Skip to main content
Environmental Health Perspectives logoLink to Environmental Health Perspectives
. 2023 Oct 4;131(10):107002. doi: 10.1289/EHP11646

Ambient PM2.5 Exposure and Bone Homeostasis: Analysis of UK Biobank Data and Experimental Studies in Mice and in Vitro

Qinwen Ge 1,*, Sijia Yang 2,*, Yu Qian 3,4,*, Jiali Chen 1, Wenhua Yuan 1, Sanduo Li 2, Pinger Wang 1, Ran Li 2,5, Lu Zhang 2,5, Guobo Chen 6, Haidong Kan 7, Sanjay Rajagopalan 8, Qinghua Sun 2,5, Hou-Feng Zheng 3,4,, Hongting Jin 1,, Cuiqing Liu 2,5,
PMCID: PMC10549986  PMID: 37792558

Abstract

Background:

Previous evidence has identified exposure to fine ambient particulate matter (PM2.5) as a leading risk factor for adverse health outcomes. However, to date, only a few studies have examined the potential association between long-term exposure to PM2.5 and bone homeostasis.

Objective:

We sought to examine the relationship between long-term PM2.5 exposure and bone health and explore its potential mechanism.

Methods:

This research included both observational and experimental studies. First, based on human data from UK Biobank, linear regression was used to explore the associations between long-term exposure to PM2.5 (i.e., annual average PM2.5 concentration for 2010) and bone mineral density [BMD; i.e., heel BMD (n=37,440) and femur neck and lumbar spine BMD (n=29,766)], which were measured during 2014–2020. For the experimental animal study, C57BL/6 male mice were assigned to ambient PM2.5 or filtered air for 6 months via a whole-body exposure system. Micro-computed tomography analyses were applied to measure BMD and bone microstructures. Biomarkers for bone turnover and inflammation were examined with histological staining, immunohistochemistry staining, and enzyme-linked immunosorbent assay. We also performed tartrate-resistant acid phosphatase (TRAP) staining and bone resorption assay to determine the effect of PM2.5 exposure on osteoclast activity in vitro. In addition, the potential downstream regulators were assessed by real-time polymerase chain reaction and western blot.

Results:

We observed that long-term exposure to PM2.5 was significantly associated with lower BMD at different anatomical sites, according to the analysis of UK Biobank data. In experimental study, mice exposed long-term to PM2.5 exhibited excessive osteoclastogenesis, dysregulated osteogenesis, higher tumor necrosis factor-alpha (TNF-α) expression, and shorter femur length than control mice, but they demonstrated no significant differences in femur structure or BMD. In vitro, cells stimulated with conditional medium of PM2.5-stimulated macrophages had aberrant osteoclastogenesis and differences in the protein/mRNA expression of members of the TNF-α/Traf6/c-Fos pathway, which could be partially rescued by TNF-α inhibition.

Discussion:

Our prospective observational evidence suggested that long-term exposure to PM2.5 is associated with lower BMD and further experimental results demonstrated exposure to PM2.5 could disrupt bone homeostasis, which may be mediated by inflammation-induced osteoclastogenesis. https://doi.org/10.1289/EHP11646

Introduction

Age-associated skeletal diseases, such as osteoporosis and fragility fracture,1 have become important public health concerns with high prevalence and health expenditures around the world.2 Osteoporosis is a common skeletal disease characterized by decreased bone mineral density (BMD) and higher osteoporotic fracture risk.3 The risk of fracture increases 2.4- and 2-fold for every 1-standard deviation (SD) decrease in femoral bone mineral density in women (0.13g/cm2) and men (0.16g/cm2), respectively.4 It was estimated that >25 million people were affected by osteoporosis and 3.5 million incident fragility fracture cases happened in the European Union in 2010, costing 37.4 billion.5 Therefore, it is essential to identify the modifiable risk factors associated with osteoporosis and fragility fracture for the prevention of the diseases.

Bone is regarded as a dynamic organ, modeling and remodeling to grow and change shape throughout life.6 Bone turnover, consisting of osteoblastic bone formation and osteoclastic bone resorption, is essential to skeletal health by regulating bone metabolism.7 Osteoblasts and osteoclasts take part in the process of cartilage-to-bone transition for bone development, as well as in removing old bone tissue and laying down new bone tissue for bone homeostasis.8 Although these two kinds of cells demonstrate different forms of action in the bone modeling and remodeling processes,9 disturbance of their activities can induce bone disorders, such as impairment of bone growth and osteoporosis.10

Hormone- and age-related bone loss or failure to achieve optimal peak bone mass during early adulthood have been recognized as the main risk factors for osteoporosis as well as for fragility fractures.11 Epidemiological studies have identified the associations between several risk factors (e.g., smoking, excessive drinking, physical inactivity) and bone mass.11 However, there is still a long way to go to identify risk factors and elucidate the underlying mechanisms.

Fine particulate matter [PM with an aerodynamic diameter of 2.5μm (PM2.5)] exposure has been shown to be associated with a range of adverse health effects in the respiratory12 and cardiovascular systems,13 with more recent evidence indicating effects in other organ systems, such as metabolic diseases14 and neurodegenerative diseases.15 Furthermore, ambient air pollution has been reported as a potential risk factor for osteoporosis.16 However, recent epidemiological studies have provided inconsistent evidence of the association between ambient air pollution and skeletal health. Although a population study found no significant association of ambient air pollutants, including NO2, O3, and PM2.5, with BMD in women and no association with the incidence of osteoporotic fractures,17 other studies did demonstrate that PM2.5 exposure was associated with lower BMD values and increased fracture risk.1821 However, it remains unclear whether PM2.5 exposure is associated with markers of skeletal health and the potential biological mechanisms by which PM2.5 exposure can contribute to skeletal disorders.

Our previous research, as well as studies conducted by other research groups, have demonstrated exposure to PM2.5 could induce proinflammatory factor expression and cause systemic inflammation,22,23 as well as peripheral inflammation, in vivo.24 Meanwhile, inflammation has been regarded as a potent contributor to excessive osteoclastic bone resorption25 and impaired bone formation in mouse models,26 factors that may eventually cause bone homeostasis imbalance. Therefore, we hypothesized that exposure to PM2.5 may impair bone homeostasis, which is associated with increased inflammation.

In the present study, we attempted to further elucidate the relationship between long-term PM2.5 exposure and bone homeostasis, as well as the potential underlying biological mechanisms by which PM2.5 could affect bone health, by conducting both an observational epidemiological study and experimental in vivo and in vitro examination.

Methods

Epidemiological Study

Study population.

The UK Biobank data set (application number: 41376)2729 involves a large-scale longitudinal cohort with 500,000 middle-aged participants recruited from 17 centers in England, 2 in Scotland, and 3 in Wales since 2006 (original ethics committee approval number: 21/NW/0157). The UK Biobank data set was used to conduct an analysis of prospective observational data to investigate the relationship between long-term PM2.5 exposure and BMD/fracture in humans.

Outcome assessment.

The outcomes of this study were BMD, including heel BMD measured by quantitative ultrasound, and femur neck and lumbar spine BMD measured by dual energy X-ray absorptiometry (DXA) during 2014–2020. Here, as in earlier publications,30,31 to minimize the influence of residual confounding, we excluded participants who had diseases that might affect BMD (i.e., secondary fracture, rheumatoid arthritis, and lupus erythematosus; Table S1). In addition, given that the exposure (i.e., annual average PM2.5 concentration) was estimated for the year 2010,32 we included only the information of BMD estimated after 2010 (n=44,119 for heel BMD; n=36,896 for femur neck and lumbar spine BMD). After excluding participants lacking covariate information (covariates are listed in Table S2), 37,440 participants were included in the analyses of the association for heel BMD and 29,766 participants for femur neck and lumbar spine BMD.

PM2.5 exposure assessment.

In the UK Biobank, the average PM2.5 concentration in 2010 was estimated using land use regression (LUR), a model that was developed as a part of the European Study of Cohorts for Air Pollution Effects (http://www.escapeproject.eu/).33 In the LUR model, the spatial variations of annual average PM2.5 concentration for each address were obtained from Geographic Information System (GIS)–derived predictors, including traffic, land use, and topography.33 These annual average PM2.5 concentrations (available for the year 2010) were linked to each participant in the UK through participants’ residential addresses given at the baseline visit (2006–2010).

Statistical methods.

Multivariate linear regression was used to estimate the association between long-term exposure to PM2.5 (in micrograms per meter cubed) and BMD (in grams per centimeter squared). Results are presented as the change in BMD in grams per centimeter squared for a 1-μg/m3 increase in annual PM2.5 concentrations. In the linear regression model, we included potential BMD-related risk factors as covariates, as in our previous work.30,31 Specifically, two models were used: a) the basic model that adjusting for sex (categorical: male and female), age (continuous: in years), ancestry (categorical: European, Mixed, Asian, Black, Chinese, and Other ethnic groups), and body mass index (BMI; continuous: in kilograms per meter squared) (model 1); and b) the fully adjusted model, that is, the basic model with additional covariates of education level (categorical: yes and no), smoking (categorical: never, previous, and current), alcohol use (categorical: never, previous, and current), physical activity (categorical: yes and no), circulating calcium (continuous: in millimoles per liter), and calcium supplementary status (categorical: yes and no) (model 2) (Table S2). Furthermore, we conducted an analyses stratified by sex to assess the effect in males and females separately. R (version 4.1.0; R Development Core Team) was used for the statistical analyses, and values of p<0.05 were regarded as statistically significant.

Experimental Study

Reagents.

All the antibodies used in this study are listed in Table S3. The Alcian blue [Chemical Abstract Service (CAS) #75881-23-1], hematoxylin (CAS #517-28-2), orange G (CAS #1936-15-8), and tartrate-resistant acid phosphatase (TRAP) staining kit (Sigma; #387) were all obtained from Sigma-Aldrich. Cell culture-related reagents purchased from Gibco included alpha-minimum essential medium (α-MEM medium; #12561-056), fetal bovine serum (FBS; #10099-141), nonessential amino acid (NEAA; #11140-050), and GlutaMAX (#35050-061). Both macrophage colony-stimulating factor (M-CSF; #315-02) and receptor activator of nuclear factor-kappa ligand (RANKL; #315-11C) were purchased from Pepro Tech. Mouse tumor necrosis factor-alpha (TNF-α) neutralizing antibody (D2H4; #11969) was purchased from Cell Signaling Technology. The enzyme-linked immunosorbent assay (ELISA) kit for bone alkaline phosphatase (BALP; #KE1424), osteocalcin (OCN; #KE1428), and TRAP 5b (TRACP-5b; #KE1580) were obtained from ImmunoWay Biotechnology, whereas the ELISA kit for TNF-α (#MTA00B) was purchased from R&D.

Animals and animal care.

In this study, twelve 5-wk-old male C57BL/6 mice were obtained from Shanghai Laboratory Animal Co., Ltd (SLAC). All the mice were housed at 24±2°C with 60±10% humidity and maintained on a 12-h light/12-h dark cycle. In addition, water and food were freely available. The animals were treated humanely during the experiment to minimize suffering, and the animal experimental protocols were approved by the Zhejiang Chinese Medical University Animal Care and Use Committee.

PM2.5 inhalational exposure protocol.

The mice were randomly divided into two groups (n=6 per group) and then consecutively exposed to filtered air (FA) or PM2.5 using the Zhejiang Whole-body Exposure System 1 (ZJ-WES1) for 6 months from May to November in 2018 (6 mice per cage). The ZJ-WES1 is located at the campus of Zhejiang Chinese Medical University, as described in detail previously.34 Briefly, the exposure system consists of two temperature-controlled chambers with the same volume. Specifically, the PM chamber was filled with ambient air except for the particles >2.5μm, which were cleared by a cyclone. The FA chamber is able to eliminate PM2.5 from the air stream with a high-efficiency particulate air filter (Shandong JuKang Technology Co., Ltd. H14; #JKKJ-200) placed in the inlet valve.

PM2.5 concentration measurement and component analysis.

As in our previous studies,35,36 the PM2.5 concentration in the chambers was reflected in real time by an aerosol monitor model pDR-1500 (Thermo Scientific). To confirm the accurate concentration of PM2.5 and analyze the main components, the collection of PM2.5 samples from the chambers was implemented with Teflon filter membranes (37mm; GE HealthCare) or a quartz filter (47mm; MTL). The weight of the membranes was measured using an Excellence Plus XP microbalance (Mettler Toledo) in a temperature- and humidity-controlled room.34 Membrane weight before and after sampling was used to calculate the exposure concentration.

To analyze trace metal elements, the sampling filter membrane was cut with ceramic scissors and immersed in 10mL of nitric acid solution (5%) followed by an ultrasonic water bath at 70°C for 3 h. Then, the suspension was centrifuged at 4,500 rpm for 5 min, and the supernatant was filtered with a 0.45-μm strainer and collected for measurement. The concentrations of corresponding trace metal elements [mercury (Hg), antimony (Sb), aluminum (Al), arsenic (As), beryllium (Be), cadmium (Cd), chromium (Cr), lead (Pb), manganese (Mn), nickel (Ni), selenium (Se), and thallium (Tl)] were determined with inductively coupled plasma mass spectrometry (iCAP Qc ICP-MS; Thermo Scientific) and analyzed with Qtegra Instrument Control software. The stock solution of metal standards [1,000μg/mLHg, 1,000μg/mL Se, and 100μg/mL multielement (Sb, Al, As, Be, Cd, Cr, Pb, Mn, Ni, and Tl)] was diluted in a 95% nitric acid solution and used to draw standard curves. The curves with linear correlation coefficients of 0.999 were deemed available for sample concentration calculation. A solution containing scandium (Sc), yttrium (Y), germanium (Ge), and rhodium (Rh) was applied to provide internal standards. In addition, quality control measures included device calibration before analysis, reagent and procedural blanks, and synchronous analysis of 20% samples. The same settings of availability of standard curve and quality control were applied to the analysis of the other two components.

For water-soluble inorganic ions analysis, the sampling filter membrane was cut with ceramic scissors and immersed in ultrapure water (25mL) at 20°C for 40 min. The extract was centrifuged at 11,000 rpm for 1 min, and the supernatant was filtered with a 0.22-μm filter. Various water-soluble inorganic ions [fluoride ion (F), chloride ion (Cl), nitrate (NO3), sulfate (SO42), and ammonium ion (NH4+)] were determined by ion chromatography (ICS-600; Thermo Scientific) equipped with a dual-piston pulse infusion pump system. The separations were performed with a chromatographic column (4×250mm) at 25°C, and the eluent solution contained 20mM methanesulfonic acid for cations and 4.5mM sodium carbonate and 0.8mM sodium bicarbonate for anions and was used at a flow rate of 1mL/min. The injection volume was 200μL. The standard stock solution of F, Cl, NO3, SO42, and NH4+ was diluted to 0.1, 0.2, 0.5, 1, 2, 5, 10, and 20μg/mL with ultrapure water and used for standard curve drawing. Finally, the data were analyzed using Chromeleon 7 software (version 7.1.2.1478).

As for the detection of polycyclic aromatic hydrocarbons (PAHs), the sampling filter membrane was cut up and immersed in 5mL decafluorobiphenyl solution (0.2μg/mL, dissolved in acetonitrile). The samples were sonicated for 30 min and centrifuged at 8,000 rpm for 5 min. Next, the supernatant was collected and filtered with a 0.45-μm strainer for examination of the 16 PAHs, which included naphthalene (NAP), acenaphthylene (ANY), fluorene (FLU), acenaphthene (ANA), phenanthrene (PHE), anthracene (ANT), fluoranthene (FLT), pyrene (PYR), chrysene (CHR), benzo(a)anthracene (BaA), benzo(b)fluoranthene (BbF), benzo(k)fluoranthene (BkF), benzo(a)pyrene (BaP), dibenzo(a,h)anthracene (DBA), benzo(g,h,i)perylene (BPE), and indeno(1,2,3-cd)pyrene (IPY). The separations were conducted with a column (C18; 250mm×4.6mm, 3.5-μm particle size) at 35°C, with a flow rate of 1mL/min acetonitrile as the mobile phase. The injection volume was 20μL. The 20-μg/mL standard stock solution containing the 16 targeted PAHs was diluted to 0.01, 0.02, 0.05, 0.1, 0.2, and 0.3 ng/mL with acetonitrile for standard curves. Then the content of the total 16 PAHs was determined by liquid chromatography (Agilent1260) and analyzed using OpenLAB CDS software (version C.01.06).

Sample collection.

After long-term PM2.5 exposure (6 months), the mice were euthanized with isoflurane and the blood sample was collected by eyeball enucleation. Each blood sample was placed at room temperature for 30 min after centrifugation with 2,000 relative centrifugal force (RCF) for 10 min at 4°C to obtain serum samples, which were stored at 80°C for ELISA analysis. After blood sample collection, the mice were sacrificed and livers were collected and washed with phosphate buffered saline (PBS) and then stored at 80°C for further analysis. In addition, intact femurs were uniformly dissected from the right hind limb followed by fixation with 4% paraformaldehyde (PFA) for 3 d at room temperature for further analysis [micro-computed tomography (micro-CT) and all the histological examinations].

Micro-CT analyses.

All the fixed femur samples were evaluated using micro-CT scanning, as described in the previous study.37 In brief, each sample was scanned with a micro-CT scanner (SkyScan; #1176) with the parameters of 45-kV voltage, 500-μA current, 780-ms integration time, 4,000×2,672 resolution, and 9-μm slice thickness. The scanned images were performed successively by reconstruction software (NRecon; version 1.6, SkyScan), reposition software (Dataviewer; version 1.5; SkyScan), and analysis software (CTAn; version 1.15; SkyScan), in sequence, to obtain the analyzed femoral parameters.

In addition, three-dimensional (3D) reconstruction images were provided by visualization software (CTVolx; version 3.0; SkyScan). Referring to the previous study,38 in brief, the evaluated region of interest (ROI) for analysis was drawn beginning from the distal femoral metaphyseal growth plate and extending proximally for 100 CT slices. For quantitative analyses, bone parameters, such as BMD, trabecular bone volume fraction (BV/TV), trabecular number (Tb. N), trabecular separation (Tb. Sp), and trabecular thickness (Tb. Th) were collected from the analysis software. After micro-CT analyses, the measurements of femur length and width were precisely conducted by reposition software (Dataviewer; version 1.2; SkyScan) rather than manual measurements. According to the anteroposterior radiograph of full-length femur projection, the length was measured from the greater trochanter to the femoral distal lateral condyle. The femoral width was measured at the traverse position, extending proximally for 15% of the total femur length from the distal femoral growth plate. All the metrical data were accurate to within 0.01mm.

Histomorphology analyses.

For histological analyses, all the femurs were decalcified using 14% ethylenediaminetetraacetic acid solution at pH 7.2 for 2 wk following micro-CT scanning. Subsequently, the samples were dehydrated with gradient alcohol. After dehydration, the femur samples were embedded in paraffin with a sagittal orientation. Each paraffin block was sectioned with microtome (Thermo Fisher Scientific; #HM 355S) at a 3-μm thickness for all staining and immunohistochemistry. Then, Alcian blue hematoxylin/orange G (ABH/OG) staining was carried out for the histological evaluation on the distal femoral metaphysis. Images were captured using a microscope (Zeiss AxioCam HRc Scope.A1) and ImageJ software (version 1.5)39 was used for quantification analyses.

TRAP staining.

In addition, TRAP staining was performed to determine the osteoclast activity. For the tissue detection, the staining was conducted on the 3-μm thickness paraffin sections from femur tissue. Briefly, sections were rehydrated and incubated with basic stock solution containing napthol AS-BI phosphate (0.2mg/mL) for an hour at 37°C. Then, sample sections were incubated for 10 min at 37°C in a mixture solution that included sodium nitrite (0.8mg/mL) and pararosaniline dye (1.0mg/mL). Next, all the sections were counterstained with Fast Green. Ultimately, multinuclear TRAP-positive cells around the distal femoral growth plate, the key site for bone remodeling in mice,25,40 were calculated using ImageJ software (version 1.5).39

Immunohistochemistry.

A series of markers for osteoblasts, osteoclasts, and inflammation were examined, as previously reported.41 In brief, deparaffinized sections of femur tissue were treated by sodium citrate (pH 6.0) at 95°C for antigen retrieval followed by eliminating the activity of endogenous peroxidase using 0.3% hydrogen peroxide, and then blocked for 20 min with normal goat serum at room temperature. After that, the samples were incubated with the primary antibody at 4°C overnight and with the corresponding secondary antibody at room temperature for 20 min. The positive signal was then visible with diaminobenzidine solution (Invitrogen). Ultimately, hematoxylin was used for counterstaining.

In the present study, anti-alkaline phosphatase (ALP) antibody (Arigo; #ARG57422; 1:300), anti-OCN antibody (Abcam; #ab13420; 1:300), anti-cathepsin K (Ctsk) antibody (Abcam; #ab188604; 1:200), anti-RANKL antibody (Abcam; #ab45039; 1:300), anti-interleukin-1β (IL-1β) antibody (Abcam; #ab9722; 1:300), and anti-TNF-α antibody (Arigo; #ARG56080; 1:300) were used to detect the expression level of the corresponding proteins. The positively stained area was measured using ImageJ software (version 5.0).39

Cell culture and conditional medium collection from PM2.5-stimulated macrophages.

Primary bone marrow macrophages (BMMs) or murine monocytic macrophage RAW264.7 cells (Cell Bank of the Chinese Academy of Sciences) were cultured for conditional medium collection. For isolation of primary BMMs, 10-wk-old male C57BL/6 mice exposed to FA were sacrificed and disinfected with 75% alcohol. Next, the femurs and tibias were separated and the bone marrow was flushed with α-MEM medium (Gibco; #12561-056) containing 2% FBS (Gibco; #10099-141) and 1% penicillin/streptomycin (P/S; NorthEnd; #15140). The cell suspension was then filtered with a 70-μm cell sieve (BIOLOGIX; #15-1070) and centrifuged at 1,000 rpm for 5 min. Cells were suspended in complete cell culture medium [α-MEM supplemented with 10% FBS, 1% P/S, 1% NEAA (Gibco; #11140-050) and 1% GlutaMAX (Gibco; #35050-061) with 10 ng/mL of M-CSF (Pepro Tech; #315-02). Two days later, the medium was replaced with the same formula except for the final concentration of M-CSF up to 30 ng/mL for another 2 d (Figure S1A and Table S4). RAW264.7 cells suspended in α-MEM medium containing 10% FBS and 1% P/S were allowed to adhere and form a monolayer for 2 d (Figure S1A and Table S5).

As previously described,42 the PM2.5 suspension (Standard Reference Material 1648a, ID160705; National Institute of Standards and Technology) was made fresh at a concentration of 10mg/mL in α-MEM medium and sonicated for 1 h at room temperature. Both BMMs and RAW264.7 cells were stimulated with PM2.5 suspensions at final doses of 25 or 50μg/mL, based on preliminary experiments. After a 24-h incubation, the supernatant from the two kinds of cells was respectively collected as conditional medium and filtered through 0.22-μm sieves. The supernatant was temporarily stored at 80°C for further experiments.

Osteoclastogenic induction and conditional medium incubation.

To determine the indirect effects of PM2.5 on osteoclast differentiation, as shown in Figure S1B, primary BMMs extracted from the bones were initially suspended in complete culture medium in the presence of M-CSF (10 ng/mL) and seeded in 96-well plates (2×104 cells per well), 24-well plates (8×105 cells per well), 12-well plates (4×105 cells per well), and 6-well plates (1×106 cells per well) for adherence. Two days later, the medium was exchanged with complete culture medium containing 30 ng/mL M-CSF and 30 ng/mL RANKL (Pepro Tech; #315-11C), and the cells were cultured for another 2 d.

Then, the culture media were respectively replaced and the cells were stimulated with 20% conditioned medium from PM-treated BMMs or 5% from RAW264.7 cells for 2 d (Figure S1B and Table S6). When the role of TNF-α in the PM2.5-mediated osteoclastogenesis was explored, TNF-α neutralizing antibody (D2H4) at a final dose of 1μg/mL (Cell Signaling Technology; #11969) was simultaneously employed with conditioned medium from BMMs (Figure S1C and Table S7). At the end of the 2-d incubation, cells were collected for further examination, including cell TRAP staining, bone resorption assay, real-time polymerase chain reaction (RT-PCR), and western blotting.

Cell TRAP staining and bone resorption assay.

Following the full process of BMMs culture, osteoclastogenic induction, in presence of or absence of conditional medium/TNF-α neutralizing antibody D2H4 incubation, multinucleated cells in 96-well plates were identified as osteoclasts using TRAP staining. First, the medium was aspirated and the cells were washed with PBS and then fixed in 4% PFA for 20 min at room temperature. Then, a TRAP staining kit (Sigma; #387A) was used to assess the TRAP activity of the cells. TRAP-positive cells that had three or more nuclei were deemed to be osteoclasts. The number of osteoclasts were calculated in each well under a light microscope (OLYMPUS; #IX71).

We carried out a bone resorption assay to analyze the effect of PM2.5 on the demineralization function mediated by osteoclasts in vitro. Briefly, primary BMMs were isolated and then seeded into 24-well plates (8×104 cells per well) coated with hydroxyapatite (CLS3989; #Corning) and cultured with complete culture medium containing M-CSF (10 ng/mL). The cell culture was performed as shown in Table S7. Next, the conditioned medium was removed and the wells were treated with ammonium hydroxide (1 mol/L) for 5 min to remove the attached cells. Finally, the areas resorbed by osteoclasts were observed by light microscopy (OLYMPUS; #IX71) and calculated using ImageJ software (version 1.5).39

RNA extraction and RT-PCR.

In this study, the total RNA of the cells seeded in 12-well plates (4×105 cells per well) was extracted with TRIzol Reagent (TaKaRa). The RNA concentrations were measured using a NanoDrop2000 ultra microspectrophotometer (Thermo Fisher), and the absorbance ratio at 260/280 nm was between 1.8 and 2.0 to ensure RNA quality purity. Then the RNA was reverse transcribed into complementary DNA with PrimeScript RT Master Mix (TaKaRa). As in the previous study,35 the program was completed by the QuantstudioQ7 (Applied Biosystems) using SYBR Green mix (Applied Biosystems) with the following cycling program: 50°C for 2 min, 95°C for 2 min (activation), 40 cycles of 95°C for 15 s (denaturation), and 60°C for 1 min (extension). Here, we adopted the CTΔΔ method to analyze the relative mRNA expression. All the primer sequences of target genes are listed in Table S8.

Western blotting.

Proteins from cells in 6-well plates and mouse liver tissue were extracted with radioimmunoprecipitation assay lysis buffer (Boster Biological Technology Co., Ltd.) on ice. Next, total protein was quantified using a bicinchoninic acid protein assay kit (Thermo Fisher Scientific; #23227), and the equivalent amount of harvested protein, 15μg, was separated on 10% sodium dodecyl sulfate–polyacrylamide gel electrophoresis (SDS-PAGE) gel and then transferred to polyvinylidene fluoride (PVDF) membranes. The membranes were blocked with 5% bovine serum albumin in Tris-buffered saline-Tween (TBST) and incubated at 4°C overnight with primary antibodies and with the corresponding secondary antibodies [Proteintech; #SA00001-1 (1:5,000) and #SA00001-2 (1:5,000)] at room temperature for 2 h.

The primary antibodies used for the western blot included ALP (Arigo; #ARG57422; 1:2,000), c-Fos (Huabio; #ET1701-95; 1:500), and Traf6 (Huabio; #R1311-2; 1:1,000). We used the ChemiDoc Imaging System (Bio-Rad) to visualize the bands through enhanced chemiluminescence (4A Biotech; #4AW011-200) and quantitated the autoradiograph by densitometric analysis with Image Lab software matching the system. β-actin (CST; #4970; 1:1,000) or glyceraldehyde 3-phosphate dehydrogenase (GAPDH; Abcam; #ab8245; 1:1,000) was used as the control reference.

ELISA.

Serum samples were collected from mouse whole blood and centrifuged at 2,000 RCF for 10 min at 4°C. In the present study, the concentrations of BALP, OCN, and TRACP-5b in serum were determined with the corresponding ELISA kits (ImmunoWay Biotechnology; BALP, #KE1424; OCN, #KE1428; TRACP-5b, #KE1580) according to the manufacturer’s instructions. In addition, PM-treated conditioned medium was filtered through a 0.22-μm strainer (Millex; #ROAB40604) and the TNF-α levels in the medium were examined with the Mouse TNF-α Quantikine ELISA kit (R&D; #MTA00B) according to the manufacturer’s instructions.

Statistical analysis.

All experimental data are presented as means±SEMs and assessed through unpaired t-tests or one-way analyses of variance (ANOVAs). In addition, GraphPad Prism software (version 5.0) was used for the statistical analyses, and values of p<0.05 were regarded as statistically significant.

Results

Associations between Long-Term PM2.5 Exposure and BMD in Human

We used UK Biobank data to analyze the association between long-term exposure to PM2.5 (estimated at 2010) and heel BMD (measured during 2014–2020). After excluding participants lacking covariate information and those with diseases that might affect BMD, 37,440 participants (n=18,849 for females and n=18,591 for males) remained (Table S9). The mean±SD age of these participants was 55.80±7.54 y, ranging from 40 to 73 y. The mean estimate±SD of PM2.5 concentrations was 9.918±1.031μg/m3. For the femur neck and lumbar spine BMD analyses, 29,766 participants were included (Table S9) (the mean age was 55.05±7.48 y, range: 40–70 y; the mean concentration was 9.913±1.039μg/m3 for PM2.5; n=14,968 for females, n=14,798 for males). We observed that long-term exposure to PM2.5 was associated with decreased heel BMD as estimated by quantitative ultrasound in the basic linear regression model adjusting for sex, age, ancestry, and BMI (model 1) [β-coefficient±standard error (SE) was 0.003±0.001, p=2.70×108] (Table 1). Here the β-coefficient represents the change in BMD in grams per centimeter squared for a 1-μg/m3 increase in annual PM2.5 concentrations. The associations remained statistically significant in the fully adjusted model, which included additional covariates such as education level, smoking, alcohol use, physical activity, circulating calcium and calcium supplementary status (model 2) (β-coefficient±SE was 0.003±0.001, p=4.53×105; Table 1). We also assessed the relationship between long-term exposure to PM2.5 and BMD assessed by DXA at the lumbar spine (β-coefficient±SE was 0.006±0.001, p=4.61×109 in model 1, and β-coefficient±SE was 0.005±0.001, p=4.18×106 in model 2) and at the femur neck (β-coefficient±SE was 0.003±0.001, p=8.89×106 in model 1, and β-coefficient±SE was 0.002±0.001, p=5.60×104 in model 2; Table 1). In the stratified analyses by sex, the trends were similar in both males and females (Table 2).

Table 1.

The observational association of long-term exposure to PM2.5 with BMD measured after the year 2010 at different anatomical sites (n=37,440 for heel BMD; n=29,766 for lumbar spine 1 to 4 BMD; n=29,766 for femur neck BMD) in the UK Biobank.

β-coefficient a SE p-Valueb
Heel BMD
 Model 1 0.003 0.001 2.70×108
 Model 2 0.003 0.001 4.53×105
Lumbar spine 1 to 4 BMD
 Model 1 0.006 0.001 4.61×109
 Model 2 0.005 0.001 4.18×106
Femur neck BMD (right)
 Model 1 0.003 0.001 8.89×106
 Model 2 0.002 0.001 0.001

Note: Model 1 was adjusted for confounders, including sex, age, body mass index, and ancestry; model 2=model 1+education+smoking+alcohol+physical activity+circulating calcium+calcium supplementary status. BMD, bone mineral density; PM2.5, particulate matter with aerodynamic diameter of 2.5μm; SE, standard error.

a

The β-coefficient represents the change in BMD in grams per centimeter squared for a 1-μg/m3 increase in annual PM2.5 concentrations.

b

All p-values were derived from multiple linear regression.

Table 2.

The observational association of long-term exposure to PM2.5 with BMD measured after the year 2010 in males (n=18,591 for heel BMD; n=14,798 for lumbar spine 1 to 4 BMD; n=14,798 for femur neck BMD) and females (n=18,849 for heel BMD; n=14,968 for lumbar spine 1 to 4 BMD; n=14,968 for femur neck BMD) in the UK Biobank.

β-coefficient a SE p-Valueb
Heel BMD
 Male
  Model 1 0.004 0.001 1.68×105
  Model 2 0.003 0.001 0.001
 Female
  Model 1 0.002 0.001 0.001
  Model 2 0.002 0.001 0.036
Lumbar spine 1 to 4 BMD
 Male
  Model 1 0.007 0.001 1.54×106
  Model 2 0.006 0.002 2.19×104
 Female
  Model 1 0.004 0.001 0.004
  Model 2 0.003 0.001 0.027
Femur neck BMD (right)
 Male
  Model 1 0.004 0.001 6.84×105
  Model 2 0.003 0.001 0.004
 Female
  Model 1 0.002 0.001 0.055
  Model 2 0.002 0.001 0.113

Note: Model 1 was adjusted for confounders, including age, body mass index, and ancestry; model 2=model 1+education+smoking+alcohol+physical activity+circulating calcium+calcium supplementary status. BMD, bone mineral density; PM2.5, particulate matter with aerodynamic diameter of 2.5μm; SE, standard error.

a

The β-coefficient represents the change in BMD in grams per centimeter squared for a 1-μg/m3 increase in annual PM2.5 concentrations.

b

All p-values were derived from multiple linear regression.

PM2.5 Concentration Measurement and Component Analyses of Animal Study

As shown in Figure 1A, the mean PM2.5 concentration of ambient air at the PM chamber was 29.27μg/m3, whereas in the FA chamber it was 1.30μg/m3. The lowest PM2.5 concentration was in July and August, whereas the highest concentration was in May and October (Figure 1B). As for the component analysis, the major water-soluble inorganic ions in the composition of PM2.5 included SO42, NO3, NH4+, and Cl (Figure 1C), whereas F was not detected. Regarding the 12 kinds of trace metals in the PM2.5, the major trace metals were Al, Pb, and Mn, which were >10 ng/m3, whereas Ni, As, Se, Sb, and Cr were 15 ng/m3 and only tiny amounts of Cd, Tl, Hg, and Be were detected (Figure 1E). In the 16 PAHs tested in the PM2.5 (Figure 1G), the most abundant PAHs was BbF, followed by IPY, PYR, DBA, and BPE. In addition, the traces of the PM2.5 compositions during the exposure duration are shown in Figure 1D,F,H. Generally, most of the main components remained at lower levels in July and August and increased in the fall, reaching a peak in November.

Figure 1.

Figure 1A is a bar graph, plotting particulate matter begin subscript 2.5 end subscript concentration (micrograms per meter cubed), ranging from 0 to 40 in increments of 10 (y-axis) across filtered air and particulate matter (x-axis). Figures 1B, 1D, 1F, 1H are line graphs, plotting particulate matter begin subscript 2.5 end subscript concentration (micrograms per meter cubed), ranging from 0 to 50 in increments of 10; concentration of water-soluble inorganic ions (micrograms per meter cubed), ranging from 0 to 16 in increments of 4; concentration of trace metals (nanograms per meter cubed), ranging from 0 to 50 in increments of 10; concentration of polycyclic aromatic hydrocarbons (nanograms per meter cubed), ranging from 0.0 to 2.5 in increments of 0.5 (y-axis) across Months, including May, June, July, August, September, October, and November (x-axis) for filtered air and particulate matter; sulfate, nitrate, ammonium ion, chloride ion; aluminum, lead, manganese, nickel, arsenic, selenium, antimony, chromium, cadmium, thallium, mercury, beryllium; benzo(b)fluoranthene, indeno pyrene, pyrene, dibenzo anthracene, benzo perylene, acenaphthylene, phenanthrene, fluoranthene, chrysene, benzo(a)anthracene, benzo(a)pyrene, benzo(k)fluoranthene, naphthalene, fluorene, acenaphthene, anthracene. Figure 1C is a bar graph, plotting concentration of water-soluble inorganic ions (micrograms per meter cubed), ranging from 0 to 5 in unit increments (y-axis) across sulfate, nitrate, ammonium ion, chloride ion (x-axis). Figures 1E and 1G are bar graphs, plotting concentration of trace metals (nanograms per meter cubed), ranging from 0.0 to 0.2 in increments of 0.2, 2 to 4 in increments of 2, 20 to 30 in increments of 10 and concentrations of polycyclic aromatic hydrocarbons (nanograms per meter cubed), ranging from 0.0 to 0.2 in increments of 0.1 and 0.4 to 1.0 in increments of 0.2 (y-axis) across aluminum, lead, manganese, nickel, arsenic, selenium, antimony, chromium, cadmium, titanium, mercury, beryllium; and benzo(b)fluoranthene, indeno pyrene, pyrene, dibenzo anthracene, benzo perylene, acenaphthylene, phenanthrene, fluoranthene, chrysene, benzo(a)anthracene, benzo(a)pyrene, benzo(k)fluoranthene, naphthalene, fluorene, acenaphthene, anthracene, respectively (x-axis).

PM2.5 concentration and component analysis during the mice exposure period. (A) Average concentrations and (B) monthly concentrations in the filtered air (FA), PM2.5 (PM) groups (n10). (C) Average concentrations and (D) monthly concentrations of water-soluble inorganic ions in the PM2.5 group (n6). (E) Average concentrations and (F) monthly concentrations of trace metals in the PM2.5 group (n6). (G) Average concentrations and (H) monthly concentrations of PAHs in the PM2.5 group (n6). Numeric data can be found in Table S10. Data are presented as means±SEMs. Note: Al, aluminum; ANA, acenaphthene; ANT, anthracene; ANY, acenaphthylene; As, arsenic; Aug, August; BaA, benzo(a)anthracene; BaP, benzo(a)pyrene; BbF, benzo(b)fluoranthene; Be, beryllium; BkF, benzo(k)fluoranthene; BPE, benzo perylene; Cd, cadmium; CHR, chrysene; Cl, chloride ion; Cr, chromium; DBA, dibenzo anthracene; FLT, fluoranthene; FLU, fluorene; Hg, mercury; IPY, indeno pyrene; Jul, July; Jun, June; Mn, manganese; NAP, naphthalene; NH4+, ammonium ion; Ni, nickel; NO3, nitrate; Nov, November; Oct, October; PAHs, polycyclic aromatic hydrocarbons; Pb, lead; PHE, phenanthrene; PM, particulate matter; PM2.5, particulate matter with aerodynamic diameter of 2.5μm; PYR, pyrene; Sb, antimony; Se, selenium; SEM, standard error of the mean; Sep, September; SO42, sulfate; Tl, thallium.

Long-Term PM2.5 Exposure, Femur Bone Growth, and Osteogenesis in C57BL/6 Mice

After 6 months of FA or PM2.5 exposure, no apparent difference in appearance was observed between two groups. Initially, we examined the length and width of the mouse femur, the most representative bone in the skeletal system, to explore the relationship between air pollution and skeletal health. To avoid the interference from soft tissue attached to the surface of the bone, anteroposterior radiograph of the full-length femur was performed to measure precisely (Figure 2A). As shown in Figure 2B, femoral length was shorter in the PM2.5-exposed mice compared with the mice exposed to FA. In addition, femoral width was a bit narrower with long-term PM2.5 exposure, although there was no statistical difference (Figure 2C). These data suggest that PM2.5 air pollution exposure affected the femur growth in length.

Figure 2.

Figure 2A is a coronal image displaying two rows, namely, filtered air and particulate matter under 2D-coronal plane. Figures 2B, 2C, 2E, 2F, 2H, 2I, 2K, 2L, 2N are bar graphs, plotting Femur length (millimeters), ranging from 12 to 16 in unit increments, femur width (millimeters), ranging from 1.0 to 3.0 in increments of 0.5; cortical bone mineral density (grams per centimeter cubed), ranging from 0.8 to 1.2 in increments of 0.2; trabecular bone mineral density (grams per centimeter cubed), ranging from 0.00 to 0.12 in increments of 0.03; osteocalcin-positive ratio (percentage), ranging from 0 to 20 in increments of 5; osteocalcin levels in serum (nanograms per milliliter), ranging from 0 to 200 in increments of 0 to 200 in increment of 50; alkaline phosphatase area ratio (percentage), ranging from 0 to 15 in increments of 5; bone alkaline phosphatase levels in serum (nanograms per milliliter), ranging from 0 to 32 in increments of 8; relative protein expression of alkaline phosphatase, ranging from 0.0 to 2.0 in increments of 0.5 (y-axis) across filtered air and particulate matter (x-axis). Figure 2D is a stained tissue displaying two columns, namely, filtered air and particulate matter under 3D-reconstruction. Figure 2G is a stained tissue displaying two columns, namely, filtered air and particulate matter and one row, namely, osteocalcin. Figure 2J is a stained tissue displaying two columns, namely, filtered air and particulate matter and one row, namely, alkaline phosphatase. Figure 2M is a western blot displaying two columns, namely, filtered air and particulate matter and two rows, namely, alkaline phosphatase and lowercase beta-actin.

The effect of PM2.5 on bone morphology and osteogenesis in vivo. Mice were exposed to FA or PM2.5 for 6 months. (A) Representative coronal images of intact femur. Scale bar: 1mm. Quantitative analyses of femur (B) length and (C) width. (D) Representative images of 3D reconstruction of ROI. Scale bar: 500μm. Micro-CT quantitative analyses of (E) cortical bone mineral density and (F) trabecular bone mineral density. (G) Representative images of OCN immumohistochemical staining. Red arrows indicate positive expression. Scale bar: 50μm. (H) Quantification of OCN positive area. (I) Quantitative analyses of OCN levels in serum. (J) Representative images of ALP immumohistochemical staining. Red arrows indicate positive expression. Scale bar: 50μm. (K) Quantification of ALP positive area. (L) Quantitative analysis of BALP levels in serum. (M) Expression of ALP in liver assessed by western blotting. (N) Quantitative analysis of ALP expressions in liver. Numeric data can be found in Table S11. Data are presented as means±SEMs (unpaired Student’s t-test, n=46), *p<0.05, ***p<0.001. Note: 2D, two dimensions; 3D, three dimensions; ALP, alkaline phosphatase; BALP, bone alkaline phosphatase; BMD, bone mineral density; FA, filtered air; micro-CT, micro-computed tomography; OCN, osteocalcin; PM, particulate matter; PM2.5, particulate matter with aerodynamic diameter of 2.5μm; ROI, region of interest; SEM, standard error of the mean.

Based on the finding that PM2.5 air pollution had a slight effect on the femur length, we performed micro-CT evaluation on the distal femoral metaphysis, the most representative site reflecting BMD and bone mass in mice,43 to determine whether the skeletal structure was altered by PM2.5 exposure. As illustrated in Figure 2D–F, there was no visual (Figure 2D) or statistical (Figure 2E,F) difference in either cortical or trabecular BMD between the FA and PM groups. Parameters reflecting bony architecture, including BV/TV, Tb.N, Tb.Sp, and Tb.Th, were evaluated, and no significant difference was observed with PM2.5 exposure (Figure S2).

To determine whether air pollution modulates osteogenesis, levels of the most representative osteogenic markers, OCN and BALP,44 were examined. Immunohistochemical analysis showed the OCN expression was significantly lower in response to PM2.5 exposure in the distal femur than that in FA controls (Figure 2G,H), which was in accord with the circulating levels of OCN in the serum (Figure 2I). In addition, ALP expression around the distal femur was examined with immunohistochemistry. However, no significant discrepancy of ALP expression around the growth plate was observed between the two groups (Figure 2J,K). Surprisingly, levels of BALP were significantly higher in PM2.5-exposed mice than in mice exposed to FA (Figure 2L). Because the liver is an attributable source of ALP synthesis, we also examined ALP expression in the liver. Interestingly, protein levels of ALP in the liver of PM2.5-exposed mice were significantly higher than those of the FA group (Figure 2M,N).

Long-Term PM2.5 Exposure, Osteoclastogenesis, and Inflammation around the Femoral Distal Metaphysis in Mice

Because the femoral length was highly relevant to bone metabolism, we detected the osteoclastogenesis around the femoral growth plate. Immunohistochemical analysis revealed that mice exposed to PM2.5 expressed a higher RANKL level in the distal femur; given that RANKL is obligatory for bone resorption,45 this suggests its expression may promote osteoclastogenesis (Figure 3A,B). Then, we performed TRAP staining to confirm the osteoclast formation around growth plate. As expected, more TRAP-positive areas were observed around the growth plate of PM2.5-exposed mice compared with that of the control group (Figure 3C,D), suggesting the number of osteoclast precursors or osteoclasts increased after PM2.5 exposure. In addition, the circulating levels of TRACP-5b in serum were higher in PM2.5-exposed mice than those in FA-exposed mice as well (Figure 3E).

Figure 3.

Figure 3A is a stained tissue displaying one column, namely, Receptor activator of nuclear factor-kappa ligand and two rows, namely, filtered air and particulate matter. Figures 3B, 3D, 3E, 3H, 3J, 3L are bar graphs, plotting Receptor activator of nuclear factor-kappa ligand area ratio (percentage), ranging from 0 to 75 in increments of 15, Tartrate-resistant acid phosphatase osteoclasts (per millimeter squared), ranging from 0 to 8 in increments of 2; Tartrate-resistant acid phosphatase 5b levels in serum (nanograms per milliliter), ranging from 0 to 50 in increments of 10; Cathepsin K area ratio (percentage), ranging from 0 to 20 in increments of 5; Tumor necrosis factor-lowercase alpha area ratio (percentage), ranging from 0 to 15 in increments of 5 (y-axis) across filtered air and particulate matter (x-axis). Figure 3C is a stained tissue depicting the Tartrate-resistant acid phosphatase staining around the femoral distal growth plate. Figure 3D is a stained tissue depicting the Quantitative analysis of TRAP-positive cells around the femoral distal growth plate. Figure 3E is a stained tissue depicting the Quantitative analysis of TRACP-5b levels in serum. Figure 3F is a stained tissue depicting the images of Alcian blue Hematoxylin or Orange G staining around the femoral distal growth plate. Figure 3G is a stained tissue depicting the of Cathepsin K immunohistochemical staining. Figure 3H is a stained tissue depicting the Quantification of the Ctsk-positive area. Figure 3I is a stained tissue displaying one column, Tumor necrosis factor- lowercase alpha and two rows, namely, filtered air and particulate matter. Figure 3K is a stained tissue displaying interleukin-1 lowercase beta and two rows, namely, filtered air and particulate matter.

The effect of PM2.5 on osteoclastogenesis and inflammation in vivo. Mice were exposed to FA or PM2.5 for 6 months. (A) Representative images of RANKL immunohistochemical staining around the growth plate. Red arrows indicate positive expression. Scale bar: 100μm. (B) Quantification of RANKL-positive area around the growth plate. (C) Representative images of TRAP staining around the femoral distal growth plate. Images in the right panel are higher magnifications of images in the respective left panel. Scale bar: 200μm. (D) Quantitative analysis of TRAP-positive cells around the femoral distal growth plate. (E) Quantitative analysis of TRACP-5b levels in serum. (F) Representative images of ABH/OG staining around the femoral distal growth plate. Red arrows indicate growth plate destruction. Scale bar: 50μm. (G) Representative images of Ctsk immunohistochemical staining. Red arrows indicate positive expression. Scale bar: 50μm. (H) Quantification of the Ctsk-positive area. (I) Representative images of TNF-α immumohistochemical staining. Red arrows indicate positive expression. Scale bar: 50μm. (J) Quantification of TNF-α–positive area. (K) Representative images of IL-1β immumohistochemical staining. Red arrows indicate positive expression. Scale bar: 50μm. (L) Quantification of IL-1β–positive area. Numeric data can be found in Table S12. Data are presented as means±SEMs (unpaired Student’s t-test, n=46), *p<0.05, **p<0.01, ***p<0.001. Note: ABH/OG, Alcian blue hematoxylin/orange G; Ctsk, cathepsin K; FA, filtered air; IL-1β, interleukin-1beta; PM, particulate matter; PM2.5, particulate matter with aerodynamic diameter of 2.5μm; RANKL, receptor activator of nuclear factor-kappa ligand; SEM, standard error of the mean; TNF-α, tumor necrosis factor-alpha; TRACP-5b, tartrate-resistant acid phosphatase 5b; TRAP, tartrate-resistant acid phosphatase.

Histological analysis of the distal femur was performed by ABH/OG staining. Intriguingly, much more lacunae occupied by chondroclast-like cells around the growth plate were detected, suggesting that the structure of calcified cartilaginous template may have been impaired in PM2.5-exposed mice (Figure 3F). In concurrence with ABH/OG staining, immunohistochemical analysis of Ctsk, a pivotal indicator of mature osteoclasts, showed a higher expression in PM2.5-exposed mice (Figure 3G,H).

Considering that inflammation is one of the key factors mediating osteoclast formation,46 immunohistochemistry was performed to analyze the changes of proinflammatory cytokine (TNF-α and IL-1β) expression around the femoral growth plate. Our data showed that more positive staining areas of TNF-α were observed around the growth plate in mice exposed to PM2.5 (Figure 3I,J), whereas no difference was shown with IL-1β expression between the FA and PM groups (Figure 3K,L).

PM2.5 Stimulation and Osteoclast Differentiation in Vitro

In view of our findings suggesting more pronounced osteoclastogenesis in PM2.5-exposed mice, we next sought to gain further insight into the osteoclast differentiation from macrophages in vitro. In the present study, we treated RAW264.7 cell and BMMs with PM2.5 particles. We then employed murine primary BMMs to induce preosteoclasts/osteoclasts. We treated RAW264.7 macrophages with PM2.5 particles and the culture medium supernatant was collected for TNF-α detection by ELISA. As shown in Figure 4A, we found that there was a dose-dependent difference in TNF-α expression in cells exposed to PM2.5 compared with control cells. Then, the conditional media supernatant containing TNF-α was applied to the osteoclasts differentiated from BMMs and osteoclastogenesis in vitro. Cells exposed to PM2.5 exhibited markedly higher gene expression of osteoclast differentiation genes, including acid phosphatase 5 (Acp5), Ctsk, nuclear factor of activated T cells cytoplasmic 1 (Nfatc1), c-Fos, dendritic cell-specific transmembrane protein (Dcstamp), Atp6v0d2, and TNF receptor-associated factor 6 (Traf6) in a dose-dependent manner (Figure 4B). In addition, cells exposed to conditional media from RAW264.7 cells stimulated by PM2.5 particles demonstrated greater osteoclast differentiation from BMMs as demonstrated by TRAP staining and quantification of osteoclasts (Figure 4C,D).

Figure 4.

Figures 4A, 4D, 4E, 4H are bar graphs, plotting Tumor necrosis factor- lowercase alpha levels in a cell line of murine macrophages supernatant (picograms per milliliter), ranging from 0 to 600 in increments of 150; osteoclast number per well, ranging from 0 to 150 in increments of 30; Tumor necrosis factor- lowercase alpha levels in bone marrow macrophages supernatant (picograms per milliliter), ranging from 0 to 160 in increments of 40; osteoclast number per well, ranging from 0 to 400 in increments of 100 (y-axis) across 0 microgram per milliliter, 25 micrograms per milliliter, and 50 micrograms per milliliter (x-axis). Figures 4B and 4F are clustered bar graph, plotting relative messenger ribonucleic acid expression (fold change), ranging from 0 to 10 in increments of 2 and 0 to 8 in increments of 2 (y-axis) across acid phosphatase 5, Cathepsin K, nuclear factor of activated T cells cytoplasmic 1, F B J murine osteosarcoma viral oncogene homolog, Dendritic cell-specific transmembrane protein, ATPase, uppercase h positive transporting, lysosomal V 0 subunit D 2, and tumor necrosis factor receptor-associated factor 6 (x-axis) for 0 microgram per milliliter, 25 micrograms per milliliter, and 50 micrograms per milliliter. Figures 4C and 4G are stained tissues depicting Stimulation with conditional medium from particulate matter begin subscript 2.5 end subscript treated cell line of murine macrophages and Stimulation with conditional medium from particulate matter begin subscript 2.5 end subscript treated bone marrow macrophages displaying three columns, namely, 0 microgram per milliliter, 25 micrograms per milliliter, and 50 micrograms per milliliter and one column, namely, Tartrate-resistant acid phosphatase staining, respectively.

The effect of PM stimulation on osteoclastogenesis in vitro. Preosteoclasts were induced and treated with conditional medium from PM2.5-stimulated RAW264.7 macrophages and primary BMMs. (A) TNF-α release from RAW264.7 macrophages in the supernatant after 24-h incubation with PM2.5 particles dosed at 0, 25, and 50μg/mL. (B) Relative mRNA expression levels of molecules related to osteoclastogenesis in BMMs after treatment with 5% conditioned medium collected from supernatant of RAW264.7 macrophages. (C) Representative images of TRAP-stained cells treated with 5% conditioned medium collected from supernatant of 0, 25, and 50μg/mL PM2.5 particles–incubated RAW264.7 macrophages. Scale bar: 100μm. (D) Quantification of TRAP-positive multinucleated cells with more than three nuclei on each well. (E) TNF-α release from BMMs in the supernatant after 24-h incubation with PM2.5 particles dosed at 0, 25, and 50μg/mL. (F) Relative mRNA expression levels of molecules related to osteoclastogenesis in BMMs after treatment with 20% conditioned medium collected from supernatant of BMMs induced by PM2.5 particles. (G) Representative images of TRAP-stained cells treated with 20% conditioned medium collected from supernatant of 0, 25, and 50μg/mL PM2.5 particles–incubated BMMs. Scale bar: 100μm. (H) Quantification of TRAP-positive multinucleated cells with more than three nuclei on each well. Numeric data can be found in Table S13. Data are presented as means±SEMs (n=34) and assessed through one-way ANOVA. *p<0.05, **p<0.01, ***p<0.001 vs. the 0-μg/mL group. #p<0.05, ##p<0.01, ###p<0.001 vs. the 25-μg/mL group. Note: ACP5, acid phosphatase 5; ANOVA, analysis of variance; Atp6v0d2, ATPase, H+ transporting, lysosomal V0 subunit D2; BMMs, bone marrow macrophages; c-Fos, FBJ murine osteosarcoma viral oncogene homolog; Ctsk, cathepsin K; Dcstamp, dendritic cell-specific transmembrane protein; Nfatc1, nuclear factor of activated T cells cytoplasmic 1; PM, particulate matter; PM2.5, particulate matter with aerodynamic diameter of 2.5μm; RAW264.7, a cell line of murine macrophages; SEM, standard error of the mean; TNF-α, tumor necrosis factor-alpha; Traf6, tumor necrosis factor receptor-associated factor 6; TRAP, tartrate-resistant acid phosphatase; UD, undetectable.

We also consistently found that BMMs exposed to PM2.5 had higher levels of TNF-α (Figure 4E). In addition, BMMs exposed to PM2.5 also had greater osteoclast differentiation as demonstrated by TRAP staining and quantification of osteoclasts (Figure 4G,H). The mRNA expression of Traf6, Nfatc1, c-Fos, and Atp6v0d2 in BMMs was higher in cells exposed to PM2.5 at 50μg/mL but not at 25μg/mL (Figure 4F).

TNF-α Inhibition and PM2.5-stimulated Osteoclastogenesis in Vitro

We then further explored whether inhibition of TNF-α could attenuate the osteoclastic phenotypes induced by PM2.5 stimulation. First, we tested whether TNF-α neutralizing antibody (D2H4) could affect osteoclastogenesis in cells exposed to PM2.5. We found that cells treated with PM2.5 and D2H4 had fewer osteoclasts per well than those exposed to PM2.5 alone as determined by TRAP staining and subsequent quantification of osteoclasts (Figure 5A,B). Likewise, results from the bone resorption assay revealed a much greater resorption area formed in the PM2.5 group when compared with the control group. However, treatment with TNF-α neutralizing antibody showed a rescue in bone resorption (Figure 5C,D).

Figure 5.

Figure 5A is a stained tissue titled Tartrate-resistant acid phosphatase staining displaying three columns, namely, 0 microgram per milliliter, 50 micrograms per milliliter, and 50 micrograms per milliliter plus Tumor necrosis factor-lowercase alpha Neutralizing. Figures 5B and 5D are bar graphs, plotting osteoclast number per well, ranging from 0 to 400 in increments of 100 and bone resorption area (pixels), ranging from 0 to 40,000 in increments of 10,000 (y-axis) across 0 microgram per milliliter, 50 micrograms per milliliter, and 50 micrograms per milliliter plus Tumor necrosis factor-lowercase alpha Neutralizing (x-axis). Figure 5C is a stained tissue titled bone resorption displaying three columns, namely, 0 microgram per milliliter, 50 micrograms per milliliter, and 50 micrograms per milliliter plus Tumor necrosis factor-lowercase alpha Neutralizing. Figure 5E is a clustered bar graph, plotting relative messenger ribonucleic acid expression (fold change), ranging from 0 to 5 in unit increments (y-axis) across nuclear factor of activated T cells cytoplasmic 1, F B J murine osteosarcoma viral oncogene homolog, ATPase, uppercase h positive transporting, lysosomal V 0 subunit D 2, and tumor necrosis factor receptor-associated factor 6 (x-axis) for 0 microgram per milliliter, 50 micrograms per milliliter, and 50 micrograms per milliliter plus Tumor necrosis factor-lowercase alpha Neutralizing. Figure 5F is a western blot with four columns, namely, 0 microgram per milliliter, 25 micrograms per milliliter, 50 micrograms per milliliter, and 50 micrograms per milliliter plus Tumor necrosis factor-lowercase alpha Neutralizing and three rows, namely, tumor necrosis factor receptor-associated factor 6, F B J murine osteosarcoma viral oncogene homolog, and Glyceraldehyde 3-phosphate dehydrogenase. Figure 5G is a clustered bar graph, plotting relative protein expression (fold change), ranging from 0 to 4 in unit increments (y-axis) across tumor necrosis factor receptor-associated factor 6 and F B J murine osteosarcoma viral oncogene homolog (x-axis) for 0 microgram per milliliter, 25 micrograms per milliliter, 50 micrograms per milliliter, and 50 micrograms per milliliter plus Tumor necrosis factor-lowercase alpha Neutralizing. Figure 5H is a schematic diagram depicting the effect of particulate matter begin subscript 2.5 end subscript in bone homeostasis based on our findings, including macrophages, normal bone, and osteoporosis. The macrophages with tumor necrosis factor-lowercase alpha lead to circulation. The circulation then again leads to tumor necrosis factor-lowercase alpha. Tumor necrosis factor-lowercase alpha leads to O C N, and O C N with osteogenesis leads to osteoblast. Tumor necrosis factor-lowercase alpha leads to B M Ms, and B M Ms with osteoclastogenesis leads to osteoclast. The osteoblast and osteoclast are present between normal bone and osteoporosis.

The effect of TNF-α inhibition on PM2.5-stimulated osteoclastogenesis. Osteoclasts treated with conditional medium from PM2.5-stimulated primary BMMs were incubated with TNF-α neutralizing antibody D2H4. (A) Representative images of TRAP-stained BMMs treated with 20% conditioned medium and additional TNF-α neutralizing antibody D2H4. Scale bar: 100μm. (B) Quantification of TRAP-positive multinucleated cells with more than three nuclei on each well. (C) Representative images of bone resorption after treatment with 50μg/mL PM2.5 particles–incubated BMMs with or without D2H4. Scale bar: 50μm. (D) Quantification of bone resorbed area in plates coated with hydroxyapatite. (E) Relative mRNA expression levels of molecules related to osteoclastogenesis in BMMs after treatment with conditioned medium collected from supernatant of 50μg/mL PM2.5 particles incubation or plus TNF-α neutralizing antibody D2H4. (F) Representative bands of Traf6, c-Fos, and GAPDH in BMMs. (G) Quantification of Traf6 and c-Fos protein expression in PM-induced BMMs with or without D2H4. (H) Schematic diagram depicting the effect of PM2.5 in bone homeostasis based on our findings. Numeric data can be found in Table S14. Data are presented as means±SEMs (n=34) and assessed through ANOVA. *p<0.05, **p<0.01, ***p<0.001 vs. the 0-μg/mL group. #p<0.05, ##p<0.01 vs. the 25-μg/mL group. $p<0.05, $$p<0.01 vs. the 50-μg/mL group. Note: ANOVA, one-way analysis of variance; Atp6v0d2, ATPase, H+ transporting, lysosomal V0 subunit D2; BMMs, bone marrow macrophages; c-Fos, FBJ murine osteosarcoma viral oncogene homolog; D2H4, TNF-α neutralizing antibody; GAPDH, glyceraldehyde 3-phosphate dehydrogenase; Nfatc1, nuclear factor of activated T cells cytoplasmic 1; OCN, osteocalcin; PM, particulate matter; PM2.5, particulate matter with aerodynamic diameter of 2.5μm; SEM, standard error of the mean; TNF-α, tumor necrosis factor-alpha; Traf6, tumor necrosis factor receptor-associated factor 6; TRAP, tartrate-resistant acid phosphatase.

Next, we performed western blot and RT-PCR to detect the expression of downstream molecules expression. RT-PCR results showed that the higher mRNA expression of Nfact1, c-Fos, Atp6v0d2, and Traf6 associated with PM2.5 exposure were suppressed by D2H4 treatment (Figure 5E). In addition, both Traf6 and c-Fos protein expressions were significantly higher in cells exposed to conditional medium from RAW cells stimulated by PM2.5 in a dose-dependent manner. As expected, these elevated expressions were inhibited by TNF-α neutralizing antibody treatment as well (Figure 5F,G).

Discussion

In the present study, with the combination of an observational epidemiological study and an animal toxicological study, we assessed the association among long-term exposure to PM2.5, bone metabolism, and the pathogenesis of osteoporosis. Our analysis suggests that long-term exposure to PM2.5 was associated with lower BMD. Animal studies further presented shorter femur length, excessive osteoclast activity, inhibited osteoblastic bone formation, and higher TNF-α expression around the growth plate in response to PM2.5 challenge. In vitro experiments suggested that PM2.5 exposure promoted osteoclastogenesis through regulating the TNF-α/Traf6/c-Fos pathway by the activation of bone resorption. These data, taken together, suggest that long-term exposure to PM2.5 was associated with the dysfunction of bone metabolism and the pathogenesis of osteoporosis, which may be mediated by inflammation-induced osteoclastogensis around the growth plate.

Previous epidemiological studies reported inconsistent associations between PM2.5 exposure and BMD. For example, a cross-sectional study including 3,717 participants did not show a significant association between air pollution and lumbar spine BMD (mean difference of 0.018; 95% confidence interval: 0.038, 0.001).47 However, results from the population-based Oslo Health Study of 590 men revealed an inverse association between PM2.5 and BMD.20 The reasons for these conflicting results were complicated, such as different study designs (i.e., cross-sectional vs. cohort), or the age difference of study samples, or confounding factors that were not considered. In our study, we found that the association between PM2.5 and BMD were consistently observed at different anatomical sites in the human (such as heel, femur neck, and lumbar spine) even when we adjusted multiple BMD-related factors, including sex, age, BMI, ancestry, education, smoking, alcohol, physical activity, circulating calcium, and calcium supplementary status.

One limitation of our epidemiological study is that only a single measurement of air pollution was available. These annual average PM2.5 concentrations (available for the year 2010) were linked to each participant in the UK through participants’ residential addresses given at the baseline visit (2006–2010). Considering that air pollution emissions remained relatively stable from 2010 to 2019 in the UK,48,49 we used the annual mean PM2.5 concentrations at baseline (2010) as exposure, assuming that the pattern of PM2.5 concentrations would not have changed greatly during the period in this study. Previous studies, which used the same ambient air pollution data set from UK Biobank, have explored the influence of long-term exposure to ambient air pollution on mortality50 and mental health.49

To assess the potential mechanism for the association observed in humans, we further conducted an animal study on bone health in a setting with ambient levels of PM2.5 exposure. Mice exposed to PM2.5 for 6 months exhibited shorter average femoral length. Mice incorporated into the study at 5 wk of age were exposed to PM2.5 for 6 months, roughly spanning the life stages from childhood to adolescence and middle age. The life stages of childhood and adolescence are particularly essential for bone modeling, including the formation of bone shape and growth of bone size.51 Several population studies have demonstrated that air pollution in childhood may be associated with transiently shorter height, as well as slower growth rate, but the underlying mechanism is unknown.5254

Bone remodeling predominating in adulthood is pivotal to maintain bone homeostasis, which is achieved by the balance of bone formation and bone resorption.51 Bone remodeling is also crucial to the process of bone growth and shape alteration. As to bone formation, the most classic and sensitive markers are OCN and BALP.55 In our study, lower levels of OCN in serum and the femur in PM2.5-exposed animals suggest an inhibition of activation of osteogenesis by PM2.5 exposure. However, ALP, another bone formation marker, showed no difference in the femur but did show a higher level in serum in PM2.5-exposed mice. Bone-type ALP is a kind of tissue-nonspecific alkaline phosphatase and is expressed in both bone and liver.56 Hepatic levels of ALP were examined to address the source of BALP in serum. Although significantly higher ALP expression was detected in the liver, suggesting that the liver contributes to the accumulation of circulating BALP, the function of liver-derived ALP in the mineralization process and whether it is adaptive to long-term PM2.5 exposure require further study. The lower OCN expression may indicate that osteogenesis could be inhibited by PM2.5 exposure. Hence, even though no difference in bone structure or BMD was observed in the animal studies, the combination of our population analysis results and some experimental evidence consistently supported the effects of PM2.5 exposure on bone health.

In terms of bone resorption, RANKL is essential for osteoclast formation in osteocytes,57 TRAP staining around the growth plate and circulating TRACP-5b level are the most important biomarkers reflecting the amount and activity of osteoclasts on bone surface.58 In addition, Ctsk is highly expressed in activated osteoclasts as a biomarker of osteoclastogenesis.59 Convincingly and first, the elevated levels of all these bone resorption biomarkers were demonstrated in the present study. Moreover, the calcified cartilaginous template was destroyed with signs of osteoclast activation around the growth plate in mice exposed to PM2.5 as well, which may lead to damage to the growth plate structure and consequential aberrant long bone growth during the growing stage.60 In addition, results from cell TRAP staining and bone resorption assay also confirmed that cells exposed to PM2.5 had abnormal osteoclast formation and function in vitro. It is important to note that the excessive osteoclast activity shown in vivo and in vitro did not associate with differences in trabecular bone mass. Assessment of an earlier time point in the study should be evaluated in future work given that by the end of this study, the trabecular bone was largely gone. Unlike the human population results, our animal results did not show a significant difference in bone microarchitecture or BMD in the PM2.5-exposed individuals; this could be explained by the incomplete consistence in exposure concentration or exposure time between UK Biobank data and animal data. Taken together, the results of this study suggest that osteoclast activity could be prominently enhanced by PM2.5 exposure.

According to our experimental results and previous studies, an imbalance between bone resorption and bone formation could be induced by PM2.5 exposure through various ways, including inflammation,61 liver toxicity,62 and immunoreaction.63,64 However, the potential mechanism by which PM2.5 exposure disturbed bone metabolism remains unknown. In light of the close association of the immune system in PM2.5 effects,64 we hypothesized that inflammation may be involved in the adverse effects of PM2.5 exposure on bone health. Recent studies have indicated a role of inflammation in regulating proliferation and differentiation of osteoclasts and osteoblasts46,65,66 Therefore, we examined the expression of proinflammatory cytokines specifically around the growth plate area. TNF-α has been shown to be involved in bone homeostasis by directly potentiating the expression of RANKL to enhance the bone resorption process and impairing the osteoblast performance.67,68 Thus, the higher expression of TNF-α in PM2.5-exposed cells/animals may disturb the balance by activating osteoclastogenesis as well as by inhibiting osteogenesis.

The expression of Traf6 is closely related to TNF-α.69 In addition, Traf6 is one of the key molecules for osteoclastogenesis, as demonstrated by a study in which Traf6-knockout mice developed severe osteopetrosis.70 Briefly, during the osteoclast differentiation upon RANKL induction, the binding of Traf6 to RANK results in the activation of downstream regulators, such as Nfatc1 and c-Fos, and then induces expression of osteoclast-specific genes.7173 Here, our data show that the wells treated with PM2.5 had a higher number of osteoclasts, higher expression of Traf6 and c-Fos, as well as more bone resorption. Most importantly, neutralizing TNF-α could partially or completely block all these altered markers or molecules for osteoclast formation observed with PM2.5 stimulation. Hence, our in vitro and in vivo results suggest that PM2.5 stimulation can facilitate osteoclastogenesis through the TNF-α/Traf6/c-Fos pathway.

In conclusion, we found that long-term exposure to PM2.5 exposure was associated with an increased risk of osteoporosis with analysis of UK Biobank data, and we further demonstrated PM2.5 exposure may promote osteoclastogenesis by the TNF-α-mediated Traf6/c-Fos pathway in vivo (Figure 5H). These findings provide a novel insight into bone homeostasis in response to long-tern PM2.5 exposure.

Supplementary Material

Acknowledgments

We thank the High-Performance Computing Center at Westlake University for the facility support and technical assistance. We are also very grateful to Hui Wang from Wuhan University for technique support.

This work was supported by the National Key Research and Development Program of China (grant 2019YFE0114500, to C.L.), National Natural Science Foundation of China (grant 82273590 and 81973001, to C.L.; 82173480, to R.L.; 81973869, to H.J.; 32061143019, to H.F.Z.), the State Administration of Traditional Chinese Medicine of Zhejiang Province (grant 2021ZZ014 to H.J.), and the Natural Science Foundation of Zhejiang Province (grant LR23H270001 to H.J.). This research has been conducted using the UK Biobank Resource under application number 41376 (https://www.ukbiobank.ac.uk/enable-your-research/approved-research?anid=41376).

The individual-level phenotype data requires permission from the UK Biobank and the code is available upon request from the corresponding authors.

References

  • 1.Burge R, Dawson-Hughes B, Solomon DH, Wong JB, King A, Tosteson A. 2007. Incidence and economic burden of osteoporosis-related fractures in the United States, 2005–2025. J Bone Miner Res 22(3):465–475, PMID: , 10.1359/jbmr.061113. [DOI] [PubMed] [Google Scholar]
  • 2.Wright NC, Looker AC, Saag KG, Curtis JR, Delzell ES, Randall S, et al. 2014. The recent prevalence of osteoporosis and low bone mass in the United States based on bone mineral density at the femoral neck or lumbar spine. J Bone Miner Res 29(11):2520–2526, PMID: , 10.1002/jbmr.2269. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Zhu X, Bai W, Zheng H. 2021. Twelve years of GWAS discoveries for osteoporosis and related traits: advances, challenges and applications. Bone Res 9(1):23, PMID: , 10.1038/s41413-021-00143-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Nguyen T, Sambrook P, Kelly P, Jones G, Lord S, Freund J, et al. 1993. Prediction of osteoporotic fractures by postural instability and bone density. BMJ 307(6912):1111–1115, PMID: , 10.1136/bmj.307.6912.1111. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Hernlund E, Svedbom A, Ivergård M, Compston J, Cooper C, Stenmark J, et al. 2013. Osteoporosis in the European Union: medical management, epidemiology and economic burden. A report prepared in collaboration with the International Osteoporosis Foundation (IOF) and the European Federation of Pharmaceutical Industry Associations (EFPIA). Arch Osteoporos 8(1):136, PMID: , 10.1007/s11657-013-0136-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Langdahl B, Ferrari S, Dempster DW. 2016. Bone modeling and remodeling: potential as therapeutic targets for the treatment of osteoporosis. Ther Adv Musculoskelet Dis 8(6):225–235, PMID: , 10.1177/1759720X16670154. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Shi GX, Mao WW, Zheng XF, Jiang LS. 2016. The role of R-spondins and their receptors in bone metabolism. Prog Biophys Mol Biol 122(2):93–100, PMID: , 10.1016/j.pbiomolbio.2016.05.012. [DOI] [PubMed] [Google Scholar]
  • 8.Mödinger Y, Löffler B, Huber-Lang M, Ignatius A. 2018. Complement involvement in bone homeostasis and bone disorders. Semin Immunol 37:53–65, PMID: , 10.1016/j.smim.2018.01.001. [DOI] [PubMed] [Google Scholar]
  • 9.Al Saedi A, Stupka N, Duque G. 2020. Pathogenesis of osteoporosis. Handb Exp Pharmacol 262:353–367, PMID: , 10.1007/164_2020_358. [DOI] [PubMed] [Google Scholar]
  • 10.Franceschi R, Longhi S, Cauvin V, Fassio A, Gallo G, Lupi F, et al. 2018. Bone geometry, quality, and bone markers in children with type 1 diabetes mellitus. Calcif Tissue Int 102(6):657–665, PMID: , 10.1007/s00223-017-0381-1. [DOI] [PubMed] [Google Scholar]
  • 11.Zhu X, Zheng H. 2021. Factors influencing peak bone mass gain. Front Med 15(1):53–69, PMID: , 10.1007/s11684-020-0748-y. [DOI] [PubMed] [Google Scholar]
  • 12.Xing YF, Xu YH, Shi MH, Lian YX. 2016. The impact of PM2.5 on the human respiratory system. J Thorac Dis 8(1):E69–E74, PMID: , 10.3978/j.issn.2072-1439.2016.01.19. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Shamsa EH, Song Z, Kim H, Shamsa F, Hazlett LD, Zhang K. 2022. The links of fine airborne particulate matter exposure to occurrence of cardiovascular and metabolic diseases in Michigan, USA. PLOS Glob Public Health 2(8):e0000707, PMID: , 10.1371/journal.pgph.0000707. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Li J, Song Y, Shi L, Jiang J, Wan X, Wang Y, et al. 2023. Long-term effects of ambient PM2.5 constituents on metabolic syndrome in Chinese children and adolescents. Environ Res 220:115238, PMID: , 10.1016/j.envres.2023.115238. [DOI] [PubMed] [Google Scholar]
  • 15.Wang Y, Xiong L, Tang M. 2017. Toxicity of inhaled particulate matter on the central nervous system: neuroinflammation, neuropsychological effects and neurodegenerative disease. J Appl Toxicol 37(6):644–667, PMID: , 10.1002/jat.3451. [DOI] [PubMed] [Google Scholar]
  • 16.Chang KH, Chang MY, Muo CH, Wu TN, Hwang BF, Chen CY, et al. 2015. Exposure to air pollution increases the risk of osteoporosis: a nationwide longitudinal study. Medicine (Baltimore) 94(17):e733, PMID: , 10.1097/MD.0000000000000733. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Chen Z, Salam MT, Karim R, Toledo-Corral CM, Watanabe RM, Xiang AH, et al. 2015. Living near a freeway is associated with lower bone mineral density among Mexican Americans. Osteoporos Int 26(6):1713–1721, PMID: , 10.1007/s00198-015-3051-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Prada D, Zhong J, Colicino E, Zanobetti A, Schwartz J, Dagincourt N, et al. 2017. Association of air particulate pollution with bone loss over time and bone fracture risk: analysis of data from two independent studies. Lancet Planet Health 1(8):e337–e347, PMID: , 10.1016/S2542-5196(17)30136-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Sung JH, Kim K, Cho Y, Choi S, Chang J, Kim SM, et al. 2020. Association of air pollution with osteoporotic fracture risk among women over 50 years of age. J Bone Miner Metab 38(6):839–847, PMID: , 10.1007/s00774-020-01117-x. [DOI] [PubMed] [Google Scholar]
  • 20.Alvaer K, Meyer HE, Falch JA, Nafstad P, Søgaard AJ. 2007. Outdoor air pollution and bone mineral density in elderly men—the Oslo Health Study. Osteoporos Int 18(12):1669–1674, PMID: , 10.1007/s00198-007-0424-y. [DOI] [PubMed] [Google Scholar]
  • 21.Liu JJ, Fu SB, Jiang J, Tang XL. 2021. Association between outdoor particulate air pollution and the risk of osteoporosis: a systematic review and meta-analysis. Osteoporos Int 32(10):1911–1919, PMID: , 10.1007/s00198-021-05961-z. [DOI] [PubMed] [Google Scholar]
  • 22.Pope CA III, Bhatnagar A, McCracken JP, Abplanalp W, Conklin DJ, O’Toole T. 2016. Exposure to fine particulate air pollution is associated with endothelial injury and systemic inflammation. Circ Res 119(11):1204–1214, PMID: , 10.1161/CIRCRESAHA.116.309279. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Guan L, Geng X, Stone C, Cosky EEP, Ji Y, Du H, et al. 2019. PM2.5 exposure induces systemic inflammation and oxidative stress in an intracranial atherosclerosis rat model. Environ Toxicol 34(4):530–538, PMID: , 10.1002/tox.22707. [DOI] [PubMed] [Google Scholar]
  • 24.Liu C, Fonken LK, Wang A, Maiseyeu A, Bai Y, Wang TY, et al. 2014. Central IKKβ inhibition prevents air pollution mediated peripheral inflammation and exaggeration of type II diabetes. Part Fibre Toxicol 11:53, PMID: , 10.1186/s12989-014-0053-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Takakura N, Matsuda M, Khan M, Hiura F, Aoki K, Hirohashi Y, et al. 2020. A novel inhibitor of NF-κB-inducing kinase prevents bone loss by inhibiting osteoclastic bone resorption in ovariectomized mice. Bone 135:115316, PMID: , 10.1016/j.bone.2020.115316. [DOI] [PubMed] [Google Scholar]
  • 26.Swarnkar G, Zhang K, Mbalaviele G, Long F, Abu-Amer Y. 2014. Constitutive activation of IKK2/NF-κB impairs osteogenesis and skeletal development. PLoS One 9(3):e91421, PMID: , 10.1371/journal.pone.0091421. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Xia JW, Zhang L, Li J, Yuan CD, Zhu XW, Qian Y, et al. 2022. Both indirect maternal and direct fetal genetic effects reflect the observational relationship between higher birth weight and lower adult bone mass. BMC Med 20(1):361, PMID: , 10.1186/s12916-022-02531-w. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Zhu XW, Liu KQ, Yuan CD, Xia JW, Qian Y, Xu L, et al. 2022. General and abdominal obesity operate differently as influencing factors of fracture risk in old adults. iScience 25(6):104466, PMID: , 10.1016/j.isci.2022.104466. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Bai WY, Wang L, Ying ZM, Hu B, Xu L, Zhang GQ, et al. 2020. Identification of PIEZO1 polymorphisms for human bone mineral density. Bone 133:115247, PMID: , 10.1016/j.bone.2020.115247. [DOI] [PubMed] [Google Scholar]
  • 30.Xia J, Xie SY, Liu KQ, Xu L, Zhao PP, Gai SR, et al. 2020. Systemic evaluation of the relationship between psoriasis, psoriatic arthritis and osteoporosis: observational and Mendelian randomisation study. Ann Rheum Dis 79(11):1460–1467, PMID: , 10.1136/annrheumdis-2020-217892. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Qian Y, Xia J, Liu KQ, Xu L, Xie SY, Chen GB, et al. 2021. Observational and genetic evidence highlight the association of human sleep behaviors with the incidence of fracture. Commun Biol 4(1):1339, PMID: , 10.1038/s42003-021-02861-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Luo H, Zhang Q, Yu K, Meng X, Kan H, Chen R. 2022. Long-term exposure to ambient air pollution is a risk factor for trajectory of cardiometabolic multimorbidity: a prospective study in the UK biobank. EBioMedicine 84:104282, PMID: , 10.1016/j.ebiom.2022.104282. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Eeftens M, Beelen R, de Hoogh K, Bellander T, Cesaroni G, Cirach M, et al. 2012. Development of land use regression models for PM2.5, PM2.5 absorbance, PM10 and PMcoarse in 20 European study areas; results of the ESCAPE project. Environ Sci Technol 46(20):11195–11205, PMID: , 10.1021/es301948k. [DOI] [PubMed] [Google Scholar]
  • 34.Li R, Wang Y, Hou B, Lam SM, Zhang W, Chen R, et al. 2020. Lipidomics insight into chronic exposure to ambient air pollution in mice. Environ Pollut 262:114668, PMID: , 10.1016/j.envpol.2020.114668. [DOI] [PubMed] [Google Scholar]
  • 35.Wang Y, Li R, Chen R, Gu W, Zhang L, Gu J, et al. 2020. Ambient fine particulate matter exposure perturbed circadian rhythm and oscillations of lipid metabolism in adipose tissues. Chemosphere 251:126392, PMID: , 10.1016/j.chemosphere.2020.126392. [DOI] [PubMed] [Google Scholar]
  • 36.Yang S, Chen R, Zhang L, Sun Q, Li R, Gu W, et al. 2021. Lipid metabolic adaption to long-term ambient PM2.5 exposure in mice. Environ Pollut 269:116193, PMID: , 10.1016/j.envpol.2020.116193. [DOI] [PubMed] [Google Scholar]
  • 37.Ge Q, Ying J, Shi Z, Mao Q, Jin H, Wang PE, et al. 2021. Chlorogenic acid retards cartilaginous endplate degeneration and ameliorates intervertebral disc degeneration via suppressing NF-κB signaling. Life Sci 274:119324, PMID: , 10.1016/j.lfs.2021.119324. [DOI] [PubMed] [Google Scholar]
  • 38.Chen H, Hu B, Lv X, Zhu S, Zhen G, Wan M, et al. 2019. Prostaglandin E2 mediates sensory nerve regulation of bone homeostasis. Nat Commun 10(1):181, PMID: , 10.1038/s41467-018-08097-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Schneider CA, Rasband WS, Eliceiri KW. 2012. NIH image to ImageJ: 25 years of image analysis. Nat Methods 9(7):671–675, PMID: , 10.1038/nmeth.2089. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Kwon OC, Choi B, Lee EJ, Park JE, Lee EJ, Kim EY, et al. 2020. Negative regulation of osteoclast commitment by intracellular protein phosphatase magnesium-dependent 1A. Arthritis Rheumatol 72(5):750–760, PMID: , 10.1002/art.41180. [DOI] [PubMed] [Google Scholar]
  • 41.Hu S, Ge Q, Xia C, Ying J, Ruan H, Shi Z, et al. 2020. Bushenhuoxue formula accelerates fracture healing via upregulation of TGF-β/Smad2 signaling in mesenchymal progenitor cells. Phytomedicine 76:153256, PMID: , 10.1016/j.phymed.2020.153256. [DOI] [PubMed] [Google Scholar]
  • 42.Mitkus RJ, Powell JL, Zeisler R, Squibb KS. 2013. Comparative physicochemical and biological characterization of NIST Interim Reference Material PM2.5 and SRM 1648 in human A549 and mouse RAW264.7 cells. Toxicol In Vitro 27(8):2289–2298, PMID: , 10.1016/j.tiv.2013.09.024. [DOI] [PubMed] [Google Scholar]
  • 43.Bouxsein ML, Boyd SK, Christiansen BA, Guldberg RE, Jepsen KJ, Müller R. 2010. Guidelines for assessment of bone microstructure in rodents using micro-computed tomography. J Bone Miner Res 25(7):1468–1486, PMID: , 10.1002/jbmr.141. [DOI] [PubMed] [Google Scholar]
  • 44.Hu K, Olsen BR. 2016. Osteoblast-derived VEGF regulates osteoblast differentiation and bone formation during bone repair. J Clin Invest 126(2):509–526, PMID: , 10.1172/JCI82585. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Xiong J, Cawley K, Piemontese M, Fujiwara Y, Zhao H, Goellner JJ, et al. 2018. Soluble RANKL contributes to osteoclast formation in adult mice but not ovariectomy-induced bone loss. Nat Commun 9(1):2909, PMID: , 10.1038/s41467-018-05244-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Yang B, Sun H, Xu X, Zhong H, Wu Y, Wang J. 2020. YAP1 inhibits the induction of TNF-α-stimulated bone-resorbing mediators by suppressing the NF-κB signaling pathway in MC3T3-E1 cells. J Cell Physiol 235(5):4698–4708, PMID: , 10.1002/jcp.29348. [DOI] [PubMed] [Google Scholar]
  • 47.Ranzani OT, Milà C, Kulkarni B, Kinra S, Tonne C. 2020. Association of ambient and household air pollution with bone mineral content among adults in peri-urban South India. JAMA Netw Open 3(1):e1918504, PMID: , 10.1001/jamanetworkopen.2019.18504. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Department for Environment, Food & Rural Affairs. 2022. Air quality statistics: an annual update on concentrations of major air pollutants in the UK. Last updated 23 September 2020. https://webarchive.nationalarchives.gov.uk/ukgwa/20201225100256/https://www.gov.uk/government/statistics/air-quality-statistics [accessed 1 October 2022].
  • 49.Yang T, Wang J, Huang J, Kelly FJ, Li G. 2023. Long-term exposure to multiple ambient air pollutants and association with incident depression and anxiety. JAMA Psychiatry 80(4):305–313, PMID: , 10.1001/jamapsychiatry.2022.4812. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Wang M, Zhou T, Song Q, Ma H, Hu Y, Heianza Y, et al. 2022. Ambient air pollution, healthy diet and vegetable intakes, and mortality: a prospective UK Biobank study. Int J Epidemiol 51(4):1243–1253, PMID: , 10.1093/ije/dyac022. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Katsimbri P. 2017. The biology of normal bone remodelling. Eur J Cancer Care (Engl) 26(6):e12740, PMID: , 10.1111/ecc.12740. [DOI] [PubMed] [Google Scholar]
  • 52.Bobak M, Richards M, Wadsworth M. 2004. Relation between children’s height and outdoor air pollution from coal-burning sources in the British 1946 birth cohort. Int Arch Occup Environ Health 77(6):383–386, PMID: , 10.1007/s00420-004-0522-5. [DOI] [PubMed] [Google Scholar]
  • 53.Huang JV, Leung GM, Schooling CM. 2018. The association of air pollution with height: evidence from Hong Kong’s “Children of 1997” birth cohort. Am J Hum Biol 30(1):e23067, PMID: , 10.1002/ajhb.23067. [DOI] [PubMed] [Google Scholar]
  • 54.Nikolić M, Stanković A, Jović S, Kocić B, Bogdanović D. 2014. Effects of air pollution on growth in schoolchildren. Coll Antropol 38(2):493–497, PMID: . [PubMed] [Google Scholar]
  • 55.Diemar SS, Møllehave LT, Quardon N, Lylloff L, Thuesen BH, Linneberg A, et al. 2020. Effects of age and sex on osteocalcin and bone-specific alkaline phosphatase—reference intervals and confounders for two bone formation markers. Arch Osteoporos 15(1):26, PMID: , 10.1007/s11657-020-00715-6. [DOI] [PubMed] [Google Scholar]
  • 56.Baba TT, Terashima T, Oida S. 2019. Liver-type of tissue non-specific alkaline phosphatase is induced during mouse bone and tooth cell differentiation. Arch Oral Biol 98:32–37, PMID: , 10.1016/j.archoralbio.2018.10.036. [DOI] [PubMed] [Google Scholar]
  • 57.Xiong J, Onal M, Jilka RL, Weinstein RS, Manolagas SC, O’Brien CA. 2011. Matrix-embedded cells control osteoclast formation. Nat Med 17(10):1235–1241, PMID: , 10.1038/nm.2448. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Gao SG, Li KH, Xu M, Jiang W, Shen H, Luo W, et al. 2011. Bone turnover in passive smoking female rat: relationships to change in bone mineral density. BMC Musculoskelet Disord 12:131, PMID: , 10.1186/1471-2474-12-131. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Dai R, Wu Z, Chu HY, Lu J, Lyu A, Liu J, et al. 2020. Cathepsin K: the action in and beyond bone. Front Cell Dev Biol 8:433, PMID: , 10.3389/fcell.2020.00433. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Knowles HJ, Moskovsky L, Thompson MS, Grunhen J, Cheng X, Kashima TG, et al. 2012. Chondroclasts are mature osteoclasts which are capable of cartilage matrix resorption. Virchows Arch 461(2):205–210, PMID: , 10.1007/s00428-012-1274-3. [DOI] [PubMed] [Google Scholar]
  • 61.Prada D, López G, Solleiro-Villavicencio H, Garcia-Cuellar C, Baccarelli AA. 2020. Molecular and cellular mechanisms linking air pollution and bone damage. Environ Res 185:109465, PMID: , 10.1016/j.envres.2020.109465. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Lu K, Shi TS, Shen SY, Shi Y, Gao HL, Wu J, et al. 2022. Defects in a liver-bone axis contribute to hepatic osteodystrophy disease progression. Cell Metab 34(3):441–457.e7, PMID: , 10.1016/j.cmet.2022.02.006. [DOI] [PubMed] [Google Scholar]
  • 63.Adamopoulos IE, Bowman EP. 2008. Immune regulation of bone loss by Th17 cells. Arthritis Res Ther 10(5):225, PMID: , 10.1186/ar2502. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Castañeda AR, Pinkerton KE, Bein KJ, Magaña-Méndez A, Yang HT, Ashwood P, et al. 2018. Ambient particulate matter activates the aryl hydrocarbon receptor in dendritic cells and enhances Th17 polarization. Toxicol Lett 292:85–96, PMID: , 10.1016/j.toxlet.2018.04.020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Yan Z, Zhu S, Wang H, Wang L, Du T, Ye Z, et al. 2019. MOTS-c inhibits osteolysis in the mouse calvaria by affecting osteocyte–osteoclast crosstalk and inhibiting inflammation. Pharmacol Res 147:104381, PMID: , 10.1016/j.phrs.2019.104381. [DOI] [PubMed] [Google Scholar]
  • 66.Baum R, Gravallese EM. 2014. Impact of inflammation on the osteoblast in rheumatic diseases. Curr Osteoporos Rep 12(1):9–16, PMID: , 10.1007/s11914-013-0183-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Marahleh A, Kitaura H, Ohori F, Kishikawa A, Ogawa S, Shen WR, et al. 2019. TNF-α directly enhances osteocyte RANKL expression and promotes osteoclast formation. Front Immunol 10:2925, PMID: , 10.3389/fimmu.2019.02925. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Cavagis A, Takamori E, Granjeiro J, Oliveira R, Ferreira C, Peppelenbosch M, et al. 2014. TNFα contributes for attenuating both Y397FAK and Y416Src phosphorylations in osteophosphorylations in osteoblasts. Oral Dis 20(8):780–786, PMID: , 10.1111/odi.12202. [DOI] [PubMed] [Google Scholar]
  • 69.Li J, Yi X, Yao Z, Chakkalakal JV, Xing L, Boyce BF. 2020. TNF receptor-associated factor 6 mediates TNFα-induced skeletal muscle atrophy in mice during aging. J Bone Miner Res 35(8):1535–1548, PMID: , 10.1002/jbmr.4021. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Lomaga MA, Yeh WC, Sarosi I, Duncan GS, Furlonger C, Ho A, et al. 1999. TRAF6 deficiency results in osteopetrosis and defective interleukin-1, CD40, and LPS signaling. Genes Dev 13(8):1015–1024, PMID: , 10.1101/gad.13.8.1015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Lee JM, Kim MJ, Lee SJ, Kim BG, Choi JY, Lee SM. 2021. PDK2 deficiency prevents ovariectomy-induced bone loss in mice by regulating the RANKL-NFATc1 pathway during osteoclastogenesis. J Bone Miner Res 36(3):553–566, PMID: , 10.1002/jbmr.4202. [DOI] [PubMed] [Google Scholar]
  • 72.Shu B, Zhao Y, Zhao S, Pan H, Xie R, Yi D, et al. 2020. Inhibition of Axin1 in osteoblast precursor cells leads to defects in postnatal bone growth through suppressing osteoclast formation. Bone Res 8:31, PMID: , 10.1038/s41413-020-0104-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Taganov KD, Boldin MP, Chang KJ, Baltimore D. 2006. NF-κB-dependent induction of microRNA miR-146, an inhibitor targeted to signaling proteins of innate immune responses. Proc Natl Acad Sci USA 103(33):12481–12486, PMID: , 10.1073/pnas.0605298103. [DOI] [PMC free article] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials


Articles from Environmental Health Perspectives are provided here courtesy of National Institute of Environmental Health Sciences

RESOURCES