Caspase-8, a Double-Edged Sword for EPC Functioning.
Qingzhong Xiao,Qingbo Xu
DOI: https://doi.org/10.1161/atvbaha.108.183087
2009-01-01
Arteriosclerosis Thrombosis and Vascular Biology
Abstract:HomeArteriosclerosis, Thrombosis, and Vascular BiologyVol. 29, No. 4Caspase-8, a Double-Edged Sword for EPC Functioning Free AccessEditorialPDF/EPUBAboutView PDFView EPUBSections ToolsAdd to favoritesDownload citationsTrack citationsPermissions ShareShare onFacebookTwitterLinked InMendeleyReddit Jump toFree AccessEditorialPDF/EPUBCaspase-8, a Double-Edged Sword for EPC Functioning Qingzhong Xiao and Qingbo Xu Qingzhong XiaoQingzhong Xiao From the Cardiovascular Division, King’s College London BHF Centre, UK. Search for more papers by this author and Qingbo XuQingbo Xu From the Cardiovascular Division, King’s College London BHF Centre, UK. Search for more papers by this author Originally published1 Apr 2009https://doi.org/10.1161/ATVBAHA.108.183087Arteriosclerosis, Thrombosis, and Vascular Biology. 2009;29:444–446Endothelial progenitor cells (EPCs) were initially identified as bone marrow–derived circulating cells expressing endothelial-specific markers.1 These progenitors may contribute to repairing denuded endothelium of injured arteries in animal models2–4 and improving neovascularization and tissue perfusion after ischemia,5–7 highlighting the potential of these cells for therapy for cardiovascular diseases. Theoretically, the benefit of EPC therapy does not appear to be solely attributable to the ability of the cells to incorporate into the endothelium and differentiate into endothelial cells to provide physically contribution to new blood vessel formation in sites, but also as a result of the additional capacity to modulate the function of preexisting vascular cells via the paracrine effects of factors released from the cells.8–10 Thus, proper retention and survival of progenitor cells within the target tissue will be crucial for successful neovascularization improvement after therapeutic application of EPCs. However, various studies aimed to investigate the homing and the long term engraftment of cells demonstrated clearly only a low percentage (1% to 30%) of infused cells are recruited and survival in target tissue/organ in animal models as well as in clinical trials for short periods.11 Therefore, strategies to improve endogenous/exogenous progenitor cell, homing, retention, and survival in target tissues to improve the efficiency of cell therapy are essential.See accompanying article on page 571In this issue of Atherosclerosis, Thrombosis, and Vascular Biology, Scharner et al12 demonstrate a novel apoptosis-unrelated role of Caspase-8 for EPC-mediated neovascularization that can be applied to improve the efficiency of cell therapy. The authors first aimed to augment EPC survival for therapeutic application through inhibition of EPC apoptosis by caspase inhibitors. Surprisingly, however, they found that selective inhibition of caspase-8 abolished ex vivo EPC formation, and inhibited EPC adhesion and migration, as well as significantly reduced the capacity of EPCs to improve neovascularization in vivo after ischemia, indicating that Caspase-8 has an unexpected role in neovascularization-promoting function. Their solid and direct evidence came from the experiment in which cells isolated from Caspase-8–deficient mice exhibited a severely reduced capacity for enhancing neovascularization when transplanted into mice after hind limb ischemia. The authors further demonstrated that the expression of the fibronectin receptor subunits α5 and β1 and the SDF-1 receptor CXCR4 on EPC surface, which is involved in EPC adhesion and homing, were markedly reduced by pharmacological inhibition and genetic depletion of Caspase-8. Importantly, the authors finally identified the E3 ubiquitin ligase Cbl-b as a potential Caspase-8 substrate. This finding, at least in part, reveals the underlying mechanisms on how Caspase-8 inhibition reduced fibronectin receptor subunits α5 and β1 and the SDF-1 receptor CXCR4 expression, which in turn affected the EPC function on neovascularization. Caspase-8 essential for EPC-mediated neovascularization has important clinical implications, because this finding creates a new perspective that Caspase-8 may function as a double-edged sword for the survival/functions of EPCs. Therefore, strategies to improve retention and survival of injected progenitor cells at sites by using antiapoptotic inhibitors or drugs should be considered carefully before applying in clinical practice (Figure). Download figureDownload PowerPointFigure. A schematic illustration of the mechanism involved in Caspase-8 apoptotic and nonapoptotic functions. Caspase-8 not only mediates apoptosis as an initiator of caspase-cascade, but also has nonapoptotic functions, such as proliferation and NF-κB activation. The outcome of Caspase-8 activation for either apoptotic or nonapoptotic is probably determined by the extent of its activation, ie, threshold. DR indicates death receptor; NF-κB, nuclear factor κB; PARP-1, poly(ADP-ribose) polymerase-1; c-FLIP, cellular caspase-8 (FLICE)-like inhibitory protein; FADD, Fas-associated death domain containing protein; MST1, Mammalian Sterile Twenty-like kinase.Growing evidence implicates that Caspase-8 is also involved in plenty of nonapoptotic functions, such as NF-κB activation.13–17 The enzymatic activity of Caspase-8 required for terminal cell differentiation seems to be one of their universal features of caspase family, either through positive or negative regulators (Figure). Our previous study demonstrated that the activity of another caspase member, Caspase-3, is important for stem cell differentiation toward smooth muscle cells.18 Inhibition of Caspase-3 slightly reduced cell apoptosis but significantly increased smooth muscle cell differentiation, indicating that caspase activity plays an important role in the signal pathway controlling the switch of stem cell fate from apoptosis to terminal differentiation.18 However, the precise functional role of caspase activity in progenitor/stem cell terminal differentiation remains to be explored.Recently we demonstrated that SDF-1α is a causal determinant of the rate of circulating EPCs in the general population.19,20 One possible explanation for this finding is that high levels of serum SDF-1α activates CXCR4 expression on circulating EPCs, which in turn promotes circulating EPC homing and recruitment to target tissue resulting in reduced EPC number in blood. The current finding that inhibition of Caspase-8 results in reduction of SDF-1α receptor CXCR4 expression on EPCs, then significantly inhibits their capacity to homing and incorporate to target tissue after ischemia, strongly supports our findings. This raises an interesting issue that SDF-1α and Caspase-8 may work together to regulate EPC number and function. However, the potential relationships between Caspase-8, SDF-1α, CXCR4, and circulating EPCs in humans remain to be clarified.Although most of the experiments were performed carefully by the authors, a couple of issues regarding the exact function of Caspase-8 in neovascularization-promoting progenitor cells remain to be answered. The authors observed the dependency of adhesion and migration on Caspase-8 activity is restricted to “early EPCs” but not mature endothelial cells. It is unknown whether different therapeutic benefits exist between “stem/progenitor-derived endothelial cells” and mature endothelial cells. Thus, it would be interesting to investigate how Caspase-8 signal pathway is directly involved in such therapeutic effects of “early EPCs.” Moreover, there is no direct evidence to reveal how the ubiquitin ligase Cbl-b regulates integrin α5β1 and CXCR4 expression on EPCs. In addition, the authors suggested that Caspase-8 exerts their nonapoptotic function may be attributable to the basal Caspase-8 activity below the threshold to stimulate the apoptosis cascade. Therefore, further investigation and better understanding on the threshold of Caspase-8 activation are crucial for controlling Caspase-8 function under different physiological and pathological conditions.In summary, EPC homing and engraftment, involving chemokines, adhesion molecules, and proteases, are prerequisite for angiogenesis potential in the target tissue particularly when cells are infused via the vascular route. The authors in the current study proposed that maintaining appropriate amount of Caspase-8 activity (under a certain threshold) is crucial for EPC function. In this process, Caspase-8 displays functions in degrading the E3 ubiquitin ligase Cb1–1, enhancing integrin α5β1 and CXCR4 expression, promoting EPC adhesion and incorporation to target tissue, which eventually improves neovascularization after ischemia. Their findings not only modify our traditional views about apoptotic and nonapoptotic functions of Caspase-8, but also have some clinical implications. Thus, strategies to improve retention and survival of injected progenitor cells at sites by using antiapoptotic inhibitors or drugs should be considered cautiously.Sources of FundingThis work was supported by grants from the British Heart Foundation and Oak Foundation.DisclosuresNone.FootnotesCorrespondence to Professor Qingbo Xu, Cardiovascular Division, King’s College London, 125 Coldharbour Lane, London SE5 9NU, UK. E-mail [email protected] References 1 Asahara T, Murohara T, Sullivan A, Silver M, van der Zee R, Li T, Witzenbichler B, Schatteman G, Isner JM. Isolation of putative progenitor endothelial cells for angiogenesis. Science. 1997; 275: 964–967.CrossrefMedlineGoogle Scholar2 Xiao Q, Zeng L, Zhang Z, Margariti A, Ali ZA, Channon KM, Xu Q, Hu Y. Sca-1+ progenitors derived from embryonic stem cells differentiate into endothelial cells capable of vascular repair after arterial injury. Arterioscler Thromb Vasc Biol. 2006; 26: 2244–2251.LinkGoogle Scholar3 Werner N, Junk S, Laufs U, Link A, Walenta K, Bohm M, Nickenig G. Intravenous transfusion of endothelial progenitor cells reduces neointima formation after vascular injury. Circ Res. 2003; 93: e17–e24.LinkGoogle Scholar4 Walter DH, Rittig K, Bahlmann FH, Kirchmair R, Silver M, Murayama T, Nishimura H, Losordo DW, Asahara T, Isner JM. Statin therapy accelerates reendothelialization: a novel effect involving mobilization and incorporation of bone marrow-derived endothelial progenitor cells. Circulation. 2002; 105: 3017–3024.LinkGoogle Scholar5 Assmus B, Schachinger V, Teupe C, Britten M, Lehmann R, Dobert N, Grunwald F, Aicher A, Urbich C, Martin H, Hoelzer D, Dimmeler S, Zeiher AM. Transplantation of Progenitor Cells and Regeneration Enhancement in Acute Myocardial Infarction (TOPCARE-AMI). Circulation. 2002; 106: 3009–3017.LinkGoogle Scholar6 Schachinger V, Erbs S, Elsasser A, Haberbosch W, Hambrecht R, Holschermann H, Yu J, Corti R, Mathey DG, Hamm CW, Suselbeck T, Assmus B, Tonn T, Dimmeler S, Zeiher AM. Intracoronary bone marrow-derived progenitor cells in acute myocardial infarction. N Engl J Med. 2006; 355: 1210–1221.CrossrefMedlineGoogle Scholar7 Assmus B, Honold J, Schachinger V, Britten MB, Fischer-Rasokat U, Lehmann R, Teupe C, Pistorius K, Martin H, Abolmaali ND, Tonn T, Dimmeler S, Zeiher AM. Transcoronary transplantation of progenitor cells after myocardial infarction. N Engl J Med. 2006; 355: 1222–1232.CrossrefMedlineGoogle Scholar8 Grunewald M, Avraham I, Dor Y, Bachar-Lustig E, Itin A, Jung S, Chimenti S, Landsman L, Abramovitch R, Keshet E. VEGF-induced adult neovascularization: recruitment, retention, and role of accessory cells. Cell. 2006; 124: 175–189.CrossrefMedlineGoogle Scholar9 Jin DK, Shido K, Kopp HG, Petit I, Shmelkov SV, Young LM, Hooper AT, Amano H, Avecilla ST, Heissig B, Hattori K, Zhang F, Hicklin DJ, Wu Y, Zhu Z, Dunn A, Salari H, Werb Z, Hackett NR, Crystal RG, Lyden D, Rafii S. Cytokine-mediated deployment of SDF-1 induces revascularization through recruitment of CXCR4+ hemangiocytes. Nat Med. 2006; 12: 557–567.CrossrefMedlineGoogle Scholar10 Hur J, Yoon CH, Kim HS, Choi JH, Kang HJ, Hwang KK, Oh BH, Lee MM, Park YB. Characterization of two types of endothelial progenitor cells and their different contributions to neovasculogenesis. Arterioscler Thromb Vasc Biol. 2004; 24: 288–293.LinkGoogle Scholar11 Chavakis E, Urbich C, Dimmeler S. Homing and engraftment of progenitor cells: a prerequisite for cell therapy. J Mol Cell Cardiol. 2008; 45: 514–522.CrossrefMedlineGoogle Scholar12 Scharner D, Rössig L, Carmona G, Chavakis E, Urbich C, Fischer A, Kang TB, Wallach D, Chiang YJ, Deribe YL, Dikic I, Zeiher AM, Dimmeler S. Caspase-8 is involved in neovascularization-promoting progenitor cell functions. Arterioscler Thromb Vasc Biol. 2009; 29: 571–578.LinkGoogle Scholar13 Varfolomeev E, Maecker H, Sharp D, Lawrence D, Renz M, Vucic D, Ashkenazi A. Molecular determinants of kinase pathway activation by Apo2 ligand/tumor necrosis factor-related apoptosis-inducing ligand. J Biol Chem. 2005; 280: 40599–40608.CrossrefMedlineGoogle Scholar14 Su H, Bidere N, Zheng L, Cubre A, Sakai K, Dale J, Salmena L, Hakem R, Straus S, Lenardo M. Requirement for caspase-8 in NF-kappaB activation by antigen receptor. Science. 2005; 307: 1465–1468.CrossrefMedlineGoogle Scholar15 Jun JI, Chung CW, Lee HJ, Pyo JO, Lee KN, Kim NS, Kim YS, Yoo HS, Lee TH, Kim E, Jung YK. Role of FLASH in caspase-8-mediated activation of NF-kappaB: dominant-negative function of FLASH mutant in NF-kappaB signaling pathway. Oncogene. 2005; 24: 688–696.CrossrefMedlineGoogle Scholar16 Sordet O, Rebe C, Plenchette S, Zermati Y, Hermine O, Vainchenker W, Garrido C, Solary E, Dubrez-Daloz L. Specific involvement of caspases in the differentiation of monocytes into macrophages. Blood. 2002; 100: 4446–4453.CrossrefMedlineGoogle Scholar17 Kang TB, Ben-Moshe T, Varfolomeev EE, Pewzner-Jung Y, Yogev N, Jurewicz A, Waisman A, Brenner O, Haffner R, Gustafsson E, Ramakrishnan P, Lapidot T, Wallach D. Caspase-8 serves both apoptotic and nonapoptotic roles. J Immunol. 2004; 173: 2976–2984.CrossrefMedlineGoogle Scholar18 Xiao Q, Roberts N, Jahangiri M, Xu Q. Stem cells, progenitor cells and vascular diseases. In Stem Cell Research Development. Fong CA (ed), NOVA Science Publishers, 2007; 5–54.Google Scholar19 Xiao Q, Kiechl S, Patel S, Oberhollenzer F, Weger S, Mayr A, Metzler B, Reindl M, Hu Y, Willeit J, Xu Q. Endothelial progenitor cells, cardiovascular risk factors, cytokine levels and atherosclerosis–results from a large population-based study. PLoS ONE. 2007; 2: e975.CrossrefMedlineGoogle Scholar20 Xiao Q, Ye S, Oberhollenzer F, Mayr A, Jahangiri M, Willeit J, Kiechl S, Xu Q. SDF1 gene variation is associated with circulating SDF1alpha level and endothelial progenitor cell number: the Bruneck Study. PLoS ONE. 2008; 3: e4061.CrossrefMedlineGoogle Scholar Previous Back to top Next FiguresReferencesRelatedDetailsCited By Abebe W, Baban B and Mozaffari M (2013) Endothelial Dysfunction in Diabetes: Role of Circulating Biomarkers as Potential Diagnostic and Prognostic Tools New Strategies to Advance Pre/Diabetes Care: Integrative Approach by PPPM, 10.1007/978-94-007-5971-8_5, (117-146), . Dragu A, Schnürer S, Surmann-Schmitt C, von der Mark K, Stürzl M, Unglaub F, Wolf M, Leffler M, Beier J, Kneser U and Horch R (2011) Gene expression analysis of ischaemia and reperfusion in human microsurgical free muscle tissue transfer, Journal of Cellular and Molecular Medicine, 10.1111/j.1582-4934.2010.01061.x, 15:4, (983-993), Online publication date: 1-Apr-2011. Abebe W and Mozaffari M (2010) Endothelial dysfunction in diabetes: potential application of circulating markers as advanced diagnostic and prognostic tools, EPMA Journal, 10.1007/s13167-010-0012-7, 1:1, (32-45), Online publication date: 1-Mar-2010. April 2009Vol 29, Issue 4 Advertisement Article InformationMetrics https://doi.org/10.1161/ATVBAHA.108.183087PMID: 19299329 Originally publishedApril 1, 2009 PDF download Advertisement
What problem does this paper attempt to address?
-
Endothelial Progenitor Cells May Inhibit Apoptosis of Pulmonary Microvascular Endothelial Cells: New Insights into Cell Therapy for Pulmonary Arterial Hypertension
Liang Xia,Guo-Sheng Fu,Jin-Xiu Yang,Fu-Rong Zhang,Xing-Xiang Wang
DOI: https://doi.org/10.1080/14653240902960460
IF: 6.196
2009-01-01
Cytotherapy
Abstract:Background aimsEndothelial apoptosis underlies the pathophysiology of pulmonary arterial hypertension (PAH). Some factors/cytokines released by endothelial progenitor cells (EPC) have been revealed as potent inhibitors of apoptosis. The aim of this study was to investigate the effects of EPC on pulmonary microvascular endothelial cell (PMVEC) survival with the PAH condition.MethodsPMVEC apoptosis was induced by high shear stress (HSS) with serum starvation or pro-inflammatory factors in an artificial capillary system. EPC were delivered into monocrotaline-induced PAH nude rats.ResultsPMVEC apoptosis under HSS and serum starvation conditions was significantly inhibited by EPC conditioned medium (CM). It was attenuated by vascular endothelial growth factor (VEGF)-A or -B blocking. EPC CM promoted PMVEC proliferation, which was weakened by VEGF-A or interleukin (IL)-8 blocking. The EPC CM caused less apoptosis of PMVEC induced by HSS plus pro-inflammatory factors. The anti-apoptotic effect of EPC CM was attenuated by blockade of either vascular endothelial growth factor receptor (VEGFR)-1 or -2. However, the pro-proliferating effect appeared to be weakened only by VEGFR-2 blocking. Both Erk1/2 and Akt phosphorylation were enhanced by EPC CM. VEGFR-2 blockage led to significant inhibition of Erk1/2 and Akt activation; VEGFR-1 blockage only of Erk1/2 activation. Human-origin VEGF co-localized with incorporated EPC in small pulmonary arterioles, and EPC transplantation resulted in down-regulation of caspase-3 expression.ConclusionsThe VEGF-A/B-VEGFR-1/2-Erk1/2 signal pathway took major responsibility for the anti-apoptotic effects of EPC on PMVEC, and VEGF-A-VEGFR-2-Akt for pro-proliferating effects. Growth factors, secreted in a paracrine manner by transplanted EPC, inhibited cell apoptosis in PAH lung.
-
Caspase-8 Modulates Angiogenesis By Regulating A Cell Death Independent Pathway In Endothelial Cells
Nathalie Tisch,Aida Freire-Valls,Rosario Yerbes,Isidora Paredes,Silvia La Porta,Xiaohong Wang,Rosa Martín-Pérez,Laura Castro,Wendy Wei-Lynn Wong,Leigh Coultas,Boris Strilic,Hermann-Josef Gröne,Thomas Hielscher,Carolin Mogler,Ralf Adams,Peter Heiduschka,Lena Claesson-Welsh,Massimiliano Mazzone,Abelardo López-Rivas,Thomas Schmidt,Hellmut G. Augustin,Carmen Ruiz de Almodovar
DOI: https://doi.org/10.1101/708651
2019-01-01
Abstract:During developmental angiogenesis blood vessels grow and remodel to ultimately build a hierarchical vascular network. Whether and how cell death signaling molecules contribute to blood vessel formation is still not well understood. Caspase-8 (Casp-8), a key protease in the extrinsic cell death-signaling pathway, regulates both cell death via apoptosis and necroptosis. Here we show that expression of Casp-8 in endothelial cells (ECs) is required for proper postnatal angiogenesis. EC specific Casp-8 knockout pups (Casp-8) have reduced retinal angiogenesis, as the loss of Casp-8 reduced EC proliferation, sprouting and migration independent of its cell death function. Instead, the loss of Casp-8 caused hyperactivation of p38 mitogen-activated protein kinase (MAPK) downstream of receptorinteracting serine/threonine-protein kinase 3 (RIPK3) and destabilization of VE-cadherin at EC junctions. In a mouse model of oxygen-induced retinopathy (OIR), resembling retinopathy of prematurity (ROP), loss of Casp-8 in ECs is beneficial, as pathological neovascularization was reduced in Casp-8 pups. Taken together, we identify that Casp-8 signals in a cell-death independent manner in ECs during postnatal and pathological blood vessel formation.
-
Stromal Cell‐derived Factor 1α Reduces Senescence of Endothelial Progenitor Subpopulation in Lectin‐binding and DiLDL‐uptaking Cell Through Telomerase Activation and Telomere Elongation
Hao Zheng,Cheng-Ji Shen,Fu-Yu Qiu,Yan-Bo Zhao,Guo-Sheng Fu
DOI: https://doi.org/10.1002/jcp.22086
2010-01-01
Journal of Cellular Physiology
Abstract:Recent studies have suggested that reduced endothelial progenitor subpopulation in lectin‐binding and DiLDL‐uptaking cell (EPC subpopulation) number and activity was associated with EPC subpopulation senescence that involved telomerase activity and telomere length. Stromal cell‐derived factor‐1α (SDF‐1α) has been shown to augment a variety of cellular functions of EPC subpopulation and subsequently contribute to ischemic neovascularization. Therefore, we investigated whether SDF‐1α might be able to prevent senescence of EPC subpopulation and also investigated the effects of SDF‐1α on the telomerase activity and telomere length. EPC subpopulation were isolated from peripheral blood and characterized. After ex vivo prolonged cultivation, EPC subpopulation became senescent as determined by acidic β‐galactosidase staining. SDF‐1α dose‐dependently inhibited the onset of EPC subpopulation senescence. Moreover, SDF‐1α increased proliferation and colony‐forming activity of EPC subpopulation. SDF‐1α also increased telomerase activity and telomere length, which was accompanied with upregulation of the catalytic subunit, telomerase reverse transcriptase (TERT). Whereas these effects of SDF‐1α on telomerase activity and expression of hTERT mRNA were significantly attenuated by CXCR4‐specific peptide antagonist (AMD3100) and phosphoinositide 3‐kinase (PI3K) inhibitor (LY294002). In conclusions, SDF‐1α delays the onset of EPC subpopulation senescence, which may be related to the activation of telomerase and elongation of telomere length. The inhibition of EPC subpopulation senescence and induction of EPC subpopulation proliferation by SDF‐1α in vitro may importantly improve the functional activity of EPC subpopulation for potential cell therapy. J. Cell. Physiol. 223:757–763, 2010. © 2010 Wiley‐Liss, Inc.
-
Thymosin Β4 Promotes Endothelial Progenitor Cell Angiogenesis Via a Vascular Endothelial Growth Factor‑dependent Mechanism.
Yanbo Zhao,Jiale Song,Xukun Bi,Jing Gao,Zhida Shen,Junhui Zhu,Guosheng Fu
DOI: https://doi.org/10.3892/mmr.2018.9199
IF: 3.423
2018-01-01
Molecular Medicine Reports
Abstract:Endothelial progenitor cells (EPCs) are a promising cell source for tissue repair and regeneration, predominantly through angiogenesis promotion. Paracrine functions serve a pivotal role in EPC‑mediated angiogenesis, which may be impaired by various cardiovascular risk factors. Therefore, it is important to identify a solution that optimizes the paracrine function of EPCs. Thymosin β4 (Tβ4) is a peptide with the potential to promote tissue regeneration and wound healing. A previous study demonstrated that Tβ4 enhances the EPC‑mediated angiogenesis of the ischemic myocardium. In the present study, whether Tβ4 improved angiogenesis by enhancing the paracrine effects of EPCs was investigated. A tube formation assay was used to assess the effect of angiogenesis, and the paracrine effects were measured using an ELISA kit. The results indicated that Tβ4 improved the paracrine effects of EPCs, evidenced by an increase in the expression of vascular endothelial growth factor (VEGF). EPC‑conditioned medium (EPC‑CM) significantly promoted human umbilical vein endothelial cell angiogenesis in vitro, which was further enhanced by pretreatment with Tβ4. The effect of Tβ4 pretreated EPC‑CM on angiogenesis was abolished by VEGF neutralizing antibody in vitro, indicating that increased VEGF secretion had a pivotal role in Tβ4‑mediated EPC angiogenesis. Furthermore, transplantation of EPCs pretreated with Tβ4 into infarcted rat hearts resulted in significantly higher VEGF expression in the border zone, compared with EPC transplantation alone. To further investigate whether the Akt/eNOS pathway was involved in Tβ4‑induced VEGF secretion in EPCs, the expression levels of VEGF in EPC‑CM were significantly decreased following knockdown of Akt or eNOS by small interfering RNA transfection. In conclusion, Tβ4 significantly increased angiogenesis by enhancing the paracrine effects of EPCs, evidenced by the increased expression of VEGF. The RAC‑α serine/threonine‑protein kinase/endothelial nitric oxide synthase signal transduction pathway was involved in the regulation of Tβ4‑induced VEGF secretion in EPCs. Further studies are required to investigate the long‑term prognosis of patients with coronary heart disease following Tβ4‑pretreated EPC transplantation.
-
Palmitic Acid Promotes Endothelial Progenitor Cells Apoptosis Via P38 and Jnk Mitogen-Activated Protein Kinase Pathways
Hailong Jiang,Chun Liang,Xing Liu,Qijun Jiang,Zhiqing He,Jianxiang Wu,Xiaoming Pan,Yusheng Ren,Min Fan,Mei Li,Zonggui Wu
DOI: https://doi.org/10.1016/j.atherosclerosis.2009.10.032
IF: 5.3
2010-01-01
Atherosclerosis
Abstract:Objective: Recent studies have demonstrated that palmitic acid (PA) could regulate endothelial progenitor cells (EPCs) function (migration, proliferation, survival and angiogenesis) via various signal pathways, but the effect of PA on EPCs apoptosis and associated mechanisms are still elusive.Methods: The human EPCs were obtained by Ficoll density gradient centrifugation and cultured in M199 medium containing rh-VEGF (30 ng/mL), rh-b-FGF (6 ng/mL) and 10% fetal bovine serum for 7 days. The adhesive EPCs were harvested, then challenged with different concentrations of PA (ranging from 0 to 800 mu mol/L) for 48 h and 400 mu mol/L PA for different time periods (ranging from 0 to 60 h) after 12 h synchronization with serum-free medium. The EPCs apoptosis was determined by flow cytometry, expression of caspase-3, phosphorylated ERK1/2, JNK and p38 mitogen-activated protein kinase (MAPK) were quantified by Western blot. The effect of PA on caspase-3 activity in the absence or presence of specific MAPK pathway inhibitors was determined by colorimetry.Results: PA increased EPCs apoptosis in a dose-and time-dependent manner, upregulated phosphorylated-p38 and -JNK, caspase-3 expression of EPCs while ERK expression was not affected. PA-induced EPCs apoptosis could be partly ameliorated by p38 inhibitor SB203580 and JNK inhibitor SP600125, but not by ERK1/2 inhibitor PD98059.Conclusion: These findings suggested that PA promoted EPCs apoptosis via p38 and JNK MAPKs pathways. (C) 2010 Published by Elsevier Ireland Ltd.
-
Caspase-8 Modulates Physiological and Pathological Angiogenesis During Retina Development.
Nathalie Tisch,Aida Freire-Valls,Rosario Yerbes,Isidora Paredes,Silvia La Porta,Xiaohong Wang,Rosa Martin-Perez,Laura Castro,Wendy Wei-Lynn Wong,Leigh Coultas,Boris Strilic,Hermann-Josef Grone,Thomas Hielscher,Carolin Mogler,Ralf H. Adams,Peter Heiduschka,Lena Claesson-Welsh,Massimiliano Mazzone,Abelardo Lopez-Rivas,Thomas Schmidt,Hellmut G. Augustin,Carmen Ruiz de Almodovar
DOI: https://doi.org/10.1172/jci122767
IF: 19.456
2019-01-01
Journal of Clinical Investigation
Abstract:During developmental angiogenesis, blood vessels grow and remodel to ultimately build a hierarchical vascular network. Whether, how, cell death signaling molecules contribute to blood vessel formation is still not well understood. Caspase-8 (Casp-8), a key protease in the extrinsic cell death-signaling pathway, regulates cell death via both apoptosis and necroptosis. Here, we show that expression of Casp-8 in endothelial cells (ECs) is required for proper postnatal retina angiogenesis. EC-specific Casp-8-KO pups (Casp-8ECKO) showed reduced retina angiogenesis, as the loss of Casp-8 reduced EC proliferation, sprouting, and migration independently of its cell death function. Instead, the loss of Casp-8 caused hyperactivation of p38 MAPK downstream of receptor-interacting serine/threonine protein kinase 3 (RIPK3) and destabilization of vascular endothelial cadherin (VE-cadherin) at EC junctions. In a mouse model of oxygen-induced retinopathy (OIR) resembling retinopathy of prematurity (ROP), loss of Casp-8 in ECs was beneficial, as pathological neovascularization was reduced in Casp-8ECKO pups. Taking these data together, we show that Casp-8 acts in a cell death-independent manner in ECs to regulate the formation of the retina vasculature and that Casp-8 in ECs is mechanistically involved in the pathophysiology of ROP.
-
SDF-1α/CXCR4 Decreases Endothelial Progenitor Cells Apoptosis under Serum Deprivation by PI3K/Akt/eNOS Pathway
Hao Zheng,Tao Dai,Binquan Zhou,Junhui Zhu,He Huang,Min Wang,Guosheng Fu
DOI: https://doi.org/10.1016/j.atherosclerosis.2008.02.011
IF: 5.3
2008-01-01
Atherosclerosis
Abstract:Recent studies have demonstrated that stromal cell-derived factor-1alpha (SDF-1alpha)/CXCR4 interaction regulates multiple cell signal pathways and a variety of cellular functions such as cell migration, proliferation, survival and angiogenesis. In present study, we aimed to determine the effect of SDF-1alpha on endothelial progenitor cells (EPCs) apoptosis induced by serum deprivation and the implication of phosphoinositide 3-kinase (PI3K)/Akt and mitogen-activated protein kinases (MAPKs) signaling in this effect. EPCs were isolated and characterized. SDF-1alpha decreased EPCs apoptosis induced by serum deprivation in a dose-dependent manner and the inhibitory effect was CXCR4 dependent as confirmed by the total abolishment by AMD3100, a CXCR4-specific peptide antagonist. SDF-1alpha treatment also significant decreased caspase-3 expression and activity. The inhibitory effect of SDF-1alpha on EPCs apoptosis was nearly completely abolished by PI3K inhibitors (either Wortmannin or LY294002) and partially abolished by NOS inhibitor, N(G)-nitro-arginine methyl ester, whereas inhibitors of MAPKs had no significant effect on this inhibitory effect. The treatment of EPCs with SDF-1alpha resulted in time-dependent Akt, eNOS, extracellular-regulated kinase (ERK1/2), p38 MAPK and c-Jun N-terminal kinase (JNK) phosphorylations. These findings suggest that PI3K/Akt/eNOS activation, but not MAPKs activation, is required for the inhibitory effect of SDF-1alpha on EPCs apoptosis.
-
Caspase-8 auto-cleavage regulates programmed cell death and collaborates with RIPK3/MLKL to prevent lymphopenia
Xiaoming Li,Fang Li,Xixi Zhang,Haiwei Zhang,Qun Zhao,Ming Li,Xiaoxia Wu,Lingxia Wang,Jianling Liu,Xuanhui Wu,Yangjing Ou,Mingyan Xing,Yue Zhang,Jiangshan Deng,Xiuzhe Wang,Yan Luo,Jinbao Li,Yuwu Zhao,Haibing Zhang
DOI: https://doi.org/10.1038/s41418-022-00938-9
IF: 12.067
2022-01-21
Cell Death and Differentiation
Abstract:Abstract Caspase-8 is an initiator of death receptor-induced apoptosis and an inhibitor of RIPK3-MLKL-dependent necroptosis. In addition, caspase-8 has been implicated in diseases such as lymphoproliferation, immunodeficiency, and autoimmunity in humans. Although auto-cleavage is indispensable for caspase-8 activation, its physiological functions remain poorly understood. Here, we generated a caspase-8 mutant lacking E385 in auto-cleavage site knock-in mouse ( Casp8 ΔE385/ΔE385 ). Casp8 ΔE385/ΔE385 cells were expectedly resistant to Fas-induced apoptosis, however, Casp8 Δ E385/ΔE385 cells could switch TNF-α-induced apoptosis to necroptosis by attenuating RIPK1 cleavage. More importantly, CASP8(ΔE385) sensitized cells to RIPK3-MLKL-dependent necroptosis through promoting complex II formation and RIPK1-RIPK3 activation. Notably, Casp8 Δ E385/ΔE385 Ripk3 −/− mice partially rescued the perinatal death of Ripk1 −/− mice by blocking apoptosis and necroptosis. In contrast to the Casp8 −/− Ripk3 −/− and Casp8 −/− Mlkl −/− mice appearing autoimmune lymphoproliferative syndrome (ALPS), both Casp8 Δ E385/ΔE385 Ripk3 −/− and Casp8 Δ E385/ΔE385 Mlkl −/− mice developed transplantable lymphopenia that could be significantly reversed by RIPK1 heterozygosity, but not by RIPK1 kinase dead mutation. Collectively, these results demonstrate previously unappreciated roles for caspase-8 auto-cleavage in regulating necroptosis and maintaining lymphocytes homeostasis.
cell biology,biochemistry & molecular biology
-
Combination of Stromal-Derived Factor-1Α and Vascular Endothelial Growth Factor Gene-Modified Endothelial Progenitor Cells is More Effective for Ischemic Neovascularization
Jian-Xing Yu,Xue-Fei Huang,Wei-Ming Lv,Cai-Sheng Ye,Xin-Zhi Peng,Hui Zhang,Long-Bin Xiao,Shen-Ming Wang
DOI: https://doi.org/10.1016/j.jvs.2009.05.049
IF: 4.86
2009-01-01
Journal of Vascular Surgery
Abstract:Background: Recruitment and entrapment of bone marrow-derived endothelial progenitor cells (EPCs) is important in vascular endothelial growth factor (V-EGF)-induced angiogenesis. EPC mobilization and differentiation are modulated by stromal-derived factor-1 alpha (SDF-1 alpha/CXCL12), another important chemokine. In this study, we investigated the hypothesis that SDF-1 alpha and VEGF might act synergistically on EPC-mediated vasculogenesis.Methods: EPCs were isolated and cultured from human peripheral blood, then transduced with retroviral vectors pBabe containing human VEGF(165) complimentary DNA (Td/V-EPCs) and pBabe wild-type (Td/p-EPCs). EPC migration activity was investigated with a modified Boyden chamber assay. EPC apoptosis induced by serum starvation was studied by annexin V assays. The combined effect of local administration of SDF-1 alpha and Td/V-EPC transplantation on neovascularization was investigated in a murine model of hind limb ischemia.Results. Over-expression of hV-EGF(165) increased SDF-1 alpha-mediated EPC migration. SDF-1 alpha-mediated migration was significantly increased when EPCs were modified with VEGF (Td/V-EPCs) vs when VEGF was not present (Td/p-EPCs) or when VEGF alone was present (Td/V-EPCs; 196.8 +/- 15.2, 81.2 +/- 9.8, and 67.4 +/- 7.4/mm(2), respectively P < .001). SDF-1 alpha combined with VEGF reduced serum starvation-induced apoptosis of EPCs more than SDF-1 alpha or VEGF alone (P < .001). To determine the effect of this combination in vivo, SDF-1 alpha was locally injected alone into the ischemic hind limb muscle of nude mice or combined with systemically injected Td/V-EPCs. The SDF-1 alpha plus VEGF group showed significantly increased local accumulation of EPCs, blood-flow recovery, and capillary density compared with the other groups. The ratio of ischemic/normal blood flow in Td/V-EPCs plus SDF-1 alpha group was significantly higher (P < .01), as was capillary density (capillaries/mm(2)), an index of neovascularization (Td/V-EPCs plus SDF-1 alpha group, 863 31; no treatment, 395 +/- 13; SDF-1 alpha, 520 +/- 29; Td/p-EPCs, 448 +/- 28; Td/p-EPCs plus SDF-1 alpha., 620 +/- 29; Td/V-EPCs, 570 +/- 30; P < .01). To investigate a possible mechanistic basis, we showed that VEGF up-regulated the receptor for SDF-1 alpha, CXCR4, on EM in Nitro.Conclusion: The combination of SDF-1 alpha and VEGF greatly increases EPC-mediated angiogenesis. The use VEGF and SDF-1 alpha together, rather than alone, will be a novel and efficient angiogenesis strategy to provide therapeutic neovascularization. (J Vase Surg 2009;50:608-16.)
-
Caspase-8 is the molecular switch for apoptosis, necroptosis and pyroptosis
Melanie Fritsch,Saskia D. Günther,Robin Schwarzer,Marie-Christine Albert,Fabian Schorn,J. Paul Werthenbach,Lars M. Schiffmann,Neil Stair,Hannah Stocks,Jens M. Seeger,Mohamed Lamkanfi,Martin Krönke,Manolis Pasparakis,Hamid Kashkar
DOI: https://doi.org/10.1038/s41586-019-1770-6
IF: 64.8
2019-11-20
Nature
Abstract:Caspase-8 is the initiator caspase of extrinsic apoptosis<sup><a href="/articles/s41586-019-1770-6#ref-CR1">1</a>,<a href="/articles/s41586-019-1770-6#ref-CR2">2</a></sup> and inhibits necroptosis mediated by RIPK3 and MLKL. Accordingly, caspase-8 deficiency in mice causes embryonic lethality<sup><a href="/articles/s41586-019-1770-6#ref-CR3">3</a></sup>, which can be rescued by deletion of either <i>Ripk3</i> or <i>Mlkl</i><sup><a href="#ref-CR4">4</a>,<a href="#ref-CR5">5</a>,<a href="/articles/s41586-019-1770-6#ref-CR6">6</a></sup>. Here we show that the expression of enzymatically inactive CASP8(C362S) causes embryonic lethality in mice by inducing necroptosis and pyroptosis. Similar to <i>Casp8</i><sup><i>−/−</i></sup> mice<sup><a href="/articles/s41586-019-1770-6#ref-CR3">3</a>,<a href="/articles/s41586-019-1770-6#ref-CR7">7</a></sup>, <i>Casp8</i><sup><i>C362S/C362S</i></sup> mouse embryos died after endothelial cell necroptosis leading to cardiovascular defects. MLKL deficiency rescued the cardiovascular phenotype but unexpectedly caused perinatal lethality in <i>Casp8</i><sup><i>C362S/C362S</i></sup> mice, indicating that CASP8(C362S) causes necroptosis-independent death at later stages of embryonic development. Specific loss of the catalytic activity of caspase-8 in intestinal epithelial cells induced intestinal inflammation similar to intestinal epithelial cell-specific <i>Casp8</i> knockout mice<sup><a href="/articles/s41586-019-1770-6#ref-CR8">8</a></sup>. Inhibition of necroptosis by additional deletion of <i>Mlkl</i> severely aggravated intestinal inflammation and caused premature lethality in <i>Mlkl</i> knockout mice with specific loss of caspase-8 catalytic activity in intestinal epithelial cells. Expression of CASP8(C362S) triggered the formation of ASC specks, activation of caspase-1 and secretion of IL-1β. Both embryonic lethality and premature death were completely rescued in <i>Casp8</i><sup><i>C362S/C362S</i></sup><i>Mlkl</i><sup><i>−/−</i></sup><i>Asc</i><sup><i>−/−</i></sup> or <i>Casp8</i><sup><i>C362S/C362S</i></sup><i>Mlkl</i><sup><i>−/−</i></sup><i>Casp1</i><sup><i>−/−</i></sup> mice, indicating that the activation of the inflammasome promotes CASP8(C362S)-mediated tissue pathology when necroptosis is blocked. Therefore, caspase-8 represents the molecular switch that controls apoptosis, necroptosis and pyroptosis, and prevents tissue damage during embryonic development and adulthood.
multidisciplinary sciences
-
Inhibition of caspase-8 cascade restrains the osteoclastogenic fate of bone marrow cells
Barbora Veselá,Adéla Ševčíková,Kateřina Holomková,Alice Ramešová,Adéla Kratochvílova,Paul T Sharpe,Eva Matalová
DOI: https://doi.org/10.1007/s00424-024-02977-2
Abstract:Osteoclasts are multinucleated cells of hematopoietic origin, with a pivotal role in bone development and remodeling. Failure in osteoclast differentiation and activation leads to various bone disorders; thus, attention has focused on a search of molecules involved in osteoclast regulatory pathways. Caspase-8 appears to be an interesting candidate for further exploration, due to its potential function in bone development and homeostasis. Mouse bone marrow cells were differentiated into osteoclasts by RANKL stimulation. Increased activation of caspase-8 and its downstream executioner caspases (caspase-3 and caspase-6) was found during osteoclastogenesis. Subsequent inhibition of caspase-8, caspase-3, or caspase-6, respectively, during osteoclast differentiation showed distinct changes in the formation of TRAP-positive multinucleated cells and reduced expression of osteoclast markers including Acp5, Ctsk, Dcstamp, and Mmp9. Analysis of bone matrix resorption confirmed significantly reduced osteoclast function after caspase inhibition. The results clearly showed the role of caspases in the proper development of osteoclasts and contributed new knowledge about non-apoptotic function of caspases.
-
Phosphorylation of caspase-8 by RSKs via organ-constrained effects controls the sensitivity to TNF-induced death
Peng He,Tingting Ai,Muzhen Qiao,Zhang-Hua Yang,Jiahuai Han
DOI: https://doi.org/10.1038/s41420-024-02024-0
2024-05-25
Cell Death Discovery
Abstract:Caspase-8 (Casp8) serves as an initiator of apoptosis or a suppressor of necroptosis in context-dependent manner. Members of the p90 RSK family can phosphorylate caspase-8 at threonine-265 (T265), which can inactivate caspase-8 for bypassing caspase-8-mediated blockade of necroptosis and can also decrease caspase-8 level by promoting its degradation. Mutating T265 in caspase-8 to alanine (A) in mice blocked TNF-induced necroptotic cecum damage but resulted in unexpectedly massive injury in the small intestine. Here, we show RSK1, RSK2, and RSK3 redundantly function in caspase-8 phosphorylation, and the duodenum is the most severely affected part of the small intestine when T265 phosphorylation of caspase-8 was prevented. Eliminating caspase-8 phosphorylation resulted in a duodenum-specific increase in basal caspase-8 protein level, which shall be responsible for the increased sensitivity to TNF-induced damage. Apoptosis of intestinal epithelial cells (IECs) was predominant in the duodenum of TNF-treated Rsk1 − / − Rsk2 − / − Rsk3 − / − and Casp8 T265A/T265A mice, though necroptosis was also observed. The heightened duodenal injury amplified systemic inflammatory responses, as evidenced by the contribution of hematopoietic cells to the sensitization of TNF-induced animal death. Further analysis revealed that hematopoietic and non-hematopoietic cells contributed differentially to cytokine production in response to the increased cell death. Collectively, RSKs emerges as a previously overlooked regulator that, via tissue/organ-constrained inactivating caspase-8 and/or downregulating caspase-8 protein level, controls the sensitivity to TNF-induced organ injury and animal death.
cell biology
-
Caspase-8 Mediates Amyloid-β-induced Apoptosis in Differentiated PC12 Cells
Min-cai Qian,Jing Liu,Jia-shu Yao,Wei-min Wang,Jian-hong Yang,Li-li Wei,Yue-di Shen,Wei Chen
DOI: https://doi.org/10.1007/s12031-015-0498-5
2015-01-01
Abstract:The pathogenesis of Alzheimer’s disease (AD) is very complex and there are currently no significant treatments for the disease. Caspase-8 is known to be involved in neuronal apoptosis. To explore a possible molecular mechanisms involved in AD pathology, this study investigated the effect of caspase-8 knockdown on amyloid-β 1–40 (Aβ1–40)-induced apoptosis in PC12 cells. The proliferation of PC12 cells was significantly inhibited in Aβ-treated cells, and a high fraction of the cells underwent apoptosis in a dose- and time-dependent manner. Transfection of caspase-8 small interfering RNA (siRNA) resulted in reduced apoptosis following Aβ1–40 treatment. The activation of caspase-3, caspase-8, and caspase-9 was stimulated by Aβ1–40, an effect that was also significantly reduced by caspase-8 siRNA. Knockdown of caspase-8 increased the phosphorylation of the signaling molecules AKT and ERK1/2 relative to cells treated with Aβ1–40 alone. Caspase-8 is an important effector molecule involved in apoptosis induced by Aβ1–40 and is likely involved in AD pathology. This study suggests that targeted inhibition of caspase-8 may be a new therapeutic for preventing neuronal apoptosis and inhibiting the progression of AD.
-
Caspase-8 regulates the expression of pro- and anti-inflammatory cytokines in human bone marrow-derived mesenchymal stromal cells
Siv H Moen,Marita Westhrin,Muhammad Zahoor,Nikolai N Nørgaard,Hanne Hella,Berit Størdal,Anders Sundan,Nadra J Nilsen,Anne-Marit Sponaas,Therese Standal
DOI: https://doi.org/10.1002/iid3.117
2016-07-21
Abstract:Introduction: Mesenchymal stem cells, also called mesenchymal stromal cells, MSCs, have great potential in stem cell therapy partly due to their immunosuppressive properties. How these cells respond to chronic inflammatory stimuli is therefore of importance. Toll-like receptors (TLR)s are innate immune receptors that mediate inflammatory signals in response to infection, stress, and damage. Caspase-8 is involved in activation of NF-kB downstream of TLRs in immune cells. Here we investigated the role of caspase-8 in regulating TLR-induced cytokine production from human bone marrow-derived mesenchymal stromal cells (hBMSCs). Methods: Cytokine expression in hBMCs in response to poly(I:C) and LPS was evaluated by PCR, multiplex cytokine assay, and ELISA. TLR3, TRIF, and caspase-8 were silenced using siRNA. Caspase-8 was also inhibited using a caspase-8 inhibitor, z-IEDT. Results: We found that TLR3 agonist poly(I:C) and TLR4 agonist LPS induced secretion of several pro-inflammatory cytokines in a TLR-dependent manner which required the TLR signaling adaptor molecule TRIF. Further, poly(I:C) reduced the expression of anti-inflammatory cytokines HGF and TGFβ whereas LPS reduced HGF expression only. Notably, caspase-8 was involved in the induction of IL- IL-1β, IL-6, CXCL10, and in the inhibition of HGF and TGFβ. Conclusion: Caspase-8 appears to modulate hBMSCs into gaining a pro-inflammatory phenotype. Therefore, inhibiting caspase-8 in hBMSCs might promote an immunosuppressive phenotype which could be useful in clinical applications to treat inflammatory disorders.
-
Caspase-8 contributes to an immuno-hot microenvironment by promoting phagocytosis via an ecto-calreticulin-dependent mechanism
Zhihua Gong,Qingzhu Jia,Jinming Guo,Chongyi Li,Shouxia Xu,Zheng Jin,Han Chu,Yisong Y Wan,Bo Zhu,Yi Zhou
DOI: https://doi.org/10.1186/s40164-022-00371-1
2023-01-12
Abstract:Background: Caspase-8 (Casp8) acts as an initiator in cell apoptosis signaling. However, the role of Casp8 in tuning the tumor immune microenvironment remains controversial due to the complicated crosstalk between immune-tolerogenic apoptotic cell death and immunogenic cell death cascades. Methods: The Cancer Genome Atlas (TCGA) and publicly accessible immune checkpoint blockade (ICB)-treated cohorts were used to investigate the clinical relevance of Casp8. A tumor-bearing mouse model was used to characterize changes in the tumor microenvironment and to explore the efficacy of ICB treatment under Casp8 knockout conditions. Results: By exploring TCGA datasets, we showed that the expression level of Casp8 was associated with an immuno-hot microenvironment across various solid tumor types. Casp8 deficiency leads to decreased CD8+ T cell infiltration and resistance to anti-PD-L1 therapy in a mouse model. Mechanistically, Casp8 deficiency or pharmacological disruption results in impaired ecto-calreticulin transition in tumor cells, which in turn hampers antigen presentation in draining lymph nodes. Furthermore, radiotherapy restored sensitivity to anti-PD-L1 treatment via elevated calreticulin surface expression. Conclusions: Our data revealed a causative role of Casp8 in modulating the immunogenicity of tumor cells and responsiveness to ICB immunotherapies and proposed radiotherapy as a salvage approach to overcome Casp8 deficiency-mediated ICB resistance.
-
Caspase-8 has dual roles in regulatory T cell homeostasis balancing immunity to infection and collateral inflammatory damage
James Cooney,Liana Mackiewicz,Philip Arandjelovic,Gregor Ebert,Marcel Doerflinger,Tania Tan,Axel Kallies,Andreas Strasser,Marc Pellegrini,Charis E. Teh,Simon P. Preston,Alissa K. Robbins,Michael D. Stutz,Michelle P. Clark,Antonia N. Policheni,Cody C. Allison,Lucille C. Rankin,Peggy P. Teh,Gabrielle T. Belz,Daniel H. D. Gray
DOI: https://doi.org/10.1126/sciimmunol.abn8041
IF: 30.63
2022-03-25
Science Immunology
Abstract:Targeting the potent immunosuppressive properties of FOXP3 + regulatory T cells (T regs ) has substantial therapeutic potential for treating autoimmune and inflammatory diseases. Yet, the molecular mechanisms controlling T reg homeostasis, particularly during inflammation, remain unclear. We report that caspase-8 is a central regulator of T reg homeostasis in a context-specific manner that is decisive during immune responses. In mouse genetic models, targeting caspase-8 in T regs led to accumulation of effector T regs resistant to apoptotic cell death. Conversely, inflammation induced the MLKL-dependent necroptosis of caspase-8–deficient lymphoid and tissue T regs , which enhanced immunity to a variety of chronic infections to promote clearance of viral or parasitic pathogens. However, improved immunity came at the risk of lethal inflammation in overwhelming infections. Caspase-8 inhibition using a clinical-stage compound revealed that human T regs have heightened sensitivity to necroptosis compared with conventional T cells. These findings reveal a fundamental mechanism in T regs that could be targeted to manipulate the balance between immune tolerance versus response for therapeutic benefit.
immunology
-
Caspase-8 restricts antiviral CD8 T cell hyperaccumulation
Yanjun Feng,Lisa P Daley-Bauer,Linda Roback,Hongyan Guo,Heather S Koehler,Marc Potempa,Lewis L Lanier,Edward S Mocarski
DOI: https://doi.org/10.1073/pnas.1904319116
2019-07-23
Abstract:The magnitude of CD8 T cell responses against viruses is checked by the balance of proliferation and death. Caspase-8 (CASP8) has the potential to influence response characteristics through initiation of apoptosis, suppression of necroptosis, and modulation of cell death-independent signal transduction. Mice deficient in CASP8 and RIPK3 (Casp8-/-Ripk3-/- ) mount enhanced peak CD8 T cell levels against the natural mouse pathogen murine cytomegalovirus (MCMV) or the human pathogen herpes simplex virus-1 compared with littermate control RIPK3-deficient or WT C57BL/6 mice, suggesting an impact of CASP8 on the magnitude of antiviral CD8 T cell expansion and not on contraction. The higher peak response to MCMV in Casp8-/-Ripk3-/- mice resulted from accumulation of greater numbers of terminally differentiated KLRG1hi effector CD8 T cell subsets. Antiviral Casp8-/-Ripk3-/- T cells exhibited enhanced proliferation when splenocytes were transferred into WT recipient mice. Thus, cell-autonomous CASP8 normally restricts CD8 T cell proliferation following T cell receptor activation in response to foreign antigen. Memory inflation is a hallmark quality of the T cell response to cytomegalovirus infection. Surprisingly, MCMV-specific memory inflation was not sustained long-term in Casp8-/-Ripk3-/- mice even though these mice retained immunity to secondary challenge. In addition, the accumulation of abnormal B220+CD3+ T cells in these viable CASP8-deficient mice was reduced by chronic MCMV infection. Combined, these data brings to light the cell death-independent role of CASP8 during CD8 T cell expansion in mice lacking the confounding impact of RIPK3-mediated necroptosis.
-
Functional Role of Matrix Metalloproteinase-8 in Stem/Progenitor Cell Migration and Their Recruitment into Atherosclerotic Lesions
Qingzhong Xiao,Feng Zhang,Luyang Lin,Changcun Fang,Guanmei Wen,Tsung-Neng Tsai,Xiangyuan Pu,David Sims,Zhongyi Zhang,Binia Thomaszewski,Boris Schmidt,Manuel Mayr,Ken Suzuki,Qingbo Xu,Shu Ye
DOI: https://doi.org/10.1161/circresaha.112.274019
IF: 23.213
2013-01-01
Circulation Research
Abstract:Rationale: Accumulating evidence indicates that stem/progenitor cells (SPCs) represent an important source of cells in atheromas and contribute to lesion formation and progression. Objective: We investigated whether matrix metalloproteinase-8 (MMP8) played a role in SPC migration and their recruitment into atheromas. Methods and Results: We found that SPCs in atheromas expressed MMP8 and that MMP8 knockout significantly reduced SPC numbers in atherosclerotic lesions in apolipoprotein E (ApoE)–deficient mice fed a Western diet. Further in vivo experiments showed that ApoE−/−/MMP8−/− mice injected with stem cells isolated from bone marrows of ApoE−/−/MMP8−/− mice had fewer SPCs in atheromas and smaller lesions than ApoE−/−/MMP8−/− mice injected with stem cells isolated from bone marrows of ApoE−/−/MMP8+/+ mice. Ex vivo experiments showed that MMP8 deficiency inhibited the ability of SPCs to migrate from the arterial lumen and the adventitia into atherosclerotic lesions. In vitro assays indicated that MMP8 facilitated SPC migration across endothelial cells and through Matrigel or collagen I. We also found that MMP8 cleaved a-disintegrin-and-metalloproteinase-domain-10 and that MMP8 deficiency reduced mature a-disintegrin-and-metalloproteinase-domain-10 on SPCs. Knockdown of MMP8 or incubation with the a-disintegrin-and-metalloproteinase-domain-10 inhibitor GI254023X decreased E-cadherin shedding on SPCs. The decrease in migratory ability of SPCs with MMP8 knockdown was reduced by incubation of such cells with culture supernatant from SPCs without MMP8 knockdown, and this compensatory effect was abolished by an antibody against soluble E-cadherin. Conclusions: MMP8 plays an important role in SPC migration and their recruitment into atherosclerotic lesions.
-
Caspase-8 deficiency in mouse embryos triggers chronic RIPK1-dependent activation of inflammatory genes, independently of RIPK3
Tae-Bong Kang,Ju-Seong Jeong,Seung-Hoon Yang,Andrew Kovalenko,David Wallach
DOI: https://doi.org/10.1038/s41418-018-0104-9
IF: 12.067
2018-04-17
Cell Death and Differentiation
Abstract:Deletion of the Casp8 gene in epithelial tissues of mice results in severe inflammatory pathologies. Its ubiquitous deletion, or its specific deletion in endothelial cells, results in intrauterine death associated with capillary damage. These pathologies are all preventable by co-deletion of Casp8 and the genes encoding either the RIPK1 or the RIPK3 protein kinase. Since activation of RIPK3 in Caspase-8-deficient cells can trigger necroptotic cell death, and since RIPK1 can activate RIPK3, it is widely assumed that the inflammatory states resulting from Caspase-8 deficiency occur as a consequence of RIPK3-induced necroptosis. Here, we report that although on a Ripk3-null background Casp8 deletion in mice does not result in outright pathological changes, it triggers enhanced expression of a variety of inflammatory genes in utero, which gradually subsides after birth. Deletion of Ripk1, or even of only one of its two alleles, obliterates this activation. Resembling the embryonic pathology observed in RIPK3-expressing cells, the activation of inflammatory genes observed on a Ripk3-null background seems to be initiated in endothelial cells. Analysis of endothelial cells isolated from livers of Caspase-8-deficient embryos revealed neither an increase in the amount of RIPK1 in these cells after Casp8 deletion, nor triggering of RIPK1 phosphorylation. These findings indicate that the triggering of inflammation by Casp8 deletion in mice occurs, in part, independently of necroptosis or other functions of RIPK3, and rather reflects enhanced RIPK1-dependent signaling for activation of inflammatory genes.
cell biology,biochemistry & molecular biology
-
BAS/BSCR43 X-box Binding Protein 1 Splicing is Crucial in Endothelial Cell Proliferation
L. Zeng,Q. Xiao,H. Li,A. Margariti,Y. Hu,Q. Xu
DOI: https://doi.org/10.1136/hrt.2010.205781.54
IF: 5.7
2010-01-01
Heart
Abstract:The X-box binding protein 1 (XBP1) is an endoplasmic reticulum stress response transcription factor. Our previous study showed that sustained activation of XBP1 splicing led to atherosclerosis development (Zeng et al. PNAS 2009). However, the function of XBP1 expression and splicing in endothelial cells and angiogenesis remains unclear. To study this issue, we generated XBP1 knockout mice by deletion of exons 1 and 2 of the gene. XBP1-null (XBP1−/−) embryos at E12.5 showed growth retardation and pale colouration phenotype. The average body weight of XBP1−/−embryos was 40% less than that of wild-type (XBP1+/+) animals. The blood vessel density in XBP1−/− embryos was significantly reduced, owing to a smaller number of CD31+ and Flk1+ cells. In in vitro culture of whole embryonic cells, XBP1−/− cells grew significantly more slowly and lost response to VEGF stimulation. To study the mechanism of XBP1-mediated cell growth, human umbilical vein endothelial cells were treated with VEGF that transiently activated IRE1α phosphorylation at Ser724. The mRNA movement inhibitor, cycloheximide, ablated VEGF-induced IRE1α phosphorylation and XBP1 splicing. Co-immunoprecipitation assay showed that there was interaction among KDR, IRE1α and unspliced XBP1 (XBP1u), which could be increased by VEGF treatment. Further experiments demonstrated that the C-terminal region of KDR and the kinase domain of IRE1α are responsible for their interactions. MTT and BrdU incorporation assays indicate that transient activation of XBP1 splicing increased while long-term activation decreased endothelial survival and proliferation. Knockdown of XBP1 or IRE1α ablated VEGF-induced proliferation in endothelial cells. Immunofluorescent staining and TOP Flash reporter assay showed that overexpression of XBP1s increased β-catenin translocation into the nucleus. Thus, this study demonstrated for the first time that XBP1 is crucial for endothelial growth and angiogenesis, in which VEGF-stimulated IRE1α/XBP1 splicing system and interaction with β-catenin are key elements, indicating a potential target of XBP1 for protecting endothelial integrity.