Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 43
Filter
1.
Stem Cells ; 40(4): 423-434, 2022 04 29.
Article in English | MEDLINE | ID: mdl-35278073

ABSTRACT

Mesenchymal stem cells (MSCs) respond to environmental forces with both cytoskeletal re-structuring and activation of protein chaperones of mechanical information, ß-catenin, and yes-associated protein 1 (YAP1). To function, MSCs must differentiate between dynamic forces such as cyclic strains of extracellular matrix due to physical activity and static strains due to ECM stiffening. To delineate how MSCs recognize and respond differently to both force types, we compared effects of dynamic (200 cycles × 2%) and static (1 × 2% hold) strain on nuclear translocation of ß-catenin and YAP1 at 3 hours after force application. Dynamic strain induced nuclear accumulation of ß-catenin, and increased cytoskeletal actin structure and cell stiffness, but had no effect on nuclear YAP1 levels. Critically, both nuclear actin and nuclear stiffness increased along with dynamic strain-induced ß-catenin transport. Augmentation of cytoskeletal structure using either static strain or lysophosphatidic acid did not increase nuclear content of ß-catenin or actin, but induced robust nuclear increase in YAP1. As actin binds ß-catenin, we considered whether ß-catenin, which lacks a nuclear localization signal, was dependent on actin to gain entry to the nucleus. Knockdown of cofilin-1 (Cfl1) or importin-9 (Ipo9), which co-mediate nuclear transfer of G-actin, prevented dynamic strain-mediated nuclear transfer of both ß-catenin and actin. In sum, dynamic strain induction of actin re-structuring promotes nuclear transport of G-actin, concurrently supporting nuclear access of ß-catenin via mechanisms used for actin transport. Thus, dynamic and static strain activate alternative mechanoresponses reflected by differences in the cellular distributions of actin, ß-catenin, and YAP1.


Subject(s)
Mesenchymal Stem Cells , beta Catenin , Actins/metabolism , Cell Nucleus/metabolism , Cytoskeleton/metabolism , Mesenchymal Stem Cells/metabolism , beta Catenin/metabolism
2.
Stem Cells ; 38(1): 102-117, 2020 01.
Article in English | MEDLINE | ID: mdl-31648392

ABSTRACT

Nuclear actin plays a critical role in mediating mesenchymal stem cell (MSC) fate commitment. In marrow-derived MSCs, the principal diaphanous-related formin Diaph3 (mDia2) is present in the nucleus and regulates intranuclear actin polymerization, whereas Diaph1 (mDia1) is localized to the cytoplasm and controls cytoplasmic actin polymerization. We here show that mDia2 can be used as a tool to query actin-lamin nucleoskeletal structure. Silencing mDia2 affected the nucleoskeletal lamin scaffold, altering nuclear morphology without affecting cytoplasmic actin cytoskeleton, and promoted MSC differentiation. Attempting to target intranuclear actin polymerization by silencing mDia2 led to a profound loss in lamin B1 nuclear envelope structure and integrity, increased nuclear height, and reduced nuclear stiffness without compensatory changes in other actin nucleation factors. Loss of mDia2 with the associated loss in lamin B1 promoted Runx2 transcription and robust osteogenic differentiation and suppressed adipogenic differentiation. Hence, mDia2 is a potent tool to query intranuclear actin-lamin nucleoskeletal structure, and its presence serves to retain multipotent stromal cells in an undifferentiated state.


Subject(s)
Lamin Type B/metabolism , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Microtubule-Associated Proteins/metabolism , NADPH Dehydrogenase/metabolism , Actins/metabolism , Animals , Cell Differentiation/physiology , Core Binding Factor Alpha 1 Subunit/biosynthesis , Core Binding Factor Alpha 1 Subunit/genetics , Gene Knockdown Techniques , Mice , Microtubule-Associated Proteins/deficiency , Microtubule-Associated Proteins/genetics , NADPH Dehydrogenase/deficiency , NADPH Dehydrogenase/genetics , Nuclear Envelope/metabolism , Osteoblasts/cytology , Osteoblasts/metabolism , Osteogenesis
3.
Int J Mol Sci ; 22(12)2021 Jun 19.
Article in English | MEDLINE | ID: mdl-34205295

ABSTRACT

Mesenchymal stem cells (MSCs) maintain the musculoskeletal system by differentiating into multiple lineages, including osteoblasts and adipocytes. Mechanical signals, including strain and low-intensity vibration (LIV), are important regulators of MSC differentiation via control exerted through the cell structure. Lamin A/C is a protein vital to the nuclear architecture that supports chromatin organization and differentiation and contributes to the mechanical integrity of the nucleus. We investigated whether lamin A/C and mechanoresponsiveness are functionally coupled during adipogenesis in MSCs. siRNA depletion of lamin A/C increased the nuclear area, height, and volume and decreased the circularity and stiffness. Lamin A/C depletion significantly decreased markers of adipogenesis (adiponectin, cellular lipid content) as did LIV treatment despite depletion of lamin A/C. Phosphorylation of focal adhesions in response to mechanical challenge was also preserved during loss of lamin A/C. RNA-seq showed no major adipogenic transcriptome changes resulting from LIV treatment, suggesting that LIV regulation of adipogenesis may not occur at the transcriptional level. We observed that during both lamin A/C depletion and LIV, interferon signaling was downregulated, suggesting potentially shared regulatory mechanism elements that could regulate protein translation. We conclude that the mechanoregulation of adipogenesis and the mechanical activation of focal adhesions function independently from those of lamin A/C.


Subject(s)
Adipogenesis , Focal Adhesions/physiology , Lamin Type A/physiology , Mesenchymal Stem Cells/physiology , Animals , Elastic Modulus , Interferons/metabolism , Male , Membrane Proteins/metabolism , Mice , Signal Transduction , Telomere-Binding Proteins/metabolism , Vibration
4.
Stem Cells ; 35(6): 1624-1635, 2017 06.
Article in English | MEDLINE | ID: mdl-28371128

ABSTRACT

Actin structure contributes to physiologic events within the nucleus to control mesenchymal stromal cell (MSC) differentiation. Continuous cytochalasin D (Cyto D) disruption of the MSC actin cytoskeleton leads to osteogenic or adipogenic differentiation, both requiring mass transfer of actin into the nucleus. Cyto D remains extranuclear, thus intranuclear actin polymerization is potentiated by actin transfer: we asked whether actin structure affects differentiation. We show that secondary actin filament branching via the Arp2/3 complex is required for osteogenesis and that preventing actin branching stimulates adipogenesis, as shown by expression profiling of osteogenic and adipogenic biomarkers and unbiased RNA-seq analysis. Mechanistically, Cyto D activates osteoblast master regulators (e.g., Runx2, Sp7, Dlx5) and novel coregulated genes (e.g., Atoh8, Nr4a3, Slfn5). Formin-induced primary actin filament formation is critical for Arp2/3 complex recruitment: osteogenesis is prevented by silencing of the formin mDia1, but not its paralog mDia2. Furthermore, while inhibition of actin, branching is a potent adipogenic stimulus, silencing of either mDia1 or mDia2 blocks adipogenic gene expression. We propose that mDia1, which localizes in the cytoplasm of multipotential MSCs and traffics into the nucleus after cytoskeletal disruption, joins intranuclear mDia2 to facilitate primary filament formation before mediating subsequent branching via Arp2/3 complex recruitment. The resulting intranuclear branched actin network specifies osteogenic differentiation, while actin polymerization in the absence of Arp2/3 complex-mediated secondary branching causes adipogenic differentiation. Stem Cells 2017;35:1624-1635.


Subject(s)
Actins/metabolism , Cell Differentiation , Cell Nucleus/metabolism , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Adipogenesis/drug effects , Animals , Cell Differentiation/drug effects , Cell Nucleus/drug effects , Cytochalasin D/pharmacology , Gene Silencing , Indoles/pharmacology , Mesenchymal Stem Cells/drug effects , Mice , Osteogenesis/drug effects , PPAR gamma/metabolism , Polymerization
5.
J Biol Chem ; 291(34): 17829-47, 2016 08 19.
Article in English | MEDLINE | ID: mdl-27402842

ABSTRACT

Terminal differentiation of multipotent stem cells is achieved through a coordinated cascade of activated transcription factors and epigenetic modifications that drive gene transcription responsible for unique cell fate. Within the mesenchymal lineage, factors such as RUNX2 and PPARγ are indispensable for osteogenesis and adipogenesis, respectively. We therefore investigated genomic binding of transcription factors and accompanying epigenetic modifications that occur during osteogenic and adipogenic differentiation of mouse bone marrow-derived mesenchymal stem cells (MSCs). As assessed by ChIP-sequencing and RNA-sequencing analyses, we found that genes vital for osteogenic identity were linked to RUNX2, C/EBPß, retinoid X receptor, and vitamin D receptor binding sites, whereas adipocyte differentiation favored PPARγ, retinoid X receptor, C/EBPα, and C/EBPß binding sites. Epigenetic marks were clear predictors of active differentiation loci as well as enhancer activities and selective gene expression. These marrow-derived MSCs displayed an epigenetic pattern that suggested a default preference for the osteogenic pathway; however, these patterns were rapidly altered near the Adipoq, Cidec, Fabp4, Lipe, Plin1, Pparg, and Cebpa genes during adipogenic differentiation. Surprisingly, we found that these cells also exhibited an epigenetic plasticity that enabled them to trans-differentiate from adipocytes to osteoblasts (and vice versa) after commitment, as assessed by staining, gene expression, and ChIP-quantitative PCR analysis. The osteogenic default pathway may be subverted during pathological conditions, leading to skeletal fragility and increased marrow adiposity during aging, estrogen deficiency, and skeletal unloading. Taken together, our data provide an increased mechanistic understanding of the epigenetic programs necessary for multipotent differentiation of MSCs that may prove beneficial in the development of therapeutic strategies.


Subject(s)
Adipogenesis/physiology , Bone Marrow Cells/metabolism , Cell Differentiation/physiology , Epigenesis, Genetic/physiology , Mesenchymal Stem Cells/metabolism , Osteogenesis/physiology , Adipocytes/cytology , Adipocytes/metabolism , Animals , Bone Marrow Cells/cytology , Female , Mesenchymal Stem Cells/cytology , Mice , Osteoblasts/cytology , Osteoblasts/metabolism
6.
Trans Am Clin Climatol Assoc ; 128: 180-192, 2017.
Article in English | MEDLINE | ID: mdl-28790502

ABSTRACT

Stem cells respond to environmental signals that induce their differentiation to cells that make up specialized tissues and organs. Our laboratory has focused on bone marrow mesenchymal stem cells (MSCs) that supply bone osteoblasts and marrow adipocytes, an output that appears to be reciprocal. Case in point: exercise promotes osteogenesis and bone formation, and inhibits marrow adipose accrual. A mechanically induced signal pathway concentrating on preserving ß-catenin also causes increased structure of the actin cytoskeleton, both of which inhibit adipogenesis. Recently we showed that intranuclear actin is as important to MSC lineage decisions as cytoplasmic actin. This opens up new areas for understanding gene expression in stem cells.


Subject(s)
Actins/physiology , Cell Differentiation/physiology , Cell Nucleus/physiology , Mesenchymal Stem Cells/physiology , Animals , Bone Marrow/physiology , Bone and Bones/physiology , Humans , Motor Activity
7.
Stem Cells ; 33(10): 3065-76, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26140478

ABSTRACT

Depolymerization of the actin cytoskeleton induces nuclear trafficking of regulatory proteins and global effects on gene transcription. We here show that in mesenchymal stem cells (MSCs), cytochalasin D treatment causes rapid cofilin-/importin-9-dependent transfer of G-actin into the nucleus. The continued presence of intranuclear actin, which forms rod-like structures that stain with phalloidin, is associated with induction of robust expression of the osteogenic genes osterix and osteocalcin in a Runx2-dependent manner, and leads to acquisition of osteogenic phenotype. Adipogenic differentiation also occurs, but to a lesser degree. Intranuclear actin leads to nuclear export of Yes-associated protein (YAP); maintenance of nuclear YAP inhibits Runx2 initiation of osteogenesis. Injection of cytochalasin into the tibial marrow space of live mice results in abundant bone formation within the space of 1 week. In sum, increased intranuclear actin forces MSC into osteogenic lineage through controlling Runx2 activity; this process may be useful for clinical objectives of forming bone.


Subject(s)
Actin Cytoskeleton/metabolism , Actins/metabolism , Core Binding Factor Alpha 1 Subunit/genetics , Mesenchymal Stem Cells/metabolism , Osteogenesis/genetics , Actin Cytoskeleton/genetics , Actin Depolymerizing Factors/metabolism , Animals , Cell Lineage/genetics , Cell Nucleus/genetics , Cell Nucleus/metabolism , Core Binding Factor Alpha 1 Subunit/metabolism , Cytochalasin D/administration & dosage , Mice , Phalloidine/metabolism , Protein Transport/genetics
8.
Stem Cells ; 33(6): 2063-76, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25787126

ABSTRACT

A cell's ability to recognize and adapt to the physical environment is central to its survival and function, but how mechanical cues are perceived and transduced into intracellular signals remains unclear. In mesenchymal stem cells (MSCs), high-magnitude substrate strain (HMS, ≥2%) effectively suppresses adipogenesis via induction of focal adhesion (FA) kinase (FAK)/mTORC2/Akt signaling generated at FAs. Physiologic systems also rely on a persistent barrage of low-level signals to regulate behavior. Exposing MSC to extremely low-magnitude mechanical signals (LMS) suppresses adipocyte formation despite the virtual absence of substrate strain (<0.001%), suggesting that LMS-induced dynamic accelerations can generate force within the cell. Here, we show that MSC response to LMS is enabled through mechanical coupling between the cytoskeleton and the nucleus, in turn activating FAK and Akt signaling followed by FAK-dependent induction of RhoA. While LMS and HMS synergistically regulated FAK activity at the FAs, LMS-induced actin remodeling was concentrated at the perinuclear domain. Preventing nuclear-actin cytoskeleton mechanocoupling by disrupting linker of nucleoskeleton and cytoskeleton (LINC) complexes inhibited these LMS-induced signals as well as prevented LMS repression of adipogenic differentiation, highlighting that LINC connections are critical for sensing LMS. In contrast, FAK activation by HMS was unaffected by LINC decoupling, consistent with signal initiation at the FA mechanosome. These results indicate that the MSC responds to its dynamic physical environment not only with "outside-in" signaling initiated by substrate strain, but vibratory signals enacted through the LINC complex enable matrix independent "inside-inside" signaling.


Subject(s)
Cell Nucleus/metabolism , Cytoskeleton/metabolism , Mesenchymal Stem Cells/cytology , Adipocytes/metabolism , Adipogenesis/physiology , Animals , Cells, Cultured , Humans , Mice, Inbred C57BL
9.
Stem Cells ; 31(11): 2528-37, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23836527

ABSTRACT

Mechanical strain provides an anti-adipogenic, pro-osteogenic stimulus to mesenchymal stem cells (MSC) through generating intracellular signals and via cytoskeletal restructuring. Recently, mTORC2 has been shown to be a novel mechanical target critical for the anti-adipogenic signal leading to preservation of ß-catenin. As mechanical activation of mTORC2 requires focal adhesions (FAs), we asked whether proximal signaling involved Src and FAK, which are early responders to integrin-FA engagement. Application of mechanical strain to marrow-derived MSCs was unable to activate mTORC2 when Src family kinases were inhibited. Fyn, but not Src, was specifically required for mechanical activation of mTORC2 and was recruited to FAs after strain. Activation of mTORC2 was further diminished following FAK inhibition, and as FAK phosphorylation (Tyr-397) required Fyn activity, provided evidence of Fyn/FAK cooperativity. Inhibition of Fyn also prevented mechanical activation of RhoA as well as mechanically induced actin stress fiber formation. We thus asked whether RhoA activation by strain was dependent on mTORC2 downstream of Fyn. Inhibition of mTORC2 or its downstream substrate, Akt, both prevented mechanical RhoA activation, indicating that Fyn/FAK affects cytoskeletal structure via mTORC2. We then sought to ascertain whether this Fyn-initiated signal pathway modulated MSC lineage decisions. siRNA knockdown of Fyn, but not Src, led to rapid attainment of adipogenic phenotype with significant increases in adipocyte protein 2, peroxisome proliferator-activated receptor gamma, adiponectin, and perilipin. As such, Fyn expression in mdMSCs contributes to basal cytoskeletal architecture and, when associated with FAs, functions as a proximal mechanical effector for environmental signals that influence MSC lineage allocation.


Subject(s)
Adipogenesis/physiology , Mesenchymal Stem Cells/metabolism , Multiprotein Complexes/metabolism , Proto-Oncogene Proteins c-fyn/metabolism , TOR Serine-Threonine Kinases/metabolism , rhoA GTP-Binding Protein/metabolism , Cell Culture Techniques , Humans , MCF-7 Cells , Mechanistic Target of Rapamycin Complex 2 , Mesenchymal Stem Cells/cytology , Multiprotein Complexes/genetics , Phosphorylation , Proto-Oncogene Proteins c-fyn/genetics , Signal Transduction , TOR Serine-Threonine Kinases/genetics , Transfection
10.
Nat Commun ; 15(1): 4095, 2024 May 15.
Article in English | MEDLINE | ID: mdl-38750021

ABSTRACT

Polymerized ß-actin may provide a structural basis for chromatin accessibility and actin transport into the nucleus can guide mesenchymal stem cell (MSC) differentiation. Using MSC, we show that using CK666 to inhibit Arp2/3 directed secondary actin branching results in decreased nuclear actin structure, and significantly alters chromatin access measured with ATACseq at 24 h. The ATAC-seq results due to CK666 are distinct from those caused by cytochalasin D (CytoD), which enhances nuclear actin structure. In addition, nuclear visualization shows Arp2/3 inhibition decreases pericentric H3K9me3 marks. CytoD, alternatively, induces redistribution of H3K27me3 marks centrally. Such alterations in chromatin landscape are consistent with differential gene expression associated with distinctive differentiation patterns. Further, knockdown of the non-enzymatic monomeric actin binding protein, Arp4, leads to extensive chromatin unpacking, but only a modest increase in transcription, indicating an active role for actin-Arp4 in transcription. These data indicate that dynamic actin remodeling can regulate chromatin interactions.


Subject(s)
Actin-Related Protein 2-3 Complex , Actins , Cell Nucleus , Chromatin , Mesenchymal Stem Cells , Actins/metabolism , Chromatin/metabolism , Cell Nucleus/metabolism , Actin-Related Protein 2-3 Complex/metabolism , Actin-Related Protein 2-3 Complex/genetics , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/cytology , Animals , Cell Differentiation , Cytochalasin D/pharmacology , Histones/metabolism , Humans , Microfilament Proteins/metabolism , Microfilament Proteins/genetics , Mice , Chromatin Assembly and Disassembly
11.
Obes Facts ; 17(2): 145-157, 2024.
Article in English | MEDLINE | ID: mdl-38224679

ABSTRACT

INTRODUCTION: Longitudinal effect of diet-induced obesity on bone is uncertain. Prior work showed both no effect and a decrement in bone density or quality when obesity begins prior to skeletal maturity. We aimed to quantify long-term effects of obesity on bone and bone marrow adipose tissue (BMAT) in adulthood. METHODS: Skeletally mature, female C57BL/6 mice (n = 70) aged 12 weeks were randomly allocated to low-fat diet (LFD; 10% kcal fat; n = 30) or high-fat diet (HFD; 60% kcal fat; n = 30), with analyses at 12, 15, 18, and 24 weeks (n = 10/group). Tibial microarchitecture was analyzed by µCT, and volumetric BMAT was quantified via 9.4T MRI/advanced image analysis. Histomorphometry of adipocytes and osteoclasts, and qPCR were performed. RESULTS: Body weight and visceral white adipose tissue accumulated in response to HFD started in adulthood. Trabecular bone parameters declined with advancing experimental age. BV/TV declined 22% in LFD (p = 0.0001) and 17% in HFD (p = 0.0022) by 24 weeks. HFD failed to appreciably alter BV/TV and had negligible impact on other microarchitecture parameters. Both dietary intervention and age accounted for variance in BMAT, with regional differences: distal femoral BMAT was more responsive to diet, while proximal femoral BMAT was more attenuated by age. BMAT increased 60% in the distal metaphysis in HFD at 18 and 24 weeks (p = 0.0011). BMAT in the proximal femoral diaphysis, unchanged by diet, decreased 45% due to age (p = 0.0002). Marrow adipocyte size via histomorphometry supported MRI quantification. Osteoclast number did not differ between groups. Tibial qPCR showed attenuation of some adipose, metabolism, and bone genes. A regulator of fatty acid ß-oxidation, cytochrome C (CYCS), was 500% more abundant in HFD bone (p < 0.0001; diet effect). CYCS also increased due to age, but to a lesser extent. HFD mildly increased OCN, TRAP, and SOST. CONCLUSIONS: Long-term high fat feeding after skeletal maturity, despite upregulation of visceral adiposity, body weight, and BMAT, failed to attenuate bone microarchitecture. In adulthood, we found aging to be a more potent regulator of microarchitecture than diet-induced obesity.


Subject(s)
Adiposity , Osteoporosis , Mice , Animals , Female , Bone Marrow/metabolism , Mice, Inbred C57BL , Obesity/etiology , Obesity/metabolism , Adipose Tissue/metabolism , Body Weight , Osteoporosis/metabolism , Diet, High-Fat/adverse effects
12.
J Biol Chem ; 286(45): 39450-6, 2011 Nov 11.
Article in English | MEDLINE | ID: mdl-21956113

ABSTRACT

Mechanical signals can inactivate glycogen synthase kinase 3ß (GSK3ß), resulting in stabilization of ß-catenin. This signaling cascade is necessary for the inhibition of adipogenesis in mesenchymal stem cells (MSC) that is produced by a daily strain regimen. We investigated whether Akt is the mechanically activated kinase responsible for phosphorylation and inactivation of GSK3ß in MSC. Mechanical strain (2% magnitude, 0.17 Hz) induced phosphorylation of Akt at Ser-473 and Thr-308 in parallel with phosphorylation of GSK3ß at Ser-9. Inhibiting Akt (Akt1/2 kinase inhibitor treatment or Akt knockdown) prevented strain-induced phosphorylation of GSK3ß at Ser-9. Inhibition of PI3K prevented Thr-308 phosphorylation, but strain-induced Ser-473 phosphorylation was measurable and induced phosphorylation of GSK3ß, suggesting that Ser-473 phosphorylation is sufficient for the downstream mechanoresponse. As Rictor/mTORC2 (mammalian target of rapamycin complex 2) is known to transduce phosphorylation of Akt at Ser-473 by insulin, we investigated whether it contributes to strain-induced Ser-473 phosphorylation. Phosphorylation of Ser-473 by both mechanical and insulin treatment in MSC was prevented by the mTOR inhibitor KU0063794. When mTORC2 was blocked, mechanical GSK3ß inactivation was prevented, whereas insulin inhibition of GSK3ß was still measured in the absence of Ser-473 phosphorylation, presumably through phosphorylation of Akt at Thr-308. In sum, mechanical input initiates a signaling cascade that is uniquely dependent on mTORC2 activation and phosphorylation of Akt at Ser-473, an effect sufficient to cause inactivation of GSK3ß. Thus, mechanical regulation of GSK3ß downstream of Akt is dependent on phosphorylation of Akt at Ser-473 in a manner distinct from that of growth factors. As such, Akt reveals itself to be a pleiotropic signaling molecule whose downstream targets are differentially regulated depending upon the nature of the activating input.


Subject(s)
Glycogen Synthase Kinase 3/metabolism , Mechanotransduction, Cellular/physiology , Mesenchymal Stem Cells/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Trans-Activators/metabolism , Animals , Carrier Proteins/metabolism , Cells, Cultured , Enzyme Activation/drug effects , Enzyme Activation/physiology , Glycogen Synthase Kinase 3 beta , Mechanotransduction, Cellular/drug effects , Mesenchymal Stem Cells/cytology , Mice , Morpholines/pharmacology , Phosphorylation/drug effects , Phosphorylation/physiology , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Pyrimidines/pharmacology , Rapamycin-Insensitive Companion of mTOR Protein , Serine/metabolism , TOR Serine-Threonine Kinases/antagonists & inhibitors , TOR Serine-Threonine Kinases/metabolism , Trans-Activators/antagonists & inhibitors , Transcription Factors
13.
Stem Cells ; 29(11): 1829-36, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21898699

ABSTRACT

The fate of pluripotent mesenchymal stem cells (MSC) is determined through integration of chemical, spatial, and physical signals. The suppression of MSC adipogenesis by mechanical stimuli, which requires Akt-induced inhibition of glycogen synthase kinase 3ß (GSK3ß) with ß-catenin activation, can be enhanced by repetitive dosing within a single day. Here, we demonstrate that reapplication of cyclic strain within a 24-hour period leads to amplification of both Akt activation and its subsequent inhibition of GSK3ß, such that total cycle number can be reduced while still inhibiting adipogenesis. Amplification of Akt signaling is facilitated by a dynamic restructuring of the cell in response to mechanical signals, as evidenced by a transient increase in focal adhesion (FA) number and increased RhoA activity. Preventing FA assembly or development of tension blocks activation of Akt by mechanical signals, but not by insulin. This indicates that the FA infrastructure is essential to the physical, but not necessarily the chemical, sensitivity, and responsiveness of the cell. Exploiting the transient nature of cytoskeletal remodeling may represent a process to enhance cell responsiveness to mechanical input and ultimately define the fate of MSCs with a minimal input.


Subject(s)
Adipocytes/cytology , Focal Adhesions/metabolism , Mesenchymal Stem Cells/cytology , Pluripotent Stem Cells/cytology , Stress, Mechanical , Adipocytes/metabolism , Animals , Cells, Cultured , Mesenchymal Stem Cells/metabolism , Mice , Pluripotent Stem Cells/metabolism , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction
14.
JBMR Plus ; 5(3): e10450, 2021 Mar.
Article in English | MEDLINE | ID: mdl-33778320

ABSTRACT

Use of the selective estrogen receptor modulator Tamoxifen (TAM) is a mainstay to induce conditional expression of Cre recombinase in transgenic laboratory mice. To excise ß-catenin fl/fl in 28-day-old male and female Prrx1-CreER/ß-catenin fl/fl mice (C57BL/6), we utilized TAM at 150 mg/kg; despite ß-catenin knockout in MSC, we found a significant increase in trabecular and cortical bone volume in all genders. Because TAM was similarly anabolic in KO and control mice, we investigated a dose effect on bone formation by treating wild-type mice (WT C57BL/6, 4 weeks) with TAM (total dose 0, 20, 40, 200 mg/kg via four injections). TAM increased bone in a dose-dependent manner analyzed by micro-computed tomography (µCT), which showed that, compared to control, 20 mg/kg TAM increased femoral bone volume fraction (bone volume/total volume [BV/TV]) (21.6% ± 1.5% to 33% ± 2.5%; 153%, p < 0.005). With TAM 40 mg/kg and 200 mg/kg, BV/TV increased to 48.1% ± 4.4% (223%, p < 0.0005) and 58% ± 3.8% (269%, p < 0.0001) respectively, compared to control. Osteoblast markers increased with 200 mg/kg TAM: Dlx5 (224%, p < 0.0001), Alp (166%, p < 0.0001), Bglap (223%, p < 0.0001), and Sp7 (228%, p < 0.0001). Osteoclasts per bone surface (Oc#/BS) nearly doubled at the lowest TAM dose (20 mg/kg), but decreased to <20% control with 200 mg/kg TAM. Our data establish that use of TAM at even very low doses to excise a floxed target in postnatal mice has profound effects on trabecular and cortical bone formation. As such, TAM treatment is a major confounder in the interpretation of bone phenotypes in conditional gene knockout mouse models. © 2020 The Authors. JBMR Plus published by Wiley Periodicals LLC. on behalf of American Society for Bone and Mineral Research.

15.
Front Endocrinol (Lausanne) ; 12: 782194, 2021.
Article in English | MEDLINE | ID: mdl-35145475

ABSTRACT

Exercise, typically beneficial for skeletal health, has not yet been studied in lipodystrophy, a condition characterized by paucity of white adipose tissue, with eventual diabetes, and steatosis. We applied a mouse model of global deficiency of Bscl2 (SEIPIN), required for lipid droplet formation. Male twelve-week-old B6 knockouts (KO) and wild type (WT) littermates were assigned six-weeks of voluntary, running exercise (E) versus non-exercise (N=5-8). KO weighed 14% less than WT (p=0.01) and exhibited an absence of epididymal adipose tissue; KO liver Plin1 via qPCR was 9-fold that of WT (p=0.04), consistent with steatosis. Bone marrow adipose tissue (BMAT), unlike white adipose, was measurable, although 40.5% lower in KO vs WT (p=0.0003) via 9.4T MRI/advanced image analysis. SEIPIN ablation's most notable effect marrow adiposity was in the proximal femoral diaphysis (-56% KO vs WT, p=0.005), with relative preservation in KO-distal-femur. Bone via µCT was preserved in SEIPIN KO, though some quality parameters were attenuated. Running distance, speed, and time were comparable in KO and WT. Exercise reduced weight (-24% WT-E vs WT p<0.001) but not in KO. Notably, exercise increased trabecular BV/TV in both (+31%, KO-E vs KO, p=0.004; +14%, WT-E vs WT, p=0.006). The presence and distribution of BMAT in SEIPIN KO, though lower than WT, is unexpected and points to a uniqueness of this depot. That trabecular bone increases were achievable in both KO and WT, despite a difference in BMAT quantity/distribution, points to potential metabolic flexibility during exercise-induced skeletal anabolism.


Subject(s)
Adipose Tissue/metabolism , Bone Marrow/metabolism , Cancellous Bone/metabolism , Femur/metabolism , GTP-Binding Protein gamma Subunits/genetics , Lipodystrophy/metabolism , Physical Conditioning, Animal , Adipose Tissue/diagnostic imaging , Adipose Tissue/pathology , Animals , Body Weight , Bone Marrow/diagnostic imaging , Bone Marrow/pathology , Cancellous Bone/diagnostic imaging , Diaphyses/diagnostic imaging , Disease Models, Animal , Epididymis/metabolism , Epididymis/pathology , Femur/diagnostic imaging , Lipodystrophy/diagnostic imaging , Lipodystrophy/genetics , Lipodystrophy/pathology , Male , Mice , Mice, Knockout , Organ Size , Perilipin-1/genetics , X-Ray Microtomography
16.
J Biol Chem ; 284(50): 34607-17, 2009 Dec 11.
Article in English | MEDLINE | ID: mdl-19840939

ABSTRACT

Mechanical stimulation can prevent adipogenic and improve osteogenic lineage allocation of mesenchymal stem cells (MSC), an effect associated with the preservation of beta-catenin levels. We asked whether mechanical up-regulation of beta-catenin was critical to reduction in adipogenesis as well as other mechanical events inducing alternate MSC lineage selection. In MSC cultured under strong adipogenic conditions, mechanical load (3600 cycles/day, 2% strain) inactivated GSK3beta in a Wnt-independent fashion. Small interfering RNA targeting GSK3beta prevented both strain-induced induction of beta-catenin and an increase in COX2, a factor associated with increased osteoprogenitor phenotype. Small interfering RNA knockdown of beta-catenin blocked mechanical reduction of peroxisome proliferator-activated receptor gamma and adiponectin, implicating beta-catenin in strain inhibition of adipogenesis. In contrast, the effect of both mechanical and pharmacologic inhibition of GSK3beta on the putative beta-catenin target, COX2, was unaffected by beta-catenin knockdown. GSK3beta inhibition caused accumulation of nuclear NFATc1; mechanical strain increased nuclear NFATc1, independent of beta-catenin. NFATc1 knockdown prevented mechanical stimulation of COX2, implicating NFATc1 signaling. Finally, inhibition of GSK3beta caused association of RNA polymerase II with the COX2 gene, suggesting transcription initiation. These results demonstrate that mechanical inhibition of GSK3beta induces activation of both beta-catenin and NFATc1 signaling, limiting adipogenesis via the former and promoting osteoblastic differentiation via NFATc1/COX2. Our novel findings suggest that mechanical loading regulates mesenchymal stem cell differentiation through inhibition of GSK3beta, which in turn regulates multiple downstream effectors.


Subject(s)
Glycogen Synthase Kinase 3/metabolism , Mesenchymal Stem Cells/physiology , NFATC Transcription Factors/metabolism , Signal Transduction/physiology , Stress, Mechanical , beta Catenin/metabolism , Adipogenesis/physiology , Adiponectin/genetics , Adiponectin/metabolism , Animals , Bone Marrow Cells/cytology , Bone Marrow Cells/physiology , Calcineurin/metabolism , Calcium/metabolism , Cell Line , Cyclooxygenase 2/genetics , Cyclooxygenase 2/metabolism , Gene Expression Regulation , Glycogen Synthase Kinase 3/genetics , Mesenchymal Stem Cells/cytology , Mice , NFATC Transcription Factors/genetics , PPAR gamma/genetics , PPAR gamma/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , beta Catenin/genetics
17.
J Cell Biochem ; 111(4): 1042-50, 2010 Nov 01.
Article in English | MEDLINE | ID: mdl-20672310

ABSTRACT

Regulation of mesenchymal stem cell (MSC) lineage selection is important for the generation of bone mass. Inhibition of cyclooxygenase-2 (COX2) may increase adipogenesis at the cost of decreasing osteoprogenitor output. Here we investigated the role of COX2 and its products during MSC differentiation. Indomethacin stimulated adipogenesis (increased aP2, adiponectin and lipid droplets) of CH310T1/2 stem cells as well as marrow-derived MSCs to a degree similar to the PPARγ2 ligand, rosiglitazone. Unlike rosiglitazone, indomethacin significantly upregulated PPARγ2 expression. Indomethacin and the COX2 specific inhibitor celecoxib suppressed PGE2 production, but celecoxib did not induce adipogenesis. As well, addition of PGE2 failed to reverse indomethacin induced adipogenesis, indicating that indomethacin's effects were prostaglandin independent. In MSCs over-expressing PPARγ2 and RXRα, indomethacin did not increase PPAR-induced transcription, while rosiglitazone and 15d-PGJ2 did (1.7- and 1.3-fold, respectively, P < 0.001). We considered whether indomethacin might directly affect C/EBPß proximally to PPARγ2 induction. Indomethacin significantly increased C/EBPß expression and protein within 24 h of addition. These results indicate that indomethacin promotes adipogenesis by increasing C/EBPß and PPARγ2 expression in a prostaglandin-independent fashion. This effect of indomethacin is pertinent to potential deleterious effects of this commonly used anti-inflammatory drug on bone remodeling and tissue healing.


Subject(s)
Adipogenesis/drug effects , Indomethacin/pharmacology , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/enzymology , Animals , CCAAT-Enhancer-Binding Protein-beta/metabolism , Celecoxib , Cell Line , Cyclooxygenase 2/genetics , Cyclooxygenase 2/metabolism , Dinoprostone/metabolism , Dose-Response Relationship, Drug , Gene Expression Regulation, Enzymologic/drug effects , Ligands , Mesenchymal Stem Cells/drug effects , Mice , PPAR gamma/genetics , PPAR gamma/metabolism , Pyrazoles/pharmacology , Response Elements/genetics , Sulfonamides/pharmacology , Transcriptional Activation/drug effects , Transcriptional Activation/genetics , beta Catenin/metabolism
18.
J Bone Miner Res ; 35(1): 106-115, 2020 01.
Article in English | MEDLINE | ID: mdl-31509274

ABSTRACT

Marrow adipose tissue (MAT) and its relevance to skeletal health during caloric restriction (CR) is unknown: It remains unclear whether exercise, which is anabolic to bone in a calorie-replete state, alters bone or MAT in CR. We hypothesized that response of bone and MAT to exercise in CR differs from the calorie-replete state. Ten-week-old female B6 mice fed a regular diet (RD) or 30% CR diet were allocated to sedentary (RD, CR, n = 10/group) or running exercise (RD-E, CR-E, n = 7/group). After 6 weeks, CR mice weighed 20% less than RD, p < 0.001; exercise did not affect weight. Femoral bone volume (BV) via 3D MRI was 20% lower in CR versus RD (p < 0.0001). CR was associated with decreased bone by µCT: Tb.Th was 16% less in CR versus RD, p < 0.003, Ct.Th was 5% less, p < 0.07. In CR-E, Tb.Th was 40% less than RD-E, p < 0.0001. Exercise increased Tb.Th in RD (+23% RD-E versus RD, p < 0.003) but failed to do so in CR. Cortical porosity increased after exercise in CR (+28%, p = 0.04), suggesting exercise during CR is deleterious to bone. In terms of bone fat, metaphyseal MAT/ BV rose 159% in CR versus RD, p = 0.003 via 3D MRI. Exercise decreased MAT/BV by 52% in RD, p < 0.05, and also suppressed MAT in CR (-121%, p = 0.047). Histomorphometric analysis of adipocyte area correlated with MAT by MRI (R2 = 0.6233, p < 0.0001). With respect to bone, TRAP and Sost mRNA were reduced in CR. Intriguingly, the repressed Sost in CR rose with exercise and may underlie the failure of CR-bone quantity to increase in response to exercise. Notably, CD36, a marker of fatty acid uptake, rose 4088% in CR (p < 0.01 versus RD), suggesting that basal increases in MAT during calorie restriction serve to supply local energy needs and are depleted during exercise with a negative impact on bone. © 2019 The Authors. Journal of Bone and Mineral Research published by American Society for Bone and Mineral Research.


Subject(s)
Bone Marrow , Caloric Restriction , Adipocytes , Adipose Tissue , Animals , Bone Marrow/diagnostic imaging , Bone and Bones/diagnostic imaging , Female , Mice
19.
J Bone Miner Res ; 35(6): 1149-1162, 2020 06.
Article in English | MEDLINE | ID: mdl-32022326

ABSTRACT

During bone marrow stromal cell (BMSC) differentiation, both Wnt signaling and the development of a rigid cytoskeleton promote commitment to the osteoblastic over adipogenic lineage. ß-catenin plays a critical role in the Wnt signaling pathway to facilitate downstream effects on gene expression. We show that ß-catenin was additive with cytoskeletal signals to prevent adipogenesis, and ß-catenin knockdown promoted adipogenesis even when the actin cytoskeleton was depolymerized. ß-catenin also prevented osteoblast commitment in a cytoskeletal-independent manner, with ß-catenin knockdown enhancing lineage commitment. Chromatin immunoprecipitation (ChIP)-sequencing demonstrated binding of ß-catenin to the promoter of enhancer of zeste homolog 2 (EZH2), a key component of the polycomb repressive complex 2 (PRC2) complex that catalyzes histone methylation. Knockdown of ß-catenin reduced EZH2 protein levels and decreased methylated histone 3 (H3K27me3) at osteogenic loci. Further, when EZH2 was inhibited, ß-catenin's anti-differentiation effects were lost. These results indicate that regulating EZH2 activity is key to ß-catenin's effects on BMSCs to preserve multipotentiality. © 2020 American Society for Bone and Mineral Research.


Subject(s)
Bone Marrow Cells , Enhancer of Zeste Homolog 2 Protein , Mesenchymal Stem Cells , beta Catenin/metabolism , Animals , Bone Marrow Cells/metabolism , Catenins , Cell Differentiation , Enhancer of Zeste Homolog 2 Protein/metabolism , Mesenchymal Stem Cells/metabolism , Mice , Osteogenesis , Polycomb Repressive Complex 2/metabolism , Wnt Signaling Pathway
20.
Endocrinology ; 149(12): 6065-75, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18687779

ABSTRACT

The ability of exercise to decrease fat mass and increase bone mass may occur through mechanical biasing of mesenchymal stem cells (MSCs) away from adipogenesis and toward osteoblastogenesis. C3H10T1/2 MSCs cultured in highly adipogenic medium express peroxisome proliferator-activated receptor gamma and adiponectin mRNA and protein, and accumulate intracellular lipid. Mechanical strain applied for 6 h daily inhibited expression of peroxisome proliferator-activated receptor gamma and adiponectin mRNA by up to 35 and 50%, respectively, after 5 d. A decrease in active and total beta-catenin levels during adipogenic differentiation was entirely prevented by daily application of mechanical strain; furthermore, strain induced beta-catenin nuclear translocation. Inhibition of glycogen synthase kinase-3beta by lithium chloride or SB415286 also prevented adipogenesis, suggesting that preservation of beta-catenin levels was important to strain inhibition of adipogenesis. Indeed, mechanical strain inactivated glycogen synthase kinase-3beta, which was preceded by Akt activation, indicating that strain transmits antiadipogenic signals through this pathway. Cells grown under adipogenic conditions showed no increase in osteogenic markers runt-related transcription factor (Runx) 2 and osterix (Osx); subsequent addition of bone morphogenetic protein 2 for 2 d increased Runx2 but not Osx expression in unstrained cultures. When cultures were strained for 5 d before bone morphogenetic protein 2 addition, Runx2 mRNA increased more than in unstrained cultures, and Osx expression more than doubled. As such, mechanical strain enhanced MSC potential to enter the osteoblast lineage despite exposure to adipogenic conditions. Our results indicate that MSC commitment to adipogenesis can be suppressed by mechanical signals, allowing other signals to promote osteoblastogenesis. These data suggest that positive effects of exercise on both fat and bone may occur during mesenchymal lineage selection.


Subject(s)
Adipogenesis/physiology , Mesenchymal Stem Cells/metabolism , beta Catenin/metabolism , Adipogenesis/drug effects , Adiponectin/genetics , Adiponectin/metabolism , Animals , Blotting, Western , Bone Morphogenetic Protein 2/pharmacology , Cell Differentiation/drug effects , Cell Differentiation/physiology , Cell Line , Core Binding Factor Alpha 1 Subunit/genetics , Core Binding Factor Alpha 1 Subunit/metabolism , Culture Media/pharmacology , Fluorescent Antibody Technique , Gene Expression/drug effects , Glycogen Synthase Kinase 3/antagonists & inhibitors , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta , Lithium Chloride/pharmacology , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/drug effects , Mice , PPAR gamma/genetics , Phosphorylation/drug effects , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/drug effects , Signal Transduction/physiology , Sp7 Transcription Factor , Stress, Mechanical , Transcription Factors/genetics , Transcription Factors/metabolism , beta Catenin/genetics , beta Catenin/physiology
SELECTION OF CITATIONS
SEARCH DETAIL