Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Anneke Kuipers is active.

Publication


Featured researches published by Anneke Kuipers.


Journal of Biological Chemistry | 2004

NisT, the Transporter of the Lantibiotic Nisin, Can Transport Fully Modified, Dehydrated, and Unmodified Prenisin and Fusions of the Leader Peptide with Non-lantibiotic Peptides

Anneke Kuipers; Esther de Boef; Rick Rink; Susan Fekken; Leon D. Kluskens; Arnold J. M. Driessen; Kees Leenhouts; Oscar P. Kuipers; Gert N. Moll

Lantibiotics are lanthionine-containing peptide antibiotics. Nisin, encoded by nisA, is a pentacyclic lantibiotic produced by some Lactococcus lactis strains. Its thioether rings are posttranslationally introduced by a membrane-bound enzyme complex. This complex is composed of three enzymes: NisB, which dehydrates serines and threonines; NisC, which couples these dehydrated residues to cysteines, thus forming thioether rings; and the transporter NisT. We followed the activity of various combinations of the nisin enzymes by measuring export of secreted peptides using antibodies against the leader peptide and mass spectroscopy for detection. L. lactis expressing the nisABTC genes efficiently produced fully posttranslationally modified prenisin. Strikingly, L. lactis expressing the nisBT genes could produce dehydrated prenisin without thioether rings and a dehydrated form of a non-lantibiotic peptide. In the absence of the biosynthetic NisBC enzymes, the NisT transporter was capable of excreting unmodified prenisin and fusions of the leader peptide with non-lantibiotic peptides. Our data show that NisT specifies a broad spectrum (poly)peptide transporter that can function either in conjunction with or independently from the biosynthetic genes. NisT secretes both unmodified and partially or fully posttranslationally modified forms of prenisin and non-lantibiotic peptides. These results open the way for efficient production of a wide range of peptides with increased stability or novel bioactivities.


Journal of Pharmacology and Experimental Therapeutics | 2009

Angiotensin-(1-7) with Thioether Bridge: An Angiotensin-Converting Enzyme-Resistant, Potent Angiotensin-(1-7) Analog

Leon D. Kluskens; S. Adriaan Nelemans; Rick Rink; Louwe de Vries; Anita Meter-Arkema; Yong Wang; Thomas Walther; Anneke Kuipers; Gert N. Moll; M Haas

The in vivo efficacy of many therapeutic peptides is hampered by their rapid proteolytic degradation. Cyclization of these therapeutic peptides is an excellent way to render them more resistant against breakdown. Here, we describe the enzymatic introduction of a thioether ring in angiotensin [Ang-(1-7)], a heptapeptide that plays a pivotal role in the renin-angiotensin system and possesses important therapeutic activities. The lactic acid bacterium Lactococcus lactis, equipped with the plasmid-based nisin modification machinery, was used to produce thioether-bridged Ang-(1-7). The resulting cyclized Ang-(1-7) is fully resistant against purified angiotensin-converting enzyme, has significantly increased stability in homogenates of different organs and in plasma derived from pig, and displays a strongly (34-fold) enhanced survival in Sprague-Dawley (SD) rats in vivo. With respect to functional activity, cyclized Ang-(1-7) induces relaxation of precontracted SD rat aorta rings in vitro. The magnitude of this effect is 2-fold larger than that obtained for natural Ang-(1-7). The Ang-(1-7) receptor antagonist d-Pro7-Ang-(1-7), which completely inhibits the activity of natural Ang-(1-7), also abolishes the vasodilation by cyclized Ang-(1-7), providing evidence that cyclized Ang-(1-7) also interacts with the Ang-(1-7) receptor. Taken together, applying a highly innovative enzymatic peptide stabilization method, we generated a stable Ang-(1-7) analog with strongly enhanced therapeutic potential.


Applied and Environmental Microbiology | 2007

Dissection and modulation of the four distinct activities of nisin by mutagenesis of rings A and B and by C-terminal truncation

Rick Rink; Jenny Wierenga; Anneke Kuipers; Leon D. Kluskens; Arnold J. M. Driessen; Oscar P. Kuipers; Gert N. Moll

ABSTRACT Nisin A is a pentacyclic antibiotic peptide produced by various Lactococcus lactis strains. Nisin displays four different activities: (i) it autoinduces its own synthesis; (ii) it inhibits the growth of target bacteria by membrane pore formation; (iii) it inhibits bacterial growth by interfering with cell wall synthesis; and, in addition, (iv) it inhibits the outgrowth of spores. Here we investigate the structural requirements and relevance of the N-terminal thioether rings of nisin by randomization of the ring A and B positions. The data demonstrate that: (i) mutation of ring A results in variants with enhanced activity and a modulated spectrum of target cells; (ii) for the cell growth-inhibiting activity of nisin, ring A is rather promiscuous with respect to its amino acid composition, whereas the bulky amino acid residues in ring B abolish antimicrobial activity; (iii) C-terminally truncated nisin A mutants lacking rings D and E retain significant antimicrobial activity but are unable to permeabilize the target membrane; (iv) the dehydroalanine in ring A is not essential for the inhibition of the outgrowth of Bacillus cells; (v) some ring A mutants have significant antimicrobial activities but have decreased autoinducing activities; (vi) the opening of ring B eliminates antimicrobial activity while retaining autoinducing activity; and (vii) some ring A mutants escape the nisin immune system(s) and are toxic to the nisin-producing strain NZ9700. These data demonstrate that the various activities of nisin can be engineered independently and provide a basis for the design and synthesis of tailor-made analogs with desired activities.


Applied and Environmental Microbiology | 2003

Identification and Characterization of Two Novel Clostridial Bacteriocins, Circularin A and Closticin 574

Robèr Kemperman; Anneke Kuipers; Harma Karsens; Arjen Nauta; Oscar P. Kuipers; Jan Kok

ABSTRACT Two novel antibacterial peptides of clostridial species were purified, N-terminally sequenced, and characterized. Moreover, their structural genes were identified. Closticin 574 is an 82-amino-acid bacteriocin produced by Clostridium tyrobutyricum ADRIAT 932. The supernatant of the producing strain showed a high level of activity against the indicator strain C. tyrobutyricum. The protein is synthesized as a preproprotein that is possibly secreted via the general secretion pathway, after which it is hydrolyzed at an Asp-Pro site. Circularin A is produced by Clostridium beijerinckii ATCC 25752 as a prepeptide of 72 amino acids. Cleavage of the prepeptide between the third leucine and fourth valine residues followed by a head-to-tail ligation between the N and C termini creates a circular antimicrobial peptide of 69 amino acids. The unusually small circularin A leader peptide of three amino acids is cleaved off in this process. The supernatant of C. beijerinckii ATCC 25752 showed a broad antibacterial activity range.


Journal of Pharmacology and Experimental Therapeutics | 2008

Angiotensin-(1-7) with thioether-bridge: an ACE-resistant, potent Ang-(1-7) analogue

Leon D. Kluskens; Ad Nelemans; Rick Rink; Louwe de Vries; Anita Meter-Arkema; Yong Wang; Thomas Walther; Anneke Kuipers; Gert N. Moll; M Haas

The in vivo efficacy of many therapeutic peptides is hampered by their rapid proteolytic degradation. Cyclization of these therapeutic peptides is an excellent way to render them more resistant against breakdown. Here, we describe the enzymatic introduction of a thioether ring in angiotensin [Ang-(1-7)], a heptapeptide that plays a pivotal role in the renin-angiotensin system and possesses important therapeutic activities. The lactic acid bacterium Lactococcus lactis, equipped with the plasmid-based nisin modification machinery, was used to produce thioether-bridged Ang-(1-7). The resulting cyclized Ang-(1-7) is fully resistant against purified angiotensin-converting enzyme, has significantly increased stability in homogenates of different organs and in plasma derived from pig, and displays a strongly (34-fold) enhanced survival in Sprague-Dawley (SD) rats in vivo. With respect to functional activity, cyclized Ang-(1-7) induces relaxation of precontracted SD rat aorta rings in vitro. The magnitude of this effect is 2-fold larger than that obtained for natural Ang-(1-7). The Ang-(1-7) receptor antagonist d-Pro7-Ang-(1-7), which completely inhibits the activity of natural Ang-(1-7), also abolishes the vasodilation by cyclized Ang-(1-7), providing evidence that cyclized Ang-(1-7) also interacts with the Ang-(1-7) receptor. Taken together, applying a highly innovative enzymatic peptide stabilization method, we generated a stable Ang-(1-7) analog with strongly enhanced therapeutic potential.


Journal of Pharmacological and Toxicological Methods | 2010

To protect peptide pharmaceuticals against peptidases

Rick Rink; A. Arkema-Meter; Inge Baudoin; Eduard Post; Anneke Kuipers; Sieger Adriaan Nelemans; M. Haas Jimoh Akanbi; Gert N. Moll

INTRODUCTION The major hurdle in the application and delivery of peptide pharmaceuticals is their rapid in vivo breakdown. METHODS We here combined two approaches to stabilize peptide pharmaceuticals, introduction of D-amino acids and cyclization, by applying an innovative enzymatic method. This method yields peptides with thioether bridges between a D-amino acid and an L-amino acid. On the basis of guidelines concerning the flanking residues of serines/threonines and cysteines, a peptide of interest is designed with serine/threonine and cysteine at appropriate positions to allow their effective participation in cyclization. In Lactococcus lactis the peptide of interest is directly or via a spacer genetically fused to a lantibiotic leader peptide which induces enzyme-catalysed synthesis of a thioether-bridged peptide. The peptide is translocated via a lantibiotic transporter, analysed by mass spectrometry and the leader peptide is removed. Because of its therapeutic relevance and terminal modifications we chose the decapeptide Luteïnizing Hormone Release Hormone (LHRH) as a test case for thioether bridge introduction. The N-terminal pyroglutamate protects against aminopeptidase activity; the amidated C-terminus, which occurs in 50% of all therapeutic peptides, precludes carboxypeptidase action and is essential for optimal receptor interaction. We had Lactococcus posttranslationally introduce a thioether bridge between residues 4 and 7 of the Leu7Cys-LHRH analog QHWSYGCRPG. The N-terminal glutamine of the thioether-bridged peptide could be converted in pyroglutamate. The introduction of the thioether bridge proved to be compatible with subsequent chemical and enzymatic amidation methods. In this way biologically produced thioether LHRH was compared with LHRH isomers obtained by base-assisted sulfur extrusion. RESULTS Biologically produced thioether LHRH is the most stable thioether LHRH isomer with strongly enhanced proteolytic resistance compared to natural LHRH. DISCUSSION The data convincingly demonstrate the broad perspective of stereo- and regiospecifically generating cyclized peptide pharmaceuticals with significantly enhanced therapeutic potential.


Applied and Environmental Microbiology | 2007

Production of Dehydroamino Acid-Containing Peptides by Lactococcus lactis

Rick Rink; Jenny Wierenga; Anneke Kuipers; Leon D. Kluskens; Arnold J. M. Driessen; Oscar P. Kuipers; Gert N. Moll

ABSTRACT Nisin is a pentacyclic peptide antibiotic produced by some Lactococcus lactis strains. Nisin contains dehydroresidues and thioether rings that are posttranslationally introduced by a membrane-associated enzyme complex, composed of a serine and threonine dehydratase NisB and the cyclase NisC. In addition, the transporter NisT is necessary for export of the modified peptide. We studied the potential of L. lactis expressing NisB and NisT to produce peptides whose serines and threonines are dehydrated. L. lactis containing the nisBT genes and a plasmid coding for a specific leader peptide fusion construct efficiently produced peptides with a series of non-naturally occurring multiple flanking dehydrobutyrines. We demonstrated NisB-mediated dehydration of serines and threonines in a C-terminal nisin(1-14) extension of nisin, which implies that also residues more distant from the leader peptide than those occurring in prenisin or any other lantibiotic can be modified. Furthermore, the feasibility and efficiency of generating a library of peptides containing dehydroresidues were demonstrated. In view of the particular shape and reactivity of dehydroamino acids, such a library provides a novel source for screening for peptides with desired biological and physicochemical properties.


Applied and Environmental Microbiology | 2011

Requirements of the Engineered Leader Peptide of Nisin for Inducing Modification, Export, and Cleavage

Annechien Plat; Leon D. Kluskens; Anneke Kuipers; Rick Rink; Gert N. Moll

ABSTRACT Nisin A is a pentacyclic peptide antibiotic produced by Lactococcus lactis. The leader peptide of prenisin keeps nisin inactive and has a role in inducing NisB- and NisC-catalyzed modifications of the propeptide and NisT-mediated export. The highly specific NisP cleaves off the leader peptide from fully modified and exported prenisin. We present here a detailed mutagenesis analysis of the nisin leader peptide. For alternative cleavage, we successfully introduced a putative NisP autocleavage site and sites for thrombin, enterokinase, Glu-C, and factor Xa in the C-terminal part of the leader peptide. Replacing residue F-18 with Trp or Thr strongly reduced production. On the other hand, D-19A, F-18H, F-18M, L-16D, L-16K, and L-16A enhanced production. Substitutions within and outside the FNLD box enhanced or reduced the transport efficiency. None of the above substitutions nor even an internal 6His tag from positions −13 to −8 had any effect on the capacity of the leader peptide to induce NisB and NisC modifications. Therefore, these data demonstrate a large mutational freedom. However, simultaneous replacement of the FNLD amino acids by four alanines strongly reduced export and even led to a complete loss of the capacity to induce modifications. Reducing the leader peptide to MSTKDFNLDLR led to 3- or 4-fold dehydration. Taken together, the FNLD box is crucial for inducing posttranslational modifications.


Antimicrobial Agents and Chemotherapy | 2010

Production of a Class II Two-Component Lantibiotic of Streptococcus pneumoniae Using the Class I Nisin Synthetic Machinery and Leader Sequence

Joanna A. Majchrzykiewicz; Jacek Lubelski; Gert N. Moll; Anneke Kuipers; Jetta J. E. Bijlsma; Oscar P. Kuipers; Rick Rink

ABSTRACT Recent studies showed that the nisin modification machinery can successfully dehydrate serines and threonines and introduce lanthionine rings in small peptides that are fused to the nisin leader sequence. This opens up exciting possibilities to produce and engineer larger antimicrobial peptides in vivo. Here we demonstrate the exploitation of the class I nisin production machinery to generate, modify, and secrete biologically active, previously not-yet-isolated and -characterized class II two-component lantibiotics that have no sequence homology to nisin. The nisin synthesis machinery, composed of the modification enzymes NisB and NisC and the transporter NisT, was used to modify and secrete a putative two-component lantibiotic of Streptococcus pneumoniae. This was achieved by genetically fusing the propeptide-encoding sequences of the spr1765 (pneA1) and spr1766 (pneA2) genes to the nisin leader-encoding sequence. The chimeric prepeptides were secreted out of Lactococcus lactis, purified by cation exchange fast protein liquid chromatography, and further characterized. Mass spectrometry analyses demonstrated the presence and partial localization of multiple dehydrated serines and/or threonines and (methyl)lanthionines in both peptides. Moreover, after cleavage of the leader peptide from the prepeptides, both modified propeptides displayed antimicrobial activity against Micrococcus flavus. These results demonstrate that the nisin synthetase machinery can be successfully used to modify and produce otherwise difficult to obtain antimicrobially active lantibiotics.


Applied and Environmental Microbiology | 2006

Sec-mediated transport of posttranslationally dehydrated peptides in Lactococcus lactis.

Anneke Kuipers; Jenny Wierenga; Rick Rink; Leon D. Kluskens; Arnold J. M. Driessen; Oscar P. Kuipers; Gert N. Moll

ABSTRACT Nisin is a lanthionine-containing antimicrobial peptide produced by Lactococcus lactis. Its (methyl)lanthionines are introduced by two posttranslational enzymatic steps involving the dehydratase NisB, which dehydrates serine and threonine residues, and the cyclase NisC, which couples these dehydrated residues to cysteines, yielding thioether-bridged amino acids called lanthionines. The prenisin is subsequently exported by the ABC transporter NisT and extracellularly processed by the peptidase NisP. L. lactis expressing the nisBTC genes can modify and secrete a wide range of nonlantibiotic peptides. Here we demonstrate that in the absence of NisT and NisC, the Sec pathway of L. lactis can be exploited for the secretion of dehydrated variants of therapeutic peptides. Furthermore, posttranslational modifications by NisB and NisC still occur even when the nisin leader is preceded by a Sec signal peptide or a Tat signal peptide 27 or 44 amino acids long, respectively. However, transport of fully modified prenisin via the Sec pathway is impaired. The extent of NisB-mediated dehydration could be improved by raising the intracellular concentration NisB or by modulating the export efficiency through altering the signal sequence. These data demonstrate that besides the traditional lantibiotic transporter NisT, the Sec pathway with an established broad substrate range can be utilized for the improved export of lantibiotic enzyme-modified (poly)peptides.

Collaboration


Dive into the Anneke Kuipers's collaboration.

Top Co-Authors

Avatar

Gert N. Moll

University of Groningen

View shared research outputs
Top Co-Authors

Avatar

Rick Rink

University of Groningen

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

M Haas

University of Groningen

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Anita Meter-Arkema

University Medical Center Groningen

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge