1
|
Chen GL, Li JY, Chen X, Liu JW, Zhang Q, Liu JY, Wen J, Wang N, Lei M, Wei JP, Yi L, Li JJ, Ling YP, Yi HQ, Hu Z, Duan J, Zhang J, Zeng B. Mechanosensitive channels TMEM63A and TMEM63B mediate lung inflation-induced surfactant secretion. J Clin Invest 2024; 134:e174508. [PMID: 38127458 PMCID: PMC10904053 DOI: 10.1172/jci174508] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Accepted: 12/20/2023] [Indexed: 12/23/2023] Open
Abstract
Pulmonary surfactant is a lipoprotein complex lining the alveolar surface to decrease the surface tension and facilitate inspiration. Surfactant deficiency is often seen in premature infants and in children and adults with respiratory distress syndrome. Mechanical stretch of alveolar type 2 epithelial (AT2) cells during lung expansion is the primary physiological factor that stimulates surfactant secretion; however, it is unclear whether there is a mechanosensor dedicated to this process. Here, we show that loss of the mechanosensitive channels TMEM63A and TMEM63B (TMEM63A/B) resulted in atelectasis and respiratory failure in mice due to a deficit of surfactant secretion. TMEM63A/B were predominantly localized at the limiting membrane of the lamellar body (LB), a lysosome-related organelle that stores pulmonary surfactant and ATP in AT2 cells. Activation of TMEM63A/B channels during cell stretch facilitated the release of surfactant and ATP from LBs fused with the plasma membrane. The released ATP evoked Ca2+ signaling in AT2 cells and potentiated exocytic fusion of more LBs. Our study uncovered a vital physiological function of TMEM63 mechanosensitive channels in preparing the lungs for the first breath at birth and maintaining respiration throughout life.
Collapse
Affiliation(s)
- Gui-Lan Chen
- Key Laboratory of Medical Electrophysiology, Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, and
| | - Jing-Yi Li
- Key Laboratory of Medical Electrophysiology, Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, and
| | - Xin Chen
- Key Laboratory of Medical Electrophysiology, Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, and
| | - Jia-Wei Liu
- Key Laboratory of Medical Electrophysiology, Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, and
| | - Qian Zhang
- Key Laboratory of Medical Electrophysiology, Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, and
| | - Jie-Yu Liu
- Key Laboratory of Medical Electrophysiology, Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, and
| | - Jing Wen
- Key Laboratory of Medical Electrophysiology, Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, and
| | - Na Wang
- Key Laboratory of Medical Electrophysiology, Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, and
| | - Ming Lei
- Key Laboratory of Medical Electrophysiology, Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, and
| | - Jun-Peng Wei
- Key Laboratory of Medical Electrophysiology, Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, and
| | - Li Yi
- Key Laboratory of Medical Electrophysiology, Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, and
| | - Jia-Jia Li
- Key Laboratory of Medical Electrophysiology, Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, and
| | - Yu-Peng Ling
- Key Laboratory of Medical Electrophysiology, Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, and
| | - He-Qiang Yi
- Department of Cardiothoracic Surgery, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan, China
| | - Zhenying Hu
- Human Aging Research Institute and School of Life Sciences and
| | - Jingjing Duan
- Human Aging Research Institute and School of Life Sciences and
| | - Jin Zhang
- School of Basic Medical Sciences, Nanchang University, Nanchang, Jiangxi, China
| | - Bo Zeng
- Key Laboratory of Medical Electrophysiology, Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, and
| |
Collapse
|
2
|
Dalla E, Papanicolaou M, Park MD, Barth N, Hou R, Segura-Villalobos D, Valencia Salazar L, Sun D, Forrest ARR, Casanova-Acebes M, Entenberg D, Merad M, Aguirre-Ghiso JA. Lung-resident alveolar macrophages regulate the timing of breast cancer metastasis. Cell 2024; 187:6631-6648.e20. [PMID: 39378878 PMCID: PMC11568918 DOI: 10.1016/j.cell.2024.09.016] [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: 05/09/2023] [Revised: 06/13/2024] [Accepted: 09/11/2024] [Indexed: 10/10/2024]
Abstract
Breast disseminated cancer cells (DCCs) can remain dormant in the lungs for extended periods, but the mechanisms limiting their expansion are not well understood. Research indicates that tissue-resident alveolar macrophages suppress breast cancer metastasis in lung alveoli by inducing dormancy. Through ligand-receptor mapping and intravital imaging, it was found that alveolar macrophages express transforming growth factor (TGF)-β2. This expression, along with persistent macrophage-cancer cell interactions via the TGF-βRIII receptor, maintains cancer cells in a dormant state. Depleting alveolar macrophages or losing the TGF-β2 receptor in cancer cells triggers metastatic awakening. Aggressive breast cancer cells are either suppressed by alveolar macrophages or evade this suppression by avoiding interaction and downregulating the TGF-β2 receptor. Restoring TGF-βRIII in aggressive cells reinstates TGF-β2-mediated macrophage growth suppression. Thus, alveolar macrophages act as a metastasis immune barrier, and downregulation of TGF-β2 signaling allows cancer cells to overcome macrophage-mediated growth suppression.
Collapse
Affiliation(s)
- Erica Dalla
- Division of Hematology and Oncology, Department of Medicine and Department of Otolaryngology, Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Oncological Sciences, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Michael Papanicolaou
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, New York, NY 10461, USA; Cancer Dormancy Institute, Albert Einstein College of Medicine, Bronx, New York, NY 10461, USA; Montefiore Einstein Comprehensive Cancer Center, Albert Einstein College of Medicine, Bronx, New York, NY 10461, USA; Gruss-Lipper Biophotonics Center, Albert Einstein College of Medicine, Bronx, New York, NY 10461, USA; Ruth L. and David S. Gottesman Institute for Stem Cell Research and Regenerative Medicine, Albert Einstein College of Medicine, Bronx, New York, NY 10461, USA; Institute for Aging Research, Albert Einstein College of Medicine, Bronx, New York, NY 10461, USA
| | - Matthew D Park
- Department of Oncological Sciences, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; The Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Nicole Barth
- Cancer Dormancy Institute, Albert Einstein College of Medicine, Bronx, New York, NY 10461, USA; Montefiore Einstein Comprehensive Cancer Center, Albert Einstein College of Medicine, Bronx, New York, NY 10461, USA; Gruss-Lipper Biophotonics Center, Albert Einstein College of Medicine, Bronx, New York, NY 10461, USA; Department of Pathology, Albert Einstein College of Medicine, Bronx, New York, NY 10461, USA; Cancer Research UK Edinburgh Centre, University of Edinburgh, Edinburgh, UK
| | - Rui Hou
- Harry Perkins Institute of Medical Research, QEII Medical Centre and Centre for Medical Research, The University of Western Australia, Nedlands, WA 6009, Australia
| | - Deisy Segura-Villalobos
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, New York, NY 10461, USA; Cancer Dormancy Institute, Albert Einstein College of Medicine, Bronx, New York, NY 10461, USA; Montefiore Einstein Comprehensive Cancer Center, Albert Einstein College of Medicine, Bronx, New York, NY 10461, USA; Gruss-Lipper Biophotonics Center, Albert Einstein College of Medicine, Bronx, New York, NY 10461, USA; Ruth L. and David S. Gottesman Institute for Stem Cell Research and Regenerative Medicine, Albert Einstein College of Medicine, Bronx, New York, NY 10461, USA; Institute for Aging Research, Albert Einstein College of Medicine, Bronx, New York, NY 10461, USA
| | - Luis Valencia Salazar
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, New York, NY 10461, USA; Cancer Dormancy Institute, Albert Einstein College of Medicine, Bronx, New York, NY 10461, USA; Montefiore Einstein Comprehensive Cancer Center, Albert Einstein College of Medicine, Bronx, New York, NY 10461, USA; Gruss-Lipper Biophotonics Center, Albert Einstein College of Medicine, Bronx, New York, NY 10461, USA; Ruth L. and David S. Gottesman Institute for Stem Cell Research and Regenerative Medicine, Albert Einstein College of Medicine, Bronx, New York, NY 10461, USA; Institute for Aging Research, Albert Einstein College of Medicine, Bronx, New York, NY 10461, USA
| | - Dan Sun
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, New York, NY 10461, USA; Cancer Dormancy Institute, Albert Einstein College of Medicine, Bronx, New York, NY 10461, USA; Montefiore Einstein Comprehensive Cancer Center, Albert Einstein College of Medicine, Bronx, New York, NY 10461, USA; Gruss-Lipper Biophotonics Center, Albert Einstein College of Medicine, Bronx, New York, NY 10461, USA
| | - Alistair R R Forrest
- Harry Perkins Institute of Medical Research, QEII Medical Centre and Centre for Medical Research, The University of Western Australia, Nedlands, WA 6009, Australia
| | - Maria Casanova-Acebes
- Cancer Immunity Laboratory, Molecular Oncology Program, Spanish National Cancer Centre, Madrid, Spain
| | - David Entenberg
- Cancer Dormancy Institute, Albert Einstein College of Medicine, Bronx, New York, NY 10461, USA; Montefiore Einstein Comprehensive Cancer Center, Albert Einstein College of Medicine, Bronx, New York, NY 10461, USA; Gruss-Lipper Biophotonics Center, Albert Einstein College of Medicine, Bronx, New York, NY 10461, USA; Department of Pathology, Albert Einstein College of Medicine, Bronx, New York, NY 10461, USA
| | - Miriam Merad
- Department of Oncological Sciences, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; The Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Human Immune Monitoring Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Julio A Aguirre-Ghiso
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, New York, NY 10461, USA; Cancer Dormancy Institute, Albert Einstein College of Medicine, Bronx, New York, NY 10461, USA; Montefiore Einstein Comprehensive Cancer Center, Albert Einstein College of Medicine, Bronx, New York, NY 10461, USA; Gruss-Lipper Biophotonics Center, Albert Einstein College of Medicine, Bronx, New York, NY 10461, USA; Ruth L. and David S. Gottesman Institute for Stem Cell Research and Regenerative Medicine, Albert Einstein College of Medicine, Bronx, New York, NY 10461, USA; Institute for Aging Research, Albert Einstein College of Medicine, Bronx, New York, NY 10461, USA.
| |
Collapse
|
3
|
Zhao Q, Mertová I, Wróblová A, Žabková S, Tlapáková T, Krylov V. Immunomodulatory role of Xenopus tropicalis immature Sertoli cells in tadpole muscle regeneration via macrophage response modulation. Stem Cell Res Ther 2024; 15:421. [PMID: 39533333 PMCID: PMC11558833 DOI: 10.1186/s13287-024-04050-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Accepted: 11/06/2024] [Indexed: 11/16/2024] Open
Abstract
BACKGROUND Regenerative medicine and transplantation science continuously seek methods to circumvent immune-mediated rejection and promote tissue regeneration. Sertoli cells, with their inherent immunoprotective properties, emerge as pivotal players in this quest. However, whether Sertoli cells can play immunomodulatory role in tadpole tail regeneration and can thus benefit the regeneration process are needed to be discovered. METHODS Immature Sertoli cells from Xenopus tropicalis (XtiSCs) were transplanted into X. tropicalis tadpoles, followed by the amputation of the final third of their tails. We assessed the migration of XtiSCs, tail regeneration length, muscle degradation and growth, and macrophage counts across various regions including the entire tail, tail trunk, injection site, and regeneration site. The interactions between XtiSCs and macrophages were examined using a confocal microscope. To deplete macrophages, clodronate liposomes were administered prior to the transplantation of XtiSCs, while the administration of control liposomes acted as a negative control. Student's t-test was used to compare the effects of XtiSCs injection to those of a 2/3PBS injection across groups with no liposomes, control liposomes, and clodronate liposomes. RESULTS XtiSCs have excellent viability after transplantation to tadpole tail and remarkable homing capabilities to the regeneration site after tail amputation. XtiSCs injection increased macrophage numbers at 3 days post-amputation and 5 days post-amputation in the tail trunk, specifically at the injection site and at the regenerated tail, in a macrophage depleted environment (clodronate-liposome injection). What's more, XtiSCs injection decreased muscle fibers degradation significantly at 1 day post-amputation and facilitated new muscle growth significantly at 3 days post-amputation. In addition, whole-mount immunostaining showed that some XtiSCs co-localized with macrophages. And we observed potential mitochondria transport from XtiSCs to macrophages using MitoTracker staining in tadpole tail. CONCLUSIONS Our study delineates the novel role of XtiSCs in facilitating muscle regeneration post tadpole tail amputation, underscoring a unique interaction with macrophages that is crucial for regenerative success. This study not only highlights the therapeutic potential of Sertoli cells in regenerative medicine but also opens avenues for clinical translation, offering insights into immunoregulatory strategies that could enhance tissue regeneration and transplant acceptance.
Collapse
Affiliation(s)
- Qing Zhao
- Department of Cell Biology, Faculty of Science, Charles University, Viničná 7, Prague 2, 128 00, Czech Republic
| | - Irem Mertová
- Department of Cell Biology, Faculty of Science, Charles University, Viničná 7, Prague 2, 128 00, Czech Republic
| | - Aneta Wróblová
- Department of Cell Biology, Faculty of Science, Charles University, Viničná 7, Prague 2, 128 00, Czech Republic
| | - Světlana Žabková
- Department of Cell Biology, Faculty of Science, Charles University, Viničná 7, Prague 2, 128 00, Czech Republic
| | - Tereza Tlapáková
- Department of Cell Biology, Faculty of Science, Charles University, Viničná 7, Prague 2, 128 00, Czech Republic
| | - Vladimir Krylov
- Department of Cell Biology, Faculty of Science, Charles University, Viničná 7, Prague 2, 128 00, Czech Republic.
| |
Collapse
|
4
|
Gu Y, Taifour T, Bui T, Zuo D, Pacis A, Poirier A, Attalla S, Fortier AM, Sanguin-Gendreau V, Pan TC, Papavasiliou V, Lin NU, Hughes ME, Smith K, Park M, Tremblay ML, Chodosh LA, Jeselsohn R, Muller WJ. Osteopontin is a therapeutic target that drives breast cancer recurrence. Nat Commun 2024; 15:9174. [PMID: 39448577 PMCID: PMC11502809 DOI: 10.1038/s41467-024-53023-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Accepted: 09/29/2024] [Indexed: 10/26/2024] Open
Abstract
Recurrent breast cancers often develop resistance to standard-of-care therapies. Identifying targetable factors contributing to cancer recurrence remains the rate-limiting step in improving long-term outcomes. In this study, we identify tumor cell-derived osteopontin as an autocrine and paracrine driver of tumor recurrence. Osteopontin promotes tumor cell proliferation, recruits macrophages, and synergizes with IL-4 to further polarize them into a pro-tumorigenic state. Macrophage depletion and osteopontin inhibition decrease recurrent tumor growth. Furthermore, targeting osteopontin in primary tumor-bearing female mice prevents metastasis, permits T cell infiltration and activation, and improves anti-PD-1 immunotherapy response. Clinically, osteopontin expression is higher in recurrent metastatic tumors versus female patient-matched primary breast tumors. Osteopontin positively correlates with macrophage infiltration, increases with higher tumor grade, and its elevated pathway activity is associated with poor prognosis and long-term recurrence. Our findings suggest clinical implications and an alternative therapeutic strategy based on osteopontin's multiaxial role in breast cancer progression and recurrence.
Collapse
Affiliation(s)
- Yu Gu
- Rosalind and Morris Goodman Cancer Institute, McGill University, Montreal, QC, Canada
- Department of Biochemistry, Faculty of Medicine and Health Sciences, McGill University, Montreal, QC, Canada
| | - Tarek Taifour
- Rosalind and Morris Goodman Cancer Institute, McGill University, Montreal, QC, Canada
- Division of Experimental Medicine, Faculty of Medicine and Health Sciences, McGill University, Montreal, QC, Canada
| | - Tung Bui
- Rosalind and Morris Goodman Cancer Institute, McGill University, Montreal, QC, Canada
- Department of Biochemistry, Faculty of Medicine and Health Sciences, McGill University, Montreal, QC, Canada
| | - Dongmei Zuo
- Rosalind and Morris Goodman Cancer Institute, McGill University, Montreal, QC, Canada
| | - Alain Pacis
- Rosalind and Morris Goodman Cancer Institute, McGill University, Montreal, QC, Canada
- Canadian Centre for Computational Genomics, McGill University Genome Center, Montreal, QC, Canada
| | - Alexandre Poirier
- Rosalind and Morris Goodman Cancer Institute, McGill University, Montreal, QC, Canada
- Division of Experimental Medicine, Faculty of Medicine and Health Sciences, McGill University, Montreal, QC, Canada
| | - Sherif Attalla
- Rosalind and Morris Goodman Cancer Institute, McGill University, Montreal, QC, Canada
| | - Anne-Marie Fortier
- Rosalind and Morris Goodman Cancer Institute, McGill University, Montreal, QC, Canada
| | | | - Tien-Chi Pan
- Department of Cancer Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Vasilios Papavasiliou
- Rosalind and Morris Goodman Cancer Institute, McGill University, Montreal, QC, Canada
| | - Nancy U Lin
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Melissa E Hughes
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Kalie Smith
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Morag Park
- Rosalind and Morris Goodman Cancer Institute, McGill University, Montreal, QC, Canada
- Department of Biochemistry, Faculty of Medicine and Health Sciences, McGill University, Montreal, QC, Canada
- Faculty of Medicine, McGill University, Montreal, QC, Canada
| | - Michel L Tremblay
- Rosalind and Morris Goodman Cancer Institute, McGill University, Montreal, QC, Canada
- Department of Biochemistry, Faculty of Medicine and Health Sciences, McGill University, Montreal, QC, Canada
- Division of Experimental Medicine, Faculty of Medicine and Health Sciences, McGill University, Montreal, QC, Canada
- Faculty of Medicine, McGill University, Montreal, QC, Canada
| | - Lewis A Chodosh
- Department of Cancer Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Rinath Jeselsohn
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - William J Muller
- Rosalind and Morris Goodman Cancer Institute, McGill University, Montreal, QC, Canada.
- Department of Biochemistry, Faculty of Medicine and Health Sciences, McGill University, Montreal, QC, Canada.
- Faculty of Medicine, McGill University, Montreal, QC, Canada.
| |
Collapse
|
5
|
Abstract
PURPOSE Dental implant osseointegration comprises two types of bone formation-contact and distance osteogenesis-which result in bone formation originating from the implant surface or bone edges, respectively. The physicochemical properties of the implant surface regulate initial contact osteogenesis by directly tuning the osteoprogenitor cells in the peri-implant environment. However, whether these implant surface properties can regulate osteoprogenitor cells distant from the implant remains unclear. Innate immune cells, including neutrophils and macrophages, govern bone metabolism, suggesting their involvement in osseointegration and distance osteogenesis. This narrative review discusses the role of innate immunity in osseointegration and the effects of implant surface properties on distant osteogenesis, focusing on innate immune regulation. STUDY SELECTION The role of innate immunity in bone formation and the effects of implant surface properties on innate immune function were reviewed based on clinical, animal, and in vitro studies. RESULTS Neutrophils and macrophages are responsible for bone formation during osseointegration, via inflammatory mediators. The microroughness and hydrophilic status of titanium implants have the potential to alleviate this inflammatory response of neutrophils, and induce an anti-inflammatory response in macrophages, to tune both contact and distance osteogenesis through the activation of osteoblasts. Thus, the surface micro-roughness and hydrophilicity of implants can regulate the function of distant osteoprogenitor cells through innate immune cells. CONCLUSIONS Surface modification of implants aimed at regulating innate immunity may be useful in promoting further osteogenesis and overcoming the limitations encountered in severe situations, such as early loading protocol application.
Collapse
Affiliation(s)
- Takeru Kondo
- Division of Molecular and Regenerative Prosthodontics, Tohoku University Graduate School of Dentistry, Sendai, Japan
| | - Masahiro Yamada
- Division of Molecular and Regenerative Prosthodontics, Tohoku University Graduate School of Dentistry, Sendai, Japan
| | - Hiroshi Egusa
- Division of Molecular and Regenerative Prosthodontics, Tohoku University Graduate School of Dentistry, Sendai, Japan
| |
Collapse
|
6
|
Mameli E, Samantsidis GR, Viswanatha R, Kwon H, Hall DR, Butnaru M, Hu Y, Mohr SE, Perrimon N, Smith RC. A genome-wide CRISPR screen in Anopheles mosquito cells identifies essential genes and required components of clodronate liposome function. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.24.614595. [PMID: 39386635 PMCID: PMC11463579 DOI: 10.1101/2024.09.24.614595] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/12/2024]
Abstract
Anopheles mosquitoes are the sole vector of human malaria, the most burdensome vector-borne disease worldwide. Strategies aimed at reducing mosquito populations and limiting their ability to transmit disease show the most promise for disease control. Therefore, gaining an improved understanding of mosquito biology, and specifically that of the immune response, can aid efforts to develop new approaches that limit malaria transmission. Here, we use a genome-wide CRISPR screening approach for the first time in mosquito cells to identify essential genes in Anopheles and identify genes for which knockout confers resistance to clodronate liposomes, which have been widely used in mammals and arthropods to ablate immune cells. In the essential gene screen, we identified a set of 1280 Anopheles genes that are highly enriched for genes involved in fundamental cell processes. For the clodronate liposome screen, we identified several candidate resistance factors and confirm their roles in the uptake and processing of clodronate liposomes through in vivo validation in Anopheles gambiae, providing new mechanistic detail of phagolysosome formation and clodronate liposome function. In summary, we demonstrate the application of a genome-wide CRISPR knockout platform in a major malaria vector and the identification of genes that are important for fitness and immune-related processes.
Collapse
Affiliation(s)
- Enzo Mameli
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA, 02115, USA
| | - George-Rafael Samantsidis
- Department of Plant Pathology, Entomology and Microbiology, Iowa State University, Ames, IA 50011, USA
| | - Raghuvir Viswanatha
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA, 02115, USA
| | - Hyeogsun Kwon
- Department of Plant Pathology, Entomology and Microbiology, Iowa State University, Ames, IA 50011, USA
| | - David R. Hall
- Department of Plant Pathology, Entomology and Microbiology, Iowa State University, Ames, IA 50011, USA
| | - Matthew Butnaru
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA, 02115, USA
| | - Yanhui Hu
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA, 02115, USA
| | - Stephanie E. Mohr
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA, 02115, USA
| | - Norbert Perrimon
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA, 02115, USA
- HHMI, Harvard Medical School, Boston, MA, 02115, USA
| | - Ryan C. Smith
- Department of Plant Pathology, Entomology and Microbiology, Iowa State University, Ames, IA 50011, USA
| |
Collapse
|
7
|
Szentirmai É, Buckley K, Kapás L. The role of Kupffer cells in microbiota-brain communication: Sleep and fever signaling in response to lipopolysaccharide. Brain Behav Immun 2024; 123:306-314. [PMID: 39322087 DOI: 10.1016/j.bbi.2024.09.028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Revised: 09/04/2024] [Accepted: 09/21/2024] [Indexed: 09/27/2024] Open
Abstract
Microbial molecules translocated from the intestinal lumen into the host's internal environment play a role in various physiological functions. Previously, we identified that butyrate, a short-chain fatty acid produced by intestinal bacteria, lipoteichoic acid, a cell wall component of gram-positive bacteria, and lipopolysaccharide (LPS), a cell wall component of gram-negative bacteria, induce sleep when their naturally occurring translocation is mimicked by direct delivery into the portal vein. Our findings suggested that these microbial molecules exert their sleep-promoting effects within the hepatoportal region. In the present experiments, we tested the hypothesis that resident liver macrophages, known as Kupffer cells, play a crucial role in the LPS-responsive, sleep-promoting mechanisms within the hepatoportal region. Intraportal administration of LPS induced increased sleep and fever in control rats. Remarkably, in Kupffer cell-depleted animals, both of these responses were significantly suppressed. These findings highlight the potential role of Kupffer cells in mediating the non-rapid-eye movement sleep-promoting and febrile effects of LPS translocated from the intestinal microbiota into the portal circulation. The strategic location of Kupffer cells within the hepatoportal region, coupled with their ability to rapidly take up LPS and other microbial molecules, together with their high secretory activity of multiple signaling molecules, underlie their key role in the communication between the intestinal microbiota and the brain.
Collapse
Affiliation(s)
- Éva Szentirmai
- Elson S. Floyd College of Medicine, Department of Translational Medicine and Physiology, Washington State University, Spokane, WA, United States of America; Sleep and Performance Research Center, Washington State University, Spokane, WA, USA.
| | - Katelin Buckley
- Elson S. Floyd College of Medicine, Department of Translational Medicine and Physiology, Washington State University, Spokane, WA, United States of America
| | - Levente Kapás
- Elson S. Floyd College of Medicine, Department of Translational Medicine and Physiology, Washington State University, Spokane, WA, United States of America; Sleep and Performance Research Center, Washington State University, Spokane, WA, USA
| |
Collapse
|
8
|
Liu J, Cui Y, Cabral H, Tong A, Yue Q, Zhao L, Sun X, Mi P. Glucosylated Nanovaccines for Dendritic Cell-Targeted Antigen Delivery and Amplified Cancer Immunotherapy. ACS NANO 2024; 18:25826-25840. [PMID: 39196858 DOI: 10.1021/acsnano.4c09053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/30/2024]
Abstract
Engineering nanovaccines capable of targeting dendritic cells (DCs) is desperately required to maximize antigen cross-presentation to effector immune cells, elicit strong immune responses, and avoid adverse reactions. Here, we showed that glucose transporter 1 (Glut-1) on DCs is a reliable target for delivering antigens to DCs, and thus, a versatile antigen delivery strategy using glucosylated nanovaccines was developed for DC-targeted antigen delivery and tumor immunotherapy. The developed glucosylated ovalbumin-loaded nanovaccines highly accumulated in lymph nodes and efficiently engaged with Glut-1 on DCs to accelerate intracellular antigen delivery and promote DC maturation and antigen presentation, which elicited potent antitumor immunity to prevent and inhibit ovalbumin-expressing melanoma. Moreover, immunotherapeutic experiments in DC- and macrophage-depleted animal models confirmed that the glucosylated nanovaccines functioned mainly through DCs. In addition, the neoantigen-delivering glucosylated nanovaccines were further engineered to elicit tumor-specific immune responses against MC38 tumors. This study offers a DC-targeted antigen delivery strategy for cancer immunotherapy.
Collapse
Affiliation(s)
- Jing Liu
- Department of Radiology, State Key Laboratory of Biotherapy and Huaxi MR Research Center (HMRRC), West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Yongsheng Cui
- Department of Radiology, State Key Laboratory of Biotherapy and Huaxi MR Research Center (HMRRC), West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Horacio Cabral
- Department of Bioengineering, Graduate School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan
| | - Aiping Tong
- Department of Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Qiang Yue
- Department of Radiology, State Key Laboratory of Biotherapy and Huaxi MR Research Center (HMRRC), West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Lihong Zhao
- Department of Radiology, State Key Laboratory of Biotherapy and Huaxi MR Research Center (HMRRC), West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Xun Sun
- Department of Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China
- Key Laboratory of Drug Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Peng Mi
- Department of Radiology, State Key Laboratory of Biotherapy and Huaxi MR Research Center (HMRRC), West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| |
Collapse
|
9
|
Gao S, Li W, Huang Z, Deiuliis JA, Braunstein Z, Liu X, Li X, Kosari M, Chen J, Min X, Yang H, Gong Q, Liu Z, Wei Y, Zhang Z, Dong L, Zhong J. Deciphering the therapeutic potential of Myeloid-Specific JAK2 inhibition in acute respiratory distress syndrome. Mucosal Immunol 2024:S1933-0219(24)00089-8. [PMID: 39173745 DOI: 10.1016/j.mucimm.2024.08.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Revised: 08/09/2024] [Accepted: 08/16/2024] [Indexed: 08/24/2024]
Abstract
Acute respiratory distress syndrome (ARDS) is a life-threatening condition characterized by severe inflammation and pulmonary dysfunction. Despite advancements in critical care, effective pharmacological interventions for ARDS remain elusive. While Janus kinase 2 (JAK2) inhibitors have emerged as an innovative treatment for numerous autoinflammatory diseases, their therapeutic potential in ARDS remains unexplored. In this study, we investigated the contribution of JAK2 and its underlying mechanisms in ARDS utilizing myeloid-specific JAK2 knockout murine models alongside a pharmacological JAK2 inhibitor. Notably, myeloid-specific JAK2 knockout led to a notable attenuation of ARDS induced by intratracheal administration of LPS, accompanied by reduced levels of neutrophils and inflammatory cytokines in bronchoalveolar lavage fluid (BALF) and lung tissue. Intriguingly, the ameliorative effects were abolished upon the depletion of monocyte-derived alveolar macrophages (Mo-AMs) rather than tissue-resident alveolar macrophages (TR-AMs). JAK2 deficiency markedly reversed LPS-induced activation of STAT5 in macrophages. Remarkably, pharmacological JAK2 inhibition using baricitinib failed to substantially alleviate neutrophils infiltration, implying that specific inhibition of JAK2 in Mo-AMs is imperative for ARDS amelioration. Collectively, our data suggest that JAK2 may mitigate ARDS progression through the JAK2 pathway in Mo-AMs, underscoring JAK2 in alveolar macrophages, particularly Mo-AMs, as a promising therapeutic target for ARDS treatment.
Collapse
Affiliation(s)
- Shupei Gao
- Department of Rheumatology and Immunology, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Wenjuan Li
- Department of Rheumatology and Immunology, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Zhiwen Huang
- Department of Rheumatology and Immunology, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Jeffrey A Deiuliis
- Cardiovascular Research Institute, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Zachary Braunstein
- Wexner Medical Center, The Ohio State University, Columbus, OH 43210, USA
| | - Xinxin Liu
- Department of Rheumatology and Immunology, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Xinlu Li
- Department of Rheumatology and Immunology, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Mohammadreza Kosari
- Department of Rheumatology and Immunology, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Jun Chen
- Sinopharm Dongfeng General Hospital, Hubei University of Medicine, Hubei Key Laboratory of Wudang Local Chinese Medicine Research (Hubei University of Medicine), Shiyan, Hubei 442008, China
| | - Xinwen Min
- Sinopharm Dongfeng General Hospital, Hubei University of Medicine, Hubei Key Laboratory of Wudang Local Chinese Medicine Research (Hubei University of Medicine), Shiyan, Hubei 442008, China
| | - Handong Yang
- Sinopharm Dongfeng General Hospital, Hubei University of Medicine, Hubei Key Laboratory of Wudang Local Chinese Medicine Research (Hubei University of Medicine), Shiyan, Hubei 442008, China
| | - Quan Gong
- Department of Immunology, School of Medicine, Yangtze University, Jingzhou, Hubei 434023, China
| | - Zheng Liu
- Institute of Allergy and Clinical Immunology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China; Department of Otolaryngology-Head and Neck Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Yingying Wei
- Department of Rheumatology and Immunology, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Ziyang Zhang
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Qiaokou District, Wuhan 430030, China
| | - Lingli Dong
- Department of Rheumatology and Immunology, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China.
| | - Jixin Zhong
- Department of Rheumatology and Immunology, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China; Cardiovascular Research Institute, Case Western Reserve University, Cleveland, OH 44106, USA; Institute of Allergy and Clinical Immunology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China; Key Laboratory of Vascular Aging (HUST), Ministry of Education, Wuhan, Hubei 430030, China.
| |
Collapse
|
10
|
Jiang Y, Wang Y, Chen G, Sun F, Wu Q, Huang Q, Zeng D, Qiu W, Wang J, Yao Z, Liang B, Li S, Wu J, Huang N, Wang Y, Chen J, Zhai X, Huang L, Xu B, Yamamoto M, Tsukamoto T, Nomura S, Liao W, Shi M. Nicotinamide metabolism face-off between macrophages and fibroblasts manipulates the microenvironment in gastric cancer. Cell Metab 2024; 36:1806-1822.e11. [PMID: 38897198 DOI: 10.1016/j.cmet.2024.05.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Revised: 03/06/2024] [Accepted: 05/22/2024] [Indexed: 06/21/2024]
Abstract
Immune checkpoint blockade has led to breakthroughs in the treatment of advanced gastric cancer. However, the prominent heterogeneity in gastric cancer, notably the heterogeneity of the tumor microenvironment, highlights the idea that the antitumor response is a reflection of multifactorial interactions. Through transcriptomic analysis and dynamic plasma sample analysis, we identified a metabolic "face-off" mechanism within the tumor microenvironment, as shown by the dual prognostic significance of nicotinamide metabolism. Specifically, macrophages and fibroblasts expressing the rate-limiting enzymes nicotinamide phosphoribosyltransferase and nicotinamide N-methyltransferase, respectively, regulate the nicotinamide/1-methylnicotinamide ratio and CD8+ T cell function. Mechanistically, nicotinamide N-methyltransferase is transcriptionally activated by the NOTCH pathway transcription factor RBP-J and is further inhibited by macrophage-derived extracellular vesicles containing nicotinamide phosphoribosyltransferase via the SIRT1/NICD axis. Manipulating nicotinamide metabolism through autologous injection of extracellular vesicles restored CD8+ T cell cytotoxicity and the anti-PD-1 response in gastric cancer.
Collapse
Affiliation(s)
- Yu Jiang
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Yawen Wang
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Guofeng Chen
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Fei Sun
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Qijing Wu
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Qiong Huang
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Dongqiang Zeng
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Wenjun Qiu
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Jiao Wang
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Zhiqi Yao
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Bishan Liang
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Shaowei Li
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Jianhua Wu
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Na Huang
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Yuanyuan Wang
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Jingsong Chen
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Xiaohui Zhai
- Department of Medical Oncology, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Li Huang
- Department of Oncology, First Affiliated Hospital, Gannan Medical University, Ganzhou, China; Jiangxi Clinical Medical Research Center for Cancer, Ganzhou, China
| | - Beibei Xu
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Masami Yamamoto
- Laboratory of Physiological Pathology, School of Veterinary Nursing and Technology, Nippon Veterinary and Life Science University, Tokyo, Japan
| | - Tetsuya Tsukamoto
- Department of Diagnostic Pathology, Fujita Health University School of Medicine, Toyoake, Aichi, Japan
| | - Sachiyo Nomura
- Department of Gastrointestinal Surgery, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Wangjun Liao
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China; Cancer Center, the Sixth Affiliated Hospital, School of Medicine, South China University of Technology, Foshan, China.
| | - Min Shi
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China.
| |
Collapse
|
11
|
Li L, Zhai M, Cheng C, Cui S, Wang J, Zhang Z, Liu J, Wei F. Mechanically induced M2 macrophages are involved in bone remodeling of the midpalatal suture during palatal expansion. Prog Orthod 2024; 25:30. [PMID: 39098934 PMCID: PMC11298508 DOI: 10.1186/s40510-024-00529-z] [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: 08/02/2023] [Accepted: 05/30/2024] [Indexed: 08/06/2024] Open
Abstract
BACKGROUND Palatal expansion is a common way of treating maxillary transverse deficiency. Under mechanical force, the midpalatal suture is expanded, causing local immune responses. This study aimed to determine whether macrophages participate in bone remodeling of the midpalatal suture during palatal expansion and the effects on bone remodeling. METHODS Palatal expansion model and macrophage depletion model were established. Micro-CT, histological staining, and immunohistochemical staining were used to investigate the changes in the number and phenotype of macrophages during palatal expansion as well as the effects on bone remodeling of the midpalatal suture. Additionally, the effect of mechanically induced M2 macrophages on palatal osteoblasts was also elucidated in vitro. RESULTS The number of macrophages increased significantly and polarized toward M2 phenotype with the increase of the expansion time, which was consistent with the trend of bone remodeling. After macrophage depletion, the function of osteoblasts and bone formation at the midpalatal suture were impaired during palatal expansion. In vitro, conditioned medium derived from M2 macrophages facilitated osteogenic differentiation of osteoblasts and decreased the RANKL/OPG ratio. CONCLUSIONS Macrophages through polarizing toward M2 phenotype participated in midpalatal suture bone remodeling during palatal expansion, which may provide a new idea for promoting bone remodeling from the perspective of regulating macrophage polarization.
Collapse
Affiliation(s)
- Lan Li
- Department of Orthodontics, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, Shandong University, No.44-1 Wenhua Road West, Shandong, Jinan, 250012, China
| | - Mingrui Zhai
- Department of Orthodontics, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, Shandong University, No.44-1 Wenhua Road West, Shandong, Jinan, 250012, China
| | - Chen Cheng
- Department of Orthodontics, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, Shandong University, No.44-1 Wenhua Road West, Shandong, Jinan, 250012, China
| | - Shuyue Cui
- Department of Orthodontics, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, Shandong University, No.44-1 Wenhua Road West, Shandong, Jinan, 250012, China
| | - Jixiao Wang
- Department of Orthodontics, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, Shandong University, No.44-1 Wenhua Road West, Shandong, Jinan, 250012, China
| | - Zijie Zhang
- Department of Orthodontics, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, Shandong University, No.44-1 Wenhua Road West, Shandong, Jinan, 250012, China
| | - Jiani Liu
- Department of Orthodontics, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, Shandong University, No.44-1 Wenhua Road West, Shandong, Jinan, 250012, China
| | - Fulan Wei
- Department of Orthodontics, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, Shandong University, No.44-1 Wenhua Road West, Shandong, Jinan, 250012, China.
| |
Collapse
|
12
|
Cheng Z, Fobian SF, Gurrieri E, Amin M, D'Agostino VG, Falahati M, Zalba S, Debets R, Garrido MJ, Saeed M, Seynhaeve ALB, Balcioglu HE, Ten Hagen TLM. Lipid-based nanosystems: the next generation of cancer immune therapy. J Hematol Oncol 2024; 17:53. [PMID: 39030582 PMCID: PMC11265205 DOI: 10.1186/s13045-024-01574-1] [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: 05/23/2024] [Accepted: 07/11/2024] [Indexed: 07/21/2024] Open
Abstract
Immunotherapy has become an important part of the oncotherapy arsenal. Its applicability in various cancer types is impressive, as well as its use of endogenous mechanisms to achieve desired ends. However, off-target or on-target-off-tumor toxicity, limited activity, lack of control in combination treatments and, especially for solid tumors, low local accumulation, have collectively limited clinical use thereof. These limitations are partially alleviated by delivery systems. Lipid-based nanoparticles (NPs) have emerged as revolutionary carriers due to favorable physicochemical characteristics, with specific applications and strengths particularly useful in immunotherapeutic agent delivery. The aim of this review is to highlight the challenges faced by immunotherapy and how lipid-based NPs have been, and may be further utilized to address such challenges. We discuss recent fundamental and clinical applications of NPs in a range of areas and provide a detailed discussion of the main obstacles in immune checkpoint inhibition therapies, adoptive cellular therapies, and cytokine therapies. We highlight how lipid-based nanosystems could address these through either delivery, direct modulation of the immune system, or targeting of the immunosuppressive tumor microenvironment. We explore advanced and emerging liposomal and lipid nanoparticle (LNP) systems for nucleic acid delivery, intrinsic and extrinsic stimulus-responsive formulations, and biomimetic lipid-based nanosystems in immunotherapy. Finally, we discuss the key challenges relating to the clinical use of lipid-based NP immunotherapies, suggesting future research directions for the near term to realize the potential of these innovative lipid-based nanosystems, as they become the crucial steppingstone towards the necessary enhancement of the efficacy of immunotherapy.
Collapse
Affiliation(s)
- Ziyun Cheng
- Precision Medicine in Oncology (PrMiO), Department of Pathology, Erasmus MC Cancer Institute, Erasmus Medical Center, Rotterdam, The Netherlands
- Nanomedicine Innovation Center Erasmus (NICE), Erasmus Medical Center, Rotterdam, The Netherlands
| | - Seth-Frerich Fobian
- Precision Medicine in Oncology (PrMiO), Department of Pathology, Erasmus MC Cancer Institute, Erasmus Medical Center, Rotterdam, The Netherlands
- Nanomedicine Innovation Center Erasmus (NICE), Erasmus Medical Center, Rotterdam, The Netherlands
| | - Elena Gurrieri
- Laboratory of Biotechnology and Nanomedicine, Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, Trento, Italy
| | - Mohamadreza Amin
- Precision Medicine in Oncology (PrMiO), Department of Pathology, Erasmus MC Cancer Institute, Erasmus Medical Center, Rotterdam, The Netherlands
- Nanomedicine Innovation Center Erasmus (NICE), Erasmus Medical Center, Rotterdam, The Netherlands
| | - Vito Giuseppe D'Agostino
- Laboratory of Biotechnology and Nanomedicine, Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, Trento, Italy
| | - Mojtaba Falahati
- Precision Medicine in Oncology (PrMiO), Department of Pathology, Erasmus MC Cancer Institute, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Sara Zalba
- Department of Pharmaceutical Sciences, School of Pharmacy and Nutrition, University of Navarra, Pamplona, Spain
- Instituto de Investigación Sanitaria de Navarra (IdiSNA), Navarra Institute for Health Research, Pamplona, Spain
| | - Reno Debets
- Laboratory of Tumor Immunology, Department of Medical Oncology, Erasmus MC Cancer Institute, Erasmus Medical Center, Rotterdam, The Netherlands
| | - María J Garrido
- Department of Pharmaceutical Sciences, School of Pharmacy and Nutrition, University of Navarra, Pamplona, Spain
- Instituto de Investigación Sanitaria de Navarra (IdiSNA), Navarra Institute for Health Research, Pamplona, Spain
| | - Mesha Saeed
- Precision Medicine in Oncology (PrMiO), Department of Pathology, Erasmus MC Cancer Institute, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Ann L B Seynhaeve
- Precision Medicine in Oncology (PrMiO), Department of Pathology, Erasmus MC Cancer Institute, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Hayri E Balcioglu
- Laboratory of Tumor Immunology, Department of Medical Oncology, Erasmus MC Cancer Institute, Erasmus Medical Center, Rotterdam, The Netherlands.
| | - Timo L M Ten Hagen
- Precision Medicine in Oncology (PrMiO), Department of Pathology, Erasmus MC Cancer Institute, Erasmus Medical Center, Rotterdam, The Netherlands.
- Nanomedicine Innovation Center Erasmus (NICE), Erasmus Medical Center, Rotterdam, The Netherlands.
| |
Collapse
|
13
|
Parada-Kusz M, Clatworthy AE, Goering ER, Blackwood SM, Shigeta JY, Mashin E, Salm EJ, Choi C, Combs S, Lee JSW, Rodriguez-Osorio C, Clish C, Tomita S, Hung DT. 3-Hydroxykynurenine targets kainate receptors to promote defense against infection. Nat Chem Biol 2024:10.1038/s41589-024-01635-z. [PMID: 38898166 DOI: 10.1038/s41589-024-01635-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Accepted: 05/07/2024] [Indexed: 06/21/2024]
Abstract
Bacterial infection involves a complex interaction between the pathogen and host where the outcome of infection is not solely determined by pathogen eradication. To identify small molecules that promote host survival by altering the host-pathogen dynamic, we conducted an in vivo chemical screen using zebrafish embryos and found that treatment with 3-hydroxykynurenine (3-HK) protects from lethal bacterial infection. 3-HK, a metabolite produced through host tryptophan metabolism, has no direct antibacterial activity but enhances host survival by restricting bacterial expansion in macrophages through a systemic mechanism that targets kainate-sensitive glutamate receptors. These findings reveal a new pathway by which tryptophan metabolism and kainate-sensitive glutamate receptors function and interact to modulate immunity, with important implications for the coordination between the immune and nervous systems in pathological conditions.
Collapse
Affiliation(s)
- Margarita Parada-Kusz
- Department of Molecular Biology and Center for Computational and Integrative Biology, Massachusetts General Hospital (MGH), Boston, MA, USA
- Department of Genetics, Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Anne E Clatworthy
- Department of Molecular Biology and Center for Computational and Integrative Biology, Massachusetts General Hospital (MGH), Boston, MA, USA.
- Department of Genetics, Harvard Medical School, Boston, MA, USA.
- Broad Institute of MIT and Harvard, Cambridge, MA, USA.
| | - Emily R Goering
- Department of Molecular Biology and Center for Computational and Integrative Biology, Massachusetts General Hospital (MGH), Boston, MA, USA
- Department of Genetics, Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Stephanie M Blackwood
- Department of Molecular Biology and Center for Computational and Integrative Biology, Massachusetts General Hospital (MGH), Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Jack Y Shigeta
- Department of Molecular Biology and Center for Computational and Integrative Biology, Massachusetts General Hospital (MGH), Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | | | - Elizabeth J Salm
- Department of Cellular and Molecular Physiology and Neuroscience, Yale School of Medicine, New Haven, CT, USA
| | - Catherine Choi
- Department of Molecular Biology and Center for Computational and Integrative Biology, Massachusetts General Hospital (MGH), Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Senya Combs
- Department of Molecular Biology and Center for Computational and Integrative Biology, Massachusetts General Hospital (MGH), Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Jenny S W Lee
- Department of Molecular Biology and Center for Computational and Integrative Biology, Massachusetts General Hospital (MGH), Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Carlos Rodriguez-Osorio
- Department of Molecular Biology and Center for Computational and Integrative Biology, Massachusetts General Hospital (MGH), Boston, MA, USA
- Department of Genetics, Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Clary Clish
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Susumu Tomita
- Department of Cellular and Molecular Physiology and Neuroscience, Yale School of Medicine, New Haven, CT, USA
| | - Deborah T Hung
- Department of Molecular Biology and Center for Computational and Integrative Biology, Massachusetts General Hospital (MGH), Boston, MA, USA.
- Department of Genetics, Harvard Medical School, Boston, MA, USA.
- Broad Institute of MIT and Harvard, Cambridge, MA, USA.
| |
Collapse
|
14
|
Li Z, Duan D, Li L, Peng D, Ming Y, Ni R, Liu Y. Tumor-associated macrophages in anti-PD-1/PD-L1 immunotherapy for hepatocellular carcinoma: recent research progress. Front Pharmacol 2024; 15:1382256. [PMID: 38957393 PMCID: PMC11217528 DOI: 10.3389/fphar.2024.1382256] [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: 02/05/2024] [Accepted: 05/22/2024] [Indexed: 07/04/2024] Open
Abstract
Hepatocellular carcinoma (HCC) is one of the cancers that seriously threaten human health. Immunotherapy serves as the mainstay of treatment for HCC patients by targeting the programmed cell death protein 1/programmed cell death 1 ligand 1 (PD-1/PD-L1) axis. However, the effectiveness of anti-PD-1/PD-L1 treatment is limited when HCC becomes drug-resistant. Tumor-associated macrophages (TAMs) are an important factor in the negative regulation of PD-1 antibody targeted therapy in the tumor microenvironment (TME). Therefore, as an emerging direction in cancer immunotherapy research for the treatment of HCC, it is crucial to elucidate the correlations and mechanisms between TAMs and PD-1/PD-L1-mediated immune tolerance. This paper summarizes the effects of TAMs on the pathogenesis and progression of HCC and their impact on HCC anti-PD-1/PD-L1 immunotherapy, and further explores current potential therapeutic strategies that target TAMs in HCC, including eliminating TAMs in the TME, inhibiting TAMs recruitment to tumors and functionally repolarizing M2-TAMs (tumor-supportive) to M1-TAMs (antitumor type).
Collapse
Affiliation(s)
| | | | | | | | | | - Rui Ni
- Department of Pharmacy, Daping Hospital, Army Medical University, Chongqing, China
| | - Yao Liu
- Department of Pharmacy, Daping Hospital, Army Medical University, Chongqing, China
| |
Collapse
|
15
|
Zhong H, Zhou S, Yin S, Qiu Y, Liu B, Yu H. Tumor microenvironment as niche constructed by cancer stem cells: Breaking the ecosystem to combat cancer. J Adv Res 2024:S2090-1232(24)00251-0. [PMID: 38866179 DOI: 10.1016/j.jare.2024.06.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Revised: 05/27/2024] [Accepted: 06/09/2024] [Indexed: 06/14/2024] Open
Abstract
BACKGROUND Cancer stem cells (CSCs) are a distinct subpopulation of cancer cells with the capacity to constantly self-renew and differentiate, and they are the main driver in the progression of cancer resistance and relapse. The tumor microenvironment (TME) constructed by CSCs is the "soil" adapted to tumor growth, helping CSCs evade immune killing, enhance their chemical resistance, and promote cancer progression. AIM OF REVIEW We aim to elaborate the tight connection between CSCs and immunosuppressive components of the TME. We attempt to summarize and provide a therapeutic strategy to eradicate CSCs based on the destruction of the tumor ecological niche. KEY SCIENTIFIC CONCEPTS OF REVIEW This review is focused on three main key concepts. First, we highlight that CSCs recruit and transform normal cells to construct the TME, which further provides ecological niche support for CSCs. Second, we describe the main characteristics of the immunosuppressive components of the TME, targeting strategies and summarize the progress of corresponding drugs in clinical trials. Third, we explore the multilevel insights of the TME to serve as an ecological niche for CSCs.
Collapse
Affiliation(s)
- Hao Zhong
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China; Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, China; Haihe Laboratory of Modern Chinese Medicine, Tianjin, China
| | - Shiyue Zhou
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China; Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, China; Haihe Laboratory of Modern Chinese Medicine, Tianjin, China
| | - Shuangshuang Yin
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China; Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, China; Haihe Laboratory of Modern Chinese Medicine, Tianjin, China
| | - Yuling Qiu
- School of Pharmacy, Tianjin Medical University, Tianjin, China.
| | - Bo Liu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, China.
| | - Haiyang Yu
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China; Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, China; Haihe Laboratory of Modern Chinese Medicine, Tianjin, China.
| |
Collapse
|
16
|
Collins BC, Shapiro JB, Scheib MM, Musci RV, Verma M, Kardon G. Three-dimensional imaging studies in mice identify cellular dynamics of skeletal muscle regeneration. Dev Cell 2024; 59:1457-1474.e5. [PMID: 38569550 PMCID: PMC11153043 DOI: 10.1016/j.devcel.2024.03.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Revised: 12/06/2023] [Accepted: 03/08/2024] [Indexed: 04/05/2024]
Abstract
The function of many organs, including skeletal muscle, depends on their three-dimensional structure. Muscle regeneration therefore requires not only reestablishment of myofibers but also restoration of tissue architecture. Resident muscle stem cells (SCs) are essential for regeneration, but how SCs regenerate muscle architecture is largely unknown. We address this problem using genetic labeling of mouse SCs and whole-mount imaging to reconstruct, in three dimensions, muscle regeneration. Unexpectedly, we found that myofibers form via two distinct phases of fusion and the residual basement membrane of necrotic myofibers is critical for promoting fusion and orienting regenerated myofibers. Furthermore, the centralized myonuclei characteristic of regenerated myofibers are associated with myofibrillogenesis and endure months post injury. Finally, we elucidate two cellular mechanisms for the formation of branched myofibers, a pathology characteristic of diseased muscle. We provide a synthesis of the cellular events of regeneration and show that these differ from those used during development.
Collapse
Affiliation(s)
- Brittany C Collins
- Department of Human Genetics, University of Utah, Salt Lake City, UT, USA
| | - Jacob B Shapiro
- Department of Human Genetics, University of Utah, Salt Lake City, UT, USA
| | - Mya M Scheib
- Department of Human Genetics, University of Utah, Salt Lake City, UT, USA
| | - Robert V Musci
- Department of Human Genetics, University of Utah, Salt Lake City, UT, USA
| | - Mayank Verma
- Department of Pediatrics, Division of Neurology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Gabrielle Kardon
- Department of Human Genetics, University of Utah, Salt Lake City, UT, USA.
| |
Collapse
|
17
|
Kelly LE, El-Hodiri HM, Crider A, Fischer AJ. Protein phosphatases regulate the formation of Müller glia-derived progenitor cells in the chick retina. Mol Cell Neurosci 2024; 129:103932. [PMID: 38679247 PMCID: PMC11362962 DOI: 10.1016/j.mcn.2024.103932] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Revised: 03/26/2024] [Accepted: 04/18/2024] [Indexed: 05/01/2024] Open
Abstract
Different kinase-dependent cell signaling pathways are known to play important roles in glia-mediated neuroprotection and reprogramming of Müller glia (MG) into Müller glia-derived progenitor cells (MGPCs) in the retina. However, very little is known about the phosphatases that regulate kinase-dependent signaling in MG. Using single-cell RNA-sequencing (scRNA-seq) databases, we investigated patterns of expression of Dual Specificity Phosphatases (DUSP1/6) and other protein phosphatases in normal and damaged chick retinas. We found that DUSP1, DUSP6, PPP3CB, PPP3R1 and PPPM1A/B/D/E/G are widely expressed by many types of retinal neurons and are dynamically expressed by MG and MGPCs in retinas during the process of reprogramming. We find that inhibition of DUSP1/6 and PP2C phosphatases enhances the formation of proliferating MGPCs in damaged retinas and in retinas treated with insulin and FGF2 in the absence of damage. By contrast, inhibition of PP2B phosphatases suppressed the formation of proliferating MGPCs, but increased numbers of proliferating MGPCs in undamaged retinas treated with insulin and FGF2. In damaged retinas, inhibition of DUSP1/6 increased levels of pERK1/2 and cFos in MG whereas inhibition of PP2B's decreased levels of pStat3 and pS6 in MG. Analyses of scRNA-seq libraries identified numerous differentially activated gene modules in MG in damaged retinas versus MG in retinas treated with insulin+FGF2 suggesting significant differences in kinase-dependent signaling pathways that converge on the formation of MGPCs. Inhibition of phosphatases had no significant effects upon numbers of dying cells in damaged retinas. We conclude that the activity of different protein phosphatases acting through retinal neurons and MG "fine-tune" the cell signaling responses of MG in damaged retinas and during the reprogramming of MG into MGPCs.
Collapse
Affiliation(s)
- Lisa E Kelly
- Department of Neuroscience, College of Medicine, The Ohio State University, Columbus, OH, USA
| | - Heithem M El-Hodiri
- Department of Neuroscience, College of Medicine, The Ohio State University, Columbus, OH, USA
| | - Andrew Crider
- Department of Neuroscience, College of Medicine, The Ohio State University, Columbus, OH, USA
| | - Andy J Fischer
- Department of Neuroscience, College of Medicine, The Ohio State University, Columbus, OH, USA.
| |
Collapse
|
18
|
Talsma AD, Niemi JP, Zigmond RE. Neither injury induced macrophages within the nerve, nor the environment created by Wallerian degeneration is necessary for enhanced in vivo axon regeneration after peripheral nerve injury. J Neuroinflammation 2024; 21:134. [PMID: 38802868 PMCID: PMC11131297 DOI: 10.1186/s12974-024-03132-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Accepted: 05/13/2024] [Indexed: 05/29/2024] Open
Abstract
BACKGROUND Since the 1990s, evidence has accumulated that macrophages promote peripheral nerve regeneration and are required for enhancing regeneration in the conditioning lesion (CL) response. After a sciatic nerve injury, macrophages accumulate in the injury site, the nerve distal to that site, and the axotomized dorsal root ganglia (DRGs). In the peripheral nervous system, as in other tissues, the macrophage response is derived from both resident macrophages and recruited monocyte-derived macrophages (MDMs). Unresolved questions are: at which sites do macrophages enhance nerve regeneration, and is a particular population needed. METHODS Ccr2 knock-out (KO) and Ccr2gfp/gfp knock-in/KO mice were used to prevent MDM recruitment. Using these strains in a sciatic CL paradigm, we examined the necessity of MDMs and residents for CL-enhanced regeneration in vivo and characterized injury-induced nerve inflammation. CL paradigm variants, including the addition of pharmacological macrophage depletion methods, tested the role of various macrophage populations in initiating or sustaining the CL response. In vivo regeneration, measured from bilateral proximal test lesions (TLs) after 2 d, and macrophages were quantified by immunofluorescent staining. RESULTS Peripheral CL-enhanced regeneration was equivalent between crush and transection CLs and was sustained for 28 days in both Ccr2 KO and WT mice despite MDM depletion. Similarly, the central CL response measured in dorsal roots was unchanged in Ccr2 KO mice. Macrophages at both the TL and CL, but not between them, stained for the pro-regenerative marker, arginase 1. TL macrophages were primarily CCR2-dependent MDMs and nearly absent in Ccr2 KO and Ccr2gfp/gfp KO mice. However, there were only slightly fewer Arg1+ macrophages in CCR2 null CLs than controls due to resident macrophage compensation. Zymosan injection into an intact WT sciatic nerve recruited Arg1+ macrophages but did not enhance regeneration. Finally, clodronate injection into Ccr2gfp KO CLs dramatically reduced CL macrophages. Combined with the Ccr2gfp KO background, depleting MDMs and TL macrophages, and a transection CL, physically removing the distal nerve environment, nearly all macrophages in the nerve were removed, yet CL-enhanced regeneration was not impaired. CONCLUSIONS Macrophages in the sciatic nerve are neither necessary nor sufficient to produce a CL response.
Collapse
Affiliation(s)
- Aaron D Talsma
- Department of Neurosciences, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, OH, 44106-4975, USA
| | - Jon P Niemi
- Department of Neurosciences, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, OH, 44106-4975, USA
| | - Richard E Zigmond
- Department of Neurosciences, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, OH, 44106-4975, USA.
| |
Collapse
|
19
|
Zhang L, Tao M, Zhang H, Zhang S, Hou X, Zong C, Sun G, Feng S, Yan H, Lu Y, Yang X, Wei L, Zhang L. Lipopolysaccharide modification enhances the inhibitory effect of clodronate liposomes on hepatic fibrosis by depletion of macrophages and hepatic stellate cells. Chem Biol Interact 2024; 395:111015. [PMID: 38663797 DOI: 10.1016/j.cbi.2024.111015] [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: 12/13/2023] [Revised: 04/15/2024] [Accepted: 04/21/2024] [Indexed: 05/01/2024]
Abstract
Hepatic fibrosis is a complex chronic liver disease in which both macrophages and hepatic stellate cells (HSCs) play important roles. Many studies have shown that clodronate liposomes (CLD-lipos) effectively deplete macrophages. However, no liposomes have been developed that target both HSCs and macrophages. This study aimed to evaluate the therapeutic efficacy of lipopolysaccharide-coupled clodronate liposomes (LPS-CLD-lipos) and the effects of liposomes size on hepatic fibrosis. Three rat models of hepatic fibrosis were established in vivo; diethylnitrosamine (DEN), bile duct ligation (BDL), and carbon tetrachloride (CCl4). Hematoxylin and eosin staining and serological liver function indices were used to analyze pathological liver damage. Masson's trichrome and Sirius red staining were used to evaluate the effect of liposomes on liver collagen fibers. The hydroxyproline content in liver tissues was determined. In vitro cell counting kit-8 (CCK-8) and immunofluorescence assays were used to further explore the effects of LPS modification and liposomes size on the killing of macrophages and HSCs. Both in vitro and in vivo experiments showed that 200 nm LPS-CLD-lipos significantly inhibited hepatic fibrosis and the abnormal deposition of collagen fibers in the liver and improved the related indicators of liver function. Further results showed that 200 nm LPS-CLD-lipos increased the clearance of macrophages and induced apoptosis of hepatic stellate cells, significantly. The present study demonstrated that 200 nm LPS-CLD-lipos could significantly inhibit hepatic fibrosis and improve liver function-related indices and this study may provide novel ideas and directions for hepatic fibrosis treatment.
Collapse
Affiliation(s)
- Luyao Zhang
- School of Pharmacy, Anhui Medical University, Hefei, China; Department of Clinical Pharmacology, The Second Hospital of Anhui Medical University, Hefei, China; Clinical Research Unit, The First Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Min Tao
- School of Pharmacy, Anhui Medical University, Hefei, China; Department of Clinical Pharmacology, The Second Hospital of Anhui Medical University, Hefei, China; Clinical Research Unit, The First Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Hengyan Zhang
- Clinical Research Unit, The First Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Shichao Zhang
- Department of Hepatic Surgery, The Eastern Hepatobiliary Surgery Hospital, Naval Medical University, Shanghai, China
| | - Xiaojuan Hou
- The National Center for Liver Cancer, Shanghai, China; Tumor Immunology and Gene Therapy Center, Third Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Chen Zong
- The National Center for Liver Cancer, Shanghai, China; Tumor Immunology and Gene Therapy Center, Third Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Gangqi Sun
- Molecular Pathology Center, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Shiyao Feng
- Department of Urology, Second Affiliated Hospital, Anhui Medical University, Hefei, China
| | - Haixin Yan
- Department of Urology, Second Affiliated Hospital, Anhui Medical University, Hefei, China
| | - Ying Lu
- Department of Pharmaceutical Sciences, School of Pharmacy, Naval Medical University, Shanghai, China
| | - Xue Yang
- The National Center for Liver Cancer, Shanghai, China; Tumor Immunology and Gene Therapy Center, Third Affiliated Hospital of Naval Medical University, Shanghai, China.
| | - Lixin Wei
- The National Center for Liver Cancer, Shanghai, China; Tumor Immunology and Gene Therapy Center, Third Affiliated Hospital of Naval Medical University, Shanghai, China.
| | - Li Zhang
- School of Pharmacy, Anhui Medical University, Hefei, China; Department of Clinical Pharmacology, The Second Hospital of Anhui Medical University, Hefei, China; Clinical Research Unit, The First Affiliated Hospital of Naval Medical University, Shanghai, China.
| |
Collapse
|
20
|
Zelepukin IV, Shevchenko KG, Deyev SM. Rediscovery of mononuclear phagocyte system blockade for nanoparticle drug delivery. Nat Commun 2024; 15:4366. [PMID: 38777821 PMCID: PMC11111695 DOI: 10.1038/s41467-024-48838-5] [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: 07/31/2023] [Accepted: 05/14/2024] [Indexed: 05/25/2024] Open
Abstract
Rapid uptake of nanoparticles by mononuclear phagocyte system (MPS) significantly hampers their therapeutic efficacy. Temporal MPS blockade is one of the few ways to overcome this barrier - the approach rediscovered many times under different names but never extensively used in clinic. Using meta-analysis of the published data we prove the efficacy of this technique for enhancing particle circulation in blood and their delivery to tumours, describe a century of its evolution and potential combined mechanism behind it. Finally, we discuss future directions of the research focusing on the features essential for successful clinical translation of the method.
Collapse
Affiliation(s)
- Ivan V Zelepukin
- Department of Medicinal Chemistry, Uppsala University, 751 23, Uppsala, Sweden.
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, 117997, Moscow, Russia.
| | | | - Sergey M Deyev
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, 117997, Moscow, Russia
| |
Collapse
|
21
|
Tsissios G, Sallese A, Perez-Estrada JR, Tangeman JA, Chen W, Smucker B, Ratvasky SC, Grajales-Esquivel E, Martinez A, Visser KJ, Joven Araus A, Wang H, Simon A, Yun MH, Del Rio-Tsonis K. Macrophages modulate fibrosis during newt lens regeneration. Stem Cell Res Ther 2024; 15:141. [PMID: 38745238 PMCID: PMC11094960 DOI: 10.1186/s13287-024-03740-1] [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: 11/15/2023] [Accepted: 04/23/2024] [Indexed: 05/16/2024] Open
Abstract
BACKGROUND Previous studies have suggested that macrophages are present during lens regeneration in newts, but their role in the process is yet to be elucidated. METHODS Here we generated a transgenic reporter line using the newt, Pleurodeles waltl, that traces macrophages during lens regeneration. Furthermore, we assessed early changes in gene expression during lens regeneration using two newt species, Notophthalmus viridescens and Pleurodeles waltl. Finally, we used clodronate liposomes to deplete macrophages during lens regeneration in both species and tested the effect of a subsequent secondary injury after macrophage recovery. RESULTS Macrophage depletion abrogated lens regeneration, induced the formation of scar-like tissue, led to inflammation, decreased iris pigment epithelial cell (iPEC) proliferation, and increased rates of apoptosis in the eye. Some of these phenotypes persisted throughout the last observation period of 100 days and could be attenuated by exogenous FGF2 administration. A distinct transcript profile encoding acute inflammatory effectors was established for the dorsal iris. Reinjury of the newt eye alleviated the effects of macrophage depletion, including the resolution of scar-like tissue, and re-initiated the regeneration process. CONCLUSIONS Together, our findings highlight the importance of macrophages for facilitating a pro-regenerative environment in the newt eye by regulating fibrotic responses, modulating the overall inflammatory landscape, and maintaining the proper balance of early proliferation and late apoptosis of the iPECs.
Collapse
Affiliation(s)
- Georgios Tsissios
- Department of Biology, Miami University, Oxford, OH, USA
- Center for Visual Sciences at, Miami University, Oxford, OH, USA
- Cellular Molecular and Structural Biology Program, Miami University, Oxford, OH, USA
| | - Anthony Sallese
- Department of Biology, Miami University, Oxford, OH, USA
- Center for Visual Sciences at, Miami University, Oxford, OH, USA
| | - J Raul Perez-Estrada
- Department of Biology, Miami University, Oxford, OH, USA
- Center for Visual Sciences at, Miami University, Oxford, OH, USA
| | - Jared A Tangeman
- Department of Biology, Miami University, Oxford, OH, USA
- Center for Visual Sciences at, Miami University, Oxford, OH, USA
- Cellular Molecular and Structural Biology Program, Miami University, Oxford, OH, USA
| | - Weihao Chen
- Center for Visual Sciences at, Miami University, Oxford, OH, USA
- Cellular Molecular and Structural Biology Program, Miami University, Oxford, OH, USA
- Department of Chemical, Paper and Biomedical Engineering, Miami University, Oxford, OH, USA
| | - Byran Smucker
- Center for Visual Sciences at, Miami University, Oxford, OH, USA
- Department of Statistics, Miami University, Oxford, OH, USA
| | - Sophia C Ratvasky
- Department of Biology, Miami University, Oxford, OH, USA
- Center for Visual Sciences at, Miami University, Oxford, OH, USA
- Cellular Molecular and Structural Biology Program, Miami University, Oxford, OH, USA
| | - Erika Grajales-Esquivel
- Department of Biology, Miami University, Oxford, OH, USA
- Center for Visual Sciences at, Miami University, Oxford, OH, USA
| | - Arielle Martinez
- Department of Biology, Miami University, Oxford, OH, USA
- Center for Visual Sciences at, Miami University, Oxford, OH, USA
| | - Kimberly J Visser
- CRTD/ Center for Regenerative Therapies Dresden, Technische Universität Dresden, Dresden, Germany
| | - Alberto Joven Araus
- Department of Cell and Molecular Biology, Karolinska Institute, Stockholm, Sweden
| | - Hui Wang
- Center for Visual Sciences at, Miami University, Oxford, OH, USA
- Department of Chemical, Paper and Biomedical Engineering, Miami University, Oxford, OH, USA
| | - András Simon
- Department of Cell and Molecular Biology, Karolinska Institute, Stockholm, Sweden
| | - Maximina H Yun
- CRTD/ Center for Regenerative Therapies Dresden, Technische Universität Dresden, Dresden, Germany
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
- Cluster of Excellence Physics of Life, Technische Universität Dresden, Dresden, Germany
| | - Katia Del Rio-Tsonis
- Department of Biology, Miami University, Oxford, OH, USA.
- Center for Visual Sciences at, Miami University, Oxford, OH, USA.
- Cellular Molecular and Structural Biology Program, Miami University, Oxford, OH, USA.
| |
Collapse
|
22
|
da Silva RG, Stocks CJ, Hu G, Kline KA, Chen J. Bosutinib Stimulates Macrophage Survival, Phagocytosis, and Intracellular Killing of Bacteria. ACS Infect Dis 2024; 10:1725-1738. [PMID: 38602352 PMCID: PMC11091880 DOI: 10.1021/acsinfecdis.4c00086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 03/27/2024] [Accepted: 03/28/2024] [Indexed: 04/12/2024]
Abstract
Host-acting compounds are emerging as potential alternatives to combating antibiotic resistance. Here, we show that bosutinib, an FDA-approved chemotherapeutic for treating chronic myelogenous leukemia, does not possess any antibiotic activity but enhances macrophage responses to bacterial infection. In vitro, bosutinib stimulates murine and human macrophages to kill bacteria more effectively. In a murine wound infection with vancomycin-resistant Enterococcus faecalis, a single intraperitoneal bosutinib injection or multiple topical applications on the wound reduce the bacterial load by approximately 10-fold, which is abolished by macrophage depletion. Mechanistically, bosutinib stimulates macrophage phagocytosis of bacteria by upregulating surface expression of bacterial uptake markers Dectin-1 and CD14 and promoting actin remodeling. Bosutinib also stimulates bacterial killing by elevating the intracellular levels of reactive oxygen species. Moreover, bosutinib drives NF-κB activation, which protects infected macrophages from dying. Other Src kinase inhibitors such as DMAT and tirbanibulin also upregulate expression of bacterial uptake markers in macrophages and enhance intracellular bacterial killing. Finally, cotreatment with bosutinib and mitoxantrone, another chemotherapeutic in clinical use, results in an additive effect on bacterial clearance in vitro and in vivo. These results show that bosutinib stimulates macrophage clearance of bacterial infections through multiple mechanisms and could be used to boost the host innate immunity to combat drug-resistant bacterial infections.
Collapse
Affiliation(s)
- Ronni
A. G. da Silva
- Singapore-MIT
Alliance for Research and Technology Centre, Antimicrobial Drug Resistance Interdisciplinary Research Group, 138602 Singapore
- Singapore
Centre for Environmental Life Sciences Engineering, Nanyang Technological University, 637551 Singapore
| | - Claudia J. Stocks
- Singapore
Centre for Environmental Life Sciences Engineering, Nanyang Technological University, 637551 Singapore
| | - Guangan Hu
- Koch
Institute for Integrative Cancer Research and Department of Biology, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
| | - Kimberly A. Kline
- Singapore-MIT
Alliance for Research and Technology Centre, Antimicrobial Drug Resistance Interdisciplinary Research Group, 138602 Singapore
- Singapore
Centre for Environmental Life Sciences Engineering, Nanyang Technological University, 637551 Singapore
- Department
of Microbiology and Molecular Medicine, Faculty of Medicine, University of Geneva, Geneva1211, Switzerland
| | - Jianzhu Chen
- Singapore-MIT
Alliance for Research and Technology Centre, Antimicrobial Drug Resistance Interdisciplinary Research Group, 138602 Singapore
- Koch
Institute for Integrative Cancer Research and Department of Biology, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
| |
Collapse
|
23
|
Wong IG, Stark J, Ya V, Moye AL, Vazquez AB, Dang SM, Shehaj A, Rouhani MJ, Bronson R, Janes SM, Rowbotham SP, Paschini M, Franklin RA, Kim CF. Airway injury induces alveolar epithelial and mesenchymal responses mediated by macrophages. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.02.587596. [PMID: 38617297 PMCID: PMC11014629 DOI: 10.1101/2024.04.02.587596] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/16/2024]
Abstract
Acute injury in the airways or the lung activates local progenitors and stimulates changes in cell-cell interactions to restore homeostasis, but it is not appreciated how more distant niches are impacted. We utilized mouse models of airway-specific epithelial injury to examine secondary tissue-wide alveolar, immune, and mesenchymal responses. Single-cell transcriptomics and in vivo validation revealed transient, tissue-wide proliferation of alveolar type 2 (AT2) progenitor cells after club cell-specific ablation. The AT2 cell proliferative response was reliant on alveolar macrophages (AMs) via upregulation of Spp1 which encodes the secreted factor Osteopontin. A previously uncharacterized mesenchymal population we termed Mesenchymal Airway/Adventitial Niche Cell 2 (MANC2) also exhibited dynamic changes in abundance and a pro-fibrotic transcriptional signature after club cell ablation in an AM-dependent manner. Overall, these results demonstrate that acute airway damage can trigger distal lung responses including altered cell-cell interactions that may contribute to potential vulnerabilities for further dysregulation and disease.
Collapse
|
24
|
Metwally H, Elbrashy MM, Ozawa T, Okuyama K, White JT, Tulyeu J, Søndergaard JN, Wing JB, Muratsu A, Matsumoto H, Ikawa M, Kishi H, Taniuchi I, Kishimoto T. Threonine phosphorylation of STAT1 restricts interferon signaling and promotes innate inflammatory responses. Proc Natl Acad Sci U S A 2024; 121:e2402226121. [PMID: 38621137 PMCID: PMC11046697 DOI: 10.1073/pnas.2402226121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Accepted: 03/15/2024] [Indexed: 04/17/2024] Open
Abstract
Since its discovery over three decades ago, signal transducer and activator of transcription 1 (STAT1) has been extensively studied as a central mediator for interferons (IFNs) signaling and antiviral defense. Here, using genetic and biochemical assays, we unveil Thr748 as a conserved IFN-independent phosphorylation switch in Stat1, which restricts IFN signaling and promotes innate inflammatory responses following the recognition of the bacterial-derived toxin lipopolysaccharide (LPS). Genetically engineered mice expressing phospho-deficient threonine748-to-alanine (T748A) mutant Stat1 are resistant to LPS-induced lethality. Of note, T748A mice exhibited undisturbed IFN signaling, as well as total expression of Stat1. Further, the T748A point mutation of Stat1 recapitulates the safeguard effect of the genetic ablation of Stat1 following LPS-induced lethality, indicating that the Thr748 phosphorylation contributes inflammatory functionalities of Stat1. Mechanistically, LPS-induced Toll-like receptor 4 endocytosis activates a cell-intrinsic IκB kinase-mediated Thr748 phosphorylation of Stat1, which promotes macrophage inflammatory response while restricting the IFN and anti-inflammatory responses. Depletion of macrophages restores the sensitivity of the T748A mice to LPS-induced lethality. Together, our study indicates a phosphorylation-dependent modular functionality of Stat1 in innate immune responses: IFN phospho-tyrosine dependent and inflammatory phospho-threonine dependent. Better understanding of the Thr748 phosphorylation of Stat1 may uncover advanced pharmacologically targetable molecules and offer better treatment modalities for sepsis, a disease that claims millions of lives annually.
Collapse
Affiliation(s)
- Hozaifa Metwally
- Laboratory of Immune Regulation, The World Premier International Research Center Initiative, Immunology Frontier Research Center, Osaka University, Osaka565-0871, Japan
| | - Maha M. Elbrashy
- Laboratory of Immune Regulation, The World Premier International Research Center Initiative, Immunology Frontier Research Center, Osaka University, Osaka565-0871, Japan
- Biochemistry Department, Biotechnology Research Institute, National Research Center, GizaP.O. 12622, Egypt
| | - Tatsuhiko Ozawa
- Department of Immunology, Faculty of Medicine, Academic Assembly, University of Toyama, Toyama930-0194, Japan
| | - Kazuki Okuyama
- Laboratory for Transcriptional Regulation, RIKEN Center for Integrative Medical Sciences, Tsurumi-ku, Yokohama, Kanagawa230-0045, Japan
| | - Jason T. White
- Laboratory of Experimental Immunology, The World Premier International Research Center Initiative, Immunology Frontier Research Center, Osaka University, Osaka565-0871, Japan
| | - Janyerkye Tulyeu
- Human Immunology Team, Center for Infectious Disease Education and Research, Osaka University, Suita565-0871, Japan
| | - Jonas Nørskov Søndergaard
- Human Immunology Team, Center for Infectious Disease Education and Research, Osaka University, Suita565-0871, Japan
| | - James Badger Wing
- Laboratory of Human Single Cell Immunology, The World Premier International Research Center Initiative, Immunology Frontier Research Center, Osaka University, Osaka565-0871, Japan
| | - Arisa Muratsu
- Department of Traumatology and Acute Critical Medicine, Osaka University Graduate School of Medicine, Osaka565-0871, Japan
| | - Hisatake Matsumoto
- Department of Traumatology and Acute Critical Medicine, Osaka University Graduate School of Medicine, Osaka565-0871, Japan
| | - Masahito Ikawa
- Research Institute for Microbial Diseases, Osaka University, Osaka565-0871, Japan
| | - Hiroyuki Kishi
- Department of Immunology, Faculty of Medicine, Academic Assembly, University of Toyama, Toyama930-0194, Japan
| | - Ichiro Taniuchi
- Laboratory for Transcriptional Regulation, RIKEN Center for Integrative Medical Sciences, Tsurumi-ku, Yokohama, Kanagawa230-0045, Japan
| | - Tadamitsu Kishimoto
- Laboratory of Immune Regulation, The World Premier International Research Center Initiative, Immunology Frontier Research Center, Osaka University, Osaka565-0871, Japan
| |
Collapse
|
25
|
Zhang L, Saito H, Higashimoto T, Kaji T, Nakamura A, Iwamori K, Nagano R, Motooka D, Okuzaki D, Uezumi A, Seno S, Fukada SI. Regulation of muscle hypertrophy through granulin: Relayed communication among mesenchymal progenitors, macrophages, and satellite cells. Cell Rep 2024; 43:114052. [PMID: 38573860 DOI: 10.1016/j.celrep.2024.114052] [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: 06/07/2023] [Revised: 02/14/2024] [Accepted: 03/19/2024] [Indexed: 04/06/2024] Open
Abstract
Skeletal muscles exert remarkable regenerative or adaptive capacities in response to injuries or mechanical loads. However, the cellular networks underlying muscle adaptation are poorly understood compared to those underlying muscle regeneration. We employed single-cell RNA sequencing to investigate the gene expression patterns and cellular networks activated in overloaded muscles and compared these results with those observed in regenerating muscles. The cellular composition of the 4-day overloaded muscle, when macrophage infiltration peaked, closely resembled that of the 10-day regenerating muscle. In addition to the mesenchymal progenitor-muscle satellite cell (MuSC) axis, interactome analyses or targeted depletion experiments revealed communications between mesenchymal progenitors-macrophages and macrophages-MuSCs. Furthermore, granulin, a macrophage-derived factor, inhibited MuSC differentiation, and Granulin-knockout mice exhibited blunted muscle hypertrophy due to the premature differentiation of overloaded MuSCs. These findings reveal the critical role of granulin through the relayed communications of mesenchymal progenitors, macrophages, and MuSCs in facilitating efficient muscle hypertrophy.
Collapse
Affiliation(s)
- Lidan Zhang
- Center for Medical Epigenetics, School of Basic Medical Sciences, Chongqing Medical University, Chongqing 40016, China; Laboratory of Stem Cell Regeneration and Adaptation, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamada-oka, Suita, Osaka 565-0871, Japan
| | - Hayato Saito
- Laboratory of Stem Cell Regeneration and Adaptation, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamada-oka, Suita, Osaka 565-0871, Japan
| | - Tatsuyoshi Higashimoto
- Laboratory of Stem Cell Regeneration and Adaptation, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamada-oka, Suita, Osaka 565-0871, Japan
| | - Takayuki Kaji
- Laboratory of Stem Cell Regeneration and Adaptation, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamada-oka, Suita, Osaka 565-0871, Japan
| | - Ayasa Nakamura
- Laboratory of Stem Cell Regeneration and Adaptation, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamada-oka, Suita, Osaka 565-0871, Japan
| | - Kanako Iwamori
- Laboratory of Stem Cell Regeneration and Adaptation, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamada-oka, Suita, Osaka 565-0871, Japan
| | - Ryoko Nagano
- Laboratory of Stem Cell Regeneration and Adaptation, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamada-oka, Suita, Osaka 565-0871, Japan; Laboratory of Oral Pathology, Division of Maxillofacial Diagnostic and Surgical Sciences, Faculty of Dental Science, Kyushu University, 3-1-1 Maidashi, Higashi, Fukuoka 812-8582, Japan
| | - Daisuke Motooka
- Genome Information Research Center, Research Institute for Microbial Diseases, Osaka University, Osaka 565-0871, Japan
| | - Daisuke Okuzaki
- Genome Information Research Center, Research Institute for Microbial Diseases, Osaka University, Osaka 565-0871, Japan
| | - Akiyoshi Uezumi
- Division of Cell Heterogeneity, Medical Research Center for High Depth Omics, Medical Institute of Bioregulation, Kyushu University, 3-1-1, Maidashi, Higashi, Fukuoka 812-8582, Japan
| | - Shigeto Seno
- Department of Bioinformatic Engineering, Graduate School of Information Science and Technology, Osaka University, 1-5 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - So-Ichiro Fukada
- Laboratory of Stem Cell Regeneration and Adaptation, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamada-oka, Suita, Osaka 565-0871, Japan.
| |
Collapse
|
26
|
Ander SE, Parks MG, Davenport BJ, Li FS, Bosco-Lauth A, Carpentier KS, Sun C, Lucas CJ, Klimstra WB, Ebel GD, Morrison TE. Phagocyte-expressed glycosaminoglycans promote capture of alphaviruses from the blood circulation in a host species-specific manner. PNAS NEXUS 2024; 3:pgae119. [PMID: 38560529 PMCID: PMC10978064 DOI: 10.1093/pnasnexus/pgae119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Accepted: 03/08/2024] [Indexed: 04/04/2024]
Abstract
The magnitude and duration of vertebrate viremia are critical determinants of arbovirus transmission, geographic spread, and disease severity-yet, mechanisms determining arbovirus viremia levels are poorly defined. Previous studies have drawn associations between in vitro virion-glycosaminoglycan (GAG) interactions and in vivo clearance kinetics of virions from blood circulation. From these observations, it is commonly hypothesized that GAG-binding virions are rapidly removed from circulation due to ubiquitous expression of GAGs by vascular endothelial cells, thereby limiting viremia. Using an in vivo model for viremia, we compared the vascular clearance of low and enhanced GAG-binding viral variants of chikungunya, eastern- (EEEV), and Venezuelan- (VEEV) equine encephalitis viruses. We find GAG-binding virions are more quickly removed from circulation than their non-GAG-binding variant; however individual clearance kinetics vary between GAG-binding viruses, from swift (VEEV) to slow removal from circulation (EEEV). Remarkably, we find phagocytes are required for efficient vascular clearance of some enhanced GAG-binding virions. Moreover, transient depletion of vascular heparan sulfate impedes vascular clearance of only some GAG-binding viral variants and in a phagocyte-dependent manner, implying phagocytes can mediate vascular GAG-virion interactions. Finally, in direct contrast to mice, we find enhanced GAG-binding EEEV is resistant to vascular clearance in avian hosts, suggesting the existence of species-specificity in virion-GAG interactions. In summary, these data support a role for GAG-mediated clearance of some viral particles from the blood circulation, illuminate the potential of blood-contacting phagocytes as a site for GAG-virion binding, and suggest a role for species-specific GAG structures in arbovirus ecology.
Collapse
Affiliation(s)
- Stephanie E Ander
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, CO 80045, USA
| | - M Guston Parks
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, CO 80045, USA
| | - Bennett J Davenport
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, CO 80045, USA
| | - Frances S Li
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, CO 80045, USA
| | - Angela Bosco-Lauth
- Department of Microbiology, Immunology, and Pathology, Colorado State University, Fort Collins, CO 80523, USA
| | - Kathryn S Carpentier
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, CO 80045, USA
| | - Chengqun Sun
- Center for Vaccine Research, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Cormac J Lucas
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, CO 80045, USA
| | - William B Klimstra
- Center for Vaccine Research, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Gregory D Ebel
- Department of Microbiology, Immunology, and Pathology, Colorado State University, Fort Collins, CO 80523, USA
| | - Thomas E Morrison
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, CO 80045, USA
| |
Collapse
|
27
|
Zaid A, Ariel A. Harnessing anti-inflammatory pathways and macrophage nano delivery to treat inflammatory and fibrotic disorders. Adv Drug Deliv Rev 2024; 207:115204. [PMID: 38342241 DOI: 10.1016/j.addr.2024.115204] [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: 07/30/2023] [Revised: 12/08/2023] [Accepted: 02/05/2024] [Indexed: 02/13/2024]
Abstract
Targeting specific organs and cell types using nanotechnology and sophisticated delivery methods has been at the forefront of applicative biomedical sciences lately. Macrophages are an appealing target for immunomodulation by nanodelivery as they are heavily involved in various aspects of many diseases and are highly plastic in their nature. Their continuum of functional "polarization" states has been a research focus for many years yielding a profound understanding of various aspects of these cells. The ability of monocyte-derived macrophages to metamorphose from pro-inflammatory to reparative and consequently to pro-resolving effectors has raised significant interest in its therapeutic potential. Here, we briefly survey macrophages' ontogeny and various polarization phenotypes, highlighting their function in the inflammation-resolution shift. We review their inducing mediators, signaling pathways, and biological programs with emphasis on the nucleic acid sensing-IFN-I axis. We also portray the polarization spectrum of macrophages and the characteristics of their transition between different subtypes. Finally, we highlighted different current drug delivery methods for targeting macrophages with emphasis on nanotargeting that might lead to breakthroughs in the treatment of wound healing, bone regeneration, autoimmune, and fibrotic diseases.
Collapse
Affiliation(s)
- Ahmad Zaid
- Department of Biology and Human Biology, University of Haifa, Haifa, 3498838 Israel
| | - Amiram Ariel
- Department of Biology and Human Biology, University of Haifa, Haifa, 3498838 Israel.
| |
Collapse
|
28
|
Wang J, Zhang L, Wang L, Tang J, Wang W, Xu Y, Li Z, Ding Z, Jiang X, Xi K, Chen L, Gu Y. Ligand-Selective Targeting of Macrophage Hydrogel Elicits Bone Immune-Stem Cell Endogenous Self-Healing Program to Promote Bone Regeneration. Adv Healthc Mater 2024; 13:e2303851. [PMID: 38226706 PMCID: PMC11468030 DOI: 10.1002/adhm.202303851] [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: 11/05/2023] [Revised: 01/05/2024] [Indexed: 01/17/2024]
Abstract
Targeting macrophages can facilitate the site-specific repair of critical bone defects. Herein, a composite hydrogel, gelatin-Bletilla striata polysaccharide-mesoporous bioactive glass hydrogel (GBMgel), is constructed via the self-assembly of mesoporous bioactive glass on polysaccharide structures, through the Schiff base reaction. GBMgel can efficiently capture macrophages and drive the recruitment of seed stem cells and vascular budding required for regeneration in the early stages of bone injury, and the observed sustained release of inorganic silicon ions further enhances bone matrix deposition, mineralization, and vascular maturation. Moreover, the use of macrophage-depleted rat calvarial defect models further confirms that GBMgel, with ligand-selective macrophage targeting, increases the bone regeneration area and the proportion of mature bone. Mechanistic studies reveal that GBMgel upregulates the TLR4/NF-κB and MAPK macrophage pathways in the early stages and the JAK/STAT3 pathway in the later stages; thus initiating macrophage polarization at different time points. In conclusion, this study is based on the endogenous self-healing properties of bone macrophages, which enhances stem cell homing, and provides a research and theoretical basis upon which bone tissue can be reshaped and regenerated using the body's immune power, providing a new strategy for the treatment of critical bone defects.
Collapse
Affiliation(s)
- Jiahao Wang
- Department of Orthopedicsthe First Affiliated Hospital of Soochow University188 Shizi Road, Gusu DistrictSuzhouJiangsu215006P. R. China
| | - Liang Zhang
- Department of OrthopedicsBeijing Friendship HospitalCapital Medical UniversityNo. 95, Yong An Road, XiCheng DistrictBeijing100050P. R. China
| | - Lingjun Wang
- Department of Orthopedicsthe First Affiliated Hospital of Soochow University188 Shizi Road, Gusu DistrictSuzhouJiangsu215006P. R. China
| | - Jincheng Tang
- Department of Orthopedicsthe First Affiliated Hospital of Soochow University188 Shizi Road, Gusu DistrictSuzhouJiangsu215006P. R. China
| | - Wei Wang
- Department of Orthopedicsthe First Affiliated Hospital of Soochow University188 Shizi Road, Gusu DistrictSuzhouJiangsu215006P. R. China
| | - Yichang Xu
- Department of Orthopedicsthe First Affiliated Hospital of Soochow University188 Shizi Road, Gusu DistrictSuzhouJiangsu215006P. R. China
| | - Ziang Li
- Department of Orthopedicsthe First Affiliated Hospital of Soochow University188 Shizi Road, Gusu DistrictSuzhouJiangsu215006P. R. China
| | - Zhouye Ding
- Department of Orthopedicsthe First Affiliated Hospital of Soochow University188 Shizi Road, Gusu DistrictSuzhouJiangsu215006P. R. China
| | - Xinzhao Jiang
- Department of Orthopedicsthe First Affiliated Hospital of Soochow University188 Shizi Road, Gusu DistrictSuzhouJiangsu215006P. R. China
| | - Kun Xi
- Department of Orthopedicsthe First Affiliated Hospital of Soochow University188 Shizi Road, Gusu DistrictSuzhouJiangsu215006P. R. China
| | - Liang Chen
- Department of Orthopedicsthe First Affiliated Hospital of Soochow University188 Shizi Road, Gusu DistrictSuzhouJiangsu215006P. R. China
| | - Yong Gu
- Department of Orthopedicsthe First Affiliated Hospital of Soochow University188 Shizi Road, Gusu DistrictSuzhouJiangsu215006P. R. China
| |
Collapse
|
29
|
Peng C, Chen J, Wu R, Jiang H, Li J. Unraveling the complex roles of macrophages in obese adipose tissue: an overview. Front Med 2024; 18:205-236. [PMID: 38165533 DOI: 10.1007/s11684-023-1033-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Accepted: 09/15/2023] [Indexed: 01/03/2024]
Abstract
Macrophages, a heterogeneous population of innate immune cells, exhibit remarkable plasticity and play pivotal roles in coordinating immune responses and maintaining tissue homeostasis within the context of metabolic diseases. The activation of inflammatory macrophages in obese adipose tissue leads to detrimental effects, inducing insulin resistance through increased inflammation, impaired thermogenesis, and adipose tissue fibrosis. Meanwhile, adipose tissue macrophages also play a beneficial role in maintaining adipose tissue homeostasis by regulating angiogenesis, facilitating the clearance of dead adipocytes, and promoting mitochondrial transfer. Exploring the heterogeneity of macrophages in obese adipose tissue is crucial for unraveling the pathogenesis of obesity and holds significant potential for targeted therapeutic interventions. Recently, the dual effects and some potential regulatory mechanisms of macrophages in adipose tissue have been elucidated using single-cell technology. In this review, we present a comprehensive overview of the intricate activation mechanisms and diverse functions of macrophages in adipose tissue during obesity, as well as explore the potential of drug delivery systems targeting macrophages, aiming to enhance the understanding of current regulatory mechanisms that may be potentially targeted for treating obesity or metabolic diseases.
Collapse
Affiliation(s)
- Chang Peng
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Jun Chen
- Department of Prosthodontics, Shanghai Engineering Research Center of Advanced Dental Technology and Materials, Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Rui Wu
- Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, 310000, China
| | - Haowen Jiang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China.
| | - Jia Li
- Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, 310000, China.
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China.
| |
Collapse
|
30
|
Saladino GM, Brodin B, Kakadiya R, Toprak MS, Hertz HM. Iterative nanoparticle bioengineering enabled by x-ray fluorescence imaging. SCIENCE ADVANCES 2024; 10:eadl2267. [PMID: 38517973 DOI: 10.1126/sciadv.adl2267] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Accepted: 02/16/2024] [Indexed: 03/24/2024]
Abstract
Nanoparticles (NPs) are currently developed for drug delivery and molecular imaging. However, they often get intercepted before reaching their target, leading to low targeting efficacy and signal-to-noise ratio. They tend to accumulate in organs like lungs, liver, kidneys, and spleen. The remedy is to iteratively engineer NP surface properties and administration strategies, presently a time-consuming process that includes organ dissection at different time points. To improve this, we propose a rapid iterative approach using whole-animal x-ray fluorescence (XRF) imaging to systematically evaluate NP distribution in vivo. We applied this method to molybdenum-based NPs and clodronate liposomes for tumor targeting with transient macrophage depletion, leading to reduced accumulations in lungs and liver and eventual tumor detection. XRF computed tomography (XFCT) provided 3D insight into NP distribution within the tumor. We validated the results using a multiscale imaging approach with dye-doped NPs and gene expression analysis for nanotoxicological profiling. XRF imaging holds potential for advancing therapeutics and diagnostics in preclinical pharmacokinetic studies.
Collapse
Affiliation(s)
- Giovanni M Saladino
- Department of Applied Physics, Biomedical and X-Ray Physics, KTH Royal Institute of Technology, SE 10691, Stockholm, Sweden
| | - Bertha Brodin
- Department of Applied Physics, Biomedical and X-Ray Physics, KTH Royal Institute of Technology, SE 10691, Stockholm, Sweden
| | - Ronak Kakadiya
- Department of Applied Physics, Biomedical and X-Ray Physics, KTH Royal Institute of Technology, SE 10691, Stockholm, Sweden
| | - Muhammet S Toprak
- Department of Applied Physics, Biomedical and X-Ray Physics, KTH Royal Institute of Technology, SE 10691, Stockholm, Sweden
| | - Hans M Hertz
- Department of Applied Physics, Biomedical and X-Ray Physics, KTH Royal Institute of Technology, SE 10691, Stockholm, Sweden
| |
Collapse
|
31
|
Maryam B, Smith ME, Miller SJ, Natarajan H, Zimmerman KA. Macrophage Ontogeny, Phenotype, and Function in Ischemia Reperfusion-Induced Injury and Repair. KIDNEY360 2024; 5:459-470. [PMID: 38297436 PMCID: PMC11000738 DOI: 10.34067/kid.0000000000000376] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Accepted: 01/22/2024] [Indexed: 02/02/2024]
Abstract
AKI is characterized by a sudden, and usually reversible, decline in kidney function. In mice, ischemia-reperfusion injury (IRI) is commonly used to model the pathophysiologic features of clinical AKI. Macrophages are a unifying feature of IRI as they regulate both the initial injury response as well as the long-term outcome following resolution of injury. Initially, macrophages in the kidney take on a proinflammatory phenotype characterized by the production of inflammatory cytokines, such as CCL2 (monocyte chemoattractant protein 1), IL-6, IL-1 β , and TNF- α . Release of these proinflammatory cytokines leads to tissue damage. After resolution of the initial injury, macrophages take on a reparative role, aiding in tissue repair and restoration of kidney function. By contrast, failure to resolve the initial injury results in prolonged inflammatory macrophage accumulation and increased kidney damage, fibrosis, and the eventual development of CKD. Despite the extensive amount of literature that has ascribed these functions to M1/M2 macrophages, a recent paradigm shift in the macrophage field now defines macrophages on the basis of their ontological origin, namely monocyte-derived and tissue-resident macrophages. In this review, we focus on macrophage phenotype and function during IRI-induced injury, repair, and transition to CKD using both the classic (M1/M2) and novel (ontological origin) definition of kidney macrophages.
Collapse
Affiliation(s)
- Bibi Maryam
- Division of Nephrology, Department of Internal Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
- Department of Internal Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - Morgan E. Smith
- Division of Nephrology, Department of Internal Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
- Department of Internal Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - Sarah J. Miller
- Division of Nephrology, Department of Internal Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
- Department of Internal Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - Hariharasudan Natarajan
- Department of Internal Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - Kurt A. Zimmerman
- Division of Nephrology, Department of Internal Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
- Department of Internal Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| |
Collapse
|
32
|
Choi TH, Yoo RJ, Park JY, Kim JY, Ann YC, Park J, Kim JS, Kim K, Shin YJ, Lee YJ, Lee KC, Park J, Chung H, Seok SH, Im HJ, Lee YS. Development of finely tuned liposome nanoplatform for macrophage depletion. J Nanobiotechnology 2024; 22:83. [PMID: 38424578 PMCID: PMC10903058 DOI: 10.1186/s12951-024-02325-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Accepted: 01/30/2024] [Indexed: 03/02/2024] Open
Abstract
BACKGROUND Immunotherapy with clodronate-encapsulated liposomes, which induce macrophage depletion, has been studied extensively. However, previously reported liposomal formulation-based drugs (Clodrosome® and m-Clodrosome®) are limited by their inconsistent size and therapeutic efficacy. Thus, we aimed to achieve consistent therapeutic effects by effectively depleting macrophages with uniform-sized liposomes. RESULTS We developed four types of click chemistry-based liposome nanoplatforms that were uniformly sized and encapsulated with clodronate, for effective macrophage depletion, followed by conjugation with Man-N3 and radiolabeling. Functionalization with Man-N3 improves the specific targeting of M2 macrophages, and radioisotope labeling enables in vivo imaging of the liposome nanoplatforms. The functionalized liposome nanoplatforms are stable under physiological conditions. The difference in the biodistribution of the four liposome nanoplatforms in vivo were recorded using positron emission tomography imaging. Among the four platforms, the clodronate-encapsulated mannosylated liposome effectively depleted M2 macrophages in the normal liver and tumor microenvironment ex vivo compared to that by Clodrosome® and m-Clodrosome®. CONCLUSION The newly-developed liposome nanoplatform, with finely tuned size control, high in vivo stability, and excellent ex vivo M2 macrophage targeting and depletion effects, is a promising macrophage-depleting agent.
Collapse
Affiliation(s)
- Tae Hyeon Choi
- Department of Nuclear Medicine, Seoul National University College of Medicine, Seoul, South Korea
- Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, Seoul National University, Seoul, 08826, South Korea
| | - Ran Ji Yoo
- Department of Nuclear Medicine, Seoul National University College of Medicine, Seoul, South Korea
- Department of Nuclear Medicine, Seoul National University Hospital, 101 Daehak-Ro, Jongno-Gu, Seoul, South Korea
- Biomedical Research Institute, Seoul National University Hospital, Seoul, South Korea
| | - Ji Yong Park
- Department of Nuclear Medicine, Seoul National University College of Medicine, Seoul, South Korea
- Institute of Radiation Medicine, Medical Research Center, Seoul National University College of Medicine, Seoul, South Korea
| | - Ji Yoon Kim
- Department of Nuclear Medicine, Seoul National University College of Medicine, Seoul, South Korea
- Institute of Radiation Medicine, Medical Research Center, Seoul National University College of Medicine, Seoul, South Korea
| | - Young Chan Ann
- Department of Nuclear Medicine, Seoul National University College of Medicine, Seoul, South Korea
- School of Dentistry, Seoul National University, Seoul, South Korea
| | - Jeongbin Park
- Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, Seoul National University, Seoul, 08826, South Korea
| | - Jin Sil Kim
- Department of Nuclear Medicine, Seoul National University College of Medicine, Seoul, South Korea
| | - Kyuwan Kim
- Department of Nuclear Medicine, Seoul National University College of Medicine, Seoul, South Korea
| | - Yu Jin Shin
- Department of Nuclear Medicine, Seoul National University College of Medicine, Seoul, South Korea
- Cancer Research Institute, Seoul National University College of Medicine, Seoul, South Korea
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, South Korea
| | - Yong Jin Lee
- Division of Applied RI, Korea Institute of Radiological and Medical Sciences (KIRAMS), Seoul, South Korea
| | - Kyo Chul Lee
- Division of Applied RI, Korea Institute of Radiological and Medical Sciences (KIRAMS), Seoul, South Korea
| | - Jisu Park
- Cancer Research Institute, Seoul National University College of Medicine, Seoul, South Korea
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, South Korea
- Department of Microbiology and Immunology, and Institute of Endemic Disease, Seoul National University College of Medicine, Seoul, South Korea
| | - Hyewon Chung
- Cancer Research Institute, Seoul National University College of Medicine, Seoul, South Korea
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, South Korea
- Department of Microbiology and Immunology, and Institute of Endemic Disease, Seoul National University College of Medicine, Seoul, South Korea
| | - Seung Hyeok Seok
- Cancer Research Institute, Seoul National University College of Medicine, Seoul, South Korea
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, South Korea
- Department of Microbiology and Immunology, and Institute of Endemic Disease, Seoul National University College of Medicine, Seoul, South Korea
| | - Hyung-Jun Im
- Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, Seoul National University, Seoul, 08826, South Korea.
- Institute of Radiation Medicine, Medical Research Center, Seoul National University College of Medicine, Seoul, South Korea.
- Cancer Research Institute, Seoul National University College of Medicine, Seoul, South Korea.
| | - Yun-Sang Lee
- Department of Nuclear Medicine, Seoul National University College of Medicine, Seoul, South Korea.
- Department of Nuclear Medicine, Seoul National University Hospital, 101 Daehak-Ro, Jongno-Gu, Seoul, South Korea.
- Institute of Radiation Medicine, Medical Research Center, Seoul National University College of Medicine, Seoul, South Korea.
- Cancer Research Institute, Seoul National University College of Medicine, Seoul, South Korea.
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, South Korea.
| |
Collapse
|
33
|
Fotio Y, Mabou Tagne A, Squire E, Lee HL, Phillips CM, Chang K, Ahmed F, Greenberg AS, Villalta SA, Scarfone VM, Spadoni G, Mor M, Piomelli D. NAAA-regulated lipid signaling in monocytes controls the induction of hyperalgesic priming in mice. Nat Commun 2024; 15:1705. [PMID: 38402219 PMCID: PMC10894261 DOI: 10.1038/s41467-024-46139-5] [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: 08/14/2023] [Accepted: 02/15/2024] [Indexed: 02/26/2024] Open
Abstract
Circulating monocytes participate in pain chronification but the molecular events that cause their deployment are unclear. Using a mouse model of hyperalgesic priming (HP), we show that monocytes enable progression to pain chronicity through a mechanism that requires transient activation of the hydrolase, N-acylethanolamine acid amidase (NAAA), and the consequent suppression of NAAA-regulated lipid signaling at peroxisome proliferator-activated receptor-α (PPAR-α). Inhibiting NAAA in the 72 hours following administration of a priming stimulus prevented HP. This effect was phenocopied by NAAA deletion and depended on PPAR-α recruitment. Mice lacking NAAA in CD11b+ cells - monocytes, macrophages, and neutrophils - were resistant to HP induction. Conversely, mice overexpressing NAAA or lacking PPAR-α in the same cells were constitutively primed. Depletion of monocytes, but not resident macrophages, generated mice that were refractory to HP. The results identify NAAA-regulated signaling in monocytes as a control node in the induction of HP and, potentially, the transition to pain chronicity.
Collapse
Affiliation(s)
- Yannick Fotio
- Department of Anatomy and Neurobiology, University of California Irvine, Irvine, CA, USA
| | - Alex Mabou Tagne
- Department of Anatomy and Neurobiology, University of California Irvine, Irvine, CA, USA
| | - Erica Squire
- Department of Anatomy and Neurobiology, University of California Irvine, Irvine, CA, USA
| | - Hye-Lim Lee
- Department of Anatomy and Neurobiology, University of California Irvine, Irvine, CA, USA
| | - Connor M Phillips
- Department of Physiology and Biophysics, University of California Irvine, Irvine, CA, USA
| | - Kayla Chang
- Department of Anatomy and Neurobiology, University of California Irvine, Irvine, CA, USA
| | - Faizy Ahmed
- Department of Anatomy and Neurobiology, University of California Irvine, Irvine, CA, USA
| | | | - S Armando Villalta
- Department of Physiology and Biophysics, University of California Irvine, Irvine, CA, USA
- Department of Neurology, University of California Irvine, Irvine, CA, USA
| | - Vanessa M Scarfone
- Sue and Bill Gross Stem Cell Research Center, University of California Irvine, Irvine, CA, USA
| | - Gilberto Spadoni
- Dipartimento di Scienze Biomolecolari, Università di Urbino "Carlo Bo,", Urbino, Italy
| | - Marco Mor
- Dipartimento di Scienze degli Alimenti e del Farmaco, Università di Parma, Parma, Italy
| | - Daniele Piomelli
- Department of Anatomy and Neurobiology, University of California Irvine, Irvine, CA, USA.
- Department of Biological Chemistry, University of California Irvine, Irvine, CA, USA.
- Department of Pharmaceutical Sciences, University of California Irvine, Irvine, CA, USA.
| |
Collapse
|
34
|
Capobianco CA, Hankenson KD, Knights AJ. Temporal dynamics of immune-stromal cell interactions in fracture healing. Front Immunol 2024; 15:1352819. [PMID: 38455063 PMCID: PMC10917940 DOI: 10.3389/fimmu.2024.1352819] [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: 12/08/2023] [Accepted: 02/06/2024] [Indexed: 03/09/2024] Open
Abstract
Bone fracture repair is a complex, multi-step process that involves communication between immune and stromal cells to coordinate the repair and regeneration of damaged tissue. In the US, 10% of all bone fractures do not heal properly without intervention, resulting in non-union. Complications from non-union fractures are physically and financially debilitating. We now appreciate the important role that immune cells play in tissue repair, and the necessity of the inflammatory response in initiating healing after skeletal trauma. The temporal dynamics of immune and stromal cell populations have been well characterized across the stages of fracture healing. Recent studies have begun to untangle the intricate mechanisms driving the immune response during normal or atypical, delayed healing. Various in vivo models of fracture healing, including genetic knockouts, as well as in vitro models of the fracture callus, have been implemented to enable experimental manipulation of the heterogeneous cellular environment. The goals of this review are to (1): summarize our current understanding of immune cell involvement in fracture healing (2); describe state-of-the art approaches to study inflammatory cells in fracture healing, including computational and in vitro models; and (3) identify gaps in our knowledge concerning immune-stromal crosstalk during bone healing.
Collapse
Affiliation(s)
- Christina A. Capobianco
- Department of Orthopaedic Surgery, University of Michigan, Ann Arbor, MI, United States
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, United States
| | - Kurt D. Hankenson
- Department of Orthopaedic Surgery, University of Michigan, Ann Arbor, MI, United States
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, United States
| | - Alexander J. Knights
- Department of Orthopaedic Surgery, University of Michigan, Ann Arbor, MI, United States
| |
Collapse
|
35
|
Abbas N, You K, Getachew A, Wu F, Hussain M, Huang X, Chen Y, Pan T, Li Y. Kupffer cells abrogate homing and repopulation of allogeneic hepatic progenitors in injured liver site. Stem Cell Res Ther 2024; 15:48. [PMID: 38378583 PMCID: PMC10877762 DOI: 10.1186/s13287-024-03656-w] [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: 11/14/2023] [Accepted: 02/05/2024] [Indexed: 02/22/2024] Open
Abstract
BACKGROUND Allogeneic hepatocyte transplantation is an emerging approach to treat acute liver defects. However, durable engraftment of the transplanted cells remains a daunting task, as they are actively cleared by the recipient's immune system. Therefore, a detailed understanding of the innate or adaptive immune cells-derived responses against allogeneic transplanted hepatic cells is the key to rationalize cell-based therapies. METHODS Here, we induced an acute inflammatory regenerative niche (3-96 h) on the surface of the liver by the application of cryo-injury (CI) to systematically evaluate the innate immune response against transplanted allogeneic hepatic progenitors in a sustained micro-inflammatory environment. RESULTS The resulting data highlighted that the injured site was significantly repopulated by alternating numbers of innate immune cells, including neutrophils, monocytes and Kupffer cells (KCs), from 3 to 96 h. The transplanted allo-HPs, engrafted 6 h post-injury, were collectively eliminated by the innate immune response within 24 h of transplantation. Selective depletion of the KCs demonstrated a delayed recruitment of monocytes from day 2 to day 6. In addition, the intrasplenic engraftment of the hepatic progenitors 54 h post-transplantation was dismantled by KCs, while a time-dependent better survival and translocation of the transplanted cells into the injured site could be observed in samples devoid of KCs. CONCLUSION Overall, this study provides evidence that KCs ablation enables a better survival and integration of allo-HPs in a sustained liver inflammatory environment, having implications for rationalizing the cell-based therapeutic interventions against liver defects.
Collapse
Affiliation(s)
- Nasir Abbas
- Center for Health Research, Guangdong Provincial Key Laboratory of Biocomputing, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China
- Centre for Regenerative Medicine and Health (CRMH), Hong Kong Institute of Science and Innovation, Chinese Academy of Sciences, Hong Kong, Hong Kong SAR, China
| | - Kai You
- Center for Health Research, Guangdong Provincial Key Laboratory of Biocomputing, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China
| | - Anteneh Getachew
- Department of Biomedical Engineering, The University of Alabama at Birmingham, Birmingham, USA
| | - Feima Wu
- Center for Health Research, Guangdong Provincial Key Laboratory of Biocomputing, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China
| | - Muzammal Hussain
- Department of Biochemistry and Molecular Pharmacology, New York University Grossman School of Medicine, New York, NY, 10016, USA
| | - Xinping Huang
- Center for Health Research, Guangdong Provincial Key Laboratory of Biocomputing, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China
| | - Yan Chen
- Center for Health Research, Guangdong Provincial Key Laboratory of Biocomputing, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China
- Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China
- CAS Key Laboratory of Regenerative Biology, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China
| | - Tingcai Pan
- Department of Hepatobiliary Surgery II, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, Guangdong Province, China
| | - Yinxiong Li
- Center for Health Research, Guangdong Provincial Key Laboratory of Biocomputing, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China.
- Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China.
- CAS Key Laboratory of Regenerative Biology, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China.
- State Key Laboratory of Respiratory Disease, Guangzhou, 510000, China.
- China-New Zealand Joint Laboratory of Biomedicine and Health, Guangzhou, 510530, China.
| |
Collapse
|
36
|
Wang L, Quine S, Frickenstein AN, Lee M, Yang W, Sheth VM, Bourlon MD, He Y, Lyu S, Garcia-Contreras L, Zhao YD, Wilhelm S. Exploring and Analyzing the Systemic Delivery Barriers for Nanoparticles. ADVANCED FUNCTIONAL MATERIALS 2024; 34:2308446. [PMID: 38828467 PMCID: PMC11142462 DOI: 10.1002/adfm.202308446] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Indexed: 06/05/2024]
Abstract
Most nanomedicines require efficient in vivo delivery to elicit diagnostic and therapeutic effects. However, en route to their intended tissues, systemically administered nanoparticles often encounter delivery barriers. To describe these barriers, we propose the term "nanoparticle blood removal pathways" (NBRP), which summarizes the interactions between nanoparticles and the body's various cell-dependent and cell-independent blood clearance mechanisms. We reviewed nanoparticle design and biological modulation strategies to mitigate nanoparticle-NBRP interactions. As these interactions affect nanoparticle delivery, we studied the preclinical literature from 2011-2021 and analyzed nanoparticle blood circulation and organ biodistribution data. Our findings revealed that nanoparticle surface chemistry affected the in vivo behavior more than other nanoparticle design parameters. Combinatory biological-PEG surface modification improved the blood area under the curve by ~418%, with a decrease in liver accumulation of up to 47%. A greater understanding of nanoparticle-NBRP interactions and associated delivery trends will provide new nanoparticle design and biological modulation strategies for safer, more effective, and more efficient nanomedicines.
Collapse
Affiliation(s)
- Lin Wang
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, Oklahoma, 73019, USA
| | - Skyler Quine
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, Oklahoma, 73019, USA
| | - Alex N. Frickenstein
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, Oklahoma, 73019, USA
| | - Michael Lee
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, Oklahoma, 73019, USA
| | - Wen Yang
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, Oklahoma, 73019, USA
| | - Vinit M. Sheth
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, Oklahoma, 73019, USA
| | - Margaret D. Bourlon
- College of Pharmacy, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, 73117, USA
| | - Yuxin He
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, Oklahoma, 73019, USA
| | - Shanxin Lyu
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, Oklahoma, 73019, USA
| | - Lucila Garcia-Contreras
- College of Pharmacy, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, 73117, USA
| | - Yan D. Zhao
- Department of Biostatistics and Epidemiology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, 73012, USA
- Stephenson Cancer Center, Oklahoma City, Oklahoma, 73104, USA
| | - Stefan Wilhelm
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, Oklahoma, 73019, USA
- Stephenson Cancer Center, Oklahoma City, Oklahoma, 73104, USA
- Institute for Biomedical Engineering, Science, and Technology (IBEST), Norman, Oklahoma, 73019, USA
| |
Collapse
|
37
|
Bobotis BC, Halvorson T, Carrier M, Tremblay MÈ. Established and emerging techniques for the study of microglia: visualization, depletion, and fate mapping. Front Cell Neurosci 2024; 18:1317125. [PMID: 38425429 PMCID: PMC10902073 DOI: 10.3389/fncel.2024.1317125] [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: 10/10/2023] [Accepted: 01/15/2024] [Indexed: 03/02/2024] Open
Abstract
The central nervous system (CNS) is an essential hub for neuronal communication. As a major component of the CNS, glial cells are vital in the maintenance and regulation of neuronal network dynamics. Research on microglia, the resident innate immune cells of the CNS, has advanced considerably in recent years, and our understanding of their diverse functions continues to grow. Microglia play critical roles in the formation and regulation of neuronal synapses, myelination, responses to injury, neurogenesis, inflammation, and many other physiological processes. In parallel with advances in microglial biology, cutting-edge techniques for the characterization of microglial properties have emerged with increasing depth and precision. Labeling tools and reporter models are important for the study of microglial morphology, ultrastructure, and dynamics, but also for microglial isolation, which is required to glean key phenotypic information through single-cell transcriptomics and other emerging approaches. Strategies for selective microglial depletion and modulation can provide novel insights into microglia-targeted treatment strategies in models of neuropsychiatric and neurodegenerative conditions, cancer, and autoimmunity. Finally, fate mapping has emerged as an important tool to answer fundamental questions about microglial biology, including their origin, migration, and proliferation throughout the lifetime of an organism. This review aims to provide a comprehensive discussion of these established and emerging techniques, with applications to the study of microglia in development, homeostasis, and CNS pathologies.
Collapse
Affiliation(s)
- Bianca Caroline Bobotis
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
- Centre for Advanced Materials and Related Technology, Victoria, BC, Canada
| | - Torin Halvorson
- Department of Medicine, University of British Columbia, Vancouver, BC, Canada
- Department of Surgery, University of British Columbia, Vancouver, BC, Canada
- British Columbia Children’s Hospital Research Institute, Vancouver, BC, Canada
| | - Micaël Carrier
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
- Département de Psychiatrie et de Neurosciences, Faculté de Médecine, Université Laval, Québec City, QC, Canada
- Axe neurosciences, Centre de Recherche du CHU de Québec, Université Laval, Québec City, QC, Canada
| | - Marie-Ève Tremblay
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
- Centre for Advanced Materials and Related Technology, Victoria, BC, Canada
- Axe neurosciences, Centre de Recherche du CHU de Québec, Université Laval, Québec City, QC, Canada
- Department of Molecular Medicine, Université Laval, Québec City, QC, Canada
| |
Collapse
|
38
|
Miyahara S, Mori H, Fukuda K, Ogawa M, Saito M. Non-purulent myositis caused by direct invasion of skeletal muscle tissue by Leptospira in a hamster model. Infect Immun 2024; 92:e0042023. [PMID: 38240601 PMCID: PMC10870730 DOI: 10.1128/iai.00420-23] [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: 10/16/2023] [Accepted: 12/22/2023] [Indexed: 02/15/2024] Open
Abstract
Myalgia is a common symptom of Leptospira infection in humans. Autopsies have reported that muscle tissue shows degeneration and necrosis of the myofibers and infiltration of inflammatory cells composed mainly of macrophages and lymphocytes. It remains unclear whether Leptospira directly infects the muscle and how the infiltrating inflammatory cells are involved in muscle fiber destruction. This study evaluated the relationship between histopathological changes and leptospiral localization in the muscle tissue of a hamster model. The influence of macrophages in skeletal muscle injury was also investigated, using selective depletion of macrophages by administration of liposomal clodronate. Hamsters infected subcutaneously with Leptospira interrogans serovar Manilae strain UP-MMC-SM showed myositis of the thighs adjacent to the inoculated area beginning at 6 days post-infection. The myositis was non-purulent and showed sporadic degeneration and necrosis of muscle fibers. The degeneration of myofibers was accompanied by aggregations of macrophages. Immunofluorescence staining revealed leptospires surrounding the damaged muscle fibers. Subcutaneous injection of formalin-killed Leptospira or intraperitoneal injection of live Leptospira caused no myositis in hamster thighs. Liposomal clodronate treatment in infected hamsters reduced macrophage infiltration in muscle tissue without impacting bacterial clearance. Muscle necrosis was still observed in the infected hamsters treated with liposomal clodronate, and there was no significant change in serum creatine kinase levels compared to those in animals treated with liposomes alone. Our findings suggest that leptospiral invasion of muscle tissue from an inoculation site leads to the destruction of muscle fibers and causes non-purulent myositis, whereas the infiltrating macrophages contribute less to muscle destruction.
Collapse
Affiliation(s)
- Satoshi Miyahara
- Department of Microbiology, School of Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Hiroshi Mori
- Department of Microbiology, School of Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan
- Department of Obstetrics and Gynecology, School of Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Kazumasa Fukuda
- Department of Microbiology, School of Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Midori Ogawa
- Department of Microbiology, School of Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Mitsumasa Saito
- Department of Microbiology, School of Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan
| |
Collapse
|
39
|
Gudenschwager Basso EK, Ju J, Soliman E, de Jager C, Wei X, Pridham KJ, Olsen ML, Theus MH. Immunoregulatory and neutrophil-like monocyte subsets with distinct single-cell transcriptomic signatures emerge following brain injury. J Neuroinflammation 2024; 21:41. [PMID: 38310257 PMCID: PMC10838447 DOI: 10.1186/s12974-024-03032-8] [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: 08/25/2023] [Accepted: 01/26/2024] [Indexed: 02/05/2024] Open
Abstract
Monocytes represent key cellular elements that contribute to the neurological sequela following brain injury. The current study reveals that trauma induces the augmented release of a transcriptionally distinct CD115+/Ly6Chi monocyte population into the circulation of mice pre-exposed to clodronate depletion conditions. This phenomenon correlates with tissue protection, blood-brain barrier stability, and cerebral blood flow improvement. Uniquely, this shifted the innate immune cell profile in the cortical milieu and reduced the expression of pro-inflammatory Il6, IL1r1, MCP-1, Cxcl1, and Ccl3 cytokines. Monocytes that emerged under these conditions displayed a morphological and gene profile consistent with a subset commonly seen during emergency monopoiesis. Single-cell RNA sequencing delineated distinct clusters of monocytes and revealed a key transcriptional signature of Ly6Chi monocytes enriched for Apoe and chitinase-like protein 3 (Chil3/Ym1), commonly expressed in pro-resolving immunoregulatory monocytes, as well as granule genes Elane, Prtn3, MPO, and Ctsg unique to neutrophil-like monocytes. The predominate shift in cell clusters included subsets with low expression of transcription factors involved in monocyte conversion, Pou2f2, Na4a1, and a robust enrichment of genes in the oxidative phosphorylation pathway which favors an anti-inflammatory phenotype. Transfer of this monocyte assemblage into brain-injured recipient mice demonstrated their direct role in neuroprotection. These findings reveal a multifaceted innate immune response to brain injury and suggest targeting surrogate monocyte subsets may foster tissue protection in the brain.
Collapse
Affiliation(s)
- Erwin K Gudenschwager Basso
- Department of Biomedical Sciences and Pathobiology, Virginia Tech, 970 Washington Street SW, Life Sciences I, Rm 249 (MC0910), Blacksburg, VA, 24061, USA
- Department of Physiology and Aging, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Jing Ju
- Department of Biomedical Sciences and Pathobiology, Virginia Tech, 970 Washington Street SW, Life Sciences I, Rm 249 (MC0910), Blacksburg, VA, 24061, USA
| | - Eman Soliman
- Department of Biomedical Sciences and Pathobiology, Virginia Tech, 970 Washington Street SW, Life Sciences I, Rm 249 (MC0910), Blacksburg, VA, 24061, USA
| | - Caroline de Jager
- Translational, Biology, Medicine and Health Graduate Program, Virginia Tech, Roanoke, VA, 24016, USA
| | - Xiaoran Wei
- School of Neuroscience, Virginia Tech, Blacksburg, VA, 24061, USA
| | - Kevin J Pridham
- Department of Biomedical Sciences and Pathobiology, Virginia Tech, 970 Washington Street SW, Life Sciences I, Rm 249 (MC0910), Blacksburg, VA, 24061, USA
| | - Michelle L Olsen
- School of Neuroscience, Virginia Tech, Blacksburg, VA, 24061, USA
| | - Michelle H Theus
- Department of Biomedical Sciences and Pathobiology, Virginia Tech, 970 Washington Street SW, Life Sciences I, Rm 249 (MC0910), Blacksburg, VA, 24061, USA.
- Center for Engineered Health, Virginia Tech, Blacksburg, VA, 24061, USA.
| |
Collapse
|
40
|
Nguyen DH. Role of Endorphins in Breast Cancer Pathogenesis and Recovery. ADVANCES IN NEUROBIOLOGY 2024; 35:87-106. [PMID: 38874719 DOI: 10.1007/978-3-031-45493-6_5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2024]
Abstract
Understanding the relationship between stress and breast cancer development is essential to preventing and alleviating the cancer. Recent research has shed light on the cognitive, physiological, cellular, and molecular underpinnings of how the endorphin pathway and stress pathway affect breast cancer. This chapter consists of two parts. Part 1 will discuss the role of endorphins in breast cancer development. This includes a discussion of three topics: (1) the neurophysiological effect of endorphins on breast tumor growth in vivo, along with further experiments that will deepen our knowledge of how β-endorphin affects breast cancer; (2) how both the opioid receptor and somatostatin receptor classes alter intracellular signaling in breast cancer cells; and (3) genetic alleles in the opioid signaling pathway that are correlated with increased breast cancer risk. Part 2 will discuss the role of endorphins in recovery from breast cancer. This includes a discussion of three topics: (1) the relationship between breast cancer diagnosis and depression; (2) the effectiveness of cognitive behavioral therapy in reducing stress in breast cancer patients; and (3) the effect of psychotherapy and exercise on preserving telomere length in breast cancer patients.
Collapse
Affiliation(s)
- David H Nguyen
- BrainScanology, Inc, Concord, CA, USA.
- Molecular Biophysics and Integrated Bioimaging Division, Lawrence Berkeley National Laboratory, Berkeley, CA, USA.
| |
Collapse
|
41
|
Kelly LE, El-Hodiri HM, Crider A, Fischer AJ. Protein phosphatases regulate the formation of Müller glia-derived progenitor cells in the chick retina. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.11.570629. [PMID: 38168320 PMCID: PMC10760049 DOI: 10.1101/2023.12.11.570629] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2024]
Abstract
Different kinase-dependent cell signaling pathways are known to play important roles in glia-mediated neuroprotection and reprogramming of Müller glia (MG) into Müller glia-derived progenitor cells (MGPCs) in the retina. However, very little is known about the phosphatases that regulate kinase-dependent signaling in MG. Using single-cell RNA-sequencing (scRNA-seq) databases, we investigated patterns of expression of Dual Specificity Phosphatases (DUSP1/6) and other protein phosphatases in normal and damaged chick retinas. We found that DUSP1, DUSP6, PPP3CB, PPP3R1 and PPPM1A/B/D/E/G are dynamically expressed by MG and MGPCs in retinas during the process of reprogramming. We find that inhibition of DUSP1/6 and PP2C phosphatases enhances the formation of proliferating MGPCs in damaged retinas and in retinas treated with insulin in FGF2 in the absence of damage. By contrast, inhibition of PP2B phosphatases suppressed the formation of proliferating MGPCs, but increased numbers of proliferating MGPCs in undamaged retinas treated with insulin and FGF2. In damaged retinas, inhibition of DUSP1/6 increased levels of pERK1/2 and cFos in MG whereas inhibition of PP2B's decreased levels of pStat3 and pS6 in MG. Analyses of scRNA-seq libraries identified numerous differentially activated gene modules in MG in damaged retinas versus MG in retinas treated with insulin+FGF2 suggesting significant differences in kinase-dependent signaling pathways that converge on the formation of MGPCs. Inhibition of phosphatases had no significant effects upon numbers of dying cells in damaged retinas. We conclude that the activity of different protein phosphatases "fine-tune" the cell signaling responses of MG in damaged retinas and during the reprogramming of MG into MGPCs.
Collapse
Affiliation(s)
- Lisa E. Kelly
- Department of Neuroscience, College of Medicine, The Ohio State University, Columbus, OH
| | - Heithem M. El-Hodiri
- Department of Neuroscience, College of Medicine, The Ohio State University, Columbus, OH
| | - Andrew Crider
- Department of Neuroscience, College of Medicine, The Ohio State University, Columbus, OH
| | - Andy J. Fischer
- Department of Neuroscience, College of Medicine, The Ohio State University, Columbus, OH
| |
Collapse
|
42
|
Ivanova A, Badertscher L, O'Driscoll G, Bergman J, Gordon E, Gunnarsson A, Johansson C, Munson MJ, Spinelli C, Torstensson S, Vilén L, Voirel A, Wiseman J, Rak J, Dekker N, Lázaro‐Ibáñez E. Creating Designer Engineered Extracellular Vesicles for Diverse Ligand Display, Target Recognition, and Controlled Protein Loading and Delivery. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2304389. [PMID: 37867228 PMCID: PMC10700174 DOI: 10.1002/advs.202304389] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 09/19/2023] [Indexed: 10/24/2023]
Abstract
Efficient and targeted delivery of therapeutic agents remains a bottleneck in modern medicine. Here, biochemical engineering approaches to advance the repurposing of extracellular vesicles (EVs) as drug delivery vehicles are explored. Targeting ligands such as the sugar GalNAc are displayed on the surface of EVs using a HaloTag-fused to a protein anchor that is enriched on engineered EVs. These EVs are successfully targeted to human primary hepatocytes. In addition, the authors are able to decorate EVs with an antibody that recognizes a GLP1 cell surface receptor by using an Fc and Fab region binding moiety fused to an anchor protein, and they show that this improves EV targeting to cells that overexpress the receptor. The authors also use two different protein-engineering approaches to improve the loading of Cre recombinase into the EV lumen and demonstrate that functional Cre protein is delivered into cells in the presence of chloroquine, an endosomal escape enhancer. Lastly, engineered EVs are well tolerated upon intravenous injection into mice without detectable signs of liver toxicity. Collectively, the data show that EVs can be engineered to improve cargo loading and specific cell targeting, which will aid their transformation into tailored drug delivery vehicles.
Collapse
Affiliation(s)
- Alena Ivanova
- Discovery BiologyDiscovery SciencesBioPharmaceuticals R&D, AstraZenecaPepparedsleden 1Mölndal431 50Sweden
| | - Lukas Badertscher
- Translational GenomicsDiscovery SciencesBioPharmaceuticals R&D, AstraZenecaPepparedsleden 1Mölndal431 50Sweden
- Present address:
Myllia Biotechnology GmbHAm Kanal 27Vienna1110Austria
| | - Gwen O'Driscoll
- Discovery BiologyDiscovery SciencesBioPharmaceuticals R&D, AstraZenecaPepparedsleden 1Mölndal431 50Sweden
- Advanced Drug DeliveryPharmaceutical SciencesBioPharmaceuticals R&D, AstraZenecaPepparedsleden 1Mölndal431 50Sweden
- Present address:
Division of Radiotherapy and ImagingThe Institute of Cancer ResearchLondonUK
| | - Joakim Bergman
- Medicinal ChemistryResearch and Early Development Cardiovascular, Renal and MetabolismBioPharmaceuticals R&D, AstraZenecaPepparedsleden 1Mölndal431 50Sweden
| | - Euan Gordon
- Discovery BiologyDiscovery SciencesBioPharmaceuticals R&D, AstraZenecaPepparedsleden 1Mölndal431 50Sweden
| | - Anders Gunnarsson
- Structure and BiophysicsDiscovery SciencesBioPharmaceuticals R&D, AstraZenecaPepparedsleden 1Mölndal431 50Sweden
| | - Camilla Johansson
- Clinical Pharmacology and Safety SciencesSweden Imaging HubBioPharmaceuticals R&D, AstraZenecaPepparedsleden 1Mölndal431 50Sweden
| | - Michael J. Munson
- Advanced Drug DeliveryPharmaceutical SciencesBioPharmaceuticals R&D, AstraZenecaPepparedsleden 1Mölndal431 50Sweden
| | - Cristiana Spinelli
- Research Institute of the McGill University Health CentreGlen SiteMcGill UniversityMontrealQuebecH4A 3J1Canada
| | - Sara Torstensson
- Translational GenomicsDiscovery SciencesBioPharmaceuticals R&D, AstraZenecaPepparedsleden 1Mölndal431 50Sweden
| | - Liisa Vilén
- DMPKResearch and Early Development Cardiovascular, Renal and MetabolismBioPharmaceuticals R&D, AstraZenecaPepparedsleden 1Mölndal431 50Sweden
| | - Andrei Voirel
- Medicinal ChemistryResearch and Early Development Cardiovascular, Renal and MetabolismBioPharmaceuticals R&D, AstraZenecaPepparedsleden 1Mölndal431 50Sweden
| | - John Wiseman
- Translational GenomicsDiscovery SciencesBioPharmaceuticals R&D, AstraZenecaPepparedsleden 1Mölndal431 50Sweden
| | - Janusz Rak
- Research Institute of the McGill University Health CentreGlen SiteMcGill UniversityMontrealQuebecH4A 3J1Canada
| | - Niek Dekker
- Discovery BiologyDiscovery SciencesBioPharmaceuticals R&D, AstraZenecaPepparedsleden 1Mölndal431 50Sweden
| | - Elisa Lázaro‐Ibáñez
- Discovery BiologyDiscovery SciencesBioPharmaceuticals R&D, AstraZenecaPepparedsleden 1Mölndal431 50Sweden
- Advanced Drug DeliveryPharmaceutical SciencesBioPharmaceuticals R&D, AstraZenecaPepparedsleden 1Mölndal431 50Sweden
| |
Collapse
|
43
|
Lee UH, Park SJ, Ju SA, Lee SC, Kim BS, Ahn B, Yi J, Park J, Won YW, Han IS, Lee BJ, Cho WJ, Park JW. DRG2 in macrophages is crucial for initial inflammatory response and protection against Listeria monocytogenes infection. Clin Immunol 2023; 257:109819. [PMID: 37918467 DOI: 10.1016/j.clim.2023.109819] [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: 07/06/2023] [Accepted: 10/19/2023] [Indexed: 11/04/2023]
Abstract
Innate immune response is critical for the control of Listeria monocytogenes infection. Here, we identified developmentally regulated GTP-binding protein 2 (DRG2) in macrophages as a major regulator of the innate immune response against L. monocytogenes infection. Both whole-body DRG2 knockout (KO) mice and macrophage-specific DRG2 KO mice had low levels of IL-6 during early infection and increased susceptibility to L. monocytogenes infection. Following an initial impaired inflammatory response of macrophages upon i.p. L. monocytogenes infection, DRG2-/- mice showed delayed recruitment of neutrophils and monocytes into the peritoneal cavity, which led to elevated bacterial burden, inflammatory cytokine production at a late infection time point, and liver micro-abscesses. DRG2 deficiency decreased the transcriptional activity of NF-κB and impaired the inflammatory response of both bone marrow-derived and peritoneal macrophages upon L. monocytogenes stimulation. Our findings reveal that DRG2 in macrophages is critical for the initial inflammatory response and protection against L. monocytogenes infection.
Collapse
Affiliation(s)
- Unn Hwa Lee
- Department of Biological Sciences, University of Ulsan, Ulsan 44610, Republic of Korea
| | - Sang Jin Park
- Department of Biological Sciences, University of Ulsan, Ulsan 44610, Republic of Korea
| | - Seong A Ju
- Department of Biological Sciences, University of Ulsan, Ulsan 44610, Republic of Korea
| | - Sang Chul Lee
- CRONEX Co., Ltd., Hwaseong-si, Gyeonggi-do 18333, Republic of Korea
| | - Byung Sam Kim
- Department of Biological Sciences, University of Ulsan, Ulsan 44610, Republic of Korea
| | - Byungyong Ahn
- Department of Food Science and Nutrition, University of Ulsan, Ulsan 44610, Republic of Korea; RopheLBio, B102, Seoul Forest M Tower, Seoul 04778, Republic of Korea
| | - Jawoon Yi
- School of Life Sciences, Gwangju Institute of Science and Technology, Gwangju 61005, Republic of Korea
| | - Jihwan Park
- School of Life Sciences, Gwangju Institute of Science and Technology, Gwangju 61005, Republic of Korea
| | - Young-Wook Won
- Department of Biomedical Engineering, University of North Texas, TX 76203-5017, USA; RopheLBio, B102, Seoul Forest M Tower, Seoul 04778, Republic of Korea
| | - In Seob Han
- Department of Biological Sciences, University of Ulsan, Ulsan 44610, Republic of Korea
| | - Byung Ju Lee
- Department of Biological Sciences, University of Ulsan, Ulsan 44610, Republic of Korea; Basic-Clinical Convergence Research Institute, University of Ulsan, Ulsan 44610, Republic of Korea
| | - Wha Ja Cho
- Department of Biological Sciences, University of Ulsan, Ulsan 44610, Republic of Korea
| | - Jeong Woo Park
- Department of Biological Sciences, University of Ulsan, Ulsan 44610, Republic of Korea; Basic-Clinical Convergence Research Institute, University of Ulsan, Ulsan 44610, Republic of Korea.
| |
Collapse
|
44
|
Tsissios G, Sallese A, Perez-Estrada JR, Tangeman JA, Chen W, Smucker B, Ratvasky SC, Grajales-Esquive EL, Martinez A, Visser KJ, Araus AJ, Wang H, Simon A, Yun MH, Rio-Tsonis KD. Macrophages modulate fibrosis during newt lens regeneration. RESEARCH SQUARE 2023:rs.3.rs-3603645. [PMID: 38045376 PMCID: PMC10690311 DOI: 10.21203/rs.3.rs-3603645/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/05/2023]
Abstract
Background Previous studies indicated that macrophages play a role during lens regeneration in newts, but their function has not been tested experimentally. Methods Here we generated a transgenic newt reporter line in which macrophages can be visualized in vivo. Using this new tool, we analyzed the location of macrophages during lens regeneration. We uncovered early gene expression changes using bulk RNAseq in two newt species, Notophthalmus viridescens and Pleurodeles waltl. Next, we used clodronate liposomes to deplete macrophages, which inhibited lens regeneration in both newt species. Results Macrophage depletion induced the formation of scar-like tissue, an increased and sustained inflammatory response, an early decrease in iris pigment epithelial cell (iPEC) proliferation and a late increase in apoptosis. Some of these phenotypes persisted for at least 100 days and could be rescued by exogenous FGF2. Re-injury alleviated the effects of macrophage depletion and re-started the regeneration process. Conclusions Together, our findings highlight the importance of macrophages in facilitating a pro-regenerative environment in the newt eye, helping to resolve fibrosis, modulating the overall inflammatory landscape and maintaining the proper balance of early proliferation and late apoptosis.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | - Maximina H Yun
- Dresden University of Technology: Technische Universitat Dresden
| | | |
Collapse
|
45
|
Thomas SK, Wattenberg MM, Choi-Bose S, Uhlik M, Harrison B, Coho H, Cassella CR, Stone ML, Patel D, Markowitz K, Delman D, Chisamore M, Drees J, Bose N, Beatty GL. Kupffer cells prevent pancreatic ductal adenocarcinoma metastasis to the liver in mice. Nat Commun 2023; 14:6330. [PMID: 37816712 PMCID: PMC10564762 DOI: 10.1038/s41467-023-41771-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Accepted: 09/12/2023] [Indexed: 10/12/2023] Open
Abstract
Although macrophages contribute to cancer cell dissemination, immune evasion, and metastatic outgrowth, they have also been reported to coordinate tumor-specific immune responses. We therefore hypothesized that macrophage polarization could be modulated therapeutically to prevent metastasis. Here, we show that macrophages respond to β-glucan (odetiglucan) treatment by inhibiting liver metastasis. β-glucan activated liver-resident macrophages (Kupffer cells), suppressed cancer cell proliferation, and invoked productive T cell-mediated responses against liver metastasis in pancreatic cancer mouse models. Although excluded from metastatic lesions, Kupffer cells were critical for the anti-metastatic activity of β-glucan, which also required T cells. Furthermore, β-glucan drove T cell activation and macrophage re-polarization in liver metastases in mice and humans and sensitized metastatic lesions to anti-PD1 therapy. These findings demonstrate the significance of macrophage function in metastasis and identify Kupffer cells as a potential therapeutic target against pancreatic cancer metastasis to the liver.
Collapse
Affiliation(s)
- Stacy K Thomas
- Division of Hematology-Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Max M Wattenberg
- Division of Hematology-Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Shaanti Choi-Bose
- Division of Hematology-Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Mark Uhlik
- HiberCell Inc, Roseville, MN, USA
- OncXerna, Waltham, MA, USA
| | | | - Heather Coho
- Division of Hematology-Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Christopher R Cassella
- Division of Hematology-Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Meredith L Stone
- Division of Hematology-Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Dhruv Patel
- Division of Hematology-Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Kelly Markowitz
- Division of Hematology-Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Devora Delman
- Division of Hematology-Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | | | | | | | - Gregory L Beatty
- Division of Hematology-Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
46
|
Shen J, Zhao S, Peng M, Li Y, Zhang L, Li X, Hu Y, Wu M, Xiang S, Wu X, Liu J, Zhang B, Chen Z, Lin D, Liu H, Tang W, Chen J, Sun X, Liao Q, Hide G, Zhou Z, Lun ZR, Wu Z. Macrophage-mediated trogocytosis contributes to destroying human schistosomes in a non-susceptible rodent host, Microtus fortis. Cell Discov 2023; 9:101. [PMID: 37794085 PMCID: PMC10550985 DOI: 10.1038/s41421-023-00603-6] [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: 03/23/2023] [Accepted: 09/13/2023] [Indexed: 10/06/2023] Open
Abstract
Schistosoma parasites, causing schistosomiasis, exhibit typical host specificity in host preference. Many mammals, including humans, are susceptible to infection, while the widely distributed rodent, Microtus fortis, exhibits natural anti-schistosome characteristics. The mechanisms of host susceptibility remain poorly understood. Comparison of schistosome infection in M. fortis with the infection in laboratory mice (highly sensitive to infection) offers a good model system to investigate these mechanisms and to gain an insight into host specificity. In this study, we showed that large numbers of leukocytes attach to the surface of human schistosomes in M. fortis but not in mice. Single-cell RNA-sequencing analyses revealed that macrophages might be involved in the cell adhesion, and we further demonstrated that M. fortis macrophages could be mediated to attach and kill schistosomula with dependence on Complement component 3 (C3) and Complement receptor 3 (CR3). Importantly, we provided direct evidence that M. fortis macrophages could destroy schistosomula by trogocytosis, a previously undescribed mode for killing helminths. This process was regulated by Ca2+/NFAT signaling. These findings not only elucidate a novel anti-schistosome mechanism in M. fortis but also provide a better understanding of host parasite interactions, host specificity and the potential generation of novel strategies for schistosomiasis control.
Collapse
Affiliation(s)
- Jia Shen
- Department of Parasitology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, Guangdong, China.
- Key Laboratory of Tropical Disease Control of the Ministry of Education, Sun Yat-sen University, Guangzhou, Guangdong, China.
- Provincial Engineering Technology Research Center for Biological Vector Control, Guangzhou, Guangdong, China.
| | - Siyu Zhao
- Department of Parasitology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, Guangdong, China
- Key Laboratory of Tropical Disease Control of the Ministry of Education, Sun Yat-sen University, Guangzhou, Guangdong, China
- Provincial Engineering Technology Research Center for Biological Vector Control, Guangzhou, Guangdong, China
| | - Mei Peng
- Department of Parasitology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, Guangdong, China
- Key Laboratory of Tropical Disease Control of the Ministry of Education, Sun Yat-sen University, Guangzhou, Guangdong, China
- Provincial Engineering Technology Research Center for Biological Vector Control, Guangzhou, Guangdong, China
| | - Yanguo Li
- Institute of Drug Discovery Technology, School of Public Health, School of Medicine, Ningbo University, Ningbo, Zhejiang, China
| | - Lichao Zhang
- Department of Parasitology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, Guangdong, China
- Key Laboratory of Tropical Disease Control of the Ministry of Education, Sun Yat-sen University, Guangzhou, Guangdong, China
- Provincial Engineering Technology Research Center for Biological Vector Control, Guangzhou, Guangdong, China
| | - Xiaoping Li
- Department of Hepatic Surgery and Liver Transplantation Center, Organ Transplantation Institute, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Yunyi Hu
- Department of Parasitology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, Guangdong, China
- Key Laboratory of Tropical Disease Control of the Ministry of Education, Sun Yat-sen University, Guangzhou, Guangdong, China
- Provincial Engineering Technology Research Center for Biological Vector Control, Guangzhou, Guangdong, China
| | - Mingrou Wu
- Department of Parasitology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, Guangdong, China
- Key Laboratory of Tropical Disease Control of the Ministry of Education, Sun Yat-sen University, Guangzhou, Guangdong, China
- Provincial Engineering Technology Research Center for Biological Vector Control, Guangzhou, Guangdong, China
| | - Suoyu Xiang
- Department of Parasitology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, Guangdong, China
- Key Laboratory of Tropical Disease Control of the Ministry of Education, Sun Yat-sen University, Guangzhou, Guangdong, China
- Provincial Engineering Technology Research Center for Biological Vector Control, Guangzhou, Guangdong, China
| | - Xiaoying Wu
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Jiahua Liu
- Department of Parasitology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, Guangdong, China
- Key Laboratory of Tropical Disease Control of the Ministry of Education, Sun Yat-sen University, Guangzhou, Guangdong, China
- Provincial Engineering Technology Research Center for Biological Vector Control, Guangzhou, Guangdong, China
| | - Beibei Zhang
- Department of Parasitology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, Guangdong, China
- Key Laboratory of Tropical Disease Control of the Ministry of Education, Sun Yat-sen University, Guangzhou, Guangdong, China
- Provincial Engineering Technology Research Center for Biological Vector Control, Guangzhou, Guangdong, China
| | - Zebin Chen
- Department of Hepatic Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Datao Lin
- Department of Parasitology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, Guangdong, China
- Key Laboratory of Tropical Disease Control of the Ministry of Education, Sun Yat-sen University, Guangzhou, Guangdong, China
- Provincial Engineering Technology Research Center for Biological Vector Control, Guangzhou, Guangdong, China
| | - Huanyao Liu
- Key Laboratory of Tropical Disease Control of the Ministry of Education, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Wenyan Tang
- Key Laboratory of Tropical Disease Control of the Ministry of Education, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Jun Chen
- Department of Immunology, Center for Precision Medicine and Engineering & Technology Research Center for Disease-Model Animals, Laboratory Animal Center, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Xi Sun
- Department of Parasitology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, Guangdong, China
- Key Laboratory of Tropical Disease Control of the Ministry of Education, Sun Yat-sen University, Guangzhou, Guangdong, China
- Provincial Engineering Technology Research Center for Biological Vector Control, Guangzhou, Guangdong, China
| | - Qi Liao
- Institute of Drug Discovery Technology, School of Public Health, School of Medicine, Ningbo University, Ningbo, Zhejiang, China
| | - Geoff Hide
- Biomedical Research and Innovation Centre, School of Science, Engineering and Environment, University of Salford, Salford, UK
| | - Zhijun Zhou
- Department of Laboratory Animals, Hunan Key Laboratory of Animal Models for Human Diseases, Central South University, Changsha, Hunan, China.
| | - Zhao-Rong Lun
- Biomedical Research and Innovation Centre, School of Science, Engineering and Environment, University of Salford, Salford, UK.
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-Sen University, Guangzhou, Guangdong, China.
| | - Zhongdao Wu
- Department of Parasitology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, Guangdong, China.
- Key Laboratory of Tropical Disease Control of the Ministry of Education, Sun Yat-sen University, Guangzhou, Guangdong, China.
- Provincial Engineering Technology Research Center for Biological Vector Control, Guangzhou, Guangdong, China.
| |
Collapse
|
47
|
Jordan CKI, Brown RL, Larkinson MLY, Sequeira RP, Edwards AM, Clarke TB. Symbiotic Firmicutes establish mutualism with the host via innate tolerance and resistance to control systemic immunity. Cell Host Microbe 2023; 31:1433-1449.e9. [PMID: 37582375 DOI: 10.1016/j.chom.2023.07.008] [Citation(s) in RCA: 18] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 05/12/2023] [Accepted: 07/18/2023] [Indexed: 08/17/2023]
Abstract
The intestinal microbiota regulates immunity across organ systems. Which symbionts control systemic immunity, the mechanisms they use, and how they avoid widespread inflammatory damage are unclear. We uncover host tolerance and resistance mechanisms that allow Firmicutes from the human microbiota to control systemic immunity without inducing immunopathology. Intestinal processing releases Firmicute glycoconjugates that disseminate, resulting in release of cytokine IL-34 that stimulates macrophages and enhances defenses against pneumonia, sepsis, and meningitis. Despite systemic penetration of Firmicutes, immune homeostasis is maintained through feedback control whereby IL-34-mediated mTORC1 activation in macrophages clears polymeric glycoconjugates from peripheral tissues. Smaller glycoconjugates evading this clearance mechanism are tolerated through sequestration by albumin, which acts as an inflammatory buffer constraining their immunological impact. Without these resistance and tolerance mechanisms, Firmicutes drive catastrophic organ damage and cachexia via IL-1β. This reveals how Firmicutes are safely assimilated into systemic immunity to protect against infection without threatening host viability.
Collapse
Affiliation(s)
- Christine K I Jordan
- Centre for Bacterial Resistance Biology, Department of Infectious Disease, Imperial College London, London SW7 2AZ, UK
| | - Rebecca L Brown
- Centre for Bacterial Resistance Biology, Department of Infectious Disease, Imperial College London, London SW7 2AZ, UK
| | - Max L Y Larkinson
- Centre for Bacterial Resistance Biology, Department of Infectious Disease, Imperial College London, London SW7 2AZ, UK
| | - Richard P Sequeira
- Centre for Bacterial Resistance Biology, Department of Infectious Disease, Imperial College London, London SW7 2AZ, UK
| | - Andrew M Edwards
- Centre for Bacterial Resistance Biology, Department of Infectious Disease, Imperial College London, London SW7 2AZ, UK
| | - Thomas B Clarke
- Centre for Bacterial Resistance Biology, Department of Infectious Disease, Imperial College London, London SW7 2AZ, UK.
| |
Collapse
|
48
|
Ilahibaks NF, Roefs MT, Brans MAD, Blok CS, de Jager SCA, Schiffelers RM, Vader P, Lei Z, Sluijter JPG. Extracellular vesicle-mediated protein delivery to the liver. JOURNAL OF EXTRACELLULAR BIOLOGY 2023; 2:e97. [PMID: 38938374 PMCID: PMC11080727 DOI: 10.1002/jex2.97] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Revised: 05/27/2023] [Accepted: 06/08/2023] [Indexed: 06/29/2024]
Abstract
Extracellular vesicles (EVs) are nanoscale particles that facilitate intercellular communication. They are regarded as a promising natural drug delivery system for transporting and delivering bioactive macromolecules to target cells. Recently, researchers have engineered EVs with FKBP12/FRB heterodimerization domains that interact with rapamycin to load and deliver exogenous proteins for both in vitro and in vivo applications. In this study, we examined the tissue distribution of EVs using near-infrared fluorescent imaging. We evaluated the effectiveness of EV-mediated delivery of Cre recombinase specifically to hepatocytes in the livers of Ai9 Cre-loxP reporter mice. Intravenous injection resulted in more efficient Cre protein delivery to the liver than intraperitoneal injections. Depleting liver-resident macrophages with clodronate-encapsulated liposome pre-treatment did not enhance EV-mediated Cre delivery to hepatocytes. Moreover, we demonstrated that multiple intravenous injections of Cre-EVs facilitated functional Cre delivery to hepatocytes. To the best of our knowledge, this is the first study to simultaneously investigate the tissue distribution of FKBP12/FRB-engineered EVs and their subsequent intracellular protein delivery in Ai9 Cre-loxP reporter mice. These insights can inform preclinical research and contribute to developing next-generation EV-based platforms for delivering therapeutic proteins or genome editing technologies targeting the liver.
Collapse
Affiliation(s)
- Nazma F. Ilahibaks
- Laboratory of Experimental Cardiology, Department Heart & LungsUniversity Medical Center UtrechtUtrechtThe Netherlands
| | - Marieke T. Roefs
- Laboratory of Experimental Cardiology, Department Heart & LungsUniversity Medical Center UtrechtUtrechtThe Netherlands
| | - Maike A. D. Brans
- Laboratory of Experimental Cardiology, Department Heart & LungsUniversity Medical Center UtrechtUtrechtThe Netherlands
| | - Christian Snijders Blok
- Laboratory of Experimental Cardiology, Department Heart & LungsUniversity Medical Center UtrechtUtrechtThe Netherlands
| | - Saskia C. A. de Jager
- Laboratory of Experimental Cardiology, Department Heart & LungsUniversity Medical Center UtrechtUtrechtThe Netherlands
| | | | - Pieter Vader
- Laboratory of Experimental Cardiology, Department Heart & LungsUniversity Medical Center UtrechtUtrechtThe Netherlands
- CDL ResearchUniversity Medical Center UtrechtUtrechtThe Netherlands
| | - Zhiyong Lei
- Laboratory of Experimental Cardiology, Department Heart & LungsUniversity Medical Center UtrechtUtrechtThe Netherlands
- CDL ResearchUniversity Medical Center UtrechtUtrechtThe Netherlands
| | - Joost P. G. Sluijter
- Laboratory of Experimental Cardiology, Department Heart & LungsUniversity Medical Center UtrechtUtrechtThe Netherlands
- Circulatory Health Laboratory, Regenerative Medicine CenterUniversity Medical Center Utrecht, University UtrechtUtrechtThe Netherlands
| |
Collapse
|
49
|
Parada-Kusz M, Clatworthy AE, Goering ER, Blackwood SM, Salm EJ, Choi C, Combs S, Lee JSW, Rodriguez-Osorio C, Tomita S, Hung DT. A tryptophan metabolite modulates the host response to bacterial infection via kainate receptors. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.16.553532. [PMID: 37645903 PMCID: PMC10462041 DOI: 10.1101/2023.08.16.553532] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/31/2023]
Abstract
Bacterial infection involves a complex interaction between the pathogen and host where the outcome of infection is not solely determined by pathogen eradication. To identify small molecules that promote host survival by altering the host-pathogen dynamic, we conducted an in vivo chemical screen using zebrafish embryos and found that treatment with 3-hydroxy-kynurenine protects from lethal gram-negative bacterial infection. 3-hydroxy-kynurenine, a metabolite produced through host tryptophan metabolism, has no direct antibacterial activity but enhances host survival by restricting bacterial expansion in macrophages by targeting kainate-sensitive glutamate receptors. These findings reveal new mechanisms by which tryptophan metabolism and kainate-sensitive glutamate receptors function and interact to modulate immunity, with significant implications for the coordination between the immune and nervous systems in pathological conditions.
Collapse
Affiliation(s)
- Margarita Parada-Kusz
- Department of Molecular Biology and Center for Computational and Integrative Biology, Massachusetts General Hospital; Boston, Massachusetts, USA
- Department of Genetics, Harvard Medical School; Boston, Massachusetts, USA
- Broad Institute of MIT and Harvard; Cambridge, Massachusetts, USA
| | - Anne E. Clatworthy
- Department of Molecular Biology and Center for Computational and Integrative Biology, Massachusetts General Hospital; Boston, Massachusetts, USA
- Department of Genetics, Harvard Medical School; Boston, Massachusetts, USA
- Broad Institute of MIT and Harvard; Cambridge, Massachusetts, USA
| | - Emily R. Goering
- Department of Molecular Biology and Center for Computational and Integrative Biology, Massachusetts General Hospital; Boston, Massachusetts, USA
- Department of Genetics, Harvard Medical School; Boston, Massachusetts, USA
- Broad Institute of MIT and Harvard; Cambridge, Massachusetts, USA
| | - Stephanie M. Blackwood
- Department of Molecular Biology and Center for Computational and Integrative Biology, Massachusetts General Hospital; Boston, Massachusetts, USA
- Broad Institute of MIT and Harvard; Cambridge, Massachusetts, USA
| | - Elizabeth J. Salm
- Department of Cellular and Molecular Physiology and Neuroscience, Yale School of Medicine; New Haven, Connecticut, USA
| | - Catherine Choi
- Department of Molecular Biology and Center for Computational and Integrative Biology, Massachusetts General Hospital; Boston, Massachusetts, USA
- Broad Institute of MIT and Harvard; Cambridge, Massachusetts, USA
| | - Senya Combs
- Department of Molecular Biology and Center for Computational and Integrative Biology, Massachusetts General Hospital; Boston, Massachusetts, USA
- Broad Institute of MIT and Harvard; Cambridge, Massachusetts, USA
| | - Jenny S. W. Lee
- Department of Molecular Biology and Center for Computational and Integrative Biology, Massachusetts General Hospital; Boston, Massachusetts, USA
- Broad Institute of MIT and Harvard; Cambridge, Massachusetts, USA
| | - Carlos Rodriguez-Osorio
- Department of Molecular Biology and Center for Computational and Integrative Biology, Massachusetts General Hospital; Boston, Massachusetts, USA
- Department of Genetics, Harvard Medical School; Boston, Massachusetts, USA
- Broad Institute of MIT and Harvard; Cambridge, Massachusetts, USA
| | - Susumu Tomita
- Department of Cellular and Molecular Physiology and Neuroscience, Yale School of Medicine; New Haven, Connecticut, USA
| | - Deborah T. Hung
- Department of Molecular Biology and Center for Computational and Integrative Biology, Massachusetts General Hospital; Boston, Massachusetts, USA
- Department of Genetics, Harvard Medical School; Boston, Massachusetts, USA
- Broad Institute of MIT and Harvard; Cambridge, Massachusetts, USA
| |
Collapse
|
50
|
Ander SE, Parks MG, Davenport BJ, Li FS, Bosco-Lauth A, Carpentier KS, Sun C, Lucas CJ, Klimstra WB, Ebel GD, Morrison TE. Phagocyte-expressed glycosaminoglycans promote capture of alphaviruses from the blood circulation in a host species-specific manner. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.09.552690. [PMID: 37609165 PMCID: PMC10441409 DOI: 10.1101/2023.08.09.552690] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/24/2023]
Abstract
The magnitude and duration of vertebrate viremia are critical determinants of arbovirus transmission, geographic spread, and disease severity-yet, mechanisms determining arbovirus viremia levels are poorly defined. Previous studies have drawn associations between in vitro virion-glycosaminoglycan (GAG) interactions and in vivo clearance kinetics of virions from blood circulation. From these observations, it is commonly hypothesized that GAG-binding virions are rapidly removed from circulation due to ubiquitous expression of GAGs by vascular endothelial cells, thereby limiting viremia. Using an in vivo model for viremia, we compared the vascular clearance of low and enhanced GAG-binding viral variants of chikungunya (CHIKV), eastern-(EEEV), and Venezuelan-(VEEV) equine encephalitis viruses. We find GAG-binding virions are more quickly removed from circulation than their non-GAG-binding variant; however individual clearance kinetics vary between GAG-binding viruses, from swift (VEEV) to slow removal from circulation (EEEV). Remarkably, we find phagocytes are required for efficient vascular clearance of some enhanced GAG-binding virions. Moreover, transient depletion of vascular heparan sulfate (HS) impedes vascular clearance of only some GAG-binding viral variants and in a phagocyte-dependent manner, implying phagocytes can mediate vascular GAG-virion interactions. Finally, in direct contrast to mice, we find enhanced GAG-binding EEEV is resistant to vascular clearance in avian hosts, suggesting the existence of species-specificity in virion-GAG interactions. In summary, these data support a role for GAG-mediated clearance of some viral particles from the blood circulation, illuminate the potential of blood-contacting phagocytes as a site for GAG-virion binding, and suggest a role for species-specific GAG structures in arbovirus ecology. Significance Statement Previously, evidence of arbovirus-GAG interactions in vivo has been limited to associations between viral residues shown to promote enhanced GAG-binding phenotypes in vitro and in vivo phenotypes of viral dissemination and pathogenesis. By directly manipulating host GAG expression, we identified virion-GAG interactions in vivo and discovered a role for phagocyte-expressed GAGs in viral vascular clearance. Moreover, we observe species-specific differences in viral vascular clearance of enhanced GAG-binding virions between murine and avian hosts. These data suggest species-specific variation in GAG structure is a mechanism to distinguish amplifying from dead-end hosts for arbovirus transmission.
Collapse
|