1
|
Reddiar SB, Xie Y, Abdallah M, Han S, Hu L, Feeney OM, Gracia G, Anshabo A, Lu Z, Farooq MA, Styles IK, Phillips ARJ, Windsor JA, Porter CJH, Cao E, Trevaskis NL. Intestinal Lymphatic Biology, Drug Delivery, and Therapeutics: Current Status and Future Directions. Pharmacol Rev 2024; 76:1326-1398. [PMID: 39179383 DOI: 10.1124/pharmrev.123.001159] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 07/29/2024] [Accepted: 08/14/2024] [Indexed: 08/26/2024] Open
Abstract
Historically, the intestinal lymphatics were considered passive conduits for fluids, immune cells, dietary lipids, lipid soluble vitamins, and lipophilic drugs. Studies of intestinal lymphatic drug delivery in the late 20th century focused primarily on the drugs' physicochemical properties, especially high lipophilicity, that resulted in intestinal lymphatic transport. More recent discoveries have changed our traditional view by demonstrating that the lymphatics are active, plastic, and tissue-specific players in a range of biological and pathological processes, including within the intestine. These findings have, in turn, inspired exploration of lymph-specific therapies for a range of diseases, as well as the development of more sophisticated strategies to actively deliver drugs or vaccines to the intestinal lymph, including a range of nanotechnologies, lipid prodrugs, and lipid-conjugated materials that "hitchhike" onto lymphatic transport pathways. With the increasing development of novel therapeutics such as biologics, there has been interest in whether these therapeutics are absorbed and transported through intestinal lymph after oral administration. Here we review the current state of understanding of the anatomy and physiology of the gastrointestinal lymphatic system in health and disease, with a focus on aspects relevant to drug delivery. We summarize the current state-of-the-art approaches to deliver drugs and quantify their uptake into the intestinal lymphatic system. Finally, and excitingly, we discuss recent examples of significant pharmacokinetic and therapeutic benefits achieved via intestinal lymphatic drug delivery. We also propose approaches to advance the development and clinical application of intestinal lymphatic delivery strategies in the future. SIGNIFICANCE STATEMENT: This comprehensive review details the understanding of the anatomy and physiology of the intestinal lymphatic system in health and disease, with a focus on aspects relevant to drug delivery. It highlights current state-of-the-art approaches to deliver drugs to the intestinal lymphatics and the shift toward the use of these strategies to achieve pharmacokinetic and therapeutic benefits for patients.
Collapse
Affiliation(s)
- Sanjeevini Babu Reddiar
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia (S.B.R., Y.X., M.A., S.H., L.H., O.M.F., G.G., A.A., Z.L., M.A.F., I.K.S., C.J.H.P., E.C., N.L.T.); China Pharmaceutical University, Nanjing, China (S.H., L.H.); Applied Surgery and Metabolism Laboratory, School of Biological Sciences (A.R.J.P.) and Surgical and Translational Research Centre, Department of Surgery, Faculty of Medical and Health Sciences (A.R.J.P., J.A.W.), University of Auckland, Auckland, New Zealand; and Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia (N.L.T.)
| | - Yining Xie
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia (S.B.R., Y.X., M.A., S.H., L.H., O.M.F., G.G., A.A., Z.L., M.A.F., I.K.S., C.J.H.P., E.C., N.L.T.); China Pharmaceutical University, Nanjing, China (S.H., L.H.); Applied Surgery and Metabolism Laboratory, School of Biological Sciences (A.R.J.P.) and Surgical and Translational Research Centre, Department of Surgery, Faculty of Medical and Health Sciences (A.R.J.P., J.A.W.), University of Auckland, Auckland, New Zealand; and Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia (N.L.T.)
| | - Mohammad Abdallah
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia (S.B.R., Y.X., M.A., S.H., L.H., O.M.F., G.G., A.A., Z.L., M.A.F., I.K.S., C.J.H.P., E.C., N.L.T.); China Pharmaceutical University, Nanjing, China (S.H., L.H.); Applied Surgery and Metabolism Laboratory, School of Biological Sciences (A.R.J.P.) and Surgical and Translational Research Centre, Department of Surgery, Faculty of Medical and Health Sciences (A.R.J.P., J.A.W.), University of Auckland, Auckland, New Zealand; and Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia (N.L.T.)
| | - Sifei Han
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia (S.B.R., Y.X., M.A., S.H., L.H., O.M.F., G.G., A.A., Z.L., M.A.F., I.K.S., C.J.H.P., E.C., N.L.T.); China Pharmaceutical University, Nanjing, China (S.H., L.H.); Applied Surgery and Metabolism Laboratory, School of Biological Sciences (A.R.J.P.) and Surgical and Translational Research Centre, Department of Surgery, Faculty of Medical and Health Sciences (A.R.J.P., J.A.W.), University of Auckland, Auckland, New Zealand; and Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia (N.L.T.)
| | - Luojuan Hu
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia (S.B.R., Y.X., M.A., S.H., L.H., O.M.F., G.G., A.A., Z.L., M.A.F., I.K.S., C.J.H.P., E.C., N.L.T.); China Pharmaceutical University, Nanjing, China (S.H., L.H.); Applied Surgery and Metabolism Laboratory, School of Biological Sciences (A.R.J.P.) and Surgical and Translational Research Centre, Department of Surgery, Faculty of Medical and Health Sciences (A.R.J.P., J.A.W.), University of Auckland, Auckland, New Zealand; and Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia (N.L.T.)
| | - Orlagh M Feeney
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia (S.B.R., Y.X., M.A., S.H., L.H., O.M.F., G.G., A.A., Z.L., M.A.F., I.K.S., C.J.H.P., E.C., N.L.T.); China Pharmaceutical University, Nanjing, China (S.H., L.H.); Applied Surgery and Metabolism Laboratory, School of Biological Sciences (A.R.J.P.) and Surgical and Translational Research Centre, Department of Surgery, Faculty of Medical and Health Sciences (A.R.J.P., J.A.W.), University of Auckland, Auckland, New Zealand; and Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia (N.L.T.)
| | - Gracia Gracia
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia (S.B.R., Y.X., M.A., S.H., L.H., O.M.F., G.G., A.A., Z.L., M.A.F., I.K.S., C.J.H.P., E.C., N.L.T.); China Pharmaceutical University, Nanjing, China (S.H., L.H.); Applied Surgery and Metabolism Laboratory, School of Biological Sciences (A.R.J.P.) and Surgical and Translational Research Centre, Department of Surgery, Faculty of Medical and Health Sciences (A.R.J.P., J.A.W.), University of Auckland, Auckland, New Zealand; and Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia (N.L.T.)
| | - Abel Anshabo
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia (S.B.R., Y.X., M.A., S.H., L.H., O.M.F., G.G., A.A., Z.L., M.A.F., I.K.S., C.J.H.P., E.C., N.L.T.); China Pharmaceutical University, Nanjing, China (S.H., L.H.); Applied Surgery and Metabolism Laboratory, School of Biological Sciences (A.R.J.P.) and Surgical and Translational Research Centre, Department of Surgery, Faculty of Medical and Health Sciences (A.R.J.P., J.A.W.), University of Auckland, Auckland, New Zealand; and Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia (N.L.T.)
| | - Zijun Lu
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia (S.B.R., Y.X., M.A., S.H., L.H., O.M.F., G.G., A.A., Z.L., M.A.F., I.K.S., C.J.H.P., E.C., N.L.T.); China Pharmaceutical University, Nanjing, China (S.H., L.H.); Applied Surgery and Metabolism Laboratory, School of Biological Sciences (A.R.J.P.) and Surgical and Translational Research Centre, Department of Surgery, Faculty of Medical and Health Sciences (A.R.J.P., J.A.W.), University of Auckland, Auckland, New Zealand; and Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia (N.L.T.)
| | - Muhammad Asim Farooq
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia (S.B.R., Y.X., M.A., S.H., L.H., O.M.F., G.G., A.A., Z.L., M.A.F., I.K.S., C.J.H.P., E.C., N.L.T.); China Pharmaceutical University, Nanjing, China (S.H., L.H.); Applied Surgery and Metabolism Laboratory, School of Biological Sciences (A.R.J.P.) and Surgical and Translational Research Centre, Department of Surgery, Faculty of Medical and Health Sciences (A.R.J.P., J.A.W.), University of Auckland, Auckland, New Zealand; and Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia (N.L.T.)
| | - Ian K Styles
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia (S.B.R., Y.X., M.A., S.H., L.H., O.M.F., G.G., A.A., Z.L., M.A.F., I.K.S., C.J.H.P., E.C., N.L.T.); China Pharmaceutical University, Nanjing, China (S.H., L.H.); Applied Surgery and Metabolism Laboratory, School of Biological Sciences (A.R.J.P.) and Surgical and Translational Research Centre, Department of Surgery, Faculty of Medical and Health Sciences (A.R.J.P., J.A.W.), University of Auckland, Auckland, New Zealand; and Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia (N.L.T.)
| | - Anthony R J Phillips
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia (S.B.R., Y.X., M.A., S.H., L.H., O.M.F., G.G., A.A., Z.L., M.A.F., I.K.S., C.J.H.P., E.C., N.L.T.); China Pharmaceutical University, Nanjing, China (S.H., L.H.); Applied Surgery and Metabolism Laboratory, School of Biological Sciences (A.R.J.P.) and Surgical and Translational Research Centre, Department of Surgery, Faculty of Medical and Health Sciences (A.R.J.P., J.A.W.), University of Auckland, Auckland, New Zealand; and Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia (N.L.T.)
| | - John A Windsor
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia (S.B.R., Y.X., M.A., S.H., L.H., O.M.F., G.G., A.A., Z.L., M.A.F., I.K.S., C.J.H.P., E.C., N.L.T.); China Pharmaceutical University, Nanjing, China (S.H., L.H.); Applied Surgery and Metabolism Laboratory, School of Biological Sciences (A.R.J.P.) and Surgical and Translational Research Centre, Department of Surgery, Faculty of Medical and Health Sciences (A.R.J.P., J.A.W.), University of Auckland, Auckland, New Zealand; and Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia (N.L.T.)
| | - Christopher J H Porter
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia (S.B.R., Y.X., M.A., S.H., L.H., O.M.F., G.G., A.A., Z.L., M.A.F., I.K.S., C.J.H.P., E.C., N.L.T.); China Pharmaceutical University, Nanjing, China (S.H., L.H.); Applied Surgery and Metabolism Laboratory, School of Biological Sciences (A.R.J.P.) and Surgical and Translational Research Centre, Department of Surgery, Faculty of Medical and Health Sciences (A.R.J.P., J.A.W.), University of Auckland, Auckland, New Zealand; and Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia (N.L.T.)
| | - Enyuan Cao
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia (S.B.R., Y.X., M.A., S.H., L.H., O.M.F., G.G., A.A., Z.L., M.A.F., I.K.S., C.J.H.P., E.C., N.L.T.); China Pharmaceutical University, Nanjing, China (S.H., L.H.); Applied Surgery and Metabolism Laboratory, School of Biological Sciences (A.R.J.P.) and Surgical and Translational Research Centre, Department of Surgery, Faculty of Medical and Health Sciences (A.R.J.P., J.A.W.), University of Auckland, Auckland, New Zealand; and Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia (N.L.T.)
| | - Natalie L Trevaskis
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia (S.B.R., Y.X., M.A., S.H., L.H., O.M.F., G.G., A.A., Z.L., M.A.F., I.K.S., C.J.H.P., E.C., N.L.T.); China Pharmaceutical University, Nanjing, China (S.H., L.H.); Applied Surgery and Metabolism Laboratory, School of Biological Sciences (A.R.J.P.) and Surgical and Translational Research Centre, Department of Surgery, Faculty of Medical and Health Sciences (A.R.J.P., J.A.W.), University of Auckland, Auckland, New Zealand; and Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia (N.L.T.)
| |
Collapse
|
2
|
Kenney HM, Chen KL, Schnur L, Fox JI, Wood RW, Xing L, Ritchlin CT, Rahimi H, Schwarz EM, Awad HA. High-throughput micro-CT analysis identifies sex-dependent biomarkers of erosive arthritis in TNF-Tg mice and differential response to anti-TNF therapy. PLoS One 2024; 19:e0305623. [PMID: 38968295 PMCID: PMC11226038 DOI: 10.1371/journal.pone.0305623] [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: 02/14/2024] [Accepted: 06/03/2024] [Indexed: 07/07/2024] Open
Abstract
BACKGROUND Development of reliable disease activity biomarkers is critical for diagnostics, prognostics, and novel drug development. Although computed tomography (CT) is the gold-standard for quantification of bone erosions, there are no consensus approaches or rationales for utilization of specific outcome measures of erosive arthritis in complex joints. In the case of preclinical models, such as sexually dimorphic tumor necrosis factor transgenic (TNF-Tg) mice, disease severity is routinely quantified in the ankle through manual segmentation of the talus or small regions of adjacent bones primarily due to the ease in measurement. Herein, we sought to determine the particular hindpaw bones that represent reliable biomarkers of sex-dependent disease progression to guide future investigation and analysis. METHODS Hindpaw micro-CT was performed on wild-type (n = 4 male, n = 4 female) and TNF-Tg (n = 4 male, n = 7 female) mice at monthly intervals from 2-5 (females) and 2-8-months (males) of age, since female TNF-Tg mice exhibit early mortality from cardiopulmonary disease at approximately 5-6-months. Further, 8-month-old WT (n = 4) and TNF-Tg males treated with anti-TNF monoclonal antibodies (n = 5) or IgG placebo isotype controls (n = 6) for 6-weeks were imaged with micro-CT every 3-weeks. For image analysis, we utilized our recently developed high-throughput and semi-automated segmentation strategy in Amira software. Synovial and osteoclast histology of ankle joints was quantified using Visiopharm. RESULTS First, we demonstrated that the accuracy of automated segmentation, determined through analysis of ~9000 individual bones by a single user, was comparable in wild-type and TNF-Tg hindpaws before correction (79.2±8.9% vs 80.1±5.1%, p = 0.52). Compared to other bone compartments, the tarsal region demonstrated a sudden, specific, and significant bone volume reduction in female TNF-Tg mice, but not in males, by 5-months (4-months 4.3± 0.22 vs 5-months 3.4± 0.62 mm3, p<0.05). Specifically, the cuboid showed significantly reduced bone volumes at early timepoints compared to other tarsals (i.e., 4-months: Cuboid -24.1±7.2% vs Talus -9.0±5.9% of 2-month baseline). Additional bones localized to the anterolateral region of the ankle also exhibited dramatic erosions in the tarsal region of females, coinciding with increased synovitis and osteoclasts. In TNF-Tg male mice with severe arthritis, the talus and calcaneus exhibited the most sensitive response to anti-TNF therapy measured by effect size of bone volume change over treatment period. CONCLUSIONS We demonstrated that sexually dimorphic changes in arthritic hindpaws of TNF-Tg mice are bone-specific, where the cuboid serves as a reliable early biomarker of erosive arthritis in female mice. Adoption of automated segmentation approaches in pre-clinical or clinical models has potential to translate quantitative biomarkers to monitor bone erosions in disease and evaluate therapeutic efficacy.
Collapse
Affiliation(s)
- H. Mark Kenney
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, United States of America
- Department of Pathology & Laboratory Medicine, University of Rochester Medical Center, Rochester, NY, United States of America
| | - Kiana L. Chen
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, United States of America
- Department of Pathology & Laboratory Medicine, University of Rochester Medical Center, Rochester, NY, United States of America
| | - Lindsay Schnur
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, United States of America
| | - Jeffrey I. Fox
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, United States of America
| | - Ronald W. Wood
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, United States of America
- Department of Obstetrics and Gynecology, University of Rochester Medical Center, Rochester, NY, United States of America
- Department of Neuroscience, University of Rochester Medical Center, Rochester, NY, United States of America
- Department of Urology, University of Rochester Medical Center, Rochester, NY, United States of America
| | - Lianping Xing
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, United States of America
- Department of Pathology & Laboratory Medicine, University of Rochester Medical Center, Rochester, NY, United States of America
| | - Christopher T. Ritchlin
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, United States of America
- Department of Medicine, Division of Allergy, Immunology, Rheumatology, University of Rochester Medical Center, Rochester, NY, United States of America
| | - Homaira Rahimi
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, United States of America
- Department of Pediatrics, Pediatric Rheumatology, University of Rochester Medical Center, Rochester, NY, United States of America
| | - Edward M. Schwarz
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, United States of America
- Department of Pathology & Laboratory Medicine, University of Rochester Medical Center, Rochester, NY, United States of America
- Department of Urology, University of Rochester Medical Center, Rochester, NY, United States of America
- Department of Medicine, Division of Allergy, Immunology, Rheumatology, University of Rochester Medical Center, Rochester, NY, United States of America
- Department of Biomedical Engineering, University of Rochester Medical Center, Rochester, NY, United States of America
- Department of Orthopaedics, University of Rochester Medical Center, Rochester, NY, United States of America
| | - Hani A. Awad
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, United States of America
- Department of Biomedical Engineering, University of Rochester Medical Center, Rochester, NY, United States of America
- Department of Orthopaedics, University of Rochester Medical Center, Rochester, NY, United States of America
| |
Collapse
|
3
|
Rodolfi S, Della-Torre E, Bongiovanni L, Mehta P, Fajgenbaum DC, Selmi C. Lymphadenopathy in the rheumatology practice: a pragmatic approach. Rheumatology (Oxford) 2024; 63:1484-1493. [PMID: 38109670 PMCID: PMC11147542 DOI: 10.1093/rheumatology/kead644] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Revised: 10/31/2023] [Accepted: 11/05/2023] [Indexed: 12/20/2023] Open
Abstract
Lymphadenopathy is a common clinical finding and diagnostic challenge within general medicine and rheumatology practice. It may represent a primary manifestation of an underlying immune-mediated disease or indicate an infectious or neoplastic complication requiring differing management. Evaluating lymphadenopathy is of particular relevance in rheumatology, given that lymph node enlargement is a common finding within the clinical spectrum of several well-known rheumatologic disorders including RA, SLE and SS. In addition, lymphadenopathy represents a hallmark manifestation of rare immunological diseases such as Castleman disease and IgG4-related disease that must be considered in the differential diagnosis because effective targeted treatments can now impact the prognosis of these conditions. In this review we present an overview of the clinical significance of lymphadenopathy in common and rare rheumatologic diseases and propose a practical approach to lymphadenopathy in the rheumatology practice. Differential diagnosis of Castleman disease and therapeutic options for this condition of increasing rheumatologic interest will be discussed in detail.
Collapse
Affiliation(s)
- Stefano Rodolfi
- Department of Rheumatology and Clinical Immunology, IRCCS Humanitas Research Hospital, Milan, Italy
- Department of Biomedical Sciences, Humanitas University, Milan, Italy
| | - Emanuel Della-Torre
- Università Vita-Salute San Raffaele, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Unit of Immunology, Rheumatology, Allergy and Rare Diseases (UnIRAR), Milan, Italy
- IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Lucia Bongiovanni
- Università Vita-Salute San Raffaele, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Department of Haematopathology Diagnostic Area, Unit of Pathology, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Puja Mehta
- Division of Medicine, University College, Centre for Inflammation and Tissue Repair, UCL Respiratory, London
- Department of Rheumatology, University College London Hospital (UCLH), London, UK
| | - David C Fajgenbaum
- Department of Medicine, Division of Translational Medicine and Human Genetics, Center for Cytokine Storm Treatment and Laboratory, Philadelphia, PA, USA
| | - Carlo Selmi
- Department of Rheumatology and Clinical Immunology, IRCCS Humanitas Research Hospital, Milan, Italy
- Department of Biomedical Sciences, Humanitas University, Milan, Italy
| |
Collapse
|
4
|
Cao X, Wang Z, Jiao Y, Diao W, Geng Q, Zhao L, Wang Z, Wang X, Zhang M, Xu J, Wang B, Deng T, Xiao C. Dihydroartemisinin alleviates erosive bone destruction by modifying local Treg cells in inflamed joints: A novel role in the treatment of rheumatoid arthritis. Int Immunopharmacol 2024; 130:111795. [PMID: 38447418 DOI: 10.1016/j.intimp.2024.111795] [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: 01/11/2024] [Revised: 02/15/2024] [Accepted: 02/29/2024] [Indexed: 03/08/2024]
Abstract
Treg cell-based therapy has exhibited promising efficacy in combatting rheumatoid arthritis (RA). Dihydroartemisinin (DHA) exerts broad immunomodulatory effects across various diseases, with its recent spotlight on T-cell regulation in autoimmune conditions. The modulation of DHA on Treg cells and its therapeutic role in RA has yet to be fully elucidated. This study seeks to unveil the influence of DHA on Treg cells in RA and furnish innovative substantiation for the potential of DHA to ameliorate RA. To this end, we initially scrutinized the impact of DHA-modulated Treg cells on osteoclast (OC) formation in vitro using Treg cell-bone marrow-derived monocyte (BMM) coculture systems. Subsequently, employing the collagen-induced arthritis (CIA) rat model, we validated the efficacy of DHA and probed its influence on Treg cells in the spleen and popliteal lymph nodes (PLN). Finally, leveraging deep proteomic analysis with data-independent acquisition (DIA) and parallel accumulation-serial fragmentation (PASEF) technology, we found the alterations in the Treg cell proteome in PLN by proteomic analysis. Our findings indicate that DHA augmented suppressive Treg cells, thereby impeding OC formation in vitro. Consistently, DHA mitigated erosive joint destruction and osteoclastogenesis by replenishing splenic and joint-draining lymph node Treg cells in CIA rats. Notably, DHA induced alterations in the Treg cell proteome in PLN, manifesting distinct upregulation of alloantigen Col2a1 (Type II collagen alfa 1 chain) and CD8a (T-cell surface glycoprotein CD8 alpha chain) in Treg cells, signifying DHA's targeted modulation of Treg cells, rendering them more adept at sustaining immune tolerance and impeding bone erosion. These results unveil a novel facet of DHA in the treatment of RA.
Collapse
Affiliation(s)
- Xiaoxue Cao
- Institute of Clinical Medicine, China-Japan Friendship Hospital, Beijing, China; Graduate School of Peking Union Medical College, Chinese Academy of Medical Sciences/Peking Union Medical College, Beijing, China.
| | - Zhaoran Wang
- Institute of Clinical Medicine, China-Japan Friendship Hospital, Beijing, China; Graduate School of Peking Union Medical College, Chinese Academy of Medical Sciences/Peking Union Medical College, Beijing, China.
| | - Yi Jiao
- China-Japan Friendship Clinical Medical College, Beijing University of Chinese Medicine, Beijing, China.
| | - Wenya Diao
- China-Japan Friendship Clinical Medical College, Beijing University of Chinese Medicine, Beijing, China.
| | - Qishun Geng
- Institute of Clinical Medicine, China-Japan Friendship Hospital, Beijing, China; Graduate School of Peking Union Medical College, Chinese Academy of Medical Sciences/Peking Union Medical College, Beijing, China.
| | - Lu Zhao
- China-Japan Friendship Hospital, Capital Medical University, Beijing, China.
| | - Zihan Wang
- China-Japan Friendship Clinical Medical College, Beijing University of Chinese Medicine, Beijing, China.
| | - Xing Wang
- China-Japan Friendship Clinical Medical College, Beijing University of Chinese Medicine, Beijing, China.
| | - Mengxiao Zhang
- Institute of Clinical Medicine, China-Japan Friendship Hospital, Beijing, China.
| | - Jiahe Xu
- China-Japan Friendship School of Clinical Medicine, Peking University, Beijing, China.
| | - Bailiang Wang
- Department of Orthopaedic Surgery, China-Japan Friendship Hospital, Beijing, China.
| | - Tingting Deng
- Institute of Clinical Medicine, China-Japan Friendship Hospital, Beijing, China.
| | - Cheng Xiao
- Institute of Clinical Medicine, China-Japan Friendship Hospital, Beijing, China; Graduate School of Peking Union Medical College, Chinese Academy of Medical Sciences/Peking Union Medical College, Beijing, China; Department of Emergency, China-Japan Friendship Hospital, Beijing, China.
| |
Collapse
|
5
|
Zhou S, Zhao G, Chen R, Li Y, Huang J, Kuang L, Zhang D, Li Z, Xu H, Xiang W, Xie Y, Chen L, Ni Z. Lymphatic vessels: roles and potential therapeutic intervention in rheumatoid arthritis and osteoarthritis. Theranostics 2024; 14:265-282. [PMID: 38164153 PMCID: PMC10750203 DOI: 10.7150/thno.90940] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Accepted: 11/07/2023] [Indexed: 01/03/2024] Open
Abstract
Lymphatic vessel networks are a main part of the vertebrate cardiovascular system, which participate in various physiological and pathological processes via regulation of fluid transport and immunosurveillance. Targeting lymphatic vessels has become a potent strategy for treating various human diseases. The presence of varying degrees of inflammation in joints of rheumatoid arthritis (RA) and osteoarthritis (OA), characterized by heightened infiltration of inflammatory cells, increased levels of inflammatory factors, and activation of inflammatory signaling pathways, significantly contributes to the disruption of cartilage and bone homeostasis in arthritic conditions. Increasing evidence has demonstrated the pivotal role of lymphatic vessels in maintaining joint homeostasis, with their pathological alterations closely associated with the initiation and progression of inflammatory joint diseases. In this review, we provide a comprehensive overview of the evolving knowledge regarding the structural and functional aspects of lymphatic vessels in the pathogenesis of RA and OA. In addition, we summarized the potential regulatory mechanisms underlying the modulation of lymphatic function in maintaining joint homeostasis during inflammatory conditions, and further discuss the distinctions between RA and OA. Moreover, we describe therapeutic strategies for inflammatory arthritis based on lymphatic vessels, including the promotion of lymphangiogenesis, restoration of proper lymphatic vessel function through anti-inflammatory approaches, enhancement of lymphatic contractility and drainage, and alleviation of congestion within the lymphatic system through the elimination of inflammatory cells. At last, we envisage potential research perspectives and strategies to target lymphatic vessels in treating these inflammatory joint diseases.
Collapse
Affiliation(s)
- Siru Zhou
- War Trauma Medical Center, State Key Laboratory of Trauma and Chemical Poisoning, Army Medical Center, Daping Hospital, Army Medical University, Chongqing, 40038, People's Republic of China
- Department of Wound Repair and Rehabilitation Medicine, State Key Laboratory of Trauma and Chemical Poisoning, Army Medical Center, Daping Hospital, Army Medical University, Chongqing, 40038, People's Republic of China
| | - Guangyu Zhao
- Department of Emergency Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, People's Republic of China
- Rehabilitation Medicine Department, Army Medical Center, Daping Hospital, Army Medical University, Chongqing 400038, People's Republic of China
| | - Ran Chen
- War Trauma Medical Center, State Key Laboratory of Trauma and Chemical Poisoning, Army Medical Center, Daping Hospital, Army Medical University, Chongqing, 40038, People's Republic of China
- Department of Wound Repair and Rehabilitation Medicine, State Key Laboratory of Trauma and Chemical Poisoning, Army Medical Center, Daping Hospital, Army Medical University, Chongqing, 40038, People's Republic of China
| | - Yang Li
- War Trauma Medical Center, State Key Laboratory of Trauma and Chemical Poisoning, Army Medical Center, Daping Hospital, Army Medical University, Chongqing, 40038, People's Republic of China
| | - Junlan Huang
- Department of Wound Repair and Rehabilitation Medicine, State Key Laboratory of Trauma and Chemical Poisoning, Army Medical Center, Daping Hospital, Army Medical University, Chongqing, 40038, People's Republic of China
| | - Liang Kuang
- Department of Wound Repair and Rehabilitation Medicine, State Key Laboratory of Trauma and Chemical Poisoning, Army Medical Center, Daping Hospital, Army Medical University, Chongqing, 40038, People's Republic of China
| | - Dali Zhang
- Department of Wound Repair and Rehabilitation Medicine, State Key Laboratory of Trauma and Chemical Poisoning, Army Medical Center, Daping Hospital, Army Medical University, Chongqing, 40038, People's Republic of China
- The Department of Cardiology, General Hospital of Northern Theater Command, Shenyang 110015, People's Republic of China
| | - Zhijun Li
- Rehabilitation Medicine Department, Army Medical Center, Daping Hospital, Army Medical University, Chongqing 400038, People's Republic of China
| | - Haofeng Xu
- Rehabilitation Medicine Department, Army Medical Center, Daping Hospital, Army Medical University, Chongqing 400038, People's Republic of China
| | - Wei Xiang
- Rehabilitation Medicine Department, Army Medical Center, Daping Hospital, Army Medical University, Chongqing 400038, People's Republic of China
| | - Yangli Xie
- Department of Wound Repair and Rehabilitation Medicine, State Key Laboratory of Trauma and Chemical Poisoning, Army Medical Center, Daping Hospital, Army Medical University, Chongqing, 40038, People's Republic of China
| | - Lin Chen
- Department of Wound Repair and Rehabilitation Medicine, State Key Laboratory of Trauma and Chemical Poisoning, Army Medical Center, Daping Hospital, Army Medical University, Chongqing, 40038, People's Republic of China
| | - Zhenhong Ni
- Rehabilitation Medicine Department, Army Medical Center, Daping Hospital, Army Medical University, Chongqing 400038, People's Republic of China
| |
Collapse
|
6
|
Garhwal A, Kendya P, Soni S, Kori S, Soni V, Kashaw SK. Drug Delivery System Approaches for Rheumatoid Arthritis Treatment: A Review. Mini Rev Med Chem 2024; 24:704-720. [PMID: 37711105 DOI: 10.2174/1389557523666230913105803] [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: 04/17/2023] [Revised: 07/22/2023] [Accepted: 07/23/2023] [Indexed: 09/16/2023]
Abstract
Rheumatoid arthritis (RA) is a chronic autoimmune disease that has traditionally been treated using a variety of pharmacological compounds. However, the effectiveness of these treatments is often limited due to challenges associated with their administration. Oral and parenteral routes of drug delivery are often restricted due to issues such as low bioavailability, rapid metabolism, poor absorption, first-pass effect, and severe side effects. In recent years, nanocarrier-based delivery methods have emerged as a promising alternative for overcoming these challenges. Nanocarriers, including nanoparticles, dendrimers, micelles, nanoemulsions, and stimuli-sensitive carriers, possess unique properties that enable efficient drug delivery and targeted therapy. Using nanocarriers makes it possible to circumvent traditional administration routes' limitations. One of the key advantages of nanocarrier- based delivery is the ability to overcome resistance or intolerance to traditional antirheumatic therapies. Moreover, nanocarriers offer improved drug stability, controlled release kinetics, and enhanced solubility, optimizing the therapeutic effect. They can also protect the encapsulated drug, prolonging its circulation time and facilitating sustained release at the target site. This targeted delivery approach ensures a higher concentration of the therapeutic agent at the site of inflammation, leading to improved therapeutic outcomes. This article explores potential developments in nanotherapeutic regimens for RA while providing a comprehensive summary of current approaches based on novel drug delivery systems. In conclusion, nanocarrier-based drug delivery systems have emerged as a promising solution for improving the treatment of rheumatoid arthritis. Further advancements in nanotechnology hold promise for enhancing the efficacy and safety of RA therapies, offering new hope for patients suffering from this debilitating disease.
Collapse
Affiliation(s)
- Anushka Garhwal
- Department of Pharmaceutical Sciences, Dr. Harisingh Gour University (A Central University), Sagar (MP), India
| | - Priyadarshi Kendya
- Department of Pharmaceutical Sciences, Dr. Harisingh Gour University (A Central University), Sagar (MP), India
| | - Sakshi Soni
- Department of Pharmaceutical Sciences, Dr. Harisingh Gour University (A Central University), Sagar (MP), India
| | - Shivam Kori
- Department of Pharmaceutical Sciences, Dr. Harisingh Gour University (A Central University), Sagar (MP), India
| | - Vandana Soni
- Department of Pharmaceutical Sciences, Dr. Harisingh Gour University (A Central University), Sagar (MP), India
| | - Sushil Kumar Kashaw
- Department of Pharmaceutical Sciences, Dr. Harisingh Gour University (A Central University), Sagar (MP), India
| |
Collapse
|
7
|
Peng Y, Kenney HM, de Mesy Bentley KL, Xing L, Ritchlin CT, Schwarz EM. Distinct mast cell subpopulations within and around lymphatic vessels regulate lymph flow and progression of inflammatory-erosive arthritis in TNF-transgenic mice. Front Immunol 2023; 14:1275871. [PMID: 38155962 PMCID: PMC10752982 DOI: 10.3389/fimmu.2023.1275871] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Accepted: 11/20/2023] [Indexed: 12/30/2023] Open
Abstract
Objective Inflammatory-erosive arthritis is exacerbated by dysfunction of joint-draining popliteal lymphatic vessels (PLVs). Synovial mast cells are known to be pro-inflammatory in rheumatoid arthritis (RA). In other settings they have anti-inflammatory and tissue reparative effects. Herein, we elucidate the role of mast cells on PLV function and inflammatory-erosive arthritis in tumor necrosis factor transgenic (TNF-tg) mice that exhibit defects in PLVs commensurate with disease progression. Methods Whole mount immunofluorescent microscopy, toluidine blue stained histology, scanning electron microscopy, and in silico bioinformatics were performed to phenotype and quantify PLV mast cells. Ankle bone volumes were assessed by μCT, while corresponding histology quantified synovitis and osteoclasts. Near-infrared indocyanine green imaging measured lymphatic clearance as an outcome of PLV draining function. Effects of genetic MC depletion were assessed via comparison of 4.5-month-old WT, TNF-tg, MC deficient KitW-sh/W-sh (cKit-/-), and TNF-tg x cKit-/- mice. Pharmacological inhibition of mast cells was assessed by treating TNF-tg mice with placebo or cromolyn sodium (3.15mg/kg/day) for 3-weeks. Results PLVs are surrounded by MCT+/MCPT1+/MCPT4+ mast cells whose numbers are increased 2.8-fold in TNF-tg mice. The percentage of peri-vascular degranulating mast cells was inversely correlated with ICG clearance. A population of MCT+/MCPT1-/MCPT4- mast cells were embedded within the PLV structure. In silico single-cell RNA-seq (scRNAseq) analyses identified a population of PLV-associated mast cells (marker genes: Mcpt4, Cma1, Cpa3, Tpsb2, Kit, Fcer1a & Gata2) with enhanced TGFβ-related signaling that are phenotypically distinct from known MC subsets in the Mouse Cell Atlas. cKit-/- mice have greater lymphatic defects than TNF-tg mice with exacerbation of lymphatic dysfunction and inflammatory-erosive arthritis in TNF-tg x cKit-/- vs. TNF-Tg mice. Cromolyn sodium therapy stabilized PLV mast cells, increased TNF-induced bone loss, synovitis, and osteoclasts, and decreased ICG clearance. Conclusions Mast cells are required for normal lymphatic function. Genetic ablation and pharmacological inhibition of mast cells exacerbates TNF-induced inflammatory-erosive arthritis with decreased lymphatic clearance. Together, these findings support an inflammatory role of activated/degranulated peri-PLV mast cells during arthritic progression, and a homeostatic role of intra-PLV mast cells, in which loss of the latter dominantly exacerbates arthritis secondary to defects in joint-draining lymphatics, warranting investigation into specific cellular mechanisms.
Collapse
Affiliation(s)
- Yue Peng
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, United States
- Department of Pathology & Laboratory Medicine, University of Rochester Medical Center, Rochester, NY, United States
| | - H. Mark Kenney
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, United States
- Department of Pathology & Laboratory Medicine, University of Rochester Medical Center, Rochester, NY, United States
| | - Karen L. de Mesy Bentley
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, United States
- Department of Pathology & Laboratory Medicine, University of Rochester Medical Center, Rochester, NY, United States
| | - Lianping Xing
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, United States
- Department of Pathology & Laboratory Medicine, University of Rochester Medical Center, Rochester, NY, United States
| | - Christopher T. Ritchlin
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, United States
- Department of Medicine, Division of Allergy, Immunology, Rheumatology, University of Rochester Medical Center, Rochester, NY, United States
| | - Edward M. Schwarz
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, United States
- Department of Pathology & Laboratory Medicine, University of Rochester Medical Center, Rochester, NY, United States
| |
Collapse
|
8
|
Akdeniz Leblebicier M, Cihan E, Şahbaz Pirinççi C, Ture A, Ari B, Ozlu A, Yaman F. Does manual lymphatic drainage improve upper extremity functionality in female patients diagnosed with rheumatoid arthritis? A randomized controlled study. Int J Rheum Dis 2023; 26:1979-1986. [PMID: 37525422 DOI: 10.1111/1756-185x.14849] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 07/01/2023] [Accepted: 07/16/2023] [Indexed: 08/02/2023]
Abstract
OBJECTIVE To demonstrate the effect of manual lymphatic drainage (MLD) on upper extremity functionality in patients with rheumatoid arthritis (RA). MATERIALS AND METHODS Thirty-nine female patients diagnosed with RA who were in remission were included in the study. The study group included 19 participants (mean age 50.63 ± 9.83 years), the control group included 20 participants (mean age 55.05 ± 5.89 years). The patients were randomized into two groups as home exercise program plus MLD and home exercise program alone. Both groups received treatment for 4 weeks. The primary outcome measure was pressure pain threshold (PPT). The patients were evaluated using the PPT, visual analog scale (VAS), hand-wrist joint range of motion examination, hand dynamometer and pinchmeter measurements, and the Rheumatoid Arthritis Quality of Life Scale before the interventention and at the end of the 4th week. RESULTS The PPT evaluated at the thumbnail, wrist, and trapezius significantly increased in the study group after treatment (p < .001, p = .001, and p < .001, respectively). MLD was found to significantly increase the PPT at the thumbnail compared with home exercise program (p = .047). The VAS score significantly decreased in the study group (p = .011). The level of increase in radial deviation was statistically higher in the study group (p = .004). CONCLUSION Manual lymphatic drainage had a positive effect on pain, PPT, and range of motion in RA patients in remission. MLD can be added to medical and other conservative treatments in RA patients.
Collapse
Affiliation(s)
- Merve Akdeniz Leblebicier
- Faculty of Medicine, Department of Physical Medicine and Rehabilitation, Kutahya Health Sciences University, Kutahya, Turkey
| | - Emine Cihan
- Vocational School of Health Sciences, Department of Physiotherapy and Rehabilitation, Physiotherapy Program, Selcuk University, Konya, Turkey
| | - Cansu Şahbaz Pirinççi
- Gulhane Faculty of Physiotherapy and Rehabilitation, University of Health Sciences, Ankara, Turkey
| | - Arzu Ture
- Evliya Celebi Training and Research Hospital, Kutahya Health Sciences University, Kutahya, Turkey
| | - Büşra Ari
- Faculty of Medicine, Department of Physical Medicine and Rehabilitation, Kutahya Health Sciences University, Kutahya, Turkey
| | - Aysun Ozlu
- Faculty of Medicine, Department of Physical Medicine and Rehabilitation, Kutahya Health Sciences University, Kutahya, Turkey
| | - Fatima Yaman
- Faculty of Medicine, Department of Physical Medicine and Rehabilitation, Kutahya Health Sciences University, Kutahya, Turkey
| |
Collapse
|
9
|
Kenney HM, Rangel-Moreno J, Peng Y, Chen KL, Bruno J, Embong A, Pritchett E, Fox JI, Becerril-Villanueva E, Gamboa-Domínguez A, Quataert S, Muthukrishnan G, Wood RW, Korman BD, Anolik JH, Xing L, Ritchlin CT, Schwarz EM, Wu CL. Multi-omics analysis identifies IgG2b class-switching with ALCAM-CD6 co-stimulation in joint-draining lymph nodes during advanced inflammatory-erosive arthritis. Front Immunol 2023; 14:1237498. [PMID: 37691918 PMCID: PMC10485835 DOI: 10.3389/fimmu.2023.1237498] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Accepted: 08/04/2023] [Indexed: 09/12/2023] Open
Abstract
Introduction Defective lymphatic drainage and translocation of B-cells in inflamed (Bin) joint-draining lymph node sinuses are pathogenic phenomena in patients with severe rheumatoid arthritis (RA). However, the molecular mechanisms underlying this lymphatic dysfunction remain poorly understood. Herein, we utilized multi-omic spatial and single-cell transcriptomics to evaluate altered cellular composition (including lymphatic endothelial cells, macrophages, B-cells, and T-cells) in the joint-draining lymph node sinuses and their associated phenotypic changes and cell-cell interactions during RA development using the tumor necrosis factor transgenic (TNF-Tg) mouse model. Methods Popliteal lymph nodes (PLNs) from wild-type (n=10) and TNF-Tg male mice with "Early" (5 to 6-months of age; n=6) and "Advanced" (>8-months of age; n=12) arthritis were harvested and processed for spatial transcriptomics. Single-cell RNA sequencing (scRNAseq) was performed in PLNs from the TNF-Tg cohorts (n=6 PLNs pooled/cohort). PLN histopathology and ELISPOT along with ankle histology and micro-CT were evaluated. Histopathology of human lymph nodes and synovia was performed for clinical correlation. Results Advanced PLN sinuses exhibited an increased Ighg2b/Ighm expression ratio (Early 0.5 ± 0.1 vs Advanced 1.4 ± 0.5 counts/counts; p<0.001) that significantly correlated with reduced talus bone volumes in the afferent ankle (R2 = 0.54, p<0.001). Integration of single-cell and spatial transcriptomics revealed the increased IgG2b+ plasma cells localized in MARCO+ peri-follicular medullary sinuses. A concomitant decreased Fth1 expression (Early 2.5 ± 0.74 vs Advanced 1.0 ± 0.50 counts, p<0.001) within Advanced PLN sinuses was associated with accumulation of iron-laden Prussian blue positive macrophages in lymph nodes and synovium of Advanced TNF-Tg mice, and further validated in RA clinical samples. T-cells were increased 8-fold in Advanced PLNs, and bioinformatic pathway assessment identified the interaction between ALCAM+ macrophages and CD6+ T-cells as a plausible co-stimulatory mechanism to promote IgG2b class-switching. Discussion Collectively, these data support a model of flare in chronic TNF-induced arthritis in which loss of lymphatic flow through affected joint-draining lymph nodes facilitates the interaction between effluxing macrophages and T-cells via ALCAM-CD6 co-stimulation, initiating IgG2b class-switching and plasma cell differentiation of the expanded Bin population. Future work is warranted to investigate immunoglobulin clonality and potential autoimmune consequences, as well as the efficacy of anti-CD6 therapy to prevent these pathogenic events.
Collapse
Affiliation(s)
- H. Mark Kenney
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, United States
- Department of Pathology & Laboratory Medicine, University of Rochester Medical Center, Rochester, NY, United States
| | - Javier Rangel-Moreno
- Department of Medicine, Division of Allergy, Immunology, Rheumatology, University of Rochester Medical Center, Rochester, NY, United States
| | - Yue Peng
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, United States
- Department of Pathology & Laboratory Medicine, University of Rochester Medical Center, Rochester, NY, United States
| | - Kiana L. Chen
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, United States
- Department of Pathology & Laboratory Medicine, University of Rochester Medical Center, Rochester, NY, United States
| | - Jennifer Bruno
- Center for Vaccine Biology and Immunology, University of Rochester Medical Center, Rochester, NY, United States
- Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY, United States
| | - Abdul Embong
- Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY, United States
| | - Elizabeth Pritchett
- Genomics Research Center, University of Rochester Medical Center, Rochester, NY, United States
| | - Jeffrey I. Fox
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, United States
| | - Enrique Becerril-Villanueva
- Psychoimmunology Laboratory, Instituto Nacional de Psiquiatría “Ramón de la Fuente Muñiz”, Mexico City, Mexico
| | - Armando Gamboa-Domínguez
- Department of Pathology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Sally Quataert
- Center for Vaccine Biology and Immunology, University of Rochester Medical Center, Rochester, NY, United States
- Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY, United States
| | - Gowrishankar Muthukrishnan
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, United States
- Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY, United States
- Department of Orthopaedics, University of Rochester Medical Center, Rochester, NY, United States
| | - Ronald W. Wood
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, United States
- Department of Obstetrics and Gynecology, University of Rochester Medical Center, Rochester, NY, United States
- Department of Neuroscience, University of Rochester Medical Center, Rochester, NY, United States
- Department of Urology, University of Rochester Medical Center, Rochester, NY, United States
| | - Benjamin D. Korman
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, United States
- Department of Medicine, Division of Allergy, Immunology, Rheumatology, University of Rochester Medical Center, Rochester, NY, United States
| | - Jennifer H. Anolik
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, United States
- Department of Pathology & Laboratory Medicine, University of Rochester Medical Center, Rochester, NY, United States
- Department of Medicine, Division of Allergy, Immunology, Rheumatology, University of Rochester Medical Center, Rochester, NY, United States
| | - Lianping Xing
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, United States
- Department of Pathology & Laboratory Medicine, University of Rochester Medical Center, Rochester, NY, United States
| | - Christopher T. Ritchlin
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, United States
- Department of Medicine, Division of Allergy, Immunology, Rheumatology, University of Rochester Medical Center, Rochester, NY, United States
| | - Edward M. Schwarz
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, United States
- Department of Pathology & Laboratory Medicine, University of Rochester Medical Center, Rochester, NY, United States
- Department of Medicine, Division of Allergy, Immunology, Rheumatology, University of Rochester Medical Center, Rochester, NY, United States
- Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY, United States
- Department of Orthopaedics, University of Rochester Medical Center, Rochester, NY, United States
- Department of Urology, University of Rochester Medical Center, Rochester, NY, United States
| | - Chia-Lung Wu
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, United States
- Department of Orthopaedics, University of Rochester Medical Center, Rochester, NY, United States
| |
Collapse
|
10
|
Kenney HM, Dieudonne G, Yee S, Maki JH, Wood RW, Schwarz EM, Ritchlin CT, Rahimi H. Near-Infrared Imaging of Indocyanine Green Identifies Novel Routes of Lymphatic Drainage from Metacarpophalangeal Joints in Healthy Human Hands. Lymphat Res Biol 2023; 21:388-395. [PMID: 36809077 PMCID: PMC10460689 DOI: 10.1089/lrb.2022.0026] [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: 02/23/2023] Open
Abstract
Background: Collecting lymphatic vessel (CLV) dysfunction has been implicated in various diseases, including rheumatoid arthritis (RA). RA patients with active hand arthritis exhibit significantly reduced lymphatic clearance of the web spaces adjacent to the metacarpophalangeal (MCP) joints and a reduction in total and basilic-associated CLVs on the dorsal surface of the hand by near-infrared (NIR) imaging of indocyanine green (ICG). In this pilot study, we assessed direct lymphatic drainage from MCP joints and aimed to visualize the total lymphatic anatomy using novel dual-agent relaxation contrast magnetic resonance lymphography (DARC-MRL) in the upper extremity of healthy human subjects. Methods and Results: Two healthy male subjects >18 years old participated in the study. We performed NIR imaging along with conventional- or DARC-MRL following intradermal web space and intra-articular MCP joint injections. ICG (NIR) or gadolinium (Gd) (MRL) was administered to visualize the CLV anatomy of the upper extremity. Web space draining CLVs were associated with the cephalic side of the antecubital fossa, while MCP draining CLVs were localized to the basilic side of the forearm by near-infrared indocyanine green imaging. The DARC-MRL methods used in this study did not adequately nullify the contrast in the blood vessels, and limited Gd-filled CLVs were identified. Conclusion: MCP joints predominantly drain into basilic CLVs in the forearm, which may explain the reduction in basilic-associated CLVs in the hands of RA patients. Current DARC-MRL techniques show limited identification of healthy lymphatic structures, and further refinement in this technique is necessary. Clinical trial registration number: NCT04046146.
Collapse
Affiliation(s)
- H. Mark Kenney
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, New York, USA
- Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, New York, USA
| | - Gregory Dieudonne
- Department of Imaging Sciences, University of Rochester Medical Center, Rochester, New York, USA
| | - Seonghwan Yee
- Department of Radiology, University of Colorado Anschutz Medical Center, Aurora, Colorado, USA
| | - Jeffrey H. Maki
- Department of Radiology, University of Colorado Anschutz Medical Center, Aurora, Colorado, USA
| | - Ronald W. Wood
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, New York, USA
| | - Edward M. Schwarz
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, New York, USA
- Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, New York, USA
- Department of Orthopaedics, Pediatric Rheumatology, University of Rochester Medical Center, Rochester, New York, USA
- Division of Allergy, Immunology, and Rheumatology, Department of Medicine, Pediatric Rheumatology, University of Rochester Medical Center, Rochester, New York, USA
| | - Christopher T. Ritchlin
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, New York, USA
- Division of Allergy, Immunology, and Rheumatology, Department of Medicine, Pediatric Rheumatology, University of Rochester Medical Center, Rochester, New York, USA
| | - Homaira Rahimi
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, New York, USA
- Department of Pediatrics, Pediatric Rheumatology, University of Rochester Medical Center, Rochester, New York, USA
| |
Collapse
|
11
|
Kenney HM, Peng Y, de Mesy Bentley KL, Xing L, Ritchlin CT, Schwarz EM. The Enigmas of Lymphatic Muscle Cells: Where Do They Come From, How Are They Maintained, and Can They Regenerate? Curr Rheumatol Rev 2023; 19:246-259. [PMID: 36705238 PMCID: PMC10257750 DOI: 10.2174/1573397119666230127144711] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Revised: 10/29/2022] [Accepted: 12/02/2022] [Indexed: 01/28/2023]
Abstract
Lymphatic muscle cell (LMC) contractility and coverage of collecting lymphatic vessels (CLVs) are integral to effective lymphatic drainage and tissue homeostasis. In fact, defects in lymphatic contractility have been identified in various conditions, including rheumatoid arthritis, inflammatory bowel disease, and obesity. However, the fundamental role of LMCs in these pathologic processes is limited, primarily due to the difficulty in directly investigating the enigmatic nature of this poorly characterized cell type. LMCs are a unique cell type that exhibit dual tonic and phasic contractility with hybrid structural features of both vascular smooth muscle cells (VSMCs) and cardiac myocytes. While advances have been made in recent years to better understand the biochemistry and function of LMCs, central questions regarding their origins, investiture into CLVs, and homeostasis remain unanswered. To summarize these discoveries, unexplained experimental results, and critical future directions, here we provide a focused review of current knowledge and open questions related to LMC progenitor cells, recruitment, maintenance, and regeneration. We also highlight the high-priority research goal of identifying LMC-specific genes towards genetic conditional- inducible in vivo gain and loss of function studies. While our interest in LMCs has been focused on understanding lymphatic dysfunction in an arthritic flare, these concepts are integral to the broader field of lymphatic biology, and have important potential for clinical translation through targeted therapeutics to control lymphatic contractility and drainage.
Collapse
Grants
- R01AG059775,R01AG059775,R01AG059775 NIA NIH HHS
- R01AR056702,R01AR069000,T32AR076950,P30AR069655,R01AR056702,R01AR069000,P30AR069655,T32AR076950,R01AR056702,R01AR069000,T32AR076950,P30AR069655 NIAMS NIH HHS
- P30 AR069655 NIAMS NIH HHS
- R01 AR069000 NIAMS NIH HHS
- T32 GM007356 NIGMS NIH HHS
- R01 AG059775 NIA NIH HHS
- T32GM007356,T32GM007356,T32GM007356,T32GM007356 NIGMS NIH HHS
- T32 AR076950 NIAMS NIH HHS
- R01 AR056702 NIAMS NIH HHS
- F30 AG076326 NIA NIH HHS
Collapse
Affiliation(s)
- H. Mark Kenney
- Center for Musculoskeletal Research, University of Rochester Medical Center, 601 Elmwood Ave, Box 665, Rochester, NY, 14642, USA
- Department of Pathology & Laboratory Medicine, University of Rochester Medical Center, Rochester, NY, USA
| | - Yue Peng
- Center for Musculoskeletal Research, University of Rochester Medical Center, 601 Elmwood Ave, Box 665, Rochester, NY, 14642, USA
- Department of Pathology & Laboratory Medicine, University of Rochester Medical Center, Rochester, NY, USA
| | - Karen L. de Mesy Bentley
- Center for Musculoskeletal Research, University of Rochester Medical Center, 601 Elmwood Ave, Box 665, Rochester, NY, 14642, USA
- Department of Pathology & Laboratory Medicine, University of Rochester Medical Center, Rochester, NY, USA
- Department of Orthopaedics, University of Rochester Medical Center, Rochester, NY, USA
| | - Lianping Xing
- Center for Musculoskeletal Research, University of Rochester Medical Center, 601 Elmwood Ave, Box 665, Rochester, NY, 14642, USA
- Department of Pathology & Laboratory Medicine, University of Rochester Medical Center, Rochester, NY, USA
| | - Christopher T. Ritchlin
- Center for Musculoskeletal Research, University of Rochester Medical Center, 601 Elmwood Ave, Box 665, Rochester, NY, 14642, USA
- Department of Medicine, Division of Allergy, Immunology, Rheumatology, University of Rochester Medical Center, Rochester, NY, USA
| | - Edward M. Schwarz
- Center for Musculoskeletal Research, University of Rochester Medical Center, 601 Elmwood Ave, Box 665, Rochester, NY, 14642, USA
- Department of Pathology & Laboratory Medicine, University of Rochester Medical Center, Rochester, NY, USA
- Department of Medicine, Division of Allergy, Immunology, Rheumatology, University of Rochester Medical Center, Rochester, NY, USA
- Department of Orthopaedics, University of Rochester Medical Center, Rochester, NY, USA
| |
Collapse
|
12
|
Kraus SE, Lee E. Engineering approaches to investigate the roles of lymphatics vessels in rheumatoid arthritis. Microcirculation 2023; 30:e12769. [PMID: 35611452 PMCID: PMC9684355 DOI: 10.1111/micc.12769] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Revised: 05/12/2022] [Accepted: 05/20/2022] [Indexed: 11/30/2022]
Abstract
Rheumatoid arthritis (RA) is one of the most common chronic inflammatory joint disorders. While our understanding of the autoimmune processes that lead to synovial degradation has improved, a majority of patients are still resistant to current treatments and require new therapeutics. An understudied and promising area for therapy involves the roles of lymphatic vessels (LVs) in RA progression, which has been observed to have a significant effect on mediating chronic inflammation. RA disease progression has been shown to correlate with dramatic changes in LV structure and interstitial fluid drainage, manifesting in the retention of distinct immune cell phenotypes within the synovium. Advances in dynamic imaging technologies have demonstrated that LVs in RA undergo an initial expansion phase of increased LVs and abnormal contractions followed by a collapsed phase of reduced lymphatic function and immune cell clearance in vivo. However, current animal models of RA fail to decouple biological and biophysical factors that might be responsible for this lymphatic dysfunction in RA, and a few attempted in vitro models of the synovium in RA have not yet included the contributions from the LVs. Various methods of replicating LVs in vitro have been developed to study lymphatic biology, but these have yet not been integrated into the RA context. This review discusses the roles of LVs in RA and the current engineering approaches to improve our understanding of lymphatic pathophysiology in RA.
Collapse
Affiliation(s)
- Samantha E. Kraus
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY 14853, USA
| | - Esak Lee
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY 14853, USA
| |
Collapse
|
13
|
Lam AD, Styles IK, Senyschyn D, Cao E, Anshabo A, Abdallah M, Mikrani R, Nowell CJ, Porter CJH, Feeney OM, Trevaskis NL. Intra-articular Injection of a B Cell Depletion Antibody Enhances Local Exposure to the Joint-Draining Lymph Node in Mice with Collagen-Induced Arthritis. Mol Pharm 2023; 20:2053-2066. [PMID: 36945772 DOI: 10.1021/acs.molpharmaceut.2c01041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/23/2023]
Abstract
Changes to the number, type, and function of immune cells within the joint-draining lymphatics is a major contributor to the progression of inflammatory arthritis. In particular, there is a significant expansion in pathogenic B cells in the joint-draining lymph node (jdLN). These B cells appear to clog the lymphatic sinuses in the lymph node, inhibit lymph flow, and therefore, reduce the clearance of inflammatory fluid and cells from the joint. Taken together, there is potential to treat inflammatory arthritis more effectively, as well as reduce off-target side effects, with localized delivery of B-cell depleting therapies to the jdLNs. We recently reported that joint-draining lymphatic exposure of biologic disease-modifying anti-rheumatic drugs (DMARDs), including the B cell depletion antibody rituximab, is increased in healthy rats following intra-articular (IA) compared to subcutaneous (SC) or intravenous (IV) administration. This suggests that IA administration of B cell depleting antibodies may increase delivery to target cells in the jdLN and increase the effectiveness of B cell depletion compared to standard SC or IV administration. However, whether enhanced local delivery of DMARDs to the jdLN is also achieved after IA injection in the setting of inflammatory arthritis, where there is inflammation in the joint and jdLN B cell expansion is unknown. We, therefore, assessed the lymph node distribution, absorption and plasma pharmacokinetics, and B cell depletion at different sites after IA, SC, or IV administration of a fluorescently labeled mouse anti-CD20 B cell depleting antibody (Cy5-αCD20) in healthy mice compared to mice with collagen-induced arthritis (CIA). The absorption and plasma pharmacokinetics of Cy5-αCD20 appeared unaltered in mice with CIA whereas distribution of Cy5-αCD20 to the jdLNs was generally increased in mice with CIA, regardless of the route of administration. However, IA administration led to greater and more specific exposure to the jdLNs. Consistent with increased Cy5-αCD20 in the jdLNs of CIA compared to healthy mice, there was a greater reduction in jdLN weight and a trend toward greater jdLN B cell depletion at 24 h compared to 4 h after IA compared to SC and IV administration. Taken together, this data supports the potential to improve local efficacy of B cell depletion therapies through a jdLN-directed approach which will enable a reduction in dose and systemic toxicities.
Collapse
Affiliation(s)
- Alina D Lam
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville Campus, 399 Royal Parade, Parkville, Victoria 3052, Australia
| | - Ian K Styles
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville Campus, 399 Royal Parade, Parkville, Victoria 3052, Australia
| | - Danielle Senyschyn
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville Campus, 399 Royal Parade, Parkville, Victoria 3052, Australia
| | - Enyuan Cao
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville Campus, 399 Royal Parade, Parkville, Victoria 3052, Australia
| | - Abel Anshabo
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville Campus, 399 Royal Parade, Parkville, Victoria 3052, Australia
| | - Mohammad Abdallah
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville Campus, 399 Royal Parade, Parkville, Victoria 3052, Australia
| | - Reyaj Mikrani
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville Campus, 399 Royal Parade, Parkville, Victoria 3052, Australia
| | - Cameron J Nowell
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville Campus, 399 Royal Parade, Parkville, Victoria 3052, Australia
| | - Christopher J H Porter
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville Campus, 399 Royal Parade, Parkville, Victoria 3052, Australia
| | - Orlagh M Feeney
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville Campus, 399 Royal Parade, Parkville, Victoria 3052, Australia
| | - Natalie L Trevaskis
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville Campus, 399 Royal Parade, Parkville, Victoria 3052, Australia
| |
Collapse
|
14
|
Sevick-Muraca EM, Fife CE, Rasmussen JC. Imaging peripheral lymphatic dysfunction in chronic conditions. Front Physiol 2023; 14:1132097. [PMID: 37007996 PMCID: PMC10050385 DOI: 10.3389/fphys.2023.1132097] [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: 12/26/2022] [Accepted: 02/17/2023] [Indexed: 03/17/2023] Open
Abstract
The lymphatics play important roles in chronic diseases/conditions that comprise the bulk of healthcare worldwide. Yet the ability to routinely image and diagnose lymphatic dysfunction, using commonly available clinical imaging modalities, has been lacking and as a result, the development of effective treatment strategies suffers. Nearly two decades ago, investigational near-infrared fluorescence lymphatic imaging and ICG lymphography were developed as routine diagnostic for clinically evaluating, quantifying, and treating lymphatic dysfunction in cancer-related and primary lymphedema, chronic venous disease, and more recently, autoimmune and neurodegenerative disorders. In this review, we provide an overview of what these non-invasive technologies have taught us about lymphatic (dys) function and anatomy in human studies and in corollary animal studies of human disease. We summarize by commenting on new impactful clinical frontiers in lymphatic science that remain to be facilitated by imaging.
Collapse
Affiliation(s)
- Eva M. Sevick-Muraca
- Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Caroline E. Fife
- Department of Geriatrics, Baylor College of Medicine, Houston, TX, United States
| | - John C. Rasmussen
- Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center at Houston, Houston, TX, United States
| |
Collapse
|
15
|
Tian J, Chen T, Huang B, Liu Y, Wang C, Cui Z, Xu H, Li Q, Zhang W, Liang Q. Inflammation specific environment activated methotrexate-loaded nanomedicine to treat rheumatoid arthritis by immune environment reconstruction. Acta Biomater 2023; 157:367-380. [PMID: 36513249 DOI: 10.1016/j.actbio.2022.12.007] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 11/15/2022] [Accepted: 12/05/2022] [Indexed: 12/14/2022]
Abstract
Rheumatoid arthritis (RA), as an autoimmune inflammatory disease, is featured by enhanced vascular permeability, irreversible cartilage destroys and bone erosion. Although the pathogenesis of RA is still unclear, the immune environment, particularly the lymphatic system, which is instrumental to immune cell surveillance and interstitial fluid balance, plays vital roles in the process of RA. Herein, an inflammation specific environment activated methotrexate-encapsulated nanomedicine (MTX@NPs) was constructed for RA treatment, which accumulated in inflamed joints, and released MTX in the specific RA microenvironment. Notably, MTX@NPs could regulate the immune environment including reducing the expressions of inflammatory cytokines of macrophages and the inflammatory level of lymphatic epithelial cells (LECs), and ameliorating the lymphatic vessel contraction and drainage. In vitro and In vivo studies illustrated that MTX@NPs exhibited a high RA therapeutic efficacy and insignificant systemic toxicity owing to the suppression of the inflammation response and the improved lymphatic functions of RA joints. It suggests that the nanomedicine paves a potential way to the clinical practice of autoimmune diseases treatments via the regulation of immune environment and lymphatic functions. STATEMENT OF SIGNIFICANCE: Although 1.0% of the population in the world suffers from rheumatoid arthritis (RA), the pathogenesis of RA is still unclear and the therapeutic effect of the first-line clinical drugs is relatively low. Herein, we propose a specific RA-microenvironment triggered nanomedicine (MTX@NPs), which enhances RA treatment of a first-line antirheumatic drug (methotrexate, MTX) by immune environment reconstruction. The nanomedicine exhibits RA joints accumulation by EPR effect, and releases MTX under the specific RA environment, leading to the dramatical drop of M1-type macrophages and acceleration of lymphatic vessel contraction and drainage. Finally, the inflammatory cytokines in RA immune environment are reduced sharply, indicating the outstanding therapeutic efficacy of MTX@NPs to RA.
Collapse
Affiliation(s)
- Jia Tian
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, 725 South Wanping Road, Shanghai 200032, China; Shanghai Key Laboratory of Functional Materials Chemistry, School of Chemistry and Molecular Engineering, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, China.
| | - Tao Chen
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, 725 South Wanping Road, Shanghai 200032, China; Jing'an District Center Hospital of Shanghai, Fudan University, Shanghai 200040, China
| | - Baoxuan Huang
- Shanghai Key Laboratory of Functional Materials Chemistry, School of Chemistry and Molecular Engineering, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, China
| | - Yang Liu
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, 725 South Wanping Road, Shanghai 200032, China; Spine Institute, Shanghai University of Traditional Chinese Medicine, 725 Wan-Ping South Road, Shanghai 200032, China; Key Laboratory of Theory and Therapy of Muscles and Bones, Ministry of Education (Shanghai University of Traditional Chinese Medicine), 1200 Cailun Road, Shanghai 201203, China
| | - Chao Wang
- Shanghai Key Laboratory of Functional Materials Chemistry, School of Chemistry and Molecular Engineering, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, China
| | - Zepeng Cui
- Shanghai Key Laboratory of Functional Materials Chemistry, School of Chemistry and Molecular Engineering, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, China
| | - Hao Xu
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, 725 South Wanping Road, Shanghai 200032, China; Spine Institute, Shanghai University of Traditional Chinese Medicine, 725 Wan-Ping South Road, Shanghai 200032, China; Key Laboratory of Theory and Therapy of Muscles and Bones, Ministry of Education (Shanghai University of Traditional Chinese Medicine), 1200 Cailun Road, Shanghai 201203, China
| | - Qiang Li
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, 725 South Wanping Road, Shanghai 200032, China; Spine Institute, Shanghai University of Traditional Chinese Medicine, 725 Wan-Ping South Road, Shanghai 200032, China; Key Laboratory of Theory and Therapy of Muscles and Bones, Ministry of Education (Shanghai University of Traditional Chinese Medicine), 1200 Cailun Road, Shanghai 201203, China
| | - Weian Zhang
- Shanghai Key Laboratory of Functional Materials Chemistry, School of Chemistry and Molecular Engineering, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, China.
| | - Qianqian Liang
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, 725 South Wanping Road, Shanghai 200032, China; Spine Institute, Shanghai University of Traditional Chinese Medicine, 725 Wan-Ping South Road, Shanghai 200032, China; Key Laboratory of Theory and Therapy of Muscles and Bones, Ministry of Education (Shanghai University of Traditional Chinese Medicine), 1200 Cailun Road, Shanghai 201203, China.
| |
Collapse
|
16
|
Inflammation-responsive nanoparticles suppress lymphatic clearance for prolonged arthritis therapy. J Control Release 2022; 352:700-711. [PMID: 36347402 DOI: 10.1016/j.jconrel.2022.11.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Revised: 10/24/2022] [Accepted: 11/02/2022] [Indexed: 11/11/2022]
Abstract
The clearance of nanomedicine in inflamed joints has been accelerated due to the increased lymph angiogenesis and lymph flow in arthritic sites. To maximize the therapeutic efficacy for rheumatoid arthritis (RA), it is necessary to facilitate targeted delivery and extended drug retention in inflamed synovium simultaneously. In general, nanosized particles are more likely to achieve prolonged circulation and targeted delivery. While drug carriers with larger dimension might be more beneficial for extending drug retention. To balance the conflicting requirements, an inflammation-responsive shape transformable nanoparticle, comprised of amyloid β-derived KLVFF peptide and polysialic acid (PSA), coupled with therapeutic agent dexamethasone (Dex) via an acid-sensitive linker, was fabricated and termed as Dex-KLVFF-PSA (DKPNPs). Under physiological condition, DKPNPs can keep stable nanosized morphology, and PSA shell could endow DKPNPs with long circulation and active targeting to arthritic sites. While in inflamed joints, acidic pH-triggered Dex dissociation or macrophages-induced specific binding with PSA would induce the re-assembly of DKPNPs from nanoparticles to nanofibers. Our results reveal that intravenously injected DKPNPs display prolonged in vivo circulation and preferential distribution in inflamed joints, where DKPNPs undergo shape transition to fibrous structures, leading to declined lymphatic clearance and prolonged efficacy. Overall, our dual-stimulus responsive transformable nanoparticle offers an intelligent solution to achieve enhanced therapeutic efficacy in RA.
Collapse
|
17
|
Kenney HM, Peng Y, Bell RD, Wood RW, Xing L, Ritchlin CT, Schwarz EM. Persistent popliteal lymphatic muscle cell coverage defects despite amelioration of arthritis and recovery of popliteal lymphatic vessel function in TNF-Tg mice following anti-TNF therapy. Sci Rep 2022; 12:12751. [PMID: 35882971 PMCID: PMC9325893 DOI: 10.1038/s41598-022-16884-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Accepted: 07/18/2022] [Indexed: 11/20/2022] Open
Abstract
While rheumatoid arthritis patients and tumor necrosis factor transgenic (TNF-Tg) mice with inflammatory-erosive arthritis display lymphatic drainage deficits, the mechanisms responsible remain unknown. As ultrastructural studies of joint-draining popliteal lymphatic vessels (PLVs) in TNF-Tg mice revealed evidence of lymphatic muscle cell (LMC) damage, we aimed to evaluate PLV-LMC coverage in TNF-Tg mice. We tested the hypothesis that alpha smooth muscle actin (αSMA)+ PLV-LMC coverage decreases with severe inflammatory-erosive arthritis, and is recovered by anti-TNF therapy facilitated by increased PLV-LMC turnover during amelioration of joint disease. TNF-Tg mice with established disease received anti-TNF monoclonal antibody (mAb) or placebo IgG isotype control mAb therapy (n = 5) for 6-weeks, while wild-type (WT) littermates (n = 8) received vehicle (PBS). Bromodeoxyuridine (BrdU) was also administered daily during the treatment period to monitor PLV-LMC turnover. Effective anti-TNF therapy was confirmed by longitudinal assessment of popliteal lymph node (PLN) volume via ultrasound, PLV contraction frequency via near-infrared imaging of indocyanine green, and ankle bone volumes via micro-computed tomography (micro-CT). Terminal knee micro-CT, and ankle and knee histology were also performed. PLVs were immunostained for αSMA and BrdU to evaluate PLV-LMC coverage and turnover, respectively, via whole-mount fluorescent microscopy. Anti-TNF therapy reduced PLN volume, increased talus and patella bone volumes, and reduced tarsal and knee synovial areas compared to placebo treated TNF-Tg mice (p < 0.05), as expected. Anti-TNF therapy also increased PLV contraction frequency at 3-weeks (from 0.81 ± 1.0 to 3.2 ± 2.0 contractions per minute, p < 0.05). However, both anti-TNF and placebo treated TNF-Tg mice exhibited significantly reduced αSMA+ PLV-LMC coverage compared to WT (p < 0.05). There was no correlation of αSMA+ PLV-LMC coverage restoration with amelioration of inflammatory-erosive arthritis. Similarly, there was no difference in PLV-LMC turnover measured by BrdU labeling between WT, TNF-Tg placebo, and TNF-Tg anti-TNF groups with an average of < 1% BrdU+ PLV-LMCs incorporated per week. Taken together these results demonstrate that PLV-LMC turnover in adult mice is limited, and that recovery of PLV function during amelioration of inflammatory-erosive arthritis occurs without restoration of αSMA+ LMC coverage. Future studies are warranted to investigate the direct and indirect effects of chronic TNF exposure, and the role of proximal inflammatory cells on PLV contractility.
Collapse
Affiliation(s)
- H Mark Kenney
- Center for Musculoskeletal Research, University of Rochester Medical Center, 601 Elmwood Ave, Box 665, Rochester, NY, 14642, USA
- Department of Pathology & Laboratory Medicine, University of Rochester Medical Center, Rochester, NY, USA
| | - Yue Peng
- Center for Musculoskeletal Research, University of Rochester Medical Center, 601 Elmwood Ave, Box 665, Rochester, NY, 14642, USA
- Department of Pathology & Laboratory Medicine, University of Rochester Medical Center, Rochester, NY, USA
| | - Richard D Bell
- Department of Research, Hospital for Special Surgery, New York, NY, USA
| | - Ronald W Wood
- Center for Musculoskeletal Research, University of Rochester Medical Center, 601 Elmwood Ave, Box 665, Rochester, NY, 14642, USA
- Department of Obstetrics and Gynecology, University of Rochester Medical Center, Rochester, NY, USA
- Department of Neuroscience, University of Rochester Medical Center, Rochester, NY, USA
- Department of Urology, University of Rochester Medical Center, Rochester, NY, USA
| | - Lianping Xing
- Center for Musculoskeletal Research, University of Rochester Medical Center, 601 Elmwood Ave, Box 665, Rochester, NY, 14642, USA
- Department of Pathology & Laboratory Medicine, University of Rochester Medical Center, Rochester, NY, USA
| | - Christopher T Ritchlin
- Center for Musculoskeletal Research, University of Rochester Medical Center, 601 Elmwood Ave, Box 665, Rochester, NY, 14642, USA
- Department of Medicine, Division of Allergy, Immunology, Rheumatology, University of Rochester Medical Center, Rochester, NY, USA
| | - Edward M Schwarz
- Center for Musculoskeletal Research, University of Rochester Medical Center, 601 Elmwood Ave, Box 665, Rochester, NY, 14642, USA.
- Department of Pathology & Laboratory Medicine, University of Rochester Medical Center, Rochester, NY, USA.
- Department of Urology, University of Rochester Medical Center, Rochester, NY, USA.
- Department of Medicine, Division of Allergy, Immunology, Rheumatology, University of Rochester Medical Center, Rochester, NY, USA.
- Department of Orthopaedics, University of Rochester Medical Center, Rochester, NY, USA.
| |
Collapse
|
18
|
Michalaki E, Nepiyushchikh Z, Rudd JM, Bernard FC, Mukherjee A, McKinney JM, Doan TN, Willett NJ, Dixon JB. Effect of Human Synovial Fluid From Osteoarthritis Patients and Healthy Individuals on Lymphatic Contractile Activity. J Biomech Eng 2022; 144:071012. [PMID: 35118490 PMCID: PMC8883121 DOI: 10.1115/1.4053749] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Revised: 12/10/2021] [Indexed: 11/08/2022]
Abstract
The lymphatic system has been proposed to play a crucial role in preventing the development and progression of osteoarthritis (OA). As OA develops and progresses, inflammatory cytokines and degradation by-products of joint tissues build up in the synovial fluid (SF) providing a feedback system to exacerbate disease. The lymphatic system plays a critical role in resolving inflammation and maintaining overall joint homeostasis; however, there is some evidence that the lymphatics can become dysfunctional during OA. We hypothesized that the functional mechanics of lymphatic vessels (LVs) draining the joint could be directly compromised due to factors within SF derived from osteoarthritis patients (OASF). Here, we utilized OASF and SF derived from healthy (non-OA) individuals (healthy SF (HSF)) to investigate potential effects of SF entering the draining lymph on migration of lymphatic endothelial cells (LECs) in vitro, and lymphatic contractile activity of rat femoral LVs (RFLVs) ex vivo. Dilutions of both OASF and HSF containing serum resulted in a similar LEC migratory response to the physiologically endothelial basal medium-treated LECs (endothelial basal medium containing serum) in vitro. Ex vivo, OASF and HSF treatments were administered within the lumen of isolated LVs under controlled pressures. OASF treatment transiently enhanced the RFLVs tonic contractions while phasic contractions were significantly reduced after 1 h of treatment and complete ceased after overnight treatment. HSF treatment on the other hand displayed a gradual decrease in lymphatic contractile activity (both tonic and phasic contractions). The observed variations after SF treatments suggest that the pump function of lymphatic vessel draining the joint could be directly compromised in OA and thus might present a new therapeutic target.
Collapse
Affiliation(s)
- Eleftheria Michalaki
- George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, 801 Ferst Drive, Atlanta, GA 30332
| | - Zhanna Nepiyushchikh
- George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, 801 Ferst Drive, Atlanta, GA 30332
| | - Josephine M. Rudd
- George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, 801 Ferst Drive, Atlanta, GA 30332
| | - Fabrice C. Bernard
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, 313 Ferst Drive, Atlanta, GA 30332
| | - Anish Mukherjee
- School of Electrical and Computer Engineering, Georgia Institute of Technology, 777 Atlantic Dr NW, Atlanta, GA 30332
| | - Jay M. McKinney
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, 313 Ferst Drive, Atlanta, GA 30332
| | - Thanh N. Doan
- Department of Orthopaedics, Emory University, 59 Executive Park South, Atlanta, GA 30329
| | - Nick J. Willett
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, 313 Ferst Drive, Atlanta, GA 30332; Department of Orthopaedics, Emory University, 59 Executive Park South, Atlanta, GA 30329
| | - J. Brandon Dixon
- George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, 801 Ferst Drive, Atlanta, GA 30332; Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, 313 Ferst Drive, Atlanta, GA 30332
| |
Collapse
|
19
|
Kenney HM, Wu CL, Loiselle AE, Xing L, Ritchlin CT, Schwarz EM. Single-cell transcriptomics of popliteal lymphatic vessels and peripheral veins reveals altered lymphatic muscle and immune cell populations in the TNF-Tg arthritis model. Arthritis Res Ther 2022; 24:64. [PMID: 35255954 PMCID: PMC8900348 DOI: 10.1186/s13075-022-02730-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Accepted: 01/21/2022] [Indexed: 11/23/2022] Open
Abstract
Background Lymphatic dysfunction exists in tumor necrosis factor transgenic (TNF-Tg) mice and rheumatoid arthritis (RA) patients. While joint-draining TNF-Tg popliteal lymphatic vessels (PLVs) have deficits in contractility during end-stage arthritis, the nature of lymphatic muscle cells (LMCs) and their TNF-altered transcriptome remain unknown. Thus, we performed single-cell RNA-sequencing (scRNAseq) on TNF-Tg LMCs in PLVs efferent to inflamed joints versus wild-type (WT) controls. Methods Single-cell suspensions of PLVs were sorted for smooth muscle cells (SMCs), which was validated by Cspg4-Cre;tdTomato reporter gene expression. Single-cell RNA-seq was performed on a 10x Genomics platform and analyzed using the Seurat R package. Uniform Manifold Approximation and Projections (UMAPs) and Ingenuity Pathway Analysis software were used to assess cell clusters and functional genomics in WT vs. TNF-Tg populations. Results Fluorescent imaging of Cspg4-Cre;tdTomato vessels demonstrated dim PLVs and strong reporter gene expression in the adjacent superficial saphenous vein, which was corroborated by flow cytometry of LMCs and vascular smooth muscle cells (VSMCs) from these vessels. Due to their unique morphology, these populations could also be readily detected by scatter analysis of cells from non-fluorescent mice. Bioinformatics analysis of flow sorted WT and TNF-Tg cells identified 20 unique cell clusters that together were 22.4% LMCs, 15.0% VSMCs, and 62.6% non-muscle cells of 8879 total cells. LMCs and M2-macrophages were decreased, while inflammatory monocytes were increased in TNF-Tg lower limb vasculature. SMC populations were defined by Cald1, Tpm1, and Pdgfrb expression and were enriched in myofibroblast-like gene expression. TNF-Tg LMCs exhibited enhanced functional genomics associated with cell death, phagocyte recruitment, and joint inflammation. Among the most prominent TNF-induced genes in SMCs were Mmp3, Cxcl12, and Ccl19, and the most downregulated genes were Zbtb16, Galnt15, and Apod. Conclusions Single-cell RNA-seq can be used to investigate functional genomics of lower limb vasculature in mice. Our findings confirm the inflammatory transcriptome of TNF-Tg vessels and altered gene expression in SMC populations. This study further supports a potential role of mesenchymal stromal cells in inflammatory-erosive arthritis pathogenesis, and warrants future studies to define the effects of this TNF-altered transcriptome on PLV function and joint homeostasis. Supplementary Information The online version contains supplementary material available at 10.1186/s13075-022-02730-z.
Collapse
Affiliation(s)
- H Mark Kenney
- Center for Musculoskeletal Research, University of Rochester Medical Center, 601 Elmwood Ave, Box 665, Rochester, NY, 14642, USA.,Department of Pathology & Laboratory Medicine, University of Rochester Medical Center, Rochester, NY, USA
| | - Chia-Lung Wu
- Center for Musculoskeletal Research, University of Rochester Medical Center, 601 Elmwood Ave, Box 665, Rochester, NY, 14642, USA.,Department of Orthopaedics, University of Rochester Medical Center, Rochester, NY, USA
| | - Alayna E Loiselle
- Center for Musculoskeletal Research, University of Rochester Medical Center, 601 Elmwood Ave, Box 665, Rochester, NY, 14642, USA.,Department of Pathology & Laboratory Medicine, University of Rochester Medical Center, Rochester, NY, USA.,Department of Orthopaedics, University of Rochester Medical Center, Rochester, NY, USA
| | - Lianping Xing
- Center for Musculoskeletal Research, University of Rochester Medical Center, 601 Elmwood Ave, Box 665, Rochester, NY, 14642, USA.,Department of Pathology & Laboratory Medicine, University of Rochester Medical Center, Rochester, NY, USA
| | - Christopher T Ritchlin
- Center for Musculoskeletal Research, University of Rochester Medical Center, 601 Elmwood Ave, Box 665, Rochester, NY, 14642, USA.,Department of Medicine, Division of Allergy, Immunology, Rheumatology, University of Rochester Medical Center, Rochester, NY, USA
| | - Edward M Schwarz
- Center for Musculoskeletal Research, University of Rochester Medical Center, 601 Elmwood Ave, Box 665, Rochester, NY, 14642, USA. .,Department of Pathology & Laboratory Medicine, University of Rochester Medical Center, Rochester, NY, USA. .,Department of Orthopaedics, University of Rochester Medical Center, Rochester, NY, USA. .,Department of Medicine, Division of Allergy, Immunology, Rheumatology, University of Rochester Medical Center, Rochester, NY, USA.
| |
Collapse
|
20
|
Lam AD, Cao E, Leong N, Gracia G, J. H. Porter C, Feeney OM, Trevaskis NL. Intra-articular injection of biologic anti-rheumatic drugs enhances local exposure to the joint-draining lymphatics. Eur J Pharm Biopharm 2022; 173:34-44. [DOI: 10.1016/j.ejpb.2022.02.014] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 02/14/2022] [Accepted: 02/21/2022] [Indexed: 12/27/2022]
|
21
|
Kemp SS, Penn MR, Koller GM, Griffin CT, Davis GE. Proinflammatory mediators, TNFα, IFNγ, and thrombin, directly induce lymphatic capillary tube regression. Front Cell Dev Biol 2022; 10:937982. [PMID: 35927983 PMCID: PMC9343954 DOI: 10.3389/fcell.2022.937982] [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: 05/06/2022] [Accepted: 06/30/2022] [Indexed: 11/30/2022] Open
Abstract
In this work, we sought to investigate the direct effects of proinflammatory mediators on lymphatic endothelial cell (LEC) capillaries and whether they might induce regression. Our laboratory has developed novel in-vitro, serum-free, lymphatic tubulogenesis assay models whereby human LEC tube networks readily form in either three-dimensional collagen or fibrin matrices. These systems were initially conceptualized in the hopes of better understanding the influence of proinflammatory mediators on LEC capillaries. In this work, we have screened and identified proinflammatory mediators that cause regression of LEC tube networks, the most potent of which is TNFα (tumor necrosis factor alpha), followed by IFNγ (interferon gamma) and thrombin. When these mediators were combined, even greater and more rapid lymphatic capillary regression occurred. Surprisingly, IL-1β (interleukin-1 beta), one of the most potent and pathologic cytokines known, had no regressive effect on these tube networks. Finally, we identified new pharmacological drug combinations capable of rescuing LEC capillaries from regression in response to the potent combination of TNFα, IFNγ, and thrombin. We speculate that protecting lymphatic capillaries from regression may be an important step toward mitigating a wide variety of acute and chronic disease states, as lymphatics are believed to clear both proinflammatory cells and mediators from inflamed and damaged tissue beds. Overall, these studies identify key proinflammatory mediators, including TNFα, IFNγ, and thrombin, that induce regression of LEC tube networks, as well as identify potential therapeutic agents to diminish LEC capillary regression responses.
Collapse
Affiliation(s)
- Scott S Kemp
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida School of Medicine, Tampa, FL, United States
| | - Marlena R Penn
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida School of Medicine, Tampa, FL, United States
| | - Gretchen M Koller
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida School of Medicine, Tampa, FL, United States
| | - Courtney T Griffin
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, United States
| | - George E Davis
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida School of Medicine, Tampa, FL, United States
| |
Collapse
|
22
|
Bernard FC, Kaiser J, Raval SK, Nepiyushchikh ZV, Doan TN, Willett NJ, Dixon JB. Multichromatic near-infrared imaging to assess interstitial lymphatic and venous uptake in vivo. JOURNAL OF BIOMEDICAL OPTICS 2021; 26:JBO-210078R. [PMID: 34881527 PMCID: PMC8654485 DOI: 10.1117/1.jbo.26.12.126001] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Accepted: 10/21/2021] [Indexed: 05/12/2023]
Abstract
SIGNIFICANCE Changes in interstitial fluid clearance are implicated in many diseases. Using near-infrared (NIR) imaging with properly sized tracers could enhance our understanding of how venous and lymphatic drainage are involved in disease progression or enhance drug delivery strategies. AIM We investigated multichromatic NIR imaging with multiple tracers to assess in vivo microvascular clearance kinetics and pathways in different tissue spaces. APPROACH We used a chemically inert IR Dye 800CW (D800) to target venous capillaries and a purified conjugate of IR dye 680RD with 40 kDa PEG (P40D680) to target lymphatic capillaries in vivo. Optical imaging settings were validated and tuned in vitro using tissue phantoms. We investigated multichromatic NIR imaging's utility in two in vivo tissue beds: the mouse tail and rat knee joint. We then tested the ability of the approach to detect interstitial fluid perturbations due to exercise. RESULTS In an in vitro simulated tissue environment, free dye and PEG mixture allowed for simultaneous detection without interference. In the mouse tail, co-injected NIR tracers cleared from the interstitial space via distinct routes, suggestive of lymphatic and venous uptake mechanisms. In the rat knee, we determined that exercise after injection transiently increased lymphatic drainage as measured by lower normalized intensity immediately after exercise, whereas exercise pre-injection exhibited a transient delay in clearance from the joint. CONCLUSIONS NIR imaging enables simultaneous imaging of lymphatic and venous-mediated fluid clearance with great sensitivity and can be used to measure temporal changes in clearance rates and pathways.
Collapse
Affiliation(s)
- Fabrice C. Bernard
- Georgia Institute of Technology and Emory University, Wallace H. Coulter Department of Biomedical Engineering, Atlanta, Georgia, United States
| | - Jarred Kaiser
- Emory University, Department of Orthopaedics, Atlanta, Georgia, United States
| | - Sarvgna K. Raval
- Emory University, Department of Orthopaedics, Atlanta, Georgia, United States
| | - Zhanna V. Nepiyushchikh
- Georgia Institute of Technology, George W. Woodruff School of Mechanical Engineering, Atlanta, Georgia, United States
| | - Thanh N. Doan
- Emory University, Department of Orthopaedics, Atlanta, Georgia, United States
| | - Nick J. Willett
- Georgia Institute of Technology and Emory University, Wallace H. Coulter Department of Biomedical Engineering, Atlanta, Georgia, United States
- Emory University, Department of Orthopaedics, Atlanta, Georgia, United States
- Atlanta Veteran’s Affairs Medical Center, Department of Orthopaedics, Atlanta, Georgia, United States
- Georgia Institute of Technology, Parker H. Petit Institute for Bioengineering and Bioscience, Atlanta, Georgia, United States
| | - J. Brandon Dixon
- Georgia Institute of Technology and Emory University, Wallace H. Coulter Department of Biomedical Engineering, Atlanta, Georgia, United States
- Georgia Institute of Technology, George W. Woodruff School of Mechanical Engineering, Atlanta, Georgia, United States
- Georgia Institute of Technology, Parker H. Petit Institute for Bioengineering and Bioscience, Atlanta, Georgia, United States
| |
Collapse
|
23
|
Scallan JP, Bouta EM, Rahimi H, Kenney HM, Ritchlin CT, Davis MJ, Schwarz EM. Ex vivo Demonstration of Functional Deficiencies in Popliteal Lymphatic Vessels From TNF-Transgenic Mice With Inflammatory Arthritis. Front Physiol 2021; 12:745096. [PMID: 34646163 PMCID: PMC8503619 DOI: 10.3389/fphys.2021.745096] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Accepted: 09/01/2021] [Indexed: 12/01/2022] Open
Abstract
Background: Recent studies demonstrated lymphangiogenesis and expansion of draining lymph nodes during chronic inflammatory arthritis, and lymphatic dysfunction associated with collapse of draining lymph nodes in rheumatoid arthritis (RA) patients and TNF-transgenic (TNF-Tg) mice experiencing arthritic flare. As the intrinsic differences between lymphatic vessels afferent to healthy, expanding, and collapsed draining lymph nodes are unknown, we characterized the ex vivo behavior of popliteal lymphatic vessels (PLVs) from WT and TNF-Tg mice. We also interrogated the mechanisms of lymphatic dysfunction through inhibition of nitric oxide synthase (NOS). Methods: Popliteal lymph nodes (PLNs) in TNF-Tg mice were phenotyped as Expanding or Collapsed by in vivo ultrasound and age-matched to WT littermate controls. The PLVs were harvested and cannulated for ex vivo functional analysis over a relatively wide range of hydrostatic pressures (0.5-10 cmH2O) to quantify the end diastolic diameter (EDD), tone, amplitude (AMP), ejection fraction (EF), contraction frequency (FREQ), and fractional pump flow (FPF) with or without NOS inhibitors Data were analyzed using repeated measures two-way ANOVA with Bonferroni's post hoc test. Results: Real time videos of the cannulated PLVs demonstrated the predicted phenotypes of robust vs. weak contractions of the WT vs. TNF-Tg PLV, respectively. Quantitative analyses confirmed that TNF-Tg PLVs had significantly decreased AMP, EF, and FPF vs. WT (p < 0.05). EF and FPF were recovered by NOS inhibition, while the reduction in AMP was NOS independent. No differences in EDD, tone, or FREQ were observed between WT and TNF-Tg PLVs, nor between Expanding vs. Collapsed PLVs. Conclusion: These findings support the concept that chronic inflammatory arthritis leads to NOS dependent and independent draining lymphatic vessel dysfunction that exacerbates disease, and may trigger arthritic flare due to decreased egress of inflammatory cells and soluble factors from affected joints.
Collapse
Affiliation(s)
- Joshua P. Scallan
- Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, MO, United States
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, Tampa, FL, United States
| | - Echoe M. Bouta
- Center for Musculoskeletal Research, Rochester, NY, United States
- Department of Biomedical Engineering, Rochester, MI, United States
| | - Homaira Rahimi
- Center for Musculoskeletal Research, Rochester, NY, United States
- Department of Pediatrics, Rochester, NY, United States
- Department of Pathology and Laboratory Medicine, Rochester, NY, United States
| | - H. Mark Kenney
- Center for Musculoskeletal Research, Rochester, NY, United States
- Department of Pathology and Laboratory Medicine, Rochester, NY, United States
| | - Christopher T. Ritchlin
- Center for Musculoskeletal Research, Rochester, NY, United States
- Division of Allergy, Immunology, Rheumatology, Department of Medicine, Rochester, NY, United States
| | - Michael J. Davis
- Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, MO, United States
| | - Edward M. Schwarz
- Center for Musculoskeletal Research, Rochester, NY, United States
- Department of Biomedical Engineering, Rochester, MI, United States
- Department of Pathology and Laboratory Medicine, Rochester, NY, United States
- Department of Orthopaedics, University of Rochester School of Medicine and Dentistry, Rochester, NY, United States
| |
Collapse
|
24
|
Liang Q, Zhang L, Xu H, Li J, Chen Y, Schwarz EM, Shi Q, Wang Y, Xing L. Lymphatic muscle cells contribute to dysfunction of the synovial lymphatic system in inflammatory arthritis in mice. Arthritis Res Ther 2021; 23:58. [PMID: 33602317 PMCID: PMC7893868 DOI: 10.1186/s13075-021-02438-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2020] [Accepted: 02/07/2021] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND Our previous studies reveal that impaired draining function of the synovial lymphatic vessel (LV) contributes to the pathogenesis of inflammatory arthritis, but the cellular and molecular mechanisms involved are not fully understood. OBJECTIVE To investigate the involvement of lymphatic muscle cells (LMCs) in mediating impaired LV function in inflammatory arthritis. METHODS TNF transgenic (TNF-Tg) arthritic mice were used. The structure and function of the LVs that drained the hind limbs were examined by whole-mount immunofluorescence staining, electron microscopy, and near-infrared lymphatic imaging. Primary LMCs were treated with TNF, and the changes in proliferation, apoptosis, and functional gene expression were assessed. The roles of the herbal drug, Panax notoginseng saponins (PNS), in arthritis and LVs were studied. RESULTS TNF-Tg mice developed ankle arthritis with age, which was associated with abnormalities of LVs: (1) dilated capillary LVs with few branch points, (2) mature LVs with reduced LMC coverage and draining function, and (3) degenerative and apoptotic appearance of LMCs. TNF caused LMC apoptosis, reduced expression of muscle functional genes, and promoted the production of nitric oxide (NO) by lymphatic endothelial cells (LECs). PNS attenuated arthritis, restored LMC coverage and draining function of mature LVs, inhibited TNF-mediated NO expression, and reduced LMC apoptosis. CONCLUSION The impaired draining function of LVs in TNF-Tg mice involves LMC apoptosis. TNF promotes LMC death directly and indirectly via NO production by LECs. PNS attenuates arthritis, improves LVs, and prevents TNF-induced LMC apoptosis by inhibiting NO production of LECs. LMCs contribute to the dysfunction of synovial LVs in inflammatory arthritis.
Collapse
Affiliation(s)
- Qianqian Liang
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
- Key Laboratory of Theory and Therapy of Muscles and Bones, Ministry of Education, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
- Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, 601 Elmwood Avenue, Rochester, NY, 14642, USA
| | - Li Zhang
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
- Key Laboratory of Theory and Therapy of Muscles and Bones, Ministry of Education, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
| | - Hao Xu
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
- Key Laboratory of Theory and Therapy of Muscles and Bones, Ministry of Education, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
- Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, 601 Elmwood Avenue, Rochester, NY, 14642, USA
| | - Jinlong Li
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
- Key Laboratory of Theory and Therapy of Muscles and Bones, Ministry of Education, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
| | - Yan Chen
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
- Key Laboratory of Theory and Therapy of Muscles and Bones, Ministry of Education, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
| | - Edward M Schwarz
- Center for Musculoskeletal Research, University of Rochester Medical Center, 601 Elmwood Avenue, Rochester, NY, 14642, USA
| | - Qi Shi
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
- Key Laboratory of Theory and Therapy of Muscles and Bones, Ministry of Education, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
| | - Yongjun Wang
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China.
- Key Laboratory of Theory and Therapy of Muscles and Bones, Ministry of Education, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China.
- Institute of Spine, Shanghai University of Traditional Chinese Medicine, 725 Wan-Ping South Road, Shanghai, 200032, China.
| | - Lianping Xing
- Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, 601 Elmwood Avenue, Rochester, NY, 14642, USA.
- Center for Musculoskeletal Research, University of Rochester Medical Center, 601 Elmwood Avenue, Rochester, NY, 14642, USA.
| |
Collapse
|
25
|
Bell RD, Rahimi H, Kenney HM, Lieberman AA, Wood RW, Schwarz EM, Ritchlin CT. Altered Lymphatic Vessel Anatomy and Markedly Diminished Lymph Clearance in Affected Hands of Patients With Active Rheumatoid Arthritis. Arthritis Rheumatol 2021; 72:1447-1455. [PMID: 32420693 DOI: 10.1002/art.41311] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Accepted: 05/05/2020] [Indexed: 01/17/2023]
Abstract
OBJECTIVE To assess differences between lymphatic function in the affected hands of rheumatoid arthritis (RA) patients with active synovitis and that of healthy controls, using indocyanine green (ICG) dye and near-infrared (NIR) imaging. METHODS NIR imaging of the hands of 8 patients with active RA and 13 healthy controls was performed following web space injection of 0.1 ml of 100 μM ICG. The percentage of ICG retention in the web spaces was determined by NIR imaging at baseline and at 7 days (±1 day) after the initial injections; image analysis provided contraction frequency. ICG+ lymphatic vessel (LV) length and branching architecture were assessed. RESULTS Retention of ICG in RA hands was higher compared to controls (P < 0.01). The average contraction frequency of ICG+ LVs in RA patients and in controls did not differ (mean ± SD 0.53 ± 0.39 contractions/minute versus 0.51 ± 0.35 contractions/minute). Total ICG+ LV length in RA hands was lower compared to healthy controls (58.3 ± 15.0 cm versus 71.4 ± 16.1 cm; P < 0.001), concomitant with a decrease in the number of ICG+ basilic LVs in the hands of RA patients (P < 0.05). CONCLUSION Lymphatic drainage in the hands of RA patients with active disease was reduced compared to controls. This reduction was associated with a decrease in total length of ICG+ LVs on the dorsal surface of the hands, which continued to contract at a similar rate to that observed in controls. These findings provide a plausible mechanism for exacerbation of synovitis and joint damage, specifically the accumulation and retention of inflammatory cells and catabolic factors in RA joints due to impaired efferent lymphatic flow. NIR/ICG imaging of RA hands is feasible and warrants formal investigation as a primary outcome measure for arthritis disease severity and/or persistence in future clinical trials.
Collapse
Affiliation(s)
- Richard D Bell
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, New York
| | - Homaira Rahimi
- University of Rochester Medical Center, Rochester, New York
| | - H Mark Kenney
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, New York
| | | | - Ronald W Wood
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, New York
| | - Edward M Schwarz
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, New York
| | | |
Collapse
|
26
|
Kenney HM, Bell RD, Masters EA, Xing L, Ritchlin CT, Schwarz EM. Lineage tracing reveals evidence of a popliteal lymphatic muscle progenitor cell that is distinct from skeletal and vascular muscle progenitors. Sci Rep 2020; 10:18088. [PMID: 33093635 PMCID: PMC7581810 DOI: 10.1038/s41598-020-75190-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2020] [Accepted: 10/12/2020] [Indexed: 12/31/2022] Open
Abstract
Loss of popliteal lymphatic vessel (PLV) contractions, which is associated with damage to lymphatic muscle cells (LMCs), is a biomarker of disease progression in mice with inflammatory arthritis. Currently, the nature of LMC progenitors has yet to be formally described. Thus, we aimed to characterize the progenitors of PLV-LMCs during murine development, towards rational therapies that target their proliferation, recruitment, and differentiation onto PLVs. Since LMCs have been described as a hybrid phenotype of striated and vascular smooth muscle cells (VSMCs), we performed lineage tracing studies in mice to further clarify this enigma by investigating LMC progenitor contribution to PLVs in neonatal mice. PLVs from Cre-tdTomato reporter mice specific for progenitors of skeletal myocytes (Pax7+ and MyoD+) and VSMCs (Prrx1+ and NG2+) were analyzed via whole mount immunofluorescent microscopy. The results showed that PLV-LMCs do not derive from skeletal muscle progenitors. Rather, PLV-LMCs originate from Pax7-/MyoD-/Prrx1+/NG2+ progenitors similar to VSMCs prior to postnatal day 10 (P10), and from a previously unknown Pax7-/MyoD-/Prrx1+/NG2- muscle progenitor pathway during development after P10. Future studies of these LMC progenitors during maintenance and repair of PLVs, along with their function in other lymphatic beds, are warranted.
Collapse
Affiliation(s)
- H Mark Kenney
- Center for Musculoskeletal Research, University of Rochester Medical Center, Box 665, 601 Elmwood Ave, Rochester, 14642, NY, USA.,Department of Pathology & Laboratory Medicine, University of Rochester Medical Center, Rochester, NY, USA
| | - Richard D Bell
- Center for Musculoskeletal Research, University of Rochester Medical Center, Box 665, 601 Elmwood Ave, Rochester, 14642, NY, USA.,Department of Pathology & Laboratory Medicine, University of Rochester Medical Center, Rochester, NY, USA
| | - Elysia A Masters
- Center for Musculoskeletal Research, University of Rochester Medical Center, Box 665, 601 Elmwood Ave, Rochester, 14642, NY, USA.,Department of Biomedical Engineering, University of Rochester, Rochester, NY, USA
| | - Lianping Xing
- Center for Musculoskeletal Research, University of Rochester Medical Center, Box 665, 601 Elmwood Ave, Rochester, 14642, NY, USA.,Department of Pathology & Laboratory Medicine, University of Rochester Medical Center, Rochester, NY, USA
| | - Christopher T Ritchlin
- Center for Musculoskeletal Research, University of Rochester Medical Center, Box 665, 601 Elmwood Ave, Rochester, 14642, NY, USA.,Division of Allergy, Immunology, Rheumatology, Department of Medicine, University of Rochester Medical Center, Rochester, NY, USA
| | - Edward M Schwarz
- Center for Musculoskeletal Research, University of Rochester Medical Center, Box 665, 601 Elmwood Ave, Rochester, 14642, NY, USA. .,Department of Pathology & Laboratory Medicine, University of Rochester Medical Center, Rochester, NY, USA. .,Department of Biomedical Engineering, University of Rochester, Rochester, NY, USA. .,Division of Allergy, Immunology, Rheumatology, Department of Medicine, University of Rochester Medical Center, Rochester, NY, USA. .,Department of Orthopaedics, University of Rochester Medical Center, Rochester, NY, USA.
| |
Collapse
|
27
|
Lymph Node Stromal Cells: Mapmakers of T Cell Immunity. Int J Mol Sci 2020; 21:ijms21207785. [PMID: 33096748 PMCID: PMC7588999 DOI: 10.3390/ijms21207785] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Revised: 10/15/2020] [Accepted: 10/18/2020] [Indexed: 12/15/2022] Open
Abstract
Stromal cells (SCs) are strategically positioned in both lymphoid and nonlymphoid organs to provide a scaffold and orchestrate immunity by modulating immune cell maturation, migration and activation. Recent characterizations of SCs have expanded our understanding of their heterogeneity and suggested a functional specialization of distinct SC subsets, further modulated by the microenvironment. Lymph node SCs (LNSCs) have been shown to be particularly important in maintaining immune homeostasis and T cell tolerance. Under inflammation situations, such as viral infections or tumor development, SCs undergo profound changes in their numbers and phenotype and play important roles in contributing to either the activation or the control of T cell immunity. In this review, we highlight the role of SCs located in LNs in shaping peripheral T cell responses in different immune contexts, such as autoimmunity, viral and cancer immunity.
Collapse
|
28
|
Sintou A, Mansfield C, Iacob A, Chowdhury RA, Narodden S, Rothery SM, Podovei R, Sanchez-Alonso JL, Ferraro E, Swiatlowska P, Harding SE, Prasad S, Rosenthal N, Gorelik J, Sattler S. Mediastinal Lymphadenopathy, Class-Switched Auto-Antibodies and Myocardial Immune-Complexes During Heart Failure in Rodents and Humans. Front Cell Dev Biol 2020; 8:695. [PMID: 32850816 PMCID: PMC7426467 DOI: 10.3389/fcell.2020.00695] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Accepted: 07/08/2020] [Indexed: 11/13/2022] Open
Abstract
Mediastinal lymphadenopathy and auto-antibodies are clinical phenomena during ischemic heart failure pointing to an autoimmune response against the heart. T and B cells have been convincingly demonstrated to be activated after myocardial infarction, a prerequisite for the generation of mature auto-antibodies. Yet, little is known about the immunoglobulin isotype repertoire thus pathological potential of anti-heart auto-antibodies during heart failure. We obtained human myocardial tissue from ischemic heart failure patients and induced experimental MI in rats. We found that anti-heart autoimmunity persists during heart failure. Rat mediastinal lymph nodes are enlarged and contain active secondary follicles with mature isotype-switched IgG2a B cells. Mature IgG2a auto-antibodies specific for cardiac antigens are present in rat heart failure serum, and IgG and complement C3 deposits are evident in heart failure tissue of both rats and human patients. Previously established myocardial inflammation, and the herein provided proof of B cell maturation in lymph nodes and myocardial deposition of mature auto-antibodies, provide all the hallmark signs of an established autoimmune response in chronic heart failure.
Collapse
Affiliation(s)
- Amalia Sintou
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Catherine Mansfield
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Alma Iacob
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
- Royal Brompton Hospital, Royal Brompton and Harefield NHS Foundation Trust, London, United Kingdom
| | - Rasheda A. Chowdhury
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Salomon Narodden
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Stephen M. Rothery
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Robert Podovei
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | | | - Elisa Ferraro
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Pamela Swiatlowska
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Sian E. Harding
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Sanjay Prasad
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
- Royal Brompton Hospital, Royal Brompton and Harefield NHS Foundation Trust, London, United Kingdom
| | - Nadia Rosenthal
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
- The Jackson Laboratory, Bar Harbor, ME, United States
| | - Julia Gorelik
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Susanne Sattler
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| |
Collapse
|
29
|
Guang Y, McGrath TM, Klug NR, Nims RJ, Shih CC, Bayguinov PO, Guilak F, Pham CTN, Fitzpatrick JAJ, Setton LA. Combined Experimental Approach and Finite Element Modeling of Small Molecule Transport Through Joint Synovium to Measure Effective Diffusivity. J Biomech Eng 2020; 142:041010. [PMID: 31536113 PMCID: PMC7104772 DOI: 10.1115/1.4044892] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Revised: 08/14/2019] [Indexed: 11/08/2022]
Abstract
Trans-synovial solute transport plays a critical role in the clearance of intra-articularly (IA) delivered drugs. In this study, we present a computational finite element model (FEM) of solute transport through the synovium validated by experiments on synovial explants. Unsteady diffusion of urea, a small uncharged molecule, was measured through devitalized porcine and human synovium using custom-built diffusion chambers. A multiphasic computational model was constructed and optimized with the experimental data to extract effective diffusivity for urea within the synovium. A monotonic decrease in urea concentration was observed in the donor bath over time, with an effective diffusivity found to be an order of magnitude lower in synovium versus that measured in free solution. Parametric studies incorporating an intimal cell layer with varying thickness and varying effective diffusivities were performed, revealing a dependence of drug clearance kinetics on both parameters. The findings of this study indicate that the synovial matrix impedes urea solute transport out of the joint with little retention of the solute in the matrix.
Collapse
Affiliation(s)
- Young Guang
- Department of Biomedical Engineering, Washington University in
St. Louis, Whitaker Hall, 1 Brookings Dr., St.
Louis, MO 63130
e-mail:
| | - Tom M. McGrath
- Department of Biomedical Engineering, Washington University in
St. Louis, Whitaker Hall, 1 Brookings Dr., St.
Louis, MO 63130
e-mail:
| | - Natalie R. Klug
- Department of Biomedical Engineering, Washington University in
St. Louis, Whitaker Hall, 1 Brookings Dr., St.
Louis, MO 63130
e-mail:
| | - Robert J. Nims
- Department of Orthopaedic Surgery, Washington University School
of Medicine, St. Louis, MO 63110
e-mail:
| | - Chien-Cheng Shih
- Center for Cellular Imaging, Department of Neuroscience,
Washington University School of Medicine, St.
Louis, MO 63110
e-mail:
| | - Peter O. Bayguinov
- Center for Cellular Imaging, Department of Neuroscience,
Washington University School of Medicine, St.
Louis, MO 63110
e-mail:
| | - Farshid Guilak
- Department of Orthopaedic Surgery, Washington University
School of Medicine, St. Louis, MO
63110 e-mail:
| | - Christine T. N. Pham
- Division of Rheumatology, Washington University School of
Medicine, St. Louis, MO 63110
e-mail:
| | - James A. J. Fitzpatrick
- Scientific Director Center for Cellular Imaging, Department of
Neuroscience, Department Cell Biology & Physiology and
Neuroscience, Washington University School of Medicine,
St. Louis, MO 63110;Department of Biomedical Engineering, Washington University in
St. Louis, Whitaker Hall, 1 Brookings Dr., St.
Louis, MO 63130
e-mail:
| | - Lori A. Setton
- Department of Biomedical Engineering, Washington University in
St. Louis, Whitaker Hall, 1 Brookings Dr., St.
Louis, MO 63130
e-mail:
| |
Collapse
|
30
|
Hou T, Liu Y, Wang X, Jiao D, Xu H, Shi Q, Wang Y, Li W, Wu T, Liang Q. Ginsenoside Rg1 promotes lymphatic drainage and improves chronic inflammatory arthritis. JOURNAL OF MUSCULOSKELETAL & NEURONAL INTERACTIONS 2020; 20:526-534. [PMID: 33265080 PMCID: PMC7716681] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
OBJECTIVE The lymphatic system plays an important role in joint diseases. This study aimed to evaluate the effects of ginsenoside Rg1 on lymphatic drainage and accumulation of inflammatory products in the joints. METHODS Two-month-old transgenic mice that overexpress tumor necrosis factor alpha (TNF-α; TNF-Tg) were used as the animal models. Ginsenoside Rg1 was administered for 12 weeks and the lymphatic drainage in the mice was evaluated using near infrared-indocyanine green (NIR-ICG) lymphatic imaging system. The clinical symptoms of arthritis were evaluated weekly. The ankle and knee joints were harvested for hematoxylin-eosin (HE), alcian blue/orange G (ABOG), and tartrate-resistant acid phosphatase (TRAP) staining, and the foot dorsal skin was used for whole-mount immuno-staining. Simultaneously, the serum levels of IL-6 and TNF-α were detected using enzyme-linked immunosorbent assay (ELISA). RESULTS Ginsenoside Rg1 significantly improved the lymphatic drainage function, reduced synovial inflammation and bone erosion, decreased serum IL-6 and TNF-α concentration, and increased smooth muscle coverage on the collecting lymphatic vessels in the foot skin of the TNF-Tg mice. Furthermore, ginsenoside Rg1 treatment for 12 weeks did not cause any damage to the liver and kidney tissues. CONCLUSION Ginsenoside Rg1 improves lymphatic drainage and joint inflammation in TNF-Tg mice. Therefore, ginsenoside Rg1 has the potential to be a candidate drug for the treatment of inflammatory arthritis.
Collapse
Affiliation(s)
- Tong Hou
- Central Hospital of Jing’an District, Fudan University, Shanghai, China,Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yang Liu
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xiaoyun Wang
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Danli Jiao
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China,Tianshan traditional Chinese medicine hospital, Shanghai, China
| | - Hao Xu
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Qi Shi
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China,Key Laboratory of theory and therapy of muscles and bones, Ministry of Education (Shanghai University of Traditional Chinese Medicine), Shanghai, China
| | - Yongjun Wang
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China,Key Laboratory of theory and therapy of muscles and bones, Ministry of Education (Shanghai University of Traditional Chinese Medicine), Shanghai, China
| | - Wei Li
- Central Hospital of Jing’an District, Fudan University, Shanghai, China,Wei Li, Central Hospital of Jing’an District, 259 Xi-Kang Road, Jing’an District, Shanghai 200040, China E-mail:
| | - Tao Wu
- Huadong Hospital, Fudan University, Shanghai, China,Tao Wu, Huadong Hospital, 221 Yan-An West Road, Jing’an District, Shanghai, Shanghai 200040, China E-mail:
| | - Qianqian Liang
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China,Key Laboratory of theory and therapy of muscles and bones, Ministry of Education (Shanghai University of Traditional Chinese Medicine), Shanghai, China,Corresponding authors: Corresponding authors: Qianqian Liang, Institute of Spine, Shanghai University of Traditional Chinese Medicine, 725 Wan-Ping South Road, Shanghai 200032, China E-mail:
| |
Collapse
|
31
|
Bell RD, Slattery PN, Wu EK, Xing L, Ritchlin CT, Schwarz EM. iNOS dependent and independent phases of lymph node expansion in mice with TNF-induced inflammatory-erosive arthritis. Arthritis Res Ther 2019; 21:240. [PMID: 31727153 PMCID: PMC6854801 DOI: 10.1186/s13075-019-2039-z] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2019] [Accepted: 10/23/2019] [Indexed: 01/15/2023] Open
Abstract
Introduction A pivotal effect of lymphatic vessel (LV) function in joint homeostasis was identified in the tumor necrosis factor-transgenic (TNF-Tg) mouse model of rheumatoid arthritis (RA). Specifically, loss of LV contractions is associated with progressive synovitis and erosions. Furthermore, draining lymph node expansion is a biomarker of arthritic progression, and both macrophages and lymphatic endothelial cells express inducible nitric oxide synthase (iNOS), which disrupts LV contraction and transport of immune cells to the draining lymph nodes. Therefore, to directly assess these relationships, we tested the hypothesis that TNF-Tg mice with global genetic ablation of iNOS (iNOS−/−) will show delayed draining lymph node expansion, maintained LV contractions, and decreased synovitis and erosions. Method iNOS−/−× TNF-Tg female and male mice, and control littermates (iNOS−/−, TNF-Tg, and WT), were examined with (1) ultrasound to determine popliteal lymph node (PLN) volume and (2) near-infrared imaging (NIR) to assess popliteal LV contraction frequency, and differences between genotypes were assessed at 3, 4, 5, and 6 months of age. Knees and PLN were harvested at 4 months in females and 6 months in males, to assess synovitis, bone erosions, and cellular accumulation in PLN sinuses via histology. Results Initially, an increase in PLN volume was observed for both female and male iNOS−/−× TNF-Tg and TNF-Tg compared to their WT and iNOS−/− counterparts at 2 and 3 months, respectively. Subsequently, TNF-Tg PLNs continue to increase in volume, while iNOS−/−× TNF-Tg did not increase in volume from the initial timepoints. WT and iNOS−/− PLN volume was unchanged throughout the experiment. LV contraction frequency was increased at 4 months in females and 5 months in males, in the iNOS−/−× TNF-Tg mice compared to the TNF-Tg. Synovitis and erosions were moderately reduced in iNOS−/−× TNF-Tg versus TNF-Tg knees in females, while no differences in knee pathology were observed in males. Conclusions Genetic iNOS ablation maintains draining lymph node volume and LV function during TNF-induced inflammatory arthritis and is associated with moderately decreased joint inflammation and damage.
Collapse
Affiliation(s)
- Richard D Bell
- Center for Musculoskeletal Research, University of Rochester Medical Center, 601 Elmwood Avenue, Box 665, Rochester, NY, 14642, USA.,Department of Pathology and Laboratory Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| | - Pamelia N Slattery
- Center for Musculoskeletal Research, University of Rochester Medical Center, 601 Elmwood Avenue, Box 665, Rochester, NY, 14642, USA.,Department of Biology, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| | - Emily K Wu
- Center for Musculoskeletal Research, University of Rochester Medical Center, 601 Elmwood Avenue, Box 665, Rochester, NY, 14642, USA.,Department of Microbiology & Immunology, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| | - Lianping Xing
- Center for Musculoskeletal Research, University of Rochester Medical Center, 601 Elmwood Avenue, Box 665, Rochester, NY, 14642, USA.,Department of Pathology and Laboratory Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| | - Christopher T Ritchlin
- Center for Musculoskeletal Research, University of Rochester Medical Center, 601 Elmwood Avenue, Box 665, Rochester, NY, 14642, USA.,Division of Allergy, Immunology, Rheumatology, Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| | - Edward M Schwarz
- Center for Musculoskeletal Research, University of Rochester Medical Center, 601 Elmwood Avenue, Box 665, Rochester, NY, 14642, USA. .,Department of Pathology and Laboratory Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA. .,Department of Microbiology & Immunology, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA. .,Department of Orthopaedics, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA.
| |
Collapse
|
32
|
Evans CH, Ghivizzani SC, Robbins PD. Gene Delivery to Joints by Intra-Articular Injection. Hum Gene Ther 2019; 29:2-14. [PMID: 29160173 DOI: 10.1089/hum.2017.181] [Citation(s) in RCA: 86] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Most forms of arthritis are incurable, difficult to treat, and a major cause of disability in Western countries. Better local treatment of arthritis is impaired by the pharmacokinetics of the joint that make it very difficult to deliver drugs to joints at sustained, therapeutic concentrations. This is especially true of biologic drugs, such as proteins and RNA, many of which show great promise in preclinical studies. Gene transfer provides a strategy for overcoming this limitation. The basic concept is to deliver cDNAs encoding therapeutic products by direct intra-articular injection, leading to sustained, endogenous synthesis of the gene products within the joint. Proof of concept has been achieved for both in vivo and ex vivo gene delivery using a variety of vectors, genes, and cells in several different animal models. There have been a small number of clinical trials for rheumatoid arthritis (RA) and osteoarthritis (OA) using retrovirus vectors for ex vivo gene delivery and adeno-associated virus (AAV) for in vivo delivery. AAV is of particular interest because, unlike other viral vectors, it is able to penetrate deep within articular cartilage and transduce chondrocytes in situ. This property is of particular importance in OA, where changes in chondrocyte metabolism are thought to be fundamental to the pathophysiology of the disease. Authorities in Korea have recently approved the world's first arthritis gene therapy. This targets OA by the injection of allogeneic chondrocytes that have been transduced with a retrovirus carrying transforming growth factor-β1 cDNA. Phase III studies are scheduled to start in the United States soon. Meanwhile, two additional Phase I trials are listed on Clinicaltrials.gov , both using AAV. One targets RA by transferring interferon-β, and the other targets OA by transferring interleukin-1 receptor antagonist. The field is thus gaining momentum and promises to improve the treatment of these common and debilitating diseases.
Collapse
Affiliation(s)
- Christopher H Evans
- 1 Rehabilitation Medicine Research Center, Mayo Clinic , Rochester, Minnesota
| | - Steven C Ghivizzani
- 2 Department of Orthopedics and Rehabilitation, University of Florida College of Medicine , Gainesville, Florida
| | - Paul D Robbins
- 3 Department of Metabolism and Aging, The Scripps Research Institute , Jupiter, Florida
| |
Collapse
|
33
|
Buckley CD, McGettrick HM. Leukocyte trafficking between stromal compartments: lessons from rheumatoid arthritis. Nat Rev Rheumatol 2018; 14:476-487. [DOI: 10.1038/s41584-018-0042-4] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
34
|
Yamakawa M, Doh SJ, Santosa SM, Montana M, Qin EC, Kong H, Han KY, Yu C, Rosenblatt MI, Kazlauskas A, Chang JH, Azar DT. Potential lymphangiogenesis therapies: Learning from current antiangiogenesis therapies-A review. Med Res Rev 2018. [PMID: 29528507 DOI: 10.1002/med.21496] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
In recent years, lymphangiogenesis, the process of lymphatic vessel formation from existing lymph vessels, has been demonstrated to have a significant role in diverse pathologies, including cancer metastasis, organ graft rejection, and lymphedema. Our understanding of the mechanisms of lymphangiogenesis has advanced on the heels of studies demonstrating vascular endothelial growth factor C as a central pro-lymphangiogenic regulator and others identifying multiple lymphatic endothelial biomarkers. Despite these breakthroughs and a growing appreciation of the signaling events that govern the lymphangiogenic process, there are no FDA-approved drugs that target lymphangiogenesis. In this review, we reflect on the lessons available from the development of antiangiogenic therapies (26 FDA-approved drugs to date), review current lymphangiogenesis research including nanotechnology in therapeutic drug delivery and imaging, and discuss molecules in the lymphangiogenic pathway that are promising therapeutic targets.
Collapse
Affiliation(s)
- Michael Yamakawa
- Department of Ophthalmology and Visual Sciences, Illinois Eye and Ear Infirmary, College of Medicine, University of Illinois at Chicago, Chicago, IL
| | - Susan J Doh
- Department of Ophthalmology and Visual Sciences, Illinois Eye and Ear Infirmary, College of Medicine, University of Illinois at Chicago, Chicago, IL
| | - Samuel M Santosa
- Department of Ophthalmology and Visual Sciences, Illinois Eye and Ear Infirmary, College of Medicine, University of Illinois at Chicago, Chicago, IL
| | - Mario Montana
- Department of Ophthalmology and Visual Sciences, Illinois Eye and Ear Infirmary, College of Medicine, University of Illinois at Chicago, Chicago, IL
| | - Ellen C Qin
- Department of Materials Science and Engineering, University of Illinois at Urbana-Champaign, Urbana, IL
| | - Hyunjoon Kong
- Department of Chemical and Biomolecular Engineering, University of Illinois at Urbana-Champaign, Urbana, IL
| | - Kyu-Yeon Han
- Department of Ophthalmology and Visual Sciences, Illinois Eye and Ear Infirmary, College of Medicine, University of Illinois at Chicago, Chicago, IL
| | - Charles Yu
- Department of Ophthalmology and Visual Sciences, Illinois Eye and Ear Infirmary, College of Medicine, University of Illinois at Chicago, Chicago, IL
| | - Mark I Rosenblatt
- Department of Ophthalmology and Visual Sciences, Illinois Eye and Ear Infirmary, College of Medicine, University of Illinois at Chicago, Chicago, IL
| | - Andrius Kazlauskas
- Department of Ophthalmology and Visual Sciences, Illinois Eye and Ear Infirmary, College of Medicine, University of Illinois at Chicago, Chicago, IL.,Department of Physiology and Biophysics, College of Medicine, University of Illinois at Chicago, Chicago, IL
| | - Jin-Hong Chang
- Department of Ophthalmology and Visual Sciences, Illinois Eye and Ear Infirmary, College of Medicine, University of Illinois at Chicago, Chicago, IL
| | - Dimitri T Azar
- Department of Ophthalmology and Visual Sciences, Illinois Eye and Ear Infirmary, College of Medicine, University of Illinois at Chicago, Chicago, IL
| |
Collapse
|
35
|
CT assessment of axillary lymphadenopathy in patients with rheumatoid arthritis: association with disease activity and severity. Rheumatol Int 2018; 38:1017-1022. [DOI: 10.1007/s00296-018-3992-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2017] [Accepted: 02/04/2018] [Indexed: 11/26/2022]
|
36
|
Targeting lymphatic function as a novel therapeutic intervention for rheumatoid arthritis. Nat Rev Rheumatol 2018; 14:94-106. [PMID: 29323343 DOI: 10.1038/nrrheum.2017.205] [Citation(s) in RCA: 85] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Although clinical outcomes for patients with rheumatoid arthritis (RA) have greatly improved with the use of biologic and conventional DMARDs, approximately 40% of patients do not achieve primary clinical outcomes in randomized trials, and only a small proportion achieve lasting remission. Over the past decade, studies in murine models point to the critical role of the lymphatic system in the pathogenesis and therapy of inflammatory-erosive arthritis, presumably by the removal of catabolic factors, cytokines and inflammatory cells from the inflamed synovium. Murine studies demonstrate that lymphatic drainage increases at the onset of inflammatory-erosive arthritis but, as inflammation progresses to a more chronic phase, lymphatic clearance declines and both structural and cellular changes are observed in the draining lymph node. Specifically, chronic damage to the lymphatic vessel from persistent inflammation results in loss of lymphatic vessel contraction followed by lymph node collapse, reduced lymphatic drainage, and ultimately severe synovitis and joint erosion. Notably, clinical pilot studies in patients with RA report lymph node changes following treatment, and thus draining lymphatic vessels and nodes could represent a potential biomarker of arthritis activity and response to therapy. Most importantly, targeting lymphatics represents an innovative strategy for therapeutic intervention for RA.
Collapse
|
37
|
Benson RA, McInnes IB, Garside P, Brewer JM. Model answers: Rational application of murine models in arthritis research. Eur J Immunol 2017; 48:32-38. [PMID: 29193037 PMCID: PMC5814907 DOI: 10.1002/eji.201746938] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2017] [Revised: 11/02/2017] [Accepted: 11/22/2017] [Indexed: 12/29/2022]
Abstract
Advances in targeted immune therapeutics have profoundly improved clinical outcomes for patients with inflammatory arthropathies particularly rheumatoid arthritis. The landscape of disease that is observed and the treatment outcomes desired for the future have also progressed. As such there is an increasing move away from traditional models of end‐stage, chronic disease with recognition of the need to consider the earliest phases of pathogenesis as a target for treatment leading to resolution and/or cure. In order to continue the discovery process and enhance our understanding of disease and treatment, we therefore need to continuously revisit the animal models we employ and assess their relevance and utility in the light of contemporary therapeutic goals. In this review, we highlight the areas where we consider new developments in animal models and their application are most required. Thus, we have contextualised the relevant mouse models and their use within the current concepts of human inflammatory arthritis pathogenesis and highlight areas of need.
Collapse
Affiliation(s)
- Robert A Benson
- Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary & Life Sciences, Sir Graeme Davies Building, University of Glasgow, Glasgow, UK
| | - Iain B McInnes
- Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary & Life Sciences, Sir Graeme Davies Building, University of Glasgow, Glasgow, UK
| | - Paul Garside
- Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary & Life Sciences, Sir Graeme Davies Building, University of Glasgow, Glasgow, UK
| | - James M Brewer
- Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary & Life Sciences, Sir Graeme Davies Building, University of Glasgow, Glasgow, UK
| |
Collapse
|
38
|
Aldrich MB, Velasquez FC, Kwon S, Azhdarinia A, Pinkston K, Harvey BR, Chan W, Rasmussen JC, Ross RF, Fife CE, Sevick-Muraca EM. Lymphatic delivery of etanercept via nanotopography improves response to collagen-induced arthritis. Arthritis Res Ther 2017; 19:116. [PMID: 28566090 PMCID: PMC5452411 DOI: 10.1186/s13075-017-1323-z] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2016] [Accepted: 05/09/2017] [Indexed: 12/21/2022] Open
Abstract
Background Evidence suggests lymphatic function mediates local rheumatoid arthritis (RA) flares. Yet biologics that target the immune system are dosed systemically via the subcutaneous (SC) administration route, thereby inefficiently reaching local lymphatic compartments. Nanotopography has previously been shown to disrupt tight cellular junctions, potentially enhancing local lymphatic delivery and potentially improving overall therapeutic efficacy. Method We first characterized nanotopography (SOFUSA™) delivery of an anti-TNF drug, etanercept, by comparing pharmacokinetic profiles to those obtained by conventional SC, intravenous (IV), and intradermal (ID) routes of administration, and assessed uptake of radiolabeled etanercept in draining lymph nodes (LNs) in single dosing studies. We then compared etanercept efficacy in a progressive rat model of collagen-induced arthritis (CIA), administered systemically via SC route of administration; via the regional lymphatics through ID delivery; or through a nanotopography (SOFUSA™) device at 10, 12, and 14 days post CIA induction. Measurements of hind limb swelling and near-infrared fluorescence (NIRF) imaging of afferent lymph pumping function and reflux were conducted on days 11, 13, and 18 post CIA induction and compared to untreated CIA animals. Univariate and multivariate analysis of variance were used to compare the group differences for percentage swelling and lymphatic contractile activity. Results Even though all three modes of administration delivered an equal amount of etanercept, SOFUSA™ delivery resulted in increased lymphatic pumping and significantly reduced swelling as compared to untreated, ID, and SC groups. Pharmacokinetic profiles in serum and LN uptake studies showed that using the nanotopography device resulted in the greatest uptake and retention in draining LNs. Conclusions Locoregional lymphatic delivery of biologics that target the immune system may have more favorable pharmacodynamics than SC or IV administration. Nanotopography may provide a more efficient method for delivery of anti-TNF drugs to reverse impairment of lymphatic function and reduce swelling associated with RA flares. Electronic supplementary material The online version of this article (doi:10.1186/s13075-017-1323-z) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Melissa B Aldrich
- The Center for Molecular Imaging, The Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center, Houston, TX, 77030, USA
| | - Fred C Velasquez
- The Center for Molecular Imaging, The Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center, Houston, TX, 77030, USA
| | - Sunkuk Kwon
- The Center for Molecular Imaging, The Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center, Houston, TX, 77030, USA
| | - Ali Azhdarinia
- The Center for Molecular Imaging, The Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center, Houston, TX, 77030, USA
| | - Kenneth Pinkston
- The Center for Molecular Imaging, The Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center, Houston, TX, 77030, USA
| | - Barrett R Harvey
- The Center for Molecular Imaging, The Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center, Houston, TX, 77030, USA
| | - Wenyaw Chan
- Department of Biostatistics, The School of Public Health, The University of Texas Health Science Center, Houston, TX, 77030, USA
| | - John C Rasmussen
- The Center for Molecular Imaging, The Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center, Houston, TX, 77030, USA
| | | | - Caroline E Fife
- The Wound Care Clinic, CHI St. Luke's Health, The Woodlands Hospital, The Woodlands, TX, 77382, USA
| | - E M Sevick-Muraca
- The Center for Molecular Imaging, The Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center, Houston, TX, 77030, USA.
| |
Collapse
|
39
|
Yoo J, Lee SK, Lim M, Sheen D, Choi EH, Kim SA. Exosomal amyloid A and lymphatic vessel endothelial hyaluronic acid receptor-1 proteins are associated with disease activity in rheumatoid arthritis. Arthritis Res Ther 2017; 19:119. [PMID: 28569211 PMCID: PMC5452405 DOI: 10.1186/s13075-017-1334-9] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2017] [Accepted: 05/15/2017] [Indexed: 01/22/2023] Open
Abstract
Background Exosomes are thought to play an important role in exchanging information between cells. The proteins and lipids in exosomes play roles in mediating inflammatory and autoimmune diseases. The aim of this study was to identify exosomal candidate proteins that are related to other inflammatory parameters in rheumatoid arthritis (RA). Methods The study population consisted of 60 patients with RA: 30 in the clinical remission (CR) group with a Disease Activity Score in 28 joints based on erythrocyte sedimentation rate (DAS28-ESR) ≤2.6 and 30 in the non-clinical remission (non-CR) group with a DAS28-ESR >2.6. Preparation of exosomes from patient serum samples was performed with the ExoQuick kit, and protein identification/quantification was performed using tandem mass tag labeling/mass spectrometry and an enzyme-linked immunosorbent assay. Comparisons between groups were made using Student’s t test or the Mann-Whitney U test, as appropriate. Spearman’s correlation coefficients (ρ) were calculated. Results We identified six candidate proteins. Exosomal levels of amyloid A (AA) and lymphatic vessel endothelial hyaluronic acid receptor-1 (LYVE-1) differed between the CR and non-CR groups. Both serum and exosomal AA levels were higher in the non-CR group than in the CR group (p = 0.001). Significant positive correlations were found between exosomal AA and C-reactive protein (CRP) as well as between serum AA and CRP (ρ = 0.614, p = 0.001, and ρ = 0.624, p = 0.001, respectively). Although serum levels of LYVE-1 did not differ between the non-CR and CR groups, exosomal levels of LYVE-1 were lower in the non-CR group than in the CR group (p = 0.01). We identified positive correlations between serum/exosomal LYVE-1 and CRP only in the non-CR group (serum ρ = 0.376, p = 0.04; exosome ρ = 0.545, p = 0.002). Conclusions Exosomal LYVE-1 shows potential for use as an additional marker of disease activity in patients with RA, and exosomes may carry other useful markers for RA.
Collapse
Affiliation(s)
- Jihyung Yoo
- Department of Internal Medicine, School of Medicine, Eulji University, Daejeon, Korea
| | - Sang Kwang Lee
- Eulji Medi-Bio Research Institute, Eulji University, Daejeon, Korea
| | - Mikyung Lim
- Department of Internal Medicine, School of Medicine, Eulji University, Daejeon, Korea
| | - Donghyuk Sheen
- Department of Internal Medicine, School of Medicine, Eulji University, Daejeon, Korea
| | - Eun-Hye Choi
- Eulji Medi-Bio Research Institute, Eulji University, Daejeon, Korea
| | - Soon Ae Kim
- Department of Pharmacology, School of Medicine, Eulji University, Daejeon, Korea.
| |
Collapse
|
40
|
Abouelkheir GR, Upchurch BD, Rutkowski JM. Lymphangiogenesis: fuel, smoke, or extinguisher of inflammation's fire? Exp Biol Med (Maywood) 2017; 242:884-895. [PMID: 28346012 DOI: 10.1177/1535370217697385] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Lymphangiogenesis is a recognized hallmark of inflammatory processes in tissues and organs as diverse as the skin, heart, bowel, and airways. In clinical and animal models wherein the signaling processes of lymphangiogenesis are manipulated, most studies demonstrate that an expanded lymphatic vasculature is necessary for the resolution of inflammation. The fundamental roles that lymphatics play in fluid clearance and immune cell trafficking from the periphery make these results seemingly obvious as a mechanism of alleviating locally inflamed environments: the lymphatics are simply providing a drain. Depending on the tissue site, lymphangiogenic mechanism, or induction timeframe, however, evidence shows that inflammation-associated lymphangiogenesis (IAL) may worsen the pathology. Recent studies have identified lymphatic endothelial cells themselves to be local regulators of immune cell activity and its consequential phenotypes - a more active role in inflammation regulation than previously thought. Indeed, results focusing on the immunocentric roles of peripheral lymphatic function have revealed that the basic drainage task of lymphatic vessels is a complex balance of locally processed and transported antigens as well as interstitial cytokine and immune cell signaling: an interplay that likely defines the function of IAL. This review will summarize the latest findings on how IAL impacts a series of disease states in various tissues in both preclinical models and clinical studies. This discussion will serve to highlight some emerging areas of lymphatic research in an attempt to answer the question relevant to an array of scientists and clinicians of whether IAL helps to fuel or extinguish inflammation. Impact statement Inflammatory progression is present in acute and chronic tissue pathologies throughout the body. Lymphatic vessels play physiological roles relevant to all medical fields as important regulators of fluid balance, immune cell trafficking, and immune identity. Lymphangiogenesis is often concurrent with inflammation and can potentially aide or worsen disease progression. How new lymphatic vessels impact inflammation and by which mechanism is an important consideration in current and future clinical therapies targeting inflammation and/or vasculogenesis. This review identifies, across a range of tissue-specific pathologies, the current understanding of inflammation-associated lymphangiogenesis in the progression or resolution of inflammation.
Collapse
Affiliation(s)
- Gabriella R Abouelkheir
- 1 Division of Lymphatic Biology, Department of Medical Physiology, Texas A&M College of Medicine, College Station, TX 77843, USA
| | - Bradley D Upchurch
- 1 Division of Lymphatic Biology, Department of Medical Physiology, Texas A&M College of Medicine, College Station, TX 77843, USA
| | - Joseph M Rutkowski
- 1 Division of Lymphatic Biology, Department of Medical Physiology, Texas A&M College of Medicine, College Station, TX 77843, USA
| |
Collapse
|
41
|
Rahimi H, Dieudonne G, Kheyfits V, Bouta EM, Wood RW, Barrett R, Moorehead S, Schwarz EM, Ritchlin CT. Relationship Between Lymph Node Volume and Pain Following Certolizumab Therapy for Rheumatoid Arthritis Flare: A Pilot Study. CLINICAL MEDICINE INSIGHTS-ARTHRITIS AND MUSCULOSKELETAL DISORDERS 2016; 9:203-208. [PMID: 28008295 PMCID: PMC5158121 DOI: 10.4137/cmamd.s40237] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/25/2016] [Revised: 09/25/2016] [Accepted: 09/25/2016] [Indexed: 12/13/2022]
Abstract
OBJECTIVES The mechanisms that trigger flare in rheumatoid arthritis (RA) are unknown. In murine arthritis models, dysfunctional lymph node (LN) drainage is associated with joint flare. To examine if LN alterations are associated with RA flare, we analyzed the change in LN volume via contrast-enhanced magnetic resonance imaging (CE-MRI) in patients with active joint flare at baseline and 16 weeks after certolizumab pegol (CZP) therapy. We also assessed the changes in popliteal or epitrochlear LN volumes versus the Rheumatoid and Arthritis Outcome Score (RAOS) (knee), or the Michigan Hand Questionnaire (MHQ; wrist/hand), and Disease Activity Score 28 (DAS28), at baseline and 16 weeks. RESULTS Total LN volume in 7 of 10 patients with measurable LN on CE-MRI significantly decreased 16 weeks after CZP therapy (mean decrease 37%; P = 0.0019). Improvement in knee pain measured by the RAOS (P = 0.03) inversely correlated with a decrease in total popliteal LN volume (R2 = 0.94). All patients demonstrated significant improvement in DAS28 (mean decrease 1.48; P = 0.0002). For flare in the hand, significant improvement in activities of daily living (ADL) as measured by the MHQ was observed (left hand mean improvement 20%; P = 0.02; right hand mean improvement 37%; P = 0.03). CONCLUSION RA patients with the smallest change in LN volume during anti-tumor necrosis factor (anti-TNF) therapy experienced the greatest pain relief in symptomatic knee joints. Moreover, the remarkably linear inverse correlation between LN volume and joint pain observed in this small clinical pilot provides initial evidence to support the concept that dynamic changes in draining LN volume are a biomarker of clinical response to therapy in RA.
Collapse
Affiliation(s)
- Homaira Rahimi
- Center for Musculoskeletal Research, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA.; Department of Pediatrics, University of Rochester Medical Center, Rochester, NY, USA
| | - Gregory Dieudonne
- Department of Imaging Sciences, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| | - Valeriy Kheyfits
- Department of Imaging Sciences, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| | - Echoe M Bouta
- Center for Musculoskeletal Research, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA.; Department of Biomedical Engineering, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA.; Department of Orthopedics, University of Rochester Medical Center, Rochester, NY, USA
| | - Ronald W Wood
- Department of Obstetrics and Gynecology, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA.; Department of Urology, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| | - Rick Barrett
- Division of Allergy, Immunology, and Rheumatology, University of Rochester Medical Center, Rochester, NY, USA
| | - Sharon Moorehead
- Division of Allergy, Immunology, and Rheumatology, University of Rochester Medical Center, Rochester, NY, USA
| | - Edward M Schwarz
- Center for Musculoskeletal Research, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA.; Department of Biomedical Engineering, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA.; Department of Orthopedics, University of Rochester Medical Center, Rochester, NY, USA.; Department of Urology, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| | - Christopher T Ritchlin
- Center for Musculoskeletal Research, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA.; Division of Allergy, Immunology, and Rheumatology, University of Rochester Medical Center, Rochester, NY, USA
| |
Collapse
|
42
|
Han J, Geng Y, Deng X, Zhang Z. Subclinical Synovitis Assessed by Ultrasound Predicts Flare and Progressive Bone Erosion in Rheumatoid Arthritis Patients with Clinical Remission: A Systematic Review and Metaanalysis. J Rheumatol 2016; 43:2010-2018. [PMID: 27803342 DOI: 10.3899/jrheum.160193] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/30/2016] [Indexed: 01/06/2023]
Abstract
OBJECTIVE Subclinical synovitis can be detected by ultrasound in patients with rheumatoid arthritis (RA) who are in clinical remission. We aimed to confirm its predictive value for flare and progressive bone erosion. METHODS A systematic literature search was performed in Pubmed, Web of Science, Embase, and Cochrane Library on September 7, 2014. Baseline clinical and ultrasonographic characteristics were collected. Methodological quality was assessed. Pooled OR were calculated using Mantel-Haenszel model. We explored the source of heterogeneity through subgroup analysis and completed a cumulative metaanalysis. RESULTS Thirteen articles were included (8 with flare, 4 with bone erosion, 1 with both flare and bone erosion). Metaanalysis revealed an association between power Doppler (PD) positivity and the risk of flare (OR 4.52, 95% CI 2.61-7.84, p < 0.00001, I2 = 21%), the risk of progressive bone erosion on patient level (OR 12.80, 95% CI 1.29-126.81, p = 0.03, I2 = 52%) and the risk of progressive bone erosion on joint level (OR 11.85, 95% CI 5.01-28.03, p < 0.00001, I2 = 0%). Further subgroup analysis showed a higher risk of flare in patients with a study period < 1 year (OR 19.98 vs 3.41). No significant differences were observed in the subgroup analysis in duration of remission, disease duration, and medications. Moreover, cumulative metaanalysis indicated the validation and an increasing accuracy of PD positivity in predicting flare since 2012. CONCLUSION Ultrasound-detected subclinical synovitis can predict the risk of flare and progressive bone erosion in RA patients with clinical remission. Additionally, the flare of RA tends to occur within a followup of 1 year.
Collapse
Affiliation(s)
- Jingjing Han
- From the Department of Geriatrics, and the Department of Rheumatology and Clinical Immunology, Peking University First Hospital, Beijing, China.,J. Han, MD, Department of Geriatrics, Peking University First Hospital; Y. Geng, MD, Department of Rheumatology and Clinical Immunology, Peking University First Hospital; X. Deng, MD, Department of Rheumatology and Clinical Immunology, Peking University First Hospital; Z. Zhang, MD, PhD, Department of Rheumatology and Clinical Immunology, Peking University First Hospital
| | - Yan Geng
- From the Department of Geriatrics, and the Department of Rheumatology and Clinical Immunology, Peking University First Hospital, Beijing, China.,J. Han, MD, Department of Geriatrics, Peking University First Hospital; Y. Geng, MD, Department of Rheumatology and Clinical Immunology, Peking University First Hospital; X. Deng, MD, Department of Rheumatology and Clinical Immunology, Peking University First Hospital; Z. Zhang, MD, PhD, Department of Rheumatology and Clinical Immunology, Peking University First Hospital
| | - Xuerong Deng
- From the Department of Geriatrics, and the Department of Rheumatology and Clinical Immunology, Peking University First Hospital, Beijing, China.,J. Han, MD, Department of Geriatrics, Peking University First Hospital; Y. Geng, MD, Department of Rheumatology and Clinical Immunology, Peking University First Hospital; X. Deng, MD, Department of Rheumatology and Clinical Immunology, Peking University First Hospital; Z. Zhang, MD, PhD, Department of Rheumatology and Clinical Immunology, Peking University First Hospital
| | - Zhuoli Zhang
- From the Department of Geriatrics, and the Department of Rheumatology and Clinical Immunology, Peking University First Hospital, Beijing, China. .,J. Han, MD, Department of Geriatrics, Peking University First Hospital; Y. Geng, MD, Department of Rheumatology and Clinical Immunology, Peking University First Hospital; X. Deng, MD, Department of Rheumatology and Clinical Immunology, Peking University First Hospital; Z. Zhang, MD, PhD, Department of Rheumatology and Clinical Immunology, Peking University First Hospital.
| |
Collapse
|
43
|
Rahimi H, Bell R, Bouta EM, Wood RW, Xing L, Ritchlin CT, Schwarz EM. Lymphatic imaging to assess rheumatoid flare: mechanistic insights and biomarker potential. Arthritis Res Ther 2016; 18:194. [PMID: 27586634 PMCID: PMC5009676 DOI: 10.1186/s13075-016-1092-0] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Proliferation of draining lymphatic vessels coupled with dynamic changes in lymph node volume and flow are characteristic features in rheumatoid arthritis (RA). Furthermore, impaired lymph egress from inflamed synovium is associated with joint flare in murine models of inflammatory-erosive arthritis. Unfortunately, advances towards a greater understanding of lymphatic changes in RA pathogenesis have been slow due to the absence of outcome measures to quantify lymphatic function in vivo. While lymphoscintigraphy is the current standard to assess lymphedema and sentinel lymph nodes in cancer patients, its sensitivity and specificity are inadequate to study lymphatics in RA. The emergence of high-resolution MRI, power Doppler ultrasound, and near-infrared imaging that permits real-time quantification of lymphatic function in animal models has been a major advance, and these techniques have produced a new paradigm of altered lymphatic function that underlies both acute arthritic flare and chronic inflammation. In acute flare, lymphatic drainage increases several fold, whereas no lymphatic contractions are detected in lymph vessels draining chronic arthritic joints. Moreover, these outcomes are now being adapted to study lymphatics in RA towards the development of novel biomarkers of arthritic flare and the discovery of new therapeutic targets. In particular, interventions that directly increase lymphatic egress from diseased joints by opening collateral lymphatic vessels, and that restore lymphatic vessel contractions, provide novel therapeutic approaches with potential for minimal toxicity and immunosuppression. To summarize the origins of this field, recent advances, and future directions, we herein review: current knowledge of lymphatics in RA based on classic literature; new in-vivo imaging modalities that have elucidated how lymphatics modulate acute versus chronic joint inflammation in murine models; and how these preclinical outcome measures are being translated to study lymphatic function in RA inflammation and how effective RA therapies alter lymphatic flow and lymph nodes draining flaring joints. Trial registration: ClinicalTrials.gov NCT02680067. Registered 7 December 2015; ClinicalTrials.gov NCT01098201. Registered 30 March 2010; and ClinicalTrials.gov NCT01083563. Registered 8 March 2010.
Collapse
Affiliation(s)
- Homaira Rahimi
- Center for Musculoskeletal Research, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA. .,Department of Pediatrics, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA. .,University of Rochester Medical Center, 601 Elmwood Avenue, Box 777, Rochester, NY, 14642, USA.
| | - Richard Bell
- Center for Musculoskeletal Research, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA.,Department of Pathology and Laboratory Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| | - Echoe M Bouta
- Center for Musculoskeletal Research, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA.,Department of Biomedical Engineering, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| | - Ronald W Wood
- Department of Obstetrics and Gynecology, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA.,Department of Urology, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA.,Department of Neuroscience, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| | - Lianping Xing
- Center for Musculoskeletal Research, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA.,Department of Pathology and Laboratory Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| | - Christopher T Ritchlin
- Center for Musculoskeletal Research, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA.,Division of Allergy, Immunology, Rheumatology, Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| | - Edward M Schwarz
- Center for Musculoskeletal Research, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA.,Department of Pathology and Laboratory Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA.,Department of Biomedical Engineering, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA.,Department of Urology, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA.,Division of Allergy, Immunology, Rheumatology, Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| |
Collapse
|
44
|
Jordan-Williams KL, Ramanujam N, Farr AG, Ruddell A. The Lymphatic Endothelial mCLCA1 Antibody Induces Proliferation and Growth of Lymph Node Lymphatic Sinuses. PLoS One 2016; 11:e0156079. [PMID: 27224029 PMCID: PMC4880189 DOI: 10.1371/journal.pone.0156079] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2016] [Accepted: 05/09/2016] [Indexed: 12/17/2022] Open
Abstract
Lymphocyte- and leukocyte-mediated lymph node (LN) lymphatic sinus growth (lymphangiogenesis) is involved in immune responses and in diseases including cancer and arthritis. We previously discovered a 10.1.1 Ab that recognizes the lymphatic endothelial cell (LEC) surface protein mCLCA1, which is an interacting partner for LFA1 and Mac-1 that mediates lymphocyte adhesion to LECs. Here, we show that 10.1.1 Ab treatment specifically induces LEC proliferation, and influences migration and adhesion in vitro. Functional testing by injection of mice with 10.1.1 Ab but not control hamster Abs identified rapid induction of LN LEC proliferation and extensive lymphangiogenesis within 23 h. BrdU pulse-chase analysis demonstrated incorporation of proliferating LYVE-1-positive LEC into the growing medullary lymphatic sinuses. The 10.1.1 Ab-induced LN remodeling involved coordinate increases in LECs and also blood endothelial cells, fibroblastic reticular cells, and double negative stroma, as is observed during the LN response to inflammation. 10.1.1 Ab-induced lymphangiogenesis was restricted to LNs, as mCLCA1-expressing lymphatic vessels of the jejunum and dermis were unaffected by 23 h 10.1.1 Ab treatment. These findings demonstrate that 10.1.1 Ab rapidly and specifically induces proliferation and growth of LN lymphatic sinuses and stroma, suggesting a key role of mCLCA1 in coordinating LN remodeling during immune responses.
Collapse
Affiliation(s)
| | - Neela Ramanujam
- Fred Hutchinson Cancer Research Center, Seattle, WA, United States of America
| | - Andrew G Farr
- Department of Biological Structure, University of Washington, Seattle, WA, United States of America
| | - Alanna Ruddell
- Department of Comparative Medicine, University of Washington, Seattle, WA, United States of America.,Fred Hutchinson Cancer Research Center, Seattle, WA, United States of America
| |
Collapse
|
45
|
Kuzin II, Kates SL, Ju Y, Zhang L, Rahimi H, Wojciechowski W, Bernstein SH, Burack R, Schwarz EM, Bottaro A. Increased numbers of CD23(+) CD21(hi) Bin-like B cells in human reactive and rheumatoid arthritis lymph nodes. Eur J Immunol 2016; 46:1752-7. [PMID: 27105894 DOI: 10.1002/eji.201546266] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2015] [Revised: 03/15/2016] [Accepted: 04/19/2016] [Indexed: 12/21/2022]
Abstract
A unique population of CD23(+) CD21(high) B cells in inflamed nodes (Bin) has been shown to accumulate in lymph nodes (LNs) draining inflamed joints of TNF-transgenic (TNF-tg) mice. Bin cells contribute to arthritis flare in mice by distorting node architecture and hampering lymphatic flow, but their existence in human inflamed LNs has not yet been described. Here, we report the characterization of resident B-cell populations in fresh popliteal lymph nodes (PLNs) from patients with severe lower limb diseases (non-RA) and rheumatoid arthritis (RA) patients, and from banked, cryopreserved reactive and normal human LN single cell suspension samples. Bin-like B cells were shown to be significantly increased in reactive LNs, and strikingly elevated (>30% of total) in RA samples. Histopathology and immunofluorescence analyses were consistent with B follicular hyperplasia and histological alterations in RA vs. non-RA PLNs. This is the first description of Bin-like B cells in human inflamed LNs. Consistent with published mouse data, this population appears to be associated with inflammatory arthritis and distortion of LN architecture. Further analyses are necessary to assess the role of CD23(+) CD21(hi) Bin-like B cells in RA pathogenesis and arthritic flare.
Collapse
Affiliation(s)
- Igor I Kuzin
- Department of Biomedical Sciences, Cooper Medical School of Rowan University, Camden, NJ, USA
| | - Stephen L Kates
- Center for Musculoskeletal Research, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA.,Department of Orthopaedics, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| | - Yawen Ju
- Center for Musculoskeletal Research, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA.,Department of Pathology and Laboratory Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| | - Longze Zhang
- Center for Musculoskeletal Research, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA.,Department of Orthopaedics, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| | - Homaira Rahimi
- Center for Musculoskeletal Research, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA.,Department of Pediatrics, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| | - Wojciech Wojciechowski
- Center for Pediatric Biomed Research and Flow Cytometry Shared Resource Laboratory, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| | - Steven H Bernstein
- J.P. Wilmot Cancer Center, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| | - Richard Burack
- Department of Pathology and Laboratory Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA.,J.P. Wilmot Cancer Center, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| | - Edward M Schwarz
- Center for Musculoskeletal Research, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA.,Department of Orthopaedics, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| | - Andrea Bottaro
- Department of Biomedical Sciences, Cooper Medical School of Rowan University, Camden, NJ, USA
| |
Collapse
|
46
|
Liang Q, Ju Y, Chen Y, Wang W, Li J, Zhang L, Xu H, Wood RW, Schwarz EM, Boyce BF, Wang Y, Xing L. Lymphatic endothelial cells efferent to inflamed joints produce iNOS and inhibit lymphatic vessel contraction and drainage in TNF-induced arthritis in mice. Arthritis Res Ther 2016; 18:62. [PMID: 26970913 PMCID: PMC4789262 DOI: 10.1186/s13075-016-0963-8] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2015] [Accepted: 02/26/2016] [Indexed: 12/11/2022] Open
Abstract
Background In this study, we sought to determine the cellular source of inducible nitric oxide synthase (iNOS) induced in lymphatic endothelial cells (LECs) in response to tumor necrosis factor (TNF), the effects of iNOS on lymphatic smooth muscle cell (LSMC) function and on the development of arthritis in TNF-transgenic (TNF-Tg) mice, and whether iNOS inhibitors improve lymphatic function and reduce joint destruction in inflammatory erosive arthritis. Methods We used quantitative polymerase chain reactions, immunohistochemistry, histology, and near-infrared imaging to examine (1) iNOS expression in podoplanin + LECs and lymphatic vessels from wild-type (WT) and TNF-Tg mice, (2) iNOS induction by TNF in WT LECs, (3) the effects of iNOS inhibitors on expression of functional muscle genes in LSMCs, and (4) the effects of iNOS inhibitors on lymphatic vessel contraction and drainage, as well as the severity of arthritis, in TNF-Tg mice. Results LECs from TNF-Tg mice had eight fold higher iNOS messenger RNA levels than WT cells, and iNOS expression was confirmed immunohistochemically in podoplanin + LECs in lymphatic vessels from inflamed joints. TNF (0.1 ng/ml) increased iNOS levels 40-fold in LECs. LSMCs cocultured with LECs pretreated with TNF had reduced expression of functional muscle genes. This reduction was prevented by ferulic acid, which blocked nitric oxide production. Local injection of L-N6-(1-iminoethyl)lysine 5-tetrazole-amide into inflamed paws of TNF-Tg mice resulted in recovery of lymphatic vessel contractions and drainage. Treatment of TNF-Tg mice with ferulic acid reduced synovial inflammation as well as cartilage and bone erosion, and it also restored lymphatic contraction and drainage. Conclusions iNOS is produced primarily by LECs in lymphatic vessel efferent from inflamed joints of TNF-Tg mice in response to TNF and inhibits LSMC contraction and lymph drainage. Ferulic acid represents a potential new therapy to restore lymphatic function and thus improve inflammatory arthritis by inhibiting local production of nitric oxide by LSMCs. Electronic supplementary material The online version of this article (doi:10.1186/s13075-016-0963-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Qianqian Liang
- Department of Orthopaedics, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, 725 South Wanping Road, Shanghai, 200032, China.,Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, 601 Elmwood Avenue, Rochester, NY, 14642, USA
| | - Yawen Ju
- Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, 601 Elmwood Avenue, Rochester, NY, 14642, USA.,Center for Musculoskeletal Research, University of Rochester Medical Center, 601 Elmwood Avenue, Rochester, NY, 14642, USA
| | - Yan Chen
- Department of Orthopaedics, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, 725 South Wanping Road, Shanghai, 200032, China
| | - Wensheng Wang
- Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, 601 Elmwood Avenue, Rochester, NY, 14642, USA
| | - Jinlong Li
- Department of Orthopaedics, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, 725 South Wanping Road, Shanghai, 200032, China
| | - Li Zhang
- Department of Orthopaedics, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, 725 South Wanping Road, Shanghai, 200032, China
| | - Hao Xu
- Department of Orthopaedics, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, 725 South Wanping Road, Shanghai, 200032, China.,Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, 601 Elmwood Avenue, Rochester, NY, 14642, USA
| | - Ronald W Wood
- Departments of Obstetrics and Gynecology, University of Rochester Medical Center, 601 Elmwood Avenue, Rochester, NY, 14642, USA
| | - Edward M Schwarz
- Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, 601 Elmwood Avenue, Rochester, NY, 14642, USA.,Center for Musculoskeletal Research, University of Rochester Medical Center, 601 Elmwood Avenue, Rochester, NY, 14642, USA
| | - Brendan F Boyce
- Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, 601 Elmwood Avenue, Rochester, NY, 14642, USA.,Center for Musculoskeletal Research, University of Rochester Medical Center, 601 Elmwood Avenue, Rochester, NY, 14642, USA
| | - Yongjun Wang
- Department of Orthopaedics, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, 725 South Wanping Road, Shanghai, 200032, China. .,Institute of Spine, Shanghai University of Traditional Chinese Medicine, 725 Wan-Ping South Road, Shanghai, 200032, China.
| | - Lianping Xing
- Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, 601 Elmwood Avenue, Rochester, NY, 14642, USA. .,Center for Musculoskeletal Research, University of Rochester Medical Center, 601 Elmwood Avenue, Rochester, NY, 14642, USA. .,Departments of Obstetrics and Gynecology, University of Rochester Medical Center, 601 Elmwood Avenue, Rochester, NY, 14642, USA.
| |
Collapse
|
47
|
Padera TP, Meijer EFJ, Munn LL. The Lymphatic System in Disease Processes and Cancer Progression. Annu Rev Biomed Eng 2016; 18:125-58. [PMID: 26863922 DOI: 10.1146/annurev-bioeng-112315-031200] [Citation(s) in RCA: 147] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Advances in our understanding of the structure and function of the lymphatic system have made it possible to identify its role in a variety of disease processes. Because it is involved not only in fluid homeostasis but also in immune cell trafficking, the lymphatic system can mediate and ultimately alter immune responses. Our rapidly increasing knowledge of the molecular control of the lymphatic system will inevitably lead to new and effective therapies for patients with lymphatic dysfunction. In this review, we discuss the molecular and physiological control of lymphatic vessel function and explore how the lymphatic system contributes to many disease processes, including cancer and lymphedema.
Collapse
Affiliation(s)
- Timothy P Padera
- Edwin L. Steele Laboratories, Department of Radiation Oncology, and Cancer Center, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts 02114;
| | - Eelco F J Meijer
- Edwin L. Steele Laboratories, Department of Radiation Oncology, and Cancer Center, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts 02114;
| | - Lance L Munn
- Edwin L. Steele Laboratories, Department of Radiation Oncology, and Cancer Center, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts 02114;
| |
Collapse
|
48
|
Kuzin II, Bouta EM, Schwarz EM, Bottaro A. TNF signals are dispensable for the generation of CD23+ CD21/35-high CD1d-high B cells in inflamed lymph nodes. Cell Immunol 2015; 296:133-7. [PMID: 25959608 DOI: 10.1016/j.cellimm.2015.04.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2015] [Revised: 04/06/2015] [Accepted: 04/15/2015] [Indexed: 12/16/2022]
Abstract
Tumor necrosis factor (TNF) is a key cytokine in rheumatoid arthritis (RA) pathogenesis, as underscored by the clinical effectiveness of TNF antagonists. While several of TNF's key targets in RA are well understood, its many pleiotropic effects remain to be elucidated. TNF-transgenic mice develop inflammatory-erosive arthritis associated with disruption of draining lymph node histology and function, and accumulation of B cells with unique phenotypic and functional features consistent with contribution to pathogenesis (B cells in inflamed nodes, Bin). Bin cell induction depends on the inflamed microenvironment, but the specific signals are unknown. Using anti-TNF treatment and TNF-receptor-deficient mice, here we show that Bin cells are induced and maintained independently of B cell-intrinsic TNF signals.
Collapse
Affiliation(s)
- Igor I Kuzin
- Dept. of Biomedical Sciences, Cooper Medical School of Rowan University, Camden, NJ, USA
| | - Echoe M Bouta
- Center for Musculoskeletal Research, Dept. of Biomedical Engineering, and Dept. of Orthopaedics, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| | - Edward M Schwarz
- Center for Musculoskeletal Research, Dept. of Biomedical Engineering, and Dept. of Orthopaedics, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| | - Andrea Bottaro
- Dept. of Biomedical Sciences, Cooper Medical School of Rowan University, Camden, NJ, USA.
| |
Collapse
|
49
|
Padera TP. Editorial: rapid growth in the field of lymphatic biology. Semin Cell Dev Biol 2015; 38:53-4. [PMID: 25868083 DOI: 10.1016/j.semcdb.2015.03.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Affiliation(s)
- Timothy P Padera
- E.L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital Cancer Center and Harvard Medical School, Boston, MA 02114, USA
| |
Collapse
|