Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 36
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Cell ; 132(2): 299-310, 2008 Jan 25.
Article in English | MEDLINE | ID: mdl-18243104

ABSTRACT

Quiescent adult stem cells reside in specialized niches where they become activated to proliferate and differentiate during tissue homeostasis and injury. How stem cell quiescence is governed is poorly understood. We report here that NFATc1 is preferentially expressed by hair follicle stem cells in their niche, where its expression is activated by BMP signaling upstream and it acts downstream to transcriptionally repress CDK4 and maintain stem cell quiescence. As stem cells become activated during hair growth, NFATc1 is downregulated, relieving CDK4 repression and activating proliferation. When calcineurin/NFATc1 signaling is suppressed, pharmacologically or via complete or conditional NFATc1 gene ablation, stem cells are activated prematurely, resulting in precocious follicular growth. Our findings may explain why patients receiving cyclosporine A for immunosuppressive therapy display excessive hair growth, and unveil a functional role for calcium-NFATc1-CDK4 circuitry in governing stem cell quiescence.


Subject(s)
Cell Proliferation , NFATC Transcription Factors/metabolism , Skin/cytology , Stem Cells/cytology , Stem Cells/physiology , Animals , Antigens, CD34/metabolism , Biomarkers , Cell Nucleus/metabolism , Cells, Cultured , Cyclin-Dependent Kinase 4/antagonists & inhibitors , Cyclosporine/pharmacology , Down-Regulation , Embryo, Mammalian , Gene Deletion , Gene Expression , Gene Expression Regulation, Developmental , Genes, Reporter , Hair Follicle/cytology , Hair Follicle/physiology , Immunohistochemistry , Immunosuppressive Agents/pharmacology , Mice , Mice, Knockout , Mice, Nude , Morphogenesis , NFATC Transcription Factors/genetics , RNA, Messenger/metabolism , Retroviridae/genetics , Skin/embryology , Skin Transplantation , Stem Cells/drug effects , Transcription Factors/metabolism , Transgenes , Transplantation, Homologous
2.
Proc Natl Acad Sci U S A ; 113(47): E7554-E7563, 2016 11 22.
Article in English | MEDLINE | ID: mdl-27821775

ABSTRACT

Appreciation of the role of the gut microbiome in regulating vertebrate metabolism has exploded recently. However, the effects of gut microbiota on skeletal growth and homeostasis have only recently begun to be explored. Here, we report that colonization of sexually mature germ-free (GF) mice with conventional specific pathogen-free (SPF) gut microbiota increases both bone formation and resorption, with the net effect of colonization varying with the duration of colonization. Although colonization of adult mice acutely reduces bone mass, in long-term colonized mice, an increase in bone formation and growth plate activity predominates, resulting in equalization of bone mass and increased longitudinal and radial bone growth. Serum levels of insulin-like growth factor 1 (IGF-1), a hormone with known actions on skeletal growth, are substantially increased in response to microbial colonization, with significant increases in liver and adipose tissue IGF-1 production. Antibiotic treatment of conventional mice, in contrast, decreases serum IGF-1 and inhibits bone formation. Supplementation of antibiotic-treated mice with short-chain fatty acids (SCFAs), products of microbial metabolism, restores IGF-1 and bone mass to levels seen in nonantibiotic-treated mice. Thus, SCFA production may be one mechanism by which microbiota increase serum IGF-1. Our study demonstrates that gut microbiota provide a net anabolic stimulus to the skeleton, which is likely mediated by IGF-1. Manipulation of the microbiome or its metabolites may afford opportunities to optimize bone health and growth.


Subject(s)
Bone Development , Bone and Bones/metabolism , Gastrointestinal Microbiome , Insulin-Like Growth Factor I/metabolism , Adipose Tissue/metabolism , Animals , Fatty Acids, Volatile/metabolism , Female , Liver/metabolism , Male , Mice , Osteogenesis , Specific Pathogen-Free Organisms
3.
Proc Natl Acad Sci U S A ; 110(49): 19914-9, 2013 Dec 03.
Article in English | MEDLINE | ID: mdl-24248346

ABSTRACT

Osteoarthritis (OA) was once viewed originally as a mechanical disease of "wear and tear," but advances made during the past two decades suggest that abnormal biomechanics contribute to active dysregulation of chondrocyte biology, leading to catabolism of the cartilage matrix. A number of signaling and transcriptional mechanisms have been studied in relation to the regulation of this catabolic program, but how they specifically regulate the initiation or progression of the disease is poorly understood. Here, we demonstrate that cartilage-specific ablation of Nuclear factor of activated T cells c1 (Nfatc1) in Nfatc2(-/-) mice leads to early onset, aggressive OA affecting multiple joints. This model recapitulates features of human OA, including loss of proteoglycans, collagen and aggrecan degradation, osteophyte formation, changes to subchondral bone architecture, and eventual progression to cartilage effacement and joint instability. Consistent with the notion that NFATC1 is an OA-suppressor gene, NFATC1 expression was significantly down-regulated in paired lesional vs. macroscopically normal cartilage samples from OA patients. The highly penetrant, early onset, and severe nature of this model make it an attractive platform for the preclinical development of treatments to alter the course of OA. Furthermore, these findings indicate that NFATs are key suppressors of OA, and regulating NFATs or their transcriptional targets in chondrocytes may lead to novel disease-modifying OA therapies.


Subject(s)
Cartilage, Articular/cytology , Chondrocytes/metabolism , Gene Expression Regulation/physiology , NFATC Transcription Factors/metabolism , Osteoarthritis/metabolism , Animals , Cartilage, Articular/metabolism , Immunohistochemistry , Mice , Mice, Knockout , Models, Biological , NFATC Transcription Factors/genetics , Real-Time Polymerase Chain Reaction , X-Ray Microtomography
4.
Proc Natl Acad Sci U S A ; 110(6): 2163-8, 2013 Feb 05.
Article in English | MEDLINE | ID: mdl-23341620

ABSTRACT

Bone remodeling requires osteoclasts to generate and maintain an acidified resorption compartment between the apical membrane and the bone surface to solubilize hydroxyapatite crystals within the bone matrix. This acidification process requires (i) apical proton secretion by a vacuolar H(+)-ATPase, (ii) actin cytoskeleton reorganization into a podosome belt that forms a gasket to restrict lacunar acid leakage, and (iii) basolateral chloride uptake and bicarbonate extrusion by an anion exchanger to provide Cl(-) permissive for apical acid secretion while preventing cytoplasmic alkalinization. Here we show that osteoclast-targeted deletion in mice of solute carrier family 4 anion exchanger member 2 (Slc4a2) results in osteopetrosis. We further demonstrate a previously unrecognized consequence of SLC4A2 loss of function in the osteoclast: dysregulation of calpain-dependent podosome disassembly, leading to abnormal actin belt formation, cell spreading, and migration. Rescue of SLC4A2-deficient osteoclasts with functionally defined mutants of SLC4A2 indicates regulation of actin cytoskeletal reorganization by anion-exchange activity and intracellular pH, independent of SLC4A2's long N-terminal cytoplasmic domain. These data suggest that maintenance of intracellular pH in osteoclasts through anion exchange regulates the actin superstructures required for bone resorption.


Subject(s)
Actin Cytoskeleton/metabolism , Anion Transport Proteins/metabolism , Antiporters/metabolism , Calpain/metabolism , Chloride-Bicarbonate Antiporters/metabolism , Osteoclasts/metabolism , Animals , Anion Transport Proteins/deficiency , Anion Transport Proteins/genetics , Antiporters/deficiency , Antiporters/genetics , Cells, Cultured , Chloride-Bicarbonate Antiporters/deficiency , Chloride-Bicarbonate Antiporters/genetics , Hydrogen-Ion Concentration , Mice , Mice, Knockout , Mutant Proteins/genetics , Mutant Proteins/metabolism , Osteoclasts/pathology , Osteopetrosis/genetics , Osteopetrosis/metabolism , Osteopetrosis/pathology , SLC4A Proteins
5.
Clin Immunol ; 159(2): 163-9, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25840106

ABSTRACT

Recent advances have dramatically increased our understanding of how organ systems interact. This has been especially true for immunology and bone biology, where the term "osteoimmunology" was coined to capture this relationship. The importance of the microbiome to the immune system has also emerged as a driver of health and disease. It makes sense therefore to ask the question: how does the intestinal microbiome influence bone biology and does dysbiosis promote bone disease? Surprisingly, few studies have analyzed this connection. A broader interpretation of this question reveals many mechanisms whereby the microbiome may affect bone cells. These include effects of the microbiome on immune cells, including myeloid progenitors and Th17 cells, as well as steroid hormones, fatty acids, serotonin and vitamin D. As mechanistic interactions of the microbiome and skeletal system are revealed within and without the immune system, novel strategies to optimize skeletal fitness may emerge.


Subject(s)
Adrenal Cortex Hormones/metabolism , Bone and Bones/immunology , Cytokines/immunology , Gastrointestinal Microbiome/immunology , Gonadal Steroid Hormones/metabolism , Osteoblasts/immunology , Osteoclasts/immunology , Th17 Cells/immunology , Animals , Bone Resorption/immunology , Bone Resorption/metabolism , Bone and Bones/metabolism , Humans , Intestines/immunology , Intestines/microbiology , Mice , Myeloid Cells/immunology , Osteoblasts/metabolism , Osteoclasts/metabolism , Osteogenesis/immunology , T-Lymphocytes/immunology
6.
Proc Natl Acad Sci U S A ; 109(21): 8173-8, 2012 May 22.
Article in English | MEDLINE | ID: mdl-22573816

ABSTRACT

Mice lacking the large zinc finger protein Schnurri-3 (Shn3) display increased bone mass, in part, attributable to augmented osteoblastic bone formation. Here, we show that in addition to regulating bone formation, Shn3 indirectly controls bone resorption by osteoclasts in vivo. Although Shn3 plays no cell-intrinsic role in osteoclasts, Shn3-deficient animals show decreased serum markers of bone turnover. Mesenchymal cells lacking Shn3 are defective in promoting osteoclastogenesis in response to selective stimuli, likely attributable to reduced expression of the key osteoclastogenic factor receptor activator of nuclear factor-κB ligand. The bone phenotype of Shn3-deficient mice becomes more pronounced with age, and mice lacking Shn3 are completely resistant to disuse osteopenia, a process that requires functional osteoclasts. Finally, selective deletion of Shn3 in the mesenchymal lineage recapitulates the high bone mass phenotype of global Shn3 KO mice, including reduced osteoclastic bone catabolism in vivo, indicating that Shn3 expression in mesenchymal cells directly controls osteoblastic bone formation and indirectly regulates osteoclastic bone resorption.


Subject(s)
Bone Resorption/physiopathology , DNA-Binding Proteins/genetics , Hyperparathyroidism, Secondary/physiopathology , Osteoblasts/physiology , Osteoclasts/physiology , Aging/physiology , Animals , Bone Resorption/genetics , Cells, Cultured , Coculture Techniques , Cyclic AMP Response Element-Binding Protein/metabolism , DNA-Binding Proteins/metabolism , Hyperparathyroidism, Secondary/genetics , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/physiology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Osteoblasts/cytology , Osteoclasts/cytology , Phenotype , RANK Ligand/metabolism , Regulatory Elements, Transcriptional/physiology , Skull/cytology
7.
Arthritis Rheum ; 65(4): 981-92, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23400684

ABSTRACT

OBJECTIVE: The pathophysiology of the most common joint disease, osteoarthritis (OA), remains poorly understood. Since synovial fluid (SF) bathes joint cartilage and synovium, we reasoned that a comparative analysis of its protein constituents in health and OA could identify pathways involved in joint damage. We undertook this study to perform a proteomic analysis of knee SF from OA patients and control subjects and to compare the results to microarray expression data from cartilage and synovium. METHODS: Age-matched knee SF samples from 10 control subjects, 10 patients with early-stage OA, and 10 patients with late-stage OA were compared using 2-dimensional difference-in-gel electrophoresis and mass spectrometry (MS). MS with a multiplexed peptide selected reaction monitoring assay was used to confirm differential expression of a subset of proteins in an independent OA patient cohort. Proteomic results were analyzed by Ingenuity Pathways Analysis and compared to published synovial tissue and cartilage messenger RNA profiles. RESULTS: Sixty-six proteins were differentially present in healthy and OA SF. Three major pathways were identified among these proteins: the acute-phase response signaling pathway, the complement pathway, and the coagulation pathway. Differential expression of 5 proteins was confirmed by selected reaction monitoring assay. A focused analysis of transcripts corresponding to the differentially present proteins indicated that both synovial and cartilage tissues may contribute to the OA SF proteome. CONCLUSION: Proteins involved in the acute-phase response signaling pathway, the complement pathway, and the coagulation pathway are differentially regulated in SF from OA patients, suggesting that they contribute to joint damage. Validation of these pathways and their utility as biomarkers or therapeutic targets in OA is warranted.


Subject(s)
Cartilage/metabolism , Osteoarthritis, Knee/metabolism , Proteome/analysis , RNA, Messenger/analysis , Synovial Fluid/metabolism , Synovial Membrane/metabolism , Acute-Phase Proteins/genetics , Acute-Phase Proteins/metabolism , Acute-Phase Reaction/metabolism , Aged , Blood Coagulation Factors/genetics , Blood Coagulation Factors/metabolism , Case-Control Studies , Complement System Proteins/genetics , Complement System Proteins/metabolism , Electrophoresis, Gel, Two-Dimensional , Female , Gene Expression Profiling , Humans , Knee Joint/metabolism , Male , Mass Spectrometry , Middle Aged , Osteoarthritis, Knee/genetics , Synovial Fluid/chemistry
8.
Immunol Rev ; 233(1): 286-300, 2010 Jan.
Article in English | MEDLINE | ID: mdl-20193006

ABSTRACT

Osteoporosis and arthritis are highly prevalent diseases and a significant cause of morbidity and mortality worldwide. These diseases result from aberrant tissue remodeling leading to weak, fracture-prone bones or painful, dysfunctional joints. The nuclear factor of activated T cells (NFAT) transcription factor family controls diverse biologic processes in vertebrates. Here, we review the scientific evidence that links NFAT-regulated gene transcription to bone and joint pathology. A particular emphasis is placed on the role of NFATs in bone resorption and formation by osteoclasts and osteoblasts, respectively. In addition, emerging data that connect NFATs with cartilage biology, angiogenesis, nociception, and neurogenic inflammation are explored. The goal of this article is to highlight the importance of tissue remodeling in musculoskeletal disease and situate NFAT-driven cellular responses within this context to inspire future research endeavors.


Subject(s)
Arthritis, Rheumatoid/metabolism , Bone Remodeling/genetics , Chondrogenesis/genetics , Joints/metabolism , NFATC Transcription Factors/metabolism , Osteoarthritis/metabolism , Osteoporosis/metabolism , Transcription, Genetic , Animals , Arthritis, Rheumatoid/genetics , Arthritis, Rheumatoid/pathology , Arthritis, Rheumatoid/physiopathology , Calcineurin/metabolism , Humans , Joints/pathology , NFATC Transcription Factors/genetics , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/metabolism , Osteoarthritis/genetics , Osteoarthritis/pathology , Osteoarthritis/physiopathology , Osteoblasts/metabolism , Osteoclasts/metabolism , Osteoporosis/genetics , Osteoporosis/pathology , Osteoporosis/physiopathology , Pain/genetics , Pain/metabolism
9.
Am J Pathol ; 180(3): 1080-1094, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22245215

ABSTRACT

Development of personalized treatment regimens is hampered by lack of insight into how individual animal models reflect subsets of human disease, and autoimmune and inflammatory conditions have proven resistant to such efforts. Scleroderma is a lethal autoimmune disease characterized by fibrosis, with no effective therapy. Comparative gene expression profiling showed that murine sclerodermatous graft-versus-host disease (sclGVHD) approximates an inflammatory subset of scleroderma estimated at 17% to 36% of patients analyzed with diffuse, 28% with limited, and 100% with localized scleroderma. Both sclGVHD and the inflammatory subset demonstrated IL-13 cytokine pathway activation. Host dermal myeloid cells and graft T cells were identified as sources of IL-13 in the model, and genetic deficiency of either IL-13 or IL-4Rα, an IL-13 signal transducer, protected the host from disease. To identify therapeutic targets, we explored the intersection of genes coordinately up-regulated in sclGVHD, the human inflammatory subset, and IL-13-treated fibroblasts; we identified chemokine CCL2 as a potential target. Treatment with anti-CCL2 antibodies prevented sclGVHD. Last, we showed that IL-13 pathway activation in scleroderma patients correlated with clinical skin scores, a marker of disease severity. Thus, an inflammatory subset of scleroderma is driven by IL-13 and may benefit from IL-13 or CCL2 blockade. This approach serves as a model for personalized translational medicine, in which well-characterized animal models are matched to molecularly stratified patient subsets.


Subject(s)
Chemokine CCL2/genetics , Graft vs Host Disease/genetics , Interleukin-13/genetics , Scleroderma, Systemic/genetics , Animals , Chemokine CCL2/antagonists & inhibitors , Disease Models, Animal , Fibroblasts/metabolism , Gene Expression , Gene Expression Profiling , Humans , Macrophages/metabolism , Mice , Mice, Inbred BALB C , Receptors, Interleukin-13/metabolism , Receptors, Interleukin-4/metabolism , Signal Transduction , T-Lymphocytes/metabolism , Up-Regulation
10.
FASEB J ; 26(3): 1110-8, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22125315

ABSTRACT

A unilateral injection of botulinum toxin A (BTxA) in the calf induces paralysis and profound loss of ipsalateral trabecular bone within days. However, the cellular mechanism underlying acute muscle paralysis-induced bone loss (MPIBL) is poorly understood. We hypothesized that MPIBL arises via rapid and extensive osteoclastogenesis. We performed a series of in vivo experiments to explore this thesis. First, we observed elevated levels of the proosteoclastogenic cytokine receptor activator for nuclear factor-κB ligand (RANKL) within the proximal tibia metaphysis at 7 d after muscle paralysis (+113%, P<0.02). Accordingly, osteoclast numbers were increased 122% compared with the contralateral limb at 5 d after paralysis (P=0.04) and MPIBL was completely blocked by treatment with human recombinant osteoprotegerin (hrOPG). Further, conditional deletion of nuclear factor of activated T-cells c1 (NFATc1), the master regulator of osteoclastogenesis, completely inhibited trabecular bone loss (-2.2±11.9%, P<0.01). All experiments included negative control assessments of contralateral limbs and/or within-animal pre- and postintervention imaging. In summary, transient muscle paralysis induced acute RANKL-mediated osteoclastogenesis resulting in profound local bone resorption. Elucidation of the pathways that initiate osteoclastogenesis after paralysis may identify novel targets to inhibit bone loss and prevent fractures.


Subject(s)
Bone and Bones/metabolism , Muscle, Skeletal/metabolism , Osteoclasts/metabolism , Paralysis/metabolism , RANK Ligand/metabolism , Animals , Bone Resorption/metabolism , Bone Resorption/prevention & control , Botulinum Toxins, Type A/toxicity , Cell Count , Female , Gene Deletion , Humans , Mice , Mice, Inbred C57BL , Muscle, Skeletal/pathology , NFATC Transcription Factors/genetics , NFATC Transcription Factors/metabolism , Osteoclasts/pathology , Osteoprotegerin/genetics , Osteoprotegerin/metabolism , Osteoprotegerin/pharmacology , Paralysis/chemically induced , Quadriceps Muscle/metabolism , Quadriceps Muscle/pathology , Recombinant Proteins/pharmacology , Tibia/metabolism , Time Factors
11.
Curr Rheumatol Rep ; 15(1): 297, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23288576

ABSTRACT

The fundamental mechanisms that drive the pathogenesis of systemic sclerosis (SSc) remain elusive, despite over 50 years of investigation. Here, we review recent progress in the understanding of the immunopathogenesis of SSc. In particular, we consider interleukin-13 (IL13), and its upstream and downstream pathways, as an example of an immune system-derived mediator involved in fibrotic and vascular pathology. Emerging results linking pattern-recognition receptors and interferon pathways to SSc are also stressed. We discuss genetic data linking the immune system to SSc risk and efforts to apply animal models to subsets of patients recently resolved by gene expression profiling. These developments will help build a context for better understanding of previous observations and design of the next generation of studies that may eventually lead to effective treatment.


Subject(s)
Autoimmune Diseases/immunology , Scleroderma, Systemic/immunology , Animals , Autoimmune Diseases/genetics , Disease Models, Animal , Fibrosis , Genetic Predisposition to Disease , Graft vs Host Disease/immunology , Humans , Interleukin-13/immunology , Mice , Scleroderma, Systemic/genetics , Skin/pathology
12.
Biomed Pharmacother ; 169: 115851, 2023 Dec 31.
Article in English | MEDLINE | ID: mdl-37976891

ABSTRACT

BACKGROUND: Clesrovimab (MK-1654) is an investigational, half-life extended human monoclonal antibody (mAb) against RSV F glycoprotein in clinical trials as a prophylactic agent against RSV infection for infants. METHODS: This adult study measured clesrovimab concentrations in the serum and nasal epithelial lining fluid (ELF) to establish the partitioning of the antibody after dosing. Clesrovimab concentrations in the nasal ELF were normalized for sampling dilution using urea concentrations from ELF and serum. Furthermore, in vitro RSV neutralization of human nasal ELF following dosing was also measured to examine the activity of clesrovimab in the nasal compartment. FINDINGS: mAbs with YTE mutations are reported in literature to partition ∼1-2 % of serum antibodies into nasal mucosa. Nasal: serum ratios of 1:69-1:30 were observed for clesrovimab in two separate adult human trials after urea normalization, translating to 1.4-3.3 % of serum concentrations. The nasal PK and estimates of peripheral volume of distribution correlated with higher extravascular distribution of clesrovimab. These higher concentration of the antibody in the nasal ELF corroborated with the nasal sample's ability to neutralize RSV ex vivo. An overall trend of decreased viral plaque AUC was also noted with increasing availability of clesrovimab in the nasal ELF from a human RSV challenge study. INTERPRETATION: Along with its extended half-life, the higher penetration of clesrovimab into the nasal epithelial lining fluid and the associated local increase in RSV neutralization activity could offer infants better protection against RSV infection.


Subject(s)
Respiratory Syncytial Virus Infections , Respiratory Syncytial Virus, Human , Humans , Adult , Antibodies, Monoclonal/therapeutic use , Half-Life , Antibodies, Viral , Respiratory Syncytial Virus Infections/drug therapy , Urea
13.
Clin Transl Sci ; 15(7): 1753-1763, 2022 07.
Article in English | MEDLINE | ID: mdl-35506164

ABSTRACT

Respiratory syncytial virus (RSV) is the leading cause of lower respiratory tract infection among all infants worldwide and remains a significant cause of morbidity and mortality. To address this unmet medical need, MK-1654, a half-life extended RSV neutralizing monoclonal antibody, is in clinical development for the prevention of RSV disease in infants. This was a phase I, randomized, placebo-controlled, single-site, double-blind trial of MK-1654 in 44 healthy Japanese adults. The safety, tolerability, pharmacokinetics, antidrug antibodies (ADAs), and serum neutralizing antibody (SNA) titers against RSV were evaluated for 1 year after a single intramuscular (i.m.) or intravenous (i.v.) dose of MK-1654 or placebo in five groups (100 mg i.m., 300 mg i.m., 300 mg i.v., 1000 mg i.v., or placebo). MK-1654 was generally well-tolerated in Japanese adults. There were no serious drug-related adverse events (AEs) reported in any MK-1654 recipient and no discontinuations due to any AEs in the study. The half-life of MK-1654 ranged from 76 to 91 days across dosing groups. Estimated bioavailability was 86% for 100 mg i.m. and 77% for 300 mg i.m. One participant out of 33 (3.0%) developed detectable ADA with no apparent associated AEs. The RSV SNA titers increased in a dose-dependent manner among participants who received MK-1654. These data support the development of MK-1654 for use in Japanese infants.


Subject(s)
Respiratory Syncytial Virus Infections , Respiratory Syncytial Virus, Human , Adult , Antibodies, Monoclonal/adverse effects , Antibodies, Monoclonal, Humanized , Humans , Infant , Japan , Respiratory Syncytial Virus Infections/drug therapy , Respiratory Syncytial Virus Infections/prevention & control
14.
J Clin Invest ; 118(11): 3775-89, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18846253

ABSTRACT

Osteoporosis results from an imbalance in skeletal remodeling that favors bone resorption over bone formation. Bone matrix is degraded by osteoclasts, which differentiate from myeloid precursors in response to the cytokine RANKL. To gain insight into the transcriptional regulation of bone resorption during growth and disease, we generated a conditional knockout of the transcription factor nuclear factor of activated T cells c1 (Nfatc1). Deletion of Nfatc1 in young mice resulted in osteopetrosis and inhibition of osteoclastogenesis in vivo and in vitro. Transcriptional profiling revealed NFATc1 as a master regulator of the osteoclast transcriptome, promoting the expression of numerous genes needed for bone resorption. In addition, NFATc1 directly repressed osteoclast progenitor expression of osteoprotegerin, a decoy receptor for RANKL previously thought to be an osteoblast-derived inhibitor of bone resorption. "Cherubism mice", which carry a gain-of-function mutation in SH3-domain binding protein 2 (Sh3bp2), develop osteoporosis and widespread inflammation dependent on the proinflammatory cytokine, TNF-alpha. Interestingly, deletion of Nfatc1 protected cherubism mice from systemic bone loss but did not inhibit inflammation. Taken together, our study demonstrates that NFATc1 is required for remodeling of the growing and adult skeleton and suggests that NFATc1 may be an effective therapeutic target for osteoporosis associated with inflammatory states.


Subject(s)
Bone Diseases, Metabolic/pathology , Cherubism/metabolism , Inflammation/pathology , NFATC Transcription Factors/metabolism , Osteoclasts/physiology , Osteoprotegerin/metabolism , Animals , Cherubism/genetics , Mice , Mice, Knockout , Mice, Transgenic , Osteoclasts/metabolism , Osteoprotegerin/genetics
15.
Proc Natl Acad Sci U S A ; 105(44): 16934-9, 2008 Nov 04.
Article in English | MEDLINE | ID: mdl-18971331

ABSTRACT

As the only cell capable of bone resorption, the osteoclast is a central mediator of skeletal homeostasis and disease. To efficiently degrade mineralized tissue, these multinucleated giant cells secrete acid into a resorption lacuna formed between their apical membrane and the bone surface. For each proton pumped into this extracellular compartment, one bicarbonate ion remains in the cytoplasm. To prevent alkalinization of the cytoplasm, a basolateral bicarbonate/chloride exchanger provides egress for intracellular bicarbonate. However, the identity of this exchanger is unknown. Here, we report that the bicarbonate/chloride exchanger, solute carrier family 4, anion exchanger, member 2 (SLC4A2), is up-regulated during osteoclast differentiation. Suppression of Slc4a2 expression by RNA interference inhibits the ability of RAW cells, a mouse macrophage cell line, to differentiate into osteoclasts and resorb mineralized matrix in vitro. Accordingly, Slc4a2-deficient mice fail to remodel the primary, cartilaginous skeletal anlagen. Abnormal multinucleated giant cells are present in the bone marrow of Slc4a2-deficient mice. Though these cells express the osteoclast markers CD68, cathepsin K, and NFATc1, compared with their wild-type (WT) counterparts they are larger, fail to express tartrate-resistant acid phosphatase (TRAP) activity, and display a propensity to undergo apoptosis. In vitro Slc4a2-deficient osteoclasts are unable to resorb mineralized tissue and cannot form an acidified, extracellular resorption compartment. These data highlight SLC4A2 as a critical mediator of osteoclast differentiation and function in vitro and in vivo.


Subject(s)
Anion Transport Proteins/physiology , Antiporters/physiology , Cell Differentiation , Osteoclasts/cytology , Osteoclasts/physiology , Animals , Anion Transport Proteins/genetics , Anion Transport Proteins/metabolism , Antiporters/genetics , Antiporters/metabolism , Apoptosis , Bone Resorption/genetics , Bone Resorption/metabolism , Cells, Cultured , Chloride-Bicarbonate Antiporters , Mice , Mice, Transgenic , NFATC Transcription Factors/metabolism , Osteoclasts/metabolism , Osteopetrosis/genetics , Osteopetrosis/metabolism , SLC4A Proteins , Up-Regulation
16.
Clin Pharmacol Drug Dev ; 10(5): 556-566, 2021 05.
Article in English | MEDLINE | ID: mdl-33125189

ABSTRACT

Respiratory syncytial virus (RSV) is the leading cause of acute lower respiratory tract infection and related morbidity and mortality in infants. Passive immunization with an RSV-neutralizing antibody can provide rapid protection to this vulnerable population. Proof-of-concept for this approach has been demonstrated by palivizumab; however, the use of this antibody is generally restricted to the highest-risk infants due to monthly dosing requirements and its cost. To address the large unmet medical need for most infants, we are evaluating MK-1654, a fully human RSV-neutralizing antibody with half-life extending mutations targeting site IV of the fusion protein. In this 2-part, placebo-controlled, double-blind, first-in-human study, 152 healthy adults were randomized 3:1 to receive a single dose of MK-1654 or placebo in 5 cohorts (100 or 300 mg as an intramuscular dose or 300, 1000, or 3000 mg as an intravenous dose). Safety, pharmacokinetics, antidrug antibodies, and RSV serum-neutralizing antibody titers were evaluated through 1 year. MK-1654 serum concentrations increased proportionally with dose and resulted in corresponding elevations in RSV serum-neutralizing antibody titers. The antibody displayed a half-life of 73 to 88 days and an estimated bioavailability of 69% at the 300-mg dose. The overall safety profile of MK-1654 was similar to placebo, and treatment-emergent antidrug antibodies were low (2.6%) with no associated adverse events. These data support the continued development of MK-1654 for the prevention of RSV disease in infants.


Subject(s)
Antibodies, Monoclonal , Antibodies, Neutralizing , Antiviral Agents , Adult , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/adverse effects , Antibodies, Monoclonal/pharmacokinetics , Antibodies, Neutralizing/administration & dosage , Antibodies, Neutralizing/adverse effects , Antiviral Agents/administration & dosage , Antiviral Agents/adverse effects , Antiviral Agents/pharmacokinetics , Biological Availability , Cohort Studies , Dose-Response Relationship, Drug , Double-Blind Method , Female , Half-Life , Humans , Infusions, Intravenous , Injections, Intramuscular , Male , Middle Aged , Respiratory Syncytial Virus Infections/prevention & control , Respiratory Syncytial Viruses/immunology , Young Adult
17.
Hum Vaccin Immunother ; 17(5): 1248-1261, 2021 05 04.
Article in English | MEDLINE | ID: mdl-33121346

ABSTRACT

Respiratory Syncytial Virus (RSV) causes lower respiratory tract infections that can be severe and sometimes fatal. The risk for severe RSV infection is highest in infants and older adults. A safe and effective RSV vaccine for older adults represents a serious unmet medical need due to higher morbidity and mortality in this age group. In this randomized, partially double-blind, placebo-controlled, phase 1 dose-escalation study, we evaluated the safety, tolerability and immunogenicity of an investigational messenger ribonucleic acid (mRNA) vaccine encoding the RSV fusion protein (F) stabilized in the prefusion conformation. The study was conducted in healthy younger adults (ages ≥18 and ≤49 years) and healthy older adults (ages ≥60 and ≤79 years). Participants received mRNA-1777 (V171) or placebo as a single intramuscular dose. For each dose level, three sentinel participants were administered open-label mRNA-1777 (V171). Seventy-two younger adults were randomized and administered 25, 100, or 200 µg mRNA-1777 (V171) or placebo, and 107 older adults were randomized and administered 25, 100, 200 or 300 µg mRNA-1777 (V171) or placebo. Primary objectives were safety and tolerability and secondary objectives included humoral and cell-mediated immunogenicity. All dose levels of mRNA-1777 (V171) were generally well tolerated and no serious adverse events related to the vaccine were reported. Immunization with mRNA-1777 (V171) elicited a humoral immune response as measured by increases in RSV neutralizing antibody titers, serum antibody titers to RSV prefusion F protein, D25 competing antibody titers to RSV prefusion F protein, and cell-mediated immune responses to RSV-F peptides.


Subject(s)
Respiratory Syncytial Virus Infections , Respiratory Syncytial Virus Vaccines , Respiratory Syncytial Virus, Human , Aged , Antibodies, Neutralizing , Antibodies, Viral , Humans , Immunogenicity, Vaccine , Middle Aged , RNA, Messenger , Viral Fusion Proteins
18.
EBioMedicine ; 73: 103651, 2021 Nov.
Article in English | MEDLINE | ID: mdl-34775220

ABSTRACT

BACKGROUND: Neutralizing mAbs can prevent communicable viral diseases. MK-1654 is a respiratory syncytial virus (RSV) F glycoprotein neutralizing monoclonal antibody (mAb) under development to prevent RSV infection in infants. Development and validation of methods to predict efficacious doses of neutralizing antibodies across patient populations exposed to a time-varying force of infection (i.e., seasonal variation) are necessary. METHODS: Five decades of clinical trial literature were leveraged to build a model-based meta-analysis (MBMA) describing the relationship between RSV serum neutralizing activity (SNA) and clinical endpoints. The MBMA was validated by backward translation to animal challenge experiments and forward translation to predict results of a recent RSV mAb trial. MBMA predictions were evaluated against a human trial of 70 participants who received either placebo or one of four dose-levels of MK-1654 and were challenged with RSV [NCT04086472]. The MBMA was used to perform clinical trial simulations and predict efficacy of MK-1654 in the infant target population. FINDINGS: The MBMA established a quantitative relationship between RSV SNA and clinical endpoints. This relationship was quantitatively consistent with animal model challenge experiments and results of a recently published clinical trial. Additionally, SNA elicited by increasing doses of MK-1654 in humans reduced RSV symptomatic infection rates with a quantitative relationship that approximated the MBMA. The MBMA indicated a high probability that a single dose of ≥ 75 mg of MK-1654 will result in prophylactic efficacy (> 75% for 5 months) in infants. INTERPRETATION: An MBMA approach can predict efficacy of neutralizing antibodies against RSV and potentially other respiratory pathogens.


Subject(s)
Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , Respiratory Syncytial Virus Infections/immunology , Respiratory Syncytial Virus Infections/prevention & control , Respiratory Syncytial Virus, Human/immunology , Translational Research, Biomedical/methods , Adolescent , Adult , Aged , Algorithms , Antibodies, Monoclonal , Antibodies, Neutralizing/administration & dosage , Antibodies, Viral/administration & dosage , Clinical Trials as Topic , Female , Humans , Incidence , Male , Middle Aged , Models, Theoretical , Premedication , Respiratory Syncytial Virus Infections/epidemiology , Seasons , Young Adult
19.
Adv Exp Med Biol ; 658: 69-75, 2010.
Article in English | MEDLINE | ID: mdl-19950017

ABSTRACT

The nuclear factor of activated T-cells (NFAT) family of transcription factors specify developmental pathways and cell fate in vertebrates. NFATc1, in particular, is crucial to multiple seemingly unrelated biologic processes, including heart valve formation, T-cell activation, osteoclast development, and the mitigation of hair follicle stem cell proliferation. Here, we review how our recently generated NFATc1 conditional knockout mouse has contributed to our understanding of this transcription factor in inflammatory and musculoskeletal conditions and their treatment.


Subject(s)
Inflammation/metabolism , Musculoskeletal Diseases/metabolism , NFATC Transcription Factors/metabolism , Animals , Calcineurin/metabolism , Calcineurin Inhibitors , Hair Follicle/growth & development , Humans , Osteogenesis
20.
Nat Commun ; 9(1): 1954, 2018 05 11.
Article in English | MEDLINE | ID: mdl-29752435

ABSTRACT

In the original version of this Article, financial support was not fully acknowledged. The PDF and HTML versions of the Article have now been corrected to include support from the National Football League Players Association.

SELECTION OF CITATIONS
SEARCH DETAIL