Doxycycline restores the impaired osteogenic commitment of diabetic-derived bone marrow mesenchymal stromal cells by increasing the canonical WNT signaling

https://doi.org/10.1016/j.mce.2020.110975Get rights and content

Highlights

  • Diabetes decreases osteogenesis and increases adipogenesis of precursor cells.

  • Doxycycline restores diabetic-derived cells' viability and proliferation.

  • Doxycycline restores the osteogenic program of diabetes-derived stem cells.

  • Doxycycline increased the canonical Wnt/b-catenin pathway signaling.

Abstract

Diabetes mellitus comprehends a group of chronic metabolic disorders, associated with damage and dysfunction of distinct tissues, including bone. At the cellular level, an impaired osteoblastogenesis has been reported, affecting the viability, proliferation and functionality of osteoblasts and precursor populations, hampering the bone metabolic activity, remodeling and healing. Tetracyclines embrace a group of broad-spectrum antibacterial compounds with potential anabolic effects on the bone tissue, through antibacterial-independent mechanisms. Accordingly, this study aims to address the modulatory capability and associated molecular signaling of a low dosage doxycycline – a semi-synthetic tetracycline, in the functional activity of osteoblastic progenitor cells (bone marrow-derived mesenchymal stromal cells), established from a translational diabetic experimental model.

Bone marrow-derived mesenchymal stromal cells were isolated from streptozotocin-induced diabetic Wistar rat with proven osteopenia. Cultures were characterized, in the presence of doxycycline (1 μg ml−1) for proliferation, metabolic activity, apoptosis, collagen synthesis and relevant gene expression with the osteogenic and adipogenic program. The activation of the Wnt/β-catenin pathway was further detailed.

Doxycycline normalized the viability, proliferation and metabolic activity of the established cultures, further decreasing cell apoptosis, to levels similar to control. The addition of this drug to the culture environment further increased the osteogenic activation, upregulating the expression of osteogenic markers and collagen synthesis, at the same time that a decreased adipogenic priming was attained. These processes were found to me mediated, at least in part, by the restoration of the signaling through the Wnt/β-catenin pathway.

Introduction

Diabetes mellitus (DM) encompasses a group of metabolic disorders characterized by hyperglycemia due to the lack of insulin production, peripheral insulin resistance, or both. DM seems to affect the skeletal system and the bone metabolism, as a relation between an increased risk of fragility fractures and DM has been established, and associated with excess morbidity, mortality and health-care costs (Miao et al., 2005; Vestergaard et al., 2009; Vestergaard, 2007a). Evidences both from the bench and bedside reveal a strong correlation between DM and bone functional impairment, with type 1 diabetes mellitus (T1DM) affecting the bone tissue more severely, with established alterations not only at the cell functionality, but also at the extracellular matrix level, disturbing the organic collagen structure and the inorganic bone mineral (Pan et al., 2014; Vestergaard, 2007b). T1DM is further characterized by a diminished bone turnover, particularly at the expenses of the bone formation process (Starup-Linde, 2013), as sustained by the reduced biochemical markers of bone formation and histomorphometric indices at the trabecular and cortical bone of experimental diabetes animal models and diabetic patients (Silva et al., 2009; Massé et al., 2010; Gunczler et al., 2001; Keenan and Maddaloni, 2016; Cervino et al., 2019). Hence, at the cellular level, an impaired osteoblastogenesis has been reported, with an increased cell death and altered functional activity, impaired osteogenic priming and delayed osteoblastic differentiation (McCabe et al., 2011; Hie et al., 2011; Silva et al., 2015). This is corroborated by the downregulation of osteogenic transcription factors and downstream targets, in association with an increased adipogenic priming of precursor cell populations, in distinct T1DM models (Lu et al., 2003; Fowlkes et al., 2007; Nyman et al., 2017; Bortolin et al., 2017). Clinically, an increased fracture rate, at different bone locations, has been established for diabetic individuals, as well as an increased risk of complications during fracture healing – with significant higher rates of delayed union, nonunion and postoperative infection, comparing to nondiabetic individuals (Sundararaghavan et al., 2017). As a result, there is an urge of innovative therapeutic approaches, based on effective modulatory agents that are capable of restoring the osteogenic functionality in diabetic conditions and prime/accelerate tissue healing (Herford et al., 2013).

Tetracyclines embrace a group of broad-spectrum antibacterial compounds – highly effective against gram-positive and gram-negative bacteria, spirochetes and rickettsia – that show a high affinity to mineralized tissues, forming bioactive complexes with the inorganic mineral component (Chopra and Roberts, 2001), and an adequate cytocompatible profile (Kallala et al., 2012). Tetracyclines, and particularly the semi-synthetic minocycline and doxycycline at a low dosage regimen, display an effective modulatory activity within the bone tissue metabolism – tempering the remodeling and healing processes – through antibacterial-independent mechanisms (Golub et al., 2016; Griffin et al., 2011). Our group has previously shown that low concentrations of doxycycline increased the metabolic activity and the osteogenic potential of human osteoblastic populations, with increased expression of osteogenic markers, enhanced extracellular matrix mineralization, inducing no negative effects on calcium metabolism (Gomes and Fernandes, 1997). Bone regenerative biomaterials, releasing low levels of semi-synthetic tetracyclines, also showed an increased osteogenic activation, in vitro and in vivo (Walter et al., 2014; Silva et al., 2017, 2018). Tetracyclines were further found to normalize bone remodeling and enhance the bone formation process in compromised conditions (Zhou et al., 2010; Kinugawa et al., 2012), supporting their successful clinical application in the management of osteopenia/osteoporosis (Payne and Golub, 2011; Golub et al., 2010; de Figueiredo et al., 2019).

Minocycline and doxycycline have also been found to modulate the diabetic disease course, lessening diabetic-related complications, such as the peripheral and autonomic neuropathy (Syngle et al., 2014), retinopathy (Scott et al., 2014), nephropathy (Aggarwal et al., 2010), and periodontal disease (ŁKondzielnik et al., 2012), in a process presumably related with the management of the excessively activated inflammatory response, matrix metalloproteinases modulation and normalization of the altered oxidative stress balance (Garrido‐Mesa et al., 2013). Despite the evidence of the positive modulation of diabetic-related complications by tetracyclines, studies concerning the diabetic osteopenia and tetracyclines are still very scarce. These have been limited to the assessment, in the early 90's of the 20th century, of Golub et al. reporting that doxycycline (Golub et al., 1990) and minocycline (Sasaki et al., 1992) prevented osteopenia, in a rat diabetic model, based on the morphological characterization of the bone tissue. Therefore, and to the best of the authors' knowledge, no cellular or molecular data on the potential modulatory activity of these drugs regarding the diabetes-mediated impaired osteoblastic functionality have been addressed. In this frame, this study aims to detail the modulatory capacity and associated molecular signaling of a low dosage doxycycline regimen, in the functional activity of an osteoblastic precursor population, established from a translational T1DM experimental model.

Section snippets

Animals

This study was conducted in accordance with accepted standards for the humane animal care and manipulation. Procedures were approved by Direção Geral de Alimentação e Veterinária and encompassed the standards for the protection of experimental animals, according to the Portuguese and European legislations.

Experimental diabetic model

All animals of both SHAM and STZ groups survived throughout the duration of the experimental protocol. Whether no significant differences were found on the initial weight (SHAM, 339.5 g ± 23.1 and STZ, 317.0 g ± 33.8), 6 weeks following induction, animals of the SHAM group increased in weight (396.7 g ± 37.3), while animals of the STZ group significantly loss weight (228.1 g ± 26.41). Further, at euthanasia, all STZ-administered animals had glycemic levels over 300 mg/dl, while animals from the

Discussion

The strong correlation between DM and bone functional impairment reflects significant changes at the cellular and tissue level, namely concerning osteogenic priming, differentiation and functionality (McCabe et al., 2011; Silva et al., 2014). Underlying mechanisms include the downregulation of osteogenic transcription factors and downstream targets due to impaired molecular signaling pathways directing osteogenesis (Zhao et al., 2013; Al-Qarakhli et al., 2019) and the interconnected

Conclusions

In this work, we have shown that doxycycline, at a low dosage regimen (1 μg ml−1), enhanced the functional activity and osteogenic capability of MSCs derived from STZ-induced diabetic rats, to levels similar to those of control, i.e. MSCs cultures established from SHAM animals. Doxycycline normalized the cell proliferation and metabolic activity of the established cultures, further decreasing cell apoptosis. The addition of this drug to the culture environment increased the osteogenic

Funding

The work was supported by UIDB/50006/2020 with funding from FCT/MCTES through national funds, and by the Faculty of Dental Medicine, U. Porto.

Ethics approval

This study was conducted in compliance with Portuguese and European legislations (Directive, 2010/63/EU in Europe) with accepted standards of humane animal care and was approved by the local IACUC.

Consent to participate

Not applicable.

Availability of data and materials

The datasets used and/or analyzed during the current study are available from the corresponding author on reasonable request.

Code availability

Not applicable.

Authors' contributions

MHF was responsible for conception of the study, made substantial contributions for the interpretation of data and substantively revised the manuscript. PSG was responsible for the study design, data analysis, interpretation and draft the manuscript. MR was responsible for data acquisition, analysis and interpretation. All authors have approved the submitted version and have agreed both to be personally accountable for the their own contributions and to ensure that questions related to the

CRediT authorship contribution statement

Pedro Sousa Gomes: Conceptualization, Supervision, Writing - review & editing. Marta Resende: Investigation, Formal analysis, Writing - original draft. Maria Helena Fernandes: Resources, Methodology, Validation, Funding acquisition.

Declaration of competing interest

The authors declare that they have no competing interests.

References (77)

  • J. Payne et al.

    Using tetracyclines to treat osteoporotic/osteopenic bone loss: from the basic science laboratory to the clinic

    Pharmacol. Res.

    (2011)
  • M. Walter et al.

    Bioactive implant surface with electrochemically bound doxycycline promotes bone formation markers in vitro and in vivo

    Dent. Mater.

    (2014)
  • M. Wei et al.

    The streptozotocin-diabetic rat as a model of the chronic complications of human diabetes

    Heart Lung Circ.

    (2003)
  • E. Weinberg et al.

    Streptozotocin-induced diabetes in rats diminishes the size of the osteoprogenitor pool in bone marrow

    Diabetes Res. Clin. Pract.

    (2014)
  • B.M. Abdallah

    Marrow adipocytes inhibit the differentiation of mesenchymal stem cells into osteoblasts via suppressing BMP-signaling

    J. Biomed. Sci.

    (2017)
  • H. Aggarwal et al.

    Evaluation of role of doxycycline (a matrix metalloproteinase inhibitor) on renal functions in patients of diabetic nephropathy

    Ren. Fail.

    (2010)
  • A. Al-Qarakhli et al.

    Effects of high glucose conditions on the expansion and differentiation capabilities of mesenchymal stromal cells derived from rat endosteal niche

    BMC Mol Cell Biol

    (2019)
  • C. Bennett et al.

    Regulation of osteoblastogenesis and bone mass by Wnt10b. PNAS

  • P. Bodine et al.

    Wnt signaling and osteoblastogenesis

    Rev. Endocr. Metab. Disord.

    (2006)
  • R.H. Bortolin et al.

    Anabolic effect of insulin therapy on the bone: osteoprotegerin and osteocalcin up-regulation in streptozotocin-induced diabetic rats

    Basic Clin Pharmacol

    (2017)
  • D. Cao et al.

    Doxycycline masks the genuine effect of the doxycycline-inducible transgene by promoting dopaminergic neuron differentiation from human pluripotent stem cells

    Stem Cell. Dev.

    (2019)
  • G. Cervino et al.

    Diabetes: oral health related quality of life and oral alterations

    BioMed Res. Int.

    (2019)
  • I. Chopra et al.

    Tetracycline antibiotics: mode of action, applications, molecular biology, and epidemiology of bacterial resistance

    Mol Biol Rev

    (2001)
  • F. de Figueiredo et al.

    Doxycycline reduces osteopenia in female rats

    Sci. Rep.

    (2019)
  • K. do Nascimento Gomes et al.

    Doxycycline induces bone repair and changes in Wnt signalling

    Int. J. Oral Sci.

    (2017)
  • S. Durão et al.

    The biomaterial-mediated healing of critical size bone defects in the ovariectomized rat

    Osteoporos. Int.

    (2014)
  • J.L.B.R. Fowlkes et al.

    Runt-related transcription factor 2 (RUNX2) and RUNX2-related osteogenic genes are down-regulated throughout osteogenesis in type 1 diabetes mellitus

    Endocrinology

    (2007)
  • N. Garrido Mesa et al.

    Minocycline: far beyond an antibiotic

    Br. J. Pharmacol.

    (2013)
  • L. Golub et al.

    Tetracycline administration prevents diabetes-induced osteopenia in the rat: initial observations

    Res. Commun. Chem. Pathol. Pharmacol.

    (1990)
  • L. Golub et al.

    Doxycycline effects on serum bone biomarkers in postmenopausal women

    J. Dent. Res.

    (2010)
  • P. Gomes et al.

    Effect of therapeutic levels of doxycycline and minocycline in the proliferation and differentiation of human bone marrow osteoblastic cells

    Arch. Oral Biol.

    (1997)
  • P.S. Gomes et al.

    Rodent models in bone-related research: the relevance of calvarial defects in the assessment of bone regeneration strategies

    Lab Animal-UK

    (2011)
  • P. Gunczler et al.

    Decreased bone mineral density and bone formation markers shortly after diagnosis of clinical type 1 diabetes mellitus

    J Pediatric End Ocrinol Metabolism

    (2001)
  • B. Gustafson et al.

    The WNT inhibitor dickkopf 1 and bone morphogenetic protein 4 rescue adipogenesis in hypertrophic obesity in humans

    Diabetes

    (2012)
  • A. Herford et al.

    Immediate distraction osteogenesis: the sandwich technique in combination with rhBMP-2 for anterior maxillary and mandibular defects

    J Cranifac Surg

    (2013)
  • M. Hie et al.

    Insulin-dependent diabetes mellitus decreases osteoblastogenesis associated with the inhibition of Wnt signaling through increased expression of Sost and Dkk1 and inhibition of Akt activation

    Int. J. Mol. Med.

    (2011)
  • E. Kalaitzoglou et al.

    Effects of type 1 diabetes on osteoblasts, osteocytes, and osteoclasts

    Curr. Osteoporos. Rep.

    (2016)
  • R. Kallala et al.

    In vitro and in vivo effects of antibiotics on bone cell metabolism and fracture healing

    Expet Opin. Drug Saf.

    (2012)
  • Cited by (8)

    View all citing articles on Scopus
    View full text