Fluoroquinolone-Mediated Tendinopathy and Tendon Rupture
Abstract
:1. Introduction
1.1. Foreword
1.2. Search Strategy
2. Fluoroquinolones
2.1. General Information
2.2. Mode of Action and Implementation
2.3. Side Effects
3. Tendon Tissue
3.1. Tendon Composition and Structure
3.2. Tendon-Lineage Cells
3.3. Other Tendon Resident Cell Types
4. Fluoroquinolones and Tendon Diseases
4.1. Fluoroquinolones and Tendinopathy
4.2. Fluoroquinolones Link to Tendinopathy and Tendon Rupture
4.3. Fluoroquinolones Effects on Tendon-Lineage Cells
4.4. Fluoroquinolones Effects on Immune Cells, Endothelial Cells, and Neurons
4.5. Studies on FQ-Mediated Tendon Rupture Using In Vivo Models
4.6. Preventative Measures and Treatment
5. Conclusions
6. Future Directions
Author Contributions
Funding
Acknowledgments
Conflicts of Interest
Abbreviations
CPX | Ciprofloxacin |
CD | Cluster of differentiation |
CDK | Cyclin-dependent kinase |
CSF1 | Colony stimulating factor 1 |
CX3CL1 | C-X3-C motif chemokine ligand 1 |
CX3CR1 | C-X3-C motif chemokine receptor 1 |
ECM | Extracellular matrix |
EGR | Early growth response factor |
EMA | European Medicines Agency |
ENR | Enrofloxacin |
FACS | Flow cytometry |
FAK | Focal adhesion kinase |
FDA | Food and Drug Administration |
FGF | Fibroblast Growth Factor |
FLX | Fleroxacin |
FQ | Fluoroquinolone |
FQAD | Fluoroquinolone-associated disability |
IL | Interleukin |
LFX | Levofloxacin |
MMP | Matrix metalloproteinase |
Mrp4 | Multidrug resistance protein 4 |
MSC | Mesenchymal stem cell |
Mkx | Mohawk |
MOX | Moxifloxacin |
Nes | Nestin |
NOR | Norfloxacin |
PFX | Pefloxacin |
PDGF | Platelet-derived growth factor |
PDGFRA | Platelet-derived growth factor receptor alpha |
qPCR | Quantitative real-time PCR |
ROS | Reactive oxygen species |
Scx | Scleraxis |
SLRP | Small Leucin-Rich Proteoglycans |
SMOC | SPARC Related Modular Calcium Binding |
SLC | Solute carrier family 40 member 1 |
SPX | Sparfloxacin |
TSPC | Tendon stem/progenitor cells |
TNMD | Tenomodulin |
TGF | Transforming growth factor |
Tppp3 | Tubulin Polymerization Promoting Protein Family Member 3 |
References
- Bhatt, S.; Chatterjee, S. Fluoroquinolone antibiotics: Occurrence, mode of action, resistance, environmental detection, and remediation—A comprehensive review. Environ. Pollut. 2022, 315, 120440. [Google Scholar] [CrossRef] [PubMed]
- Freeman, M.Z.; Cannizzaro, D.N.; Naughton, L.F.; Bove, C. Fluoroquinolones-Associated Disability: It Is Not All in Your Head. NeuroSci 2021, 2, 235–253. [Google Scholar] [CrossRef]
- Anwar, A.I.; Lu, L.; Plaisance, C.J.; Daniel, C.P.; Flanagan, C.J.; Wenger, D.M.; McGregor, D.; Varrassi, G.; Kaye, A.M.; Ahmadzadeh, S.; et al. Fluoroquinolones: Neurological Complications and Side Effects in Clinical Practice. Cureus 2024, 16, e54565. [Google Scholar] [CrossRef] [PubMed]
- Morales, D.; Pacurariu, A.; Slattery, J.; Pinheiro, L.; McGettigan, P.; Kurz, X. Association Between Peripheral Neuropathy and Exposure to Oral Fluoroquinolone or Amoxicillin-Clavulanate Therapy. JAMA Neurol. 2019, 76, 827–833. [Google Scholar] [CrossRef]
- Rusu, A.; Munteanu, A.-C.; Arbănași, E.-M.; Uivarosi, V. Overview of Side-Effects of Antibacterial Fluoroquinolones: New Drugs versus Old Drugs, a Step Forward in the Safety Profile? Pharmaceutics 2023, 15, 804. [Google Scholar] [CrossRef]
- Drayson, M.T.; Bowcock, S.; Planche, T.; Iqbal, G.; Pratt, G.; Yong, K.; Wood, J.; Raynes, K.; Higgins, H.; Dawkins, B.; et al. Levofloxacin prophylaxis in patients with newly diagnosed myeloma (TEAMM): A multicentre, double-blind, placebo-controlled, randomised, phase 3 trial. Lancet Oncol. 2019, 20, 1760–1772. [Google Scholar] [CrossRef]
- Lode, H.; Aronkyto, T.; Chuchalin, A.G.; Jaaskevi, M.; Kahnovskii, I.; Kleutgens, K. A randomised, double-blind, double-dummy comparative study of gatifloxacin with clarithromycin in the treatment of community-acquired pneumonia. Clin. Microbiol. Infect. 2004, 10, 403–408. [Google Scholar] [CrossRef]
- Noel, G.J.; Bradley, J.S.; Kauffman, R.E.; Duffy, C.M.; Gerbino, P.G.; Arguedas, A.; Bagchi, P.; Balis, D.A.; Blumer, J.L. Comparative safety profile of levofloxacin in 2523 children with a focus on four specific musculoskeletal disorders. Pediatr. Infect. Dis. J. 2007, 26, 879–891. [Google Scholar] [CrossRef]
- Lewis, T.; Cook, J. Fluoroquinolones and tendinopathy: A guide for athletes and sports clinicians and a systematic review of the literature. J. Athl. Train. 2014, 49, 422–427. [Google Scholar] [CrossRef]
- Scheld, W.M. Maintaining fluoroquinolone class efficacy: Review of influencing factors. Emerg. Infect. Dis. 2003, 9, 1–9. [Google Scholar] [CrossRef]
- Rawal, S.Y.; Walters, J.D. Effect of biologic mediators on ciprofloxacin accumulation by gingival fibroblasts. J. Periodontol. 2005, 76, 2254–2259. [Google Scholar] [CrossRef] [PubMed]
- Kaguelidou, F.; Turner, M.A.; Choonara, I.; Jacqz-Aigrain, E. Ciprofloxacin use in neonates: A systematic review of the literature. Pediatr. Infect. Dis. J. 2011, 30, e29–e37. [Google Scholar] [CrossRef] [PubMed]
- Choi, S.-H.; Kim, E.Y.; Kim, Y.-J. Systemic use of fluoroquinolone in children. Korean J. Pediatr. 2013, 56, 196–201. [Google Scholar] [CrossRef] [PubMed]
- Pham, T.D.M.; Ziora, Z.M.; Blaskovich, M.A.T. Quinolone antibiotics. Medchemcomm 2019, 10, 1719–1739. [Google Scholar] [CrossRef]
- Borcherding, S.M.; Stevens, R.; Nicholas, R.A.; Corley, C.R.; Self, T. Quinolones: A practical review of clinical uses, dosing considerations, and drug interactions. J. Fam. Pract. 1996, 42, 69–78. [Google Scholar]
- Redgrave, L.S.; Sutton, S.B.; Webber, M.A.; Piddock, L.J.V. Fluoroquinolone resistance: Mechanisms, impact on bacteria, and role in evolutionary success. Trends Microbiol. 2014, 22, 438–445. [Google Scholar] [CrossRef]
- Mathis, A.S.; Chan, V.; Gryszkiewicz, M.; Adamson, R.T.; Friedman, G.S. Levofloxacin-associated Achilles tendon rupture. Ann. Pharmacother. 2003, 37, 1014–1017. [Google Scholar] [CrossRef]
- Ball, P. Quinolone generations: Natural history or natural selection? J. Antimicrob. Chemother. 2000, 46 (Suppl. S3), 17–24. [Google Scholar] [CrossRef]
- Seral, C.; Barcia-Macay, M.; Mingeot-Leclercq, M.P.; Tulkens, P.M.; van Bambeke, F. Comparative activity of quinolones (ciprofloxacin, levofloxacin, moxifloxacin and garenoxacin) against extracellular and intracellular infection by Listeria monocytogenes and Staphylococcus aureus in J774 macrophages. J. Antimicrob. Chemother. 2005, 55, 511–517. [Google Scholar] [CrossRef]
- Cormet, E.; Huneau, J.F.; Bouras, M.; Carbon, C.; Rubinstein, E.; Tomé, D. Evidence for a passive diffusion mechanism for sparfloxacin uptake at the brush-border membrane of the human intestinal cell-line Caco-2. J. Pharm. Sci. 1997, 86, 33–36. [Google Scholar] [CrossRef]
- Vallet, C.M.; Marquez, B.; Nhiri, N.; Anantharajah, A.; Mingeot-Leclercq, M.-P.; Tulkens, P.M.; Lallemand, J.-Y.; Jacquet, E.; van Bambeke, F. Modulation of the expression of ABC transporters in murine (J774) macrophages exposed to large concentrations of the fluoroquinolone antibiotic moxifloxacin. Toxicology 2011, 290, 178–186. [Google Scholar] [CrossRef] [PubMed]
- Michot, J.-M.; Seral, C.; van Bambeke, F.; Mingeot-Leclercq, M.-P.; Tulkens, P.M. Influence of efflux transporters on the accumulation and efflux of four quinolones (ciprofloxacin, levofloxacin, garenoxacin, and moxifloxacin) in J774 macrophages. Antimicrob. Agents Chemother. 2005, 49, 2429–2437. [Google Scholar] [CrossRef] [PubMed]
- Blondeau, J.M. Fluoroquinolones: Mechanism of action, classification, and development of resistance. Surv. Ophthalmol. 2004, 49 (Suppl. S2), S73–S78. [Google Scholar] [CrossRef] [PubMed]
- Belforti, R.K.; Lagu, T.; Haessler, S.; Lindenauer, P.K.; Pekow, P.S.; Priya, A.; Zilberberg, M.D.; Skiest, D.; Higgins, T.L.; Stefan, M.S.; et al. Association Between Initial Route of Fluoroquinolone Administration and Outcomes in Patients Hospitalized for Community-Acquired Pneumonia. Clin. Infect. Dis. 2016, 63, 1–9. [Google Scholar] [CrossRef] [PubMed]
- Turnidge, J. Pharmacokinetics and pharmacodynamics of fluoroquinolones. Drugs 1999, 58 (Suppl. S2), 29–36. [Google Scholar] [CrossRef]
- Shariati, A.; Arshadi, M.; Khosrojerdi, M.A.; Abedinzadeh, M.; Ganjalishahi, M.; Maleki, A.; Heidary, M.; Khoshnood, S. The resistance mechanisms of bacteria against ciprofloxacin and new approaches for enhancing the efficacy of this antibiotic. Front. Public Health 2022, 10, 1025633. [Google Scholar] [CrossRef]
- Popowski, E.; Kohl, B.; Schneider, T.; Jankowski, J.; Schulze-Tanzil, G. Uremic Toxins and Ciprofloxacin Affect Human Tenocytes In Vitro. Int. J. Mol. Sci. 2020, 21, 4241. [Google Scholar] [CrossRef]
- Wolfson, J.S.; Hooper, D.C. Pharmacokinetics of quinolones: Newer aspects. Eur. J. Clin. Microbiol. Infect. Dis. 1991, 10, 267–274. [Google Scholar] [CrossRef]
- Blum, R.A. Influence of renal function on the pharmacokinetics of lomefloxacin compared with other fluoroquinolones. Am. J. Med. 1992, 92, 18S–21S. [Google Scholar] [CrossRef]
- Bove, C.; Baldock, R.A.; Champigneulle, O.; Martin, L.; Bennett, C.L. Fluoroquinolones: Old drugs, putative new toxicities. Expert Opin. Drug Saf. 2022, 21, 1365–1378. [Google Scholar] [CrossRef]
- Sankar, A.; Swanson, K.M.; Zhou, J.; Jena, A.B.; Ross, J.S.; Shah, N.D.; Karaca-Mandic, P. Association of Fluoroquinolone Prescribing Rates with Black Box Warnings from the US Food and Drug Administration. JAMA Netw. Open 2021, 4, e2136662. [Google Scholar] [CrossRef] [PubMed]
- Ly, N.F.; Flach, C.; Lysen, T.S.; Markov, E.; van Ballegooijen, H.; Rijnbeek, P.; Duarte-Salles, T.; Reyes, C.; John, L.H.; Karimi, L.; et al. Impact of European Union Label Changes for Fluoroquinolone-Containing Medicinal Products for Systemic and Inhalation Use: Post-Referral Prescribing Trends. Drug Saf. 2023, 46, 405–416. [Google Scholar] [CrossRef] [PubMed]
- Buehrle, D.J.; Wagener, M.M.; Clancy, C.J. Outpatient Fluoroquinolone Prescription Fills in the United States, 2014 to 2020: Assessing the Impact of Food and Drug Administration Safety Warnings. Antimicrob. Agents Chemother. 2021, 65, e0015121. [Google Scholar] [CrossRef] [PubMed]
- Chinen, T.; Sasabuchi, Y.; Matsui, H.; Yasunaga, H. Association Between Third-Generation Fluoroquinolones and Achilles Tendon Rupture: A Self-Controlled Case Series Analysis. Ann. Fam. Med. 2021, 19, 212–216. [Google Scholar] [CrossRef] [PubMed]
- Bailey, R.R.; Kirk, J.A.; Peddie, B.A. Norfloxacin-induced rheumatic disease. N. Z. Med. J. 1983, 96, 590. [Google Scholar]
- Baggio, D.; Ananda-Rajah, M.R. Fluoroquinolone antibiotics and adverse events. Aust. Prescr. 2021, 44, 161–164. [Google Scholar] [CrossRef]
- Kim, G.K. The Risk of Fluoroquinolone-induced Tendinopathy and Tendon Rupture: What Does The Clinician Need To Know? J. Clin. Aesthet. Dermatol. 2010, 3, 49–54. [Google Scholar]
- Khaliq, Y.; Zhanel, G.G. Fluoroquinolone-associated tendinopathy: A critical review of the literature. Clin. Infect. Dis. 2003, 36, 1404–1410. [Google Scholar] [CrossRef]
- Parmar, C.; Meda, K.P. Achilles tendon rupture associated with combination therapy of levofloxacin and steroid in four patients and a review of the literature. Foot Ankle Int. 2007, 28, 1287–1289. [Google Scholar] [CrossRef]
- Yang, Y.; Wu, Y.; Zhou, K.; Wu, D.; Yao, X.; Heng, B.C.; Zhou, J.; Liu, H.; Ouyang, H. Interplay of Forces and the Immune Response for Functional Tendon Regeneration. Front. Cell Dev. Biol. 2021, 9, 657621. [Google Scholar] [CrossRef]
- Huang, Z.; Yin, Z.; Xu, J.; Fei, Y.; Heng, B.C.; Jiang, X.; Chen, W.; Shen, W. Tendon Stem/Progenitor Cell Subpopulations and Their Implications in Tendon Biology. Front. Cell Dev. Biol. 2021, 9, 631272. [Google Scholar] [CrossRef] [PubMed]
- Brebels, J.; Mignon, A. Polymer-Based Constructs for Flexor Tendon Repair: A Review. Polymers 2022, 14, 867. [Google Scholar] [CrossRef] [PubMed]
- Gelse, K.; Pöschl, E.; Aigner, T. Collagens—Structure, function, and biosynthesis. Adv. Drug Deliv. Rev. 2003, 55, 1531–1546. [Google Scholar] [CrossRef] [PubMed]
- Buckley, M.R.; Evans, E.B.; Matuszewski, P.E.; Chen, Y.-L.; Satchel, L.N.; Elliott, D.M.; Soslowsky, L.J.; Dodge, G.R. Distributions of types I, II and III collagen by region in the human supraspinatus tendon. Connect. Tissue Res. 2013, 54, 374–379. [Google Scholar] [CrossRef]
- Eriksen, H.A.; Pajala, A.; Leppilahti, J.; Risteli, J. Increased content of type III collagen at the rupture site of human Achilles tendon. J. Orthop. Res. 2002, 20, 1352–1357. [Google Scholar] [CrossRef]
- Connizzo, B.K.; Yannascoli, S.M.; Soslowsky, L.J. Structure-function relationships of postnatal tendon development: A parallel to healing. Matrix Biol. 2013, 32, 106–116. [Google Scholar] [CrossRef]
- Yang, Q.; Li, J.; Su, W.; Yu, L.; Li, T.; Wang, Y.; Zhang, K.; Wu, Y.; Wang, L. Electrospun aligned poly(ε-caprolactone) nanofiber yarns guiding 3D organization of tendon stem/progenitor cells in tenogenic differentiation and tendon repair. Front. Bioeng. Biotechnol. 2022, 10, 960694. [Google Scholar] [CrossRef]
- Millar, N.L.; Silbernagel, K.G.; Thorborg, K.; Kirwan, P.D.; Galatz, L.M.; Abrams, G.D.; Murrell, G.A.C.; McInnes, I.B.; Rodeo, S.A. Tendinopathy. Nat. Rev. Dis. Primers 2021, 7, 1. [Google Scholar] [CrossRef]
- Asahara, H.; Inui, M.; Lotz, M.K. Tendons and Ligaments: Connecting Developmental Biology to Musculoskeletal Disease Pathogenesis. J. Bone Miner. Res. 2017, 32, 1773–1782. [Google Scholar] [CrossRef]
- Li, Y.; Wu, T.; Liu, S. Identification and Distinction of Tenocytes and Tendon-Derived Stem Cells. Front. Cell Dev. Biol. 2021, 9, 629515. [Google Scholar] [CrossRef]
- Kendal, A.R.; Layton, T.; Al-Mossawi, H.; Appleton, L.; Dakin, S.; Brown, R.; Loizou, C.; Rogers, M.; Sharp, R.; Carr, A. Multi-omic single cell analysis resolves novel stromal cell populations in healthy and diseased human tendon. Sci. Rep. 2020, 10, 13939. [Google Scholar] [CrossRef] [PubMed]
- Fu, W.; Yang, R.; Li, J. Single-cell and spatial transcriptomics reveal changes in cell heterogeneity during progression of human tendinopathy. BMC Biol. 2023, 21, 132. [Google Scholar] [CrossRef] [PubMed]
- Barboni, B.; Curini, V.; Russo, V.; Mauro, A.; Di Giacinto, O.; Marchisio, M.; Alfonsi, M.; Mattioli, M. Indirect co-culture with tendons or tenocytes can program amniotic epithelial cells towards stepwise tenogenic differentiation. PLoS ONE 2012, 7, e30974. [Google Scholar] [CrossRef] [PubMed]
- Kohler, J.; Popov, C.; Klotz, B.; Alberton, P.; Prall, W.C.; Haasters, F.; Müller-Deubert, S.; Ebert, R.; Klein-Hitpass, L.; Jakob, F.; et al. Uncovering the cellular and molecular changes in tendon stem/progenitor cells attributed to tendon aging and degeneration. Aging Cell 2013, 12, 988–999. [Google Scholar] [CrossRef]
- Korcari, A.; Przybelski, S.J.; Gingery, A.; Loiselle, A.E. Impact of aging on tendon homeostasis, tendinopathy development, and impaired healing. Connect. Tissue Res. 2023, 64, 1–13. [Google Scholar] [CrossRef]
- Guo, J.; Tang, H.; Huang, P.; Ye, X.; Tang, C.; Shu, Z.; Kang, X.; Shi, Y.; Zhou, B.; Liang, T.; et al. Integrative single-cell RNA and ATAC sequencing reveals that the FOXO1-PRDX2-TNF axis regulates tendinopathy. Front. Immunol. 2023, 14, 1092778. [Google Scholar] [CrossRef]
- Still, C.; Chang, W.-T.; Sherman, S.L.; Sochacki, K.R.; Dragoo, J.L.; Qi, L.S. Single-cell transcriptomic profiling reveals distinct mechanical responses between normal and diseased tendon progenitor cells. Cell Rep. Med. 2021, 2, 100343. [Google Scholar] [CrossRef]
- Marr, N.; Zamboulis, D.E.; Werling, D.; Felder, A.A.; Dudhia, J.; Pitsillides, A.A.; Thorpe, C.T. The tendon interfascicular basement membrane provides a vascular niche for CD146+ cell subpopulations. Front. Cell Dev. Biol. 2023, 10, 1094124. [Google Scholar] [CrossRef]
- Ackerman, J.E.; Best, K.T.; Muscat, S.N.; Pritchett, E.M.; Nichols, A.E.C.; Wu, C.-L.; Loiselle, A.E. Defining the spatial-molecular map of fibrotic tendon healing and the drivers of Scleraxis-lineage cell fate and function. Cell Rep. 2022, 41, 111706. [Google Scholar] [CrossRef]
- Nichols, A.E.C.; Best, K.T.; Loiselle, A.E. The cellular basis of fibrotic tendon healing: Challenges and opportunities. Transl. Res. 2019, 209, 156–168. [Google Scholar] [CrossRef]
- Mimpen, J.Y.; Ramos-Mucci, L.; Paul, C.; Kurjan, A.; Hulley, P.A.; Ikwuanusi, C.T.; Cohen, C.J.; Gwilym, S.E.; Baldwin, M.J.; Cribbs, A.P.; et al. Single nucleus and spatial transcriptomic profiling of healthy human hamstring tendon. FASEB J. 2024, 38, e23629. [Google Scholar] [CrossRef] [PubMed]
- Steffen, D.; Mienaltowski, M.; Baar, K. Spatial gene expression in the adult rat patellar tendon. Matrix Biol. Plus 2023, 19–20, 100138. [Google Scholar] [CrossRef] [PubMed]
- Zhang, J.; Li, F.; Williamson, K.M.; Tan, S.; Scott, D.; Onishi, K.; Hogan, M.V.; Wang, J.H.-C. Characterization of the structure, vascularity, and stem/progenitor cell populations in porcine Achilles tendon (PAT). Cell Tissue Res. 2021, 384, 367–387. [Google Scholar] [CrossRef] [PubMed]
- Marr, N.; Meeson, R.; Kelly, E.F.; Fang, Y.; Peffers, M.J.; Pitsillides, A.A.; Dudhia, J.; Thorpe, C.T. CD146 Delineates an Interfascicular Cell Sub-Population in Tendon That Is Recruited During Injury Through Its Ligand Laminin-α4. Int. J. Mol. Sci. 2021, 22, 9729. [Google Scholar] [CrossRef]
- de Micheli, A.J.; Swanson, J.B.; Disser, N.P.; Martinez, L.M.; Walker, N.R.; Oliver, D.J.; Cosgrove, B.D.; Mendias, C.L. Single-cell transcriptomic analysis identifies extensive heterogeneity in the cellular composition of mouse Achilles tendons. Am. J. Physiol.-Cell Physiol. 2020, 319, C885–C894. [Google Scholar] [CrossRef]
- Sorkin, M.; Huber, A.K.; Hwang, C.; Carson, W.F.; Menon, R.; Li, J.; Vasquez, K.; Pagani, C.; Patel, N.; Li, S.; et al. Regulation of heterotopic ossification by monocytes in a mouse model of aberrant wound healing. Nat. Commun. 2020, 11, 722. [Google Scholar] [CrossRef]
- Harvey, T.; Flamenco, S.; Fan, C.-M. A Tppp3+Pdgfra+ tendon stem cell population contributes to regeneration and reveals a shared role for PDGF signalling in regeneration and fibrosis. Nat. Cell Biol. 2019, 21, 1490–1503. [Google Scholar] [CrossRef]
- Yin, Z.; Hu, J.-J.; Yang, L.; Zheng, Z.-F.; An, C.-R.; Wu, B.-B.; Zhang, C.; Shen, W.-L.; Liu, H.-H.; Chen, J.-L.; et al. Single-cell analysis reveals a nestin+ tendon stem/progenitor cell population with strong tenogenic potentiality. Sci. Adv. 2016, 2, e1600874. [Google Scholar] [CrossRef]
- Asai, S.; Otsuru, S.; Candela, M.E.; Cantley, L.; Uchibe, K.; Hofmann, T.J.; Zhang, K.; Wapner, K.L.; Soslowsky, L.J.; Horwitz, E.M.; et al. Tendon progenitor cells in injured tendons have strong chondrogenic potential: The CD105-negative subpopulation induces chondrogenic degeneration. Stem Cells 2014, 32, 3266–3277. [Google Scholar] [CrossRef]
- Bi, Y.; Ehirchiou, D.; Kilts, T.M.; Inkson, C.A.; Embree, M.C.; Sonoyama, W.; Li, L.; Leet, A.I.; Seo, B.-M.; Zhang, L.; et al. Identification of tendon stem/progenitor cells and the role of the extracellular matrix in their niche. Nat. Med. 2007, 13, 1219–1227. [Google Scholar] [CrossRef]
- Wang, C.; Ma, C.; Gong, L.; Guo, Y.; Fu, K.; Zhang, Y.; Zhou, H.; Li, Y. Macrophage Polarization and Its Role in Liver Disease. Front. Immunol. 2021, 12, 803037. [Google Scholar] [CrossRef] [PubMed]
- Cotechini, T.; Atallah, A.; Grossman, A. Tissue-Resident and Recruited Macrophages in Primary Tumor and Metastatic Microenvironments: Potential Targets in Cancer Therapy. Cells 2021, 10, 960. [Google Scholar] [CrossRef] [PubMed]
- Bautista, C.A.; Srikumar, A.; Tichy, E.D.; Qian, G.; Jiang, X.; Qin, L.; Mourkioti, F.; Dyment, N.A. CD206+ tendon resident macrophages and their potential crosstalk with fibroblasts and the ECM during tendon growth and maturation. Front. Physiol. 2023, 14, 1122348. [Google Scholar] [CrossRef] [PubMed]
- Lehner, C.; Spitzer, G.; Gehwolf, R.; Wagner, A.; Weissenbacher, N.; Deininger, C.; Emmanuel, K.; Wichlas, F.; Tempfer, H.; Traweger, A. Tenophages: A novel macrophage-like tendon cell population expressing CX3CL1 and CX3CR1. Dis. Model. Mech. 2019, 12, dmm041384. [Google Scholar] [CrossRef]
- Nissinen, L.M.; Kähäri, V.-M. Collagen Turnover in Wound Repair—A Macrophage Connection. J. Investig. Dermatol. 2015, 135, 2350–2352. [Google Scholar] [CrossRef]
- Muscat, S.; Nichols, A.E.C.; Gira, E.; Loiselle, A.E. CCR2 is expressed by tendon resident macrophage and T cells, while CCR2 deficiency impairs tendon healing via blunted involvement of tendon-resident and circulating monocytes/macrophages. FASEB J. 2022, 36, e22607. [Google Scholar] [CrossRef]
- Liu, X.; Zhu, B.; Li, Y.; Liu, X.; Guo, S.; Wang, C.; Li, S.; Wang, D. The Role of Vascular Endothelial Growth Factor in Tendon Healing. Front. Physiol. 2021, 12, 766080. [Google Scholar] [CrossRef]
- Merkel, M.F.R.; Hellsten, Y.; Magnusson, S.P.; Kjaer, M. Tendon blood flow, angiogenesis, and tendinopathy pathogenesis. Transl. Sports Med. 2021, 4, 756–771. [Google Scholar] [CrossRef]
- Ackermann, P.W.; Franklin, S.L.; Dean, B.J.F.; Carr, A.J.; Salo, P.T.; Hart, D.A. Neuronal pathways in tendon healing and tendinopathy—Update. Front. Biosci. 2014, 19, 1251–1278. [Google Scholar] [CrossRef]
- Steinmann, S.; Pfeifer, C.G.; Brochhausen, C.; Docheva, D. Spectrum of Tendon Pathologies: Triggers, Trails and End-State. Int. J. Mol. Sci. 2020, 21, 844. [Google Scholar] [CrossRef]
- Fu, S.-C.; Rolf, C.; Cheuk, Y.-C.; Lui, P.P.; Chan, K.-M. Deciphering the pathogenesis of tendinopathy: A three-stages process. Sports Med. Arthrosc. Rehabil. Ther. Technol. 2010, 2, 30. [Google Scholar] [CrossRef] [PubMed]
- Abate, M.; Silbernagel, K.G.; Siljeholm, C.; Di Iorio, A.; de Amicis, D.; Salini, V.; Werner, S.; Paganelli, R. Pathogenesis of tendinopathies: Inflammation or degeneration? Arthritis Res. Ther. 2009, 11, 235. [Google Scholar] [CrossRef]
- Pandey, V.; Suman, C.P.; Sharma, S.; Rao, S.P.; Kiran Acharya, K.V.; Sambaji, C. Mucoid degeneration of the anterior cruciate ligament: Management and outcome. Indian J. Orthop. 2014, 48, 197–202. [Google Scholar] [CrossRef] [PubMed]
- Tempfer, H.; Traweger, A. Tendon Vasculature in Health and Disease. Front. Physiol. 2015, 6, 330. [Google Scholar] [CrossRef] [PubMed]
- Kannus, P.; Natri, A. Etiology and pathophysiology of tendon ruptures in sports. Scand. J. Med. Sci. Sports 1997, 7, 107–112. [Google Scholar] [CrossRef]
- Xergia, S.A.; Tsarbou, C.; Liveris, N.I.; Hadjithoma, Μ.; Tzanetakou, I.P. Risk factors for Achilles tendon rupture: An updated systematic review. Phys. Sportsmed. 2023, 51, 506–516. [Google Scholar] [CrossRef]
- Persson, R.; Jick, S. Clinical implications of the association between fluoroquinolones and tendon rupture: The magnitude of the effect with and without corticosteroids. Br. J. Clin. Pharmacol. 2019, 85, 949–959. [Google Scholar] [CrossRef]
- Shu, Y.; Zhang, Q.; He, X.; Liu, Y.; Wu, P.; Chen, L. Fluoroquinolone-associated suspected tendonitis and tendon rupture: A pharmacovigilance analysis from 2016 to 2021 based on the FAERS database. Front. Pharmacol. 2022, 13, 990241. [Google Scholar] [CrossRef]
- Morales, D.R.; Slattery, J.; Pacurariu, A.; Pinheiro, L.; McGettigan, P.; Kurz, X. Relative and Absolute Risk of Tendon Rupture with Fluoroquinolone and Concomitant Fluoroquinolone/Corticosteroid Therapy: Population-Based Nested Case-Control Study. Clin. Drug Investig. 2019, 39, 205–213. [Google Scholar] [CrossRef]
- Hall, M.M.; Finnoff, J.T.; Smith, J. Musculoskeletal complications of fluoroquinolones: Guidelines and precautions for usage in the athletic population. PM R 2011, 3, 132–142. [Google Scholar] [CrossRef]
- Waters, T.L.; Ross, B.J.; Wilder, J.H.; Cole, M.W.; Collins, L.K.; Sherman, W.F. Is Fluoroquinolone Exposure after Primary Tendon Repair Associated with Higher Rates of Reoperations? A Matched Cohort Study. Orthop. Rev. 2023, 15, 67914. [Google Scholar] [CrossRef] [PubMed]
- Akali, A.U.; Niranjan, N.S. Management of bilateral Achilles tendon rupture associated with ciprofloxacin: A review and case presentation. J. Plast. Reconstr. Aesthet. Surg. 2008, 61, 830–834. [Google Scholar] [CrossRef] [PubMed]
- Bennett, A.C.; Bennett, C.L.; Witherspoon, B.J.; Knopf, K.B. An evaluation of reports of ciprofloxacin, levofloxacin, and moxifloxacin-association neuropsychiatric toxicities, long-term disability, and aortic aneurysms/dissections disseminated by the Food and Drug Administration and the European Medicines Agency. Expert Opin. Drug Saf. 2019, 18, 1055–1063. [Google Scholar] [CrossRef] [PubMed]
- Sendzik, J.; Shakibaei, M.; Schäfer-Korting, M.; Stahlmann, R. Fluoroquinolones cause changes in extracellular matrix, signalling proteins, metalloproteinases and caspase-3 in cultured human tendon cells. Toxicology 2005, 212, 24–36. [Google Scholar] [CrossRef]
- Tsai, W.-C.; Hsu, C.-C.; Chen, H.-C.; Hsu, Y.-H.; Lin, M.-S.; Wu, C.-W.; Pang, J.-H.S. Ciprofloxacin-mediated inhibition of tenocyte migration and down-regulation of focal adhesion kinase phosphorylation. Eur. J. Pharmacol. 2009, 607, 23–26. [Google Scholar] [CrossRef]
- Tsai, W.-C.; Hsu, C.-C.; Tang, F.-T.; Wong, A.M.K.; Chen, Y.-C.; Pang, J.-H.S. Ciprofloxacin-mediated cell proliferation inhibition and G2/M cell cycle arrest in rat tendon cells. Arthritis Rheum. 2008, 58, 1657–1663. [Google Scholar] [CrossRef]
- Menon, A.; Pettinari, L.; Martinelli, C.; Colombo, G.; Portinaro, N.; Dalle-Donne, I.; d’Agostino, M.C.; Gagliano, N. New insights in extracellular matrix remodeling and collagen turnover related pathways in cultured human tenocytes after ciprofloxacin administration. Muscles Ligaments Tendons J. 2013, 3, 122–131. [Google Scholar]
- Bai, Z.-L.; Chen, Q.; Yang, S.-D.; Zhang, F.; Wang, H.-Y.; Yang, D.-L.; Ding, W.-Y. Toxic effects of levofloxacin on rat annulus fibrosus cells: An in-vitro study. Med. Sci. Monit. 2014, 20, 2205–2212. [Google Scholar] [CrossRef]
- Tsai, W.-C.; Hsu, C.-C.; Chen, C.P.C.; Chang, H.-N.; Wong, A.M.K.; Lin, M.-S.; Pang, J.-H.S. Ciprofloxacin up-regulates tendon cells to express matrix metalloproteinase-2 with degradation of type I collagen. J. Orthop. Res. 2011, 29, 67–73. [Google Scholar] [CrossRef]
- Lowes, D.A.; Wallace, C.; Murphy, M.P.; Webster, N.R.; Galley, H.F. The mitochondria targeted antioxidant MitoQ protects against fluoroquinolone-induced oxidative stress and mitochondrial membrane damage in human Achilles tendon cells. Free Radic. Res. 2009, 43, 323–328. [Google Scholar] [CrossRef]
- Corps, A.N.; Harrall, R.L.; Curry, V.A.; Hazleman, B.L.; Riley, G.P. Contrasting effects of fluoroquinolone antibiotics on the expression of the collagenases, matrix metalloproteinases (MMP)-1 and -13, in human tendon-derived cells. Rheumatology 2005, 44, 1514–1517. [Google Scholar] [CrossRef] [PubMed]
- Yoon, J.H.; Brooks, R.L.; Khan, A.; Pan, H.; Bryan, J.; Zhang, J.; Budsberg, S.C.; Mueller, P.O.E.; Halper, J. The effect of enrofloxacin on cell proliferation and proteoglycans in horse tendon cells. Cell Biol. Toxicol. 2004, 20, 41–54. [Google Scholar] [CrossRef] [PubMed]
- Yoon, J.H.; Brooks, R.L.; Zhao, J.Z.; Isaacs, D.; Halper, J. The effects of enrofloxacin on decorin and glycosaminoglycans in avian tendon cell cultures. Arch. Toxicol. 2004, 78, 599–608. [Google Scholar] [CrossRef] [PubMed]
- Pouzaud, F.; Bernard-Beaubois, K.; Thevenin, M.; Warnet, J.-M.; Hayem, G.; Rat, P. In vitro discrimination of fluoroquinolones toxicity on tendon cells: Involvement of oxidative stress. J. Pharmacol. Exp. Ther. 2004, 308, 394–402. [Google Scholar] [CrossRef]
- Corps, A.N.; Curry, V.A.; Harrall, R.L.; Dutt, D.; Hazleman, B.L.; Riley, G.P. Ciprofloxacin reduces the stimulation of prostaglandin E2 output by interleukin-1β in human tendon-derived cells. Rheumatology 2003, 42, 1306–1310. [Google Scholar] [CrossRef]
- Williams, R.J.; Attia, E.; Wickiewicz, T.L.; Hannafin, J.A. The effect of ciprofloxacin on tendon, paratenon, and capsular fibroblast metabolism. Am. J. Sports Med. 2000, 28, 364–369. [Google Scholar] [CrossRef]
- Kempka, G.; Ahr, H.J.; Rüther, W.; Schlüter, G. Effects of fluoroquinolones and glucocorticoids on cultivated tendon cells in vitro. Toxicol. In Vitro 1996, 10, 743–754. [Google Scholar] [CrossRef]
- Salimiaghdam, N.; Singh, L.; Schneider, K.; Chwa, M.; Atilano, S.R.; Nalbandian, A.; Limb, G.A.; Kenney, M.C. Effects of fluoroquinolones and tetracyclines on mitochondria of human retinal MIO-M1 cells. Exp. Eye Res. 2021, 214, 108857. [Google Scholar] [CrossRef]
- Lang, L.; Zhang, Y.; Yang, A.; Dong, J.; Li, W.; Zhang, G. Macrophage polarization induced by quinolone antibiotics at environmental residue level. Int. Immunopharmacol. 2022, 106, 108596. [Google Scholar] [CrossRef]
- Bezwada, P.; Clark, L.A.; Schneider, S. Intrinsic cytotoxic effects of fluoroquinolones on human corneal keratocytes and endothelial cells. Curr. Med. Res. Opin. 2008, 24, 419–424. [Google Scholar] [CrossRef]
- Simonin, M.A.; Gegout-Pottie, P.; Minn, A.; Gillet, P.; Netter, P.; Terlain, B. Pefloxacin-induced achilles tendon toxicity in rodents: Biochemical changes in proteoglycan synthesis and oxidative damage to collagen. Antimicrob. Agents Chemother. 2000, 44, 867–872. [Google Scholar] [CrossRef] [PubMed]
- Fox, A.J.S.; Schär, M.O.; Wanivenhaus, F.; Chen, T.; Attia, E.; Binder, N.B.; Otero, M.; Gilbert, S.L.; Nguyen, J.T.; Chaudhury, S.; et al. Fluoroquinolones impair tendon healing in a rat rotator cuff repair model: A preliminary study. Am. J. Sports Med. 2014, 42, 2851–2859. [Google Scholar] [CrossRef] [PubMed]
- Kashida, Y.; Kato, M. Characterization of fluoroquinolone-induced Achilles tendon toxicity in rats: Comparison of toxicities of 10 fluoroquinolones and effects of anti-inflammatory compounds. Antimicrob. Agents Chemother. 1997, 41, 2389–2393. [Google Scholar] [CrossRef] [PubMed]
- Kaleagasioglu, F.; Olcay, E. Fluoroquinolone-induced tendinopathy: Etiology and preventive measures. Tohoku J. Exp. Med. 2012, 226, 251–258. [Google Scholar] [CrossRef] [PubMed]
- Zargar Baboldashti, N.; Poulsen, R.C.; Franklin, S.L.; Thompson, M.S.; Hulley, P.A. Platelet-rich plasma protects tenocytes from adverse side effects of dexamethasone and ciprofloxacin. Am. J. Sports Med. 2011, 39, 1929–1935. [Google Scholar] [CrossRef]
Generation | FQ | Characteristics |
---|---|---|
Generation 1 | Nalidixic acid Oxolinic acid | Used for only urinary tract infections Not active against Gram-positive bacteria No anti-Pseudomonas activity |
Generation 2 | Ciprofloxacin (CPX) Enrofloxacin (ENR) Norfloxacin (NOR) Ofloxacin (OFX) Pefloxacin (PFX) Fleroxacin (FLX) | Amplified effectiveness against Gram-negative bacteria First generation that targets Gram-positive bacteria Better tissue penetration Longer half-life |
Generation 3 | Levofloxacin (LFX) Sparfloxacin (SPX) | Enables once-a-day dosing Reduced toxicity against the nervous system |
Generation 4 | Gatifloxacin Moxifloxacin (MOX) | Overall increased activity Potent against anaerobes |
Bacteria | Gram +/− | Disease |
---|---|---|
Bacillus anthracis | + | Anthrax |
Enterococcus faecalis | + | Urinary tract infections |
Helicobacter pylori | − | Salmonellosis |
Klebsiella pneumoniae | − | Pneumonia |
Legionella pneumophilia | − | Legionnaires’ disease |
Mycobacterium tuberculosis | + | Tuberculosis |
Neisseria gonorrhoeae | − | Gonorrhea |
Cell Types | Species | Identification Methods | Characteristics and Markers | Study |
---|---|---|---|---|
Mkx+ or Platelet Derived Growth Factor Receptor Alpha (PDGFRA+) fibroblasts | Human | Single nucleus RNA sequencing, spatial transcriptomics, immunofluorescence staining, Masson’s trichrome staining, hematoxylin, and eosin staining | • Mkx+ Cells: Expression of TNMD and thrombospondin 4. Present in the entire hamstring Markers: MMAFB, ZFHX3 • PDGFRA+ Cells: High expression of neuronal growth regulator 1 and fibrillin. Increased elastin fiber formation. More abundant in areas closer to skeletal muscle Markers: HOX2, TCF7L2 | Mimpen et al., 2024 [61] |
Intrafascicular CD146+ tendon cells in healthy tendons | Horse | Network-based interaction predictions, immunolabeling, fluorescent labeling, 3D immunolabeling, FACS, immunohistochemistry, clonogenic, adipogenesis, and osteogenesis assays | No stem-cell-like characteristics, migration towards injury sites, capable of lipid production Markers: CD146, CD44 | Marr et al., 2023 [58] |
Tendon fibroblast 1 and tendon fibroblast 2 | Rat | Spatial transcriptomics | • Tendon fibroblast 1: Positioned in the central tendon and are responsible for COL1A1 deposition Markers: Comp, Cilp2, Dcn, Col1a1 • Tendon fibroblast 2: Located in the area near the tendon–bone connection and is responsible for the deposition of collagen that envelops the tendon Markers: Apoe1, Col3a11, Cfd2, Tmsb4x2 | Steffen et al., 2023 [62] |
TSPC-0 to TSPC-7 | Human | Wound healing assay, migration assay, three-way differentiation capacity evaluation, transwell assay, transposase-accessible chromatin sequencing, single-cell RNA sequencing | • TSPC-0: Subtype that senses inflammation Marker: AKR1C1 • TSPC-1: Capable of migration Markers: STC2, HMGA1 • TSPC-2: Associated with fibrosis Markers: SLIT3, LUM • TSPC-3: Capable of proliferation Marker: MK167 • TSPC-4: Expression of anti-inflammatory factors Marker: FABP5 • TSPC-5: Dominant in tendinopathy samples, adipogenic differentiation regulators expressed Markers: ADIRF, CRABP2 • TSPC-6: Important for ECM modeling Marker: MXRA5 • TSPC-7: Lacks tendon healing-related genes and related to halted cell migration Markers: MALAT1, MEG3 | Guo et al., 2023 [56] |
Tenocytes 1–10 (TC1-10), macrophages, endothelial cell subpopulations 1–5 | Human | Immunohistochemistry, multiplex immunofluorescence staining, single-cell RNA sequencing | • TC1-3: Dominant in healthy tendons, TC1 cells are normal fibroblasts, TC2 express high levels of ECM components and proliferation regulators, TC3 cells can act anti-inflammatory Markers: COL1A1, COL3A1, and PDGFRB (TC1), MEG3, COL1A2 (TC2), PLPP3, PLA2G2A (TC3) • TC4 and 5: Attachment of tendons to the bones Markers: HAS1, PRG4 (TC5) MYOC and IGFBP6 (TC5) • TC6: Inflammatory fibroblasts Markers: PTGFR, SAA1 • TC7: Expression of genes related to tendon repair Markers: TPP3, COL3A1, and COL5A1 • TC8-9 are chondrocytes. TC10 can be classified as osteocytes Markers: CRLF1 and CD55 (TC8), SOX9, OGN (TC9), SPP1, IBSP (TC10) | Fu et al., 2023 [52] |
Synthetic, native, reactive, fibrotic, inflammatory, and muscle-associated adult Scx-expressing populations in post-repair tendons | Mouse | Histology, immunofluorescence, spatial RNA sequencing, pseudotemporal ordering and lineage trajectories, cell–cell interactome analysis | • Synthetic population: Involved in ECM and collagen fibril organization Markers: Tnmd, Fmod, and Col1a1 • Native population: Found in areas distant from tissue injury Markers: Musculoskeletal markers • Reactive population: Mostly near the injury site, associated with higher collagen catabolism, and an increase in adhesion, migration, and proliferation Markers: Higher expression Mmp13 and Lox • Fibroblastic population: High levels of ECM synthesis Markers: Col3a1, Postn Inflammatory population: Expression of markers more similar to immune cells such as macrophages Markers: Saa3 and alarmins • Muscle-associated population: Characteristics Marker: Mb | Ackermann et al., 2022 [59] |
Paratenon TSPC | Human | Histochemical staining, electron microscopy, immunostaining, immunohistochemical analysis | • Expression of smooth muscle actin Markers: CD105 and CD146 | Zhang et al., 2021 [63] |
High nestin (NES+) clonogenic TSPCs, Solute carrier family 40 member 1 (SLC40A1)+ TSPCs, pro-inflammatory TSPCs (piTSPC) | Human | Drop-sequencing test, 10X chromium test on stretched cells, mitotracker staining, qPCR | • piTSPCs: Most likely involved in inflammation-mediated development of tendinopathy Markers: IL6, IL8, and CXCL1 • SLC40A1+ TSPCs: Involved in tendon development and repair due to their higher expression of TGFs Markers: TGFB2, TGFB3 • NES+ clonogenic TSPC Markers: MKI67, TOP2A | Still et al., 2021 [57] |
Interfascicular TSPCs | Human | Immunolabeling, confocal imaging, qPCR of cell culture serum | • Possible higher expression of CD146 and higher accumulation in injury sites in some TSPCs Marker: CD146 | Marr et al., 2021 [64] |
Tenocytes A–E present in diseased and healthy tendons Monocytes and endothelial cells in Achilles tendon | Human | Single-cell RNA sequencing, histology | • Tenocyte A–B: Expression of extracellular tendon microfibrils Tenocyte A Markers: CD10, CD26 and COL6A3, LY6E Tenocyte B Markers: CD90, COL4A1, and POSTN • Tenocyte C: Similar to smooth muscle mesenchymal cells Markers: MYL9, ACTA2 • Tenocyte D–E: Similar to fibro-adipogenic progenitors Tenocyte D Markers: COL6A1, COL6A2, and COL3A1 Tenocyte E Markers: TPPP3, DCN, and FMOD | Kendal et al., 2020 [51] |
Junctional fibroblasts and tendon fibroblasts 1/2 in Achilles tendon | Mouse | Single-cell RNA sequencing, cell trajectory analysis, FACS, histology | • Fibroblast 1: Expression of connective tissue growth factor Markers: Mkx, Spp1 • Fibroblast 2: Differential SPARC Related Modular Calcium Binding 2 (SMOC2) expression Markers: Dpt, Smoc2 • Junctional Fibroblast: Moderate expression of transcripts for collagen types I and Markers: Col22a1 | Micheli et al., 2020 [65] |
Four macrophage clusters termed 1,3,4, and 7 during tendon injury | Mouse | FACS, immunofluorescence, immunohistochemistry, in vivo bioluminescence, single-cell RNA sequencing | • Cluster 1: Tendon resident macrophages Markers: Cd163, Timd4, Tgfb1 • Clusters 3 and 4: Expression of M2 markers Markers: CD206 and Arg1, Tgfb1 • Cluster 7: Less expression of M2 markers Markers: IL-1b, Ccl4, Erg1 | Sorkin et al., 2020 [66] |
Tubulin Polymerization Promoting Protein Family Member 3 (Tppp3)+ Pdgfra+ and Tppp3+Pdgfra− subpopulations, macrophages, endothelial cells | Mouse | Immunofluorescence, 10X genomics scRNA-sequencing, FACS, histology, in vitro characterization, qPCR | • Tppp3+Pdgfra+ Cells: Can act as tendon stem cells • Tppp3+Pdgfra− Cells: No proliferation and most likely differentiation into Scx− cells found in the healing tendon | Harvey et al., 2019 [67] |
Nes+ perivascular TSPCs | Human | RNA interference for Nes inhibition, qPCR, microarray analysis | • Increased expression of Nes and expression of tendon-related genes and stem cell markers Markers: Nes, CD146, CD105 | Yin et al., 2016 [68] |
CD105+ and CD105- TSPCs in injured Achilles tendon | Mouse | Histology, immunoblotting | • CD105- cells: Superior chondrogenic potential Markers: Aggrecan, TGF-β1 • CD105+ cells: Superior proliferation, important for tendon regeneration Markers: High Scx levels | Asai et al., 2014 [69] |
Young and aged TSPCs | Human | FACS, immunohistochemistry, genome-wide microarray, qPCR, senescence analysis, migration experiments | • Young TSPCs: Higher proliferative and migratory capability, multipotent, low F-actin fibers • Aged TSCPs: Less proliferative and migratory, still capable of multilineage differentiation, abundant F-actin fibers, increased senescence, higher ROCK1 activity Markers: microarray identified ca. 1000 differentially expressed genes; in top dysregulated ones, change in genes related to actin dynamics, cell–cell and cell–ECM contact, and migration | Kohler et al., 2013 [54] |
TSPCs | Mouse Human | Cell labeling, Western blotting, FACS, qPCR, luciferase reporter assays | • Both mouse and human cells reside mostly in ECM protein-rich regions, with no CD18 expression Mouse Markers: Stem cell antigen marker 1, CD90 Human Markers: CD44, CD90, and CD146 | Bi et al., 2007 [70] |
Goals | Cell Types, Treatments, and Assays | Results | Studies |
---|---|---|---|
Studying effects of uremic toxins with or without CPX on metabolic activity, vitality, and collagen I expression | • Cells: Human tenocytes isolated from the hamstring tendon • Treatment: Serum-starved tenocytes in 1% FCS for 24 h before incubation with 3, 10, 30, 50, and 100 mg/L with CPX • Assays: Alamar Blue, qPCR for MMP1, IL1β and β1-integrin expression, Western blot for collagen type I detection | CPX suppressed tenocyte activity after short exposure times at higher concentrations after 72 h and this was also observed at therapeutic concentrations. MMP1 mRNA levels increased, and the protein levels of type I collagen decreased. No significant alteration in β1-integrin expression. | Popowski et al., 2020 [27] |
To determine the toxicity of LFX on annulus fibrosus cells | • Cells: Rat annulus fibrosis cells • Treatment: LFX with 10, 20, 40, and 80 μg/mL for cell viability and caspase-3 activity assay and 30, 60, and 90 μg/mL for Annexin staining • Assays: Annexin V-FITC/PI staining and caspase-3 activity for apoptosis, MTS for cell viability, Western blot for caspase 3 and MMP3 detection, qPCR for caspase and MMP3 expression levels | LFX concentrations at 30, 60, and 90 μg/mL led to cell apoptosis caused by higher caspase-3 and MMP3 expression and activity. | Bai et al., 2014 [98] |
Exploring the impact of CPX on human ligamentocytes in vitro by morphological and molecular methods | • Cells: Cells from the human anterior cruciate ligament • Treatment: 10, 20, and 50 μg/mL CPX for 48 h • Assays: qPCR for collagen I/III, slot blot for Collagen I/II and MMP1, SDS-zymography for MMP activity, fluorescence microscopy for tubulin detection, and cytoskeleton analysis | Collagen mRNA levels were not affected. MMP1 protein levels rose upon 20 μg/mL CPX treatment only. MMP2 and TGF-β1 levels were unmodified. TIMP-1 expression was downregulated. CPX caused no changes in the cytoskeleton. | Menon et al., 2013 [97] |
Investigating the alterations in MMP2/9, TIMP1/2, and collagen I expression levels after CPX use | • Cells: Tendon cells from Sprague–Dawley rats • Treatment: 5, 10, 20, or 50 µg/mL CPX for 24 h • Assays: MTT, qPCR for collagen I and MMP2, Western blot for MMP2 and collagen I protein levels, zymography for determining MMP2 and MMP9 levels in medium, reverse zymography for TIMP1/2 levels | CPX reduced viability and increased MMP2 expression and activity in a dose-dependent manner. In contrast, MMP9 expression was not affected. No change in TIMP activity was determined. | Tsai et al., 2011 [99] |
Studying the effects of CPX on tenocyte migration | • Cells: Tenocytes from male rat tendons • Treatment: Cells incubated with 5, 10, 20, and 50 μg/mL CPX for 24 h • Assays: Transwells for migration, microscopy to determine tenocyte spreading, Western blot for focal adhesion kinase (FAK) levels and phosphorylation | CPX application led to dose-dependent inhibition of cell growth compared to control. Slow cell migration was possibly due to halted lamellipodium formation in CPX-treated cells. Total FAK expression was not altered, but phosphorylation levels were lowered. | Tsai et al., 2009 [95] |
Determining if MOX and CPX cause oxidative stress in vitro in tendon cells, and investigating the protective activity of idebenone (part of the antioxidant MitoQ complex) | • Cells: Tenocytes of normal human Achilles tendons • Treatment: Cells exposed to 0–0.3 mM of CPX or MOX after 24 h • Assays: JC-1 assay for mitochondrial membrane potential, 5-(6)-carboxy-2,7′ dichlorodihydrofluorescein-diacetate for oxidative stress | Increasing concentrations of CPX or MOX damaged the mitochondrial membrane and resulted in decreased membrane potential. Oxidative stress rose after treatment. MitoQ application prevented membrane damage. | Lowes et al., 2009 [100] |
Investigating the effect of CPX on the cell cycle of tendon cells and expression of cyclin-dependent kinase (CDK) | • Cells: Rat Achilles tendon cells • Treatment: For MTT assay and cell cycle analysis, 5, 10, 20, or 500 µg/mL CPX was given for 24 h. For immunofluorescence and Western blot: 50 µg/mL CPX for 24 h • Assays: MTT, FACS for cell cycle analysis, PCR for CDK-1, Western blot for (p) CDK1, cyclin B, and checkpoint kinase 1 | CPX application lowered cell viability and stopped the cell cycle at the G2/M phase. This is due to condensed chromosomes not properly aligning during replication. Microtubules were also misaligned. (p)CDK-1 and cyclin B expression were lowered. | Tsai et al., 2008 [96] |
Examining CPX and IL-1β’s effects on MMP13 expression levels | • Cells: Human Achilles tendon cells • Treatment: 48 h initial incubation with FQs, followed by 48 h supplementation with or without IL-1β together with FQs. • Assays: qPCR for MMP1/13 expression, fluorokine E fluorimetry for MMP1/13 activity measurement | CPX treatment lowered MMP13 expression levels. This effect was stronger when CPX was supplemented together with IL-1β. CPX and NOR enhanced MMP1 expression with or without IL-1β addition. ENR decreased cell count in a dose-dependent manner. | Corps et al., 2005 [101] |
Determining ENR-mediated changes on equine superficial digital flexor tendons | • Cells: Cells from superficial digital flexor tendons of foals and older horses • Treatment: Cells exposed to 50 or 100 μg/mL ENR and incubated for 3 days • Assays: Trypan blue for viability, gel zymography for MMP1 expression, TACS Apoptotic DNA laddering for apoptosis, Western and northern blots for decorin and biglycan expression, gas chromatography and mass spectrometry for monosaccharide content determination | Cells from the younger horses were more susceptible to membrane perforations and apoptosis at lower concentrations. No difference in the expression of MMPs, DNA fragmentation, or biglycan mRNA levels was observed. Monosaccharide content decreased by 50%, and decorin amount increased. | Yoon et al., 2004 [102] |
Studying the effect of ENR on cell proliferation and proteoglycan synthesis in avian tenocytes | • Cells: Gastrocnemius tendon cells isolated from chicken embryos • Treatment: 25, 50, 100, and 300 μg/mL ENR given for 3 days for proliferation assay. For the apoptosis test, only 50 μg/mL was used. The cells were given 20 or 100 μg/mL ENR for gel zymography • Assays: Trypan blue for cell proliferation measurement, gel zymography for proteolytic activity, Western and northern blotting, electron microscopy, TACS Apoptotic DNA Laddering Kit, Western blotting for decorin detection | Cell number was reduced 3-fold under treatment with 100 and 300 μg ENR. ENR led to the formation of tightly woven collagen fibrils in the ECM. Decorin mRNA levels were lowered after 72 h, and protein levels decreased dose-dependently. Monosaccharide levels were lower and composition was also altered. | Yoon et al., 2004 [103] |
Investigating the genotoxicity potential and mechanism of PFX, LFX, CPX, and OFX | • Cells: Spontaneously immortalized clones of tenocytes from rabbit Achilles tendon • Treatment: 0.01 μM, 0.1 μM, 1 μM, 10 μM, 0.1 mM, and 1 mM FQs given for 24 h or 72 h • Assays: Neutral red, AlamarBlue for determining redox status, monobromobimane as glutathione test | All FQs were mildly toxic to tenocytes, the toxicity was higher after 72 h. CPX and PFX treatment decreased cell viability and redox status in the highest concentration after 72 h. Meanwhile, ROS levels increased at higher concentrations (>10 μM CPX). OFX and LFX caused a mild redox activity decrease. | Pouzaud et al., 2004 [104] |
Exploring the effects of CPX on signaling responses of tendon cells | • Cells: Tendon cells from a chronic human Achilles tendinopathy sample • Treatment: Initial application of 50 µg/mL CPX, afterwards 1 ng/mL IL.1ß was given to cells After 2 days, medium supplemented with either IL-1β, CPX, or both • Assays: Prostaglandin E2 ELISA, Western blot for cyclooxygenase-2 levels | IL-1β treatment heightened PGE expression, whilst CPX lowered it. In contrast, CPX did not affect the cyclooxygenase-2 expression levels even when IL-1β was present. | Corps et al., 2003 [105] |
Studying the impact of different CPX concentrations on Achilles tendon/paratenon and shoulder capsule fibroblast function | • Cells: Fibroblasts from canine Achilles tendon, Achilles paratenon, and shoulder capsule • Treatment: 5 μg/mL, 10 μg/mL, or 50 μg/mL CPX • Assays: MTT and Coulter cell counter for cell number determination, H-proline incorporation for collagen synthesis, sulfate incorporation for proteoglycan measurement, radiolabeled casein for matrix degradation quantification | CPX supplementation caused increased matrix-degrading activity, decreased matrix synthesis, and reduced cell proliferation in each cell population. Collagen synthesis was significantly lowered. CPX was most toxic at 50 μg/mL concentration. | Williams et al., 2000 [106] |
Determining the influence of FQs on tendon cells from various species and describing age-dependency | • Cells: Human Achilles tendon samples (ages 3 months to 79 years), dogs, mini-pigs, rats, marmosets • Treatment: 3 regimens (all 72 h duration): FQ+ triamcinolone acetonide (a common corticosteroid used for skin conditions), FQ treatment, then 3 days rest • Assays: Neutral red and MTT for cell viability, 5-bromo-2′-deoxyuridine for cell proliferation, collagen I immunofluorescence | CPX-treated cells showed no age or species-specific effects. The cytotoxicity of CPX was increased when combined with corticosteroids. | Kempka et al., 1996 [107] |
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content. |
© 2025 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).
Share and Cite
Duman, E.; Müller-Deubert, S.; Pattappa, G.; Stratos, I.; Sieber, S.A.; Clausen-Schaumann, H.; Sarafian, V.; Shukunami, C.; Rudert, M.; Docheva, D. Fluoroquinolone-Mediated Tendinopathy and Tendon Rupture. Pharmaceuticals 2025, 18, 184. https://doi.org/10.3390/ph18020184
Duman E, Müller-Deubert S, Pattappa G, Stratos I, Sieber SA, Clausen-Schaumann H, Sarafian V, Shukunami C, Rudert M, Docheva D. Fluoroquinolone-Mediated Tendinopathy and Tendon Rupture. Pharmaceuticals. 2025; 18(2):184. https://doi.org/10.3390/ph18020184
Chicago/Turabian StyleDuman, Ezgi, Sigrid Müller-Deubert, Girish Pattappa, Ioannis Stratos, Stephan A. Sieber, Hauke Clausen-Schaumann, Victoria Sarafian, Chisa Shukunami, Maximilian Rudert, and Denitsa Docheva. 2025. "Fluoroquinolone-Mediated Tendinopathy and Tendon Rupture" Pharmaceuticals 18, no. 2: 184. https://doi.org/10.3390/ph18020184
APA StyleDuman, E., Müller-Deubert, S., Pattappa, G., Stratos, I., Sieber, S. A., Clausen-Schaumann, H., Sarafian, V., Shukunami, C., Rudert, M., & Docheva, D. (2025). Fluoroquinolone-Mediated Tendinopathy and Tendon Rupture. Pharmaceuticals, 18(2), 184. https://doi.org/10.3390/ph18020184