Introduction
Psoriasis is a common inflammatory skin disease characterized by an abnormal hyperproliferation of keratinocytes, activated dendritic cells and infiltration of T lymphocytes in lesions (Lin et al,
2018). The incidence of psoriasis in Europeans is ∼3%, and it has been estimated that ∼30% of psoriasis patients develop psoriatic arthritis (PsA), a systemic chronic inflammatory disease with clinical features such as arthritis, enthesitis, dactylitis, tendonitis, and cutaneous psoriasis (Cafaro and McInnes,
2018). PsA is often classified into five subtypes: distal interphalangeal predominant, asymmetric oligoarticular, symmetric polyarthritis, spondylitis, and psoriatic arthritis mutilans (PAM) (Moll and Wright,
1973). PAM is the rarest and most severe form of PsA and is characterized by the shortening of one or more digits, due to severe osteolysis of the bones, a deformity known as “digital telescoping” or “opera glass finger”. PAM patients suffer from severe joint destruction causing flail joints, and the progress of the deformities is rapid once the disease starts (Laasonen et al,
2020; Mochizuki et al,
2018). The skin phenotype in PAM patients is often described as mild (Gudbjornsson et al,
2013). Even though the overall prevalence of PAM is uncertain, several case studies have reported on PAM patients in different populations (Laasonen et al,
2015; Mochizuki et al,
2018; Perrotta et al,
2019; Qin and Beach,
2019), and in a Nordic PAM study it was estimated to have a prevalence of 3.7 cases per million habitants (Gudbjornsson et al,
2013). Clinical and radiographic details of patients in the Nordic PAM study have been described (Gudbjornsson et al,
2013; Laasonen et al,
2015; Laasonen et al,
2020; Lindqvist et al,
2017; Mistegard et al,
2021).
Genetic factors play an important role in the development of psoriasis and PsA, with dozens of susceptibility genes identified, and many but not all genetic signals overlapping (Cafaro and McInnes,
2018). Most of the known susceptibility genes act via the HLA locus, IFN, NF-κB, and IL23/17 signaling pathways, with some genes involved in skin barrier integrity such as
LCE3B-LCE3C (Cafaro and McInnes,
2018). It is also thought that genetic and environmental factors such as smoking, injuries and infections play a role in the etiology of psoriatic disorders (Yan et al,
2021). Humans have suffered from PAM since ancient times, skeletal remains with characteristic lesions of PAM have been found in a Byzantine monastery in Israel (Zias and Mitchell,
1996). Today, PAM has been reported in many studies from all over the world, reviewed in (Bruzzese et al,
2013), but the true prevalence of PAM is difficult to determine due to difficulties in clinical diagnosis and lack of biomarkers.
The nicotinamide adenine dinucleotide phosphate oxidase 4 (
NOX4) gene (OMIM:
605261) encodes a protein that contains six transmembrane domains and in its cytosolic part, a flavin adenine dinucleotide (FAD) and a NADPH-binding domain. NOX4 is an enzyme involved in the production of reactive oxygen species (ROS), a group of highly reactive molecules important in the regulation of signal transduction (Brown and Griendling,
2009). NOX4 predominantly generates hydrogen peroxide (H
2O
2) and is constitutively active, unlike the other members of the oxidase family (Schroder,
2019).
NOX4 is expressed in many cell types, including keratinocytes and osteoclasts (Brown and Griendling,
2009). Several studies have linked high levels of ROS to conditions such as cancer, inflammatory diseases, vascular disease, diabetes and osteoporosis (Yang and Lian,
2020). Furthermore, during bone formation, the balance between osteoblasts (bone-forming cells) and osteoclasts (bone-resorbing cells) differentiation and activity is thought to be affected by ROS (Schroder,
2015). In vitro and in vivo studies have shown that increased production of NOX4 leads to increased osteoclastogenesis (Garrett et al,
1990; Lee et al,
2005; Schroder,
2019; Yang et al,
2001). Abnormal regulation of osteoclasts activity is involved in pathological bone resorption in osteoporosis, autoimmune arthritis and bone cancer (Takayanagi,
2021). A previous study identified an intronic SNP that increases the expression of
NOX4 being associated with reduced bone density and increased markers for bone turnover in middle-aged women compared to normal controls (Goettsch et al,
2013). Conversely, studies in mice show that depletion of NOX4 leads to increased trabecular bone density, and inhibition of NOX4 prevents bone loss (Yang et al,
2001).
In this study, we applied massive parallel sequencing to the whole PAM cohort and found that rare variants in the NOX4 gene found in four PAM patients might significantly increase the levels of ROS and specifically the levels of H2O2. To test the hypothesis, we applied in vitro and in vivo models, including patient-derived osteoclasts, stable cell lines overexpressing the variants found, direct measurement of ROS in patient blood samples, and zebrafish models. Further genetic analysis of patients without NOX4 pathogenic variants revealed other rare variants potentially pathogenic in genes related to NOX4. All the data obtained indicate a putative connection between high levels of ROS and the development of PAM.
Discussion
The NOX gene family is comprised of seven members
NOX1-NOX5,
DUOX1, and
DUOX2 (Wegner and Haudenschild,
2020). They are specialized ROS producers and differ in their cellular and tissue-specific distributions. Impairment in the regulation of NOX expression results in pathologies such as atherosclerosis, hypertension, diabetic nephropathy, lung fibrosis, cancers, and neurodegenerative diseases (Vermot et al,
2021).
NOX4 specifically has been linked to osteoporosis, inflammatory arthritis and osteoarthritis (Wegner and Haudenschild,
2020). The role of
NOX4 in those pathologies has been suggested by tissue-specific-expression studies, functional biochemical assays, and observations in animal models (Drevet et al,
2018; Goettsch et al,
2013; Gray et al,
2019), but to our knowledge no disease-causing variants in
NOX4 have yet been found in patients.
Our study identified
NOX4 as the first candidate susceptibility gene for psoriatic arthritis mutilans (PAM), the rarest and most severe form of psoriatic arthritis. Despite the severity of the disease, no specific treatment or biomarkers have been identified to date. Here, we describe three protein-coding rare variants
; two missense (
NOX4V369F and
NOX4Y512C) and one frameshift (
NOX4Y512IfsX20), in four PAM patients, all located in the cytosolic part of NOX4, affecting the FAD and NADPH-binding domains, important for the transfer of electrons and formation of ROS (Magnani et al,
2017). In our genetic analysis, we did not find any other
NOX4 rare variants in the rest of the PAM cohort (
n = 57), but we cannot exclude that these patients may carry other variants located in intergenic or intragenic regions which would escape our analysis as most of the patients were sequenced only for exomes. It is also possible that other non-ROS-related pathways could be implicated in the development of PAM. Through further genetic analysis of the three rare variants in psoriasis, PsA and control cohorts, we found three additional carriers of the
NOX4Y512C, all in the PsA group whereas none in the psoriasis group nor in healthy controls (Table
3). Of the non-PAM patients carrying
NOX4 mutations, only PsA961 was available for further clinical examination. He presented a mild PsA phenotype affecting peripheral joints without any evidence of bone destruction. It should be noted that his PsA is of short duration, and it was his cutaneous psoriasis that motivated anti-TNF therapy. Thus, we cannot know whether he would have developed a more advanced phenotype without systemic therapeutic intervention. Also, the pathogenetic architecture in PAM is likely complex and we do not have a complete picture.
Further analysis of PAM patient sequences revealed rare variants in other genes potentially altering the levels of ROS and/or affecting osteoclast differentiation including the transcription factor
NFATc1, CSF1R, NOXO, DUOX1, RYR1, RYR2, and
RYR3. Interestingly, the
NFATc1 gene is a master regulator of RANKL-induced osteoclastogenesis and the
Nfatc1 conditional knockout mouse develops osteopetrosis, a condition characterized by increased bone density due to decreased or absent osteoclast activity (Aliprantis et al,
2008).
CSF1R is involved in osteoclast proliferation, and its suppression has been shown to attenuate pathological bone resorption in inflammatory arthritis, inflammatory bone destruction, and osteoporosis (Mun et al,
2020).
DUOX1 is part of the NADPH family and, like
NOX4, also produces H
2O
2. Duox1 forms heterodimers with dual oxidase maturation factor-1 (Duoxa1), which was recently shown to be involved in osteoclast differentiation and ROS production in bone (Cheon et al,
2020). Ryanodine receptors (RyRs) are calcium (Ca
2+) channels that are responsible for Ca
2+ release from the sarcoplasmic reticulum (Liu et al,
2017). In cancer-associated bone metastasis in a mouse model, upregulation of
Nox4 results in elevated oxidization of skeletal muscle proteins, including RyR1 (Waning et al,
2015). Also,
NOXO is involved in ROS formation, and shown to play role in angiogenesis (Brandes et al,
2016). Altogether, the variants affecting NOX4/ROS levels pathways are found in ∼20% of the PAM patients.
It is interesting to note that the
NOX4 intronic SNP rs11018628, previously linked to reduced bone density and elevated plasma markers for bone turnover (Goettsch et al,
2013) is found in the three PAM patients carrying NOX4 missense rare variants (
NOX4Y512C, NOX4V369F), but not in the PAM patient carrying the frameshift variant (
NOX4Y512IfsX20). We also found the SNP rs11018628 in the PsA961 carrier of
NOX4Y512C. Perhaps, there could be an additive or synergistic effect of these variants on
NOX4 expression at the transcriptional level, leading to increased generation of ROS. Our analysis of the ROS levels in patient PsA961 using electron paramagnetic resonance (EPR) showed significantly increased ROS levels compared to other individuals in PsA, psoriasis and healthy controls groups (Fig.
4A). The ROS levels were similar to the levels observed in the PAM12 patient. Unfortunately, we were not able to recruit any of the other patients for the measurement.
Several factors are involved in the regulation of
NOX4, including NF-κB, TGF-β, TNF-α, endoplasmic reticulum (ER) stress, hypoxia, and ischemia but the underlying mechanisms behind the regulation are not fully understood (Lou et al,
2018). Our results are in line with the previous observations showing that upregulation of NOX4 is linked to several pathogenic conditions, such as idiopathic pulmonary fibrosis (Amara et al,
2010), chronic obstructive pulmonary disease (Hollins et al,
2016) several cardiovascular conditions (Chen et al,
2012) and osteoporosis (Goettsch et al,
2013). In addition,
NOX4 is important for osteoclast differentiation. Osteoclasts are multinucleated cells derived from the monocyte-macrophage lineage, important for bone remodeling. They resorb the bone and its hyperactivated function is implicated in diseases such as osteoporosis, periprosthetic osteolysis, Paget’s bone disease, and rheumatoid arthritis (Bi et al,
2017). Here, we evaluated osteoclast differentiation and generation of ROS in one PAM patient (PAM12) and one PsA patient carrying a
NOX4 rare variant (PsA961 carrier of
NOX4Y512C) and found that the patients showed a similar pattern compared to a PsA individual without
NOX4 rare variants and two healthy controls.
In summary, we here present novel genetic findings, supported by several lines of functional evidence for the involvement of ROS in the etiology of PAM: (i) using stably transfected HEK293 cells, we show that the rare variants result in elevated
NOX4 transcript expression and ROS generation (Fig.
3A–C), (ii) measurement of ROS in patient PAM12 (a patient without identified
NOX4 mutations) and patient PsA961 (carrier of
NOX4Y512C) showed a significant increase of ROS compared to control, psoriasis and PsA samples by EPR (Fig.
4A,C), (iii) patient-derived cells from PAM12 showed increased osteoclast differentiation with increased ROS activity compared to cells from a healthy control (Fig.
5), (iv) patient-derived cells from PsA961 (carrier of
NOX4Y512C) showed increased osteoclast differentiation and increased H
2O
2 activity compared to cells from a PsA individual without
NOX4 rare variants and healthy control (Fig.
6), and finally (v) using a zebrafish model, we show in vivo that the generation of ROS is significantly enhanced by all three
NOX4 rare variants found in PAM patients (Fig.
7C,D).
A limitation of this study is the lack of access to fresh blood samples from the majority of PAM patients, needed for the measurement of ROS by EPR and for osteoclast differentiation from CD14+ monocytes. Our study would have benefited from exploring ROS levels in more patients. Nevertheless, we had the possibility of testing a couple of PAM patients and a PsA (carrier of NOX4Y512C) by EPR as a proof of concept that the generation of ROS is indeed affected in both patients. Another consideration is that the rare variants found in NOX4 are observed in just a few PAM patients (4 out of 61). Additional genetic analysis indicates potentially pathogenic variants in other genes found in PAM patients also affecting osteoclast differentiation and activity. Further functional validation experiments are required to test the pathogenicity of those variants.
Interestingly, the patients at risk of developing PAM may benefit from existing biological therapies applied in moderate and severe psoriasis which may reduce the generation of ROS. Another commonly used drug for treating psoriasis, methotrexate, inhibits osteoclast differentiation by inhibiting RANKL (Kanagawa et al,
2016). With the advent of effective therapies for psoriasis and psoriatic arthritis, PAM has become increasingly rare, still early diagnosis is important to avoid irreversible damage.
This study reveals a direct link to NOX4 and ROS/H2O2 production in PAM pathology and gives the first strong indication of where to search for specific disease identifiers in this destructive disease. Would early intervention with existing biologic treatments be sufficient or is precision therapy essential? The disease process can be rapid in PAM resulting in irreparable damage. Early identification of those at risk and initiation of effective therapy would constitute a game changer.
Methods
Human samples
In this study, genomic DNA was isolated from peripheral blood mononuclear cells (PBMCs) from the Nordic PAM patient’s cohort of 61 well-characterized patients previously described (Gudbjornsson et al,
2013; Laasonen et al,
2015; Laasonen et al,
2020; Lindqvist et al,
2017; Mistegard et al,
2021; Nikamo et al,
2020). The cohort consists of patients from Sweden (
n = 27), Denmark (
n = 21), Norway (
n = 10), and Iceland (
n = 3). The patients’ clinical and radiographic presentations follow the consensus from the Group for Research and Assessment of Psoriasis and Psoriatic Arthritis (GRAPPA) group (Ritchlin et al,
2009). In addition, for the genotyping of
NOX4 variants and for ROS measurement in blood samples we recruited psoriasis (
n = 1382) and psoriatic arthritis patients (
n = 492) and normal healthy controls (
n = 484). Caucasian origin was ascertained through ethnicity SNP genotyping (Giardina et al,
2008). Blood samples from one PAM patient (PAM12, male, 60 years old), one PsA patient carrying one of the rare variants found in PAM (PsA961, male, 39 years old), one PsA patient not carrying any of the variants investigated (PsA77, male, 43 years old) and age-gender-matched healthy controls were used for in vitro osteoclast differentiation.
DNA isolation
DNA from whole blood was purified by Gentra Puregene Blood Kit (158489, Qiagen, USA). Briefly, three volumes RBC Lysis Solution was added to blood and centrifuged at 4000×g for 10 min to pellet the white blood cells (WBS), supernatant was discarded. The WBS were lysed with one volume of cell lysis solution by vortexing. The cell lysates were treated with RNase A, and proteins were precipitated. The supernatant containing DNA was then extracted with isopropanol, followed by ethanol precipitation. After purification, the DNA was measured by Qubit.
Whole-genome and whole-exome sequence (WGS and WES)
We applied whole-genome sequencing (WGS) and whole-exome sequencing (WES) to 5 and 56 PAM patients, respectively. In addition, the parents of one PAM patient were sequenced by WGS. We applied Somalier, a tool to measure relatedness in cohorts (
https://github.com/brentp/somalier) to identify cryptic relatedness among all the samples (Fig.
EV3).
WGS was performed at the Science for Life Laboratory’s (SciLifeLab) national genomics infrastructure (NGI). The sequencing libraries were constructed using 1 µg of high-quality genomic DNA using the Illumina (San Diego, CA) TruSeq PCR-free kits (350 bp insert size) and sequenced on a single Illumina HiSeqX PE 2x150bp lane. WES was conducted at Uppsala’s SNP & SEQ technological platform. We utilized 300 ng of genomic DNA for WES; the DNA quality was determined using the FragmentAnalyzer, and the DNA concentration was determined using the Qubit/Quant-iT test. The sequencing libraries were constructed using the Twist Human Core Exome (Twist Bioscience), and the sequencing was carried out in a single S4 lane using the Illumina NovaSeq equipment and v1 sequencing chemicals (150 cycles paired-end). The data were processed, and the sequence reads were aligned to the human genome build GRCh37 Single nucleotide variants (SNVs) and insertions/deletions (INDELs) were called using the GATK v3.8. and v 4.1.4.1 pipeline and the called variants were annotated using VEP (v.91). The variants were loaded into the GEMINI database to query and filter the variants. Variants with a minor allele frequency (MAF) of <0.0001 were filtered for further investigation. The variants found were inspected manually with the integrative genomics viewer (IGV) tool in the other patients. The impact of variants was evaluated using the prediction tools SIFT, Polyphen-2, CADD and GERP + +. Selected variants were examined manually in the BAM files using Integrated Genomics Viewer.
Structural variants
Structural variants (SV) were analyzed using FindSV, a pipeline that performs SV detection using TIDDIT and CNVnator, as well as variant filtering and annotation using VEP and SVDB (Eisfeldt et al,
2017). Selected variants were visualized by the Integrative Genomics Viewer (IGV) tool.
SNP genotyping
Genotyping of three Single Nucleotide Polymorphisms (SNPs) within the
NOX4 gene (rs781430033, rs144215891, and rs765662279) was performed by using allele-specific Taqman MGB probes labeled with fluorescent dyes FAM and VIC (Applied Biosystems, Foster City, CA, USA), according to the manufacturer’s protocols. Allelic discrimination was made with the QuantStudioTM Real-Time PCR Software (Applied Biosystems). All three mutations had to be custom-made by using Custom TaqMan®Assay Design Tool (
https://www.thermofisher.com/order/custom-genomic-products/tools/cadt/); The success rate for genotyping exceeded 99% for all SNPs in the total sample set. We ran ten percent of the samples as duplicates to identify errors in genotyping and we could confirm assay accuracy of all three variations by WES and WGS of 61 PAM samples. The PCR procedure has been done using a total volume of 10 µl containing 15 ng of genomic DNA, 5 ul TaqMan® Universal PCR Master Mix (2×) and 0.5 µl TaqMan® genotyping assay mix (20×). Sequences of TapMan probes and primers are listed in Appendix Table
S3. Following an initial denaturation step at 50 °C for 2 min and 95 °C for 10 min starting all PCR procedures comprised 40 cycles of denaturation at 95 °C for 15 s, and primer annealing at 60 °C (55 °C for rs10065172) for 1 min and saved at 4 °C. We performed an endpoint plate read comprising the last step with an increasing temperature to a maximum of 60 °C (1.6 °C per second) and accompanying measurement of fluorescence intensity on a real‐time PCR on the QuantStudio 7 Flex Real-Time PCR System Instrument.
Sanger sequencing
Genomic DNA from peripheral blood samples was extracted by standard procedures. Sanger sequencing was performed by KIGene using the ABI 3730 PRISM® DNA Analyzer (Zianni et al,
2006). The primers used are shown in Appendix Table
S4.
Expression constructs and stable HEK293 cell lines
To test the effect of the variants in cells, we obtained plasmid -pcDNA3.1-hNox4 (#69352) from the Addgene repository. Primers with the alternate alleles for each SNP were designed using the “QuikChange Primer Design” (Agilent technologies) platform. Then,
NOX4Y512IfsX20,
NOX4V369F and
NOX4Y512C variants were introduced to the construct by using QuikChange XL Site-Directed Mutagenesis Kit (Agilent) according to manufacturer instructions with the primer pairs in Appendix Table
S5, which were transformed into Escherichia coli and identified by Sanger dideoxy sequencing (Appendix Table
S6). To obtain stable transfectants, we linearized plasmids with 1 µl BglII Enzyme (10 unit) and 5 µl 10× NEB buffer with the incubation at 37 °C for 15 min and 65 °C for 20 min. Human embryonic kidney 293 (HEK293) cells were kindly provided by Stefano Gastaldello (Karolinska Institutet, Stockholm, Sweden). HEK293 cells were cultured in Dulbecco’s modified Eagle medium (DMEM; Gibco) supplemented with 10% fetal bovine serum (FBS; Gibco) and 100 μg/ml Primocin (InvivoGen) at 37 °C in a humidified 5% CO
2 atmosphere, and periodically checked for mycoplasma contamination. HEK293 cells were transfected with pcDNA3.1-hNOX4 and three constructs carrier
NOX4 variants by the Lipofectamine 2000 Reagent (Thermo Fisher Scientific, USA), and G418 at 200 μg/ml was used as positive cell selection. Culture media containing the selection antibiotic was changed every 2–3 days until Geneticin®-resistant foci were identified. Next, we screened single-colony cells in the 96-well tissue culture plate and expanded the selected cells for future use.
Quantitative real-time PCR analysis
The extraction of total RNA from HEK293 stable cell lines was isolated by RNeasy mini kit (QIAGEN), and cDNA was reversed with Maxima First Strand cDNA Synthesis Kit (Thermo Fisher Scientific). Real-time quantitative PCR (RT-qPCR) was performed with SYBR® Green Master Mix according to the manufacturer’s protocol. Primer sequences are provided at Appendix Table
S2. The 2 − ΔΔCt method was utilized to achieve comparative quantification of the gene of interest between the two genotypes using actin as a reference gene.
Western blotting
HEK293 cells were harvested and lysed in RIPA lysis with 1X Halt™ Protease and Phosphatase Inhibitor Cocktail (Thermo Fisher Scientific). The concentration of total protein was determined using the BCA Protein Assay Kit (Thermo Fisher Scientific). In total, 10 μg or 20 μg of proteins were loaded in 10% SDS-PAGE gel and transferred to PVDF membranes. Membranes were blocked in Tris-buffered saline containing 5% skim milk for 1.5 h at room temperature. Then incubated at 4 °C overnight with recombinant anti-NADPH oxidase 4 antibody (1:2000, ab133303, abcam), and mouse anti-GAPDH monoclonal antibody was used for normalization (1:3000, 60004-1, Proteintech Group Inc). Immunoblots of protein bands were visualized with ECL (1705060, Clarity™ Western ECL Substrate, Biorad), and proteins were quantified with ImageJ software. The data are presented as mean ± SD of independent experiments performed in triplicate.
Osteoclast studies from patient-derived peripheral blood mononuclear cells (PBMCs)
To study the effect of the mutations on osteoclasts differentiation in cell culture, we obtained patient-derived mononuclear cells. PBMCs were isolated from whole blood using Ficoll-Paque density centrifugation. For positive selection of the osteoclast precursors, i.e., the CD14+ mononuclear cells, the EasySepTM Human CD14 positive Selection kit II was used according to the manufacturer’s instructions. Purified CD14+ cells were seeded in 24-well and 96-well plates containing Gibco DMEM supplemented with 10% FBS, 0.2% PrimocinTM and macrophage colony-stimulating factor (M-CSF) (20 ng/mL; R&D systems; USA) and receptor activator of nuclear factor kappa-B ligand (RANKL) (2 ng/mL; R&D systems; USA) to induce osteoclastogenesis. Every third day, media was refreshed. The osteoclasts were fixed and stained with tartrate-resistant acid phosphatase (TRAP)-positive cells based on a leukocyte acid phosphatase kit (387A; Sigma; USA) according to the manufacturer’s instructions. TRAP-stained cells containing three or more nuclei were defined as osteoclasts (Makitie et al,
2021).
Measurement of ROS by electron paramagnetic resonance (EPR)
The levels of ROS in the human blood and cultured cells were measured by EPR Spectroscopy (Zheng et al,
2022). Following ~36 h after transfection, cell culture media was removed, and the cells were rinsed twice with PBS. Seven hundred microliters of cyclichydroxylamine (CMH, 200 μM) in EPR-grade Krebs HEPES buffer supplemented with 25 μM Deferoxamine (DFX) and 5 μM diethyldithiocarbamate (DETC) were added to the cells and were incubated for 30 min at 37 °C. The cells are collected in the 1-mL syringes and frozen in liquid nitrogen prior to measurement. To analyze ROS levels in human blood, blood samples were incubated with CMH spin probe as mentioned above and ROS was measured using the EPR spectrometer (Noxygen, Elzach, Germany). ROS levels were converted to the concentration of CP radical using the standard curve method. Briefly, blood samples were combined with cyclichydroxylamine (CMH) spin probe and ROS was measured by a CP radical standard curve, using EPR spectrometer (Noxygen, Elzach, Germany).
Measurement of ROS production
2′,7′-dichlorofluorescein diacetate (4091-99-0, DCFH-DA, Sigma) was used as a sensitive and rapid identification of ROS in response to oxidative metabolism. First, we reconstituted in DMSO for stock, and then DCFH-DA S0033) was diluted with the serum-free cell culture medium. After washing osteoclasts with PBS twice at designated time points—day 8 and day 12, osteoclasts (at the density of 1 × 105cells/well) in a 96-well plate were incubated with 10 μM DCFH-DA in the incubator for 30 min and thereafter immediately analyzed using a fluorescence microscope (magnification ×10; EVOSTM FL, Invitrogen). The relative fluorescence intensity of DCFH-DA was analyzed using ImageJ.
Measurement of H2O2
Intracellular levels of H2O2 were detected utilizing the BioTracker Green H2O2 live-cell dye according to the manufacturer’s recommendation (SCT039, Sigma). For HEK293 stable cell lines, as well as osteoclasts derived from patients and controls, the culture medium was removed, and cells were rinsed twice with Hank’s Balanced Salt Solution (HBSS). Subsequently, a 1 mM concentration of the solution was added and the cells were then incubated for 20 min at 37 °C, 5% CO2. After removing the solution, cells underwent two additional rinses with HBSS. The visualization of living cells was conducted utilizing a fluorescence microscope (EVOSTM FL, Invitrogen).
Zebrafish assay for oxidative stress
The pcDNA3.1-h
NOX4,
NOX4Y512IfsX20,
NOX4V369F and
NOX4Y512C plasmids were linearized by restriction digestion with XhoI enzyme, and capped mRNA was transcribed in vitro using the mMESSAGE mMACHINE kit (Ambion, Thermo Fisher Scientific, Waltham, MA, USA). Zebrafish embryos (AB strain) at 1–2-cell stage were co-injected with
NOX4 mRNA and an antisense oligonucleotide used to knockdown endogenous
nox4 expression (
nox4 atg MO) (300 pg). For in vivo ROS detection, 30 h post fertilization, old embryos were exposed to 20 μM 2′,7′-dichlorofluorescein diacetate (DCFH-DA, Sigma-Aldrich) for 1 h at 28.5 °C in the dark followed by washing with embryo water a minimum of three times (Zhang et al,
2014). ROS production was visualized by mounting each embryo in a drop of low melting agarose (Hirsinger and Steventon,
2017) and imaged using a confocal microscope (Zeiss LSM700 coupled with a water-dripping lens). Importantly, every embryo was imaged using the same settings (e.g., laser intensity and optical slice thickness). The average fluorescence density (normalized to area) was analyzed using ImageJ. To this end, maximum intensity projections were produced and the total fluorescent intensity within the defined area was quantified. For each experimental group 10–16 embryos were quantified, and the experiment was repeated three times.
Statistics
Blinding was only applied for counting the number of TRAP-positive osteoclasts by two independent individuals. The assignment of TRAP figures was randomized to eliminate systematic errors, ensuring an unbiased analysis where each figure is represented in the evaluation. Images of cells located in the center of the wells were included in the analysis, while images of cells at the borders, where the distribution was uneven, were excluded.
Quantification and Statistical Analysis were performed using GraphPad Prism9. All experiments were performed with at least three independent biological replicates and expressed as the means ± standard deviation (SD). Student’s t test was applied to assess the statistical differences between experimental groups. The one-way analysis of variance (ANOVA) was used to assess the statistically significant differences between the means of three unrelated groups. Multiple comparisons were evaluated for all pairs of means by Two-way ANOVA with Tukey’s correction. P < 0.05 was considered significant (*P < 0.05, **P < 0.01, ***P < 0.001, ****P < 0.0001).
Study approval
The study was approved by ethical review boards at each institution and conducted according to the Declaration of Helsinki Principles. Written informed consent was obtained from all the participants in the study. Ethical permits:2007/1088-31/4. D.nr:00-448, 2008/4:5, Dnr 02-241, and Dnr 2022-04253-02. The Stockholm Ethical Board for Animal Experiments authorized standard operating procedures for all treatments involving zebrafish (Ethical approval: Dnr 14049-2019).
Graphics
Synopsis image was created with BioRender.com.