1
|
Li W, Zhang L, Li Y, Chen Y, Xie Z, Zhuang QK. Clinical Application Study of External Fixation Treatment Using Removed Locking Plates in Patients with Failed Debridement and Antimicrobial Therapy for Infection after Internal Fixation of Fractures. Surg Infect (Larchmt) 2024. [PMID: 39694532 DOI: 10.1089/sur.2024.261] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2024] Open
Abstract
Purpose: The main focus of this study is to investigate the clinical efficacy of external fixation treatment using removed locking plates in patients with failed debridement and antimicrobial therapy of infection after internal fixation of fractures. Patients and Methods: From January 2019 to January 2023, our medical institution treated 13 patients who had failed debridement and antimicrobial therapy for infection after internal fixation of fractures. All patients had their internal fixation devices removed, underwent thorough debridement, and then the removed locking plates were repurposed for external fixation treatment. Post-operatively, we observed the infection control status, fracture healing, and complications following locking plate external fixation. Results: Among all patients, there were seven males and six females, with an average age of 45.92 ± 13.19 years. The time interval from fracture internal fixation surgical procedure to the onset of infection was 7.08 ± 1.89 weeks, and the average debridement frequency was 1.62 ± 0.51 times. After treatment with external fixation using a removed locking plate, infection was controlled in 12 patients, ultimately achieving clinical bone healing. These 12 patients achieved clinical bone healing at a time of 7.50 ± 1.00 months, and the locking plate external fixation time was 9.83 ± 1.11 months. Conclusions: External fixation using removed locking plates appears to be an effective treatment option for patients who have failed debridement and antimicrobial therapy for infection after internal fixation of fractures. This approach effectively controls infection symptoms while not interfering with the fracture healing process. Additionally, this treatment option using a removed locked plate may potentially contribute to reducing patients' medical expenses.
Collapse
Affiliation(s)
- Wei Li
- Department of Orthopedics, No. 2 People's Hospital of Fuyang City, Fuyang City, People's Republic of China
| | - Lei Zhang
- Department of Orthopedics, No. 2 People's Hospital of Fuyang City, Fuyang City, People's Republic of China
| | - Yang Li
- Department of Orthopedics, No. 2 People's Hospital of Fuyang City, Fuyang City, People's Republic of China
| | - Yong Chen
- Department of Orthopedics, No. 2 People's Hospital of Fuyang City, Fuyang City, People's Republic of China
| | - Zhao Xie
- Department of Orthopaedics, First Affiliated Hospital (Southwest Hospital), Army Medical University, Chongqing, People's Republic of China
| | - Quan-Kui Zhuang
- Department of Orthopedics, No. 2 People's Hospital of Fuyang City, Fuyang City, People's Republic of China
| |
Collapse
|
2
|
Wan Y, Jiang G, Shan H, Lin Y, Xia W, Yin F, Jiang C, Shi Z. F-Box and WD repeat domain containing 7 induces infectious osteomyelitis by regulating MYB stability and ubiquitination. Scand J Immunol 2024; 100:e13414. [PMID: 39487565 DOI: 10.1111/sji.13414] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Revised: 08/29/2024] [Accepted: 10/13/2024] [Indexed: 11/04/2024]
Abstract
Osteomyelitis is a bone inflammation initiated by invading pathogens. Macrophages and inflammation play essential roles in osteomyelitis. F-Box and WD repeat domain containing 7 (Fbxw7) is a tumour suppressor and E3 ubiquitin ligase. In the present study, the potential roles of Fbxw7 in osteomyelitis were explored. The mRNA level of Fbxw7 was measured in bone marrow cells from patients with osteomyelitis and Staphylococcus aureus (S. aureus)-infected macrophages. The conditional knockout mice with Fbxw7 deficiency in myeloid cells were generated. The expression of interleukin (IL)-6, IL-23a and nitric oxide synthase 2 (Nos2) was measured in S. aureus-infected Fbxw7-deficient bone marrow-derived macrophages (BMDMs). The body weight loss, bacterial burden, bone loss and formation and serum level of IL-6, IL-23 and TNF-α were measured in S. aureus-infected Fbxw7 conditional KO mice. The interacting partners of Fbxw7 were predicted using STRING and the interaction were tested. Elevated expression of Fbxw7 was observed in bone marrow cells from patients with osteomyelitis and in S. aureus-infected macrophages. The expression of IL-6, IL-23a and Nos2 was remarkably suppressed in S. aureus-infected Fbxw7-deficient BMDMs. Fbxw7 conditional knockout mice had less body weight loss, higher bacterial burden, less bone loss and formation and decreased serum level of cytokines. Fbxw7 interacted with MYB. S. aureus-infected Fbxw7-deficient BMDMs had higher level of MYB and less ubiquitination of MYB. Fbxw7 promotes osteomyelitis symptoms by regulating ubiquitination and stability of MYB.
Collapse
Affiliation(s)
- Yongbo Wan
- Department of Orthopaedic Surgery, Haikou Orthopedic and Diabetes Hospital of Shanghai Sixth People's Hospital, Haikou, Hainan, China
| | - Gehan Jiang
- Department of Orthopaedic Surgery, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Haojie Shan
- Department of Orthopaedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yiwei Lin
- Department of Orthopaedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wenyang Xia
- Department of Orthopaedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Fuli Yin
- Department of Orthopaedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chaolai Jiang
- Department of Orthopaedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhongmin Shi
- Department of Orthopaedic Surgery, Haikou Orthopedic and Diabetes Hospital of Shanghai Sixth People's Hospital, Haikou, Hainan, China
- Department of Orthopaedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
3
|
Yekani M, Dizaj SM, Sharifi S, Sedaghat H, Saffari M, Memar MY. Nano-scaffold-based delivery systems of antimicrobial agents in the treatment of osteomyelitis ; a narrative review. Heliyon 2024; 10:e38392. [PMID: 39559197 PMCID: PMC11570522 DOI: 10.1016/j.heliyon.2024.e38392] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Revised: 09/16/2024] [Accepted: 09/23/2024] [Indexed: 11/20/2024] Open
Abstract
Osteomyelitis caused by drug-resistant pathogens is one of the most important medical challenges due to high rates of mortality and morbidity, and limited therapeutical options. The application of novel nano-scaffolds loaded with antibiotics has widely been studied and extensively evaluated for in vitro and in vivo inhibition of pathogens, regenerating damaged bone tissue, and increasing bone cell proliferation. The treatment of bone infections using the local osteogenic scaffolds loaded with antimicrobial agents may efficiently overcome the problems of the systemic use of antimicrobial agents and provide a controlled release and sufficient local levels of antibiotics in the infected sites. The present study reviewed various nano-scaffolds delivery systems of antimicrobial drugs evaluated to treat osteomyelitis. Nano-scaffolds offer promising approaches because they simulate natural tissue regeneration in terms of their mechanical, structural, and sometimes chemical properties. The potential of several nano-scaffolds prepared by natural polymers such as silk, collagen, gelatin, fibrinogen, chitosan, cellulose, hyaluronic, alginate, and synthetic compounds such as polylactic acid, polyglycolic acid, poly (lactic acid-co-glycolic acid), poly-ɛ-caprolactone have been studied for usage as drug delivery systems of antimicrobial agents to treat osteomyelitis. In addition to incorporated antimicrobial agents and the content of scaffolds, the physical and chemical characteristics of the prepared delivery systems are a determining factor in their effectiveness in treating osteomyelitis.
Collapse
Affiliation(s)
- Mina Yekani
- Department of Microbiology, Faculty of Medicine, Kashan University of Medical Sciences, Kashan, Iran
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
- Dental and Periodontal Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Solmaz Maleki Dizaj
- Dental and Periodontal Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Simin Sharifi
- Dental and Periodontal Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hossein Sedaghat
- Department of Microbiology, Faculty of Medicine, Kashan University of Medical Sciences, Kashan, Iran
| | - Mahmood Saffari
- Department of Microbiology, Faculty of Medicine, Kashan University of Medical Sciences, Kashan, Iran
| | - Mohammad Yousef Memar
- Infectious and Tropical Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
4
|
Duda-Madej A, Viscardi S, Pacyga K, Kupczyński R, Mączka W, Grabarczyk M, Pacyga P, Topola E, Ostrówka M, Bania J, Szumny A, Wińska K. Antibiofilm and Antimicrobial Potentials of Novel Synthesized Sulfur Camphor Derivatives. Int J Mol Sci 2024; 25:10895. [PMID: 39456678 PMCID: PMC11507198 DOI: 10.3390/ijms252010895] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 09/19/2024] [Accepted: 10/07/2024] [Indexed: 10/28/2024] Open
Abstract
The question being posed by scientists around the world is how different chemical modifications of naturally occurring compounds will affect their antimicrobial properties. In the current study, sulfur derivatives of camphor containing a sulfur atom were tested to detect their antimicrobial and antibiofilm potentials. The new compounds were tested on eight Gram-positive strains (S. aureus (3 isolates), S. epidermidis (4 isolates), and E. faecalis (1 isolate)) and eight Gram-negative strains (E. coli (6 isolates), A. baumannii (1 isolate), and P. aeruginosa (1 isolate)). The ability of the strains to eradicate a biofilm was evaluated under standard stationary and flow-through conditions using the Bioflux system. Two synthesized compounds, namely rac-thiocamphor (1a) and (S, S)-(+)-thiocamphor (2a), exhibited an effect on the 24 h biofilm formed by the Gram-positive strains. Our results are an important contribution to the science of natural compounds and allow us to classify our sulfur derivatives of camphor as potential prophylactic agents in treating skin infections, antiseptics, and disinfectants. The Gram-negative strains were excluded from further stages of the tests due to their high activity (MIC ≥ 512 µg/mL). On the other hand, the compound with the strongest antimicrobial activity against the Gram-positive strains was 2a, as it led led to a reductions in cell viability of 17-52% (for MIC), 37-66% (for 2MIC), and 40-94% (for 4MIC). In addition, the experimental retention index of thiocamphor was calculated for the first time.
Collapse
Affiliation(s)
- Anna Duda-Madej
- Department of Microbiology, Faculty of Medicine, Wroclaw Medical University, Chałubińskiego 4, 50-368 Wrocław, Poland
| | - Szymon Viscardi
- Faculty of Medicine, Wroclaw Medical University, Ludwika Pasteura 1, 50-367 Wrocław, Poland; (S.V.)
| | - Katarzyna Pacyga
- Department of Environment Hygiene and Animal Welfare, Faculty of Biology and Animal Science, Wrocław University of Environmental and Life Sciences, 50-375 Wrocław, Poland (R.K.)
| | - Robert Kupczyński
- Department of Environment Hygiene and Animal Welfare, Faculty of Biology and Animal Science, Wrocław University of Environmental and Life Sciences, 50-375 Wrocław, Poland (R.K.)
| | - Wanda Mączka
- Department of Food Chemistry and Biocatalysis, Faculty of Biotechnology and Food Science, Wrocław University of Environmental and Life Sciences, C.K. Norwida 25, 50-375 Wrocław, Poland; (W.M.); (A.S.); (K.W.)
| | - Małgorzata Grabarczyk
- Department of Food Chemistry and Biocatalysis, Faculty of Biotechnology and Food Science, Wrocław University of Environmental and Life Sciences, C.K. Norwida 25, 50-375 Wrocław, Poland; (W.M.); (A.S.); (K.W.)
| | - Paweł Pacyga
- Department of Thermodynamics and Renewable Energy Sources, Faculty of Mechanical and Power Engineering, Wrocław University of Science and Technology, 50-370 Wrocław, Poland;
| | - Ewa Topola
- Faculty of Medicine, Wroclaw Medical University, Ludwika Pasteura 1, 50-367 Wrocław, Poland; (S.V.)
| | - Michał Ostrówka
- Faculty of Biotechnology, University of Wrocław, Fryderyka Joliot-Curie 14a, 50-137 Wrocław, Poland;
| | - Jacek Bania
- Department of Food Hygiene and Consumer Health Protection, Wrocław University of Environmental and Life Sciences, 50-375 Wrocław, Poland
| | - Antoni Szumny
- Department of Food Chemistry and Biocatalysis, Faculty of Biotechnology and Food Science, Wrocław University of Environmental and Life Sciences, C.K. Norwida 25, 50-375 Wrocław, Poland; (W.M.); (A.S.); (K.W.)
| | - Katarzyna Wińska
- Department of Food Chemistry and Biocatalysis, Faculty of Biotechnology and Food Science, Wrocław University of Environmental and Life Sciences, C.K. Norwida 25, 50-375 Wrocław, Poland; (W.M.); (A.S.); (K.W.)
| |
Collapse
|
5
|
Vanvelk N, de Mesy Bentley KL, Verhofstad MHJ, Metsemakers WJ, Moriarty TF, Siverino C. Development of an ex vivo model to study Staphylococcus aureus invasion of the osteocyte lacuno-canalicular network. J Orthop Res 2024. [PMID: 39380444 DOI: 10.1002/jor.25988] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Revised: 09/08/2024] [Accepted: 09/18/2024] [Indexed: 10/10/2024]
Abstract
Staphylococcus aureus has multiple mechanisms to evade the host's immune system and antibiotic treatment. One such mechanism is the invasion of the osteocyte lacuno-canalicular network (OLCN), which may be particularly important in recurrence of infection after debridement and antibiotic therapy. The aim of this study was to develop an ex vivo model to facilitate further study of S. aureus invasion of the OLCN and early-stage testing of antibacterial strategies against bacteria in this niche. The diameter of the canaliculi of non-infected human, sheep, and mouse bones was measured microscopically on Schmorl's picrothionin stained sections, showing a large overlap in canalicular diameter. S. aureus successfully invaded the OLCN in all species in vitro as revealed by presence in osteocyte lacunae in Brown and Brenn-stained sections and by scanning electron microscopy. Murine bones were then selected for further experiments, and titanium pins with either a wild-type or ΔPBP4 mutant S. aureus USA300 were placed trans-cortically and incubated for 2 weeks in tryptic soy broth. Wild-type S. aureus readily invaded the osteocyte lacunae in mouse bones while the ΔPBP4 showed a significantly lower invasion of the OLCN (p = 0.0005). Bone specimens were then treated with gentamicin, sitafloxacin, R14 bacteriophages, or left untreated. Gentamicin (p = 0.0027) and sitafloxacin (p = 0.0280) significantly reduced the proportion of S. aureus-occupied lacunae, whilst bacteriophage treatment had no effect. This study shows that S. aureus is able to invade the OLCN in an ex vivo model. This ex vivo model can be used for future early-stage studies before proceeding to in vivo studies.
Collapse
Affiliation(s)
- Niels Vanvelk
- AO Research Institute Davos, Davos, Switzerland
- Trauma Research Unit, Department of Surgery, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Karen L de Mesy Bentley
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, New York, USA
- Center for Advanced Research Technology (CART), University of Rochester Medical Center Rochester, Rochester, New York, USA
| | - Michael H J Verhofstad
- Trauma Research Unit, Department of Surgery, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Willem-Jan Metsemakers
- Department of Trauma Surgery, University Hospitals Leuven, Leuven, Belgium
- Department of Development and Regeneration, KU Leuven-University of Leuven, Leuven, Belgium
| | - Thomas F Moriarty
- AO Research Institute Davos, Davos, Switzerland
- Center for Muskuloskeletal Infections (ZMSI), University Hospital Basel, Basel, Switzerland
| | | |
Collapse
|
6
|
Giraldo-Osorno PM, Wirsig K, Asa'ad F, Omar O, Trobos M, Bernhardt A, Palmquist A. Macrophage-to-osteocyte communication: Impact in a 3D in vitro implant-associated infection model. Acta Biomater 2024; 186:141-155. [PMID: 39142531 DOI: 10.1016/j.actbio.2024.08.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 07/23/2024] [Accepted: 08/04/2024] [Indexed: 08/16/2024]
Abstract
Macrophages and osteocytes are important regulators of inflammation, osteogenesis and osteoclastogenesis. However, their interactions under adverse conditions, such as biomaterial-associated infection (BAI) are not fully understood. We aimed to elucidate how factors released from macrophages modulate osteocyte responses in an in vitro indirect 3D co-culture model. Human monocyte-derived macrophages were cultured on etched titanium disks and activated with either IL-4 cytokine (anti-inflammatory M2 phenotype) or Staphylococcus aureus secreted virulence factors to simulate BAI (pro-inflammatory M1 phenotype). Primary osteocytes in collagen gels were then stimulated with conditioned media (CM) from these macrophages. The osteocyte response was analyzed by gene expression, protein secretion, and immunostaining. M1 phenotype macrophages were confirmed by IL-1β and TNF-α secretion, and M2 macrophages by ARG-1 and MRC-1.Osteocytes receiving M1 CM revealed bone inhibitory effects, denoted by reduced secretion of bone formation osteocalcin (BGLAP) and increased secretion of the bone inhibitory sclerostin (SOST). These osteocytes also downregulated the pro-mineralization gene PHEX and upregulated the anti-mineralization gene MEPE. Additionally, exhibited pro-osteoclastic potential by upregulating pro-osteoclastic gene RANKL expression. Nonetheless, M1-stimulated osteocytes expressed a higher level of the potent pro-osteogenic factor BMP-2 in parallel with the downregulation of the bone inhibitor genes DKK1 and SOST, suggesting a compensatory feedback mechanisms. Conversely, M2-stimulated osteocytes mainly upregulated anti-osteoclastic gene OPG expression, suggesting an anti-catabolic effect. Altogether, our findings demonstrate a strong communication between M1 macrophages and osteocytes under M1 (BAI)-simulated conditions, suggesting that the BAI adverse effects on osteoblastic and osteoclastic processes in vitro are partly mediated via this communication. STATEMENT OF SIGNIFICANCE: Biomaterial-associated infections are major challenges and the underlying mechanisms in the cellular interactions are missing, especially among the major cells from the inflammatory side (macrophages as the key cell in bacterial clearance) and the regenerative side (osteocyte as main regulator of bone). We evaluated the effect of macrophage polarization driven by the stimulation with bacterial virulence factors on the osteocyte function using an indirect co-culture model, hence mimicking the scenario of a biomaterial-associated infection. The results suggest that at least part of the adverse effects of biomaterial associated infection on osteoblastic and osteoclastic processes in vitro are mediated via macrophage-to-osteocyte communication.
Collapse
Affiliation(s)
- Paula Milena Giraldo-Osorno
- Department of Biomaterials, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Katharina Wirsig
- Faculty of Medicine, Centre for Translational Bone, Joint and Soft Tissue Research, Technische Universität Dresden, Germany
| | - Farah Asa'ad
- Department of Biomaterials, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden; Department of Oral Biochemistry, Institute of Odontology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Omar Omar
- Department of Biomedical Dental Sciences, College of Dentistry, Imam Abdulrahman Bin Faisal University, Dammam, Saudi Arabia
| | - Margarita Trobos
- Department of Biomaterials, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Anne Bernhardt
- Faculty of Medicine, Centre for Translational Bone, Joint and Soft Tissue Research, Technische Universität Dresden, Germany.
| | - Anders Palmquist
- Department of Biomaterials, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.
| |
Collapse
|
7
|
Kong Y, Yang Y, Hou Y, Wang Y, Li W, Song Y. Advance in the application of organoids in bone diseases. Front Cell Dev Biol 2024; 12:1459891. [PMID: 39291264 PMCID: PMC11406180 DOI: 10.3389/fcell.2024.1459891] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Accepted: 08/20/2024] [Indexed: 09/19/2024] Open
Abstract
Bone diseases such as osteoporosis and osteoarthritis have become important human health problems, requiring a deeper understanding of the pathogenesis of related diseases and the development of more effective treatments. Bone organoids are three-dimensional tissue masses that are useful for drug screening, regenerative medicine, and disease modeling because they may mimic the structure and physiological activities of organs. Here, we describe various potential methods for culturing bone-related organoids from different stem cells, detailing the construction processes and highlighting the main applications of these bone organoid models. The application of bone organoids in different skeletal diseases is highlighted, and current and promising bone organoids for drug screening and regenerative medicine as well as the latest technological advancements in bone organoids are discussed, while the future development of bone organoids is discussed. Looking forward, it will provide a reference for constructing bone organoids with more complete structures and functions and applying them to biomedical research.
Collapse
Affiliation(s)
- Yajie Kong
- Department of Orthopedics, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
- Hebei Medical University-National University of Ireland Galway Stem Cell Research Center, Hebei Medical University, Shijiazhuang, Hebei, China
- Hebei Research Center for Stem Cell Medical Translational Engineering, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Yujia Yang
- Hebei Medical University-National University of Ireland Galway Stem Cell Research Center, Hebei Medical University, Shijiazhuang, Hebei, China
- Hebei Research Center for Stem Cell Medical Translational Engineering, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Yu Hou
- Hebei Medical University-National University of Ireland Galway Stem Cell Research Center, Hebei Medical University, Shijiazhuang, Hebei, China
- Hebei Research Center for Stem Cell Medical Translational Engineering, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Yuzhong Wang
- Department of Orthopedics, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Wenjing Li
- Department of Oral Medicine, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Yongzhou Song
- Department of Orthopedics, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
- Hebei Medical University-National University of Ireland Galway Stem Cell Research Center, Hebei Medical University, Shijiazhuang, Hebei, China
- Hebei Research Center for Stem Cell Medical Translational Engineering, Hebei Medical University, Shijiazhuang, Hebei, China
- Hebei Key Laboratory of Rare Disease, Shijiazhuang, Hebei, China
| |
Collapse
|
8
|
Budi HS, Fadhila O, Oki AS, Anitasari S, Shen YK, Dewi PS, Amalina DGR. A promising poly (e-caprolactone)/graphene-based scaffold as an antibacterial in regenerating bone tissue. BRAZ J BIOL 2024; 84:e279967. [PMID: 39140500 DOI: 10.1590/1519-6984.279967] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2023] [Accepted: 05/14/2024] [Indexed: 08/15/2024] Open
Abstract
Scaffolds are 3D biomaterials that provide an environment for cell regeneration. In the context of bone remodeling, poly(e-caprolactone) (PCL) combined with graphene has been developed as the scaffold. It is imperative for scaffolds to possess antibacterial properties in order to properly reduce the risk of potential infections.Therefore, this study aims to analyze the antibacterial characteristics of PCL/graphene scaffolds against Staphylococcus aureus (S. aureus) and Porphyromonas gingivalis (P. gingivalis) in vitro. In this study, five different groups were used, including PCL (K-), Amoxicillin (K+), PCL/Graphene 0.5 wt%, PCL/graphene 1 wt% and PCL/Graphene 1.5 wt%. All experiments were performed in triplicates and were repeated three times, and the diffusion method by Kirby-Bauer test was used. The disc was incubated with S. aureus and P. gingivalis for 24 hours and then the diameter of the inhibition zone was measured. The results showed that the PCL/graphene scaffolds exhibited dose-dependent antibacterial activity against S. aureus and P. gingivalis. The inhibition zone diameter (IZD) against S. aureus of PCL/graphene 1 wt% was 9.53 ± 0.74 mm, and increased to 11.93 ± 0.92 mm at a concentration of 1.5 wt% of graphene. The PCL/graphene scaffold with 1.5 wt% exhibited a greater inhibitory effect, with an IZD of 12.56 ± 0.06 mm against P. gingivalis, while the inhibitory activity of the 1 wt% variant was relatively lower at 10.46 ± 0.24 mm. The negative control, PCL, and PCL/graphene 0.5 wt% exhibited no antibacterial activity sequentially (p = 1). Scaffolds of poly(e-caprolactone)/graphene exhibited an antibacterial activity at 1, and 1.5 wt% on S. aureus and P. gingivalis. The antibacterial properties of this scaffold make it a promising candidate for regenerating bone tissue.
Collapse
Affiliation(s)
- H S Budi
- Universitas Airlangga, Faculty of Dental Medicine, Department of Oral Biology, Dental Pharmacology, Surabaya, Indonesia
- Surabaya Science Laboratory, Cell and Biology Research, Surabaya, Indonesia
| | - O Fadhila
- Universitas Airlangga, Faculty of Dental Medicine, Department of Oral Biology, Surabaya, Indonesia
| | - A S Oki
- Universitas Airlangga, Faculty of Dental Medicine, Department of Oral Biology, Surabaya, Indonesia
| | - S Anitasari
- Universitas Mulawarman, Faculty of Medicine, Department Medical Microbiology, Medical Program, Samarinda, Indonesia
| | - Y K Shen
- Taipei Medical University, College of Oral Medicine, School of Dental Technology, Taipei, Taiwan
| | - P S Dewi
- Universitas Mahasaraswati, Faculty of Dentistry, Department of Oral and Maxillofacial Surgery, Denpasar, Indonesia
| | - D G R Amalina
- Universitas Airlangga, Faculty of Dental Medicine, Department of Oral Biology, Surabaya, Indonesia
| |
Collapse
|
9
|
Riehakainen L, Mota-Silva E, Kusmic C, Panetta D, Petroni D, Fragnito D, Salvadori S, Menichetti L. Assessment of tissue response in vivo: PET-CT imaging of titanium and biodegradable magnesium implants. Acta Biomater 2024; 184:461-472. [PMID: 38871201 DOI: 10.1016/j.actbio.2024.06.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Revised: 06/01/2024] [Accepted: 06/06/2024] [Indexed: 06/15/2024]
Abstract
To study in vivo the bioactivity of biodegradable magnesium implants and other possible biomaterials, we are proposing a previously unexplored application of PET-CT imaging, using available tracers to follow soft tissue and bone remodelling and immune response in the presence of orthopaedic implants. Female Wistar rats received either implants (Ti6Al7Nb titanium or WE43 magnesium) or corresponding transcortical sham defects into the diaphyseal area of the femurs. Inflammatory response was followed with [18F]FDG and osteogenesis with [18F]NaF, over the period of 1.5 months after surgery. An additional pilot study with [68Ga]NODAGA-RGD tracer specific to αvβ3 integrin expression was performed to follow the angiogenesis for one month. [18F]FDG tracer uptake peaked on day 3 before declining in all groups, with Mg and Ti groups exhibiting overall higher uptake compared to sham. This suggests increased cellular activity and tissue response in the presence of Mg during the initial weeks, with Ti showing a subsequent increase in tracer uptake on day 45, indicating a foreign body reaction. [18F]NaF uptake demonstrated the superior osteogenic potential of Mg compared to Ti, with peak uptake on day 7 for all groups. [68Ga]NODAGA-RGD pilot study revealed differences in tracer uptake trends between groups, particularly the prolonged expression of αvβ3 integrin in the presence of implants. Based on the observed differences in the uptake trends of radiotracers depending on implant material, we suggest that PET-CT is a suitable modality for long-term in vivo assessment of orthopaedic biomaterial biocompatibility and underlying tissue reactions. STATEMENT OF SIGNIFICANCE: The study explores the novel use of positron emission tomography for the assessment of the influence that biomaterials have on the surrounding tissues. Previous related studies have mostly focused on material-related effects such as implant-associated infections or to follow the osseointegration in prosthetics, but the use of PET to evaluate the materials has not been reported before. The approach tests the feasibility of using repeated PET-CT imaging to follow the tissue response over time, potentially improving the methodology for adopting new biomaterials for clinical use.
Collapse
Affiliation(s)
- Leon Riehakainen
- The Sant'Anna School of Advanced Studies, Pisa, Italy; Institute of Clinical Physiology, National Research Council (IFC-CNR), Pisa, Italy.
| | - Eduarda Mota-Silva
- The Sant'Anna School of Advanced Studies, Pisa, Italy; Institute of Clinical Physiology, National Research Council (IFC-CNR), Pisa, Italy
| | - Claudia Kusmic
- Institute of Clinical Physiology, National Research Council (IFC-CNR), Pisa, Italy
| | - Daniele Panetta
- Institute of Clinical Physiology, National Research Council (IFC-CNR), Pisa, Italy
| | - Debora Petroni
- Institute of Clinical Physiology, National Research Council (IFC-CNR), Pisa, Italy
| | - Davide Fragnito
- Institute of Clinical Physiology, National Research Council (IFC-CNR), Pisa, Italy
| | - Stefano Salvadori
- Institute of Clinical Physiology, National Research Council (IFC-CNR), Pisa, Italy
| | - Luca Menichetti
- Institute of Clinical Physiology, National Research Council (IFC-CNR), Pisa, Italy
| |
Collapse
|
10
|
Liao S, Wu S, Mao C, Wang C, Cui Z, Zheng Y, Li Z, Jiang H, Zhu S, Liu X. Electron Aggregation and Oxygen Fixation Reinforced Microwave Dynamic and Thermal Therapy for Effective Treatment of MRSA-Induced Osteomyelitis. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2312280. [PMID: 38312094 DOI: 10.1002/smll.202312280] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/30/2023] [Revised: 01/21/2024] [Indexed: 02/06/2024]
Abstract
Antibiotics are frequently used to clinically treat osteomyelitis caused by bacterial infections. However, extended antibiotic use may result in drug resistance, which can be life threatening. Here, a heterojunction comprising Fe2O3/Fe3S4 magnetic composite is constructed to achieve short-term and efficient treat osteomyelitis caused by methicillin-resistant Staphylococcus aureus (MRSA). The Fe2O3/Fe3S4 composite exhibits powerful microwave (MW) absorption properties, thereby effectively converting incident electromagnetic energy into thermal energy. Density functional theory calculations demonstrate that Fe2O3/Fe3S4 possesses significant charge accumulation and oxygen-fixing capacity at the heterogeneous interface, which provides more active sites and oxygen sources for trapping electromagnetic hotspots. The finite element analysis indicates that Fe2O3/Fe3S4 displays a larger electromagnetism field enhancement parameter than Fe2O3 owing to a significant increase in electromagnetic hotspots. These hotspots contribute to charge differential accumulation and depletion motions at the interface, thereby augmenting the release of free electrons that subsequently combine with the oxygen adsorbed by Fe2O3/Fe3S4 to generate reactive oxygen species (ROS) and heat. This research, which achieves extraordinary bacterial eradication through the synergistic effect of microwave thermal therapy (MWTT) and microwave dynamic therapy (MDT), presents a novel strategy for treating deep-tissue bacterial infections.
Collapse
Affiliation(s)
- Shasha Liao
- Biomedical Materials Engineering Research Center, Hubei Key Laboratory of Polymer Materials, Ministry-of-Education Key Laboratory for the Green Preparation and Application of Functional Materials, School of Materials Science & Engineering, State Key Laboratory of Biocatalysis and Enzyme Engineering, Hubei University, Wuhan, 430062, China
- School of Health Science & Biomedical Engineering, Hebei University of Technology, Xiping Avenue 5340#, Tianjin, 300401, China
- School of Materials Science & Engineering, Peking University, Yiheyuan Road 5#, Beijing, 100871, China
| | - Shuilin Wu
- Biomedical Materials Engineering Research Center, Hubei Key Laboratory of Polymer Materials, Ministry-of-Education Key Laboratory for the Green Preparation and Application of Functional Materials, School of Materials Science & Engineering, State Key Laboratory of Biocatalysis and Enzyme Engineering, Hubei University, Wuhan, 430062, China
- School of Materials Science & Engineering, Peking University, Yiheyuan Road 5#, Beijing, 100871, China
- School of Materials Science & Engineering, the Key Laboratory of Advanced Ceramics and Machining Technology by the Ministry of Education of China, Tianjin University, Yaguan Road 135#, Tianjin, 300072, China
| | - Congyang Mao
- Biomedical Materials Engineering Research Center, Hubei Key Laboratory of Polymer Materials, Ministry-of-Education Key Laboratory for the Green Preparation and Application of Functional Materials, School of Materials Science & Engineering, State Key Laboratory of Biocatalysis and Enzyme Engineering, Hubei University, Wuhan, 430062, China
| | - Chaofeng Wang
- School of Health Science & Biomedical Engineering, Hebei University of Technology, Xiping Avenue 5340#, Tianjin, 300401, China
| | - Zhenduo Cui
- School of Materials Science & Engineering, the Key Laboratory of Advanced Ceramics and Machining Technology by the Ministry of Education of China, Tianjin University, Yaguan Road 135#, Tianjin, 300072, China
| | - Yufeng Zheng
- School of Materials Science & Engineering, Peking University, Yiheyuan Road 5#, Beijing, 100871, China
| | - Zhaoyang Li
- School of Materials Science & Engineering, the Key Laboratory of Advanced Ceramics and Machining Technology by the Ministry of Education of China, Tianjin University, Yaguan Road 135#, Tianjin, 300072, China
| | - Hui Jiang
- School of Materials Science & Engineering, the Key Laboratory of Advanced Ceramics and Machining Technology by the Ministry of Education of China, Tianjin University, Yaguan Road 135#, Tianjin, 300072, China
| | - Shengli Zhu
- School of Materials Science & Engineering, the Key Laboratory of Advanced Ceramics and Machining Technology by the Ministry of Education of China, Tianjin University, Yaguan Road 135#, Tianjin, 300072, China
| | - Xiangmei Liu
- Biomedical Materials Engineering Research Center, Hubei Key Laboratory of Polymer Materials, Ministry-of-Education Key Laboratory for the Green Preparation and Application of Functional Materials, School of Materials Science & Engineering, State Key Laboratory of Biocatalysis and Enzyme Engineering, Hubei University, Wuhan, 430062, China
- School of Health Science & Biomedical Engineering, Hebei University of Technology, Xiping Avenue 5340#, Tianjin, 300401, China
| |
Collapse
|
11
|
Shi X, Li M, Ni H, Wu Y, Li Y, Chen X, Xu Y. Integrative gene expression analysis and animal model reveal immune- and autophagy-related biomarkers in osteomyelitis. Immun Inflamm Dis 2024; 12:e1339. [PMID: 38990187 PMCID: PMC11238574 DOI: 10.1002/iid3.1339] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Revised: 04/26/2024] [Accepted: 06/29/2024] [Indexed: 07/12/2024] Open
Abstract
BACKGROUND Osteomyelitis (OM) is recognized as a significant challenge in orthopedics due to its complex immune and inflammatory responses. The prognosis heavily depends on timely diagnosis, accurate classification, and assessment of severity. Thus, the identification of diagnostic and classification-related genes from an immunological standpoint is crucial for the early detection and tailored treatment of OM. METHODS Transcriptomic data for OM was sourced from the Gene Expression Omnibus (GEO) database, leading to the identification of autophagy- and immune-related differentially expressed genes (AIR-DEGs) through differential expression analysis. Diagnostic and classification models were subsequently developed. The CIBERSORT algorithm was utilized to examine immune cell infiltration in OM, and the relationship between OM clusters and various immune cells was explored. Key AIR-DEGs were further validated through the creation of OM animal models. RESULTS Analysis of the transcriptomic data revealed three AIR-DEGs that played a significant role in immune responses and pathways. Nomogram and receiver operating characteristic curve analyses were performed, demonstrating excellent diagnostic capability for differentiating between OM patients and healthy individuals, with an area under the curve of 0.814. An unsupervised clustering analysis discerned two unique patterns of autophagy- and immune-related genes, as well as gene patterns. Further exploration into immune infiltration exhibited notable variances across different subtypes, especially between OM cluster 1 and gene cluster A, highlighting their potential role in mitigating inflammatory responses by regulating immune activities. Moreover, the mRNA and protein expression levels of three AIR-DEGs in the animal model were aligned with those in the training and validation data sets. CONCLUSIONS From an immunological perspective, a diagnostic model was successfully developed, and two distinct clustering patterns were identified. These contributions offer a significant resource for the early detection and personalized immunotherapy of patients with OM.
Collapse
Affiliation(s)
- Xiangwen Shi
- Kunming Medical University, Kunming, China
- Laboratory of Yunnan Traumatology and Orthopedics Clinical Medical Center, Yunnan Orthopedics and Sports Rehabilitation Clinical Medical Research Center, Kunming, China
- Department of Orthopedic Surgery, 920th Hospital of Joint Logistics Support Force of PLA, Kunming, Yunnan, China
| | - Mingjun Li
- Laboratory of Yunnan Traumatology and Orthopedics Clinical Medical Center, Yunnan Orthopedics and Sports Rehabilitation Clinical Medical Research Center, Kunming, China
- Department of Orthopedic Surgery, 920th Hospital of Joint Logistics Support Force of PLA, Kunming, Yunnan, China
| | - Haonan Ni
- Orthopedic Department, First People's Hospital of Huzhou, First Affiliated Hospital of Huzhou University, Huzhou, China
| | - Yipeng Wu
- Kunming Medical University, Kunming, China
- Laboratory of Yunnan Traumatology and Orthopedics Clinical Medical Center, Yunnan Orthopedics and Sports Rehabilitation Clinical Medical Research Center, Kunming, China
- Department of Orthopedic Surgery, 920th Hospital of Joint Logistics Support Force of PLA, Kunming, Yunnan, China
| | - Yang Li
- Laboratory of Yunnan Traumatology and Orthopedics Clinical Medical Center, Yunnan Orthopedics and Sports Rehabilitation Clinical Medical Research Center, Kunming, China
- Department of Orthopedic Surgery, 920th Hospital of Joint Logistics Support Force of PLA, Kunming, Yunnan, China
| | - Xianjun Chen
- Department of Neurosurgery, Nanping First Hospital Affiliated to Fujian Medical University, Nanping, Fujian, China
| | - Yongqing Xu
- Laboratory of Yunnan Traumatology and Orthopedics Clinical Medical Center, Yunnan Orthopedics and Sports Rehabilitation Clinical Medical Research Center, Kunming, China
- Department of Orthopedic Surgery, 920th Hospital of Joint Logistics Support Force of PLA, Kunming, Yunnan, China
| |
Collapse
|
12
|
Sun Q, Huynh K, Muratovic D, Gunn NJ, Zelmer AR, Solomon LB, Atkins GJ, Yang D. Rapid bacterial evaluation beyond the colony forming unit in osteomyelitis. eLife 2024; 13:RP93698. [PMID: 38910553 PMCID: PMC11196105 DOI: 10.7554/elife.93698] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/25/2024] Open
Abstract
Examination of bacteria/host cell interactions is important for understanding the aetiology of many infectious diseases. The colony forming unit (CFU) has been the standard for quantifying bacterial burden for the past century, however, this suffers from low sensitivity and is dependent on bacterial culturability in vitro. Our data demonstrate the discrepancy between the CFU and bacterial genome copy number in an osteomyelitis-relevant co-culture system and we confirm diagnosis and quantify bacterial load in clinical bone specimens. This study provides an improved workflow for the quantification of bacterial burden in such cases.
Collapse
Affiliation(s)
- Qi Sun
- Centre for Orthopaedic & Trauma Research, Faculty of Health and Medical Sciences, University of AdelaideAdelaideAustralia
| | - Kimberley Huynh
- Centre for Orthopaedic & Trauma Research, Faculty of Health and Medical Sciences, University of AdelaideAdelaideAustralia
| | - Dzenita Muratovic
- Centre for Orthopaedic & Trauma Research, Faculty of Health and Medical Sciences, University of AdelaideAdelaideAustralia
| | - Nicholas J Gunn
- Centre for Orthopaedic & Trauma Research, Faculty of Health and Medical Sciences, University of AdelaideAdelaideAustralia
| | - Anja R Zelmer
- Centre for Orthopaedic & Trauma Research, Faculty of Health and Medical Sciences, University of AdelaideAdelaideAustralia
| | - Lucian Bogdan Solomon
- Centre for Orthopaedic & Trauma Research, Faculty of Health and Medical Sciences, University of AdelaideAdelaideAustralia
- Department of Orthopaedics and Trauma, Royal Adelaide HospitalAdelaideAustralia
| | - Gerald J Atkins
- Centre for Orthopaedic & Trauma Research, Faculty of Health and Medical Sciences, University of AdelaideAdelaideAustralia
| | - Dongqing Yang
- Centre for Orthopaedic & Trauma Research, Faculty of Health and Medical Sciences, University of AdelaideAdelaideAustralia
| |
Collapse
|
13
|
Mishra AH, Mohan S, Gutti P, Krishna S, Sundaraman S, Chakraborti S, Jaiswal AK, Nambi Raj NA, Mishra D. Bioselective and Radiopaque Zinc-Biopolymeric Complex-Based Porous Biomaterials Promote Mammalian Tissue Ingrowth In Vivo While Inhibiting Microbial Biofilm Gene Expression and Biofilm Formation. ACS APPLIED BIO MATERIALS 2024; 7:3701-3713. [PMID: 38748449 DOI: 10.1021/acsabm.4c00013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/18/2024]
Abstract
Metal-organic complexes have shown astounding bioactive properties; however, they are rarely explored as biomaterials. Recent studies showed that carboxymethyl-chitosan (CMC) genipin-conjugated zinc biomimetic scaffolds have unique bioselective properties. The biomaterial was reported to be mammalian cell-friendly; at the same time, it was found to discourage microbial biofilm formation on its surface, which seemed to be a promising solution to addressing the problem of trauma-associated biofilm formation and development of antimicrobial resistance. However, the mechanically frail characteristics and zinc overload raise concerns and limit the potential of the said biomaterials. Hence, the present work is focused on improving the strength of the earlier scaffold formulations, testing its in vivo efficacy and reaffirming its action against biofilm-forming microbe Staphylococcus aureus. Scaling up of CMC proportion increased rigidity, and 8% CMC was found to be the ideal concentration for robust scaffold fabrication. Freeze-dried CMC scaffolds with or without genipin (GP) cross-linking were conjugated with zinc using 2 M zinc acetate solution. Characterization results indicated that the CMC-Zn scaffolds, without genipin, showed mechanical properties close to bone fillers, resist in vitro enzymatic degradation until 4 weeks, are porous in nature, and have radiopacity close to mandibular bones. Upon implantation in a subcutaneous pocket of Wistar rats, the scaffolds showed tissue in-growth with simultaneous degradation without any signs of toxicity past 28 days. Neither were there any signs of toxicity in any of the vital organs. Considering many superior properties among the other formulations, the CMC-Zn scaffolds were furthered for biofilm studies. CMC-Zn showed negligible S. aureus biofilm formation on its surface as revealed by an alamar blue-based study. RT-PCR analysis revealed that CMC-Zn downregulated the expression of pro-biofilm effector genes such as icaC and clfB. A protein docking study predicted the inhibitory mechanism of CMC-Zn. Although it binds strongly when alone, at high density, it may cause inactivation of the transmembrane upstream activators of the said genes, thereby preventing their dimerization and subsequent inactivation of the effector genes. In conclusion, zinc-conjugated carboxymethyl-chitosan scaffolds are mechanically robust, porous, yet biodegradable, harmless to the host in the long term, they are radiopaque and prevent biofilm gene expression in notorious microbes; hence, they could be a suitable candidate for bone filler applications.
Collapse
Affiliation(s)
- Arushi Hitendra Mishra
- Bioinspired Design Lab, School of Bio-Sciences and Technology (SBST), Vellore Institute of Technology (VIT), Vellore Campus, Vellore, Tamil Nadu 632014, India
| | - Sanjukta Mohan
- Bioinspired Design Lab, School of Bio-Sciences and Technology (SBST), Vellore Institute of Technology (VIT), Vellore Campus, Vellore, Tamil Nadu 632014, India
| | - Pavan Gutti
- Bioinspired Design Lab, School of Bio-Sciences and Technology (SBST), Vellore Institute of Technology (VIT), Vellore Campus, Vellore, Tamil Nadu 632014, India
| | - Sreevatsan Krishna
- Bioinspired Design Lab, School of Bio-Sciences and Technology (SBST), Vellore Institute of Technology (VIT), Vellore Campus, Vellore, Tamil Nadu 632014, India
| | - Sugunapriyadarshini Sundaraman
- Bioinspired Design Lab, School of Bio-Sciences and Technology (SBST), Vellore Institute of Technology (VIT), Vellore Campus, Vellore, Tamil Nadu 632014, India
| | - Sourangshu Chakraborti
- Centre for Biomaterials, Cellular and Molecular Theranostics (CBCMT), Vellore Institute of Technology (VIT), Vellore Campus, Vellore, Tamil Nadu 632014, India
| | - Amit Kumar Jaiswal
- Centre for Biomaterials, Cellular and Molecular Theranostics (CBCMT), Vellore Institute of Technology (VIT), Vellore Campus, Vellore, Tamil Nadu 632014, India
| | - N Arunai Nambi Raj
- School of Advanced Sciences (SAS), Vellore Institute of Technology (VIT), Vellore Campus, Vellore, Tamil Nadu 632014, India
| | - Debasish Mishra
- Bioinspired Design Lab, School of Bio-Sciences and Technology (SBST), Vellore Institute of Technology (VIT), Vellore Campus, Vellore, Tamil Nadu 632014, India
| |
Collapse
|
14
|
Shi X, Ni H, Tang L, Li M, Wu Y, Xu Y. Comprehensive Gene Analysis Reveals Cuproptosis-Related Gene Signature Associated with M2 Macrophage in Staphylococcus aureus-Infected Osteomyelitis. J Inflamm Res 2024; 17:3057-3077. [PMID: 38770176 PMCID: PMC11104443 DOI: 10.2147/jir.s457414] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Accepted: 05/08/2024] [Indexed: 05/22/2024] Open
Abstract
Objective Osteomyelitis is a challenging disease in the field of bone infections, with its immune and molecular regulatory mechanisms still poorly understood. The aim of this study is to explore the value and potential mechanisms of cuproptosis-related genes (CRGs) in Staphylococcus aureus (S. aureus)-infected osteomyelitis from an immunological perspective. Methods Initially, three transcriptomic datasets from public databases were integrated and analyzed, and consistent expression of CRGs in S. aureus-infected osteomyelitis was identified. Subsequently, immune infiltration analysis was performed, and M2 macrophage-related CRGs (M2R-CRGs) were further identified. Their potential molecular mechanisms were evaluated using Gene Set Variation Analysis (GSVA) and Gene Set Enrichment Analysis (GSEA). Finally, distinct osteomyelitis subtypes and diagnostic models based on characteristic M2R-CRGs were constructed. Results Through correlation analysis with immune cell infiltration, three characteristic M2R-CRGs (SLC31A1, DLD, and MTF1) were identified. Further analysis using unsupervised clustering and immune microenvironment analysis indicated that cluster 1 might activate pro-inflammatory responses, while cluster 2 was shown to exhibit anti-inflammatory effects in osteomyelitis. Compared to Cluster A, Cluster B demonstrated higher levels and a greater diversity of immune cell infiltrations in CRG-related molecular patterns, suggesting a potential anti-inflammatory role in osteomyelitis. A diagnostic model for S. aureus-infected osteomyelitis, based on the three M2R-CRGs, was constructed, exhibiting excellent diagnostic performance and validated with an independent dataset. Significant upregulation in mRNA and protein expression levels of the three M2R-CRGs was observed in rat models of S. aureus-infected osteomyelitis, aligning with bioinformatic results. Conclusion The M2R-CRGs (SLC31A1, DLD, and MTF1) may be considered characteristic genes for early diagnosis and personalized immune therapy in patients with S. aureus-infected osteomyelitis.
Collapse
Affiliation(s)
- Xiangwen Shi
- Graduate School, Kunming Medical University, Kunming, People’s Republic of China
- Laboratory of Yunnan Traumatology and Orthopedics Clinical Medical Center, Yunnan Orthopedics and Sports Rehabilitation Clinical Medical Research Center, Department of Orthopedic Surgery, 920th Hospital of Joint Logistics Support Force of PLA, Kunming, People’s Republic of China
| | - Haonan Ni
- First People’s Hospital of Huzhou, the First affiliated Hospital of Huzhou University, Huzhou, People’s Republic of China
| | - Linmeng Tang
- Department of Radiology, the Second Affiliated Hospital of Kunming Medical University, Kunming, People’s Republic of China
| | - Mingjun Li
- Laboratory of Yunnan Traumatology and Orthopedics Clinical Medical Center, Yunnan Orthopedics and Sports Rehabilitation Clinical Medical Research Center, Department of Orthopedic Surgery, 920th Hospital of Joint Logistics Support Force of PLA, Kunming, People’s Republic of China
| | - Yipeng Wu
- Laboratory of Yunnan Traumatology and Orthopedics Clinical Medical Center, Yunnan Orthopedics and Sports Rehabilitation Clinical Medical Research Center, Department of Orthopedic Surgery, 920th Hospital of Joint Logistics Support Force of PLA, Kunming, People’s Republic of China
| | - Yongqing Xu
- First People’s Hospital of Huzhou, the First affiliated Hospital of Huzhou University, Huzhou, People’s Republic of China
| |
Collapse
|
15
|
Zhang Y, Cheng Y, Zhao Z, Jiang S, Zhang Y, Li J, Huang S, Wang W, Xue Y, Li A, Tao Z, Wu Z, Zhang X. Enhanced Chemoradiotherapy for MRSA-Infected Osteomyelitis Using Immunomodulatory Polymer-Reinforced Nanotherapeutics. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2304991. [PMID: 38408365 DOI: 10.1002/adma.202304991] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Revised: 12/27/2023] [Indexed: 02/28/2024]
Abstract
The eradication of osteomyelitis caused by methicillin-resistant Staphylococcus aureus (MRSA) poses a significant challenge due to its development of biofilm-induced antibiotic resistance and impaired innate immunity, which often leads to frequent surgical failure. Here, the design, synthesis, and performance of X-ray-activated polymer-reinforced nanotherapeutics that modulate the immunological properties of infectious microenvironments to enhance chemoradiotherapy against multidrug-resistant bacterial deep-tissue infections are reported. Upon X-ray radiation, the proposed polymer-reinforced nanotherapeutic generates reactive oxygen species and reactive nitrogen species. To robustly eradicate MRSA biofilms at deep infection sites, these species can specifically bind to MRSA and penetrate biofilms for enhanced chemoradiotherapy treatment. X-ray-activated nanotherapeutics modulate the innate immunity of macrophages to prevent the recurrence of osteomyelitis. The remarkable anti-infection effects of these nanotherapeutics are validated using a rat osteomyelitis model. This study demonstrates the significant potential of a synergistic chemoradiotherapy and immunotherapy method for treating MRSA biofilm-infected osteomyelitis.
Collapse
Affiliation(s)
- Yufei Zhang
- Key Laboratory of Functional Polymer Materials of Ministry of Education, Institute of Polymer Chemistry, Tianjin Key Laboratory of functional polymer materials College of Chemistry, Nankai University, Tianjin, 300071, China
| | - Yijie Cheng
- Key Laboratory of Functional Polymer Materials of Ministry of Education, Institute of Polymer Chemistry, Tianjin Key Laboratory of functional polymer materials College of Chemistry, Nankai University, Tianjin, 300071, China
| | - Zhe Zhao
- Department of Surgery of Traditional Chinese Medicine, Tianjin Hospital, Tianjin, 300211, China
| | - Shengpeng Jiang
- Department of Radiation Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, 300060, China
| | - Yuhan Zhang
- Department of Radiation Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, 300060, China
| | - Jie Li
- Key Laboratory of Functional Polymer Materials of Ministry of Education, Institute of Polymer Chemistry, Tianjin Key Laboratory of functional polymer materials College of Chemistry, Nankai University, Tianjin, 300071, China
| | - Siyuan Huang
- Key Laboratory of Functional Polymer Materials of Ministry of Education, Institute of Polymer Chemistry, Tianjin Key Laboratory of functional polymer materials College of Chemistry, Nankai University, Tianjin, 300071, China
| | - Wenbo Wang
- Key Laboratory of Functional Polymer Materials of Ministry of Education, Institute of Polymer Chemistry, Tianjin Key Laboratory of functional polymer materials College of Chemistry, Nankai University, Tianjin, 300071, China
| | - Yun Xue
- Key Laboratory of Functional Polymer Materials of Ministry of Education, Institute of Polymer Chemistry, Tianjin Key Laboratory of functional polymer materials College of Chemistry, Nankai University, Tianjin, 300071, China
| | - Anran Li
- Key Laboratory of Functional Polymer Materials of Ministry of Education, Institute of Polymer Chemistry, Tianjin Key Laboratory of functional polymer materials College of Chemistry, Nankai University, Tianjin, 300071, China
| | - Zhen Tao
- Department of Radiation Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, 300060, China
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
| | - Zhongming Wu
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education, Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China
| | - Xinge Zhang
- Key Laboratory of Functional Polymer Materials of Ministry of Education, Institute of Polymer Chemistry, Tianjin Key Laboratory of functional polymer materials College of Chemistry, Nankai University, Tianjin, 300071, China
| |
Collapse
|
16
|
Surendar J, Hackenberg RK, Schmitt-Sánchez F, Ossendorff R, Welle K, Stoffel-Wagner B, Sage PT, Burger C, Wirtz DC, Strauss AC, Schildberg FA. Osteomyelitis is associated with increased anti-inflammatory response and immune exhaustion. Front Immunol 2024; 15:1396592. [PMID: 38736874 PMCID: PMC11082283 DOI: 10.3389/fimmu.2024.1396592] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Accepted: 04/12/2024] [Indexed: 05/14/2024] Open
Abstract
Introduction Osteomyelitis (OMS) is a bone infection causing bone pain and severe complications. A balanced immune response is critical to eradicate infection without harming the host, yet pathogens manipulate immunity to establish a chronic infection. Understanding OMS-driven inflammation is essential for disease management, but comprehensive data on immune profiles and immune cell activation during OMS are lacking. Methods Using high-dimensional flow cytometry, we investigated the detailed innate and adaptive systemic immune cell populations in OMS and age- and sex-matched controls. Results Our study revealed that OMS is associated with increased levels of immune regulatory cells, namely T regulatory cells, B regulatory cells, and T follicular regulatory cells. In addition, the expression of immune activation markers HLA-DR and CD86 was decreased in OMS, while the expression of immune exhaustion markers TIM-3, PD-1, PD-L1, and VISTA was increased. Members of the T follicular helper (Tfh) cell family as well as classical and typical memory B cells were significantly increased in OMS individuals. We also found a strong correlation between memory B cells and Tfh cells. Discussion We conclude that OMS skews the host immune system towards the immunomodulatory arm and that the Tfh memory B cell axis is evident in OMS. Therefore, immune-directed therapies may be a promising alternative for eradication and recurrence of infection in OMS, particularly in individuals and areas where antibiotic resistance is a major concern.
Collapse
Affiliation(s)
- Jayagopi Surendar
- Department of Orthopedics and Trauma Surgery, University Hospital Bonn, Bonn, Germany
| | - Roslind K. Hackenberg
- Department of Orthopedics and Trauma Surgery, University Hospital Bonn, Bonn, Germany
- Department of Hand, Plastic and Reconstructive Surgery, Burn Center, BG Trauma Center Ludwigshafen, University of Heidelberg, Ludwigshafen, Germany
| | - Fabio Schmitt-Sánchez
- Department of Orthopedics and Trauma Surgery, University Hospital Bonn, Bonn, Germany
| | - Robert Ossendorff
- Department of Orthopedics and Trauma Surgery, University Hospital Bonn, Bonn, Germany
| | - Kristian Welle
- Department of Orthopedics and Trauma Surgery, University Hospital Bonn, Bonn, Germany
| | - Birgit Stoffel-Wagner
- Institute of Clinical Chemistry and Clinical Pharmacology, University Hospital Bonn, Bonn, Germany
| | - Peter T. Sage
- Transplantation Research Center, Renal Division, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
| | - Christof Burger
- Department of Orthopedics and Trauma Surgery, University Hospital Bonn, Bonn, Germany
| | - Dieter C. Wirtz
- Department of Orthopedics and Trauma Surgery, University Hospital Bonn, Bonn, Germany
| | - Andreas C. Strauss
- Department of Orthopedics and Trauma Surgery, University Hospital Bonn, Bonn, Germany
| | - Frank A. Schildberg
- Department of Orthopedics and Trauma Surgery, University Hospital Bonn, Bonn, Germany
| |
Collapse
|
17
|
Liu CG, Li DY, Gao X, Ma T, Zhang K, Liu DY. Examining the causal relationship between circulating immune cells and the susceptibility to osteomyelitis: A Mendelian randomization study. Int Immunopharmacol 2024; 131:111815. [PMID: 38492335 DOI: 10.1016/j.intimp.2024.111815] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2024] [Revised: 03/04/2024] [Accepted: 03/05/2024] [Indexed: 03/18/2024]
Abstract
BACKGROUND Osteomyelitis is considered as a deleterious inflammatory condition affecting the bone, primarily attributed to pathogenic infection. However, the underlying factors predisposing individuals to osteomyelitis remain incompletely elucidated. The immune system plays a multifaceted role in the progression of this condition, yet previous observational studies and randomized controlled trials investigating the association between circulating immune cell counts and osteomyelitis have been constrained. In order to address this knowledge gap, we conducted a Mendelian randomization (MR) analysis to evaluate the impact of diverse immune cell counts on the risk of developing osteomyelitis. METHODS In our study, we utilized single nucleotide polymorphisms (SNPs) that have been strongly linked to circulating immune cells or specific lymphocyte subtypes, as identified in large-scale genome-wide association studies (GWAS). These SNPs served as instrumental variables (IVs) for our MR analysis. We employed a more relaxed clumping threshold to conduct MR analysis on several related lymphocyte subtypes. To estimate causal effects, we utilized the Wald ratio, as well as the random-effects inverse variance weighted (IVW) and weighted median (WM) methods. To enhance the credibility of our results, we performed F-statistic calculations and a series of sensitivity analyses. RESULTS Our findings revealed a significant correlation between the absolute count of circulating lymphocytes and the risk of osteomyelitis [odds ratio(OR) 1.20;95 % confidence interval (CI), 1.08-1.32;P = 0.0005]. Furthermore, we identified a causal relationship between the absolute count of CD8+ T cells and susceptibility to osteomyelitis (OR 1.16; 95 % CI, 1.04-1.30; P = 0.0098). Importantly, these findings remained robust across a wide range of sensitivity analyses. CONCLUSION Through our MR analysis, we have provided evidence supporting a causal relationship between genetic predisposition to higher circulating immune cell counts and an increased risk of osteomyelitis. Specifically, our findings highlight the association between elevated CD8+ T cell counts and a heightened susceptibility to osteomyelitis. These results offer valuable insights for the future exploration of immunotherapy approaches in the management of osteomyelitis.
Collapse
Affiliation(s)
- Chun-Gui Liu
- Severe & Poly-trauma Division, Orthopedic Trauma Department, Hong-Hui Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Dong-Yang Li
- Severe & Poly-trauma Division, Orthopedic Trauma Department, Hong-Hui Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Xi Gao
- Severe & Poly-trauma Division, Orthopedic Trauma Department, Hong-Hui Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Teng Ma
- Severe & Poly-trauma Division, Orthopedic Trauma Department, Hong-Hui Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Kun Zhang
- Severe & Poly-trauma Division, Orthopedic Trauma Department, Hong-Hui Hospital, Xi'an Jiaotong University, Xi'an, China
| | - De-Yin Liu
- Severe & Poly-trauma Division, Orthopedic Trauma Department, Hong-Hui Hospital, Xi'an Jiaotong University, Xi'an, China.
| |
Collapse
|
18
|
Sánchez-Ortiz E, Blanco Gutiérrez MDM, Calvo-Fernandez C, Mencía-Gutiérrez A, Pastor Tiburón N, Alvarado Piqueras A, Pablos-Tanarro A, Martín-Maldonado B. Addressing Challenges in Wildlife Rehabilitation: Antimicrobial-Resistant Bacteria from Wounds and Fractures in Wild Birds. Animals (Basel) 2024; 14:1151. [PMID: 38672299 PMCID: PMC11047587 DOI: 10.3390/ani14081151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2024] [Revised: 04/03/2024] [Accepted: 04/09/2024] [Indexed: 04/28/2024] Open
Abstract
Injuries and bone fractures are the most frequent causes of admission at wildlife rescue centers. Wild birds are more susceptible to open fractures due to their anatomical structure, which can lead to osteomyelitis and necrosis. Antibiotic therapy in these cases is indispensable, but the increase of antimicrobial-resistant isolates in wildlife has become a significant concern in recent years. In this context, the likelihood of antibiotic failure and death of animals with infectious issues is high. This study aimed to isolate, identify, and assess the antimicrobial resistance pattern of bacteria in wounds and open fractures in wild birds. To this end, injured birds admitted to a wildlife rescue center were sampled, and bacterial isolation and identification were performed. Then, antimicrobial susceptibility testing was assessed according to the disk diffusion method. In total, 36 isolates were obtained from 26 different birds. The genera detected were Staphylococcus spp. (63.8%), Escherichia (13.9%), Bacillus (11.1%), Streptococcus (8.3%), and Micrococcus (2.8%). Among Staphylococcus isolates, S. lentus and S. aureus were the most frequent species. Antimicrobial resistance was detected in 82.6% of the isolates, among which clindamycin resistance stood out, and 31.6% of resistant isolates were considered multidrug-resistant. Results from this study highlight the escalating scope of antimicrobial resistance in wildlife. This level of resistance poses a dual concern for wildlife: firstly, the risk of therapeutic failure in species of significant environmental value, and, secondly, the circulation of resistant bacteria in ecosystems.
Collapse
Affiliation(s)
- Esther Sánchez-Ortiz
- Departamento de Sanidad Animal, Facultad de Veterinaria, Universidad Complutense, Avenida de Puerta de Hierro s/n, 28040 Madrid, Spain; (E.S.-O.); (M.d.M.B.G.)
- Grupo de Rehabilitación de la Fauna Autóctona y su Hábitat, Calle Monte del Pilar s/n, 28220 Majadahonda, Spain; (A.M.-G.); (N.P.T.); (A.A.P.)
| | - María del Mar Blanco Gutiérrez
- Departamento de Sanidad Animal, Facultad de Veterinaria, Universidad Complutense, Avenida de Puerta de Hierro s/n, 28040 Madrid, Spain; (E.S.-O.); (M.d.M.B.G.)
| | - Cristina Calvo-Fernandez
- Research Group for Food Microbiology and Hygiene, National Food Institute, Technical University of Denmark, Henrik Dams Allé, 204, 2800 Kongens Lyngby, Denmark;
- Research Group for Foodborne Pathogens and Epidemiology, National Food Institute, Technical University of Denmark, Henrik Dams Allé, 204, 2800 Kongens Lyngby, Denmark
| | - Aida Mencía-Gutiérrez
- Grupo de Rehabilitación de la Fauna Autóctona y su Hábitat, Calle Monte del Pilar s/n, 28220 Majadahonda, Spain; (A.M.-G.); (N.P.T.); (A.A.P.)
| | - Natalia Pastor Tiburón
- Grupo de Rehabilitación de la Fauna Autóctona y su Hábitat, Calle Monte del Pilar s/n, 28220 Majadahonda, Spain; (A.M.-G.); (N.P.T.); (A.A.P.)
| | - Alberto Alvarado Piqueras
- Grupo de Rehabilitación de la Fauna Autóctona y su Hábitat, Calle Monte del Pilar s/n, 28220 Majadahonda, Spain; (A.M.-G.); (N.P.T.); (A.A.P.)
| | - Alba Pablos-Tanarro
- Departamento de Veterinaria, Facultad de Ciencias Biomédicas y de la Salud, Universidad Europea de Madrid, Calle Tajo s/n, 28760 Villaviciosa de Odón, Spain;
| | - Bárbara Martín-Maldonado
- Departamento de Veterinaria, Facultad de Ciencias Biomédicas y de la Salud, Universidad Europea de Madrid, Calle Tajo s/n, 28760 Villaviciosa de Odón, Spain;
| |
Collapse
|
19
|
Shen Y, Xu Y, Yu Z, Chen G, Chen B, Liao L. Multifunctional Injectable Microspheres Containing "Naturally-Derived" Photothermal Transducer for Synergistic Physical and Chemical Treating of Acute Osteomyelitis through Sequential Immunomodulation. ACS NANO 2024. [PMID: 38335113 DOI: 10.1021/acsnano.3c10697] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/12/2024]
Abstract
Osteomyelitis induced by Staphylococcus aureus (S. aureus) is a persistent and deep-seated infection that affects bone tissue. The main challenges in treating osteomyelitis include antibiotic resistance, systemic toxicity, and the need for multiple recurrent surgeries. An ideal therapeutic strategy involves the development of materials that combine physical, chemical, and immunomodulatory synergistic effects. In this work, we prepared injectable microspheres consisting of an interpenetrating network of ionic-cross-linked sodium alginate (SA) and genipin (Gp)-cross-linked gelatin (Gel) incorporated with tannic acid (TA) and copper ions (Cu2+). The Gp-cross-linked Gel acted as a "naturally-derived" photothermal therapy (PTT) agent. The results showed that the microspheres exhibited efficient and rapid bactericidal effects against both S. aureus and Escherichia coli (E. coli) under the irradiation of near-infrared light at 808 nm wavelength; moreover, the release of Cu2+ also induced sustained inhibitory effects against bacteria during the nonirradiation period. The in vitro cell culture results indicated that when combined with PTT, the microspheres could adaptively modulate macrophage M1 and M2 phenotypes in sequence. Additionally, these microspheres were found to enhance the osteogenic differentiation of bone marrow mesenchymal stem cells (BMSCs). In vivo studies conducted in a rat femur osteomyelitis model with bone defects showed that under multiple laser irradiation the microspheres effectively controlled bacterial infection, improved the pathological immune microenvironment, and significantly enhanced the repair and regeneration of bone tissues in the affected area.
Collapse
Affiliation(s)
- Yang Shen
- Biomaterials Research Center, School of Biomedical Engineering, Southern Medical University, Guangzhou 510515, China
| | - Yaowen Xu
- Division of Orthopaedics and Traumatology, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Ziqian Yu
- Biomaterials Research Center, School of Biomedical Engineering, Southern Medical University, Guangzhou 510515, China
| | - Guo Chen
- Division of Orthopaedics and Traumatology, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Bin Chen
- Division of Orthopaedics and Traumatology, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Liqiong Liao
- Biomaterials Research Center, School of Biomedical Engineering, Southern Medical University, Guangzhou 510515, China
| |
Collapse
|
20
|
Afaque SF, Agrawal U, Shankhwar DK, Chand S, Verma V. A Rare Case of Fungal Osteomyelitis of the Distal Tibia in a Pediatric Patient. Cureus 2024; 16:e54648. [PMID: 38524028 PMCID: PMC10959727 DOI: 10.7759/cureus.54648] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/20/2024] [Indexed: 03/26/2024] Open
Abstract
Staphylococcus aureus infection is the most common cause of osteomyelitis. Over 100,000 fungal species have been described; only 150 are pathogenic to humans. These opportunistic infections frequently enter the body due to a decrease in host defense or through an invasive gateway, such as a dental extraction or skin discontinuity due to trauma. Symptoms and radiological examination often mimic those of other etiologies, which can lead to substantial delays in treatment. Our case is a 13-year-old healthy boy with no history of immune incompetency who presented to us with complaints of pain and swelling over his left ankle and leg with an on-and-off history of fever for 15 days. Based on his history and examination, he is diagnosed as having sub-acute osteomyelitis of the distal tibia with septic arthritis. The bacterial culture has no growth; however, the potassium hydroxide mount came positive for fungal elements having hyphae and pseudohyphae, and the fungal culture came positive for Candida. Management of fungal infections is challenging as they have infrequent involvement in bones. Fungal osteomyelitis is considered a rare entity in the literature, and the current case is studied for the management and diagnosis of a rare variant of osteomyelitis in the pediatric population. The treatment guidelines vary based on the identified organism and the duration of treatment. Debridement of fungal osteomyelitis or septic arthritis includes removing sinus tracts, evaluation for squamous cell carcinoma, bony and soft-tissue debridement, and antibiotic or antifungal bead placement. The spectrum of osteomyelitis ranges from Staphylococcus aureus organisms to tumors; therefore, it is necessary to investigate every spectrum of the disease, and fungal infections should be considered differential even though they are a rare entity. Early diagnosis, surgical debridement, and proper antifungal treatment based on fungal species lead to better clinical outcomes and results.
Collapse
Affiliation(s)
| | - Udit Agrawal
- Paediatric Orthopaedics, King George's Medical University, Lucknow, IND
| | | | - Suresh Chand
- Paediatric Orthopaedics, King George's Medical University, Lucknow, IND
| | - Vikas Verma
- Paediatric Orthopaedics, King George's Medical University, Lucknow, IND
| |
Collapse
|
21
|
Voss JO, Heiland M, Preissner R, Preissner S. The risk of osteomyelitis after mandibular fracture is doubled in men versus women: analysis of 300,000 patients. Sci Rep 2023; 13:20871. [PMID: 38012360 PMCID: PMC10682452 DOI: 10.1038/s41598-023-48235-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Accepted: 11/23/2023] [Indexed: 11/29/2023] Open
Abstract
Postoperative complications following mandibular fracture treatment vary from local wound infections to severe conditions including osteomyelitis and impaired fracture healing. Several risk factors have been associated with the development healing disorders, including fracture localisation, treatment modality and substance abuse. However, limited research on the sex-specific influence of these complications exists. A total of about 300,000 female and male patients with mandibular fractures were examined in two cohorts. After matching for confounders (age, nicotine and alcohol dependence, malnutrition, overweight, anaemia, diabetes, osteoporosis and vitamin D deficiency), two cohorts were compared with propensity-score-matched patients according to outcomes (osteomyelitis, pseudoarthrosis and disruption of the wound) within 1 year after fracture. There were significant differences between female and male patients regarding the occurrence of osteomyelitis (odds ratio [OR] [95% confidence interval]: 0.621 [0.563; 0.686]) and disruption of the wound (OR [95% confidence interval]: 0.703 [0.632; 0.782]). Surprisingly, matching for the expected confounders did not change the results substantially. Sex plays a dominant role in determining the risk stratification for postoperative osteomyelitis and disruption of the wound, after accounting for other potential confounding factors. Additional research is needed to understand the underlying mechanisms and to develop sex-specific strategies to prevent these complications.
Collapse
Affiliation(s)
- Jan Oliver Voss
- Department of Oral and Maxillofacial Surgery, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität Zu Berlin and Berlin Institute of Health, Augustenburger Platz 1, 13353, Berlin, Germany.
- Berlin Institute of Health (BIH), Anna-Louisa-Karsch-Straße 2, 10178, Berlin, Germany.
| | - Max Heiland
- Department of Oral and Maxillofacial Surgery, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität Zu Berlin and Berlin Institute of Health, Augustenburger Platz 1, 13353, Berlin, Germany
| | - Robert Preissner
- Institute of Physiology and Science-IT, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität Zu Berlin and Berlin Institute of Health, Philippstr. 12, 10115, Berlin, Germany
| | - Saskia Preissner
- Department of Oral and Maxillofacial Surgery, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität Zu Berlin and Berlin Institute of Health, Augustenburger Platz 1, 13353, Berlin, Germany
| |
Collapse
|
22
|
Mariano LC, Grenho L, Fernandes MH, de Sousa Gomes P. Integrative tissue, cellular and molecular responsiveness of an innovative ex vivo model of the Staphylococcus aureus-mediated bone infection. FASEB J 2023; 37:e23166. [PMID: 37650876 DOI: 10.1096/fj.202300287rr] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Revised: 07/21/2023] [Accepted: 08/14/2023] [Indexed: 09/01/2023]
Abstract
Osteomyelitis is a pathological condition of the bone, frequently associated with the presence of infectious agents - namely Staphylococcus aureus - that induce inflammation and tissue destruction. Recent advances in the understanding of its pathophysiology and the identification of innovative therapeutic approaches were gathered from experimental in vitro and in vivo systems. However, cell culture models offer limited representativeness of the cellular functionality and the cell-cell and cell-matrix interactions, further failing to mimic the three-dimensional tissue organization; and animal models allow for limited mechanistic assessment given the complex nature of systemic and paracrine regulatory systems and are endorsed with ethical constraints. Accordingly, this study aims at the establishment and assessment of a new ex vivo bone infection model, upon the organotypic culture of embryonic chicken femurs colonized with S. aureus, highlighting the model responsiveness at the molecular, cellular, and tissue levels. Upon infection with distinct bacterial inoculums, data reported an initial exponential bacterial growth, followed by diminished metabolic activity. At the tissue level, evidence of S. aureus-mediated tissue destruction was attained and demonstrated through distinct methodologies, conjoined with decreased osteoblastic/osteogenic and increased osteoclastic/osteoclastogenic functionalities-representative of the osteomyelitis clinical course. Overall, the establishment and characterization of an innovative bone tissue infection model that is simple, reproducible, easily manipulated, cost-effective, and simulates many features of human osteomyelitis, further allowing the maintenance of the bone tissue's three-dimensional morphology and cellular arrangement, was achieved. Model responsiveness was further demonstrated, showcasing the capability to improve the research pipeline in bone tissue infection-related research.
Collapse
Affiliation(s)
- Lorena Castro Mariano
- BoneLab-Laboratory for Bone Metabolism and Regeneration, Faculty of Dental Medicine, University of Porto, Porto, Portugal
- LAQV/REQUIMTE, University of Porto, Porto, Portugal
| | - Liliana Grenho
- BoneLab-Laboratory for Bone Metabolism and Regeneration, Faculty of Dental Medicine, University of Porto, Porto, Portugal
- LAQV/REQUIMTE, University of Porto, Porto, Portugal
| | - Maria Helena Fernandes
- BoneLab-Laboratory for Bone Metabolism and Regeneration, Faculty of Dental Medicine, University of Porto, Porto, Portugal
- LAQV/REQUIMTE, University of Porto, Porto, Portugal
| | - Pedro de Sousa Gomes
- BoneLab-Laboratory for Bone Metabolism and Regeneration, Faculty of Dental Medicine, University of Porto, Porto, Portugal
- LAQV/REQUIMTE, University of Porto, Porto, Portugal
| |
Collapse
|
23
|
Jothipandiyan S, Suresh D, Sekaran S, Paramasivam N. Palladium(II) Metal Complex Fabricated Titanium Implant Mitigates Dual-Species Biofilms in Artificial Synovial Fluid. Antibiotics (Basel) 2023; 12:1296. [PMID: 37627716 PMCID: PMC10451766 DOI: 10.3390/antibiotics12081296] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Revised: 08/03/2023] [Accepted: 08/05/2023] [Indexed: 08/27/2023] Open
Abstract
Metallodrugs have a potent application in various medical fields. In the current study, we used a novel Palladium(II) thiazolinyl picolinamide complex that was directly fabricated over the titanium implant to examine its potency in inhibiting dual-species biofilms and exopolysaccharides. Additionally, inhibition of mono- and dual-species biofilms by coated titanium plates in an in vitro joint microcosm was performed. The study was carried out for 7 days by cultivating mono- and dual-species biofilms on titanium plates placed in both growth media and artificial synovial fluid (ASF). By qPCR analysis, the interaction of co-cultured biofilms in ASF and the alteration in gene expression of co-cultured biofilms were studied. Remarkable alleviation of biofilm accumulation and EPS secretion was observed on the coated titanium plates. The effective impairment of biofilms and EPS matrix of biofilms on Pd(II)-E-coated titanium plates were visualized by Scanning Electron Microscopy. Moreover, coated titanium plates improved the adhesion of osteoblast cells, which is crucial for a bone biomaterial. The potential bioactivity of coated plates was also confirmed at the molecular level using qPCR analysis. The stability of coated plates in ASF for 7 days was examined with FESEM-EDAX analysis. Collectively, the present study provided an excellent anti-infective effect on Pd(II)-E-coated titanium plates without affecting their biocompatibility with bone cells.
Collapse
Affiliation(s)
- Sowndarya Jothipandiyan
- Biofilm Biology Laboratory, Centre for Research on Infectious Diseases (CRID), School of Chemical and Biotechnology, SASTRA Deemed University, Tirumalaisamudram, Thanjavur 613 401, Tamil Nadu, India;
| | - Devarajan Suresh
- Department of Chemistry, School of Chemical and Biotechnology, SASTRA Deemed University, Thanjavur 613 401, Tamil Nadu, India;
| | - Saravanan Sekaran
- Department of Prosthodontics, Saveetha Dental College and Hospitals, Saveetha Institute for Medical and Technical Sciences, Chennai 600 077, Tamil Nadu, India
| | - Nithyanand Paramasivam
- Biofilm Biology Laboratory, Centre for Research on Infectious Diseases (CRID), School of Chemical and Biotechnology, SASTRA Deemed University, Tirumalaisamudram, Thanjavur 613 401, Tamil Nadu, India;
| |
Collapse
|
24
|
Kebriaei R, Lehman SM, Shah RM, Stamper KC, Kunz Coyne AJ, Holger D, El Ghali A, Rybak MJ. Optimization of Phage-Antibiotic Combinations against Staphylococcus aureus Biofilms. Microbiol Spectr 2023; 11:e0491822. [PMID: 37199616 PMCID: PMC10269792 DOI: 10.1128/spectrum.04918-22] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Accepted: 04/21/2023] [Indexed: 05/19/2023] Open
Abstract
Phage therapy has gained attention due to the spread of antibiotic-resistant bacteria and narrow pipeline of novel antibiotics. Phage cocktails are hypothesized to slow the overall development of resistance by challenging the bacteria with more than one phage. Here, we have used a combination of plate-, planktonic-, and biofilm-based screening assays to try to identify phage-antibiotic combinations that will eradicate preformed biofilms of Staphylococcus aureus strains that are otherwise difficult to kill. We have focused on methicillin-resistant S aureus (MRSA) strains and their daptomycin-nonsusceptible vancomycin-intermediate (DNS-VISA) derivatives to understand whether the phage-antibiotic interactions are altered by the changes associated with evolution from MRSA to DNS-VISA (which is known to occur in patients receiving antibiotic therapy). We evaluated the host range and cross-resistance patterns of five obligately lytic S. aureus myophages to select a three-phage cocktail. We screened these phages for their activity against 24-h bead biofilms and found that biofilms of two strains, D712 (DNS-VISA) and 8014 (MRSA), were the most resistant to killing by single phages. Specifically, even initial phage concentrations of 107 PFU per well could not prevent visible regrowth of bacteria from the treated biofilms. However, when we treated biofilms of the same two strains with phage-antibiotic combinations, we prevented bacterial regrowth when using up to 4 orders of magnitude less phage and antibiotic concentrations that were lower than our measured minimum biofilm inhibitory concentration. We did not see a consistent association between phage activity and the evolution of DNS-VISA genotypes in this small number of bacterial strains. IMPORTANCE The extracellular polymeric matrix of biofilms presents an impediment to antibiotic diffusion, facilitating the emergence of multidrug-resistant populations. While most phage cocktails are designed for the planktonic state of bacteria, it is important to take the biofilm mode of growth (the predominant mode of bacterial growth in nature) into consideration, as it is unclear how interactions between any specific phage and its bacterial hosts will depend on the physical properties of the growth environment. In addition, the extent of bacterial sensitivity to any given phage may vary from the planktonic to the biofilm state. Therefore, phage-containing treatments targeting biofilm infections such as catheters and prosthetic joint material may not be merely based on host range characteristics. Our results open avenues to new questions regarding phage-antibiotic treatment efficiency in the eradication of topologically structured biofilm settings and the extent of eradication efficacy relative to the single agents in biofilm populations.
Collapse
Affiliation(s)
- Razieh Kebriaei
- Anti-Infective Research Laboratory, College of Pharmacy and Health Sciences, Wayne State University, Detroit, Michigan, USA
| | - Susan M. Lehman
- Center for Biologics Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, Maryland, USA
| | - Rahi M. Shah
- Anti-Infective Research Laboratory, College of Pharmacy and Health Sciences, Wayne State University, Detroit, Michigan, USA
| | - Kyle C. Stamper
- Anti-Infective Research Laboratory, College of Pharmacy and Health Sciences, Wayne State University, Detroit, Michigan, USA
| | - Ashlan J. Kunz Coyne
- Anti-Infective Research Laboratory, College of Pharmacy and Health Sciences, Wayne State University, Detroit, Michigan, USA
| | - Dana Holger
- Anti-Infective Research Laboratory, College of Pharmacy and Health Sciences, Wayne State University, Detroit, Michigan, USA
| | - Amer El Ghali
- Anti-Infective Research Laboratory, College of Pharmacy and Health Sciences, Wayne State University, Detroit, Michigan, USA
| | - Michael J. Rybak
- Anti-Infective Research Laboratory, College of Pharmacy and Health Sciences, Wayne State University, Detroit, Michigan, USA
- School of Medicine, Wayne State University, Detroit, Michigan, USA
| |
Collapse
|
25
|
Wang X, Zhang M, Zhu T, Wei Q, Liu G, Ding J. Flourishing Antibacterial Strategies for Osteomyelitis Therapy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2206154. [PMID: 36717275 PMCID: PMC10104653 DOI: 10.1002/advs.202206154] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 12/05/2022] [Indexed: 06/18/2023]
Abstract
Osteomyelitis is a destructive disease of bone tissue caused by infection with pathogenic microorganisms. Because of the complex and long-term abnormal conditions, osteomyelitis is one of the refractory diseases in orthopedics. Currently, anti-infective therapy is the primary modality for osteomyelitis therapy in addition to thorough surgical debridement. However, bacterial resistance has gradually reduced the benefits of traditional antibiotics, and the development of advanced antibacterial agents has received growing attention. This review introduces the main targets of antibacterial agents for treating osteomyelitis, including bacterial cell wall, cell membrane, intracellular macromolecules, and bacterial energy metabolism, focuses on their mechanisms, and predicts prospects for clinical applications.
Collapse
Affiliation(s)
- Xukai Wang
- Department of Thoracic SurgeryChina‐Japan Union Hospital of Jilin University126 Xiantai StreetChangchun130033P. R. China
- Key Laboratory of Polymer EcomaterialsChangchun Institute of Applied ChemistryChinese Academy of Sciences5625 Renmin StreetChangchun130022P. R. China
| | - Mingran Zhang
- Department of Thoracic SurgeryChina‐Japan Union Hospital of Jilin University126 Xiantai StreetChangchun130033P. R. China
| | - Tongtong Zhu
- Department of Thoracic SurgeryChina‐Japan Union Hospital of Jilin University126 Xiantai StreetChangchun130033P. R. China
| | - Qiuhua Wei
- Department of Disinfection and Infection ControlChinese PLA Center for Disease Control and Prevention20 Dongda StreetBeijing100071P. R. China
| | - Guangyao Liu
- Department of Thoracic SurgeryChina‐Japan Union Hospital of Jilin University126 Xiantai StreetChangchun130033P. R. China
| | - Jianxun Ding
- Key Laboratory of Polymer EcomaterialsChangchun Institute of Applied ChemistryChinese Academy of Sciences5625 Renmin StreetChangchun130022P. R. China
| |
Collapse
|
26
|
Pashapour S, Seneca S, Schröter M, Frischknecht F, Platzman I, Spatz J. Design and Development of Extracellular Matrix Protein-Based Microcapsules as Tools for Bacteria Investigation. Adv Healthc Mater 2023; 12:e2202789. [PMID: 36599129 PMCID: PMC11468930 DOI: 10.1002/adhm.202202789] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2022] [Revised: 12/19/2022] [Indexed: 01/06/2023]
Abstract
The extracellular matrix (ECM) plays an immense role in the homeostasis of tissues and organs, can function as a barrier for infectious agents, but is also exploited by pathogens during infection. Therefore, the development of well-defined 3D ECM models in the form of microcapsules to elucidate the interactions between ECM components and pathogens in confinement and study disease infectivity is important, albeit challenging. Current limitations are mainly attributed to the lack of biocompatible methods for the production of protein-based microcapsules. Herein, hollow ECM-based microcapsules from laminin-111 or laminin-111/collagen IV are generated to investigate the behavior of organisms within confined 3D extracellular matrices. Microcapsules are created using water-in-oil emulsion droplets stabilized by block copolymer surfactants as templates for the charge-mediated attraction of laminin or laminin-collagen proteins to the droplets' inner periphery, allowing for the formation of modular ECM-based microcapsules with tunable biophysical and biochemical properties and organism encapsulation. The release of E. coli-laden ECM-based protein microcapsules into a physiological environment revealed differences in the dynamic behavior of E. coli depending on the constitution of the surrounding ECM protein matrix. The developed ECM-based protein microcapsules have the potential to be implemented in several biomedical applications, including the design of in vitro infection models.
Collapse
Affiliation(s)
- Sadaf Pashapour
- Department of Cellular BiophysicsMax Planck Institute for Medical ResearchJahnstraße 29D‐69120HeidelbergGermany
- Institute for Molecular Systems Engineering and Advanced Materials (IMSEAM)Heidelberg UniversityIm Neuenheimer Feld 225D‐69120HeidelbergGermany
| | - Senne Seneca
- Department of Cellular BiophysicsMax Planck Institute for Medical ResearchJahnstraße 29D‐69120HeidelbergGermany
- Institute for Molecular Systems Engineering and Advanced Materials (IMSEAM)Heidelberg UniversityIm Neuenheimer Feld 225D‐69120HeidelbergGermany
| | - Martin Schröter
- Department of Cellular BiophysicsMax Planck Institute for Medical ResearchJahnstraße 29D‐69120HeidelbergGermany
- Institute for Molecular Systems Engineering and Advanced Materials (IMSEAM)Heidelberg UniversityIm Neuenheimer Feld 225D‐69120HeidelbergGermany
- Department of Chemistry and Earth SciencesHeidelberg UniversityIm Neuenheimer Feld 225D‐69120HeidelbergGermany
| | - Friedrich Frischknecht
- Center for Infectious DiseasesHeidelberg University Medical SchoolIm Neuenheimer Feld 344D‐69120HeidelbergGermany
- German Center for Infection ResearchDZIFPartner Site HeidelbergIm Neuenheimer Feld 344D‐69120HeidelbergGermany
| | - Ilia Platzman
- Department of Cellular BiophysicsMax Planck Institute for Medical ResearchJahnstraße 29D‐69120HeidelbergGermany
- Institute for Molecular Systems Engineering and Advanced Materials (IMSEAM)Heidelberg UniversityIm Neuenheimer Feld 225D‐69120HeidelbergGermany
| | - Joachim Spatz
- Department of Cellular BiophysicsMax Planck Institute for Medical ResearchJahnstraße 29D‐69120HeidelbergGermany
- Institute for Molecular Systems Engineering and Advanced Materials (IMSEAM)Heidelberg UniversityIm Neuenheimer Feld 225D‐69120HeidelbergGermany
- Max Planck School Matter to LifeJahnstraße 29D‐69120HeidelbergGermany
| |
Collapse
|
27
|
Borges MHR, Nagay BE, Costa RC, Souza JGS, Mathew MT, Barão VAR. Recent advances of polypyrrole conducting polymer film for biomedical application: Toward a viable platform for cell-microbial interactions. Adv Colloid Interface Sci 2023; 314:102860. [PMID: 36931199 DOI: 10.1016/j.cis.2023.102860] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Revised: 02/04/2023] [Accepted: 02/13/2023] [Indexed: 02/18/2023]
Abstract
Polypyrrole (PPy) is one of the most studied conductive polymers due to its electrical conductivity and biological properties, which drive the possibility of numerous applications in the biomedical area. The physical-chemical features of PPy allow the manufacture of biocompatible devices, enhancing cell adhesion and proliferation. Furthermore, owing to the electrostatic interactions between the negatively charged bacterial cell wall and the positive charges in the polymer structure, PPy films can perform an effective antimicrobial activity. PPy is also frequently associated with biocompatible agents and antimicrobial compounds to improve the biological response. Thus, this comprehensive review appraised the available evidence regarding the PPy-based films deposited on metallic implanted devices for biomedical applications. We focus on understanding key concepts that could influence PPy attributes regarding antimicrobial effect and cell behavior under in vitro and in vivo settings. Furthermore, we unravel the several agents incorporated into the PPy film and strategies to improve its functionality. Our findings suggest that incorporating other elements into the PPy films, such as antimicrobial agents, biomolecules, and other biocompatible polymers, may improve the biological responses. Overall, the basic properties of PPy, when combined with other composites, electrostimulation techniques, or surface treatment methods, offer great potential in biocompatibility and/or antimicrobial activities. However, challenges in synthesis standardization and potential limitations such as low adhesion and mechanical strength of the film must be overcome to improve and broaden the application of PPy film in biomedical devices.
Collapse
Affiliation(s)
- Maria H R Borges
- Department of Prosthodontics and Periodontology, Piracicaba Dental School, University of Campinas (UNICAMP), Piracicaba, São Paulo 13414-903, Brazil
| | - Bruna E Nagay
- Department of Prosthodontics and Periodontology, Piracicaba Dental School, University of Campinas (UNICAMP), Piracicaba, São Paulo 13414-903, Brazil
| | - Raphael C Costa
- Department of Prosthodontics and Periodontology, Piracicaba Dental School, University of Campinas (UNICAMP), Piracicaba, São Paulo 13414-903, Brazil
| | - João Gabriel S Souza
- Dental Research Division, Guarulhos University (UNG), Guarulhos, Sāo Paulo 07023-070, Brazil; Dental Science School (Faculdade de Ciências Odontológicas - FCO), Montes Claros, Minas Gerais 39401-303, Brazil
| | - Mathew T Mathew
- Department of Biomedical Sciences, University of Illinois, College of Medicine, Rockford, IL 61107, USA
| | - Valentim A R Barão
- Department of Prosthodontics and Periodontology, Piracicaba Dental School, University of Campinas (UNICAMP), Piracicaba, São Paulo 13414-903, Brazil.
| |
Collapse
|
28
|
Guo X. Antibacterial and anti-inflammatory effects of genistein in Staphylococcus aureus induced osteomyelitis in rats. J Biochem Mol Toxicol 2023; 37:e23298. [PMID: 36727417 DOI: 10.1002/jbt.23298] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 10/29/2022] [Accepted: 01/05/2023] [Indexed: 02/03/2023]
Abstract
Methicillin-resistant Staphylococcus aureus (MRSA) is a highly infectious Gram-positive pathogen known to cause severe diseases such as endocarditis, food poisoning, pneumonia, osteomyelitis, and septicemia. MRSA is a major public health issue. Among these, osteomyelitis is inflammation of the bone caused by the invasion of the bacterial pathogen in the bones. Its prominent symptoms include fever, pain, and redness of bones. In the case of children, it affects the long bones of arms and legs, whereas in the case of adults it affects the hip, feet, and spine. Bacterial osteomyelitis can trigger pathological remodeling of bones and hence causes substantial morbidity and mortality. The present study aims to evaluate the isoflavone genistein's (5,7-dihydroxy-3-(4-hydroxyphenyl)-4H-1-benzopyran-4-one,4',5,7 trihydroxyisoflavone) antimicrobial and anti-inflammatory effects against osteomyelitis induced by MRSA in male Wistar rats. Classification of the animals was into the following: sham (Group I), osteomyelitis (Group II, control), genistein (25 mg/kg body weight, Group III), and genistein (50 mg/kg body weight, Group IV). The rats did not receive any treatment for 4 weeks after bacterial inoculation. Genistein was then administered twice daily for 2 weeks. Bacterial growth, mean body weight bone infection status, and side effects of genistein treatment were assessed. Furthermore, lipid peroxidation, superoxide dismutase, glutathione (GSH) peroxidase, catalase, reduced GSH, tumor necrosis factor-α (TNF-α), and interleukin (IL)-6 were also determined. Two days after treatment, it was found that genistein significantly suppressed bacterial growth and reduced serum pro-inflammatory cytokines TNF-α and IL-6. Therefore, the study suggests that genistein could be a promising lead against MRSA-induced osteomyelitis.
Collapse
Affiliation(s)
- Xinyi Guo
- Department of Orthopaedic Surgery, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hosptial of Shanxi Medical University, Taiyuan, China.,Department of Orthopaedic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
29
|
Zhang B, Lu D, Duan H. Recent advances in responsive antibacterial materials: design and application scenarios. Biomater Sci 2023; 11:356-379. [PMID: 36408610 DOI: 10.1039/d2bm01573k] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Bacterial infection is one of the leading causes of death globally, although modern medicine has made considerable strides in the past century. As traditional antibiotics are suffering from the emergence of drug resistance, new antibacterial strategies are of great interest. Responsive materials are appealing alternatives that have shown great potential in combating resistant bacteria and avoiding the side effects of traditional antibiotics. In this review, the responsive antibacterial materials are introduced in terms of stimulus signals including intrinsic (pH, enzyme, ROS, etc.) and extrinsic (light, temperature, magnetic fields, etc.) stimuli. Their biomedical applications in therapeutics and medical devices are then discussed. Finally, the author's perspective of the challenge and the future of such a system is provided.
Collapse
Affiliation(s)
- Bo Zhang
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, 70 Nanyang Drive, Singapore 637457, Singapore.
| | - Derong Lu
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, 70 Nanyang Drive, Singapore 637457, Singapore.
| | - Hongwei Duan
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, 70 Nanyang Drive, Singapore 637457, Singapore.
| |
Collapse
|
30
|
Ashar H, Singh A, Ektate K, More S, Ranjan A. Treating methicillin-resistant Staphylococcus aureus (MRSA) bone infection with focused ultrasound combined thermally sensitive liposomes. Int J Hyperthermia 2023; 40:2211278. [PMID: 37437891 DOI: 10.1080/02656736.2023.2211278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 05/02/2023] [Accepted: 05/02/2023] [Indexed: 07/14/2023] Open
Abstract
OBJECTIVE Chronic bone infection caused by Staphylococcus aureus biofilms in children and adults is characterized by reduced antibiotic sensitivity. In this study, we assessed 'heat-targeted, on-demand' antibiotic delivery for S. aureus killing by combining ciprofloxacin (CIP)-laden low-temperature sensitive liposomes (LTSLs) with local high-intensity focused ultrasound (HIFU) induced bone heating in a rat model of bone infection. METHODS CIP-LTSLs were prepared using the thin-film hydration and extrusion method. Bone infection was established by surgically implanting an orthopedic K-wire colonized with methicillin-resistant S. aureus (MRSA) strain into rat's femurs. For bone heating, ultrasound-guided HIFU exposures were performed to achieve a local temperature of 40-42 °C (∼15 min) concurrently with intravenous injection of CIP-LTSLs or CIP. CIP biodistribution was determined spectrophotometrically and therapeutic efficacy was determined by bacteriological, histological and scanning electron microscopy (SEM) analyses. RESULTS CIP-LTSLs in the range of 183.5 nm ± 1.91 showed an encapsulation efficiency of >70% at 37 °C and a complete release at ∼42 °C. The metal implantation method yielded medullary osteomyelitis characterized by suppurative changes (bacterial and pus pockets) by day 10 in bones and adjoining muscle tissues. HIFU heating significantly improved CIP delivery from LTSLs in bones, resulting in a significant reduction in MRSA load compared to HIFU and CIP alone groups. These were also verified by histology and SEM, wherein a distinct reduction in S. aureus population in the infected metal wires and tissues from the combinatorial therapy was noted. CONCLUSION HIFU improved CIP delivery to bones, achieving clearance of hard-to-treat MRSA biofilms.
Collapse
Affiliation(s)
- Harshini Ashar
- Department of Physiological Sciences, College of Veterinary Medicine, OK State University, Stillwater, OK, USA
| | - Akansha Singh
- Department of Physiological Sciences, College of Veterinary Medicine, OK State University, Stillwater, OK, USA
| | - Kalyani Ektate
- Department of Physiological Sciences, College of Veterinary Medicine, OK State University, Stillwater, OK, USA
| | - Sunil More
- Department of Veterinary Pathobiology, College of Veterinary Medicine, Oklahoma State University, Stillwater, OK, USA
| | - Ashish Ranjan
- Department of Physiological Sciences, College of Veterinary Medicine, OK State University, Stillwater, OK, USA
| |
Collapse
|
31
|
Costa RC, Nagay BE, Dini C, Borges MHR, Miranda LFB, Cordeiro JM, Souza JGS, Sukotjo C, Cruz NC, Barão VAR. The race for the optimal antimicrobial surface: perspectives and challenges related to plasma electrolytic oxidation coating for titanium-based implants. Adv Colloid Interface Sci 2023; 311:102805. [PMID: 36434916 DOI: 10.1016/j.cis.2022.102805] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 10/01/2022] [Accepted: 10/20/2022] [Indexed: 01/24/2023]
Abstract
Plasma electrolytic oxidation (PEO) is a low-cost, structurally reliable, and environmentally friendly surface modification method for orthopedic and dental implants. This technique is successful for the formation of porous, corrosion-resistant, and bioactive coatings, besides introducing antimicrobial compounds easily. Given the increase in implant-related infections, antimicrobial PEO-treated surfaces have been widely proposed to surmount this public health concern. This review comprehensively discusses antimicrobial implant surfaces currently produced by PEO in terms of their in vitro and in vivo microbiological and biological properties. We present a critical [part I] and evidence-based [part II] review about the plethora of antimicrobial PEO-treated surfaces. The mechanism of microbial accumulation on implanted devices and the principles of PEO technology to ensure antimicrobial functionalization by one- or multi-step processes are outlined. Our systematic literature search showed that particular focus has been placed on the metallic and semi-metallic elements incorporated into PEO surfaces to facilitate antimicrobial properties, which are often dose-dependent, without leading to cytotoxicity in vitro. Meanwhile, there are concerns over the biocompatibility of PEO and its long-term antimicrobial effects in animal models. We clearly highlight the importance of using clinically relevant infection models and in vivo long-term assessments to guarantee the rational design of antimicrobial PEO-treated surfaces to identify the 'finish line' in the race for antimicrobial implant surfaces.
Collapse
Affiliation(s)
- Raphael C Costa
- Department of Prosthodontics and Periodontology, Piracicaba Dental School, University of Campinas (UNICAMP), Piracicaba, Sāo Paulo 13414-903, Brazil
| | - Bruna E Nagay
- Department of Prosthodontics and Periodontology, Piracicaba Dental School, University of Campinas (UNICAMP), Piracicaba, Sāo Paulo 13414-903, Brazil
| | - Caroline Dini
- Department of Prosthodontics and Periodontology, Piracicaba Dental School, University of Campinas (UNICAMP), Piracicaba, Sāo Paulo 13414-903, Brazil
| | - Maria H R Borges
- Department of Prosthodontics and Periodontology, Piracicaba Dental School, University of Campinas (UNICAMP), Piracicaba, Sāo Paulo 13414-903, Brazil
| | - Luís F B Miranda
- Department of Prosthodontics and Periodontology, Piracicaba Dental School, University of Campinas (UNICAMP), Piracicaba, Sāo Paulo 13414-903, Brazil
| | - Jairo M Cordeiro
- Department of Dentistry, Centro Universitário das Faculdades Associadas de Ensino (UNIFAE), Sāo Joāo da Boa Vista, Sāo Paulo 13870-377, Brazil
| | - Joāo G S Souza
- Dental Research Division, Guarulhos University, Guarulhos, Sāo Paulo 07023-070, Brazil; Dentistry Science School (Faculdade de Ciências Odontológicas - FCO), Montes Claros, Minas Gerais 39401-303, Brazil
| | - Cortino Sukotjo
- Department of Restorative Dentistry, University of Illinois at Chicago College of Dentistry, Chicago, IL 60612, USA
| | - Nilson C Cruz
- Laboratory of Technological Plasmas, Institute of Science and Technology, Sāo Paulo State University (UNESP), Sorocaba, Sāo Paulo 18087-180, Brazil
| | - Valentim A R Barão
- Department of Prosthodontics and Periodontology, Piracicaba Dental School, University of Campinas (UNICAMP), Piracicaba, Sāo Paulo 13414-903, Brazil.
| |
Collapse
|
32
|
Hexapeptide decorated β-cyclodextrin delivery system for targeted therapy of bone infection. J Control Release 2023; 353:337-349. [PMID: 36462641 DOI: 10.1016/j.jconrel.2022.11.047] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 11/25/2022] [Accepted: 11/28/2022] [Indexed: 12/12/2022]
Abstract
Successfully treating bone infections is a major orthopedic challenge. Clinically, oral, intravenous, or intramuscular injections of drugs are usually used for direct or complementary treatment. However, once the drug enters the system, it circulates throughout the body, leading to an insufficient local dose and limiting the therapeutic effect because of the lack of targeting in the drug system. In this study, β-cyclodextrin, modified with poly (ethylene glycol) [PEG] and aspartic acid hexapeptide (Asp6-β-CD), was used to specifically target the hydroxyapatite (HA) component of the bone. It was then loaded with norfloxacin (NFX) to treat bone infections. The antibacterial ability of NFX was enhanced by loading it into Asp6-β-CD, because the solubility of Asp6-β-CD@NFX increased significantly. Moreover, Asp6-β-CD could target bone tissue in nude mice and showed significantly enhanced accumulation (10 times) than the unmodified β-CD. In addition, in a rat model of osteomyelitis, Asp6-β-CD@NFX targeted HA well and exerted its antibacterial activity, which reduced inflammation and promoted bone tissue repair. This study indicates that the Asp6-β-CD based drug delivery system can efficiently target bone tissue to enable potential applications for treating bone-related diseases.
Collapse
|
33
|
Devascularized Bone Surface Culture: A Novel Strategy for Identifying Osteomyelitis-Related Pathogens. J Pers Med 2022; 12:jpm12122050. [PMID: 36556270 PMCID: PMC9781165 DOI: 10.3390/jpm12122050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Revised: 12/07/2022] [Accepted: 12/08/2022] [Indexed: 12/14/2022] Open
Abstract
The gold standard for identifying pathogens causing osteomyelitis (OM) is intraoperative tissue sampling culture (TSC). However, its positive rate remains inadequate. Here, we evaluated the efficiency of a novel strategy, known as devitalized bone surface culture (BSC), for detecting OM-related microorganisms and compared it to TSC. Between December 2021 and July 2022, patients diagnosed with OM and received both methods for bacterial identification were screened for analysis. In total, 51 cases were finally recruited for analysis. The mean age was 43.6 years, with the tibia as the top infection site. The positive rate of BSC was relatively higher than that of TSC (74.5% vs. 58.8%, p = 0.093), though no statistical difference was achieved. Both BSC and TSC detected definite pathogens in 29 patients, and their results were in accordance with each other. The most frequent microorganism identified by the BSC method was Staphylococcus aureus. Moreover, BSC took a significantly shorter median culture time than TSC (1.0 days vs. 3.0 days, p < 0.001). In summary, BSC may be superior to TSC for identifying OM-associated pathogens, with a higher detectable rate and a shorter culture time.
Collapse
|
34
|
González-Martín M, Silva V, Poeta P, Corbera JA, Tejedor-Junco MT. Microbiological aspects of osteomyelitis in veterinary medicine: drawing parallels to the infection in human medicine. Vet Q 2022; 42:1-11. [PMID: 34936853 PMCID: PMC8725753 DOI: 10.1080/01652176.2021.2022244] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Revised: 11/22/2021] [Accepted: 12/20/2021] [Indexed: 11/29/2022] Open
Abstract
Osteomyelitis is a challenging infectious disease affecting humans and animals. It is difficult to diagnose because, in many cases, symptoms are non-specific and, for example in implant-related cases, can appear long time after surgery. In addition to this, it is also difficult to treat due to the need to find the appropriate antibiotic regime and delivery system to reach the site of infection and to avoid development of bacterial resistance. The central purpose of this review is to compare the microbiological aspects of osteomyelitis in human and veterinary medicine, with the aim of improving the microbiological diagnosis and treatment of this infection in animals. Furthermore, the study of osteomyelitis in animals may help to improve the development of animal models for testing new treatments in humans. Host factors and underlying conditions have been studied mainly in humans, although aspects as immunodeficiency have been described in some veterinary cases. Even when Staphylococcus aureus is still considered the most prevalent causing microorganism, this prevalence should be reviewed using molecular diagnostic techniques, and this could affect treatment options. New approaches to treatment include local delivery of antibiotics using different biomaterials, antimicrobial photodynamic therapy, and new antimicrobial compounds. We would like to remark the need of large, high-quality clinical trials and of the development of guides for the diagnosis and treatment of osteomyelitis in different animal species.
Collapse
Affiliation(s)
- Margarita González-Martín
- Research Institute of Biomedical and Health Sciences, University of Las Palmas de Gran Canaria, Las Palmas de Gran Canaria, Canary Islands, Spain
- Department of Clinical Sciences, University of Las Palmas de Gran Canaria, Las Palmas de Gran Canaria, Canary Islands, Spain
| | - Vanessa Silva
- Microbiology and Antibiotic Resistance Team (MicroART), Department of Veterinary Sciences, University of Trás-os-Montes and Alto Douro (UTAD), Vila Real, Portugal
- Associated Laboratory for Green Chemistry (LAQV-REQUIMTE), University NOVA of Lisboa, Caparica, Portugal
- Veterinary and Animal Research Centre, Associate Laboratory for Animal and Veterinary Science (AL4AnimalS), University of Trás-os-Montes and Alto Douro (UTAD), Vila Real, Portugal
| | - Patricia Poeta
- Microbiology and Antibiotic Resistance Team (MicroART), Department of Veterinary Sciences, University of Trás-os-Montes and Alto Douro (UTAD), Vila Real, Portugal
- Associated Laboratory for Green Chemistry (LAQV-REQUIMTE), University NOVA of Lisboa, Caparica, Portugal
- Veterinary and Animal Research Centre, Associate Laboratory for Animal and Veterinary Science (AL4AnimalS), University of Trás-os-Montes and Alto Douro (UTAD), Vila Real, Portugal
| | - Juan Alberto Corbera
- Research Institute of Biomedical and Health Sciences, University of Las Palmas de Gran Canaria, Las Palmas de Gran Canaria, Canary Islands, Spain
- Department of Animal Pathology, Animal Production and Food Science and Technology, University of Las Palmas de Gran Canaria, Las Palmas de Gran Canaria, Canary Islands, Spain
| | - María Teresa Tejedor-Junco
- Research Institute of Biomedical and Health Sciences, University of Las Palmas de Gran Canaria, Las Palmas de Gran Canaria, Canary Islands, Spain
- Department of Clinical Sciences, University of Las Palmas de Gran Canaria, Las Palmas de Gran Canaria, Canary Islands, Spain
| |
Collapse
|
35
|
Granata V, Possetti V, Parente R, Bottazzi B, Inforzato A, Sobacchi C. The osteoblast secretome in Staphylococcus aureus osteomyelitis. Front Immunol 2022; 13:1048505. [PMID: 36483565 PMCID: PMC9723341 DOI: 10.3389/fimmu.2022.1048505] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Accepted: 11/03/2022] [Indexed: 11/23/2022] Open
Abstract
Osteomyelitis (OM) is an infectious disease of the bone predominantly caused by the opportunistic bacterium Staphylococcus aureus (S. aureus). Typically established upon hematogenous spread of the pathogen to the musculoskeletal system or contamination of the bone after fracture or surgery, osteomyelitis has a complex pathogenesis with a critical involvement of both osteal and immune components. Colonization of the bone by S. aureus is traditionally proposed to induce functional inhibition and/or apoptosis of osteoblasts, alteration of the RANKL/OPG ratio in the bone microenvironment and activation of osteoclasts; all together, these events locally subvert tissue homeostasis causing pathological bone loss. However, this paradigm has been challenged in recent years, in fact osteoblasts are emerging as active players in the induction and orientation of the immune reaction that mounts in the bone during an infection. The interaction with immune cells has been mostly ascribed to osteoblast-derived soluble mediators that add on and synergize with those contributed by professional immune cells. In this respect, several preclinical and clinical observations indicate that osteomyelitis is accompanied by alterations in the local and (sometimes) systemic levels of both pro-inflammatory (e.g., IL-6, IL-1α, TNF-α, IL-1β) and anti-inflammatory (e.g., TGF-β1) cytokines. Here we revisit the role of osteoblasts in bacterial OM, with a focus on their secretome and its crosstalk with cellular and molecular components of the bone microenvironment and immune system.
Collapse
Affiliation(s)
- Valentina Granata
- IRCCS Humanitas Research Hospital, Rozzano, Italy,Milan Unit, National Research Council - Institute for Genetic and Biomedical Research (CNR-IRGB), Milan, Italy
| | - Valentina Possetti
- IRCCS Humanitas Research Hospital, Rozzano, Italy,Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Italy
| | | | | | - Antonio Inforzato
- IRCCS Humanitas Research Hospital, Rozzano, Italy,Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Italy
| | - Cristina Sobacchi
- IRCCS Humanitas Research Hospital, Rozzano, Italy,Milan Unit, National Research Council - Institute for Genetic and Biomedical Research (CNR-IRGB), Milan, Italy,*Correspondence: Cristina Sobacchi,
| |
Collapse
|
36
|
Omar AE, Ahmed MM, Abd-Allah WM. Effect of Gamma Irradiation on Silica Nanoparticles for Ciprofloxacin Drug Delivery. SILICON 2022; 14:11171-11180. [DOI: 10.1007/s12633-022-01838-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Accepted: 03/20/2022] [Indexed: 09/02/2023]
|
37
|
Mohd Yusof N, Saleh AK, Abuomira IEAA, Attallah AA, Elshal EA, khames AAA. Mono-Lateral External Fixation for Treatment of Femoral Osteomyelitis. Orthop Res Rev 2022; 14:437-443. [DOI: 10.2147/orr.s383863] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Accepted: 10/21/2022] [Indexed: 11/23/2022] Open
|
38
|
Smith M, Roberts M, Al-Kassas R. Implantable drug delivery systems for the treatment of osteomyelitis. Drug Dev Ind Pharm 2022; 48:511-527. [PMID: 36222433 DOI: 10.1080/03639045.2022.2135729] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
Osteomyelitis is an infection of the bone tissue and bone marrow which is becoming increasingly difficult to treat due to the infection causing pathogens associated. Staphylococcus aureus is one of the main bacteria that causes this infection, which has a broad spectrum of antibiotic resistance making it extremely difficult to treat. Conventional metal implants used in orthopaedic applications often have the drawback of implant induced osteomyelitis as well as the requirement of a second surgery to remove the implant once it is no longer required. Recently, attention has been focused on the design and fabrication of biodegradable implants for the treatment of bone infection. The main benefit of biodegradable implants over polymethylmethacrylate (PMMA) based non-degradable systems is that they do not require a second surgery for removal and so making degradable implants safer and easier to use. The main purpose of a biodegradable implant is to provide the necessary support and conductivity to allow the bone to regenerate whilst themselves degrading at a rate that is compatible with the rate of formation of new bone. They must be highly biocompatible to ensure there is no inflammation or irritation within the surrounding tissue. During this review, the latest research into antibiotic loaded biodegradable implants will be explored. Their benefits and drawbacks will be compared with those non-degradable PMMA beads, which is the stable material used within antibiotic loaded implants. Biodegradable implants most frequently used are based on biodegradable natural and synthetic polymers. Implants can take the form of many different structures; the most commonly fabricated structure is a scaffold. Other structures that will be explored within this review are hydrogels, nanoparticles and surface coatings, all with their own benefits/drawbacks.
Collapse
Affiliation(s)
- Megan Smith
- School of Pharmacy and Biomolecular Sciences, Faculty of Science, Liverpool John Moores University, James Parsons Building, Byrom St, Liverpool, L3 3AF, UK
| | - Matthew Roberts
- School of Pharmacy and Biomolecular Sciences, Faculty of Science, Liverpool John Moores University, James Parsons Building, Byrom St, Liverpool, L3 3AF, UK
| | - Raida Al-Kassas
- School of Pharmacy and Biomolecular Sciences, Faculty of Science, Liverpool John Moores University, James Parsons Building, Byrom St, Liverpool, L3 3AF, UK
| |
Collapse
|
39
|
Jensen N, Jensen HE, Aalbaek B, Blirup-Plum SA, Soto SM, Cepas V, López Y, Gabasa Y, Gutiérrez-del-Río I, Villar CJ, Lombó F, Iglesias MJ, Soengas R, López Ortiz F, Jensen LK. Synthesis of the cyanobacterial halometabolite Chlorosphaerolactylate B and demonstration of its antimicrobial effect in vitro and in vivo. Front Microbiol 2022; 13:950855. [PMID: 36246241 PMCID: PMC9557163 DOI: 10.3389/fmicb.2022.950855] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Accepted: 08/18/2022] [Indexed: 12/02/2022] Open
Abstract
Chlorosphaerolactylate B, a newly discovered antimicrobial halometabolite from the cyanobacterium Sphaerospermopsis sp. LEGE 00249 has been synthesized in three steps by using 12-bromododecanoic acid as starting material. A total of 0.5 g was produced for in vitro and in vivo antimicrobial efficacy testing. In vitro, the minimal inhibitory concentration (MIC) was estimated to be 256 mg/L for Staphylococcus aureus, while the minimal biofilm inhibitory concentration (MBIC) was estimated to be 74 mg/L. The in vivo study utilized a porcine model of implant-associated osteomyelitis. In total, 12 female pigs were allocated into 3 groups based on inoculum (n = 4 in each group). An implant cavity (IC) was drilled in the right tibia and followed by inoculation and insertion of a steel implant. All pigs were inoculated with 10 μL containing either: 11.79 mg synthetic Chlorosphaerolactylate B + 104 CFU of S. aureus (Group A), 104 CFU of S. aureus (Group B), or pure saline (Group C), respectively. Pigs were euthanized five days after inoculation. All Group B animals showed macroscopic and microscopic signs of bone infection and both tissue and implant harbored S. aureus bacteria (mean CFU on implants = 1.9 × 105). In contrast, S. aureus could not be isolated from animals inoculated with saline. In Group A, two animals had a low number of S. aureus (CFU = 6.7 × 101 and 3.8 × 101, respectively) on the implants, otherwise all Group A animals were similar to Group C animals. In conclusion, synthetic Chlorosphaerolactylate B holds potential to be a novel antimicrobial and antibiofilm compound.
Collapse
Affiliation(s)
- Nikoline Jensen
- Section for Pathobiological Sciences, Department of Veterinary and Animal Science, University of Copenhagen, Copenhagen, Denmark
| | - Henrik Elvang Jensen
- Section for Pathobiological Sciences, Department of Veterinary and Animal Science, University of Copenhagen, Copenhagen, Denmark
| | - Bent Aalbaek
- Section for Pathobiological Sciences, Department of Veterinary and Animal Science, University of Copenhagen, Copenhagen, Denmark
| | - Sophie Amalie Blirup-Plum
- Section for Pathobiological Sciences, Department of Veterinary and Animal Science, University of Copenhagen, Copenhagen, Denmark
| | - Sara M. Soto
- ISGlobal, Hospital Clínic, Universitat de Barcelona, Barcelona, Spain
- CIBER Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, Madrid, Spain
| | - Virginio Cepas
- ISGlobal, Hospital Clínic, Universitat de Barcelona, Barcelona, Spain
| | - Yuly López
- ISGlobal, Hospital Clínic, Universitat de Barcelona, Barcelona, Spain
| | - Yaiza Gabasa
- ISGlobal, Hospital Clínic, Universitat de Barcelona, Barcelona, Spain
| | - Ignacio Gutiérrez-del-Río
- Biotechnology in Nutraceuticals and Bioactive Compounds-BIONUC, Department of Functional Biology, University of Oviedo, Oviedo, Spain
- Instituto Universitario de Oncología del Principado de Asturias (IUOPA), Oviedo, Spain
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Oviedo, Spain
| | - Claudio J. Villar
- Biotechnology in Nutraceuticals and Bioactive Compounds-BIONUC, Department of Functional Biology, University of Oviedo, Oviedo, Spain
- Instituto Universitario de Oncología del Principado de Asturias (IUOPA), Oviedo, Spain
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Oviedo, Spain
| | - Felipe Lombó
- Biotechnology in Nutraceuticals and Bioactive Compounds-BIONUC, Department of Functional Biology, University of Oviedo, Oviedo, Spain
- Instituto Universitario de Oncología del Principado de Asturias (IUOPA), Oviedo, Spain
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Oviedo, Spain
| | - María José Iglesias
- Instituto Universitario de Oncología del Principado de Asturias (IUOPA), Oviedo, Spain
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Oviedo, Spain
| | - Raquel Soengas
- Área de Química Orgánica, Centro de Investigación CIAIMBITAL, Universidad de Almería, Almería, Spain
| | - Fernando López Ortiz
- Área de Química Orgánica, Centro de Investigación CIAIMBITAL, Universidad de Almería, Almería, Spain
- *Correspondence: Fernando López Ortiz,
| | - Louise Kruse Jensen
- Section for Pathobiological Sciences, Department of Veterinary and Animal Science, University of Copenhagen, Copenhagen, Denmark
- Louise Kruse Jensen,
| |
Collapse
|
40
|
Lata J, Pansotra N. Osteomyelitis of Maxilla: A Rare Presentation Yet Not So Rare. J Maxillofac Oral Surg 2022; 21:1023-1031. [PMID: 36274895 PMCID: PMC9474749 DOI: 10.1007/s12663-021-01607-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Accepted: 06/12/2021] [Indexed: 11/26/2022] Open
Abstract
Objective To retrospectively study the patients diagnosed with osteomyelitis of jaw in Punjab Government Dental College and Hospital, Amritsar. Material and Methods A total of 21 case records of patients with osteomyelitis of jaw were analysed retrospectively from January 2018 to December 2020 at Department of oral and maxillofacial surgery, Punjab Government Dental College and Hospital, Amritsar, India. Results Of the 21 patients, maxilla was involved in 14 (66.6%) patients, whereas 6 patients (28.6%) had mandibular osteomyelitis and 1 patient (4.8%) had osteomyelitis of zygoma. 18 patients had underlying systemic disease with diabetes present in 10 patients (47.6%). History of alcoholism was present in 5 patients (23.8%) as one of the predisposing factors. 14 patients had odontogenic cause as predominant aetiology followed by sinusitis in 3 patients. Osteomyelitis due to post-herpetic and hematogenous infection, trauma and unknown aetiology was found in 1 patient each. With adequate medical and surgical intervention, most of our patients had satisfactory outcome. Conclusions High incidence of maxillary osteomyelitis was found in our study in contrary to previous studies and literature. As this is a small study, further prospective studies with longitudinal follow up of larger number of patients would be desirable to confirm these findings.
Collapse
Affiliation(s)
- Jeevan Lata
- Department of Oral and Maxillofacial Surgery, Punjab Government Dental College and Hospital, Amritsar, Punjab India
| | - Neetu Pansotra
- Department of Oral and Maxillofacial Surgery, Punjab Government Dental College and Hospital, Amritsar, Punjab India
| |
Collapse
|
41
|
Zelmer AR, Nelson R, Richter K, Atkins GJ. Can intracellular Staphylococcus aureus in osteomyelitis be treated using current antibiotics? A systematic review and narrative synthesis. Bone Res 2022; 10:53. [PMID: 35961964 PMCID: PMC9374758 DOI: 10.1038/s41413-022-00227-8] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 05/26/2022] [Accepted: 06/15/2022] [Indexed: 11/09/2022] Open
Abstract
Approximately 40% of treatments of chronic and recurrent osteomyelitis fail in part due to bacterial persistence. Staphylococcus aureus, the predominant pathogen in human osteomyelitis, is known to persist by phenotypic adaptation as small-colony variants (SCVs) and by formation of intracellular reservoirs, including those in major bone cell types, reducing susceptibility to antibiotics. Intracellular infections with S. aureus are difficult to treat; however, there are no evidence-based clinical guidelines addressing these infections in osteomyelitis. We conducted a systematic review of the literature to determine the demonstrated efficacy of all antibiotics against intracellular S. aureus relevant to osteomyelitis, including protein biosynthesis inhibitors (lincosamides, streptogramins, macrolides, oxazolidines, tetracyclines, fusidic acid, and aminoglycosides), enzyme inhibitors (fluoroquinolones and ansamycines), and cell wall inhibitors (beta-lactam inhibitors, glycopeptides, fosfomycin, and lipopeptides). The PubMed and Embase databases were screened for articles related to intracellular S. aureus infections that compared the effectiveness of multiple antibiotics or a single antibiotic together with another treatment, which resulted in 34 full-text articles fitting the inclusion criteria. The combined findings of these studies were largely inconclusive, most likely due to the plethora of methodologies utilized. Therefore, the reported findings in the context of the models employed and possible solutions for improved understanding are explored here. While rifampicin, oritavancin, linezolid, moxifloxacin and oxacillin were identified as the most effective potential intracellular treatments, the scientific evidence for these is still relatively weak. We advocate for more standardized research on determining the intracellular effectiveness of antibiotics in S. aureus osteomyelitis to improve treatments and patient outcomes.
Collapse
Affiliation(s)
- Anja R Zelmer
- Centre for Orthopaedic and Trauma Research, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, SA, 5000, Australia
| | - Renjy Nelson
- Department of Infectious Diseases, Central Adelaide Local Health Network, Adelaide, SA, 5000, Australia.,Royal Adelaide Hospital, Adelaide, SA, 5000, Australia
| | - Katharina Richter
- Richter Lab, Department of Surgery, Basil Hetzel Institute for Translational Health Research, University of Adelaide, Adelaide, SA, 5011, Australia
| | - Gerald J Atkins
- Centre for Orthopaedic and Trauma Research, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, SA, 5000, Australia.
| |
Collapse
|
42
|
Cordeiro JM, Barão VAR, de Avila ED, Husch JFA, Yang F, van den Beucken JJJP. Tailoring Cu 2+-loaded electrospun membranes with antibacterial ability for guided bone regeneration. BIOMATERIALS ADVANCES 2022; 139:212976. [PMID: 35882133 DOI: 10.1016/j.bioadv.2022.212976] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Revised: 05/23/2022] [Accepted: 06/04/2022] [Indexed: 06/15/2023]
Abstract
Copper (Cu)-loaded electrospun membranes were tailored for guided bone regeneration (GBR), targeting the stimulation of innate cells active in bone growth and the prevention of bacterial infections. Functional GBR membranes were produced via an electrospinning set-up using a silk-based solution associated with polyethylene oxide (Silk/PEO - control). Experimental groups were loaded with copper oxide using varying weight percentages (0.05 % to 1 % of CuO). The morphological, structural, chemical, and mechanical properties of membranes were evaluated. Direct and indirect in vitro cytocompatibility experiments were performed with primary human bone mesenchymal stem cells and primary human umbilical vein endothelial cells. The antibacterial potential of membranes was tested with Staphylococcus aureus and Fusobacterium nucleatum biofilm. CuO was successfully incorporated into membranes as clusters without compromising their mechanical properties for clinical applicability. Increased Cu concentrations generated membranes with thinner nanofibers, greater pore areas, and stronger antimicrobial effect (p < 0.01). Cu2+ ion was released from the nanofiber membranes during 1 week, showing higher release in acidic conditions. CuO 0.1 % and CuO 0.05 % membranes were able to support and stimulate cell adhesion and proliferation (p < 0.05), and favor angiogenic responses of vascular cells. In addition, detailed quantitative and qualitative analysis determined that amount of the attached biofilm was reduced on the tailored functional Cu2+-loaded GBR membrane. Importantly, these qualities represent a valuable strategy to improve the bone regeneration process and diminish the risk of bacterial infections.
Collapse
Affiliation(s)
- Jairo M Cordeiro
- Department of Prosthodontics and Periodontology, Piracicaba Dental School, University of Campinas (UNICAMP), Piracicaba, São Paulo, Brazil; Dentistry - Regenerative Biomaterials, Radboudumc, Nijmegen, the Netherlands.
| | - Valentim A R Barão
- Department of Prosthodontics and Periodontology, Piracicaba Dental School, University of Campinas (UNICAMP), Piracicaba, São Paulo, Brazil.
| | - Erica D de Avila
- Department of Dental Materials and Prosthodontics, School of Dentistry at Araraquara, São Paulo State University (UNESP), Araraquara, São Paulo, Brazil
| | - Johanna F A Husch
- Dentistry - Regenerative Biomaterials, Radboudumc, Nijmegen, the Netherlands
| | - Fang Yang
- Dentistry - Regenerative Biomaterials, Radboudumc, Nijmegen, the Netherlands
| | | |
Collapse
|
43
|
Radzieta M, Malone M, Ahmad M, Dickson HG, Schwarzer S, Jensen SO, Lavery LA. Metatranscriptome sequencing identifies Escherichia are major contributors to pathogenic functions and biofilm formation in diabetes related foot osteomyelitis. Front Microbiol 2022; 13:956332. [PMID: 35979499 PMCID: PMC9376677 DOI: 10.3389/fmicb.2022.956332] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Accepted: 07/11/2022] [Indexed: 11/26/2022] Open
Abstract
Osteomyelitis in the feet of persons with diabetes is clinically challenging and is associated with high rates of amputation. In this study RNA-sequencing was employed to explore microbial metatranscriptomes with a view to understand the relative activity and functions of the pathogen/s responsible for diabetes foot osteomyelitis (DFO). We obtained 25 intraoperative bone specimens from persons with confirmed DFO, observing that Escherichia spp. (7%), Streptomyces spp. (7%), Staphylococcus spp. (6%), Klebsiella spp. (5%) and Proteus spp. (5%) are the most active taxa on average. Data was then subset to examine functions associated with pathogenesis (virulence and toxins), biofilm formation and antimicrobial/multi-drug resistance. Analysis revealed Escherichia spp. are the most active taxa relative to pathogenic functions with K06218 (mRNA interferase relE), K03699 (membrane damaging toxin tlyC) and K03980 (putative peptidoglycan lipid II flippase murJ), K01114 (membrane damaging toxin plc) and K19168 (toxin cptA) being the most prevalent pathogenic associated transcripts. The most abundant transcripts associated with biofilm pathways included components of the biofilm EPS matrix including glycogen synthesis, cellulose synthesis, colonic acid synthesis and flagella synthesis. We further observed enrichment of a key enzyme involved in the biosynthesis of L-rhamnose (K01710 -dTDP-glucose 4,6-dehydratase rfbB, rmlB, rffG) which was present in all but four patients with DFO.
Collapse
Affiliation(s)
- Michael Radzieta
- South West Sydney Limb Preservation and Wound Research, South Western Sydney Local Health District (LHD), Sydney, NSW, Australia
- Infectious Diseases and Microbiology, School of Medicine, Western Sydney University, Sydney, NSW, Australia
| | - Matthew Malone
- South West Sydney Limb Preservation and Wound Research, South Western Sydney Local Health District (LHD), Sydney, NSW, Australia
- Infectious Diseases and Microbiology, School of Medicine, Western Sydney University, Sydney, NSW, Australia
- *Correspondence: Matthew Malone
| | - Mehtab Ahmad
- Department of Vascular Surgery, Liverpool Hospital, South Western Sydney Local Health District (LHD), Sydney, NSW, Australia
| | - Hugh G. Dickson
- South West Sydney Limb Preservation and Wound Research, South Western Sydney Local Health District (LHD), Sydney, NSW, Australia
- South Western Clinical School, University of New South Wales, Sydney, NSW, Australia
| | - Saskia Schwarzer
- South West Sydney Limb Preservation and Wound Research, South Western Sydney Local Health District (LHD), Sydney, NSW, Australia
- Infectious Diseases and Microbiology, School of Medicine, Western Sydney University, Sydney, NSW, Australia
| | - Slade O. Jensen
- South West Sydney Limb Preservation and Wound Research, South Western Sydney Local Health District (LHD), Sydney, NSW, Australia
- South Western Clinical School, University of New South Wales, Sydney, NSW, Australia
| | - Lawrence A. Lavery
- Department of Plastic Surgery, University of Texas Southwestern Medical Center, Dallas, TX, United States
| |
Collapse
|
44
|
Nanotechnology in the Diagnosis and Treatment of Osteomyelitis. Pharmaceutics 2022; 14:pharmaceutics14081563. [PMID: 36015188 PMCID: PMC9412360 DOI: 10.3390/pharmaceutics14081563] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Revised: 07/15/2022] [Accepted: 07/18/2022] [Indexed: 11/23/2022] Open
Abstract
Infection remains one of the largest threats to global health. Among those infections that are especially troublesome, osteomyelitis, or inflammation of the bone, typically due to infection, is a particularly difficult condition to diagnose and treat. This difficulty stems not only from the biological complexities of opportunistic infections designed to avoid the onslaught of both the host immune system as well as exogenous antibiotics, but also from changes in the host vasculature and the heterogeneity of infectious presentations. While several groups have attempted to classify and stage osteomyelitis, controversy remains, often delaying diagnosis and treatment. Despite a host of preclinical treatment advances being incubated in academic and company research and development labs worldwide, clinical treatment strategies remain relatively stagnant, including surgical debridement and lengthy courses of intravenous antibiotics, both of which may compromise the overall health of the bone and the patient. This manuscript reviews the current methods for diagnosing and treating osteomyelitis and then contemplates the role that nanotechnology might play in the advancement of osteomyelitis treatment.
Collapse
|
45
|
Kar AK, Singh A, Singh D, Shraogi N, Verma R, Saji J, Jagdale P, Ghosh D, Patnaik S. Biopolymeric composite hydrogel loaded with silver NPs and epigallocatechin gallate (EGCG) effectively manages ROS for rapid wound healing in type II diabetic wounds. Int J Biol Macromol 2022; 218:506-518. [PMID: 35817241 DOI: 10.1016/j.ijbiomac.2022.06.196] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Revised: 06/29/2022] [Accepted: 06/29/2022] [Indexed: 11/05/2022]
Abstract
Delayed wound healing in patients having type-II diabetes mellitus (T2DM) often results in a high rate of amputation. We report an innovative Guar Gum-based macroporous hydrogel (HG) infused with an antibacterial agent (Ag NPs), and antioxidant, epigallocatechin gallate (EGCG) to address rapid wound healing and interestingly could inhibit the associated pathophysical bone infection in a high-fat-diet-induced T2DM C57BL/6 mice model. The HG-Ag-EGCG elicits scar-free wound healing in subcutaneous wounds and histopathological evidence confirmed HG-Ag-EGCG hydrogel patch elicits better wound healing through enhanced cell proliferation, mature connecting tissue fiber formation, minimum void spaces formation, and better re-epithelialization when compared with a market available hydrogel patch material (Luofucon®). Supportive of the in vivo outcomes, in vitro experiments delineated better-wound closure due to improved management of ROS by the HG-Ag-EGCG. Additionally, a favorable non-toxicity outcome assessed through both in vitro and in vivo conditions confirmed its potential applicability in clinical wound care management.
Collapse
Affiliation(s)
- Aditya K Kar
- Water Analysis Laboratory, System Toxicology, and Health Risk Assessment Group, Vishvigyan Bhawan, 31, Mahatma Gandhi Marg, Lucknow 226001, Uttar Pradesh, India; CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Vishvigyan Bhawan, 31, Mahatma Gandhi Marg, Lucknow 226001, Uttar Pradesh, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Amrita Singh
- Water Analysis Laboratory, System Toxicology, and Health Risk Assessment Group, Vishvigyan Bhawan, 31, Mahatma Gandhi Marg, Lucknow 226001, Uttar Pradesh, India; CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Vishvigyan Bhawan, 31, Mahatma Gandhi Marg, Lucknow 226001, Uttar Pradesh, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Divya Singh
- Water Analysis Laboratory, System Toxicology, and Health Risk Assessment Group, Vishvigyan Bhawan, 31, Mahatma Gandhi Marg, Lucknow 226001, Uttar Pradesh, India; CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Vishvigyan Bhawan, 31, Mahatma Gandhi Marg, Lucknow 226001, Uttar Pradesh, India
| | - Nikita Shraogi
- Water Analysis Laboratory, System Toxicology, and Health Risk Assessment Group, Vishvigyan Bhawan, 31, Mahatma Gandhi Marg, Lucknow 226001, Uttar Pradesh, India; CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Vishvigyan Bhawan, 31, Mahatma Gandhi Marg, Lucknow 226001, Uttar Pradesh, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Rahul Verma
- Water Analysis Laboratory, System Toxicology, and Health Risk Assessment Group, Vishvigyan Bhawan, 31, Mahatma Gandhi Marg, Lucknow 226001, Uttar Pradesh, India; CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Vishvigyan Bhawan, 31, Mahatma Gandhi Marg, Lucknow 226001, Uttar Pradesh, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Joel Saji
- Water Analysis Laboratory, System Toxicology, and Health Risk Assessment Group, Vishvigyan Bhawan, 31, Mahatma Gandhi Marg, Lucknow 226001, Uttar Pradesh, India; CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Vishvigyan Bhawan, 31, Mahatma Gandhi Marg, Lucknow 226001, Uttar Pradesh, India
| | - Pankaj Jagdale
- Regulatory Toxicology Group, Vishvigyan Bhawan, 31, Mahatma Gandhi Marg, Lucknow 226001, Uttar Pradesh, India; CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Vishvigyan Bhawan, 31, Mahatma Gandhi Marg, Lucknow 226001, Uttar Pradesh, India
| | - Debabrata Ghosh
- Immunotoxicology laboratory, Food, Drug, and Chemical Toxicology Group, Vishvigyan Bhawan, 31, Mahatma Gandhi Marg, Lucknow, 226001, Uttar Pradesh, India; CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Vishvigyan Bhawan, 31, Mahatma Gandhi Marg, Lucknow 226001, Uttar Pradesh, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India.
| | - Satyakam Patnaik
- Water Analysis Laboratory, System Toxicology, and Health Risk Assessment Group, Vishvigyan Bhawan, 31, Mahatma Gandhi Marg, Lucknow 226001, Uttar Pradesh, India; CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Vishvigyan Bhawan, 31, Mahatma Gandhi Marg, Lucknow 226001, Uttar Pradesh, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India.
| |
Collapse
|
46
|
Yang Y, Li M, Zhou B, Jiang X, Zhang D, Luo H. Graphene oxide/gallium nanoderivative as a multifunctional modulator of osteoblastogenesis and osteoclastogenesis for the synergistic therapy of implant-related bone infection. Bioact Mater 2022; 25:594-614. [PMID: 37056253 PMCID: PMC10087081 DOI: 10.1016/j.bioactmat.2022.07.015] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 06/24/2022] [Accepted: 07/12/2022] [Indexed: 11/30/2022] Open
Abstract
Currently, implant-associated bacterial infections account for most hospital-acquired infections in patients suffering from bone fractures or defects. Poor osseointegration and aggravated osteolysis remain great challenges for the success of implants in infectious scenarios. Consequently, developing an effective surface modification strategy for implants is urgently needed. Here, a novel nanoplatform (GO/Ga) consisting of graphene oxide (GO) and gallium nanoparticles (GaNPs) was reported, followed by investigations of its in vitro antibacterial activity and potential bacterium inactivation mechanisms, cytocompatibility and regulatory actions on osteoblastogenesis and osteoclastogenesis. In addition, the possible molecular mechanisms underlying the regulatory effects of GO/Ga nanocomposites on osteoblast differentiation and osteoclast formation were clarified. Moreover, an in vivo infectious microenvironment was established in a rat model of implant-related femoral osteomyelitis to determine the therapeutic efficacy and biosafety of GO/Ga nanocomposites. Our results indicate that GO/Ga nanocomposites with excellent antibacterial potency have evident osteogenic potential and inhibitory effects on osteoclast differentiation by modulating the BMP/Smad, MAPK and NF-κB signaling pathways. The in vivo experiments revealed that the administration of GO/Ga nanocomposites significantly inhibited bone infections, reduced osteolysis, promoted osseointegration located in implant-bone interfaces, and resulted in satisfactory biocompatibility. In summary, this synergistic therapeutic system could accelerate the bone healing process in implant-associated infections and can significantly guide the future surface modification of implants used in bacteria-infected environments.
Collapse
Affiliation(s)
- Ying Yang
- Department of Plastic Surgery, Xiangya Hospital, Central South University, Changsha, 410008, China
- State Key Laboratory of Powder Metallurgy, Central South University, Changsha, 410083, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, China
- Corresponding author. Department of Plastic Surgery, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan, PR China.
| | - Min Li
- Department of Oncology, Changsha Central Hospital, University of South China, Changsha, 410006, China
| | - Bixia Zhou
- Department of Plastic Surgery, Xiangya Hospital, Central South University, Changsha, 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Xulei Jiang
- Department of Plastic Surgery, Xiangya Hospital, Central South University, Changsha, 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Dou Zhang
- State Key Laboratory of Powder Metallurgy, Central South University, Changsha, 410083, China
| | - Hang Luo
- State Key Laboratory of Powder Metallurgy, Central South University, Changsha, 410083, China
- Corresponding author. State Key Laboratory of Powder Metallurgy, Central South University, 932 South Lushan Road, Changsha, 410083, Hunan, China.
| |
Collapse
|
47
|
Nicolas A, Deplanche M, Commere PH, Diot A, Genthon C, Marques da Silva W, Azevedo V, Germon P, Jamme H, Guédon E, Le Loir Y, Laurent F, Bierne H, Berkova N. Transcriptome Architecture of Osteoblastic Cells Infected With Staphylococcus aureus Reveals Strong Inflammatory Responses and Signatures of Metabolic and Epigenetic Dysregulation. Front Cell Infect Microbiol 2022; 12:854242. [PMID: 35531332 PMCID: PMC9067450 DOI: 10.3389/fcimb.2022.854242] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Accepted: 03/03/2022] [Indexed: 11/21/2022] Open
Abstract
Staphylococcus aureus is an opportunistic pathogen that causes a range of devastating diseases including chronic osteomyelitis, which partially relies on the internalization and persistence of S. aureus in osteoblasts. The identification of the mechanisms of the osteoblast response to intracellular S. aureus is thus crucial to improve the knowledge of this infectious pathology. Since the signal from specifically infected bacteria-bearing cells is diluted and the results are confounded by bystander effects of uninfected cells, we developed a novel model of long-term infection. Using a flow cytometric approach we isolated only S. aureus-bearing cells from mixed populations that allows to identify signals specific to intracellular infection. Here we present an in-depth analysis of the effect of long-term S. aureus infection on the transcriptional program of human osteoblast-like cells. After RNA-seq and KEGG and Reactome pathway enrichment analysis, the remodeled transcriptomic profile of infected cells revealed exacerbated immune and inflammatory responses, as well as metabolic dysregulations that likely influence the intracellular life of bacteria. Numerous genes encoding epigenetic regulators were downregulated. The later included genes coding for components of chromatin-repressive complexes (e.g., NuRD, BAHD1 and PRC1) and epifactors involved in DNA methylation. Sets of genes encoding proteins of cell adhesion or neurotransmission were also deregulated. Our results suggest that intracellular S. aureus infection has a long-term impact on the genome and epigenome of host cells, which may exert patho-physiological dysfunctions additionally to the defense response during the infection process. Overall, these results not only improve our conceptual understanding of biological processes involved in the long-term S. aureus infections of osteoblast-like cells, but also provide an atlas of deregulated host genes and biological pathways and identify novel markers and potential candidates for prophylactic and therapeutic approaches.
Collapse
Affiliation(s)
- Aurélie Nicolas
- Institut National de Recherche pour l’agriculture, l’alimentation et l’environnement (INRAE), Institut Agro, Science et Technologie du Lait et de l’OEuf (STLO), Rennes, France
| | - Martine Deplanche
- Institut National de Recherche pour l’agriculture, l’alimentation et l’environnement (INRAE), Institut Agro, Science et Technologie du Lait et de l’OEuf (STLO), Rennes, France
| | - Pierre-Henri Commere
- Cytometry and Biomarkers Centre de Ressources et Recherches Technologiques (C2RT), Institut Pasteur, Paris, France
| | - Alan Diot
- Centre International de Recherche en Infectiologie, CIRI, Inserm U1111, Centre National de la Recherche Scientifique (CNRS) Unité Mixte de Recherche 5308 (UMR5308), Ecole Normale Supérieure (ENS) de Lyon, Universit´ Claude Bernard Lyon 1 (UCBL1), Lyon, France
- Hospices Civils de Lyon, French National Reference Centre for Staphylococci, Lyon, France
| | - Clemence Genthon
- Institut National de Recherche pour l’agriculture, l’alimentation et l’environnement (INRAE), Unité Service 1426 (US1426), Transcriptome Plateforme Technologique (GeT-PlaGe), Genotoul, Castanet-Tolosan, France
| | - Wanderson Marques da Silva
- Institut National de Recherche pour l’agriculture, l’alimentation et l’environnement (INRAE), Institut Agro, Science et Technologie du Lait et de l’OEuf (STLO), Rennes, France
- Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Brazil
| | - Vasco Azevedo
- Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Brazil
| | - Pierre Germon
- Institut National de Recherche pour l’agriculture, l’alimentation et l’environnement (INRAE), Université François Rabelais, Infectiologie et Santé Publique (ISP), Tours, France
| | - Hélène Jamme
- Université Paris-Saclay, Université de Versailles Saint-Quentin-en-Yvelines (UVSQ), Institut National de Recherche pour l’agriculture, l’alimentation et l’environnement (INRAE), Biologie de la Reproduction, Environnement, Epigénétique et Développement (BREED), Jouy-en-Josas, France
- Ecole Nationale Vétérinaire d’Alfort, Biologie de la Reproduction, Environnement, Epigénétique et Développement (BREED), Maisons-Alfort, France
| | - Eric Guédon
- Institut National de Recherche pour l’agriculture, l’alimentation et l’environnement (INRAE), Institut Agro, Science et Technologie du Lait et de l’OEuf (STLO), Rennes, France
| | - Yves Le Loir
- Institut National de Recherche pour l’agriculture, l’alimentation et l’environnement (INRAE), Institut Agro, Science et Technologie du Lait et de l’OEuf (STLO), Rennes, France
| | - Fréderic Laurent
- Centre International de Recherche en Infectiologie, CIRI, Inserm U1111, Centre National de la Recherche Scientifique (CNRS) Unité Mixte de Recherche 5308 (UMR5308), Ecole Normale Supérieure (ENS) de Lyon, Universit´ Claude Bernard Lyon 1 (UCBL1), Lyon, France
- Hospices Civils de Lyon, French National Reference Centre for Staphylococci, Lyon, France
| | - Hélène Bierne
- Université Paris-Saclay, Institut National de Recherche pour l’agriculture, l’alimentation et l’environnement (INRAE), AgroParisTech, Micalis Institute, Jouy-en-Josas, France
| | - Nadia Berkova
- Institut National de Recherche pour l’agriculture, l’alimentation et l’environnement (INRAE), Institut Agro, Science et Technologie du Lait et de l’OEuf (STLO), Rennes, France
- *Correspondence: Nadia Berkova,
| |
Collapse
|
48
|
[Research progress of antibacterial modification of orthopaedic implants surface]. ZHONGGUO XIU FU CHONG JIAN WAI KE ZA ZHI = ZHONGGUO XIUFU CHONGJIAN WAIKE ZAZHI = CHINESE JOURNAL OF REPARATIVE AND RECONSTRUCTIVE SURGERY 2022; 36:511-516. [PMID: 35426294 PMCID: PMC9011072 DOI: 10.7507/1002-1892.202112109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
OBJECTIVE To summarize the related research progress of antibacterial modification of orthopaedic implants surface in recent years. METHODS The domestic and foreign related literature in recent years was extensively consulted, the research progress on antibacterial modification of orthopaedic implants surface was discussed from two aspects of characteristics of infection in orthopedic implants and surface anti-infection modification. RESULTS The orthopaedic implants infections are mainly related to aspects of bacterial adhesion, decreased host immunity, and surface biofilm formation. At present, the main antimicrobial coating methods of orthopaedic implants are antibacterial adhesion coating, antibiotic coating, inorganic antimicrobial coating, composite antimicrobial coating, nitric oxide coating, immunomodulation, three-dimensional printing, polymer antimicrobial coating, and "smart" coating. CONCLUSION The above-mentioned antibacterial coating methods of orthopedic implants can not only inhibit bacterial adhesion, but also solve the problems of low immunity and biofilm formation. However, its mechanism of action and modification are still controversial and require further research.
Collapse
|
49
|
Jothipandiyan S, Suresh D, Sankaran SV, Thamotharan S, Shanmugasundaram K, Vincent P, Sekaran S, Gowrishankar S, Pandian SK, Paramasivam N. Heteroleptic pincer palladium(II) complex coated orthopedic implants impede the AbaI/AbaR quorum sensing system and biofilm development by Acinetobacter baumannii. BIOFOULING 2022; 38:55-70. [PMID: 34961388 DOI: 10.1080/08927014.2021.2015336] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Revised: 12/01/2021] [Accepted: 12/01/2021] [Indexed: 06/14/2023]
Abstract
Implant-associated infections mediated by Acinetobacter baumannii biofilms have become a major concern in the healthcare sector. As biofilm formation by this important pathogen is mediated by quorum sensing, quorum sensing inhibitors (QSI) have gained much attention. The present study confirms that novel thiazolinyl-picolinamide based palladium(II) complexes had good biofilm disruptive and QSI properties against A. baumannii. Key QS-mediated virulence factors like pili mediated surface motility and polysaccharide production were inhibited by the best Pd(II) complex (E). This also showed potent inhibitory activity against both the standard and clinical strains of A. baumannii. Molecular docking analysis also proved the potent binding affinity of Pd(II)-E with the virulence targets. The Pd(II) complex also disrupted preformed biofilms and down-regulated the expression of QS mediated virulence genes in the biofilms established on implant material (titanium plates). As a whole, the present study showed that the novel thiazolinyl-picolinamide based Pd(II) complexes offer a promising anti-infective strategy to combat biofilm-mediated implant infections.
Collapse
Affiliation(s)
- Sowndarya Jothipandiyan
- Biofilm Biology Laboratory, Centre for Research on Infectious Diseases (CRID), School of Chemical and Biotechnology, SASTRA Deemed University, Thanjavur, Tamil Nadu, India
| | - Devarajan Suresh
- Department of Chemistry, School of Chemical and Biotechnology, SASTRA Deemed University, Thanjavur, Tamil Nadu, India
| | - Sankaran Venkatachalam Sankaran
- Biomolecular Crystallography Laboratory, Department of Bioinformatics, School of Chemical and Biotechnology, SASTRA Deemed University, Thanjavur, Tamil Nadu, India
| | - Subbiah Thamotharan
- Biomolecular Crystallography Laboratory, Department of Bioinformatics, School of Chemical and Biotechnology, SASTRA Deemed University, Thanjavur, Tamil Nadu, India
| | | | - Preethi Vincent
- Bone Biology and Repair laboratory, School of Chemical and Biotechnology, SASTRA Deemed University, Thanjavur, Tamil Nadu, India
| | - Saravanan Sekaran
- Bone Biology and Repair laboratory, School of Chemical and Biotechnology, SASTRA Deemed University, Thanjavur, Tamil Nadu, India
- Department of Pharmacology, Saveetha Dental college and hospitals, Saveetha institute for medical and Technical sciences, Saveetha University, Chennai, India
| | | | | | - Nithyanand Paramasivam
- Biofilm Biology Laboratory, Centre for Research on Infectious Diseases (CRID), School of Chemical and Biotechnology, SASTRA Deemed University, Thanjavur, Tamil Nadu, India
| |
Collapse
|
50
|
Gunn NJ, Zelmer AR, Kidd SP, Solomon LB, Roscioli E, Yang D, Atkins GJ. A Human Osteocyte Cell Line Model for Studying Staphylococcus aureus Persistence in Osteomyelitis. Front Cell Infect Microbiol 2021; 11:781022. [PMID: 34805001 PMCID: PMC8597899 DOI: 10.3389/fcimb.2021.781022] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Accepted: 10/19/2021] [Indexed: 11/25/2022] Open
Abstract
Infectious osteomyelitis associated with periprosthetic joint infections is often recalcitrant to treatment and has a high rate of recurrence. In the case of Staphylococcus aureus, the most common pathogen in all forms of osteomyelitis, this may be attributed in part to residual intracellular infection of host cells, yet this is not generally considered in the treatment strategy. Osteocytes represent a unique cell type in this context due to their abundance, their formation of a syncytium throughout the bone that could facilitate bacterial spread and their relative inaccessibility to professional immune cells. As such, there is potential value in studying the host-pathogen interactions in the context of this cell type in a replicable and scalable in vitro model. Here, we examined the utility of the human osteosarcoma cell line SaOS2 differentiated to an osteocyte-like stage (SaOS2-OY) as an intracellular infection model for S. aureus. We demonstrate that S. aureus is capable of generating stable intracellular infections in SaOS2-OY cells but not in undifferentiated, osteoblast-like SaOS2 cells (SaOS2-OB). In SaOS2-OY cells, S. aureus transitioned towards a quasi-dormant small colony variant (SCV) growth phenotype over a 15-day post-infection period. The infected cells exhibited changes in the expression of key immunomodulatory mediators that are consistent with the infection response of primary osteocytes. Thus, SaOS2-OY is an appropriate cell line model that may be predictive of the interactions between S. aureus and human osteocytes, and this will be useful for studying mechanisms of persistence and for testing the efficacy of potential antimicrobial strategies.
Collapse
Affiliation(s)
- Nicholas J Gunn
- Centre for Orthopaedic & Trauma Research, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, SA, Australia
| | - Anja R Zelmer
- Centre for Orthopaedic & Trauma Research, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, SA, Australia
| | - Stephen P Kidd
- Australian Centre for Antimicrobial Resistance Ecology, University of Adelaide, Adelaide, SA, Australia.,Research Centre for Infectious Disease, School of Biological Science, University of Adelaide, Adelaide, SA, Australia
| | - Lucian B Solomon
- Centre for Orthopaedic & Trauma Research, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, SA, Australia.,Department of Orthopaedics and Trauma, Royal Adelaide Hospital, Adelaide, SA, Australia
| | - Eugene Roscioli
- Department of Thoracic Medicine, Royal Adelaide Hospital, Adelaide, SA, Australia.,Department of Medicine, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, SA, Australia
| | - Dongqing Yang
- Centre for Orthopaedic & Trauma Research, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, SA, Australia
| | - Gerald J Atkins
- Centre for Orthopaedic & Trauma Research, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, SA, Australia
| |
Collapse
|