1
|
Chen Z, Jin Q, Zhong J, Xie Z, Chen Q, Li L, Li J, Zhao C, Wang J, Tang X, Han M, Li R, Li Z, Tong Z, Wang M, Du H, Zhang H. Staphylococcus aureus blocks host autophagy through circSyk/miR-5106/Sik3 axis to promote progression of bone infection. PLoS Pathog 2025; 21:e1012896. [PMID: 39869636 PMCID: PMC11781720 DOI: 10.1371/journal.ppat.1012896] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Revised: 01/30/2025] [Accepted: 01/09/2025] [Indexed: 01/29/2025] Open
Abstract
With the rapid increase in the number of implant operations, the incidence of bone infections has increased. Methicillin-resistant Staphylococcus aureus (S. aureus) and other emerging fully drug-resistant strains make the management of bone infections even more challenging. Bone infections are mainly caused by S. aureus and require extensive surgical intervention and long-term antibiotic therapy. The host autophagy response is critical to the elimination of S. aureus infections. In this study, we demonstrate that a circular RNA (circRNA), circSyk, is a potential biological target for the treatment of S. aureus-induced bone infection. Most importantly, S. aureus regulates circSyk to block autophagy and promote bone destruction via the circSyk/miR-5106/Sik3 axis in a nonclassical pathway, which is involved in the S. aureus infection process through a competitive endogenous RNA network. In summary, this study proposes a novel perspective on the immune escape of S. aureus in bone infections, based on circRNA.
Collapse
Affiliation(s)
- Zhihao Chen
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Qiyuan Jin
- Department of Clinical Laboratory, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Jinqi Zhong
- Department of Clinical Laboratory, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Zonggang Xie
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Qi Chen
- Department of Clinical Laboratory, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Liubing Li
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Jijie Li
- Department of Clinical Laboratory, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Chenhao Zhao
- Department of Clinical Laboratory, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Junfeng Wang
- Department of Clinical Laboratory, The Nuclear Industry 417 Hospital, Xi’an, China
| | - Xiaoying Tang
- Department of Clinical Laboratory, The Second Affiliated Hospital of Soochow University, Suzhou, China
- Department of Clinical Laboratory, Kunshan Municipal Third People’s Hospital, Suzhou, China
| | - Mingxiao Han
- Department of Clinical Laboratory, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Ru Li
- Department of Clinical Laboratory, The Nuclear Industry 417 Hospital, Xi’an, China
| | - Ziyuan Li
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Zelei Tong
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Min Wang
- Department of Clinical Laboratory, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Hong Du
- Department of Clinical Laboratory, The Second Affiliated Hospital of Soochow University, Suzhou, China
- MOE Key Laboratory of Geriatric Diseases and Immunology, Soochow University, Suzhou, China
| | - Haifang Zhang
- Department of Clinical Laboratory, The Second Affiliated Hospital of Soochow University, Suzhou, China
- MOE Key Laboratory of Geriatric Diseases and Immunology, Soochow University, Suzhou, China
| |
Collapse
|
2
|
Dong Q, Zhou J, Feng M, Kong L, Fang B, Zhang Z. A review of bacterial and osteoclast differentiation in bone infection. Microb Pathog 2024; 197:107102. [PMID: 39505086 DOI: 10.1016/j.micpath.2024.107102] [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: 03/29/2024] [Revised: 10/18/2024] [Accepted: 11/03/2024] [Indexed: 11/08/2024]
Abstract
Bone infections are characterized by bacterial invasion of the bone microenvironment and subsequent bone structure deterioration. This holds significance because osteoclasts, which are the only cells responsible for bone resorption, are abnormally stimulated during bone infections. Multiple communication factors secreted by bone stromal cells regulate the membrane of osteoclast progenitor cells, thereby maintaining bone homeostasis through the expression of many types of receptors. During infection, the immunoinflammatory response triggered by bacterial invasion and multiple virulence factors of bacterial origin can disrupt osteoclast homeostasis. Therefore, clarifying the pathways through which bacteria affect osteoclasts can offer a theoretical basis for preventing and treating bone infections. This review summarizes studies investigating bone destruction caused by different bacterial infections. In conclusion, bacteria can affect osteoclast metabolic activity through multiple pathways, including direct contact, release of virulence factors, induction of immunoinflammatory responses, influence on bone stromal cell metabolism, and intracellular infections.
Collapse
Affiliation(s)
- Qi Dong
- Department of Spinal Surgery, Honghui Hospital, Xi'an Jiaotong University, Xi'an, 710054, China
| | - Jiuqin Zhou
- Department of Infectious Disease of Honghui Hospital, Xi'an Jiaotong University, Xi'an, 710054, China
| | - Mingzhe Feng
- Department of Spinal Surgery, Honghui Hospital, Xi'an Jiaotong University, Xi'an, 710054, China
| | - Lingqiang Kong
- Department of Orthopedics, the Central Hospital Affiliated to Shaoxing University, Shaoxing, 312030, China.
| | - Bin Fang
- Department of Orthopedics, the First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, 310000, China.
| | - Zhen Zhang
- Department of Spinal Surgery, Honghui Hospital, Xi'an Jiaotong University, Xi'an, 710054, China.
| |
Collapse
|
3
|
Villegas M, Bayat F, Kramer T, Schwarz E, Wilson D, Hosseinidoust Z, Didar TF. Emerging Strategies to Prevent Bacterial Infections on Titanium-Based Implants. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2404351. [PMID: 39161205 DOI: 10.1002/smll.202404351] [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: 05/29/2024] [Revised: 07/15/2024] [Indexed: 08/21/2024]
Abstract
Titanium and titanium alloys remain the gold standard for dental and orthopedic implants. These materials are heavily used because of their bioinert nature, robust mechanical properties, and seamless integration with bone. However, implant-associated infections (IAIs) remain one of the leading causes of implant failure. Eradicating an IAI can be difficult since bacteria can form biofilms on the medical implant, protecting the bacterial cells against systemic antibiotics and the host's immune system. If the infection is not treated promptly and aggressively, device failure is inevitable, leading to costly multi-step revision surgeries. To circumvent this dire situation, scientists and engineers continue to develop novel strategies to protect the surface of medical implants from bacteria. In this review, details on emerging strategies to prevent infection in titanium implants are reported. These strategies include anti-adhesion properties provided by polymers, superhydrophobic, superhydrophilic, and liquid-infused surface coatings, as well as strategies and coatings employed to lyse the bacteria. Additionally, commercially available technologies and those under preclinical trials are examined while discussing current and future trends.
Collapse
Affiliation(s)
- Martin Villegas
- School of Biomedical Engineering, McMaster University, 1280 Main Street West, Hamilton, ON, L8S 4L8, Canada
| | - Fereshteh Bayat
- School of Biomedical Engineering, McMaster University, 1280 Main Street West, Hamilton, ON, L8S 4L8, Canada
| | - Taylor Kramer
- Department of Mechanical Engineering, McMaster University, 1280 Main Street West, Hamilton, ON, L8S 4L8, Canada
| | - Elise Schwarz
- Department of Mechanical Engineering, McMaster University, 1280 Main Street West, Hamilton, ON, L8S 4L8, Canada
| | - David Wilson
- Division of Orthopedic Surgery, Halifax Infirmary, Halifax, NS, B3H3A6, Canada
| | - Zeinab Hosseinidoust
- School of Biomedical Engineering, McMaster University, 1280 Main Street West, Hamilton, ON, L8S 4L8, Canada
- Department of Chemical Engineering, McMaster University, 1280 Main Street West, Hamilton, ON, L8S 4L8, Canada
| | - Tohid F Didar
- School of Biomedical Engineering, McMaster University, 1280 Main Street West, Hamilton, ON, L8S 4L8, Canada
- Department of Mechanical Engineering, McMaster University, 1280 Main Street West, Hamilton, ON, L8S 4L8, Canada
| |
Collapse
|
4
|
Chang J, Kerr D, Zheng M, Seyler T. Chondrocyte Invasion May Be a Mechanism for Persistent Staphylococcus Aureus Infection In Vitro. Clin Orthop Relat Res 2024; 482:1839-1847. [PMID: 38662927 PMCID: PMC11419450 DOI: 10.1097/corr.0000000000003074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Accepted: 03/11/2024] [Indexed: 06/23/2024]
Abstract
BACKGROUND Recurrent bone and joint infection with Staphylococcus aureus is common. S. aureus can invade and persist in osteoblasts and fibroblasts, but little is known about this mechanism in chondrocytes. If S. aureus were able to invade and persist within chondrocytes, this could be a difficult compartment to treat. QUESTION/PURPOSE Can S. aureus infiltrate and persist intracellularly within chondrocytes in vitro? METHODS Cell lines were cultured in vitro and infected with S. aureus. Human chondrocytes (C20A4) were compared with positive controls of human osteoblasts (MG63) and mouse fibroblasts (NIH3T3), which have previously demonstrated S. aureus invasion and persistence (human fibroblasts were not available to us). Six replicates per cell type were followed for 6 days after infection. Cells were treated daily with antibiotic media for extracellular killing. To determine whether S. aureus can infiltrate chondrocytes, fluorescence microscopy was performed to qualitatively assess the presence of intracellular bacteria, and intracellular colony-forming units (CFU) were enumerated 2 hours after infection. To determine whether S. aureus can persist within chondrocytes, intracellular CFUs were enumerated from infected host cells each day postinfection. RESULTS S. aureus invaded human chondrocytes (C20A4) at a level (2.8 x 10 5 ± 5.5 x 10 4 CFUs/mL) greater than positive controls of human osteoblasts (MG63) (9.5 x 10 2 ± 2.5 x 10 2 CFUs/mL; p = 0.01) and mouse fibroblasts (NIH3T3) (9.1 x 10 4 ± 2.5 x 10 4 CFUs/mL; p = 0.02). S. aureus also persisted within human chondrocytes (C20A4) for 6 days at a level (1.4 x 10 3 ± 5.3 x 10 2 CFUs/mL) greater than that of human osteoblasts (MG63) (4.3 x 10 2 ± 3.5 x 10 1 CFUs/mL; p = 0.02) and mouse fibroblasts (NIH3T3) (0 CFUs/mL; p < 0.01). S. aureus was undetectable within mouse fibroblasts (NIH3T3) after 4 days. There were 0 CFUs yielded from cell media, confirming extracellular antibiotic treatment was effective. CONCLUSION S. aureus readily invaded human chondrocytes (C20A4) in vitro and persisted viably for 6 days after infection, evading extracellular antibiotics. Chondrocytes demonstrated a greater level of intracellular invasion and persistence by S. aureus than positive control human osteoblast (MG63) and mouse fibroblast (NIH3T3) cell lines. CLINICAL RELEVANCE Chondrocyte invasion and persistence may contribute to recurrent bone and joint infections. Additional research should assess longer periods of persistence and whether this mechanism is present in vivo.
Collapse
Affiliation(s)
- Jerry Chang
- Department of Orthopaedic Surgery, Duke University, Durham, NC, USA
| | - David Kerr
- Department of Orthopaedic Surgery, Duke University, Durham, NC, USA
| | - Megan Zheng
- Department of Orthopaedic Surgery, Duke University, Durham, NC, USA
| | - Thorsten Seyler
- Department of Orthopaedic Surgery, Duke University, Durham, NC, USA
| |
Collapse
|
5
|
Wang R, Wu N, Zhan D, Chen F. Naringin exerts antibacterial and anti-inflammatory effects on mice with Staphylococcus aureus-induced osteomyelitis. J Biochem Mol Toxicol 2024; 38:e23753. [PMID: 38923626 DOI: 10.1002/jbt.23753] [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: 12/22/2023] [Revised: 05/10/2024] [Accepted: 05/31/2024] [Indexed: 06/28/2024]
Abstract
Osteomyelitis is an invasive bone infection that can lead to severe pain and even disability, posing a challenge for orthopedic surgery. Naringin can reduce bone-related inflammatory conditions. This study aimed to elucidate the function and mechanism of naringin in a Staphylococcus aureus-induced mouse model of osteomyelitis. Femurs of S. aureus-infected mice were collected after naringin administration and subjected to microcomputed tomography to analyze cortical bone destruction and bone loss. Bacterial growth in femurs was also assessed. Proinflammatory cytokine levels in mouse femurs were measured using enzyme-linked immunosorbent assays. Pathological changes and bone resorption were analyzed using hematoxylin and eosin staining and tartrate-resistant acid phosphatase staining, respectively. Quantitative reverse transcription polymerase chain reaction and western blot analysis were used to quantify the messenger RNA and protein expression of osteogenic differentiation-associated genes in the femurs. The viability of human bone marrow-derived stem cells (hBMSCs) was determined using cell counting kit-8. Alizarin Red S staining and alkaline phosphatase staining were performed to assess the formation of mineralization nodules and bone formation in vitro. Notch signaling-related protein levels in femur tissues and hBMSCs were assessed using western blot analysis. Experimental results revealed that naringin alleviated S. aureus-induced cortical bone destruction and bone loss in mice by increasing the bone volume/total volume ratio. Naringin suppressed S. aureus-induced bacterial growth and inflammation in femurs. Moreover, it alleviated histopathological changes, inhibited bone resorption, and increased the expression of osteogenic markers in osteomyelitic mice. It increased the viability of hBMSCs and promoted their differentiation and bone mineralization in vitro. Furthermore, naringin activated Notch signaling by upregulating the protein levels of Notch1, Jagged1, and Hes1 in the femurs of model mice and S. aureus-stimulated hBMSCs. In conclusion, naringin reduces bacterial growth, inflammation, and bone resorption while upregulating the expression of osteogenic markers in S. aureus-infected mice and hBMSCs by activating Notch signaling.
Collapse
Affiliation(s)
- Rong Wang
- Department of Clinical Laboratory Medicine, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, China
| | - NongXin Wu
- Department of Orthopedics, Xiangyang Central HospitaI, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, China
| | - Dong'ang Zhan
- Department of Hospital Infection Management Office, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, China
| | - Fengwen Chen
- Department of Orthopedics, Xiangyang Central HospitaI, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, China
| |
Collapse
|
6
|
Zhou X, Ma S, Xu Y, Sun C, Liao J, Song M, Li G, Yuchen L, Chen P, Hu Y, Wang Y, Yu B. Nicotine promotes Staphylococcus aureus-induced osteomyelitis by activating the Nrf2/Slc7a11 signaling axis. Int Immunopharmacol 2024; 135:112223. [PMID: 38772295 DOI: 10.1016/j.intimp.2024.112223] [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: 12/13/2023] [Revised: 05/03/2024] [Accepted: 05/05/2024] [Indexed: 05/23/2024]
Abstract
Although smoking is a significant risk factor for osteomyelitis, there is limited experimental evidence that nicotine, a key tobacco constituent, is associated with this condition, leaving its mechanistic implications uncharacterized. This study revealed that nicotine promotes Staphylococcus aureus-induced osteomyelitis by increasing Nrf2 and Slc7a11 expression in vivo and in vitro. Inhibition of Slc7a11 using Erastin augmented bacterial phagocytosis/killing capabilities and fortified antimicrobial responses in an osteomyelitis model. Moreover, untargeted metabolomic analysis demonstrated that Erastin mitigated the effects of nicotine on S. aureus-induced osteomyelitis by altering glutamate/glutathione metabolism. These findings suggest that nicotine aggravates S. aureus-induced osteomyelitis by activating the Nrf2/Slc7a11 signaling pathway and that Slc7a11 inhibition can counteract the detrimental health effects of nicotine.
Collapse
Affiliation(s)
- Xuyou Zhou
- Guangdong Provincial Key Laboratory of Bone and Cartilage Regenerative Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China; Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Sushuang Ma
- Guangdong Provincial Key Laboratory of Bone and Cartilage Regenerative Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China; Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, China; Department of Orthopaedics, The Fifth Affiliated Hospital, Southerm Medical University, Guangzhou, China
| | - Yuan Xu
- Guangdong Provincial Key Laboratory of Bone and Cartilage Regenerative Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China; Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Chongkai Sun
- Guangdong Provincial Key Laboratory of Bone and Cartilage Regenerative Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China; Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Juncheng Liao
- Guangdong Provincial Key Laboratory of Bone and Cartilage Regenerative Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China; Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Mingrui Song
- Guangdong Provincial Key Laboratory of Bone and Cartilage Regenerative Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China; Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Guanzhi Li
- Guangdong Provincial Key Laboratory of Bone and Cartilage Regenerative Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China; Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Liu Yuchen
- Guangdong Provincial Key Laboratory of Bone and Cartilage Regenerative Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China; Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Peng Chen
- Guangdong Provincial Key Laboratory of Bone and Cartilage Regenerative Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China; Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, China; Department of Orthopedics, Hainan General Hospital (Hainan Affiliated Hospital of Hainan Medical University), Haikou, China
| | - Yanjun Hu
- Guangdong Provincial Key Laboratory of Bone and Cartilage Regenerative Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China; Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yutian Wang
- Guangdong Provincial Key Laboratory of Bone and Cartilage Regenerative Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China; Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, China.
| | - Bin Yu
- Guangdong Provincial Key Laboratory of Bone and Cartilage Regenerative Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China; Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, China.
| |
Collapse
|
7
|
Piuzzi NS, Klika AK, Lu Q, Higuera-Rueda CA, Stappenbeck T, Visperas A. Periprosthetic joint infection and immunity: Current understanding of host-microbe interplay. J Orthop Res 2024; 42:7-20. [PMID: 37874328 DOI: 10.1002/jor.25723] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 09/19/2023] [Accepted: 10/17/2023] [Indexed: 10/25/2023]
Abstract
Periprosthetic joint infection (PJI) is a major complication of total joint arthroplasty. Even with current treatments, failure rates are unacceptably high with a 5-year mortality rate of 26%. Majority of the literature in the field has focused on development of better biomarkers for diagnostics and treatment strategies including innovate antibiotic delivery systems, antibiofilm agents, and bacteriophages. Nevertheless, the role of the immune system, our first line of defense during PJI, is not well understood. Evidence of infection in PJI patients is found within circulation, synovial fluid, and tissue and include numerous cytokines, metabolites, antimicrobial peptides, and soluble receptors that are part of the PJI diagnosis workup. Macrophages, neutrophils, and myeloid-derived suppressor cells (MDSCs) are initially recruited into the joint by chemokines and cytokines produced by immune cells and bacteria and are activated by pathogen-associated molecular patterns. While these cells are efficient killers of planktonic bacteria by phagocytosis, opsonization, degranulation, and recruitment of adaptive immune cells, biofilm-associated bacteria are troublesome. Biofilm is not only a physical barrier for the immune system but also elicits effector functions. Additionally, bacteria have developed mechanisms to evade the immune system by inactivating effector molecules, promoting killing or anti-inflammatory effector cell phenotypes, and intracellular persistence and dissemination. Understanding these shortcomings and the mechanisms by which bacteria can subvert the immune system may open new approaches to better prepare our own immune system to combat PJI. Furthermore, preoperative immune system assessment and screening for dysregulation may aid in developing preventative interventions to decrease PJI incidence.
Collapse
Affiliation(s)
- Nicolas S Piuzzi
- Department of Orthopaedic Surgery, Cleveland Clinic Adult Reconstruction Research (CCARR), Cleveland Clinic, Cleveland, Ohio, USA
- Department of Biomedical Engineering, Cleveland Clinic, Cleveland, Ohio, USA
| | - Alison K Klika
- Department of Orthopaedic Surgery, Cleveland Clinic Adult Reconstruction Research (CCARR), Cleveland Clinic, Cleveland, Ohio, USA
| | - Qiuhe Lu
- Department of Inflammation and Immunity, Cleveland Clinic, Cleveland, Ohio, USA
| | | | | | - Anabelle Visperas
- Department of Orthopaedic Surgery, Cleveland Clinic Adult Reconstruction Research (CCARR), Cleveland Clinic, Cleveland, Ohio, USA
| |
Collapse
|
8
|
Li M, Shi X, Wu Y, Qi B, Zhang C, Wang B, Zhang B, Xu Y. Pmepa1 knockdown alleviates SpA-induced pyroptosis and osteogenic differentiation inhibition of hBMSCs via p38MAPK/NLRP3 axis. Int Immunopharmacol 2023; 124:110843. [PMID: 37634444 DOI: 10.1016/j.intimp.2023.110843] [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: 04/07/2023] [Revised: 08/12/2023] [Accepted: 08/20/2023] [Indexed: 08/29/2023]
Abstract
BACKGROUND Osteomyelitis is a refractory bone infectious disease, which usually results in progressive bone destruction and bone loss. The invasion of pathogens and subsequent inflammatory response could damage bone marrow mesenchymal stem cells (BMSCs) and inhibit osteogenic differentiation, and finally aggravate uncontrolled bone remodeling in osteomyelitis by affecting bone formation. Exploring the mechanisms of BMSCs injury and osteogenic differentiation inhibition may would help us to find potential therapeutic targets. METHOD Firstly, staphylococcal protein A (SpA)-treated human bone marrow mesenchymal stem cells (hBMSCs) were used to construct cell models of osteomyelitis. Secondly, transcriptome sequencing was performed to screen differentially expressed genes and then verified the expression of target genes. Next, in vitro experiments were conducted to explore the functions and mechanisms of prostate transmembrane protein androgen induced 1 (Pmepa1) in SpA-treated hBMSCs. Finally, the rat model of osteomyelitis was established to provide an auxiliary validation of the in vitro experimental results. RESULTS We found that SpA treatment induced inflammatory injury and inhibited osteogenic differentiation in hBMSCs, then the transcriptome sequencing and further detection results showed that Pmepa1 was significantly upregulated in this process. Functionally, Pmepa1 knockdown alleviated inflammatory injury and promoted osteogenic differentiation in SpA-treated hBMSCs. Among them, it was demonstrated that Pmepa1 knockdown exerted cytoprotective effects by alleviating pyroptosis of SpA-infected hBMSCs. Furthermore, recovery experiments revealed that Pmepa1 knockdown reversed SpA-mediated adverse effects by downregulating the p38MAPK/NLRP3 axis. Finally, the detection results of rat femoral osteomyelitis showed that the expression of Pmepa1 was up-regulated, and the expression trends of other indicators including p38MAPK, NLRP3, and caspase-1 were also consistent with the in vitro model. CONCLUSION Pmepa1 knockdown alleviates SpA-induced pyroptosis and inhibition of osteogenic differentiation in hBMSCs by downregulating p38MAPK/NLRP3 signaling axis. Modulating the expression of Pmepa1 may be a potential strategy to ameliorate osteomyelitis.
Collapse
Affiliation(s)
- Mingjun Li
- Kunming Medical University, Kunming, Yunnan 650500, China
| | - Xiangwen Shi
- Kunming Medical University, Kunming, Yunnan 650500, China
| | - Yipeng Wu
- Department of Orthopedic Surgery, 920th Hospital of Joint Logistics Support Force, Kunming, China
| | - Baochuang Qi
- Kunming Medical University, Kunming, Yunnan 650500, China
| | - Chaoqun Zhang
- Kunming Medical University, Kunming, Yunnan 650500, China
| | - Bin Wang
- Kunming Medical University, Kunming, Yunnan 650500, China
| | - Bihuan Zhang
- Kunming Medical University, Kunming, Yunnan 650500, China
| | - Yongqing Xu
- Department of Orthopedic Surgery, 920th Hospital of Joint Logistics Support Force, Kunming, China.
| |
Collapse
|
9
|
Hinz N, Butscheidt S, Jandl NM, Rohde H, Keller J, Beil FT, Hubert J, Rolvien T. Increased local bone turnover in patients with chronic periprosthetic joint infection. Bone Joint Res 2023; 12:644-653. [PMID: 37813394 PMCID: PMC10562080 DOI: 10.1302/2046-3758.1210.bjr-2023-0071.r1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/11/2023] Open
Abstract
Aims The management of periprosthetic joint infection (PJI) remains a major challenge in orthopaedic surgery. In this study, we aimed to characterize the local bone microstructure and metabolism in a clinical cohort of patients with chronic PJI. Methods Periprosthetic femoral trabecular bone specimens were obtained from patients suffering from chronic PJI of the hip and knee (n = 20). Microbiological analysis was performed on preoperative joint aspirates and tissue specimens obtained during revision surgery. Microstructural and cellular bone parameters were analyzed in bone specimens by histomorphometry on undecalcified sections complemented by tartrate-resistant acid phosphatase immunohistochemistry. Data were compared with control specimens obtained during primary arthroplasty (n = 20) and aseptic revision (n = 20). Results PJI specimens exhibited a higher bone volume, thickened trabeculae, and increased osteoid parameters compared to both control groups, suggesting an accelerated bone turnover with sclerotic microstructure. On the cellular level, osteoblast and osteoclast parameters were markedly increased in the PJI cohort. Furthermore, a positive association between serum (CRP) but not synovial (white blood cell (WBC) count) inflammatory markers and osteoclast indices could be detected. Comparison between different pathogens revealed increased osteoclastic bone resorption parameters without a concomitant increase in osteoblasts in bone specimens from patients with Staphylococcus aureus infection, compared to those with detection of Staphylococcus epidermidis and Cutibacterium spp. Conclusion This study provides insights into the local bone metabolism in chronic PJI, demonstrating osteosclerosis with high bone turnover. The fact that Staphylococcus aureus was associated with distinctly increased osteoclast indices strongly suggests early surgical treatment to prevent periprosthetic bone alterations.
Collapse
Affiliation(s)
- Nico Hinz
- Department of Trauma and Orthopaedic Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Department of Trauma, Surgery and Sports Traumatology, BG Trauma Hospital Hamburg, Hamburg, Germany
| | - Sebastian Butscheidt
- Department of Trauma and Orthopaedic Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Nico M. Jandl
- Department of Trauma and Orthopaedic Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Holger Rohde
- Instiute of Medical Microbiology, Virology and Hygiene, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Johannes Keller
- Department of Trauma and Orthopaedic Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Frank T. Beil
- Department of Trauma and Orthopaedic Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Jan Hubert
- Department of Trauma and Orthopaedic Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Tim Rolvien
- Department of Trauma and Orthopaedic Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
10
|
Chen Y, Liu Z, Lin Z, Lu M, Fu Y, Liu G, Yu B. The effect of Staphylococcus aureus on innate and adaptive immunity and potential immunotherapy for S. aureus-induced osteomyelitis. Front Immunol 2023; 14:1219895. [PMID: 37744377 PMCID: PMC10517662 DOI: 10.3389/fimmu.2023.1219895] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Accepted: 08/18/2023] [Indexed: 09/26/2023] Open
Abstract
Osteomyelitis is a chronic inflammatory bone disease caused by infection of open fractures or post-operative implants. Particularly in patients with open fractures, the risk of osteomyelitis is greatly increased as the soft tissue damage and bacterial infection are often more severe. Staphylococcus aureus, one of the most common pathogens of osteomyelitis, disrupts the immune response through multiple mechanisms, such as biofilm formation, virulence factor secretion, and metabolic pattern alteration, which attenuates the effectiveness of antibiotics and surgical debridement toward osteomyelitis. In osteomyelitis, immune cells such as neutrophils, macrophages and T cells are activated in response to pathogenic bacteria invasion with excessive inflammatory factor secretion, immune checkpoint overexpression, and downregulation of immune pathway transcription factors, which enhances osteoclastogenesis and results in bone destruction. Therefore, the study of the mechanisms of abnormal immunity will be a new breakthrough in the treatment of osteomyelitis.
Collapse
Affiliation(s)
- Yingqi Chen
- Division of Orthopaedics and Traumatology, Department of Orthopaedics, Southern Medical University Nanfang Hospital, Guangzhou, China
- Guangdong Provincial Key Laboratory of Bone and Cartilage Regenerative Medicine, Southern Medical University Nanfang Hospital, Guangzhou, China
| | - Zixian Liu
- Division of Orthopaedics and Traumatology, Department of Orthopaedics, Southern Medical University Nanfang Hospital, Guangzhou, China
- Guangdong Provincial Key Laboratory of Bone and Cartilage Regenerative Medicine, Southern Medical University Nanfang Hospital, Guangzhou, China
| | - Zexin Lin
- Division of Orthopaedics and Traumatology, Department of Orthopaedics, Southern Medical University Nanfang Hospital, Guangzhou, China
- Guangdong Provincial Key Laboratory of Bone and Cartilage Regenerative Medicine, Southern Medical University Nanfang Hospital, Guangzhou, China
| | - Mincheng Lu
- Division of Orthopaedics and Traumatology, Department of Orthopaedics, Southern Medical University Nanfang Hospital, Guangzhou, China
- Guangdong Provincial Key Laboratory of Bone and Cartilage Regenerative Medicine, Southern Medical University Nanfang Hospital, Guangzhou, China
| | - Yong Fu
- Division of Orthopaedics and Traumatology, Department of Orthopaedics, Southern Medical University Nanfang Hospital, Guangzhou, China
- Guangdong Provincial Key Laboratory of Bone and Cartilage Regenerative Medicine, Southern Medical University Nanfang Hospital, Guangzhou, China
- Trauma Center, Department of Orthopaedic Trauma, The Second Affiliated Hospital of Hengyang Medical College, South China University, Hengyang, China
| | - Guanqiao Liu
- Division of Orthopaedics and Traumatology, Department of Orthopaedics, Southern Medical University Nanfang Hospital, Guangzhou, China
- Guangdong Provincial Key Laboratory of Bone and Cartilage Regenerative Medicine, Southern Medical University Nanfang Hospital, Guangzhou, China
| | - Bin Yu
- Division of Orthopaedics and Traumatology, Department of Orthopaedics, Southern Medical University Nanfang Hospital, Guangzhou, China
- Guangdong Provincial Key Laboratory of Bone and Cartilage Regenerative Medicine, Southern Medical University Nanfang Hospital, Guangzhou, China
| |
Collapse
|
11
|
Chen Z, Xie Z, Han M, Jin Q, Li Z, Zhai Y, Zhang M, Hu G, Zhang H. Global Transcriptomic Study of Circular-RNA Expression Profile in Osteoclasts Infected by Intracellular Staphylococcus aureus. Infect Immun 2023; 91:e0035722. [PMID: 37212691 PMCID: PMC10269070 DOI: 10.1128/iai.00357-22] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2023] [Accepted: 05/01/2023] [Indexed: 05/23/2023] Open
Abstract
Osteomyelitis is difficult to cure, and the rapidly rising morbidity is a thorny problem accompanied by a large number of joint replacement applications. Staphylococcus aureus is the main pathogen of osteomyelitis. Circular RNAs (circRNAs), as emerging noncoding RNAs, play important roles in multiple physiopathological processes which could provide novel insights into osteomyelitis. However, little is known about the roles of circRNAs in the pathogenesis of osteomyelitis. Osteoclasts, considered bone sentinels, are the resident macrophages in bone and may play the immune defense roles in osteomyelitis. It has been reported that S. aureus can survive in osteoclasts, but the function of osteoclast circRNAs in response to intracellular S. aureus infection remains unclear. In this study, we investigated the profile of circRNAs in osteoclasts infected by intracellular S. aureus through high-throughput RNA sequencing. In total, 24 upregulated and 62 downregulated differentially expressed circRNAs were identified and subsequently analyzed to demonstrate their potential functions. On this basis, three circRNAs (chr4:130718154-130728164+, chr8:77409548-77413627-, and chr1:190871592-190899571-) were confirmed as potential novel biomarkers for the diagnosis of osteomyelitis through the murine model of osteomyelitis. Most importantly, we verified that the circRNA chr4:130718154-130728164+ named circPum1 could regulate the host autophagy to affect the intracellular infection of S. aureus through miR-767. In addition, circPum1 could serve as a promising serum biomarker in osteomyelitis patients caused by S. aureus infection. Taken together, this study provided the first global transcriptomic profile analysis of circRNAs in osteoclasts infected by intracellular S. aureus and first proposed a novel perspective for the pathogenesis and immunotherapy of S. aureus-induced osteomyelitis from the term of circRNAs.
Collapse
Affiliation(s)
- Zhihao Chen
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, Suzhou, People’s Republic of China
| | - Zonggang Xie
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, Suzhou, People’s Republic of China
| | - Mingxiao Han
- Department of Clinical Laboratory, The Second Affiliated Hospital of Soochow University, Suzhou, People’s Republic of China
| | - Qiyuan Jin
- Department of Clinical Laboratory, The Second Affiliated Hospital of Soochow University, Suzhou, People’s Republic of China
| | - Ziyuan Li
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, Suzhou, People’s Republic of China
| | - Yaxuan Zhai
- Department of Clinical Laboratory, The Second Affiliated Hospital of Soochow University, Suzhou, People’s Republic of China
| | - Minxing Zhang
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, Suzhou, People’s Republic of China
| | - Gangfeng Hu
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, Suzhou, People’s Republic of China
| | - Haifang Zhang
- Department of Clinical Laboratory, The Second Affiliated Hospital of Soochow University, Suzhou, People’s Republic of China
| |
Collapse
|
12
|
Wang C, Xu P, Li X, Zheng Y, Song Z. Research progress of stimulus-responsive antibacterial materials for bone infection. Front Bioeng Biotechnol 2022; 10:1069932. [PMID: 36636700 PMCID: PMC9831006 DOI: 10.3389/fbioe.2022.1069932] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Accepted: 12/09/2022] [Indexed: 12/24/2022] Open
Abstract
Infection is one of the most serious complications harmful to human health, which brings a huge burden to human health. Bone infection is one of the most common and serious complications of fracture and orthopaedic surgery. Antibacterial treatment is the premise of bone defect healing. Among all the antibacterial strategies, irritant antibacterial materials have unique advantages and the ability of targeted therapy. In this review, we focus on the research progress of irritating materials, the development of antibacterial materials and their advantages and disadvantages potential applications in bone infection.
Collapse
Affiliation(s)
| | | | | | - Yuhao Zheng
- Department of Sports Medicine, Orthopaedic Center, The First Hospital of Jilin University, Changchun, China
| | - Zhiming Song
- Department of Sports Medicine, Orthopaedic Center, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
13
|
Cui Y, Liu H, Tian Y, Fan Y, Li S, Wang G, Wang Y, Peng C, Wu D. Dual-functional composite scaffolds for inhibiting infection and promoting bone regeneration. Mater Today Bio 2022; 16:100409. [PMID: 36090611 PMCID: PMC9449864 DOI: 10.1016/j.mtbio.2022.100409] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 08/18/2022] [Accepted: 08/20/2022] [Indexed: 12/14/2022] Open
Abstract
The treatment of infected bone defects is an intractable problem in orthopedics. It comprises two critical parts, namely that of infection control and bone defect repair. According to these two core tasks during treatment, the ideal approach of simultaneously controlling infection and repairing bone defects is promising treatment strategy. Several engineered biomaterials and drug delivery systems with dual functions of anti-bacterial action and ostogenesis-promotion have been developed and demonstrated excellent therapeutic effects. Compared with the conventional treatment method, the dual-functional composite scaffold can provide one-stage treatment avoiding multiple surgeries, thereby remarkably simplifying the treatment process and reducing the treatment time, overcoming the disadvantages of conventional bone transplantation. In this review, the impaired bone repair ability and its specific mechanisms in the microenvironment of pathogen infection and excessive inflammation were analyzed, providing a theoretical basis for the treatment of infectious bone defects. Furthermore, we discussed the composite dual-functional scaffold composed of a combination of antibacterial and osteogenic material. Finally, a series of advanced drug delivery systems with antibacterial and bone-promoting capabilities were summarized and discussed. This review provides a comprehensive understanding for the microenvironment of infectious bone defects and leading-edge design strategies for the antibacterial and bone-promoting dual-function scaffold, thus providing clinically significant treatment methods for infectious bone defects. Antibacterial and bone-promoting dual-function scaffolds are ideal strategies for treatment of infectious bone defects. The effect of infection on bone repair was summarized in detail from four important aspects. A variety of dual-function scaffolds based on antibacterial and osteogenic materials were discussed. Dual-function drug delivery systems promoting repair of infectious bone defects by locally releasing functional agents. Leading-edge design strategies, challenges and prospects for dual-functional biomaterials were provided.
Collapse
|
14
|
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
|
15
|
Petronglo JR, Putnam NE, Ford CA, Cruz-Victorio V, Curry JM, Butrico CE, Fulbright LE, Johnson JR, Peck SH, Fatah SR, Cassat JE. Context-Dependent Roles for Toll-Like Receptors 2 and 9 in the Pathogenesis of Staphylococcus aureus Osteomyelitis. Infect Immun 2022; 90:e0041722. [PMID: 36226943 PMCID: PMC9670883 DOI: 10.1128/iai.00417-22] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Staphylococcus aureus is the major causative agent of bacterial osteomyelitis, an invasive infection of bone. Inflammation generated by the immune response to S. aureus contributes to bone damage by altering bone homeostasis. Increases in the differentiation of monocyte lineage cells into bone-resorbing osteoclasts (osteoclastogenesis) promote bone loss in the setting of osteomyelitis. In this study, we sought to define the role of Toll-like receptor (TLR) signaling in the pathogenesis of S. aureus osteomyelitis. We hypothesized that S. aureus-sensing TLRs 2 and 9, both of which are known to alter osteoclastogenesis in vitro, promote pathological changes to bone, including increased osteoclast abundance, bone loss, and altered callus formation during osteomyelitis. Stimulation of osteoclast precursors with S. aureus supernatant increased osteoclastogenesis in a TLR2-dependent, but not a TLR9-dependent, manner. However, in vivo studies using a posttraumatic murine model of osteomyelitis revealed that TLR2-null mice experienced similar bone damage and increased osteoclastogenesis compared to wild type (WT) mice. Therefore, we tested the hypothesis that compensation between TLR2 and TLR9 contributes to osteomyelitis pathogenesis. We found that mice deficient in both TLR2 and TLR9 (Tlr2/9-/-) have decreased trabecular bone loss in response to infection compared to WT mice. However, osteoclastogenesis is comparable between WT and Tlr2/9-/- mice, suggesting that alternative mechanisms enhance osteoclastogenesis in vivo during osteomyelitis. Indeed, we discovered that osteoclast precursors intracellularly infected with S. aureus undergo significantly increased osteoclast formation, even in the absence of TLR2 and TLR9. These results suggest that TLR2 and TLR9 have context-dependent roles in the alteration of bone homeostasis during osteomyelitis.
Collapse
Affiliation(s)
- Jenna R. Petronglo
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Centergrid.412807.8, Nashville, Tennessee, USA
| | - Nicole E. Putnam
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Centergrid.412807.8, Nashville, Tennessee, USA
| | - Caleb A. Ford
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee, USA
| | - Virginia Cruz-Victorio
- Department of Pediatrics, Division of Pediatric Infectious Diseases, Vanderbilt University Medical Centergrid.412807.8, Nashville, Tennessee, USA
| | - Jacob M. Curry
- Department of Pediatrics, Division of Pediatric Infectious Diseases, Vanderbilt University Medical Centergrid.412807.8, Nashville, Tennessee, USA
| | - Casey E. Butrico
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Centergrid.412807.8, Nashville, Tennessee, USA
| | - Laura E. Fulbright
- Department of Pediatrics, Division of Pediatric Infectious Diseases, Vanderbilt University Medical Centergrid.412807.8, Nashville, Tennessee, USA
| | - Joshua R. Johnson
- Vanderbilt Center for Bone Biology, Vanderbilt University Medical Centergrid.412807.8, Nashville, Tennessee, USA
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Centergrid.412807.8, Nashville, Tennessee, USA
| | - Sun H. Peck
- Vanderbilt Center for Bone Biology, Vanderbilt University Medical Centergrid.412807.8, Nashville, Tennessee, USA
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Centergrid.412807.8, Nashville, Tennessee, USA
| | - Sana R. Fatah
- Department of Pediatrics, Division of Pediatric Infectious Diseases, Vanderbilt University Medical Centergrid.412807.8, Nashville, Tennessee, USA
| | - James E. Cassat
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Centergrid.412807.8, Nashville, Tennessee, USA
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee, USA
- Department of Pediatrics, Division of Pediatric Infectious Diseases, Vanderbilt University Medical Centergrid.412807.8, Nashville, Tennessee, USA
- Vanderbilt Center for Bone Biology, Vanderbilt University Medical Centergrid.412807.8, Nashville, Tennessee, USA
- Vanderbilt Institute for Infection, Immunology, and Inflammation (VI4), Vanderbilt University Medical Centergrid.412807.8, Nashville, Tennessee, USA
| |
Collapse
|
16
|
Tong Z, Chen Z, Li Z, Xie Z, Zhang H. Mechanisms of promoting the differentiation and bone resorption function of osteoclasts by Staphylococcus aureus infection. Int J Med Microbiol 2022; 312:151568. [PMID: 36240531 DOI: 10.1016/j.ijmm.2022.151568] [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: 03/19/2022] [Revised: 08/24/2022] [Accepted: 09/26/2022] [Indexed: 01/18/2023] Open
Abstract
Bone infection is a common and serious complication in the field of orthopedics, which frequently leads to excessive bone destruction and fracture nonunion. Staphylococcus aureus (S. aureus) infection affects bone cell function which, in turn, causes bone destruction. Bone is mainly regulated by osteoblasts and osteoclasts. Osteoclasts are the only cell type with bone resorptive function. Their over-activation is closely associated with excessive bone loss. Understanding how S. aureus changes the functional state of osteoclasts is the key to effective treatment. By reviewing the literature, this paper summarizes several mechanisms of bone destruction caused by S. aureus influencing osteoclasts, thereby stimulating new ideas for the treatment of bone infection.
Collapse
Affiliation(s)
- Zelei Tong
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Zhihao Chen
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Ziyuan Li
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Zonggang Xie
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, Suzhou, China.
| | - Haifang Zhang
- Department of Clinical Laboratory, The Second Affiliated Hospital of Soochow University, Suzhou, China.
| |
Collapse
|
17
|
Wu S, Wu B, Liu Y, Deng S, Lei L, Zhang H. Mini Review Therapeutic Strategies Targeting for Biofilm and Bone Infections. Front Microbiol 2022; 13:936285. [PMID: 35774451 PMCID: PMC9238355 DOI: 10.3389/fmicb.2022.936285] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Accepted: 05/25/2022] [Indexed: 12/21/2022] Open
Abstract
Bone infection results in a complex inflammatory response and bone destruction. A broad spectrum of bacterial species has been involved for jaw osteomyelitis, hematogenous osteomyelitis, vertebral osteomyelitis or diabetes mellitus, such as Staphylococcus aureus (S. aureus), coagulase-negative Staphylococcus species, and aerobic gram-negative bacilli. S. aureus is the major pathogenic bacterium for osteomyelitis, which results in a complex inflammatory response and bone destruction. Although various antibiotics have been applied for bone infection, the emergence of drug resistance and biofilm formation significantly decrease the effectiveness of those agents. In combination with gram-positive aerobes, gram-negative aerobes and anaerobes functionally equivalent pathogroups interact synergistically, developing as pathogenic biofilms and causing recurrent infections. The adhesion of biofilms to bone promotes bone destruction and protects bacteria from antimicrobial agent stress and host immune system infiltration. Moreover, bone is characterized by low permeability and reduced blood flow, further hindering the therapeutic effect for bone infections. To minimize systemic toxicity and enhance antibacterial effectiveness, therapeutic strategies targeting on biofilm and bone infection can serve as a promising modality. Herein, we focus on biofilm and bone infection eradication with targeting therapeutic strategies. We summarize recent targeting moieties on biofilm and bone infection with peptide-, nucleic acid-, bacteriophage-, CaP- and turnover homeostasis-based strategies. The antibacterial and antibiofilm mechanisms of those therapeutic strategies include increasing antibacterial agents’ accumulation by bone specific affinity, specific recognition of phage-bacteria, inhibition biofilm formation in transcription level. As chronic inflammation induced by infection can trigger osteoclast activation and inhibit osteoblast functioning, we additionally expand the potential applications of turnover homeostasis-based therapeutic strategies on biofilm or infection related immunity homeostasis for host-bacteria. Based on this review, we expect to provide useful insights of targeting therapeutic efficacy for biofilm and bone infection eradication.
Collapse
Affiliation(s)
- Shizhou Wu
- Department of Orthopedic Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Binjie Wu
- Department of Orthopedic Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Yunjie Liu
- West China School of Public Health, Sichuan University, Chengdu, China
| | - Shu Deng
- Boston University Henry M. Goldman School of Dental Medicine, Boston, MA, United States
| | - Lei Lei
- West China Hospital of Stomatology, Sichuan University, Chengdu, China
- *Correspondence: Lei Lei,
| | - Hui Zhang
- Department of Orthopedic Surgery, West China Hospital, Sichuan University, Chengdu, China
- Hui Zhang,
| |
Collapse
|
18
|
Massaccesi L, Galliera E, Pellegrini A, Banfi G, Corsi Romanelli MM. Osteomyelitis, Oxidative Stress and Related Biomarkers. Antioxidants (Basel) 2022; 11:antiox11061061. [PMID: 35739958 PMCID: PMC9220672 DOI: 10.3390/antiox11061061] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2022] [Revised: 05/24/2022] [Accepted: 05/25/2022] [Indexed: 12/30/2022] Open
Abstract
Bone is a very dynamic tissue, subject to continuous renewal to maintain homeostasis through bone remodeling, a process promoted by two cell types: osteoblasts, of mesenchymal derivation, are responsible for the deposition of new material, and osteoclasts, which are hematopoietic cells, responsible for bone resorption. Osteomyelitis (OM) is an invasive infectious process, with several etiological agents, the most common being Staphylococcus aureus, affecting bone or bone marrow, and severely impairing bone homeostasis, resulting in osteolysis. One of the characteristic features of OM is a strong state of oxidative stress (OS) with severe consequences on the delicate balance between osteoblastogenesis and osteoclastogenesis. Here we describe this, analyzing the effects of OS in bone remodeling and discussing the need for new, easy-to-measure and widely available OS biomarkers that will provide valid support in the management of the disease.
Collapse
Affiliation(s)
- Luca Massaccesi
- Department of Biomedical Sciences for Health, Università degli Studi di Milano, 20133 Milan, Italy; (E.G.); (M.M.C.R.)
- Correspondence: ; Tel.: +39-0250316027
| | - Emanuela Galliera
- Department of Biomedical Sciences for Health, Università degli Studi di Milano, 20133 Milan, Italy; (E.G.); (M.M.C.R.)
- IRCCS Galeazzi Orthopaedic Institute, 20161 Milan, Italy;
| | - Antonio Pellegrini
- Centre for Reconstructive Surgery and Osteoarticular Infections, IRCCS Galeazzi Orthopaedic Institute, 20161 Milan, Italy;
| | - Giuseppe Banfi
- IRCCS Galeazzi Orthopaedic Institute, 20161 Milan, Italy;
| | - Massimiliano Marco Corsi Romanelli
- Department of Biomedical Sciences for Health, Università degli Studi di Milano, 20133 Milan, Italy; (E.G.); (M.M.C.R.)
- Service of Laboratory Medicine1-Clinical Pathology, IRCCS Policlinico San Donato, San Donato Milanese, 20097 Milan, Italy
| |
Collapse
|
19
|
Profile analysis of circRNAs in human THP-1 derived macrophages infected with intracellular Staphylococcus aureus. Microb Pathog 2022; 165:105466. [PMID: 35247499 DOI: 10.1016/j.micpath.2022.105466] [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: 08/05/2021] [Revised: 01/13/2022] [Accepted: 02/28/2022] [Indexed: 11/24/2022]
Abstract
BACKGROUND Intracellular Staphylococcus aureus (S. aureus) infection is generally persistent, recurrent and difficult to treat due to the poor availability of antibiotics within macrophages cells and the lack of ideal diagnostic markers. Circular RNAs (circRNAs), with covalently closed circular structures, exists in the serum stably and is not easily degraded by nucleases. Besides, circRNAs play a pivotal in the eukaryotic regulation of genes expression and served as biomarkers in variety disease including microbial infections. However, the function of host circRNAs in intracellular S. aureus infection remains largely unclear. METHODS In this study, the circRNAs expression profile was investigated by RNA sequencing technology in both S. aureus-infected THP-1 derived macrophages and mock control cells. The differentially expressed circRNAs (DE circRNAs) with a fold-change >1.5 (p < 0.05) are analyzed using functional pathway clustering prediction. Then, RT-qPCR was performed to verify the top 2 up-regulated circRNAs in the THP-1 cell and human serum samples so as to evaluate the value of circRNAs for S. aureus diagnosis. RESULTS An intracellular survival THP-1 derived macrophages model of S. aureus infection was established. A total of 5,299 circRNAs were identified in human THP-1 derived macrophages infected with intracellular S. aureus. There were 61 DE circRNAs with a fold-change >1.5 (p < 0.05) after S. aureus infection. Among them, 22 circRNAs were up-regulated while 39 circRNAs down-regulated. GO and KEGG pathway analysis demonstrated that DE circRNAs were enriched in the processes such as Neurotrophin, Pyruvate metabolism and Notch signaling pathway. Moreover, hsa_circ_0000311 and chr13:43500472-43544806-(novel) were verified to be significantly upregulated in THP-1 derived macrophages and human serum samples between two groups. Finally, the networks of circRNA-miRNA-mRNA based on these two circRNAs were constructed respectively. CONCLUSION Our study provides the first profile analysis of host circRNAs involved in intracellular S. aureus infection, which may serve as biomarkers for S. aureus diagnosis and contribute to the understanding of S. aureus evasion mechanisms.
Collapse
|
20
|
Accioni F, Vázquez J, Merinero M, Begines B, Alcudia A. Latest Trends in Surface Modification for Dental Implantology: Innovative Developments and Analytical Applications. Pharmaceutics 2022; 14:455. [PMID: 35214186 PMCID: PMC8876580 DOI: 10.3390/pharmaceutics14020455] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Revised: 02/15/2022] [Accepted: 02/18/2022] [Indexed: 12/27/2022] Open
Abstract
An increase in the world population and its life expectancy, as well as the ongoing concern about our physical appearance, have elevated the relevance of dental implantology in recent decades. Engineering strategies to improve the survival rate of dental implants have been widely investigated, focusing on implant material composition, geometry (usually guided to reduce stiffness), and interface surrounding tissues. Although efforts to develop different implant surface modifications are being applied in commercial dental prostheses today, the inclusion of surface coatings has gained special interest, as they can be tailored to efficiently enhance osseointegration, as well as to reduce bacterial-related infection, minimizing peri-implantitis appearance and its associated risks. The use of biomaterials to replace teeth has highlighted the need for the development of reliable analytical methods to assess the therapeutic benefits of implants. This literature review considers the state-of-the-art strategies for surface modification or coating and analytical methodologies for increasing the survival rate for teeth restoration.
Collapse
Affiliation(s)
- Francesca Accioni
- Departamento de Química Orgánica y Farmacéutica, Universidad de Sevilla, 41012 Seville, Spain; (F.A.); (M.M.)
| | - Juan Vázquez
- Departamento de Química Orgánica, Universidad de Sevilla, 41012 Seville, Spain;
| | - Manuel Merinero
- Departamento de Química Orgánica y Farmacéutica, Universidad de Sevilla, 41012 Seville, Spain; (F.A.); (M.M.)
- Departamento de Citología e Histología Normal y Patológica, Universidad de Sevilla, 41012 Seville, Spain
| | - Belén Begines
- Departamento de Química Orgánica y Farmacéutica, Universidad de Sevilla, 41012 Seville, Spain; (F.A.); (M.M.)
| | - Ana Alcudia
- Departamento de Química Orgánica y Farmacéutica, Universidad de Sevilla, 41012 Seville, Spain; (F.A.); (M.M.)
| |
Collapse
|
21
|
Chen Y, Huang C, Chen X, Cai Y, Li W, Fang X, Zhang W. Bone protein analysis via label-free quantitative proteomics in patients with periprosthetic joint infection. J Proteomics 2022; 252:104448. [PMID: 34883267 DOI: 10.1016/j.jprot.2021.104448] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2021] [Revised: 11/18/2021] [Accepted: 11/23/2021] [Indexed: 11/17/2022]
Abstract
Periprosthetic joint infection (PJI) is a catastrophic complication of arthroplasty. The treatment of PJI often requires multiple operations and long-term use of antibiotics, making PJI a substantial health and economic burden for patients. Therefore, there is an urgent need to elucidate the pathological mechanism of PJI to explore new therapeutic methods. This study aimed to explore proteomics changes in bone tissue around the prosthesis during PJI development, to explain the pathological mechanism and to provide new treatment ideas. Ten patients who underwent revision surgery at our institution were included: 5 patients with Staphylococcus aureus PJI and 5 patients with aseptic failure. The proteomics changes in bone tissues after PJI were investigated by label-free quantitative proteomics, and the pathways affected by the differential proteins were analyzed by GO annotation, GO enrichment analysis, KEGG enrichment analysis and protein-protein interaction network analysis. We identified 435 differentially expressed proteins (DEPs), with 213 upregulated and 222 downregulated proteins. Analysis revealed activation of immune-related pathways, such as complement and coagulation cascades, phagocytosis, and neutrophil activation, and inhibition of energy metabolism pathways represented by the TCA cycle. We also observed an altered balance between osteoblasts and osteoclasts during S. aureus PJI. We hope that these processes will reveal new treatment ideas. SIGNIFICANCE: PJI is a catastrophic complication of arthroplasty. When infection occurs, bacteria may invade periprosthetic bone tissue to escape immunity and cause damage. So far, only few studies focused on the changes of proteomics associated to PJI. This is the first study to describe the proteomics changes of periprosthetic bone tissue of patients with PJI. We found that the pathological process of S. aureus PJI mainly involves activation of the immune system, decreased energy metabolism, and an altered balance of osteoblasts and osteoclasts.
Collapse
Affiliation(s)
- Yang Chen
- Department of Orthopedic Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Changyu Huang
- Department of Orthopedic Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Xiaoqing Chen
- Department of Orthopedic Surgery, Quanzhou First Hospital Affiliated to Fujian Medical University, Quanzhou, China
| | - Yuanqing Cai
- Department of Orthopedic Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Wenbo Li
- Department of Orthopedic Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Xinyu Fang
- Department of Orthopedic Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China.
| | - Wenming Zhang
- Department of Orthopedic Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China.
| |
Collapse
|
22
|
Sun Y, Li J, Xie X, Gu F, Sui Z, Zhang K, Yu T. Macrophage-Osteoclast Associations: Origin, Polarization, and Subgroups. Front Immunol 2021; 12:778078. [PMID: 34925351 PMCID: PMC8672114 DOI: 10.3389/fimmu.2021.778078] [Citation(s) in RCA: 101] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Accepted: 11/15/2021] [Indexed: 12/31/2022] Open
Abstract
Cellular associations in the bone microenvironment are involved in modulating the balance between bone remodeling and resorption, which is necessary for maintaining a normal bone morphology. Macrophages and osteoclasts are both vital components of the bone marrow. Macrophages can interact with osteoclasts and regulate bone metabolism by secreting a variety of cytokines, which make a significant contribution to the associations. Although, recent studies have fully explored either macrophages or osteoclasts, indicating the significance of these two types of cells. However, it is of high importance to report the latest discoveries on the relationships between these two myeloid-derived cells in the field of osteoimmunology. Therefore, this paper reviews this topic from three novel aspects of the origin, polarization, and subgroups based on the previous work, to provide a reference for future research and treatment of bone-related diseases.
Collapse
Affiliation(s)
- Yang Sun
- Department of Orthopedics, The First Hospital of Jilin University, Changchun, China
| | - Jiangbi Li
- Department of Orthopedics, The First Hospital of Jilin University, Changchun, China
| | - Xiaoping Xie
- Department of Orthopedics, The First Hospital of Jilin University, Changchun, China
| | - Feng Gu
- Department of Orthopedics, The First Hospital of Jilin University, Changchun, China
| | - Zhenjiang Sui
- Department of Orthopedics, The First Hospital of Jilin University, Changchun, China
| | - Ke Zhang
- Department of Orthopedics, The First Hospital of Jilin University, Changchun, China
| | - Tiecheng Yu
- Department of Orthopedics, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
23
|
Li Y, Wang S, Li S, Fei J. Clindamycin-loaded titanium prevents implant-related infection through blocking biofilm formation. J Biomater Appl 2021; 36:1231-1242. [PMID: 34723682 DOI: 10.1177/08853282211051183] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Implant-related infection is a disastrous complication. Surface modification of titanium is considered as an important strategy to prevent implant-related infection. However, there is no recognized surface modification strategy that can be applied in clinic so far. We explored a new strategy of coating. The clindamycin-loaded titanium was constructed by layer-by-layer self-assembly. The release of clindamycin from titanium was detected through high performance liquid chromatography. Different titanium was co-cultured with Staphylococcus aureus for 24 h in vitro, then the effect of different titanium on bacterial colonization and biofilm formation was determined by spread plate method and scanning electron microscopy. Cytotoxicity and cytocompatibility of clindamycin-loaded titanium on MC3T3-E1 cells were measured by CCK8. The antibacterial ability of clindamycin-loaded titanium in vivo was also evaluated using a rat model of osteomyelitis. The number of osteoclasts in bone defect was observed by tartrate-resistant acid phosphatase staining. Bacterial burden of surrounding tissues around the site of infection was calculated by tissue homogenate and colony count. Clindamycin-loaded titanium could release clindamycin slowly within 160 h. It reduced bacterial colonization by three orders of magnitude compare to control (p < .05) and inhibits biofilm formation in vitro. Cells proliferation and adhesion were similar on three titanium surfaces (p > .05). In vivo, clindamycin-loaded titanium improved bone healing, reduced microbial burden, and decreased the number of osteoclasts compared control titanium in the rat model of osteomyelitis. This study demonstrated that clindamycin-loaded titanium exhibited good biocompatibility, and showed antibacterial activity both in vivo and in vitro. It is promising and might have potential for clinical application.
Collapse
Affiliation(s)
- Youbin Li
- 12525Department of Emergency Medicine of Army Medical Center, Army Medical University, Chongqing, China
| | - Shaochuan Wang
- 12525Department of Emergency Medicine of Army Medical Center, Army Medical University, Chongqing, China
| | - Shidan Li
- 12525Department of Emergency Medicine of Army Medical Center, Army Medical University, Chongqing, China
| | - Jun Fei
- 12525Department of Emergency Medicine of Army Medical Center, Army Medical University, Chongqing, China.,12525State Key Laboratory of Trauma, Burns and Combined Injury, Army Medical University, Chongqing, China
| |
Collapse
|
24
|
Staphylococcus aureus internalization impairs osteoblastic activity and early differentiation process. Sci Rep 2021; 11:17685. [PMID: 34480054 PMCID: PMC8417294 DOI: 10.1038/s41598-021-97246-y] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Accepted: 06/07/2021] [Indexed: 11/13/2022] Open
Abstract
Staphylococcus aureus is the most frequent aetiology of bone and joint infections (BJI) and can cause relapsing and chronic infections. One of the main factors involved in the chronicization of staphylococcal BJIs is the internalization of S. aureus into osteoblasts, the bone-forming cells. Previous studies have shown that S. aureus triggers an impairment of osteoblasts function that could contribute to bone loss. However, these studies focused mainly on the extracellular effects of S. aureus. Our study aimed at understanding the intracellular effects of S. aureus on the early osteoblast differentiation process. In our in vitro model of osteoblast lineage infection, we first observed that internalized S. aureus 8325-4 (a reference lab strain) significantly impacted RUNX2 and COL1A1 expression compared to its non-internalized counterpart 8325-4∆fnbAB (with deletion of fnbA and fnbB). Then, in a murine model of osteomyelitis, we reported no significant effect for S. aureus 8325-4 and 8325-4∆fnbAB on bone parameters at 7 days post-infection whereas S. aureus 8325-4 significantly decreased trabecular bone thickness at 14 days post-infection compared to 8325-4∆fnbAB. When challenged with two clinical isogenic strains isolated from initial and relapse phase of the same BJI, significant impairments of bone parameters were observed for both initial and relapse strain, without differences between the two strains. Finally, in our in vitro osteoblast infection model, both clinical strains impacted alkaline phosphatase activity whereas the expression of bone differentiation genes was significantly decreased only after infection with the relapse strain. Globally, we highlighted that S. aureus internalization into osteoblasts is responsible for an impairment of the early differentiation in vitro and that S. aureus impaired bone parameters in vivo in a strain-dependent manner.
Collapse
|
25
|
Hersh A, Young R, Pennington Z, Ehresman J, Ding A, Kopparapu S, Cottrill E, Sciubba DM, Theodore N. Removal of instrumentation for postoperative spine infection: systematic review. J Neurosurg Spine 2021; 35:376-388. [PMID: 34243152 DOI: 10.3171/2020.12.spine201300] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Accepted: 12/14/2020] [Indexed: 11/06/2022]
Abstract
OBJECTIVE Currently, no consensus exists as to whether patients who develop infection of the surgical site after undergoing instrumented fusion should have their implants removed at the time of wound debridement. Instrumentation removal may eliminate a potential infection nidus, but removal may also destabilize the patient's spine. The authors sought to summarize the existing evidence by systematically reviewing published studies that compare outcomes between patients undergoing wound washout and instrumentation removal with outcomes of patients undergoing wound washout alone. The primary objectives were to determine 1) whether instrumentation removal from an infected wound facilitates infection clearance and lowers morbidity, and 2) whether the chronicity of the underlying infection affects the decision to remove instrumentation. METHODS PRISMA guidelines were used to review the PubMed/MEDLINE, Embase, Cochrane Library, Scopus, Web of Science, and ClinicalTrials.gov databases to identify studies that compared patients with implants removed and patients with implants retained. Outcomes of interest included mortality, rate of repeat wound washout, and loss of correction. RESULTS Fifteen articles were included. Of 878 patients examined in these studies, 292 (33%) had instrumentation removed. Patient populations were highly heterogeneous, and outcome data were limited. Available data suggested that rates of reoperation, pseudarthrosis, and death were higher in patients who underwent instrumentation removal at the time of initial washout. Three studies recommended that instrumentation be uniformly removed at the time of wound washout. Five studies favored retaining the original instrumentation. Six studies favored retention in early infections but removal in late infections. CONCLUSIONS The data on this topic remain heterogeneous and low in quality. Retention may be preferred in the setting of early infection, when the risk of underlying spine instability is still high and the risk of mature biofilm formation on the implants is low. However, late infections likely favor instrumentation removal. Higher-quality evidence from large, multicenter, prospective studies is needed to reach generalizable conclusions capable of guiding clinical practice.
Collapse
|
26
|
Dufrançais O, Mascarau R, Poincloux R, Maridonneau-Parini I, Raynaud-Messina B, Vérollet C. Cellular and molecular actors of myeloid cell fusion: podosomes and tunneling nanotubes call the tune. Cell Mol Life Sci 2021; 78:6087-6104. [PMID: 34296319 PMCID: PMC8429379 DOI: 10.1007/s00018-021-03875-x] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 05/25/2021] [Accepted: 06/05/2021] [Indexed: 12/22/2022]
Abstract
Different types of multinucleated giant cells (MGCs) of myeloid origin have been described; osteoclasts are the most extensively studied because of their importance in bone homeostasis. MGCs are formed by cell-to-cell fusion, and most types have been observed in pathological conditions, especially in infectious and non-infectious chronic inflammatory contexts. The precise role of the different MGCs and the mechanisms that govern their formation remain poorly understood, likely due to their heterogeneity. First, we will introduce the main populations of MGCs derived from the monocyte/macrophage lineage. We will then discuss the known molecular actors mediating the early stages of fusion, focusing on cell-surface receptors involved in the cell-to-cell adhesion steps that ultimately lead to multinucleation. Given that cell-to-cell fusion is a complex and well-coordinated process, we will also describe what is currently known about the evolution of F-actin-based structures involved in macrophage fusion, i.e., podosomes, zipper-like structures, and tunneling nanotubes (TNT). Finally, the localization and potential role of the key fusion mediators related to the formation of these F-actin structures will be discussed. This review intends to present the current status of knowledge of the molecular and cellular mechanisms supporting multinucleation of myeloid cells, highlighting the gaps still existing, and contributing to the proposition of potential disease-specific MGC markers and/or therapeutic targets.
Collapse
Affiliation(s)
- Ophélie Dufrançais
- Institut de Pharmacologie et Biologie Structurale, IPBS, Université de Toulouse, CNRS, UPS, Toulouse, France
| | - Rémi Mascarau
- Institut de Pharmacologie et Biologie Structurale, IPBS, Université de Toulouse, CNRS, UPS, Toulouse, France
- International Associated Laboratory (LIA) CNRS "IM-TB/HIV" (1167), Toulouse, France
- International Associated Laboratory (LIA) CNRS "IM-TB/HIV" (1167), Buenos Aires, Argentina
| | - Renaud Poincloux
- Institut de Pharmacologie et Biologie Structurale, IPBS, Université de Toulouse, CNRS, UPS, Toulouse, France
| | - Isabelle Maridonneau-Parini
- Institut de Pharmacologie et Biologie Structurale, IPBS, Université de Toulouse, CNRS, UPS, Toulouse, France
- International Associated Laboratory (LIA) CNRS "IM-TB/HIV" (1167), Toulouse, France
| | - Brigitte Raynaud-Messina
- Institut de Pharmacologie et Biologie Structurale, IPBS, Université de Toulouse, CNRS, UPS, Toulouse, France.
- International Associated Laboratory (LIA) CNRS "IM-TB/HIV" (1167), Toulouse, France.
- International Associated Laboratory (LIA) CNRS "IM-TB/HIV" (1167), Buenos Aires, Argentina.
| | - Christel Vérollet
- Institut de Pharmacologie et Biologie Structurale, IPBS, Université de Toulouse, CNRS, UPS, Toulouse, France.
- International Associated Laboratory (LIA) CNRS "IM-TB/HIV" (1167), Toulouse, France.
- International Associated Laboratory (LIA) CNRS "IM-TB/HIV" (1167), Buenos Aires, Argentina.
| |
Collapse
|
27
|
CHI3L1 promotes Staphylococcus aureus-induced osteomyelitis by activating p38/MAPK and Smad signaling pathways. Exp Cell Res 2021; 403:112596. [PMID: 33826950 DOI: 10.1016/j.yexcr.2021.112596] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Revised: 03/21/2021] [Accepted: 04/01/2021] [Indexed: 12/26/2022]
Abstract
AIMS Staphylococcus aureus (S. aureus) is the most common causative bacterial pathogen involved in promoting infection-induced osteomyelitis, a disease resulting in severe bone degradation. In this study, we aimed to identify the mechanism behind inhibition of osteoclast survival and differentiation by CHI3L1, a lectin previously reported to regulate S. aureus-induced osteomyelitis. MAIN METHODS The role of CHI3L1 in osteoclast survival, proliferation, and differentiation was studied ex vivo using primary human bone marrow derived stem cells (HBMSCs) and transducing them with lentiviral expression vectors or shRNA knockdown constructs. Cell apoptosis was analyzed by flow cytometry using annexin V-fluorescein isothiocyanate/propidium iodide staining. Cell proliferation was assessed using alkaline phosphatase, Alcian Blue, and TRAP staining. The qRT-PCR was used to measure mRNA levels of osteoclast maturation markers, and western blotting was used to analyze protein expression. An in vivo murine model for osteomyelitis and microcomputed tomography analyses of infected femurs were used to study the effects of CHI3L1 on bone erosion. KEY FINDINGS Overexpression of CHI3L1 significantly reduced HBMSC cell viability, proliferation, and differentiation, and knockdown improved these effects in the presence of S. aureus infection. More specifically, CHI3L1 constitutively activated the p38/MAPK pathway to promote apoptosis. Furthermore, CHI3L1 induced activation of the Smad pathway by promoting phosphorylation of Smad-1/5 proteins. Finally, overexpression of CHI3L1 significantly induced bone erosion upon S. aureus infection in a murine osteomyelitis model, and knockdown of CHI3L1 significantly alleviated this effect. SIGNIFICANCE CHI3L1 played a vital role in osteoblast differentiation and proliferation by regulating the p38/MAPK and Smad signaling pathways to promote S. aureus-induced osteomyelitis.
Collapse
|
28
|
López-Valverde N, Macedo-de-Sousa B, López-Valverde A, Ramírez JM. Effectiveness of Antibacterial Surfaces in Osseointegration of Titanium Dental Implants: A Systematic Review. Antibiotics (Basel) 2021; 10:antibiotics10040360. [PMID: 33800702 PMCID: PMC8066819 DOI: 10.3390/antibiotics10040360] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 03/23/2021] [Accepted: 03/26/2021] [Indexed: 12/15/2022] Open
Abstract
Titanium (Ti) dental implant failure as a result of infection has been established at 40%, being regarded as one of the most habitual and untreatable problems. Current research is focused on the design of new surfaces that can generate long-lasting, infection-free osseointegration. The purpose of our study was to assess studies on Ti implants coated with different antibacterial surfaces, assessing their osseointegration. The PubMed, Web of Science and Scopus databases were electronically searched for in vivo studies up to December 2020, selecting six studies that met the inclusion criteria. The quality of the selected studies was assessed using the ARRIVE (Animal Research: Reporting of In Vivo Experiments) criteria and Systematic Review Center for Laboratory animal Experimentation's (SYRCLE's) risk of bias tool. Although all the included studies, proved greater osseointegration capacity of the different antibacterial surfaces studied, the methodological quality and experimental models used in some of them make it difficult to draw predictable conclusions. Because of the foregoing, we recommend caution when interpreting the results obtained.
Collapse
Affiliation(s)
- Nansi López-Valverde
- Department of Surgery, Instituto de Investigación Biomédica de Salamanca (IBSAL), University of Salamanca, 37007 Salamanca, Spain;
| | - Bruno Macedo-de-Sousa
- Institute for Occlusion and Orofacial Pain, Faculty of Medicine, University of Coimbra, Polo I-Edifício Central Rua Larga, 3004-504 Coimbra, Portugal;
| | - Antonio López-Valverde
- Department of Surgery, Instituto de Investigación Biomédica de Salamanca (IBSAL), University of Salamanca, 37007 Salamanca, Spain;
- Correspondence:
| | - Juan Manuel Ramírez
- Department of Morphological Sciences, University of Cordoba, Avenida Menéndez Pidal S/N, 14071 Cordoba, Spain;
| |
Collapse
|
29
|
Chen ZY, Gao S, Zhang YW, Zhou RB, Zhou F. Antibacterial biomaterials in bone tissue engineering. J Mater Chem B 2021; 9:2594-2612. [PMID: 33666632 DOI: 10.1039/d0tb02983a] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Bone infection is a devastating disease characterized by recurrence, drug-resistance, and high morbidity, that has prompted clinicians and scientists to develop novel approaches to combat it. Currently, although numerous biomaterials that possess excellent biocompatibility, biodegradability, porosity, and mechanical strength have been developed, their lack of effective antibacterial ability substantially limits bone-defect treatment efficacy. There is, accordingly, a pressing need to design antibacterial biomaterials for effective bone-infection prevention and treatment. This review focuses on antibacterial biomaterials and strategies; it presents recently reported biomaterials, including antibacterial implants, antibacterial scaffolds, antibacterial hydrogels, and antibacterial bone cement types, and aims to provide an overview of these antibacterial materials for application in biomedicine. The antibacterial mechanisms of these materials are discussed as well.
Collapse
Affiliation(s)
- Zheng-Yang Chen
- Orthopedic Department, Peking University Third Hospital, Beijing 100191, China.
| | | | | | | | | |
Collapse
|
30
|
Extracellular matrix scaffold crosslinked with vancomycin for multifunctional antibacterial bone infection therapy. Biomaterials 2020; 268:120603. [PMID: 33378735 DOI: 10.1016/j.biomaterials.2020.120603] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2020] [Revised: 12/10/2020] [Accepted: 12/11/2020] [Indexed: 12/22/2022]
Abstract
The treatment of acute and chronic bone infections remains a major clinical challenge. The various factors released by the bacteria, acidic environment, and bacterial colonies in the bone grooves and implanted synthetic materials collectively promote the formation of biofilms. Dormant bacteria and biofilms cause infections that are difficult to cure and that can develop chronically. Therefore, a new antibacterial material was synthesized in the present study for multifunctional bone infection therapy and consists of specific demineralized extracellular cancellous bone (SDECM) crosslinked with vancomycin (Van) by means of electrostatic interactions and chemical bonds. It was verified in vitro that the new material (Van-SDECM) not only has pH-sensitive release and biofilm inhibition properties, but also maintains sustained bactericidal ability accompanied by the degradation of the scaffold, which does not affect its favorable osteogenic performance. The infectious bone defect in vivo model further confirms the comprehensive anti-infective and osteogenic ability of the Van-SDECM. Further, these favorable properties are due to the pH-sensitive sustained release sterilization and scaffold contact antibacterial properties, accompanied by osteoclast activity inhibition, osteogenesis promotion and immunoregulation effects. This study provides a new drug-scaffold composite preparation method based on a native-derived extracellular matrix scaffold.
Collapse
|
31
|
Alder KD, Lee I, Munger AM, Kwon HK, Morris MT, Cahill SV, Back J, Yu KE, Lee FY. Intracellular Staphylococcus aureus in bone and joint infections: A mechanism of disease recurrence, inflammation, and bone and cartilage destruction. Bone 2020; 141:115568. [PMID: 32745687 DOI: 10.1016/j.bone.2020.115568] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2020] [Revised: 07/19/2020] [Accepted: 07/26/2020] [Indexed: 02/06/2023]
Abstract
Bone and joint infections are devastating afflictions. Although medical interventions and advents have improved their care, bone and joint infections still portend dismal outcomes. Indeed, bone and joint infections are associated with extremely high mortality and morbidity rates and, generally, occur secondary to the aggressive pathogen Staphylococcus aureus. The consequences of bone and joint infections are further compounded by the fact that although they are aggressively treated, they frequently recur and result in massive bone and articular cartilage loss. Here, we review the literature and chronicle the fact that the fundamental cellular components of the musculoskeletal system can be internally infected with Staphylococcus aureus, which explains the ready recurrence of bone and joint infections even after extensive administration of antibiotic therapy and debridement and offer potential treatment solutions for further study. Moreover, we review the ramifications of intracellular infection and expound that the massive bone and articular cartilage loss is caused by the sustained proinflammatory state induced by infection and offer potential combination therapies for further study to protect bone and cartilage.
Collapse
Affiliation(s)
- Kareme D Alder
- Department of Orthopædics & Rehabilitation, Yale University, School of Medicine, New Haven, CT, USA; Yale University School of Medicine, Department of Orthopaedics and Rehabilitation, 330 Cedar St, TMP 523, PO Box 208071, New Haven, CT 06520-8071, USA.
| | - Inkyu Lee
- Department of Orthopædics & Rehabilitation, Yale University, School of Medicine, New Haven, CT, USA; Department of Life Science, Chung-Ang University, Seoul, Republic of Korea; Yale University School of Medicine, Department of Orthopaedics and Rehabilitation, 330 Cedar St, TMP 523, PO Box 208071, New Haven, CT 06520-8071, USA.
| | - Alana M Munger
- Department of Orthopædics & Rehabilitation, Yale University, School of Medicine, New Haven, CT, USA; Yale University School of Medicine, Department of Orthopaedics and Rehabilitation, 330 Cedar St, TMP 523, PO Box 208071, New Haven, CT 06520-8071, USA.
| | - Hyuk-Kwon Kwon
- Department of Orthopædics & Rehabilitation, Yale University, School of Medicine, New Haven, CT, USA; Yale University School of Medicine, Department of Orthopaedics and Rehabilitation, 330 Cedar St, TMP 523, PO Box 208071, New Haven, CT 06520-8071, USA.
| | - Montana T Morris
- Department of Orthopædics & Rehabilitation, Yale University, School of Medicine, New Haven, CT, USA; Yale University School of Medicine, Department of Orthopaedics and Rehabilitation, 330 Cedar St, TMP 523, PO Box 208071, New Haven, CT 06520-8071, USA.
| | - Sean V Cahill
- Department of Orthopædics & Rehabilitation, Yale University, School of Medicine, New Haven, CT, USA; Yale University School of Medicine, Department of Orthopaedics and Rehabilitation, 330 Cedar St, TMP 523, PO Box 208071, New Haven, CT 06520-8071, USA.
| | - JungHo Back
- Department of Orthopædics & Rehabilitation, Yale University, School of Medicine, New Haven, CT, USA; Yale University School of Medicine, Department of Orthopaedics and Rehabilitation, 330 Cedar St, TMP 523, PO Box 208071, New Haven, CT 06520-8071, USA.
| | - Kristin E Yu
- Department of Orthopædics & Rehabilitation, Yale University, School of Medicine, New Haven, CT, USA; Yale University School of Medicine, Department of Orthopaedics and Rehabilitation, 330 Cedar St, TMP 523, PO Box 208071, New Haven, CT 06520-8071, USA.
| | - Francis Y Lee
- Department of Orthopædics & Rehabilitation, Yale University, School of Medicine, New Haven, CT, USA; Yale University School of Medicine, Department of Orthopaedics and Rehabilitation, 330 Cedar St, TMP 523, PO Box 208071, New Haven, CT 06520-8071, USA.
| |
Collapse
|
32
|
Tang RH, Yang J, Fei J. New perspectives on traumatic bone infections. Chin J Traumatol 2020; 23:314-318. [PMID: 32847694 PMCID: PMC7718542 DOI: 10.1016/j.cjtee.2020.05.009] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Revised: 02/25/2020] [Accepted: 03/02/2020] [Indexed: 02/04/2023] Open
Abstract
In this paper, we review the results of previous studies and summarize the effects of various factors on the regulation of bone metabolism in traumatic bone infections. Infection-related bone destruction incorporates pathogens and iatrogenic factors in the process of bone resorption dominated by the skeletal and immune systems. The development of bone immunology has established a bridge of communication between the skeletal system and the immune system. Exploring the effects of pathogens, skeletal systems, immune systems, and antibacterials on bone repair in infectious conditions can help improve the treatment of these diseases.
Collapse
Affiliation(s)
- Ruo-Hui Tang
- Health Team of 96824 Troops of the Chinese People's Liberation Army, Kunming, China
| | - Jing Yang
- Emergency Department, Daping Hospital, Third Military Medical University, Chongqing, China
| | - Jun Fei
- Emergency Department, Daping Hospital, Third Military Medical University, Chongqing, China,State Key Laboratory of Trauma, Burns and Combined Injury, Third Military Medical University, Chongqing, China,Corresponding author. Emergency Department, Daping Hospital, Third Military Medical University, Chongqing, China.
| |
Collapse
|
33
|
Oliveira TC, Gomes MS, Gomes AC. The Crossroads between Infection and Bone Loss. Microorganisms 2020; 8:microorganisms8111765. [PMID: 33182721 PMCID: PMC7698271 DOI: 10.3390/microorganisms8111765] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Revised: 11/05/2020] [Accepted: 11/09/2020] [Indexed: 01/18/2023] Open
Abstract
Bone homeostasis, based on a tight balance between bone formation and bone degradation, is affected by infection. On one hand, some invading pathogens are capable of directly colonizing the bone, leading to its destruction. On the other hand, immune mediators produced in response to infection may dysregulate the deposition of mineral matrix by osteoblasts and/or the resorption of bone by osteoclasts. Therefore, bone loss pathologies may develop in response to infection, and their detection and treatment are challenging. Possible biomarkers of impaired bone metabolism during chronic infection need to be identified to improve the diagnosis and management of infection-associated osteopenia. Further understanding of the impact of infections on bone metabolism is imperative for the early detection, prevention, and/or reversion of bone loss. Here, we review the mechanisms responsible for bone loss as a direct and/or indirect consequence of infection.
Collapse
Affiliation(s)
- Tiago Carvalho Oliveira
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal; (T.C.O.); (M.S.G.)
- Faculdade de Ciências da Universidade do Porto, 4169-007 Porto, Portugal
- Instituto de Ciências Biomédicas de Abel Salazar da Universidade do Porto, 4050-313 Porto, Portugal
| | - Maria Salomé Gomes
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal; (T.C.O.); (M.S.G.)
- Instituto de Ciências Biomédicas de Abel Salazar da Universidade do Porto, 4050-313 Porto, Portugal
| | - Ana Cordeiro Gomes
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal; (T.C.O.); (M.S.G.)
- Correspondence:
| |
Collapse
|
34
|
Three-Dimensional In Vitro Staphylococcus aureus Abscess Communities Display Antibiotic Tolerance and Protection from Neutrophil Clearance. Infect Immun 2020; 88:IAI.00293-20. [PMID: 32817328 DOI: 10.1128/iai.00293-20] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Accepted: 08/06/2020] [Indexed: 12/22/2022] Open
Abstract
Staphylococcus aureus is a prominent human pathogen in bone and soft-tissue infections. Pathophysiology involves abscess formation, which consists of central staphylococcal abscess communities (SACs), surrounded by a fibrin pseudocapsule and infiltrating immune cells. Protection against the ingress of immune cells such as neutrophils, or tolerance to antibiotics, remains largely unknown for SACs and is limited by the lack of availability of in vitro models. We describe a three-dimensional in vitro model of SACs grown in a human plasma-supplemented collagen gel. The in vitro SACs reached their maximum size by 24 h and elaborated a fibrin pseudocapsule, as confirmed by electron and immunofluorescence microscopy. The in vitro SACs tolerated 100× the MIC of gentamicin alone and in combination with rifampin, while planktonic controls and mechanically dispersed SACs were efficiently killed. To simulate a host response, SACs were exposed to differentiated PLB-985 neutrophil-like (dPLB) cells and to primary human neutrophils at an early stage of SAC formation or after maturation at 24 h. Both cell types were unable to clear mature in vitro SACs, but dPLB cells prevented SAC growth upon early exposure before pseudocapsule maturation. Neutrophil exposure after plasmin pretreatment of the SACs resulted in a significant decrease in the number of bacteria within the SACs. The in vitro SAC model mimics key in vivo features, offers a new tool to study host-pathogen interactions and drug efficacy assessment, and has revealed the functionality of the S. aureus pseudocapsule in protecting the bacteria from host phagocytic responses and antibiotics.
Collapse
|
35
|
A comprehensive review of bacterial osteomyelitis with emphasis on Staphylococcus aureus. Microb Pathog 2020; 148:104431. [PMID: 32801004 DOI: 10.1016/j.micpath.2020.104431] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Revised: 07/29/2020] [Accepted: 07/29/2020] [Indexed: 02/04/2023]
Abstract
Osteomyelitis, a significant infection of bone tissue, gives rise to two main groups of infection: acute and chronic. These groups are further categorized in terms of the duration of infection. Usually, children and adults are more susceptible to acute and chronic infections, respectively. The aforementioned groups of osteomyelitis share almost 80% of the corresponding bacterial pathogens. Among all bacteria, Staphylococcus aureus (S. aureus) is a significant pathogen and is associated with a high range of osteomyelitis symptoms. S. aureus has many strategies for interacting with host cells including Small Colony Variant (SCV), biofilm formation, and toxin secretion. In addition, it induces an inflammatory response and causes host cell death by apoptosis and necrosis. However, any possible step to take in this respect is dependent on the conditions and host responses. In the absence of any immune responses and antibiotics, bacteria actively duplicate themselves; however, in the presence of phagocytic cell and harassing conditions, they turn into a SCV, remaining sustainable for a long time. SCV is characterized by notable advantages such as (a) intracellular life that mediates a dam against immune cells and (b) low ATP production that mediates resistance against antibiotics.
Collapse
|
36
|
Abstract
Osteomyelitis, or inflammation of bone, is most commonly caused by invasion of bacterial pathogens into the skeleton. Bacterial osteomyelitis is notoriously difficult to treat, in part because of the widespread antimicrobial resistance in the preeminent etiologic agent, the Gram-positive bacterium Staphylococcus aureus Bacterial osteomyelitis triggers pathological bone remodeling, which in turn leads to sequestration of infectious foci from innate immune effectors and systemically delivered antimicrobials. Treatment of osteomyelitis therefore typically consists of long courses of antibiotics in conjunction with surgical debridement of necrotic infected tissues. Even with these extreme measures, many patients go on to develop chronic infection or sustain disease comorbidities. A better mechanistic understanding of how bacteria invade, survive within, and trigger pathological remodeling of bone could therefore lead to new therapies aimed at prevention or treatment of osteomyelitis as well as amelioration of disease morbidity. In this minireview, we highlight recent developments in our understanding of how pathogens invade and survive within bone, how bacterial infection or resulting innate immune responses trigger changes in bone remodeling, and how model systems can be leveraged to identify new therapeutic targets. We review the current state of osteomyelitis epidemiology, diagnostics, and therapeutic guidelines to help direct future research in bacterial pathogenesis.
Collapse
|
37
|
Baude J, Bastien S, Gillet Y, Leblanc P, Itzek A, Tristan A, Bes M, Duguez S, Moreau K, Diep BA, Norrby-Teglund A, Henry T, Vandenesch F. Necrotizing Soft Tissue Infection Staphylococcus aureus but not S. pyogenes Isolates Display High Rates of Internalization and Cytotoxicity Toward Human Myoblasts. J Infect Dis 2020; 220:710-719. [PMID: 31001627 DOI: 10.1093/infdis/jiz167] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Accepted: 04/08/2019] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Necrotizing soft tissue infections (NSTIs) caused by group A Streptococcus (GAS) and occasionally by Staphylococcus aureus (SA) frequently involve the deep fascia and often lead to muscle necrosis. METHODS To assess the pathogenicity of GAS and S. aureus for muscles in comparison to keratinocytes, adhesion and invasion of NSTI-GAS and NSTI-SA isolates were assessed in these cells. Bloodstream infections (BSI-SA) and noninvasive coagulase-negative staphylococci (CNS) isolates were used as controls. RESULTS NSTI-SA and BSI-SA exhibited stronger internalization into human keratinocytes and myoblasts than NSTI-GAS or CNS. S. aureus internalization reached over 30% in human myoblasts due to a higher percentage of infected myoblasts (>11%) as compared to keratinocytes (<3%). Higher cytotoxicity for myoblasts of NSTI-SA as compared to BSI-SA was attributed to higher levels of psmα and RNAIII transcripts in NSTI-SA. However, the 2 groups were not discriminated at the genomic level. The cellular basis of high internalization rate in myoblasts was attributed to higher expression of α5β1 integrin in myoblasts. Major contribution of FnbpAB-integrin α5β1 pathway to internalization was confirmed by isogenic mutants. CONCLUSIONS Our findings suggest a factor in NSTI-SA severity is the strong invasiveness of S. aureus in muscle cells, a property not shared by NSTI-GAS isolates.
Collapse
Affiliation(s)
- Jessica Baude
- Centre International de Recherche en Infectiologie, Université de Lyon; Inserm U1111; Ecole Normale Supérieure de Lyon; Université Lyon 1, CNRS, UMR5308; Lyon, France
| | - Sylvère Bastien
- Centre International de Recherche en Infectiologie, Université de Lyon; Inserm U1111; Ecole Normale Supérieure de Lyon; Université Lyon 1, CNRS, UMR5308; Lyon, France
| | - Yves Gillet
- Centre International de Recherche en Infectiologie, Université de Lyon; Inserm U1111; Ecole Normale Supérieure de Lyon; Université Lyon 1, CNRS, UMR5308; Lyon, France.,Centre National de Référence des Staphylocoques, Institut des Agents Infectieux, Hospices Civils de Lyon, France
| | - Pascal Leblanc
- NeuroMyoGene Institute, Université de Lyon, CNRS UMR5310, INSERM U1217, France
| | - Andreas Itzek
- Helmholtz-Zentrum für Infektionsforschung GmbH, Braunschweig, Germany
| | - Anne Tristan
- Centre International de Recherche en Infectiologie, Université de Lyon; Inserm U1111; Ecole Normale Supérieure de Lyon; Université Lyon 1, CNRS, UMR5308; Lyon, France.,Centre National de Référence des Staphylocoques, Institut des Agents Infectieux, Hospices Civils de Lyon, France
| | - Michèle Bes
- Centre International de Recherche en Infectiologie, Université de Lyon; Inserm U1111; Ecole Normale Supérieure de Lyon; Université Lyon 1, CNRS, UMR5308; Lyon, France.,Centre National de Référence des Staphylocoques, Institut des Agents Infectieux, Hospices Civils de Lyon, France
| | - Stephanie Duguez
- Northern Ireland Center for Stratified Medicine, Biomedical Sciences Research Institute, Londonderry, United Kingdom
| | - Karen Moreau
- Centre International de Recherche en Infectiologie, Université de Lyon; Inserm U1111; Ecole Normale Supérieure de Lyon; Université Lyon 1, CNRS, UMR5308; Lyon, France
| | - Binh An Diep
- Division of HIV, Infectious Diseases, and Global Medicine, Department of Medicine, University of California, San Francisco
| | - Anna Norrby-Teglund
- Center for Infectious Medicine, Karolinska Institutet, Karolinska University Hospital, Huddinge, Sweden
| | - Thomas Henry
- Centre International de Recherche en Infectiologie, Université de Lyon; Inserm U1111; Ecole Normale Supérieure de Lyon; Université Lyon 1, CNRS, UMR5308; Lyon, France
| | - François Vandenesch
- Centre International de Recherche en Infectiologie, Université de Lyon; Inserm U1111; Ecole Normale Supérieure de Lyon; Université Lyon 1, CNRS, UMR5308; Lyon, France.,Centre National de Référence des Staphylocoques, Institut des Agents Infectieux, Hospices Civils de Lyon, France
| | | |
Collapse
|
38
|
Interaction of Brucella abortus with Osteoclasts: a Step toward Understanding Osteoarticular Brucellosis and Vaccine Safety. Infect Immun 2020; 88:IAI.00822-19. [PMID: 31932325 DOI: 10.1128/iai.00822-19] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2019] [Accepted: 01/02/2020] [Indexed: 02/07/2023] Open
Abstract
Osteoarticular disease is a frequent complication of human brucellosis. Vaccination remains a critical component of brucellosis control, but there are currently no vaccines for use in humans, and no in vitro models for assessing the safety of candidate vaccines in reference to the development of bone lesions currently exist. While the effect of Brucella infection on osteoblasts has been extensively evaluated, little is known about the consequences of osteoclast infection. Murine bone marrow-derived macrophages were derived into mature osteoclasts and infected with B. abortus 2308, the vaccine strain S19, and attenuated mutants S19vjbR and B. abortus ΔvirB2 While B. abortus 2308 and S19 replicated inside mature osteoclasts, the attenuated mutants were progressively killed, behavior that mimics infection kinetics in macrophages. Interestingly, B. abortus 2308 impaired the growth of osteoclasts without reducing resorptive activity, while osteoclasts infected with B. abortus S19 and S19vjbR were significantly larger and exhibited enhanced resorption. None of the Brucella strains induced apoptosis or stimulated nitric oxide or lactose dehydrogenase production in mature osteoclasts. Finally, infection of macrophages or osteoclast precursors with B. abortus 2308 resulted in generation of smaller osteoclasts with decreased resorptive activity. Overall, Brucella exhibits similar growth characteristics in mature osteoclasts compared to the primary target cell, the macrophage, but is able to impair the maturation and alter the resorptive capacity of these cells. These results suggest that osteoclasts play an important role in osteoarticular brucellosis and could serve as a useful in vitro model for both analyzing host-pathogen interactions and assessing vaccine safety.
Collapse
|
39
|
Hofstee MI, Muthukrishnan G, Atkins GJ, Riool M, Thompson K, Morgenstern M, Stoddart MJ, Richards RG, Zaat SAJ, Moriarty TF. Current Concepts of Osteomyelitis: From Pathologic Mechanisms to Advanced Research Methods. THE AMERICAN JOURNAL OF PATHOLOGY 2020; 190:1151-1163. [PMID: 32194053 DOI: 10.1016/j.ajpath.2020.02.007] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Revised: 02/18/2020] [Accepted: 02/27/2020] [Indexed: 01/18/2023]
Abstract
Osteomyelitis is an inflammation of the bone and bone marrow that is most commonly caused by a Staphylococcus aureus infection. Much of our understanding of the underlying pathophysiology of osteomyelitis, from the perspective of both host and pathogen, has been revised in recent years, with notable discoveries including the role played by osteocytes in the recruitment of immune cells, the invasion and persistence of S. aureus in submicron channels of cortical bone, and the diagnostic role of polymorphonuclear cells in implant-associated osteomyelitis. Advanced in vitro cell culture models, such as ex vivo culture models or organoids, have also been developed over the past decade, and have become widespread in many fields, including infectious diseases. These models better mimic the in vivo environment, allow the use of human cells, and can reduce our reliance on animals in osteomyelitis research. In this review, we provide an overview of the main pathologic concepts in osteomyelitis, with a focus on the new discoveries in recent years. Furthermore, we outline the value of modern in vitro cell culture techniques, with a focus on their current application to infectious diseases and osteomyelitis in particular.
Collapse
Affiliation(s)
- Marloes I Hofstee
- AO Research Institute Davos, Davos, Switzerland; Department of Medical Microbiology, Amsterdam UMC, University of Amsterdam, Amsterdam Infection and Immunity Institute, Amsterdam, the Netherlands
| | - Gowrishankar Muthukrishnan
- Center for Musculoskeletal Research and Department of Orthopaedics, University of Rochester Medical Center, Rochester, New York
| | - Gerald J Atkins
- Centre for Orthopaedic and Trauma Research, University of Adelaide, Adelaide, South Australia, Australia
| | - Martijn Riool
- Department of Medical Microbiology, Amsterdam UMC, University of Amsterdam, Amsterdam Infection and Immunity Institute, Amsterdam, the Netherlands
| | | | - Mario Morgenstern
- Department of Orthopedic Surgery and Traumatology, University Hospital Basel, Basel, Switzerland
| | | | | | - Sebastian A J Zaat
- Department of Medical Microbiology, Amsterdam UMC, University of Amsterdam, Amsterdam Infection and Immunity Institute, Amsterdam, the Netherlands
| | | |
Collapse
|
40
|
Lüthje FL, Jensen LK, Jensen HE, Skovgaard K. The inflammatory response to bone infection - a review based on animal models and human patients. APMIS 2020; 128:275-286. [PMID: 31976582 DOI: 10.1111/apm.13027] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2019] [Accepted: 01/14/2020] [Indexed: 12/17/2022]
Abstract
Bone infections are difficult to diagnose and treat, especially when a prosthetic joint replacement or implant is involved. Bone loss is a major complication of osteomyelitis, but the mechanism behind has mainly been investigated in cell cultures and has not been confirmed in human settings. Inflammation is important in initiating an appropriate immune response to invading pathogens. However, many of the signaling molecules used by the immune system can also modulate bone remodeling and contribute to bone resorption during osteomyelitis. Our current knowledge of the inflammatory response relies heavily on animal models as research based on human samples is scarce. Staphylococcus aureus is one of the most common causes of bone infections and is the pathogen of choice in animal models. The regulation of inflammatory genes during prosthetic joint infections and implant-associated osteomyelitis has only been studied in rodent models. It is important to consider the validity of an animal model when results are extrapolated to humans, and both bone composition and the immune system of pigs has been shown to be more similar to humans, than to rodents. Here in vivo studies on the inflammatory response to prosthetic joint infections and implant-associated osteomyelitis are reviewed.
Collapse
Affiliation(s)
- Freja Lea Lüthje
- Department of Veterinary and Animal Science, University of Copenhagen, Frederiksberg C, Denmark.,Department of Biotechnology and Biomedicine, Technical University of Denmark, Lyngby, Denmark
| | - Louise Kruse Jensen
- Department of Veterinary and Animal Science, University of Copenhagen, Frederiksberg C, Denmark
| | - Henrik Elvang Jensen
- Department of Veterinary and Animal Science, University of Copenhagen, Frederiksberg C, Denmark
| | - Kerstin Skovgaard
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Lyngby, Denmark
| |
Collapse
|
41
|
Yu K, Song L, Kang HP, Kwon HK, Back J, Lee FY. Recalcitrant methicillin-resistant Staphylococcus aureus infection of bone cells: Intracellular penetration and control strategies. Bone Joint Res 2020; 9:49-59. [PMID: 32435455 PMCID: PMC7229311 DOI: 10.1302/2046-3758.92.bjr-2019-0131.r1] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/02/2022] Open
Abstract
AIMS To characterize the intracellular penetration of osteoblasts and osteoclasts by methicillin-resistant Staphylococcus aureus (MRSA) and the antibiotic and detergent susceptibility of MRSA in bone. METHODS Time-lapse confocal microscopy was used to analyze the interaction of MRSA strain USA300 with primary murine osteoblasts and osteoclasts. The effects of early and delayed antibiotic treatments on intracellular and extracellular bacterial colony formation and cell death were quantified. We tested the effects of cefazolin, gentamicin, vancomycin, tetracycline, rifampicin, and ampicillin, as well as agents used in surgical preparation and irrigation. RESULTS MRSA infiltrated bone-resident cells within 15 to 30 minutes. Penetration was most effectively prevented with early (i.e. 30 minutes) antibiotic administration. The combined administration of rifampicin with other antibiotics potentiated their protective effects against MRSA-induced cytotoxicity and most significantly reduced extracellular bacterial bioburden. Gentamicin-containing compounds were most effective in reducing intracellular MRSA bioburden. Of the surgical preparation agents evaluated, betadine reduced in vitro MRSA growth to the greatest extent. CONCLUSION The standard of care for open fractures involves debridement and antibiotics within the first six hours of injury but does not account for the window in which bacteria penetrate cells. Antibiotics must be administered as early as possible after injury or prior to incision to prevent intracellular infestation. Rifampicin can potentiate the capacity of antibiotic regimens to reduce MRSA-induced cytotoxicity.Cite this article: Bone Joint Res. 2020;9(2):49-59.
Collapse
Affiliation(s)
- Kristin Yu
- Department of Orthopaedics and Rehabilitation, Yale University, New Haven, Connecticut, USA
| | - Lee Song
- Department of Orthopaedics, Columbia University, New York, New York, USA
| | - Hyunwoo Paco Kang
- Department of Orthopedic Surgery, University of Southern California, Los Angeles, California, USA
| | - Hyuk-Kwon Kwon
- Department of Orthopaedics and Rehabilitation, Yale University, New Haven, Connecticut, USA
| | - Jungho Back
- Department of Orthopaedics and Rehabilitation, Yale University, New Haven, Connecticut, USA
| | - Francis Y Lee
- Department of Orthopaedics and Rehabilitation, Yale University, New Haven, Connecticut, USA
| |
Collapse
|
42
|
Schröder ML, Angrisani N, Fadeeva E, Hegermann J, Reifenrath J. Laser-structured spike surface shows great bone integrative properties despite infection in vivo. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2019; 109:110573. [PMID: 32228937 DOI: 10.1016/j.msec.2019.110573] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/23/2019] [Revised: 11/25/2019] [Accepted: 12/18/2019] [Indexed: 12/18/2022]
Abstract
Implant associated infections can result in devastating consequences for patients. One major cause is the formation of bacterial biofilms, which result in increased resistance against antimicrobial therapeutics. A reduction of implant associated infections can be achieved by functionalization of implant surfaces. The generation of three dimensional surface structures by femtosecond laser ablation is one method to fabricate bacterial repellent large scaled surfaces without altering the material chemical composition. The challenge is to reduce bacterial growth while improving cellular ongrowth. For this purpose, spike structures were created as small as possible by used fabrication method on cubic Ti90/Al6/V4-rods and their effectiveness against bacterial colonization was compared to unstructured Ti90/Al6/V4-rods. Rods were implanted in the rat tibia and infected intraoperatively with 103 CFU of Staphylococcus aureus. Besides clinical behaviour and lameness, the vital bacterial biomass, morphological appearance and the volume of eukaryotic cells were determined on the implant surface after 21 days. Bone alterations were examined by radiological and histological techniques. Unexpectedly, the laser-structured implants did not show a lower bacterial load on the implant surface and less severe infection related bone and tissue alterations compared to the group without structuring. Simultaneously, a better bony integration and a higher cellular colonization with eukaryotic cells was detected on the laser-structured implants. These findings don't support the previous in vitro results. Nevertheless, the strong integration into the bone is a powerful argument for further surface modifications focussing on the improvement of the antibacterial effect. Additionally, our results underline the need for in vivo testing of new materials prior to clinical use.
Collapse
Affiliation(s)
- M-L Schröder
- Hannover Medical School, Clinic for Orthopedic Surgery, Anna-von-Borries Str. 1-9, 30625 Hannover, Germany; University of Veterinary Medicine Hannover, Foundation, Small Animal Clinic, Bünteweg 9, 30559 Hannover, Germany
| | - N Angrisani
- Hannover Medical School, Clinic for Orthopedic Surgery, Anna-von-Borries Str. 1-9, 30625 Hannover, Germany
| | - E Fadeeva
- Laser Zentrum Hannover e.V., Hollerithallee 8, 30419 Hannover, Germany
| | - J Hegermann
- Hannover Medical School, Institute of Functional an Applied Anatomy, Research Core Unit Electron Microscopy, Carl-Neuberg-Str. 1, 30625 Hannover, Germany
| | - J Reifenrath
- Hannover Medical School, Clinic for Orthopedic Surgery, Anna-von-Borries Str. 1-9, 30625 Hannover, Germany.
| |
Collapse
|
43
|
Li H, Zhang S, Huo S, Tang H, Nie B, Qu X, Yue B. Effects of staphylococcal infection and aseptic inflammation on bone mass and biomechanical properties in a rabbit model. J Orthop Translat 2019; 21:66-72. [PMID: 32099806 PMCID: PMC7029375 DOI: 10.1016/j.jot.2019.11.006] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Revised: 10/28/2019] [Accepted: 11/25/2019] [Indexed: 01/18/2023] Open
Abstract
Background/objective Orthopaedic implants are important devices aimed at relieving pain and improving mobility. Staphylococcal infection and aseptic loosening are two common events associated with inflammatory osteolysis that lead to implant failures. Bone mass and biomechanical properties are important indicators that could influence patient outcomes after revision surgery. However, the dynamics of bacterial infections and their influence on bone mass and biomechanical properties remain unclear. Hence, in this study, we developed rabbit aseptic inflammation and staphylococcal infection models to determine the effects of coagulase-positive and coagulase-negative bacterial infection, as well as aseptic inflammation, on the mass and biomechanical properties of the bone. Methods Sixty New Zealand white rabbits were randomly assigned to 6 groups, and each group had 10 rabbits. The medullary cavities in rabbits of each group were injected with phosphate-buffered saline (100 μL), titanium (Ti)-wear particles (300 μg/100 μL), a low concentration of Staphylococcus epidermidis (105/100 μL), a high concentration of S. epidermidis (108/100 μL), a low concentration of Staphylococcus aureus (105/100 μL), and a high concentration of S. aureus (108/100 μL), respectively. At four and eight weeks after surgery, the rabbits were sacrificed, and the tibias on the surgical side were analysed via histopathology, microcomputed tomography, and nanoindentation testing. Results Histopathological analysis demonstrated that inflammatory responses and bacterial loads caused by high concentrations of staphylococcal infections, particularly coagulase-positive staphylococci, are more detrimental than low concentrations of bacterial infection and Ti-wear particles. Meanwhile, microcomputed tomography and nanoindentation testing showed that high concentrations of S. aureus caused the highest loss in bone mass and most biomechanical function impairment in rabbits experiencing aseptic inflammation and staphylococcal infections. Conclusions Inflammatory osteolysis caused by a high concentration of coagulase-positive staphylococci is significantly associated with low bone mass and impaired biomechanical properties. The translational potential of this article It is necessary to obtain an overall assessment of the bone mass and biomechanical properties before revision surgery, especially when S. aureus infection is involved. In addition, a better understanding of these two parameters might help develop a reasonable treatment regimen and reduce the risk of adverse events after revision surgery.
Collapse
Affiliation(s)
- Hui Li
- Department of Bone and Joint Surgery, Renji Hospital, School of Medicine, Shanghai Jiaotong University, China
| | - Shutao Zhang
- Department of Bone and Joint Surgery, Renji Hospital, School of Medicine, Shanghai Jiaotong University, China
| | - Shicheng Huo
- Department of Bone and Joint Surgery, Renji Hospital, School of Medicine, Shanghai Jiaotong University, China
| | - Haozheng Tang
- Department of Bone and Joint Surgery, Renji Hospital, School of Medicine, Shanghai Jiaotong University, China
| | - Bin'en Nie
- Department of Bone and Joint Surgery, Renji Hospital, School of Medicine, Shanghai Jiaotong University, China
| | - Xinhua Qu
- Department of Bone and Joint Surgery, Renji Hospital, School of Medicine, Shanghai Jiaotong University, China
| | - Bing Yue
- Department of Bone and Joint Surgery, Renji Hospital, School of Medicine, Shanghai Jiaotong University, China
| |
Collapse
|
44
|
Asensio G, Vázquez-Lasa B, Rojo L. Achievements in the Topographic Design of Commercial Titanium Dental Implants: Towards Anti-Peri-Implantitis Surfaces. J Clin Med 2019; 8:E1982. [PMID: 31739615 PMCID: PMC6912779 DOI: 10.3390/jcm8111982] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Revised: 11/04/2019] [Accepted: 11/11/2019] [Indexed: 12/12/2022] Open
Abstract
Titanium and its alloys constitute the gold standard materials for oral implantology in which their performance is mainly conditioned by their osseointegration capacity in the host's bone. We aim to provide an overview of the advances in surface modification of commercial dental implants analyzing and comparing the osseointegration capacity and the clinical outcome exhibited by different surfaces. Besides, the development of peri-implantitis constitutes one of the most common causes of implant loss due to bacteria colonization. Thus, a synergic response from industry and materials scientists is needed to provide reliable technical and commercial solutions to this issue. The second part of the review focuses on an update of the recent findings toward the development of new materials with osteogenic and antibacterial capacity that are most likely to be marketed, and their correlation with implant geometry, biomechanical behavior, biomaterials features, and clinical outcomes.
Collapse
Affiliation(s)
- Gerardo Asensio
- Instituto de Ciencia y Tecnología de Polímeros, Consejo Superior de Investigaciones Científicas, CSIC, 28006 Madrid, Spain; (G.A.); (B.V.-L.)
| | - Blanca Vázquez-Lasa
- Instituto de Ciencia y Tecnología de Polímeros, Consejo Superior de Investigaciones Científicas, CSIC, 28006 Madrid, Spain; (G.A.); (B.V.-L.)
- Consorcio Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina, CIBER-BBN, 28029 Madrid, Spain
| | - Luis Rojo
- Instituto de Ciencia y Tecnología de Polímeros, Consejo Superior de Investigaciones Científicas, CSIC, 28006 Madrid, Spain; (G.A.); (B.V.-L.)
- Consorcio Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina, CIBER-BBN, 28029 Madrid, Spain
| |
Collapse
|
45
|
Abstract
The inflammation of bone tissue is called osteomyelitis, and most cases are caused by an infection with the bacterium Staphylococcus aureus. To date, the bone-building cells, osteoblasts, have been implicated in the progression of these infections, but not much is known about how the bone-resorbing cells, osteoclasts, participate. In this study, we show that S. aureus can infect osteoclasts and proliferate inside these cells, whereas bone-residing macrophages, immune cells related to osteoclasts, destroy the bacteria. These findings elucidate a unique role for osteoclasts to harbor bacteria during infection, providing a possible mechanism by which bacteria could evade destruction by the immune system. Osteomyelitis (OM), or inflammation of bone tissue, occurs most frequently as a result of bacterial infection and severely perturbs bone structure. OM is predominantly caused by Staphylococcus aureus, and even with proper treatment, OM has a high rate of recurrence and chronicity. While S. aureus has been shown to infect osteoblasts, it remains unclear whether osteoclasts (OCs) are also a target of intracellular infection. Here, we demonstrate the ability of S. aureus to intracellularly infect and divide within OCs. OCs were differentiated from bone marrow macrophages (BMMs) by exposure to receptor activator of nuclear factor kappa-B ligand (RANKL). By utilizing an intracellular survival assay and flow cytometry, we found that at 18 h postinfection the intracellular burden of S. aureus increased dramatically in cells with at least 2 days of RANKL exposure, while the bacterial burden decreased in BMMs. To further explore the signals downstream of RANKL, we manipulated factors controlling OC differentiation, NFATc1 and alternative NF-κB, and found that intracellular bacterial growth correlates with NFATc1 levels in RANKL-treated cells. Confocal and time-lapse microscopy in mature OCs showed a range of intracellular infection that correlated inversely with S. aureus-phagolysosome colocalization. The propensity of OCs to become infected, paired with their diminished bactericidal capacity compared to BMMs, could promote OM progression by allowing S. aureus to evade initial immune regulation and proliferate at the periphery of lesions where OCs are most abundant.
Collapse
|
46
|
Raynaud-Messina B, Verollet C, Maridonneau-Parini I. The osteoclast, a target cell for microorganisms. Bone 2019; 127:315-323. [PMID: 31233933 DOI: 10.1016/j.bone.2019.06.023] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Revised: 06/11/2019] [Accepted: 06/21/2019] [Indexed: 02/02/2023]
Abstract
Bone is a highly adaptive tissue with regenerative properties that is subject to numerous diseases. Infection is one of the causes of altered bone homeostasis. Bone infection happens subsequently to bone surgery or to systemic spreading of microorganisms. In addition to osteoblasts, osteoclasts (OCs) also constitute cell targets for pathogens. OCs are multinucleated cells that have the exclusive ability to resorb bone mineral tissue. However, the OC is much more than a bone eater. Beyond its role in the control of bone turnover, the OC is an immune cell that produces and senses inflammatory cytokines, ingests microorganisms and presents antigens. Today, increasing evidence shows that several pathogens use OC as a host cell to grow, generating debilitating bone defects. In this review, we exhaustively inventory the bacteria and viruses that infect OC and report the present knowledge in this topic. We point out that most of the microorganisms enhance the bone resorption activity of OC. We notice that pathogen interactions with the OC require further investigation, in particular to validate the OC as a host cell in vivo and to identify the cellular mechanisms involved in altered bone resorption. Thus, we conclude that the OC is a new cell target for pathogens; this new research area paves the way for new therapeutic strategies in the infections causing bone defects.
Collapse
Affiliation(s)
- Brigitte Raynaud-Messina
- Institut de Pharmacologie et Biologie Structurale, IPBS, Université de Toulouse, CNRS, UPS, Toulouse, France; International Associated Laboratory (LIA) CNRS "IM-TB/HIV" (1167), Toulouse, France; International Associated Laboratory (LIA) CNRS "IM-TB/HIV" (1167), Buenos Aires, Argentina
| | - Christel Verollet
- Institut de Pharmacologie et Biologie Structurale, IPBS, Université de Toulouse, CNRS, UPS, Toulouse, France; International Associated Laboratory (LIA) CNRS "IM-TB/HIV" (1167), Toulouse, France; International Associated Laboratory (LIA) CNRS "IM-TB/HIV" (1167), Buenos Aires, Argentina
| | - Isabelle Maridonneau-Parini
- Institut de Pharmacologie et Biologie Structurale, IPBS, Université de Toulouse, CNRS, UPS, Toulouse, France; International Associated Laboratory (LIA) CNRS "IM-TB/HIV" (1167), Toulouse, France; International Associated Laboratory (LIA) CNRS "IM-TB/HIV" (1167), Buenos Aires, Argentina.
| |
Collapse
|
47
|
Tomecka MJ, Ethiraj LP, Sánchez LM, Roehl HH, Carney TJ. Clinical pathologies of bone fracture modelled in zebrafish. Dis Model Mech 2019; 12:dmm.037630. [PMID: 31383797 PMCID: PMC6765199 DOI: 10.1242/dmm.037630] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2018] [Accepted: 07/24/2019] [Indexed: 01/24/2023] Open
Abstract
Reduced bone quality or mineral density predict susceptibility to fracture and also attenuate subsequent repair. Bone regrowth is also compromised by bacterial infection, which exacerbates fracture site inflammation. Because of the cellular complexity of fracture repair, as well as genetic and environmental influences, there is a need for models that permit visualisation of the fracture repair process under clinically relevant conditions. To characterise the process of fracture repair in zebrafish, we employed a crush fracture of fin rays, coupled with histological and transgenic labelling of cellular responses; the results demonstrate a strong similarity to the phased response in humans. We applied our analysis to a zebrafish model of osteogenesis imperfecta (OI), which shows reduced bone quality, spontaneous fractures and propensity for non-unions. We found deficiencies in the formation of a bone callus during fracture repair in our OI model and showed that clinically employed antiresorptive bisphosphonates can reduce spontaneous fractures in OI fish and also measurably reduce fracture callus remodelling in wild-type fish. The csf1ra mutant, which has reduced osteoclast numbers, also showed reduced callus remodelling. Exposure to excessive bisphosphonate, however, disrupted callus repair. Intriguingly, neutrophils initially colonised the fracture site, but were later completely excluded. However, when fractures were infected with Staphylococcus aureus, neutrophils were retained and compromised repair. This work elevates the zebrafish bone fracture model and indicates its utility in assessing conditions of relevance to an orthopaedic setting with medium throughput. This article has an associated First Person interview with the first author of the paper. Summary: The effect of osteogenesis imperfecta, bisphosphonate treatment and bacterial infection on phases of bone fracture repair are determined using a zebrafish fracture model.
Collapse
Affiliation(s)
- Monika J Tomecka
- Institute of Molecular and Cell Biology (IMCB), A*STAR (Agency for Science, Technology and Research), 61 Biopolis Drive, Proteos, 138673, Singapore.,Department of Biomedical Science, Firth Court, Western Bank, The University of Sheffield, Sheffield, S10 2TN, United Kingdom
| | - Lalith P Ethiraj
- Lee Kong Chian School of Medicine, Experimental Medicine Building, Yunnan Garden Campus, 59 Nanyang Drive, Nanyang Technological University 636921, Singapore
| | - Luis M Sánchez
- Department of Biomedical Science, Firth Court, Western Bank, The University of Sheffield, Sheffield, S10 2TN, United Kingdom
| | - Henry H Roehl
- Department of Biomedical Science, Firth Court, Western Bank, The University of Sheffield, Sheffield, S10 2TN, United Kingdom
| | - Tom J Carney
- Institute of Molecular and Cell Biology (IMCB), A*STAR (Agency for Science, Technology and Research), 61 Biopolis Drive, Proteos, 138673, Singapore .,Lee Kong Chian School of Medicine, Experimental Medicine Building, Yunnan Garden Campus, 59 Nanyang Drive, Nanyang Technological University 636921, Singapore
| |
Collapse
|
48
|
Josse J, Valour F, Maali Y, Diot A, Batailler C, Ferry T, Laurent F. Interaction Between Staphylococcal Biofilm and Bone: How Does the Presence of Biofilm Promote Prosthesis Loosening? Front Microbiol 2019; 10:1602. [PMID: 31379772 PMCID: PMC6653651 DOI: 10.3389/fmicb.2019.01602] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Accepted: 06/26/2019] [Indexed: 12/19/2022] Open
Abstract
With the aging of population, the number of indications for total joint replacement is continuously increasing. However, prosthesis loosening can happen and is related to two major mechanisms: (1) aseptic loosening due to prosthesis micromotion and/or corrosion and release of wear particles from the different components of the implanted material and (2) septic loosening due to chronic prosthetic joint infection (PJI). The “aseptic” character of prosthesis loosening has been challenged over the years, especially considering that bacteria can persist in biofilms and be overlooked during diagnosis. Histological studies on periprosthetic tissue samples reported that macrophages are the principle cells associated with aseptic loosening due to wear debris. They produce cytokines and favor an inflammatory environment that induces formation and activation of osteoclasts, leading to bone resorption and periprosthetic osteolysis. In PJIs, the presence of infiltrates of polymorphonuclear neutrophils is a major criterion for histological diagnosis. Neutrophils are colocalized with osteoclasts and zones of osteolysis. A similar inflammatory environment also develops, leading to bone resorption through osteoclasts. Staphylococcus aureus, Staphylococcus epidermidis, and Staphylococcus lugdunensis are the main staphylococci observed in PJIs. They share the common feature to form biofilm. For S. aureus and S. epidermidis, the interaction between biofilm and immunes cells (macrophages and polymorphonuclear neutrophils) differs regarding the species. Indeed, the composition of extracellular matrix of biofilm seems to impact the interaction with immune cells. Recent papers also reported the major role of myeloid-derived suppressor cells in biofilm-associated PJIs with S. aureus. These cells prevent lymphocyte infiltration and facilitate biofilm persistence. Moreover, the role of T lymphocytes is still unclear and potentially underestimates. In this review, after introducing the cellular mechanism of aseptic and septic loosening, we will focus on the interrelationships between staphylococcal biofilm, immune cells, and bone cells.
Collapse
Affiliation(s)
- Jérôme Josse
- CIRI - Centre International de Recherche en Infectiologie, Inserm U1111, CNRS UMR5308, ENS Lyon, Université Claude Bernard Lyon 1, Lyon, France.,Université Claude Bernard Lyon 1, Université de Lyon, Lyon, France.,Centre Interrégional de Référence des Infections Ostéo-articulaires Complexes (CRIOAc Lyon), Hospices Civils de Lyon, Lyon, France
| | - Florent Valour
- CIRI - Centre International de Recherche en Infectiologie, Inserm U1111, CNRS UMR5308, ENS Lyon, Université Claude Bernard Lyon 1, Lyon, France.,Université Claude Bernard Lyon 1, Université de Lyon, Lyon, France.,Centre Interrégional de Référence des Infections Ostéo-articulaires Complexes (CRIOAc Lyon), Hospices Civils de Lyon, Lyon, France.,Service de Chirurgie Orthopédique, Hôpital de la Croix-Rousse, Hospices Civils de Lyon, Lyon, France
| | - Yousef Maali
- CIRI - Centre International de Recherche en Infectiologie, Inserm U1111, CNRS UMR5308, ENS Lyon, Université Claude Bernard Lyon 1, Lyon, France.,Université Claude Bernard Lyon 1, Université de Lyon, Lyon, France
| | - Alan Diot
- CIRI - Centre International de Recherche en Infectiologie, Inserm U1111, CNRS UMR5308, ENS Lyon, Université Claude Bernard Lyon 1, Lyon, France.,Université Claude Bernard Lyon 1, Université de Lyon, Lyon, France
| | - Cécile Batailler
- Université Claude Bernard Lyon 1, Université de Lyon, Lyon, France.,Centre Interrégional de Référence des Infections Ostéo-articulaires Complexes (CRIOAc Lyon), Hospices Civils de Lyon, Lyon, France.,Service de Maladies Infectieuses, Hôpital de la Croix-Rousse, Hospices Civils de Lyon, Lyon, France
| | - Tristan Ferry
- CIRI - Centre International de Recherche en Infectiologie, Inserm U1111, CNRS UMR5308, ENS Lyon, Université Claude Bernard Lyon 1, Lyon, France.,Université Claude Bernard Lyon 1, Université de Lyon, Lyon, France.,Centre Interrégional de Référence des Infections Ostéo-articulaires Complexes (CRIOAc Lyon), Hospices Civils de Lyon, Lyon, France.,Service de Chirurgie Orthopédique, Hôpital de la Croix-Rousse, Hospices Civils de Lyon, Lyon, France
| | - Frédéric Laurent
- CIRI - Centre International de Recherche en Infectiologie, Inserm U1111, CNRS UMR5308, ENS Lyon, Université Claude Bernard Lyon 1, Lyon, France.,Université Claude Bernard Lyon 1, Université de Lyon, Lyon, France.,Centre Interrégional de Référence des Infections Ostéo-articulaires Complexes (CRIOAc Lyon), Hospices Civils de Lyon, Lyon, France.,Laboratoire de Bactériologie, Institut des Agents Infectieux, Hôpital de la Croix-Rousse, Hospices Civils de Lyon, Lyon, France
| |
Collapse
|
49
|
Putnam NE, Fulbright LE, Curry JM, Ford CA, Petronglo JR, Hendrix AS, Cassat JE. MyD88 and IL-1R signaling drive antibacterial immunity and osteoclast-driven bone loss during Staphylococcus aureus osteomyelitis. PLoS Pathog 2019; 15:e1007744. [PMID: 30978245 PMCID: PMC6481883 DOI: 10.1371/journal.ppat.1007744] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Revised: 04/24/2019] [Accepted: 04/01/2019] [Indexed: 01/18/2023] Open
Abstract
Staphylococcus aureus is able to infect virtually all organ systems and is a frequently isolated etiologic agent of osteomyelitis, a common and debilitating invasive infection of bone. Treatment of osteomyelitis requires invasive surgical procedures and prolonged antibiotic therapy, yet is frequently unsuccessful due to extensive pathogen-induced bone damage that can limit antibiotic penetration and immune cell influx to the infectious focus. We previously established that S. aureus triggers profound alterations in bone remodeling in a murine model of osteomyelitis, in part through the production of osteolytic toxins. However, staphylococcal strains lacking osteolytic toxins still incite significant bone destruction, suggesting that host immune responses are also major drivers of pathologic bone remodeling during osteomyelitis. The objective of this study was to identify host immune pathways that contribute to antibacterial immunity during S. aureus osteomyelitis, and to define how these immune responses alter bone homeostasis and contribute to bone destruction. We specifically focused on the interleukin-1 receptor (IL-1R) and downstream adapter protein MyD88 given the prominent role of this signaling pathway in both antibacterial immunity and osteo-immunologic crosstalk. We discovered that while IL-1R signaling is necessary for local control of bacterial replication during osteomyelitis, it also contributes to bone loss during infection. Mechanistically, we demonstrate that S. aureus enhances osteoclastogenesis of myeloid precursors in vitro, and increases the abundance of osteoclasts residing on bone surfaces in vivo. This enhanced osteoclast abundance translates to trabecular bone loss, and is dependent on intact IL-1R signaling. Collectively, these data define IL-1R signaling as a critical component of the host response to S. aureus osteomyelitis, but also demonstrate that IL-1R-dependent immune responses trigger collateral bone damage through activation of osteoclast-mediated bone resorption.
Collapse
Affiliation(s)
- Nicole E. Putnam
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
| | - Laura E. Fulbright
- Department of Pediatrics, Division of Pediatric Infectious Diseases, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
| | - Jacob M. Curry
- Department of Pediatrics, Division of Pediatric Infectious Diseases, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
| | - Caleb A. Ford
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee, United States of America
| | - Jenna R. Petronglo
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
| | - Andrew S. Hendrix
- Department of Pediatrics, Division of Pediatric Infectious Diseases, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
| | - James E. Cassat
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
- Department of Pediatrics, Division of Pediatric Infectious Diseases, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee, United States of America
- Vanderbilt Institute for Infection, Immunology and Inflammation (VI4), Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
- Vanderbilt Center for Bone Biology, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
| |
Collapse
|
50
|
Wang L, Shang X, Hao Y, Wan G, Dong L, Huang D, Yang X, Sun J, Wang Q, Zha G, Yang X. Bi-functional titanium-polydopamine-zinc coatings for infection inhibition and enhanced osseointegration. RSC Adv 2019; 9:2892-2905. [PMID: 35518964 PMCID: PMC9059936 DOI: 10.1039/c8ra09112a] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2018] [Accepted: 01/09/2019] [Indexed: 01/12/2023] Open
Abstract
The ideal orthopedic implant coating is expected to both inhibit microbial infection and promote osseointegration. In this study, Zn ions were immobilized on a Ti substrate via a polydopamine (PDA) chemical surface modification to prepare Ti-PDA-Zn coatings. Scanning electron microscopy (SEM), atomic force microscopy (AFM), energy-dispersive X-ray spectroscope (EDS), X-ray photoelectron spectroscopy (XPS), contact analysis system, and inductively coupled plasma atomic emission spectrometry (ICP-AES) were used to analyze the morphology, composition, wettability, and zinc ions release of the coatings. The Ti-PDA-Zn coatings demonstrated excellent antibacterial activities in vitro against both Staphylococcus aureus and Escherichia coli. The coatings additionally displayed good biocompatibility, as confirmed by cytoskeletal observations and cell viability assays. Furthermore, the in vivo results confirmed the excellent antibacterial properties and improved osseointegration capability of the Ti-PDA-Zn coating in the presence of S. aureus. The present findings indicate that the Ti-PDA-Zn coatings prepared herein have potential application in orthopedic implantation. The ideal orthopedic implant coating is expected to both inhibit microbial infection and promote osseointegration.![]()
Collapse
|