1
|
Zhou Y, Chen L, Wang M, Yang Y, Hu B, Li G, Wei F. Paroxetine promotes longevity via ser-7-dop-4-IIS axis in Caenorhabditis elegans. GeroScience 2024:10.1007/s11357-024-01492-7. [PMID: 39729241 DOI: 10.1007/s11357-024-01492-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2024] [Accepted: 12/17/2024] [Indexed: 12/28/2024] Open
Abstract
Paroxetine, a selective serotonin reuptake inhibitor, is widely used in the clinical treatment of depression. While several antidepressants show promise as geroprotectors, the role of paroxetine in aging remains unclear. In this study, we evaluated the lifespan extension effect of paroxetine in Caenorhabditis elegans (C. elegans) and elucidated the underlying mechanisms. The results showed that paroxetine can prolong lifespan concomitant extension of healthspan as indicated by increasing mobility and reducing lipofuscin accumulation, as well as confer protection to nematodes against different abiotic stresses. Paroxetine upregulated ser-7 expression and downregulated dop-4 expression. dop-4 RNA interference (RNAi) mimicked the beneficial effect of paroxetine on lifespan. Conversely, ser-7 RNAi abolished paroxetine-induced lifespan extension and the expression changes of dop-4 and genes related to insulin/insulin-like growth factor 1 signaling (IIS). Moreover, paroxetine exhibited a comparable lifespan extension effect to that observed in daf-2 or age-1 mutants; however, this effect was abolished in daf-16 mutant. Taken together, these results suggest that paroxetine promotes health and longevity in C. elegans through the ser-7-dop-4-IIS pathway, underscoring its potential as a geroprotector.
Collapse
Affiliation(s)
- Yiming Zhou
- Center for Aging Biomedicine, College of Life Sciences, National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, Hunan Normal University, 36 Lushan Road, Changsha, 410081, Hunan, China
| | - Lijuan Chen
- Center for Aging Biomedicine, College of Life Sciences, National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, Hunan Normal University, 36 Lushan Road, Changsha, 410081, Hunan, China
| | - Meijing Wang
- Center for Aging Biomedicine, College of Life Sciences, National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, Hunan Normal University, 36 Lushan Road, Changsha, 410081, Hunan, China
| | - Yang Yang
- Center for Aging Biomedicine, College of Life Sciences, National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, Hunan Normal University, 36 Lushan Road, Changsha, 410081, Hunan, China
| | - Bin Hu
- Center for Aging Biomedicine, College of Life Sciences, National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, Hunan Normal University, 36 Lushan Road, Changsha, 410081, Hunan, China
| | - Guolin Li
- Center for Aging Biomedicine, College of Life Sciences, National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, Hunan Normal University, 36 Lushan Road, Changsha, 410081, Hunan, China
- Key Laboratory of Hunan Province for Model Animal and Stem Cell Biology, School of Medicine, Hunan Normal University, Changsha, Hunan, China
| | - Fang Wei
- Center for Aging Biomedicine, College of Life Sciences, National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, Hunan Normal University, 36 Lushan Road, Changsha, 410081, Hunan, China.
| |
Collapse
|
2
|
Ferrao Blanco MN, Lesage R, Kops N, Fahy N, Bekedam FT, Chavli A, Bastiaansen-Jenniskens YM, Geris L, Chambers MG, Pitsillides AA, Narcisi R, van Osch GJ. A multi-model approach identifies ALW-II-41-27 as a promising therapy for osteoarthritis-associated inflammation and endochondral ossification. Heliyon 2024; 10:e40871. [PMID: 39717596 PMCID: PMC11664402 DOI: 10.1016/j.heliyon.2024.e40871] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Revised: 11/29/2024] [Accepted: 12/02/2024] [Indexed: 12/25/2024] Open
Abstract
Low-grade inflammation and pathological endochondral ossification are key processes underlying the progression of osteoarthritis, the most prevalent joint disease worldwide. In this study, we employed a multi-faceted approach, integrating publicly available datasets, in silico analyses, in vitro experiments and in vivo models to identify new therapeutic candidates targeting these processes. Data mining of transcriptomic datasets identified EPHA2, a receptor tyrosine kinase associated with cancer, as being linked to both inflammation and endochondral ossification in osteoarthritis. A computational model of cellular signaling networks in chondrocytes predicted that in silico activation of EPHA2 in healthy chondrocytes increases inflammatory mediators and induces hypertrophic differentiation, a hallmark of endochondral ossification. We then evaluated the effect of EPHA2 inhibition using the tyrosine kinase inhibitor ALW-II-41-27 in cultured human chondrocytes from individuals with osteoarthritis, demonstrating significant reductions in both inflammation and hypertrophy. Additionally, systemic subcutaneous administration of ALW-II-41-27 in a mouse osteoarthritic model attenuated joint degeneration by reducing local inflammation and pathological endochondral ossification. Collectively, this study demonstrates a novel drug discovery pipeline that integrates computational, experimental, and animal models, paving the way for the development of disease-modifying treatments for osteoarthritis.
Collapse
Affiliation(s)
- Mauricio N. Ferrao Blanco
- Department of Orthopaedics and Sports Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Raphaelle Lesage
- Prometheus, Division of Skeletal Tissue Engineering, KU Leuven, Belgium
- Biomechanics Section, KU Leuven, Belgium
| | - Nicole Kops
- Department of Orthopaedics and Sports Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Niamh Fahy
- Department of Orthopaedics and Sports Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
- Department of Oral and Maxillofacial Surgery, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
- Department of Applied Science, Technological University of the Shannon: Midlands Midwest, Limerick, Ireland
| | - Fjodor T. Bekedam
- Department of Orthopaedics and Sports Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Athina Chavli
- Department of Orthopaedics and Sports Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | | | - Liesbet Geris
- Prometheus, Division of Skeletal Tissue Engineering, KU Leuven, Belgium
- Biomechanics Section, KU Leuven, Belgium
- GIGA In Silico Medicine, University of Liège, Belgium
| | - Mark G. Chambers
- Lilly Research Laboratories, Eli Lilly Pharmaceuticals, Indianapolis, USA
| | | | - Roberto Narcisi
- Department of Orthopaedics and Sports Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Gerjo J.V.M. van Osch
- Department of Orthopaedics and Sports Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
- Department of Otorhinolaryngology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
- Department of Biomechanical Engineering, University of Technology Delft, Delft, the Netherlands
| |
Collapse
|
3
|
Dong F, He K, Zhang S, Song K, Jiang L, Hu LP, Li Q, Zhang XL, Zhang N, Li BT, Zhu LL, Li J, Feng M, Gao Y, Chen J, Hu X, Wang J, Jiang C, Wang C, Zhu HH, Da LT, Ji J, Zhang ZG, Bao Z, Jiang SH. SSRI antidepressant citalopram reverses the Warburg effect to inhibit hepatocellular carcinoma by directly targeting GLUT1. Cell Rep 2024; 43:114818. [PMID: 39388353 DOI: 10.1016/j.celrep.2024.114818] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Revised: 07/20/2024] [Accepted: 09/17/2024] [Indexed: 10/12/2024] Open
Abstract
Selective serotonin reuptake inhibitors (SSRIs) have shown promise in cancer therapy, particularly for hepatocellular carcinoma (HCC), but their molecular targets and mechanisms remain unclear. Here, we show that SSRIs exhibit significant anti-HCC effects independent of their classical target, the serotonin reuptake transporter (SERT). Using global inverse gene expression profiling, drug affinity responsive target stability assays, and in silico molecular docking, we demonstrate that citalopram targets glucose transporter 1 (GLUT1), resulting in reduced glycolytic flux. A mutant GLUT1 variant at the citalopram binding site (E380) diminishes the drug's inhibitory effects on the Warburg effect and tumor growth. In preclinical models, citalopram dampens the growth of GLUT1high liver tumors and displays a synergistic effect with anti-PD-1 therapy. Retrospective analysis reveals that SSRI use correlates with a lower risk of metastasis among patients with HCC. Our study describes a role for SSRIs in cancer metabolism, establishing a rationale for their repurposing as potential anti-cancer drugs for HCC.
Collapse
Affiliation(s)
- Fangyuan Dong
- Department of Gastroenterology, Huadong Hospital, Shanghai Medical College, Fudan University, Shanghai 200040, China; Shanghai Key Laboratory of Clinical Geriatric Medicine, Shanghai 200040, China; Shanghai Institute of Geriatrics and Gerontology, Shanghai 200040, China; Department of Geriatrics, Huadong Hospital, Shanghai Medical College, Fudan University, Shanghai 200040, China
| | - Kang He
- Department of Liver Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Shan Zhang
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Kaiyuan Song
- Key Laboratory of Systems Biomedicine (Ministry of Education), Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Luju Jiang
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Li-Peng Hu
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Qing Li
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Xue-Li Zhang
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Naiqi Zhang
- Center for Primary Health Care Research, Lund University, Region Skåne, Sweden
| | - Bo-Tai Li
- Shanghai Immune Therapy Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Li-Li Zhu
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Jun Li
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Mingxuan Feng
- Department of Liver Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Yunchen Gao
- Shanghai United International School Qingpu Campus, Shanghai 201799, China
| | - Jie Chen
- Department of Gastroenterology, Huadong Hospital, Shanghai Medical College, Fudan University, Shanghai 200040, China; Shanghai Key Laboratory of Clinical Geriatric Medicine, Shanghai 200040, China; Shanghai Institute of Geriatrics and Gerontology, Shanghai 200040, China; Department of Geriatrics, Huadong Hospital, Shanghai Medical College, Fudan University, Shanghai 200040, China
| | - Xiaona Hu
- Department of Gastroenterology, Huadong Hospital, Shanghai Medical College, Fudan University, Shanghai 200040, China; Shanghai Key Laboratory of Clinical Geriatric Medicine, Shanghai 200040, China; Shanghai Institute of Geriatrics and Gerontology, Shanghai 200040, China; Department of Geriatrics, Huadong Hospital, Shanghai Medical College, Fudan University, Shanghai 200040, China
| | - Jiaofeng Wang
- Department of Gastroenterology, Huadong Hospital, Shanghai Medical College, Fudan University, Shanghai 200040, China; Shanghai Key Laboratory of Clinical Geriatric Medicine, Shanghai 200040, China; Shanghai Institute of Geriatrics and Gerontology, Shanghai 200040, China; Department of Geriatrics, Huadong Hospital, Shanghai Medical College, Fudan University, Shanghai 200040, China
| | - Chongyi Jiang
- Department of General Surgery, Hepato-Biliary-Pancreatic Center, Huadong Hospital, Fudan University, Shanghai 200040, China
| | - Cun Wang
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Helen He Zhu
- State Key Laboratory of Systems Medicine for Cancer, Renji-Med-X Stem Cell Research Center, Shanghai Cancer Institute & Department of Urology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China
| | - Lin-Tai Da
- Key Laboratory of Systems Biomedicine (Ministry of Education), Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Jianguang Ji
- Center for Primary Health Care Research, Lund University, Region Skåne, Sweden; Department of Public Health and Medicinal Administration, Faculty of Health Sciences, University of Macau, Macao, Macao SAR, China.
| | - Zhi-Gang Zhang
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200240, China.
| | - Zhijun Bao
- Department of Gastroenterology, Huadong Hospital, Shanghai Medical College, Fudan University, Shanghai 200040, China; Shanghai Key Laboratory of Clinical Geriatric Medicine, Shanghai 200040, China; Shanghai Institute of Geriatrics and Gerontology, Shanghai 200040, China; Department of Geriatrics, Huadong Hospital, Shanghai Medical College, Fudan University, Shanghai 200040, China.
| | - Shu-Heng Jiang
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200240, China.
| |
Collapse
|
4
|
Li Y, Gao S, Guo Z, Chen Z, Wei Y, Li Y, Ba Y, Liu Z, Bao H. Screening of potential drugs for the treatment of diabetic kidney disease using single-cell transcriptome sequencing and connectivity map data. Biochem Biophys Res Commun 2024; 725:150263. [PMID: 38905995 DOI: 10.1016/j.bbrc.2024.150263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Revised: 05/31/2024] [Accepted: 06/12/2024] [Indexed: 06/23/2024]
Abstract
OBJECTIVE To explore the feasibility of screening potential drugs for the treatment of diabetic kidney disease (DKD) using a single-cell transcriptome sequencing dataset and Connectivity Map (CMap) database screening. METHODS A DKD single-nucleus transcriptome sequencing dataset was analyzed using Seurat 4.0 to obtain specific podocyte subclusters and differentially expressed genes (DEGs) related to DKD. These DEGs were subsequently subjected to a search against the CMap database to screen for drug candidates. Cell and animal experiments were conducted to evaluate the efficacy of the top 3 drug candidates. RESULTS Initially, we analyzed the DKD single-nucleus transcriptome sequencing dataset to obtain intrinsic renal cells such as podocytes, endothelial cells, mesangial cells, proximal tubular cells, collecting duct cells and immune cells. Podocytes were further divided into four subclusters, among which the proportion of POD_1 podcytes was significantly greater in DKD kidneys than in control kidneys (34.0 % vs. 3.4 %). The CMap database was searched using the identified DEGs in the POD_1 subcluster, and the drugs, including tozasertib, paroxetine, and xylazine, were obtained. Cell-based experiments showed that tozasertib, paroxetine and xylazine had no significant podocyte toxicity in the concentration range of 0.01-50 μM. Tozasertib, paroxetine, and xylazine all reversed the advanced glycation end products (AGEs)-induced decrease in podocyte marker levels, but the effect of paroxetine was more prominent. Animal experiments showed that paroxetine decreased urine ALB/Cr levels in DKD model mice by approximately 51.5 % (115.7 mg/g vs. 238.8 mg/g, P < 0.05). Histopathological assessment revealed that paroxetine attenuated basement membrane thickening, restored the number of foot processes of podocytes, and reduced foot process fusion. In addition, paroxetine also attenuated renal tubular-interstitial fibrosis. Mechanistically, paroxetine inhibited the expression of GRK2 and NLRP3, decreased the phosphorylation level of p65, restored NRF2 expression, and relieved inflammation and oxidative stress. CONCLUSION This strategy based on single-cell transcriptome sequencing and CMap data can facilitate the identification and aid the rapid development of clinical DKD drugs. Paroxetine, screened by this strategy, has excellent renoprotective effects.
Collapse
Affiliation(s)
- Yi Li
- National Clinical Research Center for Kidney Diseases, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210016, China
| | - Shaohui Gao
- National Clinical Research Center for Kidney Diseases, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210016, China
| | - Zhaochen Guo
- National Clinical Research Center for Kidney Diseases, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210016, China
| | - Zige Chen
- National Clinical Research Center for Kidney Diseases, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210016, China
| | - Yihan Wei
- National Clinical Research Center for Kidney Diseases, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210016, China
| | - Yutong Li
- National Clinical Research Center for Kidney Diseases, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210016, China
| | - Yani Ba
- National Clinical Research Center for Kidney Diseases, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210016, China
| | - Zhihong Liu
- National Clinical Research Center for Kidney Diseases, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210016, China
| | - Hao Bao
- National Clinical Research Center for Kidney Diseases, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210016, China; State Key Laboratory of Pharmaceutical Biotechnology, Medical School, Nanjing University, Nanjing, 210093, China.
| |
Collapse
|
5
|
Han D, Jiang C, Xu H, Chu R, Zhang R, Fang R, Ge H, Lu M, Wang M, Tai Y, Yan S, Wei W, Wang Q. Inhibition of GRK2 ameliorates the pristane-induced mouse SLE model by suppressing plasma cells differentiation. Int Immunopharmacol 2024; 138:112557. [PMID: 38936060 DOI: 10.1016/j.intimp.2024.112557] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Revised: 06/21/2024] [Accepted: 06/21/2024] [Indexed: 06/29/2024]
Abstract
Systemic lupus erythematosus (SLE) is a multifaceted autoimmune disorder characterized by diverse clinical manifestations and organ damage. Despite its elusive etiology, dysregulated subsets and functions of B cells are pivotal in SLE pathogenesis. Peoniflorin-6'-O-benzene sulfonate (CP-25), an esterification modification of Paeoniflorin, exhibits potent anti-inflammatory and immunomodulatory properties in autoimmune diseases (AID). However, the involvement of CP-25 and its target, GRK2, in SLE development has not been explored. In this study, we demonstrate that both genetic deficiency and pharmacological inhibition of GRK2 attenuate autoantibodies production, reduce systemic inflammation, and mitigate histopathological alterations in the spleen and kidney in the pristane-induced mouse SLE model. Importantly, our findings highlight that both genetic deficiency and pharmacological inhibition of GRK2 suppress plasma cells generation and restore dysregulated B-cell subsets by modulating two crucial transcription factors, Blimp1 and IRF4. Collectively, targeting GRK2 with CP-25 emerges as a promising therapeutic approach for SLE.
Collapse
Affiliation(s)
- Dafei Han
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Center of Anti-Inflammatory and Immune Medicine, Hefei, China
| | - Chunru Jiang
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Center of Anti-Inflammatory and Immune Medicine, Hefei, China
| | - Huihui Xu
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Center of Anti-Inflammatory and Immune Medicine, Hefei, China
| | - Rui Chu
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Center of Anti-Inflammatory and Immune Medicine, Hefei, China
| | - Renhao Zhang
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Center of Anti-Inflammatory and Immune Medicine, Hefei, China
| | - Ruhong Fang
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Center of Anti-Inflammatory and Immune Medicine, Hefei, China
| | - Hui Ge
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Center of Anti-Inflammatory and Immune Medicine, Hefei, China
| | - Meiyue Lu
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Center of Anti-Inflammatory and Immune Medicine, Hefei, China
| | - Mingzhu Wang
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Center of Anti-Inflammatory and Immune Medicine, Hefei, China
| | - Yu Tai
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Center of Anti-Inflammatory and Immune Medicine, Hefei, China
| | - Shangxue Yan
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Center of Anti-Inflammatory and Immune Medicine, Hefei, China
| | - Wei Wei
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Center of Anti-Inflammatory and Immune Medicine, Hefei, China.
| | - Qingtong Wang
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Center of Anti-Inflammatory and Immune Medicine, Hefei, China.
| |
Collapse
|
6
|
Celik B, Leal AF, Tomatsu S. Potential Targeting Mechanisms for Bone-Directed Therapies. Int J Mol Sci 2024; 25:8339. [PMID: 39125906 PMCID: PMC11312506 DOI: 10.3390/ijms25158339] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Revised: 07/23/2024] [Accepted: 07/24/2024] [Indexed: 08/12/2024] Open
Abstract
Bone development is characterized by complex regulation mechanisms, including signal transduction and transcription factor-related pathways, glycobiological processes, cellular interactions, transportation mechanisms, and, importantly, chemical formation resulting from hydroxyapatite. Any abnormal regulation in the bone development processes causes skeletal system-related problems. To some extent, the avascularity of cartilage and bone makes drug delivery more challenging than that of soft tissues. Recent studies have implemented many novel bone-targeting approaches to overcome drawbacks. However, none of these strategies fully corrects skeletal dysfunction, particularly in growth plate-related ones. Although direct recombinant enzymes (e.g., Vimizim for Morquio, Cerezyme for Gaucher, Elaprase for Hunter, Mepsevii for Sly diseases) or hormone infusions (estrogen for osteoporosis and osteoarthritis), traditional gene delivery (e.g., direct infusion of viral or non-viral vectors with no modifications on capsid, envelope, or nanoparticles), and cell therapy strategies (healthy bone marrow or hematopoietic stem cell transplantation) partially improve bone lesions, novel delivery methods must be addressed regarding target specificity, less immunogenicity, and duration in circulation. In addition to improvements in bone delivery, potential regulation of bone development mechanisms involving receptor-regulated pathways has also been utilized. Targeted drug delivery using organic and inorganic compounds is a promising approach in mostly preclinical settings and future clinical translation. This review comprehensively summarizes the current bone-targeting strategies based on bone structure and remodeling concepts while emphasizing potential approaches for future bone-targeting systems.
Collapse
Affiliation(s)
- Betul Celik
- Department of Biological Sciences, University of Delaware, Newark, DE 19716, USA;
- Nemours Children’s Health, 1600 Rockland Rd., Wilmington, DE 19803, USA;
| | - Andrés Felipe Leal
- Nemours Children’s Health, 1600 Rockland Rd., Wilmington, DE 19803, USA;
- Institute for the Study of Inborn Errors of Metabolism, Faculty of Science, Pontificia Universidad Javeriana, Bogotá 110231, Colombia
| | - Shunji Tomatsu
- Nemours Children’s Health, 1600 Rockland Rd., Wilmington, DE 19803, USA;
- Department of Pediatrics, Graduate School of Medicine, Gifu University, Gifu 501-1193, Japan
- Department of Pediatrics, Thomas Jefferson University, Philadelphia, PA 19144, USA
| |
Collapse
|
7
|
Sanchez GA, Smrcka AV, Jutkiewicz EM. Biasing G βγ Downstream Signaling with Gallein Inhibits Development of Morphine Tolerance and Potentiates Morphine-Induced Nociception in a Tolerant State. Mol Pharmacol 2024; 106:47-55. [PMID: 38769020 PMCID: PMC11187686 DOI: 10.1124/molpharm.124.000875] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Revised: 05/03/2024] [Accepted: 05/09/2024] [Indexed: 05/22/2024] Open
Abstract
Opioid analgesics are widely used as a treatment option for pain management and relief. However, the misuse of opioid analgesics has contributed to the current opioid epidemic in the United States. Prescribed opioids such as morphine, codeine, oxycodone, and fentanyl are mu-opioid receptor (MOR) agonists primarily used in the clinic to treat pain or during medical procedures, but development of tolerance limits their utility for treatment of chronic pain. Here we explored the effects of biasing Gβγ signaling on tolerance development after chronic morphine treatment in vivo. We hypothesized that biasing Gβγ signaling with gallein could prevent activation of regulatory signaling pathways that result in tolerance to antinociceptive effects of MOR agonists. Gallein has been shown to bind to Gβγ and inhibit interactions of Gβγ with phospholipase-Cβ3 (PLCβ3) or G-protein-coupled receptor kinase 2 (GRK2) but not G-protein inwardly rectifying potassium (GIRK) channels. In mice, morphine-induced antinociception was evaluated in the 55°C warm water tail withdrawal assay. We used two paradigms for gallein treatment: administration during and after three times-daily morphine administration. Our results show that gallein cotreatment during repeated administration of morphine decreased opioid tolerance development and that gallein treatment in an opioid-tolerant state enhanced the potency of morphine. Mechanistically, our data suggest that PLCβ3 is necessary for potentiating effects of gallein in an opioid-tolerant state but not in preventing the development of tolerance. These studies demonstrate that small molecules that target Gβγ signaling could reduce the need for large doses of opioid analgesics to treat pain by producing an opioid-sparing effect. SIGNIFICANCE STATEMENT: Biasing Gβγ signaling prevents tolerance to repeated morphine administration in vivo and potentiates the antinociceptive effects of morphine in an opioid-tolerant state. Mechanistically, phospholipase-Cβ is necessary for potentiating effects of gallein in an opioid-tolerant state but not in preventing the development of tolerance. This study identifies a novel treatment strategy to decrease the development of tolerance to the analgesic effects of mu-opioid receptor agonists, which are necessary to improve pain treatment and decrease the incidence of opioid use disorder.
Collapse
Affiliation(s)
- Gissell A Sanchez
- Department of Pharmacology, University of Michigan, Ann Arbor, Michigan
| | - Alan V Smrcka
- Department of Pharmacology, University of Michigan, Ann Arbor, Michigan
| | | |
Collapse
|
8
|
Zou Z, Hu W, Kang F, Xu Z, Li Y, Zhang J, Li J, Zhang Y, Dong S. Interplay between lipid dysregulation and ferroptosis in chondrocytes and the targeted therapy effect of metformin on osteoarthritis. J Adv Res 2024:S2090-1232(24)00155-3. [PMID: 38621621 DOI: 10.1016/j.jare.2024.04.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Revised: 04/03/2024] [Accepted: 04/13/2024] [Indexed: 04/17/2024] Open
Abstract
INTRODUCTION Osteoarthritis (OA) is a devastating whole-joint disease affecting a large population worldwide; the role of lipid dysregulation in OA and mechanisms underlying targeted therapy effect of lipid-lowering metformin on OA remains poorly defined. OBJECTIVES To investigate the effects of lipid dysregulation on OA progression and to explore lipid dysregulation-targeting OA treatment of metformin. METHODS RNA-Seq data, biochemical, and histochemical assays in human and murine OA cartilage as well as primary chondrocytes were utilized to determine lipid dysregulation. Effects of metformin, a potent lipid-lowering medication, on ACSL4 expression and chondrocyte metabolism were determined. Further molecular experiments, including RT-qPCR, western blotting, flow cytometry, and immunofluorescence staining, were performed to investigate underlying mechanisms. Mice with intra-articular injection of metformin were utilized to determine the effects on ACLT-induced OA progression. RESULTS ACSL4 and 4-HNE expressions were elevated in human and ACLT-induced mouse OA cartilage and IL-1β-treated chondrocytes (P < 0.05). Ferrostatin-1 largely rescued IL-1β-induced MDA, lipid peroxidation, and ferroptotic mitochondrial morphology (P < 0.05). Metformin decreased the levels of OA-related genes (P < 0.05) and increased the levels of p-AMPK and p-ACC in IL-1β-treated chondrocytes. Intra-articular injection of metformin alleviated ACLT-induced OA lesions in mice, and reverted the percentage of chondrocytes positive for MMP13, Col2a1, ACSL4 and 4-HNE in ACLT mice (P < 0.05). Ferroptotic chondrocytes promoted the recruitment and chemotaxis of RAW264.7 cells via CCL2, which was blocked by metformin in vitro (P < 0.05). CONCLUSION We establish a critical role of polyunsaturated fatty acids metabolic process in OA cartilage degradation and define metformin as a potential OA treatment. Metformin reshapes lipid availability and ameliorates chondrocyte ferroptosis sensitivity via the AMPK/ACC pathway. In the future, gene-edited animals and extensive omics technologies will be utilized to reveal detailed lipids' involvement in cartilage lesions.
Collapse
Affiliation(s)
- Zhi Zou
- College of Bioengineering, Chongqing University, Chongqing 400044, China; Department of Biomedical Materials Science, College of Biomedical Engineering, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Wenhui Hu
- Department of Biomedical Materials Science, College of Biomedical Engineering, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Fei Kang
- Department of Biomedical Materials Science, College of Biomedical Engineering, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Zhonghua Xu
- Joint Disease & Sport Medicine Center, Department of Orthopedics, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing 400037, China
| | - Yuheng Li
- Department of Biomedical Materials Science, College of Biomedical Engineering, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Jing Zhang
- College of Bioengineering, Chongqing University, Chongqing 400044, China
| | - Jianmei Li
- Department of Biomedical Materials Science, College of Biomedical Engineering, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Yuan Zhang
- Joint Disease & Sport Medicine Center, Department of Orthopedics, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing 400037, China.
| | - Shiwu Dong
- Department of Biomedical Materials Science, College of Biomedical Engineering, Third Military Medical University (Army Medical University), Chongqing 400038, China; State Key Laboratory of Trauma and Chemical Poisoning, Third Military Medical University (Army Medical University), Chongqing 400038, China.
| |
Collapse
|
9
|
Lv Z, Wang P, Li W, Xie Y, Sun W, Jin X, Jiang R, Fei Y, Liu Y, Shi T, Guo H, Sun Z, Lin J, Wang X, Tan G, Wu Y, Bao N, Shi D. Bifunctional TRPV1 Targeted Magnetothermal Switch to Attenuate Osteoarthritis Progression. RESEARCH (WASHINGTON, D.C.) 2024; 7:0316. [PMID: 38371274 PMCID: PMC10871150 DOI: 10.34133/research.0316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Accepted: 01/20/2024] [Indexed: 02/20/2024]
Abstract
Transient receptor potential vanilloid family member 1 (TRPV1) has been revealed as a therapeutic target of osteoarthritis (OA), the most common deteriorating whole joint disease, by impeding macrophagic inflammation and chondrocytes ferroptosis. However, the clinical application for capsaicin as the TRPV1 agonist is largely limited by its chronic toxicity. To address this issue, we developed a bifunctional controllable magnetothermal switch targeting TRPV1 for the alleviation of OA progression by coupling of magnetic nanoparticles (MNPs) to TRPV1 monoclonal antibodies (MNPs-TRPV1). Under the alternating magnetic field (AMF) stimulation, MNPs-TRPV1 locally dissipated heat, which was sufficient to trigger the opening and activation of TRPV1, and effectively impeded macrophagic inflammation and chondrocyte ferroptosis. This magnetothermal modulation of TRPV1 simultaneously attenuated synovitis and cartilage degeneration in mice incurred by destabilization of medial meniscus surgery, indicating the delayed OA progression. Furthermore, MNPs-TRPV1 with AMF exposure remarkably reduced knee pain sensitivity, alleviated the crippled gait, and improved spontaneous ambulatory activity performance in the mice OA model. Overall, this work provides a potential pathogenesis-based precise OA therapy with temporally and spatially magnetothermal modulation of TRPV1 in a controllable manner.
Collapse
Affiliation(s)
- Zhongyang Lv
- Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School,
Nanjing University, Nanjing 210008, China
- Department of Orthopedics, Nanjing Jinling Hospital, Affiliated Hospital of Medical School,
Nanjing University, Nanjing 210002, China
| | - Peng Wang
- Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School,
Nanjing University, Nanjing 210008, China
| | - Weitong Li
- Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery,
Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, 321 Zhongshan Road, Nanjing 210008, Jiangsu, China
| | - Ya Xie
- Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery,
Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, 321 Zhongshan Road, Nanjing 210008, Jiangsu, China
| | - Wei Sun
- Department of orthopedic,
The Jiangyin Clinical College of Xuzhou Medical University, Jiangyin 214400, China
| | - Xiaoyu Jin
- Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery,
Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, 321 Zhongshan Road, Nanjing 210008, Jiangsu, China
| | - Ruiyang Jiang
- Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery,
Nanjing Drum Tower Hospital Clinical College of Xuzhou Medical University, 321 Zhongshan Road, Nanjing 210008, Jiangsu, China
| | - Yuxiang Fei
- Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School,
Nanjing University, Nanjing 210008, China
| | - Yuan Liu
- Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School,
Nanjing University, Nanjing 210008, China
| | - Tianshu Shi
- Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School,
Nanjing University, Nanjing 210008, China
| | - Hu Guo
- Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School,
Nanjing University, Nanjing 210008, China
| | - Ziying Sun
- Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School,
Nanjing University, Nanjing 210008, China
- Department of Orthopedics, Nanjing Jinling Hospital, Affiliated Hospital of Medical School,
Nanjing University, Nanjing 210002, China
| | - Jintao Lin
- Department of Orthopedics, Nanjing Jinling Hospital, Affiliated Hospital of Medical School,
Nanjing University, Nanjing 210002, China
| | - Xucai Wang
- Co-Innovation Center for Efficient Processing and Utilization of Forest Resources, College of Chemical Engineering,
Nanjing Forestry University, Nanjing, 210037, China
| | - Guihua Tan
- Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School,
Nanjing University, Nanjing 210008, China
| | - Yizhang Wu
- Department of Applied Physical Sciences,
The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Nirong Bao
- Department of Orthopedics, Nanjing Jinling Hospital, Affiliated Hospital of Medical School,
Nanjing University, Nanjing 210002, China
| | - Dongquan Shi
- Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School,
Nanjing University, Nanjing 210008, China
| |
Collapse
|
10
|
Yang X, Zhao Y, Wei Q, Zhu X, Wang L, Zhang W, Liu X, Kuai J, Wang F, Wei W. GRK2 inhibits Flt-1 + macrophage infiltration and its proangiogenic properties in rheumatoid arthritis. Acta Pharm Sin B 2024; 14:241-255. [PMID: 38261818 PMCID: PMC10792976 DOI: 10.1016/j.apsb.2023.09.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Revised: 09/04/2023] [Accepted: 09/15/2023] [Indexed: 01/25/2024] Open
Abstract
Rheumatoid arthritis (RA) is an autoimmune disease with a complex etiology. Monocyte-derived macrophages (MDMs) infiltration are associated with RA severity. We have reported the deletion of G-protein-coupled receptor kinase 2 (GRK2) reprograms macrophages toward an anti-inflammatory phenotype by recovering G-protein-coupled receptor signaling. However, as more GRK2-interacting proteins were discovered, the GRK2 interactome mechanisms in RA have been understudied. Thus, in the collagen-induced arthritis mouse model, we performed genetic GRK2 deletion using GRK2f/fLyz2-Cre+/- mice. Synovial inflammation and M1 polarization were improved in GRK2f/fLyz2-Cre+/- mice. Supporting experiments with RNA-seq and dual-luciferase reporter assays identified peroxisome proliferator-activated receptor γ (PPARγ) as a new GRK2-interacting protein. We further confirmed that fms-related tyrosine kinase 1 (Flt-1), which promoted macrophage migration to induce angiogenesis, was inhibited by GRK2-PPARγ signaling. Mechanistically, excess GRK2 membrane recruitment in CIA MDMs reduced the activation of PPARγ ligand-binding domain and enhanced Flt-1 transcription. Furthermore, the treatment of mice with GRK2 activity inhibitor resulted in significantly diminished CIA pathology, Flt-1+ macrophages induced-synovial inflammation, and angiogenesis. Altogether, we anticipate to facilitate the elucidation of previously unappreciated details of GRK2-specific intracellular signaling. Targeting GRK2 activity is a viable strategy to inhibit MDMs infiltration, affording a distinct way to control joint inflammation and angiogenesis of RA.
Collapse
Affiliation(s)
- Xuezhi Yang
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-Inflammatory and Immune Medicine, Anhui Collaborative Innovation Center of Anti-Inflammatory and Immune Medicine, Ministry of Education, Hefei 230032, China
| | - Yingjie Zhao
- Department of Clinical Pharmacology, the Second Affiliated Hospital of Anhui Medical University, Hefei 230601, China
| | - Qi Wei
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-Inflammatory and Immune Medicine, Anhui Collaborative Innovation Center of Anti-Inflammatory and Immune Medicine, Ministry of Education, Hefei 230032, China
| | - Xuemin Zhu
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-Inflammatory and Immune Medicine, Anhui Collaborative Innovation Center of Anti-Inflammatory and Immune Medicine, Ministry of Education, Hefei 230032, China
| | - Luping Wang
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-Inflammatory and Immune Medicine, Anhui Collaborative Innovation Center of Anti-Inflammatory and Immune Medicine, Ministry of Education, Hefei 230032, China
| | - Wankang Zhang
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-Inflammatory and Immune Medicine, Anhui Collaborative Innovation Center of Anti-Inflammatory and Immune Medicine, Ministry of Education, Hefei 230032, China
| | - Xiaoyi Liu
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-Inflammatory and Immune Medicine, Anhui Collaborative Innovation Center of Anti-Inflammatory and Immune Medicine, Ministry of Education, Hefei 230032, China
| | - Jiajie Kuai
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-Inflammatory and Immune Medicine, Anhui Collaborative Innovation Center of Anti-Inflammatory and Immune Medicine, Ministry of Education, Hefei 230032, China
| | - Fengling Wang
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-Inflammatory and Immune Medicine, Anhui Collaborative Innovation Center of Anti-Inflammatory and Immune Medicine, Ministry of Education, Hefei 230032, China
| | - Wei Wei
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-Inflammatory and Immune Medicine, Anhui Collaborative Innovation Center of Anti-Inflammatory and Immune Medicine, Ministry of Education, Hefei 230032, China
| |
Collapse
|
11
|
Karuppagounder V, Chung J, Abdeen A, Thompson A, Bouboukas A, Pinamont WJ, Yoshioka NK, Sepulveda DE, Raup-Konsavage WM, Graziane NM, Vrana KE, Elbarbary RA, Kamal F. Therapeutic Effects of Non-Euphorigenic Cannabis Extracts in Osteoarthritis. Cannabis Cannabinoid Res 2023; 8:1030-1044. [PMID: 35994012 PMCID: PMC10714119 DOI: 10.1089/can.2021.0244] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Introduction: Osteoarthritis (OA) is disabling and degenerative disease of the joints that is clinically characterized by pain and loss of function. With no disease-modifying treatment available, current therapies aim at pain management but are of limited efficacy. Cannabis products, specifically cannabinoids, are widely used to control pain and inflammation in many diseases with no scientific evidence demonstrating their efficacy in OA. Objective: We investigated the effects of non-euphorigenic cannabis extracts, CBD oil and cannabigerol oil (CBG oil), on pain and disease progression in OA mice. Methods and Results: Twelve-week-old male C57BL/6J mice received either sham or destabilization of the medial meniscus (DMM) surgery. DMM mice were treated with vehicle, CBD oil, or CBG oil. The gait of DMM mice was impaired as early as 2 weeks following surgery and continued deteriorating until week 8, which was restored by CBD oil and CBG oil treatments throughout the disease course. Mechanical allodynia developed in DMM mice, however, was not ameliorated by any of the treatments. On the other hand, both CBD oil and CBG oil ameliorated cold allodynia. In open field test, both oil treatments normalized changes in the locomotor activity of DMM mice. CBD oil and CBG oil treatments significantly reduced synovitis in DMM mice. Only CBG oil reduced cartilage degeneration, chondrocyte loss, and matrix metalloproteinase 13 expression, with a significant increase in the number of anabolic chondrocytes. Subchondral bone remodeling found in vehicle-treated DMM mice was not ameliorated by either CBD or CBG oil. Conclusions: Our results show evidence for the therapeutic efficacy of CBD oil and CBG oil, where both oils ameliorate pain and inflammation, and improve gait and locomotor activity in OA mice, representing clinical pain and function. Importantly, only CBG oil is chondroprotective, which may provide superior efficacy in future studies in OA patients.
Collapse
Affiliation(s)
- Vengadeshprabhu Karuppagounder
- Center for Orthopedic Research and Translational Science (CORTS), Penn State College of Medicine, Hershey, Pennsylvania, USA
- Department of Orthopedics and Rehabilitation, Departments of Penn State College of Medicine, Hershey, Pennsylvania, USA
| | - Juliet Chung
- Center for Orthopedic Research and Translational Science (CORTS), Penn State College of Medicine, Hershey, Pennsylvania, USA
- Department of Orthopedics and Rehabilitation, Departments of Penn State College of Medicine, Hershey, Pennsylvania, USA
| | - Ahmed Abdeen
- Center for Orthopedic Research and Translational Science (CORTS), Penn State College of Medicine, Hershey, Pennsylvania, USA
- Department of Orthopedics and Rehabilitation, Departments of Penn State College of Medicine, Hershey, Pennsylvania, USA
| | - Amy Thompson
- Center for Orthopedic Research and Translational Science (CORTS), Penn State College of Medicine, Hershey, Pennsylvania, USA
- Department of Orthopedics and Rehabilitation, Departments of Penn State College of Medicine, Hershey, Pennsylvania, USA
| | - Andreas Bouboukas
- Center for Orthopedic Research and Translational Science (CORTS), Penn State College of Medicine, Hershey, Pennsylvania, USA
- Department of Orthopedics and Rehabilitation, Departments of Penn State College of Medicine, Hershey, Pennsylvania, USA
| | - William J. Pinamont
- Center for Orthopedic Research and Translational Science (CORTS), Penn State College of Medicine, Hershey, Pennsylvania, USA
- Department of Orthopedics and Rehabilitation, Departments of Penn State College of Medicine, Hershey, Pennsylvania, USA
| | - Natalie K. Yoshioka
- Center for Orthopedic Research and Translational Science (CORTS), Penn State College of Medicine, Hershey, Pennsylvania, USA
- Department of Orthopedics and Rehabilitation, Departments of Penn State College of Medicine, Hershey, Pennsylvania, USA
| | - Diana E. Sepulveda
- Pharmacology, Penn State College of Medicine, Hershey, Pennsylvania, USA
- Anesthesiology and Perioperative Medicine, and Penn State College of Medicine, Hershey, Pennsylvania, USA
| | | | - Nicholas M. Graziane
- Pharmacology, Penn State College of Medicine, Hershey, Pennsylvania, USA
- Anesthesiology and Perioperative Medicine, and Penn State College of Medicine, Hershey, Pennsylvania, USA
| | - Kent E. Vrana
- Pharmacology, Penn State College of Medicine, Hershey, Pennsylvania, USA
| | - Reyad A. Elbarbary
- Center for Orthopedic Research and Translational Science (CORTS), Penn State College of Medicine, Hershey, Pennsylvania, USA
- Department of Orthopedics and Rehabilitation, Departments of Penn State College of Medicine, Hershey, Pennsylvania, USA
- Biochemistry and Molecular Biology, Penn State College of Medicine, Hershey, Pennsylvania, USA
| | - Fadia Kamal
- Center for Orthopedic Research and Translational Science (CORTS), Penn State College of Medicine, Hershey, Pennsylvania, USA
- Department of Orthopedics and Rehabilitation, Departments of Penn State College of Medicine, Hershey, Pennsylvania, USA
- Pharmacology, Penn State College of Medicine, Hershey, Pennsylvania, USA
| |
Collapse
|
12
|
Khajuria DK, Karuppagounder V, Nowak I, Sepulveda DE, Lewis GS, Norbury CC, Raup-Konsavage WM, Vrana KE, Kamal F, Elbarbary RA. Cannabidiol and Cannabigerol, Nonpsychotropic Cannabinoids, as Analgesics that Effectively Manage Bone Fracture Pain and Promote Healing in Mice. J Bone Miner Res 2023; 38:1560-1576. [PMID: 37597163 PMCID: PMC10864058 DOI: 10.1002/jbmr.4902] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Revised: 06/30/2023] [Accepted: 08/09/2023] [Indexed: 08/21/2023]
Abstract
Bone fractures are among the most prevalent musculoskeletal injuries, and pain management is an essential part of fracture treatment. Fractures heal through an early inflammatory phase, followed by repair and remodeling. Nonsteroidal anti-inflammatory drugs (NSAIDs) are not recommended for fracture pain control as they potently inhibit the inflammatory phase and, thus, impair the healing. Opioids do not provide a better alternative for several reasons, including abuse potential. Accordingly, there is an unmet clinical need for analgesics that effectively ameliorate postfracture pain without impeding the healing. Here, we investigated the analgesic efficacy of two nonpsychotropic cannabinoids, cannabidiol (CBD) and cannabigerol (CBG), in a mouse model for tibial fracture. Mice with fractured tibiae exhibited increased sensitivity to mechanical, cold, and hot stimuli. Both CBD and CBG normalized pain sensitivity to all tested stimuli, and their analgesic effects were comparable to those of the NSAIDs. Interestingly, CBD and CBG promoted bone healing via multiple mechanisms during the early and late phases. During the early inflammatory phase, both cannabinoids increased the abundance of periosteal bone progenitors in the healing hematoma and promoted the osteogenic commitment of these progenitors. During the later phases of healing, CBD and CBG accelerated the fibrocartilaginous callus mineralization and enhanced the viability and proliferation of bone and bone-marrow cells. These effects culminated in higher bone volume fraction, higher bone mineral density, and improved mechanical quality of the newly formed bone. Together, our data suggest CBD and CBG as therapeutic agents that can replace NSAIDs in managing postfracture pain as both cannabinoids exert potent analgesic effects and, at the same time, promote bone healing. © 2023 The Authors. Journal of Bone and Mineral Research published by Wiley Periodicals LLC on behalf of American Society for Bone and Mineral Research (ASBMR).
Collapse
Affiliation(s)
- Deepak Kumar Khajuria
- Department of Orthopaedics and Rehabilitation, The Pennsylvania State University College of Medicine, Hershey, Pennsylvania 17033, USA
- Center for Orthopaedic Research and Translational Science (CORTS), The Pennsylvania State University College of Medicine, Hershey, Pennsylvania 17033, USA
| | - Vengadeshprabhu Karuppagounder
- Department of Orthopaedics and Rehabilitation, The Pennsylvania State University College of Medicine, Hershey, Pennsylvania 17033, USA
- Center for Orthopaedic Research and Translational Science (CORTS), The Pennsylvania State University College of Medicine, Hershey, Pennsylvania 17033, USA
| | - Irena Nowak
- Department of Orthopaedics and Rehabilitation, The Pennsylvania State University College of Medicine, Hershey, Pennsylvania 17033, USA
- Center for Orthopaedic Research and Translational Science (CORTS), The Pennsylvania State University College of Medicine, Hershey, Pennsylvania 17033, USA
| | - Diana E. Sepulveda
- Department of Pharmacology, The Pennsylvania State University College of Medicine, Hershey, Pennsylvania 17033, USA
- Department of Anesthesiology and Perioperative Medicine, The Pennsylvania State College of Medicine, Hershey, Pennsylvania 17033, USA
| | - Gregory S. Lewis
- Department of Orthopaedics and Rehabilitation, The Pennsylvania State University College of Medicine, Hershey, Pennsylvania 17033, USA
- Center for Orthopaedic Research and Translational Science (CORTS), The Pennsylvania State University College of Medicine, Hershey, Pennsylvania 17033, USA
| | - Christopher C Norbury
- Department of Microbiology and Immunology, The Pennsylvania State University College of Medicine, Hershey, Pennsylvania 17033, USA
| | - Wesley M. Raup-Konsavage
- Department of Pharmacology, The Pennsylvania State University College of Medicine, Hershey, Pennsylvania 17033, USA
| | - Kent E. Vrana
- Department of Pharmacology, The Pennsylvania State University College of Medicine, Hershey, Pennsylvania 17033, USA
| | - Fadia Kamal
- Department of Orthopaedics and Rehabilitation, The Pennsylvania State University College of Medicine, Hershey, Pennsylvania 17033, USA
- Center for Orthopaedic Research and Translational Science (CORTS), The Pennsylvania State University College of Medicine, Hershey, Pennsylvania 17033, USA
- Department of Pharmacology, The Pennsylvania State University College of Medicine, Hershey, Pennsylvania 17033, USA
| | - Reyad A. Elbarbary
- Department of Orthopaedics and Rehabilitation, The Pennsylvania State University College of Medicine, Hershey, Pennsylvania 17033, USA
- Center for Orthopaedic Research and Translational Science (CORTS), The Pennsylvania State University College of Medicine, Hershey, Pennsylvania 17033, USA
- Department of Biochemistry and Molecular Biology, The Pennsylvania State University College of Medicine, Hershey, Pennsylvania 17033, USA
- Center for RNA Molecular Biology, Pennsylvania State University, University Park, Pennsylvania 16802, USA
| |
Collapse
|
13
|
Zhao K, Nie H, Tang Z, Chen G, Huang J. Paroxetine protects against bleomycin-induced pulmonary fibrosis by blocking GRK2/Smad3 pathway. Aging (Albany NY) 2023; 15:10524-10539. [PMID: 37815883 PMCID: PMC10599755 DOI: 10.18632/aging.205092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Accepted: 05/09/2023] [Indexed: 10/12/2023]
Abstract
G protein-coupled receptor kinase-2 (GRK2) is involved in TGF-β1-induced activation of lung fibroblasts, which could give rise to the pathogenesis of pulmonary fibrosis. Paroxetine (PRXT) serves as a selective GRK2 inhibitor which is widely used to treat anxiety and depression for several decades. However, whether PRXT could inhibit TGF-β1-induced activation of lung fibroblasts and combat bleomycin-induced pulmonary fibrosis remains unclear. Here, we investigated the effects of PRXT on pulmonary fibrosis in C57/BL6 caused by bleomycin as well as on the activation of murine primary lung fibroblasts stimulated with TGF-β1. The results demonstrated that PRXT markedly improved the pulmonary function and 21-day survival in bleomycin-induced mice. Meanwhile, PRXT significantly decreased collagen deposition, inflammation, and oxidative stress in lung tissues from bleomycin-induced mice. Furthermore, we found that PRXT could inhibit the protein and mRNA expression of GRK2 and Smad3 in lung tissues from bleomycin-induced mice. In vitro experiments also PRXT could inhibit cell activation and collagen synthesis in a concentration-dependent manner in TGF-β1-induced lung fibroblasts. In addition, we found that Smad3 overexpression by adenovirus transfection could offset anti-fibrotic and antioxidative effects from PRXT in TGF-β1-induced lung fibroblasts, which showed no effects on the protein expression of GRK2. In conclusion, PRXT mediates the inhibition of GRK2, which further blocks the transcription of Smad3 in TGF-β1-induced lung fibroblasts, providing an attractive therapeutic target for pulmonary fibrosis.
Collapse
Affiliation(s)
- Kaochang Zhao
- Department of Respiratory and Critical Care Medicine, Renmin Hospital of Wuhan University, Wuhan 430060, Hubei Province, China
| | - Hanxiang Nie
- Department of Respiratory and Critical Care Medicine, Renmin Hospital of Wuhan University, Wuhan 430060, Hubei Province, China
| | - Zheng Tang
- Department of Thoracic Surgery, Zhongnan Hospital of Wuhan University, Wuhan 430060, Hubei Province, China
| | - Guozhong Chen
- Department of Respiratory and Critical Care Medicine, Renmin Hospital of Wuhan University, Wuhan 430060, Hubei Province, China
| | - Jizhen Huang
- Department of Respiratory and Critical Care Medicine, Renmin Hospital of Wuhan University, Wuhan 430060, Hubei Province, China
| |
Collapse
|
14
|
Zheng X, Qiu J, Gao N, Jiang T, Li Z, Zhang W, Gong Y, Hong Z, Hong H. Paroxetine Attenuates Chondrocyte Pyroptosis and Inhibits Osteoclast Formation by Inhibiting NF-κB Pathway Activation to Delay Osteoarthritis Progression. Drug Des Devel Ther 2023; 17:2383-2399. [PMID: 37605762 PMCID: PMC10440089 DOI: 10.2147/dddt.s417598] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Accepted: 08/06/2023] [Indexed: 08/23/2023] Open
Abstract
Background Osteoarthritis (OA), a common chronic joint disease, is characterized by cartilage degeneration and subchondral bone reconstruction. NF-κB signaling pathway-activated inflammation and NLRP3-induced pyroptosis play essential roles in the development of OA. In this study, we examine whether paroxetine can inhibit pyroptosis and reduce osteoclast formation, thereby delaying the destruction of knee joints. Methods We employed high-density cultures, along with quantitative polymerase chain reactions and Western blotting techniques, to investigate the effects of paroxetine on extracellular matrix synthesis and degradation. The expression levels of NF-κB and pyroptosis-related signaling pathway proteins were examined by Western blotting and immunofluorescence. Furthermore, the impact of paroxetine on RANKL-induced osteoclast formation was evaluated through TRAP staining and F-actin ring fluorescence detection. To investigate the role of paroxetine in vivo, we constructed a mouse model with destabilization of the medial meniscus (DMM) surgery. Safranin O-Fast Green staining, Hematoxylin-Eosin staining, and immunohistochemistry were conducted to observe the extent of knee joint cartilage deformation. In addition, TRAP staining was used to observe the formation of osteoclasts in the subchondral bone. Results In the in vitro experiments with ATDC5, paroxetine treatment attenuated IL-1β-induced activation of the pyroptosis-related pathway and suppressed extracellular matrix catabolism by inhibiting the NF-kB signaling pathway. In addition, paroxetine treatment decreased the expression of RANKL-induced osteoclast marker genes and reduced osteoclast formation. In animal experiments conducted in vivo, mice treated with paroxetine exhibited thicker knee cartilage with a smoother surface compared to the DMM group. Additionally, the formation of osteoclasts in the subchondral bone was reduced in the paroxetine-treated mice. Further analysis revealed that paroxetine treatment played a role in preserving the balance of the extracellular matrix and delaying knee joint degeneration. Conclusion Paroxetine can inhibit pyroptosis and reduce osteoclast formation via inhibiting the NF-κB signaling pathway, suggesting that it may have therapeutic effects in patients with OA.
Collapse
Affiliation(s)
- Xiaohang Zheng
- Orthopedic Department, Taizhou Hospital Affiliated to Wenzhou Medical University, Linhai, People’s Republic of China
- Enze Medical Research Center, Taizhou Hospital Affiliated to Wenzhou Medical University, Linhai, People’s Republic of China
| | - Jianxin Qiu
- Orthopedic Department, Taizhou Hospital Affiliated to Wenzhou Medical University, Linhai, People’s Republic of China
- Enze Medical Research Center, Taizhou Hospital Affiliated to Wenzhou Medical University, Linhai, People’s Republic of China
| | - Ning Gao
- Department of Cardiovascular Surgery, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, People’s Republic of China
| | - Ting Jiang
- Orthopedic Department, Taizhou Hospital Affiliated to Wenzhou Medical University, Linhai, People’s Republic of China
- Enze Medical Research Center, Taizhou Hospital Affiliated to Wenzhou Medical University, Linhai, People’s Republic of China
| | - Ze Li
- Orthopedic Department, Taizhou Hospital Affiliated to Wenzhou Medical University, Linhai, People’s Republic of China
- Enze Medical Research Center, Taizhou Hospital Affiliated to Wenzhou Medical University, Linhai, People’s Republic of China
| | - Weikang Zhang
- Orthopedic Department, Taizhou Hospital Affiliated to Wenzhou Medical University, Linhai, People’s Republic of China
- Enze Medical Research Center, Taizhou Hospital Affiliated to Wenzhou Medical University, Linhai, People’s Republic of China
| | - Yuhang Gong
- Orthopedic Department, Taizhou Hospital Affiliated to Wenzhou Medical University, Linhai, People’s Republic of China
- Enze Medical Research Center, Taizhou Hospital Affiliated to Wenzhou Medical University, Linhai, People’s Republic of China
| | - Zhenghua Hong
- Orthopedic Department, Taizhou Hospital Affiliated to Wenzhou Medical University, Linhai, People’s Republic of China
- Enze Medical Research Center, Taizhou Hospital Affiliated to Wenzhou Medical University, Linhai, People’s Republic of China
| | - Huaxing Hong
- Orthopedic Department, Taizhou Hospital Affiliated to Wenzhou Medical University, Linhai, People’s Republic of China
- Enze Medical Research Center, Taizhou Hospital Affiliated to Wenzhou Medical University, Linhai, People’s Republic of China
| |
Collapse
|
15
|
Xiong B, Chen L, Huang Y, Lu G, Chen C, Nong J, Pan H. ZBTB16 eases lipopolysaccharide‑elicited inflammation, apoptosis and degradation of extracellular matrix in chondrocytes during osteoarthritis by suppressing GRK2 transcription. Exp Ther Med 2023; 25:276. [PMID: 37206562 PMCID: PMC10189728 DOI: 10.3892/etm.2023.11975] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Accepted: 01/23/2023] [Indexed: 05/21/2023] Open
Abstract
Osteoarthritis (OA) is a chronic degenerative disease of the bone that is a major contributor of disability in the elderly population. Zinc finger and BTB domain-containing 16 (ZBTB16) is a transcription factor that has been previously revealed to be impaired in human OA tissues. The present study was designed to elaborate the potential impact of ZBTB16 on OA and to possibly assess any latent regulatory mechanism. ZBTB16 expression in human OA tissues was examined using the Gene Expression Series (GSE) database (https://www.ncbi.nlm.nih.gov/geo/query/acc.cgi?acc=GSE169077) whereas ZBTB16 expression in chondrocytes was examined using reverse transcription-quantitative PCR (RT-qPCR) and western blotting. Cell viability was examined using a Cell Counting Kit-8 assay. A TUNEL assay and western blotting were used to assess cell apoptosis and apoptosis-related markers, including Bcl-2, Bax and cleaved caspase-3. The levels and expression of inflammatory factors, including TNF-α, IL-1β and IL-6, were determined by ELISA and western blotting. RT-qPCR and western blotting were also used to analyze the expression levels of extracellular matrix (ECM)-degrading enzymes, including MMP-13, a disintegrin-like and metalloproteinase with thrombospondin type-1 motifs-5, aggrecan and collagen type II α1. After the potential binding of ZBTB16 with the G protein coupled receptor kinase type 2 (GRK2) promoter was predicted using the Cistrome DB database, GRK2 expression was confirmed by RT-qPCR and western blotting. Chromatin immunoprecipitation and luciferase reporter assays were then used to determine the potential interaction between ZBTB16 and the GRK2 promoter. Following GRK2 overexpression in ZBTB16-overexpressing chondrocytes by co-transfection of GRK2 and ZBTB16 overexpression plasmids, the aforementioned functional experiments were performed again. ZBTB16 expression was found to be reduced in human OA tissues compared with in normal cartilage tissues and lipopolysaccharide (LPS)-stimulated chondrocytes. ZBTB16 overexpression increased cell viability whilst decreasing apoptosis, inflammation and ECM degradation by LPS-treated chondrocytes. In addition, GRK2 expression was found to be increased in LPS-stimulated chondrocytes. ZBTB16 successfully bound to the GRK2 promoter, which negatively modulated GRK2 expression. GRK2 upregulation reversed the effects of ZBTB16 overexpression on the viability, apoptosis, inflammation and ECM degradation by LPS-challenged chondrocytes. In conclusion, these data suggest that ZBTB16 may inhibit the development of OA through the transcriptional inactivation of GRK2.
Collapse
Affiliation(s)
- Bo Xiong
- Department of Orthopaedics and Traumatology, First Affiliated Hospital of The Guangxi University of Chinese Medicine, Nanning, Guangxi 530000, P.R. China
| | - Lihua Chen
- Department of Orthopaedics and Traumatology, First Affiliated Hospital of The Guangxi University of Chinese Medicine, Nanning, Guangxi 530000, P.R. China
| | - Yue Huang
- Department of Orthopaedics and Traumatology, First Affiliated Hospital of The Guangxi University of Chinese Medicine, Nanning, Guangxi 530000, P.R. China
| | - Guanyu Lu
- Department of Orthopaedics and Traumatology, First Affiliated Hospital of The Guangxi University of Chinese Medicine, Nanning, Guangxi 530000, P.R. China
| | - Cai Chen
- Department of Orthopaedics and Traumatology, First Affiliated Hospital of The Guangxi University of Chinese Medicine, Nanning, Guangxi 530000, P.R. China
| | - Jiao Nong
- Teaching Department, First Affiliated Hospital of The Guangxi University of Chinese Medicine, Nanning, Guangxi 530000, P.R. China
| | - Haida Pan
- Department of Orthopaedics and Traumatology, Yongjia County Traditional Chinese Medicine Hospital, Wenzhou, Zhejiang 325100, P.R. China
- Correspondence to: Dr Haida Pan, Department of Orthopaedics and Traumatology, Yongjia County Traditional Chinese Medicine Hospital, 6 Park Road, Jiangbei Street, Yongjia, Wenzhou, Zhejiang 325100, P.R. China
| |
Collapse
|
16
|
Zhang J, Zhang X, Lu M, Chang Y, Wang Q, Tu J, Wu H, Wang C, Hong Z, Xiong M, Song L, Wei W. GRK2 Mediates Macrophage Polarization by Regulating EP4-cAMP-pCREB Signaling in Ulcerative Colitis and the Therapeutic Effect of Paroxetine on Mice with DSS-Induced Colitis. Pharmaceuticals (Basel) 2023; 16:ph16050664. [PMID: 37242446 DOI: 10.3390/ph16050664] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Revised: 04/15/2023] [Accepted: 04/25/2023] [Indexed: 05/28/2023] Open
Abstract
G protein-coupled receptor kinase 2 (GRK2) is one of the cytosolic enzymes, and GRK2 translocation induces prostaglandin E2 receptor 4 (EP4) over-desensitization and reduces the level of cyclic adenosine monophosphate (cAMP) to regulate macrophage polarization. However, the role of GRK2 in the pathophysiology of ulcerative colitis (UC) remains unclear. In this study, we investigated the role of GRK2 in macrophage polarization in UC, using biopsies from patients, a GRK2 heterozygous mouse model with dextran sulfate sodium (DSS)-induced colitis, and THP-1 cells. The results showed that a high level of prostaglandin E2 (PGE2) stimulated the receptor EP4 and enhanced the transmembrane activity of GRK2 in colonic lamina propria mononuclear cells (LPMCs), resulting in a down-regulation of membrane EP4 expression. Then, the suppression of cAMP-cyclic AMP responsive element-binding (CREB) signal inhibited M2 polarization in UC. Paroxetine is acknowledged as one of the selective serotonin reuptake inhibitors (SSRI), which is also considered as a potent GRK2 inhibitor with a high selectivity for GRK2. We found that paroxetine could alleviate symptoms of DSS-induced colitis in mice by regulating GPCR signaling to affect macrophage polarization. Taken together, the current results show that GRK2 may act as a novel therapeutic target in UC by regulating macrophage polarization, and paroxetine as a GRK2 inhibitor may have therapeutic effect on mice with DSS-induced colitis.
Collapse
Affiliation(s)
- Jiawei Zhang
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Centre of Anti-Inflammatory and Immune Medicine, Hefei 230032, China
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei 230022, China
| | - Xianzheng Zhang
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Centre of Anti-Inflammatory and Immune Medicine, Hefei 230032, China
| | - Mingdian Lu
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei 230022, China
| | - Yan Chang
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Centre of Anti-Inflammatory and Immune Medicine, Hefei 230032, China
| | - Qingtong Wang
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Centre of Anti-Inflammatory and Immune Medicine, Hefei 230032, China
| | - Jiajie Tu
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Centre of Anti-Inflammatory and Immune Medicine, Hefei 230032, China
| | - Huaxun Wu
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Centre of Anti-Inflammatory and Immune Medicine, Hefei 230032, China
| | - Chun Wang
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Centre of Anti-Inflammatory and Immune Medicine, Hefei 230032, China
| | - Zhongyang Hong
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Centre of Anti-Inflammatory and Immune Medicine, Hefei 230032, China
| | - Maoming Xiong
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei 230022, China
| | - Lihua Song
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Centre of Anti-Inflammatory and Immune Medicine, Hefei 230032, China
| | - Wei Wei
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Centre of Anti-Inflammatory and Immune Medicine, Hefei 230032, China
| |
Collapse
|
17
|
Wang T, Zhang H, Han Y, Zheng Q, Liu H, Han M, Li Z. Reversing T Cell Dysfunction to Boost Glioblastoma Immunotherapy by Paroxetine-Mediated GRK2 Inhibition and Blockade of Multiple Checkpoints through Biomimetic Nanoparticles. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2204961. [PMID: 36698265 PMCID: PMC10037995 DOI: 10.1002/advs.202204961] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 12/02/2022] [Indexed: 05/19/2023]
Abstract
T cell dysfunction-induced tumor immune escape is particularly severe in glioblastoma (GBM), and significantly affects the efficacy of immunotherapy. It is crucial to innovatively reverse the T cell dysfunction for improving GBM immunotherapy. Herein, T cell dysfunction is remarkably reversed and immunotherapy of GBM is boosted by repurposing the U. S. Food and Drug Administration-approved antidepressant paroxetine (PX) with biomimetic nanoparticles (CS-J@CM/6 NPs). The PX is successfully applied to abrogate T cell sequestration in the bone marrow of GBM-bearing mice and increase their infiltration in tumor. The biomimetic NPs are composed of ultrasmall Cu2- x Se NPs, JQ1, and tumor cell membrane modified with CD6, and are efficiently delivered into tumor through the specific interactions between CD6 and activated leukocyte cell adhesion molecule. They ameliorate the T cell dysfunction through the double roles of loaded JQ1, which simultaneously decreases the expression of PD-1 and TIM-3 on T cells, and the expression of PD-L1 on tumor cells. The NP also induces the immunogenic cell death of tumor cells to activate immune response. The synergistic roles of PX and biomimetic CS-J@CM/6 NPs notably enhance the survival of GBM-bearing mice. This work provides new insights into tumor immunotherapy by repurposing "old drugs" with advanced NPs.
Collapse
Affiliation(s)
- Tingting Wang
- Center for Molecular Imaging and Nuclear MedicineState Key Laboratory of Radiation Medicine and ProtectionSchool for Radiological and Interdisciplinary Sciences (RAD‐X)Suzhou Medical College of Soochow UniversityCollaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education InstitutionsSuzhou215123P. R. China
| | - Hao Zhang
- Center for Molecular Imaging and Nuclear MedicineState Key Laboratory of Radiation Medicine and ProtectionSchool for Radiological and Interdisciplinary Sciences (RAD‐X)Suzhou Medical College of Soochow UniversityCollaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education InstitutionsSuzhou215123P. R. China
| | - Yaobao Han
- Center for Molecular Imaging and Nuclear MedicineState Key Laboratory of Radiation Medicine and ProtectionSchool for Radiological and Interdisciplinary Sciences (RAD‐X)Suzhou Medical College of Soochow UniversityCollaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education InstitutionsSuzhou215123P. R. China
| | - Qing Zheng
- Center for Molecular Imaging and Nuclear MedicineState Key Laboratory of Radiation Medicine and ProtectionSchool for Radiological and Interdisciplinary Sciences (RAD‐X)Suzhou Medical College of Soochow UniversityCollaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education InstitutionsSuzhou215123P. R. China
| | - Hanghang Liu
- Center for Molecular Imaging and Nuclear MedicineState Key Laboratory of Radiation Medicine and ProtectionSchool for Radiological and Interdisciplinary Sciences (RAD‐X)Suzhou Medical College of Soochow UniversityCollaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education InstitutionsSuzhou215123P. R. China
| | - Mengxiao Han
- Center for Molecular Imaging and Nuclear MedicineState Key Laboratory of Radiation Medicine and ProtectionSchool for Radiological and Interdisciplinary Sciences (RAD‐X)Suzhou Medical College of Soochow UniversityCollaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education InstitutionsSuzhou215123P. R. China
| | - Zhen Li
- Center for Molecular Imaging and Nuclear MedicineState Key Laboratory of Radiation Medicine and ProtectionSchool for Radiological and Interdisciplinary Sciences (RAD‐X)Suzhou Medical College of Soochow UniversityCollaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education InstitutionsSuzhou215123P. R. China
| |
Collapse
|
18
|
Dravid AA, Dhanabalan KM, Naskar S, Vashistha A, Agarwal S, Padhan B, Dewani M, Agarwal R. Sustained release resolvin D1 liposomes are effective in the treatment of osteoarthritis in obese mice. J Biomed Mater Res A 2023; 111:765-777. [PMID: 36773024 DOI: 10.1002/jbm.a.37512] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 01/24/2023] [Accepted: 01/31/2023] [Indexed: 02/12/2023]
Abstract
Osteoarthritis (OA) is the most common joint disorder and currently affects >500 million patients worldwide, with ~60% of them also suffering from obesity. There is no drug approved for human use that changes the course of OA progression. OA is one of the most common comorbidities of obesity, and obesity-related OA (ObOA) is a serious health concern because it shows heightened severity of tissue damage and also predominantly affects the working population. Unresolved inflammation is a major driver of ObOA, thus, resolving disease-associated inflammation is a viable strategy to treat ObOA. Resolvins are highly potent molecules that play a role in the resolution of inflammation and promote tissue healing. However, small molecules (like Resolvin D1; RvD1) have to be administered frequently or prior to injury because they lose their in vivo activity rapidly either by lymphatic clearance, or oxidation-mediated deactivation. In this study, we have encapsulated RvD1 in liposomes and established its efficacy in the mouse model of ObOA at much lower dosages than freely administered RvD1. Liposomal RvD1 (lipo-RvD1) acted as a source of the RvD1 molecules for ~11 days in vitro in synovial fluid derived from patients. When administered prophylactically or therapeutically, lipo-RvD1 suppressed cartilage damage in male C57BL/6 mice compared to untreated and free RvD1 treatments. This efficacy was achieved by increasing the proportion of the proresolution M2 macrophages over proinflammatory M1 macrophages in the synovial membrane. These results show the potential of lipo-RvD1 as an anti-OA agent.
Collapse
|
19
|
Han S. Osteoarthritis year in review 2022: biology. Osteoarthritis Cartilage 2022; 30:1575-1582. [PMID: 36150676 DOI: 10.1016/j.joca.2022.09.003] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2022] [Revised: 09/07/2022] [Accepted: 09/08/2022] [Indexed: 02/02/2023]
Abstract
The field of osteoarthritis (OA) biology is rapidly evolving and brilliant progress has been made this year as well. Landmark studies of OA biology published in 2021 and early 2022 were selected through PubMed search by personal opinion. These papers were classified by their molecular mechanisms, and it was largely divided into the intracellular signaling mechanisms and the inter-compartment interaction in chondrocyte homeostasis and OA progression. The intracellular signaling mechanisms involving OA progression included (1) Piezo1/transient receptor potential channels of the vanilloid subtype (TRPV) 4-mediated calcium signaling, (2) mechanical load-F-box and WD repeat domain containing 7 (FBXW7) in chondrocyte senescence, (3) mechanical loading-primary cilia-hedgehog signaling, (4) low grade inflammation by toll-like receptor (TLR)-CD14-lipopolysaccharide-binding protein (LBP) complex and inhibitor of NF-κB kinase (IKK) β-nuclear factor kappa B (NF-κB) signaling, (5) selenium pathway and reactive oxygen species (ROS) production, (6) G protein-coupled receptor (GPCR) and cyclic adenosine monophosphate (cAMP) signaling, (7) peroxisome proliferator-activated receptor α (PPARα)-acyl-CoA thioesterase 12 (ACOT12)-mediated de novo lipogenesis and (8) hypoxia-disruptor of telomeric silencing 1-like (DOT1L)-H3-lysine 79 (H3K79) methylation pathway. The studies on inter-compartment or intercellular interaction in OA progression included the following subjects; (1) the anabolic role of lubricin, glycoprotein from superficial zone cells, (2) osteoclast-chondrocyte interaction via exosomal miRNA and sphingosine 1-phosphate (S1P), (3) senescent fibroblast-like synoviocyte and chondrocyte interaction, (4) synovial macrophage and chondrocyte interaction through Flightless I, (5) αV integrin-mediated transforming growth factor beta (TGFβ) activation by mechanical loading, and (6) osteocytic TGFβ in subchondral bone thickening. Despite the disastrous Covid-19 pandemic, many outstanding studies have expanded the boundary of OA biology. They provide both critical insight into the pathophysiology as well as clues for the treatment of OA.
Collapse
Affiliation(s)
- S Han
- Laboratory for for Arthritis and Cartilage Biology, Research Institute of Aging and Metabolism, Kyungpook National University, Daegu, Republic of Korea; Department of Internal Medicine, School of Medicine, Kyungpook National University, Daegu, Republic of Korea.
| |
Collapse
|
20
|
Hong Z, Tie Q, Zhang L. Targeted inhibition of the GRK2/HIF-1α pathway is an effective strategy to alleviate synovial hypoxia and inflammation. Int Immunopharmacol 2022; 113:109271. [PMID: 36461590 DOI: 10.1016/j.intimp.2022.109271] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Revised: 09/19/2022] [Accepted: 09/20/2022] [Indexed: 11/11/2022]
Abstract
G-protein coupled receptor (GPCR) kinases (GRKs) and hypoxia-inducible factor-1α (HIF-1α) play key roles in rheumatoid arthritis (RA). Several studies have demonstrated that HIF-1α expression is positively regulated by GRK2, suggesting its posttranscriptional effects on HIF-1α. In this study, we review the role of HIF-1α and GRK2 in RA pathophysiology, focusing on their proinflammatory roles in immune cells and fibroblast-like synoviocytes (FLS).We then introduce several drugs that inhibit GRK2 and HIF-1α, and briefly outline their molecular mechanisms. We conclude by presenting gaps in knowledge and our prospects for the pharmacological potential of targeting these proteins and the relevant downstream signaling pathways.Future research is warranted and paramount for untangling these novel and promising roles for GRK2 and HIF-1α in RA.
Collapse
Affiliation(s)
- Zhongyang Hong
- Department of Pharmacy, Affiliated the Jianhu People's Hospital, Yancheng 224700, China.
| | - Qingsong Tie
- Department of Pharmacy, Affiliated the Jianhu People's Hospital, Yancheng 224700, China.
| | - Lingling Zhang
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Centre of Anti-Inflammatory and Immune Medicine, Center of Rheumatoid Arthritis of Anhui Medical University, Hefei 230032, China.
| |
Collapse
|
21
|
Wang W, Niu Y, Jia Q. Physical therapy as a promising treatment for osteoarthritis: A narrative review. Front Physiol 2022; 13:1011407. [PMID: 36311234 PMCID: PMC9614272 DOI: 10.3389/fphys.2022.1011407] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Accepted: 09/23/2022] [Indexed: 11/13/2022] Open
Abstract
Osteoarthritis (OA) is the most prevalent joint disease and a leading cause of disability in older adults. With an increasing population ageing and obesity, OA is becoming even more prevalent than it was in previous decades. Evidence indicates that OA is caused by the breakdown of joint tissues from mechanical loading and inflammation, but the deeper underlying mechanism of OA pathogenesis remains unclear, hindering efforts to prevent and treat this disease. Pharmacological treatments are mostly related to relieving symptoms, and there is no drug for radical cure. However, compelling evidence suggests that regular practice of resistance exercise may prevent and control the development of several musculoskeletal chronic diseases including OA, which may result in improved quality of life of the patients. In this review, we introduced the current understanding of the mechanism and clinical treatments of OA pathogenesis. We also reviewed the recent study of physical therapy in the treatment of skeletal system disorders, especially in OA. Finally, we discuss the present challenges and promising advantages of physical therapy in OA treatment.
Collapse
Affiliation(s)
- Wei Wang
- School of Physical Education, Anyang Normal University, Anyang, China
- Anyang Key Laboratory of Fitness Training and Assessment, Anyang Normal University, Anyang, China
| | - Yonggang Niu
- School of Physical Education, Anyang Normal University, Anyang, China
- Anyang Key Laboratory of Fitness Training and Assessment, Anyang Normal University, Anyang, China
| | - Qingxiu Jia
- School of Physical Education, Anyang Normal University, Anyang, China
- Anyang Key Laboratory of Fitness Training and Assessment, Anyang Normal University, Anyang, China
- *Correspondence: Qingxiu Jia,
| |
Collapse
|
22
|
Thapaliya M, Amponnawarat A, Tesmer JJG, Ali H. GRK2 inhibitors, paroxetine and CCG258747, attenuate IgE-mediated anaphylaxis but activate mast cells via MRGPRX2 and MRGPRB2. Front Immunol 2022; 13:1032497. [PMID: 36275707 PMCID: PMC9583242 DOI: 10.3389/fimmu.2022.1032497] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Accepted: 09/21/2022] [Indexed: 11/16/2022] Open
Abstract
G protein-coupled receptor (GPCR) kinase 2 (GRK2), which phosphorylates agonist-occupied GPCRs to promote their desensitization, has been investigated as an attractive therapeutic target for cardiovascular and metabolic diseases. Several GRK2-targeted inhibition strategies have been reported including the use of direct pharmacological inhibitors such as paroxetine (a widely prescribed antidepressant) and its analogs such as compound CCG258747. Cross-linking of high affinity IgE receptor (FcϵRI) on mast cells (MCs) and the resulting degranulation causes anaphylaxis and allergic asthma. Using gene silencing strategy, we recently showed that GRK2 contributes to FcεRI signaling and MC degranulation. The purpose of this study was to determine if the GRK2 inhibitors paroxetine and CCG258747 modulate FcεRI-mediated MC responses in vitro and in vivo. Utilizing rat basophilic leukemia (RBL-2H3) cells and primary mouse lung MCs (LMCs), we found that paroxetine and CCG258747 inhibit FcϵRI-mediated calcium mobilization and degranulation. Furthermore, intravenous administration of paroxetine and CCG258747 in mice resulted in substantial reduction of IgE-mediated passive cutaneous anaphylaxis. Unlike LMCs, human cutaneous MCs abundantly express a novel GPCR known as MRGPRX2 (mouse; MRGPRB2). We found that in contrast to their inhibitory effects on FcεRI-mediated MC responses, both paroxetine and CCG258747 induce calcium mobilization and degranulation in RBL-2H3 cells stably expressing MRGPRX2 but not in untransfected cells. Furthermore, paroxetine and CCG258747 induced degranulation in peritoneal MCs from Wild-type (WT) mice in vitro and caused increased cutaneous vascular permeability in vivo, but these responses were substantially reduced in Mrgprb2-/- mice. Additionally, upon intradermal injection, paroxetine also induced neutrophil recruitment in WT but not Mrgprb2-/- mice. These findings suggest that in addition to their potential therapeutic utility against cardiovascular and metabolic disorders, paroxetine-based GRK2-inhibitors may serve to modulate IgE-mediated anaphylaxis and to enhance cutaneous host defense by harnessing MC's immunomodulatory property through the activation of MRGPRX2/MRGPRB2.
Collapse
Affiliation(s)
- Monica Thapaliya
- Department of Basic and Translational Sciences, University of Pennsylvania, School of Dental Medicine, Philadelphia, PA, United States
| | - Aetas Amponnawarat
- Department of Basic and Translational Sciences, University of Pennsylvania, School of Dental Medicine, Philadelphia, PA, United States,Department of Family and Community Dentistry, Faculty of Dentistry, Chiang Mai University, Chiang Mai, Thailand
| | - John J. G. Tesmer
- Departments of Biological Sciences and of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN, United States
| | - Hydar Ali
- Department of Basic and Translational Sciences, University of Pennsylvania, School of Dental Medicine, Philadelphia, PA, United States,*Correspondence: Hydar Ali,
| |
Collapse
|
23
|
Single cell RNA-seq analysis identifies ferroptotic chondrocyte cluster and reveals TRPV1 as an anti-ferroptotic target in osteoarthritis. EBioMedicine 2022; 84:104258. [PMID: 36137413 PMCID: PMC9494174 DOI: 10.1016/j.ebiom.2022.104258] [Citation(s) in RCA: 60] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Revised: 08/25/2022] [Accepted: 08/25/2022] [Indexed: 12/29/2022] Open
|
24
|
Kwak JS, Lee Y, Yang J, Kim SK, Shin Y, Kim HJ, Choi JH, Im YJ, Kim MJ, Lee Yu K, Chang You J, Chun JS. Characterization of rhodanine derivatives as potential disease-modifying drugs for experimental mouse osteoarthritis. Osteoarthritis Cartilage 2022; 30:1210-1221. [PMID: 35513246 DOI: 10.1016/j.joca.2022.04.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Revised: 04/22/2022] [Accepted: 04/26/2022] [Indexed: 02/02/2023]
Abstract
OBJECTIVE This study was performed to characterize selected rhodanine derivatives as potential preclinical disease-modifying drugs for experimental osteoarthritis (OA) in mice. METHODS Three rhodanine derivatives, designated rhodanine (R)-501, R-502, and R-503, were selected as candidate OA disease-modifying drugs. Their effects were evaluated by intra-articular (IA) injection in OA mouse models induced by DMM (destabilization of the medial meniscus) or adenoviral overexpression in joint tissues of hypoxia-inducible factor (HIF)-2α or zinc importer ZIP8. The regulatory mechanisms impacted by the rhodanine derivatives were examined in primary-culture chondrocytes and fibroblast-like synoviocytes (FLS). RESULTS All three rhodanine derivatives inhibited OA development caused by DMM or overexpression of HIF-2α or ZIP8. Compared to vehicle-treated group, for example, IA injection of R-501 in DMM-operated mice reduced median OARSI grade from 3.78 (IQR 3.00-5.00) to 1.89 (IQR 0.94-2.00, P = 0.0001). R-502 and R-503 also reduced from 3.67 (IQR 2.11-4.56) to 2.00 (IQR 1.00-2.00, P = 0.0030) and 2.00 (IQR 1.83-2.67, P = 0.0378), respectively. Mechanistically, the rhodanine derivatives inhibited the nuclear localization and transcriptional activity of HIF-2α in chondrocytes and FLS. They did not bind to Zn2+ or modulate Zn2+ homeostasis in chondrocytes or FLS; instead, they inhibited the nuclear localization and transcriptional activity of the Zn2+-dependent transcription factor, MTF1. HIF-2α, ZIP8, and interleukin-1β could upregulate matrix-degrading enzymes in chondrocytes and FLS, and the rhodanine derivatives inhibited these effects. CONCLUSION IA administration of rhodanine derivatives significantly reduced OA pathogenesis in various mouse models, demonstrating that these derivatives have disease-modifying therapeutic potential against OA pathogenesis.
Collapse
Affiliation(s)
- J-S Kwak
- National Creative Research Initiatives Center for Osteoarthritis Pathogenesis and School of Life Sciences, Gwangju Institute of Science and Technology, Gwangju, 61005, Republic of Korea
| | - Y Lee
- National Creative Research Initiatives Center for Osteoarthritis Pathogenesis and School of Life Sciences, Gwangju Institute of Science and Technology, Gwangju, 61005, Republic of Korea
| | - J Yang
- National Creative Research Initiatives Center for Osteoarthritis Pathogenesis and School of Life Sciences, Gwangju Institute of Science and Technology, Gwangju, 61005, Republic of Korea
| | - S K Kim
- National Creative Research Initiatives Center for Osteoarthritis Pathogenesis and School of Life Sciences, Gwangju Institute of Science and Technology, Gwangju, 61005, Republic of Korea
| | - Y Shin
- National Creative Research Initiatives Center for Osteoarthritis Pathogenesis and School of Life Sciences, Gwangju Institute of Science and Technology, Gwangju, 61005, Republic of Korea
| | - H-J Kim
- National Creative Research Initiatives Center for Osteoarthritis Pathogenesis and School of Life Sciences, Gwangju Institute of Science and Technology, Gwangju, 61005, Republic of Korea
| | - J H Choi
- National Creative Research Initiatives Center for Osteoarthritis Pathogenesis and School of Life Sciences, Gwangju Institute of Science and Technology, Gwangju, 61005, Republic of Korea
| | - Y J Im
- College of Pharmacy, Chonnam National University, Gwangju, 61186, Republic of Korea
| | - M-J Kim
- Avixgen Inc., Seoul, 06649, Republic of Korea
| | - K Lee Yu
- National Research Laboratory for Molecular Virology, Department of Pathology, School of Medicine, The Catholic University of Korea, Seoul, 06591, Republic of Korea
| | - J Chang You
- Avixgen Inc., Seoul, 06649, Republic of Korea; National Research Laboratory for Molecular Virology, Department of Pathology, School of Medicine, The Catholic University of Korea, Seoul, 06591, Republic of Korea
| | - J-S Chun
- National Creative Research Initiatives Center for Osteoarthritis Pathogenesis and School of Life Sciences, Gwangju Institute of Science and Technology, Gwangju, 61005, Republic of Korea.
| |
Collapse
|
25
|
Mucke HA. Drug Repurposing Patent Applications March–June 2022. Assay Drug Dev Technol 2022; 20:286-293. [DOI: 10.1089/adt.2022.065] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
|
26
|
Loucks A, Maerz T, Hankenson K, Moeser A, Colbath A. WITHDRAWN: The Multifaceted Role of Mast Cells in Joint Inflammation and Arthritis. OSTEOARTHRITIS AND CARTILAGE OPEN 2022. [DOI: 10.1016/j.ocarto.2022.100309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
|
27
|
Karuppagounder V, Pinamont W, Yoshioka N, Elbarbary R, Kamal F. Early Gβγ-GRK2 Inhibition Ameliorates Osteoarthritis Development by Simultaneous Anti-Inflammatory and Chondroprotective Effects. Int J Mol Sci 2022; 23:ijms23147933. [PMID: 35887281 PMCID: PMC9323311 DOI: 10.3390/ijms23147933] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Revised: 07/14/2022] [Accepted: 07/17/2022] [Indexed: 12/12/2022] Open
Abstract
The G-protein-coupled receptor kinase 2 (GRK2) is an important regulator of inflammation and pathological macrophage phenotype in a variety of diseases. We hypothesize that Gβγ-GRK2 signaling promotes the early inflammatory response and chondrocyte loss in osteoarthritis (OA). Using the destabilization of the medial meniscus (DMM) model in 12-week-old male C57BL/6 mice, we determined the role of Gβγ-GRK2 signaling in synovitis, macrophage activation, and OA development. We achieved Gβγ-GRK2 inhibition at the time of DMM by administering the Gβγ inhibitor “gallein” and the GRK2 inhibitor “paroxetine” daily, starting from 2 days before DMM surgery, for a duration of 1 or 12 weeks. Synovial and cartilage structural changes were evaluated by histomorphometry, and molecular events and macrophage activation were examined. We studied the direct role of Gβγ-GRK2 in synovitis and macrophage activation in vitro using SW982 and THP1 cells. Continuous Gβγ-GRK2 inhibition initiated at the time of DMM attenuated OA development and decreased chondrocyte loss more effectively than delayed treatment. GRK2 expression and the M1 macrophage phenotype were elevated in the inflamed synovium, while early gallein and paroxetine treatment for 1 and 12 weeks following DMM resulted in their reduction and an upregulated M2 macrophage phenotype. In vitro experiments showed that Gβγ-GRK2 inhibition attenuated synoviocyte inflammation and the M1 phenotype. We show that early Gβγ-GRK2 inhibition is of higher therapeutic efficacy in OA than delayed inhibition, as it prevents OA development by inhibiting the early inflammatory response.
Collapse
Affiliation(s)
- Vengadeshprabhu Karuppagounder
- Center for Orthopaedic Research and Translational Science (CORTS), Penn State College of Medicine, 500 University Drive, Hershey, PA 17033-0850, USA; (V.K.); (W.P.); (N.Y.)
- Department of Orthopaedics and Rehabilitation, Penn State College of Medicine, 500 University Drive, Hershey, PA 17033-0850, USA
| | - William Pinamont
- Center for Orthopaedic Research and Translational Science (CORTS), Penn State College of Medicine, 500 University Drive, Hershey, PA 17033-0850, USA; (V.K.); (W.P.); (N.Y.)
- Department of Orthopaedics and Rehabilitation, Penn State College of Medicine, 500 University Drive, Hershey, PA 17033-0850, USA
| | - Natalie Yoshioka
- Center for Orthopaedic Research and Translational Science (CORTS), Penn State College of Medicine, 500 University Drive, Hershey, PA 17033-0850, USA; (V.K.); (W.P.); (N.Y.)
- Department of Orthopaedics and Rehabilitation, Penn State College of Medicine, 500 University Drive, Hershey, PA 17033-0850, USA
| | - Reyad Elbarbary
- Center for Orthopaedic Research and Translational Science (CORTS), Penn State College of Medicine, 500 University Drive, Hershey, PA 17033-0850, USA; (V.K.); (W.P.); (N.Y.)
- Department of Orthopaedics and Rehabilitation, Penn State College of Medicine, 500 University Drive, Hershey, PA 17033-0850, USA
- Correspondence: (R.E.); (F.K.); Tel.: +717-531-4808 (F.K.)
| | - Fadia Kamal
- Center for Orthopaedic Research and Translational Science (CORTS), Penn State College of Medicine, 500 University Drive, Hershey, PA 17033-0850, USA; (V.K.); (W.P.); (N.Y.)
- Department of Orthopaedics and Rehabilitation, Penn State College of Medicine, 500 University Drive, Hershey, PA 17033-0850, USA
- Correspondence: (R.E.); (F.K.); Tel.: +717-531-4808 (F.K.)
| |
Collapse
|
28
|
Yoshioka NK, Young GM, Khajuria DK, Karuppagounder V, Pinamont WJ, Fanburg-Smith JC, Abraham T, Elbarbary RA, Kamal F. Structural changes in the collagen network of joint tissues in late stages of murine OA. Sci Rep 2022; 12:9159. [PMID: 35650306 PMCID: PMC9160297 DOI: 10.1038/s41598-022-13062-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Accepted: 04/28/2022] [Indexed: 11/23/2022] Open
Abstract
Osteoarthritis (OA) is the most prevalent degenerative joint disease, resulting in joint pain, impaired movement, and structural changes. As the ability of joint tissue to resist stress is mainly imparted by fibrillar collagens in the extracellular matrix, changes in the composition and structure of collagen fibers contribute to the pathological remodeling observed in OA joints that includes cartilage degeneration, subchondral bone (SCB) sclerosis, and meniscal damage. Using the established OA model of destabilization of the medial meniscus (DMM) in C57BL/6J mice, we performed a comprehensive analysis of the content and structure of collagen fibers in the articular cartilage, subchondral bone, and menisci using complementary techniques, which included second harmonic generation microscopy and immunofluorescence staining. We found that regions exposed to increased mechanical stress in OA mice, typically closest to the site of injury, had increased collagen fiber thickness, dysregulated fiber formation, and tissue specific changes in collagen I and II (Col I and Col II) expression. In cartilage, OA was associated with decreased Col II expression in all regions, and increased Col I expression in the anterior and posterior regions. Col I fiber thickness was increased in all regions with disorganization in the center region. In the superficial SCB, all regions exhibited increased Col I expression and fiber thickness in OA mice; no changes were detected in the deeper regions of the subchondral bone except for increased Col I fiber thickness. In the menisci, OA led to increased Col I and Col II expression in the vascular and avascular regions of the anterior meniscus with increased Col I fiber thickness in these regions. Similar changes were observed only in the vascular region of the posterior meniscus. Our findings provide, for the first time, comprehensive insights into the microarchitectural changes of extracellular matrix in OA and serve as guidelines for studies investigating therapies that target collagenous changes as means to impede the progression of osteoarthritis.
Collapse
Affiliation(s)
- Natalie K Yoshioka
- Center for Orthopedic Research and Translational Sciences (CORTS), Penn State College of Medicine, Hershey, PA, USA
- Department of Orthopedics and Rehabilitation, Penn State College of Medicine, Hershey, PA, USA
| | - Gregory M Young
- Center for Orthopedic Research and Translational Sciences (CORTS), Penn State College of Medicine, Hershey, PA, USA
- Department of Orthopedics and Rehabilitation, Penn State College of Medicine, Hershey, PA, USA
| | - Deepak Kumar Khajuria
- Center for Orthopedic Research and Translational Sciences (CORTS), Penn State College of Medicine, Hershey, PA, USA
- Department of Orthopedics and Rehabilitation, Penn State College of Medicine, Hershey, PA, USA
| | - Vengadeshprabhu Karuppagounder
- Center for Orthopedic Research and Translational Sciences (CORTS), Penn State College of Medicine, Hershey, PA, USA
- Department of Orthopedics and Rehabilitation, Penn State College of Medicine, Hershey, PA, USA
| | - William J Pinamont
- Center for Orthopedic Research and Translational Sciences (CORTS), Penn State College of Medicine, Hershey, PA, USA
- Department of Orthopedics and Rehabilitation, Penn State College of Medicine, Hershey, PA, USA
| | - Julie C Fanburg-Smith
- Department of Pathology, Penn State Health/Milton S. Hershey Medical Center, Hershey, PA, USA
| | - Thomas Abraham
- Department of Neural and Behavioral Science, Penn State University College of Medicine, Hershey, PA, USA
- Microscopy Imaging Facility, Penn State University College of Medicine, Hershey, PA, USA
| | - Reyad A Elbarbary
- Center for Orthopedic Research and Translational Sciences (CORTS), Penn State College of Medicine, Hershey, PA, USA.
- Department of Orthopedics and Rehabilitation, Penn State College of Medicine, Hershey, PA, USA.
- Department of Biochemistry and Molecular Biology, Penn State College of Medicine, Hershey, PA, USA.
| | - Fadia Kamal
- Center for Orthopedic Research and Translational Sciences (CORTS), Penn State College of Medicine, Hershey, PA, USA.
- Department of Orthopedics and Rehabilitation, Penn State College of Medicine, Hershey, PA, USA.
- Department of Pharmacology, Penn State College of Medicine, Hershey, PA, USA.
| |
Collapse
|
29
|
Dravid AA, M. Dhanabalan K, Agarwal S, Agarwal R. Resolvin D1-loaded nanoliposomes promote M2 macrophage polarization and are effective in the treatment of osteoarthritis. Bioeng Transl Med 2022; 7:e10281. [PMID: 35600665 PMCID: PMC9115708 DOI: 10.1002/btm2.10281] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2021] [Revised: 09/19/2021] [Accepted: 09/22/2021] [Indexed: 11/24/2022] Open
Abstract
Current treatments for osteoarthritis (OA) offer symptomatic relief but do not prevent or halt the disease progression. Chronic low-grade inflammation is considered a significant driver of OA. Specialized proresolution mediators are powerful agents of resolution but have a short in vivo half-life. In this study, we have engineered a Resolvin D1 (RvD1)-loaded nanoliposomal formulation (Lipo-RvD1) that targets and resolves the OA-associated inflammation. This formulation creates a depot of the RvD1 molecules that allows the controlled release of the molecule for up to 11 days in vitro. In surgically induced mice model of OA, only controlled-release formulation of Lipo-RvD1 was able to treat the progressing cartilage damage when administered a month after the surgery, while the free drug was unable to prevent cartilage damage. We found that Lipo-RvD1 functions by damping the proinflammatory activity of synovial macrophages and recruiting a higher number of M2 macrophages at the site of inflammation. Our Lipo-RvD1 formulation was able to target and suppress the formation of the osteophytes and showed analgesic effect, thus emphasizing its ability to treat clinical symptoms of OA. Such controlled-release formulation of RvD1 could represent a patient-compliant treatment for OA.
Collapse
Affiliation(s)
- Ameya A. Dravid
- BioSystems Science and EngineeringIndian Institute of ScienceBangaloreKarnatakaIndia
| | - Kaamini M. Dhanabalan
- BioSystems Science and EngineeringIndian Institute of ScienceBangaloreKarnatakaIndia
| | - Smriti Agarwal
- BioSystems Science and EngineeringIndian Institute of ScienceBangaloreKarnatakaIndia
| | - Rachit Agarwal
- BioSystems Science and EngineeringIndian Institute of ScienceBangaloreKarnatakaIndia
| |
Collapse
|
30
|
Jin Y, Liu Q, Chen P, Zhao S, Jiang W, Wang F, Li P, Zhang Y, Lu W, Zhong TP, Ma X, Wang X, Gartland A, Wang N, Shah KM, Zhang H, Cao X, Yang L, Liu M, Luo J. A novel prostaglandin E receptor 4 (EP4) small molecule antagonist induces articular cartilage regeneration. Cell Discov 2022; 8:24. [PMID: 35256606 PMCID: PMC8901748 DOI: 10.1038/s41421-022-00382-6] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Accepted: 01/28/2022] [Indexed: 01/15/2023] Open
Abstract
Articular cartilage repair and regeneration is an unmet clinical need because of the poor self-regeneration capacity of the tissue. In this study, we found that the expression of prostaglandin E receptor 4 (PTGER4 or EP4) was largely increased in the injured articular cartilage in both humans and mice. In microfracture (MF) surgery-induced cartilage defect (CD) and destabilization of the medial meniscus (DMM) surgery-induced CD mouse models, cartilage-specific deletion of EP4 remarkably promoted tissue regeneration by enhancing chondrogenesis and cartilage anabolism, and suppressing cartilage catabolism and hypertrophy. Importantly, knocking out EP4 in cartilage enhanced stable mature articular cartilage formation instead of fibrocartilage, and reduced joint pain. In addition, we identified a novel selective EP4 antagonist HL-43 for promoting chondrocyte differentiation and anabolism with low toxicity and desirable bioavailability. HL-43 enhanced cartilage anabolism, suppressed catabolism, prevented fibrocartilage formation, and reduced joint pain in multiple pre-clinical animal models including the MF surgery-induced CD rat model, the DMM surgery-induced CD mouse model, and an aging-induced CD mouse model. Furthermore, HL-43 promoted chondrocyte differentiation and extracellular matrix (ECM) generation, and inhibited matrix degradation in human articular cartilage explants. At the molecular level, we found that HL-43/EP4 regulated cartilage anabolism through the cAMP/PKA/CREB/Sox9 signaling. Together, our findings demonstrate that EP4 can act as a promising therapeutic target for cartilage regeneration and the novel EP4 antagonist HL-43 has the clinical potential to be used for cartilage repair and regeneration.
Collapse
Affiliation(s)
- Yunyun Jin
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, China
| | - Qianqian Liu
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, China
| | - Peng Chen
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, China
| | - Siyuan Zhao
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, China
| | - Wenhao Jiang
- Yangzhi Rehabilitation Hospital (Sunshine Rehabilitation Centre), Tongji University School of Medicine, Shanghai, China
| | - Fanhua Wang
- Yangzhi Rehabilitation Hospital (Sunshine Rehabilitation Centre), Tongji University School of Medicine, Shanghai, China
| | - Peng Li
- Orthopedic Institute, Soochow University, Suzhou, Jiangsu, China
| | - Yuanjin Zhang
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, China
| | - Weiqiang Lu
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, China
| | - Tao P Zhong
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, China
| | - Xinran Ma
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, China
| | - Xin Wang
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, China
| | - Alison Gartland
- Department of Oncology and Metabolism, The University of Sheffield, Sheffield, UK
| | - Ning Wang
- Department of Oncology and Metabolism, The University of Sheffield, Sheffield, UK
| | - Karan Mehul Shah
- Department of Oncology and Metabolism, The University of Sheffield, Sheffield, UK
| | - Hankun Zhang
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, China
| | - Xu Cao
- Departments of Orthopaedic Surgery and Biomedical Engineering and Institute of Cell Engineering, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Lei Yang
- Orthopedic Institute, Soochow University, Suzhou, Jiangsu, China.,Center for Health Science and Engineering, School of Materials Science and Engineering, Hebei University of Technology, Tianjin, China
| | - Mingyao Liu
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, China
| | - Jian Luo
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, China. .,Yangzhi Rehabilitation Hospital (Sunshine Rehabilitation Centre), Tongji University School of Medicine, Shanghai, China.
| |
Collapse
|
31
|
Wang F, Liu M, Wang N, Luo J. G Protein-Coupled Receptors in Osteoarthritis. Front Endocrinol (Lausanne) 2022; 12:808835. [PMID: 35154008 PMCID: PMC8831737 DOI: 10.3389/fendo.2021.808835] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Accepted: 12/15/2021] [Indexed: 11/13/2022] Open
Abstract
Osteoarthritis (OA) is the most common chronic joint disease characterized, for which there are no available therapies being able to modify the progression of OA and prevent long-term disability. Critical roles of G-protein coupled receptors (GPCRs) have been established in OA cartilage degeneration, subchondral bone sclerosis and chronic pain. In this review, we describe the pathophysiological processes targeted by GPCRs in OA, along with related preclinical model and/or clinical trial data. We review examples of GPCRs which may offer attractive therapeutic strategies for OA, including receptors for cannabinoids, hormones, prostaglandins, fatty acids, adenosines, chemokines, and discuss the main challenges for developing these therapies.
Collapse
Affiliation(s)
- Fanhua Wang
- Yangzhi Rehabilitation Hospital (Shanghai Sunshine Rehabilitation Center), Tongji University School of Medicine, Shanghai, China
| | - Mingyao Liu
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, China
| | - Ning Wang
- Department of Oncology and Metabolism, The University of Sheffield, Sheffield, United Kingdom
| | - Jian Luo
- Yangzhi Rehabilitation Hospital (Shanghai Sunshine Rehabilitation Center), Tongji University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, China
| |
Collapse
|
32
|
Antidepressant identified as potential DMOAD. Nat Rev Rheumatol 2021; 17:188. [PMID: 33664517 DOI: 10.1038/s41584-021-00594-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|