1
|
Hagan ML, Tuladhar A, Yu K, Alhamad DW, Bensreti H, Dorn J, Piedra VM, Cantu N, Stokes EG, Blumenthal D, Roberts RL, Balayan V, Bass SM, Dickerson T, Cartelle AL, Montesinos-Cartagena M, Awad ME, Castro AA, Garland T, Cooley MA, Johnson M, Hamrick MW, McNeil PL, McGee-Lawrence ME. Osteocyte Sptbn1 Deficiency Alters Cell Survival and Mechanotransduction Following Formation of Plasma Membrane Disruptions (PMD) from Mechanical Loading. Calcif Tissue Int 2024; 115:725-743. [PMID: 39276238 DOI: 10.1007/s00223-024-01285-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Accepted: 08/30/2024] [Indexed: 09/16/2024]
Abstract
We and others have shown that application of high-level mechanical loading promotes the formation of transient plasma membrane disruptions (PMD) which initiate mechanotransduction. We hypothesized that increasing osteocyte cell membrane fragility, by disrupting the cytoskeleton-associated protein β2-spectrin (Sptbn1), could alter osteocytic responses and bone adaptation to loading in a PMD-related fashion. In MLO-Y4 cells, treatment with the spectrin-disrupting agent diamide or knockdown of Sptbn1 via siRNA increased the number of PMD formed by fluid shear stress. Primary osteocytes from an osteocyte-targeted DMP1-Cre Sptbn1 conditional knockout (CKO) model mimicked trends seen with diamide and siRNA treatment and suggested the creation of larger PMD, which repaired more slowly, for a given level of stimulus. Post-wounding cell survival was impaired in all three models, and calcium signaling responses from the wounded osteocyte were mildly altered in Sptbn1 CKO cultures. Although Sptbn1 CKO mice did not demonstrate an altered skeletal phenotype as compared to WT littermates under baseline conditions, they showed a blunted increase in cortical thickness when subjected to an osteogenic tibial loading protocol as well as evidence of increased osteocyte death (increased lacunar vacancy) in the loaded limb after 2 weeks of loading. The impaired post-wounding cell viability and impaired bone adaptation seen with Sptbn1 disruption support the existence of an important role for Sptbn1, and PMD formation, in osteocyte mechanotransduction and bone adaptation to mechanical loading.
Collapse
Affiliation(s)
- Mackenzie L Hagan
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, 1460 Laney Walker Blvd, CB1101, Augusta, GA, 30912, USA
| | - Anik Tuladhar
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, 1460 Laney Walker Blvd, CB1101, Augusta, GA, 30912, USA
| | - Kanglun Yu
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, 1460 Laney Walker Blvd, CB1101, Augusta, GA, 30912, USA
| | - Dima W Alhamad
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, 1460 Laney Walker Blvd, CB1101, Augusta, GA, 30912, USA
| | - Husam Bensreti
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, 1460 Laney Walker Blvd, CB1101, Augusta, GA, 30912, USA
| | - Jennifer Dorn
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, 1460 Laney Walker Blvd, CB1101, Augusta, GA, 30912, USA
| | - Victor M Piedra
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, 1460 Laney Walker Blvd, CB1101, Augusta, GA, 30912, USA
| | - Nicholas Cantu
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, 1460 Laney Walker Blvd, CB1101, Augusta, GA, 30912, USA
| | - Eric G Stokes
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, 1460 Laney Walker Blvd, CB1101, Augusta, GA, 30912, USA
| | - Daniel Blumenthal
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, 1460 Laney Walker Blvd, CB1101, Augusta, GA, 30912, USA
| | - Rachel L Roberts
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, 1460 Laney Walker Blvd, CB1101, Augusta, GA, 30912, USA
| | - Vanshika Balayan
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, 1460 Laney Walker Blvd, CB1101, Augusta, GA, 30912, USA
| | - Sarah M Bass
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, 1460 Laney Walker Blvd, CB1101, Augusta, GA, 30912, USA
| | - Thomas Dickerson
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, 1460 Laney Walker Blvd, CB1101, Augusta, GA, 30912, USA
| | - Anabel Liyen Cartelle
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, 1460 Laney Walker Blvd, CB1101, Augusta, GA, 30912, USA
| | - Marlian Montesinos-Cartagena
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, 1460 Laney Walker Blvd, CB1101, Augusta, GA, 30912, USA
| | - Mohamed E Awad
- Department of Oral Biology and Diagnostic Sciences, Dental College of Georgia, Augusta University, Augusta, GA, USA
| | - Alberto A Castro
- Evolution Ecology & Organismal Biology Department, University of California Riverside, Riverside, USA
| | - Theodore Garland
- Evolution Ecology & Organismal Biology Department, University of California Riverside, Riverside, USA
| | - Marion A Cooley
- Department of Oral Biology and Diagnostic Sciences, Dental College of Georgia, Augusta University, Augusta, GA, USA
| | - Maribeth Johnson
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Mark W Hamrick
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, 1460 Laney Walker Blvd, CB1101, Augusta, GA, 30912, USA
| | - Paul L McNeil
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, 1460 Laney Walker Blvd, CB1101, Augusta, GA, 30912, USA
| | - Meghan E McGee-Lawrence
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, 1460 Laney Walker Blvd, CB1101, Augusta, GA, 30912, USA.
- Department of Orthopaedic Surgery, Augusta University, Augusta, GA, USA.
| |
Collapse
|
2
|
Wee NKY, McGregor NE, Walker EC, Poulton IJ, Dang MKM, Gooi JH, Phillips WA, Sims NA. Direct activation of PI3K in osteoblasts and osteocytes strengthens murine bone through sex-specific actions on cortical surfaces. J Bone Miner Res 2024; 39:1174-1187. [PMID: 38959852 DOI: 10.1093/jbmr/zjae102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Revised: 06/06/2024] [Accepted: 07/01/2024] [Indexed: 07/05/2024]
Abstract
Intracellular phosphoinositide 3-kinase (PI3K) signaling is activated by multiple bone-active receptors. Genetic mutations activating PI3K signaling are associated with clinical syndromes of tissue overgrowth in multiple organs, often including the skeleton. While one formation is increased by removing the PI3K inhibitor (phosphatase and TENsin homolog deleted on chromosome 10 (PTEN)), the effect of direct PI3K activation in the osteoblast lineage has not been reported. We introduced a known gain-of-function mutation in Pik3ca, the gene encoding the p110α catalytic subunit of PI3K, in osteocytes and late osteoblasts using the dentin matrix protein-1 Cre (Dmp1Cre) mouse and assessed the skeletal phenotype. Femur shape was grossly normal, but cortical thickness was significantly greater in both male and female Dmp1Cre.Pik3caH1047R mice, leading to almost doubled bone strength at 12 wk of age. Both sexes had smaller marrow areas from 6 wk of age. Female mice also exhibited greater cross-sectional area, which continued to increase until 24 wk of age, resulting in a further increase in bone strength. Although both male and female mice had increased endocortical mineralizing surface, only female mice had increased periosteal mineralizing surface. The bone formed in the Dmp1Cre.Pik3caH1047R mice showed no increase in intracortical remodeling nor any defect in cortical bone consolidation. In contrast, on both endocortical and periosteal surfaces, there was more lamellar bone formation, including highly organized osteocyte networks extending along the entire surface at a greater thickness than in control mice. In conclusion, direct activation of PI3Kα in cells targeted by Dmp1Cre leads to high cortical bone mass and strength with abundant lamellar cortical bone in female and male mice with no increase in intracortical remodeling. This differs from the effect of PTEN deletion in the same cells, suggesting that activating PI3Kα in osteoblasts and osteocytes may be a more suitable target to promote formation of lamellar bone.
Collapse
Affiliation(s)
- Natalie K Y Wee
- Bone Cell Biology and Disease Unit, St Vincent's Institute of Medical Research, 9 Princes St, Fitzroy 3065, Victoria, Australia
| | - Narelle E McGregor
- Bone Cell Biology and Disease Unit, St Vincent's Institute of Medical Research, 9 Princes St, Fitzroy 3065, Victoria, Australia
| | - Emma C Walker
- Bone Cell Biology and Disease Unit, St Vincent's Institute of Medical Research, 9 Princes St, Fitzroy 3065, Victoria, Australia
| | - Ingrid J Poulton
- Bone Cell Biology and Disease Unit, St Vincent's Institute of Medical Research, 9 Princes St, Fitzroy 3065, Victoria, Australia
| | - Michelle Kieu Mi Dang
- Bone Cell Biology and Disease Unit, St Vincent's Institute of Medical Research, 9 Princes St, Fitzroy 3065, Victoria, Australia
| | - Jonathan H Gooi
- Structural Biology Unit, St Vincent's Institute of Medical Research, 9 Princes St, Fitzroy 3065, Victoria, Australia
| | - Wayne A Phillips
- Cancer Biology and Surgical Oncology Laboratory, Peter MacCallum Cancer Centre, 305 Grattan St, Melbourne 3000, Victoria, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville 3010, Victoria, Australia
| | - Natalie A Sims
- Bone Cell Biology and Disease Unit, St Vincent's Institute of Medical Research, 9 Princes St, Fitzroy 3065, Victoria, Australia
- Department of Medicine at St. Vincent's Hospital, The University of Melbourne, Fitzroy 3065, Victoria, Australia
- Mary Mackillop Institute for Health Research, Australian Catholic University, Melbourne 3065, Victoria, Australia
| |
Collapse
|
3
|
Makareeva E, Sousa M, Kent T, de Castro LF, Collins MT, Leikin S. RNA-based bone histomorphometry: method and its application to explaining postpubertal bone gain in a G610C mouse model of osteogenesis imperfecta. J Bone Miner Res 2024; 39:177-189. [PMID: 38477760 PMCID: PMC11207954 DOI: 10.1093/jbmr/zjad004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Revised: 11/03/2023] [Accepted: 11/10/2023] [Indexed: 03/14/2024]
Abstract
Bone histomorphometry is a well-established approach to assessing skeletal pathology, providing a standard evaluation of the cellular components, architecture, mineralization, and growth of bone tissue. However, it depends in part on the subjective interpretation of cellular morphology by an expert, which introduces bias. In addition, diseases like osteogenesis imperfecta (OI) and fibrous dysplasia are accompanied by changes in the morphology and function of skeletal tissue and cells, hindering consistent evaluation of some morphometric parameters and interpretation of the results. For instance, traditional histomorphometry combined with collagen turnover markers suggested that reduced bone formation in classical OI is accompanied by increased bone resorption. In contrast, the well-documented postpubertal reduction in fractures would be easier to explain by reduced bone resorption after puberty, highlighting the need for less ambiguous measurements. Here we propose an approach to histomorphometry based on in situ mRNA hybridization, which uses Col1a1 as osteoblast and Ctsk as osteoclast markers. This approach can be fully automated and eliminates subjective identification of bone surface cells. We validate these markers based on the expression of Bglap, Ibsp, and Acp5. Comparison with traditional histological and tartrate-resistant acid phosphatase staining of the same sections suggests that mRNA-based analysis is more reliable. Unlike inconclusive traditional histomorphometry of mice with α2(I)-Gly610 to Cys substitution in the collagen triple helix, mRNA-based measurements reveal reduced osteoclastogenesis in 11-wk-old animals consistent with the postpubertal catch-up osteogenesis observed by microCT. We optimize the technique for cryosections of mineralized bone and sections of paraffin-embedded decalcified tissue, simplifying and broadening its applications. We illustrate the application of the mRNA-based approach to human samples using the example of a McCune-Albright syndrome patient. By eliminating confounding effects of altered cellular morphology and the need for subjective morphological evaluation, this approach may provide a more reproducible and accessible evaluation of bone pathology.
Collapse
Affiliation(s)
- Elena Makareeva
- Eunice Kennedy Shriver National institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, United States
| | - Megan Sousa
- Eunice Kennedy Shriver National institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, United States
| | - Tristan Kent
- National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD 20892, United States
| | - Luis F de Castro
- National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD 20892, United States
| | - Michael T Collins
- National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD 20892, United States
| | - Sergey Leikin
- Eunice Kennedy Shriver National institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, United States
| |
Collapse
|
4
|
Shahrajabian MH, Sun W. The Significance and Importance of dPCR, qPCR, and SYBR Green PCR Kit in the Detection of Numerous Diseases. Curr Pharm Des 2024; 30:169-179. [PMID: 38243947 DOI: 10.2174/0113816128276560231218090436] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 10/27/2023] [Accepted: 11/07/2023] [Indexed: 01/22/2024]
Abstract
Digital PCR (dPCR) is the latest technique that has become commercially accessible for various types of research. This method uses Taq polymerase in a standard polymerase chain reaction (PCR) to amplify a target DNA fragment from a complex sample, like quantitative PCR (qPCR) and droplet digital PCR (dd- PCR). ddPCR may facilitate microRNA (miRNA) measurement, particularly in liquid biopsy, because it has been proven to be more effective and sensitive, and in this method, ddPCR can provide an unprecedented chance for deoxyribonucleic acid (DNA) methylation research because of its capability to increase sensitivity and precision over conventional PCR-based methods. qPCR has also been found to be a valuable standard technique to measure both copy DNA (cDNA) and genomic DNA (gDNA) levels, although the finding data can be significantly variable and non-reproducible without relevant validation and verification of both primers and samples. The SYBR green quantitative real-time PCR (qPCR) method has been reported as an appropriate technique for quantitative detection and species discrimination, and has been applied profitably in different experiments to determine, quantify, and discriminate species. Although both TaqMan qRT-PCR and SYBR green qRT-PCR are sensitive and rapid, the SYBR green qRT-PCR assay is easy and the TaqMan qRT-PCR assay is specific but expensive due to the probe required. This review aimed to introduce dPCR, qPCR, SYBR green PCR kit, and digital PCR, compare them, and also introduce their advantages in the detection of different diseases.
Collapse
Affiliation(s)
- Mohamad Hesam Shahrajabian
- National Key Laboratory of Agricultural Microbiology, Biotechnology Research Institute, Chinese Academy of Agricultural Sciences, Beijing 100086, China
| | - Wenli Sun
- National Key Laboratory of Agricultural Microbiology, Biotechnology Research Institute, Chinese Academy of Agricultural Sciences, Beijing 100086, China
| |
Collapse
|
5
|
Abstract
Although osteoblasts and osteocytes are descended from the same lineage, they each have unique and essential roles in bone. Targeting gene deletion to osteoblasts and osteocytes using the Cre/loxP system has greatly increased our current understanding of how these cells function. Additionally, the use of the Cre/loxP system in conjunction with cell-specific reporters has enabled lineage tracing of these bone cells both in vivo and ex vivo. However, concerns have been raised regarding the specificity of the promoters used and the resulting off-target effects on cells within and outside of the bone. In this review, we have summarized the main mouse models that have been used to determine the functions of specific genes in osteoblasts and osteocytes. We discuss the expression patterns and specificity of the different promoter fragments during osteoblast to osteocyte differentiation in vivo. We also highlight how their expression in non-skeletal tissues may complicate the interpretation of study results. A thorough understanding of when and where these promoters are activated will enable improved study design and greater confidence in data interpretation.
Collapse
Affiliation(s)
- Y Kitase
- Indiana Center for Musculoskeletal Health, Department of Anatomy, Cell Biology and Physiology, School of Medicine, Indiana University, Indianapolis, IN 46202, United States of America
| | - M Prideaux
- Indiana Center for Musculoskeletal Health, Department of Anatomy, Cell Biology and Physiology, School of Medicine, Indiana University, Indianapolis, IN 46202, United States of America.
| |
Collapse
|
6
|
Cunningham CJ, Choi RB, Bullock WA, Robling AG. Perspective: The current state of Cre driver mouse lines in skeletal research: Challenges and opportunities. Bone 2023; 170:116719. [PMID: 36868507 PMCID: PMC10087282 DOI: 10.1016/j.bone.2023.116719] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 02/15/2023] [Accepted: 02/19/2023] [Indexed: 03/04/2023]
Abstract
The Cre/Lox system has revolutionized the ability of biomedical researchers to ask very specific questions about the function of individual genes in specific cell types at specific times during development and/or disease progression in a variety of animal models. This is true in the skeletal biology field, and numerous Cre driver lines have been created to foster conditional gene manipulation in specific subpopulations of bone cells. However, as our ability to scrutinize these models increases, an increasing number of issues have been identified with most driver lines. All existing skeletal Cre mouse models exhibit problems in one or more of the following three areas: (1) cell type specificity-avoiding Cre expression in unintended cell types; (2) Cre inducibility-improving the dynamic range for Cre in inducible models (negligible Cre activity before induction and high Cre activity after induction); and (3) Cre toxicity-reducing the unwanted biological effects of Cre (beyond loxP recombination) on cellular processes and tissue health. These issues are hampering progress in understanding the biology of skeletal disease and aging, and consequently, identification of reliable therapeutic opportunities. Skeletal Cre models have not advanced technologically in decades despite the availability of improved tools, including multi-promoter-driven expression of permissive or fragmented recombinases, new dimerization systems, and alternative forms of recombinases and DNA sequence targets. We review the current state of skeletal Cre driver lines, and highlight some of the successes, failures, and opportunities to improve fidelity in the skeleton, based on successes pioneered in other areas of biomedical science.
Collapse
Affiliation(s)
- Connor J Cunningham
- Department of Anatomy, Cell Biology & Physiology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Roy B Choi
- Department of Anatomy, Cell Biology & Physiology, Indiana University School of Medicine, Indianapolis, IN, USA
| | | | - Alexander G Robling
- Department of Anatomy, Cell Biology & Physiology, Indiana University School of Medicine, Indianapolis, IN, USA; Richard L. Roudebush Veterans Affairs Medical Center, Indianapolis, IN, USA; Department of Biomedical Engineering, Indiana University-Purdue University at Indianapolis, Indianapolis, IN, USA; Indiana Center for Musculoskeletal Health, Indianapolis, IN, USA.
| |
Collapse
|
7
|
Rajpar I, Kumar G, Fortina P, Tomlinson RE. Toll-like receptor 4 signaling in osteoblasts is required for load-induced bone formation in mice. iScience 2023; 26:106304. [PMID: 36950122 PMCID: PMC10025993 DOI: 10.1016/j.isci.2023.106304] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Revised: 01/06/2023] [Accepted: 02/24/2023] [Indexed: 03/06/2023] Open
Abstract
In mature bone, NGF is produced by osteoblasts following mechanical loading and signals through resident sensory nerves expressing its high affinity receptor, neurotrophic tyrosine kinase receptor type 1 (TrkA), to support bone formation. Here, we investigated whether osteoblastic expression of Toll-like receptor 4 (TLR4), a key receptor in the NF-κB signaling pathway, is required to initiate NGF-TrkA signaling required for load-induced bone formation. Although Tlr4 conditional knockout mice have normal skeletal mass and strength in adulthood, the loss of TLR4 signaling significantly reduced lamellar bone formation following loading. Inhibition of TLR4 signaling reduced Ngf expression in primary osteoblasts and RNA sequencing of bones from Tlr4 conditional knockout mice and wild-type littermates revealed dysregulated inflammatory signaling three days after osteogenic mechanical loading. In total, our study reveals an important role for osteoblastic TLR4 in the skeletal adaptation of bone to mechanical forces.
Collapse
Affiliation(s)
- Ibtesam Rajpar
- Department of Orthopaedic Surgery, Thomas Jefferson University, Philadelphia, PA, USA
| | - Gaurav Kumar
- Department of Cancer Biology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Paolo Fortina
- Department of Cancer Biology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Ryan E. Tomlinson
- Department of Orthopaedic Surgery, Thomas Jefferson University, Philadelphia, PA, USA
| |
Collapse
|
8
|
Xu X, Yang H, Bullock WA, Gallant MA, Ohlsson C, Bellido TM, Main RP. Osteocyte Estrogen Receptor β (Ot-ERβ) Regulates Bone Turnover and Skeletal Adaptive Response to Mechanical Loading Differently in Male and Female Growing and Adult Mice. J Bone Miner Res 2023; 38:186-197. [PMID: 36321245 PMCID: PMC10108310 DOI: 10.1002/jbmr.4731] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Revised: 10/15/2022] [Accepted: 10/29/2022] [Indexed: 11/06/2022]
Abstract
Age-related bone loss is a failure of balanced bone turnover and diminished skeletal mechanoadaptation. Estrogen receptors, ERα and ERβ, play critical roles in osteoprotective regulation activated by estrogen and mechanical signals. Previous studies mainly focused on ERα and showed that osteocyte-ERα (Ot-ERα) regulated trabecular, but not cortical bone, and played a minor role in load-induced cortical adaptation. However, the role of Ot-ERβ in bone mass regulation remains unrevealed. To address this issue, we characterized bone (re)modeling and gene expression in male and female mice with Ot-ERβ deletion (ERβ-dOT) and littermate control (LC) at 10 weeks (young) or 28 weeks (adult) of age, as well as their responses to in vivo tibial compressive loading. Increased cancellous bone mass appeared in the L4 vertebral body of young male ERβ-dOT mice. At the same time, femoral cortical bone gene expression showed signs consistent with elevated osteoblast and osteoclast activities (type-I collagen, Cat K, RANKL). Upregulated androgen receptor (AR) expression was observed in young male ERβ-dOT mice relative to LC, suggesting a compensatory effect of testosterone on male bone protection. In contrast, bone mass in L4 decreased in adult male ERβ-dOT mice, attributed to potentially increased bone resorption activity (Cat K) with no change in bone formation. There was no effect of ERβ-dOT on bone mass or gene expression in female mice. Sex-dependent regulation of Ot-ERβ also appeared in load-induced cortical responsiveness. Young female ERβ-dOT mice showed an enhanced tibial cortical anabolic adaptation compared with LC. In contrast, an attenuated cortical anabolic response presented at the proximal tibia in male ERβ-dOT mice at both ages. For the first time, our findings suggest that Ot-ERβ regulates bone (re)modeling and the response to mechanical signals through different mechanisms in males and females. © 2022 The Authors. Journal of Bone and Mineral Research published by Wiley Periodicals LLC on behalf of American Society for Bone and Mineral Research (ASBMR).
Collapse
Affiliation(s)
- Xiaoyu Xu
- Weldon School of Biomedical EngineeringPurdue UniversityWest LafayetteINUSA
- Musculoskeletal Biology and Mechanics Lab, Department of Basic Medical SciencesPurdue UniversityWest LafayetteINUSA
| | - Haisheng Yang
- Department of Biomedical Engineering, Faculty of Environment and LifeBeijing University of TechnologyBeijingChina
| | | | - Maxim A. Gallant
- Musculoskeletal Biology and Mechanics Lab, Department of Basic Medical SciencesPurdue UniversityWest LafayetteINUSA
| | - Claes Ohlsson
- Sahlgrenska Osteoporosis Centre, Centre for Bone and Arthritis Research, Department of Internal Medicine and Clinical NutritionInstitute of Medicine, Sahlgrenska Academy, University of GothenburgGothenburgSweden
- Department of Drug TreatmentSahlgrenska University HospitalGothenburgSweden
| | - Teresita M. Bellido
- Department of Physiology and Cell BiologyUniversity of Arkansas for Medical SciencesLittle RockARUSA
| | - Russell P. Main
- Weldon School of Biomedical EngineeringPurdue UniversityWest LafayetteINUSA
- Musculoskeletal Biology and Mechanics Lab, Department of Basic Medical SciencesPurdue UniversityWest LafayetteINUSA
| |
Collapse
|
9
|
Chai W, Hao W, Liu J, Han Z, Chang S, Cheng L, Sun M, Yan G, Liu Z, Liu Y, Zhang G, Xing L, Chen H, Liu P. Visualizing Cathepsin K-Cre Expression at the Single-Cell Level with GFP Reporters. JBMR Plus 2022; 7:e10706. [PMID: 36699636 PMCID: PMC9850439 DOI: 10.1002/jbm4.10706] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Revised: 11/10/2022] [Accepted: 11/16/2022] [Indexed: 11/23/2022] Open
Abstract
The Cre/lox system is a fundamental tool for functional genomic studies, and a number of Cre lines have been generated to target genes of interest spatially and temporally in defined cells or tissues; this approach has greatly expanded our knowledge of gene functions. However, the limitations of this system have recently been recognized, and we must address the challenge of so-called nonspecific/off-target effects when a Cre line is utilized to investigate a gene of interest. For example, cathepsin K (Ctsk) has been used as a specific osteoclast marker, and Cre driven by its promoter is widely utilized for osteoclast investigations. However, Ctsk-Cre expression has recently been identified in other cell types, such as osteocytes, periosteal stem cells, and tenocytes. To better understand Ctsk-Cre expression and ensure appropriate use of this Cre line, we performed a comprehensive analysis of Ctsk-Cre expression at the single-cell level in major organs and tissues using two green fluorescent protein (GFP) reporters (ROSA nT-nG and ROSA tdT) and a tissue clearing technique in young and aging mice. The expression profile was further verified by immunofluorescence staining and droplet digital RT-PCR. The results demonstrate that Ctsk-Cre is expressed not only in osteoclasts but also at various levels in osteoblast lineage cells and other major organs/tissues, particularly in the brain, kidney, pancreas, and blood vessels. Furthermore, Ctsk-Cre expression increases markedly in the bone marrow, skeletal muscle, and intervertebral discs in aging mice. These data will be valuable for accurately interpreting data obtained from in vivo studies using Ctsk-Cre mice to avoid potentially misleading conclusions. © 2022 The Authors. JBMR Plus published by Wiley Periodicals LLC on behalf of American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Wenhuan Chai
- Laboratory of Bone & Adipose BiologyShanxi Medical UniversityTaiyuanChina
| | - Weiwei Hao
- Laboratory of Bone & Adipose BiologyShanxi Medical UniversityTaiyuanChina
| | - Jintao Liu
- Laboratory of Bone & Adipose BiologyShanxi Medical UniversityTaiyuanChina
| | - Zhenglin Han
- Laboratory of Bone & Adipose BiologyShanxi Medical UniversityTaiyuanChina
| | - Shiyu Chang
- Laboratory of Bone & Adipose BiologyShanxi Medical UniversityTaiyuanChina
| | - Liben Cheng
- Laboratory of Bone & Adipose BiologyShanxi Medical UniversityTaiyuanChina
| | - Mingxin Sun
- Laboratory of Bone & Adipose BiologyShanxi Medical UniversityTaiyuanChina
| | - Guofang Yan
- Laboratory of Bone & Adipose BiologyShanxi Medical UniversityTaiyuanChina
| | - Zemin Liu
- Laboratory of Bone & Adipose BiologyShanxi Medical UniversityTaiyuanChina
| | - Yin Liu
- Laboratory of Bone & Adipose BiologyShanxi Medical UniversityTaiyuanChina
| | - Guodong Zhang
- Laboratory of Bone & Adipose BiologyShanxi Medical UniversityTaiyuanChina
| | - Li Xing
- Laboratory of Bone & Adipose BiologyShanxi Medical UniversityTaiyuanChina
| | - Hongqian Chen
- Laboratory of Bone & Adipose BiologyShanxi Medical UniversityTaiyuanChina
| | - Peng Liu
- Laboratory of Bone & Adipose BiologyShanxi Medical UniversityTaiyuanChina
| |
Collapse
|
10
|
Chicana B, Abbasizadeh N, Burns C, Taglinao H, Spencer JA, Manilay JO. Deletion of Vhl in Dmp1-Expressing Cells Causes Microenvironmental Impairment of B Cell Lymphopoiesis. Front Immunol 2022; 13:780945. [PMID: 35250971 PMCID: PMC8889104 DOI: 10.3389/fimmu.2022.780945] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Accepted: 01/11/2022] [Indexed: 12/25/2022] Open
Abstract
The contributions of skeletal cells to the processes of B cell development in the bone marrow (BM) have not been completely described. The von-Hippel Lindau protein (VHL) plays a key role in cellular responses to hypoxia. Previous work showed that Dmp1-Cre;Vhl conditional knockout mice (VhlcKO), which deletes Vhl in subsets of mesenchymal stem cells, late osteoblasts and osteocytes, display dysregulated bone growth and reduction in B cells. Here, we investigated the mechanisms underlying the B cell defects using flow cytometry and high-resolution imaging. In the VhlcKO BM, B cell progenitors were increased in frequency and number, whereas Hardy Fractions B-F were decreased. VhlcKO Fractions B-C cells showed increased apoptosis and quiescence. Reciprocal BM chimeras confirmed a B cell-extrinsic source of the VhlcKO B cell defects. In support of this, VhlcKO BM supernatant contained reduced CXCL12 and elevated EPO levels. Intravital and ex vivo imaging revealed VhlcKO BM blood vessels with increased diameter, volume, and a diminished blood-BM barrier. Staining of VhlcKO B cells with an intracellular hypoxic marker indicated the natural existence of distinct B cell microenvironments that differ in local oxygen tensions and that the B cell developmental defects in VhlcKO BM are not initiated by hypoxia. Our studies identify novel mechanisms linking altered bone homeostasis with drastic BM microenvironmental changes that dysregulate B cell development.
Collapse
Affiliation(s)
- Betsabel Chicana
- Department of Molecular and Cell Biology, School of Natural Sciences, University of California, Merced, Merced, CA, United States.,Quantitative and Systems Biology Graduate Program, University of California, Merced, Merced, CA, United States
| | - Nastaran Abbasizadeh
- Quantitative and Systems Biology Graduate Program, University of California, Merced, Merced, CA, United States.,Department of Bioengineering, School of Engineering, University of California, Merced, Merced, CA, United States
| | - Christian Burns
- Quantitative and Systems Biology Graduate Program, University of California, Merced, Merced, CA, United States.,Department of Bioengineering, School of Engineering, University of California, Merced, Merced, CA, United States
| | - Hanna Taglinao
- Department of Molecular and Cell Biology, School of Natural Sciences, University of California, Merced, Merced, CA, United States
| | - Joel A Spencer
- Quantitative and Systems Biology Graduate Program, University of California, Merced, Merced, CA, United States.,Department of Bioengineering, School of Engineering, University of California, Merced, Merced, CA, United States.,Bioengineering Graduate Program, University of California, Merced, Merced, CA, United States
| | - Jennifer O Manilay
- Department of Molecular and Cell Biology, School of Natural Sciences, University of California, Merced, Merced, CA, United States.,Quantitative and Systems Biology Graduate Program, University of California, Merced, Merced, CA, United States
| |
Collapse
|
11
|
Couasnay G, Madel MB, Lim J, Lee B, Elefteriou F. Sites of Cre-recombinase activity in mouse lines targeting skeletal cells. J Bone Miner Res 2021; 36:1661-1679. [PMID: 34278610 DOI: 10.1002/jbmr.4415] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 07/12/2021] [Accepted: 07/15/2021] [Indexed: 12/22/2022]
Abstract
The Cre/Lox system is a powerful tool in the biologist's toolbox, allowing loss-of-function and gain-of-function studies, as well as lineage tracing, through gene recombination in a tissue-specific and inducible manner. Evidence indicates, however, that Cre transgenic lines have a far more nuanced and broader pattern of Cre activity than initially thought, exhibiting "off-target" activity in tissues/cells other than the ones they were originally designed to target. With the goal of facilitating the comparison and selection of optimal Cre lines to be used for the study of gene function, we have summarized in a single manuscript the major sites and timing of Cre activity of the main Cre lines available to target bone mesenchymal stem cells, chondrocytes, osteoblasts, osteocytes, tenocytes, and osteoclasts, along with their reported sites of "off-target" Cre activity. We also discuss characteristics, advantages, and limitations of these Cre lines for users to avoid common risks related to overinterpretation or misinterpretation based on the assumption of strict cell-type specificity or unaccounted effect of the Cre transgene or Cre inducers. © 2021 American Society for Bone and Mineral Research (ASBMR).
Collapse
Affiliation(s)
- Greig Couasnay
- Department of Orthopedic Surgery, Baylor College of Medicine, Houston, TX, USA
| | | | - Joohyun Lim
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - Brendan Lee
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - Florent Elefteriou
- Department of Orthopedic Surgery, Baylor College of Medicine, Houston, TX, USA
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| |
Collapse
|
12
|
Kwang D, Tjin G, Purton LE. Regulation of murine B lymphopoiesis by stromal cells. Immunol Rev 2021; 302:47-67. [PMID: 34002391 DOI: 10.1111/imr.12973] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 04/21/2021] [Accepted: 04/30/2021] [Indexed: 12/21/2022]
Abstract
B lymphocytes are crucial for the body's humoral immune response, secreting antibodies generated against foreign antigens to fight infection. Adult murine B lymphopoiesis is initiated in the bone marrow and additional maturation occurs in the spleen. In both these organs, B lymphopoiesis involves interactions with numerous different non-hematopoietic cells, also known as stromal or microenvironment cells, which provide migratory, maturation, and survival signals. A variety of conditional knockout and transgenic mouse models have been used to identify the roles of distinct microenvironment cell types in the regulation of B lymphopoiesis. These studies have revealed that mesenchymal lineage cells and endothelial cells comprise the non-hematopoietic microenvironment cell types that support B lymphopoiesis in the bone marrow. In the spleen, various types of stromal cells and endothelial cells contribute to B lymphocyte maturation. More recently, comprehensive single cell RNA-seq studies have also been used to identify clusters of stromal cell types in the bone marrow and spleen, which will aid in further identifying key regulators of B lymphopoiesis. Here, we review the different types of microenvironment cells and key extrinsic regulators that are known to be involved in the regulation of murine B lymphopoiesis in the bone marrow and spleen.
Collapse
Affiliation(s)
- Diannita Kwang
- Stem Cell Regulation Unit, St. Vincent's Institute of Medical Research, Fitzroy, Vic., Australia
| | - Gavin Tjin
- Stem Cell Regulation Unit, St. Vincent's Institute of Medical Research, Fitzroy, Vic., Australia
| | - Louise E Purton
- Stem Cell Regulation Unit, St. Vincent's Institute of Medical Research, Fitzroy, Vic., Australia.,Department of Medicine at St. Vincent's Hospital, The University of Melbourne, Fitzroy, Vic., Australia
| |
Collapse
|
13
|
Blei F. Update December 2020. Lymphat Res Biol 2020. [DOI: 10.1089/lrb.2020.29096.fb] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
|