LaeA, a regulator of secondary metabolism in Aspergillus spp.
Journal: 2004/December - Eukaryotic cell
ISSN: 1535-9778
PUBMED: 15075281
Abstract:
Secondary metabolites, or biochemical indicators of fungal development, are of intense interest to humankind due to their pharmaceutical and/or toxic properties. We present here a novel Aspergillus nuclear protein, LaeA, as a global regulator of secondary metabolism in this genus. Deletion of laeA (DeltalaeA) blocks the expression of metabolic gene clusters, including the sterigmatocystin (carcinogen), penicillin (antibiotic), and lovastatin (antihypercholesterolemic agent) gene clusters. Conversely, overexpression of laeA triggers increased penicillin and lovastatin gene transcription and subsequent product formation. laeA expression is negatively regulated by AflR, a sterigmatocystin Zn2Cys6 transcription factor, in a unique feedback loop, as well as by two signal transduction elements, protein kinase A and RasA. Although these last two proteins also negatively regulate sporulation, DeltalaeA strains show little difference in spore production compared to the wild type, indicating that the primary role of LaeA is to regulate metabolic gene clusters.
Relations:
Content
Citations
(203)
References
(36)
Drugs
(2)
Chemicals
(7)
Organisms
(1)
Processes
(5)
Anatomy
(2)
Affiliates
(1)
Similar articles
Articles by the same authors
Discussion board
Eukaryot Cell 3(2): 527-535

LaeA, a Regulator of Secondary Metabolism in <em>Aspergillus</em> spp.

Department of Plant Pathology, University of Wisconsin—Madison, Madison, Wisconsin 53706
Corresponding author. Mailing address: Department of Plant Pathology, University of Wisconsin, 1630 Linden Dr., Madison, WI 53706. Phone: (608) 262-9795. Fax: (608) 263-2626. E-mail: ude.csiw.htaptnalp@kpn.
Received 2003 Nov 5; Accepted 2003 Dec 16.

Abstract

Secondary metabolites, or biochemical indicators of fungal development, are of intense interest to humankind due to their pharmaceutical and/or toxic properties. We present here a novel Aspergillus nuclear protein, LaeA, as a global regulator of secondary metabolism in this genus. Deletion of laeA (ΔlaeA) blocks the expression of metabolic gene clusters, including the sterigmatocystin (carcinogen), penicillin (antibiotic), and lovastatin (antihypercholesterolemic agent) gene clusters. Conversely, overexpression of laeA triggers increased penicillin and lovastatin gene transcription and subsequent product formation. laeA expression is negatively regulated by AflR, a sterigmatocystin Zn2Cys6 transcription factor, in a unique feedback loop, as well as by two signal transduction elements, protein kinase A and RasA. Although these last two proteins also negatively regulate sporulation, ΔlaeA strains show little difference in spore production compared to the wild type, indicating that the primary role of LaeA is to regulate metabolic gene clusters.

Abstract

A complex and fascinating aspect of fungal development is the production of secondary metabolites. These compounds, frequently associated with sporulation processes, are considered part of the chemical arsenal required for niche specialization and have garnered intense interest by virtue of their biotechnological and pharmaceutical applications (9, 11). Many of them display a broad range of useful antibiotic, antiviral, antitumor, antihypercholesterolemic, and immunosuppressant activities as well as less desirable phyto- and mycotoxic activities. Hawksworth's studies of fungal biodiversity led to the conclusion that nearly 1.5 million fungal species exist on Earth, with only 5% identified thus far (16). Thus, the potential for fungal secondary metabolite discovery is vast. Furthermore, the discovery of global regulators for fungal secondary metabolite production is critical, as it would allow for universal manipulation of secondary metabolite production.

A large number of known fungal secondary metabolites have been ascribed to the Ascomycete genus Aspergillus. Studies of Aspergillus nidulans have demonstrated the power of using a model system to elucidate the biochemistry and molecular genetics of fungal secondary metabolism, principally penicillin (PN, an antibiotic) and sterigmatocystin (ST, a carcinogen biochemically related to the agricultural contaminant aflatoxin) biosynthesis (6, 17). These studies have established several characteristics of fungal secondary metabolism, including the clustering of biosynthetic and regulatory genes and a genetic connection linking secondary metabolite biosynthesis with sporulation through a shared signal transduction pathway (9).

The discovery of the G protein-cyclic AMP (cAMP)-protein kinase A regulation of ST, aflatoxin, and other fungal secondary metabolites (4, 17, 29, 34) has been helpful for establishing a model of global regulation of secondary metabolism. However, all of the signal transduction mutants described in the literature have pleiotropic effects on fungi, the most notable effect being the gross impact on spore production and vegetative hyphal growth (1, 9, 18, 29). Similarly, mutations in other major regulators of ST biosynthesis, such as RcoA, a WD protein (19), and SpdA, or spermidine synthase (20), also have gross effects on fungal morphology. Thus, currently available Aspergillus mutants are so impaired in fungal development that further elucidation of genes that are specific for the regulation of secondary metabolism is difficult.

In a previous mutagenesis hunt, 23 A. nidulans mutants that displayed a phenotype of loss of ST production but had normal sporulation were isolated (8). Here we describe the identification of a gene called laeA that complements one of these mutants. laeA encodes a nuclear protein that is required for the expression of secondary metabolite genes. We propose that LaeA is a regulator of secondary metabolism in Aspergillus, as it is required not only for ST biosynthesis but also for PN biosynthesis and the biosynthesis of mycelial pigments in A. nidulans and gliotoxin and mycelial pigments in Aspergillus fumigatus. Furthermore, this protein is required for expression of the heterologous lovastatin (LOV) gene cluster in A. nidulans as well as for native LOV expression in Aspergillus terreus. Interestingly, the protein appears to be conserved in filamentous fungi, but it is not present in Saccharomyces cerevisiae, a fungus devoid of secondary metabolites. Unlike other genes that regulate secondary metabolism, the loss of laeA has a negligible impact on morphological developmental processes.

Acknowledgments

This research was funded by NSF grant MCB-9874646 to N.P.K.

Acknowledgments

REFERENCES

REFERENCES

References

  • 1. Adams, T. H., and J. H. Yu. 1998. Coordinate control of secondary metabolite production and asexual sporulation in Aspergillus nidulans.Curr. Opin. Microbiol.1:674-677. [[PubMed]
  • 2. Adams, T. H., and W. E. Timberlake. 1990. Developmental repression of growth and gene expression in Aspergillus.Proc. Natl. Acad. Sci. USA87:5405-5409.
  • 3. Alexander, N. J., T. H. Hohn, and S. P. McCormick. 1998. The TRI11 gene of Fusarium sporotrichioides encodes a cytochrome P-450 monooxygenase required for C-15 hydroxylation in trichothecene biosynthesis. Appl. Environ. Microbiol.64:221-225.
  • 4. Alspaugh, J. A., J. R. Perfect, and J. Heitman. 1997. Cryptococcus neoformans mating and virulence are regulated by the G-protein alpha subunit GPA1 and cAMP. Genes Dev.11:3206-3217.
  • 5. Belkacemi, L., R. C. Barton, V. Hopwood, and E. G. V. Evans. 1999. Determination of optimum growth conditions for gliotoxin production by Aspergillus fumigatus and development of a novel method for gliotoxin detection. Med. Mycol.37:227-233. [[PubMed]
  • 6. Brakhage, AA. 1998. Molecular regulation of beta-lactam biosynthesis in filamentous fungi. Microbiol. Mol. Biol. Rev.62:547-585. [Google Scholar]
  • 7. Brown, D. W., J. H. Yu, H. S. Kelkar, M. Fernandes, T. C. Nesbitt, N. P. Keller, T. H. Adams, and T. J. Leonard. 1996. Twenty-five coregulated transcripts define a sterigmatocystin gene cluster in Aspergillus nidulans.Proc. Natl. Acad. Sci. USA93:1418-1422.
  • 8. Butchko, R. A., T. H. Adams, and N. P. Keller. 1999. Aspergillus nidulans mutants defective in stc gene cluster regulation. Genetics153:715-720.
  • 9. Calvo, A. M., R. A. Wilson, J. W. Bok, and N. P. Keller. 2002. Relationship between secondary metabolism and fungal development. Microbiol. Mol. Biol. Rev.66:447-459.
  • 10. Chalfie, M., Y. Tu, G. Euskirchen, W. W. Ward, and D. C. Prasher. 1994. Green fluorescent protein as a marker for gene expression. Science263:802-805. [[PubMed]
  • 11. Demain, A., and AFang. 2000. The natural functions of secondary metabolites. Adv. Biochem. Eng. Biotechnol.69:1-39. [[PubMed][Google Scholar]
  • 12. Fernandes, M., N. P. Keller, and T. H. Adams. 1998. Sequence-specific binding by Aspergillus nidulans AflR, a C6 zinc cluster protein regulating mycotoxin biosynthesis. Mol. Microbiol.28:1355-1365. [[PubMed]
  • 13. Fernandez-Abalos, J. M., H. Fox, C. Pitt, B. Wells, and J. H. Doonan. 1998. Plant-adapted green fluorescent protein is a versatile vital reporter for gene expression, protein localization and mitosis in the filamentous fungus Aspergillus nidulans.Mol. Microbiol.27:121-130. [[PubMed]
  • 14. Hah, H. 1993. Differential action of cercoran and topsin-M on sensitive and tolerant strains of toxic fungi. Cryptogamie Mycologie14:61-68. [PubMed]
  • 15. Hamahata, A., Y. Takata, T. Gomi, and M. Fujioka. 1996. Probing the S-adenosylmethionine-binding site of rat guanidinoacetate methyltransferase. Effect of site-directed mutagenesis of residues that are conserved across mammalian non-nucleic acid methyltransferases. Biochem. J.317:141-145.
  • 16. Hawksworth, DL. 2001. The magnitude of fungal diversity: the 1.5 million species estimate revisited. Mycol. Res.105:1422-1432. [PubMed][Google Scholar]
  • 17. Hicks, J. K., K. Shimizu, and N. P. Keller. 2002. Genetics and biosynthesis of aflatoxins and sterigmatocystin, p. 55-69. In F. Kempken and J. W. Bennett (ed.), The mycota, vol. XI. Springer-Verlag, Berlin, Germany.
  • 18. Hicks, J. K., J.-H. Yu, N. P. Keller, and T. H. Adams. 1997. Aspergillus sporulation and mycotoxin production both require inactivation of the FadA G alpha protein-dependent signaling pathway. EMBO J.16:4916-4923.
  • 19. Hicks, J., R. A. Lockington, J. Strauss, D. Dieringer, C. P. Kubicek, J. Kelly, and N. Keller. 2001. RcoA has pleiotropic effects on Aspergillus nidulans cellular development. Mol. Microbiol.39:1482-1493. [[PubMed]
  • 20. Jin, Y., J. W. Bok, D. Guzman-de-Peña, and N. P. Keller. 2002. Requirement of spermidine for developmental transitions in Aspergillus nidulans.Mol. Microbiol.46:801-812. [[PubMed]
  • 21. Kennedy, J., K. Auclair, S. G. Kendrew, C. Park, J. C. Vederas, and C. R. Hutchinson. 1999. Modulation of polyketide synthase activity by accessory proteins during lovastatin biosynthesis. Science284:1368-1372. [[PubMed]
  • 22. Langfelder, K., M. Streibel, B. Jahn, G. Haase, and A. A. Brakhage. 2003. Biosynthesis of fungal melanins and their importance for human pathogenic fungi. Fungal Genet. Biol.38:143-158. [[PubMed]
  • 23. Lee, B. N., and T. H. Adams. 1994. Overexpression of flbA, an early regulator of Aspergillus asexual sporulation, leads to activation of brlA and premature initiation of development. Mol. Microbiol.14:323-334. [[PubMed]
  • 24. Miller, B. L., K. Y. Miller, and W. E. Timberlake. 1985. Direct and indirect gene replacements in Aspergillus nidulans.Mol. Cell. Biol.5:1714-1721.
  • 25. Mowen, K. A., J. Tang, W. Zhu, B. T. Schurter, K. Shuai, H. R. Herschman, and M. David. 2001. Arginine methylation of STAT1 modulates IFN alpha/beta-induced transcription. Cell104:731-741. [[PubMed]
  • 26. Pontecorvo, G., J. A. Roper, L. M. Hemmons, K. P. Macdonald, and A. W. J. Bufton. 1953. The genetics of Aspergillus nidulans.Adv. Genet.5:141-238. [[PubMed]
  • 27. Rosén, S., J.-H. Yu, and T. H. Adams. 1999. The Aspergillus nidulans sfaD gene encodes a G protein beta subunit that is required for normal growth and repression of sporulation. EMBO J.18:5592-5600.
  • 28. Sambrook, J., E. F. Fritsch, and T. Maniatis. 1989. Molecular cloning: a laboratory manual. Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y.
  • 29. Shimizu, K., and N. P. Keller. 2001. Genetic involvement of a cAMP-dependent protein kinase in a G protein signaling pathway regulating morphological and chemical transitions in Aspergillus nidulans.Genetics157:591-600.
  • 30. Shimizu, K., J.-K. Hicks, T.-P. Huang, and N. P. Keller. 2003. Pka, Ras and RGS protein interactions regulate activity of AflR, a Zn(II)2Cys6 transcription factor in Aspergillus nidulans.Genetics165:1095-1104.
  • 31. Sienko, M., and APaszewski. 1999. The metG gene of Aspergillus nidulans encoding cystathionine beta-lyase: cloning and analysis. Curr. Genet.35:638-646. [[PubMed][Google Scholar]
  • 32. Skory, C. D., P. K. Chang, J. Cary, and J. E. Linz. 1992. Isolation and characterization of a gene from Aspergillus parasiticus associated with the conversion of versicolorin A to sterigmatocystin in aflatoxin biosynthesis. Appl. Environ. Microbiol.58:3527-3537.
  • 33. Som, T., and V. S. Kolaparthi. 1994. Developmental decisions in Aspergillus nidulans are modulated by Ras activity. Mol. Cell. Biol.14:5333-5348.
  • 34. Tag, A., J. Hicks, G. Garifullina, M. Beremand, and N. Keller. 2000. G-protein signalling mediates differential production of toxic secondary metabolites. Mol. Microbiol.38:658-665. [[PubMed]
  • 35. Tamaru, H., and E. U. Selker. 2001. A histone H3 methyltransferase controls DNA methylation in Neurospora crassa.Nature414:277-283. [[PubMed]
  • 36. Tilburn, J., S. Sarkar, D. A. Widdick, E. A. Espeso, M. Orejas, J. Mungroo, M. A. Penalva, and H. N. Arst, Jr. 1995. The Aspergillus PacC zinc finger transcription factor mediates regulation of both acid- and alkaline-expressed genes by ambient pH. EMBO J.14:779-790.
  • 37. Umeyama, T., P. C. Lee, and S. Horinouchi. 2002. Protein serine/threonine kinases in signal transduction for secondary metabolism and morphogenesis in Streptomyces.Appl. Microbiol. Biotechnol.59:419-425. [[PubMed]
  • 38. Wang, L., and L. C. Vining. 2003. Control of growth, secondary metabolism and sporulation in Streptomyces venezuelae ISP5230 by jadW (1), a member of the afsA family of gamma-butyrolactone regulatory genes. Microbiology149:1991-2004. [[PubMed]
  • 39. Wieser, J., and T. H. Adams. 1995. flbD encodes a Myb-like DNA-binding protein that coordinates initiation of Aspergillus nidulans conidiophore development. Genes Dev.9:491-502. [[PubMed]
  • 40. Yu, J.-H., J. Wieser, and T. H. Adams. 1996. The Aspergillus FlbA RGS domain protein antagonizes G protein signaling to block proliferation and allow development. EMBO J.15:5184-5190.
Collaboration tool especially designed for Life Science professionals.Drag-and-drop any entity to your messages.