Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Microbiol Spectr ; 11(3): e0374422, 2023 06 15.
Article in English | MEDLINE | ID: mdl-37022154

ABSTRACT

Small-scale studies investigating the relationship between pigs' intestinal microbiota and growth performance have generated inconsistent results. We hypothesized that on farms under favorable environmental conditions (e.g., promoting sow nest-building behavior, high colostrum production, low incidence of diseases and minimal use of antimicrobials), the piglet gut microbiota may develop toward a population that promotes growth and reduces pathogenic bacteria. Using 16S rRNA gene amplicon sequencing, we sampled and profiled the fecal microbiota from 170 individual piglets throughout suckling and postweaning periods (in total 670 samples) to track gut microbiota development and its potential association with growth. During the suckling period, the dominant genera were Lactobacillus and Bacteroides, the latter being gradually replaced by Clostridium sensu scricto 1 as piglets aged. The gut microbiota during the nursery stage, not the suckling period, predicted the average daily growth (ADG) of piglets. The relative abundances of SCFA-producing genera, in particular Faecalibacterium, Megasphaera, Mitsuokella, and Subdoligranulum, significantly correlated with high ADG of weaned piglets. In addition, the succession of the gut microbiota in high-ADG piglets occurred faster and stabilized sooner upon weaning, whereas the gut microbiota of low-ADG piglets continued to mature after weaning. Overall, our findings suggest that weaning is the major driver of gut microbiota variation in piglets with different levels of overall growth performance. This calls for further research to verify if promotion of specific gut microbiota, identified here at weaning transition, is beneficial for piglet growth. IMPORTANCE The relationship between pigs' intestinal microbiota and growth performance is of great importance for improving piglets' health and reducing antimicrobial use. We found that gut microbiota variation is significantly associated with growth during weaning and the early nursery period. Importantly, transitions toward a mature gut microbiota enriched with fiber-degrading bacteria mostly complete upon weaning in piglets with better growth. Postponing the weaning age may therefore favor the development of fiber degrading gut bacteria, conferring the necessary capacity to digest and harvest solid postweaning feed. The bacterial taxa associated with piglet growth identified herein hold potential to improve piglet growth and health.


Subject(s)
Gastrointestinal Microbiome , Swine , Animals , Female , Weaning , Gastrointestinal Microbiome/genetics , RNA, Ribosomal, 16S/genetics , Bacteria/genetics , Feces/microbiology
2.
BMC Genomics ; 20(1): 286, 2019 Apr 11.
Article in English | MEDLINE | ID: mdl-30975085

ABSTRACT

BACKGROUND: Cattle populations are highly amenable to the genetic mapping of male reproductive traits because longitudinal data on ejaculate quality and dense microarray-derived genotypes are available for thousands of artificial insemination bulls. Two young Nordic Red bulls delivered sperm with low progressive motility (i.e., asthenospermia) during a semen collection period of more than four months. The bulls were related through a common ancestor on both their paternal and maternal ancestry. Thus, a recessive mode of inheritance of asthenospermia was suspected. RESULTS: Both bulls were genotyped at 54,001 SNPs using the Illumina BovineSNP50 Bead chip. A scan for autozygosity revealed that they were identical by descent for a 2.98 Mb segment located on bovine chromosome 25. This haplotype was not found in the homozygous state in 8557 fertile bulls although five homozygous haplotype carriers were expected (P = 0.018). Whole genome-sequencing uncovered that both asthenospermic bulls were homozygous for a mutation that disrupts a canonical 5' splice donor site of CCDC189 encoding the coiled-coil domain containing protein 189. Transcription analysis showed that the derived allele activates a cryptic splice site resulting in a frameshift and premature termination of translation. The mutated CCDC189 protein is truncated by more than 40%, thus lacking the flagellar C1a complex subunit C1a-32 that is supposed to modulate the physiological movement of the sperm flagella. The mutant allele occurs at a frequency of 2.5% in Nordic Red cattle. CONCLUSIONS: Our study in cattle uncovered that CCDC189 is required for physiological movement of sperm flagella thus enabling active progression of spermatozoa and fertilization. A direct gene test may be implemented to monitor the asthenospermia-associated allele and prevent the birth of homozygous bulls that are infertile. Our results have been integrated in the Online Mendelian Inheritance in Animals (OMIA) database ( https://omia.org/OMIA002167/9913/ ).


Subject(s)
Dairying , Infertility, Male/genetics , Animals , Cattle , Chromosomes, Mammalian/genetics , Genotype , Homozygote , Male , Mitochondria/metabolism , Polymorphism, Single Nucleotide , Protein Isoforms/genetics
3.
Int J Mol Sci ; 20(3)2019 Jan 25.
Article in English | MEDLINE | ID: mdl-30691059

ABSTRACT

Mating or cervical deposition of spermatozoa or seminal plasma (SP) modifies the expression of genes affecting local immune defense processes at the oviductal sperm reservoir in animals with internal fertilization, frequently by down-regulation. Such responses may occur alongside sperm transport to or even beyond the reservoir. Here, immune-related gene expression was explored with cDNA microarrays on porcine cervix-to-infundibulum tissues, pre-/peri-ovulation. Samples were collected 24 h post-mating or cervical deposition of sperm-peak spermatozoa or SP (from the sperm-peak fraction or the whole ejaculate). All treatments of this interventional study affected gene expression. The concerted action of spermatozoa and SP down-regulated chemokine and cytokine (P00031), interferon-gamma signaling (P00035), and JAK/STAT (P00038) pathways in segments up to the sperm reservoir (utero-tubal junction (UTJ)/isthmus). Spermatozoa in the vanguard sperm-peak fraction (P1-AI), uniquely displayed an up-regulatory effect on these pathways in the ampulla and infundibulum. Sperm-free SP, on the other hand, did not lead to major effects on gene expression, despite the clinical notion that SP mitigates reactivity by the female immune system after mating or artificial insemination.


Subject(s)
Cervix Uteri/metabolism , Oligonucleotide Array Sequence Analysis/veterinary , Pituitary Gland/metabolism , Semen/immunology , Spermatozoa/immunology , Animals , Cytokines/genetics , Female , Gene Expression Profiling/veterinary , Gene Expression Regulation , Gene Ontology , Insemination, Artificial/veterinary , Interferon-gamma/genetics , Janus Kinases/genetics , Male , STAT Transcription Factors/genetics , Sexual Behavior, Animal , Signal Transduction , Swine
4.
Theriogenology ; 127: 7-14, 2019 Mar 15.
Article in English | MEDLINE | ID: mdl-30611014

ABSTRACT

Artificial insemination (AI) with cooled stallion semen has increased markedly during the last decades in all countries, but fertility is often lower than when fresh semen or natural mating is used. The objective of this study was to examine field data (1634 ,cycles 523 Standardbred (SB) mares, 575 Finnhorse (FH) mares, and 90 stallions) using multivariable logistic regression for factors influencing the pregnancy rate (PR) after AI with cooled transported semen from SB and FH stallions. The PR per cycle for the material was 47%: Finnhorses 42% and Standardbreds 53%. When assessed with multivariable logistic regression analyses with a generalized linear mixed model, variables that affected the PR were breed, the number of inseminated estrus cycles, the percentage of progressively motile sperm (PMOT) in the ejaculate/AI dose at the time of shipment, and the number of progressively motile sperm in the AI dose at the time of insemination. In Standardbreds, variables that increased the per cycle PR were the number of AI per estrus cycle (multiple inseminations increasing the probability of pregnancy compared to only one insemination), the number of inseminated cycles, and PMOT in the AI dose at the time of insemination. In Finnhorses, the number of AI per estrus cycle (two and three inseminations increasing the probability of pregnancy compared to only one), the number of spermatozoa in the ejaculate and in the AI dose, and PMOT in the ejaculate/AI dose at the time of shipment increased the per cycle PR. Non-significant factors for the whole material included the type of artificial vagina (open-ended or closed), transport time, place of AI (stud farm or home stable), insemination done by veterinarian or technician, weekday, month, age of the mare (all age classes combined), age of the stallion, ejaculate parameters (sperm concentration, total number of sperm), and insemination dose parameters (volume proportion of seminal plasma, sperm concentration, PMOT, total number of sperm). In conclusion, breed, breeding opportunity in more than one cycle, more than one insemination/estrus, PMOT of the ejaculate/AI dose and the number of progressively motile sperm in the AI dose at the time of insemination are important for the outcome of inseminations with cooled semen.


Subject(s)
Horses/physiology , Insemination, Artificial/veterinary , Animals , Female , Logistic Models , Male , Multivariate Analysis , Pregnancy , Pregnancy Rate , Semen Analysis/veterinary , Semen Preservation/methods , Semen Preservation/veterinary
5.
Biol Reprod ; 97(2): 249-257, 2017 Aug 01.
Article in English | MEDLINE | ID: mdl-28679164

ABSTRACT

Intrauterine growth restriction (IUGR) is caused by dysregulation of placental metabolism. Paternally inherited IUGR mutations in the fetus influence maternal physiology via the placenta. However, it is not known whether the maternal placenta also affects the extent of IUGR in such fetuses. In cattle and other ruminants, maternal-fetal communication occurs primarily at the placentomes. We previously identified a 3΄ deletion in the noncoding MER1 repeat containing imprinted transcript 1 (MIMT1) gene that, when inherited from the sire, causes IUGR and late abortion in Ayshire cattle with variable levels of severity. Here, we compared the transcriptome and genomic imprinting in fetal and maternal placentome components of wild-type and MIMT1Del/WT fetuses before IUGR became apparent, to identify key early events. Transcriptome analysis revealed fewer differentially expressed genes in maternal than fetal MIMT1Del/WT placentome. AST1, within the PEG3 domain, was the only gene consistently reduced in IUGR in both fetal and maternal samples. Several genes showed an imprinting pattern associated with IUGR, of which only secernin 3 (SCRN3) and paternally expressed 3 (PEG3) were differentially imprinted in both placentome components. Loss of strictly monoallelic, allele-specific expression (∼80:20) of PEG3 in the maternal MIMT1Del/WT placenta could be associated with incomplete penetrance of MIMT1Del. Our data show that dysregulation of the PEG3 domain is involved in IUGR, but also reveal that maternal placental tissues may affect the penetrance of the paternally inherited IUGR mutation.


Subject(s)
Cattle Diseases/genetics , Fetal Growth Retardation/veterinary , Gene Expression Regulation, Developmental/physiology , Animals , Cattle , Cattle Diseases/pathology , DNA Methylation , Female , Fetal Growth Retardation/genetics , Genetic Predisposition to Disease , Genomic Imprinting , Kruppel-Like Transcription Factors/genetics , Kruppel-Like Transcription Factors/metabolism , Mutation , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Placenta/metabolism , Pregnancy
6.
BMC Genet ; 17: 49, 2016 Feb 29.
Article in English | MEDLINE | ID: mdl-26923438

ABSTRACT

BACKGROUND: Artificial insemination is widely used in many cattle breeding programs. Semen samples of breeding bulls are collected and closely examined immediately after collection at artificial insemination centers. Only ejaculates without anomalous findings are retained for artificial insemination. Although morphological aberrations of the spermatozoa are a frequent reason for discarding ejaculates, the genetic determinants underlying poor semen quality are scarcely understood. RESULTS: A tail stump sperm defect was observed in three bulls of the Swedish Red cattle breed. The spermatozoa of affected bulls were immotile because of severely disorganized tails indicating disturbed spermatogenesis. We genotyped three affected bulls and 18 unaffected male half-sibs at 46,035 SNPs and performed homozygosity mapping to map the fertility disorder to an 8.42 Mb interval on bovine chromosome 13. The analysis of whole-genome re-sequencing data of an affected bull and 300 unaffected animals from eleven cattle breeds other than Swedish Red revealed a 1 bp deletion (Chr13: 24,301,425 bp, ss1815612719) in the eleventh exon of the armadillo repeat containing 3-encoding gene (ARMC3) that was compatible with the supposed recessive mode of inheritance. The deletion is expected to alter the reading frame and to induce premature translation termination (p.A451fs26). The mutated protein is shortened by 401 amino acids (46 %) and lacks domains that are likely essential for normal protein function. CONCLUSIONS: We report the phenotypic and genetic characterization of a sterilizing tail stump sperm defect in the Swedish Red cattle breed. Exploiting high-density genotypes and massive re-sequencing data enabled us to identify the most likely causal mutation for the fertility disorder in bovine ARMC3. Our results provide the basis for monitoring the mutated variant in the Swedish Red cattle population and for the early identification of infertile animals.


Subject(s)
Armadillo Domain Proteins/genetics , Cattle/genetics , Frameshift Mutation , Infertility/genetics , Spermatozoa/pathology , Animals , Breeding , Chromosomes, Mammalian/genetics , Fertility/genetics , Genetic Variation , Genotyping Techniques , Homozygote , Infertility/diagnosis , Infertility/veterinary , Male , Semen Analysis , Spermatogenesis/genetics , Spermatozoa/metabolism , Sweden
7.
Anim Genet ; 47(1): 106-9, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26537866

ABSTRACT

We used a genetic (MIMT1(Del)) model of intrauterine growth restriction to investigate dysregulation of PEG3 domain gene expression in bovine foetal and maternal placenta. ZIM2, APEG3 and PEG3 expressions were similarly reduced in MIMT1(Del/) (WT) foetal placenta, suggesting coordinated regulation. Methylation of DNA CpG sites associated with these genes showed no differences, but differences in the levels of MIMT1 RNA methylation at three CpG sites were found in foetal placenta. Our data are consistent with the presence of a bidirectional promoter 5' of MIMT1 and suggest a regulatory role for the MIMT1 non-coding transcript. PEG3 domain expression on the maternal placenta side was not affected by the foetal mutation.


Subject(s)
Cattle/genetics , Fetal Growth Retardation/genetics , Gene Expression Regulation, Developmental , Placenta/metabolism , Animals , CpG Islands , DNA Methylation , Female , Fetus , Mutation , Pregnancy , Promoter Regions, Genetic
8.
BMC Genomics ; 15: 890, 2014 Oct 12.
Article in English | MEDLINE | ID: mdl-25306138

ABSTRACT

BACKGROUND: Inherited developmental diseases can cause severe animal welfare and economic problems in dairy cattle. The use of a small number of bulls for artificial insemination (AI) carries a risk that recessive defects rapidly enrich in the population. In recent years, an increasing number of Finnish Ayrshire calves have been identified with signs of ptosis, intellectual disability, retarded growth and mortality, which constitute an inherited disorder classified as PIRM syndrome. RESULTS: We established a cohort of nine PIRM-affected calves and 38 unaffected half-siblings and performed a genome-wide association study (GWAS) to map the disease to a 700-kb region on bovine chromosome 17 (p = 1.55 × 10-9). Whole genome re-sequencing of an unaffected carrier, its affected progeny and 43 other unaffected animals from another breed identified a G > A substitution mutation at the last nucleotide of exon 23 in the ubiquitin protein ligase E3B encoding gene (UBE3B). UBE3B transcript analysis revealed in-frame exon skipping in the affected animals resulting in an altered protein lacking 40 amino acids, of which 20 are located in the conserved HECT-domain, the catalytic site of the UBE3B protein. Mutation screening in 129 Ayrshire AI bulls currently used in Finland indicated a high carrier frequency (17.1%). We also found that PIRM syndrome might be connected to the recently identified AH1 haplotype, which has a frequency of 26.1% in the United States Ayrshire population. CONCLUSION: We describe PIRM syndrome in cattle, which is associated with the mutated UBE3B gene. The bovine phenotype resembles human Kaufman oculocerebrofacial syndrome, which is also caused by mutations in UBE3B. PIRM syndrome might be connected with the recently identified AH1 haplotype, which is associated with reduced fertility in the US Ayrshire population. This study enables the development of a genetic test to efficiently reduce the high frequency of mutant UBE3B in Ayrshires, significantly improving animal health and reducing economic loss.


Subject(s)
Eye Abnormalities/genetics , Intellectual Disability/genetics , Limb Deformities, Congenital/genetics , Microcephaly/genetics , Ubiquitin-Protein Ligases/genetics , Amino Acid Sequence , Animals , Cattle , Chromosome Mapping , Exons , Eye Abnormalities/pathology , Eye Abnormalities/veterinary , Facies , Genome-Wide Association Study , Haplotypes , Intellectual Disability/pathology , Intellectual Disability/veterinary , Limb Deformities, Congenital/pathology , Limb Deformities, Congenital/veterinary , Male , Microcephaly/pathology , Microcephaly/veterinary , Molecular Sequence Data , Phenotype , Polymorphism, Single Nucleotide , RNA Splicing , Sequence Alignment , Ubiquitin-Protein Ligases/metabolism
9.
Biochem Biophys Res Commun ; 444(3): 391-4, 2014 Feb 14.
Article in English | MEDLINE | ID: mdl-24480436

ABSTRACT

The existence of non-CpG methylation in mammalian DNA has mainly been observed in embryonic stem cells, but its functional significance is uncertain. We found an age-dependent non-CpG hypermethylation in DMR at the 3' end of the MIMT1 in the placenta of intrauterine growth restricted foetuses in cattle. Data suggest that this DMR play a role in epigenetic regulation of the PEG3 domain.


Subject(s)
CpG Islands , DNA Methylation , Fetal Growth Retardation/metabolism , Mutation , Placenta/metabolism , Animals , Base Sequence , Cattle , DNA Primers , Female , Polymerase Chain Reaction , Pregnancy
10.
PLoS One ; 8(9): e75659, 2013.
Article in English | MEDLINE | ID: mdl-24086604

ABSTRACT

Impaired migration of primordial germ cells during embryonic development causes hereditary gonadal hypoplasia in both sexes of Northern Finncattle and Swedish Mountain cattle. The affected gonads exhibit a lack of or, in rare cases, a reduced number of germ cells. Most affected animals present left-sided gonadal hypoplasia. However, right-sided and bilateral cases are also found. This type of gonadal hypoplasia prevails in animals with white coat colour. Previous studies indicated that gonadal hypoplasia is inherited in an autosomal recessive fashion with incomplete penetrance. In order to identify genetic regions underlying gonadal hypoplasia, a genome-wide association study (GWAS) and a copy number variation (CNV) analysis were performed with 94 animals, including 21 affected animals, using bovine 777,962 SNP arrays. The GWAS and CNV results revealed two significantly associated regions on bovine chromosomes (BTA) 29 and 6, respectively (P=2.19 x 10(-13) and P=5.65 x 10(-6)). Subsequent cytogenetic and PCR analyses demonstrated that homozygosity of a ~500 kb chromosomal segment translocated from BTA6 to BTA29 (Cs29 allele) is the underlying genetic mechanism responsible for gonadal hypoplasia. The duplicated segment includes the KIT gene that is known to regulate the migration of germ cells and precursors of melanocytes. This duplication is also one of the two translocations associated with colour sidedness in various cattle breeds.


Subject(s)
Cell Movement/genetics , DNA Copy Number Variations/genetics , Germ Cells/pathology , Gonads/pathology , Alleles , Animals , Case-Control Studies , Cattle , Chromosome Mapping/veterinary , Genome-Wide Association Study/methods , Melanocytes/pathology , Polymorphism, Single Nucleotide/genetics
11.
Int J Dev Neurosci ; 31(7): 463-7, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23726833

ABSTRACT

We examined levels of gene expression in the brains of bovine fetuses carrying a truncated MIMT1 allele, MIMT1(Del), shown to cause late abortion and stillbirth as a result of fetal growth restriction. MIMT1 is a non-protein coding gene that forms part of the imprinted PEG3 (paternally expressed gene 3) domain. Microarray analysis of brain cortex samples from mid-gestation MIMT1(Del/WT) bovine fetuses and wild-type siblings was performed to study the effect of fetal growth restriction on brain gene expression. Statistical analysis revealed 134 genes with increased mRNA levels and 22 with reduced levels in MIMT1(Del/WT) fetuses. Gene set enrichment analysis identified a relatively small number of significant functional clusters representing three major biological processes: response to oxidative stress, angiogenesis, and epithelial cell proliferation. Gene expression microarray analyses identified increased expression of VIPR2, HTRA1, S100A4 and MYH8 in fetuses carrying the deletion and decreased expression of DRD2, ADAM18, miR345, ZNF585A. ADAM18, DRD2 and S100A4 are known to be involved in prenatal brain development. ZNF585A, miR-345, VIPR2, HTRA1, and MYH8 are known to be involved in cell growth and differentiation, but any role in neural developmental has yet to be elucidated.


Subject(s)
Brain/embryology , Brain/metabolism , Cattle Diseases/genetics , Gene Expression Regulation, Developmental , Kruppel-Like Transcription Factors/genetics , Transcriptome/genetics , ADAM Proteins/genetics , ADAM Proteins/metabolism , Animals , Cattle , Cattle Diseases/pathology , Embryo, Mammalian , Female , High-Temperature Requirement A Serine Peptidase 1 , Microarray Analysis , Myosin Heavy Chains/genetics , Myosin Heavy Chains/metabolism , Pregnancy , RNA, Messenger/metabolism , Receptors, Dopamine D2/genetics , Receptors, Dopamine D2/metabolism , Receptors, Vasoactive Intestinal Peptide, Type II/genetics , Receptors, Vasoactive Intestinal Peptide, Type II/metabolism , Serine Endopeptidases/genetics , Serine Endopeptidases/metabolism
12.
Biol Reprod ; 87(6): 140, 2012 Jun.
Article in English | MEDLINE | ID: mdl-23100617

ABSTRACT

We previously identified a microdeletion (Del) in the maternally imprinted PEG3 domain in cattle that results in loss of paternal MIMT1 expression and causes late-term abortion and stillbirth. The mutation, when inherited from the sire, is semilethal for his progeny, with 85% mortality. Here we precisely delineate the deletion and describe comparative analyses of fetuses carrying the deletion with wild-type (WT) siblings. Global DNA methylation analysis of cotyledon tissue revealed greater global CpG methylation in fetuses with the deletion (P = 0.003). Gene expression microarray analyses identified increased NPSR1A, IL1RN, NOS3, IL4R, ZDHHC22, and SMOC2 expression in fetuses carrying the deletion and decreased GRID1, PLG, and IGF1 expression. Involvement of the NPSR1A, IL1RN, NOS3, and IL4R genes suggests that some form of restriction related to blood supply, perhaps hypoxemia, may play a role in the pathological mechanism. Also, imprinted genes known to play a role in mammalian prenatal development, specifically IGF2, DLK1, MEST, AST1, PEG3, APEG3, and H19, showed differential expression. The most striking difference was abundant abnormal expression of the neuropeptide S receptor 1 (NPSR1) gene in placental cotyledon tissue of 7 of 11 MIMT1(Del/WT) fetuses. The similarity of this proportion to that of the semilethal mortality rate suggests that abnormal NPSR1 expression may be linked to death or survival of MIMT1(Del/WT) fetuses. NPSR1 is expressed as two isoforms (A and B), and isoform A was detected in MIMT1(Del/WT) cotyledons. NPSR1A is normally not expressed in placenta. Its role in the stillbirth phenomenon has yet to be elucidated, but it may provide a useful prognostic indicator.


Subject(s)
Abortion, Veterinary/genetics , Cattle Diseases/genetics , Gene Deletion , Gene Expression Regulation, Developmental , Kruppel-Like Transcription Factors/genetics , Placenta/metabolism , Stillbirth/veterinary , Animals , Cattle , Cattle Diseases/metabolism , Cattle Diseases/physiopathology , DNA Methylation , Female , Fetal Hypoxia/etiology , Fetal Hypoxia/physiopathology , Heterozygote , Kruppel-Like Transcription Factors/metabolism , Male , Oligonucleotide Array Sequence Analysis/veterinary , Placenta/blood supply , Pregnancy , Pregnancy Proteins/genetics , Pregnancy Proteins/metabolism , Protein Isoforms/genetics , Protein Isoforms/metabolism , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Stillbirth/genetics
13.
BMC Genomics ; 12: 591, 2011 Dec 02.
Article in English | MEDLINE | ID: mdl-22136159

ABSTRACT

BACKGROUND: Male infertility is an increasing problem in all domestic species including man. Localization and identification of genes involved in defects causing male infertility provide valuable information of specific events in sperm development. Sperm development is a complex process, where diploid spermatogonia develop into haploid, highly specialized spermatozoa. Correct expression and function of various genes and their protein products are required for production of fertile sperm. We have identified an infertility defect in Finnish Yorkshire boars caused by spermatogenic arrest. The aim of this study was to locate the disease associated region using genome wide screen with the PorcineSNP60 Beadchip and identify the causal mutation by candidate gene approach. RESULTS: In the Finnish Yorkshire pig population the spermatogenic arrest (SA) defect appears to be of genetic origin and causes severe degeneration of germ cells and total absence of spermatozoa. Genome wide scan with the PorcineSNP60 Beadchip localized the SA defect to porcine chromosome 12 in a 2 Mbp region. Sequencing of a candidate gene Tex14 revealed a 51 bp insertion within exon 27, which caused differential splicing of the exon and created a premature translation stop codon. The expression of Tex14 was markedly down regulated in the testis of a SA affected boar compared to control boars and no protein product was identified by Western blotting. The SA insertion sequence was also found within intron 27 in all analyzed animals, thus the insertion appears to be a possible duplication event. CONCLUSION: In this study we report the identification of a causal mutation for infertility caused by spermatogenic arrest at an early meiotic phase. Our results highlight the role of TEX14 specifically in spermatogenesis and the importance of specific genomic remodeling events as causes for inherited defects.


Subject(s)
Exons , Infertility, Male/genetics , Oligospermia/genetics , Swine/genetics , Transcription Factors/genetics , Animals , Azoospermia/congenital , INDEL Mutation , Male , Meiosis , Sequence Analysis, DNA
14.
PLoS One ; 5(11): e15116, 2010 Nov 30.
Article in English | MEDLINE | ID: mdl-21152099

ABSTRACT

Congenital malformations resulting in late abortions and stillbirths affect the economic wellbeing of producers and the welfare of cattle in breeding programs. An extremely high incidence of stillbirths of "half-sized" calves of normal karyotype and uninflated lungs was diagnosed in the progeny of the Finnish Ayrshire (Bos taurus) bull--YN51. No other visible anatomical abnormalities were apparent in the stillborn calves. We herein describe the positional identification of a 110 kb microdeletion in the maternally imprinted PEG3 domain that results in a loss of paternal MIMT1 expression and causes late term abortion and stillbirth in cattle. Using the BovineSNP50 BeadChip we performed a genome-wide half-sib linkage analysis that identified a 13.3 Mb associated region on BTA18 containing the maternally imprinted PEG3 domain. Within this cluster we found a 110 kb microdeletion that removes a part of the non-protein coding MER1 repeat containing imprinted transcript 1 gene (MIMT1). To confirm the elimination of gene expression in calves inheriting this deletion, we examined the mRNA levels of the three maternally imprinted genes within the PEG3 domain, in brain and cotyledon tissue collected from eight fetuses sired by the proband. None of the fetuses that inherited the microdeletion expressed MIMT1 in either tissue. The mutation, when inherited from the sire, is semi-lethal for his progeny with an observed mortality rate of 85%. The survival of 15% is presumably due to the incomplete silencing of maternally inherited MIMT1 alleles. We designed a PCR-based assay to confirm the existence of the microdeletion in the MIMT1 region that can be used to assist cattle breeders in preventing the stillbirths.


Subject(s)
Abortion, Veterinary/genetics , Kruppel-Like Transcription Factors/genetics , Mutation , Stillbirth/genetics , Animals , Animals, Newborn , Cattle , Cells, Cultured , Chromosome Deletion , Chromosome Mapping , Female , Gene Expression , Genetic Predisposition to Disease , Genomic Imprinting , Genotype , Insemination, Artificial/veterinary , Male , Pedigree , Polymorphism, Single Nucleotide , Pregnancy , Reverse Transcriptase Polymerase Chain Reaction
15.
Int J Mol Med ; 18(5): 901-8, 2006 Nov.
Article in English | MEDLINE | ID: mdl-17016620

ABSTRACT

The efficacy of the most commonly used form of suicide gene therapy, the HSV-TK/GCV method, utilizing herpes simplex virus thymidine kinase (HSV-TK) and antiviral drug ganciclovir (GCV) has been demonstrated in clinical trials. However, safer delivery of the therapeutic gene and more controlled regulation of the transgene expression, the essential prerequisites for successful therapeutic use, are still needed. We describe improved suicide gene therapy against cancer through transcripitional targeting by a strong and selective tumor-specific human hexokinase II promoter (hHKII). We examined the targeting properties of the human hHKII promoter in different human non-small cell lung cancer (NSCLC) and other human cancer cell lines using self-inactivating, VSV-G pseudotyped lentiviral vector. To confirm accurate transcriptional targeting of the hHKII promoter, the lack of transgene expression was verified in human primary bronchial epithelial and bronchial fibroblast cells. Furthermore, tissue-specific expression of the promoter was confirmed using transgenic mouse lines carrying the hHKII promoter driven luciferase reporter gene. We also tested the efficacy of the HSV-TK/GCV suicide gene therapy with the hHKII targeted lentiviral vector to NSCLC cells. Our results show that the hHKII promoter is strongly expressed in cancer cells. The targeted vector with the shortest hHKII promoter fragment (352 bp) appeared to have the best targeting properties because it efficiently governed the expression of the therapeutic gene in cancer cell lines, especially in certain non-small cell lung cancer cell lines, the transgene expression in human primary cells was virtually undetectable, and expression of the proximal hHKII promoter in transgenic mice was very low in most tissues. Also, the anti-cancer efficacy of HSV-TK/GCV therapy with the hHKII-targeted vector was comparable to that obtained with the control vector that utilized a commonly used constitutive promoter from the human elongation factor 1 alpha (hEF1alpha) gene. In conclusion, the transcriptionally targeted lentivirus vector with hHKII promoter can successfully direct HSV-TK/GCV suicide gene therapy to non-small cell lung cancer and other tumor cell types. These results warrant further studies with orthotopic animal tumor models and primary human cancer material.


Subject(s)
Genes, Transgenic, Suicide , Genetic Therapy/methods , Hexokinase/genetics , Neoplasms/therapy , Promoter Regions, Genetic/genetics , Animals , Carcinoma, Non-Small-Cell Lung/therapy , Cell Line, Tumor , Gene Transfer Techniques , Genetic Vectors , Humans , Lentivirus/genetics , Lung Neoplasms/therapy , Mice , Mice, Transgenic , Thymidine Kinase/genetics , Transduction, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL
...