1
|
Li Z, Rius Rigau A, Xie W, Huang L, Ye W, Li YN, Matei AE, Bergmann C, Shao X, Zou H, Wang J, Pinello L, Distler JHW, He R, Liang M. Spatial multiomics decipher fibroblast-macrophage dynamics in systemic sclerosis. Ann Rheum Dis 2025:S0003-4967(25)00953-7. [PMID: 40410053 DOI: 10.1016/j.ard.2025.04.025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2025] [Revised: 04/18/2025] [Accepted: 04/27/2025] [Indexed: 05/25/2025]
Abstract
OBJECTIVES Stromal-immune crosstalk shapes the pathogenic microenvironment of systemic sclerosis (SSc), but the spatial regulatory networks underlying fibrogenesis remain poorly defined. We aimed to explore tissue organisation and cell coordination in SSc skin, providing spatiotemporal insights into disease mechanisms and bridging the gap between omics discovery and precision medicine. METHODS We performed spatial transcriptomics on skin biopsies from 10 patients with diffuse cutaneous SSc and 4 healthy controls using the 10× Visium platform. These findings were confirmed using higher-resolution Stereo-seq transcriptomics, spatial proteomics, and single-cell RNA sequencing data from patients with SSc, SSc mouse models, and wound-healing reindeer models. In vivo and in vitro studies were conducted to validate the key regulatory pathways. RESULTS Fourteen skin biopsies were analysed, revealing significant expansion of fibrotic niches enriched with fibroblasts and macrophages in SSc, correlating with clinical severity. We revealed disease-specific cell states of fibroblasts and macrophages and evaluated their spatial dependency and cell-cell communication. Stratification based on signature genes enabled the identification of patients with SSc with progressive disease and treatment-nonresponsive phenotype. ACKR3 (a CXCL12 decoy receptor) was selectively expressed in myofibroblast progenitors, which diminished during differentiation towards mature myofibroblast, potentially serving to regulate CXCL12/CXCR4-mediated proinflammatory macrophage recruitment. Inhibition of CXCR4 attenuated skin and lung fibrosis in experimental fibrosis mouse models. CONCLUSIONS Our spatially resolved atlas uncovered dynamic fibroblast-macrophage interplay as a hallmark of fibrotic niche expansion. These findings offer spatiotemporal insights into disease mechanisms and pave the way for advanced mechanistic and therapeutic studies, bridging the gap between omics discovery and precision medicine.
Collapse
Affiliation(s)
- Zhijian Li
- Broad Institute of MIT and Harvard, Cambridge, MA, USA; Molecular Pathology Unit, Center for Cancer Research, Massachusetts General Hospital, Boston, MA, USA
| | - Aleix Rius Rigau
- Department of Internal Medicine 3, Rheumatology and Clinical Immunology, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and University Hospital Erlangen, Erlangen, Germany; Deutsches Zentrum Immuntherapie (DZI), FAU Erlangen-Nürnberg and University Hospital Erlangen, Erlangen, Germany
| | - Wenjie Xie
- Department of Immunology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Linlin Huang
- Department of Rheumatology, Huashan Hospital, Fudan University, Shanghai, China; Institute of Rheumatology, Immunology and Allergy, Fudan University, Shanghai, China; Huashan Rare Disease Center, Huashan Hospital, Fudan University, Shanghai, China
| | - Wenjing Ye
- Department of Rheumatology, Huashan Hospital, Fudan University, Shanghai, China; Institute of Rheumatology, Immunology and Allergy, Fudan University, Shanghai, China; Huashan Rare Disease Center, Huashan Hospital, Fudan University, Shanghai, China
| | - Yi-Nan Li
- Clinic for Rheumatology, University Hospital Düsseldorf, Medical Faculty of Heinrich Heine University, Düsseldorf, Germany; Hiller Research Center, University Hospital Düsseldorf, Medical Faculty of Heinrich Heine University, Düsseldorf, Germany
| | - Alexandru-Emil Matei
- Clinic for Rheumatology, University Hospital Düsseldorf, Medical Faculty of Heinrich Heine University, Düsseldorf, Germany; Hiller Research Center, University Hospital Düsseldorf, Medical Faculty of Heinrich Heine University, Düsseldorf, Germany
| | - Christina Bergmann
- Department of Internal Medicine 3, Rheumatology and Clinical Immunology, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and University Hospital Erlangen, Erlangen, Germany; Deutsches Zentrum Immuntherapie (DZI), FAU Erlangen-Nürnberg and University Hospital Erlangen, Erlangen, Germany
| | - Xiaohang Shao
- Department of Nephrology and Rheumatology Immunology, Ningbo Medical Center Lihuili Hospital, Ningbo, China
| | - Hejian Zou
- Department of Rheumatology, Huashan Hospital, Fudan University, Shanghai, China; Institute of Rheumatology, Immunology and Allergy, Fudan University, Shanghai, China; Huashan Rare Disease Center, Huashan Hospital, Fudan University, Shanghai, China
| | - Jiucun Wang
- Department of Rheumatology, Huashan Hospital, Fudan University, Shanghai, China; Institute of Rheumatology, Immunology and Allergy, Fudan University, Shanghai, China; State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, School of Life Sciences, and Human Phenome Institute, Fudan University, Shanghai, China; Research Unit of Dissecting the Population Genetics and Developing New Technologies for Treatment and Prevention of Skin Phenotypes and Dermatological Diseases (2019RU058), Chinese Academy of Medical Sciences, Shanghai, China
| | - Luca Pinello
- Broad Institute of MIT and Harvard, Cambridge, MA, USA; Molecular Pathology Unit, Center for Cancer Research, Massachusetts General Hospital, Boston, MA, USA; Department of Pathology, Harvard Medical School, Boston, MA, USA
| | - Jörg H W Distler
- Clinic for Rheumatology, University Hospital Düsseldorf, Medical Faculty of Heinrich Heine University, Düsseldorf, Germany; Hiller Research Center, University Hospital Düsseldorf, Medical Faculty of Heinrich Heine University, Düsseldorf, Germany.
| | - Rui He
- Department of Immunology, School of Basic Medical Sciences, Fudan University, Shanghai, China; National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, China.
| | - Minrui Liang
- Department of Rheumatology, Huashan Hospital, Fudan University, Shanghai, China; Institute of Rheumatology, Immunology and Allergy, Fudan University, Shanghai, China; Huashan Rare Disease Center, Huashan Hospital, Fudan University, Shanghai, China.
| |
Collapse
|
2
|
Vijayan V, M Unagolla J, Panchal D, John JE, Menon SS, Menon JU. Biomimetic nanoparticles for targeted therapy of liver disease. RSC PHARMACEUTICS 2025:d5pm00044k. [PMID: 40321406 PMCID: PMC12045541 DOI: 10.1039/d5pm00044k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/14/2025] [Accepted: 04/25/2025] [Indexed: 05/08/2025]
Abstract
Liver fibrosis is a progressive and fatal condition characterized by stiffness and scarring of the liver due to excessive buildup of extracellular matrix (ECM) proteins. If left untreated, it can progress to liver cirrhosis and hepatocellular carcinoma (HCC)-one of the fastest-rising causes of cancer mortality in the United States. Despite the increased prevalence of liver fibrosis due to infections, exposure to toxins, and unhealthy lifestyles, there are no effective treatments available. Recent advances in nanomedicine can lead to more targeted and effective strategies for treating liver diseases than existing treatments. In particular, the use of biomimetic nanoparticles (NPs) such as liposomes and cell-membrane-coated NPs is of interest. NPs functionalized with cell membranes mimic the properties of the source cell used and provide inherent immune evasion ability, homologous adhesion, and prolonged circulation. This review explores the types of biomimetic coatings, different cargoes delivered through biomimetic NPs for various treatment modalities, and the type of core NPs used for targeting liver fibrosis and HCC.
Collapse
Affiliation(s)
- Veena Vijayan
- Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island Kingston RI 02881 USA
| | - Janitha M Unagolla
- Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island Kingston RI 02881 USA
| | - Dhruvisha Panchal
- Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island Kingston RI 02881 USA
| | - Judith Eloyi John
- Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island Kingston RI 02881 USA
| | | | - Jyothi U Menon
- Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island Kingston RI 02881 USA
- Department of Chemical Engineering, University of Rhode Island Kingston RI 02881 USA
| |
Collapse
|
3
|
Menachem R, Nudelman I, Vorontsova A, Livneh I, Sela M, Benguigui M, Manobla B, Shammai Y, Deo A, Buxbaum C, Bessler R, Raviv Z, Shklover J, Sznitman J, Ciechanover A, Schroeder A, Shaked Y. Bone Marrow-Targeted Liposomes Loaded with Bortezomib Overcome Multiple Myeloma Resistance. ACS NANO 2025; 19:11684-11701. [PMID: 40117329 PMCID: PMC11966756 DOI: 10.1021/acsnano.4c10597] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/04/2024] [Revised: 03/11/2025] [Accepted: 03/12/2025] [Indexed: 03/23/2025]
Abstract
Multiple myeloma (MM) poses a significant therapeutic challenge due to its persistent progression and low survival rate. Although the proteasome inhibitor bortezomib has revolutionized MM treatment, MM aggressiveness and drug resistance remain critical concerns. To tackle this problem, we developed AMD3100-targeted Bortezomib Liposomes (ATBL) designed for the targeted delivery of bortezomib to MM cells. Uptake of ATBL into MM cells was dependent on CXCR4 and was enhanced compared to nontargeted liposomes, both in vitro and in vivo. Treating MM-bearing mice with ATBL achieved superior therapeutic efficacy compared to treatment with free bortezomib or nontargeted bortezomib-loaded liposomes. Notably, the therapeutic activity of ATBL was limited in mice inoculated with CXCR4-knockdown MM cells, highlighting CXCR4 as a potential biomarker for ATBL response. Importantly, ATBL was effective against an aggressive and bortezomib-resistant MM clone both in vitro and in vivo. Toxicity and biodistribution profiles demonstrated the safety and bone marrow-targeting ability of ATBL. Collectively, this study highlights ATBL as a promising next-generation proteasome inhibitor-based therapy that incorporates bone marrow-targeting ability and sensitizing elements to overcome drug resistance in MM.
Collapse
Affiliation(s)
- Rotem Menachem
- Department
of Cell Biology and Cancer Science, Rappaport Faculty of Medicine, Technion−Israel Institute of Technology, Haifa 3525422, Israel
- Rappaport
Technion Integrated Cancer Center, Technion
− Israel Institute of Technology, Haifa 3525422, Israel
- Faculty
of Chemical Engineering, Technion −
Israel Institute of Technology, Haifa 3200003, Israel
| | - Igor Nudelman
- Rappaport
Technion Integrated Cancer Center, Technion
− Israel Institute of Technology, Haifa 3525422, Israel
- Faculty
of Chemical Engineering, Technion −
Israel Institute of Technology, Haifa 3200003, Israel
| | - Avital Vorontsova
- Department
of Cell Biology and Cancer Science, Rappaport Faculty of Medicine, Technion−Israel Institute of Technology, Haifa 3525422, Israel
- Rappaport
Technion Integrated Cancer Center, Technion
− Israel Institute of Technology, Haifa 3525422, Israel
| | - Ido Livneh
- Department
of Cell Biology and Cancer Science, Rappaport Faculty of Medicine, Technion−Israel Institute of Technology, Haifa 3525422, Israel
- Rappaport
Technion Integrated Cancer Center, Technion
− Israel Institute of Technology, Haifa 3525422, Israel
| | - Mor Sela
- Rappaport
Technion Integrated Cancer Center, Technion
− Israel Institute of Technology, Haifa 3525422, Israel
- Faculty
of Chemical Engineering, Technion −
Israel Institute of Technology, Haifa 3200003, Israel
| | - Madeleine Benguigui
- Department
of Cell Biology and Cancer Science, Rappaport Faculty of Medicine, Technion−Israel Institute of Technology, Haifa 3525422, Israel
- Rappaport
Technion Integrated Cancer Center, Technion
− Israel Institute of Technology, Haifa 3525422, Israel
| | - Bar Manobla
- Department
of Cell Biology and Cancer Science, Rappaport Faculty of Medicine, Technion−Israel Institute of Technology, Haifa 3525422, Israel
- Rappaport
Technion Integrated Cancer Center, Technion
− Israel Institute of Technology, Haifa 3525422, Israel
| | - Yael Shammai
- Rappaport
Technion Integrated Cancer Center, Technion
− Israel Institute of Technology, Haifa 3525422, Israel
- Faculty
of Chemical Engineering, Technion −
Israel Institute of Technology, Haifa 3200003, Israel
| | - Abhilash Deo
- Department
of Cell Biology and Cancer Science, Rappaport Faculty of Medicine, Technion−Israel Institute of Technology, Haifa 3525422, Israel
- Rappaport
Technion Integrated Cancer Center, Technion
− Israel Institute of Technology, Haifa 3525422, Israel
| | - Chen Buxbaum
- Department
of Cell Biology and Cancer Science, Rappaport Faculty of Medicine, Technion−Israel Institute of Technology, Haifa 3525422, Israel
- Rappaport
Technion Integrated Cancer Center, Technion
− Israel Institute of Technology, Haifa 3525422, Israel
| | - Ron Bessler
- Faculty
of Biomedical Engineering, Technion−Israel
Institute of Technology, Haifa 3200001, Israel
| | - Ziv Raviv
- Department
of Cell Biology and Cancer Science, Rappaport Faculty of Medicine, Technion−Israel Institute of Technology, Haifa 3525422, Israel
- Rappaport
Technion Integrated Cancer Center, Technion
− Israel Institute of Technology, Haifa 3525422, Israel
| | - Jeny Shklover
- Rappaport
Technion Integrated Cancer Center, Technion
− Israel Institute of Technology, Haifa 3525422, Israel
- Faculty
of Chemical Engineering, Technion −
Israel Institute of Technology, Haifa 3200003, Israel
| | - Josué Sznitman
- Faculty
of Biomedical Engineering, Technion−Israel
Institute of Technology, Haifa 3200001, Israel
| | - Aaron Ciechanover
- Department
of Cell Biology and Cancer Science, Rappaport Faculty of Medicine, Technion−Israel Institute of Technology, Haifa 3525422, Israel
- Rappaport
Technion Integrated Cancer Center, Technion
− Israel Institute of Technology, Haifa 3525422, Israel
| | - Avi Schroeder
- Rappaport
Technion Integrated Cancer Center, Technion
− Israel Institute of Technology, Haifa 3525422, Israel
- Faculty
of Chemical Engineering, Technion −
Israel Institute of Technology, Haifa 3200003, Israel
| | - Yuval Shaked
- Department
of Cell Biology and Cancer Science, Rappaport Faculty of Medicine, Technion−Israel Institute of Technology, Haifa 3525422, Israel
- Rappaport
Technion Integrated Cancer Center, Technion
− Israel Institute of Technology, Haifa 3525422, Israel
| |
Collapse
|
4
|
Wang H, Yang Y, Xu Y, Chen Y, Zhang W, Liu T, Chen G, Wang K. Phage-based delivery systems: engineering, applications, and challenges in nanomedicines. J Nanobiotechnology 2024; 22:365. [PMID: 38918839 PMCID: PMC11197292 DOI: 10.1186/s12951-024-02576-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Accepted: 05/21/2024] [Indexed: 06/27/2024] Open
Abstract
Bacteriophages (phages) represent a unique category of viruses with a remarkable ability to selectively infect host bacteria, characterized by their assembly from proteins and nucleic acids. Leveraging their exceptional biological properties and modifiable characteristics, phages emerge as innovative, safe, and efficient delivery vectors. The potential drawbacks associated with conventional nanocarriers in the realms of drug and gene delivery include a lack of cell-specific targeting, cytotoxicity, and diminished in vivo transfection efficiency. In contrast, engineered phages, when employed as cargo delivery vectors, hold the promise to surmount these limitations and attain enhanced delivery efficacy. This review comprehensively outlines current strategies for the engineering of phages, delineates the principal types of phages utilized as nanocarriers in drug and gene delivery, and explores the application of phage-based delivery systems in disease therapy. Additionally, an incisive analysis is provided, critically examining the challenges confronted by phage-based delivery systems within the domain of nanotechnology. The primary objective of this article is to furnish a theoretical reference that contributes to the reasoned design and development of potent phage-based delivery systems.
Collapse
Affiliation(s)
- Hui Wang
- School of Pharmacy, Nantong University, Nantong, 226001, China
- Qingdao Central Hospital, University of Health and Rehabilitation Sciences (Qingdao Central Medical Group), Qingdao, 266024, China
- School of Rehabilitation Sciences and Engineering, University of Health and Rehabilitation Sciences, Qingdao, 266024, China
| | - Ying Yang
- School of Pharmacy, Nantong University, Nantong, 226001, China
| | - Yan Xu
- School of Pharmacy, Nantong University, Nantong, 226001, China
| | - Yi Chen
- School of Pharmacy, Nantong University, Nantong, 226001, China
| | - Wenjie Zhang
- School of Pharmacy, Nantong University, Nantong, 226001, China
| | - Tianqing Liu
- NICM Health Research Institute, Western Sydney University, Sydney, NSW, 2145, Australia.
| | - Gang Chen
- Qingdao Central Hospital, University of Health and Rehabilitation Sciences (Qingdao Central Medical Group), Qingdao, 266024, China.
- School of Rehabilitation Sciences and Engineering, University of Health and Rehabilitation Sciences, Qingdao, 266024, China.
| | - Kaikai Wang
- School of Pharmacy, Nantong University, Nantong, 226001, China.
| |
Collapse
|
5
|
Chen L, Guo W, Mao C, Shen J, Wan M. Liver fibrosis: pathological features, clinical treatment and application of therapeutic nanoagents. J Mater Chem B 2024; 12:1446-1466. [PMID: 38265305 DOI: 10.1039/d3tb02790b] [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: 01/25/2024]
Abstract
Liver fibrosis is a reversible damage-repair response, the pathological features of which mainly include damage to hepatocytes, sinusoid capillarization, hepatic stellate cells activation, excessive accumulation of extracellular matrix and inflammatory response. Although some treatments (including drugs and stem cell therapy) for these pathological features have been shown to be effective, more clinical trials are needed to confirm their effectiveness. In recent years, nanomaterials-based therapies have emerged as an innovative and promising alternative to traditional drugs, being explored for the treatment of liver fibrosis diseases. Natural nanomaterials (including extracellular vesicles) and synthetic nanomaterials (including inorganic nanomaterials and organic nanomaterials) are developed to facilitate drug targeting delivery and combination therapy. In this review, the pathological features of liver fibrosis and the current anti-fibrosis drugs in clinical trials are briefly introduced, followed by a detailed introduction of the therapeutic nanoagents for the precise delivery of anti-fibrosis drugs. Finally, the future development trend in this field is discussed.
Collapse
Affiliation(s)
- Lin Chen
- National and Local Joint Engineering Research Center of Biomedical Functional Materials, School of Chemistry and Materials Science, Nanjing Normal University, Nanjing 210023, China.
| | - Wenyan Guo
- National and Local Joint Engineering Research Center of Biomedical Functional Materials, School of Chemistry and Materials Science, Nanjing Normal University, Nanjing 210023, China.
| | - Chun Mao
- National and Local Joint Engineering Research Center of Biomedical Functional Materials, School of Chemistry and Materials Science, Nanjing Normal University, Nanjing 210023, China.
| | - Jian Shen
- National and Local Joint Engineering Research Center of Biomedical Functional Materials, School of Chemistry and Materials Science, Nanjing Normal University, Nanjing 210023, China.
| | - Mimi Wan
- National and Local Joint Engineering Research Center of Biomedical Functional Materials, School of Chemistry and Materials Science, Nanjing Normal University, Nanjing 210023, China.
| |
Collapse
|
6
|
Essola JM, Zhang M, Yang H, Li F, Xia B, Mavoungou JF, Hussain A, Huang Y. Exosome regulation of immune response mechanism: Pros and cons in immunotherapy. Bioact Mater 2024; 32:124-146. [PMID: 37927901 PMCID: PMC10622742 DOI: 10.1016/j.bioactmat.2023.09.018] [Citation(s) in RCA: 18] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Revised: 09/06/2023] [Accepted: 09/25/2023] [Indexed: 11/07/2023] Open
Abstract
Due to its multiple features, including the ability to orchestrate remote communication between different tissues, the exosomes are the extracellular vesicles arousing the highest interest in the scientific community. Their size, established as an average of 30-150 nm, allows them to be easily uptaken by most cells. According to the type of cells-derived exosomes, they may carry specific biomolecular cargoes used to reprogram the cells they are interacting with. In certain circumstances, exosomes stimulate the immune response by facilitating or amplifying the release of foreign antigens-killing cells, inflammatory factors, or antibodies (immune activation). Meanwhile, in other cases, they are efficiently used by malignant elements such as cancer cells to mislead the immune recognition mechanism, carrying and transferring their cancerous cargoes to distant healthy cells, thus contributing to antigenic invasion (immune suppression). Exosome dichotomic patterns upon immune system regulation present broad advantages in immunotherapy. Its perfect comprehension, from its early biogenesis to its specific interaction with recipient cells, will promote a significant enhancement of immunotherapy employing molecular biology, nanomedicine, and nanotechnology.
Collapse
Affiliation(s)
- Julien Milon Essola
- School of Life Science, Advanced Research Institute of Multidisciplinary Science, School of Medical Technology, Key Laboratory of Molecular Medicine and Biotherapy, Key Laboratory of Medical Molecule Science and Pharmaceutics Engineering, Beijing Institute of Technology, Beijing, 100081, China
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, No. 11, First North Road, Zhongguancun, Beijing, 100190, PR China
- University of Chinese Academy of Sciences. Beijing 100049, PR China
| | - Mengjie Zhang
- School of Life Science, Advanced Research Institute of Multidisciplinary Science, School of Medical Technology, Key Laboratory of Molecular Medicine and Biotherapy, Key Laboratory of Medical Molecule Science and Pharmaceutics Engineering, Beijing Institute of Technology, Beijing, 100081, China
| | - Haiyin Yang
- School of Life Science, Advanced Research Institute of Multidisciplinary Science, School of Medical Technology, Key Laboratory of Molecular Medicine and Biotherapy, Key Laboratory of Medical Molecule Science and Pharmaceutics Engineering, Beijing Institute of Technology, Beijing, 100081, China
| | - Fangzhou Li
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, No. 11, First North Road, Zhongguancun, Beijing, 100190, PR China
| | - Bozhang Xia
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, No. 11, First North Road, Zhongguancun, Beijing, 100190, PR China
- University of Chinese Academy of Sciences. Beijing 100049, PR China
| | - Jacques François Mavoungou
- Université Internationale de Libreville, Libreville, 20411, Gabon
- Central and West African Virus Epidemiology, Libreville, 2263, Gabon
- Département de phytotechnologies, Institut National Supérieur d’Agronomie et de Biotechnologie, Université des Sciences et Techniques de Masuku, Franceville, 901, Gabon
- Institut de Recherches Agronomiques et Forestiers, Centre National de la Recherche Scientifique et du développement Technologique, Libreville, 16182, Gabon
| | - Abid Hussain
- School of Life Science, Advanced Research Institute of Multidisciplinary Science, School of Medical Technology, Key Laboratory of Molecular Medicine and Biotherapy, Key Laboratory of Medical Molecule Science and Pharmaceutics Engineering, Beijing Institute of Technology, Beijing, 100081, China
| | - Yuanyu Huang
- School of Life Science, Advanced Research Institute of Multidisciplinary Science, School of Medical Technology, Key Laboratory of Molecular Medicine and Biotherapy, Key Laboratory of Medical Molecule Science and Pharmaceutics Engineering, Beijing Institute of Technology, Beijing, 100081, China
- Rigerna Therapeutics Co. Ltd., China
| |
Collapse
|
7
|
Raza F, Zafar H, Jiang L, Su J, Yuan W, Qiu M, Paiva-Santos AC. Progress of cell membrane-derived biomimetic nanovesicles for cancer phototherapy. Biomater Sci 2023; 12:57-91. [PMID: 37902579 DOI: 10.1039/d3bm01170d] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2023]
Abstract
In recent years, considerable attention has been given to phototherapy, including photothermal and photodynamic therapy to kill tumor cells by producing heat or reactive oxygen species (ROS). It has the high merits of noninvasiveness and limited drug resistance. To fully utilize this therapy, an extraordinary nanovehicle is required to target phototherapeutic agents in the tumor cells. Nanovesicles embody an ideal strategy for drug delivery applications. Cell membrane-derived biomimetic nanovesicles represent a developing type of nanocarrier. Combining this technique with cancer phototherapy could enable a novel strategy. Herein, efforts are made to describe a comprehensive overview of cell membrane-derived biomimetic nanovesicles for cancer phototherapy. The description in this review is mainly based on representative examples of exosome-derived biomimetic nanomedicine research, ranging from their comparison with traditional nanocarriers to extensive applications in cancer phototherapy. Additionally, the challenges and future prospectives for translating these for clinical application are discussed.
Collapse
Affiliation(s)
- Faisal Raza
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, P.R. China.
| | - Hajra Zafar
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, P.R. China.
| | - Liangdi Jiang
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, P.R. China.
| | - Jing Su
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, P.R. China.
| | - Weien Yuan
- Engineering Research Center of Cell & Therapeutic Antibody, Ministry of Education, and School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Mingfeng Qiu
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, P.R. China.
| | - Ana Cláudia Paiva-Santos
- Department of Pharmaceutical Technology, Faculty of Pharmacy of the University of Coimbra, University of Coimbra, Azinhaga Sta. Comba, 3000-548 Coimbra, Portugal
- LAQV, REQUIMTE, Department of Pharmaceutical Technology, Faculty of Pharmacy of the University of Coimbra, University of Coimbra, Azinhaga Sta. Comba, 3000-548 Coimbra, Portugal
| |
Collapse
|
8
|
Li F, Zhao Y, Nie G. Nanotechnology-based combinational strategies toward the regulation of myofibroblasts and diseased microenvironment in liver fibrosis and hepatic carcinoma. NANO RESEARCH 2023; 16:13042-13055. [DOI: 10.1007/s12274-023-5809-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/04/2023] [Revised: 04/29/2023] [Accepted: 05/05/2023] [Indexed: 01/03/2025]
|
9
|
Dwivedi NV, Datta S, El-Kersh K, Sadikot RT, Ganti AK, Batra SK, Jain M. GPCRs and fibroblast heterogeneity in fibroblast-associated diseases. FASEB J 2023; 37:e23101. [PMID: 37486603 PMCID: PMC10916681 DOI: 10.1096/fj.202301091] [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: 06/01/2023] [Accepted: 07/06/2023] [Indexed: 07/25/2023]
Abstract
G protein-coupled receptors (GPCRs) are the largest and most diverse class of signaling receptors. GPCRs regulate many functions in the human body and have earned the title of "most targeted receptors". About one-third of the commercially available drugs for various diseases target the GPCRs. Fibroblasts lay the architectural skeleton of the body, and play a key role in supporting the growth, maintenance, and repair of almost all tissues by responding to the cellular cues via diverse and intricate GPCR signaling pathways. This review discusses the dynamic architecture of the GPCRs and their intertwined signaling in pathological conditions such as idiopathic pulmonary fibrosis, cardiac fibrosis, pancreatic fibrosis, hepatic fibrosis, and cancer as opposed to the GPCR signaling of fibroblasts in physiological conditions. Understanding the dynamics of GPCR signaling in fibroblasts with disease progression can help in the recognition of the complex interplay of different GPCR subtypes in fibroblast-mediated diseases. This review highlights the importance of designing and adaptation of next-generation strategies such as GPCR-omics, focused target identification, polypharmacology, and effective personalized medicine approaches to achieve better therapeutic outcomes for fibrosis and fibrosis associated malignancies.
Collapse
Affiliation(s)
- Nidhi V Dwivedi
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Souvik Datta
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Karim El-Kersh
- Division of Pulmonary, Critical Care and Sleep Medicine, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Ruxana T Sadikot
- Division of Pulmonary, Critical Care and Sleep Medicine, University of Nebraska Medical Center, Omaha, Nebraska, USA
- VA Nebraska Western Iowa Health Care System
| | - Apar K. Ganti
- VA Nebraska Western Iowa Health Care System
- Division of Oncology and Hematology, University of Nebraska Medical Center, Omaha, Nebraska, USA
- Fred and Pamela Buffett Cancer Center, Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Surinder K Batra
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, Nebraska, USA
- Fred and Pamela Buffett Cancer Center, Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Maneesh Jain
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, Nebraska, USA
- Fred and Pamela Buffett Cancer Center, Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, Nebraska, USA
| |
Collapse
|
10
|
Ai W, Liu T, Lv C, Feng X, Wang Q. Modulation of cancer-associated fibroblasts by nanodelivery system to enhance efficacy of tumor therapy. Nanomedicine (Lond) 2023; 18:1025-1039. [PMID: 37584613 DOI: 10.2217/nnm-2023-0088] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/17/2023] Open
Abstract
Cancer-associated fibroblasts (CAFs) are the most common cells in the tumor stroma and are essential for tumor development and metastasis. While decreasing the release and infiltration of nanomedicine through nonspecific internalization, CAFs specifically increase solid tumor pressure and interstitial fluid pressure by secreting tumor growth- and migration-promoting cytokines, which increases vascular and organ pressure caused by solid tumor pressure. Nanoparticles have good permeability and can penetrate tumor tissue to reach the lesion area, inhibiting tumor growth. Thus, CAFs are used as modifiable targets. Here, the authors review the biological functions, origins and biomarkers of CAFs and summarize strategies for modulating CAFs in nanodelivery systems. This study provides a prospective guide to modulating CAFs to enhance oncology treatment.
Collapse
Affiliation(s)
- Wei Ai
- College of Life Science & Technology, Changchun University of Science & Technology, Changchun, Jilin, 130022, China
| | - Tianhui Liu
- College of Life Science & Technology, Changchun University of Science & Technology, Changchun, Jilin, 130022, China
| | - Changshun Lv
- College of Life Science & Technology, Changchun University of Science & Technology, Changchun, Jilin, 130022, China
| | - Xiangru Feng
- College of Life Science & Technology, Changchun University of Science & Technology, Changchun, Jilin, 130022, China
| | - Qingshuang Wang
- College of Life Science & Technology, Changchun University of Science & Technology, Changchun, Jilin, 130022, China
| |
Collapse
|
11
|
Mbugua SN. Targeting Tumor Microenvironment by Metal Peroxide Nanoparticles in Cancer Therapy. Bioinorg Chem Appl 2022; 2022:5041399. [PMID: 36568636 PMCID: PMC9788889 DOI: 10.1155/2022/5041399] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Revised: 12/07/2022] [Accepted: 12/10/2022] [Indexed: 12/23/2022] Open
Abstract
Solid tumors have a unique tumor microenvironment (TME), which includes hypoxia, low acidity, and high hydrogen peroxide and glutathione (GSH) levels, among others. These unique factors, which offer favourable microenvironments and nourishment for tumor development and spread, also serve as a gateway for specific and successful cancer therapies. A good example is metal peroxide structures which have been synthesized and utilized to enhance oxygen supply and they have shown great promise in the alleviation of hypoxia. In a hypoxic environment, certain oxygen-dependent treatments such as photodynamic therapy and radiotherapy fail to respond and therefore modulating the hypoxic tumor microenvironment has been found to enhance the antitumor impact of certain drugs. Under acidic environments, the hydrogen peroxide produced by the reaction of metal peroxides with water not only induces oxidative stress but also produces additional oxygen. This is achieved since hydrogen peroxide acts as a reactive substrate for molecules such as catalyse enzymes, alleviating tumor hypoxia observed in the tumor microenvironment. Metal ions released in the process can also offer distinct bioactivity in their own right. Metal peroxides used in anticancer therapy are a rapidly evolving field, and there is good evidence that they are a good option for regulating the tumor microenvironment in cancer therapy. In this regard, the synthesis and mechanisms behind the successful application of metal peroxides to specifically target the tumor microenvironment are highlighted in this review. Various characteristics of TME such as angiogenesis, inflammation, hypoxia, acidity levels, and metal ion homeostasis are addressed in this regard, together with certain forms of synergistic combination treatments.
Collapse
Affiliation(s)
- Simon Ngigi Mbugua
- Department of Chemistry, Kisii University, P.O. Box 408-40200, Kisii, Kenya
| |
Collapse
|
12
|
Zhang M, Hussain A, Yang H, Zhang J, Liang XJ, Huang Y. mRNA-based modalities for infectious disease management. NANO RESEARCH 2022; 16:672-691. [PMID: 35818566 PMCID: PMC9258466 DOI: 10.1007/s12274-022-4627-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Revised: 06/01/2022] [Accepted: 06/03/2022] [Indexed: 06/15/2023]
Abstract
The novel coronavirus disease 2019 (COVID-19) is still rampant all over the world, causing incalculable losses to the world. Major pharmaceutical organizations around the globe are focusing on vaccine research and drug development to prevent further damage caused by the pandemic. The messenger RNA (mRNA) technology has got ample of attention after the success of the two very effective mRNA vaccines during the recent pandemic of COVID-19. mRNA vaccine has been promoted to the core stage of pharmaceutical industry, and the rapid development of mRNA technology has exceeded expectations. Beyond COVID-19, the mRNA vaccine has been tested for various infectious diseases and undergoing clinical trials. Due to the ability of constant mutation, the viral infections demand abrupt responses and immediate production, and therefore mRNA-based technology offers best answers to sudden outbreaks. The need for mRNA-based vaccine became more obvious due to the recent emergence of new Omicron variant. In this review, we summarized the unique properties of mRNA-based vaccines for infectious diseases, delivery technologies, discussed current challenges, and highlighted the prospects of this promising technology in the future. We also discussed various clinical studies as well preclinical studies conducted on mRNA therapeutics for diverse infectious diseases.
Collapse
Affiliation(s)
- Mengjie Zhang
- School of Life Science, Advanced Research Institute of Multidisciplinary Science, School of Medical Technology (Institute of Engineering Medicine), Key Laboratory of Molecular Medicine and Biotherapy, Key Laboratory of Medical Molecule Science and Pharmaceutics Engineering, Beijing Institute of Technology, Beijing, 100081 China
| | - Abid Hussain
- School of Life Science, Advanced Research Institute of Multidisciplinary Science, School of Medical Technology (Institute of Engineering Medicine), Key Laboratory of Molecular Medicine and Biotherapy, Key Laboratory of Medical Molecule Science and Pharmaceutics Engineering, Beijing Institute of Technology, Beijing, 100081 China
| | - Haiyin Yang
- School of Life Science, Advanced Research Institute of Multidisciplinary Science, School of Medical Technology (Institute of Engineering Medicine), Key Laboratory of Molecular Medicine and Biotherapy, Key Laboratory of Medical Molecule Science and Pharmaceutics Engineering, Beijing Institute of Technology, Beijing, 100081 China
| | - Jinchao Zhang
- Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of the Ministry of Education, College of Chemistry and Environmental Science, Hebei University, Baoding, 071002 China
| | - Xing-Jie Liang
- Chinese Academy of Sciences (CAS) Center for Excellence in Nanoscience, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology of China, Beijing, 100190 China
| | - Yuanyu Huang
- School of Life Science, Advanced Research Institute of Multidisciplinary Science, School of Medical Technology (Institute of Engineering Medicine), Key Laboratory of Molecular Medicine and Biotherapy, Key Laboratory of Medical Molecule Science and Pharmaceutics Engineering, Beijing Institute of Technology, Beijing, 100081 China
- School of Materials and the Environment, Beijing Institute of Technology, Zhuhai, 519085 China
| |
Collapse
|