1
|
Zhong X, Patel A, Sun Y, Saeboe AM, Dennis AM. Multiplexed Shortwave Infrared Imaging Highlights Anatomical Structures in Mice. Angew Chem Int Ed Engl 2024; 63:e202410936. [PMID: 39014295 PMCID: PMC11473221 DOI: 10.1002/anie.202410936] [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: 06/10/2024] [Revised: 07/12/2024] [Accepted: 07/14/2024] [Indexed: 07/18/2024]
Abstract
Multiplexed fluorescence in vivo imaging remains challenging due to the attenuation and scattering of visible and traditional near infrared (NIR-I, 650-950 nm) wavelengths. Fluorescence imaging using shortwave infrared (SWIR, 1000-1700 nm, a.k.a. NIR-II) light enables deeper tissue penetration due to reduced tissue scattering as well as minimal background autofluorescence. SWIR-emitting semiconductor quantum dots (QDs) with tunable emission peaks and optical stability are powerful contrast agents, yet few imaging demonstrations exclusively use SWIR emission beyond two-color imaging schemes. In this study, we engineered three high quality lead sulfide/cadmium sulfide (PbS/CdS) core/shell QDs with distinct SWIR emission peaks ranging from 1100-1550 nm for simultaneous three-color imaging in mice. We first use the exceptional photostability of QDs to non-invasively track lymphatic drainage with longitudinal imaging, highlighting the detailed networks of lymphatic vessels with widefield imaging over a 2 hr period. We then perform multiplexed imaging with all three QDs to distinctly visualize the lymphatic system and spatially overlapping vasculature networks, including clearly distinguishing the liver and spleen. This work establishes optimized SWIR QDs for next generation multiplexed and longitudinal preclinical imaging, unlocking numerous opportunities for preclinical studies of disease progression, drug biodistribution, and cell trafficking dynamics in living organisms.
Collapse
Affiliation(s)
- Xingjian Zhong
- Department of Chemical Engineering, Northeastern University, Boston, MA, USA
- Department of Biomedical Engineering, Boston University, Boston, MA, USA
| | - Amish Patel
- Department of Chemical Engineering, Northeastern University, Boston, MA, USA
- Department of Biomedical Engineering, Boston University, Boston, MA, USA
| | - Yidan Sun
- Department of Chemical Engineering, Northeastern University, Boston, MA, USA
| | - Alexander M Saeboe
- Division of Material Science & Engineering, Boston University, Boston, MA, USA
| | - Allison M Dennis
- Department of Chemical Engineering, Northeastern University, Boston, MA, USA
- Department of Bioengineering, Northeastern University, Boston, MA, USA
| |
Collapse
|
2
|
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
|
3
|
Hansen C, Jagtap J, Parchur A, Sharma G, Shafiee S, Sinha S, Himburg H, Joshi A. Dynamic multispectral NIR/SWIR for in vivo lymphovascular architectural and functional quantification. JOURNAL OF BIOMEDICAL OPTICS 2024; 29:106001. [PMID: 39347012 PMCID: PMC11425400 DOI: 10.1117/1.jbo.29.10.106001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Revised: 08/30/2024] [Accepted: 08/30/2024] [Indexed: 10/01/2024]
Abstract
Significance Although the lymphatic system is the second largest circulatory system in the body, there are limited techniques available for characterizing lymphatic vessel function. We report shortwave-infrared (SWIR) imaging for minimally invasive in vivo quantification of lymphatic circulation with superior contrast and resolution compared with near-infrared first window imaging. Aim We aim to study the lymphatic structure and function in vivo via SWIR fluorescence imaging. Approach We evaluated subsurface lymphatic circulation in healthy, adult immunocompromised salt-sensitive Sprague-Dawley rats using two fluorescence imaging modalities: near-infrared first window (NIR-I, 700 to 900 nm) and SWIR (900 to 1800 nm) imaging. We also compared two fluorescent imaging probes: indocyanine green (ICG) and silver sulfide quantum dots (QDs) as SWIR lymphatic contrast agents following intradermal footpad delivery in these rats. Results SWIR imaging exhibits reduced scattering and autofluorescence background relative to NIR-I imaging. SWIR imaging with ICG provides 1.7 times better resolution and sensitivity than NIR-I, and SWIR imaging with QDs provides nearly two times better resolution and sensitivity with enhanced vessel distinguishability. SWIR images thus provide a more accurate estimation of in vivo vessel size than conventional NIR-I images. Conclusions SWIR imaging of silver sulfide QDs into the intradermal footpad injection provides superior image resolution compared with conventional imaging techniques using NIR-I imaging with ICG dye.
Collapse
Affiliation(s)
- Christopher Hansen
- Medical College of Wisconsin, Department of Biomedical Engineering, Milwaukee, Wisconsin, United States
| | - Jaidip Jagtap
- Mayo Clinic, Department of Radiology, Rochester, Minnesota, United States
| | - Abdul Parchur
- Medical College of Wisconsin, Department of Radiation Oncology, Milwaukee, Wisconsin, United States
| | - Gayatri Sharma
- Amity University, Amity Institute of Biotechnology, Centre for Medical Biotechnology, Noida, Uttar Pradesh, India
| | - Shayan Shafiee
- Medical College of Wisconsin, Department of Biomedical Engineering, Milwaukee, Wisconsin, United States
| | - Sayantan Sinha
- Medical College of Wisconsin, Department of Biomedical Engineering, Milwaukee, Wisconsin, United States
| | - Heather Himburg
- Medical College of Wisconsin, Department of Biomedical Engineering, Milwaukee, Wisconsin, United States
- Medical College of Wisconsin, Department of Radiation Oncology, Milwaukee, Wisconsin, United States
| | - Amit Joshi
- Medical College of Wisconsin, Department of Biomedical Engineering, Milwaukee, Wisconsin, United States
| |
Collapse
|
4
|
Tay RE, P L, Pang ST, Low KE, Tay HC, Ho CM, Malleret B, Rötzschke O, Olivo M, Tay ZW. High-efficiency magnetophoretic labelling of adoptively-transferred T cells for longitudinal in vivo Magnetic Particle Imaging. Theranostics 2024; 14:6138-6160. [PMID: 39431019 PMCID: PMC11488102 DOI: 10.7150/thno.95527] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Accepted: 08/07/2024] [Indexed: 10/22/2024] Open
Abstract
While adoptive cell therapies (ACT) have been successful as therapies for blood cancers, they have limited efficacy in treating solid tumours, where the tumour microenvironment excludes and suppresses adoptively transferred tumour-specific immune cells. A major obstacle to improving cell therapies for solid tumours is a lack of accessible and quantitative imaging modalities capable of tracking the migration and immune functional activity of ACT products for an extended duration in vivo. Methods: A high-efficiency magnetophoretic method was developed for facile magnetic labelling of hard-to-label immune cells, which were then injected into tumour-bearing mice and imaged over two weeks with a compact benchtop Magnetic Particle Imager (MPI) design. Results: Labelling efficiency was improved more than 10-fold over prior studies enabling longer-term tracking for at least two weeks in vivo of the labelled immune cells and their biodistribution relative to the tumour. The new imager showed 5-fold improved throughput enabling much larger density of data (up to 20 mice per experiment). Conclusions: Taken together, our innovations enable the convenient and practical use of MPI to visualise the localisation of ACT products in in vivo preclinical models for longitudinal, non-invasive functional evaluation of therapeutic efficacy.
Collapse
Affiliation(s)
- Rong En Tay
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), 8A Biomedical Grove, #04-06 Immunos, Singapore 138648, Republic of Singapore
| | - Lokamitra P
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), 8A Biomedical Grove, #04-06 Immunos, Singapore 138648, Republic of Singapore
- Institute of Bioengineering and Bioimaging (IBB), Agency for Science, Technology and Research (A*STAR), 11 Biopolis Way, #02-02 Helios Building, Singapore 138667, Republic of Singapore
| | - Shun Toll Pang
- Institute of Bioengineering and Bioimaging (IBB), Agency for Science, Technology and Research (A*STAR), 11 Biopolis Way, #02-02 Helios Building, Singapore 138667, Republic of Singapore
| | - Kay En Low
- Electron Microscopy Unit, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Republic of Singapore
| | - Hui Chien Tay
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), 8A Biomedical Grove, #04-06 Immunos, Singapore 138648, Republic of Singapore
| | - Charmaine Min Ho
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), 8A Biomedical Grove, #04-06 Immunos, Singapore 138648, Republic of Singapore
| | - Benoit Malleret
- Electron Microscopy Unit, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Republic of Singapore
- Department of Microbiology and Immunology, Immunology Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Republic of Singapore
| | - Olaf Rötzschke
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), 8A Biomedical Grove, #04-06 Immunos, Singapore 138648, Republic of Singapore
- Department of Microbiology and Immunology, Immunology Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Republic of Singapore
- School of Biological Sciences, Nanyang Technological University, 50 Nanyang Avenue, Singapore 639798, Republic of Singapore
| | - Malini Olivo
- A*STAR Skin Research Labs (A*SRL), Agency for Science, Technology and Research (A*STAR), 31 Biopolis Way, #07-01 Nanos, Singapore 138669, Republic of Singapore
| | - Zhi Wei Tay
- Institute of Bioengineering and Bioimaging (IBB), Agency for Science, Technology and Research (A*STAR), 11 Biopolis Way, #02-02 Helios Building, Singapore 138667, Republic of Singapore
- National Institute of Advanced Industrial Science and Technology (AIST), Health and Medical Research Institute (HMRI), 1-2-1 Namiki, Tsukuba, Ibaraki 305-8564, Japan
| |
Collapse
|
5
|
Choi Y, Saron WA, O'Neill A, Senanayake M, Wilder-Smith A, Rathore AP, St John AL. NKT cells promote Th1 immune bias to dengue virus that governs long-term protective antibody dynamics. J Clin Invest 2024; 134:e169251. [PMID: 39088280 PMCID: PMC11405039 DOI: 10.1172/jci169251] [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: 01/31/2023] [Accepted: 07/23/2024] [Indexed: 08/03/2024] Open
Abstract
NKT cells are innate-like T cells, recruited to the skin during viral infection, yet their contributions to long-term immune memory to viruses are unclear. We identified granzyme K, a product made by cytotoxic cells including NKT cells, as linked to induction of Th1-associated antibodies during primary dengue virus (DENV) infection in humans. We examined the role of NKT cells in vivo using DENV-infected mice lacking CD1d-dependent (CD1ddep) NKT cells. In CD1d-KO mice, Th1-polarized immunity and infection resolution were impaired, which was dependent on intrinsic NKT cell production of IFN-γ, since it was restored by adoptive transfer of WT but not IFN-γ-KO NKT cells. Furthermore, NKT cell deficiency triggered immune bias, resulting in higher levels of Th2-associated IgG1 than Th1-associated IgG2a, which failed to protect against a homologous DENV rechallenge and promoted antibody-dependent enhanced disease during secondary heterologous infections. Similarly, Th2 immunity, typified by a higher IgG4/IgG3 ratio, was associated with worsened human disease severity during secondary infections. Thus, CD1ddep NKT cells establish Th1 polarity during the early innate response to DENV, which promotes infection resolution, memory formation, and long-term protection from secondary homologous and heterologous infections in mice, with consistent associations observed in humans. These observations illustrate how early innate immune responses during primary infections can influence secondary infection outcomes.
Collapse
Affiliation(s)
- Youngjoo Choi
- Programme in Emerging Infectious Diseases, Duke-National University of Singapore Medical School, Singapore
| | - Wilfried Aa Saron
- Programme in Emerging Infectious Diseases, Duke-National University of Singapore Medical School, Singapore
| | - Aled O'Neill
- Programme in Emerging Infectious Diseases, Duke-National University of Singapore Medical School, Singapore
| | - Manouri Senanayake
- Department of Paediatrics, Faculty of Medicine, University of Colombo, Colombo, Sri Lanka
- Lady Ridgeway Children's Hospital, Colombo, Sri Lanka
| | - Annelies Wilder-Smith
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore
- Department of Disease Control, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Abhay Ps Rathore
- Programme in Emerging Infectious Diseases, Duke-National University of Singapore Medical School, Singapore
- Department of Pathology, Duke University Medical Center, Durham, North Carolina, USA
| | - Ashley L St John
- Programme in Emerging Infectious Diseases, Duke-National University of Singapore Medical School, Singapore
- Department of Pathology, Duke University Medical Center, Durham, North Carolina, USA
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- SingHealth Duke-NUS Global Health Institute, Singapore
| |
Collapse
|
6
|
Brook B, Duval V, Barman S, Speciner L, Sweitzer C, Khanmohammed A, Menon M, Foster K, Ghosh P, Abedi K, Koster J, Nanishi E, Baden LR, Levy O, VanCott T, Micol R, Dowling DJ. Adjuvantation of a SARS-CoV-2 mRNA vaccine with controlled tissue-specific expression of an mRNA encoding IL-12p70. Sci Transl Med 2024; 16:eadm8451. [PMID: 39047117 DOI: 10.1126/scitranslmed.adm8451] [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: 11/08/2023] [Revised: 03/18/2024] [Accepted: 06/24/2024] [Indexed: 07/27/2024]
Abstract
Messenger RNA (mRNA) vaccines were pivotal in reducing severe acute respiratory syndrome 2 (SARS-CoV-2) infection burden, yet they have not demonstrated robust durability, especially in older adults. Here, we describe a molecular adjuvant comprising a lipid nanoparticle (LNP)-encapsulated mRNA encoding interleukin-12p70 (IL-12p70). The bioactive adjuvant was engineered with a multiorgan protection (MOP) sequence to restrict transcript expression to the intramuscular injection site. Admixing IL-12-MOP (CTX-1796) with the BNT162b2 SARS-CoV-2 vaccine increased spike protein-specific immune responses in mice. Specifically, the benefits of IL-12-MOP adjuvantation included amplified humoral and cellular immunity and increased immune durability for 1 year after vaccination in mice. An additional benefit included the restoration of immunity in aged mice to amounts comparable to those achieved in young adult animals, alongside amplification with a single immunization. Associated enhanced dendritic cell and germinal center responses were observed. Together, these data demonstrate that an LNP-encapsulated IL-12-MOP mRNA-encoded adjuvant can amplify immunogenicity independent of age, demonstrating translational potential to benefit vulnerable populations.
Collapse
Affiliation(s)
- Byron Brook
- Precision Vaccines Program, Department of Pediatrics, Boston Children's Hospital, Boston, MA 02115, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA
| | - Valerie Duval
- Combined Therapeutics Incorporated, Boston, MA 02135, USA
| | - Soumik Barman
- Precision Vaccines Program, Department of Pediatrics, Boston Children's Hospital, Boston, MA 02115, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA
| | | | - Cali Sweitzer
- Precision Vaccines Program, Department of Pediatrics, Boston Children's Hospital, Boston, MA 02115, USA
| | | | - Manisha Menon
- Precision Vaccines Program, Department of Pediatrics, Boston Children's Hospital, Boston, MA 02115, USA
| | | | - Pallab Ghosh
- Combined Therapeutics Incorporated, Boston, MA 02135, USA
| | - Kimia Abedi
- Precision Vaccines Program, Department of Pediatrics, Boston Children's Hospital, Boston, MA 02115, USA
| | - Jacob Koster
- Precision Vaccines Program, Department of Pediatrics, Boston Children's Hospital, Boston, MA 02115, USA
| | - Etsuro Nanishi
- Precision Vaccines Program, Department of Pediatrics, Boston Children's Hospital, Boston, MA 02115, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA
| | - Lindsey R Baden
- Department of Medicine, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Ofer Levy
- Precision Vaccines Program, Department of Pediatrics, Boston Children's Hospital, Boston, MA 02115, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Thomas VanCott
- Combined Therapeutics Incorporated, Boston, MA 02135, USA
| | - Romain Micol
- Combined Therapeutics Incorporated, Boston, MA 02135, USA
| | - David J Dowling
- Precision Vaccines Program, Department of Pediatrics, Boston Children's Hospital, Boston, MA 02115, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
7
|
Andriana P, Fair-Mäkelä R, Liljenbäck H, Kärnä S, Iqbal I, Makrypidi K, Rajander J, Pirmettis I, Li XG, Jalkanen S, Saraste A, Salmi M, Roivainen A. Macrophage mannose receptor CD206 targeting of fluoride-18 labeled mannosylated dextran: A validation study in mice. Eur J Nucl Med Mol Imaging 2024; 51:2216-2228. [PMID: 38532026 PMCID: PMC11178572 DOI: 10.1007/s00259-024-06686-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Accepted: 03/10/2024] [Indexed: 03/28/2024]
Abstract
PURPOSE Aluminum fluoride-18-labeled 1,4,7-triazacyclononane-1,4,7-triacetic acid-conjugated mannosylated dextran derivative (Al[18F]F-NOTA-D10CM) is a new tracer for PET imaging. We report here on in vitro and in vivo validation of the tracer's ability to target the macrophage mannose receptor CD206. METHODS First, the uptake of intravenously (i.v.) administered Al[18F]F-NOTA-D10CM was compared between wild-type (WT) and CD206-/- knockout (KO) mice. C57BL/6N mice were injected with complete Freund's adjuvant (CFA) in the left hind leg and the uptake of Al[18F]F-NOTA-D10CM after i.v. or intradermal (i.d.) injection was studied at 5 and 14 days after CFA induction of inflammation. Healthy C57BL/6N mice were studied as controls. Mice underwent PET/CT on consecutive days with [18F]FDG, i.v. Al[18F]F-NOTA-D10CM, and i.d. Al[18F]F-NOTA-D10CM. After the last imaging, Al[18F]F-NOTA-D10CM was i.v. injected for an ex vivo biodistribution study and autoradiography of inflamed tissues. Blood plasma samples were analyzed using high-performance liquid chromatography. To evaluate the specificity of Al[18F]F-NOTA-D10CM binding, an in vitro competitive displacement study was performed on inflamed tissue sections using autoradiography. CD206 expression was assessed by immunohistochemical staining. RESULTS Compared with WT mice, the uptake of Al[18F]F-NOTA-D10CM was significantly lower in several CD206-/- KO mice tissues, including liver (SUV 8.21 ± 2.51 vs. 1.06 ± 0.16, P < 0.001) and bone marrow (SUV 1.63 ± 0.37 vs. 0.22 ± 0.05, P < 0.0001). The uptake of i.v. injected Al[18F]F-NOTA-D10CM was significantly higher in inflamed ankle joint (SUV 0.48 ± 0.13 vs. 0.18 ± 0.05, P < 0.0001) and inflamed foot pad skin (SUV 0.41 ± 0.10 vs. 0.04 ± 0.01, P < 0.0001) than in the corresponding tissues in healthy mice. The i.d.-injected Al[18F]F-NOTA-D10CM revealed differences between CFA-induced lymph node activation and lymph nodes in healthy mice. Ex vivo γ-counting, autoradiography, and immunohistochemistry supported the results, and a decrease of ~ 80% in the binding of Al[18F]F-NOTA-D10CM in the displacement study with excess NOTA-D10CM confirmed that tracer binding was specific. At 60 min after i.v. injection, an average 96.70% of plasma radioactivity was derived from intact Al[18F]F-NOTA-D10CM, indicating good in vivo stability. The uptake of Al[18F]F-NOTA-D10CM into inflamed tissues was positively associated with the area percentage of CD206-positive staining. CONCLUSION The uptake of mannosylated dextran derivative Al[18F]F-NOTA-D10CM correlated with CD206 expression and the tracer appears promising for inflammation imaging.
Collapse
Affiliation(s)
- Putri Andriana
- Turku PET Centre, University of Turku, Kiinamyllynkatu 4-8, 20520, Turku, Finland
| | - Ruth Fair-Mäkelä
- Institute of Biomedicine, University of Turku, Turku, Finland
- InFLAMES Research Flagship Center, University of Turku, Turku, Finland
| | - Heidi Liljenbäck
- Turku PET Centre, University of Turku, Kiinamyllynkatu 4-8, 20520, Turku, Finland
- Turku Center of Disease Modeling, University of Turku, Turku, Finland
| | - Salli Kärnä
- Turku PET Centre, University of Turku, Kiinamyllynkatu 4-8, 20520, Turku, Finland
| | - Imran Iqbal
- Turku PET Centre, University of Turku, Kiinamyllynkatu 4-8, 20520, Turku, Finland
| | - Konstantina Makrypidi
- Institute of Nuclear and Radiological Science and Technology, Energy and Safety, NCSR "Demokritos", Athens, Greece
| | - Johan Rajander
- Turku PET Centre, Accelerator Laboratory, Åbo Akademi University, Turku, Finland
| | - Ioannis Pirmettis
- Institute of Nuclear and Radiological Science and Technology, Energy and Safety, NCSR "Demokritos", Athens, Greece
| | - Xiang-Guo Li
- Turku PET Centre, University of Turku, Kiinamyllynkatu 4-8, 20520, Turku, Finland
- InFLAMES Research Flagship Center, University of Turku, Turku, Finland
- Department of Chemistry, University of Turku, Turku, Finland
| | - Sirpa Jalkanen
- InFLAMES Research Flagship Center, University of Turku, Turku, Finland
- MediCity Research Laboratory, University of Turku, Turku, Finland
| | - Antti Saraste
- Turku PET Centre, University of Turku, Kiinamyllynkatu 4-8, 20520, Turku, Finland
- Turku PET Centre, Turku University Hospital, Turku, Finland
- Heart Center, Turku University Hospital and University of Turku, Turku, Finland
| | - Marko Salmi
- Institute of Biomedicine, University of Turku, Turku, Finland
- InFLAMES Research Flagship Center, University of Turku, Turku, Finland
- MediCity Research Laboratory, University of Turku, Turku, Finland
| | - Anne Roivainen
- Turku PET Centre, University of Turku, Kiinamyllynkatu 4-8, 20520, Turku, Finland.
- InFLAMES Research Flagship Center, University of Turku, Turku, Finland.
- Turku Center of Disease Modeling, University of Turku, Turku, Finland.
- Turku PET Centre, Turku University Hospital, Turku, Finland.
| |
Collapse
|
8
|
Nakkala JR, Li Y, Akter L, Kang X, Chen X. Differential Regulation of DC Function, Adaptive Immunity, and MyD88 Dependence by MF59 and AS03-like Adjuvants. Vaccines (Basel) 2024; 12:531. [PMID: 38793782 PMCID: PMC11125884 DOI: 10.3390/vaccines12050531] [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: 04/01/2024] [Revised: 04/26/2024] [Accepted: 05/10/2024] [Indexed: 05/26/2024] Open
Abstract
MF59 and AS03 are squalene emulsion-based vaccine adjuvants with similar compositions and droplet sizes. Despite their broad use in licensed influenza vaccines, few studies compared their adjuvant effects and action mechanisms side by side. Considering the majority of adjuvants act on dendritic cells (DCs) to achieve their adjuvant effects, this study compared MF59 and AS03-like adjuvants (AddaVax and AddaS03, respectively) to enhance antigen uptake, DC maturation, ovalbumin (OVA) and seasonal influenza vaccine-induced immune responses. Considering MF59 was reported to activate MyD88 to mediate its adjuvant effects, this study also investigated whether the above-explored adjuvant effects of AddaVax and AddaS03 depended on MyD88. We found AddaVax more potently enhanced antigen uptake at the local injection site, while AddaS03 more potently enhanced antigen uptake in the draining lymph nodes. AddaS03 but not AddaVax stimulated DC maturation. Adjuvant-enhanced antigen uptake was MyD88 independent, while AddaS03-induced DC maturation was MyD88 dependent. AddaVax and AddaS03 similarly enhanced OVA-induced IgG and subtype IgG1 antibody responses as well as influenza vaccine-induced hemagglutination inhibition antibody titers, whileAddaS03 more potently enhanced OVA-specific IgG2c antibody responses. Both adjuvants depended on MyD88 to enhance vaccine-induced antibody responses, while AddaVax depended more on MyD88 to achieve its adjuvant effects. Our study reveals similarities and differences of the two squalene emulsion-based vaccine adjuvants, contributing to our improved understanding of their action mechanisms.
Collapse
Affiliation(s)
| | | | | | | | - Xinyuan Chen
- Biomedical & Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, 7 Greenhouse Road, Avedisian Hall, Room 480, Kingston, RI 02881, USA; (J.R.N.); (Y.L.); (L.A.); (X.K.)
| |
Collapse
|
9
|
Holloway AJ, Saito TB, Naqvi KF, Huante MB, Fan X, Lisinicchia JG, Gelman BB, Endsley JJ, Endsley MA. Inhibition of caspase pathways limits CD4 + T cell loss and restores host anti-retroviral function in HIV-1 infected humanized mice with augmented lymphoid tissue. Retrovirology 2024; 21:8. [PMID: 38693565 PMCID: PMC11064318 DOI: 10.1186/s12977-024-00641-2] [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: 03/06/2023] [Accepted: 04/05/2024] [Indexed: 05/03/2024] Open
Abstract
The study of HIV infection and pathogenicity in physical reservoirs requires a biologically relevant model. The human immune system (HIS) mouse is an established model of HIV infection, but defects in immune tissue reconstitution remain a challenge for examining pathology in tissues. We utilized exogenous injection of the human recombinant FMS-like tyrosine kinase 3 ligand (rFLT-3 L) into the hematopoietic stem cell (HSC) cord blood HIS mouse model to significantly expand the total area of lymph node (LN) and the number of circulating human T cells. The results enabled visualization and quantification of HIV infectivity, CD4 T cell depletion and other measures of pathogenesis in the secondary lymphoid tissues of the spleen and LN. Treatment with the Caspase-1/4 inhibitor VX-765 limited CD4+ T cell loss in the spleen and reduced viral load in both the spleen and axillary LN. In situ hybridization further demonstrated a decrease in viral RNA in both the spleen and LN. Transcriptomic analysis revealed that in vivo inhibition of caspase-1/4 led to an upregulation in host HIV restriction factors including SAMHD1 and APOBEC3A. These findings highlight the use of rFLT-3 L to augment human immune system characteristics in HIS mice to support investigations of HIV pathogenesis and test host directed therapies, though further refinements are needed to further augment LN architecture and cellular populations. The results further provide in vivo evidence of the potential to target inflammasome pathways as an avenue of host-directed therapy to limit immune dysfunction and virus replication in tissue compartments of HIV+ persons.
Collapse
Affiliation(s)
- Alex J Holloway
- Department of Microbiology and Immunology, University of Texas Medical Branch, 77555, Galveston, TX, USA
| | - Tais B Saito
- Department of Pathology, University of Texas Medical Branch, 77555, Galveston, TX, USA
- Current at the Laboratory of Bacteriology, Rocky Mountain Laboratories, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 59840, Hamilton, MT, USA
| | - Kubra F Naqvi
- Department of Microbiology and Immunology, University of Texas Medical Branch, 77555, Galveston, TX, USA
- Department of Internal Medicine, University of Texas Southwestern Medical Center, 75390, Dallas, TX, USA
| | - Matthew B Huante
- Department of Microbiology and Immunology, University of Texas Medical Branch, 77555, Galveston, TX, USA
| | - Xiuzhen Fan
- Department of Microbiology and Immunology, University of Texas Medical Branch, 77555, Galveston, TX, USA
- Department of Medicine, University of Toledo, 43614, Toledo, OH, USA
| | - Joshua G Lisinicchia
- Department of Pathology, University of Texas Medical Branch, 77555, Galveston, TX, USA
| | - Benjamin B Gelman
- Department of Pathology, University of Texas Medical Branch, 77555, Galveston, TX, USA
| | - Janice J Endsley
- Department of Microbiology and Immunology, University of Texas Medical Branch, 77555, Galveston, TX, USA
| | - Mark A Endsley
- Department of Microbiology and Immunology, University of Texas Medical Branch, 77555, Galveston, TX, USA.
| |
Collapse
|
10
|
Mondal DK, Xie C, Pascal GJ, Buraschi S, Iozzo RV. Decorin suppresses tumor lymphangiogenesis: A mechanism to curtail cancer progression. Proc Natl Acad Sci U S A 2024; 121:e2317760121. [PMID: 38652741 PMCID: PMC11067011 DOI: 10.1073/pnas.2317760121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Accepted: 03/25/2024] [Indexed: 04/25/2024] Open
Abstract
The complex interplay between malignant cells and the cellular and molecular components of the tumor stroma is a key aspect of cancer growth and development. These tumor-host interactions are often affected by soluble bioactive molecules such as proteoglycans. Decorin, an archetypical small leucine-rich proteoglycan primarily expressed by stromal cells, affects cancer growth in its soluble form by interacting with several receptor tyrosine kinases (RTK). Overall, decorin leads to a context-dependent and protracted cessation of oncogenic RTK activity by attenuating their ability to drive a prosurvival program and to sustain a proangiogenic network. Through an unbiased transcriptomic analysis using deep RNAseq, we identified that decorin down-regulated a cluster of tumor-associated genes involved in lymphatic vessel (LV) development when systemically delivered to mice harboring breast carcinoma allografts. We found that Lyve1 and Podoplanin, two established markers of LVs, were markedly suppressed at both the mRNA and protein levels, and this suppression correlated with a significant reduction in tumor LVs. We further identified that soluble decorin, but not its homologous proteoglycan biglycan, inhibited LV sprouting in an ex vivo 3D model of lymphangiogenesis. Mechanistically, we found that decorin interacted with vascular endothelial growth factor receptor 3 (VEGFR3), the main lymphatic RTK, and its activity was required for the decorin-mediated block of lymphangiogenesis. Finally, we identified that Lyve1 was in part degraded via decorin-evoked autophagy in a nutrient- and energy-independent manner. These findings implicate decorin as a biological factor with antilymphangiogenic activity and provide a potential therapeutic agent for curtailing breast cancer growth and metastasis.
Collapse
Affiliation(s)
- Dipon K. Mondal
- Department of Pathology and Genomic Medicine, and the Translational Cellular Oncology Program, Sidney Kimmel Cancer Center, Sidney Kimmel Medical College at Thomas Jefferson University, Philadelphia, PA19107
| | - Christopher Xie
- Department of Pathology and Genomic Medicine, and the Translational Cellular Oncology Program, Sidney Kimmel Cancer Center, Sidney Kimmel Medical College at Thomas Jefferson University, Philadelphia, PA19107
| | - Gabriel J. Pascal
- Department of Pathology and Genomic Medicine, and the Translational Cellular Oncology Program, Sidney Kimmel Cancer Center, Sidney Kimmel Medical College at Thomas Jefferson University, Philadelphia, PA19107
| | - Simone Buraschi
- Department of Pathology and Genomic Medicine, and the Translational Cellular Oncology Program, Sidney Kimmel Cancer Center, Sidney Kimmel Medical College at Thomas Jefferson University, Philadelphia, PA19107
| | - Renato V. Iozzo
- Department of Pathology and Genomic Medicine, and the Translational Cellular Oncology Program, Sidney Kimmel Cancer Center, Sidney Kimmel Medical College at Thomas Jefferson University, Philadelphia, PA19107
| |
Collapse
|
11
|
Dhakal S, Park HS, Seddu K, Lee JS, Creisher PS, Seibert B, Davis KM, Hernandez IR, Maul RW, Klein SL. Estradiol mediates greater germinal center responses to influenza vaccination in female than male mice. mBio 2024; 15:e0032624. [PMID: 38441028 PMCID: PMC11005424 DOI: 10.1128/mbio.00326-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Accepted: 02/09/2024] [Indexed: 03/06/2024] Open
Abstract
Adult females of reproductive age develop greater antibody responses to inactivated influenza vaccines (IIV) than males. How sex, age, and sex steroid concentrations impact B cells and durability of IIV-induced immunity and protection over 4 months post-vaccination (mpv) was analyzed. Vaccinated adult females had greater germinal center B cell and plasmablast frequencies in lymphoid tissues, higher neutralizing antibody responses 1-4 mpv, and better protection against live H1N1 challenge than adult males. Aged mice, regardless of sex, had reduced B cell frequencies, less durable antibody responses, and inferior protection after challenge than adult mice, which correlated with diminished estradiol among aged females. To confirm that greater IIV-induced immunity was caused by sex hormones, four core genotype (FCG) mice were used, in which the testes-determining gene, Sry, was deleted from chromosome Y (ChrY) and transferred to Chr3 to separate gonadal sex (i.e., ovaries or testes) from sex chromosome complement (i.e., XX or XY complement). Vaccinated, gonadal female FCG mice (XXF and XYF) had greater numbers of B cells, higher antiviral antibody titers, and reduced pulmonary virus titers following live H1N1 challenge than gonadal FCG males (XYM and XXM). To establish that lower estradiol concentrations cause diminished immunity, adult and aged females received either a placebo or estradiol replacement therapy prior to IIV. Estradiol replacement significantly increased IIV-induced antibody responses and reduced morbidity after the H1N1 challenge among aged females. These data highlight that estradiol is a targetable mechanism mediating greater humoral immunity following vaccination among adult females.IMPORTANCEFemales of reproductive ages develop greater antibody responses to influenza vaccines than males. We hypothesized that female-biased immunity and protection against influenza were mediated by estradiol signaling in B cells. Using diverse mouse models ranging from advanced-age mice to transgenic mice that separate sex steroids from sex chromosome complement, those mice with greater concentrations of estradiol consistently had greater numbers of antibody-producing B cells in lymphoid tissue, higher antiviral antibody titers, and greater protection against live influenza virus challenge. Treatment of aged female mice with estradiol enhanced vaccine-induced immunity and protection against disease, suggesting that estradiol signaling in B cells is critical for improved vaccine outcomes in females.
Collapse
Affiliation(s)
- Santosh Dhakal
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Han-Sol Park
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Kumba Seddu
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - John S. Lee
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Patrick S. Creisher
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Brittany Seibert
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Kimberly M. Davis
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Isabella R. Hernandez
- Laboratory of Molecular Biology and Immunology, National Institute on Aging, National Institutes of Health, Baltimore, Maryland, USA
| | - Robert W. Maul
- Laboratory of Molecular Biology and Immunology, National Institute on Aging, National Institutes of Health, Baltimore, Maryland, USA
| | - Sabra L. Klein
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| |
Collapse
|
12
|
Dhillon-LaBrooy A, Braband KL, Tantawy E, Rampoldi F, Kao YS, Boukhallouk F, Velasquez LN, Mamareli P, Silva L, Damasceno LEA, Weidenthaler-Barth B, Berod L, Almeida L, Sparwasser T. Inhibition of Mitochondrial Translation Ameliorates Imiquimod-Induced Psoriasis-Like Skin Inflammation by Targeting Vγ4+ γδ T Cells. J Invest Dermatol 2024; 144:844-854.e2. [PMID: 37832844 DOI: 10.1016/j.jid.2023.09.275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Revised: 09/20/2023] [Accepted: 09/21/2023] [Indexed: 10/15/2023]
Abstract
Psoriasis is an inflammatory skin disorder that is characterized by keratinocyte hyperproliferation in response to immune cell infiltration and cytokine secretion in the dermis. γδ T cells expressing the Vγ4 TCR chain are among the highest contributors of IL-17A, which is a major cytokine that drives a psoriasis flare, making Vγ4+ γδ T cells a suitable target to restrict psoriasis progression. In this study, we demonstrate that mitochondrial translation inhibition within Vγ4+ γδ T cells effectively reduced erythema, scaling, and skin thickening in a murine model of psoriatic disease. The antibiotic linezolid, which blocks mitochondrial translation, inhibited the production of mitochondrial-encoded protein cytochrome c oxidase in Vγ4+ γδ T cells and systemically reduced the frequencies of IL-17A+ Vγ4+ γδ T cells, effectively resolving IL-17A-dependent inflammation. Inhibiting mitochondrial translation could be a novel metabolic approach to interrupt IL-17A signaling in Vγ4+ T cells and reduce psoriasis-like skin pathophysiology.
Collapse
Affiliation(s)
- Ayesha Dhillon-LaBrooy
- Institute of Medical Microbiology and Hygiene, University Medical Center of the Johannes University Gutenberg, Mainz, Germany
| | - Kathrin L Braband
- Institute of Medical Microbiology and Hygiene, University Medical Center of the Johannes University Gutenberg, Mainz, Germany
| | - Eshraq Tantawy
- Institute of Medical Microbiology and Hygiene, University Medical Center of the Johannes University Gutenberg, Mainz, Germany
| | - Francesca Rampoldi
- Institute of Medical Microbiology and Hygiene, University Medical Center of the Johannes University Gutenberg, Mainz, Germany
| | - Yu-San Kao
- Institute of Medical Microbiology and Hygiene, University Medical Center of the Johannes University Gutenberg, Mainz, Germany
| | - Fatima Boukhallouk
- Institute of Medical Microbiology and Hygiene, University Medical Center of the Johannes University Gutenberg, Mainz, Germany
| | - Lis Noelia Velasquez
- Institute of Medical Microbiology and Hygiene, University Medical Center of the Johannes University Gutenberg, Mainz, Germany
| | - Panagiota Mamareli
- Institute of Medical Microbiology and Hygiene, University Medical Center of the Johannes University Gutenberg, Mainz, Germany
| | - Luana Silva
- Institute of Medical Microbiology and Hygiene, University Medical Center of the Johannes University Gutenberg, Mainz, Germany
| | - Luis Eduardo Alves Damasceno
- Institute of Medical Microbiology and Hygiene, University Medical Center of the Johannes University Gutenberg, Mainz, Germany; Center for Research in Inflammatory Diseases (CRID), Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, São Paulo, Brazil
| | - Beate Weidenthaler-Barth
- Department of Dermatology, University Medical Center of the Johannes University Gutenberg, Mainz, Germany
| | - Luciana Berod
- Institute of Molecular Medicine, University Medical Center of the Johannes University Gutenberg, Mainz, Germany; Research Center for Immunotherapy, University Medical Centre of the Johannes University Gutenberg, Mainz, Germany
| | - Luís Almeida
- Institute of Medical Microbiology and Hygiene, University Medical Center of the Johannes University Gutenberg, Mainz, Germany
| | - Tim Sparwasser
- Institute of Medical Microbiology and Hygiene, University Medical Center of the Johannes University Gutenberg, Mainz, Germany; Research Center for Immunotherapy, University Medical Centre of the Johannes University Gutenberg, Mainz, Germany.
| |
Collapse
|
13
|
Aguilar CC, Kalia A, Brisse ME, Dowd KA, Wise-Dent O, Burgomaster KE, Droppo J, Pierson TC, Hickman HD. Subcapsular sinus macrophages maximize germinal center development in non-draining lymph nodes during blood-borne viral infection. Sci Immunol 2024; 9:eadi4926. [PMID: 38457515 DOI: 10.1126/sciimmunol.adi4926] [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: 04/28/2023] [Accepted: 01/29/2024] [Indexed: 03/10/2024]
Abstract
Lymph node (LN) germinal centers (GCs) are critical sites for B cell activation and differentiation. GCs develop after specialized CD169+ macrophages residing in LN sinuses filter antigens (Ags) from the lymph and relay these Ags into proximal B cell follicles. Many viruses, however, first reach LNs through the blood during viremia (virus in the blood), rather than through lymph drainage from infected tissue. How LNs capture viral Ag from the blood to allow GC development is not known. Here, we followed Zika virus (ZIKV) dissemination in mice and subsequent GC formation in both infected tissue-draining and non-draining LNs. From the footpad, ZIKV initially disseminated through two LN chains, infecting LN macrophages and leading to GC formation. Despite rapid ZIKV viremia, non-draining LNs were not infected for several days. Non-draining LN infection correlated with virus-induced vascular leakage and neutralization of permeability reduced LN macrophage attrition. Depletion of non-draining LN macrophages significantly decreased GC B cells in these nodes. Thus, although LNs inefficiently captured viral Ag directly from the blood, GC formation in non-draining LNs proceeded similarly to draining LNs through LN sinus CD169+ macrophages. Together, our findings reveal a conserved pathway allowing LN macrophages to activate antiviral B cells in LNs distal from infected tissue after blood-borne viral infection.
Collapse
Affiliation(s)
- Cynthia C Aguilar
- Viral Immunity and Pathogenesis Unit, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Anurag Kalia
- Viral Immunity and Pathogenesis Unit, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Morgan E Brisse
- Viral Immunity and Pathogenesis Unit, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Kimberly A Dowd
- Arbovirus Immunity Section, Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Olivia Wise-Dent
- Viral Immunity and Pathogenesis Unit, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Katherine E Burgomaster
- Arbovirus Immunity Section, Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Joanna Droppo
- Viral Immunity and Pathogenesis Unit, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Theodore C Pierson
- Arbovirus Immunity Section, Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Heather D Hickman
- Viral Immunity and Pathogenesis Unit, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
14
|
Li Y, Yuan W, Zhong M, Qi J, Zheng X, Xie X, Li T, Zhang H, Jiang X, Peng L, Dai H. A murine groin site cardiac transplantation model-applicable tool for studying roles of peripheral lymph nodes in transplantation. Xenotransplantation 2024; 31:e12817. [PMID: 37548057 DOI: 10.1111/xen.12817] [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: 04/25/2023] [Revised: 07/22/2023] [Accepted: 07/26/2023] [Indexed: 08/08/2023]
Abstract
The murine heterotopic cardiac transplantation model has been widely used to study antigen-specific immune responses or new immunosuppressive agents, which have a strong correlation with peripheral lymph nodes. Thus, a new organ transplantation model that is applicable to related studies is needed. Here, we describe a groin-site murine heart transplantation model using a cuff technique, in which the donor aorta and pulmonary artery are anastomosed to the truncated femoral vessels of the recipient. The mean survival time (MST) of the grafts in BALB/c-to-C57BL/6 allo-transplant group was 7.2 ± 0.3 days, and 1.9 ± 0.2 days in BALB/c-to-Sprague-Dawley (SD) rat xeno-transplant group. H&E results show that donor hearts from both groups demonstrate typical pathological features at the endpoint. Evans Blue tracing revealed that the popliteal lymph nodes of the grafted side hindlimb are larger than those of the contralateral side. Moreover, IHC staining for CD3, CD20 shows that the germinal center and cortex region of the grafted side of popliteal lymph nodes is apparently increased than that of the contralateral side. To sum up, this model may serve as an ideal model to study the role of peripheral lymph nodes in organ transplant rejection. In addition, extra-peritoneal grafting makes a step forward in animal welfare under the 3Rs' principle (Replacement, Reduction, Refinement).
Collapse
Affiliation(s)
- Yaguang Li
- Department of Kidney Transplantation, The Second Xiangya Hospital, of Central South University, Changsha, China
- Clinical Research Center for Organ Transplantation in Hunan Province, Central South University, Changsha, China
| | - Wenjia Yuan
- Department of Kidney Transplantation, The Second Xiangya Hospital, of Central South University, Changsha, China
- Clinical Research Center for Organ Transplantation in Hunan Province, Central South University, Changsha, China
| | - Mingda Zhong
- Department of Kidney Transplantation, The Second Xiangya Hospital, of Central South University, Changsha, China
| | - Julia Qi
- Peking University Health Science Center, Beijing, China
| | - Xinguo Zheng
- Department of Kidney Transplantation, The Second Xiangya Hospital, of Central South University, Changsha, China
- Clinical Research Center for Organ Transplantation in Hunan Province, Central South University, Changsha, China
| | - Xubiao Xie
- Department of Kidney Transplantation, The Second Xiangya Hospital, of Central South University, Changsha, China
- Clinical Research Center for Organ Transplantation in Hunan Province, Central South University, Changsha, China
| | - Tengfang Li
- Department of Kidney Transplantation, The Second Xiangya Hospital, of Central South University, Changsha, China
- Clinical Research Center for Organ Transplantation in Hunan Province, Central South University, Changsha, China
| | - Hedong Zhang
- Department of Kidney Transplantation, The Second Xiangya Hospital, of Central South University, Changsha, China
- Clinical Research Center for Organ Transplantation in Hunan Province, Central South University, Changsha, China
| | - Xin Jiang
- Department of Organ Transplantation, The Fifth Clinical Medical College of Henan University of Chinese Medicine (Zhengzhou People's Hospital), Zhengzhou, China
| | - Longkai Peng
- Department of Kidney Transplantation, The Second Xiangya Hospital, of Central South University, Changsha, China
- Clinical Research Center for Organ Transplantation in Hunan Province, Central South University, Changsha, China
| | - Helong Dai
- Department of Kidney Transplantation, The Second Xiangya Hospital, of Central South University, Changsha, China
- Clinical Research Center for Organ Transplantation in Hunan Province, Central South University, Changsha, China
- Clinical Immunology Center, Central South University, Changsha, China
| |
Collapse
|
15
|
Zhong X, Patel A, Sun Y, Saeboe AM, Dennis AM. Multiplexed Short-wave Infrared Imaging Highlights Anatomical Structures in Mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.29.577849. [PMID: 38352582 PMCID: PMC10862713 DOI: 10.1101/2024.01.29.577849] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/23/2024]
Abstract
While multiplexed fluorescence imaging is frequently used for in vitro microscopy, extending the technique to whole animal imaging in vivo has remained challenging due to the attenuation and scattering of visible and traditional near infrared (NIR-I) wavelengths. Fluorescence imaging using short-wave infrared (SWIR, 1000 - 1700 nm, a.k.a. NIR-II) light enables deeper tissue penetration for preclinical imaging compared to previous methods due to reduced tissue scattering and minimal background autofluorescence in this optical window. Combining NIR-I excitation wavelengths with multiple distinct SWIR emission peaks presents a tremendous opportunity to distinguish multiple fluorophores with high precision for non-invasive, multiplexed anatomical imaging in small animal models. SWIR-emitting semiconductor quantum dots (QDs) with tunable emission peaks and optical stability have emerged as powerful contrast agents, but SWIR imaging demonstrations have yet to move beyond two-color imaging schemes. In this study, we engineered a set of three high quantum yield lead sulfide/cadmium sulfide (PbS/CdS) core/shell QDs with distinct SWIR emissions ranging from 1100 - 1550 nm and utilize these for simultaneous three-color imaging in mice. We first use QDs to non-invasively track lymphatic drainage, highlighting the detailed network of lymphatic vessels with high-resolution with a widefield imaging over a 2 hr period. We then perform multiplexed imaging with all three QDs to distinctly visualize the lymphatic system and spatially overlapping vasculature network. This work establishes optimized SWIR QDs for next-generation multiplexed preclinical imaging, moving beyond the capability of previous dual-labeling techniques. The capacity to discriminate several fluorescent labels through non-invasive NIR-I excitation and SWIR detection unlocks numerous opportunities for studies of disease progression, drug biodistribution, and cell trafficking dynamics in living organisms.
Collapse
Affiliation(s)
- Xingjian Zhong
- Department of Biomedical Engineering, Boston University, Boston, MA, 02215, USA
- Department of Chemical Engineering, Northeastern University, Boston, MA, 02115, USA
| | - Amish Patel
- Department of Biomedical Engineering, Boston University, Boston, MA, 02215, USA
- Department of Chemical Engineering, Northeastern University, Boston, MA, 02115, USA
| | - Yidan Sun
- Department of Chemical Engineering, Northeastern University, Boston, MA, 02115, USA
| | - Alexander M. Saeboe
- Division of Materials Science & Engineering, Boston University, Boston, MA, 02215, USA
| | - Allison M. Dennis
- Department of Biomedical Engineering, Boston University, Boston, MA, 02215, USA
- Department of Chemical Engineering, Northeastern University, Boston, MA, 02115, USA
- Division of Materials Science & Engineering, Boston University, Boston, MA, 02215, USA
| |
Collapse
|
16
|
Fazli S, Thomas A, Estrada AE, Ross HA, Xthona Lee D, Kazmierczak S, Slifka MK, Montefiori D, Messer WB, Curlin ME. Contralateral second dose improves antibody responses to a 2-dose mRNA vaccination regimen. J Clin Invest 2024; 134:e176411. [PMID: 38227381 PMCID: PMC10940087 DOI: 10.1172/jci176411] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Accepted: 01/09/2024] [Indexed: 01/17/2024] Open
Abstract
BACKGROUNDVaccination is typically administered without regard to site of prior vaccination, but this factor may substantially affect downstream immune responses.METHODSWe assessed serological responses to initial COVID-19 vaccination in baseline seronegative adults who received second-dose boosters in the ipsilateral or contralateral arm relative to initial vaccination. We measured serum SARS-CoV-2 spike-specific Ig, receptor-binding domain-specific (RBD-specific) IgG, SARS-CoV-2 nucleocapsid-specific IgG, and neutralizing antibody titers against SARS-CoV-2.D614G (early strain) and SARS-CoV-2.B.1.1.529 (Omicron) at approximately 0.6, 8, and 14 months after boosting.RESULTSIn 947 individuals, contralateral boosting was associated with higher spike-specific serum Ig, and this effect increased over time, from a 1.1-fold to a 1.4-fold increase by 14 months (P < 0.001). A similar pattern was seen for RBD-specific IgG. Among 54 pairs matched for age, sex, and relevant time intervals, arm groups had similar antibody levels at study visit 2 (W2), but contralateral boosting resulted in significantly higher binding and neutralizing antibody titers at W3 and W4, with progressive increase over time, ranging from 1.3-fold (total Ig, P = 0.007) to 4.0-fold (pseudovirus neutralization to B.1.1.529, P < 0.001).CONCLUSIONSIn previously unexposed adults receiving an initial vaccine series with the BNT162b2 mRNA COVID-19 vaccine, contralateral boosting substantially increases antibody magnitude and breadth at times beyond 3 weeks after vaccination. This effect should be considered during arm selection in the context of multidose vaccine regimens.FUNDINGM.J. Murdock Charitable Trust, OHSU Foundation, NIH.
Collapse
Affiliation(s)
| | - Archana Thomas
- Oregon National Primate Research Center, Division of Neuroscience, and
| | - Abram E. Estrada
- Department of Molecular Microbiology and Immunology, Oregon Health & Science University, Portland, Oregon, USA
| | | | - David Xthona Lee
- Department of Molecular Microbiology and Immunology, Oregon Health & Science University, Portland, Oregon, USA
| | - Steven Kazmierczak
- Department of Pathology, Oregon Health & Science University, Portland, Oregon, USA
| | - Mark K. Slifka
- Oregon National Primate Research Center, Division of Neuroscience, and
| | - David Montefiori
- Department of Surgery, Duke University Medical Center, Durham, North Carolina, USA
| | - William B. Messer
- Department of Molecular Microbiology and Immunology, Oregon Health & Science University, Portland, Oregon, USA
- Department of Medicine, Division of Infectious Diseases, Oregon Health & Science University, Portland, Oregon, USA
- Program in Epidemiology, Oregon Health & Science University, Portland State University School of Public Health, Portland, Oregon, USA
| | - Marcel E. Curlin
- Department of Occupational Health
- Department of Medicine, Division of Infectious Diseases, Oregon Health & Science University, Portland, Oregon, USA
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, Portland, Oregon, USA
| |
Collapse
|
17
|
Lucas CJ, Sheridan RM, Reynoso GV, Davenport BJ, McCarthy MK, Martin A, Hesselberth JR, Hickman HD, Tamburini BA, Morrison TE. Chikungunya virus infection disrupts lymph node lymphatic endothelial cell composition and function via MARCO. JCI Insight 2024; 9:e176537. [PMID: 38194268 PMCID: PMC11143926 DOI: 10.1172/jci.insight.176537] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Accepted: 01/05/2024] [Indexed: 01/10/2024] Open
Abstract
Infection with chikungunya virus (CHIKV) causes disruption of draining lymph node (dLN) organization, including paracortical relocalization of B cells, loss of the B cell-T cell border, and lymphocyte depletion that is associated with infiltration of the LN with inflammatory myeloid cells. Here, we found that, during the first 24 hours of infection, CHIKV RNA accumulated in MARCO-expressing lymphatic endothelial cells (LECs) in both the floor and medullary LN sinuses. The accumulation of viral RNA in the LN was associated with a switch to an antiviral and inflammatory gene expression program across LN stromal cells, and this inflammatory response - including recruitment of myeloid cells to the LN - was accelerated by CHIKV-MARCO interactions. As CHIKV infection progressed, both floor and medullary LECs diminished in number, suggesting further functional impairment of the LN by infection. Consistent with this idea, antigen acquisition by LECs, a key function of LN LECs during infection and immunization, was reduced during pathogenic CHIKV infection.
Collapse
Affiliation(s)
- Cormac J. Lucas
- Department of Immunology & Microbiology and
- RNA Bioscience Initiative, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Ryan M. Sheridan
- RNA Bioscience Initiative, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Glennys V. Reynoso
- Viral Immunity & Pathogenesis Unit, Laboratory of Clinical Immunology & Microbiology, National Institutes of Allergy & Infectious Disease, NIH, Bethesda, Maryland, USA
| | | | | | - Aspen Martin
- Department of Biochemistry & Molecular Genetics and
| | - Jay R. Hesselberth
- RNA Bioscience Initiative, University of Colorado School of Medicine, Aurora, Colorado, USA
- Department of Biochemistry & Molecular Genetics and
| | - Heather D. Hickman
- Viral Immunity & Pathogenesis Unit, Laboratory of Clinical Immunology & Microbiology, National Institutes of Allergy & Infectious Disease, NIH, Bethesda, Maryland, USA
| | - Beth A.J. Tamburini
- Department of Immunology & Microbiology and
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Colorado School of Medicine, Aurora, Colorado, USA
| | | |
Collapse
|
18
|
Li C, Lu G, Jiang Y, Su H, Li C. A Novel Etanercept-loaded Nano-emulsion for Targeted Treatment of Inflammatory Arthritis via Draining Lymph Node. Curr Drug Deliv 2024; 21:1106-1113. [PMID: 37565561 DOI: 10.2174/1567201821666230810115230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Revised: 04/23/2023] [Accepted: 05/16/2023] [Indexed: 08/12/2023]
Abstract
BACKGROUND Rheumatoid arthritis (RA) is a systemic autoimmune disease (AD), and the global incidence rate is 0.5 ~ 1%. Existing medications might reduce symptoms, however, there is no known cure for this illness. Etanercept (EN) can competitively inhibit TNF-α binding to the TNF receptor on the cell surface to treat RA. However, subcutaneous injection of free EN predisposes to systemic distribution and induces immune system hypofunction. Draining lymph nodes (LNs) play a significant role in the onset, maintenance, and progression of RA as they are the primary sites of aberrant immune response and inflammatory cytokine production. AIM The purpose of this study was to successfully treat RA with etanercept by encapsulating it in nanoemulsions (NEs/EN) and then delivering it specifically to draining LNs. The EN-loaded NEs were prepared by high-pressure homogenization method and modified with DSPE-mPEG2000 and Ca(OH)2. METHODS A novel nano-emulsion (NE) was constructed to deliver EN (NE/EN) to RA-draining LNs. To decrease aggregation and load EN, DSPE-mPEG2000 and Ca(OH)2 were successively decorated on the surface of the lipid injectable emulsions. The hydrodynamic diameter and morphology of NEs/EN were investigated by using a laser particle size analyzer and transmission electron microscopy, respectively. The in vivo fluorescence imaging system was used to study the in vivo LN targeting ability of the formulation. In the therapeutic experiment, NEs/EN was subcutaneously administrated to inhibit the development of the mouse arthritis model. RESULTS Circular dichroism spectrum and L929 cell experiment confirmed that NEs encapsulation had no impact on the biological activity of EN. In vivo investigation on collagen-induced arthritis (CIA) mouse model showed that NEs/EN have good inguinal lymph node targeting capabilities, as well as, anti-inflammatory effect against RA. Compared with the free group, the paw thickness and arthritic score in NEs/EN group were significantly alleviated. Moreover, the concentration of pro-inflammatory cytokines TNF-α and IL-1β in NEs/EN-treated mice was lower than that in free EN. CONCLUSION NEs/EN effectively improve the effectiveness of EN in the treatment of RA. Our work provides an experimental foundation for expanding the clinical application of EN.
Collapse
Affiliation(s)
- Chenglong Li
- Department of Pharmacy, Deyang People's Hospital, Deyang 618000, China
| | - Guanting Lu
- Laboratory of Translational Medicine Research, Department of Pathology, Deyang People's Hospital, Deyang 618000, China
- Key Laboratory of Tumor Molecular Research of Deyang, Deyang People's Hospital, Deyang 618000, China
| | - Yue Jiang
- Department of Pharmacy, Deyang People's Hospital, Deyang 618000, China
| | - Huaiyu Su
- Department of Pharmacy, Deyang People's Hospital, Deyang 618000, China
| | - Chen Li
- Centre for Translational Research in Cancer, Sichuan Cancer Hospital & Institute, Chengdu 610000, China
- School of Medicine, University of Electronic Science and Technology, Chengdu 610000, China
| |
Collapse
|
19
|
Dhakal S, Park HS, Seddu K, Lee J, Creisher PS, Davis KM, Hernandez IR, Maul RW, Klein SL. Estradiol Mediates Greater Germinal Center Responses to Influenza Vaccination in Female than Male Mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.11.27.568847. [PMID: 38077071 PMCID: PMC10705292 DOI: 10.1101/2023.11.27.568847] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/24/2023]
Abstract
Adult females of reproductive ages develop greater antibody responses to inactivated influenza vaccine (IIV) than males. How sex, age, and sex steroid changes impact B cells and durability of IIV-induced immunity and protection over 4-months post-vaccination (mpv) was analyzed. Vaccinated adult females had greater germinal center (GC) B cell and plasmablast frequencies in lymphoid tissues, higher neutralizing antibody responses 1-4 mpv, and better protection against live H1N1 challenge than adult males. Aged mice, regardless of sex, had reduced B cell frequencies, less durable antibody responses, and inferior protection after challenge than adult mice, which correlated with diminished estradiol among aged females. To confirm that greater IIV-induced immunity was caused by sex hormones, four core genotype (FCG) mice were used, in which the testes determining gene, Sry, was deleted from ChrY and transferred to Chr3, to separate gonadal sex (i.e., ovaries or testes) from sex chromosome complement (i.e., XX or XY complement). Vaccinated, gonadal female FCG mice (XXF and XYF) had greater numbers of B cells, higher antiviral antibody titers, and reduced pulmonary virus titers following live H1N1 challenge than gonadal FCG males (XYM and XXM). To establish that lower estradiol concentrations cause diminished immunity, adult and aged females received either a placebo or estradiol replacement therapy prior to IIV. Estradiol replacement significantly increased IIV-induced antibody responses and reduced morbidity after the H1N1 challenge among aged females. These data highlight that estradiol is a targetable mechanism mediating greater humoral immunity following vaccination among adult females.
Collapse
Affiliation(s)
- Santosh Dhakal
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Han-Sol Park
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Kumba Seddu
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - John Lee
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Patrick S. Creisher
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Kimberly M. Davis
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Isabella R. Hernandez
- Laboratory of Molecular Biology and Immunology, National Institute on Aging, National Institutes of Health, Baltimore, Maryland, USA
| | - Robert W. Maul
- Laboratory of Molecular Biology and Immunology, National Institute on Aging, National Institutes of Health, Baltimore, Maryland, USA
| | - Sabra L. Klein
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| |
Collapse
|
20
|
Machado H, Temudo A, Niz MD. The lymphatic system favours survival of a unique T. brucei population. Biol Open 2023; 12:bio059992. [PMID: 37870927 PMCID: PMC10651106 DOI: 10.1242/bio.059992] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Accepted: 10/13/2023] [Indexed: 10/25/2023] Open
Abstract
Trypanosoma brucei colonise and multiply in the blood vasculature, as well as in various organs of the host's body. Lymph nodes have been previously shown to harbour large numbers of parasites, and the lymphatic system has been proposed as a key site that allows T. brucei distribution through, and colonization of the mammalian body. However, visualization of host-pathogen interactions in the lymphatic system has never captured dynamic events with high spatial and temporal resolution throughout infection. In our work, we used a mixture of tools including intravital microscopy and ex vivo imaging to study T. brucei distribution in 20 sets of lymph nodes. We demonstrate that lymph node colonization by T. brucei is different across lymph node sets, with the most heavily colonised being the draining lymph nodes of main tissue reservoirs: the gonadal white adipose tissue and pancreas. Moreover, we show that the lymphatic vasculature is a pivotal site for parasite dispersal, and altering this colonization by blocking LYVE-1 is detrimental for parasite survival. Additionally, parasites within the lymphatic vasculature have unique morphological and behavioural characteristics, different to those found in the blood, demonstrating that across both types of vasculature, these environments are physically separated. Finally, we demonstrate that the lymph nodes and the lymphatic vasculature undergo significant alterations during T. brucei infection, resulting in oedema throughout the host's body.
Collapse
Affiliation(s)
- Henrique Machado
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa 1649-028, Portugal
| | - António Temudo
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa 1649-028, Portugal
- Bioimaging Unit, Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa 1649-028, Portugal
| | - Mariana De Niz
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa 1649-028, Portugal
| |
Collapse
|
21
|
Lucas CJ, Sheridan RM, Reynoso GV, Davenport BJ, McCarthy MK, Martin A, Hesselberth JR, Hickman HD, Tamburini BAJ, Morrison TE. Chikungunya virus infection disrupts lymph node lymphatic endothelial cell composition and function via MARCO. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.12.561615. [PMID: 37873393 PMCID: PMC10592756 DOI: 10.1101/2023.10.12.561615] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/25/2023]
Abstract
Infection with chikungunya virus (CHIKV) causes disruption of draining lymph node (dLN) organization, including paracortical relocalization of B cells, loss of the B cell-T cell border, and lymphocyte depletion that is associated with infiltration of the LN with inflammatory myeloid cells. Here, we find that during the first 24 h of infection, CHIKV RNA accumulates in MARCO-expressing lymphatic endothelial cells (LECs) in both the floor and medullary LN sinuses. The accumulation of viral RNA in the LN was associated with a switch to an antiviral and inflammatory gene expression program across LN stromal cells, and this inflammatory response, including recruitment of myeloid cells to the LN, was accelerated by CHIKV-MARCO interactions. As CHIKV infection progressed, both floor and medullary LECs diminished in number, suggesting further functional impairment of the LN by infection. Consistent with this idea, we find that antigen acquisition by LECs, a key function of LN LECs during infection and immunization, was reduced during pathogenic CHIKV infection.
Collapse
|
22
|
Jung JH, Kim KA, Choi YS, Kim ST. Effect of intralymphatic allergen-specific immunotherapy on house dust mite in a murine model of allergic rhinitis. Acta Otolaryngol 2023; 143:867-875. [PMID: 38059621 DOI: 10.1080/00016489.2023.2273405] [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: 08/09/2023] [Accepted: 10/11/2023] [Indexed: 12/08/2023]
Abstract
BACKGROUND Intralymphatic immunotherapy (ILIT) is a promising alternative for the treatment of patients with allergic rhinitis, providing similar therapeutic efficacy to conventional allergen-specific immunotherapy (AIT). However, the allergic mechanism of ILIT is not completely known. AIM The aim of this study was to determine the efficacy of ILIT in a house dust mite (HDM) mouse model of allergic rhinitis. METHODS BALB/c mice were divided into four groups: G1, control without allergy; G2, allergy sensitized with HDM; G3, allergy with ILIT (starting with HDM 1.25 μg/mL); and G4, allergy with ILIT (starting with HDM 2.5 μg/mL). After the murine model of allergic rhinitis with HDM was established, mice were administered an intralymphatic injection through the inguinal lymph nodes with HDM. RESULTS ILIT decreased serum total IgE level and eosinophil infiltration in the nasal mucosa. ILIT also decreased the expression levels of IL-13, IL-25, IL-33, IFN-γ, IL-6, and IL-17, and increased the expression of FoxP3(+) T reg cells. CONCLUSIONS AND SIGNIFICANCE Our results suggest that ILIT regulates the specific immunotherapy immunologic mechanism by downregulating Th1, Th2, and Th17 cytokines and upregulating FoxP3(+) T reg cells in the HDM allergic mouse model.
Collapse
Affiliation(s)
- Joo Hyun Jung
- Department of Otolaryngology-Head & Neck Surgery, Gachon University Gil Medical Center, Incheon, Republic of Korea
| | - Kyeong Ah Kim
- Gachon Medical Research Institute, Gachon University Gil Medical Center, Incheon, Republic of Korea
| | - Yun Sook Choi
- Gachon Medical Research Institute, Gachon University Gil Medical Center, Incheon, Republic of Korea
| | - Seon Tae Kim
- Department of Otolaryngology-Head & Neck Surgery, Gachon University Gil Medical Center, Incheon, Republic of Korea
| |
Collapse
|
23
|
Dhamdhere MR, Spiegelman DV, Schneper L, Erbe AK, Sondel PM, Spiegelman VS. Generation of Novel Immunocompetent Mouse Cell Lines to Model Experimental Metastasis of High-Risk Neuroblastoma. Cancers (Basel) 2023; 15:4693. [PMID: 37835389 PMCID: PMC10571844 DOI: 10.3390/cancers15194693] [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: 08/29/2023] [Revised: 09/18/2023] [Accepted: 09/21/2023] [Indexed: 10/15/2023] Open
Abstract
NB, being a highly metastatic cancer, is one of the leading causes of cancer-related deaths in children. Increased disease recurrence and clinical resistance in patients with metastatic high-risk NBs (HR-NBs) result in poor outcomes and lower overall survival. However, the paucity of appropriate in vivo models for HR-NB metastasis has limited investigations into the underlying biology of HR-NB metastasis. This study was designed to address this limitation and develop suitable immunocompetent models for HR-NB metastasis. Here, we developed several highly metastatic immunocompetent murine HR-NB cell lines. Our newly developed cell lines show 100% efficiency in modeling experimental metastasis in C57BL6 mice and feature metastasis to the sites frequently observed in humans with HR-NB (liver and bone). In vivo validation demonstrated their specifically gained metastatic phenotype. The in vitro characterization of the cell lines showed increased cell invasion, acquired anchorage-independent growth ability, and resistance to MHC-I induction upon IFN-γ treatment. Furthermore, RNA-seq analysis of the newly developed cells identified a differentially regulated gene signature and an enrichment of processes consistent with their acquired metastatic phenotype, including extracellular matrix remodeling, angiogenesis, cell migration, and chemotaxis. The presented newly developed cell lines are, thus, suitable and promising tools for HR-NB metastasis and microenvironment studies in an immunocompetent system.
Collapse
Affiliation(s)
- Mayura R. Dhamdhere
- Division of Pediatric Hematology and Oncology, Department of Pediatrics, The Pennsylvania State University College of Medicine, Hershey, PA 17033, USA
| | - Dan V. Spiegelman
- Department of Human Oncology, School of Medicine and Public Health, University of Wisconsin, Madison, WI 53792, USA; (D.V.S.); (P.M.S.)
| | - Lisa Schneper
- Department of Biochemistry and Molecular Biology, The Pennsylvania State University College of Medicine, Hershey, PA 17033, USA
| | - Amy K. Erbe
- Department of Human Oncology, School of Medicine and Public Health, University of Wisconsin, Madison, WI 53792, USA; (D.V.S.); (P.M.S.)
| | - Paul M. Sondel
- Department of Human Oncology, School of Medicine and Public Health, University of Wisconsin, Madison, WI 53792, USA; (D.V.S.); (P.M.S.)
| | - Vladimir S. Spiegelman
- Division of Pediatric Hematology and Oncology, Department of Pediatrics, The Pennsylvania State University College of Medicine, Hershey, PA 17033, USA
| |
Collapse
|
24
|
Ding L, Liang M, Li Y, Zeng M, Liu M, Ma W, Chen F, Li C, Reis RL, Li F, Wang Y. Zinc-Organometallic Framework Vaccine Controlled-Release Zn 2+ Regulates Tumor Extracellular Matrix Degradation Potentiate Efficacy of Immunotherapy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2302967. [PMID: 37439462 PMCID: PMC10520680 DOI: 10.1002/advs.202302967] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/21/2023] [Indexed: 07/14/2023]
Abstract
Tumor extracellular matrix (ECM) not only forms a physical barrier for T cells infiltration, but also regulates multiple immunosuppressive pathways, which is an important reason for immunotherapy failure. The cyclic guanosine monophosphate-adenosine monophosphate synthase-stimulator of interferon genes (cGAS-STING) pathway plays a key role in activating CD8+ T cells, maintaining CD8+ T cells stemness and enhancing the antitumor effect. Herein, a zinc-organometallic framework vaccine (ZPM@OVA-CpG) prepared by self-assembly, which achieves site-directed release of Zn2+ in dendritic cell (DC) lysosomes and tumor microenvironment under acidic conditions, is reported. The vaccine actively targets DC, significantly enhances cGAS-STING signal, promotes DC maturation and antigen cross-presentation, and induces strong activation of CD8+ T cells. Meanwhile, the vaccine reaches the tumor site, releasing Zn2+ , significantly up-regulates the activity of matrix metalloproteinase-2, degrades various collagen components of tumor ECM, effectively alleviates immune suppression, and significantly enhances the tumor infiltration and killing of CD8+ T cells. ZPM@OVA-CpG vaccine not only solves the problem of low antigen delivery efficiency and weak CD8+ T cells activation ability, but also achieves the degradation of tumor ECM via the vaccine for the first time, providing a promising therapeutic platform for the development of efficient novel tumor vaccines.
Collapse
Affiliation(s)
- Lin Ding
- The First Affiliated Hospital (Shenzhen People's Hospital)Southern University of Science and TechnologyShenzhen518055China
- Translational Medicine Collaborative Innovation CenterThe First Affiliated Hospital (Shenzhen People's Hospital)Southern University of Science and TechnologyShenzhen518020China
- Guangdong Engineering Technology Research Center of Stem Cell and Cell TherapyShenzhen Key Laboratory of Stem Cell Research and Clinical TransformationShenzhen Immune Cell Therapy Public Service PlatformShenzhen518020China
| | - Minli Liang
- The First Affiliated Hospital (Shenzhen People's Hospital)Southern University of Science and TechnologyShenzhen518055China
- Translational Medicine Collaborative Innovation CenterThe First Affiliated Hospital (Shenzhen People's Hospital)Southern University of Science and TechnologyShenzhen518020China
- Guangdong Engineering Technology Research Center of Stem Cell and Cell TherapyShenzhen Key Laboratory of Stem Cell Research and Clinical TransformationShenzhen Immune Cell Therapy Public Service PlatformShenzhen518020China
| | - Yuanyuan Li
- Clinical Laboratory, Jiaozuo Women's and Child's HospitalJiaozuo454001China
| | - Mei Zeng
- School of PharmacyGuangdong Medical UniversityDongguan523109China
| | - Meiting Liu
- School of PharmacyGuangdong Medical UniversityDongguan523109China
| | - Wei Ma
- Translational Medicine Collaborative Innovation CenterThe First Affiliated Hospital (Shenzhen People's Hospital)Southern University of Science and TechnologyShenzhen518020China
- Guangdong Engineering Technology Research Center of Stem Cell and Cell TherapyShenzhen Key Laboratory of Stem Cell Research and Clinical TransformationShenzhen Immune Cell Therapy Public Service PlatformShenzhen518020China
| | - Fuming Chen
- Translational Medicine Collaborative Innovation CenterThe First Affiliated Hospital (Shenzhen People's Hospital)Southern University of Science and TechnologyShenzhen518020China
- Guangdong Engineering Technology Research Center of Stem Cell and Cell TherapyShenzhen Key Laboratory of Stem Cell Research and Clinical TransformationShenzhen Immune Cell Therapy Public Service PlatformShenzhen518020China
| | - Chenchen Li
- Key Laboratory of Tropical Translational Medicine of Ministry of EducationSchool of Pharmacy and The First Affiliated HospitalHainan Medical UniversityHaikou570228China
| | - Rui L. Reis
- 3B's Research GroupI3Bs‐Research Institute on Biomaterials Biodegradables and BiomimeticsUniversity of MinhoGuimarães4805–017Portugal
| | - Fu‐Rong Li
- The First Affiliated Hospital (Shenzhen People's Hospital)Southern University of Science and TechnologyShenzhen518055China
- Translational Medicine Collaborative Innovation CenterThe First Affiliated Hospital (Shenzhen People's Hospital)Southern University of Science and TechnologyShenzhen518020China
- Guangdong Engineering Technology Research Center of Stem Cell and Cell TherapyShenzhen Key Laboratory of Stem Cell Research and Clinical TransformationShenzhen Immune Cell Therapy Public Service PlatformShenzhen518020China
| | - Yanli Wang
- Key Laboratory of Tropical Translational Medicine of Ministry of EducationSchool of Pharmacy and The First Affiliated HospitalHainan Medical UniversityHaikou570228China
| |
Collapse
|
25
|
Mondal DK, Xie C, Buraschi S, Iozzo RV. Decorin suppresses tumor lymphangiogenesis: A mechanism to curtail cancer progression. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.28.555187. [PMID: 37693608 PMCID: PMC10491239 DOI: 10.1101/2023.08.28.555187] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/12/2023]
Abstract
The complex interplay between malignant cells and the cellular and molecular components of the tumor stroma is a key aspect of cancer growth and development. These tumor-host interactions are often affected by soluble bioactive molecules such as proteoglycans. Decorin, an archetypical small leucine-rich proteoglycan primarily expressed by stromal cells, affects cancer growth in its soluble form by interacting with several receptor tyrosine kinases (RTK). Overall, decorin leads to a context-dependent and protracted cessation of oncogenic RTK activity by attenuating their ability to drive a pro-survival program and to sustain a pro-angiogenic network. Through an unbiased transcriptomic analysis using deep RNAseq, we discovered that decorin downregulated a cluster of tumor-associated genes involved in lymphatic vessel development when systemically delivered to mice harboring breast carcinoma allografts. We found that Lyve1 and Podoplanin, two established markers of lymphatic vessels, were markedly suppressed at both the mRNA and protein levels and this suppression correlated with a significant reduction in tumor lymphatic vessels. We further discovered that soluble decorin, but not its homologous proteoglycan biglycan, inhibited lymphatic vessel sprouting in an ex vivo 3D model of lymphangiogenesis. Mechanistically, we found that decorin interacted with VEGFR3, the main lymphatic RTK, and its activity was required for the decorin-mediated block of lymphangiogenesis. Finally, we discovered that Lyve1 was in part degraded via decorin-evoked autophagy in a nutrient- and energy-independent manner. These findings implicate decorin as a new biological factor with anti-lymphangiogenic activity and provide a potential therapeutic agent for curtailing breast cancer growth and metastasis.
Collapse
|
26
|
McMorrow R, Zambito G, Nigg A, Lila K, van den Bosch TPP, Lowik CWGM, Mezzanotte L. Whole-body bioluminescence imaging of T-cell response in PDAC models. Front Immunol 2023; 14:1207533. [PMID: 37497236 PMCID: PMC10367003 DOI: 10.3389/fimmu.2023.1207533] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Accepted: 06/12/2023] [Indexed: 07/28/2023] Open
Abstract
Introduction The location of T-cells during tumor progression and treatment provides crucial information in predicting the response in vivo. Methods Here, we investigated, using our bioluminescent, dual color, T-cell reporter mouse, termed TbiLuc, T-cell location and function during murine PDAC tumor growth and checkpoint blockade treatment with anti-PD-1 and anti-CTLA-4. Using this model, we could visualize T-cell location and function in the tumor and the surrounding tumor microenvironment longitudinally. We used murine PDAC clones that formed in vivo tumors with either high T-cell infiltration (immunologically 'hot') or low T-cell infiltration (immunologically 'cold'). Results Differences in total T-cell bioluminescence could be seen between the 'hot' and 'cold' tumors in the TbiLuc mice. During checkpoint blockade treatment we could see in the tumor-draining lymph nodes an increase in bioluminescence on day 7 after treatment. Conclusions In the current work, we showed that the TbiLuc mice can be used to monitor T-cell location and function during tumor growth and treatment.
Collapse
Affiliation(s)
- Roisin McMorrow
- Erasmus Medical Centre, Department of Radiology and Nuclear Medicine, Rotterdam, Netherlands
- Erasmus Medical Centre, Department of Molecular Genetics, Rotterdam, Netherlands
- Percuros BV, Leiden, Netherlands
| | - Giorgia Zambito
- Erasmus Medical Centre, Department of Radiology and Nuclear Medicine, Rotterdam, Netherlands
- Erasmus Medical Centre, Department of Molecular Genetics, Rotterdam, Netherlands
| | - Alex Nigg
- Erasmus Medical Centre, Department of Pathology, Erasmus MC Cancer Institute, Rotterdam, Netherlands
| | - Karishma Lila
- Erasmus Medical Centre, Department of Pathology, Erasmus MC Cancer Institute, Rotterdam, Netherlands
| | | | - Clemens W. G. M. Lowik
- Erasmus Medical Centre, Department of Radiology and Nuclear Medicine, Rotterdam, Netherlands
| | - Laura Mezzanotte
- Erasmus Medical Centre, Department of Radiology and Nuclear Medicine, Rotterdam, Netherlands
- Erasmus Medical Centre, Department of Molecular Genetics, Rotterdam, Netherlands
| |
Collapse
|
27
|
Zhang Y, Li Q, Ding M, Xiu W, Shan J, Yuwen L, Yang D, Song X, Yang G, Su X, Mou Y, Teng Z, Dong H. Endogenous/Exogenous Nanovaccines Synergistically Enhance Dendritic Cell-Mediated Tumor Immunotherapy. Adv Healthc Mater 2023; 12:e2203028. [PMID: 36807733 PMCID: PMC11468714 DOI: 10.1002/adhm.202203028] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 01/20/2023] [Indexed: 02/20/2023]
Abstract
Traditional dendritic cell (DC)-mediated immunotherapy is usually suppressed by weak immunogenicity in tumors and generally leads to unsatisfactory outcomes. Synergistic exogenous/endogenous immunogenic activation can provide an alternative strategy for evoking a robust immune response by promoting DC activation. Herein, Ti3 C2 MXene-based nanoplatforms (termed MXP) are prepared with high-efficiency near-infrared photothermal conversion and immunocompetent loading capacity to form endogenous/exogenous nanovaccines. Specifically, the immunogenic cell death of tumor cells induced by the photothermal effects of the MXP can generate endogenous danger signals and antigens release to boost vaccination for DC maturation and antigen cross-presentation. In addition, MXP can deliver model antigen ovalbumin (OVA) and agonists (CpG-ODN) as an exogenous nanovaccine (MXP@OC), which further enhances DC activation. Importantly, the synergistic strategy of photothermal therapy and DC-mediated immunotherapy by MXP significantly eradicates tumors and enhances adaptive immunity. Hence, the present work provides a two-pronged strategy for improving immunogenicity and killing tumor cells to achieve a favorable outcome in tumor patients.
Collapse
Affiliation(s)
- Yu Zhang
- Nanjing Stomatological HospitalMedical School of Nanjing University30 Zhongyang RoadNanjingJiangsu210008P. R. China
| | - Qiang Li
- Nanjing Stomatological HospitalMedical School of Nanjing University30 Zhongyang RoadNanjingJiangsu210008P. R. China
| | - Meng Ding
- Nanjing Stomatological HospitalMedical School of Nanjing University30 Zhongyang RoadNanjingJiangsu210008P. R. China
| | - Weijun Xiu
- Key Laboratory for Organic Electronics and Information DisplaysJiangsu Key Laboratory for BiosensorsInstitute of Advanced MaterialsJiangsu National Synergetic Innovation Centre for Advanced MaterialsNanjing University of Posts and Telecommunications9 Wenyuan RoadNanjingJiangsu210023P. R. China
| | - Jingyang Shan
- Department of NeurologyShenzhen Institute of Translational MedicineThe First Affiliated Hospital of Shenzhen UniversityShenzhen Second People's HospitalShenzhen518000P. R. China
| | - Lihui Yuwen
- Key Laboratory for Organic Electronics and Information DisplaysJiangsu Key Laboratory for BiosensorsInstitute of Advanced MaterialsJiangsu National Synergetic Innovation Centre for Advanced MaterialsNanjing University of Posts and Telecommunications9 Wenyuan RoadNanjingJiangsu210023P. R. China
| | - Dongliang Yang
- School of Physical and Mathematical SciencesNanjing Tech University30 South Puzhu RoadNanjingJiangsu211816P. R. China
| | - Xuejiao Song
- School of Physical and Mathematical SciencesNanjing Tech University30 South Puzhu RoadNanjingJiangsu211816P. R. China
| | - Guangwen Yang
- Nanjing Stomatological HospitalMedical School of Nanjing University30 Zhongyang RoadNanjingJiangsu210008P. R. China
| | - Xiaodan Su
- Key Laboratory for Organic Electronics and Information DisplaysJiangsu Key Laboratory for BiosensorsInstitute of Advanced MaterialsJiangsu National Synergetic Innovation Centre for Advanced MaterialsNanjing University of Posts and Telecommunications9 Wenyuan RoadNanjingJiangsu210023P. R. China
| | - Yongbin Mou
- Nanjing Stomatological HospitalMedical School of Nanjing University30 Zhongyang RoadNanjingJiangsu210008P. R. China
| | - Zhaogang Teng
- Key Laboratory for Organic Electronics and Information DisplaysJiangsu Key Laboratory for BiosensorsInstitute of Advanced MaterialsJiangsu National Synergetic Innovation Centre for Advanced MaterialsNanjing University of Posts and Telecommunications9 Wenyuan RoadNanjingJiangsu210023P. R. China
| | - Heng Dong
- Nanjing Stomatological HospitalMedical School of Nanjing University30 Zhongyang RoadNanjingJiangsu210008P. R. China
| |
Collapse
|
28
|
Arroz-Madeira S, Bekkhus T, Ulvmar MH, Petrova TV. Lessons of Vascular Specialization From Secondary Lymphoid Organ Lymphatic Endothelial Cells. Circ Res 2023; 132:1203-1225. [PMID: 37104555 PMCID: PMC10144364 DOI: 10.1161/circresaha.123.322136] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Revised: 03/31/2023] [Accepted: 03/31/2023] [Indexed: 04/29/2023]
Abstract
Secondary lymphoid organs, such as lymph nodes, harbor highly specialized and compartmentalized niches. These niches are optimized to facilitate the encounter of naive lymphocytes with antigens and antigen-presenting cells, enabling optimal generation of adaptive immune responses. Lymphatic vessels of lymphoid organs are uniquely specialized to perform a staggering variety of tasks. These include antigen presentation, directing the trafficking of immune cells but also modulating immune cell activation and providing factors for their survival. Recent studies have provided insights into the molecular basis of such specialization, opening avenues for better understanding the mechanisms of immune-vascular interactions and their applications. Such knowledge is essential for designing better treatments for human diseases given the central role of the immune system in infection, aging, tissue regeneration and repair. In addition, principles established in studies of lymphoid organ lymphatic vessel functions and organization may be applied to guide our understanding of specialization of vascular beds in other organs.
Collapse
Affiliation(s)
- Silvia Arroz-Madeira
- Department of Oncology, University of Lausanne, Switzerland (S.A.M., T.V.P.)
- Ludwig Institute for Cancer Research Lausanne, Switzerland (S.A.M., T.V.P.)
| | - Tove Bekkhus
- Department of Medical Biochemistry and Microbiology, Uppsala University, Sweden (T.B., M.H.U.)
| | - Maria H. Ulvmar
- Department of Medical Biochemistry and Microbiology, Uppsala University, Sweden (T.B., M.H.U.)
| | - Tatiana V. Petrova
- Department of Oncology, University of Lausanne, Switzerland (S.A.M., T.V.P.)
- Ludwig Institute for Cancer Research Lausanne, Switzerland (S.A.M., T.V.P.)
| |
Collapse
|
29
|
Peng J, Li H, Yang S, Zhang X, Li PZ, Nie X, Zhang L, Zhang Z. Individual variation in and lateral asymmetry of mouse epididymal draining lymph nodes. Am J Reprod Immunol 2023; 89:e13678. [PMID: 36648083 DOI: 10.1111/aji.13678] [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: 11/18/2022] [Revised: 12/24/2022] [Accepted: 01/10/2023] [Indexed: 01/18/2023] Open
Abstract
PROBLEM Draining lymph nodes (LNs) are pivotal sites for maintaining tolerance to self-antigens as well as eliciting immune responses to exogenous antigens. The epididymis is a male reproductive organ with a unique local immune environment. Although mice are the most commonly used laboratory animals for immunology research, there are no detailed descriptions of the anatomical location and function of LNs that drain the epididymis. METHOD OF STUDY Evans blue labeling was utilized to explore lymphatic drainage of the epididymis in eight- to ten-week-old male C57BL/6 mice. We confirmed the lymphatic drainage of the epididymis in mice using the objective technique of carboxyfluorescein succinimidyl ester (CFSE)-labeled cells. RESULTS By combined Evans blue labeling and fluorescent labeling, we found that 1) the patterns of epididymal LN drainage are highly heterogeneous between individual mice; 2) the leftside LNs participate in drainage more frequently than the right-side LNs; and 3) epididymal lymphatic drainage bypasses both the paraaortic and renal LNs in some mice. CONCLUSIONS These data highlighted the need to consider the individual variation in and lateral asymmetry of draining LNs when characterizing the regional immunology of the mouse epididymis.
Collapse
Affiliation(s)
- Jing Peng
- Andrology Center, Department of Urology, Peking University First Hospital, Peking University, Beijing, China
| | - Huixi Li
- Andrology Center, Department of Urology, Peking University First Hospital, Peking University, Beijing, China
| | - Shaojun Yang
- Key Laboratory of Infection and Immunity, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Xuyuan Zhang
- Key Laboratory of Infection and Immunity, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Patrick Z Li
- Key Laboratory of Infection and Immunity, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Xiaohua Nie
- Key Laboratory of Infection and Immunity, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Liguo Zhang
- Key Laboratory of Infection and Immunity, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Zhichao Zhang
- Andrology Center, Department of Urology, Peking University First Hospital, Peking University, Beijing, China
| |
Collapse
|
30
|
Zheng P, Wei X, Cao X, Ma P, Dong R, Tang H, Meng X, Liu X, Zhang C, Zhang S, Ming L. Antigen clearance at the peak of the primary immune response induces experimental autoimmune encephalomyelitis. Eur J Immunol 2023; 53:e2250122. [PMID: 36597350 DOI: 10.1002/eji.202250122] [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: 08/10/2022] [Revised: 12/02/2022] [Accepted: 01/03/2023] [Indexed: 01/05/2023]
Abstract
Autoimmune demyelinating diseases can be induced by an immune response against myelin peptides; however, the exact mechanism underlying the development of such diseases remains unclear. In experimental autoimmune encephalomyelitis, we found that the clearance of exogenous myelin antigen at the peak of the primary immune response is key to the pathogenesis of the disease. The generation of effector T cells requires continuous antigen stimulation, whereas redundant antigen traps and exhausts effector T cells in the periphery, which induces resistance to the disease. Moreover, insufficient antigenic stimulation fails to induce disease efficiently owing to insufficient numbers of effector T cells. When myelin antigen is entirely cleared, the number of effector T cells reaches a peak, which facilitates infiltration of more effector T cells into the central nervous system. The peripheral antigen clearance initiates the first wave of effector T cell entry into the central nervous system and induces chronic inflammation. The inflamed central nervous system recruits the second wave of effector T cells that worsen inflammation, resulting in loss of self-tolerance. These findings provide new insights into the mechanism underlying the development of autoimmune demyelinating diseases, which may potentially impact future treatments.
Collapse
Affiliation(s)
- Peiguo Zheng
- Department of Clinical Laboratory, The First Affiliated Hospital of Zhengzhou University, Key Clinical Laboratory of Henan Province, Zhengzhou, P. R. China
| | - Xufeng Wei
- Department of Clinical Laboratory, The First Affiliated Hospital of Zhengzhou University, Key Clinical Laboratory of Henan Province, Zhengzhou, P. R. China
| | - Xuezhen Cao
- Department of Clinical Laboratory, The First Affiliated Hospital of Zhengzhou University, Key Clinical Laboratory of Henan Province, Zhengzhou, P. R. China
| | - Panhong Ma
- Department of Clinical Laboratory, The First Affiliated Hospital of Zhengzhou University, Key Clinical Laboratory of Henan Province, Zhengzhou, P. R. China
| | - Rui Dong
- Department of Clinical Laboratory, The First Affiliated Hospital of Zhengzhou University, Key Clinical Laboratory of Henan Province, Zhengzhou, P. R. China
| | - Hongwei Tang
- Key Laboratory of Hepatobiliary and Pancreatic Surgery & Digestive Organ Transplantation, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, P. R. China
| | - Xianchun Meng
- Department of Clinical Laboratory, The First Affiliated Hospital of Zhengzhou University, Key Clinical Laboratory of Henan Province, Zhengzhou, P. R. China
| | - Xinjing Liu
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, P. R. China
| | - Cai Zhang
- Department of Clinical Laboratory, The First Affiliated Hospital of Zhengzhou University, Key Clinical Laboratory of Henan Province, Zhengzhou, P. R. China
| | - Shuijun Zhang
- Key Laboratory of Hepatobiliary and Pancreatic Surgery & Digestive Organ Transplantation, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, P. R. China
| | - Liang Ming
- Department of Clinical Laboratory, The First Affiliated Hospital of Zhengzhou University, Key Clinical Laboratory of Henan Province, Zhengzhou, P. R. China
| |
Collapse
|
31
|
Chen J, Guo W, Du P, Cui T, Yang Y, Wang Y, Kang P, Zhang Z, Wang Q, Ye Z, Liu L, Jian Z, Gao T, Bian H, Li S, Li C. MIF inhibition alleviates vitiligo progression by suppressing CD8 + T cell activation and proliferation. J Pathol 2023; 260:84-96. [PMID: 36852981 DOI: 10.1002/path.6073] [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: 10/03/2022] [Revised: 01/18/2023] [Accepted: 02/22/2023] [Indexed: 03/01/2023]
Abstract
In vitiligo, autoreactive CD8+ T cells have been established as the main culprit considering its pathogenic role in mediating epidermal melanocyte-specific destruction. Macrophage migration inhibitory factor (MIF) is a pleiotropic molecule that plays a central role in various immune processes including the activation and proliferation of T cells; but whether MIF is intertwined in vitiligo development and progression and its involvement in aberrantly activated CD8+ T cells remains ill-defined. In this study, we found that MIF was overabundant in vitiligo patients and a mouse model for human vitiligo. Additionally, inhibiting MIF ameliorated the disease progression in vitiligo mice, which manifested as less infiltration of CD8+ T cells and more retention of epidermal melanocytes in the tail skin. More importantly, in vitro experiments indicated that MIF-inhibition suppressed the activation and proliferation of CD8+ T cells from the lymph nodes of vitiligo mice, and the effect extended to CD8+ T cells in peripheral blood mononuclear cells of vitiligo patients. Finally, CD8+ T cells derived from MIF-inhibited vitiligo mice also exhibited an impaired capacity for activation and proliferation. Taken together, our results show that MIF might be clinically targetable in vitiligo treatment, and its inhibition might ameliorate vitiligo progression by suppressing autoreactive CD8+ T cell activation and proliferation. © 2023 The Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Jianru Chen
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, PR China
| | - Weinan Guo
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, PR China
| | - Pengran Du
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, PR China
| | - Tingting Cui
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, PR China
| | - Yuqi Yang
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, PR China
| | - Yinghan Wang
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, PR China
| | - Pan Kang
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, PR China
| | - Zhe Zhang
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, PR China
| | - Qi Wang
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, PR China
| | - Zhubiao Ye
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, PR China
| | - Ling Liu
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, PR China
| | - Zhe Jian
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, PR China
| | - Tianwen Gao
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, PR China
| | - Huijie Bian
- National Translational Science Center for Molecular Medicine and Department of Cell Biology, Fourth Military Medical University, Xi'an, PR China
| | - Shuli Li
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, PR China
| | - Chunying Li
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, PR China
| |
Collapse
|
32
|
Reynoso GV, Gordon DN, Kalia A, Aguilar CC, Malo CS, Aleshnick M, Dowd KA, Cherry CR, Shannon JP, Vrba SM, Holmes AC, Alippe Y, Maciejewski S, Asano K, Diamond MS, Pierson TC, Hickman HD. Zika virus spreads through infection of lymph node-resident macrophages. Cell Rep 2023; 42:112126. [PMID: 36795561 PMCID: PMC10425566 DOI: 10.1016/j.celrep.2023.112126] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Revised: 01/03/2023] [Accepted: 02/01/2023] [Indexed: 02/17/2023] Open
Abstract
To disseminate through the body, Zika virus (ZIKV) is thought to exploit the mobility of myeloid cells, in particular monocytes and dendritic cells. However, the timing and mechanisms underlying shuttling of the virus by immune cells remains unclear. To understand the early steps in ZIKV transit from the skin, at different time points, we spatially mapped ZIKV infection in lymph nodes (LNs), an intermediary site en route to the blood. Contrary to prevailing hypotheses, migratory immune cells are not required for the virus to reach the LNs or blood. Instead, ZIKV rapidly infects a subset of sessile CD169+ macrophages in the LNs, which release the virus to infect downstream LNs. Infection of CD169+ macrophages alone is sufficient to initiate viremia. Overall, our experiments indicate that macrophages that reside in the LNs contribute to initial ZIKV spread. These studies enhance our understanding of ZIKV dissemination and identify another anatomical site for potential antiviral intervention.
Collapse
Affiliation(s)
- Glennys V Reynoso
- Viral Immunity and Pathogenesis Unit, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD, USA
| | - David N Gordon
- Viral Pathogenesis Section, Laboratory of Viral Diseases (LVD), NIAID, NIH, Bethesda, MD, USA
| | - Anurag Kalia
- Viral Immunity and Pathogenesis Unit, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Cynthia C Aguilar
- Viral Immunity and Pathogenesis Unit, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Courtney S Malo
- Viral Immunity and Pathogenesis Unit, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Maya Aleshnick
- Viral Pathogenesis Section, Laboratory of Viral Diseases (LVD), NIAID, NIH, Bethesda, MD, USA
| | - Kimberly A Dowd
- Viral Pathogenesis Section, Laboratory of Viral Diseases (LVD), NIAID, NIH, Bethesda, MD, USA
| | - Christian R Cherry
- Viral Immunity and Pathogenesis Unit, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD, USA
| | - John P Shannon
- Viral Immunity and Pathogenesis Unit, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Sophia M Vrba
- Viral Immunity and Pathogenesis Unit, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Autumn C Holmes
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Yael Alippe
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Sonia Maciejewski
- Viral Pathogenesis Section, Laboratory of Viral Diseases (LVD), NIAID, NIH, Bethesda, MD, USA
| | - Kenichi Asano
- Laboratory of Immune Regulation, School of Life Science, Tokyo University of Pharmacy and Life Sciences, Tokyo 192-0392, Japan
| | - Michael S Diamond
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA; Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA; Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO, USA; The Andrew M. and Jane M. Bursky Center for Human Immunology and Immunotherapy Programs, Washington University School of Medicine, St. Louis, MO, USA
| | - Theodore C Pierson
- Viral Pathogenesis Section, Laboratory of Viral Diseases (LVD), NIAID, NIH, Bethesda, MD, USA
| | - Heather D Hickman
- Viral Immunity and Pathogenesis Unit, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD, USA.
| |
Collapse
|
33
|
Biswas L, Chen J, De Angelis J, Singh A, Owen-Woods C, Ding Z, Pujol JM, Kumar N, Zeng F, Ramasamy SK, Kusumbe AP. Lymphatic vessels in bone support regeneration after injury. Cell 2023; 186:382-397.e24. [PMID: 36669473 DOI: 10.1016/j.cell.2022.12.031] [Citation(s) in RCA: 76] [Impact Index Per Article: 76.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Revised: 10/05/2022] [Accepted: 12/19/2022] [Indexed: 01/20/2023]
Abstract
Blood and lymphatic vessels form a versatile transport network and provide inductive signals to regulate tissue-specific functions. Blood vessels in bone regulate osteogenesis and hematopoiesis, but current dogma suggests that bone lacks lymphatic vessels. Here, by combining high-resolution light-sheet imaging and cell-specific mouse genetics, we demonstrate presence of lymphatic vessels in mouse and human bones. We find that lymphatic vessels in bone expand during genotoxic stress. VEGF-C/VEGFR-3 signaling and genotoxic stress-induced IL6 drive lymphangiogenesis in bones. During lymphangiogenesis, secretion of CXCL12 from proliferating lymphatic endothelial cells is critical for hematopoietic and bone regeneration. Moreover, lymphangiocrine CXCL12 triggers expansion of mature Myh11+ CXCR4+ pericytes, which differentiate into bone cells and contribute to bone and hematopoietic regeneration. In aged animals, such expansion of lymphatic vessels and Myh11-positive cells in response to genotoxic stress is impaired. These data suggest lymphangiogenesis as a therapeutic avenue to stimulate hematopoietic and bone regeneration.
Collapse
Affiliation(s)
- Lincoln Biswas
- Tissue and Tumor Microenvironments Group, MRC Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine, Medical Sciences Division, University of Oxford, Oxford OX3 9DS, UK
| | - Junyu Chen
- Tissue and Tumor Microenvironments Group, MRC Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine, Medical Sciences Division, University of Oxford, Oxford OX3 9DS, UK; State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Jessica De Angelis
- Tissue and Tumor Microenvironments Group, MRC Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine, Medical Sciences Division, University of Oxford, Oxford OX3 9DS, UK
| | - Amit Singh
- Tissue and Tumor Microenvironments Group, MRC Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine, Medical Sciences Division, University of Oxford, Oxford OX3 9DS, UK; Heidelberg University Biochemistry Center, Im Neuenheimer Feld 328, Heidelberg D-69120, Germany
| | - Charlotte Owen-Woods
- Tissue and Tumor Microenvironments Group, MRC Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine, Medical Sciences Division, University of Oxford, Oxford OX3 9DS, UK
| | - Zhangfan Ding
- Tissue and Tumor Microenvironments Group, MRC Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine, Medical Sciences Division, University of Oxford, Oxford OX3 9DS, UK; State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Joan Mane Pujol
- Tissue and Tumor Microenvironments Group, MRC Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine, Medical Sciences Division, University of Oxford, Oxford OX3 9DS, UK
| | - Naveen Kumar
- Tissue and Tumor Microenvironments Group, MRC Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine, Medical Sciences Division, University of Oxford, Oxford OX3 9DS, UK
| | - Fanxin Zeng
- Department of Clinic Medical Center, Dazhou Central Hospital, Dazhou, China
| | - Saravana K Ramasamy
- MRC London Institute of Medical Sciences, Imperial College London, London W12 0NN, UK
| | - Anjali P Kusumbe
- Tissue and Tumor Microenvironments Group, MRC Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine, Medical Sciences Division, University of Oxford, Oxford OX3 9DS, UK.
| |
Collapse
|
34
|
Anatomy and pathology of lymphatic vessels under physiological and inflammatory conditions in the mouse diaphragm. Microvasc Res 2023; 145:104438. [PMID: 36122645 DOI: 10.1016/j.mvr.2022.104438] [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: 05/09/2022] [Revised: 08/28/2022] [Accepted: 09/13/2022] [Indexed: 02/03/2023]
Abstract
The lymphatic vessels in the parietal pleura drain fluids. Impaired drainage function and excessive fluid entry in the pleural cavity accumulate effusion. The rat diaphragmatic lymphatics drain fluids from the pleura to the muscle layer. Lymphatic subtypes are characterized by the major distribution of discontinuous button-like endothelial junctions (buttons) in initial lymphatics and continuous zipper-like junctions (zippers) in the collecting lymphatics. Inflammation replaced buttons with zippers in tracheal lymphatics. In the mouse diaphragm, the structural relationship between the lymphatics and blood vessels, the presence of lymphatics in the muscle layer, and the distributions of initial and collecting lymphatics are unclear. Moreover, the endothelial junctional alterations and effects of vascular endothelial growth factor receptor (VEGFR) inhibition under pleural inflammation are unclear. We subjected the whole-mount mouse diaphragms to immunohistochemistry. The lymphatics and blood vessels were distributed in different layers of the pleural membrane. Major lymphatic subtypes were initial lymphatics in the pleura and collecting lymphatics in the muscle layer. Chronic pleural inflammation disorganized the stratified layers of the lymphatics and blood vessels and replaced buttons with zippers in the pleural lymphatics, which impaired drainage function. VEGFR inhibition under inflammation maintained the vascular structures and drainage function. In addition, VEGFR inhibition maintained the lymphatic endothelial junctions and reduced the blood vessel permeability under inflammation. These findings may provide new targets for managing pleural effusions caused by inflammation, such as pleuritis and empyema, which are common pneumonia comorbidities.
Collapse
|
35
|
Huang P, Jiang L, Pan H, Ding L, Zhou B, Zhao M, Zou J, Li B, Qi M, Deng H, Zhou Y, Chen X. An Integrated Polymeric mRNA Vaccine without Inflammation Side Effects for Cellular Immunity Mediated Cancer Therapy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2023; 35:e2207471. [PMID: 36326183 DOI: 10.1002/adma.202207471] [Citation(s) in RCA: 25] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Revised: 10/22/2022] [Indexed: 06/16/2023]
Abstract
Among the few available mRNA delivery vehicles, lipid nanoparticles (LNPs) are the most clinically advanced but they require cumbersome four components and suffer from inflammation-related side effects that should be minimized for safety. Yet, a certain level of proinflammatory responses and innate immune activation are required to evoke T-cell immunity for mRNA cancer vaccination. To address these issues and develop potent yet low-inflammatory mRNA cancer vaccine vectors, a series of alternating copolymers "PHTA" featured with ortho-hydroxy tertiary amine (HTA) repeating units for mRNA delivery is synthesized, which can play triple roles of condensing mRNA, enhancing the polymeric nanoparticle (PNP) stability, and prolonging circulation time. Unlike LNPs exhibiting high levels of inflammation, the PHTA-based PNPs show negligible inflammatory side effects in vivo. Importantly, the top candidate PHTA-C18 enables successful mRNA cancer vaccine delivery in vivo and leads to a robust CD8+ T cell mediated antitumor cellular immunity. Such PHTA-based integrated PNP provides a potential approach for establishing mRNA cancer vaccines with good inflammatory safety profiles.
Collapse
Affiliation(s)
- Pei Huang
- School of Chemistry and Chemical Engineering, Frontiers Science Center for Transformative Molecules, Shanghai Jiao Tong University, Shanghai, 200240, China
- Departments of Diagnostic Radiology, Surgery, Chemical and Biomolecular Engineering, and Biomedical Engineering, Yong Loo Lin School of Medicine and Faculty of Engineering, National University of Singapore, Singapore, 119074, Singapore
| | - Lingsheng Jiang
- School of Chemistry and Chemical Engineering, Frontiers Science Center for Transformative Molecules, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Hui Pan
- School of Chemistry and Chemical Engineering, Frontiers Science Center for Transformative Molecules, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Lingwen Ding
- Nanomedicine Translational Research Program, NUS Center for Nanomedicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
- Department of Pathology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119074, Singapore
| | - Bo Zhou
- Departments of Diagnostic Radiology, Surgery, Chemical and Biomolecular Engineering, and Biomedical Engineering, Yong Loo Lin School of Medicine and Faculty of Engineering, National University of Singapore, Singapore, 119074, Singapore
| | - Mengyao Zhao
- Departments of Diagnostic Radiology, Surgery, Chemical and Biomolecular Engineering, and Biomedical Engineering, Yong Loo Lin School of Medicine and Faculty of Engineering, National University of Singapore, Singapore, 119074, Singapore
| | - Jianhua Zou
- Departments of Diagnostic Radiology, Surgery, Chemical and Biomolecular Engineering, and Biomedical Engineering, Yong Loo Lin School of Medicine and Faculty of Engineering, National University of Singapore, Singapore, 119074, Singapore
| | - Benhao Li
- Departments of Diagnostic Radiology, Surgery, Chemical and Biomolecular Engineering, and Biomedical Engineering, Yong Loo Lin School of Medicine and Faculty of Engineering, National University of Singapore, Singapore, 119074, Singapore
| | - Meiwei Qi
- School of Chemistry and Chemical Engineering, Frontiers Science Center for Transformative Molecules, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Hongzhang Deng
- Departments of Diagnostic Radiology, Surgery, Chemical and Biomolecular Engineering, and Biomedical Engineering, Yong Loo Lin School of Medicine and Faculty of Engineering, National University of Singapore, Singapore, 119074, Singapore
| | - Yongfeng Zhou
- School of Chemistry and Chemical Engineering, Frontiers Science Center for Transformative Molecules, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Xiaoyuan Chen
- Departments of Diagnostic Radiology, Surgery, Chemical and Biomolecular Engineering, and Biomedical Engineering, Yong Loo Lin School of Medicine and Faculty of Engineering, National University of Singapore, Singapore, 119074, Singapore
- Nanomedicine Translational Research Program, NUS Center for Nanomedicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
- Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117599, Singapore
- Institute of Molecular and Cell Biology, Agency for Science, Technology, and Research (A*STAR), 61 Biopolis Drive, Proteos, Singapore, 138673, Singapore
| |
Collapse
|
36
|
Dai T, Rosario SR, Katsuta E, Dessai AS, Paterson EJ, Novickis AT, Cortes Gomez E, Zhu B, Liu S, Wang H, Abrams SI, Seshadri M, Bshara W, Dasgupta S. Hypoxic activation of PFKFB4 in breast tumor microenvironment shapes metabolic and cellular plasticity to accentuate metastatic competence. Cell Rep 2022; 41:111756. [PMID: 36476868 PMCID: PMC9807018 DOI: 10.1016/j.celrep.2022.111756] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Revised: 09/27/2022] [Accepted: 11/08/2022] [Indexed: 12/12/2022] Open
Abstract
Cancer cells encounter a hostile tumor microenvironment (TME), and their adaptations to metabolic stresses determine metastatic competence. Here, we show that the metabolic enzyme 6-phosphofructo-2-kinase/fructose-2,6-biphosphatase-4 (PFKFB4) is induced in hypoxic tumors acquiring metabolic plasticity and invasive phenotype. In mouse models of breast cancer, genetic ablation of PFKFB4 significantly delays distant organ metastasis, reducing local lymph node invasion by suppressing expression of invasive gene signature including integrin β3. Photoacoustic imaging followed by metabolomics analyses of hypoxic tumors show that PFKFB4 drives metabolic flexibility, enabling rapid detoxification of reactive oxygen species favoring survival under selective pressure. Mechanistically, hypoxic induction triggers nuclear translocation of PFKFB4 accentuating non-canonical transcriptional activation of HIF-1α, and breast cancer patients with increased nuclear PFKFB4 in their tumors are found to be significantly associated with poor prognosis. Our findings imply that PFKFB4 induction is crucial for tumor cell adaptation in the hypoxic TME that determines metastatic competence.
Collapse
Affiliation(s)
- Tao Dai
- Department of Cell Stress Biology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
| | - Spencer R. Rosario
- Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
| | - Eriko Katsuta
- Department of Cell Stress Biology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
| | - Abhisha Sawant Dessai
- Department of Cell Stress Biology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
| | - Emily J. Paterson
- Department of Cell Stress Biology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
| | - Aaron T. Novickis
- Department of Cell Stress Biology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
| | - Eduardo Cortes Gomez
- Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
| | - Bokai Zhu
- Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219, USA
| | - Song Liu
- Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
| | - Hai Wang
- Department of Molecular and Cellular Biology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
| | - Scott I. Abrams
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
| | - Mukund Seshadri
- Department of Oral Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
| | - Wiam Bshara
- Department of Pathology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
| | - Subhamoy Dasgupta
- Department of Cell Stress Biology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA,Lead contact,Correspondence:
| |
Collapse
|
37
|
Li Y, Li Y, Xu S, Chen Y, Zhou P, Hu T, Li H, Liu Y, Xu Y, Ren J, Qiu Y, Lu C. N-Acylethanolamine acid amidase (NAAA) exacerbates psoriasis inflammation by enhancing dendritic cell (DCs) maturation. Pharmacol Res 2022; 185:106491. [DOI: 10.1016/j.phrs.2022.106491] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Revised: 09/13/2022] [Accepted: 10/05/2022] [Indexed: 11/30/2022]
|
38
|
Macromolecular assembly of bioluminescent protein nanoparticles for enhanced imaging. Mater Today Bio 2022; 17:100455. [PMID: 36304975 PMCID: PMC9593766 DOI: 10.1016/j.mtbio.2022.100455] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Revised: 10/05/2022] [Accepted: 10/07/2022] [Indexed: 11/11/2022] Open
Abstract
Bioluminescence imaging has advantages over fluorescence imaging, such as minimal photobleaching and autofluorescence, and greater signal-to-noise ratios in many complex environments. Although significant achievements have been made in luciferase engineering for generating bright and stable reporters, the full capability of luciferases for nanoparticle tracking has not been comprehensively examined. In biocatalysis, enhanced enzyme performance after immobilization on nanoparticles has been reported. Thus, we hypothesized that by assembling luciferases onto a nanoparticle, the resulting complex could lead to substantially improved imaging properties. Using a modular bioconjugation strategy, we attached NanoLuc (NLuc) or Akaluc bioluminescent proteins to a protein nanoparticle platform (E2), yielding nanoparticles NLuc-E2 and Akaluc-E2, both with diameters of ∼45 nm. Although no significant differences were observed between different conditions involving Akaluc and Akaluc-E2, free NLuc at pH 5.0 showed significantly lower emission values than free NLuc at pH 7.4. Interestingly, NLuc immobilization on E2 nanoparticles (NLuc-E2) emitted increased luminescence at pH 7.4, and at pH 5.0 showed over two orders of magnitude (>200-fold) higher luminescence (than free NLuc), expanding the potential for imaging detection using the nanoparticle even upon endocytic uptake. After uptake by macrophages, the resulting luminescence with NLuc-E2 nanoparticles was up to 7-fold higher than with free NLuc at 48 h. Cells incubated with NLuc-E2 could also be imaged using live bioluminescence microscopy. Finally, biodistribution of nanoparticles into lymph nodes was detected through imaging using NLuc-E2, but not with conventionally-labeled fluorescent E2. Our data demonstrate that NLuc-bound nanoparticles have advantageous properties that can be utilized in applications ranging from single-cell imaging to in vivo biodistribution.
Collapse
|
39
|
O'Melia MJ, Rohner NA, Thomas SN. Tumor Vascular Remodeling Affects Molecular Dissemination to Lymph Node and Systemic Leukocytes. Tissue Eng Part A 2022; 28:781-794. [PMID: 35442085 PMCID: PMC9508451 DOI: 10.1089/ten.tea.2022.0020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Accepted: 04/13/2022] [Indexed: 11/13/2022] Open
Abstract
Angiogenic and lymphangiogenic remodeling has long been accepted as a hallmark of cancer development and progression; however, the impacts of this remodeling on immunological responses, which are paramount to the responses to immunotherapeutic treatments, are underexplored. As immunotherapies represent one of the most promising new classes of cancer therapy, in this study, we explore the effects of angiogenic and lymphangiogenic normalization on dissemination of molecules injected into the tumor microenvironment to immune cells in lymph nodes draining the tumor as well as in systemically distributed tissues. A system of fluorescent tracers, size-matched to biomolecules of interest, was implemented to track different mechanisms of tumor transport and access to immune cells. This revealed that the presence of a tumor, and either angiogenic or lymphangiogenic remodeling, altered local retention of model biomolecules, trended toward normalizing dissemination to systemic organs, and modified access to lymph node-resident immune cells in manners dependent on mechanism of transport. More specifically, active cell migration by skin-derived antigen presenting cells was enhanced by both the presence of a tumor and lymphangiogenic normalization, while both angiogenic and lymphangiogenic normalization restored patterns of immune cell access to passively draining species. As a whole, this work uncovers the potential ramifications of tumor-induced angiogenesis and lymphangiogenesis, along with impacts of interrogation into these pathways, on access of tumor-derived species to immune cells. Impact Statement Angiogenic and lymphangiogenic normalization strategies have been utilized clinically to interrogate tumor vasculature with some success. In the age of immunotherapy, the impacts of these therapeutic interventions on immune remodeling are unclear. This work utilizes mouse models of angiogenic and lymphangiogenic normalization, along with a system of fluorescently tagged tracers, to uncover the impacts of angiogenesis and lymphangiogenesis on access of tumor-derived species to immune cell subsets within various organs.
Collapse
Affiliation(s)
- Meghan J. O'Melia
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, Georgia, USA
| | - Nathan A. Rohner
- George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, Georgia, USA
- Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, Georgia, USA
| | - Susan Napier Thomas
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, Georgia, USA
- George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, Georgia, USA
- Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, Georgia, USA
- Winship Cancer Institute, Emory University School of Medicine, Atlanta, Georgia, USA
| |
Collapse
|
40
|
Li Z, He P, Xu Y, Deng Y, Gao Y, Chen SL. In vivo evaluation of a lipopolysaccharide-induced ear vascular leakage model in mice using photoacoustic microscopy. BIOMEDICAL OPTICS EXPRESS 2022; 13:4802-4816. [PMID: 36187238 PMCID: PMC9484413 DOI: 10.1364/boe.471079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Revised: 08/05/2022] [Accepted: 08/05/2022] [Indexed: 06/16/2023]
Abstract
Sepsis is caused by dysregulated host inflammatory response to infection. During sepsis, early identification and monitoring of vascular leakage are pivotal for improved diagnosis, treatment, and prognosis. However, there is a lack of research on noninvasive observation of inflammation-related vascular leakage. Here, we investigate the use of photoacoustic microscopy (PAM) for in vivo visualization of lipopolysaccharide (LPS)-induced ear vascular leakage in mice using Evans blue (EB) as an indicator. A model combining needle pricking on the mouse ear, topical smearing of LPS on the mouse ear, and intravenous tail injection of EB is developed. Topical application of LPS is expected to induce local vascular leakage in skin. Inflammatory response is first validated by ex vivo histology and enzyme-linked immunosorbent assay. Then, local ear vascular leakage is confirmed by ex vivo measurement of swelling, thickening, and EB leakage. Finally, PAM for in vivo identification and evaluation of early vascular leakage using the model is demonstrated. For PAM, common excitation wavelength of 532 nm is used, and an algorithm is developed to extract quantitative metrics for EB leakage. The results show potential of PAM for noninvasive longitudinal monitoring of peripheral skin vascular leakage, which holds promise for clinical sepsis diagnosis and management.
Collapse
Affiliation(s)
- Zhe Li
- Department of Critical Care Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
- These authors contributed equally to this work
| | - Pengbo He
- University of Michigan-Shanghai Jiao Tong University Joint Institute, Shanghai Jiao Tong University, Shanghai 200240, China
- These authors contributed equally to this work
| | - Yuqing Xu
- Department of Critical Care Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Yuxiao Deng
- Department of Critical Care Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Yuan Gao
- Department of Critical Care Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Sung-Liang Chen
- University of Michigan-Shanghai Jiao Tong University Joint Institute, Shanghai Jiao Tong University, Shanghai 200240, China
- Engineering Research Center of Digital Medicine and Clinical Translation, Ministry of Education, Shanghai 200030, China
- State Key Laboratory of Advanced Optical Communication Systems and Networks, Shanghai Jiao Tong University, Shanghai 200240, China
| |
Collapse
|
41
|
Jha B, Reverte M, Ronet C, Prevel F, Morgenthaler FD, Desponds C, Lye LF, Owens KL, Scarpellino L, Dubey LK, Sabine A, Petrova TV, Luther SA, Beverley SM, Fasel N. In and out: Leishmania metastasis by hijacking lymphatic system and migrating immune cells. Front Cell Infect Microbiol 2022; 12:941860. [PMID: 36034709 PMCID: PMC9414205 DOI: 10.3389/fcimb.2022.941860] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Accepted: 07/19/2022] [Indexed: 11/22/2022] Open
Abstract
The lymphatic system plays a crucial role in mounting immune response against intracellular pathogens, and recent studies have documented its role in facilitating tumor dissemination linked largely with cancer cells. However, in mucocutaneous leishmaniasis (MCL) caused by Leishmania Viannia subgenus showing infectious metastasis and resulting in severe distant secondary lesions, the route of escape of these parasites to secondary sites has not yet been investigated in detail. Our results demonstrated that when infection was associated with inflammation and additionally exacerbated by the presence of dsRNA viral endosymbiont (LRV1), lymphatic vessels could serve as efficient routes for infected cells to egress from the primary site and colonize distant organs. We challenged this hypothesis by using the intracellular Leishmania protozoan parasites Leishmania guyanensis (Lgy) associated with or without a dsRNA viral endosymbiont, exacerbating the infection and responsible for a strong inflammatory response, and favoring metastasis of the infection. We analyzed possible cargo cells and the routes of dissemination through flow cytometry, histological analysis, and in vivo imaging in our metastatic model to show that parasites disseminated not only intracellularly but also as free extracellular parasites using migrating immune cells, lymph nodes (LNs), and lymph vessels, and followed intricate connections of draining and non-draining lymph node to finally end up in the blood and in distant skin, causing new lesions.
Collapse
Affiliation(s)
- Baijayanti Jha
- Department of Immunobiology, University of Lausanne, Epalinges, Switzerland
| | - Marta Reverte
- Department of Immunobiology, University of Lausanne, Epalinges, Switzerland
| | - Catherine Ronet
- Department of Immunobiology, University of Lausanne, Epalinges, Switzerland
- Department of Oncology, Ludwig Institute for Cancer Research Lausanne, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Florence Prevel
- Department of Immunobiology, University of Lausanne, Epalinges, Switzerland
| | | | - Chantal Desponds
- Department of Immunobiology, University of Lausanne, Epalinges, Switzerland
| | - Lon-Fye Lye
- Department of Molecular Microbiology, School of Medicine, Washington University, St. Louis, MO, United States
| | - Katherine L. Owens
- Department of Molecular Microbiology, School of Medicine, Washington University, St. Louis, MO, United States
| | | | - Lalit Kumar Dubey
- Department of Immunobiology, University of Lausanne, Epalinges, Switzerland
- Centre for Microvascular Research, John Vane Science Centre, Queen Mary University of London, London, United Kingdom
| | - Amélie Sabine
- Department of Oncology and Ludwig Institute for Cancer Research, University of Lausanne and Centre Hospitalier Universitaire Vaudois, Epalinges, Switzerland
| | - Tatiana V. Petrova
- Department of Oncology and Ludwig Institute for Cancer Research, University of Lausanne and Centre Hospitalier Universitaire Vaudois, Epalinges, Switzerland
| | - Sanjiv A. Luther
- Department of Immunobiology, University of Lausanne, Epalinges, Switzerland
| | - Stephen M. Beverley
- Department of Molecular Microbiology, School of Medicine, Washington University, St. Louis, MO, United States
| | - Nicolas Fasel
- Department of Immunobiology, University of Lausanne, Epalinges, Switzerland
- *Correspondence: Nicolas Fasel,
| |
Collapse
|
42
|
Li Q, Teng Z, Tao J, Shi W, Yang G, Zhang Y, Su X, Chen L, Xiu W, Yuwen L, Dong H, Mou Y. Elastic Nanovaccine Enhances Dendritic Cell-Mediated Tumor Immunotherapy. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2022; 18:e2201108. [PMID: 35734820 DOI: 10.1002/smll.202201108] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/19/2022] [Revised: 05/12/2022] [Indexed: 06/15/2023]
Abstract
Nanovaccine-based immunotherapy (NBI) has the ability to initiate dendritic cell (DC)-mediated tumor-specific immune responses and maintain long-term antitumor immune memory. To date, the mechanism by which the mechanical properties of nanoparticles alter the functions of DCs in NBI remains largely unclear. Here, a soft mesoporous organosilica-based nanovaccine (SMONV) is prepared and the elasticity-dependent effect of the nanovaccine on the underlying DC-mediated immune responses is studied. It is found that the elasticity results in greater internalization of SMONV by DCs, followed by the induction of substantial cytosolic delivery of antigens via endosomal escape, leading to effective DC maturation and antigen cross-presentation. Impressively, elasticity enables SMONV to enhance lymphatic drainage of antigens in vivo, thus stimulating robust humoral and cellular immunity. The results from therapeutic tumor vaccination further reveal that subcutaneously administered SMONV effectively suppresses tumor growth in tumor-bearing mice by evoking antigen-specific CD8+ T-cell immune responses, mitigating regulatory T-cell-mediated immunosuppression, and increasing central memory and effector memory T-cell populations. Furthermore, combinatorial immunization with SMONV and anti-PD-L1 blocking antibodies results in an amplified therapeutic effect on tumor-bearing mice. These findings reveal the elastic effect of the nanovaccine on DC-mediated immune responses, and the prepared SMONV represents a facile and powerful strategy for antitumor immunotherapy.
Collapse
Affiliation(s)
- Qiang Li
- Nanjing Stomatological Hospital, Medical School of Nanjing University, 30 Zhongyang Road, Nanjing, Jiangsu, 210008, P. R. China
| | - Zhaogang Teng
- Key Laboratory for Organic Electronics and Information Displays, Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials, Jiangsu National Synergetic Innovation Centre for Advanced Materials, Nanjing University of Posts and Telecommunications, 9 Wenyuan Road, Nanjing, Jiangsu, 210023, P. R. China
| | - Jun Tao
- Key Laboratory for Organic Electronics and Information Displays, Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials, Jiangsu National Synergetic Innovation Centre for Advanced Materials, Nanjing University of Posts and Telecommunications, 9 Wenyuan Road, Nanjing, Jiangsu, 210023, P. R. China
| | - Wenhui Shi
- Key Laboratory for Organic Electronics and Information Displays, Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials, Jiangsu National Synergetic Innovation Centre for Advanced Materials, Nanjing University of Posts and Telecommunications, 9 Wenyuan Road, Nanjing, Jiangsu, 210023, P. R. China
| | - Guangwen Yang
- Nanjing Stomatological Hospital, Medical School of Nanjing University, 30 Zhongyang Road, Nanjing, Jiangsu, 210008, P. R. China
| | - Yu Zhang
- Nanjing Stomatological Hospital, Medical School of Nanjing University, 30 Zhongyang Road, Nanjing, Jiangsu, 210008, P. R. China
| | - Xiaodan Su
- Key Laboratory for Organic Electronics and Information Displays, Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials, Jiangsu National Synergetic Innovation Centre for Advanced Materials, Nanjing University of Posts and Telecommunications, 9 Wenyuan Road, Nanjing, Jiangsu, 210023, P. R. China
| | - Lin Chen
- Nanjing Stomatological Hospital, Medical School of Nanjing University, 30 Zhongyang Road, Nanjing, Jiangsu, 210008, P. R. China
| | - Weijun Xiu
- Key Laboratory for Organic Electronics and Information Displays, Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials, Jiangsu National Synergetic Innovation Centre for Advanced Materials, Nanjing University of Posts and Telecommunications, 9 Wenyuan Road, Nanjing, Jiangsu, 210023, P. R. China
| | - Lihui Yuwen
- Key Laboratory for Organic Electronics and Information Displays, Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials, Jiangsu National Synergetic Innovation Centre for Advanced Materials, Nanjing University of Posts and Telecommunications, 9 Wenyuan Road, Nanjing, Jiangsu, 210023, P. R. China
| | - Heng Dong
- Nanjing Stomatological Hospital, Medical School of Nanjing University, 30 Zhongyang Road, Nanjing, Jiangsu, 210008, P. R. China
| | - Yongbin Mou
- Nanjing Stomatological Hospital, Medical School of Nanjing University, 30 Zhongyang Road, Nanjing, Jiangsu, 210008, P. R. China
| |
Collapse
|
43
|
Saddawi-Konefka R, O'Farrell A, Faraji F, Clubb L, Allevato MM, Jensen SM, Yung BS, Wang Z, Wu VH, Anang NA, Msari RA, Schokrpur S, Pietryga IF, Molinolo AA, Mesirov JP, Simon AB, Fox BA, Bui JD, Sharabi A, Cohen EEW, Califano JA, Gutkind JS. Lymphatic-preserving treatment sequencing with immune checkpoint inhibition unleashes cDC1-dependent antitumor immunity in HNSCC. Nat Commun 2022; 13:4298. [PMID: 35879302 PMCID: PMC9314425 DOI: 10.1038/s41467-022-31941-w] [Citation(s) in RCA: 63] [Impact Index Per Article: 31.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Accepted: 07/11/2022] [Indexed: 12/30/2022] Open
Abstract
Despite the promise of immune checkpoint inhibition (ICI), therapeutic responses remain limited. This raises the possibility that standard of care treatments delivered in concert may compromise the tumor response. To address this, we employ tobacco-signature head and neck squamous cell carcinoma murine models in which we map tumor-draining lymphatics and develop models for regional lymphablation with surgery or radiation. We find that lymphablation eliminates the tumor ICI response, worsening overall survival and repolarizing the tumor- and peripheral-immune compartments. Mechanistically, within tumor-draining lymphatics, we observe an upregulation of conventional type I dendritic cells and type I interferon signaling and show that both are necessary for the ICI response and lost with lymphablation. Ultimately, we provide a mechanistic understanding of how standard oncologic therapies targeting regional lymphatics impact the tumor response to immune-oncology therapy in order to define rational, lymphatic-preserving treatment sequences that mobilize systemic antitumor immunity, achieve optimal tumor responses, control regional metastatic disease, and confer durable antitumor immunity.
Collapse
Affiliation(s)
- Robert Saddawi-Konefka
- Department of Otolaryngology-Head and Neck Surgery, UC San Diego School of Medicine, San Diego, CA, USA.
- Moores Cancer Center, UC San Diego, La Jolla, CA, USA.
- Gleiberman Head and Neck Cancer Center, UC San Diego, La Jolla, CA, USA.
| | - Aoife O'Farrell
- Moores Cancer Center, UC San Diego, La Jolla, CA, USA
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
| | - Farhoud Faraji
- Department of Otolaryngology-Head and Neck Surgery, UC San Diego School of Medicine, San Diego, CA, USA
- Moores Cancer Center, UC San Diego, La Jolla, CA, USA
- Gleiberman Head and Neck Cancer Center, UC San Diego, La Jolla, CA, USA
| | - Lauren Clubb
- Moores Cancer Center, UC San Diego, La Jolla, CA, USA
| | | | - Shawn M Jensen
- Earle A Chiles Research Institute, Robert W Franz Cancer Research Center, Providence Portland Medical Center, Portland, OR, USA
| | - Bryan S Yung
- Moores Cancer Center, UC San Diego, La Jolla, CA, USA
| | - Zhiyong Wang
- Moores Cancer Center, UC San Diego, La Jolla, CA, USA
| | - Victoria H Wu
- Moores Cancer Center, UC San Diego, La Jolla, CA, USA
| | | | | | - Shiruyeh Schokrpur
- Moores Cancer Center, UC San Diego, La Jolla, CA, USA
- Gleiberman Head and Neck Cancer Center, UC San Diego, La Jolla, CA, USA
- Department of Medicine, Division of Hematology-Oncology, UC San Diego School of Medicine, San Diego, CA, USA
| | | | | | - Jill P Mesirov
- Moores Cancer Center, UC San Diego, La Jolla, CA, USA
- Department of Medicine, UC San Diego School of Medicine, La Jolla, CA, USA
| | - Aaron B Simon
- Moores Cancer Center, UC San Diego, La Jolla, CA, USA
- Department of Radiation Oncology, UC Irvine School of Medicine, Irvine, CA, USA
| | - Bernard A Fox
- Earle A Chiles Research Institute, Robert W Franz Cancer Research Center, Providence Portland Medical Center, Portland, OR, USA
- Department of Molecular Microbiology and Immunology, Oregon Health Science University, Portland, OR, USA
| | - Jack D Bui
- Moores Cancer Center, UC San Diego, La Jolla, CA, USA
- Department of Pathology, UC San Diego School of Medicine, La Jolla, CA, USA
| | - Andrew Sharabi
- Moores Cancer Center, UC San Diego, La Jolla, CA, USA
- Gleiberman Head and Neck Cancer Center, UC San Diego, La Jolla, CA, USA
- Department of Radiation Medicine and Applied Sciences, UC San Diego School of Medicine, San Diego, CA, USA
| | - Ezra E W Cohen
- Moores Cancer Center, UC San Diego, La Jolla, CA, USA
- Gleiberman Head and Neck Cancer Center, UC San Diego, La Jolla, CA, USA
- Department of Medicine, Division of Hematology-Oncology, UC San Diego School of Medicine, San Diego, CA, USA
| | - Joseph A Califano
- Department of Otolaryngology-Head and Neck Surgery, UC San Diego School of Medicine, San Diego, CA, USA
- Moores Cancer Center, UC San Diego, La Jolla, CA, USA
- Gleiberman Head and Neck Cancer Center, UC San Diego, La Jolla, CA, USA
| | - J Silvio Gutkind
- Moores Cancer Center, UC San Diego, La Jolla, CA, USA.
- Gleiberman Head and Neck Cancer Center, UC San Diego, La Jolla, CA, USA.
- Department of Pharmacology, UC San Diego, La Jolla, CA, USA.
| |
Collapse
|
44
|
Abstract
The traditional method for generating polyclonal and monoclonal antibodies requires the immunization of an animal. Selecting the best species of animal and getting that animal's immune system to respond to a target antigen with an antibody response are essential to obtaining good-quality antibodies and hybridomas. There are only a limited number of opportunities for a researcher to intervene to manipulate and tailor the response to a particular antigen. Here we present advice and methods for designing the way in which the antigen is presented to the immune system (i.e., the immunization protocol), including the choice of animal, the antigen dose, the use of adjuvants, the route and number of injections, and the period between injections.
Collapse
|
45
|
Ozawa M, Nakajima S, Kobayashi D, Tomii K, Li NJ, Watarai T, Suzuki R, Watanabe S, Kanda Y, Takeuchi A, Katakai T. Micro- and Macro-Anatomical Frameworks of Lymph Nodes Indispensable for the Lymphatic System Filtering Function. Front Cell Dev Biol 2022; 10:902601. [PMID: 35794860 PMCID: PMC9251010 DOI: 10.3389/fcell.2022.902601] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Accepted: 05/19/2022] [Indexed: 01/01/2023] Open
Abstract
In the lymphatic vascular system, lymph nodes (LNs) play a pivotal role in filtering and removing lymph-borne substances. The filtering function of LNs involves resident macrophages tightly associated with unique lymphatic sinus structures. Moreover, an intermittently arranged LN in the lymphatic pathway is considered to cooperatively prevent lymph-borne substances from entering blood circulation. However, the functional significance of tissue microarchitecture, cellular composition, and individual LNs in the “LN chain” system is not fully understood. To explore the mechanistic and histo-anatomical significance of LNs as lymph fluid filters, we subcutaneously injected fluorescent tracers into mice and examined the details of lymphatic transport to the LNs qualitatively and quantitatively. Lymph-borne tracers were selectively accumulated in the MARCO+ subcapsular-medullary sinus border (SMB) region of the LN, in which reticular lymphatic endothelial cells and CD169+F4/80+ medullary sinus macrophages construct a dense meshwork of the physical barrier, forming the main body to capture the tracers. We also demonstrated stepwise filtration via the LN chain in the lymphatic basin, which prevented tracer leakage into the blood. Furthermore, inflammatory responses that induce the remodeling of LN tissue as well as the lymphatic pathway reinforce the overall filtering capacity of the lymphatic basin. Taken together, specialized tissue infrastructure in the LNs and their systematic orchestration constitute an integrated filtering system for lymphatic recirculation.
Collapse
Affiliation(s)
- Madoka Ozawa
- Department of Immunology, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Shihori Nakajima
- Department of Immunology, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Daichi Kobayashi
- Department of Immunology, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Koichi Tomii
- Department of Immunology, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Nan-Jun Li
- Department of Immunology, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Tomoya Watarai
- Department of Immunology, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Ryo Suzuki
- Department of Respiratory Medicine and Infectious Diseases, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Satoshi Watanabe
- Department of Respiratory Medicine and Infectious Diseases, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Yasuhiro Kanda
- Department of Immunology, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Arata Takeuchi
- Department of Immunology, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
- Department of Immunology, Tokyo Medical University, Tokyo, Japan
| | - Tomoya Katakai
- Department of Immunology, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
- *Correspondence: Tomoya Katakai,
| |
Collapse
|
46
|
Blair TC, Bambina S, Kramer GF, Dowdell AK, Alice AF, Baird JR, Lund AW, Piening BD, Crittenden MR, Gough MJ. Fluorescent tracking identifies key migratory dendritic cells in the lymph node after radiotherapy. Life Sci Alliance 2022; 5:5/9/e202101337. [PMID: 35487695 PMCID: PMC9058260 DOI: 10.26508/lsa.202101337] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 04/19/2022] [Accepted: 04/20/2022] [Indexed: 11/24/2022] Open
Abstract
Radiation therapy impacts all cells within the treatment field. Using novel technology, we track dendritic cells from the tumor to lymph nodes and demonstrate their importance in immune control of tumors. Radiation therapy generates extensive cancer cell death capable of promoting tumor-specific immunity. Within the tumor, conventional dendritic cells (cDCs) are known to carry tumor-associated antigens to the draining lymph node (TdLN) where they initiate T-cell priming. How radiation influences cDC migration is poorly understood. Here, we show that immunological efficacy of radiation therapy is dependent on cDC migration in radioimmunogenic tumors. Using photoconvertible mice, we demonstrate that radiation impairs cDC migration to the TdLN in poorly radioimmunogenic tumors. Comparative transcriptional analysis revealed that cDCs in radioimmunogenic tumors express genes associated with activation of endogenous adjuvant signaling pathways when compared with poorly radioimmunogenic tumors. Moreover, an exogenous adjuvant combined with radiation increased the number of migrating cDCs in these poorly radioimmunogenic tumors. Taken together, our data demonstrate that cDC migration play a critical role in the response to radiation therapy.
Collapse
Affiliation(s)
- Tiffany C Blair
- Earle A Chiles Research Institute, Robert W Franz Cancer Center, Providence Portland Medical Center, Portland, OR, USA
| | - Shelly Bambina
- Earle A Chiles Research Institute, Robert W Franz Cancer Center, Providence Portland Medical Center, Portland, OR, USA
| | - Gwen F Kramer
- Earle A Chiles Research Institute, Robert W Franz Cancer Center, Providence Portland Medical Center, Portland, OR, USA
| | - Alexa K Dowdell
- Earle A Chiles Research Institute, Robert W Franz Cancer Center, Providence Portland Medical Center, Portland, OR, USA
| | - Alejandro F Alice
- Earle A Chiles Research Institute, Robert W Franz Cancer Center, Providence Portland Medical Center, Portland, OR, USA
| | - Jason R Baird
- Earle A Chiles Research Institute, Robert W Franz Cancer Center, Providence Portland Medical Center, Portland, OR, USA
| | - Amanda W Lund
- Ronald O Perelman Department of Dermatology, Department of Pathology, NYU Grossman School of Medicine, New York, NY, USA
| | - Brian D Piening
- Earle A Chiles Research Institute, Robert W Franz Cancer Center, Providence Portland Medical Center, Portland, OR, USA
| | - Marka R Crittenden
- Earle A Chiles Research Institute, Robert W Franz Cancer Center, Providence Portland Medical Center, Portland, OR, USA.,The Oregon Clinic, Portland, OR, USA
| | - Michael J Gough
- Earle A Chiles Research Institute, Robert W Franz Cancer Center, Providence Portland Medical Center, Portland, OR, USA
| |
Collapse
|
47
|
Sonar SA, Uhrlaub JL, Coplen CP, Sempowski GD, Dudakov JA, van den Brink MRM, LaFleur BJ, Jergović M, Nikolich-Žugich J. Early age-related atrophy of cutaneous lymph nodes precipitates an early functional decline in skin immunity in mice with aging. Proc Natl Acad Sci U S A 2022; 119:e2121028119. [PMID: 35439062 PMCID: PMC9169949 DOI: 10.1073/pnas.2121028119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Accepted: 03/09/2022] [Indexed: 11/18/2022] Open
Abstract
Secondary lymphoid organs (SLOs) (including the spleen and lymph nodes [LNs]) are critical both for the maintenance of naive T (TN) lymphocytes and for the initiation and coordination of immune responses. How they age, including the exact timing, extent, physiological relevance, and the nature of age-related changes, remains incompletely understood. We used “time stamping” to indelibly mark newly generated naive T cells (also known as recent thymic emigrants) (RTEs) in mice, and followed their presence, phenotype, and retention in SLOs. We found that SLOs involute asynchronously. Skin-draining LNs atrophied by 6 to 9 mo in life, whereas deeper tissue-draining LNs atrophied by 18 to 20 mo, as measured by the loss of both TN numbers and the fibroblastic reticular cell (FRC) network. Time-stamped RTEs at all ages entered SLOs and successfully completed postthymic differentiation, but the capacity of older SLOs to maintain TN numbers was reduced with aging, and that trait did not depend on the age of TNs. However, in SLOs of older mice, these cells exhibited an emigration phenotype (CCR7loS1P1hi), which correlated with an increase of the cells of the same phenotype in the blood. Finally, upon intradermal immunization, RTEs generated in mice barely participated in de novo immune responses and failed to produce well-armed effector cells detectable in blood as early as by 7 to 8 mo of age. These results highlight changes in structure and function of superficial secondary lymphoid organs in laboratory mice that are earlier than expected and are consistent with the long-appreciated reduction of cutaneous immunity with aging.
Collapse
Affiliation(s)
- Sandip Ashok Sonar
- Department of Immunobiology, University of Arizona College of Medicine-Tucson, Tucson, AZ 85724
- Arizona Center on Aging, University of Arizona College of Medicine-Tucson, Tucson, AZ 85724
| | - Jennifer L. Uhrlaub
- Department of Immunobiology, University of Arizona College of Medicine-Tucson, Tucson, AZ 85724
- Arizona Center on Aging, University of Arizona College of Medicine-Tucson, Tucson, AZ 85724
| | - Christopher P. Coplen
- Department of Immunobiology, University of Arizona College of Medicine-Tucson, Tucson, AZ 85724
- Arizona Center on Aging, University of Arizona College of Medicine-Tucson, Tucson, AZ 85724
| | | | - Jarrod A. Dudakov
- Program in Immunology, Fred Hutchinson Cancer Center, Department of Immunology, University of Washington, Seattle, WA 98109
| | | | | | - Mladen Jergović
- Department of Immunobiology, University of Arizona College of Medicine-Tucson, Tucson, AZ 85724
- Arizona Center on Aging, University of Arizona College of Medicine-Tucson, Tucson, AZ 85724
| | - Janko Nikolich-Žugich
- Department of Immunobiology, University of Arizona College of Medicine-Tucson, Tucson, AZ 85724
- Arizona Center on Aging, University of Arizona College of Medicine-Tucson, Tucson, AZ 85724
- BIO5 Institute, University of Arizona, Tucson, AZ 85719
| |
Collapse
|
48
|
Wang L, Liu G, Hu Y, Gou S, He T, Feng Q, Cai K. Doxorubicin-loaded polypyrrole nanovesicles for suppressing tumor metastasis through combining photothermotherapy and lymphatic system-targeted chemotherapy. NANOSCALE 2022; 14:3097-3111. [PMID: 35141740 DOI: 10.1039/d2nr00186a] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
The lymphatic system provides a main route for the dissemination of most malignancies, which was related to high mortality in cancer patients. Traditional intravenous chemotherapy is of limited effectiveness on lymphatic metastasis due to the difficulty in accessing the lymphatic system. Herein, a novel lymphatic-targeting nanoplatform is prepared by loading doxorubicin (DOX) into sub-50 nm polypyrrole nanovesicles (PPy NVs). The PPy NVs possessed hollow spherical morphologies and a negative surface charge, leading to high drug loading capacity. These vesicles can also convert near-infrared (NIR) light into heat and thus can be used for tumor thermal ablation. DOX loaded PPy NVs (PPy@DOX NVs) along with NIR illumination are highly effective against 4T1 breast cancer cells in vitro. More importantly, following subcutaneous (SC) injection, a direct lymphatic migration of PPy@DOX NVs is confirmed through fluorescence observation of the isolated draining nodes. The acidic conditions in metastatic nodes might subsequently trigger the release of the encapsulated DOX NVs based on their pH-sensitive release profile. In a mouse model bearing 4T1 breast cancer, lymphatic metastases, as well as lung metastases, are significantly inhibited by nanocarrier-mediated trans-lymphatic drug delivery in combination with photothermal ablation. In conclusion, this platform holds great potential in impeding tumor growth and metastasis.
Collapse
Affiliation(s)
- Lu Wang
- Key Laboratory of Biorheological Science and Technology, Ministry of Education College of Bioengineering, Chongqing University, Chongqing 400044, China.
| | - Genhua Liu
- Key Laboratory of Biorheological Science and Technology, Ministry of Education College of Bioengineering, Chongqing University, Chongqing 400044, China.
| | - Yunping Hu
- Fujian Provincial Key Laboratory of Advanced Materials Oriented Chemical Engineering, College of Chemistry and Materials Science, Fujian Normal University Fuzhou, Fujian 350007, China
| | - Shuangquan Gou
- Key Laboratory of Biorheological Science and Technology, Ministry of Education College of Bioengineering, Chongqing University, Chongqing 400044, China.
| | - Tingting He
- Key Laboratory of Biorheological Science and Technology, Ministry of Education College of Bioengineering, Chongqing University, Chongqing 400044, China.
| | - Qian Feng
- Key Laboratory of Biorheological Science and Technology, Ministry of Education College of Bioengineering, Chongqing University, Chongqing 400044, China.
| | - Kaiyong Cai
- Key Laboratory of Biorheological Science and Technology, Ministry of Education College of Bioengineering, Chongqing University, Chongqing 400044, China.
| |
Collapse
|
49
|
Development of Preclinical Ultrasound Imaging Techniques to Identify and Image Sentinel Lymph Nodes in a Cancerous Animal Model. Cancers (Basel) 2022; 14:cancers14030561. [PMID: 35158829 PMCID: PMC8833694 DOI: 10.3390/cancers14030561] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Accepted: 01/19/2022] [Indexed: 02/06/2023] Open
Abstract
Simple Summary Bowel cancer is the fourth most common cancer in the UK. Treatment is dominated by major surgery because current imaging modalities cannot accurately determine lymph node involvement or vascular invasion. Although potentially curative, surgery carries a high risk of short- and long-term morbidity, including stoma formation. Optimized pre-treatment imaging would decrease the number of bowel cancer patients requiring major surgery. Such imaging would also be equally applicable to other cancers where local resection could significantly improve patient quality of life without compromising long-term outcomes (e.g., melanoma, head and neck cancers, gastro-esophageal, bladder). In this study, we created two mouse models (tumor and control) and used the resolution of high-frequency ultrasound imaging and parameters calculated from dynamically contrast-enhanced ultrasound to predict the likelihood of draining lymph nodes to be involved in the disease. Abstract Lymph nodes (LNs) are believed to be the first organs targeted by colorectal cancer cells detached from a primary solid tumor because of their role in draining interstitial fluids. Better detection and assessment of these organs have the potential to help clinicians in stratification and designing optimal design of oncological treatments for each patient. Whilst highly valuable for the detection of primary tumors, CT and MRI remain limited for the characterization of LNs. B-mode ultrasound (US) and contrast-enhanced ultrasound (CEUS) can improve the detection of LNs and could provide critical complementary information to MRI and CT scans; however, the European Federation of Societies for Ultrasound in Medicine and Biology (EFSUMB) guidelines advise that further evidence is required before US or CEUS can be recommended for clinical use. Moreover, knowledge of the lymphatic system and LNs is relatively limited, especially in preclinical models. In this pilot study, we have created a mouse model of metastatic cancer and utilized 3D high-frequency ultrasound to assess the volume, shape, and absence of hilum, along with CEUS to assess the flow dynamics of tumor-free and tumor-bearing LNs in vivo. The aforementioned parameters were used to create a scoring system to predict the likelihood of a disease-involved LN before establishing post-mortem diagnosis with histopathology. Preliminary results suggest that a sum score of parameters may provide a more accurate diagnosis than the LN size, the single parameter currently used to predict the involvement of an LN in disease.
Collapse
|
50
|
Cooper TK, Meyerholz DK, Beck AP, Delaney MA, Piersigilli A, Southard TL, Brayton CF. Research-Relevant Conditions and Pathology of Laboratory Mice, Rats, Gerbils, Guinea Pigs, Hamsters, Naked Mole Rats, and Rabbits. ILAR J 2022; 62:77-132. [PMID: 34979559 DOI: 10.1093/ilar/ilab022] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Accepted: 05/12/2021] [Indexed: 12/31/2022] Open
Abstract
Animals are valuable resources in biomedical research in investigations of biological processes, disease pathogenesis, therapeutic interventions, safety, toxicity, and carcinogenicity. Interpretation of data from animals requires knowledge not only of the processes or diseases (pathophysiology) under study but also recognition of spontaneous conditions and background lesions (pathology) that can influence or confound the study results. Species, strain/stock, sex, age, anatomy, physiology, spontaneous diseases (noninfectious and infectious), and neoplasia impact experimental results and interpretation as well as animal welfare. This review and the references selected aim to provide a pathology resource for researchers, pathologists, and veterinary personnel who strive to achieve research rigor and validity and must understand the spectrum of "normal" and expected conditions to accurately identify research-relevant experimental phenotypes as well as unusual illness, pathology, or other conditions that can compromise studies involving laboratory mice, rats, gerbils, guinea pigs, hamsters, naked mole rats, and rabbits.
Collapse
Affiliation(s)
- Timothy K Cooper
- Department of Comparative Medicine, Penn State Hershey Medical Center, Hershey, PA, USA
| | - David K Meyerholz
- Department of Pathology, University of Iowa Roy J. and Lucille A. Carver College of Medicine, Iowa City, Iowa, USA
| | - Amanda P Beck
- Department of Pathology, Yeshiva University Albert Einstein College of Medicine, Bronx, New York, USA
| | - Martha A Delaney
- Zoological Pathology Program, University of Illinois at Urbana-Champaign College of Veterinary Medicine, Urbana-Champaign, Illinois, USA
| | - Alessandra Piersigilli
- Laboratory of Comparative Pathology and the Genetically Modified Animal Phenotyping Service, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Teresa L Southard
- Department of Biomedical Sciences, Cornell University College of Veterinary Medicine, Ithaca, New York, USA
| | - Cory F Brayton
- Department of Molecular and Comparative Pathobiology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|