1
|
Strasser AS, Gonzalez-Reiche AS, Zhou X, Valdebenito-Maturana B, Ye X, Zhang B, Wu M, van Bakel H, Jabs EW. Limb reduction in an Esco2 cohesinopathy mouse model is mediated by p53-dependent apoptosis and vascular disruption. Nat Commun 2024; 15:7154. [PMID: 39168984 PMCID: PMC11339411 DOI: 10.1038/s41467-024-51328-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Accepted: 08/01/2024] [Indexed: 08/23/2024] Open
Abstract
Roberts syndrome (RBS) is an autosomal recessive disorder with profound growth deficiency and limb reduction caused by ESCO2 loss-of-function variants. Here, we elucidate the pathogenesis of limb reduction in an Esco2fl/fl;Prrx1-CreTg/0 mouse model using bulk- and single-cell-RNA-seq and gene co-expression network analyses during embryogenesis. Our results reveal morphological and vascular defects culminating in hemorrhage of mutant limbs at E12.5. Underlying this abnormal developmental progression is a pre-apoptotic, mesenchymal cell population specific to mutant limb buds enriched for p53-related signaling beginning at E9.5. We then characterize these p53-related processes of cell cycle arrest, DNA damage, cell death, and the inflammatory leukotriene signaling pathway in vivo. In utero treatment with pifithrin-α, a p53 inhibitor, rescued the hemorrhage in mutant limbs. Lastly, significant enrichments were identified among genes associated with RBS, thalidomide embryopathy, and other genetic limb reduction disorders, suggesting a common vascular etiology among these conditions.
Collapse
Affiliation(s)
- Arielle S Strasser
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, USA
| | - Ana Silvia Gonzalez-Reiche
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, USA
| | - Xianxiao Zhou
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, USA
- Mount Sinai Center for Transformative Disease Modeling, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, USA
| | - Braulio Valdebenito-Maturana
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, USA
| | - Xiaoqian Ye
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, USA
| | - Bin Zhang
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, USA
- Mount Sinai Center for Transformative Disease Modeling, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, USA
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, USA
| | - Meng Wu
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, USA.
- Department of Clinical Genomics, Mayo Clinic, 200 First Street, Rochester, MN, USA.
- Department of Biochemistry and Molecular Biology, Mayo Clinic, 200 First Street, Rochester, MN, USA.
| | - Harm van Bakel
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, USA.
- Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, USA.
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, USA.
- Department of Artificial Intelligence and Human Health, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, USA.
| | - Ethylin Wang Jabs
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, USA.
- Department of Clinical Genomics, Mayo Clinic, 200 First Street, Rochester, MN, USA.
- Department of Biochemistry and Molecular Biology, Mayo Clinic, 200 First Street, Rochester, MN, USA.
- Department of Cell, Development and Regenerative Biology, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, USA.
- Department of Pediatrics, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, USA.
| |
Collapse
|
2
|
Liu Y, Li X, Zhou X, Wang J, Ao X. FADD as a key molecular player in cancer progression. Mol Med 2022; 28:132. [DOI: 10.1186/s10020-022-00560-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Revised: 10/05/2022] [Accepted: 10/19/2022] [Indexed: 11/11/2022] Open
Abstract
AbstractCancer is a leading disease-related cause of death worldwide. Despite advances in therapeutic interventions, cancer remains a major global public health problem. Cancer pathogenesis is extremely intricate and largely unknown. Fas-associated protein with death domain (FADD) was initially identified as an adaptor protein for death receptor-mediated extrinsic apoptosis. Recent evidence suggests that FADD plays a vital role in non-apoptotic cellular processes, such as proliferation, autophagy, and necroptosis. FADD expression and activity of are modulated by a complicated network of processes, such as DNA methylation, non-coding RNA, and post-translational modification. FADD dysregulation has been shown to be closely associated with the pathogenesis of numerous types of cancer. However, the detailed mechanisms of FADD dysregulation involved in cancer progression are still not fully understood. This review mainly summarizes recent findings on the structure, functions, and regulatory mechanisms of FADD and focuses on its role in cancer progression. The clinical implications of FADD as a biomarker and therapeutic target for cancer patients are also discussed. The information reviewed herein may expand researchers’ understanding of FADD and contribute to the development of FADD-based therapeutic strategies for cancer patients.
Collapse
|
3
|
Aravani D, Foote K, Figg N, Finigan A, Uryga A, Clarke M, Bennett M. Cytokine regulation of apoptosis-induced apoptosis and apoptosis-induced cell proliferation in vascular smooth muscle cells. Apoptosis 2020; 25:648-662. [PMID: 32627119 PMCID: PMC7527356 DOI: 10.1007/s10495-020-01622-4] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Vascular smooth muscle cells (VSMCs) are the main structural cell of blood vessels, and VSMC apoptosis occurs in vascular disease, after injury, and in vessel remodeling during development. Although VSMC apoptosis is viewed as silent, recent studies show that apoptotic cells can promote apoptosis-induced compensatory proliferation (AICP), apoptosis-induced apoptosis (AIA), and migration of both local somatic and infiltrating inflammatory cells. However, the effects of VSMC apoptosis on adjacent VSMCs, and their underlying signaling and mechanisms are unknown. We examined the consequences of VSMC apoptosis after activating extrinsic and intrinsic death pathways. VSMCs undergoing apoptosis through Fas/CD95 or the protein kinase inhibitor staurosporine transcriptionally activated interleukin 6 (IL-6) and granulocyte-macrophage colony stimulating factor (GM-CSF), leading to their secretion. Apoptosis induced activation of p38MAPK, JNK, and Akt, but neither p38 and JNK activation nor IL-6 or GM-CSF induction required caspase cleavage. IL-6 induction depended upon p38 activity, while Fas-induced GM-CSF expression required p38 and JNK. Conditioned media from apoptotic VSMCs induced VSMC apoptosis in vitro, and IL-6 and GM-CSF acted as pro-survival factors for AIA. VSMC apoptosis was studied in vivo using SM22α-DTR mice that express the diphtheria toxin receptor in VSMCs only. DT administration induced VSMC apoptosis and VSMC proliferation, and also signficantly induced IL-6 and GM-CSF. We conclude that VSMC apoptosis activates multiple caspase-independent intracellular signaling cascades, leading to release of soluble cytokines involved in regulation of both cell proliferation and apoptosis. VSMC AICP may ameliorate while AIA may amplify the effects of pro-apoptotic stimuli in vessel remodeling and disease.
Collapse
Affiliation(s)
- Dimitra Aravani
- Division of Cardiovascular Medicine, University of Cambridge, ACCI, Addenbrooke's Hospital, Box 110, CB2 0QQ, Cambridge, UK
| | - Kirsty Foote
- Division of Cardiovascular Medicine, University of Cambridge, ACCI, Addenbrooke's Hospital, Box 110, CB2 0QQ, Cambridge, UK
| | - Nichola Figg
- Division of Cardiovascular Medicine, University of Cambridge, ACCI, Addenbrooke's Hospital, Box 110, CB2 0QQ, Cambridge, UK
| | - Alison Finigan
- Division of Cardiovascular Medicine, University of Cambridge, ACCI, Addenbrooke's Hospital, Box 110, CB2 0QQ, Cambridge, UK
| | - Anna Uryga
- Division of Cardiovascular Medicine, University of Cambridge, ACCI, Addenbrooke's Hospital, Box 110, CB2 0QQ, Cambridge, UK
| | - Murray Clarke
- Division of Cardiovascular Medicine, University of Cambridge, ACCI, Addenbrooke's Hospital, Box 110, CB2 0QQ, Cambridge, UK
| | - Martin Bennett
- Division of Cardiovascular Medicine, University of Cambridge, ACCI, Addenbrooke's Hospital, Box 110, CB2 0QQ, Cambridge, UK.
| |
Collapse
|
4
|
Lu Y, Guo J, Zhu S, Zhang H, Zhu Q, Li Y. LncRNA HCG18 is critical for vascular smooth muscle cell proliferation and phenotypic switching. Hum Cell 2020; 33:537-544. [PMID: 32449112 DOI: 10.1007/s13577-020-00366-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Accepted: 04/21/2020] [Indexed: 11/25/2022]
Abstract
Previous studies have shown that some specific long non-coding RNAs are dysregulated in vascular walls and abnormally expressed in vascular disease. LncRNA HLA complex group 18 (HCG18) is a member of the HLA complex group, which has been rarely investigated in human diseases. In this study, we aimed to investigate the role of HCG in vascular smooth muscle cells. HCG18 was over-expressed by adenovirus transfection and knocked down in vascular smooth muscle cells by shRNA. Cell proliferation was detected by CCK-8 assays. Flow cytometry was employed to test the impacts of HCG18 on vascular smooth muscle apoptotic cells. The expression of associated genes in protein and mRNA levels was detected by western blotting, immunofluorescence and qRT-PCR. The interactions between HCG18 and fused in sarcoma (FUS) were confirmed by RNA EMSA and RIP assays. The expression of serum HCG18 was decreased in hypertensive patients and PDGF-BB-treated vascular smooth muscle cells. HCG18 inhibited proliferation and induced apoptotic cells in vascular smooth muscle cells. In addition, we also found that HCG18 can inhibit vascular smooth muscle cell phenotypic switching from a contractile to a secretory phenotype. Finally, our results showed that HCG18 enhanced apoptotic cells by directly binding with FUS. Our findings reveal that HCG18 is involved in the regulation of proliferation, apoptosis and the expression levels of markers of the contractile and synthetic phenotype.
Collapse
Affiliation(s)
- Yanjiao Lu
- Department of Cardiology, Huaihe Hospital, Henan University, Kaifeng, 475000, China
| | - Jingjing Guo
- Department of Cardiology, Huaihe Hospital, Henan University, Kaifeng, 475000, China
| | - Shengnan Zhu
- Department of Cardiology, Huaihe Hospital, Henan University, Kaifeng, 475000, China
| | - Han Zhang
- Department of Cardiology, Huaihe Hospital, Henan University, Kaifeng, 475000, China
| | - Qing Zhu
- Department of Cardiology, Huaihe Hospital, Henan University, Kaifeng, 475000, China
| | - Yanming Li
- Department of Cardiology, Huaihe Hospital, Henan University, Kaifeng, 475000, China.
| |
Collapse
|
5
|
Li H, Chen T, Sun H, Wu X, Jiang X, Ren C. Functional characterisation of Holothuria leucospilota Fas-associated death domain in the innate immune-related signalling pathways. Innate Immun 2019; 26:138-145. [PMID: 31550955 PMCID: PMC7016406 DOI: 10.1177/1753425919877680] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
In this study, the functions of Holothuria leucospilota Fas-associated death domain (HLFADD) in the innate immune-related signalling pathways were investigated. The results showed that over-expression of HLFADD in HEK293T cells could activate the transcription factors NF-κB and activator protein-1 (AP-1), and induce the secretion of downstream pro-inflammatory cytokines IL-6, IL-8 and IL-18, suggesting the involvement of the sea cucumber FADD in activating the NF-κB and c-Jun NH2-terminal kinase-dependent pathways. On the other hand, HLFADD could down-regulate the activations of NF-κB and AP-1 that induced by over-expression of H. leucospilota myeloid differentiation factor 88 (HLMyD88), which is supposed to be mediated through its interaction with HLMyD88 to keep the MyD88-dependent TLR signalling at a proper magnitude. The interaction of HLFADD and HLMyD88 were further supported by a co-immunoprecipitation assay. Moreover, HLFADD could activate transcription factor IFN regulatory factor-3 and induced the secretion of downstream IFN-α and IFN-β, indicating that the sea cucumber FADD may also activate the antiviral IFN signalling pathway. In summary, our study may give new insights on the functions of sea cucumber FADD in the innate immune-related signalling pathways.
Collapse
Affiliation(s)
- Haipeng Li
- School of Environmental Science and Engineering, Guangzhou University, PR China
| | - Ting Chen
- CAS Key laboratory of Tropical Marine Bio-Resources and Ecology (LMB), Guangdong Provincial Key Laboratory of Applied Marine Biology (LAMB), South China Sea Institute of Oceanology, Chinese Academy of Sciences, PR China.,Institution of South China Sea Ecology and Environmental Engineering, Chinese Academy of Sciences, ISEE, CAS, PR China
| | - Hongyan Sun
- College of Marine Sciences, South China Agricultural University, PR China
| | - Xiaofen Wu
- CAS Key laboratory of Tropical Marine Bio-Resources and Ecology (LMB), Guangdong Provincial Key Laboratory of Applied Marine Biology (LAMB), South China Sea Institute of Oceanology, Chinese Academy of Sciences, PR China.,Institution of South China Sea Ecology and Environmental Engineering, Chinese Academy of Sciences, ISEE, CAS, PR China
| | - Xiao Jiang
- CAS Key laboratory of Tropical Marine Bio-Resources and Ecology (LMB), Guangdong Provincial Key Laboratory of Applied Marine Biology (LAMB), South China Sea Institute of Oceanology, Chinese Academy of Sciences, PR China.,Institution of South China Sea Ecology and Environmental Engineering, Chinese Academy of Sciences, ISEE, CAS, PR China
| | - Chunhua Ren
- CAS Key laboratory of Tropical Marine Bio-Resources and Ecology (LMB), Guangdong Provincial Key Laboratory of Applied Marine Biology (LAMB), South China Sea Institute of Oceanology, Chinese Academy of Sciences, PR China.,Institution of South China Sea Ecology and Environmental Engineering, Chinese Academy of Sciences, ISEE, CAS, PR China
| |
Collapse
|
6
|
Kural MH, Wang J, Gui L, Yuan Y, Li G, Leiby KL, Quijano E, Tellides G, Saltzman WM, Niklason LE. Fas ligand and nitric oxide combination to control smooth muscle growth while sparing endothelium. Biomaterials 2019; 212:28-38. [PMID: 31102854 DOI: 10.1016/j.biomaterials.2019.05.011] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Revised: 04/21/2019] [Accepted: 05/06/2019] [Indexed: 01/08/2023]
Abstract
Metallic stents cause vascular wall damage with subsequent smooth muscle cell (SMC) proliferation, neointimal hyperplasia, and treatment failure. To combat in-stent restenosis, drug-eluting stents (DES) delivering mTOR inhibitors such as sirolimus or everolimus have become standard for coronary stenting. However, the relatively non-specific action of mTOR inhibitors prevents efficient endothelium recovery and mandates dual antiplatelet therapy to prevent thrombosis. Unfortunately, long-term dual antiplatelet therapy leads to increased risk of bleeding/stroke and, paradoxically, myocardial infarction. Here, we took advantage of the fact that nitric oxide (NO) increases Fas receptors on the SMC surface. Fas forms a death-inducing complex upon binding to Fas ligand (FasL), while endothelial cells (ECs) are relatively resistant to this pathway. Selected doses of FasL and NO donor synergistically increased SMC apoptosis and inhibited SMC growth more potently than did everolimus or sirolimus, while having no significant effect on EC viability and proliferation. This differential effect was corroborated in ex vivo pig coronaries, where the neointimal formation was inhibited by the drug combination, but endothelial viability was retained. We also deployed FasL-NO donor-releasing ethylene-vinyl acetate copolymer (EVAc)-coated stents into pig coronary arteries, and cultured them in perfusion bioreactors for one week. FasL and NO donor, released from the stent coating, killed SMCs close to the stent struts, even in the presence of flow rates mimicking those of native arteries. Thus, the FasL-NO donor-combination has a potential to prevent intimal hyperplasia and in-stent restenosis, without harming endothelial restoration, and hence may be a superior drug delivery strategy for DES.
Collapse
Affiliation(s)
- Mehmet H Kural
- Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, CT 06520, USA; Department of Anesthesiology, Yale University, New Haven, CT 06519, USA.
| | - Juan Wang
- Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, CT 06520, USA; Department of Anesthesiology, Yale University, New Haven, CT 06519, USA
| | - Liqiong Gui
- Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, CT 06520, USA; Department of Anesthesiology, Yale University, New Haven, CT 06519, USA
| | - Yifan Yuan
- Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, CT 06520, USA; Department of Anesthesiology, Yale University, New Haven, CT 06519, USA
| | - Guangxin Li
- Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, CT 06520, USA; Department of Surgery, Yale University, New Haven, CT 06519, USA
| | - Katherine L Leiby
- Department of Biomedical Engineering, Yale University, New Haven, CT 06519, USA
| | - Elias Quijano
- Department of Biomedical Engineering, Yale University, New Haven, CT 06519, USA
| | - George Tellides
- Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, CT 06520, USA; Department of Surgery, Yale University, New Haven, CT 06519, USA
| | - W Mark Saltzman
- Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, CT 06520, USA; Department of Biomedical Engineering, Yale University, New Haven, CT 06519, USA
| | - Laura E Niklason
- Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, CT 06520, USA; Department of Anesthesiology, Yale University, New Haven, CT 06519, USA; Department of Biomedical Engineering, Yale University, New Haven, CT 06519, USA; Yale Stem Cell Center, New Haven, CT 06520, USA
| |
Collapse
|
7
|
Maifrino LBM, Lima NEAD, Marques MR, Cardoso CG, Souza LBD, Tomé TDC, Quintana HT, Oliveira FD, Reis BDCAA, Fonseca FLA. Evaluation of Collagen Fibers, MMP2, MMP9, 8-OHdG and Apoptosis in the Aorta of Ovariectomized LDL Knockout Mice Submitted to Aerobic Exercise. Arq Bras Cardiol 2018; 112:180-188. [PMID: 30570072 PMCID: PMC6371828 DOI: 10.5935/abc.20180263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2017] [Accepted: 07/23/2018] [Indexed: 11/20/2022] Open
Abstract
Background In menopause, there is greater cellular exposure to oxidative stress, related
to the decreased antioxidative effects of estrogen. These metabolic changes
favor the progression of cardiovascular diseases, such as atherosclerosis.
Abnormal function of the aorta - the most important artery - is associated
with many cardiovascular diseases. Collagen, especially types I and III, is
one of the most important aortic wall components and it can be affected by
many factors, including menopause. The 8-OHdG is one of the main markers of
DNA oxidative damage induced by reactive oxygen species (ROS). Objective We aimed to investigate effects of moderate aerobic training on the ascending
aorta of LDL-knockout (LDL-KO) and ovariectomized female mice. Methods A total of 15 C57BL/6 mice and 15 LDL-KO mice were divided into experimental
groups. The thickness and volume density of types I and III collagen fibers
were performed by morphoquantitative analysis, whereas the MMP-2 and MMP-9
and 8-OHdG were detected by immunohistochemistry and apoptosis was detected
by the TUNEL assay. The significance level for all tests was p <
0.05. Results Exercise causes an increase in the thickness of the aorta in LDL-KO groups,
particularly accentuated in the ovariectomized groups. The type I collagen
fibers showed an increase in volume density influenced by training in both
Control groups and in the LDL-KO group. Type III collagen density decreased
in both groups. The MMP-2 showed moderade immunostaining in the tunica media
in LDL-KO groups, which did not occur in the control groups and the MMP-9
stained irregularly in all tissues. The marker 8-OhdG was stronger in the
exercise training groups. Additionally, the ovariectomy, the exercise
training and the LDL-KO treatments increased apoptosis. Conclusion These results suggest that moderate-intensity aerobic exercise in
ovariectomized mice associated to an increase in LDL rate possibly increases
oxidative stress and apoptosis induction.
Collapse
Affiliation(s)
| | | | | | | | - Lidiane B de Souza
- Departamento de Biociências da Universidade Federal de São Paulo, São Paulo, SP - Brazil
| | | | | | - Flavia de Oliveira
- Departamento de Biociências da Universidade Federal de São Paulo, São Paulo, SP - Brazil
| | | | | |
Collapse
|
8
|
Mouasni S, Tourneur L. FADD at the Crossroads between Cancer and Inflammation. Trends Immunol 2018; 39:1036-1053. [PMID: 30401514 DOI: 10.1016/j.it.2018.10.005] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2018] [Revised: 10/02/2018] [Accepted: 10/09/2018] [Indexed: 12/19/2022]
Abstract
Initially described as an adaptor molecule for death receptor (DR)-mediated apoptosis, Fas-associated death domain (FADD) was later implicated in nonapoptotic cellular processes. During the last decade, FADD has been shown to participate and regulate most of the signalosome complexes, including necrosome, FADDosome, innateosome, and inflammasome. Given the role of these signaling complexes, FADD has emerged as a new actor in innate immunity, inflammation, and cancer development. Concomitant to these new roles, a surprising number of mechanisms deemed to regulate FADD functions have been identified, including post-translational modifications of FADD protein and FADD secretion. This review focuses on recent knowledge of the biological roles of FADD, a pleiotropic molecule having multiple partners, and its impact in cancer, innate immunity, and inflammation.
Collapse
Affiliation(s)
- Sara Mouasni
- Department of Infection, Immunity and Inflammation, Cochin Institute, 75014 Paris, France; INSERM, U1016, Paris, France; CNRS, UMR8104, Paris, France; Paris Descartes University, Sorbonne Paris Cité, Paris, France
| | - Léa Tourneur
- Department of Infection, Immunity and Inflammation, Cochin Institute, 75014 Paris, France; INSERM, U1016, Paris, France; CNRS, UMR8104, Paris, France; Paris Descartes University, Sorbonne Paris Cité, Paris, France.
| |
Collapse
|
9
|
Osonoi Y, Mita T, Azuma K, Nakajima K, Masuyama A, Goto H, Nishida Y, Miyatsuka T, Fujitani Y, Koike M, Mitsumata M, Watada H. Defective autophagy in vascular smooth muscle cells enhances cell death and atherosclerosis. Autophagy 2018; 14:1991-2006. [PMID: 30025494 DOI: 10.1080/15548627.2018.1501132] [Citation(s) in RCA: 106] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Macroautophagy/autophagy is considered as an evolutionarily conserved cellular catabolic process. In this study, we aimed to elucidate the role of autophagy in vascular smooth muscle cells (SMCs) on atherosclerosis. SMCs cultured from mice with SMC-specific deletion of the essential autophagy gene atg7 (Atg7cKO) showed reduced serum-induced cell growth, increased cell death, and decreased cell proliferation rate. Furthermore, 7-ketocholestrerol enhanced apoptosis and the expression of CCL2 (chemokine [C-C motif] ligand 2) with the activation of TRP53, the mouse ortholog of human and rat TP53, in SMCs from Atg7cKO mice. In addition, Atg7cKO mice crossed with Apoe (apolipoprotein E)-deficient mice (apoeKO; Atg7cKO:apoeKO) showed reduced medial cellularity and increased TUNEL-positive cells in the descending aorta at 10 weeks of age. Intriguingly, Atg7cKO: apoeKO mice fed a Western diet containing 1.25% cholesterol for 14 weeks showed a reduced survival rate. Autopsy of the mice demonstrated the presence of aortic rupture. Analysis of the descending aorta in Atg7cKO:apoeKO mice showed increased plaque area, increased TUNEL-positive area, decreased SMC-positive area, accumulation of macrophages in the media, and adventitia and perivascular tissue, increased CCL2 expression in SMCs in the vascular wall, medial disruption, and aneurysm formation. In conclusion, our data suggest that defective autophagy in SMCs enhances atherosclerotic changes with outward arterial remodeling.
Collapse
Affiliation(s)
- Yusuke Osonoi
- a Department of Metabolism & Endocrinology , Juntendo University Graduate School of Medicine , 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421 , Japan.,b Center for Identification of Diabetic Therapeutic Targets , Juntendo University Graduate School of Medicine , 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421 , Japan
| | - Tomoya Mita
- a Department of Metabolism & Endocrinology , Juntendo University Graduate School of Medicine , 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421 , Japan.,b Center for Identification of Diabetic Therapeutic Targets , Juntendo University Graduate School of Medicine , 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421 , Japan.,c Center for Therapeutic Innovations in Diabetes , Juntendo University Graduate School of Medicine , 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421 , Japan
| | - Kosuke Azuma
- a Department of Metabolism & Endocrinology , Juntendo University Graduate School of Medicine , 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421 , Japan
| | - Kenichi Nakajima
- a Department of Metabolism & Endocrinology , Juntendo University Graduate School of Medicine , 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421 , Japan
| | - Atsushi Masuyama
- a Department of Metabolism & Endocrinology , Juntendo University Graduate School of Medicine , 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421 , Japan
| | - Hiromasa Goto
- a Department of Metabolism & Endocrinology , Juntendo University Graduate School of Medicine , 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421 , Japan
| | - Yuya Nishida
- a Department of Metabolism & Endocrinology , Juntendo University Graduate School of Medicine , 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421 , Japan
| | - Takeshi Miyatsuka
- a Department of Metabolism & Endocrinology , Juntendo University Graduate School of Medicine , 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421 , Japan.,c Center for Therapeutic Innovations in Diabetes , Juntendo University Graduate School of Medicine , 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421 , Japan
| | - Yoshio Fujitani
- a Department of Metabolism & Endocrinology , Juntendo University Graduate School of Medicine , 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421 , Japan.,d Laboratory of Developmental Biology and Metabolism, Institute for Molecular and Cellular Regulation , Gunma University , Maebashi , Japan
| | - Masato Koike
- e Department of Cell Biology and Neuroscience , Juntendo University Graduate School of Medicine , Tokyo , Japan
| | - Masako Mitsumata
- f Division of Cardiology, Department of Medicine , Nihon University School of Medicine , Tokyo , Japan
| | - Hirotaka Watada
- a Department of Metabolism & Endocrinology , Juntendo University Graduate School of Medicine , 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421 , Japan.,b Center for Identification of Diabetic Therapeutic Targets , Juntendo University Graduate School of Medicine , 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421 , Japan.,c Center for Therapeutic Innovations in Diabetes , Juntendo University Graduate School of Medicine , 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421 , Japan
| |
Collapse
|
10
|
Signorelli F, Sela S, Gesualdo L, Chevrel S, Tollet F, Pailler-Mattei C, Tacconi L, Turjman F, Vacca A, Schul DB. Hemodynamic Stress, Inflammation, and Intracranial Aneurysm Development and Rupture: A Systematic Review. World Neurosurg 2018; 115:234-244. [DOI: 10.1016/j.wneu.2018.04.143] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2018] [Revised: 04/18/2018] [Accepted: 04/19/2018] [Indexed: 10/17/2022]
|
11
|
The Impact of Uremic Toxins on Vascular Smooth Muscle Cell Function. Toxins (Basel) 2018; 10:toxins10060218. [PMID: 29844272 PMCID: PMC6024314 DOI: 10.3390/toxins10060218] [Citation(s) in RCA: 65] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Revised: 05/26/2018] [Accepted: 05/27/2018] [Indexed: 12/18/2022] Open
Abstract
Chronic kidney disease (CKD) is associated with profound vascular remodeling, which accelerates the progression of cardiovascular disease. This remodeling is characterized by intimal hyperplasia, accelerated atherosclerosis, excessive vascular calcification, and vascular stiffness. Vascular smooth muscle cell (VSMC) dysfunction has a key role in the remodeling process. Under uremic conditions, VSMCs can switch from a contractile phenotype to a synthetic phenotype, and undergo abnormal proliferation, migration, senescence, apoptosis, and calcification. A growing body of data from experiments in vitro and animal models suggests that uremic toxins (such as inorganic phosphate, indoxyl sulfate and advanced-glycation end products) may directly impact the VSMCs’ physiological functions. Chronic, low-grade inflammation and oxidative stress—hallmarks of CKD—are also strong inducers of VSMC dysfunction. Here, we review current knowledge about the impact of uremic toxins on VSMC function in CKD, and the consequences for pathological vascular remodeling.
Collapse
|
12
|
Wu S, Wang J, Li F. Dysregulation of PTEN caused by the underexpression of microRNA‑130b is associated with the severity of lupus nephritis. Mol Med Rep 2018; 17:7966-7972. [PMID: 29620214 DOI: 10.3892/mmr.2018.8839] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2016] [Accepted: 08/31/2017] [Indexed: 11/05/2022] Open
Abstract
There are several reports in the literature regarding microRNA (miR)‑130b. It has been reported that miR‑130b is involved in several diseases. The present study aimed to understand the association between the levels of miR‑130b and lupus nephritis in patients. A total of 61 blood samples were collected and the expression level of miR‑130b was determined. The online miRNA database was then searched using the 'seed sequence' located within the 3'‑untranslated region of the target gene. Linear analysis and a luciferase assay were performed to understand the regulatory association between miR‑130b and phosphatase and tensin homolog (PTEN). In addition, reverse transcription‑polymerase chain reaction and western blot analyses were performed to examine the mRNA and protein expression levels of PTEN among individuals with lupus nephritis (n=28) and those without lupus nephritis (n=31), and in mesangial cells treated with scramble control, miR‑130b mimics, PTEN small interfering (si)RNA and miR‑130b inhibitors. In addition mesangial cells were treated with scramble control, miR‑130b mimics, PTEN siRNA and miR‑130b inhibitors to investigate the affect of miR‑130b and PTEN on the viability and apoptosis of mesangial cells. The results demonstrated that miR‑130b was downregulated in the hormone‑resistant group of lupus nephritis patients. PTEN was a virtual target of miR‑130b. There was a negative regulatory association between miR‑130b and PTEN. The mRNA and protein expression levels of PTEN were increased in the hormone‑resistant group. miR‑130b decreased the expression of PTEN. miR‑130b negatively interfered with the viability of mesangial cells and PTEN positively interfered with the viability of mesangial cells. miR‑130b accelerated apoptosis and PTEN inhibited apoptosis. Taken together, the results showed that miR‑130b was upregulated in the lupus nephritis group. PTEN was a virtual target of miR‑130b, and there was a negative regulatory association between miR‑130b and PTEN. miR‑130b and PTEN interfered with the viability and apoptosis of the mesangial cells.
Collapse
Affiliation(s)
- Shupeng Wu
- Department of Rheumatism and Immunology, Tai'an Central Hospital, Tai'an, Shandong 271000, P.R. China
| | - Jing Wang
- Department of Geriatric Diseases, Tai'an Central Hospital, Tai'an, Shandong 271000, P.R. China
| | - Fang Li
- Department of Rheumatism and Immunology, Tai'an Central Hospital, Tai'an, Shandong 271000, P.R. China
| |
Collapse
|
13
|
Adly AA, Ismail EA, Andrawes NG, Mahmoud MM, Eladawy R. Soluble Fas/FasL ratio as a marker of vasculopathy in children and adolescents with sickle cell disease. Cytokine 2016; 79:52-8. [PMID: 26765484 DOI: 10.1016/j.cyto.2015.12.022] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2015] [Revised: 12/12/2015] [Accepted: 12/29/2015] [Indexed: 01/19/2023]
Abstract
OBJECTIVES Sickle cell disease (SCD) is characterized by chronic inflammation due to ischemic tissue damage, accentuated during acute complications. Fas and its ligand (FasL) are members of tumor necrosis factor receptor superfamily and a major pathway for induction of apoptosis. Fas/FasL interactions may be related to augmentation of inflammatory response. We assessed the levels of sFas and sFasL in 35 children and adolescents with SCD compared with 35 healthy controls in relation to hemolysis, iron overload, sickle vasculopathy including kidney disease. METHODS SCD patients, in steady state and asymptomatic for pulmonary hypertension, were studied stressing on hydroxyurea therapy, serum ferritin, urinary albumin creatinine ratio (UACR), high-sensitivity C-reactive protein (hs-CRP) and sFas/sFasL levels. RESULTS sFas/sFasL ratio was significantly higher in patients compared with controls. sFas/sFasL ratio was elevated in patients with pulmonary hypertension, nephropathy and those who had history of frequent sickling crisis or serum ferritin ⩾2500. SCD patients treated with hydroxyurea had lower sFas/sFasL ratio than untreated patients. sFas/sFasL ratio was positively correlated to transfusion index, white blood cells, hs-CRP, serum ferritin and UACR. The cutoff value of sFas/sFasL at 8.75pg/mL could differentiate SCD patients with and without nephropathy while the cutoff value at 22pg/mL could differentiate SCD patients with and without pulmonary hypertension risk with high sensitivity and specificity. CONCLUSION sFas/sFasL ratio may be considered as a marker for vascular dysfunction in SCD patients and is related to inflammation, iron overload and albuminuria level. Thus, it may be a reliable method to assess renal impairment in SCD.
Collapse
Affiliation(s)
- Amira A Adly
- Department of Pediatric, Faculty of Medicine, Ain Shams University, Egypt
| | - Eman A Ismail
- Department of Clinical Pathology, Faculty of Medicine, Ain Shams University, Egypt.
| | - Nevine G Andrawes
- Department of Pediatric, Faculty of Medicine, Ain Shams University, Egypt
| | - Mai M Mahmoud
- Department of Pediatric, Faculty of Medicine, Ain Shams University, Egypt
| | - Rasha Eladawy
- Department of Pediatric, Faculty of Medicine, Ain Shams University, Egypt
| |
Collapse
|
14
|
Mill C, Monk BA, Williams H, Simmonds SJ, Jeremy JY, Johnson JL, George SJ. Wnt5a-induced Wnt1-inducible secreted protein-1 suppresses vascular smooth muscle cell apoptosis induced by oxidative stress. Arterioscler Thromb Vasc Biol 2014; 34:2449-56. [PMID: 25212236 DOI: 10.1161/atvbaha.114.303922] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE Apoptosis of vascular smooth muscle cells (VSMCs) contributes to thinning and rupture of the atherosclerotic plaque fibrous cap and is thereby associated with myocardial infarction. Wnt protein activation of β-catenin regulates numerous genes that are associated with cell survival. We therefore investigated Wnt/β-catenin survival signaling in VSMCs and assessed the presence of this pathway in human atherosclerotic plaques at various stages of the disease process. APPROACH AND RESULTS Wnt5a induced β-catenin/T-cell factor signaling and retarded oxidative stress (H₂O₂)-induced apoptosis in mouse aortic VSMCs. Quantification of mRNA levels revealed a >4-fold (P<0.05; n=9) increase in the expression of the Wnt/β-catenin responsive gene, Wnt1-inducible secreted protein-1 (WISP-1), which was dependent on cAMP response element-binding protein and sustained in the presence of H₂O₂. Exogenous WISP-1 significantly reduced H₂O₂-induced apoptosis by 43% (P<0.05; n=3) and was shown using silencing small interfering RNA, to be important for Wnt5a-dependent survival responses to H₂O₂ (P<0.05; n=3). WISP-1 protein levels were significantly lower (≈50%) in unstable atherosclerosis compared with stable plaques (n=11 and n=14). CONCLUSIONS These results indicate for the first time that Wnt5a induces β-catenin survival signaling in VSMCs via WISP-1. The deficiency of the novel survival factor, WISP-1 in intimal VSMCs of unstable coronary plaques, suggests that there is altered Wnt/β-catenin/ T-cell factor signaling with progressive atherosclerosis, and restoration of WISP-1 protein might be an effective stabilization factor for vulnerable atherosclerotic plaques.
Collapse
Affiliation(s)
- Carina Mill
- From the School of Clinical Sciences, University of Bristol, Bristol Royal Infirmary, Bristol, United Kingdom
| | - Bethan Alice Monk
- From the School of Clinical Sciences, University of Bristol, Bristol Royal Infirmary, Bristol, United Kingdom
| | - Helen Williams
- From the School of Clinical Sciences, University of Bristol, Bristol Royal Infirmary, Bristol, United Kingdom
| | - Steven John Simmonds
- From the School of Clinical Sciences, University of Bristol, Bristol Royal Infirmary, Bristol, United Kingdom
| | - Jamie Yancey Jeremy
- From the School of Clinical Sciences, University of Bristol, Bristol Royal Infirmary, Bristol, United Kingdom
| | - Jason Lee Johnson
- From the School of Clinical Sciences, University of Bristol, Bristol Royal Infirmary, Bristol, United Kingdom
| | - Sarah Jane George
- From the School of Clinical Sciences, University of Bristol, Bristol Royal Infirmary, Bristol, United Kingdom.
| |
Collapse
|
15
|
Musumeci G, Coleman R, Imbesi R, Magro G, Parenti R, Szychlinska MA, Scuderi R, Cinà CS, Castorina S, Castrogiovanni P. ADAM-10 could mediate cleavage of N-cadherin promoting apoptosis in human atherosclerotic lesions leading to vulnerable plaque: a morphological and immunohistochemical study. Acta Histochem 2014; 116:1148-58. [PMID: 24985126 DOI: 10.1016/j.acthis.2014.06.002] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2014] [Revised: 06/01/2014] [Accepted: 06/02/2014] [Indexed: 01/03/2023]
Abstract
Atherosclerosis remains a major cause of mortality. Whereas the histopathological progression of atherosclerotic lesions is well documented, much less is known about the development of unstable or vulnerable plaque, which can rupture leading to thrombus, luminal occlusion and infarct. Apoptosis in the fibrous cap, which is rich in vascular smooth muscle cells (VSMCs) and macrophages, and its subsequent weakening or erosion seems to be an important regulator of plaque stability. The aim of our study was to improve our knowledge on the biological mechanisms that cause plaque instability in order to develop new therapies to maintain atherosclerotic plaque stability and avoid its rupture. In our study, we collected surgical specimens from atherosclerotic plaques in the right or left internal carotid artery of 62 patients with evident clinical symptoms. Histopathology and histochemistry were performed on wax-embedded sections. Immunohistochemical localization of caspase-3, N-cadherin and ADAM-10 was undertaken in order to highlight links between apoptosis, as expressed by caspase-3 immunostaining, and possible roles of N-cadherin, a cell-cell junction protein in VSMCs and macrophages that provides a pro-survival signal reducing apoptosis, and ADAM-10, a "disintegrin and metalloproteases" that is able to cleave N-cadherin in glioblastomas. Our results showed that when apoptosis, expressed by caspase-3 immunostaining, increased in the fibrous cap, rich in VSMCs and macrophages, the expression of N-cadherin decreased. The decreased N-cadherin expression, in turn, was linked to increased ADAM-10 expression. This study shows that apoptotic events are probably involved in the vulnerability of atherosclerotic plaque.
Collapse
|
16
|
Woollard KJ, Lumsden NG, Andrews KL, Aprico A, Harris E, Irvine JC, Jefferis AM, Fang L, Kanellakis P, Bobik A, Chin-Dusting JPF. Raised soluble P-selectin moderately accelerates atherosclerotic plaque progression. PLoS One 2014; 9:e97422. [PMID: 24846287 PMCID: PMC4028245 DOI: 10.1371/journal.pone.0097422] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2014] [Accepted: 04/17/2014] [Indexed: 01/13/2023] Open
Abstract
Soluble P-selectin (sP-selectin), a biomarker of inflammatory related pathologies including cardiovascular and peripheral vascular diseases, also has pro-atherosclerotic effects including the ability to increase leukocyte recruitment and modulate thrombotic responses in vivo. The current study explores its role in progressing atherosclerotic plaque disease. Apoe−/− mice placed on a high fat diet (HFD) were given daily injections of recombinant dimeric murine P-selectin (22.5 µg/kg/day) for 8 or 16 weeks. Saline or sE-selectin injections were used as negative controls. In order to assess the role of sP-selectin on atherothrombosis an experimental plaque remodelling murine model, with sm22α-hDTR Apoe−/− mice on a HFD in conjunction with delivery of diphtheria toxin to induce targeted vascular smooth muscle apoptosis, was used. These mice were similarly given daily injections of sP-selectin for 8 or 16 weeks. While plaque mass and aortic lipid content did not change with sP-selectin treatment in Apoe−/− or SM22α-hDTR Apoe−/− mice on HFD, increased plasma MCP-1 and a higher plaque CD45 content in Apoe−/− HFD mice was observed. As well, a significant shift towards a more unstable plaque phenotype in the SM22α-hDTR Apoe−/− HFD mice, with increased macrophage accumulation and lower collagen content, leading to a lower plaque stability index, was observed. These results demonstrate that chronically raised sP-selectin favours progression of an unstable atherosclerotic plaque phenotype.
Collapse
Affiliation(s)
- Kevin J. Woollard
- Baker IDI Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | | | - Karen L. Andrews
- Baker IDI Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | - Andrea Aprico
- Baker IDI Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | - Emma Harris
- Baker IDI Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | | | | | - Lu Fang
- Baker IDI Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | - Peter Kanellakis
- Baker IDI Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | - Alex Bobik
- Baker IDI Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | | |
Collapse
|
17
|
Lyon CA, Johnson JL, White S, Sala-Newby GB, George SJ. EC4, a truncation of soluble N-cadherin, reduces vascular smooth muscle cell apoptosis and markers of atherosclerotic plaque instability. Mol Ther Methods Clin Dev 2014; 1:14004. [PMID: 26015951 PMCID: PMC4362368 DOI: 10.1038/mtm.2014.4] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2013] [Accepted: 01/05/2014] [Indexed: 11/21/2022]
Abstract
Atherosclerotic plaque instability is precipitated by vascular smooth muscle cell apoptosis in the fibrous cap, weakening it and leading to plaque rupture. We previously showed that reducing smooth muscle cell apoptosis with soluble N-cadherin (SNC) increased features of plaque stability. We have now identified the active site of SNC and examined whether a truncated form containing this site retains the antiapoptotic effect. SNC was mutated to prevent interaction with N-cadherin or fibroblast growth factor receptor (FGFR). Interaction with FGFR in the extracellular (EC) 4 domain of SNC was essential for the antiapoptotic effect. Therefore, we made a truncated form consisting of the EC4 domain. EC4 significantly reduced smooth muscle cell, macrophage, and endothelial cell apoptosis in vitro by ~70%, similar to SNC. Elevation of plasma levels of EC4 in male apolipoprotein E-deficient mice with existing atherosclerosis significantly reduced apoptosis in brachiocephalic artery plaques by ~50%. EC4 reduced plaque size and the incidence of buried fibrous layers and the macrophage:smooth muscle cell ratio (surrogate markers of plaque instability). Interaction of EC4 with FGFR induced potent antiapoptotic signaling in vitro and in vivo. EC4 modulates atherosclerosis in mice demonstrating its therapeutic potential for retarding plaque size and instability.
Collapse
Affiliation(s)
- Cressida A Lyon
- Bristol Heart Institute, Bristol Royal Infirmary, Bristol, UK
| | - Jason L Johnson
- Bristol Heart Institute, Bristol Royal Infirmary, Bristol, UK
| | - Stephen White
- Bristol Heart Institute, Bristol Royal Infirmary, Bristol, UK
| | | | - Sarah J George
- Bristol Heart Institute, Bristol Royal Infirmary, Bristol, UK
| |
Collapse
|
18
|
Bossaller L, Rathinam VAK, Bonegio R, Chiang PI, Busto P, Wespiser AR, Caffrey DR, Li QZ, Mohan C, Fitzgerald KA, Latz E, Marshak-Rothstein A. Overexpression of membrane-bound fas ligand (CD95L) exacerbates autoimmune disease and renal pathology in pristane-induced lupus. THE JOURNAL OF IMMUNOLOGY 2013; 191:2104-14. [PMID: 23918976 DOI: 10.4049/jimmunol.1300341] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Loss-of-function mutations in the Fas death receptor or its ligand result in a lymphoproliferative syndrome and exacerbate clinical disease in most lupus-prone strains of mice. One exception is mice injected with 2,6,10,14-tetramethylpentadecane (TMPD), a hydrocarbon oil commonly known as pristane, which induces systemic lupus erythematosus-like disease. Although Fas/Fas ligand (FasL) interactions have been strongly implicated in the activation-induced cell death of both lymphocytes and other APCs, FasL can also trigger the production of proinflammatory cytokines. FasL is a transmembrane protein with a matrix metalloproteinase cleavage site in the ectodomain. Matrix metalloproteinase cleavage inactivates membrane-bound FasL and releases a soluble form reported to have both antagonist and agonist activity. To better understand the impact of FasL cleavage on both the proapoptotic and proinflammatory activity of FasL, its cleavage site was deleted through targeted mutation to produce the deleted cleavage site (ΔCS) mouse line. ΔCS mice express higher levels of membrane-bound FasL than do wild-type mice and fail to release soluble FasL. To determine to what extent FasL promotes inflammation in lupus mice, TMPD-injected FasL-deficient and ΔCS BALB/c mice were compared with control TMPD-injected BALB/c mice. We found that FasL deficiency significantly reduced the early inflammatory exudate induced by TMPD injection. In contrast, ΔCS mice developed a markedly exacerbated disease profile associated with a higher frequency of splenic neutrophils and macrophages, a profound change in anti-nuclear Ab specificity, and markedly increased proteinuria and kidney pathology compared with controls. These results demonstrate that FasL promotes inflammation in TMPD-induced autoimmunity, and its cleavage limits FasL proinflammatory activity.
Collapse
Affiliation(s)
- Lukas Bossaller
- Division of Infectious Diseases and Immunology, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Atherosclerosis and interferon-γ: new insights and therapeutic targets. Trends Cardiovasc Med 2013; 24:45-51. [PMID: 23916809 DOI: 10.1016/j.tcm.2013.06.003] [Citation(s) in RCA: 110] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2013] [Revised: 06/13/2013] [Accepted: 06/14/2013] [Indexed: 11/23/2022]
Abstract
Atherosclerosis is considered to be a chronic inflammatory disease of the arterial wall. Atherogenesis is accompanied by local production and release of inflammatory mediators, for which the macrophage is a major source. The proinflammatory cytokine, interferon (IFN)-γ derived from T cells, is expressed at high levels in atherosclerotic lesions. IFN-γ is the classic macrophage-activating factor, vital for both innate and adaptive immunity. It primes macrophages to produce chemokines and cytotoxic molecules and induces expression of genes that regulate lipid uptake. IFN-γ is a key trigger for the formation and release of reactive oxygen species. IFN-γ has important effects on endothelial cells, promoting expression of adhesion molecules. Atherogenic effects of IFN-γ have been shown in murine models where exogenous administration enhances atherosclerotic lesion formation while knockout of IFN-γ or its receptor reduces lesion size. IFN-γ signaling is largely mediated by a Janus kinase (JAK) to signal transduction and activator of transcription (STAT)1 cytosolic factor pathway. A clear understanding of IFN-γ effects on atherogenesis should enable development of novel targeted interventions for clinical use in the prevention and treatment of atherosclerosis. This review will discuss the actions of the cytokine IFN-γ and its complex effects on cells involved in atherosclerosis.
Collapse
|
20
|
Cullen SP, Henry CM, Kearney CJ, Logue SE, Feoktistova M, Tynan GA, Lavelle EC, Leverkus M, Martin SJ. Fas/CD95-induced chemokines can serve as "find-me" signals for apoptotic cells. Mol Cell 2013; 49:1034-48. [PMID: 23434371 DOI: 10.1016/j.molcel.2013.01.025] [Citation(s) in RCA: 260] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2012] [Revised: 12/14/2012] [Accepted: 01/15/2013] [Indexed: 11/29/2022]
Abstract
Apoptosis is commonly thought to represent an immunologically silent or even anti-inflammatory mode of cell death, resulting in cell clearance in the absence of explicit activation of the immune system. However, here we show that Fas/CD95-induced apoptosis is associated with the production of an array of cytokines and chemokines, including IL-6, IL-8, CXCL1, MCP-1, and GMCSF. Fas-induced production of MCP-1 and IL-8 promoted chemotaxis of phagocytes toward apoptotic cells, suggesting that these factors serve as "find-me" signals in this context. We also show that RIPK1 and IAPs are required for optimal production of cytokines and chemokines in response to Fas receptor stimulation. Consequently, a synthetic IAP antagonist potently suppressed Fas-dependent expression of multiple proinflammatory mediators and inhibited Fas-induced chemotaxis. Thus, in addition to provoking apoptosis, Fas receptor stimulation can trigger the secretion of chemotactic factors and other immunologically active proteins that can influence immune responsiveness toward dying cells.
Collapse
Affiliation(s)
- Sean P Cullen
- Molecular Cell Biology Laboratory, Department of Genetics, The Smurfit Institute, Trinity College, Dublin 2, Ireland
| | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Dounousi E, Koliousi E, Papagianni A, Ioannou K, Zikou X, Katopodis K, Kelesidis A, Tsakiris D, Siamopoulos KC. Mononuclear leukocyte apoptosis and inflammatory markers in patients with chronic kidney disease. Am J Nephrol 2012; 36:531-6. [PMID: 23258075 DOI: 10.1159/000345352] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2012] [Accepted: 10/19/2012] [Indexed: 01/30/2023]
Abstract
BACKGROUND/AIM Increased apoptosis along with enhanced inflammation has been reported in hemodialysis and pre-dialysis patients. However, there is limited information at which stage during the progression of chronic kidney disease (CKD) the balance between pro- and anti-apoptotic mechanisms is disturbed and inflammatory response is activated. The aim of this study was to investigate possible alterations in apoptotic and inflammatory markers during CKD (stages 1-4) progression and the probable interactions between them. METHODS In a cross-sectional study, 152 steady-state CKD outpatients (83 males, 55%) with mean estimated glomerular filtration rate 46 (29-76) ml/min/1.73 m(2) were studied. Apoptosis was assessed in peripheral blood mononuclear cells by estimating Bcl-2 expression, annexin V-propidium iodine staining and serum soluble Fas (sFas) and Fas-ligand. Serum levels of C-reactive protein, tumor necrosis factor-α (TNF-α), interleukin-6 and plasma levels of fibrinogen were measured as markers of inflammation. RESULTS Bcl-2 expression was found to decrease significantly in both lymphocytes and monocytes from CKD stage 1 to 4. In contrast, the activity of sFas increased significantly and so did the levels of TNF-α and fibrinogen. The majority of these alterations occurred as soon as patients entered stage 3 of CKD. A multivariate regression analysis demonstrated that CKD remained a significant predictor of the aggregate of the assessed markers. CONCLUSIONS Apoptosis appeared to increase across CKD stages 1-4, and this was associated with increased proinflammatory activity.
Collapse
Affiliation(s)
- Evangelia Dounousi
- Department of Nephrology of University Hospital of Ioannina, Ioannina, Greece
| | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Rapold RA, Wueest S, Knoepfel A, Schoenle EJ, Konrad D. Fas activates lipolysis in a Ca2+-CaMKII-dependent manner in 3T3-L1 adipocytes. J Lipid Res 2012; 54:63-70. [PMID: 23089915 DOI: 10.1194/jlr.m028035] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Fas (CD95) is a member of the tumor necrosis factor (TNF) receptor superfamily and plays a crucial role in the induction of apoptosis. However, like TNF, Fas can induce nonapoptotic signaling pathways. We previously demonstrated that mice lacking Fas specifically in adipocytes are partly protected from diet-induced insulin resistance, potentially via decreased delivery of FAs to the liver, as manifested by lower total liver ceramide content. In the present study, we aimed to delineate the signaling pathway involved in Fas-mediated adipocyte lipid mobilization. Treatment of differentiated 3T3-L1 adipocytes with membrane-bound Fas ligand (FasL) significantly increased lipolysis after 12 h without inducing apoptosis. In parallel, Fas activation increased phosphorylation of ERK1/2, and FasL-induced lipolysis was blunted in the presence of the ERK-inhibitor U0126 or in ERK1/2-depleted adipocytes. Furthermore, Fas activation increased phosphorylation of the Ca(2+)/calmodulin-dependent protein kinases II (CaMKII), and blocking of the CaMKII-pathway (either by the Ca(2+) chelator BAPTA or by the CaMKII inhibitor KN62) blunted FasL-induced ERK1/2 phosphorylation and glycerol release. In conclusion, we propose a novel role for CaMKII in promoting lipolysis in adipocytes.
Collapse
Affiliation(s)
- Reto A Rapold
- Division of Pediatric Endocrinology and Diabetology, University Children's Hospital, CH-8032 Zurich, Switzerland
| | | | | | | | | |
Collapse
|
23
|
Sgarra L, Addabbo F, Potenza MA, Montagnani M. Determinants of evolving metabolic and cardiovascular benefit/risk profiles of rosiglitazone therapy during the natural history of diabetes: molecular mechanisms in the context of integrated pathophysiology. Am J Physiol Endocrinol Metab 2012; 302:E1171-82. [PMID: 22374753 DOI: 10.1152/ajpendo.00038.2012] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Rosiglitazone is a thiazolidinedione, a synthetic PPARγ receptor agonist with insulin-sensitizing properties that is used as an antidiabetic drug. In addition to improving glycemic control through actions in metabolic target tissues, rosiglitazone has numerous biological actions that impact on cardiovascular homeostasis. Some of these actions are helpful (e.g., improving endothelial function), whereas others are potentially harmful (e.g., promoting fluid retention). Since cardiovascular morbidity and mortality are major endpoints for diabetes, it is essential to understand how the natural history of diabetes alters the net benefits and risks of rosiglitazone therapy. This complex issue is an important determinant of optimal use of rosiglitazone and is critical for understanding cardiovascular safety issues. We give special attention to the effects of rosiglitazone in diabetic patients with stable coronary artery disease and the impact of rosiglitazone actions on atherosclerosis and plaque instability. This provides a rational conceptual framework for predicting evolving benefit/risk profiles that inform optimal use of rosiglitazone in the clinical setting and help explain the results of recent large clinical intervention trials where rosiglitazone had disappointing cardiovascular outcomes. Thus, in this perspective, we describe what is known about the molecular mechanisms of action of rosiglitazone on cardiovascular targets in the context of the evolving pathophysiology of diabetes over its natural history.
Collapse
Affiliation(s)
- Luca Sgarra
- Department of Biomedical Sciences and Human Oncology, Medical School, University of Bari, Bari, Italy
| | | | | | | |
Collapse
|
24
|
MFG-E8 released by apoptotic endothelial cells triggers anti-inflammatory macrophage reprogramming. PLoS One 2012; 7:e36368. [PMID: 22558449 PMCID: PMC3340380 DOI: 10.1371/journal.pone.0036368] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2012] [Accepted: 04/05/2012] [Indexed: 01/08/2023] Open
Abstract
Apoptotic endothelial cells are an important component of the “response to injury” process. Several atherosclerosis risk factors such as hyperglycemia and oxidized low-density lipoproteins, and immune injuries, such as antibodies and complement, induce endothelial cell apoptosis. While endothelial cell apoptosis is known to affect neighboring vascular wall cell biology, its consequences on macrophage reprogramming are ill defined. In this study, we report that apoptosis of human and mouse endothelial cells triggers the release of milk fat globule-epidermal growth factor 8 (MFG-E8) and reprograms macrophages into an anti-inflammatory cells. We demonstrated that MFG-E8 is released by apoptotic endothelial cells in a caspase-3-dependent manner. When macrophages were exposed to conditioned media from serum-starved apoptotic endothelial cells, they adopt a high anti-inflammatory, low pro-inflammatory cytokine/chemokine secreting phenotype that is lost if MFG-E8 is absent from the media. Macrophage treatment with recombinant MFG-E8 recapitulates the effect of conditioned media. Finally, we showed that MFG-E8-mediated reprogramming of macrophages occurs through increased phosphorylation of signal transducer and activator of transcription-3 (STAT-3). Taken together, our study suggests a key role of MFG-E8 release from apoptotic endothelial cells in macrophage reprogramming and demonstrates the importance of the apoptotic microenvironment in anti-inflammatory macrophage responses.
Collapse
|
25
|
Bennett M, Yu H, Clarke M. Signalling from dead cells drives inflammation and vessel remodelling. Vascul Pharmacol 2012; 56:187-92. [PMID: 22306421 DOI: 10.1016/j.vph.2012.01.006] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2011] [Revised: 01/17/2012] [Accepted: 01/21/2012] [Indexed: 11/16/2022]
Abstract
Death of vascular smooth muscle cells (VSMCs) has been demonstrated in vessel development and in disease, most notably in atherosclerosis, but also after injury and remodelling. VSMC death promotes multiple features of vulnerable plaques, but also induces features of normal vessel ageing and cystic medial necrosis, including loss of VSMCs, elastin fragmentation and loss, increased glycosaminoglycans and speckled calcification. VSMC apoptosis in the absence of efficient phagocytosis also produces inflammation due to secondary necrosis; in contrast, VSMC apoptosis in normal vessels can be silent. We have investigated the consequences of VSMC apoptosis in both disease and during vessel remodelling. We find that VSMCs release specific cytokines dependent upon the mode of cell death; IL-1β predominates during apoptosis, whilst IL-1α predominates during necrosis. Both IL-1α and β promote release of further cytokines from adjacent live cells, in particular IL-6 and MCP-1. The balance of cytokines results in pathology with differing compositions, including inflammation or neointima formation/vascular repair, via direct promotion of VSMC proliferation and migration. Thus, VSMC death can promote either pathology or repair, depending upon the context and cytokine signalling.
Collapse
Affiliation(s)
- Martin Bennett
- Division of Cardiovascular Medicine, University of Cambridge, Addenbrooke's Centre for Clinical Investigation, Addenbrooke's Hospital, Cambridge, United Kingdom.
| | | | | |
Collapse
|
26
|
Zheng Y, Gardner SE, Clarke MCH. Cell death, damage-associated molecular patterns, and sterile inflammation in cardiovascular disease. Arterioscler Thromb Vasc Biol 2012; 31:2781-6. [PMID: 22096097 DOI: 10.1161/atvbaha.111.224907] [Citation(s) in RCA: 135] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Cell death and inflammation are ancient processes of fundamental biological importance in both normal physiology and pathology. This is evidenced by the profound conservation of mediators, with ancestral homologues identified from plants to humans, and the number of diseases driven by aberrant control of either process. Apoptosis is the most well-studied cell death, but many forms exist, including autophagy, necrosis, pyroptosis, paraptosis, and the obscure dark cell death. Cell death occurs throughout the cardiovascular system, from initial shaping of the heart and vasculature during development to involvement in pathologies, including atherosclerosis, aneurysm, cardiomyopathy, restenosis, and vascular graft rejection. However, determining whether cell death primarily drives pathology or is a secondary bystander effect is difficult. Inflammation, the primary response of innate immunity, is considered essential in initiating and driving vascular diseases. Cell death and inflammation are inextricably linked with their effectors modulating the other process. Indeed, an evolutionary link between cell death and inflammation occurs at caspase-1 (which activates interleukin-1β), which can induce death by pyroptosis, and is a member of the caspase family vital for apoptosis. This review examines cell death in vascular disease, how it can induce inflammation, and finally the emergence of inflammasomes in vascular pathology.
Collapse
Affiliation(s)
- Yue Zheng
- Division of Cardiovascular Medicine, University of Cambridge, Box 110, ACCI, Addenbrooke's Hospital, Cambridge CB2 2QQ, United Kingdom
| | | | | |
Collapse
|
27
|
Teoh H, Quan A, Creighton AK, Annie Bang KW, Singh KK, Shukla PC, Gupta N, Pan Y, Lovren F, Leong-Poi H, Al-Omran M, Verma S. BRCA1 gene therapy reduces systemic inflammatory response and multiple organ failure and improves survival in experimental sepsis. Gene Ther 2012; 20:51-61. [DOI: 10.1038/gt.2011.214] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
28
|
Agouni A, Ducluzeau PH, Benameur T, Faure S, Sladkova M, Duluc L, Leftheriotis G, Pechanova O, Delibegovic M, Martinez MC, Andriantsitohaina R. Microparticles from patients with metabolic syndrome induce vascular hypo-reactivity via Fas/Fas-ligand pathway in mice. PLoS One 2011; 6:e27809. [PMID: 22110764 PMCID: PMC3217000 DOI: 10.1371/journal.pone.0027809] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2011] [Accepted: 10/25/2011] [Indexed: 11/18/2022] Open
Abstract
Microparticles are membrane vesicles with pro-inflammatory properties. Circulating levels of microparticles have previously been found to be elevated in patients with metabolic syndrome (MetS). The present study aimed to evaluate the effects of in vivo treatment with microparticles, from patients with MetS and from healthy subjects (HS), on ex vivo vascular function in mice. Microparticles isolated from MetS patients or HS, or a vehicle were intravenously injected into mice, following which vascular reactivity in response to vasoconstrictor agonists was assessed by myography with respect to cyclo-oxygenase pathway, oxidative and nitrosative stress. Injection of microparticles from MetS patients into mice induced vascular hypo-reactivity in response to serotonin. Hypo-reactivity was associated with up-regulation of inducible NO-synthase and increased production of NO, and was reversed by the NO-synthase inhibitor (NG-nitro-L-arginine). The selective COX-2 inhibitor (NS398) reduced the contractile effect of serotonin in aortas from mice treated with vehicle or HS microparticles; however, this was not observed within mice treated with MetS microparticles, probably due to the ability of MetS microparticles to enhance prostacyclin. MetS microparticle-mediated vascular dysfunction was associated with increased reactive oxygen species (ROS) and enhanced expression of the NADPH oxidase subunits. Neutralization of the pro-inflammatory pathway Fas/FasL completely prevented vascular hypo-reactivity and the ability of MetS microparticles to enhance both inducible NO-synthase and monocyte chemoattractant protein-1 (MCP-1). Our data provide evidence that microparticles from MetS patients induce ex vivo vascular dysfunction by increasing both ROS and NO release and by altering cyclo-oxygenase metabolites and MCP-1 through the Fas/FasL pathway.
Collapse
Affiliation(s)
- Abdelali Agouni
- INSERM, U694, Angers, France; Université d'Angers, Angers, France
- Institute of Biological and Environmental Sciences, University of Aberdeen, Aberdeen, Scotland, United Kingdom
| | - Pierre-Henri Ducluzeau
- INSERM, U694, Angers, France; Université d'Angers, Angers, France
- Département d'Endocrinologie et Diabétologie, CHU d'Angers, Angers, France
| | - Tarek Benameur
- INSERM, U694, Angers, France; Université d'Angers, Angers, France
| | - Sébastien Faure
- INSERM, U694, Angers, France; Université d'Angers, Angers, France
| | - Martina Sladkova
- INSERM, U694, Angers, France; Université d'Angers, Angers, France
- Institute of Normal and Pathological Physiology, Slovak Academy of Sciences, Bratislava, Slovak Republic
| | - Lucie Duluc
- INSERM, U694, Angers, France; Université d'Angers, Angers, France
| | | | | | - Mirela Delibegovic
- Institute of Biological and Environmental Sciences, University of Aberdeen, Aberdeen, Scotland, United Kingdom
| | | | | |
Collapse
|
29
|
Yu H, Clarke MCH, Figg N, Littlewood TD, Bennett MR. Smooth muscle cell apoptosis promotes vessel remodeling and repair via activation of cell migration, proliferation, and collagen synthesis. Arterioscler Thromb Vasc Biol 2011; 31:2402-9. [PMID: 21885847 DOI: 10.1161/atvbaha.111.235622] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE Although vascular smooth muscle cell (VSMC) apoptosis occurs after vessel injury and during remodeling, the direct role of VSMC death in determining final vessel structure is unclear. We sought to determine the role of VSMC apoptosis in vessel remodeling, medial repair, and neointima formation and to identify the mediators involved. METHODS AND RESULTS The left common carotid artery was ligated in SM22α-human diphtheria toxin receptor mice, in which diphtheria toxin treatment selectively induces VSMC apoptosis. Apoptosis induced from day 7 to day 14 after ligation significantly increased neointimal and medial areas, cell proliferation, migration, and vessel size. Neointima formation depended on VSMCs, as VSMC depletion before ligation significantly reduced neointimal area and cellularity. In culture, conditioned media from apoptotic VSMCs promoted VSMC migration, proliferation, and collagen synthesis. Interleukin-6 (IL-6) secretion increased 5-fold and IL-1α 1.5-fold after apoptosis, whereas IL-6 inhibition negated the effect of apoptotic VSMC supernatants on VSMC migration, proliferation, and matrix synthesis. CONCLUSION Signaling from apoptotic VSMCs directly promotes vessel remodeling, medial repair, and neointima formation after flow reduction. Although lumen size appears to depend on flow, VSMC apoptosis is an important determinant of vessel, medial, and neointimal size after flow reduction.
Collapse
Affiliation(s)
- Haixiang Yu
- Division of Cardiovascular Medicine, University of Cambridge, Addenbrooke's Hospital, Cambridge, United Kingdom
| | | | | | | | | |
Collapse
|
30
|
Kenagy RD, Min SK, Mulvihill E, Clowes AW. A link between smooth muscle cell death and extracellular matrix degradation during vascular atrophy. J Vasc Surg 2011; 54:182-191.e24. [PMID: 21493032 PMCID: PMC3129478 DOI: 10.1016/j.jvs.2010.12.070] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2010] [Revised: 12/07/2010] [Accepted: 12/11/2010] [Indexed: 12/12/2022]
Abstract
OBJECTIVE High blood flow induces neointimal atrophy in polytetrafluoroethylene (PTFE) aortoiliac grafts and a tight external PTFE wrap of the iliac artery induces medial atrophy. In both nonhuman primate models, atrophy with loss of smooth muscle cells and extracellular matrix (ECM) begins at ≤4 days. We hypothesized that matrix loss would be linked to cell death, but the factors and mechanisms involved are not known. The purpose of this study was to determine commonly regulated genes in these two models, which we hypothesized would be a small set of genes that might be key regulators of vascular atrophy. METHODS DNA microarray analysis (Sentrix Human Ref 8; Illumina, San Diego, Calif; ∼23,000 genes) was performed on arterial tissue from the wrap model (n = 9) and graft neointima from the graft model (n = 5) 1 day after wrapping or the switch to high flow, respectively. Quantitative reverse-transcription polymerase chain reaction (qRT-PCR) was also performed. Expression of this vascular atrophy gene set was also studied after Fas ligand-induced cell death in cultured smooth muscle cells and organ cultured arteries. RESULTS Microarray analysis showed 15 genes were regulated in the same direction in both atrophy models: 9 upregulated and 6 downregulated. Seven of nine upregulated genes were confirmed by qRT-PCR in both models. Upregulated genes included the ECM-degrading enzymes ADAMTS4, tissue plasminogen activator (PLAT), and hyaluronidase 2; possible growth regulatory factors, including chromosome 8 open reading frame 4 and leucine-rich repeat family containing 8; a differentiation regulatory factor (musculoskeletal embryonic nuclear protein 1); a dead cell removal factor (ficolin 3); and a prostaglandin transporter (solute carrier organic anion transporter family member 2A1). Five downregulated genes were confirmed but only in one or the other model. Of the seven upregulated genes, ADAMTS4, PLAT, hyaluronidase 2, solute carrier organic anion transporter family member 2A1, leucine-rich repeat family containing 8, and chromosome 8 open reading frame 4 were also upregulated in vitro in cultured smooth muscle cells or cultured iliac artery by treatment with FasL, which causes cell death. However, blockade of caspase activity with Z-VAD inhibited FasL-mediated cell death, but not gene induction. CONCLUSION Seven gene products were upregulated in two distinctly different in vivo nonhuman primate vascular atrophy models. Induction of cell death by FasL in vitro induced six of these genes, including the ECM-degrading factors ADAMTS4, hyaluronidase 2, and PLAT, suggesting a mechanism by which the program of tissue atrophy coordinately removes extracellular matrix as cells die. These genes may be key regulators of vascular atrophy.
Collapse
MESH Headings
- Animals
- Apoptosis
- Arteriovenous Shunt, Surgical/adverse effects
- Atrophy
- Blood Vessel Prosthesis Implantation/adverse effects
- Cells, Cultured
- Disease Models, Animal
- Extracellular Matrix/metabolism
- Fas Ligand Protein/metabolism
- Femoral Artery/metabolism
- Femoral Artery/pathology
- Femoral Artery/surgery
- Femoral Vein/metabolism
- Femoral Vein/pathology
- Femoral Vein/surgery
- Gene Expression Profiling/methods
- Gene Expression Regulation
- Iliac Artery/metabolism
- Iliac Artery/pathology
- Iliac Artery/surgery
- Male
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Muscle, Smooth, Vascular/surgery
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Oligonucleotide Array Sequence Analysis
- Papio
- Postoperative Complications/etiology
- Postoperative Complications/genetics
- Postoperative Complications/metabolism
- Postoperative Complications/pathology
- Reverse Transcriptase Polymerase Chain Reaction
- Time Factors
Collapse
Affiliation(s)
- Richard D Kenagy
- Department of Surgery, University of Washington, Seattle, WA 98195-6410, USA
| | | | | | | |
Collapse
|
31
|
Farnand AW, Eastman AJ, Herrero R, Hanson JF, Mongovin S, Altemeier WA, Matute-Bello G. Fas activation in alveolar epithelial cells induces KC (CXCL1) release by a MyD88-dependent mechanism. Am J Respir Cell Mol Biol 2011; 45:650-8. [PMID: 21257927 DOI: 10.1165/rcmb.2010-0153oc] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Activation of the Fas/Fas ligand (FasL) system is associated with activation of apoptotic and proinflammatory pathways that lead to the development of acute lung injury. Previous studies in chimeric mice and macrophage-depleted mice suggested that the main effector cell in Fas-mediated lung injury is not a myeloid cell, but likely an epithelial cell. The goal of this study was to determine whether epithelial cells release proinflammatory cytokines after Fas activation, and to identify the relevant pathways. Incubation of the murine alveolar epithelial cell line, MLE-12, with the Fas-activating monoclonal antibody, Jo2, resulted in release of the CXC chemokine, KC, in a dose-dependent manner. KC release was not prevented by the pan-caspase inhibitor, zVAD.fmk. Silencing of the adaptor protein, MyD88, with small interfering (si)RNA resulted in attenuation of KC release in response to Jo2. Fas activation resulted in phosphorylation of the mitogen-activated kinases extracellular signal-regulated kinase (ERK) and c-Jun-N-terminal kinase (JNK), and pharmacologic inhibition of ERK and JNK attenuated KC release in a dose-response manner. Similarly, primary human small airways epithelial cells released IL-8 in response to soluble FasL, and this was abrogated by inhibition of JNK and ERK. In vivo confirmatory studies showed that MyD88-null mice are protected from Fas-induced acute lung injury. In summary, we conclude that Fas induces KC release in MLE-12 cells by a mechanism requiring MyD88, mitogen-activated protein kinases, and likely activator protein-1.
Collapse
Affiliation(s)
- Alex W Farnand
- Center for Lung Biology, Division of Pulmonary & Critical Care Medicine, University of Washington School of Medicine, Seattle, Washington, USA
| | | | | | | | | | | | | |
Collapse
|
32
|
Tekabe Y, Li Q, Luma J, Weisenberger D, Sedlar M, Harja E, Narula J, Johnson LL. Noninvasive monitoring the biology of atherosclerotic plaque development with radiolabeled annexin V and matrix metalloproteinase inhibitor in spontaneous atherosclerotic mice. J Nucl Cardiol 2010; 17:1073-81. [PMID: 20700679 DOI: 10.1007/s12350-010-9276-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2010] [Accepted: 07/18/2010] [Indexed: 10/19/2022]
Abstract
OBJECTIVES To compare the ability of (99m)Tc-labeled broad-based matrix metalloproteinase inhibitor (RP805) (MPI) and (99m)Tc-annexin V to identify more advanced atherosclerotic disease in apolipoprotein E-null (apoE(-/-)) mice. BACKGROUND Both MMP expression and apoptotic cell death occur in both early and in advanced atherosclerotic plaques. METHODS Eight 6-9-week-old apoE(-/-) mice, 10 apoE(-/-) mice at 20 weeks, and 12 apoE(-/-) at 40 weeks were injected with both tracers in alternating sequence separated by 48 h, underwent planar imaging and were killed. Radiotracer uptake was quantified from the scans as percent whole body and from tissue as percent injected dose per gram (%ID/g). Quantitative immunohistopathology of the aorta and carotids for macrophages, MMPs, and caspase was performed. RESULTS At 6 weeks, mice showed no tracer uptake in the chest or neck and had minimal lesion. At 20 weeks, uptake of annexin V as %ID was borderline higher than MPI (1.10 ± .48% vs .77 ± .31%, P = .09), between 20 and 40 weeks aortic lesion area increased from 37.4 ± 12.0% to 46.2 ± 7.4% and at 40 weeks MPI was significantly greater than annexin V uptake (1.11 ± .66% vs .70 ± .16%, P = .05). On histology there were greater increases in % MMP-2 and -9 than % caspase positive cells. Carotid uptake of MPI was greater than annexin V at both 20 and 40 weeks (1.25 ± .48% vs .78 ± .25%, P = .02 and 3.70 ± 1.45% vs 2.25 ± .66%, P = .005). The carotid lesion area at 40 weeks was 74 ± 9% with greater % cells positive for MMP's than caspase. %ID/g annexin V correlated significantly with % macrophages and with caspase-3 positive cells and %ID/g MPI correlated significantly with % macrophages and with MMP-2 and -9 positive cells. CONCLUSIONS In apoE(-/-) mice, MMP expression is greater than apoptosis as the disease progresses and MPI may be a better imaging agent for more advanced disease.
Collapse
Affiliation(s)
- Yared Tekabe
- Division of Cardiology, Department of Medicine, Columbia University Medical Center, New York, NY 10032, USA.
| | | | | | | | | | | | | | | |
Collapse
|
33
|
Hattori Y, Takano KI, Teramae H, Yamamoto S, Yokoo H, Matsuda N. Insights into sepsis therapeutic design based on the apoptotic death pathway. J Pharmacol Sci 2010; 114:354-65. [PMID: 21081836 DOI: 10.1254/jphs.10r04cr] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022] Open
Abstract
Sepsis remains the leading cause of death in critically ill patients. A major problem contributing to sepsis-related high mortality is the lack of effective medical treatment. Thus, the key goal in critical care medicine is to develop novel therapeutic strategies that will impact favorably on septic patient outcome. While it is generally accepted that sepsis is an inflammatory state resulting from the systemic response to infection, apoptosis is implicated to be an important mechanism of the death of lymphocytes, gastrointestinal and lung epithelial cells, and vascular endothelial cells associated with the development of multiple organ failure in sepsis. The pivotal role of cell apoptosis is now highlighted by multiple studies demonstrating that prevention of cell apoptosis can improve survival in clinically relevant animal models of sepsis. In this review article, we address the scientific rationale for remedying apoptotic cell death in sepsis and propose that therapeutic efforts aimed at blocking cell signaling pathways leading to apoptosis may represent an attractive target for sepsis therapy.
Collapse
Affiliation(s)
- Yuichi Hattori
- Department of Molecular and Medical Pharmacology, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, 2630 Sugitani, Toyama 930-0194, Japan.
| | | | | | | | | | | |
Collapse
|
34
|
Hu WT, Chen-Plotkin A, Grossman M, Arnold SE, Clark CM, Shaw LM, McCluskey L, Elman L, Hurtig HI, Siderowf A, Lee VMY, Soares H, Trojanowski JQ. Novel CSF biomarkers for frontotemporal lobar degenerations. Neurology 2010; 75:2079-86. [PMID: 21048198 DOI: 10.1212/wnl.0b013e318200d78d] [Citation(s) in RCA: 82] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
OBJECTIVE To identify antemortem CSF diagnostic biomarkers that can potentially distinguish between the 2 main causes of frontotemporal lobar degeneration (FTLD), i.e., FTLD with TDP-43 pathology (FTLD-TDP) and FTLD with tau pathology (FTLD-tau). METHODS CSF samples were collected antemortem from 23 patients with FTLD with known pathology to form a autopsy cohort as part of a comparative biomarker study that additionally included 33 living cognitively normal subjects and 66 patients with autopsy-confirmed Alzheimer disease (AD). CSF samples were also collected from 80 living patients clinically diagnosed with frontotemporal dementia (FTD). Levels of 151 novel analytes were measured via a targeted multiplex panel enriched in neuropeptides, cytokines, and growth factors, along with levels of CSF biomarkers for AD. RESULTS CSF levels of multiple analytes differed between FTLD-TDP and FTLD-tau, including Fas, neuropeptides (agouti-related peptide and adrenocorticotropic hormone), and chemokines (IL-23, IL-17). Classification by random forest analysis achieved high sensitivity for FTLD-TDP (86%) with modest specificity (78%) in the autopsy cohort. When the classification algorithm was applied to a living FTD cohort, semantic dementia was the phenotype with the highest predicted proportion of FTLD-TDP. When living patients with behavioral variant FTD were examined in detail, those predicted to have FTLD-TDP demonstrated neuropsychological differences vs those predicted to have FTLD-tau in a pattern consistent with previously reported trends in autopsy-confirmed cases. CONCLUSIONS Clinical cases with FTLD-TDP and FTLD-tau pathology can be potentially identified antemortem by assaying levels of specific analytes that are well-known and readily measurable in CSF.
Collapse
Affiliation(s)
- W T Hu
- Department of Neurology, University of Pennsylvania School of Medicine, Philadelphia 19104-4283, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Kim-Kaneyama JR, Takeda N, Sasai A, Miyazaki A, Sata M, Hirabayashi T, Shibanuma M, Yamada G, Nose K. Hic-5 deficiency enhances mechanosensitive apoptosis and modulates vascular remodeling. J Mol Cell Cardiol 2010; 50:77-86. [PMID: 20933520 DOI: 10.1016/j.yjmcc.2010.09.024] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2010] [Revised: 09/01/2010] [Accepted: 09/29/2010] [Indexed: 12/28/2022]
Abstract
Forces associated with blood flow are crucial not only for blood vessel development but also for regulation of vascular pathology. Although there have been many studies characterizing the responses to mechanical stimuli, molecular mechanisms linking biological responses to mechanical forces remain unclear. Hic-5 (hydrogen peroxide-inducible clone-5) is a focal adhesion adaptor protein proposed as a candidate for a mediator of mechanotransduction. In the present study, we generated Hic-5-deficient mice by targeted mutation. Mice lacking Hic-5 are viable and fertile, and show no obvious histological abnormalities including vasculature. However, after wire injury of the femoral artery in Hic-5 deficient mice, histological recovery of arterial media was delayed due to enhanced apoptosis of vascular wall cells, whereas neointima formation was enhanced. Stretch-induced apoptosis was enhanced in cultured vascular smooth muscle cells (vascular SMCs) from Hic-5 deficient mice. Mechanical stress also induced the alteration of intracellular distribution of vinculin from focal adhesions to the whole cytoplasm in SMCs. Immunoelectron microscopic study of vascular SMCs from a wire-injured artery demonstrated that vinculin was dispersed in the nucleus and the cytoplasm in Hic-5-deficient mice whereas vinculin was localized mainly in the sub-plasma membrane region in wild type mice. Our findings indicate that Hic-5 may serve as a key regulator in mechanosensitive vascular remodeling.
Collapse
Affiliation(s)
- Joo-ri Kim-Kaneyama
- Department of Microbiology, Showa University School of Pharmacy, Tokyo, Japan.
| | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Ahmad U, Ali R, Lebastchi AH, Qin L, Lo SFL, Yakimov AO, Khan SF, Choy JC, Geirsson A, Pober JS, Tellides G. IFN-gamma primes intact human coronary arteries and cultured coronary smooth muscle cells to double-stranded RNA- and self-RNA-induced inflammatory responses by upregulating TLR3 and melanoma differentiation-associated gene 5. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2010; 185:1283-94. [PMID: 20562257 PMCID: PMC2902797 DOI: 10.4049/jimmunol.0902283] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Atherosclerosis of native coronary arteries and graft arteriosclerosis in transplanted hearts are characterized by activation of innate and adaptive immune responses. Nucleic acids generated by infections or cell death have been detected within arteriosclerotic lesions, and it is known that microbial and synthetic nucleic acids evoke inflammatory responses in cultured vascular cells. In this study, we report that model RNA, but not DNA, instigated robust cytokine and chemokine production from intact human coronary arteries containing both intrinsic vascular cells and resident/infiltrating leukocytes. An ssRNA analog induced TNF-alpha and IFN-gamma-induced protein of 10 kDa secretion by isolated human PBMCs, but not vascular cells. Conversely, synthetic dsRNA induced these inflammatory mediators by vascular cells, but not PBMCs. IFN-gamma, a cytokine linked to atherosclerosis and graft arteriosclerosis, potentiated the inflammatory responses of intact arteries and cultured vascular smooth muscle cells (VSMCs) to polyinosinic:polycytidylic acid [poly(I:C)] and was necessary for inflammatory responses of VSMC to self-RNA derived from autologous cells. IFN-gamma also induced the expression of TLR3, melanoma differentiation-associated gene 5, and retinoic acid-inducible gene I dsRNA receptors. Small interfering RNA knockdown revealed that TLR3 mediated VSMC activation by poly(I:C), whereas melanoma differentiation-associated gene 5 was more important for VSMC stimulation by self-RNA. IFN-gamma-mediated induction of dsRNA receptors and priming for inflammatory responses to poly(I:C) was confirmed in vivo using immunodeficient mice bearing human coronary artery grafts. These findings suggest that IFN-gamma, and by inference adaptive immunity, sensitizes the vasculature to innate immune activators, such as RNA, and activation of IFN-gamma-primed vascular cells by exogenous or endogenous sources of RNA may contribute to the inflammatory milieu of arteriosclerosis.
Collapse
MESH Headings
- Animals
- Cells, Cultured
- Chemokine CXCL10/metabolism
- Coronary Vessels/drug effects
- Coronary Vessels/immunology
- Coronary Vessels/transplantation
- DEAD-box RNA Helicases/genetics
- DEAD-box RNA Helicases/metabolism
- Enzyme-Linked Immunosorbent Assay
- Female
- Humans
- Immunoblotting
- In Vitro Techniques
- Interferon-Induced Helicase, IFIH1
- Interferon-gamma/genetics
- Interferon-gamma/immunology
- Interferon-gamma/metabolism
- Mice
- Mice, SCID
- Muscle, Smooth, Vascular/cytology
- Muscle, Smooth, Vascular/immunology
- Muscle, Smooth, Vascular/metabolism
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/immunology
- Myocytes, Smooth Muscle/metabolism
- Poly I-C/pharmacology
- RNA Interference
- RNA, Double-Stranded/immunology
- RNA, Double-Stranded/metabolism
- RNA, Double-Stranded/pharmacology
- Reverse Transcriptase Polymerase Chain Reaction
- Toll-Like Receptor 2/genetics
- Toll-Like Receptor 2/metabolism
- Transplantation, Heterologous
- Tumor Necrosis Factor-alpha/metabolism
- Up-Regulation
Collapse
Affiliation(s)
- Usman Ahmad
- Department of Surgery, Interdepartmental Program in Vascular Biology and Therapeutics, Yale University School of Medicine, New Haven, Connecticut 06510
| | - Rahmat Ali
- Department of Surgery, Interdepartmental Program in Vascular Biology and Therapeutics, Yale University School of Medicine, New Haven, Connecticut 06510
| | - Amir H. Lebastchi
- Department of Surgery, Interdepartmental Program in Vascular Biology and Therapeutics, Yale University School of Medicine, New Haven, Connecticut 06510
| | - Lingfeng Qin
- Department of Surgery, Interdepartmental Program in Vascular Biology and Therapeutics, Yale University School of Medicine, New Haven, Connecticut 06510
| | - Sheng-fu L. Lo
- Department of Surgery, Interdepartmental Program in Vascular Biology and Therapeutics, Yale University School of Medicine, New Haven, Connecticut 06510
| | - Alexander O. Yakimov
- Department of Surgery, Interdepartmental Program in Vascular Biology and Therapeutics, Yale University School of Medicine, New Haven, Connecticut 06510
| | - Salman F. Khan
- Department of Surgery, Interdepartmental Program in Vascular Biology and Therapeutics, Yale University School of Medicine, New Haven, Connecticut 06510
| | - Jonathan C. Choy
- Department of Immunobiology, Interdepartmental Program in Vascular Biology and Therapeutics, Yale University School of Medicine, New Haven, Connecticut 06510
| | - Arnar Geirsson
- Department of Surgery, Interdepartmental Program in Vascular Biology and Therapeutics, Yale University School of Medicine, New Haven, Connecticut 06510
- Veterans Affairs Connecticut Healthcare System, West Haven, Connecticut 06516
| | - Jordan S. Pober
- Department of Immunobiology, Interdepartmental Program in Vascular Biology and Therapeutics, Yale University School of Medicine, New Haven, Connecticut 06510
| | - George Tellides
- Department of Surgery, Interdepartmental Program in Vascular Biology and Therapeutics, Yale University School of Medicine, New Haven, Connecticut 06510
- Veterans Affairs Connecticut Healthcare System, West Haven, Connecticut 06516
| |
Collapse
|
37
|
de Paepe ME, Haley SA, Lacourse Z, Mao Q. Effects of Fas-ligand overexpression on alveolar type II cell growth kinetics in perinatal murine lungs. Pediatr Res 2010; 68:57-62. [PMID: 20375852 PMCID: PMC2888646 DOI: 10.1203/pdr.0b013e3181e084af] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
We determined the time-specific effects of FasL overexpression on perinatal alveolar type II cell growth kinetics. To achieve temporal overexpression of respiratory epithelium-specific FasL expression, tetracycline inducible CCSP-rtTA/FasL-TetOp transgenic mice were given doxycycline (Dox) from gestational d 14 (E14) to E19 (antenatal treatment group), from postnatal d 1 (P1) to P7 (postnatal group), or from E14 to P7 (combined antenatal and postnatal group). Antenatal Dox administration induced an increase of pulmonary FasL mRNA levels in double transgenic animals up to >300-fold over single transgenic littermate controls, associated with massive fetal respiratory epithelial apoptosis and excessive postnatal lethality. Although animals from the combined antenatal/postnatal Dox treatment group continued to display evidence of increased apoptosis, there was a paradoxical increase in alveolar type II cell proliferation, resulting in a net increase in type II cell density, elevated pulmonary surfactant protein C levels and improved postnatal survival. Postnatal Dox administration was also associated with increased type II cell density, although FasL up-regulation was more variable. In conclusion, these results, and our previous studies, suggest that FasL signaling has dual timing-dependent proapoptotic and proproliferative effects on postcanalicular type II cell kinetics.
Collapse
Affiliation(s)
- Monique E de Paepe
- Department of Pathology, Women and Infants Hospital, and Department of Pathology and Laboratory Medicine, Alpert Medical School of Brown University, Providence, Rhode Island 02905, USA.
| | | | | | | |
Collapse
|
38
|
Adipocytokines in atherothrombosis: focus on platelets and vascular smooth muscle cells. Mediators Inflamm 2010; 2010:174341. [PMID: 20652043 PMCID: PMC2905911 DOI: 10.1155/2010/174341] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2009] [Revised: 03/14/2010] [Accepted: 04/29/2010] [Indexed: 12/11/2022] Open
Abstract
Visceral obesity is a relevant pathological condition closely associated with high risk of atherosclerotic vascular disease including myocardial infarction and stroke. The increased vascular risk is related also to peculiar dysfunction in the endocrine activity of adipose tissue responsible of vascular impairment (including endothelial dysfunction), prothrombotic tendency, and low-grade chronic inflammation. In particular, increased synthesis and release of different cytokines, including interleukins and tumor necrosis factor-α (TNF-α), and adipokines—such as leptin—have been reported as associated with future cardiovascular events. Since vascular cell dysfunction plays a major role in the atherothrombotic complications in central obesity, this paper aims at focusing, in particular, on the relationship between platelets and vascular smooth muscle cells, and the impaired secretory pattern of adipose tissue.
Collapse
|
39
|
FADD: a regulator of life and death. Trends Immunol 2010; 31:260-9. [PMID: 20576468 DOI: 10.1016/j.it.2010.05.005] [Citation(s) in RCA: 153] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2010] [Revised: 05/11/2010] [Accepted: 05/12/2010] [Indexed: 12/30/2022]
Abstract
FAS-associated protein with death domain (FADD) is the key adaptor protein transmitting apoptotic signals mediated by the main death receptors (DRs). Besides being an essential instrument in cell death, FADD is also implicated in proliferation, cell cycle progression, tumor development, inflammation, innate immunity, and autophagy. Recently, many of these new functions of FADD were shown to be independent of DRs. Moreover, FADD function is dictated by protein localization and phosphorylation state. Thus, FADD is a crucial and unique controller of many essential cellular processes. The full understanding of the networks dictating the ultimate function of FADD may provide a new paradigm for other multifaceted proteins.
Collapse
|
40
|
Yamanouchi D, Morgan S, Kato K, Lengfeld J, Zhang F, Liu B. Effects of caspase inhibitor on angiotensin II-induced abdominal aortic aneurysm in apolipoprotein E-deficient mice. Arterioscler Thromb Vasc Biol 2010; 30:702-7. [PMID: 20075419 DOI: 10.1161/atvbaha.109.200527] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE The presence of apoptotic markers is a prominent histological feature of abdominal aortic aneurysm. To understand the role of apoptosis in the pathogenesis of this common vascular disease, we tested the effect of the pan-caspase inhibitor quinoline-Val-Asp-difluorophenoxymethylketone (Q-VD-OPh) on aneurysm formation using a mouse angiotensin II (Ang II) model. METHODS AND RESULTS Ang II in apolipoprotein E-deficient mice significantly induced medial cell apoptosis 3 days after infusion at the aortic region, eventually becoming aneurismal. A daily administration of 20 mg/kg per day Q-VD-OPh starting 6 hours before Ang II infusion reduced aneurysm incidence from 83.3% to 16.7% and maximal aortic diameter from 2.43+/-0.29 mm to 1.58+/-0.18 mm. The caspase inhibitor treated mice showed profoundly diminished levels of medial apoptosis and inflammation. In contrast, administration of Q-VD-OPh starting 7 days after Ang II infusion had no significant impact on aneurysm development. In vitro, media conditioned by Ang II-treated smooth muscle cells (SMCs) stimulated macrophage chemotaxis in a caspase-dependent manner. Inhibition of monocyte chemoattractant protein-1 (MCP-1) in the conditioned media via a neutralizing antibody completely blocked the ability of conditioned media to attract macrophages. CONCLUSIONS These results indicate that medial SMC apoptosis may contribute to vascular inflammation and thus aneurysm formation, in part through production of MCP-1.
Collapse
Affiliation(s)
- Dai Yamanouchi
- Division of Vascular Surgery, Department of Surgery, University of Wisconsin-Madison, 1111 Highland Avenue, WIMR 5120, Madison, WI 53705, USA
| | | | | | | | | | | |
Collapse
|
41
|
Ginestier C, Liu S, Diebel ME, Korkaya H, Luo M, Brown M, Wicinski J, Cabaud O, Charafe-Jauffret E, Birnbaum D, Guan JL, Dontu G, Wicha MS. CXCR1 blockade selectively targets human breast cancer stem cells in vitro and in xenografts. J Clin Invest 2010; 120:485-97. [PMID: 20051626 DOI: 10.1172/jci39397] [Citation(s) in RCA: 591] [Impact Index Per Article: 39.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2009] [Accepted: 11/11/2009] [Indexed: 12/14/2022] Open
Abstract
Recent evidence suggests that breast cancer and other solid tumors possess a rare population of cells capable of extensive self-renewal that contribute to metastasis and treatment resistance. We report here the development of a strategy to target these breast cancer stem cells (CSCs) through blockade of the IL-8 receptor CXCR1. CXCR1 blockade using either a CXCR1-specific blocking antibody or repertaxin, a small-molecule CXCR1 inhibitor, selectively depleted the CSC population in 2 human breast cancer cell lines in vitro. Furthermore, this was followed by the induction of massive apoptosis in the bulk tumor population via FASL/FAS signaling. The effects of CXCR1 blockade on CSC viability and on FASL production were mediated by the FAK/AKT/FOXO3A pathway. In addition, repertaxin was able to specifically target the CSC population in human breast cancer xenografts, retarding tumor growth and reducing metastasis. Our data therefore suggest that CXCR1 blockade may provide a novel means of targeting and eliminating breast CSCs.
Collapse
Affiliation(s)
- Christophe Ginestier
- University of Michigan Comprehensive Cancer Center, Department of Internal Medicine/Oncology, Ann Arbor, Michigan 48109-0015, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Haider N, Hartung D, Fujimoto S, Petrov A, Kolodgie FD, Virmani R, Ohshima S, Liu H, Zhou J, Fujimoto A, Tahara A, Hofstra L, Narula N, Reutelingsperger C, Narula J. Dual molecular imaging for targeting metalloproteinase activity and apoptosis in atherosclerosis: molecular imaging facilitates understanding of pathogenesis. J Nucl Cardiol 2009; 16:753-62. [PMID: 19662466 PMCID: PMC2746297 DOI: 10.1007/s12350-009-9107-8] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2009] [Revised: 04/29/2009] [Accepted: 05/23/2009] [Indexed: 11/27/2022]
Abstract
BACKGROUND Macrophage apoptosis and MMP activity contribute to vulnerability of atherosclerotic plaques to rupture. By employing molecular imaging techniques, we investigated if apoptosis and MMP release are interlinked. METHODS Atherosclerosis was produced in rabbits receiving high-cholesterol diet (HC), who underwent dual radionuclide imaging with (99m)Tc-labeled matrix metalloproteinase inhibitor (MPI) and (111)In-labeled annexin A5 (AA5) using micro-SPECT/CT. %ID/g MPI and AA5 uptake was measured, followed by histological characterization. Unmanipulated animals were used as disease controls. Correlation between MPI and AA5 uptake was undertaken and relationship confirmed in culture study of activated THP-1 monocytes. RESULTS MPI and AA5 uptake was best visualized in HC diet animals (n = 6) and reduced significantly after fluvastatin treatment (n = 4) or diet withdrawal (n = 3). %ID/g MPI (.087 +/- .018%) and AA5 (.03 +/- .01%) uptake was higher in HC than control (n = 6) animals (.014 +/- .004%, P < .0001; .0007 +/- .0002%, P < .0001), and reduced substantially after diet or statin intervention. There was a significant correlation between MPI and AA5 uptake (r = .62, P < .0001), both correlated with pathologically verified MMP-9 activity, macrophage content, and TUNEL staining. In vitro studies demonstrated MMP-9 release in culture medium from apoptotic THP-1 monocytes. CONCLUSIONS The present study suggests that apoptosis and MMP are interrelated in atherosclerotic lesions and the targeting of more than one molecular candidate is feasible by molecular imaging.
Collapse
Affiliation(s)
- Nezam Haider
- Department of Cardiology, Irvine School of Medicine, University of California, C116 Med Sci I, Irvine, CA 92697 USA
| | - Dagmar Hartung
- Department of Cardiology, Irvine School of Medicine, University of California, C116 Med Sci I, Irvine, CA 92697 USA
| | - Shinichiro Fujimoto
- Department of Cardiology, Irvine School of Medicine, University of California, C116 Med Sci I, Irvine, CA 92697 USA
| | - Artiom Petrov
- Department of Cardiology, Irvine School of Medicine, University of California, C116 Med Sci I, Irvine, CA 92697 USA
| | | | - Renu Virmani
- Cardiovascular Pathology Institute, Gaithersburg, MD USA
| | - Satoru Ohshima
- Department of Cardiology, Irvine School of Medicine, University of California, C116 Med Sci I, Irvine, CA 92697 USA
| | - Han Liu
- Department of Cardiology, Irvine School of Medicine, University of California, C116 Med Sci I, Irvine, CA 92697 USA
| | - Jun Zhou
- Department of Cardiology, Irvine School of Medicine, University of California, C116 Med Sci I, Irvine, CA 92697 USA
| | - Ai Fujimoto
- Department of Cardiology, Irvine School of Medicine, University of California, C116 Med Sci I, Irvine, CA 92697 USA
| | - Atsuko Tahara
- Department of Cardiology, Irvine School of Medicine, University of California, C116 Med Sci I, Irvine, CA 92697 USA
| | - Leo Hofstra
- Department of Cardiology, University Hospital Maastricht, Academic University of Maastricht, Maastricht, The Netherlands
| | - Navneet Narula
- Department of Pathology, Irvine School of Medicine, University of California, C116 Med Sci I, Irvine, CA 92697 USA
| | - Chris Reutelingsperger
- Department of Biochemistry, University Hospital Maastricht, Academic University of Maastricht, Maastricht, The Netherlands
| | - Jagat Narula
- Department of Cardiology, Irvine School of Medicine, University of California, C116 Med Sci I, Irvine, CA 92697 USA
| |
Collapse
|
43
|
Kenagy RD, Min SK, Clowes AW, Sandy JD. Cell death-associated ADAMTS4 and versican degradation in vascular tissue. J Histochem Cytochem 2009; 57:889-97. [PMID: 19506088 DOI: 10.1369/jhc.2009.953901] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
High blood flow through baboon polytetrafluorethylene aorto-iliac grafts increases neointimal vascular smooth muscle cell (SMC) death, neointimal atrophy, and cleavage of versican to generate the DPEAAE neoepitope, a marker of ADAMTS-mediated proteolysis. In this study, we have determined the effect of high blood flow on transcript abundance in the neointima for ADAMTS1, -4, -5, -8, -9, -15, and -20. We found that after 24 hr of flow, the mRNA for ADAMTS4 was significantly increased, whereas that for the other family members was unchanged. Because vascular SMC death is markedly increased in the graft after 24 hr of high flow, we next examined the possibility that the ADAMTS4 induction and the cell death are causally related. The addition of Fas ligand to SMC cultures increased both ADAMTS4 mRNA and cell death approximately 5-fold, consistent with the idea that ADAMTS4-dependent cleavage of versican may be partly responsible for cell death and tissue atrophy under these conditions.
Collapse
Affiliation(s)
- Richard D Kenagy
- Center for Cardiovascular Biology, PO Box 358050, University of Washington School of Medicine, 815 Mercer St., Seattle, WA 98109.
| | | | | | | |
Collapse
|
44
|
Zacharioudaki V, Androulidaki A, Arranz A, Vrentzos G, Margioris AN, Tsatsanis C. Adiponectin promotes endotoxin tolerance in macrophages by inducing IRAK-M expression. THE JOURNAL OF IMMUNOLOGY 2009; 182:6444-51. [PMID: 19414798 DOI: 10.4049/jimmunol.0803694] [Citation(s) in RCA: 61] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
High levels of plasma adiponectin are associated with low levels of inflammatory markers and cardioprotection. The mechanism via which adiponectin exerts its anti-inflammatory effect is yet unknown. In the present study, we demonstrate that globular adiponectin (gAd) induces the expression of the inactive isoform of IL-1R-associated kinases (IRAK), IRAK-M. Homologous deletion of IRAK-M in IRAK-M(-/-) mice abolished the tolerogenic properties of gAd because pretreatment of IRAK-M(-/-) macrophages with gAd did not suppress LPS-induced proinflammatory cytokine production. GAd activated the MAPKs MEK1/2 and ERK1/2 in macrophages via their upstream regulator Tpl2. Activation of ERK1/2 via Tpl2 appeared necessary for the induction of IRAK-M because gAd did not induce IRAK-M in Tpl2(-/-) macrophages or in macrophages pretreated with the MEK1/2 inhibitor UO126. In addition, activation of PI3K and Akt1 also appeared necessary for the induction of IRAK-M by gAd, because treatment of Akt1(-/-) macrophages or pretreatment of macrophages with the PI3K inhibitor wortmannin abolished gAd-induced IRAK-M expression. Analysis of IRAK-M expression in human peripheral blood cells confirmed that serum adiponectin was negatively associated with IRAK-M and responsiveness to LPS. In conclusion, our data demonstrate that IRAK-M is a major mediator of gAd-induced endotoxin tolerance in primary macrophages, expression of which depends on the activation of Tpl2/ERK and PI3K/Akt1 signaling pathways.
Collapse
Affiliation(s)
- Vassiliki Zacharioudaki
- Department of Clinical Chemistry, Graduate Program on Molecular Basis of Human Disease, School of Medicine, University of Crete, Crete, Greece
| | | | | | | | | | | |
Collapse
|
45
|
Tahara N, Imaizumi T, Virmani R, Narula J. Clinical feasibility of molecular imaging of plaque inflammation in atherosclerosis. J Nucl Med 2009; 50:331-4. [PMID: 19223412 DOI: 10.2967/jnumed.108.060376] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Despite substantial advances in the diagnosis and management of coronary artery disease, acute coronary events continue to occur in many patients. It has been increasingly realized that the lesions responsible for acute events may not necessarily be critically obstructive and hence not be associated with inducible ischemia. Various morphologic features of plaque vulnerability have been described by CT angiography, intravascular ultrasound, and optical coherence tomography. The culprit plaques often demonstrate large plaque and necrotic core volumes, positive vascular remodeling, and attenuation of fibrous plaque caps. The remaining obligatory component of plaque vulnerability is fibrous cap inflammation; molecular imaging is best suited for identification of monocyte-macrophage infiltration. Whereas multiple candidate targets have been evaluated in preclinical molecular imaging studies, only (18)F-FDG and (99m)Tc-annexin-A5 have been recently used in the settings of acute vascular events. These 2 imaging strategies have demonstrated the clinical feasibility of imaging for detection of inflammation.
Collapse
|
46
|
Matsuda N, Yamamoto S, Takano KI, Kageyama SI, Kurobe Y, Yoshihara Y, Takano Y, Hattori Y. Silencing of fas-associated death domain protects mice from septic lung inflammation and apoptosis. Am J Respir Crit Care Med 2009; 179:806-15. [PMID: 19201926 DOI: 10.1164/rccm.200804-534oc] [Citation(s) in RCA: 66] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
RATIONALE A better understanding of the molecular mechanisms involved in the pathogenesis of sepsis and its resultant organ failure and new therapeutic approaches and targets are urgently needed. Accumulating evidence suggests that apoptosis plays an important role in the pathophysiology of sepsis and that apoptosis may be detrimental in septic acute lung injury (ALI). OBJECTIVES We tested the hypothesis that systemic administration of small interfering RNA (siRNA) targeting Fas-associated death domain (FADD), which recruits procaspase-8 into the death-inducing signaling complex, may be protective in septic ALI and mortality. METHODS Polymicrobial sepsis was induced by cecal ligation and puncture (CLP) in BALB/c mice. In vivo delivery of siRNA was performed by using a transfection reagent at 10 hours after CLP. As a negative control, animals received nonsense (scrambled) siRNA. MEASUREMENTS AND MAIN RESULTS In CLP-induced septic mice, surface expression of death receptors was up-regulated, and FADD was highly expressed. DNA fragmentation ladder and transferase-mediated dUTP nick end labeling assays showed that treatment with FADD siRNA suppressed apoptosis induction in septic lungs. This siRNA treatment prevented the ALI development in CLP mice, as indicated by the findings that blood-gas derangements, histologic lung damage, and increased pulmonary inflammatory cells were greatly improved. Finally, FADD siRNA administration dramatically improved the survival of CLP mice. CONCLUSIONS These results indicate the pathophysiologic significance of the death receptor apoptotic pathway, including FADD, in septic ALI and the potential usefulness of FADD siRNA for gene therapy of the septic syndrome.
Collapse
Affiliation(s)
- Naoyuki Matsuda
- Department of Molecular and Medical Pharmacology, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama 930-0194, Japan
| | | | | | | | | | | | | | | |
Collapse
|
47
|
Lyon CA, Johnson JL, Williams H, Sala-Newby GB, George SJ. Soluble N-cadherin overexpression reduces features of atherosclerotic plaque instability. Arterioscler Thromb Vasc Biol 2009; 29:195-201. [PMID: 19008530 PMCID: PMC2853707 DOI: 10.1161/atvbaha.108.178087] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE Vascular smooth muscle cell (VSMC) apoptosis contributes to atherosclerotic plaque instability and myocardial infarction. Consequently, reducing VSMC apoptosis may be beneficial for reducing plaque instability and acute coronary events. We previously demonstrated that N-cadherin, a cell-cell adhesion molecule, reduces VSMC apoptosis in vitro. In this study, we examined whether a soluble form of N-cadherin (SNC) affected VSMC apoptosis and plaque stability. METHODS AND RESULTS SNC significantly inhibited VSMC apoptosis in vitro by approximately 50% via activation of fibroblast growth factor receptor, phosphoinositide-3 kinase, and Akt signaling. SNC also significantly reduced macrophage and foam cell-macrophage apoptosis in vitro by >50%, without affecting monocyte invasion or macrophage proliferation. Elevation of plasma levels of SNC in male apolipoprotein E-deficient mice with existing atherosclerosis via adenoviral delivery significantly reduced VSMC and macrophage apoptosis in brachiocephalic artery plaques by approximately 60%. Additionally, SNC promoted plaques of a more stable phenotype by elevating VSMC:macrophage ratio and presence of VSMC-rich fibrous cap, as well as attenuating macrophage number and incidence of buried fibrous caps (a surrogate plaque rupture marker). CONCLUSIONS In summary, this study demonstrates that SNC suppressed plaque instability by attenuation of apoptosis, suggesting that SNC may have a therapeutic potential for retarding plaque instability.
Collapse
MESH Headings
- Adenoviridae/genetics
- Animals
- Apolipoproteins E/deficiency
- Apolipoproteins E/genetics
- Apoptosis
- Atherosclerosis/genetics
- Atherosclerosis/metabolism
- Atherosclerosis/pathology
- Atherosclerosis/therapy
- Brachiocephalic Trunk/metabolism
- Brachiocephalic Trunk/pathology
- CHO Cells
- Cadherins/biosynthesis
- Cadherins/genetics
- Cell Adhesion
- Cricetinae
- Cricetulus
- Disease Models, Animal
- Foam Cells/metabolism
- Foam Cells/pathology
- Genetic Therapy/methods
- Genetic Vectors
- Humans
- Male
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Monocytes/metabolism
- Monocytes/pathology
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Peptide Fragments/biosynthesis
- Signal Transduction
- Time Factors
- Transduction, Genetic
- Up-Regulation
Collapse
|
48
|
Halvorsen B, Otterdal K, Dahl TB, Skjelland M, Gullestad L, Øie E, Aukrust P. Atherosclerotic plaque stability--what determines the fate of a plaque? Prog Cardiovasc Dis 2008; 51:183-94. [PMID: 19026853 DOI: 10.1016/j.pcad.2008.09.001] [Citation(s) in RCA: 112] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Although the understanding of the underlying pathology of atherosclerosis has improved in recent years, the disease is still the main cause of death globally. Current evidence has implicated the role of inflammation in atherogenesis and plaque destabilization. Thus, inflammatory cytokines may attenuate interstitial collagen synthesis, increase matrix degradation, and promote apoptosis in several atheroma-associated cell types, and all these cellular events may enhance plaque vulnerability. Several cell types found within the lesion (ie, monocyte/macrophages, T cells, mast cells, platelets) contribute to this immune-mediated plaque destabilization, and a better understanding of these processes is a prerequisite for the development of new treatment strategies in these individuals. Such knowledge could also facilitate a better identification of high-risk individuals. In the present study, these issues will be discussed in more detail, particularly focusing on the interactions between matrix degradation, apoptotic, and inflammatory processes in plaque destabilization.
Collapse
Affiliation(s)
- Bente Halvorsen
- Research Institute for Internal Medicine, Department of Neurology, Rikshospitalet Medical Center, Oslo, Norway.
| | | | | | | | | | | | | |
Collapse
|
49
|
Perl M, Lomas-Neira J, Chung CS, Ayala A. Epithelial cell apoptosis and neutrophil recruitment in acute lung injury-a unifying hypothesis? What we have learned from small interfering RNAs. Mol Med 2008; 14:465-75. [PMID: 18368145 PMCID: PMC2274893 DOI: 10.2119/2008-00011.perl] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2008] [Accepted: 03/17/2008] [Indexed: 01/11/2023] Open
Abstract
In spite of protective ventilatory strategies, Acute Lung Injury (ALI) remains associated with high morbidity and mortality. One reason for the lack of therapeutic options might be that ALI is a co-morbid event associated with a diverse family of diseases and, thus, may be the result of distinct pathological processes. Among them, activated neutrophil- (PMN-) induced tissue injury and epithelial cell apoptosis mediated lung damage represent two potentially important candidate pathomechanisms that have been put forward. Several approaches have been undertaken to test these hypotheses, with substantial success in the treatment of experimental forms of ALI. With this in mind, we will summarize these two current hypotheses of ALI briefly, emphasizing the role of apoptosis in regulating PMN and/or lung epithelial cell responses. In addition, the contribution that Fas-mediated inflammation may play as a potential biological link between lung cell apoptosis and PMN recruitment will be considered, as well as the in vivo application of small interfering RNA (siRNA) as a novel approach to the inhibition of ALI and its therapeutic implications.
Collapse
Affiliation(s)
- Mario Perl
- Department of Traumatology, Hand- and Reconstructive Surgery, University of Ulm Medical School, Ulm, Germany
| | | | | | | |
Collapse
|
50
|
Mao Q, Gundavarapu S, Patel C, Tsai A, Luks FI, De Paepe ME. The Fas system confers protection against alveolar disruption in hyperoxia-exposed newborn mice. Am J Respir Cell Mol Biol 2008; 39:717-29. [PMID: 18587053 DOI: 10.1165/rcmb.2008-0052oc] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
The functional significance of the Fas/Fas-ligand (FasL) system in hyperoxia-induced lung injury and alveolar disruption in newborn lungs in vivo remains undetermined. To assess the role of the Fas/FasL system, we compared the effects of hyperoxia (95% O2 from birth to Postnatal Day [P]7) in Fas-deficient lpr mice and wild-type mice. Alveolar disruption was more severe in hyperoxic lpr mice than in wild-type mice. In addition, a transient alveolarization defect was noted in normoxic lpr mice. Hyperoxia induced marked up-regulation of pulmonary Fas expression in wild-type mice, as well as elevated mRNA levels of pro-apoptotic Bax, Bad, and Bak. Pulmonary apoptotic activity was similar in hyperoxic wild-type and lpr mice. In contrast, lung growth and proliferation, assessed by stereologic volumetry and Ki67 proliferation studies, were significantly higher in hyperoxic wild-type mice compared with lpr mice, suggesting the Fas/FasL system has a pro-proliferative role in hyperoxic conditions. Levels of the prosurvival MAPkinase, pERK1/2, were significantly higher in hyperoxic wild-type mice compared with lpr mice, while pAkt levels were similar. These data suggest that the primary role of the Fas/FasL system in hyperoxic newborn lungs is pro-proliferative, rather than pro-apoptotic, and likely mediated through a Fas-ERK1/2 pathway. Fas-induced proliferation and lung growth in hyperoxic newborn lungs may counteract, in part, the detrimental effects of apoptosis mediated by non-Fas pathways, such as pro-apoptotic Bax/Bcl-2 family members. The capacity of the Fas/FasL signaling pathway to mediate protective rather than destructive functions in hyperoxic newborn lungs highlights the versatility of this complex pathway.
Collapse
Affiliation(s)
- Quanfu Mao
- Department of Pathology, Women and Infants Hospital, Providence, Rhode Island 02905, USA
| | | | | | | | | | | |
Collapse
|