1
|
Hanafy BI, Munson MJ, Soundararajan R, Pereira S, Gallud A, Sanaullah SM, Carlesso G, Mazza M. Advancing Cellular-Specific Delivery: Machine Learning Insights into Lipid Nanoparticles Design and Cellular Tropism. Adv Healthc Mater 2025:e2500383. [PMID: 40326205 DOI: 10.1002/adhm.202500383] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2025] [Revised: 03/26/2025] [Indexed: 05/07/2025]
Abstract
Lipid nanoparticles (LNPs) have gained significant attention as effective nucleic acid delivery vehicles. Despite their success, LNPs are predominantly liver-targeted which limits their broader application. To expand the therapeutic potential of LNPs, this work implements a data-driven approach that combines design of experiments (DoE), high throughput screening (HTS), and machine learning (ML) to tailor LNP formulations for preferential immune cell targeting. This methodology involves the generation of 180 LNP formulations, with varying lipid molar ratios and lipid chemistries, to explore a diverse design space. This work aims to identify LNP properties that enhance immune cell specificity while reducing hepatic uptake. The in vitro screening of these LNPs provided a rich dataset for ML analysis, leading to the identification of promising candidates with improved immune cellular selectivity profiles. These findings are validated in vivo where it is demonstrated that selected LNPs achieved preferential spleen expression with a successful redirection of LNP tropism beyond hepatic cells. This workflow highlights the importance of tailoring LNP compositions for the development of LNPs with selective cellular tropism.
Collapse
Affiliation(s)
- Belal I Hanafy
- Advanced Drug Delivery, Pharmaceutical Sciences, Biopharmaceuticals R&D, AstraZeneca, Cambridge, CB2 0AA, United Kingdom
| | - Michael J Munson
- Advanced Drug Delivery, Pharmaceutical Sciences, Biopharmaceuticals R&D, AstraZeneca, Gothenburg, 431 83, Sweden
| | - Ramesh Soundararajan
- Advanced Drug Delivery, Pharmaceutical Sciences, Biopharmaceuticals R&D, AstraZeneca, Cambridge, CB2 0AA, United Kingdom
| | - Sara Pereira
- Advanced Drug Delivery, Pharmaceutical Sciences, Biopharmaceuticals R&D, AstraZeneca, Cambridge, CB2 0AA, United Kingdom
| | - Audrey Gallud
- Advanced Drug Delivery, Pharmaceutical Sciences, Biopharmaceuticals R&D, AstraZeneca, Gothenburg, 431 83, Sweden
| | - Sajib Md Sanaullah
- Early ICC Discovery, R&D Oncology, AstraZeneca, Gaithersburg, MD, 20878, USA
| | - Gianluca Carlesso
- Early ICC Discovery, R&D Oncology, AstraZeneca, Gaithersburg, MD, 20878, USA
| | - Mariarosa Mazza
- Advanced Drug Delivery, Pharmaceutical Sciences, Biopharmaceuticals R&D, AstraZeneca, Cambridge, CB2 0AA, United Kingdom
| |
Collapse
|
2
|
Heiser BJ, Veyssi A, Ghosh D. Recent strategies for enhanced delivery of mRNA to the lungs. Nanomedicine (Lond) 2025; 20:1043-1069. [PMID: 40190037 PMCID: PMC12051540 DOI: 10.1080/17435889.2025.2485669] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Accepted: 03/25/2025] [Indexed: 05/02/2025] Open
Abstract
mRNA-based therapies have emerged as a transformative tool in modern medicine, gaining significant attention following their successful use in COVID-19 vaccines. Delivery to the lungs offers several compelling advantages for mRNA delivery. The lungs are one of the most vascularized organs in the body, which provides an extensive surface area that can facilitate efficient drug transport. Local delivery to the lungs bypasses gastrointestinal degradation, potentially enhancing therapeutic efficacy. In addition, the extensive capillary network of the lungs provides an ideal target for systemic delivery. However, developing effective mRNA therapies for the lungs presents significant challenges. The complex anatomy of the lungs and the body's immune response to foreign particles create barriers to delivery. This review discusses key approaches for overcoming these challenges and improving mRNA delivery to the lungs. It examines both local and systemic delivery strategies aimed at improving lung delivery while mitigating off-target effects. Although substantial progress has been made in lung-targeted mRNA therapies, challenges remain in optimizing cellular uptake and achieving therapeutic efficacy within pulmonary tissues. The continued refinement of delivery strategies that enhance lung-specific targeting while minimizing degradation is critical for the clinical success of mRNA-based pulmonary therapies.
Collapse
Affiliation(s)
- Brittany J. Heiser
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX, USA
| | - Arian Veyssi
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX, USA
| | - Debadyuti Ghosh
- Division of Molecular Pharmaceutics and Drug Delivery, College of Pharmacy, The University of Texas at Austin, Austin, TX, USA
| |
Collapse
|
3
|
Shin YJ, Safina D, Zheng Y, Levenberg S. Microvascularization in 3D Human Engineered Tissue and Organoids. Annu Rev Biomed Eng 2025; 27:473-498. [PMID: 40310885 DOI: 10.1146/annurev-bioeng-103023-115236] [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: 05/03/2025]
Abstract
The microvasculature, a complex network of small blood vessels, connects systemic circulation with local tissues, facilitating the nutrient and oxygen exchange that is critical for homeostasis and organ function. Engineering these structures is paramount for advancing tissue regeneration, disease modeling, and drug testing. However, replicating the intricate architecture of native vascular systems-characterized by diverse vessel diameters, cellular constituents, and dynamic perfusion capabilities-presents significant challenges. This complexity is compounded by the need to precisely integrate biomechanical, biochemical, and cellular cues. Recent breakthroughs in microfabrication, organoids, bioprinting, organ-on-a-chip platforms, and in vivo vascularization techniques have propelled the field toward faithfully replicating vascular complexity. These innovations not only enhance our understanding of vascular biology but also enable the generation of functional, perfusable tissue constructs. Here, we explore state-of-the-art technologies and strategies in microvascular engineering, emphasizing key advancements and addressing the remaining challenges to developing fully functional vascularized tissues.
Collapse
Affiliation(s)
- Yu Jung Shin
- Department of Bioengineering, University of Washington, Seattle, Washington, USA;
- Institute of Stem Cell and Regenerative Medicine, University of Washington, Seattle, Washington, USA
| | - Dina Safina
- Department of Biomedical Engineering, Technion - Israel Institute of Technology, Haifa, Israel;
| | - Ying Zheng
- Department of Bioengineering, University of Washington, Seattle, Washington, USA;
- Institute of Stem Cell and Regenerative Medicine, University of Washington, Seattle, Washington, USA
| | - Shulamit Levenberg
- Department of Biomedical Engineering, Technion - Israel Institute of Technology, Haifa, Israel;
| |
Collapse
|
4
|
Puri M, Sonawane S. Liver Sinusoidal Endothelial Cells in the Regulation of Immune Responses and Fibrosis in Metabolic Dysfunction-Associated Fatty Liver Disease. Int J Mol Sci 2025; 26:3988. [PMID: 40362227 PMCID: PMC12071881 DOI: 10.3390/ijms26093988] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2025] [Revised: 04/08/2025] [Accepted: 04/18/2025] [Indexed: 05/15/2025] Open
Abstract
Liver Sinusoidal Endothelial Cells (LSECs) play a crucial role in maintaining liver homeostasis, regulating immune responses, and fibrosis in liver diseases. This review explores the unique functions of LSECs in liver pathology, particularly their roles in immune tolerance, antigen presentation, and the modulation of hepatic stellate cells (HSCs) during fibrosis. LSECs act as key regulators of immune balance in the liver by preventing excessive immune activation while also filtering antigens and interacting with immune cells, including Kupffer cells and T cells. Metabolic Dysfunction-Associated Fatty Liver Disease(MAFLD) is significant because it can lead to advanced liver dysfunction, such as cirrhosis and liver cancer. The prevalence of Metabolic Associated Steatohepatitis (MASH) is increasing globally, particularly in the United States, and is closely linked to rising rates of obesity and type 2 diabetes. Early diagnosis and intervention are vital to prevent severe outcomes, highlighting the importance of studying LSECs in liver disease. However, during chronic liver diseases, LSECs undergo dysfunction, leading to their capillarization, loss of fenestrations, and promotion of pro-fibrotic signaling pathways such as Transforming growth factor-beta (TGF-β), which subsequently activates HSCs and contributes to the progression of liver fibrosis. The review also discusses the dynamic interaction between LSECs, HSCs, and other hepatic cells during the progression of liver diseases, emphasizing how changes in LSEC phenotype contribute to liver scarring and fibrosis. Furthermore, it highlights the potential of LSECs as therapeutic targets for modulating immune responses and preventing fibrosis in liver diseases. By restoring LSECs' function and targeting pathways associated with their dysfunction, novel therapies could be developed to halt or reverse liver disease progression. The findings of this review reinforce the importance of LSECs in liver pathology and suggest that they hold significant promises as targets for future treatment strategies aimed at addressing chronic liver diseases.
Collapse
Affiliation(s)
- Munish Puri
- Onco-Immunology, Magnit Global, Folsom, CA 95630, USA
| | - Snehal Sonawane
- Department of Pathology, University of Illinois, Chicago, IL 60612, USA;
| |
Collapse
|
5
|
Borah D, Blacharczyk O, Szafranska K, Czyzynska-Cichon I, Metwally S, Szymanowski K, Hübner W, Kotlinowski J, Dobosz E, McCourt P, Huser T, Lekka M, Zapotoczny B. Mimicking the Liver Sinusoidal Endothelial Cell Niche In Vitro to Enhance Fenestration in a Genetic Model of Systemic Inflammation. Cells 2025; 14:621. [PMID: 40277946 PMCID: PMC12025456 DOI: 10.3390/cells14080621] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2025] [Revised: 03/26/2025] [Accepted: 04/07/2025] [Indexed: 04/26/2025] Open
Abstract
Liver sinusoidal endothelial cells (LSECs) play a crucial role in hepatic homeostasis, clearance, and microcirculatory regulation. Their fenestrations-patent transcellular pores-are essential for proper liver function, yet disappear in pathological conditions such as liver fibrosis and inflammation through a process known as defenestration. Defenestrated sinusoids are often linked to the liver stiffening that occurs through mechanotransduction-regulated processes. We performed a detailed characterization of polyacrylamide (PAA) hydrogels using atomic force microscopy (AFM), rheometry, scanning electron microscopy, and fluorescence microscopy to assess their potential as biomimetic substrates for LSECs. We additionally implemented AFM; quantitative fluorescence microscopy, including high-resolution structured illumination microscopy (HR-SIM); and an endocytosis assay to characterize the morphology and function of LSECs. Our results revealed significant local variations in hydrogel stiffness and differences in pore sizes. The primary LSECs cultured on these substrates had a range of stiffnesses and were analyzed with regard to their number of fenestrations, cytoskeletal organization, and endocytic function. To explore mechanotransduction in inflammatory liver disease, we investigated LSECs from a genetic model of systemic inflammation triggered by the deletion of Mcpip1 in myeloid leukocytes and examined their ability to restore their fenestrations on soft substrates. Our study demonstrates the beneficial effect of soft hydrogels on LSECs. Control cells exhibited a similar fenestrated morphology and function compared to cells cultured on plastic substrates. However, the pathological LSECs from the genetic model of systemic inflammation regained their fenestrations when cultured on soft hydrogels. This observation supports previous findings on the beneficial effects of soft substrates on LSEC fenestration status.
Collapse
Affiliation(s)
- Dibakar Borah
- Department of Biophysical Microstructures, Institute of Nuclear Physics, Polish Academy of Sciences, ul. Radzikowskiego 142, 31-342 Krakow, Poland; (D.B.); (S.M.); (M.L.)
| | - Oliwia Blacharczyk
- Department of Biophysical Microstructures, Institute of Nuclear Physics, Polish Academy of Sciences, ul. Radzikowskiego 142, 31-342 Krakow, Poland; (D.B.); (S.M.); (M.L.)
| | - Karolina Szafranska
- Vascular Biology Research Group, Department of Medical Biology University of Tromsø—The Arctic University of Norway, 9019 Tromsø, Norway; (K.S.); (P.M.)
| | - Izabela Czyzynska-Cichon
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, Bobrzynskiego 14, 30-348 Krakow, Poland;
| | - Sara Metwally
- Department of Biophysical Microstructures, Institute of Nuclear Physics, Polish Academy of Sciences, ul. Radzikowskiego 142, 31-342 Krakow, Poland; (D.B.); (S.M.); (M.L.)
| | - Konrad Szymanowski
- Department of Biophysical Microstructures, Institute of Nuclear Physics, Polish Academy of Sciences, ul. Radzikowskiego 142, 31-342 Krakow, Poland; (D.B.); (S.M.); (M.L.)
| | - Wolfgang Hübner
- Biomolecular Photonics, Faculty of Physics, Bielefeld University, 33615 Bielefeld, Germany; (W.H.); (T.H.)
| | - Jerzy Kotlinowski
- Department of General Biochemistry, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387 Krakow, Poland;
| | - Ewelina Dobosz
- Department of Microbiology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387 Krakow, Poland;
| | - Peter McCourt
- Vascular Biology Research Group, Department of Medical Biology University of Tromsø—The Arctic University of Norway, 9019 Tromsø, Norway; (K.S.); (P.M.)
| | - Thomas Huser
- Biomolecular Photonics, Faculty of Physics, Bielefeld University, 33615 Bielefeld, Germany; (W.H.); (T.H.)
| | - Malgorzata Lekka
- Department of Biophysical Microstructures, Institute of Nuclear Physics, Polish Academy of Sciences, ul. Radzikowskiego 142, 31-342 Krakow, Poland; (D.B.); (S.M.); (M.L.)
| | - Bartlomiej Zapotoczny
- Department of Biophysical Microstructures, Institute of Nuclear Physics, Polish Academy of Sciences, ul. Radzikowskiego 142, 31-342 Krakow, Poland; (D.B.); (S.M.); (M.L.)
| |
Collapse
|
6
|
Park SH, Song SJ, Lee JA, Shin JA. Effects of Aging on the Severity of Liver Injury in Mice With Iron Overload. J Gastroenterol Hepatol 2025; 40:1016-1025. [PMID: 39971277 DOI: 10.1111/jgh.16908] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Revised: 01/15/2025] [Accepted: 02/05/2025] [Indexed: 02/21/2025]
Abstract
Although iron is a vital component in the body, excessive iron leads to iron toxicity, which affects vital organs. In particular, the liver is considerably affected by iron toxicity because it stores the highest amount of iron in the body. Nonetheless, the relationship between iron overload and aging in the liver has not yet been clearly identified. This study aimed to observe the effects of aging on iron overload in the liver. Female C57BL/6J mice were randomly divided into vehicle control and iron overload groups (n = 7-22 per group). The iron overload group was injected with iron-dextran (Fe-dextran, ferric hydroxide dextran complex) (0.5 g/kg) for 4 weeks. After the experimental period, liver and blood samples were obtained from 2-, 15-, and 22-month-old mice. Liver weight, iron deposition, structural changes, cell death, extracellular matrix deposition, and fenestration of sinusoidal vessels were analyzed and compared between the groups. Additionally, biochemical analyses (aspartate aminotransferase, alanine aminotransferase, and serum total iron levels) were performed. The iron overload group exhibited significant differences compared with the control group with age. In the elderly iron overload model, iron deposition, inflammatory cell infiltration, and cell death were significantly increased (p < 0.0001). Moreover, deposition of the extracellular matrix and defenestration of sinusoidal fenestrae were observed among 22-month-old mice in the iron overload group. These results suggest that aging is a risk factor for iron-induced liver injury. Therefore, caution should be exercised when performing iron-related treatments in the elderly.
Collapse
Affiliation(s)
- So-Hyun Park
- Department of Anatomy, Ewha Womans University College of Medicine, Seoul, South Korea
| | - Soo-Jin Song
- Department of Anatomy, Ewha Womans University College of Medicine, Seoul, South Korea
| | - Jin-A Lee
- Department of Anatomy, Ewha Womans University College of Medicine, Seoul, South Korea
- Department of Urology, Ewha Womans University Seoul Hospital, Seoul, South Korea
| | - Jung-A Shin
- Department of Anatomy, Ewha Womans University College of Medicine, Seoul, South Korea
| |
Collapse
|
7
|
Jin Z, Li Y, Yi H, Wang M, Wang C, Du S, Zeng W, Zong Z. Pathogenetic development, diagnosis and clinical therapeutic approaches for liver metastasis from colorectal cancer (Review). Int J Oncol 2025; 66:22. [PMID: 39950314 PMCID: PMC11844340 DOI: 10.3892/ijo.2025.5728] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Accepted: 01/10/2025] [Indexed: 02/23/2025] Open
Abstract
Colorectal cancer (CRC) is a prevalent malignancy and a significant proportion of patients with CRC develop liver metastasis (CRLM), which is a major contributor to CRC‑related mortality. The present review aimed to comprehensively examine the pathogenetic development and diagnosis of CRLM and the clinical therapeutic approaches for treatment of this disease. The molecular mechanisms underlying CRLM were discussed, including the role of the tumour microenvironment and epithelial‑mesenchymal transition. The present review also highlighted the importance of early detection and the current challenges in predicting the development of CRLM. Various treatment strategies were reviewed, including surgical resection, chemotherapy and immunotherapy, and the potential of novel therapies, such as selective internal radiation therapy and Traditional Chinese Medicine. Despite recent advancements in treatment options, the treatment of CRLM remains a therapeutic challenge due to the complexity of the liver microenvironment and the heterogeneity of CRC. The present review emphasized the need for a multidisciplinary approach and the integration of emerging therapies to improve patient outcomes.
Collapse
Affiliation(s)
- Zhenhua Jin
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
- The Second Clinical Medical College of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Yin Li
- Huan Kui Academy, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Hao Yi
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
- The Second Clinical Medical College of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Menghui Wang
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
- Huan Kui Academy, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Chaofeng Wang
- Queen Mary School, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Shaokun Du
- The Second Clinical Medical College of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Wenjuan Zeng
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
- Huan Kui Academy, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Zhen Zong
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| |
Collapse
|
8
|
Sun Z, Li X. A promising mesoporous silica carrier material for the diagnosis and treatment of liver diseases: recent research advances. J Mater Chem B 2025; 13:1935-1960. [PMID: 39801308 DOI: 10.1039/d4tb01822b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2025]
Abstract
The therapeutic diagnosis of liver diseases has garnered significant interest within the medical community. In recent years, mesoporous silica nanoparticles (MSNs) have emerged as crucial nanocarriers for the treatment of liver ailments. Their remarkable diagnostic capabilities enable them to be used in techniques such as high-throughput mass spectrometry (MS), magnetic resonance imaging (MRI), near-infrared (NIR) fluorescence imaging, photoacoustic imaging (PAI), and ultrasonography (US), attracting considerable attention. Furthermore, the introduction of amino and carboxyl group modifications in MSNs has facilitated their use as drug delivery carriers for treating liver diseases, including hepatocellular carcinoma. This paper reviews the preparation methods, in vitro diagnostic capabilities, and in vivo therapeutic delivery systems of MSNs for liver disease treatment. It also summarizes relevant toxicity studies, aiming to provide a comprehensive overview of the diagnostic and therapeutic applications of MSNs in the treatment of liver diseases, particularly hepatocellular carcinoma. Through this review, we seek to offer theoretical insights into the potential of MSNs for diagnostic and therapeutic applications in liver disease treatment.
Collapse
Affiliation(s)
- Zihao Sun
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, China.
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Xiaofang Li
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, China.
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| |
Collapse
|
9
|
Kruse LD, Holte C, Zapotoczny B, Struck EC, Schürstedt J, Hübner W, Huser T, Szafranska K. Hydrogen peroxide damage to rat liver sinusoidal endothelial cells is prevented by n-acetyl-cysteine but not GSH. Hepatol Commun 2025; 9:e0617. [PMID: 40163767 PMCID: PMC11737494 DOI: 10.1097/hc9.0000000000000617] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Accepted: 11/01/2024] [Indexed: 04/02/2025] Open
Abstract
BACKGROUND Reactive oxygen species (ROS) are prevalent in the liver during intoxication, infection, inflammation, and aging. Changes in liver sinusoidal endothelial cells (LSEC) are associated with various liver diseases. METHODS Isolated rat LSEC were studied under oxidative stress induced by H2O2 at different concentrations (0.5-1000 µM) and exposure times (10-120 min). LSEC functions were tested in a dose-dependent and time-dependent manner. RESULTS (1) Cell viability, reducing potential, and scavenging function decreased as H2O2 concentration and exposure time increased; (2) intracellular ROS levels rose with higher H2O2 concentrations; (3) fenestrations exhibited a dynamic response, initially closing but partially reopening at H2O2 concentrations above 100 µM after about 1 hour; (4) scavenging function was affected after just 10 minutes of exposure, with the impact being irreversible and primarily affecting degradation rather than receptor-mediated uptake; (5) the tubulin network was disrupted in high H2O2 concentration while the actin cytoskeleton appears to remain largely intact. Finally, we found that reducing agents and thiol donors such as n-acetyl cysteine and glutathione (GSH) could protect cells from ROS-induced damage but could not reverse existing damage as pretreatment with n-acetyl cysteine, but not GSH, reduced the negative effects of ROS exposure. CONCLUSIONS The results suggest that LSEC does not store an excess amount of GSH but rather can readily produce it in the occurrence of oxidative stress conditions. Moreover, the observed thresholds in dose-dependent and time-dependent changes, as well as the treatments with n-acetyl cysteine/GSH, confirm the existence of a ROS-depleting system in LSEC.
Collapse
Affiliation(s)
- Larissa D. Kruse
- Vascular Biology Research Group, Department of Medical Biology, University of Tromsø—The Arctic University of Norway, Tromsø, Norway
| | - Christopher Holte
- Vascular Biology Research Group, Department of Medical Biology, University of Tromsø—The Arctic University of Norway, Tromsø, Norway
| | | | - Eike C. Struck
- Translational Vascular Research Group, Department of Clinical Medicine, University of Tromsø—The Arctic University of Norway, Tromsø, Norway
| | - Jasmin Schürstedt
- Biomolecular Photonics Research Group, Department of Physics, Bielefeld University, Bielefeld, Germany
| | - Wolfgang Hübner
- Biomolecular Photonics Research Group, Department of Physics, Bielefeld University, Bielefeld, Germany
| | - Thomas Huser
- Biomolecular Photonics Research Group, Department of Physics, Bielefeld University, Bielefeld, Germany
| | - Karolina Szafranska
- Vascular Biology Research Group, Department of Medical Biology, University of Tromsø—The Arctic University of Norway, Tromsø, Norway
| |
Collapse
|
10
|
Henriques-Pons A, Vacani-Martins N, dos Santos CDLP, Meuser-Batista M. The liver's dilemma: sensing real danger in a sea of PAMPs: the (arterial) sinusoidal segment theory. Front Immunol 2025; 15:1503063. [PMID: 39931578 PMCID: PMC11808282 DOI: 10.3389/fimmu.2024.1503063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2024] [Accepted: 12/30/2024] [Indexed: 02/13/2025] Open
Abstract
The liver is susceptible to viruses and bacterial infections, tumors, and sterile tissue damage, but immunological danger recognition in the liver is highly unconventional. When analyzing innate and adaptive immunity in the organ, the valid concepts that guide danger recognition and immune response in the periphery should be put aside. In the liver, the vascular anatomy is a game changer, as about 80% of the blood that percolates the organ arrives from the hepatic portal vein, draining blood rich in molecules from the intestinal flora. This 24/7 exposure to high amounts of pathogen-associated molecular pattern (PAMPs) molecules results in hepatic immunological tolerance. In the liver, dendritic, Kupffer (KC), liver sinusoidal endothelial cells (LSECs), and even hepatocytes express PD-L1, a T lymphocyte downregulatory molecule. Most cells express Fas-L, IL-10, TGF-β, low levels of co-stimulatory molecules, lack of or have low levels of MHC-I and/or MHC-II expression. Moreover, other negative regulators such as CTLA-4, IDO-1, and prostaglandin E2 (PGE2) are regularly expressed. Then, how can real danger be discerned and recognized in this sea of PAMPs? This is an open question. Here, we hypothesize that conventional immunological danger recognition can occur in the liver but in specific and minor arterial sinusoidal segments,. Then, in the portal triad, where the hepatic artery ramificates into the stroma and carries arterial blood with no gut-derived PAMPs, there is no evolutive or environmental pressure for immunosuppressive pathways, and conventional immunological danger recognition could occur. Therefore, in arterial sinusoidal segments with no sea of PAMPs, the liver could recognize real danger and support innate and adaptive immunity.
Collapse
Affiliation(s)
- Andrea Henriques-Pons
- Laboratorio de Inovações em Terapias, Ensino e Bioprodutos, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro, Brazil
| | - Natália Vacani-Martins
- Laboratorio de Inovações em Terapias, Ensino e Bioprodutos, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro, Brazil
| | | | - Marcelo Meuser-Batista
- Laboratório de Educação Profissional em Técnicas Laboratoriais em Saúde, Escola Politecnica de Saúde Joaquim Venâncio, Fundação Oswaldo Cruz, Rio de Janeiro, Brazil
| |
Collapse
|
11
|
Czyzynska-Cichon I, Kotlinowski J, Blacharczyk O, Giergiel M, Szymanowski K, Metwally S, Wojnar-Lason K, Dobosz E, Koziel J, Lekka M, Chlopicki S, Zapotoczny B. Early and late phases of liver sinusoidal endothelial cell (LSEC) defenestration in mouse model of systemic inflammation. Cell Mol Biol Lett 2024; 29:139. [PMID: 39528938 PMCID: PMC11556108 DOI: 10.1186/s11658-024-00655-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Accepted: 10/21/2024] [Indexed: 11/16/2024] Open
Abstract
BACKGROUND Liver sinusoidal endothelial cells (LSECs) have transcellular pores, called fenestrations, participating in the bidirectional transport between the vascular system and liver parenchyma. Fenestrated LSECs indicate a healthy phenotype of liver while loss of fenestrations (defenestration) in LSECs is associated with liver pathologies. METHODS We introduce a unique model of systemic inflammation triggered by the deletion of Mcpip1 in myeloid leukocytes (Mcpip1fl/flLysMCre) characterised by progressive alterations in LSEC phenotype. We implement multiparametric characterisation of LSECs by using novel real-time atomic force microscopy supported with scanning electron microscopy and quantitative fluorescence microscopy. In addition, we provide genetic profiling, searching for characteristic genes encoding proteins that might be connected with the structure of fenestrations. RESULTS We demonstrate that LSECs in Mcpip1fl/flLysMCre display two phases of defenestration: the early phase, with modest defenestration that was fully reversible using cytochalasin B and the late phase, with severe defenestration that is mostly irreversible. By thorough analysis of LSEC porosity, elastic modulus and actin abundance in Mcpip1fl/flLysMCre and in response to cytochalasin B, we demonstrate that proteins other than actin must be additionally responsible for inducing open fenestrations. We highlight several genes that were severely affected in the late but not in the early phase of LSEC defenestration shedding a light on complex structure of individual fenestrations. CONCLUSIONS The presented model of LSEC derived from Mcpip1fl/flLysMCre provides a valuable reference for developing novel strategies for LSEC refenestration in the early and late phases of liver pathology.
Collapse
Affiliation(s)
- Izabela Czyzynska-Cichon
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, Bobrzynskiego 14, 30-348, Krakow, Poland
| | - Jerzy Kotlinowski
- Department of General Biochemistry, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387, Krakow, Poland
| | - Oliwia Blacharczyk
- Institute of Nuclear Physics Polish Academy of Sciences, 31342, Krakow, Poland
| | - Magdalena Giergiel
- Centre for Nanometer-Scale Science and Advanced Materials (NANOSAM), Faculty of Physics, Astronomy, and Applied Computer Science, Jagiellonian University, Krakow, Poland
| | - Konrad Szymanowski
- Institute of Nuclear Physics Polish Academy of Sciences, 31342, Krakow, Poland
| | - Sara Metwally
- Institute of Nuclear Physics Polish Academy of Sciences, 31342, Krakow, Poland
| | - Kamila Wojnar-Lason
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, Bobrzynskiego 14, 30-348, Krakow, Poland
- Department of Pharmacology, Jagiellonian University Medical College, Grzegorzecka 16, 31-531, Krakow, Poland
| | - Ewelina Dobosz
- Department of Microbiology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387, Krakow, Poland
| | - Joanna Koziel
- Department of Microbiology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387, Krakow, Poland
| | - Malgorzata Lekka
- Institute of Nuclear Physics Polish Academy of Sciences, 31342, Krakow, Poland
| | - Stefan Chlopicki
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, Bobrzynskiego 14, 30-348, Krakow, Poland
- Department of Pharmacology, Jagiellonian University Medical College, Grzegorzecka 16, 31-531, Krakow, Poland
| | | |
Collapse
|
12
|
Bhandari S, Kyrrestad I, Simón-Santamaría J, Li R, Szafranska KJ, Dumitriu G, Sánchez Romano J, Smedsrød B, Sørensen KK. Mouse liver sinusoidal endothelial cell responses to the glucocorticoid receptor agonist dexamethasone. Front Pharmacol 2024; 15:1377136. [PMID: 39439887 PMCID: PMC11494038 DOI: 10.3389/fphar.2024.1377136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Accepted: 09/13/2024] [Indexed: 10/25/2024] Open
Abstract
Liver sinusoidal endothelial cells (LSECs) which make up the fenestrated wall of the hepatic sinusoids, are active scavenger cells involved in blood waste clearance and liver immune functions. Dexamethasone is a synthetic glucocorticoid commonly used in the clinic and as cell culture supplement. However, the response is dependent on tissue, cell type, and cell state. The aim of this study was to investigate the effect of dexamethasone on primary mouse LSECs (C57BL/6J); their viability (live-dead, LDH release, caspase 3/7 assays), morphology (scanning electron microscopy), release of inflammatory markers (ELISA), and scavenging functions (endocytosis assays), and associated biological processes and pathways. We have characterized and catalogued the proteome of LSECs cultured for 1, 10, or 48 h to elucidate time-dependent and dexamethasone-specific cell responses. More than 6,000 protein IDs were quantified using tandem mass tag technology and advanced mass spectrometry (synchronous precursor selection multi-notch MS3). Enrichment analysis showed a culture-induced upregulation of stress and inflammatory markers, and a significant shift in cell metabolism already at 10 h, with enhancement of glycolysis and concomitant repression of oxidative phosphorylation. At 48 h, changes in metabolic pathways were more pronounced with dexamethasone compared to time-matched controls. Dexamethasone repressed the activation of inflammatory pathways (IFN-gamma response, TNF-alpha signaling via NF-kB, Cell adhesion molecules), and culture-induced release of interleukin-6, VCAM-1, and ICAM-1, and improved cell viability partly through inhibition of apoptosis. The mouse LSECs did not proliferate in culture. Dexamethasone treated cells showed upregulation of xanthine dehydrogenase/oxidase (Xdh), and the transcription regulator Foxo1. The drug further delayed but did not block the culture-induced loss of LSEC fenestration. The LSEC capacity for endocytosis was significantly reduced at 48 h, independent of dexamethasone, which correlated with diminished expression of several scavenger receptors and C-type lectins and altered expression of proteins in the endocytic machinery. The glucocorticoid receptor (NR3C1) was suppressed by dexamethasone at 48 h, suggesting limited effect of the drug in prolonged LSEC culture. Conclusion: The study presents a detailed overview of biological processes and pathways affected by dexamethasone in mouse LSECs in vitro.
Collapse
|
13
|
Somnay K, Wadgaonkar P, Sridhar N, Roshni P, Rao N, Wadgaonkar R. Liver Fibrosis Leading to Cirrhosis: Basic Mechanisms and Clinical Perspectives. Biomedicines 2024; 12:2229. [PMID: 39457542 PMCID: PMC11505165 DOI: 10.3390/biomedicines12102229] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Revised: 09/20/2024] [Accepted: 09/23/2024] [Indexed: 10/28/2024] Open
Abstract
Liver fibrosis is the pathological deposition of extracellular matrix rich in fibrillar collagen within the hepatocytes in response to chronic liver injury due to various causes. As the condition advances, it can progress to cirrhosis, the late stages of which are irreversible. Multiple pathophysiological mechanisms and cell types are responsible for the progression of liver fibrosis and cirrhosis. Hepatic stellate cells and myofibroblast activation represent a key event in fibrosis. Capillarization of liver sinusoidal endothelial cells further contributes to extracellular matrix deposition and an increase in portal pressure. Macrophages and neutrophils produce inflammatory cytokines and participate in activating hepatic stellate cells. Although initially believed to be irreversible, early stages of fibrosis are now found to be reversible. Furthermore, advances in noninvasive imaging and serum studies have changed and improved how cirrhosis can be evaluated and monitored. Although there are currently no specific approved therapies to reverse liver fibrosis, management of underlying diseases has been found to halt the progression, and to an extent, even reverse liver fibrosis, preventing further liver injury and cirrhosis-related complications.
Collapse
Affiliation(s)
- Kaumudi Somnay
- New York Presbyterian Hospital, Queens, New York, NY 11355, USA
- New York Digestive Disease Center, Queens, New York, NY 11355, USA
| | | | | | - Prarath Roshni
- New York Digestive Disease Center, Queens, New York, NY 11355, USA
| | - Nachiketh Rao
- New York Digestive Disease Center, Queens, New York, NY 11355, USA
| | - Raj Wadgaonkar
- SUNY Downstate Medical Center, Brooklyn, New York, NY 11203, USA;
| |
Collapse
|
14
|
Mou X, Leeman SM, Roye Y, Miller C, Musah S. Fenestrated Endothelial Cells across Organs: Insights into Kidney Function and Disease. Int J Mol Sci 2024; 25:9107. [PMID: 39201792 PMCID: PMC11354928 DOI: 10.3390/ijms25169107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Revised: 08/07/2024] [Accepted: 08/19/2024] [Indexed: 09/03/2024] Open
Abstract
In the human body, the vascular system plays an indispensable role in maintaining homeostasis by supplying oxygen and nutrients to cells and organs and facilitating the removal of metabolic waste and toxins. Blood vessels-the key constituents of the vascular system-are composed of a layer of endothelial cells on their luminal surface. In most organs, tightly packed endothelial cells serve as a barrier separating blood and lymph from surrounding tissues. Intriguingly, endothelial cells in some tissues and organs (e.g., choroid plexus, liver sinusoids, small intestines, and kidney glomerulus) form transcellular pores called fenestrations that facilitate molecular and ionic transport across the vasculature and mediate immune responses through leukocyte transmigration. However, the development and unique functions of endothelial cell fenestrations across organs are yet to be fully uncovered. This review article provides an overview of fenestrated endothelial cells in multiple organs. We describe their development and organ-specific roles, with expanded discussions on their contributions to glomerular health and disease. We extend these discussions to highlight the dynamic changes in endothelial cell fenestrations in diabetic nephropathy, focal segmental glomerulosclerosis, Alport syndrome, and preeclampsia, and how these unique cellular features could be targeted for therapeutic development. Finally, we discuss emerging technologies for in vitro modeling of biological systems, and their relevance for advancing the current understanding of endothelial cell fenestrations in health and disease.
Collapse
Affiliation(s)
- Xingrui Mou
- Department of Biomedical Engineering, Pratt School of Engineering, Duke University, Durham, NC 27710, USA
| | - Sophia M. Leeman
- Department of Biomedical Engineering, Pratt School of Engineering, Duke University, Durham, NC 27710, USA
- Department of Computer Science, Duke University, Durham, NC 27710, USA
| | - Yasmin Roye
- Department of Biomedical Engineering, Pratt School of Engineering, Duke University, Durham, NC 27710, USA
| | - Carmen Miller
- Department of Biology, Duke University, Durham, NC 27710, USA
| | - Samira Musah
- Department of Biomedical Engineering, Pratt School of Engineering, Duke University, Durham, NC 27710, USA
- Center for Biomolecular and Tissue Engineering, Duke University, Durham, NC 27710, USA
- Division of Nephrology, Department of Medicine, School of Medicine, Duke University, Durham, NC 27710, USA
- Department of Cell Biology, Duke University, Durham, NC 27710, USA
- Faculty of the Developmental and Stem Cell Biology Program, Duke Regeneration Center, Duke MEDx Initiative, Duke University, Durham, NC 27710, USA
| |
Collapse
|
15
|
Liu P, Liang WL, Huang RT, Chen XX, Zou DH, Kurihara H, Li YF, Xu YH, Ouyang SH, He RR. Hepatic microcirculatory disturbance in liver diseases: intervention with traditional Chinese medicine. Front Pharmacol 2024; 15:1399598. [PMID: 39108760 PMCID: PMC11300221 DOI: 10.3389/fphar.2024.1399598] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Accepted: 06/25/2024] [Indexed: 04/18/2025] Open
Abstract
The liver, a complex parenchymal organ, possesses a distinctive microcirculatory system crucial for its physiological functions. An intricate interplay exists between hepatic microcirculatory disturbance and the manifestation of pathological features in diverse liver diseases. This review updates the main characteristics of hepatic microcirculatory disturbance, including hepatic sinusoidal capillarization, narrowing of sinusoidal space, portal hypertension, and pathological angiogenesis, as well as their formation mechanisms. It also summarized the detection methods for hepatic microcirculation. Simultaneously, we have also reviewed the characteristics of microcirculatory disturbance in diverse liver diseases such as acute liver failure, hepatic ischemia-reperfusion injury, viral hepatitis, non-alcoholic fatty liver disease, hepatic fibrosis, hepatic cirrhosis, and hepatocellular carcinoma. Finally, this review also summarizes the advancement in hepatic microcirculation attributed to traditional Chinese medicine (TCM) and its active metabolites, providing novel insights into the application of TCM in treating liver diseases.
Collapse
Affiliation(s)
- Pei Liu
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau SAR, China
- Guangdong Engineering Research Center of Traditional Chinese Medicine & Disease Susceptibility, Guangdong-Hong Kong-Macao Universities Joint Laboratory for the Internationalization of Traditional Chinese Medicine, Guangzhou Key Laboratory of Traditional Chinese Medicine & Disease Susceptibility, International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Chinese Ministry of Education (MOE), Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou, China
| | - Wan-Li Liang
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau SAR, China
- Guangdong Engineering Research Center of Traditional Chinese Medicine & Disease Susceptibility, Guangdong-Hong Kong-Macao Universities Joint Laboratory for the Internationalization of Traditional Chinese Medicine, Guangzhou Key Laboratory of Traditional Chinese Medicine & Disease Susceptibility, International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Chinese Ministry of Education (MOE), Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou, China
| | - Rui-Ting Huang
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau SAR, China
- Guangdong Engineering Research Center of Traditional Chinese Medicine & Disease Susceptibility, Guangdong-Hong Kong-Macao Universities Joint Laboratory for the Internationalization of Traditional Chinese Medicine, Guangzhou Key Laboratory of Traditional Chinese Medicine & Disease Susceptibility, International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Chinese Ministry of Education (MOE), Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou, China
| | - Xin-Xing Chen
- Guangdong Engineering Research Center of Traditional Chinese Medicine & Disease Susceptibility, Guangdong-Hong Kong-Macao Universities Joint Laboratory for the Internationalization of Traditional Chinese Medicine, Guangzhou Key Laboratory of Traditional Chinese Medicine & Disease Susceptibility, International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Chinese Ministry of Education (MOE), Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou, China
| | - De-Hua Zou
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau SAR, China
- Guangdong Engineering Research Center of Traditional Chinese Medicine & Disease Susceptibility, Guangdong-Hong Kong-Macao Universities Joint Laboratory for the Internationalization of Traditional Chinese Medicine, Guangzhou Key Laboratory of Traditional Chinese Medicine & Disease Susceptibility, International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Chinese Ministry of Education (MOE), Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou, China
| | - Hiroshi Kurihara
- Guangdong Engineering Research Center of Traditional Chinese Medicine & Disease Susceptibility, Guangdong-Hong Kong-Macao Universities Joint Laboratory for the Internationalization of Traditional Chinese Medicine, Guangzhou Key Laboratory of Traditional Chinese Medicine & Disease Susceptibility, International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Chinese Ministry of Education (MOE), Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou, China
| | - Yi-Fang Li
- Guangdong Engineering Research Center of Traditional Chinese Medicine & Disease Susceptibility, Guangdong-Hong Kong-Macao Universities Joint Laboratory for the Internationalization of Traditional Chinese Medicine, Guangzhou Key Laboratory of Traditional Chinese Medicine & Disease Susceptibility, International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Chinese Ministry of Education (MOE), Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou, China
| | - You-Hua Xu
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau SAR, China
| | - Shu-Hua Ouyang
- Guangdong Engineering Research Center of Traditional Chinese Medicine & Disease Susceptibility, Guangdong-Hong Kong-Macao Universities Joint Laboratory for the Internationalization of Traditional Chinese Medicine, Guangzhou Key Laboratory of Traditional Chinese Medicine & Disease Susceptibility, International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Chinese Ministry of Education (MOE), Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou, China
| | - Rong-Rong He
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau SAR, China
- Guangdong Engineering Research Center of Traditional Chinese Medicine & Disease Susceptibility, Guangdong-Hong Kong-Macao Universities Joint Laboratory for the Internationalization of Traditional Chinese Medicine, Guangzhou Key Laboratory of Traditional Chinese Medicine & Disease Susceptibility, International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Chinese Ministry of Education (MOE), Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou, China
| |
Collapse
|
16
|
Wang L, He L, Yi W, Wang M, Xu F, Liu H, Nie J, Pan YH, Dang S, Zhang W. ADAMTS18-fibronectin interaction regulates the morphology of liver sinusoidal endothelial cells. iScience 2024; 27:110273. [PMID: 39040056 PMCID: PMC11261151 DOI: 10.1016/j.isci.2024.110273] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Revised: 05/12/2024] [Accepted: 06/12/2024] [Indexed: 07/24/2024] Open
Abstract
Liver sinusoidal endothelial cells (LSECs) have a unique morphological structure known as "fenestra" that plays a crucial role in liver substance exchange and homeostasis maintenance. In this study, we demonstrate that ADAMTS18 protease is primarily secreted by fetal liver endothelial cells. ADAMTS18 deficiency leads to enlarged fenestrae and increased porosity of LSECs, microthrombus formation in liver vessels, and an imbalance of liver oxidative stress. These defects worsen carbon tetrachloride (CCl4)-induced liver fibrosis and diethylnitrosamine (DEN)/high-fat-induced hepatocellular carcinoma (HCC) in adult Adamts18-deficient mice. Mechanically, ADAMTS18 functions as a modifier of fibronectin (FN) to regulate the morphological acquisition of LSECs via the vascular endothelial growth factor A (VEGFA) signaling pathways. Collectively, a mechanism is proposed for LSEC morphogenesis and liver homeostasis maintenance via ADAMTS18-FN-VEGFA niches.
Collapse
Affiliation(s)
- Liya Wang
- Key Laboratory of Brain Functional Genomics (Ministry of Education and Shanghai), School of Life Sciences, East China Normal University, Shanghai, China
| | - Li He
- Key Laboratory of Brain Functional Genomics (Ministry of Education and Shanghai), School of Life Sciences, East China Normal University, Shanghai, China
| | - Weijia Yi
- Key Laboratory of Brain Functional Genomics (Ministry of Education and Shanghai), School of Life Sciences, East China Normal University, Shanghai, China
| | - Min Wang
- Key Laboratory of Brain Functional Genomics (Ministry of Education and Shanghai), School of Life Sciences, East China Normal University, Shanghai, China
| | - Fangmin Xu
- Key Laboratory of Brain Functional Genomics (Ministry of Education and Shanghai), School of Life Sciences, East China Normal University, Shanghai, China
| | - Hanlin Liu
- Key Laboratory of Brain Functional Genomics (Ministry of Education and Shanghai), School of Life Sciences, East China Normal University, Shanghai, China
| | - Jiahui Nie
- Key Laboratory of Brain Functional Genomics (Ministry of Education and Shanghai), School of Life Sciences, East China Normal University, Shanghai, China
| | - Yi-Hsuan Pan
- Key Laboratory of Brain Functional Genomics (Ministry of Education and Shanghai), School of Life Sciences, East China Normal University, Shanghai, China
| | - Suying Dang
- Department of Biochemistry and Molecular Cell Biology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wei Zhang
- Key Laboratory of Brain Functional Genomics (Ministry of Education and Shanghai), School of Life Sciences, East China Normal University, Shanghai, China
| |
Collapse
|
17
|
Ciriaci N, Rautou PE, Poisson J. Loss of fenestrae in liver sinusoidal endothelial cells contributes to MASLD. NATURE CARDIOVASCULAR RESEARCH 2024; 3:622-624. [PMID: 39196234 DOI: 10.1038/s44161-024-00490-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/29/2024]
Affiliation(s)
- Nadia Ciriaci
- Université Paris-Cité, Inserm, Centre de Recherche sur l'Inflammation, UMR 1149, Paris, France
| | - Pierre-Emmanuel Rautou
- Université Paris-Cité, Inserm, Centre de Recherche sur l'Inflammation, UMR 1149, Paris, France
- AP-HP, Hôpital Beaujon, Service d'Hépatologie, DMU DIGEST, Centre de Référence des Maladies Vasculaires du Foie, FILFOIE, ERN RARE-LIVER, Clichy, France
| | - Johanne Poisson
- Université Paris-Cité, Inserm, Centre de Recherche sur l'Inflammation, UMR 1149, Paris, France.
- AP-HP, Hôpital Européen George Pompidou, Hôpital Corentin-Celton, Service de Gériatrie, Institut CARPEM, Paris, France.
| |
Collapse
|
18
|
Miatmoko A, Octavia RT, Araki T, Annoura T, Sari R. Advancing liposome technology for innovative strategies against malaria. Saudi Pharm J 2024; 32:102085. [PMID: 38690211 PMCID: PMC11059525 DOI: 10.1016/j.jsps.2024.102085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2023] [Accepted: 04/23/2024] [Indexed: 05/02/2024] Open
Abstract
This review discusses the potential of liposomes as drug delivery systems for antimalarial therapies. Malaria continues to be a significant cause of mortality and morbidity, particularly among children and pregnant women. Drug resistance due to patient non-compliance and troublesome side effects remains a significant challenge in antimalarial treatment. Liposomes, as targeted and efficient drug carriers, have garnered attention owing to their ability to address these issues. Liposomes encapsulate hydrophilic and/or hydrophobic drugs, thus providing comprehensive and suitable therapeutic drug delivery. Moreover, the potential of passive and active drug delivery enables drug concentration in specific target tissues while reducing adverse effects. However, successful liposome formulation is influenced by various factors, including drug physicochemical characteristics and physiological barriers encountered during drug delivery. To overcome these challenges, researchers have explored modifications in liposome nanocarriers to achieve efficient drug loading, controlled release, and system stability. Computational approaches have also been adopted to predict liposome system stability, membrane integrity, and drug-liposome interactions, improving formulation development efficiency. By leveraging computational methods, optimizing liposomal drug delivery systems holds promise for enhancing treatment efficacy and minimizing side effects in malaria therapy. This review consolidates the current understanding and highlights the potential of liposome strategies against malaria.
Collapse
Affiliation(s)
- Andang Miatmoko
- Department of Pharmaceutical Science, Faculty of Pharmacy, Universitas Airlangga, Campus C UNAIR Mulyorejo, Surabaya 60115, Indonesia
- Stem Cell Research and Development Center, Universitas Airlangga, 2 Floor Institute of Tropical Disease Building, Campus C UNAIR Mulyorejo, Surabaya 60115, Indonesia
- Nanotechnology and Drug Delivery System Research Group, Faculty of Pharmacy, Universitas Airlangga, Campus C UNAIR Mulyorejo, Surabaya 60115, Indonesia
| | - Rifda Tarimi Octavia
- Master Program of Pharmaceutical Sciences, Faculty of Pharmacy, Universitas Airlangga, Campus C UNAIR Mulyorejo, Surabaya 60115, Indonesia
| | - Tamasa Araki
- Department of Parasitology, National Institute of Infectious Diseases (NIID), 1-23-1 Toyama, Shinju-ku, Tokyo 162-8640, Japan
| | - Takeshi Annoura
- Department of Parasitology, National Institute of Infectious Diseases (NIID), 1-23-1 Toyama, Shinju-ku, Tokyo 162-8640, Japan
| | - Retno Sari
- Department of Pharmaceutical Science, Faculty of Pharmacy, Universitas Airlangga, Campus C UNAIR Mulyorejo, Surabaya 60115, Indonesia
| |
Collapse
|
19
|
Czyzynska-Cichon I, Giergiel M, Kwiatkowski G, Kurpinska A, Wojnar-Lason K, Kaczara P, Szymonski M, Lekka M, Kalvins I, Zapotoczny B, Chlopicki S. Protein disulfide isomerase A1 regulates fenestration dynamics in primary mouse liver sinusoidal endothelial cells (LSECs). Redox Biol 2024; 72:103162. [PMID: 38669864 PMCID: PMC11068635 DOI: 10.1016/j.redox.2024.103162] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Revised: 04/06/2024] [Accepted: 04/15/2024] [Indexed: 04/28/2024] Open
Abstract
Protein disulfide isomerases (PDIs) are involved in many intracellular and extracellular processes, including cell adhesion and cytoskeletal reorganisation, but their contribution to the regulation of fenestrations in liver sinusoidal endothelial cells (LSECs) remains unknown. Given that fenestrations are supported on a cytoskeleton scaffold, this study aimed to investigate whether endothelial PDIs regulate fenestration dynamics in primary mouse LSECs. PDIA3 and PDIA1 were found to be the most abundant among PDI isoforms in LSECs. Taking advantage of atomic force microscopy, the effects of PDIA1 or PDIA3 inhibition on the fenestrations in LSECs were investigated using a classic PDIA1 inhibitor (bepristat) and novel aromatic N-sulfonamides of aziridine-2-carboxylic acid derivatives as PDIA1 (C-3389) or PDIA3 (C-3399) inhibitors. The effect of PDIA1 inhibition on liver perfusion was studied in vivo using dynamic contrast-enhanced magnetic resonance imaging. Additionally, PDIA1 inhibitors were examined in vitro in LSECs for effects on adhesion, cytoskeleton organisation, bioenergetics, and viability. Inhibition of PDIA1 with bepristat or C-3389 significantly reduced the number of fenestrations in LSECs, while inhibition of PDIA3 with C-3399 had no effect. Moreover, the blocking of free thiols by the cell-penetrating N-ethylmaleimide, but not by the non-cell-penetrating 4-chloromercuribenzenesulfonate, resulted in LSEC defenestration. Inhibition of PDIA1 did not affect LSEC adhesion, viability, and bioenergetics, nor did it induce a clear-cut rearrangement of the cytoskeleton. However, PDIA1-dependent defenestration was reversed by cytochalasin B, a known fenestration stimulator, pointing to the preserved ability of LSECs to form new pores. Importantly, systemic inhibition of PDIA1 in vivo affected intra-parenchymal uptake of contrast agent in mice consistent with LSEC defenestration. These results revealed the role of intracellular PDIA1 in the regulation of fenestration dynamics in LSECs, and in maintaining hepatic sinusoid homeostasis.
Collapse
Affiliation(s)
- Izabela Czyzynska-Cichon
- Jagiellonian University, Jagiellonian Centre for Experimental Therapeutics (JCET), Bobrzynskiego 14, 30-348, Krakow, Poland
| | - Magdalena Giergiel
- Jagiellonian University, Centre for Nanometer-Scale Science and Advanced Materials, NANOSAM, Faculty of Physics, Astronomy, and Applied Computer Science, Lojasiewicza 11, 30-348, Krakow, Poland
| | - Grzegorz Kwiatkowski
- Jagiellonian University, Jagiellonian Centre for Experimental Therapeutics (JCET), Bobrzynskiego 14, 30-348, Krakow, Poland
| | - Anna Kurpinska
- Jagiellonian University, Jagiellonian Centre for Experimental Therapeutics (JCET), Bobrzynskiego 14, 30-348, Krakow, Poland
| | - Kamila Wojnar-Lason
- Jagiellonian University, Jagiellonian Centre for Experimental Therapeutics (JCET), Bobrzynskiego 14, 30-348, Krakow, Poland; Jagiellonian University Medical College, Faculty of Medicine, Department of Pharmacology, Grzegorzecka 16, 31-531, Krakow, Poland
| | - Patrycja Kaczara
- Jagiellonian University, Jagiellonian Centre for Experimental Therapeutics (JCET), Bobrzynskiego 14, 30-348, Krakow, Poland
| | - Marek Szymonski
- Jagiellonian University, Centre for Nanometer-Scale Science and Advanced Materials, NANOSAM, Faculty of Physics, Astronomy, and Applied Computer Science, Lojasiewicza 11, 30-348, Krakow, Poland
| | - Malgorzata Lekka
- Institute of Nuclear Physics Polish Academy of Sciences, Radzikowskiego 152, 31-342, Krakow, Poland
| | - Ivars Kalvins
- Laboratory of Carbofunctional Compounds, Latvian Institute of Organic Synthesis, LV-1006, Riga, Latvia
| | - Bartlomiej Zapotoczny
- Jagiellonian University, Centre for Nanometer-Scale Science and Advanced Materials, NANOSAM, Faculty of Physics, Astronomy, and Applied Computer Science, Lojasiewicza 11, 30-348, Krakow, Poland; Institute of Nuclear Physics Polish Academy of Sciences, Radzikowskiego 152, 31-342, Krakow, Poland.
| | - Stefan Chlopicki
- Jagiellonian University, Jagiellonian Centre for Experimental Therapeutics (JCET), Bobrzynskiego 14, 30-348, Krakow, Poland; Jagiellonian University Medical College, Faculty of Medicine, Department of Pharmacology, Grzegorzecka 16, 31-531, Krakow, Poland.
| |
Collapse
|
20
|
Gomes F, Ribeiro AC, Sanches GS, Borges HS, Takahashi LAU, Daniel-Ribeiro CT, Tedesco AC, Nascimento JWL, Carvalho LJM. A nanochitosan-D-galactose formulation increases the accumulation of primaquine in the liver. Antimicrob Agents Chemother 2024; 68:e0091523. [PMID: 38517190 PMCID: PMC11064505 DOI: 10.1128/aac.00915-23] [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: 07/11/2023] [Accepted: 02/25/2024] [Indexed: 03/23/2024] Open
Abstract
Primaquine is the mainstream antimalarial drug to prevent Plasmodium vivax relapses. However, this drug can induce hemolysis in patients with glucose-6-phosphate dehydrogenase deficiency. Nanostructure formulations of primaquine loaded with D-galactose were used as a strategy to target the drug to the liver and decrease the hemolytic risks. Nanoemulsion (NE-Pq) and nanochitosan (NQ-Pq) formulations of primaquine diphosphate containing D-galactose were prepared and characterized by their physicochemistry properties. Pharmacokinetic and biodistribution studies were conducted using Swiss Webster mice. A single dose of 10 mg/kg of each nanoformulation or free primaquine solution was administered by gavage to the animals, which were killed at 0.5, 1, 2, 4, 8, and 24 hours. Blood samples and tissues were collected, processed, and analyzed by high-performance liquid chromatography. The nanoformulation showed sizes around 200 nm (NE-Pq) and 400 nm (NQ-Pq) and physicochemical stability for over 30 days. Free primaquine solution achieved higher primaquine Cmax in the liver than NE-Pq or NQ-Pq at 0.5 hours. However, the half-life and mean residence time (MRT) of primaquine in the liver were three times higher with the NQ-Pq formulation than with free primaquine, and the volume distribution was four times higher. Conversely, primaquine's half-life, MRT, and volume distribution in the plasma were lower for NQ-Pq than for free primaquine. NE-Pq, on the other hand, accumulated more in the lungs but not in the liver. Galactose-coated primaquine nanochitosan formulation showed increased drug targeting to the liver compared to free primaquine and may represent a promising strategy for a more efficient and safer radical cure for vivax malaria.
Collapse
Affiliation(s)
- F. Gomes
- Laboratory of Malaria Research, Oswaldo Cruz Institute (IOC/Fiocruz), Reference Center for Malaria Research, Diagnosis and Training, Rio de Janeiro, Brazil
| | - A. C. Ribeiro
- Laboratory of Malaria Research, Oswaldo Cruz Institute (IOC/Fiocruz), Reference Center for Malaria Research, Diagnosis and Training, Rio de Janeiro, Brazil
- Department of Pharmacology (LaFaCE) - ICB, Federal University of Juiz de Fora (UFJF), Juiz de Fora, Minas Gerais, Brazil
| | - G. S. Sanches
- Laboratory of Malaria Research, Oswaldo Cruz Institute (IOC/Fiocruz), Reference Center for Malaria Research, Diagnosis and Training, Rio de Janeiro, Brazil
| | - H. S. Borges
- Department of Chemistry, Center of Nanotechnology and Tissue Engineering - Photobiology and Photomedicine Research Group, Faculty of Philosophy, Sciences and Letters, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - L. A. U. Takahashi
- Department of Chemistry, Center of Nanotechnology and Tissue Engineering - Photobiology and Photomedicine Research Group, Faculty of Philosophy, Sciences and Letters, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - C. T. Daniel-Ribeiro
- Laboratory of Malaria Research, Oswaldo Cruz Institute (IOC/Fiocruz), Reference Center for Malaria Research, Diagnosis and Training, Rio de Janeiro, Brazil
| | - A. C. Tedesco
- Department of Chemistry, Center of Nanotechnology and Tissue Engineering - Photobiology and Photomedicine Research Group, Faculty of Philosophy, Sciences and Letters, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - J. W. L. Nascimento
- Department of Pharmacology (LaFaCE) - ICB, Federal University of Juiz de Fora (UFJF), Juiz de Fora, Minas Gerais, Brazil
| | - L. J. M. Carvalho
- Laboratory of Malaria Research, Oswaldo Cruz Institute (IOC/Fiocruz), Reference Center for Malaria Research, Diagnosis and Training, Rio de Janeiro, Brazil
| |
Collapse
|
21
|
McCourt P. Go with the flow: Hemodynamic changes affect liver ultrastructure. Acta Physiol (Oxf) 2024; 240:e14141. [PMID: 38523468 DOI: 10.1111/apha.14141] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Accepted: 03/18/2024] [Indexed: 03/26/2024]
Affiliation(s)
- Peter McCourt
- Department of Medical Biology, UiT Norges arktiske universitet, Tromsø, Norway
| |
Collapse
|
22
|
Zhang X, Li P, Zhou J, Zhang Z, Wu H, Shu X, Li W, Wu Y, Du Y, Lü D, Lü S, Li N, Long M. FAK-p38 signaling serves as a potential target for reverting matrix stiffness-modulated liver sinusoidal endothelial cell defenestration. Biomaterials 2024; 305:122462. [PMID: 38171118 DOI: 10.1016/j.biomaterials.2023.122462] [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: 10/17/2023] [Revised: 12/26/2023] [Accepted: 12/31/2023] [Indexed: 01/05/2024]
Abstract
Liver sinusoidal endothelial cells (LSECs) are highly specific endothelial cells which play an essential role in the maintenance of liver homeostasis. During the progression of liver fibrosis, matrix stiffening promotes LSEC defenestration, however, the underlying mechanotransduction mechanism remains poorly understood. Here, we applied stiffness-tunable hydrogels to assess the matrix stiffening-induced phenotypic changes in primary mouse LSECs. Results indicated that increased stiffness promoted LSEC defenestration through cytoskeletal reorganization. LSECs sensed the increased matrix stiffness via focal adhesion kinase (FAK), leading to the activation of p38-mitogen activated protein kinase activated protein kinase 2 (MK2) pathway, thereby inducing actin remodeling via LIM Kinase 1 (LIMK1) and Cofilin. Interestingly, inhibition of FAK or p38-MK2 pathway was able to effectively restore the fenestrae to a certain degree in LSECs isolated from early to late stages of liver fibrosis mice. Thus, this study highlights the impact of mechanotransduction in LSEC defenestration, and provides novel insights for potential therapeutic interventions for liver fibrosis.
Collapse
Affiliation(s)
- Xiaoyu Zhang
- Center for Biomechanics and Bioengineering, Beijing Key Laboratory of Engineered Construction and Mechanobiology and Key Laboratory of Microgravity (National Microgravity Laboratory), Institute of Mechanics, Chinese Academy of Sciences, Beijing, 100190, China; School of Engineering Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Peiwen Li
- Center for Biomechanics and Bioengineering, Beijing Key Laboratory of Engineered Construction and Mechanobiology and Key Laboratory of Microgravity (National Microgravity Laboratory), Institute of Mechanics, Chinese Academy of Sciences, Beijing, 100190, China
| | - Jin Zhou
- Center for Biomechanics and Bioengineering, Beijing Key Laboratory of Engineered Construction and Mechanobiology and Key Laboratory of Microgravity (National Microgravity Laboratory), Institute of Mechanics, Chinese Academy of Sciences, Beijing, 100190, China
| | - Ziliang Zhang
- Center for Biomechanics and Bioengineering, Beijing Key Laboratory of Engineered Construction and Mechanobiology and Key Laboratory of Microgravity (National Microgravity Laboratory), Institute of Mechanics, Chinese Academy of Sciences, Beijing, 100190, China; Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, 250117, China
| | - Huan Wu
- Center for Biomechanics and Bioengineering, Beijing Key Laboratory of Engineered Construction and Mechanobiology and Key Laboratory of Microgravity (National Microgravity Laboratory), Institute of Mechanics, Chinese Academy of Sciences, Beijing, 100190, China
| | - Xinyu Shu
- Center for Biomechanics and Bioengineering, Beijing Key Laboratory of Engineered Construction and Mechanobiology and Key Laboratory of Microgravity (National Microgravity Laboratory), Institute of Mechanics, Chinese Academy of Sciences, Beijing, 100190, China; School of Engineering Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Wang Li
- Center for Biomechanics and Bioengineering, Beijing Key Laboratory of Engineered Construction and Mechanobiology and Key Laboratory of Microgravity (National Microgravity Laboratory), Institute of Mechanics, Chinese Academy of Sciences, Beijing, 100190, China; School of Engineering Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Yi Wu
- Center for Biomechanics and Bioengineering, Beijing Key Laboratory of Engineered Construction and Mechanobiology and Key Laboratory of Microgravity (National Microgravity Laboratory), Institute of Mechanics, Chinese Academy of Sciences, Beijing, 100190, China; School of Engineering Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Yu Du
- Center for Biomechanics and Bioengineering, Beijing Key Laboratory of Engineered Construction and Mechanobiology and Key Laboratory of Microgravity (National Microgravity Laboratory), Institute of Mechanics, Chinese Academy of Sciences, Beijing, 100190, China
| | - Dongyuan Lü
- Center for Biomechanics and Bioengineering, Beijing Key Laboratory of Engineered Construction and Mechanobiology and Key Laboratory of Microgravity (National Microgravity Laboratory), Institute of Mechanics, Chinese Academy of Sciences, Beijing, 100190, China; School of Engineering Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Shouqin Lü
- Center for Biomechanics and Bioengineering, Beijing Key Laboratory of Engineered Construction and Mechanobiology and Key Laboratory of Microgravity (National Microgravity Laboratory), Institute of Mechanics, Chinese Academy of Sciences, Beijing, 100190, China; School of Engineering Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Ning Li
- Center for Biomechanics and Bioengineering, Beijing Key Laboratory of Engineered Construction and Mechanobiology and Key Laboratory of Microgravity (National Microgravity Laboratory), Institute of Mechanics, Chinese Academy of Sciences, Beijing, 100190, China; School of Engineering Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China.
| | - Mian Long
- Center for Biomechanics and Bioengineering, Beijing Key Laboratory of Engineered Construction and Mechanobiology and Key Laboratory of Microgravity (National Microgravity Laboratory), Institute of Mechanics, Chinese Academy of Sciences, Beijing, 100190, China; School of Engineering Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China.
| |
Collapse
|
23
|
Szafranska K, Sørensen KK, Lalor PF, McCourt P. Sinusoidal cells and liver immunology. SINUSOIDAL CELLS IN LIVER DISEASES 2024:53-75. [DOI: 10.1016/b978-0-323-95262-0.00003-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
24
|
Tavakoli Pirzaman A, Alishah A, Babajani B, Ebrahimi P, Sheikhi SA, Moosaei F, Salarfar A, Doostmohamadian S, Kazemi S. The Role of microRNAs in Hepatocellular Cancer: A Narrative Review Focused on Tumor Microenvironment and Drug Resistance. Technol Cancer Res Treat 2024; 23:15330338241239188. [PMID: 38634139 PMCID: PMC11025440 DOI: 10.1177/15330338241239188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Revised: 01/26/2024] [Accepted: 02/26/2024] [Indexed: 04/19/2024] Open
Abstract
Globally, hepatic cancer ranks fourth in terms of cancer-related mortality and is the sixth most frequent kind of cancer. Around 80% of liver cancers are hepatocellular carcinomas (HCC), which are the leading cause of cancer death. It is well known that HCC may develop resistance to the available chemotherapy treatments very fast. One of the biggest obstacles in providing cancer patients with appropriate care is drug resistance. According to reports, more than 90% of cancer-specific fatalities are caused by treatment resistance. By binding to the 3'-untranslated region of target messenger RNAs (mRNAs), microRNAs (miRNAs), a group of noncoding RNAs which are around 17 to 25 nucleotides long, regulate target gene expression. Moreover, they play role in the control of signaling pathways, cell proliferation, and cell death. As a result, miRNAs play an important role in the microenvironment of HCC by changing immune phenotypes, hypoxic conditions, and acidification, as well as angiogenesis and extracellular matrix components. Moreover, changes in miRNA levels in HCC can effectively resist cancer cells to chemotherapy by affecting various cellular processes such as autophagy, apoptosis, and membrane transporter activity. In the current work, we narratively reviewed the role of miRNAs in HCC, with a special focus on tumor microenvironment and drug resistance.
Collapse
Affiliation(s)
| | - Ali Alishah
- Student Research Committee, Babol University of Medical Sciences, Babol, Iran
| | - Bahareh Babajani
- Student Research Committee, Babol University of Medical Sciences, Babol, Iran
| | - Pouyan Ebrahimi
- Student Research Committee, Babol University of Medical Sciences, Babol, Iran
| | - Seyyed Ali Sheikhi
- Student Research Committee, Babol University of Medical Sciences, Babol, Iran
| | - Farhad Moosaei
- Student Research Committee, Babol University of Medical Sciences, Babol, Iran
| | | | | | - Sohrab Kazemi
- Cellular and Molecular Biology Research Center, Health Research Center, Babol University of Medical Sciences, Babol, Iran
| |
Collapse
|
25
|
Kyrrestad I, Larsen AK, Sánchez Romano J, Simón-Santamaría J, Li R, Sørensen KK. Infection of liver sinusoidal endothelial cells with Muromegalovirus muridbeta1 involves binding to neuropilin-1 and is dynamin-dependent. Front Cell Infect Microbiol 2023; 13:1249894. [PMID: 38029264 PMCID: PMC10665495 DOI: 10.3389/fcimb.2023.1249894] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Accepted: 10/23/2023] [Indexed: 12/01/2023] Open
Abstract
Liver sinusoidal endothelial cells (LSEC) are scavenger cells with a remarkably high capacity for clearance of several blood-borne macromolecules and nanoparticles, including some viruses. Endocytosis in LSEC is mainly via the clathrin-coated pit mediated route, which is dynamin-dependent. LSEC can also be a site of infection and latency of betaherpesvirus, but mode of virus entry into these cells has not yet been described. In this study we have investigated the role of dynamin in the early stage of muromegalovirus muridbeta1 (MuHV-1, murid betaherpesvirus 1, murine cytomegalovirus) infection in mouse LSECs. LSEC cultures were freshly prepared from C57Bl/6JRj mouse liver. We first examined dose- and time-dependent effects of two dynamin-inhibitors, dynasore and MitMAB, on cell viability, morphology, and endocytosis of model ligands via different LSEC scavenger receptors to establish a protocol for dynamin-inhibition studies in these primary cells. LSECs were challenged with MuHV-1 (MOI 0.2) ± dynamin inhibitors for 1h, then without inhibitors and virus for 11h, and nuclear expression of MuHV-1 immediate early antigen (IE1) measured by immune fluorescence. MuHV-1 efficiently infected LSECs in vitro. Infection was significantly and independently inhibited by dynasore and MitMAB, which block dynamin function via different mechanisms, suggesting that initial steps of MuHV-1 infection is dynamin-dependent in LSECs. Infection was also reduced in the presence of monensin which inhibits acidification of endosomes. Furthermore, competitive binding studies with a neuropilin-1 antibody blocked LSEC infection. This suggests that MuHV-1 infection in mouse LSECs involves virus binding to neuropilin-1 and occurs via endocytosis.
Collapse
Affiliation(s)
- Ingelin Kyrrestad
- Department of Medical Biology, Faculty of Health Sciences, UiT - The Arctic University of Norway, Tromsø, Norway
| | | | | | | | | | | |
Collapse
|
26
|
Manandhar S, Chambers S, Miller A, Ishii I, Bhatia M. Pharmacological Inhibition and Genetic Deletion of Cystathionine Gamma-Lyase in Mice Protects against Organ Injury in Sepsis: A Key Role of Adhesion Molecules on Endothelial Cells. Int J Mol Sci 2023; 24:13650. [PMID: 37686458 PMCID: PMC10487872 DOI: 10.3390/ijms241713650] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Revised: 08/29/2023] [Accepted: 08/30/2023] [Indexed: 09/10/2023] Open
Abstract
Hydrogen sulfide (H2S), synthesized by cystathionine gamma-lyase (Cth), contributes to the inflammatory response observed in sepsis. This study examines the effect of Cth-derived H2S in adhesion molecules on endothelial cells of vital organs in mice in a cecal ligation puncture (CLP)-induced model of sepsis, using two different and complementary approaches: Cth gene deletion and pharmacological inhibition. Our findings revealed a decreased level of H2S-synthesizing activity (via Cth) in both Cth-/- mice and PAG-treated wild-type (WT) mice following CLP-induced sepsis. Both treatment groups had reduced MPO activity and expression of chemokines (MCP-1 and MIP-2α), adhesion molecules (ICAM-1 and VCAM-1), ERK1/2 phosphorylation, and NF-κB in the liver and lung compared with in CLP-WT mice. Additionally, we found that PAG treatment in Cth-/- mice had no additional effect on the expression of ERK1/2 phosphorylation, NF-κB, or the production of chemokines and adhesion molecules in the liver and lung compared to Cth-/- mice following CLP-induced sepsis. The WT group with sepsis had an increased immunoreactivity of adhesion molecules on endothelial cells in the liver and lung than the WT sham-operated control. The Cth-/-, PAG-treated WT, and Cth-/- groups of mice showed decreased immunoreactivity of adhesion molecules on endothelial cells in the liver and lung following sepsis. Inhibition of H2S production via both approaches reduced adhesion molecule expression on endothelial cells and reduced liver and lung injury in mice with sepsis. In conclusion, this study demonstrates that H2S has an important role in the pathogenesis of sepsis and validates PAG use as a suited tool for investigating the Cth/H2S-signalling axis in sepsis.
Collapse
Affiliation(s)
- Sumeet Manandhar
- Department of Pathology and Biomedical Science, University of Otago, Christchurch 8140, New Zealand; (S.M.); (S.C.); (A.M.)
| | - Stephen Chambers
- Department of Pathology and Biomedical Science, University of Otago, Christchurch 8140, New Zealand; (S.M.); (S.C.); (A.M.)
| | - Andrew Miller
- Department of Pathology and Biomedical Science, University of Otago, Christchurch 8140, New Zealand; (S.M.); (S.C.); (A.M.)
| | - Isao Ishii
- Department of Health Chemistry, Showa Pharmaceutical University, Machida, Tokyo 194-8543, Japan;
| | - Madhav Bhatia
- Department of Pathology and Biomedical Science, University of Otago, Christchurch 8140, New Zealand; (S.M.); (S.C.); (A.M.)
| |
Collapse
|
27
|
Mao H, Szafranska K, Kruse L, Holte C, Wolfson DL, Ahluwalia BS, Whitchurch CB, Cole L, Lockwood GP, Diekmann R, Le Couteur D, Cogger VC, McCourt PAG. Effect of caffeine and other xanthines on liver sinusoidal endothelial cell ultrastructure. Sci Rep 2023; 13:13390. [PMID: 37591901 PMCID: PMC10435486 DOI: 10.1038/s41598-023-40227-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Accepted: 08/07/2023] [Indexed: 08/19/2023] Open
Abstract
Xanthines such as caffeine and theobromine are among the most consumed psychoactive stimulants in the world, either as natural components of coffee, tea and chocolate, or as added ingredients. The present study assessed if xanthines affect liver sinusoidal endothelial cells (LSEC). Cultured primary rat LSEC were challenged with xanthines at concentrations typically obtained from normal consumption of xanthine-containing beverages, food or medicines; and at higher concentrations below the in vitro toxic limit. The fenestrated morphology of LSEC were examined with scanning electron and structured illumination microscopy. All xanthine challenges had no toxic effects on LSEC ultrastructure as judged by LSEC fenestration morphology, or function as determined by endocytosis studies. All xanthines in high concentrations (150 μg/mL) increased fenestration frequency but at physiologically relevant concentrations, only theobromine (8 μg/mL) showed an effect. LSEC porosity was influenced only by high caffeine doses which also shifted the fenestration distribution towards smaller pores. Moreover, a dose-dependent increase in fenestration number was observed after caffeine treatment. If these compounds induce similar changes in vivo, age-related reduction of LSEC porosity can be reversed by oral treatment with theobromine or with other xanthines using targeted delivery.
Collapse
Affiliation(s)
- Hong Mao
- Vascular Biology Research Group, Department of Medical Biology, Faculty of Health Sciences, University of Tromsø, The Arctic University of Norway, 9037, Tromsø, Norway.
- Optical Nanoscopy Research Group, Department of Physics and Technology, Faculty of Science and Technology, University of Tromsø, The Arctic University of Norway, Tromsø, Norway.
| | - Karolina Szafranska
- Vascular Biology Research Group, Department of Medical Biology, Faculty of Health Sciences, University of Tromsø, The Arctic University of Norway, 9037, Tromsø, Norway.
| | - Larissa Kruse
- Vascular Biology Research Group, Department of Medical Biology, Faculty of Health Sciences, University of Tromsø, The Arctic University of Norway, 9037, Tromsø, Norway
| | - Christopher Holte
- Vascular Biology Research Group, Department of Medical Biology, Faculty of Health Sciences, University of Tromsø, The Arctic University of Norway, 9037, Tromsø, Norway
| | - Deanna L Wolfson
- Optical Nanoscopy Research Group, Department of Physics and Technology, Faculty of Science and Technology, University of Tromsø, The Arctic University of Norway, Tromsø, Norway
| | - Balpreet Singh Ahluwalia
- Optical Nanoscopy Research Group, Department of Physics and Technology, Faculty of Science and Technology, University of Tromsø, The Arctic University of Norway, Tromsø, Norway
| | - Cynthia B Whitchurch
- Microbial Imaging Facility, The ithree Institute, University of Technology Sydney, Ultimo, NSW, Australia
| | - Louise Cole
- Microbial Imaging Facility, The ithree Institute, University of Technology Sydney, Ultimo, NSW, Australia
| | - Glen P Lockwood
- Centre for Education and Research, ANZAC Research Institute, Concord Repatriation General Hospital, Concord, NSW, Australia
- The Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia
| | - Robin Diekmann
- Cell Biology and Biophysics Unit, European Molecular Biology Laboratory (EMBL), Heidelberg, Germany
- LaVision BioTec GmbH, Bielefeld, Germany
| | - David Le Couteur
- Centre for Education and Research, ANZAC Research Institute, Concord Repatriation General Hospital, Concord, NSW, Australia
- The Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia
| | - Victoria C Cogger
- Centre for Education and Research, ANZAC Research Institute, Concord Repatriation General Hospital, Concord, NSW, Australia
- The Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia
| | - Peter A G McCourt
- Vascular Biology Research Group, Department of Medical Biology, Faculty of Health Sciences, University of Tromsø, The Arctic University of Norway, 9037, Tromsø, Norway
- Centre for Education and Research, ANZAC Research Institute, Concord Repatriation General Hospital, Concord, NSW, Australia
- The Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia
| |
Collapse
|
28
|
Kaps L, Limeres MJ, Schneider P, Svensson M, Zeyn Y, Fraude S, Cacicedo ML, Galle PR, Gehring S, Bros M. Liver Cell Type-Specific Targeting by Nanoformulations for Therapeutic Applications. Int J Mol Sci 2023; 24:11869. [PMID: 37511628 PMCID: PMC10380755 DOI: 10.3390/ijms241411869] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Revised: 06/21/2023] [Accepted: 07/21/2023] [Indexed: 07/30/2023] Open
Abstract
Hepatocytes exert pivotal roles in metabolism, protein synthesis and detoxification. Non-parenchymal liver cells (NPCs), largely comprising macrophages, dendritic cells, hepatic stellate cells and liver sinusoidal cells (LSECs), serve to induce immunological tolerance. Therefore, the liver is an important target for therapeutic approaches, in case of both (inflammatory) metabolic diseases and immunological disorders. This review aims to summarize current preclinical nanodrug-based approaches for the treatment of liver disorders. So far, nano-vaccines that aim to induce hepatitis virus-specific immune responses and nanoformulated adjuvants to overcome the default tolerogenic state of liver NPCs for the treatment of chronic hepatitis have been tested. Moreover, liver cancer may be treated using nanodrugs which specifically target and kill tumor cells. Alternatively, nanodrugs may target and reprogram or deplete immunosuppressive cells of the tumor microenvironment, such as tumor-associated macrophages. Here, combination therapies have been demonstrated to yield synergistic effects. In the case of autoimmune hepatitis and other inflammatory liver diseases, anti-inflammatory agents can be encapsulated into nanoparticles to dampen inflammatory processes specifically in the liver. Finally, the tolerance-promoting activity especially of LSECs has been exploited to induce antigen-specific tolerance for the treatment of allergic and autoimmune diseases.
Collapse
Affiliation(s)
- Leonard Kaps
- I. Department of Medicine, University Medical Center Mainz, Langenbeckstrasse 1, 55131 Mainz, Germany
| | - María José Limeres
- Children's Hospital, University Medical Center, Langenbeckstrasse 1, 55131 Mainz, Germany
| | - Paul Schneider
- I. Department of Medicine, University Medical Center Mainz, Langenbeckstrasse 1, 55131 Mainz, Germany
| | - Malin Svensson
- Children's Hospital, University Medical Center, Langenbeckstrasse 1, 55131 Mainz, Germany
| | - Yanira Zeyn
- Department of Dermatology, University Medical Center Mainz, Langenbeckstrasse 1, 55131 Mainz, Germany
| | - Silvia Fraude
- Children's Hospital, University Medical Center, Langenbeckstrasse 1, 55131 Mainz, Germany
| | - Maximiliano L Cacicedo
- Children's Hospital, University Medical Center, Langenbeckstrasse 1, 55131 Mainz, Germany
| | - Peter R Galle
- I. Department of Medicine, University Medical Center Mainz, Langenbeckstrasse 1, 55131 Mainz, Germany
| | - Stephan Gehring
- Children's Hospital, University Medical Center, Langenbeckstrasse 1, 55131 Mainz, Germany
| | - Matthias Bros
- Department of Dermatology, University Medical Center Mainz, Langenbeckstrasse 1, 55131 Mainz, Germany
| |
Collapse
|
29
|
Wang Y, Zhong X, He X, Hu Z, Huang H, Chen J, Chen K, Zhao S, Wei P, Li D. Liver metastasis from colorectal cancer: pathogenetic development, immune landscape of the tumour microenvironment and therapeutic approaches. J Exp Clin Cancer Res 2023; 42:177. [PMID: 37480104 PMCID: PMC10362774 DOI: 10.1186/s13046-023-02729-7] [Citation(s) in RCA: 41] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Accepted: 06/05/2023] [Indexed: 07/23/2023] Open
Abstract
Colorectal cancer liver metastasis (CRLM) is one of the leading causes of death among patients with colorectal cancer (CRC). Although immunotherapy has demonstrated encouraging outcomes in CRC, its benefits are minimal in CRLM. The complex immune landscape of the hepatic tumour microenvironment is essential for the development of a premetastatic niche and for the colonisation and metastasis of CRC cells; thus, an in-depth understanding of these mechanisms can provide effective immunotherapeutic targets for CRLM. This review summarises recent studies on the immune landscape of the tumour microenvironment of CRLM and highlights therapeutic prospects for targeting the suppressive immune microenvironment of CRLM.
Collapse
Affiliation(s)
- Yaxian Wang
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College of Fudan University, Shanghai, China
| | - Xinyang Zhong
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College of Fudan University, Shanghai, China
| | - Xuefeng He
- ZJU-UCLA Joint Center for Medical Education and Research, Cancer Institute, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Zijuan Hu
- Department of Oncology, Shanghai Medical College of Fudan University, Shanghai, China
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, China
- Cancer Institute, Fudan University Shanghai Cancer Center, Shanghai, China
- Institute of Pathology, Fudan University, Shanghai, China
| | - Huixia Huang
- Department of Oncology, Shanghai Medical College of Fudan University, Shanghai, China
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, China
- Cancer Institute, Fudan University Shanghai Cancer Center, Shanghai, China
- Institute of Pathology, Fudan University, Shanghai, China
| | - Jiayu Chen
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College of Fudan University, Shanghai, China
| | - Keji Chen
- Department of Oncology, Shanghai Medical College of Fudan University, Shanghai, China
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, China
- Cancer Institute, Fudan University Shanghai Cancer Center, Shanghai, China
- Institute of Pathology, Fudan University, Shanghai, China
| | - Senlin Zhao
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College of Fudan University, Shanghai, China
| | - Ping Wei
- Department of Oncology, Shanghai Medical College of Fudan University, Shanghai, China.
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, China.
- Cancer Institute, Fudan University Shanghai Cancer Center, Shanghai, China.
- Institute of Pathology, Fudan University, Shanghai, China.
| | - Dawei Li
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, Shanghai, China.
- Department of Oncology, Shanghai Medical College of Fudan University, Shanghai, China.
| |
Collapse
|
30
|
Han DW, Xu K, Jin ZL, Xu YN, Li YH, Wang L, Cao Q, Kim KP, Ryu D, Hong K, Kim NH. Customized liver organoids as an advanced in vitro modeling and drug discovery platform for non-alcoholic fatty liver diseases. Int J Biol Sci 2023; 19:3595-3613. [PMID: 37497008 PMCID: PMC10367556 DOI: 10.7150/ijbs.85145] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Accepted: 06/12/2023] [Indexed: 07/28/2023] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) and its progressive form non-alcoholic steatohepatitis (NASH) have presented a major and common health concern worldwide due to their increasing prevalence and progressive development of severe pathological conditions such as cirrhosis and liver cancer. Although a large number of drug candidates for the treatment of NASH have entered clinical trial testing, all have not been released to market due to their limited efficacy, and there remains no approved treatment for NASH available to this day. Recently, organoid technology that produces 3D multicellular aggregates with a liver tissue-like cytoarchitecture and improved functionality has been suggested as a novel platform for modeling the human-specific complex pathophysiology of NAFLD and NASH. In this review, we describe the cellular crosstalk between each cellular compartment in the liver during the pathogenesis of NAFLD and NASH. We also summarize the current state of liver organoid technology, describing the cellular diversity that could be recapitulated in liver organoids and proposing a future direction for liver organoid technology as an in vitro platform for disease modeling and drug discovery for NAFLD and NASH.
Collapse
Affiliation(s)
- Dong Wook Han
- Guangdong Provincial Key Laboratory of Large Animal Models for Biomedicine, School of Biotechnology and Health Sciences, Wuyi University, Jiangmen, China
- International Healthcare Innovation Institute (Jiangmen), Jianghai, Jiangmen, Guangdong Province, China
- Research and Development, Qingdao Haier Biotech Co. Ltd, Qingdao, China
- Guangdong ORGANOID Biotechnology Co. Ltd, Jiangmen, China
| | - KangHe Xu
- Department of Surgery, College of Medicine, Chungbuk National University, Cheongju, Republic of Korea
| | - Zhe-Long Jin
- Guangdong Provincial Key Laboratory of Large Animal Models for Biomedicine, School of Biotechnology and Health Sciences, Wuyi University, Jiangmen, China
- International Healthcare Innovation Institute (Jiangmen), Jianghai, Jiangmen, Guangdong Province, China
- Guangdong ORGANOID Biotechnology Co. Ltd, Jiangmen, China
| | - Yong-Nan Xu
- Guangdong Provincial Key Laboratory of Large Animal Models for Biomedicine, School of Biotechnology and Health Sciences, Wuyi University, Jiangmen, China
- International Healthcare Innovation Institute (Jiangmen), Jianghai, Jiangmen, Guangdong Province, China
| | - Ying-Hua Li
- Guangdong Provincial Key Laboratory of Large Animal Models for Biomedicine, School of Biotechnology and Health Sciences, Wuyi University, Jiangmen, China
- International Healthcare Innovation Institute (Jiangmen), Jianghai, Jiangmen, Guangdong Province, China
| | - Lin Wang
- Research and Development, Qingdao Haier Biotech Co. Ltd, Qingdao, China
| | - Qilong Cao
- Research and Development, Qingdao Haier Biotech Co. Ltd, Qingdao, China
| | - Kee-Pyo Kim
- Department of Life Sciences, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - DongHee Ryu
- Department of Surgery, College of Medicine, Chungbuk National University, Cheongju, Republic of Korea
| | - Kwonho Hong
- Department of Stem Cell and Regenerative Biotechnology, The institute of advanced regenerative science, Konkuk University, Seoul, Republic of Korea
| | - Nam-Hyung Kim
- Guangdong Provincial Key Laboratory of Large Animal Models for Biomedicine, School of Biotechnology and Health Sciences, Wuyi University, Jiangmen, China
- International Healthcare Innovation Institute (Jiangmen), Jianghai, Jiangmen, Guangdong Province, China
- Research and Development, Qingdao Haier Biotech Co. Ltd, Qingdao, China
- Guangdong ORGANOID Biotechnology Co. Ltd, Jiangmen, China
| |
Collapse
|
31
|
Kwak G, Lee D, Suk JS. Advanced approaches to overcome biological barriers in respiratory and systemic routes of administration for enhanced nucleic acid delivery to the lung. Expert Opin Drug Deliv 2023; 20:1531-1552. [PMID: 37946533 PMCID: PMC10872418 DOI: 10.1080/17425247.2023.2282535] [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: 07/11/2023] [Accepted: 11/08/2023] [Indexed: 11/12/2023]
Abstract
INTRODUCTION Numerous delivery strategies, primarily novel nucleic acid delivery carriers, have been developed and explored to enable therapeutically relevant lung gene therapy. However, its clinical translation is yet to be achieved despite over 30 years of efforts, which is attributed to the inability to overcome a series of biological barriers that hamper efficient nucleic acid transfer to target cells in the lung. AREAS COVERED This review is initiated with the fundamentals of nucleic acid therapy and a brief overview of previous and ongoing efforts on clinical translation of lung gene therapy. We then walk through the nature of biological barriers encountered by nucleic acid carriers administered via respiratory and/or systemic routes. Finally, we introduce advanced strategies developed to overcome those barriers to achieve therapeutically relevant nucleic acid delivery efficiency in the lung. EXPERT OPINION We are now stepping close to the clinical translation of lung gene therapy, thanks to the discovery of novel delivery strategies that overcome biological barriers via comprehensive preclinical studies. However, preclinical findings should be cautiously interpreted and validated to ultimately realize meaningful therapeutic outcomes with newly developed delivery strategies in humans. In particular, individual strategies should be selected, tailored, and implemented in a manner directly relevant to specific therapeutic applications and goals.
Collapse
Affiliation(s)
- Gijung Kwak
- Department of Neurosurgery and Medicine Institute for Neuroscience Discovery (UM-MIND), University of Maryland School of Medicine, Baltimore, MD, USA
- Center for Nanomedicine, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Ophthalmology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Daiheon Lee
- Department of Neurosurgery and Medicine Institute for Neuroscience Discovery (UM-MIND), University of Maryland School of Medicine, Baltimore, MD, USA
- Center for Nanomedicine, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Ophthalmology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Jung Soo Suk
- Department of Neurosurgery and Medicine Institute for Neuroscience Discovery (UM-MIND), University of Maryland School of Medicine, Baltimore, MD, USA
- Center for Nanomedicine, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Ophthalmology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Chemical & Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, USA
| |
Collapse
|
32
|
Li X, Souilhol C, Canham L, Jia X, Diagbouga M, Ayllon BT, Serbanovic-Canic J, Evans PC. DLL4 promotes partial endothelial-to-mesenchymal transition at atherosclerosis-prone regions of arteries. Vascul Pharmacol 2023; 150:107178. [PMID: 37137436 DOI: 10.1016/j.vph.2023.107178] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 04/25/2023] [Accepted: 04/26/2023] [Indexed: 05/05/2023]
Abstract
Flowing blood regulates vascular development, homeostasis and disease by generating wall shear stress which has major effects on endothelial cell (EC) physiology. Low oscillatory shear stress (LOSS) induces a form of cell plasticity called endothelial-to-mesenchymal transition (EndMT). This process has divergent effects; in embryos LOSS-induced EndMT drives the development of atrioventricular valves, whereas in adult arteries it is associated with inflammation and atherosclerosis. The Notch ligand DLL4 is essential for LOSS-dependent valve development; here we investigated whether DLL4 is required for responses to LOSS in adult arteries. Analysis of cultured human coronary artery EC revealed that DLL4 regulates the transcriptome to induce markers of EndMT and inflammation under LOSS conditions. Consistently, genetic deletion of Dll4 from murine EC reduced SNAIL (EndMT marker) and VCAM-1 (inflammation marker) at a LOSS region of the murine aorta. We hypothesized that endothelial Dll4 is pro-atherogenic but this analysis was confounded because endothelial Dll4 negatively regulated plasma cholesterol levels in hyperlipidemic mice. We conclude that endothelial DLL4 is required for LOSS-induction of EndMT and inflammation regulators at atheroprone regions of arteries, and is also a regulator of plasma cholesterol.
Collapse
Affiliation(s)
- Xiuying Li
- Department of Pharmacy, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, PR China; School of Pharmacy, Southwest Medical University, LuZhou, Sichuan 646000, PR China; Department of Infection, Immunity and Cardiovascular Disease, INSIGNEO Institute, Bateson Centre, University of Sheffield, UK
| | - Celine Souilhol
- Department of Infection, Immunity and Cardiovascular Disease, INSIGNEO Institute, Bateson Centre, University of Sheffield, UK.
| | - Lindsay Canham
- Department of Infection, Immunity and Cardiovascular Disease, INSIGNEO Institute, Bateson Centre, University of Sheffield, UK
| | - Xueqi Jia
- Department of Infection, Immunity and Cardiovascular Disease, INSIGNEO Institute, Bateson Centre, University of Sheffield, UK
| | - Mannekomba Diagbouga
- Department of Infection, Immunity and Cardiovascular Disease, INSIGNEO Institute, Bateson Centre, University of Sheffield, UK
| | - Blanca Tardajos Ayllon
- Department of Infection, Immunity and Cardiovascular Disease, INSIGNEO Institute, Bateson Centre, University of Sheffield, UK
| | - Jovana Serbanovic-Canic
- Department of Infection, Immunity and Cardiovascular Disease, INSIGNEO Institute, Bateson Centre, University of Sheffield, UK
| | - Paul C Evans
- Department of Infection, Immunity and Cardiovascular Disease, INSIGNEO Institute, Bateson Centre, University of Sheffield, UK; Centre for Biochemical Pharmacology, William Harvey Research Institute, Faculty of Medicine and Dentistry, Barts and The London, Queen Mary University of London Charterhouse Square, London EC1M 6BQ, UK.
| |
Collapse
|
33
|
Gupta D, Wiklander OP, Wood MJ, El-Andaloussi S. Biodistribution of therapeutic extracellular vesicles. EXTRACELLULAR VESICLES AND CIRCULATING NUCLEIC ACIDS 2023; 4:170-190. [PMID: 39697988 PMCID: PMC11648525 DOI: 10.20517/evcna.2023.12] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 03/27/2023] [Accepted: 04/03/2023] [Indexed: 12/20/2024]
Abstract
The field of extracellular vesicles (EVs) has seen a tremendous paradigm shift in the past two decades, from being regarded as cellular waste bags to being considered essential mediators in intercellular communication. Their unique ability to transfer macromolecules across cells and biological barriers has made them a rising star in drug delivery. Mounting evidence suggests that EVs can be explored as efficient drug delivery vehicles for a range of therapeutic macromolecules. In contrast to many synthetic delivery systems, these vesicles appear exceptionally well tolerated in vivo. This tremendous development in the therapeutic application of EVs has been made through technological advancement in labelling and understanding the in vivo biodistribution of EVs. Here in this review, we have summarised the recent findings in EV in vivo pharmacokinetics and discussed various biological barriers that need to be surpassed to achieve tissue-specific delivery.
Collapse
Affiliation(s)
- Dhanu Gupta
- Department of Paediatrics. University of Oxford, Oxford OX3 7TY, UK
- Biomolecular Medicine, Division of Biomolecular and Cellular Medicine, Department of Laboratory Medicine, Karolinska Institutet, Huddinge 14151, Sweden
| | - Oscar P.B Wiklander
- Biomolecular Medicine, Division of Biomolecular and Cellular Medicine, Department of Laboratory Medicine, Karolinska Institutet, Huddinge 14151, Sweden
| | - Matthew J.A Wood
- Department of Paediatrics. University of Oxford, Oxford OX3 7TY, UK
| | - Samir El-Andaloussi
- Biomolecular Medicine, Division of Biomolecular and Cellular Medicine, Department of Laboratory Medicine, Karolinska Institutet, Huddinge 14151, Sweden
| |
Collapse
|
34
|
The Association of Integrins β3, β4, and αVβ5 on Exosomes, CTCs and Tumor Cells with Localization of Distant Metastasis in Breast Cancer Patients. Int J Mol Sci 2023; 24:ijms24032929. [PMID: 36769251 PMCID: PMC9918050 DOI: 10.3390/ijms24032929] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Revised: 01/11/2023] [Accepted: 01/30/2023] [Indexed: 02/05/2023] Open
Abstract
Integrins are cell adhesion receptors, which play a role in breast cancer invasion, angiogenesis, and metastasis. Moreover, it has been shown that exosomal integrins provide organotropic metastasis in a mouse model. In our study, we aimed to investigate the expression of integrins β3, β4, and αVβ5 on exosomes and tumor cells (circulating tumor cells and primary tumor) and their association with the localization of distant metastasis. We confirmed the association of exosomal integrin β4 with lung metastasis in breast cancer patients. However, we were unable to evaluate the role of integrin β3 in brain metastasis due to the rarity of this localization. We established no association of exosomal integrin αVβ5 with liver metastasis in our cohort of breast cancer patients. The further evaluation of β3, β4, and αVβ5 integrin expression on CTCs revealed an association of integrin β4 and αVβ5 with liver, but not the lung metastases. Integrin β4 in the primary tumor was associated with liver metastasis. Furthermore, an in-depth analysis of phenotypic characteristics of β4+ tumor cells revealed a significantly increased proportion of E-cadherin+ and CD44+CD24- cells in patients with liver metastases compared to patients with lung or no distant metastases.
Collapse
|
35
|
Abstract
Gene therapy is poised to revolutionize modern medicine, with seemingly unlimited potential for treating and curing genetic disorders. For otherwise incurable indications, including most inherited metabolic liver disorders, gene therapy provides a realistic therapeutic option. In this Review, we discuss gene supplementation and gene editing involving the use of recombinant adeno-associated virus (rAAV) vectors for the treatment of inherited liver diseases, including updates on several ongoing clinical trials that are producing promising results. Clinical testing has been essential in highlighting many key translational challenges associated with this transformative therapy. In particular, the interaction of a patient's immune system with the vector raises issues of safety and the duration of treatment efficacy. Furthermore, several serious adverse events after the administration of high doses of rAAVs suggest greater involvement of innate immune responses and pre-existing hepatic conditions than initially anticipated. Finally, permanent modification of the host genome associated with rAAV genome integration and gene editing raises concerns about the risk of oncogenicity that require careful evaluation. We summarize the main progress, challenges and pathways forward for gene therapy for liver diseases.
Collapse
|
36
|
Liu GW, Guzman EB, Menon N, Langer RS. Lipid Nanoparticles for Nucleic Acid Delivery to Endothelial Cells. Pharm Res 2023; 40:3-25. [PMID: 36735106 PMCID: PMC9897626 DOI: 10.1007/s11095-023-03471-7] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Accepted: 01/12/2023] [Indexed: 02/04/2023]
Abstract
Endothelial cells play critical roles in circulatory homeostasis and are also the gateway to the major organs of the body. Dysfunction, injury, and gene expression profiles of these cells can cause, or are caused by, prevalent chronic diseases such as diabetes, cardiovascular disease, and cancer. Modulation of gene expression within endothelial cells could therefore be therapeutically strategic in treating longstanding disease challenges. Lipid nanoparticles (LNP) have emerged as potent, scalable, and tunable carrier systems for delivering nucleic acids, making them attractive vehicles for gene delivery to endothelial cells. Here, we discuss the functions of endothelial cells and highlight some receptors that are upregulated during health and disease. Examples and applications of DNA, mRNA, circRNA, saRNA, siRNA, shRNA, miRNA, and ASO delivery to endothelial cells and their targets are reviewed, as well as LNP composition and morphology, formulation strategies, target proteins, and biomechanical factors that modulate endothelial cell targeting. Finally, we discuss FDA-approved LNPs as well as LNPs that have been tested in clinical trials and their challenges, and provide some perspectives as to how to surmount those challenges.
Collapse
Affiliation(s)
- Gary W Liu
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Edward B Guzman
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
- School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, 02138, USA
| | - Nandita Menon
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
- Strand Therapeutics, MA, 02215, Boston, USA
| | - Robert S Langer
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA.
| |
Collapse
|
37
|
Zhao X, Xue X, Cui Z, Kwame Amevor F, Wan Y, Fu K, Wang C, Peng C, Li Y. microRNAs-based diagnostic and therapeutic applications in liver fibrosis. WILEY INTERDISCIPLINARY REVIEWS. RNA 2022:e1773. [PMID: 36585388 DOI: 10.1002/wrna.1773] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Revised: 12/05/2022] [Accepted: 12/12/2022] [Indexed: 01/01/2023]
Abstract
Liver fibrosis is a process of over-extracellular matrix (ECM) aggregation and angiogenesis, which develops into cirrhosis and hepatocellular carcinoma (HCC). With the increasing pressure of liver fibrosis, new therapeutics to cure this disease requires much attention. Exosome-cargoed microRNAs (miRNAs) are emerging approaches in the precision of the liver fibrotic paradigm. In this review, we outlined the different types of hepatic cells derived miRNAs that drive intra-/extra-cellular interactive communication in liver fibrosis with different physiological and pathological processes. Specifically, we highlighted the possible mechanism of liver fibrosis pathogenesis associated with immune response and angiogenesis. In addition, potential clinical biomarkers and different stem cell transplant-derived miRNAs-based therapeutic strategies in liver fibrosis were summarized in this review. miRNAs-based approaches might help researchers devise new candidates for the cell-free treatment of liver fibrosis. This article is categorized under: RNA in Disease and Development > RNA in Disease.
Collapse
Affiliation(s)
- Xingtao Zhao
- State Key Laboratory of Southwestern Chinese Medicine Resources, Ministry of Education, Chengdu, China.,School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xinyan Xue
- State Key Laboratory of Southwestern Chinese Medicine Resources, Ministry of Education, Chengdu, China.,School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Zhifu Cui
- College Science and Technology, Southwest University, Chongqing, China
| | - Felix Kwame Amevor
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, China
| | - Yan Wan
- State Key Laboratory of Southwestern Chinese Medicine Resources, Ministry of Education, Chengdu, China.,School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Ke Fu
- State Key Laboratory of Southwestern Chinese Medicine Resources, Ministry of Education, Chengdu, China.,School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Cheng Wang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Ministry of Education, Chengdu, China.,School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Cheng Peng
- State Key Laboratory of Southwestern Chinese Medicine Resources, Ministry of Education, Chengdu, China.,School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yunxia Li
- State Key Laboratory of Southwestern Chinese Medicine Resources, Ministry of Education, Chengdu, China.,School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
38
|
Chen S, Zhu J, Xue J, Wang X, Jing P, Zhou L, Cui Y, Wang T, Gong X, Lü S, Long M. Numerical simulation of flow characteristics in a permeable liver sinusoid with leukocytes. Biophys J 2022; 121:4666-4678. [PMID: 36271623 PMCID: PMC9748252 DOI: 10.1016/j.bpj.2022.10.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Revised: 08/31/2022] [Accepted: 10/17/2022] [Indexed: 02/07/2023] Open
Abstract
Double-layered channels of sinusoid lumen and Disse space separated by fenestrated liver sinusoidal endothelial cells (LSECs) endow the unique mechanical environment of the liver sinusoid network, which further guarantees its biological function. It is also known that this mechanical environment changes dramatically under liver fibrosis and cirrhosis, including the reduced plasma penetration and metabolite exchange between the two flow channels and the reduced Disse space deformability. The squeezing of leukocytes through narrow sinusoid lumen also affects the mechanical environment of liver sinusoid. To date, the detailed flow-field profile of liver sinusoid is still far from clear due to experimental limitations. It also remains elusive whether and how the varied physical properties of the pathological liver sinusoid regulate the fluid flow characteristics. Here a numerical model based on the immersed boundary method was established, and the effects of Disse space and leukocyte elasticities, endothelium permeability, and sinusoidal stenosis degree on fluid flow as well as leukocyte trafficking were specified upon a mimic liver sinusoid structure. Results showed that endothelium permeability dominantly controlled the plasma penetration velocity across the endothelium, whereas leukocyte squeezing promoted local penetration and significantly regulated wall shear stress on hepatocytes, which was strongly related to the Disse space and leukocyte deformability. Permeability and elasticity cooperatively regulated the process of leukocytes trafficking through the liver sinusoid, especially for stiffer leukocytes. This study will offer new insights into deeper understanding of the elaborate mechanical features of liver sinusoid and corresponding biological function.
Collapse
Affiliation(s)
- Shenbao Chen
- Center of Biomechanics and Bioengineering, Key Laboratory of Microgravity (National Microgravity Laboratory), Beijing Key Laboratory of Engineered Construction and Mechanobiology, and CAS Center for Excellence in Complex System Mechanics, Institute of Mechanics, Chinese Academy of Sciences, Beijing, China; School of Engineering Science, University of Chinese Academy of Sciences, Beijing, China
| | - Jingchen Zhu
- Center of Biomechanics and Bioengineering, Key Laboratory of Microgravity (National Microgravity Laboratory), Beijing Key Laboratory of Engineered Construction and Mechanobiology, and CAS Center for Excellence in Complex System Mechanics, Institute of Mechanics, Chinese Academy of Sciences, Beijing, China; School of Engineering Science, University of Chinese Academy of Sciences, Beijing, China
| | - Jian Xue
- School of Engineering Science, University of Chinese Academy of Sciences, Beijing, China; State Key Laboratory of Nonlinear Mechanics (LNM), Institute of Mechanics, Chinese Academy of Sciences, Beijing, China
| | - Xiaolong Wang
- Key Laboratory of Hydrodynamics (Ministry of Education), Department of Engineering Mechanics, School of Naval Architecture, Ocean and Civil Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Peng Jing
- Key Laboratory of Hydrodynamics (Ministry of Education), Department of Engineering Mechanics, School of Naval Architecture, Ocean and Civil Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Lüwen Zhou
- Center of Biomechanics and Bioengineering, Key Laboratory of Microgravity (National Microgravity Laboratory), Beijing Key Laboratory of Engineered Construction and Mechanobiology, and CAS Center for Excellence in Complex System Mechanics, Institute of Mechanics, Chinese Academy of Sciences, Beijing, China; School of Engineering Science, University of Chinese Academy of Sciences, Beijing, China
| | - Yuhong Cui
- Department of Mechanics, Tianjin University, Tianjin, China
| | - Tianhao Wang
- Department of Mechanics, Tianjin University, Tianjin, China
| | - Xiaobo Gong
- Key Laboratory of Hydrodynamics (Ministry of Education), Department of Engineering Mechanics, School of Naval Architecture, Ocean and Civil Engineering, Shanghai Jiao Tong University, Shanghai, China.
| | - Shouqin Lü
- Center of Biomechanics and Bioengineering, Key Laboratory of Microgravity (National Microgravity Laboratory), Beijing Key Laboratory of Engineered Construction and Mechanobiology, and CAS Center for Excellence in Complex System Mechanics, Institute of Mechanics, Chinese Academy of Sciences, Beijing, China; School of Engineering Science, University of Chinese Academy of Sciences, Beijing, China.
| | - Mian Long
- Center of Biomechanics and Bioengineering, Key Laboratory of Microgravity (National Microgravity Laboratory), Beijing Key Laboratory of Engineered Construction and Mechanobiology, and CAS Center for Excellence in Complex System Mechanics, Institute of Mechanics, Chinese Academy of Sciences, Beijing, China; School of Engineering Science, University of Chinese Academy of Sciences, Beijing, China.
| |
Collapse
|
39
|
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: 6] [Impact Index Per Article: 2.0] [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
|
40
|
Huu Hoang T, Sato-Matsubara M, Yuasa H, Matsubara T, Thuy LTT, Ikenaga H, Phuong DM, Hanh NV, Hieu VN, Hoang DV, Hai H, Okina Y, Enomoto M, Tamori A, Daikoku A, Urushima H, Ikeda K, Dat NQ, Yasui Y, Shinkawa H, Kubo S, Yamagishi R, Ohtani N, Yoshizato K, Gracia-Sancho J, Kawada N. Cancer cells produce liver metastasis via gap formation in sinusoidal endothelial cells through proinflammatory paracrine mechanisms. SCIENCE ADVANCES 2022; 8:eabo5525. [PMID: 36170363 PMCID: PMC9519040 DOI: 10.1126/sciadv.abo5525] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/12/2022] [Accepted: 08/04/2022] [Indexed: 06/10/2023]
Abstract
Intracellular gap (iGap) formation in liver sinusoidal endothelial cells (LSECs) is caused by the destruction of fenestrae and appears under pathological conditions; nevertheless, their role in metastasis of cancer cells to the liver remained unexplored. We elucidated that hepatotoxin-damaged and fibrotic livers gave rise to LSECs-iGap formation, which was positively correlated with increased numbers of metastatic liver foci after intrasplenic injection of Hepa1-6 cells. Hepa1-6 cells induced interleukin-23-dependent tumor necrosis factor-α (TNF-α) secretion by LSECs and triggered LSECs-iGap formation, toward which their processes protruded to transmigrate into the liver parenchyma. TNF-α triggered depolymerization of F-actin and induced matrix metalloproteinase 9 (MMP9), intracellular adhesion molecule 1, and CXCL expression in LSECs. Blocking MMP9 activity by doxycycline or an MMP2/9 inhibitor eliminated LSECs-iGap formation and attenuated liver metastasis of Hepa1-6 cells. Overall, this study revealed that cancer cells induced LSEC-iGap formation via proinflammatory paracrine mechanisms and proposed MMP9 as a favorable target for blocking cancer cell metastasis to the liver.
Collapse
Affiliation(s)
- Truong Huu Hoang
- Department of Hepatology, Graduate School of Medicine, Osaka Metropolitan University, Osaka, Japan
- Department of Pain Medicine and Palliative Care, Cancer Institute, 108 Military Central Hospital, Hanoi, Vietnam
| | - Misako Sato-Matsubara
- Department of Hepatology, Graduate School of Medicine, Osaka Metropolitan University, Osaka, Japan
- Endowed Laboratory of Synthetic Biology, Graduate School of Medicine, Osaka Metropolitan University, Osaka, Japan
| | - Hideto Yuasa
- Department of Anatomy and Regenerative Biology, Graduate School of Medicine, Osaka Metropolitan University, Osaka, Japan
| | - Tsutomu Matsubara
- Department of Anatomy and Regenerative Biology, Graduate School of Medicine, Osaka Metropolitan University, Osaka, Japan
| | - Le Thi Thanh Thuy
- Department of Hepatology, Graduate School of Medicine, Osaka Metropolitan University, Osaka, Japan
| | - Hiroko Ikenaga
- Department of Hepatology, Graduate School of Medicine, Osaka Metropolitan University, Osaka, Japan
| | - Dong Minh Phuong
- Department of Hepatology, Graduate School of Medicine, Osaka Metropolitan University, Osaka, Japan
| | - Ngo Vinh Hanh
- Department of Hepatology, Graduate School of Medicine, Osaka Metropolitan University, Osaka, Japan
| | - Vu Ngoc Hieu
- Department of Hepatology, Graduate School of Medicine, Osaka Metropolitan University, Osaka, Japan
| | - Dinh Viet Hoang
- Department of Anesthesiology, Cho Ray Hospital, Ho Chi Minh City, Vietnam
| | - Hoang Hai
- Department of Hepatology, Graduate School of Medicine, Osaka Metropolitan University, Osaka, Japan
| | - Yoshinori Okina
- Department of Hepatology, Graduate School of Medicine, Osaka Metropolitan University, Osaka, Japan
| | - Masaru Enomoto
- Department of Hepatology, Graduate School of Medicine, Osaka Metropolitan University, Osaka, Japan
| | - Akihiro Tamori
- Department of Hepatology, Graduate School of Medicine, Osaka Metropolitan University, Osaka, Japan
| | - Atsuko Daikoku
- Department of Anatomy and Regenerative Biology, Graduate School of Medicine, Osaka Metropolitan University, Osaka, Japan
| | - Hayato Urushima
- Department of Anatomy and Regenerative Biology, Graduate School of Medicine, Osaka Metropolitan University, Osaka, Japan
| | - Kazuo Ikeda
- Department of Anatomy and Regenerative Biology, Graduate School of Medicine, Osaka Metropolitan University, Osaka, Japan
| | - Ninh Quoc Dat
- Department of Pediatrics, Hanoi Medical University, Hanoi, Vietnam
| | - Yutaka Yasui
- Department of Gastroenterology and Hepatology, Musashino Red Cross Hospital, Tokyo, Japan
| | - Hiroji Shinkawa
- Department of Hepato-Biliary-Pancreatic Surgery, Graduate School of Medicine, Osaka Metropolitan University, Osaka, Japan
| | - Shoji Kubo
- Department of Hepato-Biliary-Pancreatic Surgery, Graduate School of Medicine, Osaka Metropolitan University, Osaka, Japan
| | - Ryota Yamagishi
- Department of Pathophysiology, Graduate School of Medicine, Osaka Metropolitan University, Osaka, Japan
| | - Naoko Ohtani
- Department of Pathophysiology, Graduate School of Medicine, Osaka Metropolitan University, Osaka, Japan
| | - Katsutoshi Yoshizato
- Endowed Laboratory of Synthetic Biology, Graduate School of Medicine, Osaka Metropolitan University, Osaka, Japan
- BioIntegrence Co. Ltd., Osaka, Japan
| | - Jordi Gracia-Sancho
- Liver Vascular Biology Research Group, IDIBAPS Biomedical Research Institute, CIBEREHD, Barcelona, Spain
| | - Norifumi Kawada
- Department of Hepatology, Graduate School of Medicine, Osaka Metropolitan University, Osaka, Japan
| |
Collapse
|
41
|
Li R, Bhandari S, Martinez-Zubiaurre I, Bruun JA, Urbarova I, Smedsrød B, Simón-Santamaría J, Sørensen KK. Changes in the proteome and secretome of rat liver sinusoidal endothelial cells during early primary culture and effects of dexamethasone. PLoS One 2022; 17:e0273843. [PMID: 36054185 PMCID: PMC9439253 DOI: 10.1371/journal.pone.0273843] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2021] [Accepted: 08/16/2022] [Indexed: 11/28/2022] Open
Abstract
Introduction Liver sinusoidal endothelial cells (LSECs) are specialized fenestrated scavenger endothelial cells involved in the elimination of modified plasma proteins and tissue turnover waste macromolecules from blood. LSECs also participate in liver immune responses. A challenge when studying LSEC biology is the rapid loss of the in vivo phenotype in culture. In this study, we have examined biological processes and pathways affected during early-stage primary culture of rat LSECs and checked for cell responses to the pro-inflammatory cytokine interleukin (IL)-1β and the anti-inflammatory drug dexamethasone. Methods LSECs from male Sprague Dawley rats were cultured on type I collagen in 5% oxygen atmosphere in DMEM with serum-free supplements for 2 and 24 h. Quantitative proteomics using tandem mass tag technology was used to examine proteins in cells and supernatants. Validation was done with qPCR, ELISA, multiplex immunoassay, and caspase 3/7 assay. Cell ultrastructure was examined by scanning electron microscopy, and scavenger function by quantitative endocytosis assays. Results LSECs cultured for 24 h showed a characteristic pro-inflammatory phenotype both in the presence and absence of IL-1β, with upregulation of cellular responses to cytokines and interferon-γ, cell-cell adhesion, and glycolysis, increased expression of fatty acid binding proteins (FABP4, FABP5), and downregulation of several membrane receptors (STAB1, STAB2, LYVE1, CLEC4G) and proteins in pyruvate metabolism, citric acid cycle, fatty acid elongation, amino acid metabolism, and oxidation-reduction processes. Dexamethasone inhibited apoptosis and improved LSEC viability in culture, repressed inflammatory and immune regulatory pathways and secretion of IL-1β and IL-6, and further upregulated FABP4 and FABP5 compared to time-matched controls. The LSEC porosity and endocytic activity were reduced at 24 h both with and without dexamethasone but the dexamethasone-treated cells showed a less stressed phenotype. Conclusion Rat LSECs become activated towards a pro-inflammatory phenotype during early culture. Dexamethasone represses LSEC activation, inhibits apoptosis, and improves cell viability.
Collapse
Affiliation(s)
- Ruomei Li
- Department of Medical Biology, UiT–The Arctic University of Norway, Tromsø, Norway
| | - Sabin Bhandari
- Department of Medical Biology, UiT–The Arctic University of Norway, Tromsø, Norway
| | | | - Jack-Ansgar Bruun
- Department of Medical Biology, UiT–The Arctic University of Norway, Tromsø, Norway
| | - Ilona Urbarova
- Department of Community Medicine, UiT–The Arctic University of Norway, Tromsø, Norway
| | - Bård Smedsrød
- Department of Medical Biology, UiT–The Arctic University of Norway, Tromsø, Norway
| | | | | |
Collapse
|
42
|
Tuning of Liver Sieve: The Interplay between Actin and Myosin Regulatory Light Chain Regulates Fenestration Size and Number in Murine Liver Sinusoidal Endothelial Cells. Int J Mol Sci 2022; 23:ijms23179850. [PMID: 36077249 PMCID: PMC9456121 DOI: 10.3390/ijms23179850] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 08/23/2022] [Accepted: 08/25/2022] [Indexed: 12/02/2022] Open
Abstract
Liver sinusoidal endothelial cells (LSECs) facilitate the efficient transport of macromolecules and solutes between the blood and hepatocytes. The efficiency of this transport is realized via transcellular nanopores, called fenestrations. The mean fenestration size is 140 ± 20 nm, with the range from 50 nm to 350 nm being mostly below the limits of diffraction of visible light. The cellular mechanisms controlling fenestrations are still poorly understood. In this study, we tested a hypothesis that both Rho kinase (ROCK) and myosin light chain (MLC) kinase (MLCK)-dependent phosphorylation of MLC regulates fenestrations. We verified the hypothesis using a combination of several molecular inhibitors and by applying two high-resolution microscopy modalities: structured illumination microscopy (SIM) and scanning electron microscopy (SEM). We demonstrated precise, dose-dependent, and reversible regulation of the mean fenestration diameter within a wide range from 120 nm to 220 nm and the fine-tuning of the porosity in a range from ~0% up to 12% using the ROCK pathway. Moreover, our findings indicate that MLCK is involved in the formation of new fenestrations—after inhibiting MLCK, closed fenestrations cannot be reopened with other agents. We, therefore, conclude that the Rho-ROCK pathway is responsible for the control of the fenestration diameter, while the inhibition of MLCK prevents the formation of new fenestrations.
Collapse
|
43
|
Li N, Zhang X, Zhou J, Li W, Shu X, Wu Y, Long M. Multiscale biomechanics and mechanotransduction from liver fibrosis to cancer. Adv Drug Deliv Rev 2022; 188:114448. [PMID: 35820602 DOI: 10.1016/j.addr.2022.114448] [Citation(s) in RCA: 46] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Revised: 05/08/2022] [Accepted: 07/06/2022] [Indexed: 02/06/2023]
Abstract
A growing body of multiscale biomechanical studies has been proposed to highlight the mechanical cues in the development of hepatic fibrosis and cancer. At the cellular level, changes in mechanical microenvironment induce phenotypic and functional alterations of hepatic cells, initiating a positive feedback loop that promotes liver fibrogenesis and hepatocarcinogenesis. Tumor mechanical microenvironment of hepatocellular carcinoma facilitates tumor cell growth and metastasis, and hinders the drug delivery and immunotherapy. At the molecular level, mechanical forces are sensed and transmitted into hepatic cells via allosteric activation of mechanoreceptors on the cell membrane, leading to the activation of various mechanotransduction pathways including integrin and YAP signaling and then regulating cell function. Thus, the application of mechanomedicine concept in the treatment of liver diseases is promising for rational design and cell-specific delivery of therapeutic drugs. This review mainly discusses the correlation between biomechanical cues and liver diseases from the viewpoint of mechanobiology.
Collapse
Affiliation(s)
- Ning Li
- Center for Biomechanics and Bioengineering, Key Laboratory of Microgravity (National Microgravity Laboratory), and Beijing Key Laboratory of Engineered Construction and Mechanobiology, Institute of Mechanics, Chinese Academy of Sciences, Beijing 100190, China; School of Engineering Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xiaoyu Zhang
- Center for Biomechanics and Bioengineering, Key Laboratory of Microgravity (National Microgravity Laboratory), and Beijing Key Laboratory of Engineered Construction and Mechanobiology, Institute of Mechanics, Chinese Academy of Sciences, Beijing 100190, China; School of Engineering Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jin Zhou
- Center for Biomechanics and Bioengineering, Key Laboratory of Microgravity (National Microgravity Laboratory), and Beijing Key Laboratory of Engineered Construction and Mechanobiology, Institute of Mechanics, Chinese Academy of Sciences, Beijing 100190, China
| | - Wang Li
- Center for Biomechanics and Bioengineering, Key Laboratory of Microgravity (National Microgravity Laboratory), and Beijing Key Laboratory of Engineered Construction and Mechanobiology, Institute of Mechanics, Chinese Academy of Sciences, Beijing 100190, China; School of Engineering Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xinyu Shu
- Center for Biomechanics and Bioengineering, Key Laboratory of Microgravity (National Microgravity Laboratory), and Beijing Key Laboratory of Engineered Construction and Mechanobiology, Institute of Mechanics, Chinese Academy of Sciences, Beijing 100190, China; School of Engineering Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yi Wu
- Center for Biomechanics and Bioengineering, Key Laboratory of Microgravity (National Microgravity Laboratory), and Beijing Key Laboratory of Engineered Construction and Mechanobiology, Institute of Mechanics, Chinese Academy of Sciences, Beijing 100190, China; School of Engineering Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Mian Long
- Center for Biomechanics and Bioengineering, Key Laboratory of Microgravity (National Microgravity Laboratory), and Beijing Key Laboratory of Engineered Construction and Mechanobiology, Institute of Mechanics, Chinese Academy of Sciences, Beijing 100190, China; School of Engineering Sciences, University of Chinese Academy of Sciences, Beijing 100049, China.
| |
Collapse
|
44
|
Haroon H, Hunter A, Farhangrazi Z, Moghimi S. A brief history of long circulating nanoparticles. Adv Drug Deliv Rev 2022; 188:114396. [DOI: 10.1016/j.addr.2022.114396] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2022] [Revised: 05/26/2022] [Accepted: 05/29/2022] [Indexed: 12/21/2022]
|
45
|
Enrichment Methods for Murine Liver Non-Parenchymal Cells Differentially Affect Their Immunophenotype and Responsiveness towards Stimulation. Int J Mol Sci 2022; 23:ijms23126543. [PMID: 35742987 PMCID: PMC9223567 DOI: 10.3390/ijms23126543] [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: 05/20/2022] [Revised: 06/07/2022] [Accepted: 06/09/2022] [Indexed: 12/12/2022] Open
Abstract
Hepatocytes comprise the majority of the liver and largely exert metabolic functions, whereas non-parenchymal cells (NPCs)—comprising Kupffer cells, dendritic cells and liver sinusoidal endothelial cells—control the immunological state within this organ. Here, we compared the suitability of two isolation methods for murine liver NPCs. Liver perfusion (LP) with collagenase/DNase I applied via the portal vein leads to efficient liver digestion, whereas the modified liver dissociation (LD) method combines mechanical dissociation of the retrieved organ with enzymatic degradation of the extracellular matrix. In cases of both LP and LD, NPCs were enriched by subsequent gradient density centrifugation. Our results indicate that LP and LD are largely comparable with regards to the yield, purity, and composition of liver NPCs. However, LD-enriched liver NPCs displayed a higher degree of activation after overnight cultivation, and accordingly were less responsive towards stimulation with toll-like receptor ligands that are frequently used as adjuvants, e.g., in nano-vaccines. We conclude that LP is more suitable for obtaining liver NPCs for subsequent in vitro studies, whereas LD as the less laborious method, is more convenient for parallel isolation of larger numbers of samples for ex vivo analysis.
Collapse
|
46
|
Szafranska K, Neuman T, Baster Z, Rajfur Z, Szelest O, Holte C, Kubisiak A, Kus E, Wolfson DL, Chlopicki S, Ahluwalia BS, Lekka M, Szymonski M, McCourt P, Zapotoczny B. From fixed-dried to wet-fixed to live - comparative super-resolution microscopy of liver sinusoidal endothelial cell fenestrations. NANOPHOTONICS (BERLIN, GERMANY) 2022; 11:2253-2270. [PMID: 39678082 PMCID: PMC11636152 DOI: 10.1515/nanoph-2021-0818] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Revised: 02/21/2022] [Accepted: 04/06/2022] [Indexed: 12/17/2024]
Abstract
Fenestrations in liver sinusoidal endothelial cells (LSEC) are transcellular nanopores of 50-350 nm diameter that facilitate bidirectional transport of solutes and macromolecules between the bloodstream and the parenchyma of the liver. Liver diseases, ageing, and various substances such as nicotine or ethanol can negatively influence LSECs fenestrations and lead to defenestration. Over the years, the diameter of fenestrations remained the main challenge for imaging of LSEC in vitro. Several microscopy, or rather nanoscopy, approaches have been used to quantify fenestrations in LSEC to assess the effect of drugs and, and toxins in different biological models. All techniques have their limitations, and measurements of the "true" size of fenestrations are hampered because of this. In this study, we approach the comparison of different types of microscopy in a correlative manner. We combine scanning electron microscopy (SEM) with optical nanoscopy methods such as structured illumination microscopy (SIM) or stimulated emission depletion (STED) microscopy. In addition, we combined atomic force microscopy (AFM) with SEM and STED, all to better understand the previously reported differences between the reports of fenestration dimensions. We conclude that sample dehydration alters fenestration diameters. Finally, we propose the combination of AFM with conventional microscopy that allows for easy super-resolution observation of the cell dynamics with additional chemical information that can be traced back for the whole experiment. Overall, by pairing the various types of imaging techniques that provide topological 2D/3D/label-free/chemical information we get a deeper insight into both limitations and strengths of each type microscopy when applied to fenestration analysis.
Collapse
Affiliation(s)
- Karolina Szafranska
- Department of Medical Biology, Vascular Biology Research Group, University of Tromsø (UiT), The Arctic University of Norway, Tromsø, Norway
| | - Tanja Neuman
- JPK BioAFM Business, Nano Surfaces and Metrology Division, Bruker Nano GmbH, Berlin, Germany
| | - Zbigniew Baster
- Marian Smoluchowski Institute of Physics, Faculty of Physics, Astronomy and Applied Computer Sciences, Jagiellonian University, Krakow, Poland
| | - Zenon Rajfur
- Marian Smoluchowski Institute of Physics, Faculty of Physics, Astronomy and Applied Computer Sciences, Jagiellonian University, Krakow, Poland
| | | | - Christopher Holte
- Department of Medical Biology, Vascular Biology Research Group, University of Tromsø (UiT), The Arctic University of Norway, Tromsø, Norway
| | - Agata Kubisiak
- Marian Smoluchowski Institute of Physics, Faculty of Physics, Astronomy and Applied Computer Sciences, Jagiellonian University, Krakow, Poland
| | - Edyta Kus
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, Krakow, Poland
| | - Deanna L. Wolfson
- Department of Physics and Technology, UiT-The Arctic University of Norway, Tromsø, Norway
| | - Stefan Chlopicki
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, Krakow, Poland
- Chair of Pharmacology, Jagiellonian University Medical College, Krakow, Poland
| | - Balpreet S. Ahluwalia
- Department of Physics and Technology, UiT-The Arctic University of Norway, Tromsø, Norway
| | - Malgorzata Lekka
- Institute of Nuclear Physics, Polish Academy of Sciences, Kraków, Poland
| | - Marek Szymonski
- Marian Smoluchowski Institute of Physics, Faculty of Physics, Astronomy and Applied Computer Sciences, Jagiellonian University, Krakow, Poland
| | - Peter McCourt
- Department of Medical Biology, Vascular Biology Research Group, University of Tromsø (UiT), The Arctic University of Norway, Tromsø, Norway
| | - Bartlomiej Zapotoczny
- Department of Medical Biology, Vascular Biology Research Group, University of Tromsø (UiT), The Arctic University of Norway, Tromsø, Norway
- Institute of Nuclear Physics, Polish Academy of Sciences, Kraków, Poland
| |
Collapse
|