#PAGE_PARAMS# #ADS_HEAD_SCRIPTS# #MICRODATA#

Distinct and Atypical Intrinsic and Extrinsic Cell Death Pathways between Photoreceptor Cell Types upon Specific Ablation of in Cone Photoreceptors


Non-autonomous cell-death is a cardinal feature of the disintegration of neural networks in neurodegenerative diseases, but the molecular bases of this process are poorly understood. The neural retina comprises a mosaic of rod and cone photoreceptors. Cone and rod photoreceptors degenerate upon rod-specific expression of heterogeneous mutations in functionally distinct genes, whereas cone-specific mutations are thought to cause only cone demise. Here we show that conditional ablation in cone photoreceptors of Ran-binding protein-2 (Ranbp2), a cell context-dependent pleiotropic protein linked to neuroprotection, familial necrotic encephalopathies, acute transverse myelitis and tumor-suppression, promotes early electrophysiological deficits, subcellular erosive destruction and non-apoptotic death of cones, whereas rod photoreceptors undergo cone-dependent non-autonomous apoptosis. Cone-specific Ranbp2 ablation causes the temporal activation of a cone-intrinsic molecular cascade highlighted by the early activation of metalloproteinase 11/stromelysin-3 and up-regulation of Crx and CoREST, followed by the down-modulation of cone-specific phototransduction genes, transient up-regulation of regulatory/survival genes and activation of caspase-7 without apoptosis. Conversely, PARP1+-apoptotic rods develop upon sequential activation of caspase-9 and caspase-3 and loss of membrane permeability. Rod photoreceptor demise ceases upon cone degeneration. These findings reveal novel roles of Ranbp2 in the modulation of intrinsic and extrinsic cell death mechanisms and pathways. They also unveil a novel spatiotemporal paradigm of progression of neurodegeneration upon cell-specific genetic damage whereby a cone to rod non-autonomous death pathway with intrinsically distinct cell-type death manifestations is triggered by cell-specific loss of Ranbp2. Finally, this study casts new light onto cell-death mechanisms that may be shared by human dystrophies with distinct retinal spatial signatures as well as with other etiologically distinct neurodegenerative disorders.


Vyšlo v časopise: Distinct and Atypical Intrinsic and Extrinsic Cell Death Pathways between Photoreceptor Cell Types upon Specific Ablation of in Cone Photoreceptors. PLoS Genet 9(6): e32767. doi:10.1371/journal.pgen.1003555
Kategorie: Research Article
prolekare.web.journal.doi_sk: https://doi.org/10.1371/journal.pgen.1003555

Souhrn

Non-autonomous cell-death is a cardinal feature of the disintegration of neural networks in neurodegenerative diseases, but the molecular bases of this process are poorly understood. The neural retina comprises a mosaic of rod and cone photoreceptors. Cone and rod photoreceptors degenerate upon rod-specific expression of heterogeneous mutations in functionally distinct genes, whereas cone-specific mutations are thought to cause only cone demise. Here we show that conditional ablation in cone photoreceptors of Ran-binding protein-2 (Ranbp2), a cell context-dependent pleiotropic protein linked to neuroprotection, familial necrotic encephalopathies, acute transverse myelitis and tumor-suppression, promotes early electrophysiological deficits, subcellular erosive destruction and non-apoptotic death of cones, whereas rod photoreceptors undergo cone-dependent non-autonomous apoptosis. Cone-specific Ranbp2 ablation causes the temporal activation of a cone-intrinsic molecular cascade highlighted by the early activation of metalloproteinase 11/stromelysin-3 and up-regulation of Crx and CoREST, followed by the down-modulation of cone-specific phototransduction genes, transient up-regulation of regulatory/survival genes and activation of caspase-7 without apoptosis. Conversely, PARP1+-apoptotic rods develop upon sequential activation of caspase-9 and caspase-3 and loss of membrane permeability. Rod photoreceptor demise ceases upon cone degeneration. These findings reveal novel roles of Ranbp2 in the modulation of intrinsic and extrinsic cell death mechanisms and pathways. They also unveil a novel spatiotemporal paradigm of progression of neurodegeneration upon cell-specific genetic damage whereby a cone to rod non-autonomous death pathway with intrinsically distinct cell-type death manifestations is triggered by cell-specific loss of Ranbp2. Finally, this study casts new light onto cell-death mechanisms that may be shared by human dystrophies with distinct retinal spatial signatures as well as with other etiologically distinct neurodegenerative disorders.


Zdroje

1. PalopJJ, ChinJ, MuckeL (2006) A network dysfunction perspective on neurodegenerative diseases. Nature 443: 768–773.

2. IlievaH, PolymenidouM, ClevelandDW (2009) Non-cell autonomous toxicity in neurodegenerative disorders: ALS and beyond. J Cell Biol 187: 761–772.

3. BramallAN, WrightAF, JacobsonSG, McInnesRR (2010) The genomic, biochemical, and cellular responses of the retina in inherited photoreceptor degenerations and prospects for the treatment of these disorders. Annu Rev Neurosci 33: 441–472.

4. Portera-CailliauC, SungCH, NathansJ, AdlerR (1994) Apoptotic photoreceptor cell death in mouse models of retinitis pigmentosa. Proc Natl Acad Sci USA 91: 974–978.

5. KedzierskiW, BokD, TravisGH (1998) Non-cell-autonomous photoreceptor degeneration in rds mutant mice mosaic for expression of a rescue transgene. J Neurosci 18: 4076–4082.

6. Mohand-SaidS, HicksD, LeveillardT, PicaudS, PortoF, et al. (2001) Rod-cone interactions: developmental and clinical significance. Prog Retin Eye Res 20: 451–467.

7. HuangPC, GaitanAE, HaoY, PettersRM, WongF (1993) Cellular interactions implicated in the mechanism of photoreceptor degeneration in transgenic mice expressing a mutant rhodopsin gene. Proc Natl Acad Sci USA 90: 8484–8488.

8. LewisA, WilliamsP, LawrenceO, WongRO, BrockerhoffSE (2010) Wild-type cone photoreceptors persist despite neighboring mutant cone degeneration. J Neurosci 30: 382–389.

9. Carter-DawsonLD, LaVailMM, SidmanRL (1978) Differential effect of the rd mutation on rods and cones in the mouse retina. Invest Ophthalmol Vis Sci 17: 489–498.

10. HartongDT, BersonEL, DryjaTP (2006) Retinitis pigmentosa. Lancet 368: 1795–1809.

11. JaissleGB, MayCA, ReinhardJ, KohlerK, FauserS, et al. (2001) Evaluation of the rhodopsin knockout mouse as a model of pure cone function. Invest Ophthalmol Vis Sci 42: 506–513.

12. CideciyanAV, HoodDC, HuangY, BaninE, LiZY, et al. (1998) Disease sequence from mutant rhodopsin allele to rod and cone photoreceptor degeneration in man. Proc Natl Acad Sci U S A 95: 7103–7108.

13. JohnSK, SmithJE, AguirreGD, MilamAH (2000) Loss of cone molecular markers in rhodopsin-mutant human retinas with retinitis pigmentosa. Mol Vis 6: 204–215.

14. HumphriesMM, RancourtD, FarrarGJ, KennaP, HazelM, et al. (1997) Retinopathy induced in mice by targeted disruption of the rhodopsin gene. Nat Genet 15: 216–219.

15. DryjaTP, FinnJT, PengYW, McGeeTL, BersonEL, et al. (1995) Mutations in the gene encoding the alpha subunit of the rod cGMP-gated channel in autosomal recessive retinitis pigmentosa. Proc Natl Acad Sci USA 92: 10177–10181.

16. BareilC, HamelCP, DelagueV, ArnaudB, DemailleJ, et al. (2001) Segregation of a mutation in CNGB1 encoding the beta-subunit of the rod cGMP-gated channel in a family with autosomal recessive retinitis pigmentosa. Hum Genet 108: 328–334.

17. ChangB, GrauT, DangelS, HurdR, JurkliesB, et al. (2009) A homologous genetic basis of the murine cpfl1 mutant and human achromatopsia linked to mutations in the PDE6C gene. Proc Natl Acad Sci USA 106: 19581–19586.

18. ThiadensAA, den HollanderAI, RoosingS, NabuursSB, Zekveld-VroonRC, et al. (2009) Homozygosity mapping reveals PDE6C mutations in patients with early-onset cone photoreceptor disorders. Am J Hum Genet 85: 240–247.

19. TrifunovicD, DenglerK, MichalakisS, ZrennerE, WissingerB, et al. (2010) cGMP-dependent cone photoreceptor degeneration in the cpfl1 mouse retina. J Comp Neurol 518: 3604–3617.

20. ChangB, HawesNL, HurdRE, DavissonMT, NusinowitzS, et al. (2002) Retinal degeneration mutants in the mouse. Vision Res 42: 517–525.

21. GardnerJC, WebbTR, KanugaN, RobsonAG, HolderGE, et al. (2010) X-linked cone dystrophy caused by mutation of the red and green cone opsins. Am J Hum Genet 87: 26–39.

22. GlicksteinM, HeathGG (1975) Receptors in the monochromat eye. Vision Res 15: 633–636.

23. WissingerB, GamerD, JagleH, GiordaR, MarxT, et al. (2001) CNGA3 mutations in hereditary cone photoreceptor disorders. Am J Hum Genet 69: 722–737.

24. NishiguchiKM, SandbergMA, GorjiN, BersonEL, DryjaTP (2005) Cone cGMP-gated channel mutations and clinical findings in patients with achromatopsia, macular degeneration, and other hereditary cone diseases. Hum Mutat 25: 248–258.

25. BielM, SeeligerM, PfeiferA, KohlerK, GerstnerA, et al. (1999) Selective loss of cone function in mice lacking the cyclic nucleotide-gated channel CNG3. Proc Natl Acad Sci USA 96: 7553–7557.

26. DingXQ, HarryCS, UminoY, MatveevAV, FlieslerSJ, et al. (2009) Impaired cone function and cone degeneration resulting from CNGB3 deficiency: down-regulation of CNGA3 biosynthesis as a potential mechanism. Hum Mol Genet 18: 4770–4780.

27. ShenJ, YangX, DongA, PettersRM, PengYW, et al. (2005) Oxidative damage is a potential cause of cone cell death in retinitis pigmentosa. J Cell Physiol 203: 457–464.

28. PunzoC, KornackerK, CepkoCL (2009) Stimulation of the insulin/mTOR pathway delays cone death in a mouse model of retinitis pigmentosa. Nat Neurosci 12: 44–52.

29. KomeimaK, RogersBS, LuL, CampochiaroPA (2006) Antioxidants reduce cone cell death in a model of retinitis pigmentosa. Proc Natl Acad Sci USA 103: 11300–11305.

30. LeveillardT, Mohand-SaidS, LorentzO, HicksD, FintzAC, et al. (2004) Identification and characterization of rod-derived cone viability factor. Nat Genet 36: 755–759.

31. OtaniA, DorrellMI, KinderK, MorenoSK, NusinowitzS, et al. (2004) Rescue of retinal degeneration by intravitreally injected adult bone marrow-derived lineage-negative hematopoietic stem cells. J Clin Invest 114: 765–774.

32. ZengHY, ZhuXA, ZhangC, YangLP, WuLM, et al. (2005) Identification of sequential events and factors associated with microglial activation, migration, and cytotoxicity in retinal degeneration in rd mice. Invest Ophthalmol Vis Sci 46: 2992–2999.

33. ZeissCJ, JohnsonEA (2004) Proliferation of microglia, but not photoreceptors, in the outer nuclear layer of the rd-1 mouse. Invest Ophthalmol Vis Sci 45: 971–976.

34. RippsH (2002) Cell death in retinitis pigmentosa: gap junctions and the ‘bystander’ effect. Exp Eye Res 74: 327–336.

35. DoonanF, DonovanM, CotterTG (2003) Caspase-independent photoreceptor apoptosis in mouse models of retinal degeneration. J Neurosci 23: 5723–5731.

36. Sancho-PelluzJ, Arango-GonzalezB, KustermannS, RomeroFJ, van VeenT, et al. (2008) Photoreceptor cell death mechanisms in inherited retinal degeneration. Mol Neurobiol 38: 253–269.

37. FrassonM, SahelJA, FabreM, SimonuttiM, DreyfusH, et al. (1999) Retinitis pigmentosa: rod photoreceptor rescue by a calcium-channel blocker in the rd mouse. Nat Med 5: 1183–1187.

38. PawlykBS, LiT, ScimecaMS, SandbergMA, BersonEL (2002) Absence of photoreceptor rescue with D-cis-diltiazem in the rd mouse. Invest Ophthalmol Vis Sci 43: 1912–1915.

39. Pearce-KellingSE, AlemanTS, NickleA, LatiesAM, AguirreGD, et al. (2001) Calcium channel blocker D-cis-diltiazem does not slow retinal degeneration in the PDE6B mutant rcd1 canine model of retinitis pigmentosa. Mol Vis 7: 42–47.

40. TakeuchiK, NakazawaM, MizukoshiS (2008) Systemic administration of nilvadipine delays photoreceptor degeneration of heterozygous retinal degeneration slow (rds) mouse. Exp Eye Res 86: 60–69.

41. JosephRM, LiT (1996) Overexpression of Bcl-2 or Bcl-XL transgenes and photoreceptor degeneration. Invest Ophthalmol Vis Sci 37: 2434–2446.

42. ChenJ, FlanneryJG, LaVailMM, SteinbergRH, XuJ, et al. (1996) bcl-2 overexpression reduces apoptotic photoreceptor cell death in three different retinal degenerations. Proc Natl Acad Sci U S A 93: 7042–7047.

43. ZeissCJ, NealJ, JohnsonEA (2004) Caspase-3 in postnatal retinal development and degeneration. Invest Ophthalmol Vis Sci 45: 964–970.

44. JomaryC, NealMJ, JonesSE (2001) Characterization of cell death pathways in murine retinal neurodegeneration implicates cytochrome c release, caspase activation, and bid cleavage. Mol Cell Neurosci 18: 335–346.

45. SangesD, ComitatoA, TammaroR, MarigoV (2006) Apoptosis in retinal degeneration involves cross-talk between apoptosis-inducing factor (AIF) and caspase-12 and is blocked by calpain inhibitors. Proc Natl Acad Sci USA 103: 17366–17371.

46. SchaeferhoffK, MichalakisS, TanimotoN, FischerMD, BecirovicE, et al. (2010) Induction of STAT3-related genes in fast degenerating cone photoreceptors of cpfl1 mice. Cell Mol Life Sci 67: 3173–3186.

47. AslanukovA, BhowmickR, GurujuM, OswaldJ, RazD, et al. (2006) RanBP2 Modulates Cox11 and Hexokinase I Activities and Haploinsufficiency of RanBP2 Causes Deficits in Glucose Metabolism. PLoS Genet 2: e177.

48. DawlatyMM, MalureanuL, JeganathanKB, KaoE, SustmannC, et al. (2008) Resolution of sister centromeres requires RanBP2-mediated SUMOylation of topoisomerase IIalpha. Cell 133: 103–115.

49. ChoKI, YiH, TserentsoodolN, SearleK, FerreiraPA (2010) Neuroprotection resulting from insufficiency of RANBP2 is associated with the modulation of protein and lipid homeostasis of functionally diverse but linked pathways in response to oxidative stress. Dis Model Mech 3: 595–604.

50. ChoKI, YiH, YehA, TserentsoodolN, CuadradoL, et al. (2009) Haploinsufficiency of RanBP2 is neuroprotective against light-elicited and age-dependent degeneration of photoreceptor neurons. Cell Death Differ 16: 287–297.

51. ChoKI, SearleK, WebbM, YiH, FerreiraPA (2012) Ranbp2 haploinsufficiency mediates distinct cellular and biochemical phenotypes in brain and retinal dopaminergic and glia cells elicited by the Parkinsonian neurotoxin, 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP). Cell Mol Life Sci 69: 3511–3527.

52. NeilsonDE, AdamsMD, OrrCM, SchellingDK, EibenRM, et al. (2009) Infection-Triggered Familial or Recurrent Cases of Acute Necrotizing Encephalopathy Caused by Mutations in a Component of the Nuclear Pore, RANBP2. Am J Hum Genet 84: 44–51.

53. WolfK, Schmitt-MechelkeT, KolliasS, CurtA (2013) Acute necrotizing encephalopathy (ANE1): rare autosomal-dominant disorder presenting as acute transverse myelitis. J Neurol [Epub ahead of print]

54. LonnqvistT, IsohanniP, ValanneL, Olli-LahdesmakiT, SuomalainenA, et al. (2011) Dominant encephalopathy mimicking mitochondrial disease. Neurology 76: 101–103.

55. MavlyutovTA, CaiY, FerreiraPA (2002) Identification of RanBP2- and Kinesin-Mediated Transport Pathways with Restricted Neuronal and Subcellular Localization. Traffic 3: 630–640.

56. LeYZ, AshJD, Al-UbaidiMR, ChenY, MaJX, et al. (2004) Targeted expression of Cre recombinase to cone photoreceptors in transgenic mice. Mol Vis 10: 1011–1018.

57. LeYZ, AshJD, Al-UbaidiMR, ChenY, MaJX, et al. (2006) Conditional gene knockout system in cone photoreceptors. Adv Exp Med Biol 572: 173–178.

58. NikonovSS, BrownBM, DavisJA, ZunigaFI, BraginA, et al. (2008) Mouse cones require an arrestin for normal inactivation of phototransduction. Neuron 59: 462–474.

59. JeonCJ, StrettoiE, MaslandRH (1998) The major cell populations of the mouse retina. J Neurosci 18: 8936–8946.

60. ChenJ, RattnerA, NathansJ (2005) The rod photoreceptor-specific nuclear receptor Nr2e3 represses transcription of multiple cone-specific genes. J Neurosci 25: 118–129.

61. GalluzziL, VitaleI, AbramsJM, AlnemriES, BaehreckeEH, et al. (2012) Molecular definitions of cell death subroutines: recommendations of the Nomenclature Committee on Cell Death 2012. Cell Death Differ 19: 107–120.

62. KroemerG, GalluzziL, VandenabeeleP, AbramsJ, AlnemriES, et al. (2009) Classification of cell death: recommendations of the Nomenclature Committee on Cell Death 2009. Cell Death Differ 16: 3–11.

63. KaufmannSH, DesnoyersS, OttavianoY, DavidsonNE, PoirierGG (1993) Specific proteolytic cleavage of poly(ADP-ribose) polymerase: an early marker of chemotherapy-induced apoptosis. Cancer Res 53: 3976–3985.

64. GermainM, AffarEB, D'AmoursD, DixitVM, SalvesenGS, et al. (1999) Cleavage of automodified poly(ADP-ribose) polymerase during apoptosis. Evidence for involvement of caspase-7. J Biol Chem 274: 28379–28384.

65. Paquet-DurandF, SilvaJ, TalukdarT, JohnsonLE, AzadiS, et al. (2007) Excessive activation of poly(ADP-ribose) polymerase contributes to inherited photoreceptor degeneration in the retinal degeneration 1 mouse. J Neurosci 27: 10311–10319.

66. SleeEA, HarteMT, KluckRM, WolfBB, CasianoCA, et al. (1999) Ordering the cytochrome c-initiated caspase cascade: hierarchical activation of caspases-2, -3, -6, -7, -8, and -10 in a caspase-9-dependent manner. J Cell Biol 144: 281–292.

67. KleinJA, Longo-GuessCM, RossmannMP, SeburnKL, HurdRE, et al. (2002) The harlequin mouse mutation downregulates apoptosis-inducing factor. Nature 419: 367–374.

68. LiptonSA, Bossy-WetzelE (2002) Dueling activities of AIF in cell death versus survival: DNA binding and redox activity. Cell 111: 147–150.

69. MurakamiY, MatsumotoH, RohM, SuzukiJ, HisatomiT, et al. (2012) Receptor interacting protein kinase mediates necrotic cone but not rod cell death in a mouse model of inherited degeneration. Proc Natl Acad Sci U S A 109: 14598–14603.

70. KunchithapauthamK, RohrerB (2007) Apoptosis and autophagy in photoreceptors exposed to oxidative stress. Autophagy 3: 433–441.

71. KunchithapauthamK, RohrerB (2007) Autophagy is one of the multiple mechanisms active in photoreceptor degeneration. Autophagy 3: 65–66.

72. Page-McCawA, EwaldAJ, WerbZ (2007) Matrix metalloproteinases and the regulation of tissue remodelling. Nat Rev Mol Cell Biol 8: 221–233.

73. SanukiR, OnishiA, KoikeC, MuramatsuR, WatanabeS, et al. (2011) miR-124a is required for hippocampal axogenesis and retinal cone survival through Lhx2 suppression. Nat Neurosci 14: 1125–1134.

74. HennigAK, PengGH, ChenS (2008) Regulation of photoreceptor gene expression by Crx-associated transcription factor network. Brain Res 1192: 114–133.

75. NgL, HurleyJB, DierksB, SrinivasM, SaltoC, et al. (2001) A thyroid hormone receptor that is required for the development of green cone photoreceptors. Nat Genet 27: 94–98.

76. RobertsMR, SrinivasM, ForrestD, Morreale de EscobarG, RehTA (2006) Making the gradient: thyroid hormone regulates cone opsin expression in the developing mouse retina. Proc Natl Acad Sci USA 103: 6218–6223.

77. NishidaA, FurukawaA, KoikeC, TanoY, AizawaS, et al. (2003) Otx2 homeobox gene controls retinal photoreceptor cell fate and pineal gland development. Nat Neurosci 6: 1255–1263.

78. KoikeC, NishidaA, UenoS, SaitoH, SanukiR, et al. (2007) Functional roles of Otx2 transcription factor in postnatal mouse retinal development. Mol Cell Biol 27: 8318–8329.

79. BaudetML, ZivrajKH, Abreu-GoodgerC, MuldalA, ArmisenJ, et al. (2012) miR-124 acts through CoREST to control onset of Sema3A sensitivity in navigating retinal growth cones. Nat Neurosci 15: 29–38.

80. MotrescuER, BlaiseS, EtiqueN, MessaddeqN, ChenardMP, et al. (2008) Matrix metalloproteinase-11/stromelysin-3 exhibits collagenolytic function against collagen VI under normal and malignant conditions. Oncogene 27: 6347–6355.

81. HanHB, GuJ, ZuoHJ, ChenZG, ZhaoW, et al. (2012) Let-7c functions as a metastasis suppressor by targeting MMP11 and PBX3 in colorectal cancer. J Pathol 226: 544–555.

82. Stehlin-GaonC, WillmannD, ZeyerD, SanglierS, Van DorsselaerA, et al. (2003) All-trans retinoic acid is a ligand for the orphan nuclear receptor ROR beta. Nat Struct Biol 10: 820–825.

83. FujiedaH, BremnerR, MearsAJ, SasakiH (2009) Retinoic acid receptor-related orphan receptor alpha regulates a subset of cone genes during mouse retinal development. J Neurochem 108: 91–101.

84. HelmlingerD, YvertG, PicaudS, MerienneK, SahelJ, et al. (2002) Progressive retinal degeneration and dysfunction in R6 Huntington's disease mice. Hum Mol Genet 11: 3351–3359.

85. HaradaC, GuoX, NamekataK, KimuraA, NakamuraK, et al. (2011) Glia- and neuron-specific functions of TrkB signalling during retinal degeneration and regeneration. Nat Commun 2: 189.

86. WilsonRB, KunchithapauthamK, RohrerB (2007) Paradoxical role of BDNF: BDNF+/− retinas are protected against light damage-mediated stress. Invest Ophthalmol Vis Sci 48: 2877–2886.

87. van AmerongenR, MikelsA, NusseR (2008) Alternative wnt signaling is initiated by distinct receptors. Sci Signal 1: re9.

88. SharmaS, BallSL, PeacheyNS (2005) Pharmacological studies of the mouse cone electroretinogram. Vis Neurosci 22: 631–636.

89. XuX, QuiambaoAB, RoveriL, PardueMT, MarxJL, et al. (2000) Degeneration of cone photoreceptors induced by expression of the Mas1 protooncogene. Exp Neurol 163: 207–219.

90. BaroukhN, RavierMA, LoderMK, HillEV, BounacerA, et al. (2007) MicroRNA-124a regulates Foxa2 expression and intracellular signaling in pancreatic beta-cell lines. J Biol Chem 282: 19575–19588.

91. PeiD, WeissSJ (1995) Furin-dependent intracellular activation of the human stromelysin-3 zymogen. Nature 375: 244–247.

92. MassonR, LefebvreO, NoelA, FahimeME, ChenardMP, et al. (1998) In vivo evidence that the stromelysin-3 metalloproteinase contributes in a paracrine manner to epithelial cell malignancy. J Cell Biol 140: 1535–1541.

93. AndarawewaKL, MotrescuER, ChenardMP, GansmullerA, StollI, et al. (2005) Stromelysin-3 is a potent negative regulator of adipogenesis participating to cancer cell-adipocyte interaction/crosstalk at the tumor invasive front. Cancer Res 65: 10862–10871.

94. ManesS, MiraE, BarbacidMM, CipresA, Fernandez-ResaP, et al. (1997) Identification of insulin-like growth factor-binding protein-1 as a potential physiological substrate for human stromelysin-3. J Biol Chem 272: 25706–25712.

95. WuE, MariBP, WangF, AndersonIC, SundayME, et al. (2001) Stromelysin-3 suppresses tumor cell apoptosis in a murine model. J Cell Biochem 82: 549–555.

96. BoulayA, MassonR, ChenardMP, El FahimeM, CassardL, et al. (2001) High cancer cell death in syngeneic tumors developed in host mice deficient for the stromelysin-3 matrix metalloproteinase. Cancer Res 61: 2189–2193.

97. JonesLE, HumphreysMJ, CampbellF, NeoptolemosJP, BoydMT (2004) Comprehensive analysis of matrix metalloproteinase and tissue inhibitor expression in pancreatic cancer: increased expression of matrix metalloproteinase-7 predicts poor survival. Clin Cancer Res 10: 2832–2845.

98. AsanoT, TadaM, ChengS, TakemotoN, KuramaeT, et al. (2008) Prognostic values of matrix metalloproteinase family expression in human colorectal carcinoma. J Surg Res 146: 32–42.

99. YangYH, DengH, LiWM, ZhangQY, HuXT, et al. (2008) Identification of matrix metalloproteinase 11 as a predictive tumor marker in serum based on gene expression profiling. Clin Cancer Res 14: 74–81.

100. FisherJF, MobasheryS (2006) Recent advances in MMP inhibitor design. Cancer metastasis reviews 25: 115–136.

101. MatziariM, DiveV, YiotakisA (2007) Matrix metalloproteinase 11 (MMP-11; stromelysin-3) and synthetic inhibitors. Med Res Rev 27: 528–552.

Štítky
Genetika Reprodukčná medicína

Článok vyšiel v časopise

PLOS Genetics


2013 Číslo 6
Najčítanejšie tento týždeň
Najčítanejšie v tomto čísle
Kurzy

Zvýšte si kvalifikáciu online z pohodlia domova

Získaná hemofilie - Povědomí o nemoci a její diagnostika
nový kurz

Eozinofilní granulomatóza s polyangiitidou
Autori: doc. MUDr. Martina Doubková, Ph.D.

Všetky kurzy
Prihlásenie
Zabudnuté heslo

Zadajte e-mailovú adresu, s ktorou ste vytvárali účet. Budú Vám na ňu zasielané informácie k nastaveniu nového hesla.

Prihlásenie

Nemáte účet?  Registrujte sa

#ADS_BOTTOM_SCRIPTS#