scholarly journals Induced fit on heme binding to the Pseudomonas aeruginosa cytoplasmic protein (PhuS) drives interaction with heme oxygenase (HemO)

2012 ◽  
Vol 109 (15) ◽  
pp. 5639-5644 ◽  
Author(s):  
M. J. O'Neill ◽  
M. N. Bhakta ◽  
K. G. Fleming ◽  
A. Wilks
2020 ◽  
Vol 477 (24) ◽  
pp. 4785-4796
Author(s):  
Jia Wang ◽  
Qi Guo ◽  
Xiaoyi Li ◽  
Xiao Wang ◽  
Lin Liu

Plant tetrapyrroles, including heme and bilins, are synthesized in plastids. Heme oxygenase (HO) catalyzes the oxidative cleavage of heme to the linear tetrapyrrole biliverdin as the initial step in bilin biosynthesis. Besides the canonical α-helical HO that is conserved from prokaryotes to human, a subfamily of non-canonical dimeric β-barrel HO has been found in bacteria. In this work, we discovered that the Arabidopsis locus AT3G03890 encodes a dimeric β-barrel protein that is structurally related to the putative non-canonical HO and is located in chloroplasts. The recombinant protein was able to bind and degrade heme in a manner different from known HO proteins. Crystal structure of the heme–protein complex reveals that the heme-binding site is in the interdimer interface and the heme iron is co-ordinated by a fixed water molecule. Our results identify a new protein that may function additionally in the tetrapyrrole biosynthetic pathway.


2001 ◽  
Vol 183 (21) ◽  
pp. 6394-6403 ◽  
Author(s):  
Melanie Ratliff ◽  
Wenming Zhu ◽  
Rahul Deshmukh ◽  
Angela Wilks ◽  
Igor Stojiljkovic

ABSTRACT The oxidative cleavage of heme to release iron is a mechanism by which some bacterial pathogens can utilize heme as an iron source. ThepigA gene of Pseudomonas aeruginosa is shown to encode a heme oxygenase protein, which was identified in the genome sequence by its significant homology (37%) with HemO ofNeisseria meningitidis. When the gene encoding the neisserial heme oxygenase, hemO, was replaced withpigA, we demonstrated that pigA could functionally replace hemO and allow for heme utilization by neisseriae. Furthermore, when pigA was disrupted by cassette mutagenesis in P. aeruginosa, heme utilization was defective in iron-poor media supplemented with heme. This defect could be restored both by the addition of exogenous FeSO4, indicating that the mutant did not have a defect in iron metabolism, and by in trans complementation with pigA from a plasmid with an inducible promoter. The PigA protein was purified by ion-exchange chromotography. The UV-visible spectrum of PigA reconstituted with heme showed characteristics previously reported for other bacterial and mammalian heme oxygenases. The heme-PigA complex could be converted to ferric biliverdin in the presence of ascorbate, demonstrating the need for an exogenous reductant. Acidification and high-performance liquid chromatography analysis of the ascorbate reduction products identified a major product of biliverdin IX-β. This differs from the previously characterized heme oxygenases in which biliverdin IX-α is the typical product. We conclude that PigA is a heme oxygenase and may represent a class of these enzymes with novel regiospecificity.


2018 ◽  
Vol 28 (6) ◽  
pp. 1024-1029 ◽  
Author(s):  
Dongdong Liang ◽  
Elizabeth Robinson ◽  
Kellie Hom ◽  
Wenbo Yu ◽  
Nam Nguyen ◽  
...  

2020 ◽  
Vol 295 (50) ◽  
pp. 17227-17240 ◽  
Author(s):  
Liu Liu ◽  
Arti B. Dumbrepatil ◽  
Angela S. Fleischhacker ◽  
E. Neil G. Marsh ◽  
Stephen W. Ragsdale

Heme oxygenase-2 (HO2) and -1 (HO1) catalyze heme degradation to biliverdin, CO, and iron, forming an essential link in the heme metabolism network. Tight regulation of the cellular levels and catalytic activities of HO1 and HO2 is important for maintaining heme homeostasis. HO1 expression is transcriptionally regulated; however, HO2 expression is constitutive. How the cellular levels and activity of HO2 are regulated remains unclear. Here, we elucidate the mechanism of post-translational regulation of cellular HO2 levels by heme. We find that, under heme-deficient conditions, HO2 is destabilized and targeted for degradation, suggesting that heme plays a direct role in HO2 regulation. HO2 has three heme binding sites: one at its catalytic site and the others at its two heme regulatory motifs (HRMs). We report that, in contrast to other HRM-containing proteins, the cellular protein level and degradation rate of HO2 are independent of heme binding to the HRMs. Rather, under heme deficiency, loss of heme binding to the catalytic site destabilizes HO2. Consistently, an HO2 catalytic site variant that is unable to bind heme exhibits a constant low protein level and an enhanced protein degradation rate compared with the WT HO2. Finally, HO2 is degraded by the lysosome through chaperone-mediated autophagy, distinct from other HRM-containing proteins and HO1, which are degraded by the proteasome. These results reveal a novel aspect of HO2 regulation and deepen our understanding of HO2's role in maintaining heme homeostasis, paving the way for future investigation into HO2's pathophysiological role in heme deficiency response.


2019 ◽  
Vol 129 ◽  
pp. 64-67
Author(s):  
Jingming Zhao ◽  
Dongdong Liang ◽  
Elizabeth Robinson ◽  
Fengtian Xue

2016 ◽  
Vol 59 (14) ◽  
pp. 6929-6942 ◽  
Author(s):  
Geoffrey A. Heinzl ◽  
Weiliang Huang ◽  
Wenbo Yu ◽  
Bennett J. Giardina ◽  
Yue Zhou ◽  
...  

Biochemistry ◽  
1995 ◽  
Vol 34 (41) ◽  
pp. 13407-13411 ◽  
Author(s):  
Stephan Immenschuh ◽  
Shin-ichiro Iwahara ◽  
Hiroyuki Satoh ◽  
Christina Nell ◽  
Norbert Katz ◽  
...  

2020 ◽  
Author(s):  
Liu Liu ◽  
Arti B. Dumbrepatil ◽  
Angela S. Fleischhacker ◽  
E. Neil G. Marsh ◽  
Stephen W. Ragsdale

ABSTRACTHeme oxygenase-2 (HO2) and −1 (HO1) catalyze heme degradation to biliverdin, CO, and iron, forming an essential link in the heme metabolism network. Tight regulation of the cellular levels and catalytic activities of HO1 and HO2 is important for maintaining heme homeostasis. While transcriptional control of HO1 expression has been well-studied, how the cellular levels and activity of HO2 are regulated remains unclear. Here, the mechanism of post-translational regulation of cellular HO2 level by heme is elucidated. Under heme deficient conditions, HO2 is destabilized and targeted for degradation. In HO2, three heme binding sites are potential targets of heme-dependent regulation: one at its catalytic site; the others at its two heme regulatory motifs (HRMs). We report that, in contrast to other HRM-containing proteins, the cellular protein level and degradation rate of HO2 are independent of heme binding to the HRMs. Rather, under heme deficiency, loss of heme binding to the catalytic site destabilizes HO2. Consistently, a HO2 catalytic site variant that is unable to bind heme exhibits a constant low protein level and an enhanced protein degradation rate compared to the wild-type HO2. However, cellular heme overload does not affect HO2 stability. Finally, HO2 is degraded by the lysosome through chaperone-mediated autophagy, distinct from other HRM-containing proteins and HO1, which are degraded by the proteasome. These results reveal a novel aspect of HO2 regulation and deepen our understanding of HO2’s role in maintaining heme homeostasis, paving the way for future investigation into HO2’s pathophysiological role in heme deficiency response.


Sign in / Sign up

Export Citation Format

Share Document