Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
1.
J Immunother Cancer ; 12(4)2024 Apr 04.
Article in English | MEDLINE | ID: mdl-38580330

ABSTRACT

BACKGROUND: Initiation of antitumor immunity is reliant on the stimulation of dendritic cells (DCs) to present tumor antigens to naïve T cells and generate effector T cells that can kill cancer cells. Induction of immunogenic cell death after certain types of cytotoxic anticancer therapies can stimulate T cell-mediated immunity. However, cytotoxic therapies simultaneously activate multiple types of cellular stress and programmed cell death; hence, it remains unknown what types of cancer cell death confer superior antitumor immunity. METHODS: Murine cancer cells were engineered to activate apoptotic or pyroptotic cell death after Dox-induced expression of procell death proteins. Cell-free supernatants were collected to measure secreted danger signals, cytokines, and chemokines. Tumors were formed by transplanting engineered tumor cells to specifically activate apoptosis or pyroptosis in established tumors and the magnitude of immune response measured by flow cytometry. Tumor growth was measured using calipers to estimate end point tumor volumes for Kaplan-Meier survival analysis. RESULTS: We demonstrated that, unlike apoptosis, pyroptosis induces an immunostimulatory secretome signature. In established tumors pyroptosis preferentially activated CD103+ and XCR1+ type I conventional DCs (cDC1) along with a higher magnitude and functionality of tumor-specific CD8+ T cells and reduced number of regulatory T cells within the tumor. Depletion of cDC1 or CD4+ and CD8+ T cells ablated the antitumor response leaving mice susceptible to a tumor rechallenge. CONCLUSION: Our study highlights that distinct types of cell death yield varying immunotherapeutic effect and selective activation of pyroptosis can be used to potentiate multiple aspects of the anticancer immunity cycle.


Subject(s)
CD8-Positive T-Lymphocytes , Neoplasms , Mice , Animals , Pyroptosis , Dendritic Cells , Cytokines/metabolism
2.
Vet Immunol Immunopathol ; 209: 45-52, 2019 Mar.
Article in English | MEDLINE | ID: mdl-30885305

ABSTRACT

Several molecular clonality assays have been developed to assess canine B cell proliferations. These assays were based on different sequence data, utilized different assay designs and employed different testing strategies. This has resulted in a complex body of literature and complicates evidence-based selection of primer sets. In addition, further refinement of primer sets is difficult because it is unknown how well current primer sets cover the expressed sequence repertoire. The objectives of this study were 1) to provide an overview of published IGH clonality assays that highlights key differences in assay design and testing strategy and 2) to propose a novel method for optimizing primer sets that leverages large-scale sequencing data. A review of previously published assays highlighted confounding factors that hamper a direct comparison of performance metrics between studies. These findings illustrate the need for a multi-institutional effort to harmonize veterinary clonality testing. A novel in silico analysis of primer sequences using a large dataset of expressed sequences identified shortfalls of existing primer sets and was used to guide primer optimization. Three optimized primer sets were tested and yielded qualitative sensitivity values between 80-90%. The qualitative sensitivity ranged from 1% to over 50% and was dependent on the size of the neoplastic clone and the sample DNA used. These findings illustrate that inclusion of high-throughput sequencing data for primer design can be a useful tool to guide primer design and optimization. This strategy could be applied to other antigen receptor loci or species to further improve veterinary clonality assays.


Subject(s)
B-Lymphocytes/cytology , Clone Cells , DNA Primers , Dogs/immunology , Immunoglobulin Heavy Chains/genetics , Animals , Dogs/genetics
3.
Vet Immunol Immunopathol ; 202: 181-190, 2018 Aug.
Article in English | MEDLINE | ID: mdl-30078594

ABSTRACT

The ability to mount adaptive immune responses to a diverse array of pathogens is essential to maintaining the health of an individual. The outcome of adaptive immune responses is influenced by the pool of available lymphocyte antigen receptors. Understanding the composition and dynamics of immune repertoires is hence of relevance to characterizing physiologic immunological processes as well as understanding disease pathogenesis. The dog is increasingly recognized as a model for human disease. The objective of this study was to utilize NGS for comprehensive and unbiased analysis of the IGH repertoire in healthy dogs. First, the IGH locus was searched in silico for previously unidentified genes. Second, IGH transcripts from major lymphoid organs were amplified using a 5'RACE approach without V/J primer bias. Third, amplicons were sequenced on an Illumina MiSeq platform, and data were analyzed using the ARResT/Interrogate platform. Data analysis included V/J usage, V-J pairing biases, isotype frequency, CDR3 diversity, convergent recombination, and public repertoires. The results of this study provide a comprehensive IGH repertoire analysis for healthy dogs. These data will allow further improvement of V/J gene-specific primer sets and will serve as baseline for future studies investigating immune repertoires in health and disease.


Subject(s)
Immunoglobulin Heavy Chains/genetics , Immunoglobulin Variable Region/genetics , Animals , Dogs , High-Throughput Nucleotide Sequencing , Immunoglobulin Isotypes/genetics , Receptors, Antigen/genetics , Sequence Analysis, DNA
4.
Biochem Biophys Res Commun ; 490(1): 29-35, 2017 08 12.
Article in English | MEDLINE | ID: mdl-28576498

ABSTRACT

BACKGROUND: Cancer stem cells (CSCs) share a number of properties with somatic stem cells including heightened protective mechanisms and the ability to self-renew. CSCs are a critical subpopulation of cancer cells implicated in tumor formation, metastases and recurrence. METHODS: We used serial colonosphere culture to enrich for CSCs from two human CRC cell lines. The expression of proposed colorectal CSC markers and multi-drug resistance genes were assessed via flow cytometry and RT-qPCR. Drug resistance gene expression and self-renewal ability were also determined following treatment with the chemotherapeutic 5-fluorouracil. RESULTS: Colonosphere culture successfully enriched for a subpopulation of cells with CSC-related gene expression and heightened self-renewal ability, particularly in the SW480 cell line. Chemotherapy treatment significantly reduced sphere formation however a small fraction of cells survived treatment and retained their self-renewal ability. CONCLUSIONS: Our findings support the use of the sphere formation assay to study CSCs. The ability of cells to self-renew following chemotherapy treatment highlights the importance of targeting both the bulk of tumor cells and the CSC population to prevent recurrence in colorectal cancer.


Subject(s)
Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/pathology , Drug Resistance, Neoplasm/drug effects , Fluorouracil/pharmacology , Neoplastic Stem Cells/drug effects , Cell Line, Tumor , Colorectal Neoplasms/genetics , Drug Resistance, Neoplasm/genetics , Fluorouracil/administration & dosage , Humans , Neoplastic Stem Cells/pathology , Structure-Activity Relationship
5.
Cell Stress Chaperones ; 21(6): 1065-1076, 2016 11.
Article in English | MEDLINE | ID: mdl-27631331

ABSTRACT

Heat shock proteins (HSPs) are molecular chaperones subdivided into several families based on their molecular weight. Due to their cytoprotective roles, these proteins may help protect cancer cells against chemotherapy-induced cell death. Investigation into the biologic activity of HSPs in a variety of cancers including primary bone tumors, such as osteosarcoma (OSA), is of great interest. Both human and canine OSA tumor samples have aberrant production of HSP70. This study assessed the response of canine OSA cells to inhibition of HSP70 and GRP78 by the ATP-mimetic VER-155008 and whether this treatment strategy could sensitize cells to doxorubicin chemotherapy. Single-agent VER-155008 treatment decreased cellular viability and clonogenic survival and increased apoptosis in canine OSA cell lines. However, combination schedules with doxorubicin after pretreatment with VER-155008 did not improve inhibition of cellular viability, apoptosis, or clonogenic survival. Treatment with VER-155008 prior to chemotherapy resulted in an upregulation of target proteins HSP70 and GRP78 in addition to the co-chaperone proteins Herp, C/EBP homologous transcription protein (CHOP), and BAG-1. The increased GRP78 was more cytoplasmic in location compared to untreated cells. Single-agent treatment also revealed a dose-dependent reduction in activated and total Akt. Based on these results, targeting GRP78 and HSP70 may have biologic activity in canine osteosarcoma. Further studies are required to determine if and how this strategy may impact the response of osteosarcoma cells to chemotherapy.


Subject(s)
Antibiotics, Antineoplastic/toxicity , Doxorubicin/toxicity , HSP70 Heat-Shock Proteins/antagonists & inhibitors , Heat-Shock Proteins/antagonists & inhibitors , Animals , Bone Neoplasms/metabolism , Bone Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , DNA-Binding Proteins/metabolism , Dogs , Endoplasmic Reticulum Chaperone BiP , HSP70 Heat-Shock Proteins/metabolism , Heat-Shock Proteins/metabolism , Microscopy, Fluorescence , Osteosarcoma/metabolism , Osteosarcoma/pathology , Proto-Oncogene Proteins c-akt/metabolism , Purine Nucleosides/pharmacology , Signal Transduction/drug effects , Transcription Factor CHOP/metabolism , Transcription Factors/metabolism , Unfolded Protein Response , Up-Regulation/drug effects
6.
Microvasc Res ; 108: 48-63, 2016 11.
Article in English | MEDLINE | ID: mdl-27503671

ABSTRACT

Angiogenesis is essential for mammalian development and tissue homeostasis, and is involved in several pathological processes, including tumor growth and dissemination. Many factors within the tissue microenvironment are known to modulate angiogenesis, including cytokines, such as transforming growth factor beta (TGFß), and oxygen level. TGFß exists in three different isoforms (1, 2 and 3), all of which (albeit in different contexts) might mediate angiogenesis and are able to induce endothelial-mesenchymal transition (EndoMT), a process involved in heart development, pathologic fibrosis and, as recently reported, in angiogenesis. Low oxygen level, referred to as hypoxia, has been independently shown to induce angiogenesis, modulate TGFß signalling and promote EndoMT. However, how these phenomena might be interconnected to drive angiogenesis is rather unexplored. To begin addressing the potential contribution of TGFß-induced EndoMT to angiogenesis, and to explore how microenvironmental hypoxia might influence these processes, we investigated the effect of TGFß isoforms 1 and 2 on early EndoMT response in cultured adult endothelium under standard (21 %) and hypoxic (1 %) culture conditions. Our data indicates that EndoMT-like changes, such as an increase in expression and nuclear translocation of Snail, Slug and Zeb1, and reduction of VE-cadherin expression, occur in response to TGFß1 and/or TGFß2 as early as 6h after stimulation and might be enhanced by hypoxia in an isoform-specific manner. Further, hypoxia enhances canonical TGFß signalling, and appears to be a key determinant of Snail's differential involvement in endothelial cell responses to TGFß1 versus TGFß2.


Subject(s)
Endothelial Cells/drug effects , Epithelial-Mesenchymal Transition/drug effects , Neovascularization, Physiologic/drug effects , Transforming Growth Factor beta1/pharmacology , Transforming Growth Factor beta2/pharmacology , Animals , Antigens, CD/metabolism , Cadherins/metabolism , Cattle , Cell Hypoxia , Cells, Cultured , Cellular Microenvironment , Endothelial Cells/metabolism , Endothelial Cells/pathology , Humans , Kinetics , Protein Serine-Threonine Kinases/metabolism , RNA Interference , Receptor, Transforming Growth Factor-beta Type I , Receptors, Transforming Growth Factor beta/agonists , Receptors, Transforming Growth Factor beta/metabolism , Signal Transduction/drug effects , Smad1 Protein/metabolism , Smad2 Protein/metabolism , Snail Family Transcription Factors/genetics , Snail Family Transcription Factors/metabolism , Transfection , Vascular Endothelial Growth Factor Receptor-2/genetics , Vascular Endothelial Growth Factor Receptor-2/metabolism , Zinc Finger E-box-Binding Homeobox 1/metabolism
7.
BMC Vet Res ; 12: 82, 2016 May 31.
Article in English | MEDLINE | ID: mdl-27245053

ABSTRACT

BACKGROUND: Radiation therapy is a palliative treatment modality for canine osteosarcoma, with transient improvement in analgesia observed in many cases. However there is room for improvement in outcome for these patients. It is possible that the addition of sensitizing agents may increase tumor response to radiation therapy and prolong quality of life. Epidermal growth factor receptor (EGFR) expression has been documented in canine osteosarcoma and higher EGFR levels have been correlated to a worse prognosis. However, effects of EGFR inhibition on radiation responsiveness in canine osteosarcoma have not been previously characterized. This study examined the effects of the small molecule EGFR inhibitor erlotinib on canine osteosarcoma radiation responses, target and downstream protein expression in vitro. Additionally, to assess the potential impact of treatment on tumor angiogenesis, vascular endothelial growth factor (VEGF) levels in conditioned media were measured. RESULTS: Erlotinib as a single agent reduced clonogenic survival in two canine osteosarcoma cell lines and enhanced the impact of radiation in one out of three cell lines investigated. In cell viability assays, erlotinib enhanced radiation effects and demonstrated single agent effects. Erlotinib did not alter total levels of EGFR, nor inhibit downstream protein kinase B (PKB/Akt) activation. On the contrary, erlotinib treatment increased phosphorylated Akt in these osteosarcoma cell lines. VEGF levels in conditioned media increased after erlotinib treatment as a single agent and in combination with radiation in two out of three cell lines investigated. However, VEGF levels decreased with erlotinib treatment in the third cell line. CONCLUSIONS: Erlotinib treatment promoted modest enhancement of radiation effects in canine osteosarcoma cells, and possessed activity as a single agent in some cell lines, indicating a potential role for EGFR inhibition in the treatment of a subset of osteosarcoma patients. The relative radioresistance of osteosarcoma cells does not appear to be related to EGFR signalling exclusively. Angiogenic responses to radiation and kinase inhibitors are similarly likely to be multifactorial and require further investigation.


Subject(s)
Antineoplastic Agents/therapeutic use , Dog Diseases/drug therapy , Dog Diseases/radiotherapy , ErbB Receptors/antagonists & inhibitors , Erlotinib Hydrochloride/therapeutic use , Osteosarcoma/veterinary , Protein Kinase Inhibitors/therapeutic use , Animals , Cell Line, Tumor , Cell Survival , Combined Modality Therapy/veterinary , Dogs , Osteosarcoma/drug therapy , Osteosarcoma/radiotherapy , Radiation Tolerance/drug effects , Vascular Endothelial Growth Factor A/metabolism
8.
Biosci Rep ; 36(1): e00299, 2016 Jan 06.
Article in English | MEDLINE | ID: mdl-26740252

ABSTRACT

Cancer cells heavily rely on the glycolytic pathway regardless of oxygen tension. Hexokinase II (HKII) catalyses the first irreversible step of glycolysis and is often overexpressed in cancer cells. 3-Bromopyruvate (3BP) has been shown to primarily target HKII, and is a promising anti-cancer compound capable of altering critical metabolic pathways in cancer cells. Abnormal vasculature within tumours leads to heterogeneous microenvironments, including glucose availability, which may affect drug sensitivity. The aim of the present study was to elucidate the mechanisms by which 3BP acts on colorectal cancer (CRC) cells with focus on the HKII/Akt signalling axis. High HKII-expressing cell lines were more sensitive to 3BP than low HKII-expressing cells. 3BP-induced rapid Akt phosphorylation at site Thr-308 and cell death via both apoptotic and necrotic mechanisms. Cells grown under lower glucose concentrations showed greater resistance towards 3BP. Cells with HKII knockdown showed no changes in 3BP sensitivity, suggesting the effects of 3BP are independent of HKII expression. These results emphasize the importance of the tumour microenvironment and glucose availability when considering therapeutic approaches involving metabolic modulation.


Subject(s)
Colorectal Neoplasms/metabolism , Gene Expression Regulation, Enzymologic/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Glucose/metabolism , Hexokinase/biosynthesis , Pyruvates/pharmacology , Apoptosis/drug effects , Apoptosis/genetics , Cell Line, Tumor , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/genetics , Glucose/genetics , Hexokinase/genetics , Humans , Phosphorylation/drug effects , Phosphorylation/genetics , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism
9.
J Nutr Biochem ; 26(8): 818-26, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25804133

ABSTRACT

Folate and its synthetic form, folic acid (FA), are essential vitamins for the regeneration of S-adenosyl methionine molecules, thereby maintaining adequate cellular methylation. The deregulation of DNA methylation is a contributing factor to carcinogenesis, as alterations in genetic methylation may contribute to stem cell reprogramming and dedifferentiation processes that lead to a cancer stem cell (CSC) phenotype. Here, we investigate the potential effects of FA exposure on DNA methylation and colonosphere formation in cultured human colorectal cancer (CRC) cell lines. We show for the first time that HCT116, LS174T, and SW480 cells grown without adequate FA demonstrate significantly impaired colonosphere forming ability with limited changes in CD133, CD166, and EpCAM surface expression. These differences were accompanied by concomitant changes to DNA methyltransferase (DNMT) enzyme expression and DNA methylation levels, which varied depending on cell line. Taken together, these results demonstrate an interaction between FA metabolism and CSC phenotype in vitro and help elucidate a connection between supplemental FA intake and CRC development.


Subject(s)
DNA Methylation/drug effects , Folic Acid/pharmacology , Cell Line, Tumor , Cell Proliferation , Colonic Neoplasms/metabolism , DNA (Cytosine-5-)-Methyltransferases/genetics , DNA (Cytosine-5-)-Methyltransferases/metabolism , DNA Methyltransferase 3A , DNA Modification Methylases/metabolism , HCT116 Cells , Humans , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/metabolism , Repressor Proteins/genetics , Repressor Proteins/metabolism , DNA Methyltransferase 3B
10.
Neoplasia ; 14(7): 612-23, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22904678

ABSTRACT

Receptors for the angiogenic factor VEGF are expressed by tumor cancer cells including melanoma, although their functionality remains unclear. Paired human melanoma cell lines WM115 and WM239 were used to investigate differences in expression and functionality of VEGF and VEGFR2 in vitro and in vivo with the anti-VEGF antibody bevacizumab. Both WM115 and WM239 cells expressed VEGF and VEGFR2, the levels of which were modulated by hypoxia. Detection of native and phosphorylated VEGFR2 in subcellular fractions under serum-free conditions showed the presence of a functional autocrine as well as intracrine VEGF/VEGFR2 signaling loops. Interestingly, treatment of WM115 and WM239 cells with increasing doses of bevacizumab (0-300 µg/ml) in vitro did not show any significant inhibition of VEGFR2 phosphorylation. Small-molecule tyrosine kinase inhibitor, sunitinib, caused an inhibition of VEGFR2 phosphorylation in WM239 but not in WM115 cells. An increase in cell proliferation was observed in WM115 cells treated with bevacizumab, whereas sunitinib inhibited proliferation. When xenografted to immune-deficient mice, we found bevacizumab to be an effective antiangiogenic but not antitumorigenic agent for both cell lines. Because bevacizumab is unable to neutralize murine VEGF, this supports a paracrine angiogenic response. We propose that the failure of bevacizumab to generate an antitumorigenic effect may be related to its generation of enhanced autocrine/intracrine signaling in the cancer cells themselves. Collectively, these results suggest that, for cancers with intracrine VEGF/ VEGFR2 signaling loops, small-molecule inhibitors of VEGFR2 may be more effective than neutralizing antibodies at disease control.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Antibodies, Monoclonal, Humanized/pharmacology , Autocrine Communication/drug effects , Melanoma/metabolism , Signal Transduction/drug effects , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor Receptor-2/metabolism , Animals , Bevacizumab , Cell Hypoxia , Cell Line, Tumor , Cell Proliferation/drug effects , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Melanoma/genetics , Melanoma/pathology , Mice , Mice, Nude , Phosphorylation/drug effects , Tumor Burden/drug effects , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor Receptor-2/genetics , Xenograft Model Antitumor Assays
11.
Drug Metab Dispos ; 38(9): 1612-22, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20516252

ABSTRACT

Organic anion-transporting polypeptides (OATPs), members of the SLCO/SLC21 family, mediate the transport of various endo- and xenobiotics. In human liver, OATP1B1, 1B3, and 2B1 are located at the basolateral membrane of hepatocytes and are involved in hepatic drug uptake and biliary elimination. Clinically significant drug-drug interactions (DDIs) mediated by hepatic OATPs have drawn great attention from clinical practitioners and researchers. However, there are considerable challenges to prospectively understanding the extent of OATP-mediated DDIs because of the lack of specific OATP inhibitors or substrates and the limitations of in vitro tools. In the present study, a novel RNA interference knockdown sandwich-cultured human hepatocyte model was developed and validated. Quantitative polymerase chain reaction, microarray and immunoblotting analyses, along with uptake assays, illustrated that the expression and transport activity of hepatic OATPs were reduced by small interfering (siRNA) efficiently and specifically in this model. Although OATP siRNA decreased only 20 to 30% of the total uptake of cerivastatin into human hepatocytes, it caused a 50% reduction in cerivastatin metabolism, which was observed by monitoring the formation of the two major metabolites of cerivastatin. The results suggest that coadministration of a drug that is a hepatic OATP inhibitor could significantly alter the pharmacokinetic profile of cerivastatin in clinical studies. Further studies with this novel model demonstrated that OATP and cytochrome P450 have a synergistic effect on cerivastatin-gemfibrozil interactions. The siRNA knockdown sandwich-cultured human hepatocytes may provide a new powerful model for evaluating DDIs.


Subject(s)
Drug Interactions , Hepatocytes/drug effects , Models, Biological , Organic Anion Transporters/antagonists & inhibitors , RNA Interference , Chromatography, Liquid , Hepatocytes/metabolism , Humans , Organic Anion Transporters/genetics , RNA, Messenger/genetics , Tandem Mass Spectrometry
12.
J Pharmacol Exp Ther ; 330(2): 389-402, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19414624

ABSTRACT

The effects of 1alpha,25-dihydroxyvitamin D(3) [1,25(OH)(2)D(3)] on gene expression and function were studied in Caco-2 cells. Microarray analyses, real-time quantitative polymerase chain reactions, and Western blotting were used to determine the mRNA and protein expression of transporters and enzymes after 1,25(OH)(2)D(3) or vehicle (0.1% ethanol) treatment for 1, 3, 6, and 10 days. The mRNA and protein expressions of the apical sodium-dependent bile acid transporter, oligopeptide transporter 1, multidrug resistance-associated protein (MRP) 3, and sulfotransferase 1E1 remained unchanged with 1,25(OH)(2)D(3) treatment, whereas those for CYP3A4, multidrug resistance protein 1, and MRP2 were significantly increased (P < 0.05). 1,25(OH)(2)D(3) treatment significantly enhanced MRP4 protein expression by increasing protein stability without affecting mRNA expression, as confirmed in cycloheximide experiments. Marked increase in 6beta-hydroxylation of testosterone by CYP3A4 was also observed in the 6-day 1,25(OH)(2)D(3)-treated (100 nM) cell lysate. The transport of [(3)H]digoxin, the P-glycoprotein (P-gp) substrate, after treatment with 100 nM 1,25(OH)(2)D(3) for 3 days revealed a higher apparent permeability (P(app)) value in the basal (B)-to-apical (A) direction over that of vehicle treatment (15.1 +/- 0.53 x 10(-6) versus 11.8 +/- 0.58 x 10(-6) cm/s; P < 0.05), whereas the P(app) in the A-to-B direction was unchanged; the efflux ratio was increased (from 5.8 to 8.0). Reduced cellular retention of 5-(and-6)-carboxy-2',7'-dichlorofluorescein, suggestive of higher MRP2 activity, was observed in the 3-day 100 nM 1,25(OH)(2)D(3)-treated cells over controls. Higher protein expression of CYP3A4, MRP2, P-gp, and MRP4 was also observed after a 6-day treatment with other vitamin D analogs (100 nM 1alpha-hydroxyvitamin D(3),1alpha-hydroxyvitamin D(2) or Hectorol, and 25-hydroxyvitamin D(3)) in Caco-2 cells, suggesting a role of 1,25(OH)(2)D(3) and analogs in the activation of enzymes and transporters via the vitamin D receptor.


Subject(s)
Cytochrome P-450 CYP3A/biosynthesis , Membrane Transport Proteins/biosynthesis , Receptors, Calcitriol/metabolism , Up-Regulation/physiology , Vitamin D/analogs & derivatives , Vitamin D/metabolism , Caco-2 Cells , Cytochrome P-450 CYP3A/genetics , Gene Expression Profiling , Humans , Ligands , Membrane Transport Proteins/genetics , Oligonucleotide Array Sequence Analysis
13.
Nat Genet ; 33(1): 97-101, 2003 Jan.
Article in English | MEDLINE | ID: mdl-12496757

ABSTRACT

Shwachman-Diamond syndrome (SDS; OMIM 260400) is an autosomal recessive disorder with clinical features that include pancreatic exocrine insufficiency, hematological dysfunction and skeletal abnormalities. Here, we report identification of disease-associated mutations in an uncharacterized gene, SBDS, in the interval of 1.9 cM at 7q11 previously shown to be associated with the disease. We report that SBDS has a 1.6-kb transcript and encodes a predicted protein of 250 amino acids. A pseudogene copy (SBDSP) with 97% nucleotide sequence identity resides in a locally duplicated genomic segment of 305 kb. We found recurring mutations resulting from gene conversion in 89% of unrelated individuals with SDS (141 of 158), with 60% (95 of 158) carrying two converted alleles. Converted segments consistently included at least one of two pseudogene-like sequence changes that result in protein truncation. SDBS is a member of a highly conserved protein family of unknown function with putative orthologs in diverse species including archaea and eukaryotes. Archaeal orthologs are located within highly conserved operons that include homologs of RNA-processing genes, suggesting that SDS may be caused by a deficiency in an aspect of RNA metabolism that is essential for development of the exocrine pancreas, hematopoiesis and chrondrogenesis.


Subject(s)
Exocrine Pancreatic Insufficiency/genetics , Hematologic Diseases/genetics , Musculoskeletal Abnormalities/genetics , Proteins/genetics , Alleles , Base Sequence , Chromosomes, Human, Pair 7/genetics , Conserved Sequence , DNA Mutational Analysis , Female , Gene Conversion , Gene Expression Profiling , Humans , Lod Score , Male , Molecular Sequence Data , Mutation , Pseudogenes/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Syndrome
14.
J Pediatr ; 141(2): 259-65, 2002 Aug.
Article in English | MEDLINE | ID: mdl-12183724

ABSTRACT

OBJECTIVE: To evaluate the role of serum enzymes for defining the pancreatic phenotype in Shwachman-Diamond syndrome (SDS), an inherited multisystem condition. STUDY DESIGN: Serum pancreatic trypsinogen and isoamylase were measured in 164 patients known or presumed to have SDS. The diagnosis was confirmed in 90 patients. Among 74 unconfirmed cases, 35 ("probable SDS") had hematologic dysfunction but lacked documented pancreatic dysfunction, whereas 39 patients ("improbable SDS") lacked both documented pancreatic and hematologic dysfunction. Classification and regression tree (CART) analysis was performed in 90 patients with SDS and 134 control patients to establish a rule for defining the pancreatic phenotype of SDS; the rule was then applied to the patients with unconfirmed diagnosis. RESULTS: In the control patients, serum trypsinogen showed little variation with age, whereas serum isoamylase values rose from birth on, attaining adult values by 3 years. For patients with SDS, serum trypsinogen values were low in young patients and tended to increase with age, whereas serum isoamylase values remained low at all ages. The CART rule combined results from both enzymes and classified the pancreatic phenotype in all but one SDS patient, who was <3 years of age. Excluding patients <3 years of age, CART identified the pancreatic phenotype in 82% and 7% of the "probable SDS" and "improbable SDS" cases, respectively. CONCLUSIONS: Serum pancreatic enzymes are useful for determining the pancreatic phenotype and confirming the diagnosis of SDS.


Subject(s)
Abnormalities, Multiple/enzymology , Abnormalities, Multiple/genetics , Chromosome Aberrations , Chromosomes, Human, Pair 7/enzymology , Chromosomes, Human, Pair 7/genetics , Exocrine Pancreatic Insufficiency/enzymology , Exocrine Pancreatic Insufficiency/genetics , Isoamylase/blood , Isoamylase/genetics , Pancreas/blood supply , Pancreas/enzymology , Trypsinogen/blood , Trypsinogen/genetics , Abnormalities, Multiple/blood , Adolescent , Adult , Biomarkers/blood , Child , Child Welfare , Child, Preschool , Clinical Laboratory Techniques , Exocrine Pancreatic Insufficiency/blood , Female , Hematologic Diseases/blood , Hematologic Diseases/diagnosis , Hematologic Diseases/enzymology , Humans , Infant , Infant Welfare , Intracranial Hemorrhages/blood , Intracranial Hemorrhages/enzymology , Intracranial Hemorrhages/mortality , Male , Phenotype , Retrospective Studies , Syndrome
15.
Eur J Hum Genet ; 10(4): 250-8, 2002 Apr.
Article in English | MEDLINE | ID: mdl-12032733

ABSTRACT

Shwachman-Diamond syndrome (SDS) is an autosomal recessive disorder characterised by exocrine pancreatic dysfunction, haematological and skeletal abnormalities. We have previously defined the SDS locus as a 2.7 cM interval spanning the centromere of chromosome 7. To facilitate additional analysis of this complex and poorly characterised region, a framework of ordered genetic markers at 7p11-q11, including six newly identified, has been constructed using somatic cell, radiation hybrid and STS-content mapping. We have identified shared disease haplotypes, that recur in unrelated families of common ethnic origin, and extend across the SDS locus. Detection of ancestral and intrafamilial recombination events in patients refined the SDS locus to a 1.9 cM interval at 7q11, which contains the tyrosylprotein sulfotransferase 1 (TPST1) gene. Patients with SDS were screened for mutations in TPST1 by sequencing of exons and intron-exon junctions. Two single nucleotide polymorphisms, but no disease-causing mutations, were identified. In addition, Southern blot analysis yielded no evidence of large-scale mutations, and RT-PCR analysis failed to detect alterations in expression. These results exclude TPST1 as the causative gene for SDS. The established map of the refined SDS locus will assist in the identification and characterisation of other candidate genes for SDS.


Subject(s)
Chromosomes, Human, Pair 7/genetics , Exocrine Pancreatic Insufficiency/genetics , Hematologic Diseases/genetics , Musculoskeletal Abnormalities/genetics , Physical Chromosome Mapping , Sulfotransferases/genetics , DNA Mutational Analysis , Female , Haplotypes , Humans , Linkage Disequilibrium , Male , Pedigree , Reverse Transcriptase Polymerase Chain Reaction , Sulfotransferases/metabolism , Syndrome
SELECTION OF CITATIONS
SEARCH DETAIL
...