1
|
Tamburini B, Sheridan R, Doan T, Lucas C, Forward T, Fleming I, Uecker-Martin A, Morrison T, Hesselberth J. A specific gene expression program underlies antigen archiving by lymphatic endothelial cells in mammalian lymph nodes. RESEARCH SQUARE 2024:rs.3.rs-5493746. [PMID: 39711554 PMCID: PMC11661310 DOI: 10.21203/rs.3.rs-5493746/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2024]
Abstract
Lymph node (LN) lymphatic endothelial cells (LEC) actively acquire and archive foreign antigens. Here, we address questions of how LECs achieve durable antigen archiving and whether LECs with high levels of antigen express unique transcriptional programs. We used single cell sequencing in dissociated LN tissue and spatial transcriptomics to quantify antigen levels in LEC subsets and dendritic cell populations at multiple time points after immunization and determined that ceiling and floor LECs archive antigen for the longest duration. We identify, using spatial transcriptomics, antigen positive LEC-dendritic cell interactions. Using a prime-boost strategy we find increased antigen levels within LECs after a second immunization demonstrating that LEC antigen acquisition and archiving capacity can be improved over multiple exposures. Using machine learning we defined a unique transcriptional program within archiving LECs that predicted LEC archiving capacity in mouse and human independent data sets. We validated this modeling, showing we could predict lower levels of LEC antigen archiving in chikungunya virus-infected mice and demonstrated in vivo the accuracy of our prediction. Collectively, our findings establish unique properties of LECs and a defining transcriptional program for antigen archiving that can predict antigen archiving capacity in different disease states and organisms.
Collapse
Affiliation(s)
| | | | - Thu Doan
- University of Colorado Anschutz Medical Campus
| | | | | | | | | | | | | |
Collapse
|
2
|
Yang L, Wang G, Ma Y, Zhao Q, Zhao H, Wang Q, Zhong C, Zhang C, Yang Y. TRPML1 acts as a predisposing factor in lymphedema development by regulating the subcellular localization of aquaporin-3, -5. PLoS One 2024; 19:e0310653. [PMID: 39637010 PMCID: PMC11620549 DOI: 10.1371/journal.pone.0310653] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2024] [Accepted: 09/04/2024] [Indexed: 12/07/2024] Open
Abstract
An imbalance in lymphatic fluid, whether it is caused by generation, transport, outflow, or dysfunctional vessels, can lead to lymphedema; however, the exact pathogenesis of this disease remains unclear. To explore the mechanism, we focused on the association among TRPML1, aquaporin-3 (AQP3), and aquaporin-5 (AQP5) in human lymphatic endothelial cells (HLECs). We explored the role of TRPML1 in altering the permeability of HLECs in lymphedema. Meanwhile, we constructed a disease model using gene-knockout mice to observe the effect of TRPML1 on inflammation and fibrosis in lymphedema sites. Our results indicate that TRPML1 not only regulates the localization of AQP3, -5 to the cell membrane but also increases HLEC permeability, disrupts lymphatic fluid transport, and mediates the development of chronic inflammation at the site of lymphedema. Our study suggests that TRPML1 is a precipitating factor in lymphedema. Our findings improve the understanding of TRPML1 and aquaporins in secondary lymphedema, providing valuable insights for future research.
Collapse
Affiliation(s)
- Lijie Yang
- Department of Cell Biology and Medical Genetics, College of Basic Medical Sciences, Jilin University, Changchun, Jilin Province, China
| | - Guanzheng Wang
- Department of Cell Biology and Medical Genetics, College of Basic Medical Sciences, Jilin University, Changchun, Jilin Province, China
| | - Yuan Ma
- College of Basic Medicine, Changchun University of Chinese Medicine, Changchun, Jilin Province, China
| | - Qiancheng Zhao
- Department of Cell Biology and Medical Genetics, College of Basic Medical Sciences, Jilin University, Changchun, Jilin Province, China
| | - He Zhao
- Department of Cell Biology and Medical Genetics, College of Basic Medical Sciences, Jilin University, Changchun, Jilin Province, China
| | - Qi Wang
- Department of Cell Biology and Medical Genetics, College of Basic Medical Sciences, Jilin University, Changchun, Jilin Province, China
| | - Chonghua Zhong
- College of Basic Medical Sciences, Jilin University, Changchun, Jilin Province, China
| | - Chunmei Zhang
- Department of Cell Biology and Medical Genetics, College of Basic Medical Sciences, Jilin University, Changchun, Jilin Province, China
| | - Yiming Yang
- Department of Cell Biology and Medical Genetics, College of Basic Medical Sciences, Jilin University, Changchun, Jilin Province, China
- College of Basic Medical Sciences, Jilin University, Changchun, Jilin Province, China
| |
Collapse
|
3
|
Guo R, Zhong L, Ma S, Gong B, Shen C, Wang Z, Deng L, Zhao D, Gao H, Gong T. A biomimetic solution, albumin-doxorubicin molecular complex, targeting tumor and tumor-draining lymph nodes. J Mater Chem B 2024. [PMID: 39479935 DOI: 10.1039/d4tb01917b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2024]
Abstract
Chemotherapy-induced immunologic cell death is haunted by the non-specific distribution of chemotherapeutic drugs and insignificant immune activation effects, which render efforts to inhibit the distant metastasis of tumors frustrated. Given the pivotal role that lymph nodes play in tumor metastasis, it is of vital importance whether the drug delivery to tumor-draining lymph nodes (TDLNs) succeeds. In the current study, we developed a doxorubicin-albumin complex (DOX-HSA) solution with the specific ability to simultaneously target the primary tumor and the TDLNs. DOX-HSA could effectively activate and amplify the immunogenic cell death (ICD) effect in both the tumor tissues and the TDLNs, resulting in increased release of damage-associated molecular patterns (DAMPs), which further promoted phagocytosis and maturation of dendritic cells (DCs), stimulated activation of CD8+T cells, and then significantly enhanced the therapeutic effects of doxorubicin on orthotopic 4T1 tumor-bearing model mice. Therefore, the DOX-HSA solution demonstrated a more prominent ability to control cancer cells and curb metastasis, as well as improved security by reducing cardiotoxicity and myelosuppression toxicity of doxorubicin itself. This DOX-HSA strengthened the synergistic anti-tumor effects based on the ICD effect in combination with traditional chemotherapy, thus providing promising prospects for clinical application.
Collapse
Affiliation(s)
- Rui Guo
- Key Laboratory of Drug-Targeting & Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drugs & Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610064, P. R. China.
| | - Lanlan Zhong
- Key Laboratory of Drug-Targeting & Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drugs & Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610064, P. R. China.
| | - Sirui Ma
- Key Laboratory of Drug-Targeting & Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drugs & Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610064, P. R. China.
| | - Bokai Gong
- Key Laboratory of Drug-Targeting & Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drugs & Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610064, P. R. China.
| | - Chen Shen
- Key Laboratory of Drug-Targeting & Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drugs & Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610064, P. R. China.
| | - Zijun Wang
- Key Laboratory of Drug-Targeting & Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drugs & Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610064, P. R. China.
| | - Li Deng
- Key Laboratory of Drug-Targeting & Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drugs & Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610064, P. R. China.
| | - Dong Zhao
- Sichuan Kelun Pharmaceutical Research Institute Co. Ltd., Chengdu 611130, P. R. China
| | - Huile Gao
- Key Laboratory of Drug-Targeting & Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drugs & Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610064, P. R. China.
| | - Tao Gong
- Key Laboratory of Drug-Targeting & Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drugs & Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610064, P. R. China.
| |
Collapse
|
4
|
Lu R, Lee BJ, Lee E. Three-Dimensional Lymphatics-on-a-Chip Reveals Distinct, Size-Dependent Nanoparticle Transport Mechanisms in Lymphatic Drug Delivery. ACS Biomater Sci Eng 2024; 10:5752-5763. [PMID: 39176471 DOI: 10.1021/acsbiomaterials.4c01005] [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] [Indexed: 08/24/2024]
Abstract
Although nanoparticle-based lymphatic drug delivery systems promise better treatment of cancer, infectious disease, and immune disease, their clinical translations are limited by low delivery efficiencies and unclear transport mechanisms. Here, we employed a three-dimensional (3D) lymphatics-on-a-chip featuring an engineered lymphatic vessel (LV) capable of draining interstitial fluids including nanoparticles. We tested lymphatic drainage of different sizes (30, 50, and 70 nm) of PLGA-b-PEG nanoparticles (NPs) using the lymphatics-on-a-chip device. In this study, we discovered that smaller NPs (30 and 50 nm) transported faster than larger NPs (70 nm) through the interstitial space, as expected, but the smaller NPs were captured by lymphatic endothelial cells (LECs) and accumulated within their cytosol, delaying NP transport into the lymphatic lumen, which was not observed in larger NPs. To examine the mechanisms of size-dependent NP transports, we employed four inhibitors, dynasore, nystatin, amiloride, and adrenomedullin, to selectively block dynamin-, caveolin-, macropinocytosis-mediated endocytosis-, and cell junction-mediated paracellular transport. Inhibiting dynamin using dynasore enhanced the transport of smaller NPs (30 and 50 nm) into the lymphatic lumen, minimizing cytosolic accumulation, but showed no effect on larger NP transport. Interestingly, the inhibition of caveolin by nystatin decreased the lymphatic transport of larger NPs without affecting the smaller NP transport, indicating distinct endocytosis mechanisms used by different sizes of NPs. Macropinocytosis inhibition by amiloride did not change the drainage of all sizes of NPs; however, paracellular transport inhibition by adrenomedullin blocked the lymphatic transport of NPs of all sizes. We further revealed that smaller NPs were captured in the Rab7-positive late-stage lymphatic endosomes to delay their lymphatic drainage, which was reversed by dynamin inhibition, suggesting that Rab7 is a potential target to enhance the lymphatic delivery of smaller NPs. Together, our 3D lymphatics-on-a-chip model unveils size-dependent NP transport mechanisms in lymphatic drug delivery.
Collapse
Affiliation(s)
- Renhao Lu
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, New York 14853, United States
| | - Benjamin J Lee
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, New York 14853, United States
| | - Esak Lee
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, New York 14853, United States
| |
Collapse
|
5
|
Karakousi T, Mudianto T, Lund AW. Lymphatic vessels in the age of cancer immunotherapy. Nat Rev Cancer 2024; 24:363-381. [PMID: 38605228 DOI: 10.1038/s41568-024-00681-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/27/2024] [Indexed: 04/13/2024]
Abstract
Lymphatic transport maintains homeostatic health and is necessary for immune surveillance, and yet lymphatic growth is often associated with solid tumour development and dissemination. Although tumour-associated lymphatic remodelling and growth were initially presumed to simply expand a passive route for regional metastasis, emerging research puts lymphatic vessels and their active transport at the interface of metastasis, tumour-associated inflammation and systemic immune surveillance. Here, we discuss active mechanisms through which lymphatic vessels shape their transport function to influence peripheral tissue immunity and the current understanding of how tumour-associated lymphatic vessels may both augment and disrupt antitumour immune surveillance. We end by looking forward to emerging areas of interest in the field of cancer immunotherapy in which lymphatic vessels and their transport function are likely key players: the formation of tertiary lymphoid structures, immune surveillance in the central nervous system, the microbiome, obesity and ageing. The lessons learnt support a working framework that defines the lymphatic system as a key determinant of both local and systemic inflammatory networks and thereby a crucial player in the response to cancer immunotherapy.
Collapse
Affiliation(s)
- Triantafyllia Karakousi
- Ronald O. Perelman Department of Dermatology, NYU Grossman School of Medicine, New York, NY, USA
| | - Tenny Mudianto
- Ronald O. Perelman Department of Dermatology, NYU Grossman School of Medicine, New York, NY, USA
| | - Amanda W Lund
- Ronald O. Perelman Department of Dermatology, NYU Grossman School of Medicine, New York, NY, USA.
- Department of Pathology, NYU Grossman School of Medicine, New York, NY, USA.
- Laura and Isaac Perlmutter Cancer Center, NYU Langone Health, New York, NY, USA.
| |
Collapse
|
6
|
Sheridan RM, Doan TA, Lucas C, Forward TS, Uecker-Martin A, Morrison TE, Hesselberth JR, Tamburini BAJ. A specific and portable gene expression program underlies antigen archiving by lymphatic endothelial cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.01.587647. [PMID: 38617225 PMCID: PMC11014631 DOI: 10.1101/2024.04.01.587647] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/16/2024]
Abstract
Antigens from protein subunit vaccination traffic from the tissue to the draining lymph node, either passively via the lymph or carried by dendritic cells at the local injection site. Lymph node (LN) lymphatic endothelial cells (LEC) actively acquire and archive foreign antigens, and archived antigen can be released during subsequent inflammatory stimulus to improve immune responses. Here, we answer questions about how LECs achieve durable antigen archiving and whether there are transcriptional signatures associated with LECs containing high levels of antigen. We used single cell sequencing in dissociated LN tissue to quantify antigen levels in LEC and dendritic cell populations at multiple timepoints after immunization, and used machine learning to define a unique transcriptional program within archiving LECs that can predict LEC archiving capacity in independent data sets. Finally, we validated this modeling, showing we could predict antigen archiving from a transcriptional dataset of CHIKV infected mice and demonstrated in vivo the accuracy of our prediction. Collectively, our findings establish a unique transcriptional program in LECs that promotes antigen archiving that can be translated to other systems.
Collapse
Affiliation(s)
- Ryan M. Sheridan
- Department of Biochemistry and Molecular Genetics, RNA Bioscience Initiative, University of Colorado School of Medicine
| | - Thu A. Doan
- Department of Medicine, Division of Gastroenterology and Hepatology, University of Colorado School of Medicine
- Immunology Graduate Program, University of Colorado School of Medicine
| | - Cormac Lucas
- Department of Immunology and Microbiology, Aurora, CO, USA
| | - Tadg S. Forward
- Department of Medicine, Division of Gastroenterology and Hepatology, University of Colorado School of Medicine
| | - Aspen Uecker-Martin
- Department of Biochemistry and Molecular Genetics, RNA Bioscience Initiative, University of Colorado School of Medicine
| | | | - Jay R. Hesselberth
- Department of Biochemistry and Molecular Genetics, RNA Bioscience Initiative, University of Colorado School of Medicine
| | - Beth A. Jirón Tamburini
- Department of Medicine, Division of Gastroenterology and Hepatology, University of Colorado School of Medicine
- Immunology Graduate Program, University of Colorado School of Medicine
- Department of Immunology and Microbiology, Aurora, CO, USA
| |
Collapse
|
7
|
McCright J, Yarmovsky J, Maisel K. Para- and Transcellular Transport Kinetics of Nanoparticles across Lymphatic Endothelial Cells. Mol Pharm 2024; 21:1160-1169. [PMID: 37851841 PMCID: PMC10923144 DOI: 10.1021/acs.molpharmaceut.3c00720] [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] [Indexed: 10/20/2023]
Abstract
Lymphatic vessels have received significant attention as drug delivery targets, as they shuttle materials from peripheral tissues to the lymph nodes, where adaptive immunity is formed. Delivery of immune modulatory materials to the lymph nodes via lymphatic vessels has been shown to enhance their efficacy and also improve the bioavailability of drugs when delivered to intestinal lymphatic vessels. In this study, we generated a three-compartment model of a lymphatic vessel with a set of kinematic differential equations to describe the transport of nanoparticles from the surrounding tissues into lymphatic vessels. We used previously published data and collected additional experimental parameters, including the transport efficiency of nanoparticles over time, and also examined how nanoparticle formulation affected the cellular transport mechanisms using small molecule inhibitors. These experimental data were incorporated into a system of kinematic differential equations, and nonlinear, least-squares curve fitting algorithms were employed to extrapolate transport coefficients within our model. The subsequent computational framework produced some of the first parameters to describe transport kinetics across lymphatic endothelial cells and allowed for the quantitative analysis of the driving mechanisms of transport into lymphatic vessels. Our model indicates that transcellular mechanisms, such as micro- and macropinocytosis, drive transport into lymphatics. This information is crucial to further design strategies that will modulate lymphatic transport for drug delivery, particularly in diseases like lymphedema, where normal lymphatic functions are impaired.
Collapse
Affiliation(s)
- Jacob McCright
- Department of Bioengineering, University of Maryland College Park, College Park, Maryland 20742, United States
| | - Jenny Yarmovsky
- Department of Bioengineering, University of Maryland College Park, College Park, Maryland 20742, United States
| | - Katharina Maisel
- Department of Bioengineering, University of Maryland College Park, College Park, Maryland 20742, United States
| |
Collapse
|
8
|
Zhong X, Liu Y, Ardekani AM. A compartment model for subcutaneous injection of monoclonal antibodies. Int J Pharm 2024; 650:123687. [PMID: 38103705 DOI: 10.1016/j.ijpharm.2023.123687] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2023] [Revised: 12/01/2023] [Accepted: 12/06/2023] [Indexed: 12/19/2023]
Abstract
Despite the growing popularity of subcutaneous (SC) administration for monoclonal antibodies (mAbs), there remains a limited understanding of the significance of mAb transport rate constants within the interstitial space and the lymphatic system on their pharmacokinetics. To bridge this knowledge gap, we introduce a compartmental model for subcutaneously administered mAbs. Our model differentiates FcRn-expressing cells across various sites, and the model predictions agree with experimental data from both human and rat studies. Our findings indicate that the time to reach the maximum mAb concentration in the plasma, denoted by Tmax, displays a weak positive correlation with mAb half-life and a negligible correlation with bioavailability. In contrast, the half-life of mAbs exhibits a strong positive correlation with bioavailability. Moreover, the rate of mAb transport from lymph to plasma significantly affects the mAb half-life. Increasing the transport rates of mAbs from the injection site to the lymph or from lymph to plasma enhances bioavailability. These insights, combined with our compartmental model, contribute to a deeper understanding of the pharmacokinetics of subcutaneously administered mAbs.
Collapse
Affiliation(s)
- Xiaoxu Zhong
- School of Mechanical Engineering, Purdue University, West Lafayette, IN 47906, United States
| | - Yikai Liu
- School of Mechanical Engineering, Purdue University, West Lafayette, IN 47906, United States
| | - Arezoo M Ardekani
- School of Mechanical Engineering, Purdue University, West Lafayette, IN 47906, United States.
| |
Collapse
|
9
|
Taheri A, Bremmell KE, Joyce P, Prestidge CA. Battle of the milky way: Lymphatic targeted drug delivery for pathogen eradication. J Control Release 2023; 363:507-524. [PMID: 37797891 DOI: 10.1016/j.jconrel.2023.10.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Revised: 09/14/2023] [Accepted: 10/01/2023] [Indexed: 10/07/2023]
Abstract
Many viruses, bacteria, and parasites rely on the lymphatic system for survival, replication, and dissemination. While conventional anti-infectives can combat infection-causing agents in the bloodstream, they do not reach the lymphatic system to eradicate the pathogens harboured there. This can result in ineffective drug exposure and reduce treatment effectiveness. By developing effective lymphatic delivery strategies for antiviral, antibacterial, and antiparasitic drugs, their systemic pharmacokinetics may be improved, as would their ability to reach their target pathogens within the lymphatics, thereby improving clinical outcomes in a variety of acute and chronic infections with lymphatic involvement (e.g., acquired immunodeficiency syndrome, tuberculosis, and filariasis). Here, we discuss approaches to targeting anti-infective drugs to the intestinal and dermal lymphatics, aiming to eliminate pathogen reservoirs and interfere with their survival and reproduction inside the lymphatic system. These include optimized lipophilic prodrugs and drug delivery systems that promote lymphatic transport after oral and dermal drug intake. For intestinal lymphatic delivery via the chylomicron pathway, molecules should have logP values >5 and long-chain triglyceride solubilities >50 mg/g, and for dermal lymphatic delivery via interstitial lymphatic drainage, nanoparticle formulations with particle size between 10 and 100 nm are generally preferred. Insight from this review may promote new and improved therapeutic solutions for pathogen eradication and combating infective diseases, as lymphatic system involvement in pathogen dissemination and drug resistance has been neglected compared to other pathways leading to treatment failure.
Collapse
Affiliation(s)
- Ali Taheri
- Clinical and Health Sciences, University of South Australia, Adelaide, SA 5000, Australia
| | - Kristen E Bremmell
- Clinical and Health Sciences, University of South Australia, Adelaide, SA 5000, Australia
| | - Paul Joyce
- Clinical and Health Sciences, University of South Australia, Adelaide, SA 5000, Australia
| | - Clive A Prestidge
- Clinical and Health Sciences, University of South Australia, Adelaide, SA 5000, Australia.
| |
Collapse
|
10
|
Lee E, Chan SL, Lee Y, Polacheck WJ, Kwak S, Wen A, Nguyen DHT, Kutys ML, Alimperti S, Kolarzyk AM, Kwak TJ, Eyckmans J, Bielenberg DR, Chen H, Chen CS. A 3D biomimetic model of lymphatics reveals cell-cell junction tightening and lymphedema via a cytokine-induced ROCK2/JAM-A complex. Proc Natl Acad Sci U S A 2023; 120:e2308941120. [PMID: 37782785 PMCID: PMC10576061 DOI: 10.1073/pnas.2308941120] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Accepted: 09/04/2023] [Indexed: 10/04/2023] Open
Abstract
Impaired lymphatic drainage and lymphedema are major morbidities whose mechanisms have remained obscure. To study lymphatic drainage and its impairment, we engineered a microfluidic culture model of lymphatic vessels draining interstitial fluid. This lymphatic drainage-on-chip revealed that inflammatory cytokines that are known to disrupt blood vessel junctions instead tightened lymphatic cell-cell junctions and impeded lymphatic drainage. This opposing response was further demonstrated when inhibition of rho-associated protein kinase (ROCK) was found to normalize fluid drainage under cytokine challenge by simultaneously loosening lymphatic junctions and tightening blood vessel junctions. Studies also revealed a previously undescribed shift in ROCK isoforms in lymphatic endothelial cells, wherein a ROCK2/junctional adhesion molecule-A (JAM-A) complex emerges that is responsible for the cytokine-induced lymphatic junction zippering. To validate these in vitro findings, we further demonstrated in a genetic mouse model that lymphatic-specific knockout of ROCK2 reversed lymphedema in vivo. These studies provide a unique platform to generate interstitial fluid pressure and measure the drainage of interstitial fluid into lymphatics and reveal a previously unappreciated ROCK2-mediated mechanism in regulating lymphatic drainage.
Collapse
Affiliation(s)
- Esak Lee
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA02115
- Department of Biomedical Engineering, Biological Design Center, Boston University, Boston, MA02215
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY14853
| | - Siu-Lung Chan
- Vascular Biology Program, Boston Children’s Hospital, Harvard Medical School, Boston, MA02115
| | - Yang Lee
- Vascular Biology Program, Boston Children’s Hospital, Harvard Medical School, Boston, MA02115
| | - William J. Polacheck
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA02115
- Department of Biomedical Engineering, Biological Design Center, Boston University, Boston, MA02215
| | - Sukyoung Kwak
- Vascular Biology Program, Boston Children’s Hospital, Harvard Medical School, Boston, MA02115
| | - Aiyun Wen
- Vascular Biology Program, Boston Children’s Hospital, Harvard Medical School, Boston, MA02115
| | - Duc-Huy T. Nguyen
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA02115
- Department of Biomedical Engineering, Biological Design Center, Boston University, Boston, MA02215
| | - Matthew L. Kutys
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA02115
- Department of Biomedical Engineering, Biological Design Center, Boston University, Boston, MA02215
| | - Stella Alimperti
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA02115
- Department of Biomedical Engineering, Biological Design Center, Boston University, Boston, MA02215
| | - Anna M. Kolarzyk
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY14853
| | - Tae Joon Kwak
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY14853
| | - Jeroen Eyckmans
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA02115
- Department of Biomedical Engineering, Biological Design Center, Boston University, Boston, MA02215
| | - Diane R. Bielenberg
- Vascular Biology Program, Boston Children’s Hospital, Harvard Medical School, Boston, MA02115
| | - Hong Chen
- Vascular Biology Program, Boston Children’s Hospital, Harvard Medical School, Boston, MA02115
| | - Christopher S. Chen
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA02115
- Department of Biomedical Engineering, Biological Design Center, Boston University, Boston, MA02215
| |
Collapse
|
11
|
Liu P, Ding P, Sun C, Chen S, Lowe S, Meng L, Zhao Q. Lymphangiogenesis in gastric cancer: function and mechanism. Eur J Med Res 2023; 28:405. [PMID: 37803421 PMCID: PMC10559534 DOI: 10.1186/s40001-023-01298-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2023] [Accepted: 08/18/2023] [Indexed: 10/08/2023] Open
Abstract
Increased lymphangiogenesis and lymph node (LN) metastasis are thought to be important steps in cancer metastasis, and are associated with patient's poor prognosis. There is increasing evidence that the lymphatic system may play a crucial role in regulating tumor immune response and limiting tumor metastasis, since tumor lymphangiogenesis is more prominent in tumor metastasis and diffusion. Lymphangiogenesis takes place in embryonic development, wound healing, and a variety of pathological conditions, including tumors. Tumor cells and tumor microenvironment cells generate growth factors (such as lymphangiogenesis factor VEGF-C/D), which can promote lymphangiogenesis, thereby inducing the metastasis and diffusion of tumor cells. Nevertheless, the current research on lymphangiogenesis in gastric cancer is relatively scattered and lacks a comprehensive understanding. Therefore, in this review, we aim to provide a detailed perspective on molecules and signal transduction pathways that regulate gastric cancer lymphogenesis, which may provide new insights for the diagnosis and treatment of cancer.
Collapse
Affiliation(s)
- Pengpeng Liu
- The Third Department of Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, 050011, Hebei, China
- Hebei Key Laboratory of Precision Diagnosis and Comprehensive Treatment of Gastric Cancer, Shijiazhuang, 050011, China
| | - Ping'an Ding
- The Third Department of Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, 050011, Hebei, China
- Hebei Key Laboratory of Precision Diagnosis and Comprehensive Treatment of Gastric Cancer, Shijiazhuang, 050011, China
| | - Chenyu Sun
- AMITA Health Saint Joseph Hospital Chicago, 2900 N. Lake Shore Drive, Chicago, IL, 60657, USA
| | - Shuya Chen
- Newham University Hospital, Glen Road, Plaistow, London, E13 8SL, England, UK
| | - Scott Lowe
- College of Osteopathic Medicine, Kansas City University, 1750 Independence Ave, Kansas City, MO, 64106, USA
| | - Lingjiao Meng
- Hebei Key Laboratory of Precision Diagnosis and Comprehensive Treatment of Gastric Cancer, Shijiazhuang, 050011, China.
- Research Center of the Fourth Hospital of Hebei Medical University, Shijiazhuang, 050011, China.
| | - Qun Zhao
- The Third Department of Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, 050011, Hebei, China.
- Hebei Key Laboratory of Precision Diagnosis and Comprehensive Treatment of Gastric Cancer, Shijiazhuang, 050011, China.
| |
Collapse
|
12
|
Luse MA, Jackson MG, Juśkiewicz ZJ, Isakson BE. Physiological functions of caveolae in endothelium. CURRENT OPINION IN PHYSIOLOGY 2023; 35:100701. [PMID: 37873030 PMCID: PMC10588508 DOI: 10.1016/j.cophys.2023.100701] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2023]
Abstract
Endothelial caveolae are essential for a wide range of physiological processes and have emerged as key players in vascular biology. Our understanding of caveolar biology in endothelial cells has expanded dramatically since their discovery revealing critical roles in mechanosensation, signal transduction, eNOS regulation, lymphatic transport, and metabolic disease progression. Furthermore, caveolae are involved in the organization of membrane domains, regulation of membrane fluidity, and endocytosis which contribute to endothelial function and integrity. Additionally, recent advances highlight the impact of caveolae-mediated signaling pathways on vascular homeostasis and pathology. Together, the diverse roles of caveolae discussed here represent a breadth of cellular functions presenting caveolae as a defining feature of endothelial form and function. In light of these new insights, targeting caveolae may hold potential for the development of novel therapeutic strategies to treat a range of vascular diseases.
Collapse
Affiliation(s)
- Melissa A. Luse
- Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine
- Department of Molecular Physiology and Biophysics, University of Virginia School of Medicine
| | - Madeline G. Jackson
- Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine
| | - Zuzanna J. Juśkiewicz
- Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine
- Department of Molecular Physiology and Biophysics, University of Virginia School of Medicine
| | - Brant E. Isakson
- Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine
- Department of Molecular Physiology and Biophysics, University of Virginia School of Medicine
| |
Collapse
|
13
|
Mehrara BJ, Radtke AJ, Randolph GJ, Wachter BT, Greenwel P, Rovira II, Galis ZS, Muratoglu SC. The emerging importance of lymphatics in health and disease: an NIH workshop report. J Clin Invest 2023; 133:e171582. [PMID: 37655664 PMCID: PMC10471172 DOI: 10.1172/jci171582] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/02/2023] Open
Abstract
The lymphatic system (LS) is composed of lymphoid organs and a network of vessels that transport interstitial fluid, antigens, lipids, cholesterol, immune cells, and other materials in the body. Abnormal development or malfunction of the LS has been shown to play a key role in the pathophysiology of many disease states. Thus, improved understanding of the anatomical and molecular characteristics of the LS may provide approaches for disease prevention or treatment. Recent advances harnessing single-cell technologies, clinical imaging, discovery of biomarkers, and computational tools have led to the development of strategies to study the LS. This Review summarizes the outcomes of the NIH workshop entitled "Yet to be Charted: Lymphatic System in Health and Disease," held in September 2022, with emphasis on major areas for advancement. International experts showcased the current state of knowledge regarding the LS and highlighted remaining challenges and opportunities to advance the field.
Collapse
Affiliation(s)
- Babak J. Mehrara
- Department of Plastic and Reconstructive Surgery, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Andrea J. Radtke
- Lymphocyte Biology Section and Center for Advanced Tissue Imaging, Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, Maryland, USA
| | - Gwendalyn J. Randolph
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Brianna T. Wachter
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, Maryland, USA
| | - Patricia Greenwel
- Division of Digestive Diseases & Nutrition, National Institute of Diabetes and Digestive and Kidney Diseases, and
| | - Ilsa I. Rovira
- Division of Cardiovascular Sciences, National Heart, Lung, and Blood Institute, NIH, Bethesda, Maryland, USA
| | - Zorina S. Galis
- Division of Cardiovascular Sciences, National Heart, Lung, and Blood Institute, NIH, Bethesda, Maryland, USA
| | - Selen C. Muratoglu
- Division of Cardiovascular Sciences, National Heart, Lung, and Blood Institute, NIH, Bethesda, Maryland, USA
| |
Collapse
|
14
|
Alexander S, Moghadam MG, Rothenbroker M, Y T Chou L. Addressing the in vivo delivery of nucleic-acid nanostructure therapeutics. Adv Drug Deliv Rev 2023; 199:114898. [PMID: 37230305 DOI: 10.1016/j.addr.2023.114898] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 05/02/2023] [Accepted: 05/18/2023] [Indexed: 05/27/2023]
Abstract
DNA and RNA nanostructures are being investigated as therapeutics, vaccines, and drug delivery systems. These nanostructures can be functionalized with guests ranging from small molecules to proteins with precise spatial and stoichiometric control. This has enabled new strategies to manipulate drug activity and to engineer devices with novel therapeutic functionalities. Although existing studies have offered encouraging in vitro or pre-clinical proof-of-concepts, establishing mechanisms of in vivo delivery is the new frontier for nucleic-acid nanotechnologies. In this review, we first provide a summary of existing literature on the in vivo uses of DNA and RNA nanostructures. Based on their application areas, we discuss current models of nanoparticle delivery, and thereby highlight knowledge gaps on the in vivo interactions of nucleic-acid nanostructures. Finally, we describe techniques and strategies for investigating and engineering these interactions. Together, we propose a framework to establish in vivo design principles and advance the in vivo translation of nucleic-acid nanotechnologies.
Collapse
Affiliation(s)
- Shana Alexander
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON M5S 3G9, Canada
| | | | - Meghan Rothenbroker
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON M5S 3G9, Canada
| | - Leo Y T Chou
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON M5S 3G9, Canada.
| |
Collapse
|
15
|
Ejazi SA, Louisthelmy R, Maisel K. Mechanisms of Nanoparticle Transport across Intestinal Tissue: An Oral Delivery Perspective. ACS NANO 2023. [PMID: 37410891 DOI: 10.1021/acsnano.3c02403] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/08/2023]
Abstract
Oral drug administration has been a popular choice due to patient compliance and limited clinical resources. Orally delivered drugs must circumvent the harsh gastrointestinal (GI) environment to effectively enter the systemic circulation. The GI tract has a number of structural and physiological barriers that limit drug bioavailability including mucus, the tightly regulated epithelial layer, immune cells, and associated vasculature. Nanoparticles have been used to enhance oral bioavailability of drugs, as they can act as a shield to the harsh GI environment and prevent early degradation while also increasing uptake and transport of drugs across the intestinal epithelium. Evidence suggests that different nanoparticle formulations may be transported via different intracellular mechanisms to cross the intestinal epithelium. Despite the existence of a significant body of work on intestinal transport of nanoparticles, many key questions remain: What causes the poor bioavailability of the oral drugs? What factors contribute to the ability of a nanoparticle to cross different intestinal barriers? Do nanoparticle properties such as size and charge influence the type of endocytic pathways taken? In this Review, we summarize the different components of intestinal barriers and the types of nanoparticles developed for oral delivery. In particular, we focus on the various intracellular pathways used in nanoparticle internalization and nanoparticle or cargo translocation across the epithelium. Understanding the gut barrier, nanoparticle characteristics, and transport pathways may lead to the development of more therapeutically useful nanoparticles as drug carriers.
Collapse
Affiliation(s)
- Sarfaraz Ahmad Ejazi
- Fischell Department of Bioengineering, University of Maryland, 3120 A. James Clark Hall, College Park, Maryland 20742, United States
| | - Rebecca Louisthelmy
- Fischell Department of Bioengineering, University of Maryland, 3120 A. James Clark Hall, College Park, Maryland 20742, United States
| | - Katharina Maisel
- Fischell Department of Bioengineering, University of Maryland, 3120 A. James Clark Hall, College Park, Maryland 20742, United States
| |
Collapse
|
16
|
Maisel K, McClain CA, Bogseth A, Thomas SN. Nanotechnologies for Physiology-Informed Drug Delivery to the Lymphatic System. Annu Rev Biomed Eng 2023; 25:233-256. [PMID: 37000965 PMCID: PMC10879987 DOI: 10.1146/annurev-bioeng-092222-034906] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2023]
Abstract
Accompanying the increasing translational impact of immunotherapeutic strategies to treat and prevent disease has been a broadening interest across both bioscience and bioengineering in the lymphatic system. Herein, the lymphatic system physiology, ranging from its tissue structures to immune functions and effects, is described. Design principles and engineering approaches to analyze and manipulate this tissue system in nanoparticle-based drug delivery applications are also elaborated.
Collapse
Affiliation(s)
- Katharina Maisel
- Fischell Department of Bioengineering, University of Maryland, College Park, Maryland, USA;
| | - Claire A McClain
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, Georgia, USA;
| | - Amanda Bogseth
- Fischell Department of Bioengineering, University of Maryland, College Park, Maryland, USA;
| | - Susan N 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 of Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, Georgia, USA
- Winship Cancer Institute, Emory University, Atlanta, Georgia, USA
| |
Collapse
|
17
|
Bogseth A, Ramirez A, Vaughan E, Maisel K. In Vitro Models of Blood and Lymphatic Vessels-Connecting Tissues and Immunity. Adv Biol (Weinh) 2023; 7:e2200041. [PMID: 35751460 PMCID: PMC9790046 DOI: 10.1002/adbi.202200041] [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: 02/18/2022] [Revised: 05/10/2022] [Indexed: 12/27/2022]
Abstract
Blood and lymphatic vessels are regulators of physiological processes, including oxygenation and fluid transport. Both vessels are ubiquitous throughout the body and are critical for sustaining tissue homeostasis. The complexity of each vessel's processes has limited the understanding of exactly how the vessels maintain their functions. Both vessels have been shown to be involved in the pathogenesis of many diseases, including cancer metastasis, and it is crucial to probe further specific mechanisms involved. In vitro models are developed to better understand blood and lymphatic physiological functions and their mechanisms. In this review, blood and lymphatic in vitro model systems, including 2D and 3D designs made using Transwells, microfluidic devices, organoid cultures, and various other methods, are described. Models studying endothelial cell-extracellular matrix interactions, endothelial barrier properties, transendothelial transport and cell migration, lymph/angiogenesis, vascular inflammation, and endothelial-cancer cell interactions are particularly focused. While the field has made significant progress in modeling and understanding lymphatic and blood vasculature, more models that include coculture of multiple cell types, complex extracellular matrix, and 3D morphologies, particularly for models mimicking disease states, will help further the understanding of the role of blood and lymphatic vasculature in health and disease.
Collapse
Affiliation(s)
- Amanda Bogseth
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742
| | - Ann Ramirez
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742
| | - Erik Vaughan
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742
| | - Katharina Maisel
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742
| |
Collapse
|
18
|
Abstract
Despite enormous advances, cardiovascular disorders are still a major threat to global health and are responsible for one-third of deaths worldwide. Research for new therapeutics and the investigation of their effects on vascular parameters is often limited by species-specific pathways and a lack of high-throughput methods. The complex 3-dimensional environment of blood vessels, intricate cellular crosstalks, and organ-specific architectures further complicate the quest for a faithful human in vitro model. The development of novel organoid models of various tissues such as brain, gut, and kidney signified a leap for the field of personalized medicine and disease research. By utilizing either embryonic- or patient-derived stem cells, different developmental and pathological mechanisms can be modeled and investigated in a controlled in vitro environment. We have recently developed self-organizing human capillary blood vessel organoids that recapitulate key processes of vasculogenesis, angiogenesis, and diabetic vasculopathy. Since then, this organoid system has been utilized as a model for other disease processes, refined, and adapted for organ specificity. In this review, we will discuss novel and alternative approaches to blood vessel engineering and explore the cellular identity of engineered blood vessels in comparison to in vivo vasculature. Future perspectives and the therapeutic potential of blood vessel organoids will be discussed.
Collapse
Affiliation(s)
- Kirill Salewskij
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences, Vienna (K.S., J.M.P.).,Vienna BioCenter PhD Program, Doctoral School of the University of Vienna and Medical University of Vienna, Austria (K.S.)
| | - Josef M Penninger
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences, Vienna (K.S., J.M.P.).,Department of Medical Genetics, Life Sciences Institute, University of British Columbia, Vancouver, Canada (J.M.P.)
| |
Collapse
|
19
|
Ruliffson BNK, Whittington CF. Regulating Lymphatic Vasculature in Fibrosis: Understanding the Biology to Improve the Modeling. Adv Biol (Weinh) 2023; 7:e2200158. [PMID: 36792967 DOI: 10.1002/adbi.202200158] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Revised: 12/19/2022] [Indexed: 02/17/2023]
Abstract
Fibrosis occurs in many chronic diseases with lymphatic vascular insufficiency (e.g., kidney disease, tumors, and lymphedema). New lymphatic capillary growth can be triggered by fibrosis-related tissue stiffening and soluble factors, but questions remain for how related biomechanical, biophysical, and biochemical cues affect lymphatic vascular growth and function. The current preclinical standard for studying lymphatics is animal modeling, but in vitro and in vivo outcomes often do not align. In vitro models can also be limited in their ability to separate vascular growth and function as individual outcomes, and fibrosis is not traditionally included in model design. Tissue engineering provides an opportunity to address in vitro limitations and mimic microenvironmental features that impact lymphatic vasculature. This review discusses fibrosis-related lymphatic vascular growth and function in disease and the current state of in vitro lymphatic vascular models while highlighting relevant knowledge gaps. Additional insights into the future of in vitro lymphatic vascular models demonstrate how prioritizing fibrosis alongside lymphatics will help capture the complexity and dynamics of lymphatics in disease. Overall, this review aims to emphasize that an advanced understanding of lymphatics within a fibrotic disease-enabled through more accurate preclinical modeling-will significantly impact therapeutic development toward restoring lymphatic vessel growth and function in patients.
Collapse
Affiliation(s)
- Brian N K Ruliffson
- Department of Biomedical Engineering, Worcester Polytechnic Institute, 100 Institute Rd., Worcester, MA, 01609, USA
| | - Catherine F Whittington
- Department of Biomedical Engineering, Worcester Polytechnic Institute, 100 Institute Rd., Worcester, MA, 01609, USA
| |
Collapse
|
20
|
Lymphatic uptake of biotherapeutics through a 3D hybrid discrete-continuum vessel network in the skin tissue. J Control Release 2023; 354:869-888. [PMID: 36634711 DOI: 10.1016/j.jconrel.2022.12.045] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 12/17/2022] [Accepted: 12/20/2022] [Indexed: 01/14/2023]
Abstract
Subcutaneous administration is a common approach for the delivery of biotherapeutics, which is achieved mainly through the absorption across lymphatic vessels. In this paper, the drug transport and lymphatic uptake through a three-dimensional hybrid discrete-continuum vessel network in the skin tissue are investigated through high-fidelity numerical simulations. We find that the local lymphatic uptake through the explicit vessels significantly affects macroscopic drug absorption. The diffusion of drug solute through the explicit vessel network affects the lymphatic uptake after the injection. This effect, however, cannot be captured using previously developed continuum models. The lymphatic uptake is dominated by the convection due to lymphatic drainage driven by the pressure difference, which is rarely studied in experiments and simulations. Furthermore, the effects of injection volume and depth on the lymphatic uptake are investigated in a multi-layered domain. We find that the injection volume significantly affects the rate of lymphatic uptake through the heterogeneous vessel network, while the injection depth has little influence, which is consistent with the experimental results. At last, the binding and metabolism of drug molecules are studied to bridge the simulations to the drug clearance experients. We provide a new approach to study the diffusion and convection of drug molecules into the lymphatic system through the hybrid vessel network.
Collapse
|
21
|
Goldberg AR, Ferguson M, Pal S, Cohen R, Orlicky DJ, McCullough RL, Rutkowski JM, Burchill MA, Tamburini BAJ. Oxidized low density lipoprotein in the liver causes decreased permeability of liver lymphatic- but not liver sinusoidal-endothelial cells via VEGFR-3 regulation of VE-Cadherin. Front Physiol 2022; 13:1021038. [PMID: 36338478 PMCID: PMC9626955 DOI: 10.3389/fphys.2022.1021038] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Accepted: 10/05/2022] [Indexed: 01/27/2023] Open
Abstract
The lymphatic vasculature of the liver is vital for liver function as it maintains fluid and protein homeostasis and is important for immune cell transport to the lymph node. Chronic liver disease is associated with increased expression of inflammatory mediators including oxidized low-density lipoprotein (oxLDL). Intrahepatic levels of oxLDL are elevated in nonalcoholic fatty liver disease (NAFLD), chronic hepatitis C infection (HCV), alcohol-associated liver disease (ALD), and cholestatic liver diseases. To determine if liver lymphatic function is impaired in chronic liver diseases, in which increased oxLDL has been documented, we measured liver lymphatic function in murine models of NAFLD, ALD and primary sclerosing cholangitis (PSC). We found that Mdr2-/- (PSC), Lieber-DeCarli ethanol fed (ALD) and high fat and high cholesterol diet fed (NAFLD) mice all had a significant impairment in the ability to traffic FITC labeled dextran from the liver parenchyma to the liver draining lymph nodes. Utilizing an in vitro permeability assay, we found that oxLDL decreased the permeability of lymphatic endothelial cells (LEC)s, but not liver sinusoidal endothelial cells (LSEC)s. Here we demonstrate that LECs and LSECs differentially regulate SRC-family kinases, MAPK kinase and VE-Cadherin in response to oxLDL. Furthermore, Vascular Endothelial Growth Factor (VEGF)C or D (VEGFR-3 ligands) appear to regulate VE-Cadherin expression as well as decrease cellular permeability of LECs in vitro and in vivo after oxLDL treatment. These findings suggest that oxLDL acts to impede protein transport through the lymphatics through tightening of the cell-cell junctions. Importantly, engagement of VEGFR-3 by its ligands prevents VE-Cadherin upregulation and improves lymphatic permeability. These studies provide a potential therapeutic target to restore liver lymphatic function and improve liver function.
Collapse
Affiliation(s)
- Alyssa R. Goldberg
- Department of Pediatrics, Section of Pediatric Gastroenterology, Hepatology & Nutrition. Children’s Hospital Colorado, Digestive Health Institute- Pediatric Liver Center, University of Colorado School of Medicine, Aurora, CO, United States
| | - Megan Ferguson
- Department of Medicine, Division of Gastroenterology and Hepatology, University of Colorado School of Medicine, Aurora, CO, United States
| | - Sarit Pal
- Department of Medicine, Division of Gastroenterology and Hepatology, University of Colorado School of Medicine, Aurora, CO, United States
| | - Rachel Cohen
- Department of Medicine, Division of Gastroenterology and Hepatology, University of Colorado School of Medicine, Aurora, CO, United States
| | - David J. Orlicky
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Rebecca L. McCullough
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado School of Medicine, Aurora, CO, United States
| | - Joseph M. Rutkowski
- Division of Lymphatic Biology, Department of Medical Physiology, Texas A&M University School of Medicine, Bryan, TX, United States
| | - Matthew A. Burchill
- Department of Medicine, Division of Gastroenterology and Hepatology, University of Colorado School of Medicine, Aurora, CO, United States
| | - Beth A. Jirón Tamburini
- Department of Medicine, Division of Gastroenterology and Hepatology, University of Colorado School of Medicine, Aurora, CO, United States
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, CO, United States
| |
Collapse
|
22
|
Scallan JP, Jannaway M. Lymphatic Vascular Permeability. Cold Spring Harb Perspect Med 2022; 12:a041274. [PMID: 35879102 PMCID: PMC9380735 DOI: 10.1101/cshperspect.a041274] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Blood vessels have a regulated permeability to fluid and solutes, which allows for the delivery of nutrients and signaling molecules to all cells in the body, a process essential to life. The lymphatic vasculature is the second network of vessels in the body, making up part of the immune system, yet is not typically thought of as having a permeability to fluid and solute. However, the major function of the lymphatic vasculature is to regulate tissue fluid balance to prevent edema, so lymphatic vessels must be permeable to absorb and transport fluid efficiently. Only recently were lymphatic vessels discovered to be permeable, which has had many functional implications. In this review, we will provide an overview of what is known about lymphatic vascular permeability, discuss the biophysical and signaling mechanisms regulating lymphatic permeability, and examine the disease relevance of this new property of lymphatic vessels.
Collapse
Affiliation(s)
- Joshua P Scallan
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, Tampa, Florida 33612, USA
| | - Melanie Jannaway
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, Tampa, Florida 33612, USA
| |
Collapse
|
23
|
Liu J, Yu C. Lymphangiogenesis and Lymphatic Barrier Dysfunction in Renal Fibrosis. Int J Mol Sci 2022; 23:ijms23136970. [PMID: 35805972 PMCID: PMC9267103 DOI: 10.3390/ijms23136970] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Revised: 06/20/2022] [Accepted: 06/20/2022] [Indexed: 11/16/2022] Open
Abstract
As an integral part of the vascular system, the lymphatic vasculature is essential for tissue fluid homeostasis, nutritional lipid assimilation and immune regulation. The composition of the lymphatic vasculature includes fluid-absorbing initial lymphatic vessels (LVs), transporting collecting vessels and anti-regurgitation valves. Although, in recent decades, research has drastically enlightened our view of LVs, investigations of initial LVs, also known as lymphatic capillaries, have been stagnant due to technical limitations. In the kidney, the lymphatic vasculature mainly presents in the cortex, keeping the local balance of fluid, solutes and immune cells. The contribution of renal LVs to various forms of pathology, especially chronic kidney diseases, has been addressed in previous studies, however with diverging and inconclusive results. In this review, we discuss the most recent advances in the proliferation and permeability of lymphatic capillaries as well as their influencing factors. Novel technologies to visualize and measure LVs function are described. Then, we highlight the role of the lymphatic network in renal fibrosis and the crosstalk between kidney and other organs, such as gut and heart.
Collapse
|
24
|
McCright J, Naiknavare R, Yarmovsky J, Maisel K. Targeting Lymphatics for Nanoparticle Drug Delivery. Front Pharmacol 2022; 13:887402. [PMID: 35721179 PMCID: PMC9203826 DOI: 10.3389/fphar.2022.887402] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Accepted: 05/16/2022] [Indexed: 12/25/2022] Open
Abstract
The lymphatics transport material from peripheral tissues to lymph nodes, where immune responses are formed, before being transported into systemic circulation. With key roles in transport and fluid homeostasis, lymphatic dysregulation is linked to diseases, including lymphedema. Fluid within the interstitium passes into initial lymphatic vessels where a valve system prevents fluid backflow. Additionally, lymphatic endothelial cells produce key chemokines, such as CCL21, that direct the migration of dendritic cells and lymphocytes. As a result, lymphatics are an attractive delivery route for transporting immune modulatory treatments to lymph nodes where immunotherapies are potentiated in addition to being an alternative method of reaching systemic circulation. In this review, we discuss the physiology of lymphatic vessels and mechanisms used in the transport of materials from peripheral tissues to lymph nodes. We then summarize nanomaterial-based strategies to take advantage of lymphatic transport functions for delivering therapeutics to lymph nodes or systemic circulation. We also describe opportunities for targeting lymphatic endothelial cells to modulate transport and immune functions.
Collapse
|
25
|
McCright J, Skeen C, Yarmovsky J, Maisel K. Nanoparticles with dense poly(ethylene glycol) coatings with near neutral charge are maximally transported across lymphatics and to the lymph nodes. Acta Biomater 2022; 145:146-158. [PMID: 35381399 PMCID: PMC9133124 DOI: 10.1016/j.actbio.2022.03.054] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Revised: 03/24/2022] [Accepted: 03/29/2022] [Indexed: 12/19/2022]
Abstract
Lymphatic vessels have recently been shown to effectively deliver immune modulatory therapies to the lymph nodes, which enhances their therapeutic efficacy. Prior work has shown that lymphatics transport 10-250 nm nanoparticles from peripheral tissues to the lymph node. However, the surface chemistry required to maximize this transport is poorly understood. Here, we determined the effect of surface poly(ethylene glycol) (PEG) density and size on nanoparticle transport across lymphatic endothelial cells (LECs) by differentially PEGylated model polystyrene nanoparticles. Using an established in-vitro lymphatic transport model, we found PEGylation improved the transport of 100 and 40 nm nanoparticles across LECs 50-fold compared to the unmodified nanoparticles and that transport is maximized when the PEG is in a dense brush conformation or high grafting density (Rf/D = 4.9). We also determined that these trends are not size-dependent. PEGylating 40 nm nanoparticles improved transport efficiency across LECs 68-fold compared to unmodified nanoparticles. We also found that PEGylated 100 nm and 40 nm nanoparticles accumulate in lymph nodes within 4 h after intradermal injection, while unmodified nanoparticles accumulated minimally. Densely PEGylated nanoparticles traveled the furthest distance from the injection site and densely PEGylated 40 nm nanoparticles had maximum accumulation in the lymph nodes compared to low density PEGylated and unmodified nanoparticles. Finally, we determined that nanoparticles are transported via both paracellular and transcellular mechanisms, and that PEG conformation modulates the cellular transport mechanisms. Our results suggest that PEG conformation is crucial to maximize nanoparticle transport across LECs and into lymphatic vessels, making PEG density a crucial design. Optimizing PEG density on nanoparticle formulations has the potential to enhance immunotherapeutic and vaccine outcomes. STATEMENT OF SIGNIFICANCE: Lymphatic vessels are an emerging target for drug delivery both in the context of modulating immune responses and enhancing bioavailability by avoiding first pass hepatic metabolism after oral delivery. Lymphatic vessels are the natural conduits from peripheral tissues to the lymph nodes, where the adaptive immune response is shaped, and eventually to systemic circulation via the thoracic duct. Lymphatics can be targeted via nanoparticles, but the surface chemistry required to maximize nanoparticle transport by lymphatics vessels remains poorly understood. Here, we demonstrate that coating nanoparticles with hydrophilic polyethylene glycol (PEG) effectively enhances their transport across lymphatic endothelial cells in vitro and in vivo and that both paracellular and micropinocytosis mechanisms underly this transport. We found that dense PEG coatings maximize lymphatic transport of nanoparticles, thus providing new material design criteria for lymphatic targeted drug delivery.
Collapse
Affiliation(s)
- Jacob McCright
- Department of Bioengineering, University of Maryland College Park, College Park, 8278 Paint Branch Drive, MD 20742, USA
| | - Colin Skeen
- Department of Bioengineering, University of Maryland College Park, College Park, 8278 Paint Branch Drive, MD 20742, USA
| | - Jenny Yarmovsky
- Department of Bioengineering, University of Maryland College Park, College Park, 8278 Paint Branch Drive, MD 20742, USA
| | - Katharina Maisel
- Department of Bioengineering, University of Maryland College Park, College Park, 8278 Paint Branch Drive, MD 20742, USA.
| |
Collapse
|
26
|
Cai N, Lai ACK, Liao K, Corridon PR, Graves DJ, Chan V. Recent Advances in Fluorescence Recovery after Photobleaching for Decoupling Transport and Kinetics of Biomacromolecules in Cellular Physiology. Polymers (Basel) 2022; 14:1913. [PMID: 35567083 PMCID: PMC9105003 DOI: 10.3390/polym14091913] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Revised: 04/27/2022] [Accepted: 04/29/2022] [Indexed: 12/16/2022] Open
Abstract
Among the new molecular tools available to scientists and engineers, some of the most useful include fluorescently tagged biomolecules. Tools, such as green fluorescence protein (GFP), have been applied to perform semi-quantitative studies on biological signal transduction and cellular structural dynamics involved in the physiology of healthy and disease states. Such studies focus on drug pharmacokinetics, receptor-mediated endocytosis, nuclear mechanobiology, viral infections, and cancer metastasis. In 1976, fluorescence recovery after photobleaching (FRAP), which involves the monitoring of fluorescence emission recovery within a photobleached spot, was developed. FRAP allowed investigators to probe two-dimensional (2D) diffusion of fluorescently-labelled biomolecules. Since then, FRAP has been refined through the advancements of optics, charged-coupled-device (CCD) cameras, confocal microscopes, and molecular probes. FRAP is now a highly quantitative tool used for transport and kinetic studies in the cytosol, organelles, and membrane of a cell. In this work, the authors intend to provide a review of recent advances in FRAP. The authors include epifluorescence spot FRAP, total internal reflection (TIR)/FRAP, and confocal microscope-based FRAP. The underlying mathematical models are also described. Finally, our understanding of coupled transport and kinetics as determined by FRAP will be discussed and the potential for future advances suggested.
Collapse
Affiliation(s)
- Ning Cai
- Wuhan Institute of Technology, School of Chemical Engineering and Pharmacy, Wuhan 430073, China;
| | - Alvin Chi-Keung Lai
- Department of Architecture and Civil Engineering, City University of Hong Kong, Tat Chee Avenue, Kowloon Tong, Hong Kong 999077, China;
| | - Kin Liao
- Department of Aerospace Engineering, Khalifa University of Science and Technology, Abu Dhabi P.O. Box 127788, United Arab Emirates;
| | - Peter R. Corridon
- Department of Physiology and Immunology, Khalifa University of Science and Technology, Abu Dhabi P.O. Box 127788, United Arab Emirates;
- Healthcare Engineering Innovation Center, Khalifa University of Science and Technology, Abu Dhabi P.O. Box 127788, United Arab Emirates
- Center for Biotechnology, Khalifa University of Science and Technology, Abu Dhabi P.O. Box 127788, United Arab Emirates
| | - David J. Graves
- Department of Chemical and Biomolecular Engineering, University of Pennsylvania, Philadelphia, PA 19104, USA;
| | - Vincent Chan
- Department of Biomedical Engineering, Khalifa University of Science and Technology, Abu Dhabi P.O. Box 127788, United Arab Emirates
| |
Collapse
|
27
|
Hypoxia orchestrates the lymphovascular–immune ensemble in cancer. Trends Cancer 2022; 8:771-784. [DOI: 10.1016/j.trecan.2022.04.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Revised: 03/31/2022] [Accepted: 04/25/2022] [Indexed: 11/18/2022]
|
28
|
Lee Y, Zawieja SD, Muthuchamy M. Lymphatic Collecting Vessel: New Perspectives on Mechanisms of Contractile Regulation and Potential Lymphatic Contractile Pathways to Target in Obesity and Metabolic Diseases. Front Pharmacol 2022; 13:848088. [PMID: 35355722 PMCID: PMC8959455 DOI: 10.3389/fphar.2022.848088] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Accepted: 02/17/2022] [Indexed: 01/19/2023] Open
Abstract
Obesity and metabolic syndrome pose a significant risk for developing cardiovascular disease and remain a critical healthcare challenge. Given the lymphatic system's role as a nexus for lipid absorption, immune cell trafficking, interstitial fluid and macromolecule homeostasis maintenance, the impact of obesity and metabolic disease on lymphatic function is a burgeoning field in lymphatic research. Work over the past decade has progressed from the association of an obese phenotype with Prox1 haploinsufficiency and the identification of obesity as a risk factor for lymphedema to consistent findings of lymphatic collecting vessel dysfunction across multiple metabolic disease models and organisms and characterization of obesity-induced lymphedema in the morbidly obese. Critically, recent findings have suggested that restoration of lymphatic function can also ameliorate obesity and insulin resistance, positing lymphatic targeted therapies as relevant pharmacological interventions. There remain, however, significant gaps in our understanding of lymphatic collecting vessel function, particularly the mechanisms that regulate the spontaneous contractile activity required for active lymph propulsion and lymph return in humans. In this article, we will review the current findings on lymphatic architecture and collecting vessel function, including recent advances in the ionic basis of lymphatic muscle contractile activity. We will then discuss lymphatic dysfunction observed with metabolic disruption and potential pathways to target with pharmacological approaches to improve lymphatic collecting vessel function.
Collapse
Affiliation(s)
- Yang Lee
- Department of Medical Physiology, College of Medicine, Texas A&M University, Bryan, TX, United States
| | - Scott D Zawieja
- Medical Pharmacology and Physiology, School of Medicine, University of Missouri, Columbia, MO, United States
| | - Mariappan Muthuchamy
- Department of Medical Physiology, College of Medicine, Texas A&M University, Bryan, TX, United States
| |
Collapse
|
29
|
Jones JH, Minshall RD. Endothelial Transcytosis in Acute Lung Injury: Emerging Mechanisms and Therapeutic Approaches. Front Physiol 2022; 13:828093. [PMID: 35431977 PMCID: PMC9008570 DOI: 10.3389/fphys.2022.828093] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Accepted: 02/28/2022] [Indexed: 01/08/2023] Open
Abstract
Acute Lung Injury (ALI) is characterized by widespread inflammation which in its severe form, Acute Respiratory Distress Syndrome (ARDS), leads to compromise in respiration causing hypoxemia and death in a substantial number of affected individuals. Loss of endothelial barrier integrity, pneumocyte necrosis, and circulating leukocyte recruitment into the injured lung are recognized mechanisms that contribute to the progression of ALI/ARDS. Additionally, damage to the pulmonary microvasculature by Gram-negative and positive bacteria or viruses (e.g., Escherichia coli, SARS-Cov-2) leads to increased protein and fluid permeability and interstitial edema, further impairing lung function. While most of the vascular leakage is attributed to loss of inter-endothelial junctional integrity, studies in animal models suggest that transendothelial transport of protein through caveolar vesicles, known as transcytosis, occurs in the early phase of ALI/ARDS. Here, we discuss the role of transcytosis in healthy and injured endothelium and highlight recent studies that have contributed to our understanding of the process during ALI/ARDS. We also cover potential approaches that utilize caveolar transport to deliver therapeutics to the lungs which may prevent further injury or improve recovery.
Collapse
Affiliation(s)
- Joshua H. Jones
- Department of Pharmacology, University of Illinois College of Medicine at Chicago, Chicago, IL, United States
| | - Richard D. Minshall
- Department of Pharmacology, University of Illinois College of Medicine at Chicago, Chicago, IL, United States,Department of Anesthesiology, University of Illinois College of Medicine at Chicago, Chicago, IL, United States,*Correspondence: Richard D. Minshall,
| |
Collapse
|
30
|
Summers BD, Kim K, Clement CC, Khan Z, Thangaswamy S, McCright J, Maisel K, Zamora S, Quintero S, Racanelli AC, Redmond D, D'Armiento J, Yang J, Kuang A, Monticelli L, Kahn ML, Choi AMK, Santambrogio L, Reed HO. Lung lymphatic thrombosis and dysfunction caused by cigarette smoke exposure precedes emphysema in mice. Sci Rep 2022; 12:5012. [PMID: 35322079 PMCID: PMC8943143 DOI: 10.1038/s41598-022-08617-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Accepted: 03/09/2022] [Indexed: 11/21/2022] Open
Abstract
The lymphatic vasculature is critical for lung function, but defects in lymphatic function in the pathogenesis of lung disease is understudied. In mice, lymphatic dysfunction alone is sufficient to cause lung injury that resembles human emphysema. Whether lymphatic function is disrupted in cigarette smoke (CS)-induced emphysema is unknown. In this study, we investigated the effect of CS on lung lymphatic function. Analysis of human lung tissue revealed significant lung lymphatic thrombosis in patients with emphysema compared to control smokers that increased with disease severity. In a mouse model, CS exposure led to lung lymphatic thrombosis, decreased lymphatic drainage, and impaired leukocyte trafficking that all preceded the development of emphysema. Proteomic analysis demonstrated an increased abundance of coagulation factors in the lymph draining from the lungs of CS-exposed mice compared to control mice. In addition, in vitro assays demonstrated a direct effect of CS on lymphatic endothelial cell integrity. These data show that CS exposure results in lung lymphatic dysfunction and a shift in thoracic lymph towards a prothrombic state. Furthermore, our data suggest that lymphatic dysfunction is due to effects of CS on the lymphatic vasculature that precede emphysema. These studies demonstrate a novel component of CS-induced lung injury that occurs early in the pathogenesis of emphysema.
Collapse
Affiliation(s)
| | - Kihwan Kim
- Department of Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Cristina C Clement
- Department of Radiation Oncology and Caryl and Israel Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Zohaib Khan
- Department of Radiation Oncology and Caryl and Israel Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Sangeetha Thangaswamy
- Department of Radiation Oncology and Caryl and Israel Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Jacob McCright
- Fischell Department of Bioengineering, University of Maryland, College Park, MD, USA
| | - Katharina Maisel
- Fischell Department of Bioengineering, University of Maryland, College Park, MD, USA
| | - Sofia Zamora
- Department of Medicine, Weill Cornell Medicine, New York, NY, USA
| | | | | | - David Redmond
- Ansary Stem Cell Institute, Division of Regenerative Medicine, Department of Medicine, Weill Cornell Medicine, New York, USA
| | - Jeanine D'Armiento
- Department of Medicine in Anesthesiology, Columbia University, New York, NY, USA
| | - Jisheng Yang
- Department of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Amy Kuang
- Department of Medicine, Weill Cornell Medicine, New York, NY, USA
| | | | - Mark L Kahn
- Department of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | | | - Laura Santambrogio
- Department of Radiation Oncology and Caryl and Israel Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Hasina Outtz Reed
- Department of Medicine, Weill Cornell Medicine, New York, NY, USA.
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Weill Cornell Medicine, 1300 York Ave, Room 323, New York, NY, 10065, USA.
| |
Collapse
|
31
|
Kolarzyk AM, Wong G, Lee E. Lymphatic Tissue and Organ Engineering for In Vitro Modeling and In Vivo Regeneration. Cold Spring Harb Perspect Med 2022; 12:a041169. [PMID: 35288402 PMCID: PMC9435571 DOI: 10.1101/cshperspect.a041169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
The lymphatic system has an important role in maintaining fluid homeostasis and transporting immune cells and biomolecules, such as dietary fat, metabolic products, and antigens in different organs and tissues. Therefore, impaired lymphatic vessel function and/or lymphatic vessel deficiency can lead to numerous human diseases. The discovery of lymphatic endothelial markers and prolymphangiogenic growth factors, along with a growing number of in vitro and in vivo models and technologies has expedited research in lymphatic tissue and organ engineering, advancing therapeutic strategies. In this article, we describe lymphatic tissue and organ engineering in two- and three-dimensional culture systems and recently developed microfluidics and organ-on-a-chip systems in vitro. Next, we discuss advances in lymphatic tissue and organ engineering in vivo, focusing on biomaterial and scaffold engineering and their applications for lymphatic vessels and lymphoid organ regeneration. Last, we provide expert perspective and prospects in the field of lymphatic tissue engineering.
Collapse
Affiliation(s)
- Anna M Kolarzyk
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Ithaca, New York 14853, USA
- Biomedical and Biological Sciences PhD Program, Ithaca, New York 14853, USA
| | - Gigi Wong
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Ithaca, New York 14853, USA
- Biological Sciences, Cornell University, Ithaca, New York 14853, USA
| | - Esak Lee
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Ithaca, New York 14853, USA
- Biomedical and Biological Sciences PhD Program, Ithaca, New York 14853, USA
| |
Collapse
|
32
|
Lee GH, Huang SA, Aw WY, Rathod M, Cho C, Ligler FS, Polacheck WJ. Multilayer microfluidic platform for the study of luminal, transmural, and interstitial flow. Biofabrication 2022; 14:10.1088/1758-5090/ac48e5. [PMID: 34991082 PMCID: PMC8867496 DOI: 10.1088/1758-5090/ac48e5] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Accepted: 01/06/2022] [Indexed: 01/27/2023]
Abstract
Efficient delivery of oxygen and nutrients to tissues requires an intricate balance of blood, lymphatic, and interstitial fluid pressures (IFPs), and gradients in fluid pressure drive the flow of blood, lymph, and interstitial fluid through tissues. While specific fluid mechanical stimuli, such as wall shear stress, have been shown to modulate cellular signaling pathways along with gene and protein expression patterns, an understanding of the key signals imparted by flowing fluid and how these signals are integrated across multiple cells and cell types in native tissues is incomplete due to limitations with current assays. Here, we introduce a multi-layer microfluidic platform (MμLTI-Flow) that enables the culture of engineered blood and lymphatic microvessels and independent control of blood, lymphatic, and IFPs. Using optical microscopy methods to measure fluid velocity for applied input pressures, we demonstrate varying rates of interstitial fluid flow as a function of blood, lymphatic, and interstitial pressure, consistent with computational fluid dynamics (CFD) models. The resulting microfluidic and computational platforms will provide for analysis of key fluid mechanical parameters and cellular mechanisms that contribute to diseases in which fluid imbalances play a role in progression, including lymphedema and solid cancer.
Collapse
Affiliation(s)
- Gi-hun Lee
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University
| | - Stephanie A. Huang
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University
| | - Wen Y. Aw
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University
| | - Mitesh Rathod
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University
| | - Crescentia Cho
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University
| | - Frances S. Ligler
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University
| | - William J. Polacheck
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University,Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill,McAllister Heart Institute, University of North Carolina at Chapel Hill
| |
Collapse
|
33
|
Harris NR, Nielsen NR, Pawlak JB, Aghajanian A, Rangarajan K, Serafin DS, Farber G, Dy DM, Nelson-Maney NP, Xu W, Ratra D, Hurr SH, Qian L, Scallan JP, Caron KM. VE-Cadherin Is Required for Cardiac Lymphatic Maintenance and Signaling. Circ Res 2022; 130:5-23. [PMID: 34789016 PMCID: PMC8756423 DOI: 10.1161/circresaha.121.318852] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
BACKGROUND The adherens protein VE-cadherin (vascular endothelial cadherin) has diverse roles in organ-specific lymphatic vessels. However, its physiological role in cardiac lymphatics and its interaction with lymphangiogenic factors has not been fully explored. We sought to determine the spatiotemporal functions of VE-cadherin in cardiac lymphatics and mechanistically elucidate how VE-cadherin loss influences prolymphangiogenic signaling pathways, such as adrenomedullin and VEGF (vascular endothelial growth factor)-C/VEGFR3 (vascular endothelial growth factor receptor 3) signaling. METHODS Cdh5flox/flox;Prox1CreERT2 mice were used to delete VE-cadherin in lymphatic endothelial cells across life stages, including embryonic, postnatal, and adult. Lymphatic architecture and function was characterized using immunostaining and functional lymphangiography. To evaluate the impact of temporal and functional regression of cardiac lymphatics in Cdh5flox/flox;Prox1CreERT2 mice, left anterior descending artery ligation was performed and cardiac function and repair after myocardial infarction was evaluated by echocardiography and histology. Cellular effects of VE-cadherin deletion on lymphatic signaling pathways were assessed by knockdown of VE-cadherin in cultured lymphatic endothelial cells. RESULTS Embryonic deletion of VE-cadherin produced edematous embryos with dilated cardiac lymphatics with significantly altered vessel tip morphology. Postnatal deletion of VE-cadherin caused complete disassembly of cardiac lymphatics. Adult deletion caused a temporal regression of the quiescent epicardial lymphatic network which correlated with significant dermal and cardiac lymphatic dysfunction, as measured by fluorescent and quantum dot lymphangiography, respectively. Surprisingly, despite regression of cardiac lymphatics, Cdh5flox/flox;Prox1CreERT2 mice exhibited preserved cardiac function, both at baseline and following myocardial infarction, compared with control mice. Mechanistically, loss of VE-cadherin leads to aberrant cellular internalization of VEGFR3, precluding the ability of VEGFR3 to be either canonically activated by VEGF-C or noncanonically transactivated by adrenomedullin signaling, impairing downstream processes such as cellular proliferation. CONCLUSIONS VE-cadherin is an essential scaffolding protein to maintain prolymphangiogenic signaling nodes at the plasma membrane, which are required for the development and adult maintenance of cardiac lymphatics, but not for cardiac function basally or after injury.
Collapse
Affiliation(s)
- Natalie R. Harris
- Department of Cell Biology and Physiology, University of
North Carolina at Chapel Hill; 111 Mason Farm Road, Chapel Hill, North Carolina, USA
27599
| | - Natalie R. Nielsen
- Department of Cell Biology and Physiology, University of
North Carolina at Chapel Hill; 111 Mason Farm Road, Chapel Hill, North Carolina, USA
27599
| | - John B. Pawlak
- Department of Cell Biology and Physiology, University of
North Carolina at Chapel Hill; 111 Mason Farm Road, Chapel Hill, North Carolina, USA
27599
| | - Amir Aghajanian
- Department of Medicine Division of Cardiology, University
of North Carolina at Chapel Hill; 160 Dental Circle, Chapel Hill, North Carolina,
USA 27599
| | - Krsna Rangarajan
- Department of Cell Biology and Physiology, University of
North Carolina at Chapel Hill; 111 Mason Farm Road, Chapel Hill, North Carolina, USA
27599
| | - D. Stephen Serafin
- Department of Cell Biology and Physiology, University of
North Carolina at Chapel Hill; 111 Mason Farm Road, Chapel Hill, North Carolina, USA
27599
| | - Gregory Farber
- Department of Pathology and Laboratory Medicine, University
of North Carolina at Chapel Hill; 111 Mason Farm Road, Chapel Hill, North Carolina,
USA 27599,McAllister Heart Institute, University of North Carolina,
Chapel Hill, North Carolina, USA 27599
| | - Danielle M. Dy
- Department of Cell Biology and Physiology, University of
North Carolina at Chapel Hill; 111 Mason Farm Road, Chapel Hill, North Carolina, USA
27599
| | - Nathan P. Nelson-Maney
- Department of Cell Biology and Physiology, University of
North Carolina at Chapel Hill; 111 Mason Farm Road, Chapel Hill, North Carolina, USA
27599
| | - Wenjing Xu
- Department of Cell Biology and Physiology, University of
North Carolina at Chapel Hill; 111 Mason Farm Road, Chapel Hill, North Carolina, USA
27599
| | - Disha Ratra
- Department of Cell Biology and Physiology, University of
North Carolina at Chapel Hill; 111 Mason Farm Road, Chapel Hill, North Carolina, USA
27599
| | - Sophia H. Hurr
- Department of Cell Biology and Physiology, University of
North Carolina at Chapel Hill; 111 Mason Farm Road, Chapel Hill, North Carolina, USA
27599
| | - Li Qian
- Department of Pathology and Laboratory Medicine, University
of North Carolina at Chapel Hill; 111 Mason Farm Road, Chapel Hill, North Carolina,
USA 27599
| | - Joshua P. Scallan
- Department of Molecular Pharmacology and Physiology,
University of South Florida, Tampa, Florida, USA 33612
| | - Kathleen M. Caron
- Department of Cell Biology and Physiology, University of
North Carolina at Chapel Hill; 111 Mason Farm Road, Chapel Hill, North Carolina, USA
27599
| |
Collapse
|
34
|
Kim J, Archer PA, Thomas SN. Innovations in lymph node targeting nanocarriers. Semin Immunol 2021; 56:101534. [PMID: 34836772 DOI: 10.1016/j.smim.2021.101534] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 11/11/2021] [Accepted: 11/18/2021] [Indexed: 12/19/2022]
Abstract
Lymph nodes are secondary lymphoid tissues in the body that facilitate the co-mingling of immune cells to enable and regulate the adaptive immune response. They are also tissues implicated in a variety of diseases, including but not limited to malignancy. The ability to access lymph nodes is thus attractive for a variety of therapeutic and diagnostic applications. As nanotechnologies are now well established for their potential in translational biomedical applications, their high relevance to applications that involve lymph nodes is highlighted. Herein, established paradigms of nanocarrier design to enable delivery to lymph nodes are discussed, considering the unique lymph node tissue structure as well as lymphatic system physiology. The influence of delivery mechanism on how nanocarrier systems distribute to different compartments and cells that reside within lymph nodes is also elaborated. Finally, current advanced nanoparticle technologies that have been developed to enable lymph node delivery are discussed.
Collapse
Affiliation(s)
- Jihoon Kim
- Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, 315 Ferst Dr NW, Atlanta, GA 30332, USA; George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, 315 Ferst Dr NW, Atlanta, GA 30332, USA
| | - Paul A Archer
- Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, 315 Ferst Dr NW, Atlanta, GA 30332, USA; School of Chemical and Biomolecular Engineering, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Susan N Thomas
- Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, 315 Ferst Dr NW, Atlanta, GA 30332, USA; George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, 315 Ferst Dr NW, Atlanta, GA 30332, USA; Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, 313 Ferst Dr NW, Atlanta, GA 30332, USA; Emory University, 201 Dowman Drive, Atlanta, GA 30322, USA; Winship Cancer Institute, Emory University School of Medicine, 1365-C Clifton Road NE, Atlanta, GA 30322, USA.
| |
Collapse
|
35
|
Cho Y, Na K, Jun Y, Won J, Yang JH, Chung S. Three-Dimensional In Vitro Lymphangiogenesis Model in Tumor Microenvironment. Front Bioeng Biotechnol 2021; 9:697657. [PMID: 34671596 PMCID: PMC8520924 DOI: 10.3389/fbioe.2021.697657] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Accepted: 08/30/2021] [Indexed: 11/25/2022] Open
Abstract
Lymphangiogenesis is a stage of new lymphatic vessel formation in development and pathology, such as inflammation and tumor metastasis. Physiologically relevant models of lymphatic vessels have been in demand because studies on lymphatic vessels are required for understanding the mechanism of tumor metastasis. In this study, a new three-dimensional lymphangiogenesis model in a tumor microenvironment is proposed, using a newly designed macrofluidic platform. It is verified that controllable biochemical and biomechanical cues, which contribute to lymphangiogenesis, can be applied in this platform. In particular, this model demonstrates that a reconstituted lymphatic vessel has an in vivo–like lymphatic vessel in both physical and biochemical aspects. Since biomechanical stress with a biochemical factor influences robust directional lymphatic sprouting, whether our model closely approximates in vivo, the initial lymphatics in terms of the morphological and genetic signatures is investigated. Furthermore, attempting an incorporation with a tumor spheroid, this study successfully develops a complex tumor microenvironment model for use in lymphangiogenesis and reveals the microenvironment factors that contribute to tumor metastasis. As a first attempt at a coculture model, this reconstituted model is a novel system with a fully three-dimensional structure and can be a powerful tool for pathological drug screening or disease model.
Collapse
Affiliation(s)
- Youngkyu Cho
- Department of IT Convergence, Korea University, Seoul, South Korea.,Samsung Research, Samsung Electronics Co. Ltd., Seoul, South Korea
| | - Kyuhwan Na
- School of Mechanical Engineering, Korea University, Seoul, South Korea
| | - Yesl Jun
- Departments of Pediatrics and Cellular & Molecular Medicine, Pediatric Diabetes Research Center, University of California, La Jolla, CA, United States.,Drug Discovery Platform Research Center, Therapeutics and Biotechnology Division, Korea Research Institute of Chemical Technology, Daejeon, South Korea
| | - Jihee Won
- School of Mechanical Engineering, Korea University, Seoul, South Korea
| | - Ji Hun Yang
- School of Mechanical Engineering, Korea University, Seoul, South Korea.,Next&Bio Inc., Seoul, South Korea
| | - Seok Chung
- Department of IT Convergence, Korea University, Seoul, South Korea.,School of Mechanical Engineering, Korea University, Seoul, South Korea
| |
Collapse
|
36
|
Jannaway M, Scallan JP. VE-Cadherin and Vesicles Differentially Regulate Lymphatic Vascular Permeability to Solutes of Various Sizes. Front Physiol 2021; 12:687563. [PMID: 34621180 PMCID: PMC8491776 DOI: 10.3389/fphys.2021.687563] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Accepted: 08/24/2021] [Indexed: 01/04/2023] Open
Abstract
Lymphatic vascular permeability prevents lymph leakage that is associated with lymphedema, lymphatic malformations, obesity, and inflammation. However, the molecular control of lymphatic permeability remains poorly understood. Recent studies have suggested that adherens junctions and vesicle transport may be involved in regulating lymphatic vessel permeability. To determine the contribution of each transport pathway, we utilized an ex vivo permeability assay to directly measure the solute flux of various molecular weight solutes across a range of pressures in intact murine collecting lymphatic vessels. Pharmacological and biological tools were used to probe the relative contributions of vesicles and junction proteins in the lymphatic vasculature. We show that the permeability of collecting lymphatic vessels is inversely related to the solute molecular weight. Further, our data reveal that vesicles selectively transport BSA, as an inhibitor of vesicle formation significantly decreased the permeability to BSA (∼60% decrease, n = 8, P = 0.02), but not to 3 kDa dextran (n = 7, P = 0.41), α-lactalbumin (n = 5, P = 0.26) or 70 kDa dextran (n = 8, P = 0.13). In contrast, disruption of VE-cadherin binding with a function blocking antibody significantly increased lymphatic vessel permeability to both 3 kDa dextran (5.7-fold increase, n = 5, P < 0.0001) and BSA (5.8-fold increase, n = 5, P < 0.0001). Thus, in the lymphatic vasculature, adherens junctions did not exhibit selectivity for any of the solutes tested here, whereas vesicles specifically transport BSA. Overall, the findings suggest that disease states that disrupt VE-cadherin localization or expression will cause significant leakage of solutes and fluid from the lymphatic vasculature.
Collapse
Affiliation(s)
- Melanie Jannaway
- Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, Tampa, FL, United States
| | - Joshua P Scallan
- Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, Tampa, FL, United States
| |
Collapse
|
37
|
Henderson AR, Ilan IS, Lee E. A bioengineered lymphatic vessel model for studying lymphatic endothelial cell-cell junction and barrier function. Microcirculation 2021; 28:e12730. [PMID: 34569678 DOI: 10.1111/micc.12730] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Revised: 09/09/2021] [Accepted: 09/20/2021] [Indexed: 12/15/2022]
Abstract
OBJECTIVE Lymphatic vessels (LVs) maintain fluid homeostasis by draining interstitial fluid. A failure in lymphatic drainage triggers lymphatic diseases such as lymphedema. Since lymphatic drainage is regulated by lymphatic barrier function, developing experimental models that assess lymphatic barrier function is critical for better understanding of lymphatic physiology and disease. METHODS We built a lymphatic vessel-on-chip (LV-on-chip) by fabricating a microfluidic device that includes a hollow microchannel embedded in three-dimensional (3D) hydrogel. Employing luminal flow in the microchannel, human lymphatic endothelial cells (LECs) seeded in the microchannel formed an engineered LV exhibiting 3D conduit structure. RESULTS Lymphatic endothelial cells formed relatively permeable junctions in 3D collagen 1. However, adding fibronectin to the collagen 1 apparently tightened LEC junctions. We tested lymphatic barrier function by introducing dextran into LV lumens. While LECs in collagen 1 showed permeable barriers, LECs in fibronectin/collagen 1 showed reduced permeability, which was reversed by integrin α5 inhibition. Mechanistically, LECs expressed inactivated integrin α5 in collagen 1. However, integrin α5 is activated in fibronectin and enhances barrier function. Integrin α5 activation itself also tightened LEC junctions in the absence of fibronectin. CONCLUSIONS Lymphatic vessel-on-chip reveals integrin α5 as a regulator of lymphatic barrier function and provides a platform for studying lymphatic barrier function in various conditions.
Collapse
Affiliation(s)
- Aria R Henderson
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, New York, USA
| | - Isabelle S Ilan
- College of Human Ecology, Cornell University, Ithaca, New York, USA
| | - Esak Lee
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, New York, USA
| |
Collapse
|
38
|
Geng X, Ho YC, Srinivasan RS. Biochemical and mechanical signals in the lymphatic vasculature. Cell Mol Life Sci 2021; 78:5903-5923. [PMID: 34240226 PMCID: PMC11072415 DOI: 10.1007/s00018-021-03886-8] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Revised: 06/15/2021] [Accepted: 06/18/2021] [Indexed: 12/15/2022]
Abstract
Lymphatic vasculature is an integral part of the cardiovascular system where it maintains interstitial fluid balance. Additionally, lymphatic vasculature regulates lipid assimilation and inflammatory response. Lymphatic vasculature is composed of lymphatic capillaries, collecting lymphatic vessels and valves that function in synergy to absorb and transport fluid against gravitational and pressure gradients. Defects in lymphatic vessels or valves leads to fluid accumulation in tissues (lymphedema), chylous ascites, chylothorax, metabolic disorders and inflammation. The past three decades of research has identified numerous molecules that are necessary for the stepwise development of lymphatic vasculature. However, approaches to treat lymphatic disorders are still limited to massages and compression bandages. Hence, better understanding of the mechanisms that regulate lymphatic vascular development and function is urgently needed to develop efficient therapies. Recent research has linked mechanical signals such as shear stress and matrix stiffness with biochemical pathways that regulate lymphatic vessel growth, patterning and maturation and valve formation. The goal of this review article is to highlight these innovative developments and speculate on unanswered questions.
Collapse
Affiliation(s)
- Xin Geng
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, 73013, USA
| | - Yen-Chun Ho
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, 73013, USA
| | - R Sathish Srinivasan
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, 73013, USA.
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73117, USA.
| |
Collapse
|
39
|
Wiig H. As for blood vessels, the answer regarding lymphatics is often NO. Acta Physiol (Oxf) 2021; 232:e13697. [PMID: 34057826 DOI: 10.1111/apha.13697] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Affiliation(s)
- Helge Wiig
- Department of Biomedicine University of Bergen Bergen Norway
| |
Collapse
|
40
|
Baranwal G, Creed HA, Cromer WE, Wang W, Upchurch BD, Smithhart MC, Vadlamani SS, Clark MC, Busbuso NC, Blais SN, Reyna AJ, Dongaonkar RM, Zawieja DC, Rutkowski JM. Dichotomous effects on lymphatic transport with loss of caveolae in mice. Acta Physiol (Oxf) 2021; 232:e13656. [PMID: 33793057 DOI: 10.1111/apha.13656] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Revised: 03/26/2021] [Accepted: 03/29/2021] [Indexed: 11/27/2022]
Abstract
AIM Fluid and macromolecule transport from the interstitium into and through lymphatic vessels is necessary for tissue homeostasis. While lymphatic capillary structure suggests that passive, paracellular transport would be the predominant route of macromolecule entry, active caveolae-mediated transcellular transport has been identified in lymphatic endothelial cells (LECs) in vitro. Caveolae also mediate a wide array of endothelial cell processes, including nitric oxide regulation. Thus, how does the lack of caveolae impact "lymphatic function"? METHODS Various aspects of lymphatic transport were measured in mice constitutively lacking caveolin-1 ("CavKO"), the protein required for caveolae formation in endothelial cells, and in mice with a LEC-specific Cav1 gene deletion (Lyve1-Cre x Cav1flox/flox ; "LyCav") and ex vivo in their vessels and cells. RESULTS In each model, lymphatic architecture was largely unchanged. The lymphatic conductance, or initial tissue uptake, was significantly higher in both CavKO mice and LyCav mice by quantitative microlymphangiography and the permeability to 70 kDa dextran was significantly increased in monolayers of LECs isolated from CavKO mice. Conversely, transport within the lymphatic system to the sentinel node was significantly reduced in anaesthetized CavKO and LyCav mice. Isolated, cannulated collecting vessel studies identified significantly reduced phasic contractility when lymphatic endothelium lacks caveolae. Inhibition of nitric oxide synthase was able to partially restore ex vivo vessel contractility. CONCLUSION Macromolecule transport across lymphatics is increased with loss of caveolae, yet phasic contractility reduced, resulting in reduced overall lymphatic transport function. These studies identify lymphatic caveolar biology as a key regulator of active lymphatic transport functions.
Collapse
Affiliation(s)
- Gaurav Baranwal
- Division of Lymphatic Biology Department of Medical Physiology Texas A&M University College of Medicine Bryan TX USA
| | - Heidi A. Creed
- Division of Lymphatic Biology Department of Medical Physiology Texas A&M University College of Medicine Bryan TX USA
| | - Walter E. Cromer
- Division of Lymphatic Biology Department of Medical Physiology Texas A&M University College of Medicine Bryan TX USA
| | - Wei Wang
- Division of Lymphatic Biology Department of Medical Physiology Texas A&M University College of Medicine Bryan TX USA
| | - Bradley D. Upchurch
- Division of Lymphatic Biology Department of Medical Physiology Texas A&M University College of Medicine Bryan TX USA
| | - Matt C. Smithhart
- Division of Lymphatic Biology Department of Medical Physiology Texas A&M University College of Medicine Bryan TX USA
| | - Suman S. Vadlamani
- Division of Lymphatic Biology Department of Medical Physiology Texas A&M University College of Medicine Bryan TX USA
| | - Mary‐Catherine C. Clark
- Division of Lymphatic Biology Department of Medical Physiology Texas A&M University College of Medicine Bryan TX USA
| | | | - Stephanie N. Blais
- Division of Lymphatic Biology Department of Medical Physiology Texas A&M University College of Medicine Bryan TX USA
| | - Andrea J. Reyna
- Division of Lymphatic Biology Department of Medical Physiology Texas A&M University College of Medicine Bryan TX USA
| | - Ranjeet M. Dongaonkar
- Department of Veterinary Physiology & Pharmacology Texas A&M University College of Veterinary Medicine & Biomedical Sciences College Station TX USA
| | - David C. Zawieja
- Division of Lymphatic Biology Department of Medical Physiology Texas A&M University College of Medicine Bryan TX USA
| | - Joseph M. Rutkowski
- Division of Lymphatic Biology Department of Medical Physiology Texas A&M University College of Medicine Bryan TX USA
| |
Collapse
|
41
|
Stritt S, Koltowska K, Mäkinen T. Homeostatic maintenance of the lymphatic vasculature. Trends Mol Med 2021; 27:955-970. [PMID: 34332911 DOI: 10.1016/j.molmed.2021.07.003] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Revised: 06/30/2021] [Accepted: 07/06/2021] [Indexed: 12/24/2022]
Abstract
The lymphatic vasculature is emerging as a multifaceted regulator of tissue homeostasis and regeneration. Lymphatic vessels drain fluid, macromolecules, and immune cells from peripheral tissues to lymph nodes (LNs) and the systemic circulation. Their recently uncovered functions extend beyond drainage and include direct modulation of adaptive immunity and paracrine regulation of organ growth. The developmental mechanisms controlling lymphatic vessel growth have been described with increasing precision. It is less clear how the essential functional features of lymphatic vessels are established and maintained. We discuss the mechanisms that maintain lymphatic vessel integrity in adult tissues and control vessel repair and regeneration. This knowledge is crucial for understanding the pathological vessel changes that contribute to disease, and provides an opportunity for therapy development.
Collapse
Affiliation(s)
- Simon Stritt
- Uppsala University, Department of Immunology, Genetics, and Pathology, 751 85 Uppsala, Sweden
| | - Katarzyna Koltowska
- Uppsala University, Department of Immunology, Genetics, and Pathology, 751 85 Uppsala, Sweden
| | - Taija Mäkinen
- Uppsala University, Department of Immunology, Genetics, and Pathology, 751 85 Uppsala, Sweden.
| |
Collapse
|
42
|
Liu Q, Hou C, Zhang H, Fu C, Wang W, Wang B, Li J, Zhao Y, Yang X. Impaired meningeal lymphatic vessels exacerbate early brain injury after experimental subarachnoid hemorrhage. Brain Res 2021; 1769:147584. [PMID: 34303696 DOI: 10.1016/j.brainres.2021.147584] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Revised: 06/26/2021] [Accepted: 07/14/2021] [Indexed: 11/25/2022]
Abstract
BACKGROUND AND PURPOSE Blood that enters the subarachnoid space (SAS) and its breakdown products are neurotoxic and are the principal inducers of brain injury after subarachnoid hemorrhage (SAH). Recently, meningeal lymphatic vessels (MLVs) have been proven to play an important role in clearing erythrocytes that arise from SAH, as well as other macromolecular solutes. However, evidence demonstrating the relationship between MLVs and brain injury after SAH is still limited. Therefore, we performed this study to observe the effects of meningeal lymphatic impairment on early brain injury (EBI) after experimental SAH. METHODS The MLVs of C57BL/6 male adult mice were ablated by injecting Visudyne into the cisterna magna and transcranially photoconverting it with laser light. The MLVs were then examined by immunofluorescence staining for lyve-1. Next, both the MLV-ablated group and the control group (normal mice) underwent filament perforation to model SAH or sham operation. We assessed the cortical perfusion of all the mice before SAH induction, 5 min after SAH and 24 h after SAH. In addition, we evaluated neurological function deficits by Garcia scores and measured brain water content at 24 h post SAH. Then, neuroinflammation and neural apoptosis in the mouse brain were also examined. RESULTS Visudyne and transcranial photoconversion treatment notably ablated mouse MLVs. Five minutes after SAH induction, cortical perfusion was significantly impaired, and after 24 h, this impairment was ameliorated considerably in the control group but ameliorated only slightly or worsened in the MLV-ablated group. Additionally, the MLVablated group presented worse neurological function deficits and more severe brain edema than the control group. More notably, neuroinflammation and neural apoptosis were also observed. CONCLUSION Ablation of MLVs by Visudyne treatment exacerbated EBI after experimental SAH in mice. The worsening of EBI may have arisen from limited drainage of blood and other breakdown products, which are thought to cause brain edema, neuroinflammation, neuronal apoptosis and other pathological processes.
Collapse
Affiliation(s)
- Quanlei Liu
- Tianjin Neurological Institute, Key Laboratory of Post-trauma Neuro-repair and Regeneration in Central Nervous System, Ministry of Education, Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, China
| | - Changkai Hou
- Tianjin Neurological Institute, Key Laboratory of Post-trauma Neuro-repair and Regeneration in Central Nervous System, Ministry of Education, Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, China; Department of Neurosurgery, Tianjin Medical University General Hospital, China
| | - Hao Zhang
- Tianjin Neurological Institute, Key Laboratory of Post-trauma Neuro-repair and Regeneration in Central Nervous System, Ministry of Education, Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, China
| | - Cong Fu
- Tianjin Neurological Institute, Key Laboratory of Post-trauma Neuro-repair and Regeneration in Central Nervous System, Ministry of Education, Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, China
| | - Weihan Wang
- Tianjin Neurological Institute, Key Laboratory of Post-trauma Neuro-repair and Regeneration in Central Nervous System, Ministry of Education, Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, China
| | - Bangyue Wang
- Tianjin Neurological Institute, Key Laboratory of Post-trauma Neuro-repair and Regeneration in Central Nervous System, Ministry of Education, Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, China; Department of Neurosurgery, Tianjin Medical University General Hospital, China
| | - Jian Li
- Tianjin Neurological Institute, Key Laboratory of Post-trauma Neuro-repair and Regeneration in Central Nervous System, Ministry of Education, Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, China; Department of Neurosurgery, Tianjin Medical University General Hospital, China
| | - Yan Zhao
- Tianjin Neurological Institute, Key Laboratory of Post-trauma Neuro-repair and Regeneration in Central Nervous System, Ministry of Education, Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, China; Department of Neurosurgery, Tianjin Medical University General Hospital, China
| | - Xinyu Yang
- Tianjin Neurological Institute, Key Laboratory of Post-trauma Neuro-repair and Regeneration in Central Nervous System, Ministry of Education, Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, China; Department of Neurosurgery, Tianjin Medical University General Hospital, China.
| |
Collapse
|
43
|
Wasson EM, Dubbin K, Moya ML. Go with the flow: modeling unique biological flows in engineered in vitro platforms. LAB ON A CHIP 2021; 21:2095-2120. [PMID: 34008661 DOI: 10.1039/d1lc00014d] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/19/2023]
Abstract
Interest in recapitulating in vivo phenomena in vitro using organ-on-a-chip technology has grown rapidly and with it, attention to the types of fluid flow experienced in the body has followed suit. These platforms offer distinct advantages over in vivo models with regards to human relevance, cost, and control of inputs (e.g., controlled manipulation of biomechanical cues from fluid perfusion). Given the critical role biophysical forces play in several tissues and organs, it is therefore imperative that engineered in vitro platforms capture the complex, unique flow profiles experienced in the body that are intimately tied with organ function. In this review, we outline the complex and unique flow regimes experienced by three different organ systems: blood vasculature, lymphatic vasculature, and the intestinal system. We highlight current state-of-the-art platforms that strive to replicate physiological flows within engineered tissues while introducing potential limitations in current approaches.
Collapse
Affiliation(s)
- Elisa M Wasson
- Material Engineering Division, Lawrence Livermore National Laboratory, 7000 East Ave L-222, Livermore, CA 94551, USA.
| | - Karen Dubbin
- Material Engineering Division, Lawrence Livermore National Laboratory, 7000 East Ave L-222, Livermore, CA 94551, USA.
| | - Monica L Moya
- Material Engineering Division, Lawrence Livermore National Laboratory, 7000 East Ave L-222, Livermore, CA 94551, USA.
| |
Collapse
|
44
|
Steele MM, Lund AW. Afferent Lymphatic Transport and Peripheral Tissue Immunity. THE JOURNAL OF IMMUNOLOGY 2021; 206:264-272. [PMID: 33397740 DOI: 10.4049/jimmunol.2001060] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Accepted: 11/11/2020] [Indexed: 12/30/2022]
Abstract
Lymphatic vessels provide an anatomical framework for immune surveillance and adaptive immune responses. Although appreciated as the route for Ag and dendritic cell transport, peripheral lymphatic vessels are often not considered active players in immune surveillance. Lymphatic vessels, however, integrate contextual cues that directly regulate transport, including changes in intrinsic pumping and capillary remodeling, and express a dynamic repertoire of inflammatory chemokines and adhesion molecules that facilitates leukocyte egress out of inflamed tissue. These mechanisms together contribute to the course of peripheral tissue immunity. In this review, we focus on context-dependent mechanisms that regulate fluid and cellular transport out of peripheral nonlymphoid tissues to provide a framework for understanding the effects of afferent lymphatic transport on immune surveillance, peripheral tissue inflammation, and adaptive immunity.
Collapse
Affiliation(s)
- Maria M Steele
- Ronald O. Perelman Department of Dermatology, New York University Grossman School of Medicine, New York, NY 10016
| | - Amanda W Lund
- Ronald O. Perelman Department of Dermatology, New York University Grossman School of Medicine, New York, NY 10016; .,Department of Pathology, New York University Grossman School of Medicine, New York, NY 10016; and.,Laura and Isaac Perlmutter Cancer Center, New York University Grossman School of Medicine, New York, NY 10016
| |
Collapse
|
45
|
Walsh SM, Sheridan RM, Lucas ED, Doan TA, Ware BC, Schafer J, Fu R, Burchill MA, Hesselberth JR, Tamburini BAJ. Molecular tracking devices quantify antigen distribution and archiving in the murine lymph node. eLife 2021; 10:e62781. [PMID: 33843587 PMCID: PMC8116055 DOI: 10.7554/elife.62781] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Accepted: 04/09/2021] [Indexed: 01/02/2023] Open
Abstract
The detection of foreign antigens in vivo has relied on fluorescent conjugation or indirect read-outs such as antigen presentation. In our studies, we found that these widely used techniques had several technical limitations that have precluded a complete picture of antigen trafficking or retention across lymph node cell types. To address these limitations, we developed a 'molecular tracking device' to follow the distribution, acquisition, and retention of antigen in the lymph node. Utilizing an antigen conjugated to a nuclease-resistant DNA tag, acting as a combined antigen-adjuvant conjugate, and single-cell mRNA sequencing, we quantified antigen abundance in the lymph node. Variable antigen levels enabled the identification of caveolar endocytosis as a mechanism of antigen acquisition or retention in lymphatic endothelial cells. Thus, these molecular tracking devices enable new approaches to study dynamic tissue dissemination of antigen-adjuvant conjugates and identify new mechanisms of antigen acquisition and retention at cellular resolution in vivo.
Collapse
Affiliation(s)
- Shannon M Walsh
- Department of Biochemistry and Molecular Genetics, University of Colorado School of MedicineAuroraUnited States
| | - Ryan M Sheridan
- RNA Bioscience Initiative, University of Colorado School of MedicineAuroraUnited States
| | - Erin D Lucas
- Immunology Graduate Program, University of Colorado School of MedicineAuroraUnited States
- Department of Immunology and Microbiology, University of Colorado School of MedicineAuroraUnited States
| | - Thu A Doan
- Immunology Graduate Program, University of Colorado School of MedicineAuroraUnited States
- Department of Medicine, Division of Gastroenterology and Hepatology, University of Colorado School of MedicineAuroraUnited States
| | - Brian C Ware
- Immunology Graduate Program, University of Colorado School of MedicineAuroraUnited States
- Department of Immunology and Microbiology, University of Colorado School of MedicineAuroraUnited States
| | - Johnathon Schafer
- Department of Medicine, Division of Gastroenterology and Hepatology, University of Colorado School of MedicineAuroraUnited States
| | - Rui Fu
- RNA Bioscience Initiative, University of Colorado School of MedicineAuroraUnited States
| | - Matthew A Burchill
- Department of Medicine, Division of Gastroenterology and Hepatology, University of Colorado School of MedicineAuroraUnited States
| | - Jay R Hesselberth
- Department of Biochemistry and Molecular Genetics, University of Colorado School of MedicineAuroraUnited States
- RNA Bioscience Initiative, University of Colorado School of MedicineAuroraUnited States
| | - Beth Ann Jiron Tamburini
- Immunology Graduate Program, University of Colorado School of MedicineAuroraUnited States
- Department of Immunology and Microbiology, University of Colorado School of MedicineAuroraUnited States
- Department of Medicine, Division of Gastroenterology and Hepatology, University of Colorado School of MedicineAuroraUnited States
| |
Collapse
|
46
|
Sasso MS, Mitrousis N, Wang Y, Briquez PS, Hauert S, Ishihara J, Hubbell JA, Swartz MA. Lymphangiogenesis-inducing vaccines elicit potent and long-lasting T cell immunity against melanomas. SCIENCE ADVANCES 2021; 7:eabe4362. [PMID: 33762337 PMCID: PMC7990326 DOI: 10.1126/sciadv.abe4362] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Accepted: 01/22/2021] [Indexed: 05/06/2023]
Abstract
In melanoma, the induction of lymphatic growth (lymphangiogenesis) has long been correlated with metastasis and poor prognosis, but we recently showed it can synergistically enhance cancer immunotherapy and boost T cell immunity. Here, we develop a translational approach for exploiting this "lymphangiogenic potentiation" of immunotherapy in a cancer vaccine using lethally irradiated tumor cells overexpressing vascular endothelial growth factor C (VEGF-C) and topical adjuvants. Our "VEGFC vax" induced extensive local lymphangiogenesis and promoted stronger T cell activation in both the intradermal vaccine site and draining lymph nodes, resulting in higher frequencies of antigen-specific T cells present systemically than control vaccines. In mouse melanoma models, VEGFC vax elicited potent tumor-specific T cell immunity and provided effective tumor control and long-term immunological memory. Together, these data introduce the potential of lymphangiogenesis induction as a novel immunotherapeutic strategy to consider in cancer vaccine design.
Collapse
Affiliation(s)
- Maria Stella Sasso
- Pritzker School for Molecular Engineering, University of Chicago, Chicago, IL, USA.
| | - Nikolaos Mitrousis
- Pritzker School for Molecular Engineering, University of Chicago, Chicago, IL, USA
| | - Yue Wang
- Pritzker School for Molecular Engineering, University of Chicago, Chicago, IL, USA
| | - Priscilla S Briquez
- Pritzker School for Molecular Engineering, University of Chicago, Chicago, IL, USA
| | - Sylvie Hauert
- Pritzker School for Molecular Engineering, University of Chicago, Chicago, IL, USA
| | - Jun Ishihara
- Pritzker School for Molecular Engineering, University of Chicago, Chicago, IL, USA
| | - Jeffrey A Hubbell
- Pritzker School for Molecular Engineering, University of Chicago, Chicago, IL, USA
| | - Melody A Swartz
- Pritzker School for Molecular Engineering, University of Chicago, Chicago, IL, USA.
- Ben May Department for Cancer Research, University of Chicago, Chicago, IL, USA
| |
Collapse
|
47
|
Wang Z, Tang M. Research progress on toxicity, function, and mechanism of metal oxide nanoparticles on vascular endothelial cells. J Appl Toxicol 2020; 41:683-700. [DOI: 10.1002/jat.4121] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Revised: 11/05/2020] [Accepted: 11/12/2020] [Indexed: 02/06/2023]
Affiliation(s)
- Zhihui Wang
- Key Laboratory of Environmental Medicine and Engineering, Ministry of Education, School of Public Health Southeast University Nanjing China
| | - Meng Tang
- Key Laboratory of Environmental Medicine and Engineering, Ministry of Education, School of Public Health Southeast University Nanjing China
| |
Collapse
|
48
|
Lymph-directed nitric oxide increases immune cell access to lymph-borne nanoscale solutes. Biomaterials 2020; 265:120411. [PMID: 33080460 DOI: 10.1016/j.biomaterials.2020.120411] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 09/16/2020] [Accepted: 09/18/2020] [Indexed: 12/12/2022]
Abstract
Lymph nodes (LNs) are immune organs housing high concentrations of lymphocytes, making them critical targets for therapeutic immunomodulation in a wide variety of diseases. While there is great interest in targeted drug delivery to LNs, many nanoscale drug delivery carriers have limited access to parenchymal resident immune cells compared to small molecules, limiting their efficacy. Nitric oxide (NO) is a potent regulator of vascular and lymphatic transport and a promising candidate for modulating nanocarrier access to LNs, but its lymphatic accumulation is limited by its low molecular weight and high reactivity. In this work, we employ S-nitrosated nanoparticles (SNO-NP), a lymphatic-targeted delivery system for controlled NO release, to investigate the effect of NO application on molecule accumulation and distribution within the LN. We evaluated the LN accumulation, spatial distribution, and cellular distribution of a panel of fluorescent tracers after intradermal administration alongside SNO-NP or a small molecule NO donor. While SNO-NP did not alter total tracer accumulation in draining lymph nodes (dLNs) or affect active cellular transport of large molecules from the injection site, its application enhanced the penetration of nanoscale 30 nm dextrans into the LN and their subsequent uptake by LN-resident lymphocytes, while nontargeted NO delivery did not. These results further extended to a peptide-conjugated NP drug delivery system, which showed enhanced uptake by B cells and dendritic cells when administered alongside SNO-NP. Together, these results highlight the utility of LN-targeted NO application for the enhancement of nanocarrier access to therapeutically relevant LN-resident immune cells, making NO a potentially useful tool for improving LN drug delivery and immune responses.
Collapse
|
49
|
Brain Glymphatic/Lymphatic Imaging by MRI and PET. Nucl Med Mol Imaging 2020; 54:207-223. [PMID: 33088350 DOI: 10.1007/s13139-020-00665-4] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Revised: 08/09/2020] [Accepted: 08/19/2020] [Indexed: 01/19/2023] Open
Abstract
Since glymphatic was proposed and meningeal lymphatic was discovered, MRI and even PET were introduced to investigate brain parenchymal interstitial fluid (ISF), cerebrospinal fluid (CSF), and lymphatic outflow in rodents and humans. Previous findings by ex vivo fluorescent microscopic, and in vivo two-photon imaging in rodents were reproduced using intrathecal contrast (gadobutrol and the similar)-enhanced MRI in rodents and further in humans. On dynamic MRI of meningeal lymphatics, in contrast to rodents, humans use mainly dorsal meningeal lymphatic pathways of ISF-CSF-lymphatic efflux. In mice, ISF-CSF exchange was examined thoroughly using an intra-cistern injection of fluorescent tracers during sleep, aging, and neurodegeneration yielding many details. CSF to lymphatic efflux is across arachnoid barrier cells over the dorsal dura in rodents and in humans. Meningeal lymphatic efflux to cervical lymph nodes and systemic circulation is also well-delineated especially in humans onintrathecal contrast MRI. Sleep- or anesthesia-related changes of glymphatic-lymphatic flow and the coupling of ISF-CSF-lymphatic drainage are major confounders ininterpreting brain glymphatic/lymphatic outflow in rodents. PET imaging in humans should be interpreted based on human anatomy and physiology, different in some aspects, using MRI recently. Based on the summary in this review, we propose non-invasive and longer-term intrathecal SPECT/PET or MRI studies to unravel the roles of brain glymphatic/lymphatic in diseases.
Collapse
|
50
|
Landh E, M Moir L, Bradbury P, Traini D, M Young P, Ong HX. Properties of rapamycin solid lipid nanoparticles for lymphatic access through the lungs & part I: the effect of size. Nanomedicine (Lond) 2020; 15:1927-1945. [PMID: 32820673 DOI: 10.2217/nnm-2020-0077] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Background: Lymphangioleiomyomatosis (LAM) is characterized by growth of smooth muscle-like cells in the lungs that spread to other organs via lymphatic vessels. Current oral rapamycin treatment is limited by low bioavailability of approximately 15%. Aim: The effect of inhaled rapamycin solid lipid nanoparticles (Rapa-SLNs) size on its penetration through the lymphatics. Method: Three Rapa-SLN formulations (200-1000 nm) were produced and assessed for particle characteristics and further for toxicity and performance in vitro. Results: Rapa-SLNs of 200 nm inhibited proliferation in TSC2-negative mouse embryonic fibroblast cells and penetrated the respiratory epithelium and lymphatic endothelium significantly faster compared with free rapamycin and larger Rapa-SLNs. Conclusion: Rapa-SLN approximately 200 nm allows efficient entry of rapamycin into the lymphatic system and is therefore a promising treatment for LAM patients.
Collapse
Affiliation(s)
- Emelie Landh
- Respiratory Technology, Woolcock Institute of Medical Research, Glebe, NSW, 2037, Australia.,Discipline of Pharmacology, Faculty of Medicine & Health, Sydney, 2006, Australia
| | - Lyn M Moir
- Respiratory Technology, Woolcock Institute of Medical Research, Glebe, NSW, 2037, Australia.,Discipline of Pharmacology, Faculty of Medicine & Health, Sydney, 2006, Australia
| | - Peta Bradbury
- Respiratory Technology, Woolcock Institute of Medical Research, Glebe, NSW, 2037, Australia
| | - Daniela Traini
- Respiratory Technology, Woolcock Institute of Medical Research, Glebe, NSW, 2037, Australia.,Discipline of Pharmacology, Faculty of Medicine & Health, Sydney, 2006, Australia
| | - Paul M Young
- Respiratory Technology, Woolcock Institute of Medical Research, Glebe, NSW, 2037, Australia.,Discipline of Pharmacology, Faculty of Medicine & Health, Sydney, 2006, Australia
| | - Hui Xin Ong
- Respiratory Technology, Woolcock Institute of Medical Research, Glebe, NSW, 2037, Australia.,Discipline of Pharmacology, Faculty of Medicine & Health, Sydney, 2006, Australia
| |
Collapse
|