Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
Elife ; 122023 10 17.
Article in English | MEDLINE | ID: mdl-37847154

ABSTRACT

DMRT1 is the testis-determining factor in several species of vertebrates, but its involvement in mammalian testes differentiation, where SRY is the testis-determining gene, remains ambiguous. So far, DMRT1 loss-of-function has been described in two mammalian species and induces different phenotypes: Disorders of Sex Development (46, XY DSD) in men and male infertility in mice. We thus abolished DMRT1 expression by CRISPR/Cas9 in a third species of mammal, the rabbit. First, we observed that gonads from XY DMRT1-/- rabbit fetuses differentiated like ovaries, highlighting that DMRT1 is involved in testis determination. In addition to SRY, DMRT1 is required in the supporting cells to increase the expression of the SOX9 gene, which heads the testicular genetic cascade. Second, we highlighted another function of DMRT1 in the germline since XX and XY DMRT1-/- ovaries did not undergo meiosis and folliculogenesis. XX DMRT1-/- adult females were sterile, showing that DMRT1 is also crucial for female fertility. To conclude, these phenotypes indicate an evolutionary continuum between non-mammalian vertebrates such as birds and non-rodent mammals. Furthermore, our data support the potential involvement of DMRT1 mutations in different human pathologies, such as 46, XY DSD as well as male and female infertility.


Animals that reproduce sexually have organs called gonads, the ovaries and testes, which produce eggs and sperm. These organs, which are different in males and females, originate from the same cells during the development of the embryo. As a general rule, the chromosomal sex of an embryo, which gets determined at fertilization, leads to the activation and repression of specific genes. This in turn, controls whether the cells that will form the gonads will differentiate to develop testes or ovaries. Disruption of the key genes involved in the differentiation of the gonads can lead to fertility problems, and in some cases, it can cause the gonads to develop in the 'opposite' direction, resulting in a sex reversal. Identifying these genes is therefore essential to know how to maintain or restore fertility. DMRT1 is a gene that drives the differentiation of gonadal cells into the testicular pathway in several species of animals with backbones, including species of fish, frogs and birds. However, its role in mammals ­ where testis differentiation is driven by a different gene called SRY ­ is not well understood. Indeed, when DMRT1 is disrupted in male humans it leads to disorders of sex development, while disrupting this gene in male mice causes infertility. To obtain more information about the roles of DMRT1 in mammalian species, Dujardin et al. disrupted the gene in a third species of mammal: the rabbit. Dujardin et al. observed that chromosomally-male rabbits lacking DMRT1 developed ovaries instead of testes, showing that in rabbits, both SRY and DMRT1 are both required to produce testes. Additionally, this effect is similar to what is seen in humans, suggesting that rabbits may be a better model for human gonadal differentiation than mice are. Additionally, Dujardin et al. were also able to show that in female rabbits, lack of DMRT1 led to infertility, an effect that had not been previously described in other species. The results of Dujardin et al. may lead to better models for gonadal development in humans, involving DMRT1 in the differentiation of testes. Interestingly, they also suggest the possibility that mutations in this gene may be responsible for some cases of infertility in women. Overall, these findings indicate that DMRT1 is a key fertility gene.


Subject(s)
Disorder of Sex Development, 46,XY , Testis , Animals , Female , Male , Rabbits , Disorder of Sex Development, 46,XY/genetics , Disorder of Sex Development, 46,XY/metabolism , Fertility/genetics , Gene Expression Regulation, Developmental , Gonads/metabolism , Mammals/genetics , Sex Determination Processes/genetics , SOX9 Transcription Factor/genetics , SOX9 Transcription Factor/metabolism , Testis/metabolism
2.
Biol Reprod ; 109(4): 408-414, 2023 10 13.
Article in English | MEDLINE | ID: mdl-37561421

ABSTRACT

Gene knockout experiments have shown that many genes are dispensable for a given biological function. In this review, we make an assessment of male and female germ cell-specific genes dispensable for the function of reproduction in mice, the inactivation of which does not affect fertility. In particular, we describe the deletion of a 1 Mb block containing nineteen paralogous genes of the oogenesin/Pramel family specifically expressed in female and/or male germ cells, which has no consequences in both sexes. We discuss this notion of dispensability and the experiments that need to be carried out to definitively conclude that a gene is dispensable for a function.


Subject(s)
Infertility, Male , Testis , Animals , Female , Male , Mice , Fertility/genetics , Germ Cells , Infertility, Male/genetics , Mice, Knockout , Reproduction , Spermatogenesis/genetics
3.
Genes (Basel) ; 12(11)2021 11 19.
Article in English | MEDLINE | ID: mdl-34828428

ABSTRACT

Among tetrapods, the well differentiated heteromorphic sex chromosomes of birds and mammals have been highly investigated and their master sex-determining (MSD) gene, Dmrt1 and SRY, respectively, have been identified. The homomorphic sex chromosomes of reptiles have been the least studied, but the gap with birds and mammals has begun to fill. This review describes our current knowledge of reptilian sex chromosomes at the cytogenetic and molecular level. Most of it arose recently from various studies comparing male to female gene content. This includes restriction site-associated DNA sequencing (RAD-Seq) experiments in several male and female samples, RNA sequencing and identification of Z- or X-linked genes by male/female comparative transcriptome coverage, and male/female transcriptomic or transcriptome/genome substraction approaches allowing the identification of Y- or W-linked transcripts. A few putative master sex-determining (MSD) genes have been proposed, but none has been demonstrated yet. Lastly, future directions in the field of reptilian sex chromosomes and their MSD gene studies are considered.


Subject(s)
Sex Chromosomes/genetics , Sex Determination Processes/genetics , Turtles/genetics , Animals , Female , Male , Turtles/physiology
4.
Biol Reprod ; 91(6): 153, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25395674

ABSTRACT

FOXL2 loss of function in goats leads to the early transdifferentiation of ovaries into testes, then to the full sex reversal of XX homozygous mutants. By contrast, Foxl2 loss of function in mice induces an arrest of follicle formation after birth, followed by complete female sterility. In order to understand the molecular role of FOXL2 during ovarian differentiation in the goat species, putative FOXL2 target genes were determined at the earliest stage of gonadal sex-specific differentiation by comparing the mRNA profiles of XX gonads expressing the FOXL2 protein or not. Of these 163 deregulated genes, around two-thirds corresponded to testicular genes that were up-regulated when FOXL2 was absent, and only 19 represented female-associated genes, down-regulated in the absence of FOXL2. FOXL2 should therefore be viewed as an antitestis gene rather than as a female-promoting gene. In particular, the key testis-determining gene DMRT1 was found to be up-regulated ahead of SOX9, thus suggesting in goats that SOX9 primary up-regulation may require DMRT1. Overall, our results equated to FOXL2 being an antitestis gene, allowing us to propose an alternative model for the sex-determination process in goats that differs slightly from that demonstrated in mice.


Subject(s)
46, XX Testicular Disorders of Sex Development/genetics , Forkhead Transcription Factors/genetics , Genitalia, Female/metabolism , Goats/physiology , SOX9 Transcription Factor/genetics , Transcription Factors/genetics , 46, XX Testicular Disorders of Sex Development/veterinary , Animals , Animals, Genetically Modified , Cell Transdifferentiation , Embryo, Mammalian , Female , Gene Expression Regulation, Developmental , Genitalia, Female/embryology , Goats/genetics , High-Throughput Nucleotide Sequencing , Male , Ovary/metabolism , Sex Determination Processes/genetics , Testis/metabolism , Up-Regulation
5.
J Immunol ; 193(6): 2891-901, 2014 Sep 15.
Article in English | MEDLINE | ID: mdl-25114105

ABSTRACT

The MHC is a large genetic region controlling Ag processing and recognition by T lymphocytes in vertebrates. Approximately 40% of its genes are implicated in innate or adaptive immunity. A putative proto-MHC exists in the chordate amphioxus and in the fruit fly, indicating that a core MHC region predated the emergence of the adaptive immune system in vertebrates. In this study, we identify a putative proto-MHC with archetypal markers in the most basal branch of Metazoans--the placozoan Trichoplax adhaerens, indicating that the proto-MHC is much older than previously believed--and present in the common ancestor of bilaterians (contains vertebrates) and placozoans. Our evidence for a T. adhaerens proto-MHC was based on macrosynteny and phylogenetic analyses revealing approximately one third of the multiple marker sets within the human MHC-related paralogy groups have unique counterparts in T. adhaerens, consistent with two successive whole genome duplications during early vertebrate evolution. A genetic ontologic analysis of the proto-MHC markers in T. adhaerens was consistent with its involvement in defense, showing proteins implicated in antiviral immunity, stress response, and ubiquitination/proteasome pathway. Proteasome genes psma, psmb, and psmd are present, whereas the typical markers of adaptive immunity, such as MHC class I and II, are absent. Our results suggest that the proto-MHC was involved in intracellular intrinsic immunity and provide insight into the primordial architecture and functional landscape of this region that later in evolution became associated with numerous genes critical for adaptive immunity in vertebrates.


Subject(s)
Adaptive Immunity/genetics , Major Histocompatibility Complex/genetics , Placozoa/genetics , Placozoa/immunology , Animals , Biological Evolution , Genome , Humans , Major Histocompatibility Complex/immunology , Nerve Growth Factors/genetics , Phylogeny , Proteasome Endopeptidase Complex/genetics , Stress, Physiological/genetics , T-Lymphocytes/immunology , Ubiquitination/genetics
6.
PLoS One ; 8(5): e63512, 2013.
Article in English | MEDLINE | ID: mdl-23717440

ABSTRACT

Despite massive research efforts, the molecular etiology of bovine polledness and the developmental pathways involved in horn ontogenesis are still poorly understood. In a recent article, we provided evidence for the existence of at least two different alleles at the Polled locus and identified candidate mutations for each of them. None of these mutations was located in known coding or regulatory regions, thus adding to the complexity of understanding the molecular basis of polledness. We confirm previous results here and exhaustively identify the causative mutation for the Celtic allele (PC) and four candidate mutations for the Friesian allele (PF). We describe a previously unreported eyelash-and-eyelid phenotype associated with regular polledness, and present unique histological and gene expression data on bovine horn bud differentiation in fetuses affected by three different horn defect syndromes, as well as in wild-type controls. We propose the ectopic expression of a lincRNA in PC/p horn buds as a probable cause of horn bud agenesis. In addition, we provide evidence for an involvement of OLIG2, FOXL2 and RXFP2 in horn bud differentiation, and draw a first link between bovine, ovine and caprine Polled loci. Our results represent a first and important step in understanding the genetic pathways and key process involved in horn bud differentiation in Bovidae.


Subject(s)
Cattle/growth & development , Horns/growth & development , Alleles , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Cattle/genetics , Chromosome Mapping/methods , Forkhead Transcription Factors/genetics , Gene Expression Regulation, Developmental/genetics , Genetic Variation/genetics , Genotype , Goats/genetics , Goats/growth & development , Mutation/genetics , Phenotype , Receptors, G-Protein-Coupled/genetics , Sheep/genetics , Sheep/growth & development
7.
PLoS One ; 8(4): e60451, 2013.
Article in English | MEDLINE | ID: mdl-23593221

ABSTRACT

The rabbit is an attractive species for the study of gonad differentiation because of its 31-day long gestation, the timing of female meiosis around birth and the 15-day delay between gonadal switch and the onset of meiosis in the female. The expression of a series of genes was thus determined by qPCR during foetal life until adulthood, completed by a histological analysis and whenever possible by an immunohistological one. Interesting gene expression profiles were recorded. Firstly, the peak of SRY gene expression that is observed in early differentiated XY gonads in numerous mammals was also seen in the rabbit, but this expression was maintained at a high level until the end of puberty. Secondly, a peak of aromatase gene expression was observed at two-thirds of the gestation in XX gonads as in many other species except in the mouse. Thirdly, the expression of STRA8 and DMC1 genes (which are known to be specifically expressed in germ cells during meiosis) was enhanced in XX gonads around birth but also slightly and significantly in XY gonads at the same time, even though no meiosis occurs in XY gonad at this stage. This was probably a consequence of the synchronous strong NANOS2 gene expression in XY gonad. In conclusion, our data highlighted some rabbit-specific findings with respect to the gonad differentiation process.


Subject(s)
Cell Differentiation , Ovary/cytology , Ovary/embryology , Testis/cytology , Testis/embryology , Animals , Biomarkers/metabolism , Female , Fetus/cytology , Fetus/embryology , Fetus/metabolism , Gene Expression Regulation, Developmental , Male , Meiosis , Mice , Ovary/metabolism , Ovum/cytology , Ovum/metabolism , Puberty , Rabbits , Species Specificity , Spermatozoa/cytology , Spermatozoa/metabolism , Testis/metabolism , Tretinoin/metabolism
8.
Transgenic Res ; 22(3): 489-500, 2013 Jun.
Article in English | MEDLINE | ID: mdl-22961198

ABSTRACT

RNA interference is an attractive strategy to fight against viral diseases by targeting the mRNA of viral genes. Most studies have reported the transient delivery of small interfering RNA or small hairpin (shRNA) expression constructs. Here, we present the production of transgenic mice stably expressing shRNA or miRNA targeting the IE180 mRNA (immediate early gene) of the pseudorabies virus (PRV) which infects mice and farm animals. We firstly designed non-retroviral shRNA or miRNA expression vectors. Secondly, we selected the most efficient shRNA construct that targeted either the 5'part or 3'UTR of the IE mRNA and was able to knockdown the target gene expression in cultured cells, by measuring systematically the shRNA content and comparing this with the interfering effects. We then produced four lines of transgenic mice expressing different amounts of shRNA or miRNA in the brain but without signs of stimulation of innate immunity. Lastly, we tested their resistance to PRV infection. In all transgenic lines, we observed a significant resistance to viral challenge, the best being achieved with the shRNA construct targeting the 3'UTR of the IE gene. Viral DNA levels in the brains of infected mice were always lower in transgenic mice, even in animals that did not survive. Finally, this work reports an effective strategy to generate transgenic animals producing shRNA from non-retroviral expression vectors. Moreover, these mice are the first transgenic animal models producing shRNA with a significant antiviral effect but without any apparent shRNA toxicity.


Subject(s)
Disease Resistance/genetics , Mice, Transgenic , Pseudorabies/genetics , RNA, Small Interfering/genetics , Viral Proteins/genetics , 3' Untranslated Regions , Animals , Brain/virology , Disease Resistance/immunology , Genes, Immediate-Early , Herpesvirus 1, Suid/genetics , Herpesvirus 1, Suid/pathogenicity , Immunity, Innate/genetics , Mice , MicroRNAs/genetics
9.
PLoS One ; 6(11): e26950, 2011.
Article in English | MEDLINE | ID: mdl-22069478

ABSTRACT

BACKGROUND: We had previously reported that the Suppression Subtractive Hybridization (SSH) approach was relevant for the isolation of new mammalian genes involved in oogenesis and early follicle development. Some of these transcripts might be potential new oocyte and granulosa cell markers. We have now characterized one of them, named TOPAZ1 for the Testis and Ovary-specific PAZ domain gene. PRINCIPAL FINDINGS: Sheep and mouse TOPAZ1 mRNA have 4,803 bp and 4,962 bp open reading frames (20 exons), respectively, and encode putative TOPAZ1 proteins containing 1,600 and 1653 amino acids. They possess PAZ and CCCH domains. In sheep, TOPAZ1 mRNA is preferentially expressed in females during fetal life with a peak during prophase I of meiosis, and in males during adulthood. In the mouse, Topaz1 is a germ cell-specific gene. TOPAZ1 protein is highly conserved in vertebrates and specifically expressed in mouse and sheep gonads. It is localized in the cytoplasm of germ cells from the sheep fetal ovary and mouse adult testis. CONCLUSIONS: We have identified a novel PAZ-domain protein that is abundantly expressed in the gonads during germ cell meiosis. The expression pattern of TOPAZ1, and its high degree of conservation, suggests that it may play an important role in germ cell development. Further characterization of TOPAZ1 may elucidate the mechanisms involved in gametogenesis, and particularly in the RNA silencing process in the germ line.


Subject(s)
Evolution, Molecular , Germ Cells/metabolism , Homeodomain Proteins/metabolism , Meiosis/physiology , Ovary/metabolism , Testis/metabolism , Vertebrates/physiology , Amino Acid Sequence , Animals , Blotting, Western , Expressed Sequence Tags , Female , Fluorescent Antibody Technique , Gene Expression Regulation, Developmental , Germ Cells/cytology , Homeodomain Proteins/genetics , Male , Mice , Mice, Inbred C57BL , Molecular Sequence Data , Ovary/cytology , Phylogeny , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Sequence Homology, Amino Acid , Sheep , Testis/cytology
10.
Nature ; 472(7343): 370-4, 2011 Apr 21.
Article in English | MEDLINE | ID: mdl-21471966

ABSTRACT

X-chromosome inactivation (XCI) in female mammals allows dosage compensation for X-linked gene products between the sexes. The developmental regulation of this process has been extensively investigated in mice, where the X chromosome of paternal origin (Xp) is silenced during early embryogenesis owing to imprinted expression of the regulatory RNA, Xist (X-inactive specific transcript). Paternal XCI is reversed in the inner cell mass of the blastocyst and random XCI subsequently occurs in epiblast cells. Here we show that other eutherian mammals have very different strategies for initiating XCI. In rabbits and humans, the Xist homologue is not subject to imprinting and XCI begins later than in mice. Furthermore, Xist is upregulated on both X chromosomes in a high proportion of rabbit and human embryo cells, even in the inner cell mass. In rabbits, this triggers XCI on both X chromosomes in some cells. In humans, chromosome-wide XCI has not initiated even by the blastocyst stage, despite the upregulation of XIST. The choice of which X chromosome will finally become inactive thus occurs downstream of Xist upregulation in both rabbits and humans, unlike in mice. Our study demonstrates the remarkable diversity in XCI regulation and highlights differences between mammals in their requirement for dosage compensation during early embryogenesis.


Subject(s)
Chromosomes, Mammalian/genetics , Gene Expression Regulation, Developmental/genetics , Mammals/genetics , X Chromosome Inactivation/genetics , X Chromosome/genetics , Animals , Biological Evolution , Blastocyst/metabolism , Dosage Compensation, Genetic/genetics , Embryo, Mammalian/embryology , Embryo, Mammalian/metabolism , Female , Genes, X-Linked/genetics , Genomic Imprinting/genetics , Histones/metabolism , Humans , Hypoxanthine Phosphoribosyltransferase/genetics , Male , Mammals/embryology , Mice , Parthenogenesis , RNA, Long Noncoding , RNA, Untranslated/genetics , Rabbits , Species Specificity , Up-Regulation/genetics
11.
Gene ; 401(1-2): 97-107, 2007 Oct 15.
Article in English | MEDLINE | ID: mdl-17692477

ABSTRACT

Distal control of the whey acidic protein (WAP) locus was studied using a transgenic approach. A series of pig genomic fragments encompassing increasing DNA lengths upstream of the mammary specific whey acidic protein (WAP) gene transcription start point (tsp) and 5 kb downstream were used for microinjection in mouse fertilized eggs. Our data pointed out three regions as potent regulators for WAP but not for RAMP3 gene expression (a non mammary-specific gene located 30 kb upstream of the WAP gene). WAP gene activating elements were present in the -80 kb to -30 kb and -145 kb to -130 kb regions whereas inhibitors were present in the -130 kb to -80 kb region. The stimulatory regions were characterized by peaks of histone H4 acetylation and a poor nucleosome occupancy in lactating sow mammary glands but not in liver. These data reveal for the first time the existence of several remote potent regulatory regions of the pig WAP gene.


Subject(s)
Gene Expression Regulation , Milk Proteins/genetics , Acetylation , Animals , Chromatin Immunoprecipitation , Chromosomes, Artificial, Bacterial , DNA/genetics , Female , Gene Dosage , Histones/metabolism , Lactation , Mammary Glands, Animal/metabolism , Mice , Mice, Transgenic , Microinjections , Milk Proteins/isolation & purification , Nucleosomes/metabolism , Pregnancy , RNA, Messenger/metabolism , Swine , Transcription Factors/metabolism , Transcription Initiation Site , Transcription, Genetic , Transgenes , Zygote/metabolism
12.
Reprod Nutr Dev ; 46(5): 579-88, 2006.
Article in English | MEDLINE | ID: mdl-17107647

ABSTRACT

Milk is a very abundant source of proteins for animal and human consumption. Milk composition can be modified using transgenesis, including exogenous gene addition and endogenous gene inactivation. The study of milk protein genes has provided researchers with regulatory regions capable of efficiently and specifically driving the expression of foreign genes in milk. The projects underway are aimed at modifying milk composition, improving its nutritional value, reducing mammary infections, providing consumers with antipathogen proteins and preparing purified recombinant proteins for pharmaceutical use. The present paper summarises the current progress in this field.


Subject(s)
Animals, Genetically Modified , Lactation/genetics , Lactation/metabolism , Milk Proteins/genetics , Milk/standards , Animals , Cattle , Female , Gene Expression Regulation , Genetic Vectors , Humans , Milk/chemistry , Milk Proteins/chemistry , Milk Proteins/metabolism , Nutritive Value , Transgenes
13.
Gene ; 370: 104-12, 2006 Mar 29.
Article in English | MEDLINE | ID: mdl-16483732

ABSTRACT

Whey Acidic Protein (WAP) has been identified in the milk of only a few species, including mouse, rat, rabbit, camel, pig, tammar wallaby, brushtail possum, echidna and platypus. Despite intensive studies, it has not yet been found in the milk of Ruminants. We have isolated and characterized genomic WAP clones from ewe, goat and cow, identified their chromosomal localization and examined the expression of the endogenous WAP sequence in the mammary glands of all three species. The WAP sequences were localized on chromosome 4 (4q26) as expected from comparative mapping data. The three ruminant WAP sequences reveal the same deletion of a nucleotide at the end of the first exon when compared with the pig sequence. Due to this frameshift mutation, the putative proteins encoded by these sequences do not harbor the features of a usual WAP protein with two four-disulfide core domains. Moreover, RT-PCR experiments have shown that these sequences are not transcribed and are, thus, pseudogenes. This loss of functionality of the gene in Ruminants raises the question of the biological role of the WAP. Some putative roles previously suggested for WAP are discussed.


Subject(s)
Frameshift Mutation , Gene Expression Regulation/physiology , Milk Proteins/genetics , Pseudogenes/genetics , Ruminants/genetics , Sequence Deletion , Animals , Base Sequence , Chromosomes/genetics , Exons/genetics , Female , Mammary Glands, Animal/metabolism , Milk Proteins/biosynthesis , Molecular Sequence Data , Ruminants/metabolism , Sequence Homology, Nucleic Acid
14.
J Dairy Res ; 72 Spec No: 113-9, 2005.
Article in English | MEDLINE | ID: mdl-16180729

ABSTRACT

For 10 years, the regulatory regions of the mouse and rabbit whey acidic protein gene have been used to express heterologous proteins in the milk of transgenic mice, as well as to produce pharmaceutical proteins, on a large scale, in the milk of transgenic livestock. To date, a broad range of expression levels have been detected, and elucidation of the structure-function relationship in these regulatory regions might help to achieve high levels of expression, reproducibly. An extended 5' regulatory region (17.6 kb v. 6.3 kb) of the rabbit whey acidic promoter resulted in an increased frequency of rabbit whey acidic protein expression in transgenic mice. However, the expression levels were low compared with the high expression levels achieved in both transgenic mice and rabbits using the heterologous kappa-casein in the 6.3 kb rabbit whey acidic protein 5' regulatory region. These results underline the importance of the 3' downstream regulatory regions, which still need to be better characterized in the whey acidic protein gene.


Subject(s)
Gene Expression Regulation , Milk Proteins/genetics , Rabbits/genetics , Regulatory Sequences, Nucleic Acid/physiology , Animals , Caseins/genetics , Genetic Linkage , Mice , Mice, Transgenic , Milk/chemistry , Milk/metabolism , Promoter Regions, Genetic/genetics , Transfection
15.
Biochim Biophys Acta ; 1627(1): 7-14, 2003 May 13.
Article in English | MEDLINE | ID: mdl-12759187

ABSTRACT

A 140-kb pig DNA fragment containing the whey acidic protein (WAP) gene cloned in a bacterial artificial chromosome (BAC344H5) has been shown to contain all of the cis-elements necessary for position-independent, copy-dependent and tissue-specific expression in transgenic mice. The insert from this BAC was sequenced. This revealed the presence of two other genes with quite different expression patterns in pig tissues and in transfected HC11 mouse mammary cells. The RAMP3 gene is located 15 kb upstream of the WAP gene in reverse orientation. The CPR2 gene is located 5 kb downstream of the WAP gene in the same orientation. The same locus organization was found in the human genome. The region between RAMP3 and CPR2 in the human genome contains a WAP gene-like sequence with several points of mutation which may account for the absence of WAP from human milk.


Subject(s)
Gene Expression Regulation , Genetic Linkage , Milk Proteins/genetics , Animals , Base Sequence , Chromosome Mapping , Genome , Humans , Intracellular Signaling Peptides and Proteins , Membrane Proteins/genetics , Molecular Sequence Data , Organ Specificity , Receptor Activity-Modifying Protein 3 , Receptor Activity-Modifying Proteins , Sequence Analysis, DNA , Swine , Synteny
16.
Reprod Nutr Dev ; 42(2): 117-25, 2002.
Article in English | MEDLINE | ID: mdl-12216957

ABSTRACT

The study and the control of milk synthesis are required to decipher the mechanisms of gene expression, to improve milk production, to modify milk composition, to induce a resistance to diseases in the mammary gland and to produce recombinant proteins of pharmaceutical interest. Transgenesis has become a mandatory tool to reach these goals. The use of transgenesis is still limited by the difficulty of adding foreign genes in farm animals and mainly by replacing genes by homologous recombination. Transgene expression is also often ill-controlled. The present paper summarizes the current progress in this field with a particular emphasis on expression vectors for transgenes.


Subject(s)
Animals, Genetically Modified , Gene Expression Regulation , Lactation/genetics , Lactation/physiology , Milk/metabolism , Animals , Female , Genetic Vectors , Mammary Glands, Animal/metabolism , Mammary Glands, Animal/physiology , Transgenes
SELECTION OF CITATIONS
SEARCH DETAIL
...