Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Oncogenesis ; 13(1): 13, 2024 Apr 03.
Article in English | MEDLINE | ID: mdl-38570533

ABSTRACT

Change within the intratumoral microbiome is a common feature in lung and other cancers and may influence inflammation and immunity in the tumor microenvironment, affecting growth and metastases. We previously characterized the lung cancer microbiome in patients and identified Acidovorax temperans as enriched in tumors. Here, we instilled A. temperans in an animal model driven by mutant K-ras and Tp53. This revealed A. temperans accelerates tumor development and burden through infiltration of proinflammatory cells. Neutrophils exposed to A. temperans displayed a mature, pro-tumorigenic phenotype with increased cytokine signaling, with a global shift away from IL-1ß signaling. Neutrophil to monocyte and macrophage signaling upregulated MHC II to activate CD4+ T cells, polarizing them to an IL-17A+ phenotype detectable in CD4+ and γδ populations (T17). These T17 cells shared a common gene expression program predictive of poor survival in human LUAD. These data indicate bacterial exposure promotes tumor growth by modulating inflammation.

2.
Nat Commun ; 12(1): 5551, 2021 09 21.
Article in English | MEDLINE | ID: mdl-34548489

ABSTRACT

While dysregulation of RNA splicing has been recognized as an emerging target for cancer therapy, the functional significance of RNA splicing and individual splicing factors in brain tumors is poorly understood. Here, we identify SON as a master regulator that activates PTBP1-mediated oncogenic splicing while suppressing RBFOX2-mediated non-oncogenic neuronal splicing in glioblastoma multiforme (GBM). SON is overexpressed in GBM patients and SON knockdown causes failure in intron removal from the PTBP1 transcript, resulting in PTBP1 downregulation and inhibition of its downstream oncogenic splicing. Furthermore, SON forms a complex with hnRNP A2B1 and antagonizes RBFOX2, which leads to skipping of RBFOX2-targeted cassette exons, including the PTBP2 neuronal exon. SON knockdown inhibits proliferation and clonogenicity of GBM cells in vitro and significantly suppresses tumor growth in orthotopic xenografts in vivo. Collectively, our study reveals that SON-mediated RNA splicing is a GBM vulnerability, implicating SON as a potential therapeutic target in brain tumors.


Subject(s)
Brain Neoplasms/genetics , DNA-Binding Proteins/genetics , Glioblastoma/genetics , Heterogeneous-Nuclear Ribonucleoproteins/genetics , Minor Histocompatibility Antigens/genetics , Nerve Tissue Proteins/genetics , Polypyrimidine Tract-Binding Protein/genetics , RNA Splicing Factors/genetics , RNA Splicing , Repressor Proteins/genetics , Animals , Brain Neoplasms/metabolism , Brain Neoplasms/mortality , Brain Neoplasms/pathology , Cell Cycle/genetics , Cell Line, Tumor , Cell Proliferation , DNA-Binding Proteins/antagonists & inhibitors , DNA-Binding Proteins/metabolism , Exons , Gene Expression Regulation, Neoplastic , Glioblastoma/metabolism , Glioblastoma/mortality , Glioblastoma/pathology , Heterogeneous-Nuclear Ribonucleoprotein Group A-B/genetics , Heterogeneous-Nuclear Ribonucleoprotein Group A-B/metabolism , Heterogeneous-Nuclear Ribonucleoproteins/metabolism , Heterografts , Humans , Introns , Mice , Minor Histocompatibility Antigens/metabolism , Nerve Tissue Proteins/metabolism , Neuroglia/metabolism , Neuroglia/pathology , Neurons/metabolism , Neurons/pathology , Polypyrimidine Tract-Binding Protein/metabolism , RNA Splicing Factors/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Repressor Proteins/metabolism , Signal Transduction , Spheroids, Cellular/metabolism , Spheroids, Cellular/pathology , Survival Analysis
3.
PLoS One ; 16(5): e0251515, 2021.
Article in English | MEDLINE | ID: mdl-34019552

ABSTRACT

Emerging evidence has shown that active enhancers are abundantly transcribed, generating long non-coding RNAs, called enhancer RNAs (eRNAs). While putative eRNAs are often observed from RNA sequencing, the roles of most eRNAs remain largely unknown. Previously, we identified putative enhancer regions at the MALAT1 locus that form chromatin-chromatin interactions under hypoxia, and one of these enhancers is located about 30 kb downstream of the NEAT1 gene and -20 kb upstream of the MALAT1 gene (MALAT1-20 kb enhancer). Here, we report that a novel eRNA, named eRNA of the NEAT1-MALAT1-Locus (eNEMAL), is transcribed from the MALAT1-20 kb enhancer and conserved in primates. We found that eNEMAL is upregulated in response to hypoxia in multiple breast cancer cell lines, but not in non-tumorigenic MCF10A cells. Overexpression and knockdown of eNEMAL revealed that alteration of eNEMAL level does not affect MALAT1 expression. Instead, we found that eNEMAL upregulates the long isoform of NEAT1 (NEAT1_2) without increasing the total NEAT1 transcript level in MCF7 breast cancer cells, suggesting that eNEMAL has a repressive effect on the 3'-end polyadenylation process required for generating the short isoform of NEAT1 (NEAT1_1). Altogether, we demonstrated that an eRNA transcribed from a MALAT1 enhancer regulates NEAT1 isoform expression, implicating the MALAT1-20 kb enhancer and its transcript eNEMAL in co-regulation of MALAT1 and NEAT1 in response to hypoxia in breast cancer cells.


Subject(s)
Breast Neoplasms , Enhancer Elements, Genetic , Gene Expression Regulation, Neoplastic , Genetic Loci , RNA, Long Noncoding , RNA, Neoplasm , Up-Regulation , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Cell Line, Tumor , Female , Humans , RNA, Long Noncoding/biosynthesis , RNA, Long Noncoding/genetics , RNA, Neoplasm/biosynthesis , RNA, Neoplasm/genetics
4.
Cancer Gene Ther ; 28(9): 1000-1015, 2021 09.
Article in English | MEDLINE | ID: mdl-33247227

ABSTRACT

A high incidence of acute megakaryoblastic leukemia (AMKL) in Down syndrome patients implies that chromosome 21 genes have a pivotal role in AMKL development, but the functional contribution of individual genes remains elusive. Here, we report that SON, a chromosome 21-encoded DNA- and RNA-binding protein, inhibits megakaryocytic differentiation by suppressing RUNX1 and the megakaryocytic gene expression program. As megakaryocytic progenitors differentiate, SON expression is drastically reduced, with mature megakaryocytes having the lowest levels. In contrast, AMKL cells express an aberrantly high level of SON, and knockdown of SON induced the onset of megakaryocytic differentiation in AMKL cell lines. Genome-wide transcriptome analyses revealed that SON knockdown turns on the expression of pro-megakaryocytic genes while reducing erythroid gene expression. Mechanistically, SON represses RUNX1 expression by directly binding to the proximal promoter and two enhancer regions, the known +23 kb enhancer and the novel +139 kb enhancer, at the RUNX1 locus to suppress H3K4 methylation. In addition, SON represses the expression of the AP-1 complex subunits JUN, JUNB, and FOSB which are required for late megakaryocytic gene expression. Our findings define SON as a negative regulator of RUNX1 and megakaryocytic differentiation, implicating SON overexpression in impaired differentiation during AMKL development.


Subject(s)
Core Binding Factor Alpha 2 Subunit/metabolism , DNA-Binding Proteins/metabolism , Leukemia, Megakaryoblastic, Acute/metabolism , Megakaryocytes/metabolism , Minor Histocompatibility Antigens/metabolism , Cell Differentiation , Down Syndrome/genetics , Gene Expression , Genetic Predisposition to Disease , Humans , Leukemia, Megakaryoblastic, Acute/genetics , Leukemia, Megakaryoblastic, Acute/pathology , Transfection
5.
Cancer Cell ; 38(2): 158-160, 2020 08 10.
Article in English | MEDLINE | ID: mdl-32649886

ABSTRACT

A paper published in Science probes over 1500 tumor samples from multiple cancer types for markers of a tumor microbiome and viable bacteria. Unique intertumoral and intracellular signatures associated with clinical biomarkers and metabolic pathways provide clues to the roles bacteria play in carcinogenesis.


Subject(s)
Microbiota , Neoplasms , Bacteria/genetics , Carcinogenesis , Humans
6.
J Biol Chem ; 294(29): 11213-11224, 2019 07 19.
Article in English | MEDLINE | ID: mdl-31167784

ABSTRACT

Metastasis-associated lung adenocarcinoma transcript 1 (MALAT1) is a long noncoding RNA overexpressed in various cancers that promotes cell growth and metastasis. Although hypoxia has been shown to up-regulate MALAT1, only hypoxia-inducible factors (HIFs) have been implicated in activation of the MALAT1 promoter in specific cell types and other molecular mechanisms associated with hypoxia-mediated MALAT1 up-regulation remain largely unknown. Here, we demonstrate that hypoxia induces cancer cell-specific chromatin-chromatin interactions between newly identified enhancer-like cis-regulatory elements present at the MALAT1 locus. We show that hypoxia-mediated up-regulation of MALAT1 as well as its antisense strand TALAM1 occurs in breast cancer cells, but not in nontumorigenic mammary epithelial cells. Our analyses on the MALAT1 genomic locus discovered three novel putative enhancers that are located upstream and downstream of the MALAT1 gene body. We found that parts of these putative enhancers are epigenetically modified to a more open chromatin state under hypoxia in breast cancer cells. Furthermore, our chromosome conformation capture experiment demonstrated that noncancerous cells and breast cancer cells exhibit different interaction profiles under both normoxia and hypoxia, and only breast cancer cells gain specific chromatin interactions under hypoxia. Although the HIF-2α protein can enhance the interaction between the promoter and the putative 3' enhancer, the gain of chromatin interactions associated with other upstream elements, such as putative -7 and -20 kb enhancers, were HIF-independent events. Collectively, our study demonstrates that cancer cell-specific chromatin-chromatin interactions are formed at the MALAT1 locus under hypoxia, implicating a novel mechanism of MALAT1 regulation in cancer.


Subject(s)
Breast Neoplasms/metabolism , Cell Hypoxia , Chromatin/metabolism , RNA, Long Noncoding/genetics , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Line, Tumor , Enhancer Elements, Genetic , Humans , Promoter Regions, Genetic , Protein Binding , Up-Regulation
7.
Kidney Int ; 95(6): 1494-1504, 2019 06.
Article in English | MEDLINE | ID: mdl-31005274

ABSTRACT

Although genetic testing is increasingly used in clinical nephrology, a large number of patients with congenital abnormalities of the kidney and urinary tract (CAKUT) remain undiagnosed with current gene panels. Therefore, careful curation of novel genetic findings is key to improving diagnostic yields. We recently described a novel intellectual disability syndrome caused by de novo heterozygous loss-of-function mutations in the gene encoding the splicing factor SON. Here, we show that many of these patients, including two previously unreported, exhibit a wide array of kidney abnormalities. Detailed phenotyping of 14 patients with SON haploinsufficiency identified kidney anomalies in 8 patients, including horseshoe kidney, unilateral renal hypoplasia, and renal cysts. Recurrent urinary tract infections, electrolyte disturbances, and hypertension were also observed in some patients. SON knockdown in kidney cell lines leads to abnormal pre-mRNA splicing, resulting in decreased expression of several established CAKUT genes. Furthermore, these molecular events were observed in patient-derived cells with SON haploinsufficiency. Taken together, our data suggest that the wide spectrum of phenotypes in patients with a pathogenic SON mutation is a consequence of impaired pre-mRNA splicing of several CAKUT genes. We propose that genetic testing panels designed to diagnose children with a kidney phenotype should include the SON gene.


Subject(s)
DNA-Binding Proteins/genetics , Genetic Testing/methods , Haploinsufficiency , Minor Histocompatibility Antigens/genetics , RNA Splicing/genetics , Urogenital Abnormalities/genetics , Vesico-Ureteral Reflux/genetics , Adolescent , Adult , Child , Child, Preschool , DNA-Binding Proteins/metabolism , Female , HEK293 Cells , Humans , Male , Minor Histocompatibility Antigens/metabolism , RNA Precursors/genetics , RNA Precursors/metabolism , TRPP Cation Channels/genetics , Urogenital Abnormalities/diagnosis , Vesico-Ureteral Reflux/diagnosis
8.
Appl Environ Microbiol ; 83(5)2017 03 01.
Article in English | MEDLINE | ID: mdl-27986727

ABSTRACT

During routine screening for Burkholderia pseudomallei from water wells in northern Australia in areas where it is endemic, Gram-negative bacteria (strains MSMB43T, MSMB121, and MSMB122) with a similar morphology and biochemical pattern to B. pseudomallei and B. thailandensis were coisolated with B. pseudomallei on Ashdown's selective agar. To determine the exact taxonomic position of these strains and to distinguish them from B. pseudomallei and B. thailandensis, they were subjected to a series of phenotypic and molecular analyses. Biochemical and fatty acid methyl ester analysis was unable to distinguish B. humptydooensis sp. nov. from closely related species. With matrix-assisted laser desorption ionization-time of flight analysis, all isolates grouped together in a cluster separate from other Burkholderia spp. 16S rRNA and recA sequence analyses demonstrated phylogenetic placement for B. humptydooensis sp. nov. in a novel clade within the B. pseudomallei group. Multilocus sequence typing (MLST) analysis of the three isolates in comparison with MLST data from 3,340 B. pseudomallei strains and related taxa revealed a new sequence type (ST318). Genome-to-genome distance calculations and the average nucleotide identity of all isolates to both B. thailandensis and B. pseudomallei, based on whole-genome sequences, also confirmed B. humptydooensis sp. nov. as a novel Burkholderia species within the B. pseudomallei complex. Molecular analyses clearly demonstrated that strains MSMB43T, MSMB121, and MSMB122 belong to a novel Burkholderia species for which the name Burkholderia humptydooensis sp. nov. is proposed, with the type strain MSMB43T (American Type Culture Collection BAA-2767; Belgian Co-ordinated Collections of Microorganisms LMG 29471; DDBJ accession numbers CP013380 to CP013382).IMPORTANCEBurkholderia pseudomallei is a soil-dwelling bacterium and the causative agent of melioidosis. The genus Burkholderia consists of a diverse group of species, with the closest relatives of B. pseudomallei referred to as the B. pseudomallei complex. A proposed novel species, B. humptydooensis sp. nov., was isolated from a bore water sample from the Northern Territory in Australia. B. humptydooensis sp. nov. is phylogenetically distinct from B. pseudomallei and other members of the B. pseudomallei complex, making it the fifth member of this important group of bacteria.


Subject(s)
Burkholderia pseudomallei/classification , Burkholderia/classification , Burkholderia/genetics , Burkholderia/physiology , Phylogeny , Animals , Australia , Bacterial Typing Techniques/methods , Burkholderia/isolation & purification , Burkholderia Infections/microbiology , DNA, Bacterial/genetics , Disease Models, Animal , Fatty Acids/analysis , Genes, Bacterial/genetics , Genome, Bacterial , Melioidosis/microbiology , Mice , Mice, Inbred BALB C , Microbial Sensitivity Tests , Multilocus Sequence Typing/methods , Northern Territory , Phenotype , RNA, Ribosomal, 16S/genetics , Rec A Recombinases/genetics , Sequence Analysis, DNA , Species Specificity , Virulence , Water Microbiology
9.
Am J Hum Genet ; 99(3): 711-719, 2016 09 01.
Article in English | MEDLINE | ID: mdl-27545680

ABSTRACT

The overall understanding of the molecular etiologies of intellectual disability (ID) and developmental delay (DD) is increasing as next-generation sequencing technologies identify genetic variants in individuals with such disorders. However, detailed analyses conclusively confirming these variants, as well as the underlying molecular mechanisms explaining the diseases, are often lacking. Here, we report on an ID syndrome caused by de novo heterozygous loss-of-function (LoF) mutations in SON. The syndrome is characterized by ID and/or DD, malformations of the cerebral cortex, epilepsy, vision problems, musculoskeletal abnormalities, and congenital malformations. Knockdown of son in zebrafish resulted in severe malformation of the spine, brain, and eyes. Importantly, analyses of RNA from affected individuals revealed that genes critical for neuronal migration and cortex organization (TUBG1, FLNA, PNKP, WDR62, PSMD3, and HDAC6) and metabolism (PCK2, PFKL, IDH2, ACY1, and ADA) are significantly downregulated because of the accumulation of mis-spliced transcripts resulting from erroneous SON-mediated RNA splicing. Our data highlight SON as a master regulator governing neurodevelopment and demonstrate the importance of SON-mediated RNA splicing in human development.


Subject(s)
Brain/embryology , Brain/metabolism , DNA-Binding Proteins/genetics , Genes, Essential/genetics , Intellectual Disability/genetics , Minor Histocompatibility Antigens/genetics , Mutation/genetics , RNA Splicing/genetics , Animals , Brain/abnormalities , Brain/pathology , DNA-Binding Proteins/analysis , DNA-Binding Proteins/metabolism , Developmental Disabilities/genetics , Developmental Disabilities/pathology , Developmental Disabilities/physiopathology , Eye Abnormalities/genetics , Female , Haploinsufficiency/genetics , Head/abnormalities , Heterozygote , Humans , Intellectual Disability/pathology , Intellectual Disability/physiopathology , Male , Metabolic Diseases/genetics , Metabolic Diseases/metabolism , Minor Histocompatibility Antigens/analysis , Minor Histocompatibility Antigens/metabolism , Pedigree , RNA, Messenger/analysis , Spine/abnormalities , Syndrome , Zebrafish/abnormalities , Zebrafish/embryology , Zebrafish/genetics
10.
Mol Cell ; 61(6): 859-73, 2016 Mar 17.
Article in English | MEDLINE | ID: mdl-26990989

ABSTRACT

Dysregulation of MLL complex-mediated histone methylation plays a pivotal role in gene expression associated with diseases, but little is known about cellular factors modulating MLL complex activity. Here, we report that SON, previously known as an RNA splicing factor, controls MLL complex-mediated transcriptional initiation. SON binds to DNA near transcription start sites, interacts with menin, and inhibits MLL complex assembly, resulting in decreased H3K4me3 and transcriptional repression. Importantly, alternatively spliced short isoforms of SON are markedly upregulated in acute myeloid leukemia. The short isoforms compete with full-length SON for chromatin occupancy but lack the menin-binding ability, thereby antagonizing full-length SON function in transcriptional repression while not impairing full-length SON-mediated RNA splicing. Furthermore, overexpression of a short isoform of SON enhances replating potential of hematopoietic progenitors. Our findings define SON as a fine-tuner of the MLL-menin interaction and reveal short SON overexpression as a marker indicating aberrant transcriptional initiation in leukemia.


Subject(s)
DNA-Binding Proteins/genetics , Histone-Lysine N-Methyltransferase/biosynthesis , Leukemia, Myeloid, Acute/genetics , Myeloid-Lymphoid Leukemia Protein/biosynthesis , Proto-Oncogene Proteins/genetics , Transcription, Genetic , Alternative Splicing/genetics , Cell Line, Tumor , Chromatin/genetics , DNA-Binding Proteins/biosynthesis , Gene Expression Regulation, Leukemic , Histone-Lysine N-Methyltransferase/genetics , Humans , Leukemia, Myeloid, Acute/pathology , Methylation , Minor Histocompatibility Antigens , Myeloid-Lymphoid Leukemia Protein/genetics , Protein Binding , Protein Isoforms/genetics , Proto-Oncogene Proteins/metabolism
11.
PLoS One ; 10(10): e0140274, 2015.
Article in English | MEDLINE | ID: mdl-26484663

ABSTRACT

The pangenomic diversity in Burkholderia pseudomallei is high, with approximately 5.8% of the genome consisting of genomic islands. Genomic islands are known hotspots for recombination driven primarily by site-specific recombination associated with tRNAs. However, recombination rates in other portions of the genome are also high, a feature we expected to disrupt gene order. We analyzed the pangenome of 37 isolates of B. pseudomallei and demonstrate that the pangenome is 'open', with approximately 136 new genes identified with each new genome sequenced, and that the global core genome consists of 4568±16 homologs. Genes associated with metabolism were statistically overrepresented in the core genome, and genes associated with mobile elements, disease, and motility were primarily associated with accessory portions of the pangenome. The frequency distribution of genes present in between 1 and 37 of the genomes analyzed matches well with a model of genome evolution in which 96% of the genome has very low recombination rates but 4% of the genome recombines readily. Using homologous genes among pairs of genomes, we found that gene order was highly conserved among strains, despite the high recombination rates previously observed. High rates of gene transfer and recombination are incompatible with retaining gene order unless these processes are either highly localized to specific sites within the genome, or are characterized by symmetrical gene gain and loss. Our results demonstrate that both processes occur: localized recombination introduces many new genes at relatively few sites, and recombination throughout the genome generates the novel multi-locus sequence types previously observed while preserving gene order.


Subject(s)
Burkholderia pseudomallei/genetics , Gene Order , Genes, Bacterial/genetics , Genome, Bacterial/genetics , Algorithms , Burkholderia pseudomallei/classification , Burkholderia pseudomallei/isolation & purification , Evolution, Molecular , Gene Transfer, Horizontal , Genetic Variation , Models, Genetic , Recombination, Genetic , Species Specificity
12.
Expert Rev Anti Infect Ther ; 12(12): 1487-99, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25312349

ABSTRACT

Burkholderia pseudomallei is a gram-negative bacterium that causes melioidosis, a multifaceted disease that is highly endemic in southeast Asia and northern Australia. This facultative intracellular pathogen possesses a large genome that encodes a wide array of virulence factors that promote survival in vivo by manipulating host cell processes and disarming elements of the host immune system. Antigens and systems that play key roles in B. pseudomallei virulence include capsular polysaccharide, lipopolysaccharide, adhesins, specialized secretion systems, actin-based motility and various secreted factors. This review provides an overview of the current and steadily expanding knowledge regarding the molecular mechanisms used by this organism to survive within a host and their contribution to the pathogenesis of melioidosis.


Subject(s)
Burkholderia pseudomallei/genetics , Melioidosis/microbiology , Melioidosis/physiopathology , Virulence Factors/genetics , Bacterial Adhesion , Bacterial Secretion Systems , Burkholderia pseudomallei/immunology , Burkholderia pseudomallei/pathogenicity , Drug Resistance, Bacterial/genetics , Host-Pathogen Interactions , Humans , Melioidosis/immunology , Virulence Factors/immunology , Virulence Factors/metabolism
13.
Carbohydr Res ; 386: 68-72, 2014 Mar 11.
Article in English | MEDLINE | ID: mdl-24491843

ABSTRACT

Burkholderia oklahomensis E0147 is a US isolated bacterium believed to express a similar O-antigen to type A structure of the highly pathogenic species, Burkholderia pseudomallei. Both species are genetically closely related. Lipopolysaccharide was collected from E0147 and structurally characterized to test this hypothesis. Glycosyl composition and linkage analyses in conjunction with 1D and 2D (1)H and (13)C NMR spectroscopy showed that the O-antigen was a repeating disaccharide with the following structure: [3)-ß-D-Glcp-(1→3)-2OAc-α-L-6dTalp-(1→]n NMR spectroscopy also revealed the presence of a co-extracted exopolysaccharide previously described in B. pseudomallei, with the structure: [3)-2OAc-ß-D-Galp-(1→4)-α-D-Galp-(1→3)-ß-D-Galp-(1→5)-ß-D-Kdop-(2→]n.


Subject(s)
Burkholderia/chemistry , O Antigens/chemistry , Carbohydrate Conformation , Carbohydrate Sequence , Magnetic Resonance Spectroscopy , Molecular Sequence Data
14.
Genome Announc ; 1(4)2013 Aug 22.
Article in English | MEDLINE | ID: mdl-23969058

ABSTRACT

We describe the complete genome sequence of Burkholderia pseudomallei MSHR305, a clinical isolate taken from a fatal encephalomyelitis case, a rare form of melioidosis. This sequence will be used for comparisons to identify the genes that are involved in neurological cases.

15.
Genome Announc ; 1(3)2013 May 23.
Article in English | MEDLINE | ID: mdl-23704173

ABSTRACT

Here, we describe the draft genome sequence of Burkholderia pseudomallei NCTC 13392. This isolate has been distributed as K96243, but distinct genomic differences have been identified. The genomic sequence of this isolate will provide the genomic context for previously conducted functional studies.

16.
BMC Microbiol ; 12: 250, 2012 Nov 05.
Article in English | MEDLINE | ID: mdl-23126230

ABSTRACT

BACKGROUND: Burkholderia pseudomallei is the etiological agent of melioidosis and a CDC category B select agent with no available effective vaccine. Previous immunizations in mice have utilized the lipopolysaccharide (LPS) as a potential vaccine target because it is known as one of the most important antigenic epitopes in B. pseudomallei. Complicating this strategy are the four different B. pseudomallei LPS O-antigen types: A, B, B2, and rough. Sero-crossreactivity is common among O-antigens of Burkholderia species. Here, we identified the presence of multiple B. pseudomallei O-antigen types and sero-crossreactivity in its near-neighbor species. RESULTS: PCR screening of O-antigen biosynthesis genes, phenotypic characterization using SDS-PAGE, and immunoblot analysis showed that majority of B. mallei and B. thailandensis strains contained the typical O-antigen type A. In contrast, most of B. ubonensis and B. thailandensis-like strains expressed the atypical O-antigen types B and B2, respectively. Most B. oklahomensis strains expressed a distinct and non-seroreactive O-antigen type, except strain E0147 which expressed O-antigen type A. O-antigen type B2 was also detected in B. thailandensis 82172, B. ubonensis MSMB108, and Burkholderia sp. MSMB175. Interestingly, B. thailandensis-like MSMB43 contained a novel serotype B positive O-antigen. CONCLUSIONS: This study expands the number of species which express B. pseudomallei O-antigen types. Further work is required to elucidate the full structures and how closely these are to the B. pseudomallei O-antigens, which will ultimately determine the efficacy of the near-neighbor B serotypes for vaccine development.


Subject(s)
Burkholderia/classification , Burkholderia/immunology , O Antigens/analysis , Animals , Biosynthetic Pathways/genetics , Cross Reactions , DNA, Bacterial/chemistry , DNA, Bacterial/genetics , Electrophoresis, Polyacrylamide Gel , Genes, Bacterial , Humans , Immunoblotting , Mice , Molecular Sequence Data , Polymerase Chain Reaction , Sequence Analysis, DNA , Serotyping
17.
PLoS Negl Trop Dis ; 6(1): e1453, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22235357

ABSTRACT

Lipopolysaccharide (LPS) is one of the most important virulence and antigenic components of Burkholderia pseudomallei, the causative agent of melioidosis. LPS diversity in B. pseudomallei has been described as typical, atypical or rough, based upon banding patterns on SDS-PAGE. Here, we studied the genetic and molecular basis of these phenotypic differences. Bioinformatics was used to determine the diversity of genes known or predicted to be involved in biosynthesis of the O-antigenic moiety of LPS in B. pseudomallei and its near-relative species. Multiplex-PCR assays were developed to target diversity of the O-antigen biosynthesis gene patterns or LPS genotypes in B. pseudomallei populations. We found that the typical LPS genotype (LPS genotype A) was highly prevalent in strains from Thailand and other countries in Southeast Asia, whereas the atypical LPS genotype (LPS genotype B) was most often detected in Australian strains (~13.8%). In addition, we report a novel LPS ladder pattern, a derivative of the atypical LPS phenotype, associated with an uncommon O-antigen biosynthesis gene cluster that is found in only a small B. pseudomallei sub-population. This new LPS group was designated as genotype B2. We also report natural mutations in the O-antigen biosynthesis genes that potentially cause the rough LPS phenotype. We postulate that the diversity of LPS may correlate with differential immunopathogenicity and virulence among B. pseudomallei strains.


Subject(s)
Antigenic Variation/genetics , Antigenic Variation/immunology , Burkholderia pseudomallei/genetics , Burkholderia pseudomallei/immunology , O Antigens/genetics , O Antigens/immunology , Asia, Southeastern , Australia , Burkholderia pseudomallei/isolation & purification , Computational Biology , DNA, Bacterial/chemistry , DNA, Bacterial/genetics , Genotype , Humans , Lipopolysaccharides/genetics , Lipopolysaccharides/immunology , Melioidosis/microbiology , Molecular Sequence Data , Multiplex Polymerase Chain Reaction/methods , Sequence Alignment , Sequence Analysis, DNA , Sequence Homology, Nucleic Acid
SELECTION OF CITATIONS
SEARCH DETAIL
...