Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Nature ; 603(7901): 497-502, 2022 03.
Article in English | MEDLINE | ID: mdl-35236989

ABSTRACT

Discrimination of self from non-self is fundamental to a wide range of immunological processes1. During pregnancy, the mother does not recognize the placenta as immunologically foreign because antigens expressed by trophoblasts, the placental cells that interface with the maternal immune system, do not activate maternal T cells2. Currently, these activation defects are thought to reflect suppression by regulatory T cells3. By contrast, mechanisms of B cell tolerance to trophoblast antigens have not been identified. Here we provide evidence that glycan-mediated B cell suppression has a key role in establishing fetomaternal tolerance in mice. B cells specific for a model trophoblast antigen are strongly suppressed through CD22-LYN inhibitory signalling, which in turn implicates the sialylated glycans of the antigen as key suppressive determinants. Moreover, B cells mediate the MHC-class-II-restricted presentation of antigens to CD4+ T cells, which leads to T cell suppression, and trophoblast-derived sialoglycoproteins are released into the maternal circulation during pregnancy in mice and humans. How protein glycosylation promotes non-immunogenic placental self-recognition may have relevance to immune-mediated pregnancy complications and to tumour immune evasion. We also anticipate that our findings will bolster efforts to harness glycan biology to control antigen-specific immune responses in autoimmune disease.


Subject(s)
Antigens , Placenta , Trophoblasts , Animals , Autoimmune Diseases , B-Lymphocytes , Female , Immune Tolerance , Mice , Placenta/immunology , Polysaccharides/metabolism , Pregnancy/immunology
3.
Curr Opin Immunol ; 13(5): 590-3, 2001 Oct.
Article in English | MEDLINE | ID: mdl-11544009

ABSTRACT

The long-standing question of how the fetal allograft avoids immune rejection during pregnancy has lately been generating renewed interest. Recent insights have emerged from studies in mice on uterine NK cells, NKT cells, complement inhibition and the reproductive effects of 1-methyl-tryptophan.


Subject(s)
Fetus/immunology , Graft Rejection/immunology , Abortion, Spontaneous/immunology , Animals , Female , Humans , Pregnancy
4.
Development ; 126(19): 4267-79, 1999 Oct.
Article in English | MEDLINE | ID: mdl-10477295

ABSTRACT

Transforming growth factor-beta (TGF-beta) is abundant in bone matrix and has been shown to regulate the activity of osteoblasts and osteoclasts in vitro. To explore the role of endogenous TGF-(beta) in osteoblast function in vivo, we have inhibited osteoblastic responsiveness to TGF-beta in transgenic mice by expressing a cytoplasmically truncated type II TGF-beta receptor from the osteocalcin promoter. These transgenic mice develop an age-dependent increase in trabecular bone mass, which progresses up to the age of 6 months, due to an imbalance between bone formation and resorption during bone remodeling. Since the rate of osteoblastic bone formation was not altered, their increased trabecular bone mass is likely due to decreased bone resorption by osteoclasts. Accordingly, direct evidence of reduced osteoclast activity was found in transgenic mouse skulls, which had less cavitation and fewer mature osteoclasts relative to skulls of wild-type mice. These bone remodeling defects resulted in altered biomechanical properties. The femurs of transgenic mice were tougher, and their vertebral bodies were stiffer and stronger than those of wild-type mice. Lastly, osteocyte density was decreased in transgenic mice, suggesting that TGF-beta signaling in osteoblasts is required for normal osteoblast differentiation in vivo. Our results demonstrate that endogenous TGF-beta acts directly on osteoblasts to regulate bone remodeling, structure and biomechanical properties.


Subject(s)
Bone Remodeling , Osteoblasts/metabolism , Receptors, Transforming Growth Factor beta/metabolism , Signal Transduction , Age Factors , Animals , Bone Density/physiology , Bone Development/genetics , Bone and Bones/metabolism , Female , Femur/anatomy & histology , Femur/diagnostic imaging , Genes, Dominant , Kinetics , Male , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Mice, Transgenic , Osteocalcin/genetics , Ovary/physiology , Promoter Regions, Genetic , Radiography , Receptors, Transforming Growth Factor beta/physiology , Skull/diagnostic imaging , Skull/growth & development
5.
Mol Biol Cell ; 9(7): 1903-18, 1998 Jul.
Article in English | MEDLINE | ID: mdl-9658179

ABSTRACT

Bone remodeling depends on the spatial and temporal coupling of bone formation by osteoblasts and bone resorption by osteoclasts; however, the molecular basis of these inductive interactions is unknown. We have previously shown that osteoblastic overexpression of TGF-beta2 in transgenic mice deregulates bone remodeling and leads to an age-dependent loss of bone mass that resembles high-turnover osteoporosis in humans. This phenotype implicates TGF-beta2 as a physiological regulator of bone remodeling and raises the question of how this single secreted factor regulates the functions of osteoblasts and osteoclasts and coordinates their opposing activities in vivo. To gain insight into the physiological role of TGF-beta in bone remodeling, we have now characterized the responses of osteoblasts to TGF-beta in these transgenic mice. We took advantage of the ability of alendronate to specifically inhibit bone resorption, the lack of osteoclast activity in c-fos-/- mice, and a new transgenic mouse line that expresses a dominant-negative form of the type II TGF-beta receptor in osteoblasts. Our results show that TGF-beta directly increases the steady-state rate of osteoblastic differentiation from osteoprogenitor cell to terminally differentiated osteocyte and thereby increases the final density of osteocytes embedded within bone matrix. Mice overexpressing TGF-beta2 also have increased rates of bone matrix formation; however, this activity does not result from a direct effect of TGF-beta on osteoblasts, but is more likely a homeostatic response to the increase in bone resorption caused by TGF-beta. Lastly, we find that osteoclastic activity contributes to the TGF-beta-induced increase in osteoblast differentiation at sites of bone resorption. These results suggest that TGF-beta is a physiological regulator of osteoblast differentiation and acts as a central component of the coupling of bone formation to resorption during bone remodeling.


Subject(s)
Bone Remodeling/physiology , Osteoblasts/physiology , Transforming Growth Factor beta/physiology , Alendronate/pharmacology , Animals , Bone Remodeling/drug effects , Bone Resorption/genetics , Cell Count/drug effects , Cell Differentiation/drug effects , Femur , Gene Expression Regulation/drug effects , Kinetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Minerals/metabolism , Osteoblasts/drug effects , Osteoclasts/cytology , Osteocytes/cytology , Periosteum/metabolism , Phenotype , Receptors, Transforming Growth Factor beta/physiology , Transforming Growth Factor beta/blood , Transforming Growth Factor beta/genetics
6.
J Cell Biol ; 132(1-2): 195-210, 1996 Jan.
Article in English | MEDLINE | ID: mdl-8567723

ABSTRACT

The development of the skeleton requires the coordinated activities of bone-forming osteoblasts and bone-resorbing osteoclasts. The activities of these two cell types are likely to be regulated by TGF-beta, which is abundant in bone matrix. We have used transgenic mice to evaluate the role of TGF-beta 2 in bone development and turnover. Osteoblast-specific overexpression of TGF-beta 2 from the osteocalcin promoter resulted in progressive bone loss associated with increases in osteoblastic matrix deposition and osteoclastic bone resorption. This phenotype closely resembles the bone abnormalities seen in human hyperparathyroidism and osteoporosis. Furthermore, a high level of TGF-beta 2 overexpression resulted in defective bone mineralization and severe hypoplasia of the clavicles, a hallmark of the developmental disease cleidocranial dysplasia. Our results suggest that TGF-beta 2 functions as a local positive regulator of bone remodeling and that alterations in TGF-beta 2 synthesis by bone cells, or in their responsiveness to TGF-beta 2, may contribute to the pathogenesis of metabolic bone disease.


Subject(s)
Osteoblasts/metabolism , Osteoporosis/genetics , Transforming Growth Factor beta/biosynthesis , Age Factors , Animals , Base Sequence , Bone Remodeling , Bone Resorption , Bone and Bones/embryology , Bone and Bones/pathology , Calcification, Physiologic , Clavicle/embryology , Clavicle/pathology , Immunohistochemistry , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Mice, Transgenic , Molecular Sequence Data , Osteocalcin/genetics , Osteoclasts , Osteoporosis/etiology , Phenotype , Promoter Regions, Genetic , Recombinant Fusion Proteins/biosynthesis , Transforming Growth Factor beta/genetics
8.
Dev Genet ; 12(3): 206-11, 1991.
Article in English | MEDLINE | ID: mdl-1678322

ABSTRACT

H2.0, a homeobox gene identified by homology to the Sex combs reduced homeobox of Drosophila, is expressed in all the cellular precursors of the visceral musculature. By analogy to the essential function of most other known homeobox genes in determining the fate of cells where they are expressed, we hypothesized that mutation of H2.0 would disrupt gut muscle development. In this paper, we show that a small deletion, which eliminates H2.0, has no detectable effect on normal gut morphogenesis, visceral muscle actin organization, or larval peristalsis.


Subject(s)
Drosophila/genetics , Genes, Homeobox , Mesoderm/metabolism , Muscles/cytology , Animals , Chromosome Deletion , Drosophila/embryology , Gene Expression , Homozygote , Morphogenesis/genetics , Phenotype
SELECTION OF CITATIONS
SEARCH DETAIL
...