Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
1.
Nature ; 586(7828): 281-286, 2020 10.
Article in English | MEDLINE | ID: mdl-32968276

ABSTRACT

'Dysbiosis' of the maternal gut microbiome, in response to challenges such as infection1, altered diet2 and stress3 during pregnancy, has been increasingly associated with abnormalities in brain function and behaviour of the offspring4. However, it is unclear whether the maternal gut microbiome influences neurodevelopment during critical prenatal periods and in the absence of environmental challenges. Here we investigate how depletion and selective reconstitution of the maternal gut microbiome influences fetal neurodevelopment in mice. Embryos from antibiotic-treated and germ-free dams exhibited reduced brain expression of genes related to axonogenesis, deficient thalamocortical axons and impaired outgrowth of thalamic axons in response to cell-extrinsic factors. Gnotobiotic colonization of microbiome-depleted dams with a limited consortium of bacteria prevented abnormalities in fetal brain gene expression and thalamocortical axonogenesis. Metabolomic profiling revealed that the maternal microbiome regulates numerous small molecules in the maternal serum and the brains of fetal offspring. Select microbiota-dependent metabolites promoted axon outgrowth from fetal thalamic explants. Moreover, maternal supplementation with these metabolites abrogated deficiencies in fetal thalamocortical axons. Manipulation of the maternal microbiome and microbial metabolites during pregnancy yielded adult offspring with altered tactile sensitivity in two aversive somatosensory behavioural tasks, but no overt differences in many other sensorimotor behaviours. Together, our findings show that the maternal gut microbiome promotes fetal thalamocortical axonogenesis, probably through signalling by microbially modulated metabolites to neurons in the developing brain.


Subject(s)
Brain/embryology , Brain/metabolism , Dysbiosis/microbiology , Fetus/embryology , Fetus/metabolism , Gastrointestinal Microbiome/physiology , Mothers , Animals , Axons/metabolism , Brain/cytology , Cerebral Cortex/cytology , Cerebral Cortex/embryology , Cerebral Cortex/metabolism , Computer Simulation , Dysbiosis/blood , Dysbiosis/pathology , Female , Fetus/cytology , Male , Mice , Mice, Inbred C57BL , Pregnancy , Pregnancy Complications/blood , Pregnancy Complications/microbiology , Pregnancy Complications/pathology , Principal Component Analysis , Thalamus/cytology , Thalamus/embryology , Thalamus/metabolism
2.
Am J Pathol ; 188(10): 2318-2327, 2018 10.
Article in English | MEDLINE | ID: mdl-30059656

ABSTRACT

Medication-related osteonecrosis of the jaw (MRONJ) is a rare but detrimental intraoral lesion that predominantly occurs in patients with long-term use of antiresorptive agents, such as bisphosphonate and denosumab, a human anti-receptor activator of NF-κB ligand (RANKL) monoclonal antibody (Ab). Surgical intervention, such as tooth extraction, is a known risk factor for MRONJ, which is often performed to eliminate preexiting pathologic inflammatory conditions, such as periodontal diseases. Nonetheless, it remains unknown whether pre-existing periodontal disease condition exacerbates, or removal of such condition ameliorates, MRONJ development after tooth extraction. In this study, we combined the ligature-induced periodontitis and the tooth extraction mouse models under the administration of zoledronic acid (ZOL) or anti-RANKL Ab, and provide experimental evidence that a pre-existing pathologic inflammatory condition exacerbates MRONJ development after tooth extraction in mice. Under ZOL administration, tooth extraction alone induced ONJ lesions; however, extraction of a ligature-placed tooth further exacerbated ONJ development. When the ligature was removed and the inflammatory condition was deescalated, ONJ development was ameliorated. Anti-RANKL Ab administration resulted in similar outcomes. Interestingly, unlike ZOL-administered mice, anti-RANKL Ab-administered mice exhibited complete absence of osteoclasts, suggesting that physical presence of osteoclasts is not directly involved in ONJ development. Collectively, our study demonstrated that periodontal disease is a functionally linked risk factor that predisposes ONJ development after tooth extraction in the presence of bisphosphonate and denosumab.


Subject(s)
Jaw Diseases/prevention & control , Osteonecrosis/prevention & control , Periodontitis/therapy , Tooth Extraction , Animals , Bisphosphonate-Associated Osteonecrosis of the Jaw/prevention & control , Bone Density Conservation Agents/toxicity , Denosumab/toxicity , Disease Models, Animal , Female , Jaw Diseases/chemically induced , Ligation , Mice, Inbred C57BL , Osteonecrosis/chemically induced
3.
Am J Pathol ; 184(11): 3084-93, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25173134

ABSTRACT

Drug-induced osteonecrosis of the jaw (ONJ) is a detrimental intraoral lesion that often occurs after dental-related interventions in patients undergoing treatment with bisphosphonates or denosumab, the neutralizing human anti-receptor activator of NF-κB ligand (RANKL) antibody (Ab). The cause of ONJ by these drugs has been speculated to their direct effects on osteoclasts. However, the extent to which osteoclasts contribute to ONJ pathogenesis remains controversial. Herein, by using a tooth-extraction mouse model with i.v. administration of mouse anti-RANKL Ab or the bisphosphonate zoledronate (ZOL), we show that unresorbed bone due to impaired formation or suppressed functions of osteoclasts, respectively, is associated with ONJ development. After tooth extraction, ONJ-like lesions developed 50% in the anti-RANKL Ab-treated mice and 30% in the ZOL-treated mice. Nonviable and unresorbed bone was found more in anti-RANKL Ab-treated mice compared with mice receiving ZOL. All mice receiving anti-RANKL Ab had an undetectable tartrate-resistant acid phosphatase (TRAP) level in the serum and no TRAP-positive osteoclasts at the extracted sockets, whereas ZOL-treated mice had a decreased TRAP level without altering the numbers of TRAP-positive osteoclasts. Interestingly, the absence of newly formed woven bone in the extracted sockets was evident in ONJ-like lesions from both anti-RANKL Ab- and ZOL-treated mice. Our study suggests that the lack of osteoclasts' bone-resorptive functions by these drugs and suppression of woven bone formation after dental trauma may be associated with ONJ development.


Subject(s)
Bisphosphonate-Associated Osteonecrosis of the Jaw/pathology , Bone Resorption/pathology , Osteoclasts/pathology , RANK Ligand/antagonists & inhibitors , Animals , Antibodies, Monoclonal, Humanized , Denosumab , Diphosphonates , Disease Models, Animal , Imidazoles , Mice , Osteoclasts/drug effects , Zoledronic Acid
4.
Sci Adv ; 9(40): eadk1887, 2023 10 06.
Article in English | MEDLINE | ID: mdl-37801498

ABSTRACT

The maternal microbiome is an important regulator of gestational health, but how it affects the placenta as the interface between mother and fetus remains unexplored. Here, we show that the maternal gut microbiota supports placental development in mice. Depletion of the maternal gut microbiota restricts placental growth and impairs feto-placental vascularization. The maternal gut microbiota modulates metabolites in the maternal and fetal circulation. Short-chain fatty acids (SCFAs) stimulate cultured endothelial cell tube formation and prevent abnormalities in placental vascularization in microbiota-deficient mice. Furthermore, in a model of maternal malnutrition, gestational supplementation with SCFAs prevents placental growth restriction and vascular insufficiency. These findings highlight the importance of host-microbial symbioses during pregnancy and reveal that the maternal gut microbiome promotes placental growth and vascularization in mice.


Subject(s)
Gastrointestinal Microbiome , Microbiota , Pregnancy , Mice , Female , Animals , Placentation , Placenta/metabolism , Fetus
5.
bioRxiv ; 2023 Feb 15.
Article in English | MEDLINE | ID: mdl-36824779

ABSTRACT

The maternal microbiome is an important regulator of gestational health, but how it impacts the placenta as the interface between mother and fetus remains unexplored. Here we show that the maternal gut microbiota supports placental development in mice. Depletion of the maternal gut microbiota restricts placental growth and impairs feto-placental vascularization. The maternal gut microbiota modulates metabolites in the maternal and fetal circulation. Short-chain fatty acids (SCFAs) stimulate angiogenesis-related tube formation by endothelial cells and prevent abnormalities in placental vascularization in microbiota-deficient mice. Furthermore, in a model of maternal malnutrition, gestational supplementation with SCFAs prevents placental growth restriction and vascular insufficiency. These findings highlight the importance of host-microbial symbioses during pregnancy and reveal that the maternal gut microbiome promotes placental growth and vascularization in mice.

6.
Sci Immunol ; 7(72): eabp8632, 2022 06 24.
Article in English | MEDLINE | ID: mdl-35714199

ABSTRACT

In this issue of Science Immunology, Barreto de Albuquerque et al. track immune responsiveness to the foodborne pathogen Listeria monocytogenes during oral infection. Their findings extend the notion of compartmentalized immunity within the gastrointestinal tract to the oral cavity and provide previously unkown insights into regional specialization of oral immunity.


Subject(s)
Listeria monocytogenes , Listeriosis , Gastrointestinal Tract , Humans , Immunity, Mucosal , Mouth Mucosa
7.
Int J Oral Sci ; 14(1): 16, 2022 03 21.
Article in English | MEDLINE | ID: mdl-35307731

ABSTRACT

Bacterial infection is a common finding in patients, who develop medication-related osteonecrosis of the jaw (MRONJ) by the long-term and/or high-dose use of anti-resorptive agents such as bisphosphonate (BPs). However, pathological role of bacteria in MRONJ development at the early stage remains controversial. Here, we demonstrated that commensal microbiota protects against MRONJ development in the pulp-exposed periapical periodontitis mouse model. C57/BL6 female mice were treated with intragastric broad-spectrum antibiotics for 1 week. Zoledronic acid (ZOL) through intravenous injection and antibiotics in drinking water were administered for throughout the experiment. Pulp was exposed on the left maxillary first molar, then the mice were left for 5 weeks after which bilateral maxillary first molar was extracted and mice were left for additional 3 weeks to heal. All mice were harvested, and cecum, maxilla, and femurs were collected. ONJ development was assessed using µCT and histologic analyses. When antibiotic was treated in mice, these mice had no weight changes, but developed significantly enlarged ceca compared to the control group (CTL mice). Periapical bone resorption prior to the tooth extraction was similarly prevented when treated with antibiotics, which was confirmed by decreased osteoclasts and inflammation. ZOL treatment with pulp exposure significantly increased bone necrosis as determined by empty lacunae and necrotic bone amount. Furthermore, antibiotics treatment could further exacerbate bone necrosis, with increased osteoclast number. Our findings suggest that the commensal microbiome may play protective role, rather than pathological role, in the early stages of MRONJ development.


Subject(s)
Bisphosphonate-Associated Osteonecrosis of the Jaw , Bone Density Conservation Agents , Microbiota , Periapical Diseases , Animals , Bisphosphonate-Associated Osteonecrosis of the Jaw/pathology , Bisphosphonate-Associated Osteonecrosis of the Jaw/prevention & control , Diphosphonates , Female , Humans , Mice , Zoledronic Acid
8.
Science ; 373(6561): eabi8835, 2021 Sep 17.
Article in English | MEDLINE | ID: mdl-34529475

ABSTRACT

Puel and Casanova and Kisand et al. challenge our conclusions that interferonopathy and not IL-17/IL-22 autoantibodies promote candidiasis in autoimmune polyendocrinopathy­candidiasis­ectodermal dystrophy. We acknowledge that conclusive evidence for causation is difficult to obtain in complex human diseases. However, our studies clearly document interferonopathy driving mucosal candidiasis with intact IL-17/IL-22 responses in Aire-deficient mice, with strong corroborative evidence in patients.


Subject(s)
Immunity, Mucosal , Mycoses , Humans , Mucous Membrane , Animals , Mice
9.
Science ; 371(6526)2021 01 15.
Article in English | MEDLINE | ID: mdl-33446526

ABSTRACT

Human monogenic disorders have revealed the critical contribution of type 17 responses in mucosal fungal surveillance. We unexpectedly found that in certain settings, enhanced type 1 immunity rather than defective type 17 responses can promote mucosal fungal infection susceptibility. Notably, in mice and humans with AIRE deficiency, an autoimmune disease characterized by selective susceptibility to mucosal but not systemic fungal infection, mucosal type 17 responses are intact while type 1 responses are exacerbated. These responses promote aberrant interferon-γ (IFN-γ)- and signal transducer and activator of transcription 1 (STAT1)-dependent epithelial barrier defects as well as mucosal fungal infection susceptibility. Concordantly, genetic and pharmacologic inhibition of IFN-γ or Janus kinase (JAK)-STAT signaling ameliorates mucosal fungal disease. Thus, we identify aberrant T cell-dependent, type 1 mucosal inflammation as a critical tissue-specific pathogenic mechanism that promotes mucosal fungal infection susceptibility in mice and humans.


Subject(s)
Candida albicans/immunology , Candidiasis, Chronic Mucocutaneous/genetics , Candidiasis, Chronic Mucocutaneous/immunology , Immunity, Mucosal/immunology , Polyendocrinopathies, Autoimmune/genetics , Polyendocrinopathies, Autoimmune/immunology , Adolescent , Adult , Aged , Animals , Disease Models, Animal , Female , Humans , Immunity, Mucosal/genetics , Immunologic Surveillance/genetics , Immunologic Surveillance/immunology , Interferon-gamma/genetics , Interleukins/genetics , Janus Kinases/genetics , Male , Mice , Mice, Inbred BALB C , Middle Aged , Mouth Mucosa/immunology , Mouth Mucosa/pathology , Receptors, Interleukin-17/genetics , STAT1 Transcription Factor/genetics , T-Lymphocytes/immunology , Young Adult , Interleukin-22
10.
J Bone Miner Res ; 32(2): 309-318, 2017 Feb.
Article in English | MEDLINE | ID: mdl-27567012

ABSTRACT

Long-term administration of nitrogen-containing bisphosphonates can induce detrimental side effects such as bisphosphonate-related osteonecrosis of the jaw (BRONJ) in human. Although inflammation is known to be associated with BRONJ development, the detailed underlying mechanism remains unknown. Here, we report that the pro-inflammatory cytokine IL-36α is, in part, responsible for the BRONJ development. We found a notably higher level of IL-36α and lower level of collagen in the BRONJ lesions in mice. We also found that IL-36α remarkably suppressed TGF-ß-mediated expression of Collα1 and α-Sma via the activation of Erk signaling pathway in mouse gingival mesenchymal stem cells. When IL-36 signaling was abrogated in vivo, development of BRONJ lesions was ameliorated in mice. Taken together, we showed the pathologic role of IL-36α in BRONJ development by inhibiting collagen expression and demonstrated that IL-36α could be a potential marker and a therapeutic target for the prevention and treatment of BRONJ. © 2016 American Society for Bone and Mineral Research.


Subject(s)
Bisphosphonate-Associated Osteonecrosis of the Jaw/pathology , Collagen/metabolism , Interleukin-1/adverse effects , Transforming Growth Factor beta/pharmacology , Animals , Antibodies, Neutralizing/pharmacology , Antibodies, Neutralizing/therapeutic use , Bisphosphonate-Associated Osteonecrosis of the Jaw/drug therapy , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , Gene Expression Profiling , MAP Kinase Signaling System/drug effects , Mice, Inbred C57BL , Models, Biological , Phosphorylation/drug effects , Protein Transport/drug effects , Smad Proteins/metabolism
11.
Aging Cell ; 14(5): 838-46, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26138448

ABSTRACT

p53, the guardian of the genome, is a tumor suppressor protein and critical for the genomic integrity of the cells. Many studies have shown that intracellular level of p53 is enhanced during replicative senescence in normal fibroblasts, and the enhanced level of p53 is viewed as the cause of senescence. Here, we report that, unlike in normal fibroblasts, the level of intracellular p53 reduces during replicative senescence and oncogene-induced senescence (OIS) in normal human keratinocytes (NHKs). We found that the intracellular p53 level was also decreased in age-dependent manner in normal human epithelial tissues. Senescent NHKs exhibited an enhanced level of p16(INK4A) , induced G2 cell cycle arrest, and lowered the p53 expression and transactivation activity. We found that low level of p53 in senescent NHKs was due to reduced transcription of p53. The methylation status at the p53 promoter was not altered during senescence, but senescent NHKs exhibited notably lower level of acetylated histone 3 (H3) at the p53 promoter in comparison with rapidly proliferating cells. Moreover, p53 knockdown in rapidly proliferating NHKs resulted in the disruption of fidelity in repaired DNA. Taken together, our study demonstrates that p53 level is diminished during replicative senescence and OIS and that such diminution is associated with H3 deacetylation at the p53 promoter. The reduced intracellular p53 level in keratinocytes of the elderly could be a contributing factor for more frequent development of epithelial cancer in the elderly because of the loss of genomic integrity of cells.


Subject(s)
Cellular Senescence , Keratinocytes/cytology , Keratinocytes/metabolism , Tumor Suppressor Protein p53/metabolism , Cell Proliferation , Cells, Cultured , DNA Methylation , Histones/metabolism , Humans , Tumor Suppressor Protein p53/deficiency , Tumor Suppressor Protein p53/genetics
12.
J Endod ; 39(9): 1156-60, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23953290

ABSTRACT

INTRODUCTION: Cell migration is an important step in pulpal wound healing. Although components in the resin-based dental materials are known to have adverse effects on pulp wound healing including proliferation and mineralization, their effects on cell migration have been scarcely examined. Here, we investigated the effects of 2-hydroxyethyl methacrylate (HEMA) on the migration of dental pulp stem cells (DPSC) in vitro. METHODS: Cell viability was assessed using the MTT (3-(4, 5-dimethylthiazolyl-2)-2, 5-diphenyltetrazolium bromide) assay, and cell migration was evaluated using the wound scratch assay and transwell migration assay at noncytotoxic doses. The Western blot was used to examine pathways associated with migration such as focal adhesion kinase, mitogen-activated protein kinase, and glycogen synthase kinase 3. RESULTS: There were no drastic changes in the cell viability below 3 mmol/L HEMA. When DPSCs were treated with HEMA at 0.5, 1.0, and 2.5 mmol/L, cell migration was diminished. HEMA-treated DPSCs exhibited the loss of phosphorylated focal adhesion kinase in a dose-dependent manner. The HEMA-mediated inhibition of cell migration was associated with phosphorylation of p38 but not glycogen synthase kinase 3, Extracellular signal-related kinase (ERK), or c-Jun N-terminal kinase (JNK) pathways. When we inhibited the p38 signaling pathway using a p38 inhibitor, the migration of DPSCs was suppressed. CONCLUSIONS: HEMA inhibits the migration of dental pulp cells in vitro, suggesting that poor pulpal wound healing under resin-based dental materials may be caused, in part, by the inhibition of cell migration by HEMA.


Subject(s)
Dental Materials/pharmacology , Dental Pulp/drug effects , Methacrylates/pharmacology , Stem Cells/drug effects , Blotting, Western , Cell Culture Techniques , Cell Movement/drug effects , Cell Survival/drug effects , Coloring Agents , Dental Pulp/cytology , Dose-Response Relationship, Drug , Enzyme Inhibitors/pharmacology , Focal Adhesion Kinase 1 , Glycogen Synthase Kinase 3/drug effects , Humans , Imidazoles/pharmacology , MAP Kinase Signaling System/drug effects , Mitogen-Activated Protein Kinases/drug effects , Tetrazolium Salts , Thiazoles , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , p38 Mitogen-Activated Protein Kinases/drug effects
13.
J Endod ; 39(1): 57-61, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23228258

ABSTRACT

INTRODUCTION: Camphorquinone (CQ) is a photoinitiator that triggers polymerization of light-curing materials such as dental adhesives and composites. CQ does not become a part of the polymer network, suggesting that CQ can be leached out into surrounding environment including dental pulp and exert adversary effects on tissues. In order to understand the mechanisms of CQ-induced side effects, we investigated the effect of CQ on cell viability, cytokine secretion, and odontogenic differentiation of dental pulp stem cells in vitro. METHODS: Cell viability was assessed using the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay after CQ exposure. Western blotting was performed for p16(INK4A), p21(WAF1), and p53. Secretory cytokines were evaluated using the membrane-enzyme-linked immunosorbent assay as well as conventional and quantitative reverse-transcription polymerase chain reaction. The effects of CQ on odontogenic differentiation were evaluated using alkaline phosphatase and alizarin red S staining methods. RESULTS: CQ treatment suppressed the proliferation of DPSCs and induced the expression of p16(INK4A), p21(WAF1), and p53. Levels of proinflammatory cytokines (eg, interleukin 6, interleukin 8, and matrix metalloproteinase-3 [MMP3]) were increased by CQ treatment. CQ also inhibited odontogenic differentiation and mineralization capacities of DPSC and MC3T3-E1 cells. CONCLUSIONS: Our study showed that CQ may trigger pulpal inflammation by inducing proinflammatory cytokine production from the pulpal cells and may impair odontogenic differentiation of dental pulp cells, resulting in pulpal irritation and inflammation.


Subject(s)
Camphor/analogs & derivatives , Cytokines/drug effects , Dental Pulp/drug effects , Inflammation Mediators/metabolism , Odontogenesis/drug effects , Photoinitiators, Dental/toxicity , 3T3 Cells , Alkaline Phosphatase/analysis , Animals , Anthraquinones , Blotting, Western , Camphor/toxicity , Cell Differentiation/drug effects , Cell Survival/drug effects , Coloring Agents , Cyclin-Dependent Kinase Inhibitor p16/analysis , Cyclin-Dependent Kinase Inhibitor p21/analysis , Cytokines/metabolism , Dental Materials/toxicity , Dental Pulp/cytology , Humans , Interleukin-6/analysis , Interleukin-8/analysis , Materials Testing , Matrix Metalloproteinase 3/analysis , Methacrylates/toxicity , Mice , Tetrazolium Salts , Thiazoles , Tooth Calcification/drug effects , Tumor Suppressor Protein p53/analysis
SELECTION OF CITATIONS
SEARCH DETAIL