1001
|
Li X, Bottini M, Zhang L, Zhang S, Chen J, Zhang T, Liu L, Rosato N, Ma X, Shi X, Wu Y, Guo W, Liang XJ. Core-Satellite Nanomedicines for in Vivo Real-Time Monitoring of Enzyme-Activatable Drug Release by Fluorescence and Photoacoustic Dual-Modal Imaging. ACS NANO 2019; 13:176-186. [PMID: 30592401 DOI: 10.1021/acsnano.8b05136] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/28/2023]
Abstract
It remains an unresolved challenge to achieve spatial and temporal monitoring of drug release from nanomedicines (NMs) in vivo, which is of crucial importance in disease treatment. To tackle this issue, we constructed core-satellite ICG/DOX@Gel-CuS NMs, which consist of gelatin (Gel) nanoparticles (NPs) with payloads of near-infrared fluorochrome indocyanine green (ICG) and chemo-drug doxorubicin (DOX) and surrounding CuS NPs. The fluorescence of ICG was initially shielded by satellite CuS NPs within the intact ICG/DOX@Gel-CuS NMs and increased in proportion to the amount of DOX released from NMs in response to enzyme-activated NMs degradation. For more comprehensive understanding of the drug-release profile, a theoretical model derived from computer simulation was employed to reconstruct the enzyme-activatable drug release of the ICG/DOX@Gel-CuS NMs, which demonstrated the underlying kinetics functional relationship between the released DOX amount and recovered ICG fluorescence intensity. The kinetics of drug release in vivo was assessed by administrating ICG/DOX@Gel-CuS NMs both locally and systemically into MDA-MB-231 tumor-bearing mice. Upon accumulation of ICG/DOX@Gel-CuS NMs in the tumor, overexpressed enzymes triggered the degradation of the gelatin scaffold as well as the release of DOX and ICG, which can be visually depicted with the ICG fluorescence signal increasing only in the tumor area by fluorescence imaging. Additionally, the photoacoustic signal from CuS NPs was independent from the physical status of ICG/DOX@Gel-CuS NMs and hence was utilized for real-time NMs tracking. Thus, by taking advantage of the core-satellite architecture and NMs degradability in tumor site, the DOX release profile of ICG/DOX@Gel-CuS NMs was monitored by fluorescence and photoacoustic dual-modal imaging in a real-time noninvasive manner.
Collapse
Affiliation(s)
- Xianlei Li
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience , National Center for Nanoscience and Technology of China , No. 11, First North Road , Zhongguancun, Beijing 100190 , P. R. China
- University of Chinese Academy of Sciences , Beijing 100049 , P. R. China
| | - Massimo Bottini
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience , National Center for Nanoscience and Technology of China , No. 11, First North Road , Zhongguancun, Beijing 100190 , P. R. China
- Department of Experimental Medicine and Surgery , University of Rome Tor Vergata , Via Montpellier 1 , 00133 Rome , Italy
| | - Luyao Zhang
- University of Chinese Academy of Sciences , Beijing 100049 , P. R. China
- LNM, Institute of Mechanics, Chinese Academy of Sciences , Beijing 100190 , P. R. China
| | - Shuai Zhang
- CAS Key Laboratory of Molecular Imaging , Institute of Automation, Chinese Academy of Sciences , Beijing 100190 , P. R. China
| | - Jing Chen
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience , National Center for Nanoscience and Technology of China , No. 11, First North Road , Zhongguancun, Beijing 100190 , P. R. China
| | - Tingbin Zhang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience , National Center for Nanoscience and Technology of China , No. 11, First North Road , Zhongguancun, Beijing 100190 , P. R. China
| | - Lu Liu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience , National Center for Nanoscience and Technology of China , No. 11, First North Road , Zhongguancun, Beijing 100190 , P. R. China
- University of Chinese Academy of Sciences , Beijing 100049 , P. R. China
| | - Nicola Rosato
- Department of Experimental Medicine and Surgery , University of Rome Tor Vergata , Via Montpellier 1 , 00133 Rome , Italy
| | - Xibo Ma
- University of Chinese Academy of Sciences , Beijing 100049 , P. R. China
- CAS Key Laboratory of Molecular Imaging , Institute of Automation, Chinese Academy of Sciences , Beijing 100190 , P. R. China
| | - Xinghua Shi
- University of Chinese Academy of Sciences , Beijing 100049 , P. R. China
- CAS Key Laboratory for Nanosystem and Hierarchy Fabrication, CAS Center for Excellence in Nanoscience , National Center for Nanoscience and Technology, Chinese Academy of Sciences , Beijing 100190 , P. R. China
| | - Yan Wu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience , National Center for Nanoscience and Technology of China , No. 11, First North Road , Zhongguancun, Beijing 100190 , P. R. China
- University of Chinese Academy of Sciences , Beijing 100049 , P. R. China
| | - Weisheng Guo
- Translational Medicine Center, State Key Laboratory of Respiratory Disease, The Second Affiliated Hospital , Guangzhou Medical University , Guangzhou 510260 , P. R. China
| | - Xing-Jie Liang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience , National Center for Nanoscience and Technology of China , No. 11, First North Road , Zhongguancun, Beijing 100190 , P. R. China
- University of Chinese Academy of Sciences , Beijing 100049 , P. R. China
| |
Collapse
|
1002
|
Multiscale Stem Cell Technologies for Osteonecrosis of the Femoral Head. Stem Cells Int 2019; 2019:8914569. [PMID: 30728843 PMCID: PMC6341242 DOI: 10.1155/2019/8914569] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2018] [Revised: 10/21/2018] [Accepted: 11/14/2018] [Indexed: 02/06/2023] Open
Abstract
The last couple of decades have seen brilliant progress in stem cell therapies, including native, genetically modified, and engineered stem cells, for osteonecrosis of the femoral head (ONFH). In vitro studies evaluate the effect of endogenous or exogenous factor or gene regulation on osteogenic phenotype maintenance and/or differentiation towards osteogenic lineage. The preclinical and clinical outcomes accelerate the clinical translation. Bone marrow mesenchymal stem cells and adipose-derived stem cells have demonstrated better effects in the treatment of femoral head necrosis. Various materials have been used widely in the ONFH treatment in both preclinical and clinical trials. In a word, in vivo and multiscale efforts are expected to overcome obstacles in the approaches for treating ONFH and provide clinical relevance and commercial strategies in the future. Therefore, we will discuss the above aspects in this paper and present our opinions.
Collapse
|
1003
|
Abstract
In physiologically young patients with displaced femoral neck fractures, surgical treatment is aimed at achieving fracture union while preserving native hip anatomy and biomechanics. The intracapsular environment, tenuous vascular supply, and unfavorable hip biomechanics contribute to the high complication rates seen after osteosynthesis of these fractures. Conventional fixation methods for osteosynthesis of femoral neck fractures include multiple cancellous screws, fixed-angle dynamic implants, and fixed-angle length-stable constructs. Despite several biomechanical and clinical studies evaluating various surgical options, the optimal fixation construct to allow healing and prevent nonunion of displaced femoral neck fractures is not known. This article will review the clinical data regarding conventional fixation constructs and describe the technique and rationale behind 2 novel alternative treatment options for these challenging fractures. The surgical technique and clinical examples for constructs involving multiple cannulated screws/Pauwels screw augmented with a fibular strut graft, as well as a novel fixed-angle locking plate with controlled dynamic compression, are presented.
Collapse
|
1004
|
Afewerki S, Sheikhi A, Kannan S, Ahadian S, Khademhosseini A. Gelatin-polysaccharide composite scaffolds for 3D cell culture and tissue engineering: Towards natural therapeutics. Bioeng Transl Med 2019; 4:96-115. [PMID: 30680322 PMCID: PMC6336672 DOI: 10.1002/btm2.10124] [Citation(s) in RCA: 234] [Impact Index Per Article: 39.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2018] [Revised: 11/23/2018] [Accepted: 11/26/2018] [Indexed: 12/12/2022] Open
Abstract
Gelatin is a promising material as scaffold with therapeutic and regenerative characteristics due to its chemical similarities to the extracellular matrix (ECM) in the native tissues, biocompatibility, biodegradability, low antigenicity, cost-effectiveness, abundance, and accessible functional groups that allow facile chemical modifications with other biomaterials or biomolecules. Despite the advantages of gelatin, poor mechanical properties, sensitivity to enzymatic degradation, high viscosity, and reduced solubility in concentrated aqueous media have limited its applications and encouraged the development of gelatin-based composite hydrogels. The drawbacks of gelatin may be surmounted by synergistically combining it with a wide range of polysaccharides. The addition of polysaccharides to gelatin is advantageous in mimicking the ECM, which largely contains proteoglycans or glycoproteins. Moreover, gelatin-polysaccharide biomaterials benefit from mechanical resilience, high stability, low thermal expansion, improved hydrophilicity, biocompatibility, antimicrobial and anti-inflammatory properties, and wound healing potential. Here, we discuss how combining gelatin and polysaccharides provides a promising approach for developing superior therapeutic biomaterials. We review gelatin-polysaccharides scaffolds and their applications in cell culture and tissue engineering, providing an outlook for the future of this family of biomaterials as advanced natural therapeutics.
Collapse
Affiliation(s)
- Samson Afewerki
- Biomaterials Innovation Research Center, Division of Biomedical Engineering, Dept. of MedicineBrigham and Women's Hospital, Harvard Medical SchoolCambridgeMA 02142
- Harvard‐MIT Division of Health Sciences and TechnologyMassachusetts Institute of TechnologyCambridgeMA 02139
| | - Amir Sheikhi
- Biomaterials Innovation Research Center, Division of Biomedical Engineering, Dept. of MedicineBrigham and Women's Hospital, Harvard Medical SchoolCambridgeMA 02142
- Harvard‐MIT Division of Health Sciences and TechnologyMassachusetts Institute of TechnologyCambridgeMA 02139
- Center for Minimally Invasive Therapeutics (C‐MIT)University of California‐Los AngelesLos AngelesCA 90095
- California NanoSystems Institute (CNSI)University of California‐Los AngelesLos AngelesCA 90095
- Dept. of BioengineeringUniversity of California‐Los AngelesLos AngelesCA 90095
| | - Soundarapandian Kannan
- Biomaterials Innovation Research Center, Division of Biomedical Engineering, Dept. of MedicineBrigham and Women's Hospital, Harvard Medical SchoolCambridgeMA 02142
- Harvard‐MIT Division of Health Sciences and TechnologyMassachusetts Institute of TechnologyCambridgeMA 02139
- Nanomedicine Division, Dept. of ZoologyPeriyar UniversitySalemTamil NaduIndia
| | - Samad Ahadian
- Center for Minimally Invasive Therapeutics (C‐MIT)University of California‐Los AngelesLos AngelesCA 90095
- California NanoSystems Institute (CNSI)University of California‐Los AngelesLos AngelesCA 90095
- Dept. of BioengineeringUniversity of California‐Los AngelesLos AngelesCA 90095
| | - Ali Khademhosseini
- Biomaterials Innovation Research Center, Division of Biomedical Engineering, Dept. of MedicineBrigham and Women's Hospital, Harvard Medical SchoolCambridgeMA 02142
- Harvard‐MIT Division of Health Sciences and TechnologyMassachusetts Institute of TechnologyCambridgeMA 02139
- Center for Minimally Invasive Therapeutics (C‐MIT)University of California‐Los AngelesLos AngelesCA 90095
- California NanoSystems Institute (CNSI)University of California‐Los AngelesLos AngelesCA 90095
- Dept. of BioengineeringUniversity of California‐Los AngelesLos AngelesCA 90095
- Dept. of Radiological Sciences, David Geffen School of MedicineUniversity of California‐Los AngelesLos AngelesCA 90095
- Dept. of Chemical and Biomolecular EngineeringUniversity of California‐Los AngelesLos AngelesCA 90095
- Dept. of Bioindustrial Technologies, College of Animal Bioscience and TechnologyKonkuk UniversitySeoulRepublic of Korea
| |
Collapse
|
1005
|
Dai Z, Jin Y, Zheng J, Liu K, Zhao J, Zhang S, Wu F, Sun Z. MiR-217 promotes cell proliferation and osteogenic differentiation of BMSCs by targeting DKK1 in steroid-associated osteonecrosis. Biomed Pharmacother 2019; 109:1112-1119. [PMID: 30551361 DOI: 10.1016/j.biopha.2018.10.166] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Revised: 10/26/2018] [Accepted: 10/26/2018] [Indexed: 12/18/2022] Open
Abstract
MicroRNAs (miRNAs) have recently been recognized to play an important role in bone-associated diseases. This study aims to explore the expression profile and biological function of miR-217, which is known to be related to tumor cell proliferation and migration, to the proliferation and osteogenic differentiation of MSCs from the patients with steroid-associated osteonecrosis (ONFH). Bone marrow was obtained from the proximal femur of 10 patients with ONFH and 10 patients with femoral neck fractures. Bone marrow-derived mesenchymal stem cells (MSCs) were isolated and cultured. The expression profile, biological function of miR-217 and the interaction between miR-217 and DKK1 were assayed using cell viability measurement, western blot, Real-time PCR, luciferase reporter assay, Alizarin Red S (ARS) staining. We noted that the expression level of miR-217 was significantly decreased in the ONFH samples compared to the control samples (P < 0.0001). By targeting DKK1, miR-217 promoted nuclear translocation of β-catenin, increased expression of RUNX2, COL1A1 and obviously promoted the proliferation and differentiation of MSCs. Restoring the expression of DKK1 in the MSCs partially reversed the role of miR-217. These findings suggest that miR-217 promotes cell proliferation and osteogenic differentiation by inhibiting DKK1 during the development of steroid-associated osteonecrosis.
Collapse
Affiliation(s)
- Zhipeng Dai
- Department of Orthopedics, Henan Provincial People's Hospital, Zhengzhou, Henan, China
| | - Yi Jin
- Department of Orthopedics, Henan Provincial People's Hospital, Zhengzhou, Henan, China.
| | - Jia Zheng
- Department of Orthopedics, Henan Provincial People's Hospital, Zhengzhou, Henan, China
| | - Ke Liu
- Department of Orthopedics, Henan Provincial People's Hospital, Zhengzhou, Henan, China
| | - Jiajun Zhao
- Department of Orthopedics, Henan Provincial People's Hospital, Zhengzhou, Henan, China
| | - Shanfeng Zhang
- Department of Orthopedics, Wuhan Fourth Hospital, Puai Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Fei Wu
- Department of Orthopedics, Renmin Hospital, Wuhan University, Wuhan, Hubei, China
| | - Zhibo Sun
- Department of Orthopedics, Renmin Hospital, Wuhan University, Wuhan, Hubei, China; Department of Orthopedics, Wuhan Fourth Hospital, Puai Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China.
| |
Collapse
|
1006
|
Incidence and severity of femoral head avascularity after femoral neck or intertrochanteric fractures on preoperative bone single photon emission computed tomography/computed tomography: preliminary study. Nucl Med Commun 2018; 40:199-205. [PMID: 30531406 DOI: 10.1097/mnm.0000000000000963] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
OBJECTIVE The aim of the present study was to evaluate the incidence and degree of femoral head avascularity depending on the types of femoral neck or intertrochanteric fractures using preoperative bone single photon emission computed tomography (SPECT)/computed tomography (CT). PATIENTS AND METHODS A total of 131 patients with femoral neck or intertrochanteric fractures who underwent preoperative bone SPECT/CT were enrolled. Femoral head avascularity was evaluated using bone SPECT/CT images. Visual scores ranged from 1 to 5 based on visually assessed diagnostic confidence: 1=definitely avascular femoral head, 2=likely avascular, 3=equivocal, 4=likely not avascular, and 5=definitely not avascular. In quantitative analysis, contralateral, ipsilateral, and size ratios were measured. RESULTS Among 131 patients, 39 of 54 (72.22%) with femoral neck fractures and 23 of 77 (29.87%) with intertrochanteric fractures showed avascular femoral heads. The incidence of femoral head avascularity was significantly higher in patients with femoral neck fracture than those with intertrochanteric fracture. While the incidence and severity of femoral head avascularity increased with higher Garden stage in femoral neck fracture, neither was related to AO/Orthopaedic Trauma Association classification of intertrochanteric fracture. In addition, the number of bony fragments around intertrochanteric fractures was not a significant predictor of femoral head avascularity. CONCLUSION Although avascular femoral head on bone SPECT/CT does not directly indicate avascular necrosis, assessing the incidence and severity of femoral head avascularity using qualitative and quantitative parameters could give clinically useful information related to the classification. Additional studies with larger sample sizes would be a next step to expand the clinical role of bone SPECT/CT.
Collapse
|
1007
|
Sharma S, Kumar R, Kumari P, Kharwar RN, Yadav AK, Saripella S. Mechanically magnified chitosan-based hydrogel as tissue adhesive and antimicrobial candidate. Int J Biol Macromol 2018; 125:109-115. [PMID: 30521916 DOI: 10.1016/j.ijbiomac.2018.12.018] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Revised: 11/23/2018] [Accepted: 12/01/2018] [Indexed: 12/29/2022]
Abstract
The present article reports the development of chitosan (CS) based hydrogel series by varying the concentration of cross-linking agent i.e. N,N'-methylenebisacrylamide (MBA) (0.8-1.4 wt%) via free-radical polymerization in aqueous medium. SEM image analysis confirmed the presence of porous 3D-network in the hydrogel. Prepared hydrogel series exhibited good tissue adhesive property along with antimicrobial activity against E. coli, K. pneumonia, S. aureus, C. albicans &M. gypseum bacteria with the good MIC (4-20 mm). The adhesive strength of hydrogel was found 14 kPa, which seems to be quite efficient in tissue adhesiveness applications, which was also validated and tested on Drosophila (Oregon-R) tissues, results were promising. Magnified mechanical strength i.e. storage modulus (G') and loss modulus (G″) were found 106 Pa and 104 Pa, respectively, which makes the hydrogel a potential candidate in the biomedical field. Moreover, CS hydrogel showed good swelling ratio in aqueous medium up to 390% at room temperature.
Collapse
Affiliation(s)
- Swati Sharma
- Organic Polymer Laboratory, Centre of Advanced Studies in Chemistry, Institute of Science, Banaras Hindu University, Varanasi 221005, UP, India
| | - Rajesh Kumar
- Organic Polymer Laboratory, Centre of Advanced Studies in Chemistry, Institute of Science, Banaras Hindu University, Varanasi 221005, UP, India.
| | - Puja Kumari
- Department of Botany, Institute of Science, Banaras Hindu University, Varanasi 221005, UP, India
| | - Ravindra Nath Kharwar
- Department of Botany, Institute of Science, Banaras Hindu University, Varanasi 221005, UP, India
| | - Amarish Kumar Yadav
- Department of Biochemistry, Institute of Science, Banaras Hindu University, Varanasi 221005, UP, India
| | - Srikrishna Saripella
- Department of Biochemistry, Institute of Science, Banaras Hindu University, Varanasi 221005, UP, India
| |
Collapse
|
1008
|
Ewalefo SO, Dombrowski M, Hirase T, Rocha JL, Weaver M, Kline A, Carney D, Hogan MV. Management of Posttraumatic Ankle Arthritis: Literature Review. Curr Rev Musculoskelet Med 2018; 11:546-557. [PMID: 30327933 PMCID: PMC6220012 DOI: 10.1007/s12178-018-9525-9] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
PURPOSE OF REVIEW Trauma is the principle cause of osteoarthritis in the ankle, which is associated with significant morbidity. This review highlights the current literature for the purpose of bringing the reader up-to-date on the management of posttraumatic ankle arthritis, describing treatment efficacy, indications, contraindications, and complications. RECENT FINDINGS Recent studies on osteoarthritis have demonstrated variability among anatomic locations regarding the mechanisms and rates of development for posttraumatic osteoarthritis, which are attributed to newly discovered biological differences intrinsic to each joint. Regarding surgical management of posttraumatic ankle arthritis, osteochondral allograft transplantation of the talus, and supramalleolar osteotomies have demonstrated promising results. Additionally, the outpatient setting was found to be appropriate for managing pain following total ankle arthroplasty, associated with low complication rates and no readmission. Management for posttraumatic ankle arthritis is generally progressive. Initial treatment entails nonpharmacologic options with surgery reserved for posttraumatic ankle arthritis refractory to conservative treatment. Patient demographics and lifestyles should be carefully considered when formulating a management strategy, as outcomes are dependent upon the satisfaction of each set of respective criteria. Ultimately, the management of posttraumatic ankle arthritis should be individualized to satisfy the needs and desires, which are specific to each patient.
Collapse
Affiliation(s)
- Samuel O Ewalefo
- Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.
- Texas A&M College of Medicine, Bryan, TX, USA.
| | - Malcolm Dombrowski
- Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Takashi Hirase
- Department of Orthopedics and Sports Medicine, Houston Methodist Hospital, Houston, TX, USA
| | - Jorge L Rocha
- Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Mitchell Weaver
- Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Alex Kline
- Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Dwayne Carney
- Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - MaCalus V Hogan
- Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| |
Collapse
|
1009
|
Maruyama M, Nabeshima A, Pan CC, Behn AW, Thio T, Lin T, Pajarinen J, Kawai T, Takagi M, Goodman SB, Yang YP. The effects of a functionally-graded scaffold and bone marrow-derived mononuclear cells on steroid-induced femoral head osteonecrosis. Biomaterials 2018; 187:39-46. [PMID: 30292940 PMCID: PMC6193256 DOI: 10.1016/j.biomaterials.2018.09.030] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2018] [Revised: 09/15/2018] [Accepted: 09/17/2018] [Indexed: 12/30/2022]
Abstract
Osteonecrosis of the femoral head (ONFH) is a debilitating disease that may progress to femoral head collapse and subsequently, degenerative arthritis. Although injection of bone marrow-derived mononuclear cells (BMMCs) is often performed with core decompression (CD) in the early stage of ONFH, these treatments are not always effective in prevention of disease progression and femoral head collapse. We previously described a novel 3D printed, customized functionally-graded scaffold (FGS) that improved bone growth in the femoral head after CD in a normal healthy rabbit, by providing structural and mechanical guidance. The present study demonstrates similar results of the FGS in a rabbit steroid-induced osteonecrosis model. Furthermore, the injection of BMMCs into the CD decreased the osteonecrotic area in the femoral head. Thus, the combination of FGS and BMMC provides a new therapy modality that may improve the outcome of CD for early stage of ONFH by providing both enhanced biological and biomechanical cues to promote bone regeneration in the osteonecrotic area.
Collapse
Affiliation(s)
- Masahiro Maruyama
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, CA, USA; Department of Orthopaedic Surgery, Yamagata University Faculty of Medicine, Yamagata, Japan
| | - Akira Nabeshima
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Chi-Chun Pan
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, CA, USA; Mechanical Engineering, Stanford University School of Medicine, Stanford, CA, USA
| | - Anthony W Behn
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Timothy Thio
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Tzuhua Lin
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Jukka Pajarinen
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Toshiyuki Kawai
- Department of Orthopaedic Surgery, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Michiaki Takagi
- Department of Orthopaedic Surgery, Yamagata University Faculty of Medicine, Yamagata, Japan
| | - Stuart B Goodman
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, CA, USA; Bioengineering, Stanford University School of Medicine, Stanford, CA, USA.
| | - Yunzhi Peter Yang
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, CA, USA; Material Science and Engineering, Stanford University School of Medicine, Stanford, CA, USA; Bioengineering, Stanford University School of Medicine, Stanford, CA, USA.
| |
Collapse
|
1010
|
Latfi ASA, Pramanik S, Poon CT, Gumel AM, Lai KW, Annuar MSM, Pingguan-Murphy B. Structural and bone marrow stem cell biocompatibility studies of hydrogel synthesized via chemo-enzymatic route. J Biomater Appl 2018; 33:854-865. [PMID: 30458659 DOI: 10.1177/0885328218812490] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Natural biopolymers have many attractive medical applications; however, complications due to fibrosis caused a reduction in diffusion and dispersal of nutrients and waste products. Consequently, severe immunocompatibility problems and poor mechanical and degradation properties in synthetic polymers ensue. Hence, the present study investigates a novel hydrogel material synthesized from caprolactone, ethylene glycol, ethylenediamine, polyethylene glycol, ammonium persulfate, and tetramethylethylenediamine via chemo-enzymatic route. Spectroscopic analyses indicated the formation of polyurea and polyhydroxyurethane as the primary building block of the hydrogel starting material. Biocompatibility studies showed positive observation in biosafety test using direct contact cytotoxicity assay in addition to active cellular growth on the hydrogel scaffold based on fluorescence observation. The synthesized hydrogel also exhibited (self)fluorescence properties under specific wavelength excitation. Hence, synthesized hydrogel could be a potential candidate for medical imaging as well as tissue engineering applications as a tissue expander, coating material, biosensor, and drug delivery system.
Collapse
Affiliation(s)
- Ahmad Safwan Ahmad Latfi
- 1 Department of Biomedical Engineering, Faculty of Engineering, University of Malaya, Kuala Lumpur, Malaysia
| | - Sumit Pramanik
- 2 Department of Mechanical Engineering, SRM Institute of Science and Technology, Chennai, India
| | - Chi Tat Poon
- 1 Department of Biomedical Engineering, Faculty of Engineering, University of Malaya, Kuala Lumpur, Malaysia
| | - Ahmad Mohammed Gumel
- 3 Institute of Biological Sciences, Faculty of Science, University of Malaya, Kuala Lumpur, Malaysia
| | - Khin Wee Lai
- 1 Department of Biomedical Engineering, Faculty of Engineering, University of Malaya, Kuala Lumpur, Malaysia
| | | | - Belinda Pingguan-Murphy
- 1 Department of Biomedical Engineering, Faculty of Engineering, University of Malaya, Kuala Lumpur, Malaysia
| |
Collapse
|
1011
|
Emerging role of circadian rhythm in bone remodeling. J Mol Med (Berl) 2018; 97:19-24. [PMID: 30446776 DOI: 10.1007/s00109-018-1723-9] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Revised: 11/07/2018] [Accepted: 11/09/2018] [Indexed: 12/31/2022]
Abstract
The 24-h rhythm of behavioral and physiological processes is a typical biological phenomenon regulated by a group of circadian rhythm genes. Dysfunction of the circadian rhythm can cause a wide range of problems, such as cancer and metabolic diseases. In recent decades, increased understanding of the roles of circadian rhythm genes in the bone remodeling process have been documented, including osteoblastic bone formation, osteoclastic bone resorption, and osteoblast/osteoclast communication. A timely review of the current findings may help to facilitate the new field of circadian rhythmic bone remodeling research. Targeted pharmacological modulation of circadian rhythm genes is a possible therapeutic approach through which to overcome bone remodeling problems in the future.
Collapse
|
1012
|
Study of Osteocyte Behavior by High-Resolution Intravital Imaging Following Photo-Induced Ischemia. Molecules 2018; 23:molecules23112874. [PMID: 30400346 PMCID: PMC6278482 DOI: 10.3390/molecules23112874] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Revised: 10/26/2018] [Accepted: 10/28/2018] [Indexed: 11/17/2022] Open
Abstract
Ischemic injuries and local hypoxia can result in osteocytes dysfunction and play a key role in the pathogenesis of avascular osteonecrosis. Conventional imaging techniques including magnetic resonance imaging (MRI) and computed tomography (CT) can reveal structural and functional changes within bony anatomy; however, characterization of osteocyte behavioral dynamics in the setting of osteonecrosis at the single cell resolution is limited. Here, we demonstrate an optical approach to study real-time osteocyte functions in vivo. Using nicotinamide adenine dinucleotide (NADH) as a biomarker for metabolic dynamics in osteocytes, we showed that NADH level within osteocytes transiently increase significantly after local ischemia through non-invasive photo-induced thrombosis of afferent arterioles followed by a steady decline. Our study presents a non-invasive optical approach to study osteocyte behavior through the modulation of local environmental conditions. Thus it provides a powerful toolkit to study cellular processes involved in bone pathologies in vivo.
Collapse
|
1013
|
Lane NE, Mohan G, Yao W, Shidara K, Lay YAE, Junjing J, Dubrovsky A, Kimmel DB. Prevalence of glucocorticoid induced osteonecrosis in the mouse is not affected by treatments that maintain bone vascularity. Bone Rep 2018; 9:181-187. [PMID: 30510976 PMCID: PMC6260230 DOI: 10.1016/j.bonr.2018.10.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Revised: 09/24/2018] [Accepted: 10/31/2018] [Indexed: 02/07/2023] Open
Abstract
Objective Determine if LLP2A-Ale or PTH (1–34) affects the prevalence of glucocorticoid-induced osteonecrosis (ON) in a mouse model. Methods Eight-week-old young adult male BALB/cJ mice were weight-randomized into Control (Con), glucocorticoid (GC)-only, or concurrent treatments with GC and LLP2A-Ale (250 μg/kg or 500 μg/kg, IV, Days 1, 14, 28) or parathyroid hormone hPTH (1–34) (40 μg/kg, 5×/week). Mice were necropsied after 45 days for qualitative evaluation of prevalent ON and quantitative evaluation of vascularity in the distal femoral epiphysis (DFE); and quantitative evaluation of bone mass, microarchitecture, and strength in the distal femoral metaphysis and lumbar vertebral body. Results The prevalence of ON was 14% in the Con group and 36% in the GC-only group (P = 0.07). The prevalence of ON did not differ among GC-only, GC + LLP2A-Ale, and GC + PTH groups. GC-only mice had significantly lower trabecular and cortical bone strength than Con, while GC + LLP2A-Ale (500 μg/kg) and GC + PTH (1–34) groups had significantly greater trabecular bone strength than the GC-only group. GC + LLP2A-Ale (250 μg/kg and 500 μg/kg) and GC + PTH had significantly higher trabecular bone volume than GC-only mice at the vertebrae, distal femoral epiphyses and distal femoral metaphyses. DFE vascularity was lower in GC-only mice than in all other groups. Conclusion Neither LLP2A-Ale nor hPTH (1–34) reduced the prevalence of GC-induced ON, compared to GC-only mice. However, GC-treated mice given LLP2A-Ale or hPTH (1–34) had better bone mass, microarchitecture, and strength in trabecular-rich regions, and higher levels of vascularity than GC-only mice.
Collapse
Affiliation(s)
- Nancy E Lane
- Center for Musculoskeletal Health, University of California at Davis Medical Center, Sacramento, CA 95817, USA
| | - Geetha Mohan
- Center for Musculoskeletal Health, University of California at Davis Medical Center, Sacramento, CA 95817, USA
| | - Wei Yao
- Center for Musculoskeletal Health, University of California at Davis Medical Center, Sacramento, CA 95817, USA
| | - Kie Shidara
- Center for Musculoskeletal Health, University of California at Davis Medical Center, Sacramento, CA 95817, USA
| | - Yu-An Evan Lay
- Center for Musculoskeletal Health, University of California at Davis Medical Center, Sacramento, CA 95817, USA
| | - Jia Junjing
- Facility of Animal Science, Yunnan Agricultural University, Kunming, Yunnan, 650201, China
| | - Alanna Dubrovsky
- Center for Musculoskeletal Health, University of California at Davis Medical Center, Sacramento, CA 95817, USA
| | - Donald B Kimmel
- Department of Physiological Sciences, University of Florida, Gainesville, FL 32610, USA
| |
Collapse
|
1014
|
Yoo DY, Cho SB, Jung HY, Kim W, Lee KY, Kim JW, Moon SM, Won MH, Choi JH, Yoon YS, Kim DW, Choi SY, Hwang IK. Protein disulfide-isomerase A3 significantly reduces ischemia-induced damage by reducing oxidative and endoplasmic reticulum stress. Neurochem Int 2018; 122:19-30. [PMID: 30399388 DOI: 10.1016/j.neuint.2018.11.002] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2018] [Revised: 11/02/2018] [Accepted: 11/02/2018] [Indexed: 12/19/2022]
Abstract
Ischemia causes oxidative stress in the endoplasmic reticulum (ER), accelerates the accumulation of unfolded and misfolded proteins, and may ultimately lead to neuronal cell apoptosis. In the present study, we investigated the effects of protein disulfide-isomerase A3 (PDIA3), an ER-resident chaperone that catalyzes disulfide-bond formation in a subset of glycoproteins, against oxidative damage in the hypoxic HT22 cell line and against ischemic damage in the gerbil hippocampus. We also confirmed the neuroprotective effects of PDIA3 by using PDIA3-knockout HAP1 cells. The HT22 and HAP1 cell lines showed effective (dose-dependent and time-dependent) penetration and stable expression of the Tat-PDIA3 fusion protein 24 h after Tat-PDIA3 treatment compared to that in the control-PDIA3-treated group. We observed that the fluorescence for both 2',7'-dichlorofluorescein diacetate (DCF-DA) and terminal deoxynucleotidyl transferase dUTP nick-end labeling (TUNEL), which are markers for the formation of hydrogen peroxide (H2O2)-induced reactive oxygen species and apoptosis, respectively, was higher in HAP1 cells than in HT22 cells. The administration of Tat-PDIA3 significantly reduced the (1) DCF-DA and TUNEL fluorescence in HT22 and HAP1 cells, (2) ischemia-induced hyperactivity that was observed 1 day after ischemia/reperfusion, (3) ischemia-induced neuronal damage and glial (astrocytes and microglia) activation that was observed in the hippocampal CA1 region 4 days after ischemia/reperfusion, and (4) lipid peroxidation and nitric oxide generation in the hippocampal homogenates 3-12 h after ischemia/reperfusion. Transient forebrain ischemia significantly elevated the immunoglobulin-binding protein (BiP) and C/EBP-homologous protein (CHOP) mRNA levels in the hippocampus at 12 h and 4 days after ischemia, relative to those in the time-matched sham-operated group. Administration of Tat-PDIA3 ameliorated the ischemia-induced upregulation of BiP mRNA levels versus the Tat peptide- or control-PDIA3-treated groups, and significantly reduced the induction of CHOP mRNA levels, at 12 h or 4 days after ischemia. Collectively, these results suggest that Tat-PDIA3 acts as a neuroprotective agent against ischemia by attenuating oxidative damage and blocking the apoptotic pathway that is related to the unfolded protein response in the ER.
Collapse
Affiliation(s)
- Dae Young Yoo
- Department of Anatomy and Cell Biology, College of Veterinary Medicine, Research Institute for Veterinary Science, Seoul National University, Seoul, 08826, South Korea; Department of Anatomy, College of Medicine, Soonchunhyang University, Cheonan, Chungcheongnam, 31151, South Korea
| | - Su Bin Cho
- Department of Biomedical Sciences, Research Institute for Bioscience and Biotechnology, Hallym University, Chuncheon, 24252, South Korea
| | - Hyo Young Jung
- Department of Anatomy and Cell Biology, College of Veterinary Medicine, Research Institute for Veterinary Science, Seoul National University, Seoul, 08826, South Korea
| | - Woosuk Kim
- Department of Anatomy and Cell Biology, College of Veterinary Medicine, Research Institute for Veterinary Science, Seoul National University, Seoul, 08826, South Korea
| | - Kwon Young Lee
- Department of Anatomy, College of Veterinary Medicine and Institute of Veterinary Science, Kangwon National University, Chuncheon, 24341, South Korea
| | - Jong Whi Kim
- Department of Anatomy and Cell Biology, College of Veterinary Medicine, Research Institute for Veterinary Science, Seoul National University, Seoul, 08826, South Korea
| | - Seung Myung Moon
- Department of Neurosurgery, Dongtan Sacred Heart Hospital, College of Medicine, Hallym University, Hwaseong, 18450, South Korea; Research Institute for Complementary & Alternative Medicine, Hallym University, Chuncheon, 24253, South Korea
| | - Moo-Ho Won
- Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon, 24341, South Korea
| | - Jung Hoon Choi
- Department of Anatomy, College of Veterinary Medicine and Institute of Veterinary Science, Kangwon National University, Chuncheon, 24341, South Korea
| | - Yeo Sung Yoon
- Department of Anatomy and Cell Biology, College of Veterinary Medicine, Research Institute for Veterinary Science, Seoul National University, Seoul, 08826, South Korea
| | - Dae Won Kim
- Department of Biochemistry and Molecular Biology, Research Institute of Oral Sciences, College of Dentistry, Gangneung-Wonju National University, Gangneung, 25457, South Korea
| | - Soo Young Choi
- Department of Biomedical Sciences, Research Institute for Bioscience and Biotechnology, Hallym University, Chuncheon, 24252, South Korea.
| | - In Koo Hwang
- Department of Anatomy and Cell Biology, College of Veterinary Medicine, Research Institute for Veterinary Science, Seoul National University, Seoul, 08826, South Korea.
| |
Collapse
|
1015
|
Rezvantalab S, Drude NI, Moraveji MK, Güvener N, Koons EK, Shi Y, Lammers T, Kiessling F. PLGA-Based Nanoparticles in Cancer Treatment. Front Pharmacol 2018; 9:1260. [PMID: 30450050 PMCID: PMC6224484 DOI: 10.3389/fphar.2018.01260] [Citation(s) in RCA: 331] [Impact Index Per Article: 47.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Accepted: 10/15/2018] [Indexed: 12/18/2022] Open
Abstract
Nanomedicines can be used for a variety of cancer therapies including tumor-targeted drug delivery, hyperthermia, and photodynamic therapy. Poly (lactic-co-glycolic acid) (PLGA)-based materials are frequently used in such setups. This review article gives an overview of the properties of previously reported PLGA nanoparticles (NPs), their behavior in biological systems, and their use for cancer therapy. Strategies are emphasized to target PLGA NPs to the tumor site passively and actively. Furthermore, combination therapies are introduced that enhance the accumulation of NPs and, thereby, their therapeutic efficacy. In this context, the huge number of reports on PLGA NPs used as drug delivery systems in cancer treatment highlight the potential of PLGA NPs as drug carriers for cancer therapeutics and encourage further translational research.
Collapse
Affiliation(s)
- Sima Rezvantalab
- Department of Chemical Engineering, Amirkabir University of Technology (Tehran Polytechnic), Tehran, Iran.,Institute for Experimental Molecular Imaging, University Clinic and Helmholtz Institute for Biomedical Engineering, RWTH Aachen University, Aachen, Germany
| | - Natascha Ingrid Drude
- Institute for Experimental Molecular Imaging, University Clinic and Helmholtz Institute for Biomedical Engineering, RWTH Aachen University, Aachen, Germany.,Department of Nuclear Medicine, Uniklinik RWTH Aachen University, Aachen, Germany
| | - Mostafa Keshavarz Moraveji
- Department of Chemical Engineering, Amirkabir University of Technology (Tehran Polytechnic), Tehran, Iran
| | - Nihan Güvener
- Institute for Experimental Molecular Imaging, University Clinic and Helmholtz Institute for Biomedical Engineering, RWTH Aachen University, Aachen, Germany
| | - Emily Kate Koons
- Department of Pharmacology and Toxicology, College of Pharmacy & UA Cancer Center, University of Arizona, Tucson, AZ, United States
| | - Yang Shi
- Institute for Experimental Molecular Imaging, University Clinic and Helmholtz Institute for Biomedical Engineering, RWTH Aachen University, Aachen, Germany
| | - Twan Lammers
- Institute for Experimental Molecular Imaging, University Clinic and Helmholtz Institute for Biomedical Engineering, RWTH Aachen University, Aachen, Germany
| | - Fabian Kiessling
- Institute for Experimental Molecular Imaging, University Clinic and Helmholtz Institute for Biomedical Engineering, RWTH Aachen University, Aachen, Germany
| |
Collapse
|
1016
|
Fan W, Li J, Yuan L, Chen J, Wang Z, Wang Y, Guo C, Mo X, Yan Z. Intra-articular injection of kartogenin-conjugated polyurethane nanoparticles attenuates the progression of osteoarthritis. Drug Deliv 2018; 25:1004-1012. [PMID: 29667446 PMCID: PMC6058480 DOI: 10.1080/10717544.2018.1461279] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2018] [Revised: 04/01/2018] [Accepted: 04/02/2018] [Indexed: 01/10/2023] Open
Abstract
Osteoarthritis (OA) is the most common form of joint disease and a leading cause of physical disability, there is an urgent need to attenuate the progression of OA. Intra-articular (IA) injection is an effective treatment for joints diseases, however, the therapeutic effects mostly depend on the efficacy of drug duration in joints. Drug delivery system can provide drug-controlled release and reduce the number of IA injection. In this study, amphiphilic polyurethanes with pendant amino group were synthesized and amide bonds were formed between the amine group of polyurethane and the carboxyl group of kartogenin (KGN), a small molecular reported to show both regenerative and protective effects on cartilage. Our results showed that KGN-conjugated polyurethane nanoparticles (PN-KGN) were spherical and regular in shape with an average size of 25 nm and could sustained and controlled release of KGN in vitro. PN-KGN showed no cytotoxicity and pro-inflammatory effects on chondrocytes. The therapeutic effects in OA model showed that IA injection of KGN could attenuate the progress of OA, however, the cartilage degeneration became obviously at 12 weeks with matrix loss and vertical fissures. By contrast, IA injection of PN-KGN showed less cartilage degeneration with significant lower OARSI scores even at 12 weeks, indicating PN-KGN could further arrest the development of OA. Immunohistochemistry also validated that IA injection of PN-KGN retained the normal compositions of cartilage matrix, with much stronger Col II staining and less Col I staining. In conclusion, IA injection of PN-KGN is a better potential strategy to treat OA, with long-time cartilage protection and less IA injections.
Collapse
Affiliation(s)
- Wenshuai Fan
- Department of Orthopedics, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Jinghuan Li
- Department of Hepatic Oncology, Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Liu Yuan
- Biomaterials and Tissue Engineering Lab, College of Chemistry, Chemical Engineering and Biotechnology, Donghua University, Shanghai, China
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Materials Science and Engineering, Donghua University, Shanghai, China
| | - Jifei Chen
- Department of Orthopedics, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Zhe Wang
- Department of Orthopedics, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yiming Wang
- Department of Orthopedics, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Changan Guo
- Department of Orthopedics, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Xiumei Mo
- Biomaterials and Tissue Engineering Lab, College of Chemistry, Chemical Engineering and Biotechnology, Donghua University, Shanghai, China
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Materials Science and Engineering, Donghua University, Shanghai, China
| | - Zuoqin Yan
- Department of Orthopedics, Zhongshan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
1017
|
Gharanizadeh K, Aminizadeh S, Molavi N, Darbandi A, Nadjafi S, Fadavighaffari M, Shooshtarizadeh T. Effects of Zoledronic Acid and Vitamin E on Surgical- Induced Osteonecrosis of the Femoral Head in Rabbit. THE ARCHIVES OF BONE AND JOINT SURGERY 2018; 6:547-553. [PMID: 30637311 PMCID: PMC6310187] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 06/22/2017] [Accepted: 11/02/2017] [Indexed: 06/09/2023]
Abstract
BACKGROUND Femoral head osteonecrosis is a progressive disease with disabling outcomes in hip joint if not treated. This study was designed to compare the effects of zoledronic acid plus vitamin E versus zoledronic acid alone in surgical induced femoral head osteonecrosis in rabbits. METHODS 26 Japanese white adult normal male rabbits at 28-32 weeks old were undertaken surgical femoral dislocation to devastate the femoral neck vessels; the femoral neck vessels were ligated and the hip was relocated. Next, the first 10 rabbits received zoledronic acid injections at 1st and the 4th weeks; the second group (10 rabbits) received zoledronic acid injections at 1st and the 4th week along with daily oral vitamin E for 12 weeks; and the third group was considered as non-treated control group. Radiographic and postmortem pathological assessments including the Ficat classification, epiphyseal quotient (EQ), new bone formation, and residual necrotic bone (RNB) were performed and compared after week 12. RESULTS A significant difference was found between the combination therapy group and the control group in Ficat classification at 12th weeks (P=0.048), but, the difference between monotherapy and combination therapy groups at 12th weeks was nonsignificant (P=0.37). Also, both treated groups had significant difference with the control group for RNB (P=0.015). There were no significant differences between the three groups for Ficat classification at the 6th week (P=0.65); EQ at 6th (P=0.59) and 12th week (P=0.64); and NBF (P=0.55). CONCLUSION Although zoledronic acid therapy along with vitamin E could improve some radiologic and pathological indices related to femoral head osteonecrosis, vitamin E showed a relative impact. LEVEL OF EVIDENCE I.
Collapse
Affiliation(s)
- Kaveh Gharanizadeh
- Research performed at Shaheed Rajaei Cardiovascular Medical and Research Center, Iran University of Medical Sciences (IUMS), Tehran, Iran
- Bone and Joint Reconstruction Research Center, Shafa Orthopedic Hospital, Iran University of Medical Sciences (IUMS), Tehran, Iran
- Rajaie Cardiovascular, Medical and Research Centre, Iran University of Medical Sciences (IUMS), Tehran, Iran
- Iran University of Medical Sciences (IUMS), Tehran, Iran
| | - Sina Aminizadeh
- Research performed at Shaheed Rajaei Cardiovascular Medical and Research Center, Iran University of Medical Sciences (IUMS), Tehran, Iran
- Bone and Joint Reconstruction Research Center, Shafa Orthopedic Hospital, Iran University of Medical Sciences (IUMS), Tehran, Iran
- Rajaie Cardiovascular, Medical and Research Centre, Iran University of Medical Sciences (IUMS), Tehran, Iran
- Iran University of Medical Sciences (IUMS), Tehran, Iran
| | - Nima Molavi
- Research performed at Shaheed Rajaei Cardiovascular Medical and Research Center, Iran University of Medical Sciences (IUMS), Tehran, Iran
- Bone and Joint Reconstruction Research Center, Shafa Orthopedic Hospital, Iran University of Medical Sciences (IUMS), Tehran, Iran
- Rajaie Cardiovascular, Medical and Research Centre, Iran University of Medical Sciences (IUMS), Tehran, Iran
- Iran University of Medical Sciences (IUMS), Tehran, Iran
| | - Amir Darbandi
- Research performed at Shaheed Rajaei Cardiovascular Medical and Research Center, Iran University of Medical Sciences (IUMS), Tehran, Iran
- Bone and Joint Reconstruction Research Center, Shafa Orthopedic Hospital, Iran University of Medical Sciences (IUMS), Tehran, Iran
- Rajaie Cardiovascular, Medical and Research Centre, Iran University of Medical Sciences (IUMS), Tehran, Iran
- Iran University of Medical Sciences (IUMS), Tehran, Iran
| | - Shabnam Nadjafi
- Research performed at Shaheed Rajaei Cardiovascular Medical and Research Center, Iran University of Medical Sciences (IUMS), Tehran, Iran
- Bone and Joint Reconstruction Research Center, Shafa Orthopedic Hospital, Iran University of Medical Sciences (IUMS), Tehran, Iran
- Rajaie Cardiovascular, Medical and Research Centre, Iran University of Medical Sciences (IUMS), Tehran, Iran
- Iran University of Medical Sciences (IUMS), Tehran, Iran
| | - Mahsa Fadavighaffari
- Research performed at Shaheed Rajaei Cardiovascular Medical and Research Center, Iran University of Medical Sciences (IUMS), Tehran, Iran
- Bone and Joint Reconstruction Research Center, Shafa Orthopedic Hospital, Iran University of Medical Sciences (IUMS), Tehran, Iran
- Rajaie Cardiovascular, Medical and Research Centre, Iran University of Medical Sciences (IUMS), Tehran, Iran
- Iran University of Medical Sciences (IUMS), Tehran, Iran
| | - Tina Shooshtarizadeh
- Research performed at Shaheed Rajaei Cardiovascular Medical and Research Center, Iran University of Medical Sciences (IUMS), Tehran, Iran
- Bone and Joint Reconstruction Research Center, Shafa Orthopedic Hospital, Iran University of Medical Sciences (IUMS), Tehran, Iran
- Rajaie Cardiovascular, Medical and Research Centre, Iran University of Medical Sciences (IUMS), Tehran, Iran
- Iran University of Medical Sciences (IUMS), Tehran, Iran
| |
Collapse
|
1018
|
Sobhan MR, Mahdinezhad-Yazdi M, Moghimi M, Aghili K, Jafari M, Zare-Shehneh M, Neamatzadeh H. Plasminogen Activator Inhibitor-1 4G/5G Polymorphism Contributes to Osteonecrosis of the Femoral Head Susceptibility: Evidence from a Systematic Review and Meta-analysis. THE ARCHIVES OF BONE AND JOINT SURGERY 2018; 6:468-477. [PMID: 30637301 PMCID: PMC6310193] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 05/18/2018] [Accepted: 09/23/2018] [Indexed: 06/09/2023]
Abstract
BACKGROUND The Plasminogen Activator Inhibitor-1 gene 4G/5G (PAI-1 4G/5G) polymorphism has been suggested to be associated with osteonecrosis of the femoral head (ONFH) susceptibility; however, the results are conflicting and inconclusive. We have carried out a comprehensive meta-analysis to derive a more precise estimation of the association. METHODS A comprehensive search in PubMed, EMBASE, Google Scholar, and ISI Web of Knowledge databases was conducted to identify all eligible case-control publications investigating the association between PAI-1 4G/5G polymorphism and ONFH risk. Odds ratios (OR) and corresponding 95% confidence intervals (CI) were used to assess the association. RESULTS A total of six studies with 456 cases and 1,019 controls were included in this review. Three studies were from Caucasian descendants and the three others were from East Asian descendants. Overall analysis suggests a significant association between PAI-1 4G/5G polymorphism and ONFH risk under the allele model (4G vs. 5G: OR =1.540, 95% CI =1.055-2.248, P=0.025) and the recessive model (4G4G vs. 4G5G+5G5G: OR=1.931, 95% CI: 1.162-3.207, P=0.011). When stratified by ethnicity, we have found a significant association between PAI-1 4G/5G polymorphism and ONFH risk among the Caucasian (4G5G vs. 5G5G: OR=1.806, 95% CI: 1.064-3.067, P=0.029) and East Asians (4G4G vs. 5G5G: OR=1.619, 95% CI: 1.025-2.556, P=0.039 and 4G4G vs. 4G5G+5G5G: OR=1.665, 95% CI: 1.207-2.297, P=0.002). CONCLUSION The present meta-analysis suggested that PAI-1 4G/5G (rs1799889) polymorphism is a potential risk factor for development of ONFH. However, large-scale and well-designed case-control studies in different ethnicities are required to validate these results.
Collapse
Affiliation(s)
- Mohammad R Sobhan
- Research performed at the Department of Orthopedics, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
- Department of Orthopedics, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
- Department of Orthopedics, Afshar Hospital, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
- Department of Pathology, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
- Department of Radiology, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
- Department of Emergency Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
- Department of Medical Genetics, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Masoud Mahdinezhad-Yazdi
- Research performed at the Department of Orthopedics, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
- Department of Orthopedics, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
- Department of Orthopedics, Afshar Hospital, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
- Department of Pathology, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
- Department of Radiology, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
- Department of Emergency Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
- Department of Medical Genetics, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Mansour Moghimi
- Research performed at the Department of Orthopedics, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
- Department of Orthopedics, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
- Department of Orthopedics, Afshar Hospital, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
- Department of Pathology, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
- Department of Radiology, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
- Department of Emergency Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
- Department of Medical Genetics, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Kazem Aghili
- Research performed at the Department of Orthopedics, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
- Department of Orthopedics, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
- Department of Orthopedics, Afshar Hospital, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
- Department of Pathology, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
- Department of Radiology, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
- Department of Emergency Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
- Department of Medical Genetics, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Mohammadali Jafari
- Research performed at the Department of Orthopedics, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
- Department of Orthopedics, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
- Department of Orthopedics, Afshar Hospital, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
- Department of Pathology, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
- Department of Radiology, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
- Department of Emergency Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
- Department of Medical Genetics, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Masoud Zare-Shehneh
- Research performed at the Department of Orthopedics, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
- Department of Orthopedics, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
- Department of Orthopedics, Afshar Hospital, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
- Department of Pathology, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
- Department of Radiology, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
- Department of Emergency Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
- Department of Medical Genetics, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Hossein Neamatzadeh
- Research performed at the Department of Orthopedics, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
- Department of Orthopedics, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
- Department of Orthopedics, Afshar Hospital, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
- Department of Pathology, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
- Department of Radiology, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
- Department of Emergency Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
- Department of Medical Genetics, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| |
Collapse
|
1019
|
Rivera-Reyes A, Ye S, E Marino G, Egolf S, E Ciotti G, Chor S, Liu Y, Posimo JM, Park PMC, Pak K, Babichev Y, Sostre-Colón J, Tameire F, Leli NM, Koumenis C, C Brady D, Mancuso A, Weber K, Gladdy R, Qi J, Eisinger-Mathason TSK. YAP1 enhances NF-κB-dependent and independent effects on clock-mediated unfolded protein responses and autophagy in sarcoma. Cell Death Dis 2018; 9:1108. [PMID: 30382078 PMCID: PMC6208433 DOI: 10.1038/s41419-018-1142-4] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2018] [Revised: 10/03/2018] [Accepted: 10/10/2018] [Indexed: 12/12/2022]
Abstract
Terminal differentiation opposes proliferation in the vast majority of tissue types. As a result, loss of lineage differentiation is a hallmark of aggressive cancers, including soft tissue sarcomas (STS). Consistent with these observations, undifferentiated pleomorphic sarcoma (UPS), an STS subtype devoid of lineage markers, is among the most lethal sarcomas in adults. Though tissue-specific features are lost in these mesenchymal tumors they are most commonly diagnosed in skeletal muscle, and are thought to develop from transformed muscle progenitor cells. We have found that a combination of HDAC (Vorinostat) and BET bromodomain (JQ1) inhibition partially restores differentiation to skeletal muscle UPS cells and tissues, enforcing a myoblast-like identity. Importantly, differentiation is partially contingent upon downregulation of the Hippo pathway transcriptional effector Yes-associated protein 1 (YAP1) and nuclear factor (NF)-κB. Previously, we observed that Vorinostat/JQ1 inactivates YAP1 and restores oscillation of NF-κB in differentiating myoblasts. These effects correlate with reduced tumorigenesis, and enhanced differentiation. However, the mechanisms by which the Hippo/NF-κB axis impact differentiation remained unknown. Here, we report that YAP1 and NF-κB activity suppress circadian clock function, inhibiting differentiation and promoting proliferation. In most tissues, clock activation is antagonized by the unfolded protein response (UPR). However, skeletal muscle differentiation requires both Clock and UPR activity, suggesting the molecular link between them is unique in muscle. In skeletal muscle-derived UPS, we observed that YAP1 suppresses PERK and ATF6-mediated UPR target expression as well as clock genes. These pathways govern metabolic processes, including autophagy, and their disruption shifts metabolism toward cancer cell-associated glycolysis and hyper-proliferation. Treatment with Vorinostat/JQ1 inhibited glycolysis/MTOR signaling, activated the clock, and upregulated the UPR and autophagy via inhibition of YAP1/NF-κB. These findings support the use of epigenetic modulators to treat human UPS. In addition, we identify specific autophagy, UPR, and muscle differentiation-associated genes as potential biomarkers of treatment efficacy and differentiation.
Collapse
Affiliation(s)
- Adrian Rivera-Reyes
- Abramson Family Cancer Research Institute, Department of Pathology & Laboratory Medicine, Penn Sarcoma Program, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Shuai Ye
- Abramson Family Cancer Research Institute, Department of Pathology & Laboratory Medicine, Penn Sarcoma Program, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Gloria E Marino
- Abramson Family Cancer Research Institute, Department of Pathology & Laboratory Medicine, Penn Sarcoma Program, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Shaun Egolf
- Abramson Family Cancer Research Institute, Department of Pathology & Laboratory Medicine, Penn Sarcoma Program, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Gabrielle E Ciotti
- Abramson Family Cancer Research Institute, Department of Pathology & Laboratory Medicine, Penn Sarcoma Program, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Susan Chor
- Abramson Family Cancer Research Institute, Department of Pathology & Laboratory Medicine, Penn Sarcoma Program, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Ying Liu
- Abramson Family Cancer Research Institute, Department of Pathology & Laboratory Medicine, Penn Sarcoma Program, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Jessica M Posimo
- Abramson Family Cancer Research Institute, Department of Pathology & Laboratory Medicine, Penn Sarcoma Program, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA.,Department of Cancer Biology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Paul M C Park
- Department of Medicine, Harvard Medical School, Boston, MA, 02115, USA
| | - Koreana Pak
- Abramson Family Cancer Research Institute, Department of Pathology & Laboratory Medicine, Penn Sarcoma Program, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Yael Babichev
- Department of Surgery and Institute of Medical Science, University of Toronto, Toronto, ON, Canada.,Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, ON, Canada
| | - Jaimarie Sostre-Colón
- Abramson Family Cancer Research Institute, Department of Pathology & Laboratory Medicine, Penn Sarcoma Program, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Feven Tameire
- Department of Radiation Oncology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Nektaria Maria Leli
- Department of Radiation Oncology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Constantinos Koumenis
- Department of Radiation Oncology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Donita C Brady
- Abramson Family Cancer Research Institute, Department of Pathology & Laboratory Medicine, Penn Sarcoma Program, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA.,Department of Cancer Biology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Anthony Mancuso
- Department of Radiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Kristy Weber
- Department of Orthopedic Surgery, Penn Sarcoma Program, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Rebecca Gladdy
- Department of Surgery and Institute of Medical Science, University of Toronto, Toronto, ON, Canada.,Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, ON, Canada
| | - Jun Qi
- Department of Medicine, Harvard Medical School, Boston, MA, 02115, USA
| | - T S Karin Eisinger-Mathason
- Abramson Family Cancer Research Institute, Department of Pathology & Laboratory Medicine, Penn Sarcoma Program, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA.
| |
Collapse
|
1020
|
Rogers EH, Hunt JA, Pekovic-Vaughan V. Adult stem cell maintenance and tissue regeneration around the clock: do impaired stem cell clocks drive age-associated tissue degeneration? Biogerontology 2018; 19:497-517. [PMID: 30374678 PMCID: PMC6223734 DOI: 10.1007/s10522-018-9772-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2018] [Accepted: 09/19/2018] [Indexed: 02/06/2023]
Abstract
Human adult stem cell research is a highly prolific area in modern tissue engineering as these cells have significant potential to provide future cellular therapies for the world's increasingly aged population. Cellular therapies require a smart biomaterial to deliver and localise the cell population; protecting and guiding the stem cells toward predetermined lineage-specific pathways. The cells, in turn, can provide protection to biomaterials and increase its longevity. The right combination of stem cells and biomaterials can significantly increase the therapeutic efficacy. Adult stem cells are utilised to target many changes that negatively impact tissue functions with age. Understanding the underlying mechanisms that lead to changes brought about by the ageing process is imperative as ageing leads to many detrimental effects on stem cell activation, maintenance and differentiation. The circadian clock is an evolutionarily conserved timing mechanism that coordinates physiology, metabolism and behavior with the 24 h solar day to provide temporal tissue homeostasis with the external environment. Circadian rhythms deteriorate with age at both the behavioural and molecular levels, leading to age-associated changes in downstream rhythmic tissue physiology in humans and rodent models. In this review, we highlight recent advances in our knowledge of the role of circadian clocks in adult stem cell maintenance, driven by both cell-autonomous and tissue-specific factors, and the mechanisms by which they co-opt various cellular signaling pathways to impose temporal control on stem cell function. Future research investigating pharmacological and lifestyle interventions by which circadian rhythms within adult stem niches can be manipulated will provide avenues for temporally guided cellular therapies and smart biomaterials to ameliorate age-related tissue deterioration and reduce the burden of chronic disease.
Collapse
Affiliation(s)
- Eve H Rogers
- Institute of Ageing and Chronic Disease, University of Liverpool, The William Henry Duncan Building, 6 West Derby Street, Liverpool, L7 8TX, UK
| | - John A Hunt
- School of Science and Technology, Nottingham Trent University, Clifton Campus, College Drive, Nottingham, NG11 8NS, UK
| | - Vanja Pekovic-Vaughan
- Institute of Ageing and Chronic Disease, University of Liverpool, The William Henry Duncan Building, 6 West Derby Street, Liverpool, L7 8TX, UK.
| |
Collapse
|
1021
|
Elkhenany H, AlOkda A, El-Badawy A, El-Badri N. Tissue regeneration: Impact of sleep on stem cell regenerative capacity. Life Sci 2018; 214:51-61. [PMID: 30393021 DOI: 10.1016/j.lfs.2018.10.057] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2018] [Revised: 10/23/2018] [Accepted: 10/25/2018] [Indexed: 12/31/2022]
Abstract
The circadian rhythm orchestrates many cellular functions, such as cell division, cell migration, metabolism and numerous intracellular biological processes. The physiological changes during sleep are believed to promote a suitable microenvironment for stem cells to proliferate, migrate and differentiate. These effects are mediated either directly by circadian clock genes or indirectly via hormones and cytokines. Hormones, such as melatonin and cortisol, are secreted in response to neural optic signals and act in harmony to regulate many biological functions during sleep. Herein, we correlate the effects of the main circadian genes on the expression of certain stem cell genes responsible for the regeneration of different tissues, including bone, cartilage, skin, and intestine. We also review the effects of different hormones and cytokines on stem cell activation or suppression and their relationship to the day/night cycle. The correlation of circadian rhythm with tissue regeneration could have implications in understanding the biology of sleep and tissue regeneration and in enhancing the efficacy and timing of surgical procedures.
Collapse
Affiliation(s)
- Hoda Elkhenany
- Centre of Excellence for Stem Cells and Regenerative Medicine (CESC), Zewail City of Science and Technology, 12588, Egypt; Department of Surgery, Faculty of Veterinary Medicine, Alexandria University, 22785, Egypt
| | - Abdelrahman AlOkda
- Centre of Excellence for Stem Cells and Regenerative Medicine (CESC), Zewail City of Science and Technology, 12588, Egypt
| | - Ahmed El-Badawy
- Centre of Excellence for Stem Cells and Regenerative Medicine (CESC), Zewail City of Science and Technology, 12588, Egypt
| | - Nagwa El-Badri
- Centre of Excellence for Stem Cells and Regenerative Medicine (CESC), Zewail City of Science and Technology, 12588, Egypt.
| |
Collapse
|
1022
|
Kang JS, Suh YJ, Moon KH, Park JS, Roh TH, Park MH, Ryu DJ. Clinical efficiency of bone marrow mesenchymal stem cell implantation for osteonecrosis of the femoral head: a matched pair control study with simple core decompression. Stem Cell Res Ther 2018; 9:274. [PMID: 30359323 PMCID: PMC6202854 DOI: 10.1186/s13287-018-1030-y] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2018] [Revised: 08/31/2018] [Accepted: 09/30/2018] [Indexed: 02/03/2023] Open
Abstract
Background To date, several trials have reported the use of mesenchymal stem cell (MSC) implantation for osteonecrosis of the femoral head (ONFH). However, the clinical outcomes have not been conclusive. This study compared the clinical and radiological results of bone marrow mesenchymal stem cell (BMMSC) implantation with traditional simple core decompression (CD) using a matched pair case–control design. Methods We retrospectively reviewed 100 patients with ONFH (106 hips) who had been treated by CD alone (50 patients, 53 hips) and CD + BMMSC implantation (50 patients, 53 hips) between February 2004 and October 2014. We assessed the total hip replacement arthroplasty (THA) conversion rate and ARCO (Association Research Circulation Osseous) stage progression. Survivor rate analysis was performed using the Kaplan–Meier method, and an additional THA was defined as the primary endpoints. Results The mean follow-up period was 4.28 years. There was a difference in the THA conversion rate between the CD (49%) and CD + BMMSC groups (28.3%) (p = 0.028). ARCO stage progression was noted in 20 of 53 hips (37.7%) in the CD group and 19 of 53 hips (35.8%) in the CD + BMMSC group. Among collapsed cases (ARCO stages III and IV), there was no difference in clinical failure rate between the two groups. Conversely, in the pre-collapse cases (ARCO stages I and II), only 6 of 30 hips (20%) progressed to clinical failure in the CD + BMMSC group, whereas 15 of 30 hips (50%) progressed to clinical failure in the CD group (p = 0.014). Kaplan–Meier survival analysis showed a significant difference in the time to failure between the two groups up to 10-year follow-up (log-rank test p = 0.031). There was no significant difference in terms of age (p = 0.87) and gender (p = 0.51) when comparing THA conversion rates between groups. No complication was noted. Conclusions These results suggest that implantation of MSCs into the femoral head at an early stage of ONFH lowers the THA conversion rate. However, ARCO stage progression is not affected by this treatment. Trial registration Retrospectively registered
Collapse
Affiliation(s)
- Joon Soon Kang
- Department of Orthopedic Surgery, College of Medicine, Inha University Hospital, 7-206, 3rd Street Sinheung-Dong, Jung-Gu, Incheon, 400-103, South Korea
| | - Young Ju Suh
- Department of Biomedical Sciences, College of Medicine, Inha University, Incheon, South Korea
| | - Kyoung Ho Moon
- Department of Orthopedic Surgery, College of Medicine, Inha University Hospital, 7-206, 3rd Street Sinheung-Dong, Jung-Gu, Incheon, 400-103, South Korea
| | - Jun Sung Park
- Department of Orthopedic Surgery, College of Medicine, Inha University Hospital, 7-206, 3rd Street Sinheung-Dong, Jung-Gu, Incheon, 400-103, South Korea
| | - Tae Hoon Roh
- Department of Orthopedic Surgery, College of Medicine, Inha University Hospital, 7-206, 3rd Street Sinheung-Dong, Jung-Gu, Incheon, 400-103, South Korea
| | - Myung Hoon Park
- Department of Orthopedic Surgery, College of Medicine, Inha University Hospital, 7-206, 3rd Street Sinheung-Dong, Jung-Gu, Incheon, 400-103, South Korea
| | - Dong Jin Ryu
- Department of Orthopedic Surgery, College of Medicine, Inha University Hospital, 7-206, 3rd Street Sinheung-Dong, Jung-Gu, Incheon, 400-103, South Korea.
| |
Collapse
|
1023
|
Li R, Lin QX, Liang XZ, Liu GB, Tang H, Wang Y, Lu SB, Peng J. Stem cell therapy for treating osteonecrosis of the femoral head: From clinical applications to related basic research. Stem Cell Res Ther 2018; 9:291. [PMID: 30359305 PMCID: PMC6202807 DOI: 10.1186/s13287-018-1018-7] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Osteonecrosis of the femoral head (ONFH) is a refractory disease that is associated with collapse of the femoral head, with a risk of hip arthroplasty in younger populations. Thus, there has been an increased focus on early interventions for ONFH that aim to preserve the native articulation. Stem cell therapy is a promising treatment, and an increasing number of recent studies have focused on this topic. Many clinical studies have reported positive outcomes of stem cell therapy for the treatment of ONFH. To improve the therapeutic effects of this approach, many related basic research studies have also been performed. However, some issues must be further explored, such as the appropriate patient selection procedure, the optimal stem cell selection protocol, the ideal injection number, and the safety of stem cell therapy. The purpose of this review is to summarize the available clinical studies and basic research related to stem cell therapy for ONFH.
Collapse
Affiliation(s)
- Rui Li
- Institute of Orthopedics, Beijing Key Laboratory of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries, Chinese PLA General Hospital, Beijing, 100853 China
| | - Qiu-Xia Lin
- Institute of Orthopedics, Beijing Key Laboratory of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries, Chinese PLA General Hospital, Beijing, 100853 China
| | - Xue-Zhen Liang
- The First Clinical Medical School, Shandong University of Traditional Chinese Medicine, Jinan, 250355 Shandong China
| | - Guang-Bo Liu
- Institute of Orthopedics, Beijing Key Laboratory of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries, Chinese PLA General Hospital, Beijing, 100853 China
| | - He Tang
- Institute of Orthopedics, Beijing Key Laboratory of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries, Chinese PLA General Hospital, Beijing, 100853 China
| | - Yu Wang
- Institute of Orthopedics, Beijing Key Laboratory of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries, Chinese PLA General Hospital, Beijing, 100853 China
| | - Shi-Bi Lu
- Institute of Orthopedics, Beijing Key Laboratory of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries, Chinese PLA General Hospital, Beijing, 100853 China
| | - Jiang Peng
- Institute of Orthopedics, Beijing Key Laboratory of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries, Chinese PLA General Hospital, Beijing, 100853 China
| |
Collapse
|
1024
|
Navet B, Ando K, Vargas-Franco JW, Brion R, Amiaud J, Mori K, Yagita H, Mueller CG, Verrecchia F, Dumars C, Heymann MF, Heymann D, Lézot F. The Intrinsic and Extrinsic Implications of RANKL/RANK Signaling in Osteosarcoma: From Tumor Initiation to Lung Metastases. Cancers (Basel) 2018; 10:cancers10110398. [PMID: 30355966 PMCID: PMC6265790 DOI: 10.3390/cancers10110398] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2018] [Revised: 10/08/2018] [Accepted: 10/19/2018] [Indexed: 12/24/2022] Open
Abstract
Background: Osteosarcoma is the most frequent form of malignant pediatric bone tumor. Despite the current therapeutic arsenal, patient life-expectancy remains low if metastases are detected at the time of diagnosis, justifying research into better knowledge at all stages of osteosarcoma ontogenesis and identification of new therapeutic targets. Receptor Activator of Nuclear factor κB (RANK)expression has been reported in osteosarcoma cells, raising the question of Receptor Activator of Nuclear factor κB Ligand (RANKL)/RANK signaling implications in these tumor cells (intrinsic), in addition to previously reported implications through osteoclast activation in the tumor microenvironment (extrinsic). Methods: Based on in vitro and in vivo experimentations using human and mouse osteosarcoma cell lines, the consequences on the main cellular processes of RANK expression in osteosarcoma cells were analyzed. Results: The results revealed that RANK expression had no impact on cell proliferation and tumor growth, but stimulated cellular differentiation and, in an immune-compromised environment, increased the number of lung metastases. The analysis of RANKL, RANK and osteoprotegerin (OPG) expressions in biopsies of a cohort of patients revealed that while RANK expression in osteosarcoma cells was not significantly different between patients with or without metastases at the time of diagnosis, the OPG/RANK ratio decreased significantly. Conclusion: Altogether, these results are in favor of RANKL-RANK signaling inhibition as an adjuvant for the treatment of osteosarcoma.
Collapse
Affiliation(s)
- Benjamin Navet
- INSERM, UMR 1238, Faculté de Médecine, Université de Nantes, F-44035 Nantes, France.
| | - Kosei Ando
- INSERM, UMR 1238, Faculté de Médecine, Université de Nantes, F-44035 Nantes, France.
- Department of Orthopedic Surgery, Shiga University of Medical Science, Tsukinowa-cho, Seta, Otsu, Shiga 520-2192, Japan.
| | - Jorge William Vargas-Franco
- INSERM, UMR 1238, Faculté de Médecine, Université de Nantes, F-44035 Nantes, France.
- Department of Basic Studies, Faculty of Odontology, University of Antioquia, Medellin AA 1226, Colombia.
| | - Régis Brion
- INSERM, UMR 1238, Faculté de Médecine, Université de Nantes, F-44035 Nantes, France.
- Centre Hospitalier Universitaire, Hôtel Dieu, F-44035 Nantes, France.
| | - Jérome Amiaud
- INSERM, UMR 1238, Faculté de Médecine, Université de Nantes, F-44035 Nantes, France.
| | - Kanji Mori
- Department of Orthopedic Surgery, Shiga University of Medical Science, Tsukinowa-cho, Seta, Otsu, Shiga 520-2192, Japan.
| | - Hideo Yagita
- Department of Immunology, Juntendo University School of Medicine, Tokyo 113-8421, Japan.
| | - Christopher G Mueller
- CNRS, UPR 9021, Institut de Biologie Moléculaire et Cellulaire (IBMC), Laboratoire Immunologie et Chimie Thérapeutiques, Université de Strasbourg, F-67084 Strasbourg, France.
| | - Franck Verrecchia
- INSERM, UMR 1238, Faculté de Médecine, Université de Nantes, F-44035 Nantes, France.
| | - Clotilde Dumars
- INSERM, UMR 1238, Faculté de Médecine, Université de Nantes, F-44035 Nantes, France.
- Centre Hospitalier Universitaire, Hôtel Dieu, F-44035 Nantes, France.
| | - Marie-Françoise Heymann
- INSERM, LEA Sarcoma Research Unit, Department of Oncology and Human Metabolism, Medical School, University of Sheffield, Sheffield S10 2RX, UK.
- INSERM, UMR 1232, LabCT, Université de Nantes, Institut de Cancérologie de l'Ouest, site René Gauducheau, F-44805 Saint-Herblain, France.
| | - Dominique Heymann
- INSERM, LEA Sarcoma Research Unit, Department of Oncology and Human Metabolism, Medical School, University of Sheffield, Sheffield S10 2RX, UK.
- INSERM, UMR 1232, LabCT, Université de Nantes, Institut de Cancérologie de l'Ouest, site René Gauducheau, F-44805 Saint-Herblain, France.
| | - Frédéric Lézot
- INSERM, UMR 1238, Faculté de Médecine, Université de Nantes, F-44035 Nantes, France.
| |
Collapse
|
1025
|
Wang T, Azeddine B, Mah W, Harvey EJ, Rosenblatt D, Séguin C. Osteonecrosis of the femoral head: genetic basis. INTERNATIONAL ORTHOPAEDICS 2018; 43:519-530. [PMID: 30328481 DOI: 10.1007/s00264-018-4172-8] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/07/2018] [Accepted: 09/18/2018] [Indexed: 12/19/2022]
Abstract
PURPOSE Genetic factors and hereditary forms of osteonecrosis of the femoral head (ONFH) have been elucidated through genetic association studies. The significance of these cases is that they suggest an alternative hypothesis to the development of the disease. This review presents a summary of single nucleotide polymorphisms (SNPs) and other genetic mutation variations found in association with ONFH, including our recent identification of a novel mutation in the transient receptor potential vanilloid 4 (TRPV4) gene in association with inherited ONFH. The purpose of this review is to consolidate and categorize genetic linkages according to physiological pathways. METHODS A systematic review of literature from PubMed and Google Scholar was undertaken with a focus on genetic linkages and hereditary case studies of the disease. Recent genetic analysis studies published after 2007 were the focus of genetic linkages in non-hereditary cases. RESULTS The summary of these genetic findings identifies biological processes believed to be involved in the development of ONFH, which include circulation, steroid metabolism, immunity, and the regulation of bone formation. CONCLUSION Taken together, these associations may lead to new pathways of bone repair and remodeling while opening new avenues for therapeutic targets. Knowledge of genetic variations could help identify individuals considered to be at higher risk of developing ONFH and prevent the multiple hit effect.
Collapse
Affiliation(s)
- Tracy Wang
- Vascular Biology Research lab, Research Institute (RI) McGill University Health Centre, C9 Montreal General Hospital, 1650 Cedar Avenue, Montreal, QC, H3G 1A4, Canada.
| | - Bouziane Azeddine
- Vascular Biology Research lab, Research Institute (RI) McGill University Health Centre, C9 Montreal General Hospital, 1650 Cedar Avenue, Montreal, QC, H3G 1A4, Canada
| | - Wayne Mah
- Vascular Biology Research lab, Research Institute (RI) McGill University Health Centre, C9 Montreal General Hospital, 1650 Cedar Avenue, Montreal, QC, H3G 1A4, Canada
| | - Edward J Harvey
- Department Surgery, Division Orthopaedic Surgery, McGill University Health Centre, B5 Montreal General Hospital, 1650 Cedar Avenue, Montreal, QC, H3G 1A4, Canada
| | - David Rosenblatt
- Department of Human Genetics, McGill University, Montreal, Quebec, Canada
| | - Chantal Séguin
- Vascular Biology Research lab, Research Institute (RI) McGill University Health Centre, C9 Montreal General Hospital, 1650 Cedar Avenue, Montreal, QC, H3G 1A4, Canada. .,Department of Medicine, Division of Hematology and Oncology, McGill University Health Centre, Montreal, Quebec, H4A 3J1, Canada. .,Glen Site, Cedars Cancer Centre, McGill University Health Centre, 1001 Décarie Blvd., room D02.7519, Montreal, Quebec, H4A 3J1, Canada.
| |
Collapse
|
1026
|
Yan S, Ren J, Jian Y, Wang W, Yun W, Yin J. Injectable Maltodextrin-Based Micelle/Hydrogel Composites for Simvastatin-Controlled Release. Biomacromolecules 2018; 19:4554-4564. [DOI: 10.1021/acs.biomac.8b01234] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Shifeng Yan
- Department of Polymer Materials, Shanghai University, 99 Shangda Road, Shanghai 200444, People’s Republic of China
| | - Jie Ren
- Department of Polymer Materials, Shanghai University, 99 Shangda Road, Shanghai 200444, People’s Republic of China
| | - Yuhang Jian
- Department of Polymer Materials, Shanghai University, 99 Shangda Road, Shanghai 200444, People’s Republic of China
| | - Weidong Wang
- Department of Polymer Materials, Shanghai University, 99 Shangda Road, Shanghai 200444, People’s Republic of China
| | - Wentao Yun
- Department of Polymer Materials, Shanghai University, 99 Shangda Road, Shanghai 200444, People’s Republic of China
| | - Jingbo Yin
- Department of Polymer Materials, Shanghai University, 99 Shangda Road, Shanghai 200444, People’s Republic of China
| |
Collapse
|
1027
|
Genetic Markers Can Predict Chondrogenic Differentiation Potential in Bone Marrow-Derived Mesenchymal Stromal Cells. Stem Cells Int 2018; 2018:9530932. [PMID: 30405725 PMCID: PMC6199884 DOI: 10.1155/2018/9530932] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2018] [Accepted: 08/27/2018] [Indexed: 02/06/2023] Open
Abstract
The precise predictions of the differentiation direction and potential of mesenchymal stromal cells (MSCs) are an important key to the success of regenerative medicine. The expression levels of fate-determining genes may provide tools for predicting differentiation potential. The expression levels of 95 candidate marker genes and glycosaminoglycan (GAG) contents after chondrogenic induction in 10 undifferentiated ilium and 5 jaw MSC cultures were determined, and their correlations were analyzed. The expression levels of eight genes before the induction of chondrogenic MSC differentiation were significantly correlated with the GAG levels after induction. Based on correlation patterns, the eight genes were classified into two groups: group 1 genes (AURKB, E2F1, CDKN2D, LIF, and ACLY), related to cell cycle regulation, and group 2 genes (CD74, EFEMP1, and TGM2), involved in chondrogenesis. The expression levels of the group 2 genes were significantly correlated with the ages of the cell donors. The expression levels of CDKN2D, CD74, and TGM2 were >10-fold higher in highly potent MSCs (ilium MSCs) than in MSCs with limited potential (jaw MSCs). Three-dimensional (3D) scatter plot analyses of the expression levels of these genes showed reduced variability between donors and confirmed predictive potential. These data suggest that group 2 genes are involved in age-dependent decreases in the chondrogenic differentiation potential of MSCs, and combined 3D analyses of the expression profiles of three genes, including two group 2 genes, were predictive of MSC differentiation potential.
Collapse
|
1028
|
Betker JL, Jones D, Childs CR, Helm KM, Terrell K, Nagel MA, Anchordoquy TJ. Nanoparticle uptake by circulating leukocytes: A major barrier to tumor delivery. J Control Release 2018; 286:85-93. [PMID: 30030182 PMCID: PMC6936323 DOI: 10.1016/j.jconrel.2018.07.031] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2018] [Revised: 07/05/2018] [Accepted: 07/16/2018] [Indexed: 12/18/2022]
Abstract
Decades of research into improving drug delivery to tumors has documented uptake of particulate delivery systems by resident macrophages in the lung, liver, and spleen, and correlated short circulation times with reduced tumor accumulation. An implicit assumption in these studies is that nanoparticles present in the blood are available for distribution to the tumor. This study documents significant levels of lipoplex uptake by circulating leukocytes, and its effect on distribution to the tumor and other organs. In agreement with previous studies, PEGylation dramatically extends circulation times and enhances tumor delivery. However, our studies suggest that this relationship is not straightforward, and that particle sequestration by leukocytes can significantly alter biodistribution, especially with non-PEGylated nanoparticle formulations. We conclude that leukocyte uptake should be considered in biodistribution studies, and that delivery to these circulating cells may present opportunities for treating viral infections and leukemia.
Collapse
Affiliation(s)
- Jamie L Betker
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Dallas Jones
- Department of Neurology, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Christine R Childs
- Flow Cytometry Core Facility, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Karen M Helm
- Flow Cytometry Core Facility, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Kristina Terrell
- Flow Cytometry Core Facility, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Maria A Nagel
- Department of Neurology, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Thomas J Anchordoquy
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, United States.
| |
Collapse
|
1029
|
Insights into the Role of Circadian Rhythms in Bone Metabolism: A Promising Intervention Target? BIOMED RESEARCH INTERNATIONAL 2018; 2018:9156478. [PMID: 30363685 PMCID: PMC6180976 DOI: 10.1155/2018/9156478] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/28/2018] [Accepted: 09/09/2018] [Indexed: 11/18/2022]
Abstract
Numerous physiological processes of mammals, including bone metabolism, are regulated by the circadian clock system, which consists of a central regulator, the suprachiasmatic nucleus (SCN), and the peripheral oscillators of the BMAL1/CLOCK-PERs/CRYs system. Various bone turnover markers and bone metabolism-regulating hormones such as melatonin and parathyroid hormone (PTH) display diurnal rhythmicity. According to previous research, disruption of the circadian clock due to shift work, sleep restriction, or clock gene knockout is associated with osteoporosis or other abnormal bone metabolism, showing the importance of the circadian clock system for maintaining homeostasis of bone metabolism. Moreover, common causes of osteoporosis, including postmenopausal status and aging, are associated with changes in the circadian clock. In our previous research, we found that agonism of the circadian regulators REV-ERBs inhibits osteoclast differentiation and ameliorates ovariectomy-induced bone loss in mice, suggesting that clock genes may be promising intervention targets for abnormal bone metabolism. Moreover, osteoporosis interventions at different time points can provide varying degrees of bone protection, showing the importance of accounting for circadian rhythms for optimal curative effects in clinical treatment of osteoporosis. In this review, we summarize current knowledge about circadian rhythms and bone metabolism.
Collapse
|
1030
|
Three-dimensional nanofibrous microenvironment designed for the regulation of mesenchymal stem cells. APPLIED NANOSCIENCE 2018. [DOI: 10.1007/s13204-018-0877-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
|
1031
|
Nanomedicines for developing cancer nanotherapeutics: from benchtop to bedside and beyond. Appl Microbiol Biotechnol 2018; 102:9449-9470. [DOI: 10.1007/s00253-018-9352-3] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Revised: 08/29/2018] [Accepted: 08/29/2018] [Indexed: 12/21/2022]
|
1032
|
Noguchi T, Hussein AI, Horowitz N, Carroll D, Gower AC, Demissie S, Gerstenfeld LC. Hypophosphatemia Regulates Molecular Mechanisms of Circadian Rhythm. Sci Rep 2018; 8:13756. [PMID: 30213970 PMCID: PMC6137060 DOI: 10.1038/s41598-018-31830-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2018] [Accepted: 08/13/2018] [Indexed: 12/31/2022] Open
Abstract
Transcriptomic analysis showed that the central circadian pathway genes had significantly altered expression in fracture calluses from mice fed a low phosphate diet. This led us to hypothesize that phosphate deficiency altered the circadian cycle in peripheral tissues. Analysis of the expression of the central clock genes over a 24-36 hour period in multiple peripheral tissues including fracture callus, proximal tibia growth plate and cardiac tissues after 12 days on a low phosphate diet showed higher levels of gene expression in the hypophosphatemia groups (p < 0.001) and a 3 to 6 hour elongation of the circadian cycle. A comparative analysis of the callus tissue transcriptome genes that were differentially regulated by hypophosphatemia with published data for the genes in bone that are diurnally regulated identified 1879 genes with overlapping differential regulation, which were shown by ontology assessment to be associated with oxidative metabolism and apoptosis. Network analysis of the central circadian pathway genes linked their expression to the up regulated expression of the histone methyltransferase gene EZH2, a gene that when mutated in both humans and mice controls overall skeletal growth. These data suggest that phosphate is an essential metabolite that controls circadian function in both skeletal and non skeletal peripheral tissues and associates its levels with the overall oxidative metabolism and skeletal growth of animals.
Collapse
Affiliation(s)
- Takashi Noguchi
- Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, Boston University School of Medicine, Boston, USA
| | - Amira I Hussein
- Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, Boston University School of Medicine, Boston, USA
| | - Nina Horowitz
- Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, Boston University School of Medicine, Boston, USA
| | - Deven Carroll
- Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, Boston University School of Medicine, Boston, USA
| | - Adam C Gower
- Clinical and Translational Science Institute, Boston University School of Medicine, Boston, USA
| | - Serkalem Demissie
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, 02118, USA
| | - Louis C Gerstenfeld
- Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, Boston University School of Medicine, Boston, USA.
| |
Collapse
|
1033
|
Wang T, Hu Q, Lee JY, Luo Y. Solid Lipid-Polymer Hybrid Nanoparticles by In Situ Conjugation for Oral Delivery of Astaxanthin. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2018; 66:9473-9480. [PMID: 30130387 DOI: 10.1021/acs.jafc.8b02827] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/27/2023]
Abstract
Solid lipid-polymer hybrid nanoparticles (SLPN) are nanocarriers made from a combination of polymers and lipids, integrating the advantages of biocompatible lipid-based nanoparticles and gastrointestinal (GI)-stable polymeric nanoparticles. In this study, a novel preparation strategy was proposed to fabricate GI-stable SLPN through in situ conjugation between oxidized dextran and bovine serum albumin. Effects of molecular weight of dextran (20, 40, 75, and 150 kDa), conjugation temperature (65 °C, 75 °C, and 85 °C), and time (30, 60, 120 min) on the particulate characteristics and stability were comprehensively investigated and optimized. As heating temperature increased from 65 °C to 75 °C, the particle size of SLPN increased from 139 to 180 nm with narrow size distribution, but when the temperature reached 85 °C severe aggregation was observed after 60 min. SLPN prepared with 40 kDa oxidized dextran under 85 °C/30 min heating condition exhibited excellent GI stability with no significant changes in particle size and PDI after incubation in simulated GI fluids. The prepared SLPN were then used to encapsulate astaxanthin, a lipophilic bioactive compound, studied as a model nutrient. After encapsulation in SLPN, antioxidant activity of astaxanthin was dramatically enhanced in aqueous condition and a sustained release was achieved in simulated GI fluids. Therefore, the SLPN developed in this study are a promising oral delivery system for lipophilic compounds, such as astaxanthin.
Collapse
Affiliation(s)
- Taoran Wang
- Department of Nutritional Sciences , University of Connecticut , Storrs , Connecticut 06269 , United States
| | - Qiaobin Hu
- Department of Nutritional Sciences , University of Connecticut , Storrs , Connecticut 06269 , United States
| | - Ji-Young Lee
- Department of Nutritional Sciences , University of Connecticut , Storrs , Connecticut 06269 , United States
| | - Yangchao Luo
- Department of Nutritional Sciences , University of Connecticut , Storrs , Connecticut 06269 , United States
| |
Collapse
|
1034
|
Wang A, Ren M, Wang J. The pathogenesis of steroid-induced osteonecrosis of the femoral head: A systematic review of the literature. Gene 2018; 671:103-109. [DOI: 10.1016/j.gene.2018.05.091] [Citation(s) in RCA: 77] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2017] [Revised: 05/14/2018] [Accepted: 05/23/2018] [Indexed: 12/16/2022]
|
1035
|
Rodríguez-Nogales C, González-Fernández Y, Aldaz A, Couvreur P, Blanco-Prieto MJ. Nanomedicines for Pediatric Cancers. ACS NANO 2018; 12:7482-7496. [PMID: 30071163 DOI: 10.1021/acsnano.8b03684] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/14/2023]
Abstract
Chemotherapy protocols for childhood cancers are still problematic due to the high toxicity associated with chemotherapeutic agents and incorrect dosing regimens extrapolated from adults. Nanotechnology has demonstrated significant ability to reduce toxicity of anticancer compounds. Improvement in the therapeutic index of cytostatic drugs makes this strategy an alternative to common chemotherapy in adults. However, the lack of nanomedicines specifically for pediatric cancer care raises a medical conundrum. This review highlights the current state and progress of nanomedicine in pediatric cancer and discusses the real clinical challenges and opportunities.
Collapse
Affiliation(s)
- Carlos Rodríguez-Nogales
- Pharmacy and Pharmaceutical Technology Department , University of Navarra , Pamplona 31008 , Spain
- Instituto de Investigación Sanitaria de Navarra (IdiSNA) , Pamplona 31008 , Spain
| | | | - Azucena Aldaz
- Department of Pharmacy , Clínica Universidad de Navarra , Pamplona 31008 , Spain
| | - Patrick Couvreur
- Institut Galien Paris-Sud, UMR CNRS 8612, Université Paris-Sud, Université Paris-Saclay, Châtenay-Malabry Cedex 92296 , France
| | - María J Blanco-Prieto
- Pharmacy and Pharmaceutical Technology Department , University of Navarra , Pamplona 31008 , Spain
- Instituto de Investigación Sanitaria de Navarra (IdiSNA) , Pamplona 31008 , Spain
| |
Collapse
|
1036
|
Liang X, Duan P, Gao J, Guo R, Qu Z, Li X, He Y, Yao H, Ding J. Bilayered PLGA/PLGA-HAp Composite Scaffold for Osteochondral Tissue Engineering and Tissue Regeneration. ACS Biomater Sci Eng 2018; 4:3506-3521. [PMID: 33465902 DOI: 10.1021/acsbiomaterials.8b00552] [Citation(s) in RCA: 77] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Affiliation(s)
- Xiangyu Liang
- State Key Laboratory of Molecular Engineering of Polymers, Department of Macromolecular Science, Fudan University, Shanghai 200438, China
| | - Pingguo Duan
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Nanchang University, Nanchang 330006, China
| | - Jingming Gao
- State Key Laboratory of Molecular Engineering of Polymers, Department of Macromolecular Science, Fudan University, Shanghai 200438, China
| | - Runsheng Guo
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Nanchang University, Nanchang 330006, China
| | - Zehua Qu
- State Key Laboratory of Molecular Engineering of Polymers, Department of Macromolecular Science, Fudan University, Shanghai 200438, China
| | - Xiaofeng Li
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Nanchang University, Nanchang 330006, China
| | - Yao He
- State Key Laboratory of Molecular Engineering of Polymers, Department of Macromolecular Science, Fudan University, Shanghai 200438, China
| | - Haoqun Yao
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Nanchang University, Nanchang 330006, China
| | - Jiandong Ding
- State Key Laboratory of Molecular Engineering of Polymers, Department of Macromolecular Science, Fudan University, Shanghai 200438, China
| |
Collapse
|
1037
|
Cao W, Peng Y, Zhang Y, Qiu F, Li M, Tang J, Wu Z. Novel bone wax based on tricalcium silicate cement and BGs mixtures. Biomed Mater 2018; 13:065001. [DOI: 10.1088/1748-605x/aad73c] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
|
1038
|
Takahashi T, Sato M, Toyoda E, Maehara M, Takizawa D, Maruki H, Tominaga A, Okada E, Okazaki K, Watanabe M. Rabbit xenogeneic transplantation model for evaluating human chondrocyte sheets used in articular cartilage repair. J Tissue Eng Regen Med 2018; 12:2067-2076. [PMID: 30058138 PMCID: PMC6221121 DOI: 10.1002/term.2741] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Revised: 05/27/2018] [Accepted: 07/09/2018] [Indexed: 12/18/2022]
Abstract
Research on cartilage regeneration has developed novel sources for human chondrocytes and new regenerative therapies, but appropriate animal models for translational research are needed. Although rabbit models are frequently used in such studies, the availability of immunocompromised rabbits is limited. Here, we investigated the usefulness of an immunosuppressed rabbit model to evaluate directly the efficacy of human chondrocyte sheets through xenogeneic transplantation. Human chondrocyte sheets were transplanted into knee osteochondral defects in Japanese white rabbits administered with immunosuppressant tacrolimus at a dosage of 0.8 or 1.6 mg/kg/day for 4 weeks. Histological evaluation at 4 weeks after transplantation in rabbits administered 1.6 mg/kg/day showed successful engraftment of human chondrocytes and cartilage regeneration involving a mixture of hyaline cartilage and fibrocartilage. No human chondrocytes were detected in rabbits administered 0.8 mg/kg/day, although regeneration of hyaline cartilage was confirmed. Histological evaluation at 12 weeks after transplantation (i.e., 8 weeks after termination of immunosuppression) showed strong immune rejection of human chondrocytes, which indicated that, even after engraftment, articular cartilage is not particularly immune privileged in xenogeneic transplantation. Our results suggest that Japanese white rabbits administered tacrolimus at 1.6 mg/kg/day and evaluated at 4 weeks may be useful as a preclinical model for the direct evaluation of human cell‐based therapies.
Collapse
Affiliation(s)
- Takumi Takahashi
- Department of Orthopaedic Surgery, Surgical Science, Tokai University School of Medicine, Isehara, Japan
| | - Masato Sato
- Department of Orthopaedic Surgery, Surgical Science, Tokai University School of Medicine, Isehara, Japan
| | - Eriko Toyoda
- Department of Orthopaedic Surgery, Surgical Science, Tokai University School of Medicine, Isehara, Japan
| | - Miki Maehara
- Department of Orthopaedic Surgery, Surgical Science, Tokai University School of Medicine, Isehara, Japan
| | - Daichi Takizawa
- Department of Orthopaedic Surgery, Surgical Science, Tokai University School of Medicine, Isehara, Japan
| | - Hideyuki Maruki
- Department of Orthopaedic Surgery, Tokyo Women's University, Tokyo, Japan
| | - Ayako Tominaga
- Department of Orthopaedic Surgery, Tokyo Women's University, Tokyo, Japan
| | - Eri Okada
- Department of Orthopaedic Surgery, Surgical Science, Tokai University School of Medicine, Isehara, Japan
| | - Ken Okazaki
- Department of Orthopaedic Surgery, Tokyo Women's University, Tokyo, Japan
| | - Masahiko Watanabe
- Department of Orthopaedic Surgery, Surgical Science, Tokai University School of Medicine, Isehara, Japan
| |
Collapse
|
1039
|
Bone Wax in Neurosurgery: A Review. World Neurosurg 2018; 116:72-76. [DOI: 10.1016/j.wneu.2018.04.222] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2018] [Revised: 04/28/2018] [Accepted: 04/30/2018] [Indexed: 11/20/2022]
|
1040
|
Lüring C, Benignus C, Beckmann J. [Joint-preserving operative treatment of avascular necrosis of the femoral head]. DER ORTHOPADE 2018; 47:745-750. [PMID: 30046854 DOI: 10.1007/s00132-018-3607-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
The cause of avascular necrosis of the femoral head is multifactorial. Conservative treatment is only an option in the early stages. It is only symptomatic and not causative treatment. The implantation of an artificial hip joint should be postponed as the typically affected middle-aged males are right in the middle of their working life. Therefore, some joint-preserving operative therapies might be considered in stages ARCO I-III. Those range from core decompression to osteotomies and grafts, the advantages and disadvantages of which have to be weighted in each case. More recent therapies such as additive stem cells or platelet rich plasma (PRP) combined with core decompression have yet to prove their efficacy.
Collapse
Affiliation(s)
- C Lüring
- Orthopädische Klinik, Klinikum Dortmund gGmbH, Beurhausstraße 40, 44137, Dortmund, Deutschland.
| | - C Benignus
- Sportklinik Stuttgart GmbH, Taubenheimstr. 8, 70372, Stuttgart, Deutschland
| | - J Beckmann
- Sportklinik Stuttgart GmbH, Taubenheimstr. 8, 70372, Stuttgart, Deutschland
| |
Collapse
|
1041
|
Current Status of Canine Umbilical Cord Blood-Derived Mesenchymal Stem Cells in Veterinary Medicine. Stem Cells Int 2018; 2018:8329174. [PMID: 30123294 PMCID: PMC6079340 DOI: 10.1155/2018/8329174] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2018] [Accepted: 06/19/2018] [Indexed: 12/26/2022] Open
Abstract
Stem cell therapy has prompted the expansion of veterinary medicine both experimentally and clinically, with the potential to contribute to contemporary treatment strategies for various diseases and conditions for which limited or no therapeutic options are presently available. Although the application of various types of stem cells, such as bone marrow-derived mesenchymal stem cells (BM-MSCs), adipose tissue-derived mesenchymal stem cells (AT-MSCs), and umbilical cord blood-derived mesenchymal stem cells (UCB-MSCs), has promising potential to improve the health of different species, it is crucial that the benefits and drawbacks are completely evaluated before use. Umbilical cord blood (UCB) is a rich source of stem cells; nonetheless, isolation of mesenchymal stem cells (MSCs) from UCB presents technical challenges. Although MSCs have been isolated from UCB of diverse species such as human, equine, sheep, goat, and canine, there are inherent limitations of using UCB from these species for the expansion of MSCs. In this review, we investigated canine UCB (cUCB) and compared it with UCB from other species by reviewing recent articles published from February 2003 to June 2017 to gain an understanding of the limitations of cUCB in the acquisition of MSCs and to determine other suitable sources for the isolation of MSCs from canine. Our review indicates that cUCB is not an ideal source of MSCs because of insufficient volume and ethical issues. However, canine reproductive organs discarded during neutering may help broaden our understanding of effective isolation of MSCs. We recommend exploring canine reproductive and adipose tissue rather than UCB to fulfill the current need in veterinary medicine for the well-designed and ethically approved source of MSCs.
Collapse
|
1042
|
Ali A, Ahmed S. Recent Advances in Edible Polymer Based Hydrogels as a Sustainable Alternative to Conventional Polymers. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2018; 66:6940-6967. [PMID: 29878765 DOI: 10.1021/acs.jafc.8b01052] [Citation(s) in RCA: 144] [Impact Index Per Article: 20.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
The over increasing demand of eco-friendly materials to counter various problems, such as environmental issues, economics, sustainability, biodegradability, and biocompatibility, open up new fields of research highly focusing on nature-based products. Edible polymer based materials mainly consisting of polysaccharides, proteins, and lipids could be a prospective contender to handle such problems. Hydrogels based on edible polymer offer many valuable properties compared to their synthetic counterparts. Edible polymers can contribute to the reduction of environmental contamination, advance recyclability, provide sustainability, and thereby increase its applicability along with providing environmentally benign products. This review is highly emphasizing on toward the development of hydrogels from edible polymer, their classification, properties, chemical modification, and their potential applications. The application of edible polymer hydrogels covers many areas including the food industry, agricultural applications, drug delivery to tissue engineering in the biomedical field and provide more safe and attractive products in the pharmaceutical, agricultural, and environmental fields, etc.
Collapse
Affiliation(s)
- Akbar Ali
- Department of Chemistry , Jamia Millia Islamia , New Delhi , 110025 , India
| | - Shakeel Ahmed
- Department of Chemistry , Government Degree College Mendhar , Jammu , Jammu and Kashmir , 185211 , India
- Higher Education Department , Government of Jammu and Kashmir , Jammu , 180001 , India
| |
Collapse
|
1043
|
Tan B, Shi X, Zhang J, Qin J, Zhang N, Ren H, Qian M, Siwko S, Carmon K, Liu Q, Han H, Du B, Liu M. Inhibition of Rspo-Lgr4 Facilitates Checkpoint Blockade Therapy by Switching Macrophage Polarization. Cancer Res 2018; 78:4929-4942. [DOI: 10.1158/0008-5472.can-18-0152] [Citation(s) in RCA: 87] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2018] [Revised: 05/02/2018] [Accepted: 06/18/2018] [Indexed: 11/16/2022]
|
1044
|
Xu W, Wang Z, Liu Y, Wang L, Jiang Z, Li T, Zhang W, Liang Y. Carboxymethyl chitosan/gelatin/hyaluronic acid blended-membranes as epithelia transplanting scaffold for corneal wound healing. Carbohydr Polym 2018; 192:240-250. [DOI: 10.1016/j.carbpol.2018.03.033] [Citation(s) in RCA: 61] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2017] [Revised: 01/22/2018] [Accepted: 03/13/2018] [Indexed: 11/30/2022]
|
1045
|
Xu J, Gong H, Lu S, Deasey MJ, Cui Q. Animal models of steroid-induced osteonecrosis of the femoral head-a comprehensive research review up to 2018. INTERNATIONAL ORTHOPAEDICS 2018; 42:1729-1737. [PMID: 29705870 DOI: 10.1007/s00264-018-3956-1] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/25/2018] [Accepted: 04/17/2018] [Indexed: 01/11/2023]
Abstract
Osteonecrosis of the femoral head (ONFH) is a significant cause of both pain and disability that often affects young adults during what ought to be their most productive age. Two broad categories of ONFH exist: traumatic and non-traumatic. Traumatic ONFH results from acute mechanical disruption of the femoral head's blood supply. Many factors that increase the risk of non-traumatic osteonecrosis have been identified. Steroid-induced osteonecrosis of the femoral head (SONFH) is the most common form of non-traumatic ONFH. Many hypotheses as to the pathogenesis of SONFH have been proposed, including intravascular thrombosis, abnormal fat metabolism, intramedullary adipocyte hypertrophy, and osteoporosis; however, the exact mechanism of SONFH is still not clearly understood. Animal models using rats, mice, rabbits, chickens, pigs, and emus have been used to study SONFH. Unfortunately, these models each have limitations. Therefore, it is necessary to establish a reproducible model that better simulates human disease. The present review is intended to summarize the currently available models, evaluative indicators, and application of current understanding to both the prevention and treatment of SONFH.
Collapse
Affiliation(s)
- Jianzhong Xu
- Department of Orthopedic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450000, Henan, People's Republic of China
| | - Hanpu Gong
- Department of Orthopedic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450000, Henan, People's Republic of China
| | - Shitao Lu
- Department of Orthopedic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450000, Henan, People's Republic of China
| | - Matthey J Deasey
- Department of Orthopaedic Surgery, University of Virginia School of Medicine, 400 Ray C. Hunt Drive, Suite 330, Charlottesville, VA 22903, USA
| | - Quanjun Cui
- Department of Orthopedic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450000, Henan, People's Republic of China.
- Department of Orthopaedic Surgery, University of Virginia School of Medicine, 400 Ray C. Hunt Drive, Suite 330, Charlottesville, VA 22903, USA.
| |
Collapse
|
1046
|
Construction of lentiviral RNAi vector of PPARγ gene in cashmere goats and comparison with the transcriptome analysis of adipose cells treatments. Small Rumin Res 2018. [DOI: 10.1016/j.smallrumres.2018.04.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
|
1047
|
Andriolo L, Merli G, Tobar C, Altamura SA, Kon E, Filardo G. Regenerative therapies increase survivorship of avascular necrosis of the femoral head: a systematic review and meta-analysis. INTERNATIONAL ORTHOPAEDICS 2018; 42:1689-1704. [PMID: 29411077 DOI: 10.1007/s00264-018-3787-0] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/02/2017] [Accepted: 01/18/2018] [Indexed: 02/08/2023]
Abstract
PURPOSE The aim of this study was to document the available evidence on the use of regenerative techniques for the treatment of femoral head osteonecrosis (or avascular necrosis of femoral head, AVN) and to understand their benefit compared to core decompression (CD) alone in avoiding failure and the need for total hip replacement (THR). METHODS The search was conducted on three medical electronic databases according to PRISMA guidelines. The studies reporting number and timing of failures were included in a meta-analysis calculating cumulative survivorship with a Kaplan-Mayer curve. Moreover, the results on failures in treatment groups reported in RCT were compared with those documented in control groups, in order to understand the benefit of biological therapies compared to CD for the treatment of AVN. RESULTS Forty-eight studies were included in this systematic review, reporting results of different types of regenerative techniques: mesenchymal stem cell implantation in the osteonecrotic area, intra-arterial infiltration with mesenchymal stem cells, implantation of bioactive molecules, or platelet-rich plasma. Overall, reported results were good, with a cumulative survivorship of 80% after ten year follow-up, and better results when regenerative treatments were combined to CD compared to CD alone (89.9% vs 70.6%, p < 0.0001). CONCLUSION Regenerative therapies offer good clinical results for the treatment of AVN. The combination of CD with regenerative techniques provides a significant improvement in terms of survivorship over time compared with CD alone. Further studies are needed to identify the best procedure and the most suitable patients to benefit from regenerative treatments for AVN.
Collapse
Affiliation(s)
- Luca Andriolo
- II Orthopaedic and Traumatologic Clinic, Rizzoli Orthopaedic Institute, Bologna, Italy
| | - Giulia Merli
- Nano-Biotechnology Laboratory-NaBi, Research and Innovation Technology Department, Bologna, Italy.
| | - Carlos Tobar
- Department of Orthopedics and Traumatology, Avansalud Clinic of Santiago, Santiago, Chile
| | | | - Elizaveta Kon
- Humanitas University Department of Biomedical Sciences, Milan, Italy
- Humanitas Clinical and Research Center, Milan, Italy
| | - Giuseppe Filardo
- Nano-Biotechnology Laboratory-NaBi, Research and Innovation Technology Department, Bologna, Italy
| |
Collapse
|
1048
|
Parathyroid hormone 1‑34 inhibits senescence in rat nucleus pulposus cells by activating autophagy via the m‑TOR pathway. Mol Med Rep 2018; 18:2681-2688. [PMID: 29956812 PMCID: PMC6102631 DOI: 10.3892/mmr.2018.9229] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2018] [Accepted: 06/20/2018] [Indexed: 11/15/2022] Open
Abstract
Osteoporosis is closely associated with intervertebral disc degeneration. While parathyroid hormone (PTH) 1–34, which is an established drug used to treatosteoporosis, is thought to inhibit the disc degeneration associated with osteoporosis, the precise mechanism involved remains unclear. In the present study, primary Sprague-Dawley rat nucleus pulposus cells (NPCs) were cultured, phenotyped and then treated with dexamethasone (DXM) for 48 h. Cell area analysis and β-galactosidase staining were used to investigate the effect of DXM on the senescence of NPCs. In addition, the protein levels of LC3-II, Beclin-1, P62, p-mTOR and p-p70S6k were determined by western blotting and analyzing the regulatory effect of PTH upon autophagy and the mTOR signaling pathway in cells treated with DXM. Following autophagic inhibition induced by ATG5 siRNA transfection, the regulatory effect of PTH on senescence in NPCs were investigated in addition to the potential role of autophagy. As the concentration of DXM increased, the size of the NPCs was significantly enlarged and the proportion of cells with positive β-galactosidase staining increased significantly (P<0.05). In terms of protein expression, PTH treatment led to an increase in LC3-II and Beclin-1 proteins, a reduction in P62 protein, and inhibited p-mTOR and p-p70S6k protein expression in DXM-treated NPCs (P<0.05). PTH attenuated the effect of DXM according to the cell size and percentage of β-galactosidase-positive cells. However, the inhibition of autophagy via ATG5 siRNA transfection reversed the protective effect of PTH on cell senescence (P<0.05). Collectively, the present findings suggest that PTH may inhibit the senescence of NPCs induced by DXM by activating autophagy via the mTOR pathway.
Collapse
|
1049
|
Papavasiliou AV, Triantafyllopoulos I, Paxinos O, Tsoukas D, Kostantoulakis C. The role of cell therapies and hip arthroscopy in the management of osteonecrosis: an update. J Hip Preserv Surg 2018; 5:202-208. [PMID: 30393546 PMCID: PMC6206693 DOI: 10.1093/jhps/hny021] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2018] [Revised: 05/15/2018] [Accepted: 05/22/2018] [Indexed: 12/19/2022] Open
Abstract
The exact pathophysiology of osteonecrosis of the femoral head (ONFH) is still unknown. There is evidence to suggest that in ON there is decreased population and altered function of the mesenchymal stem cells (MSCs) of the femoral head. This could influence both the actual occurrence of ON itself and the repair process that follows. Hence, in such an environment it only is rational to consider the use of cell-based treatments to potentially regenerate lost or damaged bone. The aim of this review is to provide an up-to-date, evidence-based information in the use of cell therapies in the treatment of nontraumatic ONFH and the use of hip arthroscopy in the field.
Collapse
Affiliation(s)
| | | | - Odysseas Paxinos
- Orthopedic Department, 251 Hellenic Air Force Hospital, Athens, Greece
| | - Dimitrios Tsoukas
- Hygeia Hospital, 4 Erythrou Stavrou Str. & Kifisias Av, Marousi, Athens, Greece
| | | |
Collapse
|
1050
|
de Araújo Farias V, Carrillo-Gálvez AB, Martín F, Anderson P. TGF-β and mesenchymal stromal cells in regenerative medicine, autoimmunity and cancer. Cytokine Growth Factor Rev 2018; 43:25-37. [PMID: 29954665 DOI: 10.1016/j.cytogfr.2018.06.002] [Citation(s) in RCA: 97] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2018] [Accepted: 06/12/2018] [Indexed: 12/30/2022]
Abstract
Multipotent mesenchymal stromal cells (MSCs) represent a promising cell-based therapy in regenerative medicine and for the treatment of inflammatory/autoimmune diseases. Importantly, MSCs have emerged as an important contributor to the tumor stroma with both pro- and anti-tumorigenic effects. However, the successful translation of MSCs to the clinic and the prevention of their tumorigenic and metastatic effect require a greater understanding of factors controlling their proliferation, differentiation, migration and immunomodulation in vitro and in vivo. The transforming growth factor(TGF)-β1, 2 and 3 are involved in almost every aspect of MSC function. The aim of this review is to highlight the roles that TGF-β play in the biology and therapeutic applications of MSCs. We will discuss the how TGF-β modulate MSC function as well as the paracrine effects of MSC-derived TGF-β on other cell types in the context of tissue regeneration, immune responses and cancer. Finally, taking all these aspects into consideration we discuss how modulation of TGF-β signaling/production in MSCs could be of clinical interest.
Collapse
Affiliation(s)
- Virgínea de Araújo Farias
- Centre for Genomics and Oncological Research (GENYO): Pfizer/University of Granada/Andalucian Regional Government, PTS Granada, Avenida de la Ilustración 114, 18016 Granada, Spain; Facultad de Odontología, Universidad de Granada, Campus Universitario de Cartuja, 18071 Granada, Spain
| | - Ana Belén Carrillo-Gálvez
- Centre for Genomics and Oncological Research (GENYO): Pfizer/University of Granada/Andalucian Regional Government, PTS Granada, Avenida de la Ilustración 114, 18016 Granada, Spain
| | - Francisco Martín
- Centre for Genomics and Oncological Research (GENYO): Pfizer/University of Granada/Andalucian Regional Government, PTS Granada, Avenida de la Ilustración 114, 18016 Granada, Spain
| | - Per Anderson
- Centre for Genomics and Oncological Research (GENYO): Pfizer/University of Granada/Andalucian Regional Government, PTS Granada, Avenida de la Ilustración 114, 18016 Granada, Spain.
| |
Collapse
|