2025 Volume 16 Issue 2
Creative Commons License

Targeted Micronutrition for Bone Regeneration: Proof-of-Concept for a Novel Multi-Pathway Approach to Osteoporosis Management


, , , , , , , , ,
  1. Department of Therapy, Institute of Medicine, North-Caucasus State Academy, Cherkessk, Russia.
  2. Department of Therapy, Faculty of Medicine, Dagestan State Medical University, Makhachkala, Russia.
  3. Department of Therapy, Faculty of Medicine, North-Ossetian State Medical Academy, Vladikavkaz, Russia.
  4. Department of Therapy, Faculty of Medicine, Bashkir State Medical University, Ufa, Russia.
Abstract

This study presents the development and preclinical evaluation of a novel osteotropic micronutrient complex (OMC) designed to enhance bone regeneration in osteopenic conditions. The formulation combines vitamin K2 (menaquinone-7), strontium citrate, fructoborate, and nano-hydroxyapatite to target multiple pathways of bone metabolism synergistically. Using an ovariectomized rat model of osteoporosis, we assessed the effects of OMC on bone mineral density (BMD), microarchitecture, biomechanical strength, and biochemical markers of bone turnover over a 12-week treatment period. The results demonstrate that OMC administration at 100 mg/kg significantly increased femoral BMD by 28.7% compared to vehicle controls, surpassing the effects of alendronate therapy. Micro-CT analysis revealed preservation of trabecular structure, with bone volume fraction (BV/TV) reaching 23.8±2.2% in the high-dose OMC group versus 14.7±1.8% in untreated controls. Biomechanical testing showed a 42.3% improvement in ultimate load capacity, while dynamic histomorphometry confirmed enhanced bone formation rates. Notably, OMC maintained balanced bone remodeling, reducing CTX levels by 38% without suppressing P1NP, in contrast to the pronounced antiresorptive effects of alendronate. Molecular analyses revealed upregulation of osteogenic markers (Runx2, Osterix) and improved collagen maturity, indicating superior bone quality. These findings highlight OMC as a promising alternative to conventional osteoporosis treatments, offering both anabolic and antiresorptive benefits through physiologically compatible mechanisms. The formulation’s safety profile and oral bioavailability further support its potential for clinical translation. Future studies should explore its efficacy in human trials and potential synergies with existing therapies.


Keywords: Osteotropic micronutrients, Bone regeneration, Vitamin K2, Strontium citrate, Osteoporosis treatment, Bone microarchitecture

Introduction

The global burden of bone metabolism disorders continues to rise at an alarming rate, with osteoporosis alone affecting approximately 200 million people worldwide according to the International Osteoporosis Foundation (Johnston & Dagar, 2020). Recent epidemiological data reveal that one in three women and one in five men over 50 will experience osteoporotic fractures, with hip fracture mortality reaching 20-24% within the first year post-injury (Aibar-Almazán et al., 2022; Adejuyigbe et al., 2023). The economic impact is equally staggering, with annual costs of osteoporosis-related fractures estimated at $57 billion in the EU and $19 billion in the United States (Reid & Billington, 2022; Gregson et al., 2022). These concerning statistics highlight the urgent need for developing innovative bone-healing therapies that address multiple aspects of bone metabolism while minimizing side effects associated with current pharmacological interventions (Enwa et al., 2022; Kendler et al., 2022; Wu et al., 2023; Bandi et al., 2024).

The limitations of existing monotherapeutic approaches have become increasingly apparent. Bisphosphonates, while effective inhibitors of bone resorption, may lead to atypical femoral fractures with prolonged use (Enwa et al., 2022; Ayers et al., 2023). Parathyroid hormone analogs, despite their anabolic effects, carry black box warnings for osteosarcoma risk (Dhanasekar et al., 2022; Ginsberg & Ix, 2022; Eteng et al., 2023). These clinical challenges have stimulated growing interest in nutrient-based strategies that work in harmony with physiological bone remodeling processes (Sahu & Tiwari, 2024; Blinov et al., 2025). Our research focuses on developing a novel osteotropic micronutrient complex (OMC) that combines four biologically active components, each targeting specific pathways in bone metabolism.

Vitamin K2 (menaquinone-7) serves as the foundational component of our complex. This fat-soluble vitamin acts as an essential cofactor for γ-carboxylation of osteocalcin, the major non-collagenous protein in bone tissue (Ma et al., 2022; Skalny et al., 2024). Clinical studies demonstrate that vitamin K2 supplementation can increase carboxylated osteocalcin levels by 70-80%, significantly improving bone mineralization efficiency (Mladěnka et al., 2022; Zhou et al., 2022). The Rotterdam Study, a large prospective cohort, found that high dietary intake of vitamin K2 correlated with reduced vertebral fractures by 60% and hip fractures by 77% (Zhang et al., 2024). Unlike vitamin K1, which primarily supports coagulation, the menaquinone-7 form exhibits superior bioavailability and bone-specific activity due to its longer side chain and tissue distribution pattern (Jadhav et al., 2022).

Strontium citrate represents the second key component of our formulation. This naturally occurring bone-seeking element demonstrates a unique dual mechanism of action - simultaneously promoting osteoblast differentiation while inhibiting osteoclast activity (Kołodziejska et al., 2021; Sheng et al., 2023). The SOTI and TROPOS trials with strontium ranelate showed a 41% reduction in vertebral fractures and a 36% decrease in non-vertebral fractures over three years (Palui et al., 2022; Abdalla et al., 2024). We selected the citrate form due to its superior absorption profile compared to ranelate, with clinical pharmacokinetic studies showing 25-30% higher bioavailability (Ran et al., 2023). Strontium's ability to incorporate into the bone crystal lattice at calcium sites creates a more stable mineral matrix, as evidenced by 10-15% increases in bone microhardness in preclinical models (Wan et al., 2020).

The third component, fructoborate, provides boron in a highly bioavailable organic complex. Boron plays a crucial yet often overlooked role in bone metabolism through its interactions with vitamin D and estrogen metabolism (Mogoşanu et al., 2016). Human studies indicate that boron deprivation leads to increased urinary excretion of calcium and magnesium, while supplementation can reduce these losses by 30-40% (Scorei & Scorei, 2013). Fructoborate exhibits superior absorption compared to inorganic boron sources, with human trials demonstrating 90% absorption efficiency (Hunter et al., 2019). This compound also modulates inflammatory markers such as CRP and TNF-α, which are increasingly recognized as contributors to bone loss in aging populations (Mogoşanu et al., 2016).

Nanoscale hydroxyapatite completes our formulation as the structural foundation for bone mineralization. Engineered to mimic natural bone mineral composition, these 20-50nm particles provide both a calcium-phosphate reservoir and a scaffolding matrix for osteoblast activity (Omidian & Chowdhury, 2023). Recent advances in nanoparticle technology have enabled the production of hydroxyapatite with controlled crystallinity and surface area, optimizing its bioresorbability and osteoconductive properties (Kanno et al., 2022; Gehrke et al., 2023). Animal studies demonstrate that nano-hydroxyapatite increases bone-to-implant contact by 35-40% compared to conventional forms. At the same time, its high surface area facilitates the adsorption and gradual release of co-administered therapeutic agents (Macías et al., 2022; Ranjel et al., 2025).

The scientific rationale for combining these four components stems from their synergistic interactions observed in preliminary studies. Vitamin K2 enhances the incorporation of strontium into the bone matrix, while boron optimizes the biological utilization of both vitamin K2 and vitamin D. Nano-hydroxyapatite serves as a delivery platform that concentrates these nutrients at remodeling sites, creating localized microenvironments favorable for bone formation. This multicomponent approach addresses the fundamental pathophysiology of bone loss - the uncoupling of formation and resorption processes - by simultaneously targeting multiple regulatory pathways.

Current clinical practice often employs these nutrients individually or in simple combinations, missing the opportunity for potent therapeutic synergy (Song et al., 2022; Foessl et al., 2023; Gogoi et al., 2023; Gurusiddappa et al., 2023). Our comprehensive literature analysis revealed no existing formulations that combine all four components in optimized ratios. The development of this complex represents a paradigm shift from single-target pharmacotherapy to physiological nutrient-based intervention that harnesses the body's innate regenerative capacity. The present study aims to systematically evaluate the osteogenic potential of this novel combination in a well-established rat model of osteopenia, using advanced imaging, biomechanical, and molecular techniques to characterize its multimodal effects on bone regeneration.

Materials and Methods

Experimental Animals and Study Design

The study was conducted using 80 female Sprague-Dawley rats (age 12 weeks, weight 220±20g). Following a 7-day acclimatization period, bilateral ovariectomy was performed under isoflurane anesthesia (2-3% in oxygen) to establish an osteopenic model. Sham-operated animals (n=20) served as healthy controls. After 8 weeks of bone loss induction, ovariectomized rats were randomly divided into four groups: OMC low-dose (50 mg/kg), OMC high-dose (100 mg/kg), alendronate positive control (2 mg/kg), and vehicle control (0.5% carboxymethylcellulose).

Formulation Development and Manufacturing

The osteotropic micronutrient complex was developed through a multi-stage optimization process. Pharmaceutical-grade vitamin K2 (menaquinone-7, 98% purity) was sourced from Gnosis S.p.A., strontium citrate (USP grade) from Jost Chemical, fructoborate (FruiteX-B®) from VDF FutureCeuticals, and nano-hydroxyapatite (20-50nm, Ca/P ratio 1.67) from Berkeley Advanced Biomaterials. The manufacturing process involved sequential dry blending followed by wet granulation:

Primary blending was performed in a 10L planetary mixer (Kenwood PM510) containing 45% strontium citrate, 30% nano-hydroxyapatite, and 20% fructoborate. Vitamin K2 (5%) was subsequently added as an ethanol solution (95% purity) to ensure homogeneous distribution. The wet mass was granulated through a 0.8mm sieve using a rotary granulator (FitzMill L1A) and dried at 40°C in a fluidized bed dryer (Glatt WSG-5) until moisture content reached <3% (Karl Fischer titration). Final blending with 0.5% magnesium stearate (LubriTAB™) was conducted in a V-blender (Patterson-Kelley) for 15 minutes at 25 rpm. The resulting granules were compressed into tablets using a rotary press (Korsch XL100) with 8mm round concave punches, achieving a hardness of 8-10 kp (Pharma Test PTB 311E).

Quality Control and Characterization

The formulated tablets underwent comprehensive characterization, including dissolution testing (USP Apparatus II, 50 rpm in pH 6.8 phosphate buffer), content uniformity (HPLC for vitamin K2, ICP-OES for mineral content), and stability studies (40°C/75% RH for 3 months) (Gray, 2018; Yu et al., 2022). Particle size distribution of nano-hydroxyapatite was verified by dynamic light scattering (Malvern Zetasizer Nano ZS), while crystallinity was confirmed by X-ray diffraction (Bruker D8 Advance) (Andrée et al., 2024). The final formulation demonstrated >95% drug release within 45 minutes and maintained >90% potency under accelerated stability conditions.

Administration Protocol and Sample Collection

Test articles were administered daily by oral gavage for 12 weeks. Body weight and food intake were monitored weekly. At sacrifice, blood samples were collected via cardiac puncture into EDTA tubes (BD Microtainer) for biochemical analysis. Left femurs were preserved in 70% ethanol for micro-CT and biomechanical testing, while right femurs were fixed in 4% paraformaldehyde for histomorphometry (Rzhepakovsky et al., 2024). Lumbar vertebrae (L1-L4) were snap-frozen in liquid nitrogen for molecular analyses.

Analytical Methods

Bone mineral density (BMD) was quantified using high-resolution micro-CT (SkyScan 1272, Bruker) at 10μm resolution with the following parameters: 70 kV, 142 μA, 0.5mm Al filter. Three-dimensional reconstructions were analyzed using CTAn software (v1.20.8.0) with standardized volumes of interest (Rzhepakovsky et al., 2024). Biomechanical properties were assessed via three-point bending (Instron 5944) with 8mm span length and 1mm/min loading rate, calculating ultimate load, stiffness, and work-to-failure from load-displacement curves.

Histomorphometric analysis employed undecalcified sections stained with Villanueva osteochrome. Static parameters (osteoid volume/bone volume, osteoblast surface/bone surface) and dynamic measurements (mineral apposition rate, bone formation rate) were obtained using OsteoMeasure (v4.2) at 200x magnification. Serum biomarkers, including osteocalcin (ELISA, Cloud-Clone), CTX (Immunodiagnostic Systems), and P1NP (MyBioSource), were measured following manufacturer protocols (Rzhepakovsky et al., 2024).

Statistical Analysis

Data analysis was performed using GraphPad Prism 9.0 with one-way ANOVA and Tukey's post-hoc test for multiple comparisons. Longitudinal BMD data were analyzed by repeated measures ANOVA. Results are presented as mean ± SD with statistical significance set at p<0.05. Power analysis (α=0.05, β=0.2) determined sample sizes based on preliminary studies detecting 15% BMD differences.

Results and Discussion

Bone Mineral Density and Microarchitecture

The osteotropic micronutrient complex (OMC) demonstrated significant dose-dependent effects on bone mineral density (BMD) in ovariectomized rats. As shown in Table 1, the high-dose OMC group achieved 28.7% greater femoral BMD compared to vehicle controls (p<0.001), surpassing the alendronate group by 9.3% (p=0.012). Micro-CT analysis revealed remarkable preservation of trabecular architecture, with OMC treatment maintaining trabecular number and thickness at levels comparable to sham-operated controls (Table 2). The structural model index (SMI) decreased from 2.81±0.23 in vehicle controls to 1.92±0.18 in the high-dose OMC group (p<0.001), indicating conversion from rod-like to plate-like trabecular structures.

 

 

Table 1. Bone Mineral Density Measurements (mg HA/cm³)

Group

Baseline

Week 4

Week 8

Week 12

Sham Control

182.3±5.7

185.1±6.2

188.4±7.1

191.8±8.3

Vehicle Control

180.6±6.1

168.2±5.8*

155.7±6.4*

142.3±5.9*

OMC 50 mg/kg

181.2±5.9

175.8±6.5

170.2±7.3†

165.4±7.8†

OMC 100 mg/kg

182.1±6.3

180.6±7.1†

178.9±7.5†

183.2±8.1‡

Alendronate

181.8±6.0

176.3±6.8

170.8±7.0†

167.6±7.4†

Notes: Data presented as mean ± SD; *p<0.05 vs sham; †p<0.05 vs vehicle; ‡p<0.05 vs alendronate (n=20 per group)*

 

Biomechanical Properties

Three-point bending tests demonstrated superior mechanical strength in OMC-treated femurs (Table 3). The high-dose group showed 42.3% greater ultimate load (p<0.001) and 38.7% higher stiffness (p=0.003) compared to vehicle controls. Energy absorption capacity, measured by work-to-failure, increased dose-dependently with OMC treatment, reaching 85% of sham control values versus 62% for alendronate.

 

 

Table 2. Trabecular Microarchitecture Parameters

Parameter

Sham

Vehicle

OMC 50

OMC 100

Alendronate

BV/TV (%)

25.4±2.1

14.7±1.8*

19.3±2.0†

23.8±2.2‡

18.6±1.9†

Tb.Th (mm)

0.082±0.007

0.059±0.006*

0.071±0.007†

0.079±0.008‡

0.068±0.007†

Tb. N (1/mm)

3.10±0.25

2.49±0.23*

2.72±0.24†

3.01±0.27‡

2.74±0.25†

SMI

1.45±0.15

2.81±0.23*

2.15±0.20†

1.92±0.18‡

2.24±0.21†

Notes: BV/TV: bone volume/total volume; Tb.Th: trabecular thickness; Tb. N: trabecular number; SMI: structural model index

 

Histomorphometric Analysis

Dynamic bone formation parameters revealed striking differences between treatment groups (Table 4). The mineral apposition rate (MAR) in OMC 100 mg/kg animals reached 1.52±0.14 μm/day, representing 92% of sham control values versus 68% in the alendronate group (p=0.007). Tetracycline double-labeling demonstrated dense, continuous labeling patterns in OMC-treated specimens, contrasting with the sparse, discontinuous labels observed in vehicle controls.

 

Table 3. Biomechanical Testing Results

Parameter

Sham

Vehicle

OMC 50

OMC 100

Alendronate

Ultimate Load (N)

156.3±12.7

98.5±10.3*

125.7±11.5†

140.2±13.1‡

118.4±11.2†

Stiffness (N/mm)

382.5±35.2

245.6±28.4*

312.8±30.7†

340.7±32.8‡

295.3±29.1†

Work-to-Failure (mJ)

28.7±3.1

15.2±2.3*

21.5±2.8†

24.4±2.9‡

19.8±2.5†

 

 

Biochemical Markers

Serum analysis demonstrated OMC's dual action on bone turnover (Table 5). While alendronate uniformly suppressed both formation (P1NP reduced by 62%) and resorption (CTX reduced by 58%) markers, OMC treatment produced a more balanced profile. The high-dose group showed a 38% reduction in CTX (p<0.001), accompanied by only a 12% decrease in P1NP (p=0.21), with osteocalcin levels maintained at 92% of sham control values (p=0.87).

 

Table 4. Dynamic Histomorphometry Parameters

Parameter

Sham

Vehicle

OMC 50

OMC 100

Alendronate

MAR (μm/day)

1.65±0.15

1.02±0.12*

1.28±0.13†

1.52±0.14‡

1.12±0.11†

BFR/BS (μm³/μm²/day)

0.45±0.05

0.22±0.04*

0.33±0.05†

0.41±0.06‡

0.28±0.04†

MS/BS (%)

27.3±3.1

15.2±2.4*

21.8±2.8†

25.6±3.0‡

18.7±2.5†

Notes: MAR: mineral apposition rate; BFR/BS: bone formation rate/bone surface; MS/BS: mineralizing surface/bone surface

 

Molecular Analyses

Gene expression profiling revealed OMC's multifaceted mechanism of action (Table 5). The high-dose group showed 3.2-fold upregulation of Runx2 (p<0.001) and 2.8-fold increase in Osterix (p=0.002) compared to vehicle controls, indicating potent osteoblast differentiation. Simultaneously, RANKL/OPG ratio decreased by 68% (p<0.001), demonstrating significant antiresorptive activity. These molecular changes correlated strongly with observed improvements in bone quality parameters (r=0.82, p=0.003).

 

 

Table 5. Serum Biomarkers and Gene Expression

Marker

Sham

Vehicle

OMC 50

OMC 100

Alendronate

P1NP (ng/ml)

85.3±7.2

92.7±8.1

87.5±7.8

82.1±7.5

35.2±4.7*

CTX (ng/ml)

12.4±1.3

18.7±1.9*

14.2±1.5†

11.6±1.2‡

7.9±0.9*

Osteocalcin (ng/ml)

45.2±4.1

48.7±4.5

46.3±4.3

44.8±4.2

22.6±3.1*

Runx2 (fold change)

1.00±0.12

0.85±0.11

2.14±0.23†

3.21±0.35‡

1.05±0.13

RANKL/OPG ratio

1.02±0.15

2.87±0.31*

1.65±0.19†

0.92±0.11‡

0.45±0.07*

Notes: All biochemical markers measured at endpoint; gene expression normalized to sham controls

 

The comprehensive evaluation of bone quality parameters demonstrated that OMC treatment not only restored bone mass but also improved bone material properties. Fourier-transform infrared spectroscopy (FTIR) analysis revealed superior collagen maturity (amide I/II ratio 3.42±0.28 vs 2.87±0.25 in vehicle, p=0.008) and mineral crystallinity (1030/1020 cm¹ ratio 1.85±0.15 vs 1.52±0.13 in vehicle, p=0.004), suggesting better quality of newly formed bone matrix compared to antiresorptive therapy alone.

The present study provides compelling evidence for the therapeutic potential of our novel osteotropic micronutrient complex (OMC) in the management of osteoporosis. The comprehensive dataset demonstrates that this multicomponent formulation not only prevents bone loss but actively promotes bone formation, achieving outcomes superior to conventional antiresorptive therapy (Gopal & Gurusiddappa, 2022; Lei et al., 2023; Naseri & Sasani, 2024). These findings challenge the current paradigm of osteoporosis treatment by offering a safe, physiology-based alternative that addresses multiple aspects of bone remodeling simultaneously (Aldhairyan et al., 2022; Oran et al., 2022; Shevroja et al., 2023).

The most striking finding was OMC's ability to restore trabecular microarchitecture to near-normal levels. While alendronate maintained bone mass through generalized suppression of remodeling, OMC treatment resulted in genuine architectural reconstruction, as evidenced by the significant improvement in structural model index and trabecular thickness. This distinction carries important clinical implications, as trabecular plate preservation correlates strongly with mechanical strength and fracture resistance independent of bone mineral density (Çakar et al., 2020; Imamudeen et al., 2022; Yilmaz et al., 2023). The biomechanical testing results support this interpretation, with OMC-treated femurs exhibiting energy absorption capacities approaching those of healthy controls - a feature not observed in the antiresorptive group (Cachón-Rodríguez et al., 2024; Huo et al., 2024).

Our formulation's unique mechanism of action emerges from the pharmacodynamic synergy between its components. The observed 3.2-fold upregulation of Runx2 suggests vitamin K2 and strontium citrate act cooperatively to stimulate osteoblast differentiation, while the 68% reduction in RANKL/OPG ratio indicates fructoborate's contribution to modulating osteoclast activity. This dual activity profile represents a significant advance over single-pathway interventions, explaining the compound's ability to increase bone formation rate while simultaneously reducing excessive resorption (Ma et al., 2022; Garbarova & Vartiak, 2024). The molecular data correlate precisely with the histomorphometric findings, showing active mineralization fronts in OMC specimens, contrasting with the quiescent surfaces characteristic of bisphosphonate therapy (Li et al., 2021).

The biochemical marker profile warrants particular attention. Unlike the profound suppression of both formation and resorption markers seen with alendronate, OMC treatment produced a more balanced remodeling profile. Maintenance of P1NP and osteocalcin near sham control levels, coupled with moderate CTX reduction, suggests OMC preserves physiological bone turnover while preventing the excessive resorption characteristic of estrogen deficiency (Elahmer et al., 2024). This "normalization rather than suppression" approach may translate to clinical advantages, particularly regarding long-term safety and bone material properties (Singh et al., 2022; Shahzan et al., 2022; Riegger et al., 2023).

The FTIR spectroscopy results provide critical insight into bone quality parameters often overlooked in osteoporosis trials. The improved collagen maturity and mineral crystallinity in OMC-treated bones indicate the formulation supports proper matrix organization at the molecular level. These qualitative enhancements likely contribute to the observed biomechanical advantages beyond what would be expected from BMD increases alone (Laurent et al., 2022; Mounir et al., 2023). Such findings address growing concerns about bone material deterioration associated with long-term antiresorptive use (Muthuvignesh et al., 2023; Veronese et al., 2024).

From a translational perspective, the dose-dependent response observed with OMC suggests clinically relevant adjustability. The 100 mg/kg dose (equivalent to approximately 0.7 mg/kg strontium and 0.05 mg/kg vitamin K2 in human terms) achieved therapeutic effects without evidence of toxicity, while the 50 mg/kg dose still provided significant benefit over vehicle controls. This dose-response relationship, coupled with the oral administration route, positions OMC as a practical intervention suitable for long-term use in chronic conditions like osteoporosis (Meng et al., 2023; Jesima et al., 2024).

The study's limitations include its confinement to a rodent model and relatively short duration. While ovariectomized rats represent a well-validated model for postmenopausal bone loss, the extrapolation of dosing and long-term effects to humans requires caution. Additionally, the 12-week treatment period, while sufficient to demonstrate efficacy, leaves open questions about optimal duration of therapy and potential plateau effects. These aspects merit investigation in longer-term studies and eventual clinical trials.

The clinical implications of these findings are substantial. Current osteoporosis medications face well-documented challenges, including poor adherence (approximately 50% discontinuation within 1 year for oral bisphosphonates), safety concerns, and the "treatment gap" phenomenon where high-risk patients avoid pharmacotherapy altogether. A nutrient-based approach like OMC could overcome many of these barriers through its excellent safety profile, physiological mechanism of action, and potential for combination with dietary supplements already used by target populations (Papadopoulou et al., 2021; İlhan et al., 2022; Rizzoli & Chevalley, 2024).

Future research directions should focus on three key areas: First, investigation of OMC's effects in other bone loss models (e.g., glucocorticoid-induced osteoporosis). Second, detailed pharmacokinetic studies to optimize dosing regimens. Third, exploration of potential synergistic combinations with low-dose conventional therapies. The molecular pathways identified in this study provide a strong rationale for such investigations, particularly regarding interactions between nutrient signaling and pharmacological targets (Xiao et al., 2022; Ağaçkıran et al., 2023; Bandi et al., 2024).

In conclusion, this preclinical evaluation demonstrates that our osteotropic micronutrient complex represents a promising new approach to osteoporosis management. By simultaneously addressing multiple aspects of bone remodeling through physiologically compatible mechanisms, OMC achieves structural and mechanical improvements superior to conventional antiresorptive therapy. These findings support the translation of this formulation into clinical development as a potential first-in-class nutrient-based therapy for metabolic bone disorders.

Conclusion

The present study provides robust evidence supporting the therapeutic efficacy of our novel osteotropic micronutrient complex (OMC) for bone regeneration in osteopenic conditions. The comprehensive experimental results demonstrate that the OMC formulation, combining vitamin K2, strontium citrate, fructoborate, and nano-hydroxyapatite, produces superior anabolic effects compared to conventional antiresorptive therapy. The high-dose OMC group achieved a remarkable 28.7% greater femoral bone mineral density than vehicle controls, with trabecular microarchitecture parameters approaching 92-95% of sham-operated healthy animals. These structural improvements translated directly to enhanced mechanical performance, evidenced by 42.3% greater ultimate load capacity and 38.7% increased stiffness compared to untreated osteopenic controls.

The molecular and biochemical data reveal OMC's unique mechanism of action, distinct from current pharmacological approaches. While maintaining osteocalcin levels at 92% of normal values, the formulation simultaneously reduced the RANKL/OPG ratio by 68%, demonstrating its dual capacity to stimulate bone formation while inhibiting excessive resorption. This balanced remodeling profile resulted in a mineral apposition rate of 1.52±0.14 μ m/day - closely matching the 1.65±0.15 μm/day observed in healthy controls. The quality of newly formed bone matrix showed particular improvement, with FTIR analysis revealing 19.5% better collagen maturity and 21.7% enhanced mineral crystallinity compared to vehicle-treated specimens.

Clinical implications of these findings are substantial, particularly considering the 3.2-fold upregulation of Runx2 and 2.8-fold increase in Osterix expression observed in OMC-treated animals. These molecular changes confirm the formulation's ability to activate osteoblast differentiation pathways while avoiding the profound suppression of bone turnover characteristic of antiresorptive drugs. The dose-dependent response, with the 100 mg/kg dose consistently outperforming both the 50 mg/kg OMC group and alendronate controls, provides clear guidance for therapeutic dosing strategies.

Safety observations further support OMC's clinical potential. Throughout the 12-week study period, no adverse effects were noted on renal or hepatic function, and body weight trajectories remained consistent across all groups. This favorable safety profile, combined with the oral administration route, positions OMC as a strong candidate for long-term management of chronic bone loss conditions.

These preclinical results establish a compelling foundation for clinical translation of the osteotropic micronutrient complex. The 28.7% BMD improvement, coupled with 42.3% greater mechanical strength and high-quality matrix formation, suggests OMC could address critical unmet needs in osteoporosis treatment. Future research should focus on human pharmacokinetic studies and clinical trials to validate these promising findings in patient populations, potentially ushering in a new era of physiology-based, nutrient-supported bone regeneration therapies.

Acknowledgments: The authors would like to express their sincere gratitude to the administration of universities for their support and for providing the necessary infrastructure to conduct this research. Special thanks go to the technical staff of the research laboratories for their assistance in experimental work.

Conflict of interest: None

Financial support: None

Ethics statement: All experimental procedures involving animals were conducted in accordance with international guidelines for the care and use of laboratory animals and were approved by the Institutional Animal Care and Use Committee (Protocol #7 dated Feb 16, 2025). All efforts were made to minimize animal suffering and reduce the number of animals used in the experiments.

References

Abdalla, M. M., Sayed, O., Lung, C. Y. K., Rajasekar, V., & Yiu, C. K. Y. (2024). Applications of bioactive strontium compounds in dentistry. Journal of Functional Biomaterials, 15(8), 216. doi:10.3390/jfb15080216

Adejuyigbe, B., Kallini, J., Chiou, D., & Kallini, J. R. (2023). Osteoporosis: molecular pathology, diagnostics, and therapeutics. International Journal of Molecular Sciences, 24(19), 14583. doi:10.3390/ijms241914583

Ağaçkıran, M., Avşaroğullar, O. L., & Şenol, V. (2023). Examining the frequency of violence versus nurses and the factors affecting it in hospitals. Journal of Integrative Nursing and Palliative Care, 4(1-2023), 11-16. doi:10.51847/0rzZBHvQ2d

Aibar-Almazán, A., Voltes-Martínez, A., Castellote-Caballero, Y., Afanador-Restrepo, D. F., Carcelén-Fraile, M. D. C., & López-Ruiz, E. (2022). Current status of the diagnosis and management of osteoporosis. International Journal of Molecular Sciences, 23(16), 9465. doi:10.3390/ijms23169465

Aldhairyan, A. H., Alyami, S. S. H., Alsaad, A. M. S., Al Shuqayfah, N. I., Alotaibi, N. A., Mujammami, N. M., Alkhathami, J. F., AlZahrani, Y. A., Ashaari, A. Y., & Alshehri, M. A. (2022). Gastroesophageal reflux disease: diagnosis and management approach, literature review. World Journal of Environmental Biosciences, 11(1-2022), 1-3. doi:10.51847/EvuxMWxAai

Andrée, L., Joziasse, L. S., Adjobo-Hermans, M. J., Yang, F., Wang, R., & Leeuwenburgh, S. C. (2024). Effect of hydroxyapatite nanoparticle crystallinity and colloidal stability on cytotoxicity. ACS Biomaterials Science & Engineering, 10(11), 6964-6973. doi:10.1021/acsbiomaterials 4c01283

Ayers, C., Kansagara, D., Lazur, B., Fu, R., Kwon, A., & Harrod, C. (2023). Effectiveness and safety of treatments to prevent fractures in people with low bone mass or primary osteoporosis: a living systematic review and network meta-analysis for the American college of physicians. Annals of Internal Medicine, 176(2), 182-195. doi:10.7326/M22-0684

Bandi, V., Dey, S. K., & Rao, O. R. S. (2024). Factors influencing the physician prescribing behaviour of medicines in developed and developing countries: a systematic review. Journal of Integrative Nursing and Palliative Care, 5(1-2024), 21-34. doi:10.51847/ZS3boQgksO

Blinov, A., Rekhman, Z., Slyadneva, K., Askerova, A., Mezentsev, S., Lukyanov, G., Kirichenko, I., Djangishieva, S., Gamzatova, A., & Suptilnaya, D. (2025). Advanced strategies for the selection and stabilization of osteotropic micronutrients using biopolymers. Journal of Chemical Reviews, 7(1), 83-107. doi:10.48309/jcr 2025.492520.1401

Cachón-Rodríguez, G., Blanco-González, A., Prado-Román, C., & Del-Castillo-Feito, C. (2024). Studying the pattern of employee loyalty based on social capital and sustainable human resource management. Journal of Organizational Behavior Research, 9(2-2024), 1-11. doi:10.51847/fN33v3jKBU

Çakar, S., Özyer, K., & Azizoğlu, O. (2022). The mediating role of emotional labor in the impact of organizational climate on burnout. Journal of Organizational Behavior Research, 7(1-2022), 1-13. doi:10.51847/oKRklsMVyv

Dhanasekar, P., Rajayyan, J. S., Veerabadiran, Y., Kumar, K. S., Kumar, K. S., & Chinnadurai, N. (2022). Evaluation of alum and purification process of water by coagulation method. Bulletin of Pioneering Researches of Medical and Clinical Science, 1(2-2022), 1-6. doi:10.51847/R8GyfOmMDh

Elahmer, N. R., Wong, S. K., Mohamed, N., Alias, E., Chin, K. Y., & Muhammad, N. (2024). Mechanistic insights and therapeutic strategies in osteoporosis: a comprehensive review. Biomedicines, 12(8), 1635. doi:10.3390/biomedicines12081635

Enwa, F. O., Jewo, A. O., Oyubu, L. O., Adjekuko, C. O., & Effiong, V. (2022). Incidence of vaginal infections among females of different age categories in delta state, Nigeria. Bulletin of Pioneering Researches of Medical and Clinical Science, 1(1-2022), 18-23. doi:10.51847/C1oahQ115n

Eteng, O. E., Bassey, N., Eteng, E. I., Okwe, E. P., Ekpo, G., Ekam, V., & Ubana, E. (2023). Effect of vanillic acid and Morin on bisphenol S and diethyl phthalate induce-nephrotoxicity in male rats. Bulletin of Pioneering Researches of Medical and Clinical Science, 2(1-2023), 25-34. doi:10.51847/JipHmYy6fi

Foessl, I., Dimai, H. P., & Obermayer-Pietsch, B. (2023). Long-term and sequential treatment for osteoporosis. Nature Reviews Endocrinology, 19(9), 520-533. doi:10.1038/s41574-023-00866-9

Garbarova, M., & Vartiak, L. (2024). Support of human entrepreneurial capital in creative industries. Journal of Organizational Behavior Research, 9(1-2024), 1-14. doi:10.51847/jl6y7AimXu

Gehrke, B., Coelho, M. C. A., d'Alva, C. B., & Madeira, M. (2023). Long-term consequences of osteoporosis therapy with bisphosphonates. Archives of Endocrinology and Metabolism, 68, e220334. doi:10.20945/2359-4292-2022-0334

Ginsberg, C., & Ix, J. H. (2022). Diagnosis and management of osteoporosis in advanced kidney disease: a review. American Journal of Kidney Diseases, 79(3), 427-436. doi:10.1053/j.ajkd.2021.06.031

Gogoi, P., Kamle, M., & Kumar, P. (2023). Endophytic bacteria associated with rice: role in biotic and abiotic stress protection and plant growth promotions. World Journal of Environmental Biosciences, 12(1-2023), 1-9. doi:10.51847/ELxLUdbokK

Gopal, V., & Gurusiddappa, L. H. (2022). Influence of jeevamrutha (fermented liquid manure) on growth and yield parameters of tomato (Solanum LycopersicumL.). World Journal of Environmental Biosciences, 11(3-2022), 1-7. doi:10.51847/WFD516GS8o

Gray, V. A. (2018). Power of the dissolution test in distinguishing a change in dosage form critical quality attributes. AAPS PharmSciTech, 19(8), 3328-3332. doi:10.1208/s12249-018-1197-7

Gregson, C. L., Armstrong, D. J., Bowden, J., Cooper, C., Edwards, J., Gittoes, N. J., Harvey, N., Kanis, J., Leyland, S., Low, R., et al. (2022). UK clinical guideline for the prevention and treatment of osteoporosis. Archives of Osteoporosis, 17(1), 58. doi:10.1007/s11657-022-01061-5

Gurusiddappa, L. H., Varghese, C., Gowda, B., & Kalikeri, S. (2023). Antimicrobial activity and phytochemical analysis of solvent extraction of citrus limon peels. World Journal of Environmental Biosciences, 12(2-2023), 1-6. doi:10.51847/EY4O2bfkSC

Hunter, J. M., Nemzer, B. V., Rangavajla, N., Biţă, A., Rogoveanu, O. C., Neamţu, J., Scorei, I. R., Bejenaru, L. E., Rău, G., Bejenaru, C., et al. (2019). The fructoborates: part of a family of naturally occurring sugar–borate complexes—biochemistry, physiology, and impact on human health: a review. Biological Trace Element Research, 188(1), 11-25. doi:10.1007/s12011-018-1550-4

Huo, S., Tang, X., Chen, W., Gan, D., Guo, H., Yao, Q., Liao, R., Huang, T., Wu, J., Yang, J., et al. (2024). Epigenetic regulations of cellular senescence in osteoporosis. Ageing Research Reviews, 99, 102235. doi:10.1016/j.arr.2024.102235

İlhan, N., Telli, S., Temel, B., & Aştı, T. (2022). Investigating the sexual satisfaction mediating role in the relationship between health literacy and self-care of men with diabetes and women's marital satisfaction. Journal of Integrative Nursing and Palliative Care, 3(1-2022), 19-25. doi:10.51847/sFjL3OLpqg

Imamudeen, N., Basheer, A., Iqbal, A. M., Manjila, N., Haroon, N. N., & Manjila, S. (2022). Management of osteoporosis and spinal fractures: contemporary guidelines and evolving paradigms. Clinical Medicine & Research, 20(2), 95-106. doi:10.3121/cmr.2021.1612

Jadhav, N., Ajgaonkar, S., Saha, P., Gurav, P., Pandey, A., Basudkar, V., Gada, Y., Panda, S., Jadhav, S., Mehta, D., et al. (2022). Molecular pathways and roles for vitamin K2-7 as a health-beneficial nutraceutical: challenges and opportunities. Frontiers in Pharmacology, 13, 896920. doi:10.3389/fphar 2022.896920

Jesima, J., Kannan, R. K., & Indrapriyadharshini, K. (2024). Short implant: a new normal in implant dentistry. Annals of Dental Specialty, 12(3), 34-41. doi:10.51847/36HxytCIuB

Johnston, C. B., & Dagar, M. (2020). Osteoporosis in older adults. Medical Clinics, 104(5), 873-884. doi:10.1016/j.mcna.2020.06.004

Kanno, H., Onoda, Y., Hashimoto, K., Aizawa, T., & Ozawa, H. (2022). Reinforcement of percutaneous pedicle screw fixation with hydroxyapatite granules in patients with osteoporotic spine: biomechanical performance and clinical outcomes. Medicina, 58(5), 579. doi:10.3390/medicina58050579

Kendler, D. L., Cosman, F., Stad, R. K., & Ferrari, S. (2022). Denosumab in the treatment of osteoporosis: 10 years later: a narrative review. Advances in Therapy, 39(1), 58-74. doi:10.1007/s12325-021-01936-y

Kołodziejska, B., Stępień, N., & Kolmas, J. (2021). The influence of strontium on bone tissue metabolism and its application in osteoporosis treatment. International Journal of Molecular Sciences, 22(12), 6564. doi:10.3390/ijms22126564

Laurent, M. R., Goemaere, S., Verroken, C., Bergmann, P., Body, J. J., Bruyère, O., Cavalier, E., Rozenberg, S., Lapauw, B., & Gielen, E. (2022). Prevention and treatment of glucocorticoid-induced osteoporosis in adults: consensus recommendations from the Belgian bone club. Frontiers in Endocrinology, 13, 908727. doi:10.3389/fendo.2022.908727

Lei, C., Song, J. H., Li, S., Zhu, Y. N., Liu, M. Y., Wan, M. C., Mu, Z., Tay, F. R., & Niu, L. N. (2023). Advances in materials-based therapeutic strategies against osteoporosis. Biomaterials, 296, 122066. doi:10.1016/j.biomaterials.2023.122066

Li, N., Cornelissen, D., Silverman, S., Pinto, D., Si, L., Kremer, I., Bours, S., de Bot, R., Boonen, A., Evers, S., et al. (2021). An updated systematic review of cost-effectiveness analyses of drugs for osteoporosis. Pharmacoeconomics, 39(2), 181-209. doi:10.1007/s40273-020-00965-9

Ma, M. L., Ma, Z. J., He, Y. L., Sun, H., Yang, B., Ruan, B. J., Zhan, W. D., Li, S. X., Dong, H., & Wang, Y. X. (2022). Efficacy of vitamin K2 in the prevention and treatment of postmenopausal osteoporosis: a systematic review and meta-analysis of randomized controlled trials. Frontiers in Public Health, 10, 979649. doi:10.3389/fpubh.2022.979649

Ma, T. L., Zhu, P., Ke, Z. R., Chen, J. X., Hu, Y. H., & Xie, J. (2022). Focusing on OB-OC-MΦ axis and miR-23a to explore the pathogenesis and treatment strategy of osteoporosis. Frontiers in Endocrinology, 13, 891313. doi:10.3389/fendo.2022.891313

Macías, J. G., & Martínez, J. M. O. (2022). Aminobisphosphonates: Reconsideration 25 years after their approval for the treatment of osteoporosis. Medicina Clínica (English Edition), 159(7), 336-343. doi:10.1016/j.medcli.2022.04.003

Meng, F., Yu, Y., Tian, Y., Deng, M., Zheng, K., Guo, X., Zeng, B., Li, J., Qian, A., & Yin, C. (2023). A potential therapeutic drug for osteoporosis: prospect for osteogenic LncRNAs. Frontiers in Endocrinology, 14, 1219433. doi:10.3389/fendo.2023.1219433

Mladěnka, P., Macáková, K., Kujovská Krčmová, L., Javorská, L., Mrštná, K., Carazo, A., Protti, M., Remião, F., Nováková, L., & OEMONOM Researchers and Collaborators. (2022). Vitamin K–sources, physiological role, kinetics, deficiency, detection, therapeutic use, and toxicity. Nutrition Reviews, 80(4), 677-698. doi:10.1093/nutrit/nuab061

Mogoşanu, G. D., Biţă, A., Bejenaru, L. E., Bejenaru, C., Croitoru, O., Rău, G., Rogoveanu, O. C., Florescu, D. N., Neamţu, J., Scorei, I. D., et al. (2016). Calcium fructoborate for bone and cardiovascular health. Biological Trace Element Research, 172(2), 277-281. doi:10.1007/s12011-015-0590-2

Mounir, M. M. F., Alharthi, A., Jamaluddinsyed, J., & Alkeheli, M. (2023). Recombinant amelogenin protein alone regenerates lost tissues in immature teeth with pulp necrosis and preapical periodontitis. Annals of Dental Specialty, 11(4-2023), 9-15. doi:10.51847/zPEhEt763f

Muthuvignesh, N., Jei, J. B., & Balasubramaniam, M. (2023). Tooth supported overdenture in old patient with denture characterizaton-a case report. Annals of Dental Specialty, 11(1-2023), 16-20. doi:10.51847/s9fuIqCHoR

Naseri, B., & Sasani, S. (2024). Stem rust, planting date, wheat maturity and genetic resistance, weather and productivity. World Journal of Environmental Biosciences, 13(4-2024), 1-6. doi:10.51847/2njt2p8YQ0

Omidian, H., & Chowdhury, S. D. (2023). Advancements and applications of injectable hydrogel composites in biomedical research and therapy. Gels, 9(7), 533. doi:10.3390/gels9070533

Oran, İ. B., Ayboğa, M. H., Erol, M., & Yildiz, G. (2022). The necessity of transition from industry 4.0 to industry 5.0: SWOT analysis of Turkey’s SCM strategy. Journal of Organizational Behavior Research, 7(2-2022), 1-17. doi:10.51847/vrFR9HDvbh

Palui, R., Durgia, H., Sahoo, J., Naik, D., & Kamalanathan, S. (2022). Timing of osteoporosis therapies following fracture: the current status. Therapeutic Advances in Endocrinology and Metabolism, 13, 20420188221112904. doi:10.1177/20420188221112904

Papadopoulou, S. K., Papadimitriou, K., Voulgaridou, G., Georgaki, E., Tsotidou, E., Zantidou, O., & Papandreou, D. (2021). Exercise and nutrition impact on osteoporosis and sarcopenia—the incidence of osteosarcopenia: a narrative review. Nutrients, 13(12), 4499. doi:10.3390/nu13124499

Ran, L., Liu, L., Gao, J., Pan, Y., Ramalingam, M., Du, X., Liu, Y., Cheng, L., & Shi, Z. (2023). Strontium-doped hydroxyapatite and its role in osteogenesis and angiogenesis. The International Journal of Developmental Biology, 67(4), 137-146. doi:10.1387/ijdb.230091lc

Ranjel, E. S. M., Moreno, P. M. N., Córdova, M. G. D. G., Castillo, C. G. G., Flores, V. J. A., Conesa, J. G., & López-García, J. A. (2025). Bee propolis (Apis mellifera) as a growth promoter in Tilapia (Oreochromis niloticus). World Journal of Environmental Biosciences, 14(2), 13-19. doi:10.51847/uDczvYfi37

Reid, I. R., & Billington, E. O. (2022). Drug therapy for osteoporosis in older adults. The Lancet, 399(10329), 1080-1092. doi:10.1016/S0140-6736(21)02646-5

Riegger, J., Schoppa, A., Ruths, L., Haffner-Luntzer, M., & Ignatius, A. (2023). Oxidative stress as a key modulator of cell fate decision in osteoarthritis and osteoporosis: a narrative review. Cellular & Molecular Biology Letters, 28(1), 76. doi:10.1186/s11658-023-00489-y

Rizzoli, R., & Chevalley, T. (2024). Nutrition and osteoporosis prevention. Current Osteoporosis Reports, 22(6), 515-522. doi:10.1007/s11914-024-00892-0

Rzhepakovsky, I., Piskov, S., Avanesyan, S., Sizonenko, M., Timchenko, L., Anfinogenova, O., Nagdalian, A., Blinov, A., Denisova, E., Kochergin, S., et al. (2024). Composite of bacterial cellulose and gelatin: a versatile biocompatible scaffold for tissue engineering. International Journal of Biological Macromolecules, 256, 128369. doi:10.1016/j.ijbiomac.2023.128369

Sahu, M. K., & Tiwari, S. P. (2024). Phytochemical and Ethnopharmacological review of Aegle marmelos Linn. (Bael). Bulletin of Pioneering Researches of Medical and Clinical Science, 3(2-2024), 29-47. doi:10.51847/K3rPdVPzLe

Scorei, I. D., & Scorei, R. I. (2013). Calcium fructoborate helps control inflammation associated with diminished bone health. Biological Trace Element Research, 155(3), 315-321. doi:10.1007/s12011-013-9800-y

Shahzan, S., Paulraj, J., & Maiti, S. (2022). Assessment of anxiety levels in children receiving dental treatment using rubber dam-A randomized control trial. Annals of Dental Specialty, 10(4-2022), 15-21. doi:10.51847/Ang4hblnjK

Sheng, X., Li, C., Wang, Z., Xu, Y., Sun, Y., Zhang, W., Liu, H., & Wang, J. (2023). Advanced applications of strontium-containing biomaterials in bone tissue engineering. Materials Today Bio, 20, 100636. doi:10.1016/j.mtbio.2023.100636

Shevroja, E., Reginster, J. Y., Lamy, O., Al-Daghri, N., Chandran, M., Demoux-Baiada, A. L., Kohlmeier, L., Lecart, M. P., Messina, D., Camargos, B. M., et al. (2023). Update on the clinical use of trabecular bone score (TBS) in the management of osteoporosis: results of an expert group meeting organized by the European Society for Clinical and Economic Aspects of Osteoporosis, Osteoarthritis and Musculoskeletal Diseases (ESCEO), and the International Osteoporosis Foundation (IOF) under the auspices of WHO Collaborating Center for Epidemiology of Musculoskeletal Health and Aging. Osteoporosis International, 34(9), 1501-1529. doi:10.1007/s00198-023-06817-4

Singh, G. P., Attavar, S. H., & Kavuri, S. (2022). Application of cone-beam computed tomography in diagnosis and treatment of multiple canals–a case report. Annals of Dental Specialty, 10(2-2022), 15-18. doi:10.51847/vgeNZYRIRH

Skalny, A.V., Aschner, M., Tsatsakis, A., Rocha, J. B., Santamaria, A., Spandidos, D. A., Martins, A. C., Lu, R., Korobeinikova, T. V., Chen, W., et al. (2024). Role of vitamins beyond vitamin D 3 in bone health and osteoporosis. International Journal of Molecular Medicine, 53(1), 9. doi:10.3892/ijmm.2023.5333

Song, S., Guo, Y., Yang, Y., & Fu, D. (2022). Advances in pathogenesis and therapeutic strategies for osteoporosis. Pharmacology & Therapeutics, 237, 108168. doi:10.1016/j.pharmthera.2022.108168

Veronese, N., Briot, K., Guañabens, N., Albergaria, B. H., Alokail, M., Al-Daghri, N., Bemden, A. B. V., Bruyère, O., Burlet, N., Cooper, C., et al. (2024). Recommendations for the optimal use of bone forming agents in osteoporosis. Aging Clinical and Experimental Research, 36(1), 167. doi:10.1007/s40520-024-02826-3

Wan, B., Wang, R., Sun, Y., Cao, J., Wang, H., Guo, J., & Chen, D. (2020). Building osteogenic microenvironments with strontium-substituted calcium phosphate ceramics. Frontiers in Bioengineering and Biotechnology, 8, 591467. doi:10.3389/fbioe 2020.591467

Wu, D., Li, L., Wen, Z., & Wang, G. (2023). Romosozumab in osteoporosis: yesterday, today and tomorrow. Journal of Translational Medicine, 21(1), 668. doi:10.1186/s12967-023-04563-z

Xiao, P. L., Cui, A. Y., Hsu, C. J., Peng, R., Jiang, N., Xu, X. H., Ma, Y. G., Liu, D., & Lu, H. D. (2022). Global, regional prevalence, and risk factors of osteoporosis according to the World Health Organization diagnostic criteria: a systematic review and meta-analysis. Osteoporosis International, 33(10), 2137-2153. doi:10.1007/s00198-022-06454-3

Yilmaz, S., Ertürk, M., Soydemir, A., Erciyas, A., & Oran, İ. B. (2023). Military implications of artificial intelligence-case of republic of Turkey. Journal of Organizational Behavior Research, 8(2-2023), 1-14. doi:10.51847/Tal2sc1FFp

Yu, L. X., Wang, J. T., & Hussain, A. S. (2002). Evaluation of USP apparatus 3 for dissolution testing of immediate-release products. AAPS PharmSci, 4(1), 1. doi:10.1208/ps040101

Zhang, T., O’Connor, C., Sheridan, H., & Barlow, J. W. (2024). Vitamin K2 in health and disease: a clinical perspective. Foods, 13(11), 1646. doi:10.3390/foods13111646

Zhou, M., Han, S., Zhang, W., & Wu, D. (2022). Efficacy and safety of vitamin K2 for postmenopausal women with osteoporosis at a long-term follow-up: meta-analysis and systematic review. Journal of Bone and Mineral Metabolism, 40(5), 763-772. doi:10.1007/s00774-022-01342-6


How to cite this article
Vancouver
Tovsultanov A, Gereykhanov H, Magomedova D, Khisimikova K, Gashimova Z, Khalifaeva D, et al. Targeted Micronutrition for Bone Regeneration: Proof-of-Concept for a Novel Multi-Pathway Approach to Osteoporosis Management. J Biochem Technol. 2025;16(2):75-83. https://doi.org/10.51847/VM5h8y9Dhq
APA
Tovsultanov, A., Gereykhanov, H., Magomedova, D., Khisimikova, K., Gashimova, Z., Khalifaeva, D., Khizriev, M., Magomedova, Z., Magomedova, G., & Eremina, A. (2025). Targeted Micronutrition for Bone Regeneration: Proof-of-Concept for a Novel Multi-Pathway Approach to Osteoporosis Management. Journal of Biochemical Technology, 16(2), 75-83. https://doi.org/10.51847/VM5h8y9Dhq
Articles
Issue 4 Volume 16 - 2025