Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
Add more filters










Publication year range
1.
Biomedicines ; 12(4)2024 Apr 20.
Article in English | MEDLINE | ID: mdl-38672266

ABSTRACT

Casimersen (AMONDYS 45TM) is an antisense oligonucleotide of the phosphorodiamidate morpholino oligomer subclass developed by Sarepta therapeutics. It was approved by the Food and Drug Administration (FDA) in February 2021 to treat Duchenne muscular dystrophy (DMD) in patients whose DMD gene mutation is amenable to exon 45 skipping. Administered intravenously, casimersen binds to the pre-mRNA of the DMD gene to skip a mutated region of an exon, thereby producing an internally truncated yet functional dystrophin protein in DMD patients. This is essential in maintaining the structure of a myocyte membrane. While casimersen is currently continuing in phase III of clinical trials in various countries, it was granted approval by the FDA under the accelerated approval program due to its observed increase in dystrophin production. This article discusses the pathophysiology of DMD, summarizes available treatments thus far, and provides a full drug review of casimersen (AMONDYS 45TM).

2.
J Bone Miner Res ; 39(3): 298-314, 2024 Apr 19.
Article in English | MEDLINE | ID: mdl-38477790

ABSTRACT

Osteocytes sense and respond to mechanical force by controlling the activity of other bone cells. However, the mechanisms by which osteocytes sense mechanical input and transmit biological signals remain unclear. Voltage-sensitive calcium channels (VSCCs) regulate calcium (Ca2+) influx in response to external stimuli. Inhibition or deletion of VSCCs impairs osteogenesis and skeletal responses to mechanical loading. VSCC activity is influenced by its auxiliary subunits, which bind the channel's α1 pore-forming subunit to alter intracellular Ca2+ concentrations. The α2δ1 auxiliary subunit associates with the pore-forming subunit via a glycosylphosphatidylinositol anchor and regulates the channel's calcium-gating kinetics. Knockdown of α2δ1 in osteocytes impairs responses to membrane stretch, and global deletion of α2δ1 in mice results in osteopenia and impaired skeletal responses to loading in vivo. Therefore, we hypothesized that the α2δ1 subunit functions as a mechanotransducer, and its deletion in osteocytes would impair skeletal development and load-induced bone formation. Mice (C57BL/6) with LoxP sequences flanking Cacna2d1, the gene encoding α2δ1, were crossed with mice expressing Cre under the control of the Dmp1 promoter (10 kb). Deletion of α2δ1 in osteocytes and late-stage osteoblasts decreased femoral bone quantity (P < .05) by DXA, reduced relative osteoid surface (P < .05), and altered osteoblast and osteocyte regulatory gene expression (P < .01). Cacna2d1f/f, Cre + male mice displayed decreased femoral strength and lower 10-wk cancellous bone in vivo micro-computed tomography measurements at the proximal tibia (P < .01) compared to controls, whereas Cacna2d1f/f, Cre + female mice showed impaired 20-wk cancellous and cortical bone ex vivo micro-computed tomography measurements (P < .05) vs controls. Deletion of α2δ1 in osteocytes and late-stage osteoblasts suppressed load-induced calcium signaling in vivo and decreased anabolic responses to mechanical loading in male mice, demonstrating decreased mechanosensitivity. Collectively, the α2δ1 auxiliary subunit is essential for the regulation of osteoid-formation, femur strength, and load-induced bone formation in male mice.


The ability of bone to sense and respond to forces generated during daily physical activities is essential to skeletal health. Although several bone cell types contribute to the maintenance of bone health, osteocytes are thought to be the primary mechanosensitive cells; however, the mechanisms through which these cells perceive mechanical stimuli remains unclear. Previous work has shown that voltage sensitive calcium channels are necessary for bone to sense mechanical force; yet the means by which those channels translate the physical signal into a biochemical signal is unclear. Data within this manuscript demonstrate that the extracellular α2δ1 subunit of voltage sensitive calcium channels is necessary for load-induced bone formation as well as to enable calcium influx within osteocytes. As this subunit enables physical interactions of the channel pore with the extracellular matrix, our data demonstrate the need for the α2δ1 subunit for mechanically induced bone adaptation, thus serving as a physical conduit through which mechanical signals from the bone matrix are transduced into biochemical signals by enabling calcium influx into osteocytes.


Subject(s)
Osteocytes , Osteogenesis , Mice , Male , Female , Animals , Osteocytes/metabolism , Osteogenesis/genetics , Calcium/metabolism , X-Ray Microtomography , Mice, Inbred C57BL , Osteoblasts/metabolism , Femur/diagnostic imaging , Femur/metabolism , Calcium Channels/genetics , Calcium Channels/metabolism
3.
JBMR Plus ; 8(2): ziad008, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38505532

ABSTRACT

Voltage-sensitive calcium channels (VSCCs) influence bone structure and function, including anabolic responses to mechanical loading. While the pore-forming (α1) subunit of VSCCs allows Ca2+ influx, auxiliary subunits regulate the biophysical properties of the pore. The α2δ1 subunit influences gating kinetics of the α1 pore and enables mechanically induced signaling in osteocytes; however, the skeletal function of α2δ1 in vivo remains unknown. In this work, we examined the skeletal consequences of deleting Cacna2d1, the gene encoding α2δ1. Dual-energy X-ray absorptiometry and microcomputed tomography imaging demonstrated that deletion of α2δ1 diminished bone mineral content and density in both male and female C57BL/6 mice. Structural differences manifested in both trabecular and cortical bone for males, while the absence of α2δ1 affected only cortical bone in female mice. Deletion of α2δ1 impaired skeletal mechanical properties in both sexes, as measured by three-point bending to failure. While no changes in osteoblast number or activity were found for either sex, male mice displayed a significant increase in osteoclast number, accompanied by increased eroded bone surface and upregulation of genes that regulate osteoclast differentiation. Deletion of α2δ1 also rendered the skeleton insensitive to exogenous mechanical loading in males. While previous work demonstrates that VSCCs are essential for anabolic responses to mechanical loading, the mechanism by which these channels sense and respond to force remained unclear. Our data demonstrate that the α2δ1 auxiliary VSCC subunit functions to maintain baseline bone mass and strength through regulation of osteoclast activity and also provides skeletal mechanotransduction in male mice. These data reveal a molecular player in our understanding of the mechanisms by which VSCCs influence skeletal adaptation.

4.
Int J Mol Sci ; 24(18)2023 Sep 21.
Article in English | MEDLINE | ID: mdl-37762698

ABSTRACT

Low back pain (LBP) is a common musculoskeletal complaint that can impede physical function and mobility. Current management often involves pain medication, but there is a need for non-pharmacological and non-invasive interventions. Soft tissue manipulation (STM), such as massage, has been shown to be effective in human subjects, but the molecular mechanisms underlying these findings are not well understood. In this paper, we evaluated potential changes in the soft tissue levels of more than thirty pro- or anti-inflammatory cytokines following instrument-assisted STM (IASTM) in rats with chronic, induced LBP using Complete Freund's Adjuvant. Our results indicate that IASTM is associated with reduced soft tissue levels of Regulated on Activation, Normal T cell Expressed and Secreted (RANTES)/Chemokine (C-C motif) ligand 5 (CCL5) and increased soft tissue levels of Interleukin (IL)-4, which are pro-inflammatory and anti-inflammatory factors, respectively, by 120 min post-treatment. IASTM was not associated with tissue-level changes in C-X-C Motif Chemokine Ligand (CXCL)-5/Lipopolysaccharide-Induced CXC Chemokine (LIX)-which is the murine homologue of IL-8, CXCL-7, Granulocyte-Macrophage-Colony Simulating Factor (GM-CSF), Intercellular Adhesion Molecule (ICAM)-1, IL1-Receptor Antagonist (IL-1ra), IL-6, Interferon-Inducible Protein (IP)-10/CXCL-10, L-selectin, Tumor Necrosis Factor (TNF)-α, or Vascular Endothelial Growth Factor (VEGF) at either 30 or 120 min post-treatment. Combined, our findings raise the possibility that IASTM may exert tissue-level effects associated with improved clinical outcomes and potentially beneficial changes in pro-/anti-inflammatory cytokines in circulation and at the tissue level.

5.
Life (Basel) ; 13(4)2023 Mar 31.
Article in English | MEDLINE | ID: mdl-37109447

ABSTRACT

Global loss of the neuropeptide Neuromedin-U (NMU) is associated with increased bone formation and high bone mass in male and female mice by twelve weeks of age, suggesting that NMU suppresses osteoblast differentiation and/or activity in vivo. NMU is highly expressed in numerous anatomical locations including the skeleton and the hypothalamus. This raises the possibility that NMU exerts indirect effects on bone remodeling from an extra-skeletal location such as the brain. Thus, in the present study we used microinjection to deliver viruses carrying short-hairpin RNA designed to knockdown Nmu expression in the hypothalamus of 8-week-old male rats and evaluated the effects on bone mass in the peripheral skeleton. Quantitative RT-PCR confirmed approximately 92% knockdown of Nmu in the hypothalamus. However, after six weeks, micro computed tomography on tibiae from Nmu-knockdown rats demonstrated no significant change in trabecular or cortical bone mass as compared to controls. These findings are corroborated by histomorphometric analyses which indicate no differences in osteoblast or osteoclast parameters between controls and Nmu-knockdown samples. Collectively, these data suggest that hypothalamus-derived NMU does not regulate bone remodeling in the postnatal skeleton. Future studies are necessary to delineate the direct versus indirect effects of NMU on bone remodeling.

6.
Biomolecules ; 12(12)2022 12 12.
Article in English | MEDLINE | ID: mdl-36551284

ABSTRACT

Our understanding of how osteocytes, the principal mechanosensors within bone, sense and perceive force remains unclear. Previous work identified "tethering elements" (TEs) spanning the pericellular space of osteocytes and transmitting mechanical information into biochemical signals. While we identified the heparan sulfate proteoglycan perlecan (PLN) as a component of these TEs, PLN must attach to the cell surface to induce biochemical responses. As voltage-sensitive calcium channels (VSCCs) are critical for bone mechanotransduction, we hypothesized that PLN binds the extracellular α2δ1 subunit of VSCCs to couple the bone matrix to the osteocyte membrane. Here, we showed co-localization of PLN and α2δ1 along osteocyte dendritic processes. Additionally, we quantified the molecular interactions between α2δ1 and PLN domains and demonstrated for the first time that α2δ1 strongly associates with PLN via its domain III. Furthermore, α2δ1 is the binding site for the commonly used pain drug, gabapentin (GBP), which is associated with adverse skeletal effects when used chronically. We found that GBP disrupts PLN::α2δ1 binding in vitro, and GBP treatment in vivo results in impaired bone mechanosensation. Our work identified a novel mechanosensory complex within osteocytes composed of PLN and α2δ1, necessary for bone force transmission and sensitive to the drug GBP.


Subject(s)
Heparan Sulfate Proteoglycans , Mechanotransduction, Cellular , Heparan Sulfate Proteoglycans/metabolism , Gabapentin/pharmacology , Extracellular Matrix Proteins/metabolism , Calcium Channels/genetics , Calcium Channels/metabolism
7.
Endocrinology ; 164(1)2022 11 14.
Article in English | MEDLINE | ID: mdl-36412119

ABSTRACT

Diabetes results from insufficient insulin production by pancreatic islet ß-cells or a loss of ß-cells themselves. Restoration of regulated insulin production is a predominant goal of translational diabetes research. Here, we provide a brief overview of recent advances in the fields of ß-cell proliferation, regeneration, and replacement. The discovery of therapeutic targets and associated small molecules has been enabled by improved understanding of ß-cell development and cell cycle regulation, as well as advanced high-throughput screening methodologies. Important findings in ß-cell transdifferentiation, neogenesis, and stem cell differentiation have nucleated multiple promising therapeutic strategies. In particular, clinical trials are underway using in vitro-generated ß-like cells from human pluripotent stem cells. Significant challenges remain for each of these strategies, but continued support for efforts in these research areas will be critical for the generation of distinct diabetes therapies.


Subject(s)
Diabetes Mellitus , Insulin-Secreting Cells , Humans , Regeneration/physiology , Insulin-Secreting Cells/metabolism , Cell Proliferation , Diabetes Mellitus/therapy , Diabetes Mellitus/metabolism , Insulin/metabolism , Cell Differentiation
8.
Curr Osteoporos Rep ; 20(6): 365-378, 2022 12.
Article in English | MEDLINE | ID: mdl-36149592

ABSTRACT

PURPOSE OF REVIEW: In this review, we discuss the mechanism of action of gabapentinoids and the potential consequences of long-term treatment with these drugs on the musculoskeletal system. RECENT FINDINGS: Gabapentinoids, such as gabapentin (GBP) and pregabalin (PGB) were designed as antiepileptic reagents and are now commonly used as first-line treatment for neuropathic pain and increasingly prescribed off-label for other pain disorders such as migraines and back pain. GBP and PGB exert their analgesic actions by selectively binding the α2δ1 auxiliary subunit of voltage-sensitive calcium channels, thereby inhibiting channel function. Numerous tissues express the α2δ1 subunit where GBP and PGB can alter calcium-mediated signaling events. In tissues such as bone, muscle, and cartilage, α2δ1 has important roles in skeletal formation, mechanosensation, and normal tissue function/repair that may be affected by chronic use of gabapentinoids. Long-term use of gabapentinoids is associated with detrimental musculoskeletal outcomes, including increased fracture risk. Therefore, understanding potential complications is essential for clinicians to guide appropriate treatments.


Subject(s)
Calcium , Humans , Gabapentin/pharmacology , gamma-Aminobutyric Acid/therapeutic use , gamma-Aminobutyric Acid/pharmacology , Homeostasis , Pregabalin/therapeutic use , Pregabalin/pharmacology
9.
Physiol Rep ; 10(11): e15307, 2022 06.
Article in English | MEDLINE | ID: mdl-35656701

ABSTRACT

Ferric citrate (FC) is an approved therapy for chronic kidney disease (CKD) patients as a phosphate (Pi) binder for dialysis-dependent CKD, and for iron deficiency anemia (IDA) in non-dialysis CKD. Elevated Pi and IDA both lead to increased FGF23, however, the roles of iron and FGF23 during CKD remain unclear. To this end, iron and Pi metabolism were tested in a mouse model of CKD (0.2% adenine) ± 0.5% FC for 6 weeks, with and without osteocyte deletion of Fgf23 (flox-Fgf23/Dmp1-Cre). Intact FGF23 (iFGF23) increased in all CKD mice but was lower in Cre+ mice with or without FC, thus the Dmp1-Cre effectively reduced FGF23. Cre+ mice fed AD-only had higher serum Pi than Cre- pre- and post-diet, and the Cre+ mice had higher BUN regardless of FC treatment. Total serum iron was higher in all mice receiving FC, and liver Tfrc, Bmp6, and hepcidin mRNAs were increased regardless of genotype; liver IL-6 showed decreased mRNA in FC-fed mice. The renal 1,25-dihydroxyvitamin D (1,25D) anabolic enzyme Cyp27b1 had higher mRNA and the catabolic Cyp24a1 showed lower mRNA in FC-fed mice. Finally, mice with loss of FGF23 had higher bone cortical porosity, whereas Raman spectroscopy showed no changes in matrix mineral parameters. Thus, FC- and FGF23-dependent and -independent actions were identified in CKD; loss of FGF23 was associated with higher serum Pi and BUN, demonstrating that FGF23 was protective of mineral metabolism. In contrast, FC maintained serum iron and corrected inflammation mediators, potentially providing ancillary benefit.


Subject(s)
Fibroblast Growth Factors , Iron , Renal Insufficiency, Chronic , Animals , Citric Acid , Disease Models, Animal , Electrolytes , Ferric Compounds , Fibroblast Growth Factor-23 , Fibroblast Growth Factors/metabolism , Humans , Iron/metabolism , Mice , Minerals , RNA, Messenger/metabolism , Renal Insufficiency, Chronic/metabolism
10.
Life (Basel) ; 11(10)2021 Sep 29.
Article in English | MEDLINE | ID: mdl-34685399

ABSTRACT

Neuromedin-U (NMU) is an evolutionarily conserved peptide that regulates varying physiologic effects including blood pressure, stress and allergic responses, metabolic and feeding behavior, pain perception, and neuroendocrine functions. Recently, several lines of investigation implicate NMU in regulating bone remodeling. For instance, global loss of NMU expression in male and female mice leads to high bone mass due to elevated bone formation rate with no alteration in bone resorption rate or observable defect in skeletal patterning. Additionally, NMU treatment regulates the activity of osteoblasts in vitro. The downstream pathway utilized by NMU to carry out these effects is unknown as NMU signals via two G-protein-coupled receptors (GPCRs), NMU receptor 1 (NMUR1), and NMU receptor 2 (NMUR2), and both are expressed in the postnatal skeleton. Here, we sought to address this open question and build a better understanding of the downstream pathway utilized by NMU. Our approach involved the knockdown of Nmur1 in MC3T3-E1 cells in vitro and a global knockout of Nmur1 in vivo. We detail specific cell signaling events (e.g., mTOR phosphorylation) that are deficient in the absence of NMUR1 expression yet trabecular bone volume in femora and tibiae of 12-week-old male Nmur1 knockout mice are unchanged, compared to controls. These results suggest that NMUR1 is required for NMU-dependent signaling in MC3T3-E1 cells, but it is not required for the NMU-mediated effects on bone remodeling in vivo. Future studies examining the role of NMUR2 are required to determine the downstream pathway utilized by NMU to regulate bone remodeling in vivo.

11.
Life (Basel) ; 11(6)2021 Jun 15.
Article in English | MEDLINE | ID: mdl-34203792

ABSTRACT

The authors present a stereotypical case presentation of X-linked hypophosphatemia (XLH) and provide a review of the pathophysiology and related pharmacology of this condition, primarily focusing on the FDA-approved medication burosumab. XLH is a renal phosphate wasting disorder caused by loss of function mutations in the PHEX gene (phosphate-regulating gene with homologies to endopeptidases on the X chromosome). Typical biochemical findings include elevated serum levels of bioactive/intact fibroblast growth factor 23 (FGF23) which lead to (i) low serum phosphate levels, (ii) increased fractional excretion of phosphate, and (iii) inappropriately low or normal 1,25-dihydroxyvitamin D (1,25-vitD). XLH is the most common form of heritable rickets and short stature in patients with XLH is due to chronic hypophosphatemia. Additionally, patients with XLH experience joint pain and osteoarthritis from skeletal deformities, fractures, enthesopathy, spinal stenosis, and hearing loss. Historically, treatment for XLH was limited to oral phosphate supplementation, active vitamin D supplementation, and surgical intervention for cases of severe bowed legs. In 2018, the United States Food and Drug Administration (FDA) approved burosumab for the treatment of XLH and this medication has demonstrated substantial benefit compared with conventional therapy. Burosumab binds circulating intact FGF23 and blocks its biological effects in target tissues, resulting in increased serum inorganic phosphate (Pi) concentrations and increased conversion of inactive vitamin D to active 1,25-vitD.

12.
BMC Res Notes ; 13(1): 400, 2020 Aug 27.
Article in English | MEDLINE | ID: mdl-32854782

ABSTRACT

OBJECTIVE: Soft tissue manual therapies are commonly utilized by osteopathic physicians, chiropractors, physical therapists and massage therapists. These techniques are predicated on subjecting tissues to biophysical mechanical stimulation but the cellular and molecular mechanism(s) mediating these effects are poorly understood. Previous studies established an in vitro model system for examining mechanical stimulation of dermal fibroblasts and established that cyclical strain, intended to mimic overuse injury, induces secretion of numerous pro-inflammatory cytokines. Moreover, mechanical strain intended to mimic soft tissue manual therapy reduces strain-induced secretion of pro-inflammatory cytokines. Here, we sought to partially confirm and extend these reports and provide independent corroboration of prior results. RESULTS: Using cultures of primary human dermal fibroblasts, we confirm cyclical mechanical strain increases levels of IL-6 and adding long-duration stretch, intended to mimic therapeutic soft tissue stimulation, after cyclical strain results in lower IL-6 levels. We also extend the prior work, reporting that long-duration stretch results in lower levels of IL-8. Although there are important limitations to this experimental model, these findings provide supportive evidence that therapeutic soft tissue stimulation may reduce levels of pro-inflammatory cytokines. Future work is required to address these open questions and advance the mechanistic understanding of therapeutic soft tissue stimulation.


Subject(s)
Cytokines , Musculoskeletal Manipulations , Cells, Cultured , Fibroblasts , Humans , Skin
15.
J Am Osteopath Assoc ; 120(4): 236-244, 2020 Apr 01.
Article in English | MEDLINE | ID: mdl-32227149

ABSTRACT

CONTEXT: There are limited data regarding the experiences of and attitudes toward research participation among osteopathic medical students despite rapidly increasing enrollment and expansion of the number of osteopathic medical schools. OBJECTIVE: To assess first-year osteopathic medical students' experience with research, their interest in it, their perceptions of its value, and barriers to participation. METHODS: An anonymous, online survey was sent to 868 medical students in the class of 2021 at 4 colleges of osteopathic medicine. The survey consisted of 14 multiple-choice items (7 of which offered the option of a written response) and 1 open-ended item that asked them to report their age. The survey remained open for 2 weeks, with 1 reminder email sent on the last day of the survey. Incomplete responses were excluded from the analysis. RESULTS: A total of 328 participants were included, for a response rate of 38%. A majority of respondents reported previous research experience (261 [79.6%]), consistent with a strong perception that research participation is important (315 [96.0%]). Fewer students (177 [54.0%]) were either currently participating in research or affirmed interest in performing research during medical school, with the highest level of interest in clinical research (259 [79.0%]) followed by basic science (166 [50.6%]). Regarding incentives that might encourage participation in research, students preferred monetary compensation (213 [64.9%]) or extra credit in courses (195 [59.5%]). A commonly reported barrier to performing research during medical school was the possibility of a negative impact on performance in coursework (289 [88.1%]). CONCLUSION: First-year osteopathic medical students are interested in research, view research experience as valuable, and consider research experience as beneficial to future career development. This study's findings highlight opportunities for increasing student participation in research through incentives or removal of perceived barriers.


Subject(s)
Osteopathic Medicine , Students, Medical , Humans , Osteopathic Medicine/education , Perception , Schools, Medical , Surveys and Questionnaires
16.
Biochem Biophys Res Commun ; 524(4): 890-894, 2020 04 16.
Article in English | MEDLINE | ID: mdl-32057362

ABSTRACT

Osteoporosis is a disease of low bone mass that places individuals at enhanced risk for fracture, disability, and death. Osteoporosis rates are expected to rise significantly in the coming decades yet there are limited pharmacological treatment options, particularly for long-term management of this chronic condition. The drug development pipeline is relatively bereft of new strategies, causing an urgent and unmet need for developing new strategies and targets for treating osteoporosis. Here, we examine a lesser-studied bone remodeling pathway, Neuromedin U (NMU), which is expressed in the bone microenvironment along with its cognate receptors NMU receptor 1 (NMUR1) and 2 (NMUR2). We independently corroborate a prior report that global loss of NMU expression leads to high bone mass and test the hypothesis that NMU negatively regulates osteoblast differentiation. Consistent with this, in vitro studies reveal NMU represses osteoblastic differentiation of osteogenic precursors but, in contrast, promotes osteoblastic marker expression, proliferation and activity of osteoblast-like cells. Phospho-profiling arrays were used to detail differential signaling outcomes that may underlie the opposite responses of these cell types. Collectively, our findings indicate that NMU exerts cell-type-specific responses to regulate osteoblast differentiation and activity.


Subject(s)
Neuropeptides/genetics , Osteoblasts/metabolism , Osteoporosis/genetics , Phosphoproteins/genetics , Receptors, G-Protein-Coupled/genetics , Receptors, Neuropeptide/genetics , Receptors, Neurotransmitter/genetics , Animals , Bone Density , Bone and Bones/metabolism , Bone and Bones/pathology , Cell Differentiation , Cell Line , Disease Models, Animal , Female , Gene Expression Profiling , Gene Expression Regulation , Humans , Mice , Mice, Knockout , Neuropeptides/metabolism , Osteoblasts/pathology , Osteogenesis/genetics , Osteoporosis/metabolism , Osteoporosis/pathology , Phosphoproteins/classification , Phosphoproteins/metabolism , Phosphorylation , Receptors, G-Protein-Coupled/metabolism , Receptors, Neuropeptide/metabolism , Receptors, Neurotransmitter/metabolism , Signal Transduction
17.
BMC Res Notes ; 12(1): 331, 2019 Jun 11.
Article in English | MEDLINE | ID: mdl-31186065

ABSTRACT

OBJECTIVE: The bone morphogenetic protein (BMP) signaling pathway comprises the largest subdivision of the transforming growth factor (TGFß) superfamily. BMP signaling plays essential roles in both embryonic development and postnatal tissue homeostasis. Dysregulated BMP signaling underlies human pathologies ranging from pulmonary arterial hypertension to heterotopic ossification. Thus, understanding the basic mechanisms and regulation of BMP signaling may yield translational opportunities. Unfortunately, limited tools are available to evaluate this pathway, and genetic approaches are frequently confounded by developmental requirements or ability of pathway components to compensate for one another. Specific inhibitors for type 2 receptors are poorly represented. Thus, we sought to identify and validate an antibody that neutralizes the ligand-binding function of BMP receptor type 2 (BMPR2) extracellular domain (ECD). RESULTS: Using a modified, cell-free immunoprecipitation assay, we examined the neutralizing ability of the mouse monoclonal antibody 3F6 and found a dose-dependent inhibition of BMPR2-ECD ligand-binding. Consistent with this, 3F6 blocks endogenous BMPR2 function in the BMP-responsive cell line HEK293T. The specificity of 3F6 action was confirmed by demonstrating that this antibody has no effect on BMP-responsiveness in HEK293T cells in which BMPR2 expression is knocked-down. Our results provide important proof-of-concept data for future studies interrogating BMPR2 function.


Subject(s)
Antibodies, Monoclonal/immunology , Antibodies, Neutralizing/immunology , Antibody Specificity/immunology , Bone Morphogenetic Protein Receptors, Type II/immunology , Animals , Antibodies, Monoclonal/metabolism , Antibodies, Monoclonal/pharmacology , Antibodies, Neutralizing/metabolism , Antibodies, Neutralizing/pharmacology , Bone Morphogenetic Protein Receptors, Type II/antagonists & inhibitors , Bone Morphogenetic Protein Receptors, Type II/metabolism , HEK293 Cells , Humans , Mice , Protein Binding/drug effects , Protein Binding/immunology , Signal Transduction/drug effects , Signal Transduction/immunology
18.
JCI Insight ; 4(4)2019 02 21.
Article in English | MEDLINE | ID: mdl-30830862

ABSTRACT

The phosphaturic hormone FGF23 is elevated in chronic kidney disease (CKD). The risk of premature death is substantially higher in the CKD patient population, with cardiovascular disease (CVD) as the leading mortality cause at all stages of CKD. Elevated FGF23 in CKD has been associated with increased odds for all-cause mortality; however, whether FGF23 is associated with positive adaptation in CKD is unknown. To test the role of FGF23 in CKD phenotypes, a late osteoblast/osteocyte conditional flox-Fgf23 mouse (Fgf23fl/fl/Dmp1-Cre+/-) was placed on an adenine-containing diet to induce CKD. Serum analysis showed casein-fed Cre+ mice had significantly higher serum phosphate and blood urea nitrogen (BUN) versus casein diet and Cre- genotype controls. Adenine significantly induced serum intact FGF23 in the Cre- mice over casein-fed mice, whereas Cre+ mice on adenine had 90% reduction in serum intact FGF23 and C-terminal FGF23 as well as bone Fgf23 mRNA. Parathyroid hormone was significantly elevated in mice fed adenine diet regardless of genotype, which significantly enhanced midshaft cortical porosity. Echocardiographs of the adenine-fed Cre+ hearts revealed profound aortic calcification and cardiac hypertrophy versus diet and genotype controls. Thus, these studies demonstrate that increased bone FGF23, although associated with poor outcomes in CKD, is necessary to protect against the cardio-renal consequences of elevated tissue phosphate.


Subject(s)
Bone and Bones/metabolism , Cardio-Renal Syndrome/metabolism , Fibroblast Growth Factors/metabolism , Phosphates/blood , Renal Insufficiency, Chronic/metabolism , Adenine/administration & dosage , Adenine/toxicity , Animals , Bone and Bones/cytology , Cardio-Renal Syndrome/blood , Cardio-Renal Syndrome/diagnosis , Cardio-Renal Syndrome/etiology , Disease Models, Animal , Echocardiography , Female , Fibroblast Growth Factor-23 , Fibroblast Growth Factors/blood , Fibroblast Growth Factors/genetics , Humans , Mice , Mice, Transgenic , Osteocytes/metabolism , Parathyroid Hormone/blood , Parathyroid Hormone/metabolism , Phosphates/metabolism , Renal Insufficiency, Chronic/blood , Renal Insufficiency, Chronic/chemically induced , Renal Insufficiency, Chronic/complications , Vascular Calcification/blood , Vascular Calcification/etiology , Vascular Calcification/metabolism
19.
J Appl Physiol (1985) ; 126(4): 854-862, 2019 04 01.
Article in English | MEDLINE | ID: mdl-30605400

ABSTRACT

Diabetic nephropathy (DN) is a primary cause of end-stage renal disease and is becoming more prevalent because of the global rise in type 2 diabetes. A model of DN, the db/db uninephrectomized ( db/db-uni) mouse, is characterized by obesity, as well as compromised renal function. This model also manifests defects in mineral metabolism common in DN, including hyperphosphatemia, which leads to severe endocrine disease. The FGF23 coreceptor, α-Klotho, circulates as a soluble, cleaved form (cKL) and may directly influence phosphate handling. Our study sought to test the effects of cKL on mineral metabolism in db/db-uni mice. Mice were placed into either mild or moderate disease groups on the basis of the albumin-to-creatinine ratio (ACR). Body weights of db/db-uni mice were significantly greater across the study compared with lean controls regardless of disease severity. Adeno-associated cKL administration was associated with increased serum Klotho, intact, bioactive FGF23 (iFGF23), and COOH-terminal fragments of FGF23 ( P < 0.05). Blood urea nitrogen was improved after cKL administration, and cKL corrected hyperphosphatemia in the high- and low-ACR db/db-uni groups. Interestingly, 2 wk after cKL delivery, blood glucose levels were significantly reduced in db/db-uni mice with high ACR ( P < 0.05). Interestingly, several genes associated with stabilizing active iFGF23 were also increased in the osteoblastic UMR-106 cell line with cKL treatment. In summary, delivery of cKL to a model of DN normalized blood phosphate levels regardless of disease severity, supporting the concept that targeting cKL-affected pathways could provide future therapeutic avenues in DN. NEW & NOTEWORTHY In this work, systemic and continuous delivery of the "soluble" or "cleaved" form of the FGF23 coreceptor α-Klotho (cKL) via adeno-associated virus to a rodent model of diabetic nephropathy (DN), the db/db uninephrectomized mouse, normalized blood phosphate levels regardless of disease severity. This work supports the concept that targeting cKL-affected pathways could provide future therapeutic avenues for the severe mineral metabolism defects associated with DN.


Subject(s)
Diabetic Nephropathies/blood , Glucuronidase/metabolism , Phosphates/blood , Animals , Blood Glucose/metabolism , Cell Line, Tumor , Diabetes Mellitus, Type 2/blood , Diabetes Mellitus, Type 2/metabolism , Diabetic Nephropathies/metabolism , Disease Models, Animal , Fibroblast Growth Factor-23 , Fibroblast Growth Factors/metabolism , Hyperphosphatemia/blood , Hyperphosphatemia/metabolism , Klotho Proteins , Mice , Osteoblasts/metabolism , Rats
SELECTION OF CITATIONS
SEARCH DETAIL
...