53
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Isolation and Characterization of a Hybrid Respiratory Supercomplex Consisting of Mycobacterium tuberculosis Cytochrome bcc and Mycobacterium smegmatis Cytochrome aa 3*

      research-article

      Read this article at

      Bookmark
          There is no author summary for this article yet. Authors can add summaries to their articles on ScienceOpen to make them more accessible to a non-specialist audience.

          Abstract

          Background: Mycobacteria have no soluble cytochrome c; the electron transfer chain involves a Complex III-IV “supercomplex.”

          Results: Expression of the M. tuberculosis Complex III in M. smegmatis lacking native complex yields a functional hybrid supercomplex.

          Conclusion: This supercomplex is a dimer of protomers containing two each of hemes A, B, and C.

          Significance: This is the first purification of respiratory Complex III or IV from Mycobacterium.

          Abstract

          Recently, energy production pathways have been shown to be viable antitubercular drug targets to combat multidrug-resistant tuberculosis and eliminate pathogen in the dormant state. One family of drugs currently under development, the imidazo[1,2- a]pyridine derivatives, is believed to target the pathogen's homolog of the mitochondrial bc 1 complex. This complex, denoted cytochrome bcc, is highly divergent from mitochondrial Complex III both in subunit structure and inhibitor sensitivity, making it a good target for drug development. There is no soluble cytochrome c in mycobacteria to transport electrons from the bcc complex to cytochrome oxidase. Instead, the bcc complex exists in a “supercomplex” with a cytochrome aa 3-type cytochrome oxidase, presumably allowing direct electron transfer. We describe here purification and initial characterization of the mycobacterial cytochrome bcc-aa 3 supercomplex using a strain of M. smegmatis that has been engineered to express the M. tuberculosis cytochrome bcc. The resulting hybrid supercomplex is stable during extraction and purification in the presence of dodecyl maltoside detergent. It is hoped that this purification procedure will potentiate functional studies of the complex as well as crystallographic studies of drug binding and provide structural insight into a third class of the bc complex superfamily.

          Related collections

          Most cited references26

          • Record: found
          • Abstract: found
          • Article: not found

          Genes required for mycobacterial growth defined by high density mutagenesis.

          Despite over a century of research, tuberculosis remains a leading cause of infectious death worldwide. Faced with increasing rates of drug resistance, the identification of genes that are required for the growth of this organism should provide new targets for the design of antimycobacterial agents. Here, we describe the use of transposon site hybridization (TraSH) to comprehensively identify the genes required by the causative agent, Mycobacterium tuberculosis, for optimal growth. These genes include those that can be assigned to essential pathways as well as many of unknown function. The genes important for the growth of M. tuberculosis are largely conserved in the degenerate genome of the leprosy bacillus, Mycobacterium leprae, indicating that non-essential functions have been selectively lost since this bacterium diverged from other mycobacteria. In contrast, a surprisingly high proportion of these genes lack identifiable orthologues in other bacteria, suggesting that the minimal gene set required for survival varies greatly between organisms with different evolutionary histories.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            The cytochrome bd respiratory oxygen reductases.

            Cytochrome bd is a respiratory quinol: O₂ oxidoreductase found in many prokaryotes, including a number of pathogens. The main bioenergetic function of the enzyme is the production of a proton motive force by the vectorial charge transfer of protons. The sequences of cytochromes bd are not homologous to those of the other respiratory oxygen reductases, i.e., the heme-copper oxygen reductases or alternative oxidases (AOX). Generally, cytochromes bd are noteworthy for their high affinity for O₂ and resistance to inhibition by cyanide. In E. coli, for example, cytochrome bd (specifically, cytochrome bd-I) is expressed under O₂-limited conditions. Among the members of the bd-family are the so-called cyanide-insensitive quinol oxidases (CIO) which often have a low content of the eponymous heme d but, instead, have heme b in place of heme d in at least a majority of the enzyme population. However, at this point, no sequence motif has been identified to distinguish cytochrome bd (with a stoichiometric complement of heme d) from an enzyme designated as CIO. Members of the bd-family can be subdivided into those which contain either a long or a short hydrophilic connection between transmembrane helices 6 and 7 in subunit I, designated as the Q-loop. However, it is not clear whether there is a functional consequence of this difference. This review summarizes current knowledge on the physiological functions, genetics, structural and catalytic properties of cytochromes bd. Included in this review are descriptions of the intermediates of the catalytic cycle, the proposed site for the reduction of O₂, evidence for a proton channel connecting this active site to the bacterial cytoplasm, and the molecular mechanism by which a membrane potential is generated. 2011 Elsevier B.V. All rights reserved.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Discovery of Q203, a potent clinical candidate for the treatment of tuberculosis.

              New therapeutic strategies are needed to combat the tuberculosis pandemic and the spread of multidrug-resistant (MDR) and extensively drug-resistant (XDR) forms of the disease, which remain a serious public health challenge worldwide. The most urgent clinical need is to discover potent agents capable of reducing the duration of MDR and XDR tuberculosis therapy with a success rate comparable to that of current therapies for drug-susceptible tuberculosis. The last decade has seen the discovery of new agent classes for the management of tuberculosis, several of which are currently in clinical trials. However, given the high attrition rate of drug candidates during clinical development and the emergence of drug resistance, the discovery of additional clinical candidates is clearly needed. Here, we report on a promising class of imidazopyridine amide (IPA) compounds that block Mycobacterium tuberculosis growth by targeting the respiratory cytochrome bc1 complex. The optimized IPA compound Q203 inhibited the growth of MDR and XDR M. tuberculosis clinical isolates in culture broth medium in the low nanomolar range and was efficacious in a mouse model of tuberculosis at a dose less than 1 mg per kg body weight, which highlights the potency of this compound. In addition, Q203 displays pharmacokinetic and safety profiles compatible with once-daily dosing. Together, our data indicate that Q203 is a promising new clinical candidate for the treatment of tuberculosis.
                Bookmark

                Author and article information

                Journal
                J Biol Chem
                J. Biol. Chem
                jbc
                jbc
                JBC
                The Journal of Biological Chemistry
                American Society for Biochemistry and Molecular Biology (11200 Rockville Pike, Suite 302, Rockville, MD 20852-3110, U.S.A. )
                0021-9258
                1083-351X
                5 June 2015
                10 April 2015
                10 April 2015
                : 290
                : 23
                : 14350-14360
                Affiliations
                From the []State University of New York Upstate Medical University, Syracuse, New York 13210,
                the [§ ]Institut Pasteur Korea, Sampyeong-dong, Seongnam-si, Gyeonggi-do 463-400, Korea, and
                the []Lee Kong Chian School of Medicine and School of Biological Sciences, Nanyang Technological University, Singapore 639798, Singapore
                Author notes
                [6 ] To whom correspondence may be addressed: Dept. of Biochemistry and Molecular Biology, SUNY Upstate Medical University, 750 E. Adams St., Syracuse, NY 13210. Tel.: 315-464-8751; Fax: 315-464-8750; E-mail: berrye@ 123456upstate.edu .
                [7 ] To whom correspondence may be addressed: Dept. of Biochemistry and Molecular Biology, SUNY Upstate Medical University, 750 E. Adams St., Syracuse, NY 13210. Tel.: 315-464-8751; Fax: 315-464-8750; E-mail: berryl@ 123456upstate.edu .
                [1]

                Both authors contributed equally to this work.

                [2]

                Supported by National Research Foundation of Korea Grant NRF-2013R1A6A3A03026543. Present address: College of Pharmacy, Division of Life and Pharmaceutical Sciences, Ewha Womans University, Seoul, Republic of Korea 120-750.

                [3]

                Supported by a National Research foundation of Korea (NRF) grant funded by the Korean government (MSIP) 2007-00559, Gyeonggi-do, KISTI, and National Research Foundation of Korea Grant 2014R1A1A1007464.

                [4]

                Supported by an NRF grant funded by the Korean government (MSIP) 2007-00559, Gyeonggi-do, KISTI, and a Lee Kong Chian School of Medicine, Nanyang Technological University Start-Up Grant.

                [5]

                Supported by the State University of New York.

                Author information
                http://orcid.org/0000-0003-0297-0150
                http://orcid.org/0000-0001-8929-0259
                Article
                M114.624312
                10.1074/jbc.M114.624312
                4505504
                25861988
                3fdbde28-3735-47d7-89ac-ff8fd35da012
                © 2015 by The American Society for Biochemistry and Molecular Biology, Inc.

                Author's Choice—Final version free via Creative Commons CC-BY license.

                History
                : 6 November 2014
                : 1 April 2015
                Categories
                Bioenergetics

                Biochemistry
                electron transfer complex,enzyme purification,mycobacterium smegmatis,mycobacterium tuberculosis,respiratory chain,q203,cytochrome bcc,respiratory supercomplex

                Comments

                Comment on this article