A matter of regeneration and repair: caspases as the key molecules

A matter of regeneration and repair: caspases as the key molecules

Researchers have been focused on understanding the pathogenesis of human diseases. They have been working to find the roles of caspases in the balance between apoptosis, autophagy, pyroptosis, and necroptosis, and also in the regeneration of damaged tissue. At this point, besides their death-inducing roles, new findings indicate the role of caspases in proliferation for maintaining the viability of cells in response to signals from apoptotic cells. Recently, determining cell fate has also been identified among other functions of caspases. Findings indicate that caspases direct cellular pathways to cell differentiation by suppressing stem cell self-renewal. The common opinion about the related mechanism is that low and transient caspase activation leads to terminal differentiation by affecting the expression of key genes related to differentiation. Moreover, caspases are essential proteases in the regulation and modulation of the repair process. In repair, they have roles in apoptosis, release of inflammatory cytokines and chemokines, promotion of cell migration, and immune cell infiltration. However, paracrine signaling through caspase activation leads to cell proliferation after cancer therapy and causes tumor relapse, which complicates the current therapy. In this scope, we have reviewed the main mechanisms of pathological, regenerative, and restorative effects of caspases.

___

  • Aachoui Y, Sagulenko V, Miao EV, Stacey KJ (2013). Inflammasome mediated pyroptotic and apoptotic cell death and defense against infection. Curr Opin Microbiol 16: 319–329.
  • Abdul-Ghani M, Megeney LA (2008). Rehabilitation of a contract killer: caspase-3 directs stem cell differentiation. Cell Stem Cell 2: 515–516.
  • Adamson YR, Bowden DH (1974). The type 2 cell as progenitor of alveolar epithelial regeneration. A cytodynamic study in mice after exposition to oxygen. Lab Invest 30: 35–42.
  • Ailles LE, Weissman IL (2007). Cancer stem cells in solid tumors. Curr Opin Biotechnol 18: 460–466.
  • Alt EU, Senst C, Murthy SN, Slakey DP, Dupin CL, Chaffin AE, Kadowitz PJ, Izadpanah R (2012). Aging alters tissue resident mesenchymal stem cell properties. Stem Cell Res 8: 215–225.
  • Antoniou KM, Pataka A, Bouros D, Siafakas NM. Pathogenetic pathways and novel pharmacotherapeutic targets in idiopathic pulmonary fibrosis (2007). Pulm Pharmacol Ther 20: 453–461.
  • Arama E, Bader M, Rieckhof GE, Steller H (2007). A ubiquitin ligase complex regulates caspase activation during sperm differentiation in Drosophila. PLoS Biol 5: e251.
  • Artlett CM, Sassi-Gaha S, Rieger JL, Boesteanu AC, Feghali-Bostwick CA, Katsikis PD (2011). The inflammasome activating caspase 1 mediates fibrosis and myofibroblast differentiation in systemic sclerosis. Arthritis Rheum 63: 3563–3574.
  • Bagnato GL, Roberts WN, Fiorenza A, Arcuri C, Certo R, Trimarchi F, Ruggeri RM, Bagnato GF (2015). Skin fibrosis correlates with circulating thyrotropin levels in systemic sclerosis: translational association with Hashimoto’s thyroiditis (in press).
  • Barbero S, Mielgo A, Torres V, Teitz T, Shields DJ, Mikolon D, Bogyo M, Barilà D, Lahti JM, Schlaepfer D et al. (2009). Caspase-8 association with the focal adhesion complex promotes tumor cell migration and metastasis. Cancer Res 69: 3755–3763.
  • Bataller R, Brenner DA (2005). Liver fibrosis. J Clin Invest 115: 209– 218.
  • Beisner DR, Ch’en IL, Kolla RV, Hoffmann A, Hedrick SM (2005). Cutting edge: innate immunity conferred by B cells is regulated by caspase-8. J Immunol 175: 3469–3473.
  • Berghe V, Linkermann T Jouan-Lanhauet A, Walczak S, Vandenabeele P (2014). Regulated necrosis the expanding network of non-apoptotic cell death pathways. Nat Rev Mol Cell Biol 15: 135–147.
  • Bergmann, A, Steller H (2010). Apoptosis, stem cells, and tissue regeneration. Sci Signal 3: 8.
  • Bergsbaken T, Fink SL, Cookson BT (2009). Pyroptosis: host cell death and inflammation. Nat Rev Microbiol 7: 99–109.
  • Boatright KM, Deis C, Denault JB, Sutherlin DP, Salvesen GS (2004). Activation of caspases-8 and -10 by FLIP(L). Biochem J 382: 651–657.
  • Boland K, Flanagon L, Prehnt JHM (2013). Paracrine control of tissue regeneration and cell proliferation by caspase-3. Cell Death Dis 4: e725.
  • Bonner C, Bacon S, Concannon CG, Rizvi SR, Baquié M, Farrelly AM, Kilbride SM, Dussmann H, Ward MW, Boulanger CM et al. (2010). INS-1 cells undergoing caspase-dependent apoptosis enhance the regenerative capacity of neighboring cells. Diabetes 59: 2799–2808.
  • Bosch M, Serras F, Martín-Blanco E, Baguñà J (2005). JNK signaling pathway required for wound healing in regenerating Drosophila wing imaginal discs. Dev Biol 280: 73–86.
  • Brentnall M, Weir DB, Rongvaux A, Marcus AI, Boise LH (2014). Procaspase-3 regulates fibronectin secretion and influences adhesion, migration and survival independently of catalytic function. J Cell Sci 127: 2217–2226.
  • Can A (2014). Kök Hücre. Biyolojisi, Türleri ve Tedavide Kullanımları. 1st ed. Ankara, Turkey: Akademisyen Tıp Kitapevi (in Turkish).
  • Castellone MD, Teramoto H, Williams BO, Druey KM, Gutkind JS (2005). Prostaglandin E2 promotes colon cancer cell growth through a Gs-axin-beta-catenin signaling axis. Science 310: 1504–1510.
  • Chang DW, Xing Z, Pan Y, Algeciras-Schimnich A, Barnhart BC, Yaish-Ohad S, Peter ME, Yang X (2002). c- FLIP(L) is a dual function regulator for caspase-8 activation and CD95- mediated apoptosis. EMBO J 21: 3704–3714.
  • Cheng J, Tian L, Ma J, Gong Y, Zhang Z, Chen Z, Xu B, Xiong H, Li C, Huang Q (2015). Dying tumor cells stimulate proliferation of living tumor cells via caspase-dependent protein kinase Cδ activation in pancreatic ductal adenocarcinoma. Mol Oncol 9: 105–114.
  • Chera S, Ghila L, Dobretz K, Wenger Y, Bauer C, Buzgariu W, Martinou JC, Galliot B (2009). Apoptotic cells provide an unexpected source of Wnt3 signaling to drive Hydra head regeneration. Dev Cell 17: 279–289.
  • Chun HJ, Zheng L, Ahmad M, Wang J, Speirs CK, Siegel RM, Dale JK, Puck J, Davis J, Hall CG et al (2002). Pleiotropic defects in lymphocyte activation caused by caspase-8 mutations lead to human immunodeficiency. Nature 419: 395–399.
  • Connolly PF, Jäger R, Farnhead HO (2014). New roles for old enzymes: killer caspases as the engine of cell behavior changes. Front Physiol 5: 1–8.
  • Conus S, Perozzo R, Reinheckel T, Peters C, Scapozza L, Yousefi S, Simon HU (2008). Caspase-8 is activated by cathepsin D initiating neutrophil apoptosis during the resolution of inflammation. J Exp Med 205: 685–698.
  • Cullen SP, Henry CM, Kearney CJ, Logue SE, Feoktistova M, Tynan GA, Lavelle EC, Leverkus M, Martin SJ (2013). Fas/ CD95-induced chemokines can serve as “find-me” signals for apoptotic cells. Mol Cell 49: 1034–1048.
  • D’Amelio M, Cavallucci V, Cecconi F (2010). Neuronal caspase-3 signaling: not only cell death. Cell Death Differ 17: 1104–1114.
  • Darby IA, Bisucci T, Pittet B, Garbin S, Gabbiani G, Desmouliere A (2002). Skin flap-induced regression of granulation tissue correlates with reduced growth factor and increased metalloproteinase expression. J Pathol 197: 117–127.
  • Delavallée L, Cabon L, Galán-Malo P, Lorenzo HK, Susin SA (2011). AIF-mediated caspase-independent necroptosis: a new chance for targeted therapeutics. Life 63: 221–232.
  • Denes A, Lopez-Castejon G, Brough D (2012). Caspase-1: is IL-1 just the tip of the ICEberg? Cell Death Dis 3: e338.
  • Dohrman A, Kataoka T, Cuenin S, Russell JQ, Tschopp J, Budd RC (2005). Cellular FLIP (long form) regulates CD8+ T cell activation through caspase-8-dependent NF-kappa B activation. J Immunol 174: 5270–5278.
  • Donato AL, Huang Q, Liu X, Li F, Zimmerman MA, Li CY (2014). Caspase 3 promotes surviving melanoma tumor cell growth after cytotoxic therapy. J Invest Dermatol 134: 1686–1692.
  • Duffield JS, Lupher M, Thannickal VJ, Wynn TA (2013). Host responses in issue repair and fibrosis. Annu Rev Pathol 8: 241–276.
  • Ellis HM, Horvitz HR (1986). Genetic control of programmed cell death in the nematode C. elegans. Cell. 44: 817–829.
  • Eming SA, Martin P, Tomic-Canic M (2014). Wound repair and regeneration: mechanisms, signaling, and translation. Sci Transl Med 6: 265sr6.
  • Fan W, Dai Y, Xu H, Zhu X, Cai P, Wang L, Sun C, Hu C, Zheng P, Zhao BQ (2014). Caspase-3 modulates regenerative response after stroke. Stem Cells 32: 473–486.
  • Fan Y, Bergmann A (2008a). Apoptosis-induced compensatory proliferation. The Cell is dead. Long live the Cell! Trends Cell Biol 18: 467–473.
  • Fan Y, Bergmann A (2008b). Distinct mechanisms of apoptosis induced compensatory proliferation in proliferating and differentiating tissues in the Drosophila eye. Dev Cell 14: 399– 410.
  • Fernando P, Brunette S, Megeney LA (2005). Neural stem cell differentiation is dependent upon endogenous caspase 3 activity. FASEB J 19: 1671–1673.
  • Fernando P, Kelly JF, Balazsi K, Slack RS, Megeney LA (2002). Caspase 3 activity is required for skeletal muscle differentiation. P Natl Acad Sci USA 99: 11025–11030.
  • Fernando P, Megeney LA (2007). Is caspase-dependent apoptosis only cell differentiation taken to the extreme? FASEB J 21: 8–17.
  • Fink SL, Cookson BT (2005). Apoptosis, pyroptosis and necrosis: mechanistic description of death and dying eukaryotic cells. Infect Immun 73: 1907–1916.
  • Fischer B, Metzger M, Richardson R, Knyphausen P, Ramezani T, Franzen R, Schmelzer E, Bloch W, Carney TJ, Hammerschmidt M (2014). p53 and TAp63 promote keratinocyte proliferation and differentiation in breeding tubercles of the zebrafish. PLoS Genet 10: e1004048.
  • Fuchs E, Chen T (2013). A matter of life and death: self-renewal in stem cells. EMBO Rep 14: 39–48.
  • Fuchs Y, Steller H (2011). Programmed cell death in animal development and disease. Cell 147: 742–758.
  • Fujita J, Crane AM, Souza MK, Dejosez M, Kyba M, Flavell RA, Thomson JA, Zwaka TP (2008). Caspase activity mediates the differentiation of embryonic stem cells. Cell Stem Cell 2: 595–601.
  • Fulle S, Sancilio S, Mancinelli R, Gatta V, Di Pietro R (2013). Dual role of the caspase enzymes in satellite cells from aged and young subjects. Cell Death Dis 4: e955.
  • Galluzzi L, Vitale J, Abrams JM, Alnemri ES, Baehrecke EH, Blagosklonny MV, Dawson TM, Dawson VL, El-Deiry WS, Fulda S et al. (2012). Molecular definitions of cell death subroutines: recommendations of the Nomenclature Committee on Cell Death. Cell Death Differ 19: 107–120.
  • Gdynia G, Grund K, Eckert A, Böck BC, Funke B, Macher-Goeppinger S, Sieber S, Herold-Mende C, Wiestler B, Wiestler OD et al. (2007). Basal caspase activity promotes migration and invasiveness in glioblastoma cells. Mol Cancer Res 5: 1232–1240.
  • Gezginci S, Bolkent S (2007). The effect of Z-FA.FMK on D-galactosamine/TNF-alpha-induced liver injury in mice. Cell Biochem Funct 25: 277–286.
  • Gezginci-Oktayoglu S, Bolkent S (2012). Ras signaling in NGF reduction and TNF-α-related pancreatic β cell apoptosis in hyperglycemic rats. Apoptosis 17: 14–24.
  • Gezginci-Oktayoglu S, Sacan O, Yanardag R, Karatug A, Bolkent S (2011). Exendin-4 improves hepatocyte injury by decreasing proliferation through blocking NGF/TrkA in diabetic mice. Peptides 32: 223–231.
  • Gezginci-Oktayoglu S, Tunali S, Yanardag R, Bolkent S (2008). Effects of Z-FA.FMK on D-galactosamine/tumor necrosis factor-alpha-induced kidney injury and oxidative stress in mice : effects of Z-FA.FMK on TNF-alpha-mediated kidney injury. Mol Cell Biochem 309: 9–20.
  • Goessling W, North TE, Loewer S, Lord AM, Lee S, Stoick-Cooper CL, Weidinger G, Puder M, Daley GQ, Moon RT et al. (2009). Genetic interaction of PGE2 and Wnt signaling regulates developmental specification of stem cells and regeneration. Cell 136: 1136–1147.
  • Golstein P, Kroemer G (2006). Cell death by necrosis: towards a molecular definition. Trends Biochem Sci 32: 37–42.
  • Gozuacik D, Bialik S, Raveh T, Mitou G, Shohat G, Sabanay H, Mizushima N, Yoshimon T, Kimchi A (2008). DAP-Kinase is a mediator of endoplasmic reticulum stress-induced caspase activation and autophagic cell death. Cell Death Differ 15: 1875–1886.
  • Gregory CD, Pound JD (2011). Cell death in the neighbourhood: direct microenvironmental effects of apoptosis in normal and neoplastic tissues. J Pathol 223: 177–194.
  • Gurtner GC, Werner S, Barrandon Y, Longaker MT (2008). Wound repair and regeneration. Nature 453: 314–321.
  • Hagimoto N, Kuwano K, Miyazaki H, Kunitake R, Fujita M, Kawasaki M, Kaneko Y, Hara N (1997). Induction of apoptosis and pulmonary fibrosis in mice in response to ligation of Fas antigen. Am J Respir Cell Mol Biol 17: 272–278.
  • Herold S, Tabar TS, Janssen H, Hoegner K, Cabanski M, Lewe Schlosser P, Albrecht J, Driever F, Vadasz I, Seeger W et al. (2011). Exudate macrophages attenuate lung injury by the release of IL-1 receptor antagonist in gram-negative pneumonia. Am J Respir Crit Care Med 183: 1380–1390.
  • Herz HM, Chen Z, Scherr H, Lackey M, Bolduc C, Bergmann A (2006). vps25 mosaics display non-autonomous cell survival and overgrowth, and autonomous apoptosis. Development 133: 1871–1880.
  • Hewitson TD (2012). Fibrosis in the kidney: is a problem shared a problem halved? Fibrogenesis Tissue Repair 5 (Suppl. 1): S14.
  • Huang Q, Li F, Liu X, Li W, Shi W, Liu FF, O’Sullivan B, He Z, Peng Y, Tan AC et al. (2011). Caspase 3-mediated stimulation of tumor cell repopulation during cancer radiotherapy. Nat Med 17: 860–866.
  • Huesmann GR, Clayton DF (2006). Dynamic role of postsynaptic caspase-3 and BIRC4 in zebra finch song-response habituation. Neuron 52: 1061–1072.
  • Huh JR, Guo M, Hay BA (2004). Compensatory proliferation induced by cell death in the Drosophila wing disc requires activity of the apical cell death caspase Dronc in a nonapoptotic role. Curr Biol 14: 1262–1266.
  • Ishizaki Y, Jacobson MD, Raff MC (1998). A role for caspases in lens fiber differentiation. J Cell Biol 140: 153–158.
  • Janzen V, Fleming HE, Riedt T, Karlsson G, Riese MJ, Lo Celso C, Reynolds G, Milne CD, Paige CJ, Karlsson S et al. (2008). Hematopoietic stem cell responsiveness to exogenous signals is limited by caspase-3. Cell Stem Cell 2: 584–594.
  • Jejurikar SS, Henkelman EA, Cederna PS, Marcelo CL, Urbanchek MG, Kuzon WM Jr (2006). Aging increases the susceptibility of skeletal muscle derived satellite cells to apoptosis. Exp Gerontol 41: 828–836.
  • Jiao S, Li Z (2011). Non-apoptotic function of BAD and BAX in long-term depression of synaptic transmission. Neuron 70: 758–772.
  • Jouan-Lanhouet S, Arshad MI, Piquet-Pellorce C, Martin-Chouly C, Le Moigne Muller G, Van Herreweghe F, Takahashi N, Sergent O, Laqadic-Goossman D, Vanderabeele P et al. (2012). TRAIL induces necroptosis involving RIPK1/RIPK3-dependent PARP-1 activation. Cell Death Differ 19: 2003–2014.
  • Jun JI, Lau LF (2010). Cellular senescence controls fibrosis in wound healing. Aging (Albany NY) 2: 627–631.
  • Kang TB, Ben-Moshe T, Varfolomeev EE, Pewzner-Jung Y, Yogev N, Jurewicz A, Waisman A, Brenner O, Haffner R, Gustafsson E et al. (2004). Caspase-8 serves both apoptotic and nonapoptotic roles. J Immunol 173: 2976–2984.
  • Kaplan Y, Gibbs-Bar L, Kalifa Y, Feinstein-Rotkopf Y, Arama E (2010). Gradients of a ubiquitin E3 ligase inhibitor and a caspase inhibitor determine differentiation or death in spermatids. Dev Cell 19: 160–173.
  • Kataoka T, Tschopp J (2004). N-terminal fragment of c-FLIP(L) processed by caspase 8 specifically interacts with TRAF2 and induces activation of the NF-kappaB signaling pathway. Mol Cell Biol 24: 2627–2636.
  • Kayagaki N, Warming S, Lamkanfi M, Vande Walle L, Louie S, Dong J, Newton K, Qu Y, Liu J, Heldens S et al. (2011). Non-canonical inflammasome activation targets caspase-11. Nature 479: 117– 121.
  • Kayalar O, Oztay F (2014). Retinoic acid induced repair in the lung of adult hyperoxic mice, reducing transforming growth factor-β1 mediated abnormal alterations. Acta Histochem 116: 810–819.
  • Kim KW, Hwang M, Moretti L, Jaboin JJ, Cha YI, Lu B (2008). Autophagy upregulation by inhibitors of caspase-3 and mTOR enhances radiotherapy in a mouse model of lung cancer. Autophagy 4: 659–668.
  • Koizumi K, Saitoh Y, Minami T, Takeno N, Tsuneyama K, Miyahara T, Nakayama T, Sakurai H, Takano Y, Nishimura M et al. (2009). Role of CX3CL1/fractalkine in osteoclast differentiation and bone resorption. J Immunol 183: 7825–7831.
  • Koleva M, Kappler R, Vogler M, Herwig A, Fulda S, Hahn H (2005). Pleiotropic effects of sonic hedgehog on muscle satellite cells. Cell Mol Life Sci 62: 1863–1870.
  • Kondo S, Senoo-Matsuda N, Hiromi Y, Miura M (2006). DRONC coordinates cell death and compensatory proliferation. Mol Cell Biol 26: 7258–7268.
  • Konigshoff M, Kramer M, Balsara N, Wilhelm J, Amarie OV, Jahn A, Rose F, Fink L, Seeger W, Schaefer L et al. (2009). WNT1- inducible signaling protein-1 mediates pulmonary fibrosis in mice and is upregulated in humans with idiopathic pulmonary fibrosis. J Clin Invest 119: 772–787.
  • Kroemer G, Galluzzi L, Vandenabeele P, Abrams J, Alnemri ES, Baehrecke EH, Blagosklonny MV, El-Deiry WS, Golstein P, Green DR et al. (2009). Classification of cell death: recommendations of the Nomenclature Committee on Cell Death. Cell Death Differ 16: 3–11.
  • Kulasekaran P, Scavone CA, Rogers DS, Arenberg DA, Thannickal VJ, Horowitz JC (2009). Endothelin-1 and transforming growth factor-beta1 independently induce fibroblast resistance to apoptosis via AKT activation. Am J Respir Cell Mol Biol 41: 484–493.
  • Kuranaga E, Miura M (2007). Nonapoptotic functions of caspases: caspases as regulatory molecules for immunity and cell-fate determination. Trends Cell Biol 17: 135–144.
  • Kushner EJ, MacEneaney OJ, Weil BR, Greiner JJ, Stauffer BL, DeSouza CA (2011). Aging is associated with a proapoptotic endothelial progenitor cell phenotype. J Vasc Res 48: 408–414.
  • Labbé K, Saleh M (2011). Pyroptosis: a caspase-1-dependent programmed cell death and a barrier to infection. In: Couillin I, Pétrilli V, Martinon F, editors. The Inflammasomes. Basel, Switzerland: Springer Basel AG, pp. 17–36.
  • Larsen BD, Rampalli S, Burns LE, Brunette S, Dilworth FJ, Megeney LA (2010). Caspase 3/caspase-activated DNase promote cell differentiation by inducing DNA strand breaks. P Natl Acad Sci USA 107: 4230–4235.
  • Lavrik IN, Golks A, Krammer PH (2005). Caspases: pharmacological manipulation of cell death. J Clin Invest 115: 2665–2672.
  • Lee TV, Ding T, Chen Z, Rajendran V, Scherr H, Lackey M, Bolduc C, Bergmann A (2008). The E1 ubiquitin-activating enzyme Uba1 in Drosophila controls apoptosis autonomously and tissue growth nonautonomously. Development 135: 43–52.
  • Lens SM, Kataoka T, Fortner KA, Tinel A, Ferrero I, MacDonald RH, Hahne M, Beermann F, Attinger A, Orbea HA et al. (2002). The caspase 8 inhibitor c-FLIP(L) modulates T-cell receptor-induced proliferation but not activation-induced cell death of lymphocytes. Mol Cell Biol 22: 5419–5433.
  • Li F, Huang Q, Chen J, Peng Y, Roop DR, Bedford JS, Li CY (2010). Apoptotic cells activate the “phoenix rising” pathway to promote wound healing and tissue regeneration. Sci Signal 3: ra13.
  • Li J, Brieher WM, Scimone ML, Kang SJ, Zhu H, Yin H, von Andrian UH, Mitchison T, Yuan J (2007). Caspase-11 regulates cell migration by promoting Aip1-Cofilin-mediated actin depolymerization. Nat Cell Biol 9: 276–286.
  • Li J, Yuan J (2008). Caspases in apoptosis and beyond. Oncogene 27: 6194–6206.
  • Li W, Kale A, Baker NE (2009). Oriented cell division as a response to cell death and cell competition. Curr Biol 19: 1821–1826.
  • Liu L, Nishio N, Ito S, Tanaka Y, Isobe K ( 2014). Negative regulation of GADD34 on myofibroblasts during cutaneous wound healing. Biomed Res Int 2014: 137–149.
  • Liu X, He Y, Li F, Huang Q, Kato TA, Hall RP, Li CY (2015). Caspase-3 promotes genetic instability and carcinogenesis. Mol Cell 58: 284–296.
  • Liu YR, Sun B, Zhao XL, Gu Q, Liu ZY, Dong XY, Che N, Mo J (2013). Basal caspase-3 activity promotes migration, invasion, and vasculogenic mimicry formation of melanoma cells. Melanoma Res 23: 243–253.
  • Lovric MM, Hawkins CJ (2010). TRAIL treatment provokes mutations in surviving cells. Oncogene 29: 5048–5060.
  • Luo X, Kraus WL (2012). On PAR with PARP: cellular stress signaling through poly (ADP-ribose) and PARP-1. Genes Dev 26: 417–432.
  • Ma J, Chen T, Mandelin J, Ceponis A, Miller NE, Hukkanen M, Ma GF, Konttinen YT (2003). Regulation of macrophage activation. Cell Mol Life Sci 60: 2334–2346.
  • Maeda S, Kamata H, Luo JL, Leffert H, Karin M (2005). IKKb couples hepatocyte death to cytokine-driven compensatory proliferation that promotes chemical hepatocarcinogenesis. Cell 121: 977–990.
  • Mao C, Obeid LM (2008). Ceramidases: regulators of cellular responses mediated by ceramide, sphingosine, and sphingosine 1-phosphate. Biochim Biophys Acta 1781: 424–434.
  • Mao P, Smith L, Xie W, Wang M (2013). Dying endothelial cells stimulate proliferation of malignant glioma cells via a caspase 3-mediated pathway. Oncol Lett 5: 1615–1620.
  • Marcotte R, Lacelle C, Wang E (2004). Senescent fibroblasts resist apoptosis by downregulating caspase-3. Mech Ageing Dev 125: 777–783.
  • Martinon F, Tschopp J (2007). Inflammatory caspases and inflammasomes; master switches of inflammation. Cell Death Differ 14: 10–22.
  • Matalova E, Lesot H, Svandova E, Vanden Berghe T, Sharpe PT, Healy C, Vandenabeele P, Tucker AS (2013). Caspase-7 participates in differentiation of cells forming dental hard tissues. Dev Growth Differ 55: 615–621.
  • McIlwain DR, Berger T, Mak TW (2013). Caspase functions in cell death and disease. Cold Spring Harb Perspect Biol 5: a008656.
  • Micheau O, Thome M, Schneider P, Holler N, Tschopp J, Nicholson DW, Briand C, Grütter MG (2002). The long form of FLIP is an activator of caspase-8 at the Fas death-inducing signaling complex. J Biol Chem 277: 45162–45171.
  • Miura M (2012). Apoptotic and nonapoptotic caspase functions in animal development. Cold Spring Harb Perspect Biol 4: a008664.
  • Miura M, Chen XD, Allen MR, Bi Y, Gronthos S, Seo BM, Lakhani S, Flavell RA, Feng XH, Robey PG et al. (2004). A crucial role of caspase-3 in osteogenic differentiation of bone marrow stromal stem cells. J Clin Invest 114: 1704–1713.
  • Moberg KH, Schelble S, Burdick SK, Hariharan IK (2005). Mutations in erupted, the Drosophila ortholog of mammalian tumor susceptibility gene 101, elicit non-cell-autonomous overgrowth. Dev Cell 9: 699–710.
  • Mogi M, Togari A (2003). Activation of caspases is required for osteoblastic differentiation. J Biol Chem 278: 47477–47482.
  • Monack DM, Raupach B, Hromockyj AE, Falkow S (1996). Salmonella typhimurium invasion induces apoptosis in infected macrophages. P Natl Acad Sci USA 93: 9833–9838.
  • Moodley YP, Misso NL, Scaffidi AK, Fogel-Petrovic M, McAnulty RJ, Laurent GJ, Thompson PJ, Knight DA (2003). Inverse effects of interleukin-6 on apoptosis of fibroblasts from pulmonary fibrosis and normal lungs. Am J Respir Cell Mol Biol 29: 490– 498.
  • Moretti L, Kim KW, Jung DK, Willey CD, Lu B (2009). Radiosensitization of solid tumors by Z-VAD, a pan-caspase inhibitor. Mol Cancer Ther 8: 1270–1279.
  • Murray TV, McMahon JM, Howley BA, Stanley A, Ritter T, Mohr A, Zwacka R, Fearnhead HO (2008). A non-apoptotic role for caspase-9 in muscle differentiation. J Cell Sci 121: 3786–3793.
  • Nikoletopoulou V, Markaki M, Palikaras K, Tavernarakis N (2013). Crosstalk between apoptosis, necrosis and autophagy. Biochim Biophys Acta 1833: 3448–3459.
  • Norman JM, Cohen GM, Bampton ET (2010). The in vitro cleavage of the hAtg proteins by cell death proteases. Autophagy 6: 1042–1056.
  • O’Brien LE, Soliman SS, Li X, Bilder D (2011). Altered modes of stem cell division drive adaptive intestinal growth. Cell 147: 603–614.
  • Oh J, Lee YD, Wagers AJ (2014). Stem cell aging: mechanisms, regulators and therapeutic opportunities. Nat Med 20: 870– 880.
  • Okuyama R, Nguyen BC, Talora C, Ogawa E, Tommasi di Vignano A, Lioumi M, Chiorino G, Tagami H, Woo M, Dotto GP (2004). High commitment of embryonic keratinocytes to terminal differentiation through a Notch1-caspase 3 regulatory mechanism. Dev Cell 6: 551–562.
  • Olson NE, Graves JD, Shu GL, Ryan EJ, Clark EA (2003). Caspase activity is required for stimulated B lymphocytes to enter the cell cycle. J Immunol 170: 6065–6072.
  • Oral O, Oz-Arslan D, Itah Z, Naghavi A, Deveci R, Karacali S, Gozuacik D (2012). Cleavage of tg3 protein by caspase-8 regulates autophagy during receptor-activated cell death. Apoptosis 17: 810–820.
  • Ovet H, Oztay F (2014). The copper chelator tetrathiomolybdate regressed bleomycin-induced pulmonary fibrosis in mice, by reducing lysyl oxidase expressions. Biol Trace Elem Res 162: 189–199.
  • Oztay F, Gezginci-Oktayoglu S, Bayrak BB, Yanardag R, Bolkent S (2010). Cathepsin B inhibition improves lung injury associated to D-galactosamine/tumor necrosis factor-alpha-induced liver injury in mice. Mol Cell Biochem 333: 65–72.
  • Oztay F, Yilmaz O (2015). Method of Treating Fibrotic Diseases Using Dasatinib. TR201503969A2.
  • Pellettieri J, Fitzgerald P, Watanabe S, Mancuso J, Green DR, Sánchez Alvarado A (2010). Cell death and tissue remodeling in planarian regeneration. Dev Biol 338: 76–85.
  • Pérez-Garijo A, Martín FA, Morata G (2004). Caspase inhibition during apoptosis causes abnormal signalling and developmental aberrations in Drosophila. Development 131: 5591–5598
  • Pérez-Garijo A, Shlevkov E, Morata G (2009). The role of Dpp and Wg in compensatory proliferation and in the formation of hyperplastic overgrowths caused by apoptotic cells in the Drosophila wing disc. Development 136: 1169–1177.
  • Peter ME, Krammer PH (2003). The CD95 (APO-1/Fas) DISC and beyond. Cell Death Differ 10: 26–35.
  • Pfleger CM, Harvey KF, Yan H, Hariharan IK (2007). Mutation of the gene encoding the ubiquitin activating enzyme Uba1 causes tissue overgrowth in Drosophila. Fly 1: 95–105.
  • Plenchette S, Cathelin S, Rébé C, Launay S, Ladoire S, Sordet O, Ponnelle T, Debili N, Phan TH, Padua RA et al. (2004). Translocation of the inhibitor of apoptosis protein c-IAP1 from the nucleus to the Golgi in hematopoietic cells undergoing differentiation: a nuclear export signal-mediated event. Blood 104: 2035–2043.
  • Pop C, Oberst A, Drag M, van Raam BJ, Riedl SJ, Green DR, Salvesen GS (2011). FLIPL induces caspase 8 activity in the absence of interdomain caspase 8 cleavage and alters substrate specificity. Biochem J 433: 447–457.
  • Ribeil JA, Zermati Y, Vandekerckhove J, Cathelin S, Kersual J, Dussiot M, Coulon S, Moura IC, Zeuner A, Kirkegaard-Sørensen T et al. (2007). Hsp70 regulates erythropoiesis by preventing caspase-3-mediated cleavage of GATA-1. Nature 445: 102–105.
  • Rudrapatna VA, Bangi E, Cagan RL (2013). Caspase signalling in the absence of apoptosis drives Jnk-dependent invasion. EMBO Rep 14: 172–177.
  • Ryborg AK, Johansen C, Iversen L, Kragballe K (2004). Lysophosphatidylcholine induces keratinocyte differentiation and upregulation of AP-1- and NF-kappaB DNA-binding activity. Acta Derm Venereol 84: 433–438.
  • Ryoo HD, Gorenc T, Steller H (2004). Apoptotic cells can induce compensatory cell proliferation through the JNK and the Wingless signaling pathways. Dev Cell 7: 491–501.
  • Sakurai T, Maeda S, Chang L, Karin M (2006). Loss of hepatic NF-kB activity enhances chemical hepatocarcinogenesis through sustained c-Jun N-terminal kinase 1 activation. P Natl Acad Sci USA 103: 10544–10551.
  • Salmena L, Lemmers B, Hakem A, Matysiak-Zablocki E, Murakami K, Au PY, Berry DM, Tamblyn L, Shehabeldin A, Migon E et al. (2003). Essential role for caspase 8 in T-cell homeostasis and T-cell-mediated immunity. Genes Dev 17: 883–895.
  • Santiago B, Galindo M, Palao G, Pablos JL (2004). Intracellular regulation of Fas-induced apoptosis in human fibroblasts by extracellular factors and cycloheximide. J Immunol 172: 560– 566.
  • Schwab JM, Chiang N, Arita M, Serhan CN (2007). Resolvin E1 and protectin D1 activate inflammation-resolution programmes. Nature 447: 869–874.
  • Senft J, Helfer B, Frisch SM (2007). Caspase-8 interacts with the p85 subunit of phosphatidylinositol 3-kinase to regulate cell adhesion and motility. Cancer Res 67: 11505–11509.
  • Shalini S, Dorstyn L, Dawar S, Kumar S (2015). Old, new and emerging functions of caspases. Cell Death Differ 22: 526–539.
  • Shi J, Zhao Y, Wang Y, Gao W, Ding J, Li P, Hu L, Shao F (2014). Inflammatory caspases are innate immune receptors for intracellular LPS. Nature 514: 187–192.
  • Solá S, Aranha MM, Rodrigues CM (2012). Driving apoptosis-relevant proteins toward neural differentiation. Mol Neurobiol 46: 316–331.
  • Solá S, Morgado AL, Rodrigues CMP (2013). Death receptors and mitochondria: two prime triggers of neural apoptosis. Biochim Biophys Acta 1830: 2160–2166.
  • Sollberger G, Strittmatter GE, Garstkiewicz M, Sand J, Beer HD (2014). Caspase-1: the inflammasome and beyond. Innate Immun 20: 115–125.
  • Sordet O, Rébé C, Plenchette S, Zermati Y, Hermine O, Vainchenker W, Garrido C, Solary E, Dubrez-Daloz L (2002). Specific involvement of caspases in the differentiation of monocytes into macrophages. Blood 100: 4446–4453.
  • Stoneman V, Bennett MR (2009). Role of Fas/Fas-L in vascular cell apoptosis. J Cardiovasc Pharmacol 53: 100–108.
  • Su H, Bidère N, Zheng L, Cubre A, Sakai K, Dale J, Salmena L, Hakem R, Straus S, Lenardo M (2005). Requirement for caspase-8 in NF-kappaB activation by antigen receptor. Science 307: 1465– 1468.
  • Tanaka T, Yoshimi M, Maeyama T, Hagimoto N, Kuwano K, Hara N (2002). Resistance to Fas-mediated apoptosis in human lung fibroblast. Eur Respir J 20: 359–368.
  • Thompson BJ, Mathieu J, Sung HH, Loeser E, Rørth P, Cohen SM (2005). Tumor suppressor properties of the ESCRT-II complex component Vps25 in Drosophila. Dev Cell 9: 711–720.
  • Tiwari M, Sharma LK, Vanegas D, Callaway DA, Bai Y, Lechleiter JD, Herman B (2014). A nonapoptotic role for CASP2/caspase 2: modulation of autophagy. Autophagy 10: 1054–1070.
  • Tschopp J, Irmler M, Thome M (1998). Inhibition of fas death signals by FLIPs. Curr Opin Immunol 10: 552–558.
  • Uhal BD, Joshi I, Hughes WF, Ramos C, Pardo A, Selman M (1998). Alveolar epithelial cell death adjacent to underlying myofibroblasts in advanced fibrotic human lung. Am J Physiol Lung C 275: L1192–L1199.
  • Uhal BD, Joshi I, True AL, Mundle S, Raza A, Pardo A, Selman M (1995). Fibroblasts isolated after fibrotic lung injury induce apoptosis of alveolar epithelial cells in vitro. Am J Physiol-Lung C 269: L819–L828.
  • Vaccari T, Bilder D (2005). The Drosophila tumor suppressor vps25 prevents nonautonomous overproliferation by regulating notch trafficking. Dev Cell 9: 687–698.
  • Vandenabeele P, Galluzzi L, Vanden Berghe T, Kroemer G (2010). Molecular mechanisms of necroptosis: an ordered cellular explosion. Nat Rev Mol Cell Biol. 11: 700–714.
  • Vercammen BD, Beyaert R, Denecker G, Goossens V, Loo GV, Declercq W, Grooten J, Fiers W, Vanderabeele P (1998). Inhibition of caspases increases the sensitivity of L929 cells to necrosis mediated by tumor necrosis factor. J Exp Med 187: 1477–1485.
  • Viganò E, Mortellaro A (2013). Caspase-11: the driving factor for nancanonical inflammasomes. Eur J Immunol 43: 2240–2245.
  • Wang R, Ramos C, Joshi I, Zagariya A, Pardo A, Selman M, Uhal BD (1999). Human lung myofibroblast-derived inducers of alveolar epithelial apoptosis identified as angiotensin peptides. Am J Physiol 277: L1158–L1164.
  • Watt FM, Estrach S, Ambler CA (2008). Epidermal Notch signalling: differentiation, cancer and adhesion. Curr Opin Cell Biol 20: 171–179.
  • Wilson A, Shehadeh LA, Yu H, Webster KA (2010). Age-related molecular genetic changes of murine bone marrow mesenchymal stem cells. BMC Genomics 11: 229.
  • Woo M, Hakem R, Furlonger C, Hakem A, Duncan GS, Sasaki T, Bouchard D, Lu L, Wu GE, Paige CJ et al. (2003). Caspase-3 regulates cell cycle in B cells: a consequence of substrate specificity. Nat Immunol 4: 1016–1022.
  • Wu H, Che X, Zheng Q, Wu A, Pan K, Shao A, Wu Q, Zhang J, Hong Y (2014). Caspases: a molecular switch node in the crosstalk between autophagy and apoptosis. Int J Biol Sci 10: 1072–1083.
  • Yan XX, Najbauer J, Woo CC, Dashtipour K, Ribak CE, Leon M (2001). Expression of active caspase-3 in mitotic and postmitotic cells of the rat forebrain. J Comp Neurol 433: 4–22.
  • Yang L, Cao Z, Yan H, Wood WC (2003). Coexistence of high levels of apoptotic signaling and inhibitor of apoptosis proteins in human tumor cells: implication for cancer specific therapy. Cancer Res 63: 6815–6524.
  • Yi CH, Yuan J (2009). The Jekyll and Hyde functions of caspases. Dev Cell 16: 21–34.
  • Zermati Y, Garrido C, Amsellem S, Fishelson S, Bouscary D, Valensi F, Varet B, Solary E, Hermine O (2001). Caspase activation is required for terminal erythroid differentiation. J Exp Med 193: 247–254.
  • Zhao X, Wang D, Zhao Z, Xiao Y, Sengupta S, Xiao Y, Zhang R, Lauber K, Wesselborg S, Feng L et al. (2006). Caspase-3- dependent activation of calcium-independent phospholipase A2 enhances cell migration in non-apoptotic ovarian cancer cells. J Biol Chem 281: 29357–29368.
  • Zhong Z, Schwabe RF, Kai Y, He L, Yang L, Bunzendahl H, Brenner DA, Lemasters JJ (2006). Liver regeneration is suppressed in small-for-size liver grafts after transplantation: involvement of c-Jun N-terminal kinase, cyclin D1, and defective energy supply. Transplantation 82: 241–250.