1
|
Zou Z, Liu X, Yu J, Ban T, Zhang Z, Wang P, Huang R, Zheng F, Chang Y, Peng W, Tang Y, Feng X, Zhao Z, Lv X, Huang S, Guo J, Tuo Y, Zhou Z, Liang S. Nuclear miR-204-3p mitigates metabolic dysfunction-associated steatotic liver disease in mice. J Hepatol 2024; 80:834-845. [PMID: 38331323 DOI: 10.1016/j.jhep.2024.01.029] [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: 04/06/2023] [Revised: 01/16/2024] [Accepted: 01/29/2024] [Indexed: 02/10/2024]
Abstract
BACKGROUND & AIMS Accumulating evidence has indicated the presence of mature microRNAs (miR) in the nucleus, but their effects on steatohepatitis remain elusive. We have previously demonstrated that the intranuclear miR-204-3p in macrophages protects against atherosclerosis, which shares multiple risk factors with metabolic dysfunction-associated steatotic liver disease (MASLD). Herein, we aimed to explore the functional significance of miR-204-3p in steatohepatitis. METHODS miR-204-3p levels and subcellular localization were assessed in the livers and peripheral blood mononuclear cells of patients with MASLD. Wild-type mice fed high-fat or methionine- and choline-deficient diets were injected with an adeno-associated virus system containing miR-204-3p to determine the effect of miR-204-3p on steatohepatitis. Co-culture systems were applied to investigate the crosstalk between macrophages and hepatocytes or hepatic stellate cells (HSCs). Multiple high-throughput epigenomic sequencings were performed to explore miR-204-3p targets. RESULTS miR-204-3p expression decreased in livers and macrophages in mice and patients with fatty liver. In patients with MASLD, miR-204-3p levels in peripheral blood mononuclear cells were inversely related to the severity of hepatic inflammation and damage. Macrophage-specific miR-204-3p overexpression reduced steatohepatitis in high-fat or methionine- and choline-deficient diet-fed mice. miR-204-3p-overexpressing macrophages inhibited TLR4/JNK signaling and pro-inflammatory cytokine release, thereby limiting fat deposition and inflammation in hepatocytes and fibrogenic activation in HSCs. Epigenomic profiling identified miR-204-3p as a specific regulator of ULK1 expression. ULK1 transcription and VPS34 complex activation by intranuclear miR-204-3p improved autophagic flux, promoting the anti-inflammatory effects of miR-204-3p in macrophages. CONCLUSIONS miR-204-3p inhibits macrophage inflammation, coordinating macrophage actions on hepatocytes and HSCs to ameliorate steatohepatitis. Macrophage miR-204-3p may be a therapeutic target for MASLD. IMPACT AND IMPLICATIONS Metabolic dysfunction-associated steatotic liver disease (MASLD) is a chronic inflammatory disease ranging from simple steatosis to steatohepatitis. However, the molecular mechanisms underlying the progression of MASLD remain incompletely understood. Here, we demonstrate that miR-204-3p levels in circulating peripheral blood mononuclear cells are negatively correlated with disease severity in patients with MASLD. Nuclear miR-204-3p activates ULK1 transcription and improves autophagic flux, limiting macrophage activation and hepatic steatosis. Our study provides a novel understanding of the mechanism of macrophage autophagy and inflammation in steatohepatitis and suggests that miR-204-3p may act as a potential therapeutic target for MASLD.
Collapse
Affiliation(s)
- Zhaowei Zou
- Department of General Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, China
| | - Xiu Liu
- Department of Cardiovascular Surgery, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Jie Yu
- Department of General Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, China; Department of Gastrointestinal Surgery, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510080, China
| | - Tao Ban
- Department of General Surgery, The Fourth Affiliated Hospital of Harbin Medical University, Harbin 150081, China; Department of Pharmacology (State Key Laboratory of Frigid Zone Cardiovascular Diseases, Ministry of Science and Technology; The Key Laboratory of Cardiovascular Research, Ministry of Education) at College of Pharmacy, Harbin Medical University, Harbin 150081, China; Heilongjiang Academy of Medical Sciences, Harbin 150081, China
| | - Ziyi Zhang
- Department of Pharmacology, Cardiac and Cerebral Vascular Research Center, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou 510080, China
| | - Peiqi Wang
- Department of General Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, China
| | - Renli Huang
- Department of General Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, China
| | - Fuxin Zheng
- Department of General Surgery, Beihai Hospital, Guangxi University of Chinese Medicine, Beihai 536000, China
| | - Yafei Chang
- Faculty of Forensic Medicine, Guangdong Province Translational Forensic Medicine Engineering Technology Research Center, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou 510080, China
| | - Wanli Peng
- Department of Pharmacology, Cardiac and Cerebral Vascular Research Center, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou 510080, China
| | - Yubo Tang
- Department of Pharmacy, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, China
| | - Xiaoqing Feng
- Department of Pharmacy, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, China
| | - Ziying Zhao
- Department of Pharmacy, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, China
| | - Xiaofei Lv
- Department of Pharmacology, Cardiac and Cerebral Vascular Research Center, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou 510080, China
| | - Shuai Huang
- Department of Orthopaedic Surgery, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou 510260, China
| | - Jiawei Guo
- Department of Pharmacology, School of Medicine, Yangtze University, Jingzhou 434023, China
| | - Yonghua Tuo
- Department of Neurosurgery, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou 510260, China
| | - Zhijun Zhou
- Department of Medicine, The University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, United States
| | - Sijia Liang
- Department of Pharmacology, Cardiac and Cerebral Vascular Research Center, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou 510080, China; Advanced Medical Technology Center, The First Affiliated Hospital, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou 510080, China.
| |
Collapse
|
2
|
Wang Q, Li T, Fang C, Zhang B. Bioinformatics analysis of the wheel treadmill test on motor function recovery after spinal cord injury. IBRAIN 2021; 7:265-277. [PMID: 37786556 PMCID: PMC10529348 DOI: 10.1002/ibra.12006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/17/2021] [Revised: 11/09/2021] [Accepted: 11/10/2021] [Indexed: 02/05/2023]
Abstract
This study aimed to explore the possible target and mechanism of the wheel treadmill (WTM) test for motor function recovery of spinal cord injury (SCI). Rats were divided into sham, control and WTM groups to establish an SCI mode. Rats in the WTM group were trained on the WTM test, and Basso-Beattie-Bresnahan (BBB) scores were determined. The samples were collected, and mRNA sequencing was conducted to determine the changes in gene expression. The coexpressed genes were screened to construct a protein-protein interaction (PPI), followed by the Kyoto Encyclopedia of Genes and Genomes pathway and Gene Ontology function enrichment analysis, and the differentially expressed genes (DEGs) volcano map and hub gene expression heat map were constructed using R language. The BBB scores in the control and WTM groups increased with time, with the WTM group scoring higher than the control group. The results of rat spinal cord tissue sequencing showed that a total of 1679 DEGs were screened in the sham and control groups; 928 DEGs and 731 overlapping genes were screened in the WTM and control groups. The key genes were identified by PPI analysis. One hundred and thirty-three genes were found to be overlapping by combined analysis of spinal cord sequencing data and BBB scores of rats at Week 7. The top 10 DEGs from high to low were Tyrobp, Rac2, Cd68, C1qb, Aif1, Cd74, Spi1, Fcer1g, RT1-DA, and Ccl4. The terms with the highest enrichment scores were microglia-mediated positive regulation of cytotoxicity and major histocompatibility complex class II protein complexes. Treatment with the WTM test promotes recovery of motor function after SCI in rats by modulating intercellular communication and immune function.
Collapse
Affiliation(s)
- Qiu‐Lin Wang
- School of AnesthesiologySouthwest Medical UniversityLuzhouSichuanChina
| | - Ting‐Ting Li
- Department of Anesthesiology, Institute of Neurological Disease, West China HospitalSichuan UniversityChengduChina
| | - Chang‐Le Fang
- School of AnesthesiologySouthwest Medical UniversityLuzhouSichuanChina
| | - Bao‐Lei Zhang
- Department of Experimental ZoologyKunming Medical UniversityKunmingYunnanChina
| |
Collapse
|
3
|
Han CY, Kang I, Harten IA, Gebe JA, Chan CK, Omer M, Alonge KM, den Hartigh LJ, Gomes Kjerulf D, Goodspeed L, Subramanian S, Wang S, Kim F, Birk DE, Wight TN, Chait A. Adipocyte-Derived Versican and Macrophage-Derived Biglycan Control Adipose Tissue Inflammation in Obesity. Cell Rep 2021; 31:107818. [PMID: 32610121 DOI: 10.1016/j.celrep.2020.107818] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Revised: 04/20/2020] [Accepted: 06/04/2020] [Indexed: 12/18/2022] Open
Abstract
Obesity is characterized by adipose tissue inflammation. Because proteoglycans regulate inflammation, here we investigate their role in adipose tissue inflammation in obesity. We find that adipose tissue versican and biglycan increase in obesity. Versican is produced mainly by adipocytes and biglycan by adipose tissue macrophages. Both proteoglycans are also present in adipose tissue from obese human subjects undergoing gastric bypass surgery. Deletion of adipocyte-specific versican or macrophage-specific biglycan in mice reduces macrophage accumulation and chemokine and cytokine expression, although only adipocyte-specific versican deletion leads to sustained improvement in glucose tolerance. Macrophage-derived biglycan activates inflammatory genes in adipocytes. Versican expression increases in cultured adipocytes exposed to excess glucose, and adipocyte-conditioned medium stimulates inflammation in resident peritoneal macrophages, in part because of a versican breakdown product, versikine. These findings provide insights into the role of adipocyte- and macrophage-derived proteoglycans in adipose tissue inflammation in obesity.
Collapse
Affiliation(s)
- Chang Yeop Han
- Department of Medicine, Division of Metabolism, Endocrinology, and Nutrition, University of Washington, Seattle, WA, USA
| | - Inkyung Kang
- Matrix Biology Program, Benaroya Research Institute, Seattle, WA, USA
| | - Ingrid A Harten
- Matrix Biology Program, Benaroya Research Institute, Seattle, WA, USA
| | - John A Gebe
- Matrix Biology Program, Benaroya Research Institute, Seattle, WA, USA
| | - Christina K Chan
- Matrix Biology Program, Benaroya Research Institute, Seattle, WA, USA
| | - Mohamed Omer
- Department of Medicine, Division of Metabolism, Endocrinology, and Nutrition, University of Washington, Seattle, WA, USA
| | - Kimberly M Alonge
- Department of Medicine, Division of Metabolism, Endocrinology, and Nutrition, University of Washington, Seattle, WA, USA
| | - Laura J den Hartigh
- Department of Medicine, Division of Metabolism, Endocrinology, and Nutrition, University of Washington, Seattle, WA, USA
| | - Diego Gomes Kjerulf
- Department of Medicine, Division of Metabolism, Endocrinology, and Nutrition, University of Washington, Seattle, WA, USA
| | - Leela Goodspeed
- Department of Medicine, Division of Metabolism, Endocrinology, and Nutrition, University of Washington, Seattle, WA, USA
| | - Savitha Subramanian
- Department of Medicine, Division of Metabolism, Endocrinology, and Nutrition, University of Washington, Seattle, WA, USA
| | - Shari Wang
- Department of Medicine, Division of Metabolism, Endocrinology, and Nutrition, University of Washington, Seattle, WA, USA
| | - Francis Kim
- Division of Cardiology, University of Washington, Seattle, WA, USA
| | - David E Birk
- Department of Molecular Pharmacology & Physiology, University of South Florida, Tampa, FL, USA
| | - Thomas N Wight
- Matrix Biology Program, Benaroya Research Institute, Seattle, WA, USA
| | - Alan Chait
- Department of Medicine, Division of Metabolism, Endocrinology, and Nutrition, University of Washington, Seattle, WA, USA.
| |
Collapse
|
4
|
Viaud M, Abdel-Wahab O, Gall J, Ivanov S, Guinamard R, Sore S, Merlin J, Ayrault M, Guilbaud E, Jacquel A, Auberger P, Wang N, Levine RL, Tall AR, Yvan-Charvet L. ABCA1 Exerts Tumor-Suppressor Function in Myeloproliferative Neoplasms. Cell Rep 2021; 30:3397-3410.e5. [PMID: 32160545 PMCID: PMC7473128 DOI: 10.1016/j.celrep.2020.02.056] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2019] [Revised: 11/23/2019] [Accepted: 02/12/2020] [Indexed: 12/13/2022] Open
Abstract
Defective cholesterol efflux pathways in mice promote the expansion of hematopoietic stem and progenitor cells and a bias toward the myeloid lineage, as observed in chronic myelomonocytic leukemia (CMML). Here, we identify 5 somatic missense mutations in ABCA1 in 26 patients with CMML. These mutations confer a proliferative advantage to monocytic leukemia cell lines in vitro. In vivo inactivation of ABCA1 or expression of ABCA1 mutants in hematopoietic cells in the setting of Tet2 loss demonstrates a myelosuppressive function of ABCA1. Mechanistically, ABCA1 mutations impair the tumor-suppressor functions of WT ABCA1 in myeloproliferative neoplasms by increasing the IL-3Rβ signaling via MAPK and JAK2 and subsequent metabolic reprogramming. Overexpression of a human apolipoprotein A-1 transgene dampens myeloproliferation. These findings identify somatic mutations in ABCA1 that subvert its anti-proliferative and cholesterol efflux functions and permit the progression of myeloid neoplasms. Therapeutic increases in HDL bypass these defects and restore normal hematopoiesis. Viaud et al. show that ABCA1 mutants identified in CMML patients diminish the tumor-suppressor functions of ABCA1 and cooperate with Tet2 loss to confer the hypersensitivity of myeloid progenitors to IL-3 receptor β canonical signaling, which can be prevented by raising HDL levels.
Collapse
Affiliation(s)
- Manon Viaud
- Institut National de la Santé et de la Recherche Médicale (INSERM) U1065, Université Côte d'Azur, Centre Méditerranéen de Médecine Moléculaire (C3M), Atip-Avenir, Fédération Hospitalo-Universitaire (FHU) Oncoage, 06204 Nice, France
| | - Omar Abdel-Wahab
- Human Oncology and Pathogenesis Program and Leukemia Service, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Julie Gall
- Institut National de la Santé et de la Recherche Médicale (INSERM) U1065, Université Côte d'Azur, Centre Méditerranéen de Médecine Moléculaire (C3M), Atip-Avenir, Fédération Hospitalo-Universitaire (FHU) Oncoage, 06204 Nice, France
| | - Stoyan Ivanov
- Institut National de la Santé et de la Recherche Médicale (INSERM) U1065, Université Côte d'Azur, Centre Méditerranéen de Médecine Moléculaire (C3M), Atip-Avenir, Fédération Hospitalo-Universitaire (FHU) Oncoage, 06204 Nice, France
| | - Rodolphe Guinamard
- Institut National de la Santé et de la Recherche Médicale (INSERM) U1065, Université Côte d'Azur, Centre Méditerranéen de Médecine Moléculaire (C3M), Atip-Avenir, Fédération Hospitalo-Universitaire (FHU) Oncoage, 06204 Nice, France
| | - Sophie Sore
- Institut National de la Santé et de la Recherche Médicale (INSERM) U1065, Université Côte d'Azur, Centre Méditerranéen de Médecine Moléculaire (C3M), Atip-Avenir, Fédération Hospitalo-Universitaire (FHU) Oncoage, 06204 Nice, France
| | - Johanna Merlin
- Institut National de la Santé et de la Recherche Médicale (INSERM) U1065, Université Côte d'Azur, Centre Méditerranéen de Médecine Moléculaire (C3M), Atip-Avenir, Fédération Hospitalo-Universitaire (FHU) Oncoage, 06204 Nice, France
| | - Marion Ayrault
- Institut National de la Santé et de la Recherche Médicale (INSERM) U1065, Université Côte d'Azur, Centre Méditerranéen de Médecine Moléculaire (C3M), Atip-Avenir, Fédération Hospitalo-Universitaire (FHU) Oncoage, 06204 Nice, France
| | - Emma Guilbaud
- Institut National de la Santé et de la Recherche Médicale (INSERM) U1065, Université Côte d'Azur, Centre Méditerranéen de Médecine Moléculaire (C3M), Atip-Avenir, Fédération Hospitalo-Universitaire (FHU) Oncoage, 06204 Nice, France
| | - Arnaud Jacquel
- Institut National de la Santé et de la Recherche Médicale (INSERM) U1065, Université Côte d'Azur, Centre Méditerranéen de Médecine Moléculaire (C3M), Atip-Avenir, Fédération Hospitalo-Universitaire (FHU) Oncoage, 06204 Nice, France
| | - Patrick Auberger
- Institut National de la Santé et de la Recherche Médicale (INSERM) U1065, Université Côte d'Azur, Centre Méditerranéen de Médecine Moléculaire (C3M), Atip-Avenir, Fédération Hospitalo-Universitaire (FHU) Oncoage, 06204 Nice, France
| | - Nan Wang
- Division of Molecular Medicine, Department of Medicine, Columbia University, New York, NY 10032, USA
| | - Ross L Levine
- Human Oncology and Pathogenesis Program and Leukemia Service, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Alan R Tall
- Division of Molecular Medicine, Department of Medicine, Columbia University, New York, NY 10032, USA
| | - Laurent Yvan-Charvet
- Institut National de la Santé et de la Recherche Médicale (INSERM) U1065, Université Côte d'Azur, Centre Méditerranéen de Médecine Moléculaire (C3M), Atip-Avenir, Fédération Hospitalo-Universitaire (FHU) Oncoage, 06204 Nice, France.
| |
Collapse
|
5
|
Chan MWY, Viswanathan S. Recent progress on developing exogenous monocyte/macrophage-based therapies for inflammatory and degenerative diseases. Cytotherapy 2019; 21:393-415. [PMID: 30871899 DOI: 10.1016/j.jcyt.2019.02.002] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2018] [Revised: 02/04/2019] [Accepted: 02/13/2019] [Indexed: 12/14/2022]
Abstract
Cell-based therapies are a rapidly developing area of regenerative medicine as dynamic treatments that execute therapeutic functions multimodally. Monocytes and macrophages, as innate immune cells that control inflammation and tissue repair, are increasing popular clinical candidates due to their spectrum of functionality. In this article, we review the role of monocytes and macrophages specifically in inflammatory and degenerative disease pathology and the evidence supporting the use of these cells as an effective therapeutic strategy. We compare current strategies of exogenously polarized monocyte/macrophage therapies regarding dosage, delivery and processing to identify outcomes, advances and challenges to their clinical use. Monocytes/macrophages hold the potential to be a promising therapeutic avenue but understanding and optimization of disease-specific efficacy is needed to accelerate their clinical use.
Collapse
Affiliation(s)
- Mable Wing Yan Chan
- Arthritis Program, Krembil Research Institute, University Health Network, Toronto, Ontario, Canada; Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada
| | - Sowmya Viswanathan
- Arthritis Program, Krembil Research Institute, University Health Network, Toronto, Ontario, Canada; Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada; Cell Therapy Program, University Health Network, Toronto, Ontario, Canada; Division of Hematology, Department of Medicine, University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
6
|
McCurdy S, Baumer Y, Toulmin E, Lee BH, Boisvert WA. Macrophage-Specific Expression of IL-37 in Hyperlipidemic Mice Attenuates Atherosclerosis. THE JOURNAL OF IMMUNOLOGY 2017; 199:3604-3613. [DOI: 10.4049/jimmunol.1601907] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/08/2016] [Accepted: 09/12/2017] [Indexed: 01/05/2023]
|
7
|
Alessandrini F, Pezzè L, Ciribilli Y. LAMPs: Shedding light on cancer biology. Semin Oncol 2017; 44:239-253. [PMID: 29526252 DOI: 10.1053/j.seminoncol.2017.10.013] [Citation(s) in RCA: 94] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2017] [Revised: 10/27/2017] [Accepted: 10/29/2017] [Indexed: 01/09/2023]
Abstract
Lysosomes are important cytoplasmic organelles whose critical functions in cells are increasingly being understood. In particular, despite the long-standing accepted concept about the role of lysosomes as cellular machineries solely assigned to degradation, it has been demonstrated that they play active roles in homeostasis and even in cancer biology. Indeed, it is now well documented that during the process of cellular transformation and cancer progression lysosomes are changing localization, composition, and volume and, through the release of their enzymes, lysosomes can also enhance cancer aggressiveness. LAMPs (lysosome associated membrane proteins) represent a family of glycosylated proteins present predominantly on the membrane of lysosomes whose expression can vary among different tissues, suggesting a separation of functions. In this review we focus on the functions and roles of the different LAMP family members, with a particular emphasis on cancer progression and metastatic spread. LAMP proteins are involved in many different aspects of cell biology and can influence cellular processes such as phagocytosis, autophagy, lipid transport, and aging. Interestingly, for all the five members identified so far (LAMP1, LAMP2, LAMP3, CD68/Macrosialin/LAMP4, and BAD-LAMP/LAMP5), a role in cancer has been suggested. While this is well documented for LAMP1 and LAMP2, the involvement of the other three proteins in cancer progression and aggressiveness has recently been proposed and remains to be elucidated. Here we present different examples about how LAMP proteins can influence and support tumor growth and metastatic spread, emphasizing the impact of each single member of the family.
Collapse
Affiliation(s)
- Federica Alessandrini
- Laboratory of Molecular Cancer Genetics, Centre for Integrative Biology (CIBIO), University of Trento, Povo (TN), Italy
| | - Laura Pezzè
- Laboratory of Molecular Cancer Genetics, Centre for Integrative Biology (CIBIO), University of Trento, Povo (TN), Italy
| | - Yari Ciribilli
- Laboratory of Molecular Cancer Genetics, Centre for Integrative Biology (CIBIO), University of Trento, Povo (TN), Italy.
| |
Collapse
|
8
|
Hairy/enhancer of Split Homologue-1 Suppresses Vascular Endothelial Growth Factor-induced Angiogenesis via Downregulation of Osteopontin Expression. Sci Rep 2017; 7:898. [PMID: 28420872 PMCID: PMC5429857 DOI: 10.1038/s41598-017-01018-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2016] [Accepted: 03/23/2017] [Indexed: 01/11/2023] Open
Abstract
Angiogenesis plays a critical role in the progression and vulnerability of atherosclerotic plaques; however, the orchestration of angiogenesis in atherosclerotic plaque formation remains unclear. The results of microarray analysis, real-time PCR and immunohistochemical analyses showed that Hairy/enhancer of split homologue-1 (Hes-1) expression was significantly decreased, while that of osteopontin (OPN) was increased, in atherosclerotic plaques. Meanwhile, immunofluorescence results demonstrated that both Hes-1 and OPN were expressed in endothelial cells (ECs) of neovessels in atherosclerotic plaques. The results of an in vitro study showed that Hes-1 was downregulated, while OPN was upregulated, in a time- and dose-dependent manner in human umbilical vein endothelial cells (HUVECs) by VEGF treatment. In addition, Hes-1 knockdown was found to have transcriptional promotion effect on OPN expression in HUVECs and enhance OPN-induced angiogenesis in response to VEGF. On the contrary, Hes-1 overexpression inhibited OPN expression in HUVECs and reduced angiogenesis in vitro and in vivo. The results of this study suggest that decreased Hes-1 expression in atherosclerotic plaques exaggerate VEGF-induced angiogenesis by upregulating OPN. Therefore, restoring Hes-1 expression and inhibiting OPN expression may be a promising strategy to prevent vulnerable plaque formation in patients with atherosclerosis.
Collapse
|
9
|
Jackson MF, Scatena M, Giachelli CM. Osteoclast precursors do not express CD68: results from CD68 promoter-driven RANK transgenic mice. FEBS Lett 2017; 591:728-736. [PMID: 28173622 DOI: 10.1002/1873-3468.12588] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2016] [Revised: 01/30/2017] [Accepted: 01/31/2017] [Indexed: 12/22/2022]
Abstract
Macrophages and osteoclasts are thought to derive from CD68 lineage marker-positive common myeloid precursors. We used the CD68 promoter to drive an inducible receptor activator of NF-κB (iRANK) construct that selectively activates RANK signaling in myeloid cells in vivo. The cytoplasmic portion of RANK was fused to a mutant FK506 binding domain, which selectively binds the chemical inducer of dimerization AP20187 and initiates signaling. iRANK mRNA was expressed in macrophages isolated from peritoneal cavity, spleen-, and bone marrow-derived myeloid cells. Unexpectedly, AP20187 did not induce osteoclast formation in spleen- and bone marrow-derived myeloid cells. However, AP20187-dependent RANK signaling induced ERK1/2 phosphorylation and mRNA expression of MMP9 and CathepsinK in peritoneal macrophages. Importantly, CD68 was not expressed until day 3 and day 5 in bone marrow and spleen myeloid cells, respectively. Contrary to dogma, osteoclast precursors do not express the lineage marker CD68.
Collapse
Affiliation(s)
- Melissa F Jackson
- Department of Bioengineering, University of Washington, Seattle, WA, USA
| | - Marta Scatena
- Department of Bioengineering, University of Washington, Seattle, WA, USA
| | | |
Collapse
|
10
|
Dahl M, Doyle A, Olsson K, Månsson JE, Marques ARA, Mirzaian M, Aerts JM, Ehinger M, Rothe M, Modlich U, Schambach A, Karlsson S. Lentiviral gene therapy using cellular promoters cures type 1 Gaucher disease in mice. Mol Ther 2015; 23:835-844. [PMID: 25655314 DOI: 10.1038/mt.2015.16] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2014] [Accepted: 01/22/2015] [Indexed: 12/17/2022] Open
Abstract
Gaucher disease is caused by an inherited deficiency of the enzyme glucosylceramidase. Due to the lack of a fully functional enzyme, there is progressive build-up of the lipid component glucosylceramide. Insufficient glucosylceramidase activity results in hepatosplenomegaly, cytopenias, and bone disease in patients. Gene therapy represents a future therapeutic option for patients unresponsive to enzyme replacement therapy and lacking a suitable bone marrow donor. By proof-of-principle experiments, we have previously demonstrated a reversal of symptoms in a murine disease model of type 1 Gaucher disease, using gammaretroviral vectors harboring strong viral promoters to drive glucosidase β-acid (GBA) gene expression. To investigate whether safer vectors can correct the enzyme deficiency, we utilized self-inactivating lentiviral vectors (SIN LVs) with the GBA gene under the control of human phosphoglycerate kinase (PGK) and CD68 promoter, respectively. Here, we report prevention of, as well as reversal of, manifest disease symptoms after lentiviral gene transfer. Glucosylceramidase activity above levels required for clearance of glucosylceramide from tissues resulted in reversal of splenomegaly, reduced Gaucher cell infiltration and a restoration of hematological parameters. These findings support the use of SIN-LVs with cellular promoters in future clinical gene therapy protocols for type 1 Gaucher disease.
Collapse
Affiliation(s)
- Maria Dahl
- Department of Molecular Medicine and Gene Therapy, Lund University, Lund, Sweden; Lund Strategic Center for Stem Cell Biology and Cell Therapy, Lund University Hospital, Lund, Sweden
| | - Alexander Doyle
- Department of Molecular Medicine and Gene Therapy, Lund University, Lund, Sweden; Lund Strategic Center for Stem Cell Biology and Cell Therapy, Lund University Hospital, Lund, Sweden
| | - Karin Olsson
- Department of Molecular Medicine and Gene Therapy, Lund University, Lund, Sweden; Lund Strategic Center for Stem Cell Biology and Cell Therapy, Lund University Hospital, Lund, Sweden
| | - Jan-Eric Månsson
- Department of Clinical Chemistry, Institute of Biomedicine, the Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - André R A Marques
- Department of Medical Biochemistry, University of Amsterdam, Amsterdam, The Netherlands
| | - Mina Mirzaian
- Department of Medical Biochemistry, University of Amsterdam, Amsterdam, The Netherlands
| | - Johannes M Aerts
- Department of Medical Biochemistry, University of Amsterdam, Amsterdam, The Netherlands
| | - Mats Ehinger
- Department of Pathology, Lund University Hospital, Lund, Sweden
| | - Michael Rothe
- Institute of Experimental Hematology, Hannover Medical School, Hannover, Germany
| | - Ute Modlich
- Institute of Experimental Hematology, Hannover Medical School, Hannover, Germany
| | - Axel Schambach
- Institute of Experimental Hematology, Hannover Medical School, Hannover, Germany
| | - Stefan Karlsson
- Department of Molecular Medicine and Gene Therapy, Lund University, Lund, Sweden; Lund Strategic Center for Stem Cell Biology and Cell Therapy, Lund University Hospital, Lund, Sweden.
| |
Collapse
|
11
|
Human CD68 promoter GFP transgenic mice allow analysis of monocyte to macrophage differentiation in vivo. Blood 2014; 124:e33-44. [PMID: 25030063 DOI: 10.1182/blood-2014-04-568691] [Citation(s) in RCA: 71] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
The recruitment of monocytes and their differentiation into macrophages at sites of inflammation are key events in determining the outcome of the inflammatory response and initiating the return to tissue homeostasis. To study monocyte trafficking and macrophage differentiation in vivo, we have generated a novel transgenic reporter mouse expressing a green fluorescent protein (GFP) under the control of the human CD68 promoter. CD68-GFP mice express high levels of GFP in both monocyte and embryo-derived tissue resident macrophages in adult animals. The human CD68 promoter drives GFP expression in all CD115(+) monocytes of adult blood, spleen, and bone marrow; we took advantage of this to directly compare the trafficking of bone marrow-derived CD68-GFP monocytes to that of CX3CR1(GFP) monocytes in vivo using a sterile zymosan peritonitis model. Unlike CX3CR1(GFP) monocytes, which downregulate GFP expression on differentiation into macrophages in this model, CD68-GFP monocytes retain high-level GFP expression for 72 hours after differentiation into macrophages, allowing continued cell tracking during resolution of inflammation. In summary, this novel CD68-GFP transgenic reporter mouse line represents a powerful resource for analyzing monocyte mobilization and monocyte trafficking as well as studying the fate of recruited monocytes in models of acute and chronic inflammation.
Collapse
|
12
|
Nishizawa T, Kanter JE, Kramer F, Barnhart S, Shen X, Vivekanandan-Giri A, Wall VZ, Kowitz J, Devaraj S, O'Brien KD, Pennathur S, Tang J, Miyaoka RS, Raines EW, Bornfeldt KE. Testing the role of myeloid cell glucose flux in inflammation and atherosclerosis. Cell Rep 2014; 7:356-365. [PMID: 24726364 DOI: 10.1016/j.celrep.2014.03.028] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2013] [Revised: 02/26/2014] [Accepted: 03/11/2014] [Indexed: 10/25/2022] Open
Abstract
Inflammatory activation of myeloid cells is accompanied by increased glycolysis, which is required for the surge in cytokine production. Although in vitro studies suggest that increased macrophage glucose metabolism is sufficient for cytokine induction, the proinflammatory effects of increased myeloid cell glucose flux in vivo and the impact on atherosclerosis, a major complication of diabetes, are unknown. We therefore tested the hypothesis that increased glucose uptake in myeloid cells stimulates cytokine production and atherosclerosis. Overexpression of the glucose transporter GLUT1 in myeloid cells caused increased glycolysis and flux through the pentose phosphate pathway but did not induce cytokines. Moreover, myeloid-cell-specific overexpression of GLUT1 in LDL receptor-deficient mice was ineffective in promoting atherosclerosis. Thus, increased glucose flux is insufficient for inflammatory myeloid cell activation and atherogenesis. If glucose promotes atherosclerosis by increasing cellular glucose flux, myeloid cells do not appear to be the key targets.
Collapse
Affiliation(s)
- Tomohiro Nishizawa
- Diabetes and Obesity Center of Excellence, University of Washington, Seattle, WA 98109, USA; Department of Medicine, University of Washington, Seattle, WA 98109, USA
| | - Jenny E Kanter
- Diabetes and Obesity Center of Excellence, University of Washington, Seattle, WA 98109, USA; Department of Medicine, University of Washington, Seattle, WA 98109, USA
| | - Farah Kramer
- Diabetes and Obesity Center of Excellence, University of Washington, Seattle, WA 98109, USA; Department of Medicine, University of Washington, Seattle, WA 98109, USA
| | - Shelley Barnhart
- Diabetes and Obesity Center of Excellence, University of Washington, Seattle, WA 98109, USA; Department of Medicine, University of Washington, Seattle, WA 98109, USA
| | - Xia Shen
- Diabetes and Obesity Center of Excellence, University of Washington, Seattle, WA 98109, USA; Department of Medicine, University of Washington, Seattle, WA 98109, USA
| | | | - Valerie Z Wall
- Diabetes and Obesity Center of Excellence, University of Washington, Seattle, WA 98109, USA; Department of Medicine, University of Washington, Seattle, WA 98109, USA
| | - Jason Kowitz
- Diabetes and Obesity Center of Excellence, University of Washington, Seattle, WA 98109, USA; Department of Medicine, University of Washington, Seattle, WA 98109, USA
| | - Sridevi Devaraj
- Baylor College of Medicine, Houston, TX 77030, USA; Texas Children's Hospital, Houston, TX 77030, USA
| | - Kevin D O'Brien
- Department of Medicine, University of Washington, Seattle, WA 98109, USA
| | | | - Jingjing Tang
- Department of Pathology, University of Washington, Seattle, WA 98109, USA
| | - Robert S Miyaoka
- Department of Radiology, University of Washington, Seattle, WA 98195, USA
| | - Elaine W Raines
- Department of Pathology, University of Washington, Seattle, WA 98109, USA
| | - Karin E Bornfeldt
- Diabetes and Obesity Center of Excellence, University of Washington, Seattle, WA 98109, USA; Department of Medicine, University of Washington, Seattle, WA 98109, USA; Department of Pathology, University of Washington, Seattle, WA 98109, USA.
| |
Collapse
|
13
|
Affiliation(s)
- Andrew J Murphy
- From the Haematopoiesis and Leukocyte Biology, Baker IDI Heart and Diabetes Institute, Melbourne, Australia (A.J.M.); Department of Immunology, Monash University, Melbourne, Australia (A.J.M.); and Department of Medicine, Division of Molecular Medicine, Columbia University, New York, NY (A.R.T.)
| | | |
Collapse
|
14
|
Lund SA, Wilson CL, Raines EW, Tang J, Giachelli CM, Scatena M. Osteopontin mediates macrophage chemotaxis via α4 and α9 integrins and survival via the α4 integrin. J Cell Biochem 2013. [PMID: 23192608 DOI: 10.1002/jcb.24462] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Osteopontin (OPN) is highly expressed by macrophages and plays a key role in the pathology of several chronic inflammatory diseases including atherosclerosis and the foreign body reaction. However, the molecular mechanism behind OPN regulation of macrophage functions is not well understood. OPN is a secreted molecule and interacts with several integrins via two domains: the RGD sequence binding to α(v) -containing integrins, and the SLAYGLR sequence binding to α(4) β(1), α(4) β(7), and α(9) β(1) integrins. Here we determined the role of OPN in macrophage survival, chemotaxis, and activation state. For survival studies, OPN treated-bone marrow derived macrophages (BMDMs) were challenged with growth factor withdrawal and neutralizing integrin antibodies. We found that survival in BMDMs is mediated primarily through the α(4) integrin. In chemotaxis studies, we observed that migration to OPN was blocked by neutralizing α(4) and α(9) integrin antibodies. Further, OPN did not affect macrophage activation as measured by IL-12 production. Finally, the relative contributions of the RGD and the SLAYGLR functional domains of OPN to leukocyte recruitment were evaluated in an in vivo model. We generated chimeric mice expressing mutated forms of OPN in myeloid-derived leukocytes, and found that the SLAYGLR functional domain of OPN, but not the RGD, mediates macrophage accumulation in response to thioglycollate-elicited peritonitis. Collectively, these data indicate that α(4) and α(9) integrins interacting with OPN via the SLAYGLR domain play a key role in macrophage biology by regulating migration, survival, and accumulation.
Collapse
Affiliation(s)
- Susan Amanda Lund
- Department of Bioengineering, University of Washington, Seattle, WA 98109, USA
| | | | | | | | | | | |
Collapse
|
15
|
Wood TR, Chow RY, Hanes CM, Zhang X, Kashiwagi K, Shirai Y, Trebak M, Loegering DJ, Saito N, Lennartz MR. PKC-ε pseudosubstrate and catalytic activity are necessary for membrane delivery during IgG-mediated phagocytosis. J Leukoc Biol 2013; 94:109-22. [PMID: 23670290 DOI: 10.1189/jlb.1212634] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
In RAW 264.7 cells, PKC-ε regulates FcγR-mediated phagocytosis. BMDM behave similarly; PKC-ε concentrates at phagosomes and internalization are reduced in PKC-ε⁻/⁻ cells. Two questions were asked: what is the role of PKC-ε? and what domains are necessary for PKC-ε concentration? Function was studied using BMDM and frustrated phagocytosis. On IgG surfaces, PKC-ε⁻/⁻ macrophages spread less than WT. Patch-clamping revealed that the spreading defect is a result of the failure of PKC-ε⁻/⁻ macrophages to add membrane. The defect is specific for FcγR ligation and can be reversed by expression of full-length (but not the isolated RD) PKC-ε in PKC-ε⁻/⁻ BMDM. Thus, PKC-ε function in phagocytosis requires translocation to phagosomes and the catalytic domain. The expression of chimeric PKC molecules in RAW cells identified the εPS as necessary for PKC-ε targeting. When placed into (nonlocalizing) PKC-δ, εPS was sufficient for concentration, albeit to a lesser degree than intact PKC-ε. In contrast, translocation of δ(εPSC1B) resembled that of WT PKC-ε. Thus, εPS and εC1B cooperate for optimal phagosome targeting. Finally, cells expressing εK437W were significantly less phagocytic than their PKC-ε-expressing counterparts, blocked at the pseudopod-extension phase. In summary, we have shown that εPS and εC1B are necessary and sufficient for targeting PKC-ε to phagosomes, where its catalytic activity is required for membrane delivery and pseudopod extension.
Collapse
Affiliation(s)
- Tiffany R Wood
- Centers for Cell Biology and Cancer Researchnces, Albany Medical College, Albany, New York, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Levin MC, Lidberg U, Jirholt P, Adiels M, Wramstedt A, Gustafsson K, Greaves DR, Li S, Fazio S, Linton MF, Olofsson SO, Borén J, Gjertsson I. Evaluation of macrophage-specific promoters using lentiviral delivery in mice. Gene Ther 2012; 19:1041-7. [PMID: 22130447 PMCID: PMC3697098 DOI: 10.1038/gt.2011.195] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2011] [Revised: 10/07/2011] [Accepted: 10/19/2011] [Indexed: 12/22/2022]
Abstract
In gene therapy, tissue-specific promoters are useful tools to direct transgene expression and improve efficiency and safety. Macrophage-specific promoters (MSPs) have previously been published using different delivery systems. In this study, we evaluated five different MSPs fused with green fluorescent protein (GFP) to delineate the one with highest specificity using lentiviral delivery. We compared three variants of the CD68 promoter (full length, the 343-bp proximal part and the 150-bp proximal part) and two variants (in forward and reverse orientation) of a previously characterized synthetic promoter derived from elements of transcription factor genes. We transduced a number of cell lines and primary cells in vitro. In addition, hematopoietic stem cells were transduced with MSPs and transferred into lethally irradiated recipient mice. Fluorescence activated cell sorting analysis was performed to determine the GFP expression in different cell populations both in vitro and in vivo. We showed that MSPs can efficiently be used for lentiviral gene delivery and that the 150-bp proximal part of the CD68 promoter provides primarily macrophage-specific expression of GFP. We propose that this is the best currently available MSP to use for directing transgene expression to macrophage populations in vivo using lentiviral vectors.
Collapse
Affiliation(s)
- M C Levin
- Department of Molecular and Clinical Medicine/Wallenberg Laboratory, University of Gothenburg, Göteborg, Sweden.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Genetic ablation of CD68 results in mice with increased bone and dysfunctional osteoclasts. PLoS One 2011; 6:e25838. [PMID: 21991369 PMCID: PMC3185056 DOI: 10.1371/journal.pone.0025838] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2011] [Accepted: 09/12/2011] [Indexed: 12/25/2022] Open
Abstract
CD68 is a member of the lysosome associated membrane protein (LAMP) family that is restricted in its expression to cells of the monocyte/macrophage lineage. This lineage restriction includes osteoclasts, and, while previous studies of CD68 in macrophages and dendritic cells have proposed roles in lipid metabolism, phagocytosis, and antigen presentation, the expression and function of CD68 in osteoclasts have not been explored. In this study, we investigated the expression and localization of CD68 in macrophages and osteoclasts in response to the monocyte/macrophage-colony stimulating factor (M-CSF) and the receptor activator of NF-κB ligand (RANKL). We found that M-CSF stimulates CD68 expression and RANKL alters the apparent molecular weight of CD68 as measured by Western immunoblotting. In addition, we explored the significance of CD68 expression in osteoclasts by generating mice that lack expression of CD68. These mice have increased trabecular bone, and in vitro assessment of CD68−/− osteoclasts revealed that, in the absence of CD68, osteoclasts demonstrate an accumulation of intracellular vesicle-like structures, and do not efficiently resorb bone. These findings demonstrate a role for CD68 in the function of osteoclasts, and future studies will determine the mechanistic nature of the defects seen in CD68−/− osteoclasts.
Collapse
|
18
|
Abstract
Scavenger receptors (ScRs) are a structurally unrelated family of receptors with the ability to bind modified low density lipoprotein (LDL) as well as a broad range of polyanionic ligands. CD68, whose expression is restricted to mononuclear phagocytes, is a unique ScR family member, owing to its lysosome associated membrane protein (LAMP)-like domain and predominant endosomal distribution. Knockout (ko) mice were generated to directly evaluate the role murine CD68 may play in oxidized LDL (Ox-LDL) uptake. However, CD68⁻/⁻ macrophages took up Ox-LDL robustly. Likewise, no defects were observed in the ability of CD68⁻/⁻ mononuclear phagocytes to take up or mount an effective innate response against a number of microbes. Curiously, CD68⁻/⁻ mononuclear phagocytes exhibited a trend toward enhanced antigen presentation to CD4⁺ T-cells, raising the possibility that CD68 may function either to negatively regulate antigen uptake, loading, or major histocompatibility complex class II (MHC-II) trafficking.
Collapse
Affiliation(s)
- Li Song
- Department of Microbiology, Columbia University, New York, NY, USA
| | | | | |
Collapse
|
19
|
Sather BD, Ryu BY, Stirling BV, Garibov M, Kerns HM, Humblet-Baron S, Astrakhan A, Rawlings DJ. Development of B-lineage predominant lentiviral vectors for use in genetic therapies for B cell disorders. Mol Ther 2010; 19:515-25. [PMID: 21139568 DOI: 10.1038/mt.2010.259] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
Sustained, targeted, high-level transgene expression in primary B lymphocytes may be useful for gene therapy in B cell disorders. We developed several candidate B-lineage predominant self-inactivating lentiviral vectors (LV) containing alternative enhancer/promoter elements including: the immunoglobulin β (Igβ) (B29) promoter combined with the immunoglobulin µ enhancer (EµB29); and the endogenous BTK promoter with or without Eµ (EµBtkp or Btkp). LV-driven enhanced green fluorescent protein (eGFP) reporter expression was evaluated in cell lines and primary cells derived from human or murine hematopoietic stem cells (HSC). In murine primary cells, EµB29 and EµBtkp LV-mediated high-level expression in immature and mature B cells compared with all other lineages. Expression increased with B cell maturation and was maintained in peripheral subsets. Expression in T and myeloid cells was much lower in percentage and intensity. Similarly, both EµB29 and EµBtkp LV exhibited high-level activity in human primary B cells. In contrast to EµB29, Btkp and EµBtkp LV also exhibited modest activity in myeloid cells, consistent with the expression profile of endogenous Bruton's tyrosine kinase (Btk). Notably, EµB29 and EµBtkp activity was superior in all expression models to an alternative, B-lineage targeted vector containing the EµS.CD19 enhancer/promoter. In summary, EµB29 and EµBtkp LV comprise efficient delivery platforms for gene expression in B-lineage cells.
Collapse
Affiliation(s)
- Blythe D Sather
- Center for Immunity and Immunotherapies, Seattle Children's Research Institute, Seattle, Washington 98101, USA
| | | | | | | | | | | | | | | |
Collapse
|
20
|
Abstract
After more than 1500 gene therapy clinical trials in the past two decades, the overall conclusion is that for gene therapy (GT) to be successful, the vector systems must still be improved in terms of delivery, expression and safety. The recent development of more efficient and stable vector systems has created great expectations for the future of GT. Impressive results were obtained in three primary immunodeficiencies and other inherited diseases such as congenital blindness, adrenoleukodystrophy or junctional epidermolysis bullosa. However, the development of leukemia in five children included in the GT clinical trials for X-linked severe combined immunodeficiency and the silencing of the therapeutic gene in the chronic granulomatous disease clearly showed the importance of improving safety and efficiency. In this review, we focus on the main strategies available to achieve physiological or tissue-specific expression of therapeutic transgenes and discuss the importance of controlling transgene expression to improve safety. We propose that tissue-specific and/or physiological viral vectors offer the best balance between efficiency and safety and will be the tools of choice for future clinical trials in GT of inherited diseases.
Collapse
|
21
|
Aorta transplantation in young apolipoprotein E-deficient mice: Possible model for studies on regression of atherosclerotic lesions? Open Med (Wars) 2010. [DOI: 10.2478/s11536-010-0006-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
AbstractSyngeneic transplantation of murine aorta segments with advanced atherosclerotic lesions in defined recipients is a valuable model for regression studies. To date, this model has not been used to study the regression of initial atherosclerotic lesions. The aim of this study was to evaluate a microsurgical technique of syngeneic heterotopic transplantation of the thoracic aorta of young apolipoprotein E-deficient (ApoE-/-) mice to the abdominal aorta of wild-type recipients. Stereological quantification methods were tested in order to assess changes in structure and volume of the aortic wall including the involvement of immune cells in changes of the atherosclerotic lesions. The animals were euthanised one month after surgery and histological analysis including stereological quantification of changes in both the grafts and adjacent aorta segments was performed. The overall survival rate of the recipients was 62.5%. No regression of initial atherosclerotic lesion was achieved and neointima formation and elastin degradation prevailed in all transplanted specimens. The volume of the arteriosclerotic lesions was higher (p<0.001) and elastin length density was lower (p<0.001) in transplanted ApoE-/- samples as compared to adjacent segments. In transplanted grafts, T- and B-lymphocytes, macrophages and neutrophilic granulocytes formed non-random clusters within the vessel wall and they were colocalised with the sutures. The reproducibility of the promising regression model was derogated in young mice by the striking dependence of the results upon the operation technique. Stereological assessment has proven to be accurate, correct and reproducible; it has provided us with robust quantitative estimates, which can be achieved with a reasonable effort.
Collapse
|
22
|
Wu C, Lu Y. High-titre retroviral vector system for efficient gene delivery into human and mouse cells of haematopoietic and lymphocytic lineages. J Gen Virol 2010; 91:1909-1918. [PMID: 20410313 PMCID: PMC3052536 DOI: 10.1099/vir.0.020255-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Genetically modified cells of haematopoietic and lymphocytic lineages could provide potentially curative treatments for a wide range of inherited and acquired diseases. However, this application is limited in mouse models by the low efficiency of lentiviral vectors. To facilitate the rapid production of high-titre helper-free retroviral vectors for enhanced gene delivery, multiple modifications to a prototype moloney murine leukemia virus (MoMLV)-derived vector system were made including adaptation of the vector system to simian virus 40 ori/T antigen-mediated episomal replication in packaging cells, replacement of the MoMLV 5' U3 promoter with a series of stronger composite promoters and addition of an extra polyadenylation signal downstream of the 3' long terminal repeat. These modifications enhanced vector production by 2-3 logs. High-titre vector stocks were tested for their ability to infect a variety of cells derived from humans and mice, including primary monocyte-derived macrophage cultures. Whilst the lentiviral vector was significantly restricted at the integration level, the MoMLV-based vector showed effective gene transduction of mouse cells. This high-titre retroviral vector system represents a useful tool for efficient gene delivery into human and mouse haematopoietic and lymphocytic cells, with particular application in mice as a small animal model for novel gene therapy tests.
Collapse
Affiliation(s)
- Chengxiang Wu
- Departments of Public Health Sciences and Microbiology, University of Hawaii at Manoa, Honolulu, HI 96822, USA
| | - Yuanan Lu
- Departments of Public Health Sciences and Microbiology, University of Hawaii at Manoa, Honolulu, HI 96822, USA
| |
Collapse
|
23
|
Han X, Kitamoto S, Wang H, Boisvert WA. Interleukin-10 overexpression in macrophages suppresses atherosclerosis in hyperlipidemic mice. FASEB J 2010; 24:2869-80. [PMID: 20354139 DOI: 10.1096/fj.09-148155] [Citation(s) in RCA: 97] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
In atherogenesis, macrophage foam cell formation is modulated by pathways involving both the uptake and efflux of cholesterol. We recently showed that interleukin-10 (IL-10) modulates lipid metabolism by enhancing both uptake and efflux of cholesterol in macrophages. However, the mechanistic details of these properties in vivo have been unclear. Thus, the purpose of this study was to determine whether expression of IL-10 in macrophages would alter susceptibility to atherosclerosis and whether IL-10 exerts its antiatherosclerotic properties by modulating lipid metabolism in macrophages. We utilized a macrophage-specific retroviral vector that allows long-term in vivo expression of IL-10 in macrophages through transplantation of retrovirally transduced bone marrow cells (BMCs). IL-10 expressed by macrophages derived from transduced BMCs inhibited atherosclerosis in LDLR(-/-) mice by reducing cholesteryl ester accumulation in atherosclerotic sites. Experiments with primary macrophages indicated that macrophage source of IL-10 stimulated both the uptake (by up-regulating scavenger receptors) and efflux of cholesterol (by activating the PPARgamma-LXR-ABCA1/ABCG1 pathway), thereby reducing inflammation and apoptosis in atherosclerosis. These findings indicate that BMC-transduced macrophage IL-10 production can act as a strong antiatherogenic agent, and they highlight a novel antiatherosclerotic therapy using a simple, yet effective, stem cell transduction system that facilitates long-term expression of IL-10 in macrophages.
Collapse
Affiliation(s)
- Xinbing Han
- Vascular Medicine Research Unit, Brigham Women's Hospital, Harvard Medical School, Cambridge, Massachusetts, USA
| | | | | | | |
Collapse
|
24
|
Inducible transgenes under the control of the hCD68 promoter identifies mouse macrophages with a distribution that differs from the F4/80 - and CSF-1R-expressing populations. Exp Hematol 2009; 37:1387-92. [PMID: 19772887 DOI: 10.1016/j.exphem.2009.09.003] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2009] [Revised: 08/26/2009] [Accepted: 09/16/2009] [Indexed: 02/02/2023]
Abstract
OBJECTIVE Macrophages are critical components of diverse microenvironments (ME) in adulthood, as well as during embryogenesis. Their role in development precludes the use of gene-targeting and knockout approaches for studying their function. Hence, we proposed to create a macrophage-specific inducible transgenic mouse where genes can be turned on or off at will. MATERIALS AND METHODS A transgenic mouse in which the reverse tetracycline activator (rtTA-M2) is expressed under the hCD68 promoter for macrophage-specific gene induction was developed and crossed with a second transgenic reporter mouse strain in which the gene for green fluorescent protein (GFP) is under the control of tetracycline responsive element promoter. After doxycycline induction of the double transgenic animals (designated CD68-rtTA-tet-GFP), inducible expression of GFP was characterized by multicolor flow cytometric analysis of blood, marrow, and spleen cells and by demonstration of GFP expression in fresh-frozen sections in diverse tissues. RESULTS In bone marrow, inducible GFP expression was not confined to, or inclusive of, all cells expressing the classical macrophage markers, such as F4/80. However, GFP-expressing cells in thioglycollate-elicited peritoneal macrophages were also positive for F4/80 and monocyte-macrophage-specific 2 antigen. Interestingly, flow analysis also indicated little overlap between the F4/80 and CSF-1R-positive populations. Fresh-frozen samples of tissues known to contain macrophages revealed GFP-expressing cells with variable morphologies. CONCLUSION Our results show that the hCD68 promoter directs gene expression in a macrophage population distinct from that defined by classical monocyte-macrophage markers or promoters. Whether this population is functionally distinct remains to be established.
Collapse
|
25
|
Intraclonal competition limits the fate determination of regulatory T cells in the thymus. Nat Immunol 2009; 10:610-7. [PMID: 19430476 PMCID: PMC2756247 DOI: 10.1038/ni.1739] [Citation(s) in RCA: 203] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2009] [Accepted: 04/15/2009] [Indexed: 12/11/2022]
Abstract
Because the deletion of self-reactive T cells is incomplete, thymic development of natural Foxp3+CD4+ regulatory T (Treg) cells is required for preventing autoimmunity. However, the role of T cell receptor (TCR) specificity in thymic Treg cell development remains controversial. To address this issue, we generated a transgenic line expressing a naturally occurring Treg cell-derived TCR. Surprisingly, efficient thymic Treg cell development occurred only when the antigen-specific Treg cell precursors were present at low clonal frequency (<1%) within a normal thymus. Using retroviral vectors and bone marrow chimeras, we observed similar behavior with two other Treg cell-derived TCRs. These data demonstrate that thymic Treg cell development is a TCR-instructive process involving a niche which can be saturable at much lower clonal frequencies than the niche for positive selection.
Collapse
|
26
|
Zeitouni S, Ford BS, Harris SM, Whitney MJ, Gregory CA, Prockop DJ. Pharmaceutical induction of ApoE secretion by multipotent mesenchymal stromal cells (MSCs). BMC Biotechnol 2008; 8:75. [PMID: 18823563 PMCID: PMC2596794 DOI: 10.1186/1472-6750-8-75] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2008] [Accepted: 09/29/2008] [Indexed: 12/23/2022] Open
Abstract
Background Apolipoprotein E (ApoE) is a molecular scavenger in the blood and brain. Aberrant function of the molecule causes formation of protein and lipid deposits or "plaques" that characterize Alzheimer's disease (AD) and atherosclerosis. There are three human isoforms of ApoE designated ε2, ε3, and ε4. Each isoform differentially affects the structure and function of the protein and thus the development of disease. Homozygosity for ApoE ε4 is associated with atherosclerosis and Alzheimer's disease whereas ApoE ε2 and ε3 tend to be protective. Furthermore, the ε2 form may cause forms of hyperlipoproteinemia. Therefore, introduction of ApoE ε3 may be beneficial to patients that are susceptible to or suffering from these diseases. Mesenchymal stem cells or multipotent mesenchymal stromal cells (MSCs) are adult progenitor cells found in numerous tissues. They are easily expanded in culture and engraft into host tissues when administered appropriately. Furthermore, MSCs are immunosuppressive and have been reported to engraft as allogeneic transplants. In our previous study, mouse MSCs (mMSCs) were implanted into the brains of ApoE null mice, resulting in production of small amounts of ApoE in the brain and attenuation of cognitive deficits. Therefore human MSCs (hMSCs) are a promising vector for the administration of ApoE ε3 in humans. Results Unlike mMSCs, hMSCs were found not to express ApoE in culture; therefore a molecular screen was performed for compounds that induce expression. PPARγ agonists, neural stem cell conditioned medium, osteo-inductive media, dexamethasone, and adipo-inductive media (AIM) were tested. Of the conditions tested, only AIM or dexamethasone induced sustained secretion of ApoE in MSCs and the duration of secretion was only limited by the length of time MSCs could be sustained in culture. Upon withdrawal of the inductive stimuli, the ApoE secretion persisted for a further 14 days. Conclusion The data demonstrated that pre-treatment and perhaps co-administration of MSCs homozygous for ApoE ε3 and dexamethasone may represent a novel therapy for severe instances of AD, atherosclerosis and other ApoE-related diseases.
Collapse
Affiliation(s)
- Suzanne Zeitouni
- Center for Gene Therapy, Tulane University Medical School, New Orleans, LA, 70115, USA.
| | | | | | | | | | | |
Collapse
|
27
|
Su YR, Blakemore JL, Zhang Y, Linton MF, Fazio S. Lentiviral transduction of apoAI into hematopoietic progenitor cells and macrophages: applications to cell therapy of atherosclerosis. Arterioscler Thromb Vasc Biol 2008; 28:1439-46. [PMID: 18497309 DOI: 10.1161/atvbaha.107.160093] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE We used genetically engineered mouse hematopoietic progenitor cells (HPCs) to investigate the therapeutic effects of human apoAI on atherosclerosis in apoE(-/-) mice. METHODS AND RESULTS Lentiviral constructs expressing either human apoAI (LV-apoAI) or green fluorescent protein (LV-GFP) cDNA under a macrophage specific promoter (CD68) were generated and used for ex vivo transduction of mouse HPCs and macrophages. The transduction efficiency was >25% for HPCs and >70% for macrophages. ApoAI was found in the macrophage culture media, mostly associated with the HDL fraction. Interestingly, a significant increase in mRNA and protein levels for ATP binding cassette A1 (ABCA1) and ABCG1 were found in apoAI-expressing macrophages after acLDL loading. Expression of apoAI significantly increased cholesterol efflux in wild-type and apoE(-/-) macrophages. HPCs transduced with LV-apoAI ex vivo and then transplanted into apoE(-/-) mice caused a 50% reduction in atherosclerotic lesion area compared to GFP controls, without influencing plasma HDL-C levels. CONCLUSIONS Lentiviral transduction of apoAI into HPCs reduces atherosclerosis in apoE(-/-) mice. Expression of apoAI in macrophages improves cholesterol trafficking in wild-type apoE-producing macrophages and causes upregulation of ABCA1 and ABCG1. These novel observations set the stage for a cell therapy approach to atherosclerosis regression, exploiting the cooperation between apoE and apoAI to maximize cholesterol exit from the plaque.
Collapse
Affiliation(s)
- Yan Ru Su
- Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville TN 37232-6300, USA.
| | | | | | | | | |
Collapse
|
28
|
Abstract
Osteopontin (OPN) is a multifunctional molecule highly expressed in chronic inflammatory and autoimmune diseases, and it is specifically localized in and around inflammatory cells. OPN is a secreted adhesive molecule, and it is thought to aid in the recruitment of monocytes-macrophages and to regulate cytokine production in macrophages, dendritic cells, and T-cells. OPN has been classified as T-helper 1 cytokine and thus believed to exacerbate inflammation in several chronic inflammatory diseases, including atherosclerosis. Besides proinflammatory functions, physiologically OPN is a potent inhibitor of mineralization, it prevents ectopic calcium deposits and is a potent inducible inhibitor of vascular calcification. Clinically, OPN plasma levels have been found associated with various inflammatory diseases, including cardiovascular burden. It is thus imperative to dissect the OPN proinflammatory and anticalcific functions. OPN recruitment functions of inflammatory cells are thought to be mediated through its adhesive domains, especially the arginine-glycine-aspartate (RGD) sequence that interacts with several integrin heterodimers. However, the integrin receptors and intracellular pathways mediating OPN effects on immune cells are not well established. Furthermore, several studies show that OPN is cleaved by at least 2 classes of proteases: thrombin and matrix-metalloproteases (MMPs). Most importantly, at least
in vitro
, fragments generated by cleavage not only maintain OPN adhesive functions but also expose new active domains that may impart new activities. The role for OPN proteolytic fragments
in vivo
is almost completely unexplored. We believe that further knowledge of the effects of OPN fragments on cell responses might help in designing therapeutics targeting inflammatory and cardiovascular diseases.
Collapse
Affiliation(s)
- Marta Scatena
- Department of Bioengineering, University of Washington, Box 355061, Seattle, WA 98195, USA.
| | | | | |
Collapse
|
29
|
Wisse BE, Ogimoto K, Tang J, Harris MK, Raines EW, Schwartz MW. Evidence that lipopolysaccharide-induced anorexia depends upon central, rather than peripheral, inflammatory signals. Endocrinology 2007; 148:5230-7. [PMID: 17673516 DOI: 10.1210/en.2007-0394] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Systemic inflammatory stimuli cause anorexia and weight loss by disrupting the physiological regulation of energy balance. Mice lacking MyD88, an intracellular mediator of signal transduction activated by Toll-like receptor 4 or IL-1beta receptors, are resistant to anorexia induced by the bacterial endotoxin lipopolysaccharide (LPS), despite a significant circulating cytokine response. Thus, we hypothesized that induction of a peripheral inflammatory response is insufficient to cause LPS-induced anorexia when MyD88 signaling in the central nervous system and other tissues is absent. To test this hypothesis, we used bone marrow transplantation (BMT) to determine if LPS-induced anorexia can be restored to MyD88-deficient mice by reconstituting their bone marrow with wild-type (WT) immune cells. We found that restoring WT circulating immune cells to mice lacking MyD88 conferred only a mild, short-lived anorexia in response to LPS, such that food intake was fully normalized by 20 h post injection (LPS 4.1 +/- 0.5 g vs. vehicle 4.3 +/- 0.3 g), whereas LPS-induced anorexia was profound and sustained in WT controls after either autologous BMT or sham BMT. Similarly, LPS-mediated induction of hypothalamic mRNA encoding IL-1beta and TNFalpha was robust in both WT control groups but was absent in chimeric MyD88 mice, despite comparable peripheral inflammatory responses across the three groups. We conclude that LPS reduces food intake via a mechanism dependent on MyD88 signaling within brain and/or other tissues and that in the absence of this effect, robust stimulation of circulating immune cells cannot induce sustained anorexia.
Collapse
Affiliation(s)
- Brent E Wisse
- Division of Metabolism, Harborview Medical Center, Seattle, WA 98104-2499, USA.
| | | | | | | | | | | |
Collapse
|
30
|
O'Reilly D, Greaves DR. Cell-type-specific expression of the human CD68 gene is associated with changes in Pol II phosphorylation and short-range intrachromosomal gene looping. Genomics 2007; 90:407-15. [PMID: 17583472 DOI: 10.1016/j.ygeno.2007.04.010] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2006] [Revised: 04/26/2007] [Accepted: 04/28/2007] [Indexed: 01/05/2023]
Abstract
Transcriptional regulation in higher eukaryotes frequently involves long-range interactions, up to tens of hundreds of kilobases away, of a number of cis-acting regulatory DNA elements. Using the chromosome conformation capture technique we have analyzed the expression of a small 2.5-kb gene, CD68, in different human cell types and show for the first time that short-range interactions may also be critical. In human monocytes, which produce high levels of CD68 mRNA, the gene is characterized by intramolecular ligations between the promoter and the 3' intervening region. In cells that poorly express the gene a change in architecture is apparent whereby the promoter preferentially associates with the terminator region only. Furthermore, alterations in CD68 gene structure are associated with failings in mRNA splicing and changes with the phosphorylation status of RNA Pol II across the gene. We propose that short-range intrachromosomal interactions may form the basis of coordinated control of monocyte-specific gene regulation.
Collapse
MESH Headings
- Antigens, CD/biosynthesis
- Antigens, CD/physiology
- Antigens, Differentiation, Myelomonocytic/biosynthesis
- Antigens, Differentiation, Myelomonocytic/physiology
- Cell Line
- Cell Line, Tumor
- Chromatin/metabolism
- Chromatin Immunoprecipitation
- DNA Polymerase II/metabolism
- Gene Expression Regulation
- HL-60 Cells
- Humans
- Jurkat Cells
- Models, Genetic
- Myeloid Cells/metabolism
- Phosphorylation
- RNA, Messenger/metabolism
- Transcription, Genetic
Collapse
Affiliation(s)
- Dawn O'Reilly
- Sir William Dunn School of Pathology, University of Oxford, South Parks Road, Oxford OX1 3RE, UK
| | | |
Collapse
|
31
|
Lavenu-Bombled C, Izac B, Legrand F, Cambot M, Vigier A, Massé JM, Dubart-Kupperschmitt A. Glycoprotein Ibalpha promoter drives megakaryocytic lineage-restricted expression after hematopoietic stem cell transduction using a self-inactivating lentiviral vector. Stem Cells 2007; 25:1571-7. [PMID: 17379771 DOI: 10.1634/stemcells.2006-0321] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Megakaryocytic (MK) lineage is an attractive target for cell/gene therapy approaches, aiming at correcting platelet protein deficiencies. However, MK cells are short-lived cells, and their permanent modification requires modification of hematopoietic stem cells with an integrative vector such as a lentiviral vector. Glycoprotein (Gp) IIb promoter, the most studied among the MK regulatory sequences, is also active in stem cells. To strictly limit transgene expression to the MK lineage after transduction of human CD34(+) hematopoietic cells with a lentiviral vector, we looked for a promoter activated later during MK differentiation. Human cord blood, bone marrow, and peripheral-blood mobilized CD34(+) cells were transduced with a human immunodeficiency virus-derived self-inactivating lentiviral vector encoding the green fluorescent protein (GFP) under the transcriptional control of GpIbalpha, GpIIb, or EF1alpha gene regulatory sequences. Both GpIbalpha and GpIIb promoters restricted GFP expression (analyzed by flow cytometry and immunoelectron microscopy) in MK cells among the maturing progeny of transduced cells. However, only the GpIbalpha promoter was strictly MK-specific, whereas GpIIb promoter was leaky in immature progenitor cells not yet engaged in MK cell lineage differentiation. We thus demonstrate the pertinence of using a 328-base-pair fragment of the human GpIbalpha gene regulatory sequence, in the context of a lentiviral vector, to tightly restrict transgene expression to the MK lineage after transduction of human CD34(+) hematopoietic cells. Disclosure of potential conflicts of interest is found at the end of this article.
Collapse
Affiliation(s)
- Cécile Lavenu-Bombled
- Institut Cochin, Department of Hematology, Hôpital de Port-Royal, 123 Bd de Port-Royal, Paris 75014, France
| | | | | | | | | | | | | |
Collapse
|
32
|
Bostrom MA, Boyanovsky BB, Jordan CT, Wadsworth MP, Taatjes DJ, de Beer FC, Webb NR. Group V Secretory Phospholipase A2Promotes Atherosclerosis. Arterioscler Thromb Vasc Biol 2007; 27:600-6. [PMID: 17204667 DOI: 10.1161/01.atv.0000257133.60884.44] [Citation(s) in RCA: 104] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE Group V secretory phospholipase A2 (GV sPLA2) has been detected in both human and mouse atherosclerotic lesions. This enzyme has potent hydrolytic activity towards phosphatidylcholine-containing substrates, including lipoprotein particles. Numerous studies in vitro indicate that hydrolysis of high density lipoproteins (HDL) and low density lipoproteins (LDL) by GV sPLA2 leads to the formation of atherogenic particles and potentially proinflammatory lipid mediators. However, there is no direct evidence that this enzyme promotes atherogenic processes in vivo. METHODS AND RESULTS We performed gain-of-function and loss-of-function studies to investigate the role of GV sPLA2 in atherogenesis in LDL receptor-deficient mice. Compared with control mice, animals overexpressing GV sPLA2 by retrovirus-mediated gene transfer had a 2.7 fold increase in lesion area in the ascending region of the aortic root. Increased atherosclerosis was associated with an increase in lesional collagen deposition in the same region. Mice deficient in bone marrow-derived GV sPLA2 had a 36% reduction in atherosclerosis in the aortic arch/thoracic aorta. CONCLUSIONS Our data in mouse models provide the first in vivo evidence that GV sPLA2 contributes to atherosclerotic processes, and draw attention to this enzyme as an attractive target for the treatment of atherosclerotic disease.
Collapse
Affiliation(s)
- Meredith A Bostrom
- Graduate Center for Nutritional Sciences, University of Kentucky, Lexington, KY, USA
| | | | | | | | | | | | | |
Collapse
|
33
|
Chang AH, Sadelain M. The Genetic Engineering of Hematopoietic Stem Cells: the Rise of Lentiviral Vectors, the Conundrum of the LTR, and the Promise of Lineage-restricted Vectors. Mol Ther 2007; 15:445-56. [PMID: 17228317 DOI: 10.1038/sj.mt.6300060] [Citation(s) in RCA: 69] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Recent studies on the integration patterns of different categories of retroviral vectors, the genotoxicity of long-terminal repeats (LTRs) and other genetic elements, the rise of lentiviral technology and the emergence of regulated vector systems providing tissue-restricted transgene expression and RNA interference, are profoundly changing the landscape of stem cell-based therapies. New developments in vector design and an increasing understanding of the mechanisms underlying insertional oncogenesis are ushering in a new phase in hematopoietic stem cell (HSC) engineering, thus bringing the hitherto exclusive reliance on LTR-driven, gamma-retroviral vectors to an end. Based on their ability to transduce non-dividing cells and their genomic stability, lentiviral vectors offer new prospects for the manipulation of HSCs. Tissue-specific vectors, as exemplified by globin vectors, not only provide therapeutic efficacy, but may also enhance safety, insofar that they restrict transgene expression in stem cells, progenitor cells and blood cells in all but the transcriptionally targeted lineage. This review provides a survey of these advances as well as several remaining challenges, focusing in particular on the importance of achieving adequate levels of protein expression from a limited number of vector copies per cell-ideally one to two.
Collapse
Affiliation(s)
- Alex H Chang
- Laboratory of Gene Transfer and Gene Expression, Memorial Sloan-Kettering Cancer Center, New York, New York, USA
| | | |
Collapse
|
34
|
Abstract
Despite the many studies of murine atherosclerosis, we do not yet know the relevance of the natural history of this model to the final events precipitated by plaque disruption of human atherosclerotic lesions. The literature has become particularly confused because of the common use of terms such as "instability", "vulnerable", "rupture", or even "thrombosis" for features of plaques in murine model systems not yet shown to rupture spontaneously and in an animal surprisingly resistant to formation of thrombi at sites of atherosclerosis. We suggest that use of conclusory terms like "vulnerable" and "stable" should be discouraged. Similarly, terms such as "buried fibrous caps" that imply preceding events that are unproven tend to create confusion. We will argue that such terminology may mislead readers by implying knowledge that does not yet exist. We suggest, instead, a focus on specific processes that various forms of data have implicated in plaque progression. For example, formation of the fibrous cap, protease activation, and cell death in the necrotic core can be well described and have all been modeled in well-defined experiments. The relevance of such well-defined, objective, descriptive observations in the mouse can be tested for relevance against data from human pathology.
Collapse
Affiliation(s)
- Stephen M Schwartz
- Department of Pathology, 815 Mercer Street, Room 421, University of Washington, Seattle, WA 98109-4714, USA.
| | | | | | | |
Collapse
|
35
|
He W, Qiang M, Ma W, Valente AJ, Quinones MP, Wang W, Reddick RL, Xiao Q, Ahuja SS, Clark RA, Freeman GL, Li S. Development of a synthetic promoter for macrophage gene therapy. Hum Gene Ther 2006; 17:949-59. [PMID: 16972763 DOI: 10.1089/hum.2006.17.949] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Macrophages have the potential to deliver therapeutic genes to many target tissues. Macrophage-specific synthetic promoters (SPs) generated by random ligation of myeloid/macrophage cis elements had activity up to 100-fold that of a native macrophage promoter in macrophage cell lines, but were minimally active in nonmyeloid cells. Mouse bone marrow cells (BMCs) transduced ex vivo with lentivectors expressing green fluorescent protein (GFP) driven either by an SP (SP-GFP) or a cytomegalovirus (CMV) promoter (CMV-GFP) were used for syngeneic transplantation of lethally irradiated mice. Blood leukocytes showed stable GFP expression for up to 15 months after transplantation. SP-GFP expression was selective for CD11b+ macrophages, whereas CMV-GFP expression was observed in erythrocytes, as well as in both CD11b+ and CD11b- leukocytes. Furthermore, SP-GFP expression was much stronger than CMV-GFP expression in CD11b+ macrophages. apoE-/- BMCs transduced with the lentiviral vector encoding human apoE were used to transplant apoE-/- mice. Macrophage expression of apoE from 10 to 26 weeks of age significantly reduced atherosclerotic lesions in recipient apoE-/- mice. Thus, the novel SPs, especially when combined with lentivectors, are useful for macrophage-specific delivery of therapeutic genes.
Collapse
Affiliation(s)
- Weijing He
- Department of Medicine, University of Texas Health Science Center at San Antonio, and South Texas Veterans Health Care System, Audie L. Murphy Division, San Antonio, TX 78229-3900, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
He W, Qiang M, Ma W, Valente AJ, Quinones MP, Wang W, Reddick RL, Xiao Q, Ahuja SS, Clark RA, Freeman GL, Li S. Development of a Synthetic Promoter for Macrophage Gene Therapy. Hum Gene Ther 2006. [DOI: 10.1089/hum.2006.17.ft-240] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
|
37
|
Jaalouk DE, Crosato M, Brodt P, Galipeau J. Inhibition of histone deacetylation in 293GPG packaging cell line improves the production of self-inactivating MLV-derived retroviral vectors. Virol J 2006; 3:27. [PMID: 16603064 PMCID: PMC1488828 DOI: 10.1186/1743-422x-3-27] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2005] [Accepted: 04/07/2006] [Indexed: 02/08/2023] Open
Abstract
Background Self-inactivating retroviral vectors (SIN) are often associated with very low titers. Promoter elements embedded within SIN designs may suppress transcription of packageable retroviral RNA which in turn results in titer reduction. We tested whether this dominant-negative effect involves histone acetylation state. We designed an MLV-derived SIN vector using the cytomegalovirus immediate early enhancer-promoter (CMVIE) as an embedded internal promoter (SINCMV) and transfected the pantropic 293GPG packaging cell line. Results The SINCMV retroviral producer had uniformly very low titers (~10,000 infectious retroparticles per ml). Northern blot showed low levels of expression of retroviral mRNA in producer cells in particular that of packageable RNA transcript. Treatment of the producers with the histone deacetylase (HDAC) inhibitors sodium butyrate and trichostatin A reversed transcriptional suppression and resulted in an average 106.3 ± 4.6 – fold (P = 0.002) and 15.5 ± 1.3 – fold increase in titer (P = 0.008), respectively. A histone gel assay confirmed increased histone acetylation in treated producer cells. Conclusion These results show that SIN retrovectors incorporating strong internal promoters such as CMVIE, are susceptible to transcriptional silencing and that treatment of the producer cells with HDAC inhibitors can overcome this blockade suggesting that histone deacetylation is implicated in the mechanism of transcriptional suppression.
Collapse
Affiliation(s)
- Diana E Jaalouk
- Department of Medicine, Lady Davis Institute for Medical Research, McGill University, Montreal, Canada
- Department of GU Medical Oncology, Unit 1374, The University of Texas M. D. Anderson Cancer Center, P.O. Box 301439, Houston, Texas, USA
| | - Milena Crosato
- Department of Medicine, Lady Davis Institute for Medical Research, McGill University, Montreal, Canada
| | - Pnina Brodt
- Department of Medicine, McGill University Health Center, McGill University, Montreal, Canada
- Department of Surgery, McGill University Health Center, McGill University, Montreal, Canada
| | - Jacques Galipeau
- Department of Medicine, Lady Davis Institute for Medical Research, McGill University, Montreal, Canada
- Division of Hematology/Oncology, Jewish General Hospital, McGill University, Montreal, Canada
| |
Collapse
|
38
|
Gough PJ, Gomez IG, Wille PT, Raines EW. Macrophage expression of active MMP-9 induces acute plaque disruption in apoE-deficient mice. J Clin Invest 2005; 116:59-69. [PMID: 16374516 PMCID: PMC1319218 DOI: 10.1172/jci25074] [Citation(s) in RCA: 340] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2005] [Accepted: 10/25/2005] [Indexed: 11/17/2022] Open
Abstract
The majority of acute clinical manifestations of atherosclerosis are due to the physical rupture of advanced atherosclerotic plaques. It has been hypothesized that macrophages play a key role in inducing plaque rupture by secreting proteases that destroy the extracellular matrix that provides physical strength to the fibrous cap. Despite reports detailing the expression of multiple proteases by macrophages in rupture-prone regions, there is no direct proof that macrophage-mediated matrix degradation can induce plaque rupture. We aimed to test this hypothesis by retrovirally overexpressing the candidate enzyme MMP-9 in macrophages of advanced atherosclerotic lesions of apoE-/- mice. Despite a greater than 10-fold increase in the expression of MMP-9 by macrophages, there was only a minor increase in the incidence of plaque fissuring. Subsequent analysis revealed that macrophages secrete MMP-9 predominantly as a proform, and this form is unable to degrade the matrix component elastin. Expression of an autoactivating form of MMP-9 in macrophages in vitro greatly enhances elastin degradation and induces significant plaque disruption when overexpressed by macrophages in advanced atherosclerotic lesions of apoE-/- mice in vivo. These data show that enhanced macrophage proteolytic activity can induce acute plaque disruption and highlight MMP-9 as a potential therapeutic target for stabilizing rupture-prone plaques.
Collapse
Affiliation(s)
- Peter J Gough
- Department of Pathology, University of Washington, Harborview Medical Center, Seattle, Washington 98104-2499, USA.
| | | | | | | |
Collapse
|
39
|
Abstract
Existing approaches to the treatment of refractory hypercholesterolaemia, severe hypertriglyceridaemia, low levels of high-density lipoprotein cholesterol and certain inherited disorders of intracellular lipid metabolism are ineffective in a substantial number of patients. Somatic gene therapy is considered to be a potential approach to the therapy of several of these lipid disorders. In many cases preclinical proof-of-principle studies have already been performed, and in one (homozygous familial hypercholesterolaemia) a clinical trial has been conducted. Other clinical gene therapy trials for dyslipidaemia are likely to be initiated within the next several years.
Collapse
Affiliation(s)
- Uli C Broedl
- University of Munich, Department of InternalMedicine II, Marchioninistr. 15, 81377 Munich, Germany.
| | | |
Collapse
|
40
|
Zhao S, Weinreich MA, Ihara K, Richard RE, Blau CA. In vivo selection of genetically modified erythroid cells using a jak2-based cell growth switch. Mol Ther 2005; 10:456-68. [PMID: 15336646 DOI: 10.1016/j.ymthe.2004.05.014] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2003] [Accepted: 05/07/2004] [Indexed: 10/26/2022] Open
Abstract
Cell-based therapies have potential widespread applications in clinical medicine, and methods for controlling the fate of transplanted cells are needed. We have previously described a means for directing the growth of genetically modified cells in vivo using a derivative of the thrombopoietin receptor, mpl, that is reversibly activated by a drug called a chemical inducer of dimerization (CID). Since Jak2 participates in signaling from a number of different cytokine receptors (including mpl), we tested whether direct activation of the JH1 domain of Jak2 would broaden the repertoire of hematopoietic lineages responsive to the CID. While the engineered Jak2 induced a significant rise in genetically modified red cells, as we have observed previously with mpl, it lacked mpl's ability to expand genetically modified platelets and failed to expand genetically modified granulocytes, B cells, or T cells. These findings identify a signaling molecule other than mpl that can function as a cell growth switch in vivo and demonstrate that signaling molecules used for in vivo selection need not be confined to receptors. The erythroid-restricted growth response suggests that CID-activated Jak2 may be well suited to gene therapy applications in sickle cell anemia or beta-thalassemia.
Collapse
Affiliation(s)
- Shengming Zhao
- Division of Hematology, Department of Medicine, Mail Stop 357710, K260 Health Sciences Building, University of Washington, Seattle, WA 98195, USA
| | | | | | | | | |
Collapse
|
41
|
Greenow K, Pearce NJ, Ramji DP. The key role of apolipoprotein E in atherosclerosis. J Mol Med (Berl) 2005; 83:329-42. [PMID: 15827760 DOI: 10.1007/s00109-004-0631-3] [Citation(s) in RCA: 162] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2004] [Accepted: 11/08/2004] [Indexed: 01/17/2023]
Abstract
Apolipoprotein E is a multifunctional protein that is synthesized by the liver and several peripheral tissues and cell types, including macrophages. The protein is involved in the efficient hepatic uptake of lipoprotein particles, stimulation of cholesterol efflux from macrophage foam cells in the atherosclerotic lesion, and the regulation of immune and inflammatory responses. Apolipoprotein E deficiency in mice leads to the development of atherosclerosis and re-expression of the protein reduces the extent of the disease. This review presents evidence for the potent anti-atherogenic action of apolipoprotein E and describes our current understanding of its multiple functions and regulation by factors implicated in the pathogenesis of cardiovascular disease.
Collapse
Affiliation(s)
- Kirsty Greenow
- Cardiff School of Biosciences, Cardiff University, Museum Avenue, P.O. Box 911, Cardiff CF10 3US, Wales, UK
| | | | | |
Collapse
|
42
|
Yasui K, Furuta RA, Matsumoto K, Tani Y, Fujisawa JI. HIV-1-derived self-inactivating lentivirus vector induces megakaryocyte lineage-specific gene expression. Microbes Infect 2005; 7:240-7. [PMID: 15725386 DOI: 10.1016/j.micinf.2004.11.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2004] [Revised: 11/02/2004] [Accepted: 11/02/2004] [Indexed: 11/24/2022]
Abstract
Pluripotent, self-renewing, hematopoietic stem cells are considered good targets for gene modification to treat a wide variety of disorders. However, as many genes are expressed in a stage-specific manner during the course of hematopoietic development, it is necessary to establish a lineage-specific gene expression system to ensure the proper expression of transduced genes in hematopoietic stem cells. In this study, we constructed a VSV-G-pseudotyped, human immunodeficiency virus type 1-based, self-inactivating lentivirus vector that expressed green fluorescent protein (GFP) under the control of the human CD41 (glycoprotein 2b; GP2b) promoter; this activity is restricted to megakaryocytic lineage cells. The recombinant virus was used to infect human peripheral blood CD34+ (hematopoietic stem/progenitor) cells, and lineage-specific gene expression was monitored with GFP measurements. The analysis by FACS determined that GFP expression driven by the GP2b promoter was restricted to megakaryocytic progenitors and was not present in erythrocytes. Furthermore, in the hematopoietic colony-forming assay, GFP expression was restricted to colony-forming units-megakaryocyte (CFU-Meg) colonies under the control of the GP2b promoter, whereas all myeloid colonies (burst-forming units-erythroid, colony-forming units-granulocyte-macrophage, and CFU-Meg) expressed GFP when the transgene was regulated by the cytomegalovirus promoter. These results demonstrated lineage-specific expression after gene transduction of hematopoietic stem cells. The application of this vector system should provide a useful tool for gene therapy to treat disorders associated with megakaryocyte (platelet) dysfunction.
Collapse
Affiliation(s)
- Kazuta Yasui
- Department of Microbiology, Kansai Medical University, 10-15 Fumizono-cho, Moriguchi, Osaka 570-8506, Japan
| | | | | | | | | |
Collapse
|
43
|
Moreau T, Bardin F, Imbert J, Chabannon C, Tonnelle C. Restriction of transgene expression to the B-lymphoid progeny of human lentivirally transduced CD34+ cells. Mol Ther 2005; 10:45-56. [PMID: 15233941 DOI: 10.1016/j.ymthe.2004.04.005] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2004] [Accepted: 04/05/2004] [Indexed: 11/16/2022] Open
Abstract
Development of gene transfer strategies will necessitate improved efficiency and control of transduction and transgene expression. We here provide evidence that targeting expression of the GFP reporter gene to the B-lymphoid progeny of genetically modified human hematopoietic progenitor cells can be achieved through the insertion of regulatory sequences from the human CD19 gene promoter into a lentiviral vector. Based on a bioinformatics approach, three human CD19-derived sequences were designed and inserted into a self-inactivated lentiviral vector backbone upstream of the GFP gene: S.CD19 (230 bp), M.CD19 (464 bp), and L.CD19 (1274 bp). These new lentiviral vectors efficiently transduced cord blood CD34(+) cells. The M.CD19 and especially L.CD19 sequences preferentially targeted GFP expression to in vitro and in vivo differentiated CD19(+) progeny; moreover, transgene expression was detected from the CD34(+) pro/pre-B cell to the mature peripheral IgM(+) B cell stage. In contrast, GFP expression was weak or absent in primary T-lymphoid and uncommitted progenitor cells or in erythroid, natural killer, or myeloid differentiated cells. Such B-lineage-specific lentiviral vectors may be useful for correcting inherited disorders that affect B-lymphoid cells or for deciphering the transcriptional program that controls B cell commitment and differentiation.
Collapse
Affiliation(s)
- Thomas Moreau
- Centre de Thérapie Cellulaire et Génique, Institut Paoli-Calmettes, Centre Régional de Lutte contre le Cancer Provence-Alpes-Côte d'Azur, Marseille, France
| | | | | | | | | |
Collapse
|
44
|
Dijon M, Torne-Celer C, Moreau T, Tonnelle C, Chabannon C. Expression and recombination of the EGFP and EYFP genes in lentiviral vectors carrying two heterologous promoters. Cytotherapy 2005; 7:417-26. [PMID: 16236631 DOI: 10.1080/14653240500319317] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
BACKGROUND Expressing two genes in the progeny of stem and progenitor cells that are transduced with a unique viral vector is desirable in certain situations. We tested the ability of two lentiviral vectors to transduce human cells of hematopoietic origin and concomitantly express two reporter genes, either EGFP (enhanced green fluorescent protein) and DsRed2, or EGFP and EYFP (enhanced yellow fluorescent protein), from two internal promoters. METHODS The vectors were generated from the pTRIP deltaU3 EF1alpha EGFP lentiviral vector. Following transduction of hematopoietic and non-hematopoietic cell lines, we performed FACS, PCR and Southern blot analyzes to quantify transduction, integration efficiencies and size of integrated lentiviral vectors, respectively. RESULTS The detection of DsRed2 fluorescence appeared unexpectedly low in human cells of hematopoietic origin. Alternatively, a modification in the flow cytometry assay allowed us to distinguish between the two overlapping fluorescence signals emitted by EGFP and EYFP, when transduced cells were excited with a 488-nm laser beam. However, the low frequency of double-positive EGFP+ EYFP+ cells, and the existence of single-positive, mostly EGFP- EYFP+, cells, prompted us to search for recombinations in the vector sequence. Southern blotting of DNA obtained from transduced cells indeed demonstrated that recombination had occurred between the two closely related EGFP and EYFP sequences. DISCUSSION These observations suggest that recombination occurred within the EGFP and EYFP genes, which differ by only four amino acids. We conclude that the insertion of two highly homologous sequences into a lentiviral backbone can favor recombination.
Collapse
Affiliation(s)
- M Dijon
- Centre de Thérapie Cellulaire et Génique, Institut Paoli-Calmettes, Centre Régional de Lutte Contre le Cancer Provence-Alpes-Côte d'Azur, and Unité Mixte de Recherche 599, Institut de Recherche sur le Cancer de Marseille, Marseille, France
| | | | | | | | | |
Collapse
|
45
|
Hsieh CS, Liang Y, Tyznik AJ, Self SG, Liggitt D, Rudensky AY. Recognition of the peripheral self by naturally arising CD25+ CD4+ T cell receptors. Immunity 2004; 21:267-77. [PMID: 15308106 DOI: 10.1016/j.immuni.2004.07.009] [Citation(s) in RCA: 552] [Impact Index Per Article: 26.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2004] [Revised: 06/17/2004] [Accepted: 06/23/2004] [Indexed: 12/13/2022]
Abstract
Naturally arising CD25+ CD4+ regulatory T cells (TR) play an important role in the prevention of autoimmunity. TCR specificity is thought to play a critical role in TR development and function, but the repertoire and specificity of TR TCRs remain largely unknown. We find by sequencing of TRAV14 (Valpha2) TCRalpha chains associated with a transgenic TCRbeta chain that the TRand CD25- CD4+ TCR repertoires are similarly diverse, yet only partially overlapping. Retroviral expression of TCRalpha genes in TCR transgenic RAG-deficient T cells revealed that a high frequency of TCRs derived from CD25+ but not CD25- CD4+ T cells confers the ability to rapidly expand upon transfer into a lymphopenic host. Thus, these data show that a large proportion of naturally arising TR have substantially more efficient interactions with MHC class II bound peptides from the peripheral self than CD25- T cells.
Collapse
Affiliation(s)
- Chyi-Song Hsieh
- Department of Medicine, Division of Rheumatology, University of Washington, Seattle, Washington 98195, USA
| | | | | | | | | | | |
Collapse
|
46
|
Meir KS, Leitersdorf E. Atherosclerosis in the apolipoprotein-E-deficient mouse: a decade of progress. Arterioscler Thromb Vasc Biol 2004; 24:1006-14. [PMID: 15087308 DOI: 10.1161/01.atv.0000128849.12617.f4] [Citation(s) in RCA: 347] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Arguably the most critical advancement in the elucidation of factors affecting atherogenesis has been the development of mouse models of atherosclerosis. Among available models, the apolipoprotein E-deficient (apoE-/-) mouse is particularly popular because of its propensity to spontaneously develop atherosclerotic lesions on a standard chow diet. A Medline search reveals over 645 articles dedicated to studies using this reliable and convenient "super" animal model since its inception (Piedrahita JA et al, Proc Natl Acad Sci U S A 1992;89:4471-4475; Plump AS et al, Cell 1992;71:343-353) with a more or less steady increase from year to year. This review will examine our present understanding of the pathology and progression of plaques in this animal and highlight some of the nutritional, pharmacological, and genetic studies that have enhanced this understanding.
Collapse
Affiliation(s)
- Karen S Meir
- Department of Pathology, Hadassah University Hospital, Kiryat Hadassah, Jerusalem, Israel
| | | |
Collapse
|
47
|
Gough PJ, Garton KJ, Wille PT, Rychlewski M, Dempsey PJ, Raines EW. A Disintegrin and Metalloproteinase 10-Mediated Cleavage and Shedding Regulates the Cell Surface Expression of CXC Chemokine Ligand 16. THE JOURNAL OF IMMUNOLOGY 2004; 172:3678-85. [PMID: 15004171 DOI: 10.4049/jimmunol.172.6.3678] [Citation(s) in RCA: 194] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
CXC chemokine ligand (CXCL)16 and scavenger receptor for phosphatidylserine and oxidized low-density lipoprotein were independently identified as a chemokine and a scavenger receptor, respectively, but have since been shown to be identical. CXCL16 is synthesized as a transmembrane protein with its chemokine domain at the end of a mucin-rich stalk. When expressed at the cell surface, CXCL16 functions as a scavenger receptor, binding and internalizing oxidized low-density lipoprotein and bacteria. As a soluble form, CXCL16 is a chemoattractant for activated CD4+ and CD8+ T cells through binding its receptor, CXCR6. In this study, we examined the mechanisms that regulate the conversion between these two functionally distinct forms of CXCL16. We demonstrate that murine CXCL16 is synthesized as an intracellular precursor that is rapidly transported to the cell surface where it undergoes metalloproteinase-dependent cleavage, causing the release of a fragment that constitutes the majority of the CXCL16 extracellular domain. Using a novel retroviral system for the generation of short interfering RNAs, we show that knockdown of a disintegrin and metalloproteinase (ADAM) family protease ADAM10 decreases this constitutive shedding of CXCL16. Furthermore, we show that overexpression of ADAM10 increases CXCL16 shedding, whereas overexpression of a dominant-negative form of ADAM10 lowers shedding of CXCL16 in a similar manner to short interfering RNAs. Through the modulation of ADAM10 function, we demonstrate that ADAM10-mediated constitutive shedding is a key regulator of CXCL16 cell surface expression. The identification of ADAM10 as a major protease responsible for the conversion of CXCL16 from a membrane-bound scavenger receptor to a soluble chemoattractant will provide new information for understanding the physiological function of this molecule.
Collapse
MESH Headings
- ADAM Proteins
- ADAM10 Protein
- ADAM17 Protein
- Amyloid Precursor Protein Secretases
- Animals
- Catalysis
- Cell Line
- Cell Membrane/genetics
- Cell Membrane/immunology
- Cell Membrane/metabolism
- Chemokine CXCL16
- Chemokine CXCL6
- Chemokines, CXC/antagonists & inhibitors
- Chemokines, CXC/biosynthesis
- Chemokines, CXC/genetics
- Chemokines, CXC/metabolism
- Disintegrins/physiology
- Genetic Vectors
- Hydrolysis
- Macrophages, Peritoneal/enzymology
- Macrophages, Peritoneal/immunology
- Macrophages, Peritoneal/metabolism
- Membrane Proteins/antagonists & inhibitors
- Membrane Proteins/biosynthesis
- Membrane Proteins/genetics
- Membrane Proteins/metabolism
- Membrane Proteins/physiology
- Metalloendopeptidases/antagonists & inhibitors
- Metalloendopeptidases/genetics
- Metalloendopeptidases/physiology
- Mice
- Mice, Inbred C57BL
- Protein Processing, Post-Translational/immunology
- Protein Structure, Tertiary/genetics
- RNA, Small Interfering/biosynthesis
- RNA, Small Interfering/genetics
- RNA, Small Interfering/physiology
- Receptors, Immunologic/antagonists & inhibitors
- Receptors, Immunologic/biosynthesis
- Receptors, Immunologic/genetics
- Receptors, Immunologic/metabolism
- Receptors, Scavenger
Collapse
Affiliation(s)
- Peter J Gough
- Department of Pathology, University of Washington, Harborview Medical Center, Seattle, WA 98104, USA.
| | | | | | | | | | | |
Collapse
|
48
|
Pastorino S, Carta L, Puppo M, Melillo G, Bosco MC, Varesio L. Picolinic acid- or desferrioxamine-inducible autocrine activation of macrophages engineered to produce IFNγ: an approach for gene therapy. Gene Ther 2004; 11:560-8. [PMID: 14961065 DOI: 10.1038/sj.gt.3302217] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Macrophage (Mphi)-based vectors are highly mobile cellular shuttles designed to deliver therapeutic genes within the tissues. We engineered a mouse Mphi cell line to express the murine interferon-gamma (IFNgamma) under the control of an inducible promoter containing the hypoxia-responsive element, which can be triggered by hypoxia and other stimuli. We show that this Mphi vector can be induced to produce IFNgamma under normoxic conditions by stimulation with picolinic acid (PA), a catabolite of tryptophan, or desferrioxamine (DFX), an iron-chelating drug. The Mphi vector responds to IFNgamma with the induction of IRF-1 and of other IFNgamma-inducible genes, the expression of Ia antigens and induction of phagocytic activity. Inducible nitric oxygen synthase gene expression, nitric oxide production, as well as TNFalpha secretion were enhanced by PA or DFX as the sole stimuli. None of the above responses could be triggered individually by PA or DFX in control, normal Mphi, indicating that the Mphi vector overcame the need for costimulatory molecules derived from the immune system for its full activation. Furthermore, we demonstrate that extracellular iron can downregulate such response, thereby identifying an additional tool for the fine tuning of the Mphi vector response to stimulation.
Collapse
Affiliation(s)
- S Pastorino
- Laboratory of Molecular Biology, G Gaslini Institute, Largo Gaslini, Genova, Italy
| | | | | | | | | | | |
Collapse
|
49
|
Affiliation(s)
- Minghan Wang
- Department of Cardiovascular and Metabolic Diseases, Pharmacia Corporation, 800 North Lindbergh Boulevard, St Louis, Missouri 63167, USA.
| | | |
Collapse
|
50
|
Gough PJ, Raines EW. Advances in retroviral transduction of hematopoietic stem cells for the gene therapy of atherosclerosis. Curr Opin Lipidol 2003; 14:491-7. [PMID: 14501588 DOI: 10.1097/00041433-200310000-00010] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
PURPOSE OF REVIEW Atherosclerosis is a chronic inflammatory disease that is the primary cause of morbidity and mortality in the developed world. Many studies have shown that macrophages and T-cells play critical roles in multiple aspects of the pathogenesis of the disease. Given that these cells are ultimately derived from bone marrow precursors, the concept of performing gene therapy for atherosclerosis through the retroviral transduction of hematopoietic stem cells has received much attention. This review will highlight recent advances that will help bring this goal closer. RECENT FINDINGS The clinical application of retroviral gene transfer into hematopoietic stem cells has been hampered, in part, by the absence of vectors that can direct long-lasting, cell-type specific gene expression. In this review we will detail recent developments in the design of novel retroviral and lentiviral vectors that appear to overcome these problems, offering approaches to express therapeutic genes in specific cell-types within atherosclerotic lesions. We will also highlight advances in our understanding of the pathogenesis of atherosclerosis that may offer new gene therapeutic targets. SUMMARY The use of retroviral transduction of hematopoietic stem cells for treatment of patients with atherosclerosis still remains a long-term goal. However, the recent development of retroviral vectors capable of directing expression to specific cell types within the lesion will allow more targeted therapeutic strategies to be devised. In addition, these vectors will provide powerful experimental tools to further our understanding of the pathogenesis of the disease.
Collapse
Affiliation(s)
- Peter J Gough
- Department of Pathology, University of Washington, Harborview Medical Center, Seattle, Washington 98104-2499, USA.
| | | |
Collapse
|