Cytolethal distending toxin

Last updated
Cytolethal distending toxin
PDB protein 1sr4 (HdCDT).jpg
Crystal structure of the fully assembled Haemophilus ducreyi cytolethal distending toxin
Identifiers
SymbolCDT
Pfam PF03498
Pfam clan CL0066
InterPro IPR003558
Available protein structures:
Pfam   structures / ECOD  
PDB RCSB PDB; PDBe; PDBj
PDBsum structure summary

Cytolethal distending toxins (abbreviated CDTs) are a class of hetero trimeric toxins produced by certain gram-negative bacteria that display DNase activity. [1] [2] These toxins trigger G2/M cell cycle arrest in specific mammalian cell lines, leading to the enlarged or distended cells for which these toxins are named. [3] Affected cells die by apoptosis. [1]

Contents

Each toxin consists of three distinct subunits named alphabetically in the order that their coding genes appear in the cdt operon . Cytolethal distending toxins are classified as AB toxins, with an active ("A") subunit that directly damages DNA and a binding ("B") subunit that helps the toxin attach to the target cells. CdtB is the active subunit and a homolog to mammalian DNase I, whereas CdtA and CdtC make up the binding subunit. [4]

Cytolethal distending toxins are produced by gram-negative pathogenic bacteria from the phylum Pseudomonadota . Many of these bacteria, including Shigella dysenteriae , Haemophilus ducreyi , and Escherichia coli , infect humans. Bacteria that produce CDTs often persistently colonize their host. [1]

History

The first recorded observation of a cytolethal-distending toxin was in 1987 in a pathogenic strain in E. coli isolated from a young patient. [3] Later that year, scientists W.M. Johnson and H. Lior published the journal article "Production of Shiga toxin and a cytolethal distending toxin (CLDT) by serogroups of Shigella spp." in Microbiology Letters. [1] The discovery of other bacteria producing CDT toxins continues to this day.

In 1994 Scott and Kaper cloned and sequenced a cdt operon from another E. coli strain, publishing in Infection and Immunity. [1] [5] The three genes discovered were denoted cdtA, cdtB, and cdtC. [5]

In 1997, the first paper of many to show G2/M cell cycle arrest caused by a cytolethal distending toxin was published in Molecular Microbiology . [1] The study focused on another E. coli strain. This paper was followed by a 1999 publication in Infectious Immunity, which demonstrated that H. ducreyi CDT causes cell death via apoptosis. This finding was also confirmed for other cytolethal distending toxins in subsequent studies.

The discovery of the homology of cdtB to mammalian DNase I and the current AB model for the toxin were published in early 2000. [2] [6] Further research and the publication of crystal structures for the CDT toxins from two different species continues to support this model. [1]

Sources

All known cytolethal distending toxins are produced by gram-negative bacteria in the Gammaproteobacteria and Campylobacterota . In several cases, the bacteria producing CDT are human pathogens. Medically important CDT producers include: [1]

CDT-producing bacteria are often associated with mucosal linings, such as those in the stomach and intestines, and with persistent infections. The toxins are either secreted freely or associated with the membrane of the producing bacteria. [1]

Nomenclature

Individual cytolethal distending toxins are named for the bacterial species that they are isolated from. As of 2011, most scientists have adopted the practice of placing the first letter of both the genus and species in front of the toxin name to reflect its source (i.e., the CDT from Haemaphilus ducreyi is referred to as HdCDT). [1] [7] If several subspecies produce different toxins, as in the case of E. coli, Roman numerals may be added after the second letter. [7] Both complete toxins and individual subunits are labeled using this convention.

In response to the continued discovery of additional cytolethal distending toxins, a 2011 review has proposed that the toxin names be expanded to include the first three letters of the species (i.e., HducCDT for Haemaphilus ducreyi CDT). [1]

Cellular effects

CDT toxins are genotoxins capable of directly damaging DNA in target cells. They are the only AB-type toxins discovered that display DNase activity, allowing them to introduce breaks into the target cell's DNA. [1] [4]

In many cell lines including human fibroblasts, epithelial cells, endothelial cells, and keratinocytes, CDTs cause G2/M cell cycle arrest, cytoplasmic distension, and eventual cell death via apoptosis. [1] [3] [8] Most publications attribute the G2/M cycle arrest to the buildup of irreversible DNA damage from the toxin's DNase activity as the trigger for the G2/M cell cycle arrest, but other research suggests that this model is incomplete. [8] The cytoplasmic distension is a direct result of the G2/M cell cycle arrest. The cell enlarges in preparation for mitosis, but cannot divide to restore its normal size. Aside from classical apoptosis, signs of cellular senescence has also been observed in normal and cancer cell lines (fibroblasts, HeLa and U2-OS) after CDT intoxication [9]

In lymphocytes, cell death occurs quickly and is not preceded by significant cytoplasmic distension. [8] The ability of these toxins to effect lymphocytes differently may be advantageous to the bacteria that utilize these toxins, but the mechanism behind this phenomenon is not yet well understood.

Toxin structure

The active, assembled toxin is a tripartite structure with three distinct subunits- CdtA, CdtB, and CdtC. In terms of function, it is an AB toxin. In this context, the CdtB subunit is actually the catalytically active "A" subunit, and the CdtA and CdtC together form the binding "B" subunit, which helps the toxin bind and enter target cells. [6] Some literature refers to the toxin structure as AB2 to reflect the presence of both CdtA and CdtC.

Different from all other CDTs, Salmonella enterica serovar Typhi CDT (SeCDT) has no CdtA and CdtC homologues. However, encoded closely to the active subunit cdtb, the Pertussis-like toxin A and B (pltA/pltB) have been shown to be essential for cellular intoxication. [10] PltA and PltB have a different structure from CdtA and CdtC, thus promoting CdtB activity in a different way. Both PltA and PltB have been found to bind directly to CdtB in vitro. [10] In addition, different from all other CDTs, Salmonella genotoxin is produced only upon bacterial internalization in infected cells, thus the SeCDT traffic may differ remarkably from the canonical ones.

CdtB

CdtB is considered the active subunit of the CDT holotoxin. Microinjection of CdtB into susceptible cells without CdtA or CdtC results in the G2/M cell cycle arrest and cytoplasmic distension characteristic of CDT toxins. [2] The structure of CdtB is well-conserved between different bacteria. The CdtB subunit is the most sequentially conserved between species. [4] The molecular weight of CdtB ranges from 28 kDa to 29 kDa, depending on the species. [1]

As the active subunit, CdtB is termed the "A" subunit according to the AB toxin model. [1] This confusing nomenclature is due to the naming of the toxin's subunits before their individual functions were understood.

Activity

CdtB exhibits at least two enzymatic activities- DNase activity capable of introducing double-strand breaks in DNA, and a phosphatase activity that resembles phosphatidylinositol 3,4,5-triphosphatase. [2] [8] Both activities can be demonstrated in vitro in the absence of the other two subunits. [11] The relative importance of each activity in vivo is unclear. [11] Mutations that reduce either activity also reduce the toxin's ability to induce G2/M phase arrest in at least some of the susceptible cell lines. [2] [8]

Similarities to mammalian DNase I

CdtB is functionally homologous to mammalian DNase I and contains a conserved pentapeptide sequence found in all DNase I enzymes to date. [2] In addition, several residues critical to DNase I's ability to break the phosphodiester bonds in the DNA backbone are found in the CdtB structure. A 2002 paper studying the effect of point mutations on five of these residues found that four of the five mutations tested abolished both CdtB's ability to degrade DNA in cell-free extracts and to cause G2/M arrest upon microinjection. The fifth mutation moderately reduced CdtB's activity. [2]

CdtA and CdtC

CdtA and CdtC make up the B subunit of the CDT holotoxin responsible for targeting the CdtB against susceptible cells. [6] Neither subunit appears highly conserved, with sequence identities between different species often lower than 30%. [4] The molecular weight of CdtA ranges from 23 kDa to 30 kDa, whereas CdtC ranges from 19 kDa to 21 kDa depending on the species. [1]

Activity

CdtA and CdtC are both believed to bind to the surface of target cells. The exact mechanism of this binding is unclear, and may not be conserved between CDT toxins from different species. [1] [11] Proposed targets of CdtA and CdtC binding have included cholesterol, N-linked glycans, and glycosphingolipids. [11] Current research has produced conflicting results on the actual importance of these proposed targets. [1] [11] Both CdtA and CdtC contain lectin domains, [12] suggesting that the toxin may bind via carbohydrates on the target cell's surface, whereas other research has suggested that the targets are surface proteins. [1]

Notes

  1. 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 Jinadasa RN, Bloom SE, Weiss RS, Duhamel GE (July 2011). "Cytolethal distending toxin: a conserved bacterial genotoxin that blocks cell cycle progression, leading to apoptosis of a broad range of mammalian cell lineages". Microbiology. 157 (Pt 7): 1851–75. doi:10.1099/mic.0.049536-0. PMC   3167888 . PMID   21565933.
  2. 1 2 3 4 5 6 7 Cherilyn A. Elwell; Lawrence A. Dreyfus (2000). "DNase I homologous residues in CdtB are critical for cytolethal distending toxin-mediated cell cycle arrest". Molecular Microbiology. 37 (4): 952–963. doi: 10.1046/j.1365-2958.2000.02070.x . PMID   10972814. S2CID   10087538.
  3. 1 2 3 Dreyfus, Lawrence A. (2003), "Cyotlethal Distending Toxin", in D. Burns; et al. (eds.), Bacterial Protein Toxins, Washington, DC: ASM Press, pp. 257–270
  4. 1 2 3 4 Guerra L, Cortes-Bratti X, Guidi R, Frisan T (March 2011). "The biology of the cytolethal distending toxins". Toxins. 3 (12): 172–90. doi: 10.3390/toxins3030172 . PMC   3202825 . PMID   22069704.
  5. 1 2 Scott DA, Kaper JB (January 1994). "Cloning and sequencing of the genes encoding Escherichia coli cytolethal distending toxin". Infection and Immunity. 62 (1): 244–51. doi:10.1128/iai.62.1.244-251.1994. PMC   186093 . PMID   8262635.
  6. 1 2 3 Lara-Tejero M, Galán JE (July 2001). "CdtA, CdtB, and CdtC form a tripartite complex that is required for cytolethal distending toxin activity". Infection and Immunity. 69 (7): 4358–65. doi:10.1128/IAI.69.7.4358-4365.2001. PMC   98507 . PMID   11401974.
  7. 1 2 Ximena Cortes-Bratti; Teresa Frisan; Monica Thelestam (2001). "The Cytolethal Distending Toxins Induce DNA Damage and Cell Cycle Arrest". Toxicon. 39 (11): 1729–1736. doi:10.1016/S0041-0101(01)00159-3. PMID   11595635.
  8. 1 2 3 4 5 Bruce J. Shenker; Mensur Dlakic; Lisa P. Walker; Dave Besack; Eileen Jaffe; Ed LaBelle; Kathleen Boesze-Battaglia (2007). "A Novel Mode of Action for a Microbial-Derived Immunotoxin: The Cytolethal Distending Toxin Subunit B Exhibits Phosphatidylinositol 3,4,5-Triphosphate Phosphatase Activity". The Journal of Immunology. 178 (8): 5099–5108. doi:10.4049/jimmunol.178.8.5099. PMC   4472023 . PMID   17404292.
  9. Blazkova H, Krejcikova K, Moudry P, Frisan T, Hodny Z, Bartek J (January 2010). "Bacterial intoxication evokes cellular senescence with persistent DNA damage and cytokine signalling". Journal of Cellular and Molecular Medicine. 14 (1–2): 357–67. doi:10.1111/j.1582-4934.2009.00862.x. PMC   3837606 . PMID   19650831.
  10. 1 2 Spanò S, Ugalde JE, Galán JE (January 2008). "Delivery of a Salmonella Typhi exotoxin from a host intracellular compartment". Cell Host & Microbe. 3 (1): 30–8. doi: 10.1016/j.chom.2007.11.001 . PMID   18191792.
  11. 1 2 3 4 5 Eshraghi A, Maldonado-Arocho FJ, Gargi A, Cardwell MM, Prouty MG, Blanke SR, Bradley KA (June 2010). "Cytolethal distending toxin family members are differentially affected by alterations in host glycans and membrane cholesterol". The Journal of Biological Chemistry. 285 (24): 18199–207. doi: 10.1074/jbc.m110.112912 . PMC   2881744 . PMID   20385557.
  12. Nesić D, Hsu Y, Stebbins CE (May 2004). "Assembly and function of a bacterial genotoxin". Nature. 429 (6990): 429–33. Bibcode:2004Natur.429..429N. doi:10.1038/nature02532. PMID   15164065. S2CID   4373500.

Related Research Articles

<span class="mw-page-title-main">Shiga toxin</span> Family of related toxins

Shiga toxins are a family of related toxins with two major groups, Stx1 and Stx2, expressed by genes considered to be part of the genome of lambdoid prophages. The toxins are named after Kiyoshi Shiga, who first described the bacterial origin of dysentery caused by Shigella dysenteriae. Shiga-like toxin (SLT) is a historical term for similar or identical toxins produced by Escherichia coli. The most common sources for Shiga toxin are the bacteria S. dysenteriae and some serotypes of Escherichia coli (STEC), which includes serotypes O157:H7, and O104:H4.

<span class="mw-page-title-main">FtsZ</span> Protein encoded by the ftsZ gene

FtsZ is a protein encoded by the ftsZ gene that assembles into a ring at the future site of bacterial cell division. FtsZ is a prokaryotic homologue of the eukaryotic protein tubulin. The initials FtsZ mean "Filamenting temperature-sensitive mutant Z." The hypothesis was that cell division mutants of E. coli would grow as filaments due to the inability of the daughter cells to separate from one another. FtsZ is found in almost all bacteria, many archaea, all chloroplasts and some mitochondria, where it is essential for cell division. FtsZ assembles the cytoskeletal scaffold of the Z ring that, along with additional proteins, constricts to divide the cell in two.

<span class="mw-page-title-main">Exotoxin</span> Toxin from bacteria that destroys or disrupts cells

An exotoxin is a toxin secreted by bacteria. An exotoxin can cause damage to the host by destroying cells or disrupting normal cellular metabolism. They are highly potent and can cause major damage to the host. Exotoxins may be secreted, or, similar to endotoxins, may be released during lysis of the cell. Gram negative pathogens may secrete outer membrane vesicles containing lipopolysaccharide endotoxin and some virulence proteins in the bounding membrane along with some other toxins as intra-vesicular contents, thus adding a previously unforeseen dimension to the well-known eukaryote process of membrane vesicle trafficking, which is quite active at the host–pathogen interface.

<span class="mw-page-title-main">SOS response</span> Biological process

The SOS response is a global response to DNA damage in which the cell cycle is arrested and DNA repair and mutagenesis is induced. The system involves the RecA protein. The RecA protein, stimulated by single-stranded DNA, is involved in the inactivation of the repressor (LexA) of SOS response genes thereby inducing the response. It is an error-prone repair system that contributes significantly to DNA changes observed in a wide range of species.

<span class="mw-page-title-main">Polymyxin</span>

Polymyxins are antibiotics. Polymyxins B and E are used in the treatment of Gram-negative bacterial infections. They work mostly by breaking up the bacterial cell membrane. They are part of a broader class of molecules called nonribosomal peptides.

DNA gyrase, or simply gyrase, is an enzyme within the class of topoisomerase and is a subclass of Type II topoisomerases that reduces topological strain in an ATP dependent manner while double-stranded DNA is being unwound by elongating RNA-polymerase or by helicase in front of the progressing replication fork. It is the only known enzyme to actively contribute negative supercoiling to DNA, while it also is capable of relaxing positive supercoils. It does so by looping the template to form a crossing, then cutting one of the double helices and passing the other through it before releasing the break, changing the linking number by two in each enzymatic step. This process occurs in bacteria, whose single circular DNA is cut by DNA gyrase and the two ends are then twisted around each other to form supercoils. Gyrase is also found in eukaryotic plastids: it has been found in the apicoplast of the malarial parasite Plasmodium falciparum and in chloroplasts of several plants. Bacterial DNA gyrase is the target of many antibiotics, including nalidixic acid, novobiocin, albicidin, and ciprofloxacin.

<span class="mw-page-title-main">Lysogenic cycle</span> Process of virus reproduction

Lysogeny, or the lysogenic cycle, is one of two cycles of viral reproduction. Lysogeny is characterized by integration of the bacteriophage nucleic acid into the host bacterium's genome or formation of a circular replicon in the bacterial cytoplasm. In this condition the bacterium continues to live and reproduce normally, while the bacteriophage lies in a dormant state in the host cell. The genetic material of the bacteriophage, called a prophage, can be transmitted to daughter cells at each subsequent cell division, and later events can release it, causing proliferation of new phages via the lytic cycle. Lysogenic cycles can also occur in eukaryotes, although the method of DNA incorporation is not fully understood. For instance the AIDS viruses can either infect humans lytically, or lay dormant (lysogenic) as part of the infected cells' genome, keeping the ability to return to lysis at a later time. The rest of this article is about lysogeny in bacterial hosts.

Casein kinase 2 (CK2/CSNK2) is a serine/threonine-selective protein kinase that has been implicated in cell cycle control, DNA repair, regulation of the circadian rhythm, and other cellular processes. De-regulation of CK2 has been linked to tumorigenesis as a potential protection mechanism for mutated cells. Proper CK2 function is necessary for survival of cells as no knockout models have been successfully generated.

The AB5 toxins are six-component protein complexes secreted by certain pathogenic bacteria known to cause human diseases such as cholera, dysentery, and hemolytic–uremic syndrome. One component is known as the A subunit, and the remaining five components are B subunits. All of these toxins share a similar structure and mechanism for entering targeted host cells. The B subunit is responsible for binding to receptors to open up a pathway for the A subunit to enter the cell. The A subunit is then able to use its catalytic machinery to take over the host cell's regular functions.

<span class="mw-page-title-main">ADP-ribosylation</span> Addition of one or more ADP-ribose moieties to a protein.

ADP-ribosylation is the addition of one or more ADP-ribose moieties to a protein. It is a reversible post-translational modification that is involved in many cellular processes, including cell signaling, DNA repair, gene regulation and apoptosis. Improper ADP-ribosylation has been implicated in some forms of cancer. It is also the basis for the toxicity of bacterial compounds such as cholera toxin, diphtheria toxin, and others.

<span class="mw-page-title-main">AB toxin</span>

The AB toxins are two-component protein complexes secreted by a number of pathogenic bacteria, though there is a pore-forming AB toxin found the eggs of a snail. They can be classified as Type III toxins because they interfere with internal cell function. They are named AB toxins due to their components: the "A" component is usually the "active" portion, and the "B" component is usually the "binding" portion. The "A" subunit possesses enzyme activity, and is transferred to the host cell following a conformational change in the membrane-bound transport "B" subunit. These proteins consist of two independent polypeptides, which correspond to the A/B subunit moieties. The enzyme component (A) enters the cell through endosomes produced by the oligomeric binding/translocation protein (B), and prevents actin polymerisation through ADP-ribosylation of monomeric G-actin.

<span class="mw-page-title-main">Circular chromosome</span> Type of chromosome

A circular chromosome is a chromosome in bacteria, archaea, mitochondria, and chloroplasts, in the form of a molecule of circular DNA, unlike the linear chromosome of most eukaryotes.

<span class="mw-page-title-main">Caspase-activated DNase</span> Protein-coding gene in the species Homo sapiens

Caspase-activated DNase (CAD) or DNA fragmentation factor subunit beta is a protein that in humans is encoded by the DFFB gene. It breaks up the DNA during apoptosis and promotes cell differentiation. It is usually an inactive monomer inhibited by ICAD. This is cleaved before dimerization.

<span class="mw-page-title-main">Toxin-antitoxin system</span> Biological process

A toxin-antitoxin system consists of a "toxin" and a corresponding "antitoxin", usually encoded by closely linked genes. The toxin is usually a protein while the antitoxin can be a protein or an RNA. Toxin-antitoxin systems are widely distributed in prokaryotes, and organisms often have them in multiple copies. When these systems are contained on plasmids – transferable genetic elements – they ensure that only the daughter cells that inherit the plasmid survive after cell division. If the plasmid is absent in a daughter cell, the unstable antitoxin is degraded and the stable toxic protein kills the new cell; this is known as 'post-segregational killing' (PSK).

Pathogenic <i>Escherichia coli</i> Strains of E. coli that can cause disease

Escherichia coli is a gram-negative, rod-shaped bacterium that is commonly found in the lower intestine of warm-blooded organisms (endotherms). Most E. coli strains are harmless, but pathogenic varieties cause serious food poisoning, septic shock, meningitis, or urinary tract infections in humans. Unlike normal flora E. coli, the pathogenic varieties produce toxins and other virulence factors that enable them to reside in parts of the body normally not inhabited by E. coli, and to damage host cells. These pathogenic traits are encoded by virulence genes carried only by the pathogens.

<span class="mw-page-title-main">CcdA/CcdB Type II Toxin-antitoxin system</span>

The CcdA/CcdB Type II Toxin-antitoxin system is one example of the bacterial toxin-antitoxin (TA) systems that encode two proteins, one a potent inhibitor of cell proliferation (toxin) and the other its specific antidote (antitoxin). These systems preferentially guarantee growth of plasmid-carrying daughter cells in a bacterial population by killing newborn bacteria that have not inherited a plasmid copy at cell division.

Campylobacter coli is a Gram-negative, microaerophilic, non-endospore-forming, S-shaped bacterial species within genus Campylobacter. In humans, it C. coli can cause campylobacteriosis, a diarrhoeal disease which is the most frequently reported foodborne illness in the European Union. C. coli grows slowly with an optimum temperature of 42 °C. When exposed to air for long periods, they become spherical or coccoid shaped.

Ribonuclease E is a bacterial ribonuclease that participates in the processing of ribosomal RNA and the chemical degradation of bulk cellular RNA.

Contact-dependent growth inhibition (CDI) is a phenomenon where a bacterial cell may deliver a polymorphic toxin molecule into neighbouring bacterial cells upon direct cell-cell contact, causing growth arrest or cell death.

<span class="mw-page-title-main">Bacterial therapy</span>

Bacterial therapy is the therapeutic use of bacteria to treat diseases. Bacterial therapeutics are living medicines, and may be wild type bacteria or bacteria that have been genetically engineered to possess therapeutic properties that is injected into a patient. Other examples of living medicines include cellular therapeutics, activators of anti-tumor immunity, or synergizing with existing tools and approaches. and phage therapeutics, or as delivery vehicles for treatment, diagnosis, or imaging, complementing or synergizing with existing tools and approaches.