
Hum Mutat ; 30(4): 677-87, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19204906

ABSTRACT

The UDP-glucuronosyltransferase UGT1 locus is composed of nine exon 1s, each flanked by a unique promoter region, and common exons (2, 3, 4, and the alternatively spliced exons 5a and 5b). Here, we characterized the genetic architecture of the UGT1 gene in a Caucasian sample. Overall, 98 variations in regulatory domains, exons and exon-intron boundaries were genotyped in 254 unrelated subjects, including 12 unreported UGT1 polymorphisms. We determined allele frequencies, computed pairwise linkage disequilibrium (LD), and inferred haplotypes; this thorough analysis yielding a limited number of common UGT1 haplotypes. Moreover, only 17 haplotype tagging single nucleotide polymorphisms (htSNPs) are required to capture most of the allelic diversity of the locus. Four haplotype blocks were inferred: Block 9/6 (UGT1A9, UGT1A7 and UGT1A6), Block 4 (UGT1A4), Block 3/1 (UGT1A3 and UGT1A1), and Block C (3'UTR). A high level of linkage exists between Blocks 9/6 and 3/1, while the 3'UTR SNPs are genetically isolated. The most common haplotype (16.5%) presents multiple deleterious alleles, mainly 1A1*28, 1A3*2, 1A6*2, and 1A7*4. More interestingly, we reveal the co-occurrences of multiple deleterious variations, some of which could be associated with interindividual differences in glucuronidation. Comparison with the HapMap data set demonstrated differences in haplotypic diversity between ethnic samples, but similarity between Caucasian cohorts, as observed previously. This report provides relevant data for further pharmacogenomic studies.


Subject(s)
Glucuronosyltransferase/genetics , Haplotypes , Polymorphism, Single Nucleotide , Alleles , Canada , France/ethnology , Gene Frequency , Genetic Variation , Genotype , Humans , Linkage Disequilibrium , White People/genetics
19.
Trends Genet ; 23(11): 547-56, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17963976

ABSTRACT

Mood disorders, including major depressive disorder and bipolar disorder, are influenced by both genetic and environmental factors. Hypotheses about the neurobiology of mood disorders have been postulated and putatively associated genes identified. Recently, the immune-related gene encoding purinergic receptor P2X, ligand-gated ion channel, 7 (P2RX7) has been genetically associated with major depressive disorder and bipolar disorder. New candidate genes and emerging gene networks and pathways involved in the aetiology of mood disorders point to a major role for neuronal survival and the adaptive immune systems.


Subject(s)
Depressive Disorder, Major/genetics , Gene Regulatory Networks , Bipolar Disorder/genetics , Databases, Genetic , Humans , Models, Genetic , PubMed , Receptors, Purinergic P2/genetics , Receptors, Purinergic P2X7
20.
Psychiatr Genet ; 17(1): 17-22, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17167340

ABSTRACT

OBJECTIVE: Tryptophan hydroxylase is the rate-limiting enzyme in the serotonin biosynthetic pathway and plays an important role in the regulation of serotonin levels. Recently, a brain-specific isoform, tryptophan hydroxylase 2 or n-tryptophan hydroxylase, has been discovered. Some studies reported genetic and functional associations between this isoform and bipolar disorder and/or major depressive disorder. The aim of this study was to investigate further association of genetic variants in French Canadian samples with bipolar disorders. METHODS: Genetic variants in the tryptophan hydroxylase 2 gene were genotyped in a case-control sample consisting of 225 affected individuals (191 bipolar I and 34 bipolar II) and 221 controls and in a collection of extended pedigrees and trios from the same population 357 nuclear families (201 bipolar I, 64 bipolar II, 79 recurrent major depressive disorder). RESULTS: We determined linkage disequilibrium structure in our isolated population and analyzed six tagged single nucleotide polymorphisms in the case-control sample. Whereas no single, single nucleotide polymorphism gave any significant result, a three single nucleotide polymorphism haplotype gave a global P=0.01. Family-based association showed significant association (P=0.004) of one polymorphism (rs4290270) with the major allele overtransmitted to affected offspring. CONCLUSIONS: Case-control and family-based association studies further support the presence of a susceptibility locus for bipolar disorder in tryptophan hydroxylase 2 by showing statistically significant associations with both, single nucleotide polymorphism alone and haplotype of single nucleotide polymorphism markers.


Subject(s)
Bipolar Disorder/genetics , Neurons/enzymology , Polymorphism, Single Nucleotide , Tryptophan Hydroxylase/genetics , Bipolar Disorder/enzymology , Canada , Case-Control Studies , Female , France/ethnology , Genetic Predisposition to Disease , Genotype , Humans , Male , Pedigree
SELECTION OF CITATIONS
SEARCH DETAIL
...