Skip to main content
Log in

Glucotoxic and diabetic conditions induce caspase 6-mediated degradation of nuclear lamin A in human islets, rodent islets and INS-1 832/13 cells

  • Original Paper
  • Published:
Apoptosis Aims and scope Submit manuscript

Abstract

Nuclear lamins form the lamina on the interior surface of the nuclear envelope, and regulate nuclear metabolic events, including DNA replication and organization of chromatin. The current study is aimed at understanding the role of executioner caspase 6 on lamin A integrity in islet β-cells under duress of glucotoxic (20 mM glucose; 24 h) and diabetic conditions. Under glucotoxic conditions, glucose-stimulated insulin secretion and metabolic cell viability were significantly attenuated in INS-1 832/13 cells. Further, exposure of normal human islets, rat islets and INS-1 832/13 cells to glucotoxic conditions leads to caspase 6 activation and lamin A degradation, which is also observed in islets from the Zucker diabetic fatty rat, a model for type 2 diabetes (T2D), and in islets from a human donor with T2D. Z-Val-Glu-Ile-Asp-fluoromethylketone, a specific inhibitor of caspase 6, markedly attenuated high glucose-induced caspase 6 activation and lamin A degradation, confirming that caspase 6 mediates lamin A degradation under high glucose exposure conditions. Moreover, Z-Asp-Glu-Val-Asp-fluoromethylketone, a known caspase 3 inhibitor, significantly inhibited high glucose-induced caspase 6 activation and lamin A degradation, suggesting that activation of caspase 3 might be upstream to caspase 6 activation in the islet β-cell under glucotoxic conditions. Lastly, we report expression of ZMPSTE24, a zinc metallopeptidase involved in the processing of prelamin A to mature lamin A, in INS-1 832/13 cells and human islets; was unaffected by high glucose. We conclude that caspases 3 and 6 could contribute to alterations in the integrity of nuclear lamins leading to metabolic dysregulation and failure of the islet β-cell.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Institutional subscriptions

Fig. 1
Fig. 2
Fig. 3
Fig. 4
Fig. 5
Fig. 6
Fig. 7
Fig. 8

Similar content being viewed by others

Abbreviations

ELISA:

Enzyme-linked immunosorbent assay

ER:

Endoplasmic reticulum

FTI:

Farnesyl transferase inhibitor

GSIS:

Glucose stimulated insulin secretion

IFNγ:

Interferon gamma

IL-1β:

Interleukin-1beta

iNOS:

Inducible nitric oxide synthase

LMNA:

Lamin A gene

NO:

Nitric oxide

T2D:

Type 2 Diabetes

TNF α:

Tumor necrosis factor alpha

Z-DEVD-FMK:

Caspase 3 inhibitor (Z-Asp-Glu-Val-Asp-fluoromethylketone)

ZDF:

Zucker diabetic fatty

ZLC:

Zucker lean control

ZMPSTE24:

Zinc metallopeptidase (STE24 homolog)

Z-VEID-FMK:

Caspase 6 inhibitor (Z-Val-Glu-Ile-Asp-fluoromethylketone)

References

  1. Gruenbaum Y, Wilson KL, Harel A, Goldberg M, Cohen M (2000) Review: nuclear lamins–structural proteins with fundamental functions. J Struct Biol 129:313–323

    Article  CAS  PubMed  Google Scholar 

  2. Moir RD, Spann TP (2001) The structure and function of nuclear lamins: implications for disease. Cell Mol Life Sci 58:1748–1757

    Article  CAS  PubMed  Google Scholar 

  3. Moir RD, Spann TP, Goldman RD (1995) The dynamic properties and possible functions of nuclear lamins. Int Rev Cytol 162:141–182

    Article  Google Scholar 

  4. Moir RD, Spann TP, Herrmann H, Goldman RD (2000) Disruption of nuclear lamin organization blocks the elongation phase of DNA replication. J Cell Biol 149:1179–1192

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  5. Shahzidi S, Brech A, Sioud M, Li X, Suo Z, Nesland JM, Peng Q (2013) Lamin A/C cleavage by caspase-6 activation is crucial for apoptotic induction by photodynamic therapy with hexaminolevulinate in human B-cell lymphoma cells. Cancer Lett 339:25–32

    Article  CAS  PubMed  Google Scholar 

  6. Attur M, Ben-Artzi A, Yang Q, Al-Mussawir HE, Worman HJ, Palmer G, Abramson SB (2012) Perturbation of nuclear lamin A causes cell death in chondrocytes. Arthritis Rheum 64:1940–1949

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  7. Chang KH, Multani PS, Sun KH, Vincent F, de Pablo Y, Ghosh S, Gupta R, Lee HP, Lee HG, Smith MA, Shah K (2011) Nuclear envelope dispersion triggered by deregulated Cdk5 precedes neuronal death. Mol Biol Cell 22:1452–1462

    Article  PubMed Central  PubMed  Google Scholar 

  8. Yeo HS, Shehzad A, Lee YS (2012) Prostaglandin E2 blocks menadione-induced apoptosis through the Ras/Raf/Erk signaling pathway in promonocytic leukemia cell lines. Mol Cells 33:371–378

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  9. Martínez-Poveda B, Rodríguez-Nieto S, García-Caballero M, Medina MÁ, Quesada AR (2012) The antiangiogenic compound aeroplysinin-1 induces apoptosis in endothelial cells by activating the mitochondrial pathway. Mar Drugs 10:2033–2046

    Article  PubMed Central  PubMed  Google Scholar 

  10. Burke B (2001) Lamins and apoptosis: a two-way street? J Cell Biol 153:F5–F7

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  11. Syeda K, Mohammed AM, Arora DK, Kowluru A (2013) Glucotoxic conditions induce endoplasmic reticulum stress to cause caspase 3 mediated lamin B degradation in pancreatic β-cells: protection by nifedipine. Biochem Pharmacol 86:1338–1346

    Article  CAS  PubMed  Google Scholar 

  12. Lenin R, Maria MS, Agrawal M, Balasubramanyam J, Mohan V, Balasubramanyam M (2012) Amelioration of glucolipotoxicity-induced endoplasmic reticulum stress by a “chemical chaperone” in human THP-1 monocytes. Exp Diabetes Res. doi:10.1155/2012/356487

    PubMed Central  PubMed  Google Scholar 

  13. Jing G, Wang JJ, Zhang SX (2012) ER stress and apoptosis: a new mechanism for retinal cell death. Exp Diabetes Res. doi:10.1155/2012/589589

    PubMed Central  PubMed  Google Scholar 

  14. Roy S, Trudeau K, Roy S, Tien T, Barrette KF (2013) Mitochondrial dysfunction and endoplasmic reticulum stress in diabetic retinopathy: mechanistic insights into high glucose-induced retinal cell death. Curr Clin Pharmacol 8:278–284

    Article  CAS  PubMed  Google Scholar 

  15. Newsholme P, Haber EP, Hirabara SM, Rebelato EL, Procopio J, Morgan D, Oliveira-Emilio HC, Carpinelli AR, Curi R (2007) Diabetes associated cell stress and dysfunction: role of mitochondrial and non-mitochondrial ROS production and activity. J Physiol 583:9–24

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  16. Lim S, Rashid MA, Jang M, Kim Y, Won H, Lee J, Woo JT, Kim YS, Murphy MP, Ali L, Ha J, Kim SS (2011) Mitochondria-targeted antioxidants protect pancreatic β-cells against oxidative stress and improve insulin secretion in glucotoxicity and glucolipotoxicity. Cell Physiol Biochem 28:873–886

    Article  CAS  PubMed  Google Scholar 

  17. Veluthakal R, Palanivel R, Zhao Y, McDonald P, Gruber S, Kowluru A (2005) Ceramide induces mitochondrial abnormalities in insulin-secreting INS-1 cells: potential mechanisms underlying ceramide-mediated metabolic dysfunction of the beta cell. Apoptosis 10:841–850

    Article  CAS  PubMed  Google Scholar 

  18. Inoue S, Browne G, Melino G, Cohen GM (2009) Ordering of caspases in cells undergoing apoptosis by the intrinsic pathway. Cell Death Differ 16:1053–1061

    Article  CAS  PubMed  Google Scholar 

  19. Seki K, Yoshikawa H, Shiiki K, Hamada Y, Akamatsu N, Tasaka K (2000) Cisplatin (CDDP) specifically induces apoptosis via sequential activation of caspase-8, -3 and -6 in osteosarcoma. Cancer Chemother Pharmacol 45:199–206

    Article  CAS  PubMed  Google Scholar 

  20. Jayaram B, Kowluru A (2012) Phagocytic NADPH oxidase links ARNO-Arf6 signaling pathway in glucose-stimulated insulin secretion from the pancreatic β-cell. Cell Physiol Biochem 30:1351–1362

    Article  CAS  PubMed  Google Scholar 

  21. Arora DK, Machhadieh B, Matti A, Wadzinski BE, Ramanadham S, Kowluru A (2014) High glucose exposure promotes activation of protein phosphatase 2A in rodent islets and INS-1 832/13 β-cells by increasing the posttranslational carboxylmethylation of its catalytic subunit. Endocrinology 155:380–391

    Article  CAS  PubMed  Google Scholar 

  22. Slee EA, Adrain C, Martin SJ (2001) Executioner caspase-3, -6, and -7 perform distinct, non-redundant roles during the demolition phase of apoptosis. J Biol Chem 276:7320–7326

    Article  CAS  PubMed  Google Scholar 

  23. Simon DJ, Weimer RM, McLaughlin T, Kallop D, Stanger K, Yang J, O’Leary DD, Hannoush RN, Tessier-Lavigne M (2012) A caspase cascade regulating developmental axon degeneration. J Neurosci 32:17540–17553

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  24. Okinaga T, Kasai H, Tsujisawa T, Nishihara T (2007) Role of caspases in cleavage of lamin A/C and PARP during apoptosis in macrophages infected with a periodontopathic bacterium. J Med Microbiol 56:1399–1404

    Article  CAS  PubMed  Google Scholar 

  25. Zhao H, Zhao W, Lok K, Wang Z, Yin M (2014) A synergic role of caspase-6 and caspase-3 in Tau truncation at D421 induced by H2O2. Cell Mol Neurobiol 34:369–378

    Article  CAS  PubMed  Google Scholar 

  26. Veluthakal R, Amin R, Kowluru A (2004) Interleukin-1 induces posttranslational carboxymethylation and alterations in subnuclear distribution of lamin B in insulin-secreting RINm5F cells. Am J Physiol Cell Physiol 287:1152–1162

    Article  Google Scholar 

  27. Veluthakal R, Wadzinski BE, Kowluru A (2006) Localization of a nuclear serine/threonine protein phosphatase in insulin-secreting INS-1 cells: potential regulation by IL-1beta. Apoptosis 11:1401–1411

    Article  CAS  PubMed  Google Scholar 

  28. Reddy S, Comai L (2012) Lamin A, farnesylation and aging. Exp Cell Res 318:1–7

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  29. Burke B, Stewart CL (2013) The nuclear lamins: flexibility in function. Nat Rev Mol Cell Biol 14:13–24

    Article  CAS  PubMed  Google Scholar 

  30. Dechat T, Adam SA, Taimen P, Shimi T, Goldman RD (2010) Nuclear lamins. Cold Spring Harb Perspect Biol. doi:10.1101/cshperspect.a000547

    PubMed Central  PubMed  Google Scholar 

  31. Kowluru A (2000) Evidence for the carboxyl methylation of nuclear lamin-B in the pancreatic beta cell. Biochem Biophys Res Commun 268:249–254

    Article  CAS  PubMed  Google Scholar 

  32. Poitout V, Robertson RP (2008) Glucolipotoxicity: fuel excess and beta-cell dysfunction. Endocr Rev 29:351–366

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  33. Syed I, Kyathanahalli CN, Jayaram B, Govind S, Rhodes CJ, Kowluru RA, Kowluru A (2011) Increased phagocyte-like NADPH oxidase and ROS generation in type 2 diabetic ZDF rat and human islets: role of Rac1-JNK1/2 signaling pathway in mitochondrial dysregulation in the diabetic islet. Diabetes 60:2843–2852

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  34. Mohammed AM, Syeda K, Hadden T, Kowluru A (2013) Upregulation of phagocyte-like NADPH oxidase by cytokines in pancreatic beta-cells: attenuation of oxidative and nitrosative stress by 2-bromopalmitate. Biochem Pharmacol 85:109–114

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  35. Eizirik DL, Miani M, Cardozo AK (2013) Signalling danger: endoplasmic reticulum stress and the unfolded protein response in pancreatic islet inflammation. Diabetologia 56:234–241

    Article  CAS  PubMed  Google Scholar 

  36. Wang H, Kouri G, Wollheim CB (2005) ER stress and SREBP-1 activation are implicated in beta-cell glucolipotoxicity. J Cell Sci 118:3905–3915

    Article  CAS  PubMed  Google Scholar 

  37. Jonas JC, Bensellam M, Duprez J, Elouil H, Guiot Y, Pascal SM (2009) Glucose regulation of islet stress responses and beta-cell failure in type 2 diabetes. Diabetes Obes Metab 11:65–81

    Article  CAS  PubMed  Google Scholar 

  38. Lei X, Zhang S, Bohrer A, Barbour SE, Ramanadham S (2012) Role of calcium-independent phospholipase A(2)β in human pancreatic islet β-cell apoptosis. Am J Physiol Endocrinol Metab 303:1386–1395

    Article  Google Scholar 

  39. Maedler K, Sergeev P, Ris F, Oberholzer J, Joller-Jemelka HI, Spinas GA, Kaiser N, Halban PA, Donath MY (2002) Glucose-induced beta cell production of IL-1beta contributes to glucotoxicity in human pancreatic islets. J Clin Invest 110:851–860

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  40. Donath MY, Ehses JA, Maedler K, Schumann DM, Ellingsgaard H, Eppler E, Reinecke M (2005) Mechanisms of beta-cell death in type 2 diabetes. Diabetes 54:S108–S113

    Article  CAS  PubMed  Google Scholar 

  41. Boni-Schnetzler M, Thorne J, Parnaud G, Marselli L, Ehses JA, Kerr-Conte J, Pattou F, Halban PA, Weir GC, Donath MY (2008) Increased interleukin (IL)-1beta messenger ribonucleic acid expression in beta-cells of individuals with Type 2 diabetes and regulation of IL-1beta in human islets by glucose and autostimulation. J Clin Endocrinol Metab 93:4065–4074

    Article  PubMed Central  PubMed  Google Scholar 

  42. Huo J, Luo RH, Metz SA, Li G (2002) Activation of caspase-2 mediates the apoptosis induced by GTP-depletion in insulin-secreting (HIT-T15) cells. Endocrinology 143:1695–1704

    CAS  PubMed  Google Scholar 

  43. Yamada K, Ichikawa F, Ishiyama-Shigemoto S, Yuan X, Nonaka K (1999) Essential role of caspase-3 in apoptosis of mouse beta-cells transfected with human Fas. Diabetes 48:478–483

    Article  CAS  PubMed  Google Scholar 

  44. Hirota N, Otabe S, Nakayama H, Yuan X, Yamada K (2006) Sequential activation of caspases and synergistic beta-cell cytotoxicity by palmitate and anti-Fas antibodies. Life Sci 79:1312–1316

    Article  CAS  PubMed  Google Scholar 

  45. Di Matola T, D’Ascoli F, Luongo C, Bifulco M, Rossi G, Fenzi G, Vitale M (2001) Lovastatin-induced apoptosis in thyroid cells: involvement of cytochrome c and lamin B. Eur J Endocrinol 145:645–650

    Article  PubMed  Google Scholar 

  46. Chang SY, Hudon-Miller SE, Yang SH, Jung HJ, Lee JM, Farber E, Subramanian T, Andres DA, Spielmann HP, Hrycyna CA, Young SG, Fong LG (2012) Inhibitors of protein geranylgeranyltransferase-I lead to prelamin A accumulation in cells by inhibiting ZMPSTE24. J Lipid Res 53:1176–1182

    Article  CAS  PubMed Central  PubMed  Google Scholar 

Download references

Acknowledgments

This research was supported in part by a Merit Review award (to AK; 1BX000469) from the Department of Veterans Affairs, the National Institutes of Health (DK94201 and EY022230), the Juvenile Diabetes Research Foundation (5-2012-257), and Research Stimulation Funds from the Office of Vice President for Research-Wayne State University. AK is also the recipient of a Senior Research Career Scientist Award from the Department of VA (13S-RCS-006). KS is the recipient of Rumble Fellowship from Wayne State University. We thank Prof. Chris Newgard for providing INS-1 832/13 cells.

Conflict of interest

The authors declare no conflict of interests.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Anjaneyulu Kowluru.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Khadija, S., Veluthakal, R., Sidarala, V. et al. Glucotoxic and diabetic conditions induce caspase 6-mediated degradation of nuclear lamin A in human islets, rodent islets and INS-1 832/13 cells. Apoptosis 19, 1691–1701 (2014). https://doi.org/10.1007/s10495-014-1038-4

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s10495-014-1038-4

Keywords

Navigation