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










Publication year range
1.
Mol Biol Cell ; 30(2): 209-218, 2019 01 15.
Article in English | MEDLINE | ID: mdl-30462579

ABSTRACT

Tyrosine kinase inhibitors are widely used in the clinic, but limited information is available about their toxicity in developing organisms. Here, we tested the effect of tyrosine kinase inhibitors targeting the ErbB receptors for their effects on developing zebrafish ( Danio rerio) embryos. Embryos treated with wide-spectrum pan-ErbB inhibitors or erbb4a-targeting antisense oligonucleotides demonstrated reduced locomotion, reduced diameter of skeletal muscle fibers, and reduced expression of muscle-specific genes, as well as reduced motoneuron length. The phenotypes in the skeletal muscle, as well as the defect in motility, were rescued both by microinjection of human ERBB4 mRNA and by transposon-mediated muscle-specific ERBB4 overexpression. The role of ErbB4 in regulating motility was further controlled by targeted mutation of the endogenous erbb4a locus in the zebrafish genome by CRISPR/Cas9. These observations demonstrate a potential for the ErbB tyrosine kinase inhibitors to induce neuromuscular toxicity in a developing organism via a mechanism involving inhibition of ErbB4 function.


Subject(s)
Embryo, Nonmammalian/metabolism , Muscle Development/drug effects , Neurogenesis/drug effects , Neuromuscular Junction/embryology , Protein Kinase Inhibitors/pharmacology , Receptor Protein-Tyrosine Kinases/antagonists & inhibitors , Receptor, ErbB-4/antagonists & inhibitors , Zebrafish Proteins/antagonists & inhibitors , Zebrafish/embryology , Animals , Base Sequence , Embryo, Nonmammalian/drug effects , Gene Expression Regulation, Developmental/drug effects , Morpholinos/pharmacology , Motor Neurons/drug effects , Motor Neurons/metabolism , Muscle Cells/drug effects , Muscle Cells/metabolism , Muscle Development/genetics , Muscle, Skeletal/metabolism , Mutation/genetics , Neurogenesis/genetics , Neuromuscular Junction/drug effects , Receptor Protein-Tyrosine Kinases/genetics , Receptor Protein-Tyrosine Kinases/metabolism , Receptor, ErbB-4/genetics , Receptor, ErbB-4/metabolism , Zebrafish/genetics , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism
2.
PLoS One ; 8(6): e65887, 2013.
Article in English | MEDLINE | ID: mdl-23824706

ABSTRACT

A characteristic feature of celiac disease is the presence of circulating autoantibodies targeted against transglutaminase 2 (TG2), reputed to have a function in angiogenesis. In this study we investigated whether TG2-specific autoantibodies derived from celiac patients inhibit angiogenesis in both ex vivo and in vivo models and sought to clarify the mechanism behind this phenomenon. We used the ex vivo murine aorta-ring and the in vivo mouse matrigel-plug assays to address aforementioned issues. We found angiogenesis to be impaired as a result of celiac disease antibody supplementation in both systems. Our results also showed the dynamics of endothelial cells was affected in the presence of celiac antibodies. In the in vivo angiogenesis assays, the vessels formed were able to transport blood despite impairment of functionality after treatment with celiac autoantibodies, as revealed by positron emission tomography. We conclude that celiac autoantibodies inhibit angiogenesis ex vivo and in vivo and impair vascular functionality. Our data suggest that the anti-angiogenic mechanism of the celiac disease-specific autoantibodies involves extracellular TG2 and inhibited endothelial cell mobility.


Subject(s)
Autoantibodies/immunology , Celiac Disease/immunology , GTP-Binding Proteins/immunology , Neovascularization, Pathologic/immunology , Transglutaminases/immunology , Animals , Endothelial Cells/immunology , Female , Human Umbilical Vein Endothelial Cells , Humans , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Protein Glutamine gamma Glutamyltransferase 2
3.
Mol Cell Biol ; 32(4): 868-79, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22184064

ABSTRACT

Cyclin-dependent kinase 16 (CDK16, PCTK1) is a poorly characterized protein kinase, highly expressed in the testis and the brain. Here, we report that CDK16 is activated by membrane-associated cyclin Y (CCNY). Treatment of transfected human cells with the protein kinase A (PKA) activator forskolin blocked, while kinase inhibition promoted, CCNY-dependent targeting of CDK16-green fluorescent protein (GFP) to the cell membrane. CCNY binding to CDK16 required a region upstream of the kinase domain and was found to be inhibited by phosphorylation of serine 153, a potential PKA phosphorylation site. Thus, in contrast to other CDKs, CDK16 is regulated by phosphorylation-controlled cyclin binding. CDK16 isolated from murine testis was unphosphorylated, interacted with CCNY, and exhibited kinase activity. To investigate the function of CDK16 in vivo, we established a conditional knockout allele. Mice lacking CDK16 developed normally, but male mice were infertile. Spermatozoa isolated from their epididymis displayed thinning and elongation of the annulus region, adopted a bent shape, and showed impaired motility. Moreover, CDK16-deficient spermatozoa had malformed heads and excess residual cytoplasm, suggesting a role of CDK16 in spermiation. Thus, CDK16 is a membrane-targeted CDK essential for spermatogenesis.


Subject(s)
Cyclin-Dependent Kinases/metabolism , Cyclins/metabolism , Spermatogenesis/physiology , Amino Acid Sequence , Animals , Base Sequence , Cell Line , Cell Membrane/metabolism , Cyclin-Dependent Kinases/deficiency , Cyclin-Dependent Kinases/genetics , Cyclins/genetics , DNA Primers/genetics , Enzyme Activation , Female , Humans , Infertility, Male/genetics , Infertility, Male/metabolism , Infertility, Male/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Molecular Sequence Data , Phosphorylation , Protein Binding , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sequence Homology, Amino Acid , Spermatogenesis/genetics , Spermatozoa/abnormalities
4.
PLoS One ; 6(4): e18929, 2011 Apr 18.
Article in English | MEDLINE | ID: mdl-21533109

ABSTRACT

Bacterial biofilms resist host defenses and antibiotics partly because of their decreased metabolism. Some bacteria use proinflammatory cytokines, such as interleukin (IL)-1ß, as cues to promote biofilm formation and to alter virulence. Although one potential bacterial IL-1ß receptor has been identified, current knowledge of the bacterial IL-1ß sensing mechanism is limited. In chronic biofilm infection, periodontitis, Aggregatibacter actinomycetemcomitans requires tight adherence (tad)-locus to form biofilms, and tissue destroying active lesions contain more IL-1ß than inactive ones. The effect of IL-1ß on the metabolic activity of A. actinomycetemcomitans biofilm was tested using alamarBlue™. The binding of IL-1ß to A. actinomycetemcomitans cells was investigated using transmission electron microscopy and flow cytometry. To identify the proteins which interacted with IL-1ß, different protein fractions from A. actinomycetemcomitans were run in native-PAGE and blotted using biotinylated IL-1ß and avidin-HRP, and identified using mass spectroscopy. We show that although IL-1ß slightly increases the biofilm formation of A. actinomycetemcomitans, it reduces the metabolic activity of the biofilm. A similar reduction was observed with all tad-locus mutants except the secretin mutant, although all tested mutant strains as well as wild type strains bound IL-1ß. Our results suggest that IL-1ß might be transported into the A. actinomycetemcomitans cells, and the trimeric form of intracellular ATP synthase subunit ß interacted with IL-1ß, possibly explaining the decreased metabolic activity. Because ATP synthase is highly conserved, it might universally enhance biofilm resistance to host defense by binding IL-1ß during inflammation.


Subject(s)
Bacterial Proteins/metabolism , Biopolymers/metabolism , Interleukin-1beta/metabolism , Pasteurellaceae/enzymology , Biofilms , Flow Cytometry , Humans , Microscopy, Electron, Transmission , Protein Binding
5.
Reproduction ; 139(3): 587-98, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20007639

ABSTRACT

Growth differentiation factor-9 (GDF9) and bone morphogenetic protein-15 (BMP15) are among the key regulators transmitting the signaling between the oocyte and the surrounding granulosa cells. Previously, it has been shown that a recombinant BMP type II receptor ectodomain-Fc fusion protein (BMPR2ecd-Fc) is able to inhibit the actions of GDF9 and BMP15 in vitro. Here, we have produced bioactive BMPR2ecd-Fc, which was injected i.p. into neonatal mice. Early folliculogenesis was first studied by injecting mice five times with various doses of BMPR2ecd-Fc during the postnatal days 4-12. Folliculogenesis was affected dose dependently, as evidenced by a decreased mitogenesis of granulosa cells of the growing follicles. Furthermore, we also noticed a decrease in the number of secondary and tertiary follicles as well as an increase in the oocyte size. Electron microscopic analysis revealed that the ultrastructure of the granulosa cells of the primary follicles was not affected by the BMPR2ecd-Fc treatment. A second study was conducted to investigate whether a longer treatment with 12 injections during postnatal days 4-28 would inhibit folliculogenesis. Similar effects were observed in the two studies on the early follicular developmental stages. However, in the long-term study, later stages of folliculogenesis were not blocked but rather increased numbers of antral follicles, preovulatory follicles, and corpora lutea were found. We conclude that BMPR2ecd-Fc is a potent modulator of ovarian folliculogenesis in vivo, and thus, is a valuable tool for studying the physiology and downstream effects of oocyte-derived growth factors in vivo.


Subject(s)
Bone Morphogenetic Protein 15/antagonists & inhibitors , Growth Differentiation Factor 9/antagonists & inhibitors , Oocytes/drug effects , Ovarian Follicle/drug effects , Recombinant Fusion Proteins/pharmacology , Animals , Animals, Newborn , Bone Morphogenetic Protein 15/pharmacology , Bone Morphogenetic Protein Receptors, Type II/chemistry , CHO Cells , Cricetinae , Cricetulus , Female , Growth Differentiation Factor 9/pharmacology , Hep G2 Cells , Humans , Immunoglobulin Fc Fragments/chemistry , Immunoglobulin Fc Fragments/pharmacology , Intercellular Signaling Peptides and Proteins/pharmacology , Mice , Oocytes/physiology , Oogenesis/drug effects , Oogenesis/physiology , Ovarian Follicle/physiology , Sexual Maturation/physiology
6.
Mech Dev ; 126(1-2): 80-9, 2009.
Article in English | MEDLINE | ID: mdl-18955134

ABSTRACT

The transcription factor Gata3 is an important regulator of the development of thymus, the nervous system, ear, kidney, and adrenal glands. This study analyzes the role of Gata3 in the developing heart using a mouse strain containing an nlsLacZ reporter gene fused in frame to the Gata3 gene by homologous recombination. Using in situ hybridization, RT-PCR and Gata3-LacZ histochemistry, Gata3 expression was shown in various cardiac structures up to newborn stage. During looping stages (E9.5-E11.5) Gata3-LacZ activity recapitulated endogenous Gata3 and was abundantly expressed in the endocardial ridges and endothelium of distal outflow tract. Strong reporter gene expression was also noted in the mesenchyme of ventral branchial arches, and in the epithelium. In the atrioventricular canal expression was relatively lower. In the four-chambered heart stages (E13.5-E17.5) the LacZ-staining did not recapitulate the endogenous Gata3 transcript and showed rather lineage tracing of formerly Gata3-expressing cells in the hearts. beta-Galactosidase activity was detected in the cusps of semilunar valves, aorta, pulmonary trunk, innominate and common carotid arteries, and faintly in the atrioventricular valves. Gata3-null embryos die normally between E11 and E12. Pharmacological treatment with sympathomimetic beta-adrenergic receptor agonist lengthens the survival up to E18 when malformations of the heart such as ventricular septal defect (VSD), double-outlet of right ventricle (DORV), anomalies of the aortic arch (AAA) and persistent truncus arteriosus (PTA) were detected. The specified malformations correlate with the normal developmental pattern of Gata3-LacZ expression. The short outflow tract and insufficient rotation of truncus arteriosus during looping stages might be the main reasons underlying these malformations.


Subject(s)
GATA3 Transcription Factor/deficiency , GATA3 Transcription Factor/metabolism , Heart Defects, Congenital/metabolism , Heart Defects, Congenital/physiopathology , Animals , Embryo, Mammalian/embryology , Embryo, Mammalian/metabolism , GATA3 Transcription Factor/genetics , Gene Expression Regulation, Developmental , Genes, Reporter/genetics , Heart Defects, Congenital/genetics , In Situ Hybridization , Mice , Mice, Knockout , Neural Crest/embryology , Neural Crest/metabolism , Phenotype , RNA, Messenger/genetics
8.
Int J Cardiol ; 112(2): 219-22, 2006 Sep 20.
Article in English | MEDLINE | ID: mdl-16297473

ABSTRACT

Angiotensin II (ANG-II) is a critical regulator of various signaling pathways involved in growth and remodeling of the vascular, cardiac, and renal cells and tissues. Although it contributes to several physiologic and pathologic events in the cardiovascular system, its role in growth and differentiation of the newborn heart is still unclear. We analyzed the effect of ANG-II treatment on apoptosis, DNA synthesis, and nucleolar organizer regions (AgNORs) activity in newborn rat myocardium. Injections of ANG-II for 5-days caused significant increase of the 3H-thymidine labeling index (M+/-m) in the myocardium of 7-day-old rats (from 6.95+/-0.32% to 8.53+/-0.22%, p<0.05). There was also significant increase in the cross sectional surface area of cardiomyocytes (from 686+/-57 to 872+/-54 microm2, p<0.05), number of nucleoli (from 2.5+/-0.05 to 2.8+/-0.1, p<0.05), and nucleolar surface area (from 2.6+/-0.09 to 3.2+/-0.22 microm2, p<0.05). These changes were accompanied by significant increase in the apoptotic indices analyzed by TDT-mediated dUTP-biotin nick end-labeling (TUNEL) (from 0.044+/-0.01% to 0.093+/-0.01%, p<0.05). Interestingly, we found no differences in cell proliferation between the test and control animals after 21-45 days of age, which were injected with ANG-II in the first postnatal week. However, the area of cardiomyocytes and the number of nucleoli in 21-day-old rats continued to increase significantly. Our results indicate that ANG-II modulates cardiac growth during the neonatal period via stimulation of apoptosis, cell cycle events and cellular growth of cardiomyocytes and that these effects can persist up to 15 days after injection of ANG-II has been completed.


Subject(s)
Angiotensin II/pharmacology , Antigens, Nuclear/biosynthesis , Apoptosis/drug effects , Cell Proliferation/drug effects , Heart Ventricles/drug effects , Nuclear Proteins/biosynthesis , Vasoconstrictor Agents/pharmacology , Angiotensin II/physiology , Animals , Animals, Newborn , Antigens, Nuclear/drug effects , Immunohistochemistry , In Situ Nick-End Labeling , Myocytes, Cardiac/metabolism , Nuclear Proteins/drug effects , Rats , Ventricular Function
9.
Oncogene ; 24(49): 7301-9, 2005 Nov 10.
Article in English | MEDLINE | ID: mdl-16007123

ABSTRACT

We have previously demonstrated that male transgenic (TG) mice overexpressing human chorionic gonadotropin (hCG+) develop reproductive organ defects, but no tumors, in adult age. In this study, the effects of persistently elevated hCG were followed in TG males between day 5 postpartum and adulthood. Leydig cell (LC) adenomas were found in prepubertal mice, most prominently at the age of 10 days, but not in adult age. Serum testosterone concentrations were significantly increased in TG males at all ages studied. The phenotype of the prepubertal hCG+ males resembled that found in boys upon expression of constitutively activating luteinizing hormone (LH) receptor mutations. The temporal expression patterns of the fetal LC marker gene, thrombospondin 2, and those of adult LCs, hydroxysteroid dehydrogenase-6, delta5-3-beta and prostaglandin D synthase, were similar in wild-type and hCG+ males. Hence, the postnatal adenomas resemble functionally fetal LCs, and only these cells are susceptible to hCG-induced tumorigenesis. Our findings demonstrate a novel intriguing difference between the fetal and adult LC populations and provide further insight into the potential tumorigenic effects of gonadotropins.


Subject(s)
Adenoma/etiology , Chorionic Gonadotropin, beta Subunit, Human/metabolism , Gene Expression Regulation, Developmental , Glycoprotein Hormones, alpha Subunit/metabolism , Leydig Cells/metabolism , 3-Hydroxysteroid Dehydrogenases/metabolism , Adenoma/metabolism , Adenoma/pathology , Animals , Chorionic Gonadotropin, beta Subunit, Human/genetics , Fetus , Gene Expression Regulation, Neoplastic , Glycoprotein Hormones, alpha Subunit/genetics , Humans , Intramolecular Oxidoreductases/metabolism , Leydig Cells/cytology , Lipocalins , Male , Mice , Mice, Transgenic , Phenotype , Testis/metabolism , Testis/pathology , Thrombospondins/metabolism , Up-Regulation
10.
Mol Cell Biol ; 25(12): 4892-902, 2005 Jun.
Article in English | MEDLINE | ID: mdl-15923608

ABSTRACT

The testatin gene was previously isolated in a screen focused on finding novel signaling molecules involved in sex determination and differentiation. testatin is specifically upregulated in pre-Sertoli cells in early fetal development, immediately after the onset of Sry expression, and was therefore considered a strong candidate for involvement in early testis development. testatin expression is maintained in the adult Sertoli cell, and it can also be found in a small population of germ cells. Testatin shows homology to family 2 cystatins, a group of broadly expressed small secretory proteins that are inhibitors of cysteine proteases in vitro but whose in vivo functions are unclear. testatin belongs to a novel subfamily among the cystatins, comprising genes that all show expression patterns that are strikingly restricted to reproductive tissue. To investigate a possible role of testatin in testis development and male reproduction, we have generated a mouse with targeted disruption of the testatin gene. We found no abnormalities in the testatin knockout mice with regard to fetal and adult testis morphology, cellular ultrastructure, body and testis weight, number of offspring, spermatogenesis, or hormonal parameters (testosterone, luteinizing hormone, and follicle-stimulating hormone).


Subject(s)
Cystatins/genetics , Cystatins/metabolism , Fertility/physiology , Sexual Development/physiology , Testis/growth & development , Amino Acid Sequence , Animals , Cystatins/classification , Female , Follicle Stimulating Hormone/blood , Gene Targeting , Luteinizing Hormone/blood , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Molecular Sequence Data , Phylogeny , Pregnancy , Reproduction/physiology , Sequence Alignment , Sex Determination Analysis , Spermatozoa/cytology , Spermatozoa/metabolism , Testis/metabolism , Testis/physiology , Testis/ultrastructure , Testosterone/blood
11.
Prog Retin Eye Res ; 23(4): 403-34, 2004 Jul.
Article in English | MEDLINE | ID: mdl-15219875

ABSTRACT

The three-dimensional structure of the eye plays an important role in providing a correct optical environment for vision. Much of this function is dependent on the unique structural features of ocular connective tissue, especially of the collagen types and their supramolecular structures. For example, the organization of collagen fibrils is largely responsible for transparency and refraction of cornea, lens and vitreous body, and collagens present in the sclera are largely responsible for the structural strength of the eye. Phylogenetically, most of the collagens are highly conserved between different species, which suggests that collagens also share similar functions in mice and men. Despite considerable differences between the mouse and the human eye, particularly in the proportion of the different tissue components, the difficulty of performing systematic histologic and molecular studies on the human eye has made mouse an appealing alternative to studies addressing the role of individual genes and their mutations in ocular diseases. From a genetic standpoint, the mouse has major advantages over other experimental animals as its genome is better known than that of other species and it can be manipulated by the modern techniques of genetic engineering. Furthermore, it is easy, quick and relatively cheap to produce large quantities of mice for systematic studies. Thus, transgenic techniques have made it possible to study consequences of specific mutations in genes coding for structural components of ocular connective tissues in mice. As these changes in mice have been shown to resemble those in human diseases, mouse models are likely to provide efficient tools for pathogenetic studies on human disorders affecting the extracellular matrix. This review is aimed to clarify the role of collagenous components in the mouse and human eye with a closer look at the new findings of the collagens in the cartilage and the eye, the so-called "cartilage collagens".


Subject(s)
Collagen/physiology , Extracellular Matrix/metabolism , Eye Proteins/physiology , Eye/metabolism , Animals , Cartilage/metabolism , Humans , Mice
12.
Pediatr Res ; 55(2): 197-204, 2004 Feb.
Article in English | MEDLINE | ID: mdl-14605248

ABSTRACT

Mice that lack cardiac muscle alpha-actin die during the perinatal period. Approximately 56% of mice that are homozygous null (-/-) for a functional cardiac alpha-actin gene do not survive to term, and the remainder generally die within 2 wk of birth. We found that there were neither morphologic differences nor differences in the extent of apoptosis between the mutant and normal hearts on embryonic day (E) 12 and E14 of development. However, apoptosis was greater in the hearts of homozygous null mice on E17 and postnatal day 1 when compared with wild-type hearts. The antiapoptotic factor Bcl-x/(L) was localized in regions adjacent to where apoptosis was detected. The distribution patterns of the apoptosis triggering protein p53 were similar to those of apoptotic cells. The growth of the prenatal and postnatal hearts of the cardiac alpha-actin-deficient mice was retarded, and the cytoplasmic filaments were disorganized. Although apoptotic cells were observed in both the atria and ventricles in the hearts of the homozygous null animals, the frequency was greater in the ventricles than in the atria. Our results indicate that the functional and structural disturbances in the mice with a homozygous lack of cardiac alpha-actin seem to be due to disorganized development of acto-myosin filaments in the affected cardiomyocytes. Other actin isoforms cannot compensate for the lack of cardiac alpha-actin, and this seems to induce apoptosis in defective cardiac myocytes, which are not able to cope with the increased workload in the perinatal phase.


Subject(s)
Actins/genetics , Apoptosis , Heart Defects, Congenital/pathology , Myocardium/pathology , Actin Cytoskeleton/pathology , Animals , Cause of Death , Female , Heart/embryology , Heart Defects, Congenital/mortality , Male , Mice , Mice, Mutant Strains , Pregnancy
13.
Endocrinology ; 143(11): 4477-82, 2002 Nov.
Article in English | MEDLINE | ID: mdl-12399445

ABSTRACT

We have studied male sexual differentiation of null mutant mice (-/-) for the thyroid-specific enhancer-binding protein (T/ebp or Nkx2.1) gene, a homeodomain transcription factor that plays a role in organogenesis of the thyroid, lung, ventral forebrain, and pituitary gland. Because the T/ebp/Nkx2.1 (-/-) mice do not develop the pituitary gland, their sexual differentiation, if any, must occur in the absence of action of gonadotropins and other pituitary hormones. The (-/-) mice survive only until birth (embryonic d 19-19.5 of pregnancy), and when their external and internal genitals were inspected at embryonic d 18.5, they were indistinguishable from the (+/-) and (+/+) control mice. The testis weights of (-/-) mice were 20% lower than in (+/+) and (+/-) mice. The testosterone content of the (-/-) testes (13.5 +/- 2.4 pg/gonad, mean +/- SEM, n = 11) was dramatically reduced, compared with (+/-) (165 +/- 22.5 pg, n = 14) and (+/+) (234 +/- 37.3 pg, n = 10) littermates. Light microscopy revealed no difference in seminiferous tubules, interstitial tissue, or relative proportions of the two-cell compartments between the (-/-) and (+/+) testes. However, electron microscopy confirmed that Leydig cells in the (-/-) testes were much smaller, with smaller mitochondria and proportion of smooth endoplasmic reticulum than found in the controls, which was in support of the low androgen content of the knockout testes. In conclusion, this study on T/ebp/Nkx2.1 knockout mice, devoid of the pituitary gland, demonstrates that pituitary hormone secretion is not needed for stimulation of sufficient fetal testicular androgen synthesis to induce male sexual differentiation. The endogenous testosterone level in the null mutant testes is 5-10% of the control level, which suggests that there is a considerable safety margin in the amount of testosterone that is needed for the male fetal masculinization.


Subject(s)
Mutation , Nuclear Proteins/genetics , Nuclear Proteins/physiology , Phenotype , Pituitary Hormones/physiology , Sex Differentiation , Transcription Factors/genetics , Transcription Factors/physiology , Animals , Endoplasmic Reticulum, Smooth/ultrastructure , Gestational Age , Leydig Cells/ultrastructure , Male , Mice , Mice, Knockout , Microscopy, Electron , Mitochondria/ultrastructure , Organ Size , Testis/chemistry , Testis/embryology , Testosterone/analysis , Thyroid Nuclear Factor 1
14.
Microsc Res Tech ; 58(5): 395-403, 2002 Sep 01.
Article in English | MEDLINE | ID: mdl-12226809

ABSTRACT

The transformation of the endocardial cushion into valves and septa is a critical step in cardiac morphogenesis as it initiates the development of the four-chambered heart. This transformation results from a region-specific balance between cellular proliferation, apoptosis, and differentiation. The development of the form and structure of the endocardial cushion is accompanied by precise patterns of abundant cell death having the morphological features of programmed cell death (apoptosis), which plays an important role in the elimination of redundant cells and in changes of phenotypic composition during histogenesis. Apoptosis is an essential process in morphogenesis as it balances mitosis in renewing tissues. It is controlled by one or more genetic programs that kill the targeted cell. However, the causes, role, and regulation of apoptosis in the developing endocardial cushion still remain to be determined. The clarification of the role of the apoptosis regulatory genes constitutes a major task in future studies of cell death in the developing heart. This new molecular histology of heart development awaits further experiments to clarify the interactive mechanisms that act to ensure the sculpting of the endocardial cushion into valves and septa by determining the size of the cushion cell populations. The relation between the expression of different factors and the modifications of the cushion region during cardiac development are reviewed. In addition, we review and summarize information on molecules identified in our experiments that imply the activity of a number of essential genes coinciding with the key steps in generating the overall architecture of the heart. We correlate their temporal and spatial expression with their proposed roles.


Subject(s)
Endocardium/embryology , Heart/embryology , Apoptosis , Cell Death , Cell Differentiation , Heart Septum/embryology , Heart Valves/embryology , Immunohistochemistry
15.
Endocrinology ; 143(10): 4084-95, 2002 Oct.
Article in English | MEDLINE | ID: mdl-12239120

ABSTRACT

To assess the consequences of prolonged exposure to elevated levels of LH/human chorionic gonadotropin (hCG) in the female, we developed a transgenic (TG) mouse model (hCGbeta+) that overexpresses the hCGbeta-subunit cDNA. Because of the promoter used, ubiquitin C, the transgene is expressed in multiple tissues, including the pituitary gland, in which coupling with the endogenous common alpha-subunit results in synthesis of high levels of bioactive hCG. The TG females presented with precocious puberty, infertility, enhanced ovarian steroidogenesis, and abnormal uterine structure. Pituitary enlargement was evident from the age of 2 months, which progressed to adenomas by the age of 10-12 months. Immunohistochemical studies and electron microscopy demonstrated lactotrope origin for the adenomas, associated with severe hyperprolactinemia. The mammary glands of TG females showed marked lobuloalveolar development followed by mammary tumors with characteristics of adenocarcinoma at the age of 9-12 months. More than 90% of penetrance and high frequency of metastasis (47%) was observed. Formation of the pituitary and mammary gland tumors was totally abolished by ovariectomy despite persistently elevated hCG levels. Taken together, these findings suggest that the hCG-induced aberrations of ovarian function are clearly responsible for the extragonadal tumors observed in these TG mice.


Subject(s)
Adenoma/chemically induced , Chorionic Gonadotropin, beta Subunit, Human , Infertility/chemically induced , Mammary Neoplasms, Experimental/chemically induced , Pituitary Gland, Anterior , Pituitary Neoplasms/chemically induced , Prolactin/metabolism , Adenoma/metabolism , Adenoma/pathology , Animals , Chorionic Gonadotropin, beta Subunit, Human/administration & dosage , Chorionic Gonadotropin, beta Subunit, Human/genetics , Dose-Response Relationship, Drug , Female , Genitalia, Female/pathology , Genitalia, Female/physiopathology , Hormones/biosynthesis , Humans , Hyperprolactinemia/chemically induced , Infertility/pathology , Infertility/physiopathology , Mammary Neoplasms, Experimental/pathology , Mice , Mice, Transgenic/genetics , Obesity/chemically induced , Ovary/metabolism , Pituitary Gland, Anterior/pathology , Pituitary Neoplasms/metabolism , Pituitary Neoplasms/pathology
16.
Curr Eye Res ; 24(6): 439-50, 2002 Jun.
Article in English | MEDLINE | ID: mdl-12525971

ABSTRACT

PURPOSE: Molecular genetic analyses have clearly associated vitreoretinal degeneration with mutations in the type II collagen gene, but lack of experimental models has prevented systematic analyses of the occurrence of phenotypic changes and of the pathogenetic mechanisms involved. The present study is a detailed morphological and ultrastructural analysis of the vitreoretinal consequences of a small deletion mutation in the type II collagen gene. METHODS: The eyes of Del1 mice carrying six copies of pro alpha1(II) collagen transgene with a small deletion mutation were analyzed during embryonic development, postnatal growth and aging using their nontransgenic littermates as controls. Tissue samples were processed for light and electron microscopy for morphological and ultrastructural analyses. Transcription of pro alpha1(II) collagen gene was localized by in situ hybridization, and type II collagen was detected by immunohistochemistry. RESULTS: In this mouse model most components of the eye are ultrastructurally unaltered. However, the transgenes caused a dose-dependent dominant negative effect seen as a reduced number of type II collagen fibrils in the vitreous. In concert with this, dose-dependent accumulation of amorphous material was observed in the dilated rough endoplasmic reticulum of cells responsible for the production of type II collagen molecules. In mice homozygous for the transgene locus, the vitreoretinal degenerative lesions appeared already during late embryonic development. In mice heterozygous for the locus, such changes were milder and appeared only during postnatal growth and progressed gradually upon aging. CONCLUSIONS: The observed ultrastructural changes suggest that defective structure and function of collagen fibrils in Del1 mice result from a partial block in the post-translational processing and secretion of the mutated procollagen chains, and partly from secretion of mutated procollagen molecules which interfere with normal fibrillogenesis.


Subject(s)
Collagen Type II/genetics , Gene Deletion , Retina/ultrastructure , Vitreous Body/ultrastructure , Aging/metabolism , Animals , Embryo, Mammalian/metabolism , Embryo, Mammalian/pathology , Embryonic and Fetal Development , Eye/embryology , Mice , Mice, Knockout/genetics , Mice, Transgenic/genetics , RNA, Messenger/metabolism , Retina/pathology , Tissue Distribution , Vitreous Body/metabolism , Vitreous Body/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...