Skip to main page content
U.S. flag

An official website of the United States government

Dot gov

The .gov means it’s official.
Federal government websites often end in .gov or .mil. Before sharing sensitive information, make sure you’re on a federal government site.

Https

The site is secure.
The https:// ensures that you are connecting to the official website and that any information you provide is encrypted and transmitted securely.

Access keys NCBI Homepage MyNCBI Homepage Main Content Main Navigation
. 2001 Jan 1;21(1):169-75.
doi: 10.1523/JNEUROSCI.21-01-00169.2001.

Bcl-X(L)-caspase-9 interactions in the developing nervous system: evidence for multiple death pathways

Affiliations

Bcl-X(L)-caspase-9 interactions in the developing nervous system: evidence for multiple death pathways

A U Zaidi et al. J Neurosci. .

Abstract

Programmed cell death is critical for normal nervous system development and is regulated by Bcl-2 and Caspase family members. Targeted disruption of bcl-x(L), an antiapoptotic bcl-2 gene family member, causes massive death of immature neurons in the developing nervous system whereas disruption of caspase-9, a proapoptotic caspase gene family member, leads to decreased neuronal apoptosis and neurodevelopmental abnormalities. To determine whether Bcl-X(L) and Caspase-9 interact in an obligate pathway of neuronal apoptosis, bcl-x/caspase-9 double homozygous mutants were generated. The increased apoptosis of immature neurons observed in Bcl-X(L)-deficient embryos was completely prevented by concomitant Caspase-9 deficiency. In contrast, bcl-x(-/-)/caspase-9(-/-) embryonic mice exhibited an expanded ventricular zone and neuronal malformations identical to that observed in mice lacking only Caspase-9. These results indicate both epistatic and independent actions of Bcl-X(L) and Caspase-9 in neuronal programmed cell death. To examine Bcl-2 and Caspase family-dependent apoptotic pathways in telencephalic neurons, we compared the effects of cytosine arabinoside (AraC), a known neuronal apoptosis inducer, on wild-type, Bcl-X(L)-, Bax-, Caspase-9-, Caspase-3-, and p53-deficient telencephalic neurons in vitro. AraC caused extensive apoptosis of wild-type and Bcl-X(L)-deficient neurons. p53- and Bax-deficient neurons showed marked protection from AraC-induced death, whereas Caspase-9- and Caspase-3-deficient neurons showed minimal or no protection, respectively. These findings contrast with our previous investigation of AraC-induced apoptosis of telencephalic neural precursor cells in which death was completely blocked by p53 or Caspase-9 deficiency but not Bax deficiency. In total, these results indicate a transition from Caspase-9- to Bax- and Bcl-X(L)-mediated neuronal apoptosis.

PubMed Disclaimer

Figures

Fig. 1.
Fig. 1.
Caspase-9 deficiency blocks the increased immature neuron apoptosis caused by Bcl-XL deficiency.A–C, Hematoxylin and eosin-stained sections from the E12.5 brainstem show normal healthy neurons in a wild-type embryo (A) and large numbers of apoptotic neurons in a Bcl-XL-deficient embryo (B; examples of apoptotic nuclei are indicated by arrows).Bcl-x/−/caspase-9/−brainstem (C) shows no evidence of increased apoptosis. D–F, A bisbenzimide-stained section from a Bcl-XL-deficient embryo shows frequent abnormal condensed nuclei in the anterior spinal cord (D; examples indicated by arrows), and simultaneous labeling for activated Caspase-3 immunoreactivity (E, red) reveals numerous positive cells. Dual staining for activated Caspase-3 immunoreactivity and nuclei in abcl-x/−/caspase-9/−E12.5 spinal cord (F) shows neither activated Caspase-3 immunoreactivity nor condensed nuclei. Scale bar, 25 μm.
Fig. 2.
Fig. 2.
Bcl-XL deficiency fails to prevent neurodevelopmental abnormalities in Caspase-9-deficient embryos. Toluidine blue-stained horizontal sections from the E12.5 telencephalon show well organized neuroepithelium in wild-type (A) but neithercaspase-9/−(B) norbcl-x/−/caspase-9/−(C) embryos. In both Caspase-9-deficient embryos, the telencephalic vesicle is thickened, and supernumerary cells fill the lateral ventricle (V).a, Anterior. Scale bar, 25 μm.
Fig. 3.
Fig. 3.
Quantification of Caspase-3 activation and cell death in wild-type, Caspase-9-deficient, Bcl-XL-deficient, and double-deficient telencephalic cells. E12.5 telencephalic cells were grown for 48 hr in DMEM; activated Caspase-3 immunoreactive cells were detected with CM1 antiserum (A), and cell death (B) was determined by propidium iodide labeling. Bcl-XL-deficient cells had increased Caspase-3 activation and cell death; concomitant Caspase-9 deficiency inhibited the death-promoting effects of Bcl-XL in this in vitro paradigm. No effect of gene dosage on Caspase-3 activation or cell viability was observed for eitherbcl-x or caspase-9, and therefore, data from wild-type and heterozygous mutants were pooled (+/o denotes +/+ and +/− mice). Each data point represents the mean ± SEM. *p < 0.05 compared withbcl-x+/o/caspase-9+/ocells.
Fig. 4.
Fig. 4.
Effect of proapoptotic and antiapoptotic gene disruptions on AraC-induced telencephalic neuron Caspase-3 activation and cell death. Primary telencephalic neuron cultures were prepared from control and homozygous mutant embryos. After 48 hr of AraC exposure, Caspase-3 activation and cell death was assessed using CM1 antiserum and SYTOX Green labeling. In Bcl-XL-deficient neurons, AraC treatment produced a significant increase in the number of cells exhibiting activated Caspase-3 immunoreactivity (A) and in dead cells (B). Although Bax-, Caspase-9-, Caspase-3-, and p53-deficient neurons all exhibited reduced Caspase-3 activation compared with control neurons after AraC exposure, only Bax- and p53-deficient neurons exhibited significant neuroprotection. Heterozygous bax,caspase-9, and caspase-3 mutant neurons behaved identically to wild-type neurons and therefore, data from wild-type and +/− cultures were pooled (+/o denotes +/+ and +/− mice). In contrast, p53+/− neurons exhibited an intermediate degree of neuroprotection (data not shown) and therefore, only wild-type littermates were used to compare the effects of p53 deficiency. Each data point represents mean ± SEM (n = 5–20 for the different genotypes).

Similar articles

Cited by

  • Association of Insulin-like Growth Factor-1 and Neurofilament Light Chain in Patients with Progressive Supranuclear Palsy.
    Dey S, Yelamanchi R, Mullapudi T, Holla VV, Kamble N, Mahale RR, Sathyaprabha TN, Pal PK, Debnath M, Yadav R. Dey S, et al. Ann Indian Acad Neurol. 2024 Jan-Feb;27(1):40-45. doi: 10.4103/aian.aian_507_23. Epub 2024 Jan 18. Ann Indian Acad Neurol. 2024. PMID: 38495245 Free PMC article.
  • Apoptotic cell death in disease-Current understanding of the NCCD 2023.
    Vitale I, Pietrocola F, Guilbaud E, Aaronson SA, Abrams JM, Adam D, Agostini M, Agostinis P, Alnemri ES, Altucci L, Amelio I, Andrews DW, Aqeilan RI, Arama E, Baehrecke EH, Balachandran S, Bano D, Barlev NA, Bartek J, Bazan NG, Becker C, Bernassola F, Bertrand MJM, Bianchi ME, Blagosklonny MV, Blander JM, Blandino G, Blomgren K, Borner C, Bortner CD, Bove P, Boya P, Brenner C, Broz P, Brunner T, Damgaard RB, Calin GA, Campanella M, Candi E, Carbone M, Carmona-Gutierrez D, Cecconi F, Chan FK, Chen GQ, Chen Q, Chen YH, Cheng EH, Chipuk JE, Cidlowski JA, Ciechanover A, Ciliberto G, Conrad M, Cubillos-Ruiz JR, Czabotar PE, D'Angiolella V, Daugaard M, Dawson TM, Dawson VL, De Maria R, De Strooper B, Debatin KM, Deberardinis RJ, Degterev A, Del Sal G, Deshmukh M, Di Virgilio F, Diederich M, Dixon SJ, Dynlacht BD, El-Deiry WS, Elrod JW, Engeland K, Fimia GM, Galassi C, Ganini C, Garcia-Saez AJ, Garg AD, Garrido C, Gavathiotis E, Gerlic M, Ghosh S, Green DR, Greene LA, Gronemeyer H, Häcker G, Hajnóczky G, Hardwick JM, Haupt Y, He S, Heery DM, Hengartner MO, Hetz C, Hildeman DA, Ichijo H, Inoue S, Jäättelä M, Janic A, Joseph B, Jost PJ, Kanneganti TD, Karin M, Kashkar H, Kaufmann T, Kelly … See abstract for full author list ➔ Vitale I, et al. Cell Death Differ. 2023 May;30(5):1097-1154. doi: 10.1038/s41418-023-01153-w. Epub 2023 Apr 26. Cell Death Differ. 2023. PMID: 37100955 Free PMC article. Review.
  • Active DNA demethylation promotes cell fate specification and the DNA damage response.
    Wang D, Wu W, Callen E, Pavani R, Zolnerowich N, Kodali S, Zong D, Wong N, Noriega S, Nathan WJ, Matos-Rodrigues G, Chari R, Kruhlak MJ, Livak F, Ward M, Caldecott K, Di Stefano B, Nussenzweig A. Wang D, et al. Science. 2022 Dec 2;378(6623):983-989. doi: 10.1126/science.add9838. Epub 2022 Dec 1. Science. 2022. PMID: 36454826 Free PMC article.
  • Potential of Therapeutic Small Molecules in Apoptosis Regulation in the Treatment of Neurodegenerative Diseases: An Updated Review.
    Dailah HG. Dailah HG. Molecules. 2022 Oct 25;27(21):7207. doi: 10.3390/molecules27217207. Molecules. 2022. PMID: 36364033 Free PMC article. Review.
  • Development of Conformational Antibodies to Detect Bcl-xL's Amyloid Aggregates in Metal-Induced Apoptotic Neuroblastoma Cells.
    Gonneaud A, Fakhir FZ, Landas E, Le Tallec E, Chartier-Garcia E, Almunia C, Chenal A, Forge V, Marquette C. Gonneaud A, et al. Int J Mol Sci. 2020 Oct 15;21(20):7625. doi: 10.3390/ijms21207625. Int J Mol Sci. 2020. PMID: 33076337 Free PMC article.

References

    1. Ahmad M, Srinivasula SM, Hegde R, Mukattash R, Fernandes-Alnemri T, Alnemri ES. Identification and characterization of murine caspase-14, a new member of the caspase family. Cancer Res. 1998;58:5201–5205. - PubMed
    1. Armstrong RC, Aja TJ, Hoang KD, Gaur S, Bai X, Alnemri ES, Litwack G, Karanewsky DS, Fritz LC, Tomaselli KJ. Activation of the CED3/ICE-related protease CPP32 in cerebellar granule neurons undergoing apoptosis but not necrosis. J Neurosci. 1997;17:553–562. - PMC - PubMed
    1. Boise LH, González-García M, Postema CE, Ding L, Linsten T, Turka LA, Mao X, Thompson CB. bcl-x, a bcl-2-related gene that functions as a dominant regulator of apoptotic cell death. Cell. 1993;74:597–608. - PubMed
    1. Cecconi F, Alvarez-Bolado G, Meyer BI, Roth KA, Gruss P. Apaf1 (CED-4 Homolog) regulates programmed cell death in mammalian development. Cell. 1998;94:727–737. - PubMed
    1. Chong MJ, Murray MR, Gosink EC, Russell HRC, Srinivasan A, Kapsetaki M, Korsmeyer SJ, McKinnon PJ. Atm and Bax cooperate in ionizing radiation-induced apoptosis in the central nervous system. Proc Natl Acad Sci USA. 2000;97:889–894. - PMC - PubMed

Publication types

MeSH terms

-