Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.018
Filter
1.
J Nanobiotechnology ; 22(1): 483, 2024 Aug 13.
Article in English | MEDLINE | ID: mdl-39138475

ABSTRACT

The mortality of ovarian cancer (OC) has long been the highest among gynecological malignancies. Although OC is considered to be an immunogenic tumor, the effect of immunotherapy is not satisfactory. The immunosuppressive microenvironment is one reason for this, and the absence of recognized effective antigens for vaccines is another. Chemotherapy, as one of the most commonly used treatment for OC, can produce chemotherapy-associated antigens (CAAs) during treatment and show the effect of in situ vaccine. Herein, we designed an antigen capture nano-vaccine NP-TP1@M-M with tumor targeting peptide TMTP1 and dendritic cell (DC) receptor mannose assembled on the surface and adjuvant monophosphoryl lipid A (MPLA) encapsulated in the core of poly (D, L-lactide-co-glycolide) (PLGA) nanoparticles. PLGA itself possessed the ability of antigen capture. TMTP1 was a tumor-homing peptide screened by our research team, which held extensive and excellent tumor targeting ability. After these modifications, NP-TP1@M-M could capture and enrich more tumor-specific antigens after chemotherapy, stimulate DC maturation, activate the adaptive immunity and combined with immune checkpoint blockade to maximize the release of the body's immune potential, providing an eutherapeutic strategy for the treatment of OC.


Subject(s)
Antigens, Neoplasm , B7-H1 Antigen , Cancer Vaccines , Nanoparticles , Ovarian Neoplasms , Female , Ovarian Neoplasms/drug therapy , Animals , Mice , Cancer Vaccines/therapeutic use , Nanoparticles/chemistry , Cell Line, Tumor , Antigens, Neoplasm/immunology , Humans , Dendritic Cells/drug effects , Peptides/chemistry , Peptides/pharmacology , Lipid A/analogs & derivatives , Lipid A/chemistry , Lipid A/pharmacology , Immunotherapy/methods , Polylactic Acid-Polyglycolic Acid Copolymer/chemistry , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Mice, Inbred BALB C , Immune Checkpoint Inhibitors/pharmacology , Nanovaccines
2.
Toxins (Basel) ; 16(7)2024 Jul 18.
Article in English | MEDLINE | ID: mdl-39057962

ABSTRACT

The toxic nature of bacterial endotoxins is affected by the structural details of lipid A, including the variety and position of acyl chains and phosphate group(s) on its diglucosamine backbone. Negative-ion mode tandem mass spectrometry is a primary method for the structure elucidation of lipid A, used independently or in combination with separation techniques. However, it is challenging to accurately characterize constitutional isomers of lipid A extracts by direct mass spectrometry, as the elemental composition and molecular mass of these molecules are identical. Thus, their simultaneous fragmentation leads to a composite, so-called chimera mass spectrum. The present study focuses on the phosphopositional isomers of the classical monophosphorylated, hexaacylated Escherichia coli-type lipid A. Collision-induced dissociation (CID) was performed in an HPLC-ESI-QTOF system. Energy-resolved mass spectrometry (ERMS) was applied to uncover the distinct fragmentation profiles of the phosphorylation isomers. A fragmentation strategy applying multi-levels of collision energy has been proposed and applied to reveal sample complexity, whether it contains only a 4'-phosphorylated species or a mixture of 1- and 4'-phosphorylated variants. This comparative fragmentation study of isomeric lipid A species demonstrates the high potential of ERMS-derived information for the successful discrimination of co-ionized phosphorylation isomers of hexaacylated lipid A.


Subject(s)
Lipid A , Tandem Mass Spectrometry , Lipid A/chemistry , Isomerism , Spectrometry, Mass, Electrospray Ionization , Escherichia coli , Chromatography, High Pressure Liquid , Phosphorylation
3.
Nat Commun ; 15(1): 5946, 2024 Jul 15.
Article in English | MEDLINE | ID: mdl-39009687

ABSTRACT

The ATP-binding cassette (ABC) transporter, MsbA, plays a pivotal role in lipopolysaccharide (LPS) biogenesis by facilitating the transport of the LPS precursor lipooligosaccharide (LOS) from the cytoplasmic to the periplasmic leaflet of the inner membrane. Despite multiple studies shedding light on MsbA, the role of lipids in modulating MsbA-nucleotide interactions remains poorly understood. Here we use native mass spectrometry (MS) to investigate and resolve nucleotide and lipid binding to MsbA, demonstrating that the transporter has a higher affinity for adenosine 5'-diphosphate (ADP). Moreover, native MS shows the LPS-precursor 3-deoxy-D-manno-oct-2-ulosonic acid (Kdo)2-lipid A (KDL) can tune the selectivity of MsbA for adenosine 5'-triphosphate (ATP) over ADP. Guided by these studies, four open, inward-facing structures of MsbA are determined that vary in their openness. We also report a 2.7 Å-resolution structure of MsbA in an open, outward-facing conformation that is not only bound to KDL at the exterior site, but with the nucleotide binding domains (NBDs) adopting a distinct nucleotide-free structure. The results obtained from this study offer valuable insight and snapshots of MsbA during the transport cycle.


Subject(s)
ATP-Binding Cassette Transporters , Adenosine Diphosphate , Adenosine Triphosphate , Mass Spectrometry , ATP-Binding Cassette Transporters/metabolism , ATP-Binding Cassette Transporters/chemistry , Adenosine Triphosphate/metabolism , Adenosine Diphosphate/metabolism , Mass Spectrometry/methods , Bacterial Proteins/metabolism , Bacterial Proteins/chemistry , Lipopolysaccharides/metabolism , Lipid A/metabolism , Lipid A/chemistry , Protein Binding , Models, Molecular , Crystallography, X-Ray , Lipids/chemistry , Escherichia coli/metabolism , Protein Conformation
4.
ACS Appl Mater Interfaces ; 16(32): 41810-41818, 2024 Aug 14.
Article in English | MEDLINE | ID: mdl-39084852

ABSTRACT

Anticancer chemo-immunotherapy has gained considerable attention across various scientific domains as a prospective approach for the comprehensive eradication of malignant tumors. Recent research has particularly been focused on traditional anthracycline chemo drugs, such as doxorubicin and mitoxantrone. These compounds trigger apoptosis in tumor cells and evoke immunogenic cell death (ICD). ICD is a pivotal initiator of the cancer-immunity cycle by facilitating the release of damage-associated molecular patterns (DAMPs). The resultant DAMPs released from cancer cells effectively activate the immune system, resulting in an increase in tumor-infiltrating T cells. In this study, we have innovated a co-delivery strategy involving folate-modified liposomes to deliver doxorubicin and monophosphoryl lipid A (MPLA) simultaneously to tumor tissue. The engineered liposomes exploit the overexpression of folate receptors within the tumor tissues. Delivered doxorubicin initiates ICD at the tumor cells, further enhancing the immunogenic stimulus. Additionally, MPLA helps T cell priming by activating antigen-presenting cells. This intricate interplay culminates in a synergistic effect, ultimately resulting in an augmented and potentiated anticancer chemo-immunotherapeutic liposomal treatment.


Subject(s)
Doxorubicin , Immunogenic Cell Death , Immunotherapy , Lipid A , Liposomes , Toll-Like Receptor 4 , Liposomes/chemistry , Doxorubicin/pharmacology , Doxorubicin/chemistry , Animals , Immunogenic Cell Death/drug effects , Humans , Toll-Like Receptor 4/agonists , Toll-Like Receptor 4/metabolism , Mice , Lipid A/analogs & derivatives , Lipid A/chemistry , Lipid A/pharmacology , Neoplasms/drug therapy , Neoplasms/immunology , Neoplasms/therapy , Cell Line, Tumor , Female , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Folic Acid/chemistry
5.
Nat Commun ; 15(1): 4733, 2024 Jun 03.
Article in English | MEDLINE | ID: mdl-38830951

ABSTRACT

Polymyxins are gram-negative antibiotics that target lipid A, the conserved membrane anchor of lipopolysaccharide in the outer membrane. Despite their clinical importance, the molecular mechanisms underpinning polymyxin activity remain unresolved. Here, we use surface plasmon resonance to kinetically interrogate interactions between polymyxins and lipid A and derive a phenomenological model. Our analyses suggest a lipid A-catalyzed, three-state mechanism for polymyxins: transient binding, membrane insertion, and super-stoichiometric cluster accumulation with a long residence time. Accumulation also occurs for brevicidine, another lipid A-targeting antibacterial molecule. Lipid A modifications that impart polymyxin resistance and a non-bactericidal polymyxin derivative exhibit binding that does not evolve into long-lived species. We propose that transient binding to lipid A permeabilizes the outer membrane and cluster accumulation enables the bactericidal activity of polymyxins. These findings could establish a blueprint for discovery of lipid A-targeting antibiotics and provide a generalizable approach to study interactions with the gram-negative outer membrane.


Subject(s)
Anti-Bacterial Agents , Lipid A , Polymyxin B , Surface Plasmon Resonance , Polymyxin B/pharmacology , Polymyxin B/metabolism , Lipid A/metabolism , Lipid A/chemistry , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/metabolism , Microbial Sensitivity Tests , Bacterial Outer Membrane/metabolism , Bacterial Outer Membrane/drug effects , Kinetics
6.
Int J Mol Sci ; 25(12)2024 Jun 08.
Article in English | MEDLINE | ID: mdl-38928052

ABSTRACT

Bacterial endotoxins (lipopolysaccharides (LPSs)) are important mediators of inflammatory processes induced by Gram-negative microorganisms. LPSs are the key inducers of septic shock due to a Gram-negative bacterial infection; thus, the structure and functions of LPSs are of specific interest. Often, highly purified bacterial endotoxins must be isolated from small amounts of biological material. Each of the currently available methods for LPS extraction has certain limitations. Herein, we describe a rapid and simple microscale method for extracting LPSs. The method consists of the following steps: ultrasonic destruction of the bacterial material, LPS extraction via heating, LPS purification with organic solvents, and treatment with proteinase K. LPSs that were extracted by using this method contained less than 2-3% protein and 1% total nucleic acid. We also demonstrated the structural integrity of the O-antigen and lipid A via the sodium dodecyl-sulfate polyacrylamide gel electrophoresis (SDS-PAGE) and matrix-assisted laser desorption ionization mass spectrometry (MALDI-MS) methods, respectively. We demonstrated the ability of the extracted LPSs to induce typical secretion of cytokines and chemokines by primary macrophages. Overall, this method may be used to isolate purified LPSs with preserved structures of both the O-antigen and lipid A and unchanged functional activity from small amounts of bacterial biomass.


Subject(s)
Lipopolysaccharides , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Lipopolysaccharides/isolation & purification , Lipopolysaccharides/chemistry , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization/methods , Animals , Mice , Macrophages/metabolism , Lipid A/chemistry , Lipid A/isolation & purification , Cytokines/metabolism , Endopeptidase K/metabolism , Endopeptidase K/chemistry , Electrophoresis, Polyacrylamide Gel/methods
7.
Adv Healthc Mater ; 13(18): e2304109, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38849130

ABSTRACT

Lipid vesicles are widely used for drug and gene delivery, but their structural instability reduces in vivo efficacy and requires specialized handling. To address these limitations, strategies like lipid cross-linking and polymer-lipid conjugation are suggested to enhance stability and biological efficacy. However, the in vivo metabolism of these altered lipids remains unclear, necessitating further studies. A new stabilization technique without chemical modification is urgently needed. Here, a bio-mimetic approach for fabricating robust multilamellar lipid vesicles to enhance in vivo delivery and stabilization of protein antigens is presented. This method leverages 1-O-acylceramide, a natural skin lipid, to facilitate the self-assembly of lipid nanovesicles. Incorporating 1-O-acylceramide, anchoring lipid bilayers akin to its role in the stratum corneum, provides excellent stability under environmental stresses, including freeze-thaw cycles. Encapsulating ovalbumin as a model antigen and the adjuvant monophosphoryl lipid A demonstrates the vesicle's potential as a nanovaccine platform. In vitro studies show enhanced immune responses with both unilamellar and multilamellar vesicles, but in vivo analyses highlight the superior efficiency of multilamellar vesicles in inducing higher antibody and cytokine levels. This work suggests ceramide-induced multilamellar lipid vesicles as an effective nanovaccine platform for enhanced antigen delivery and stability.


Subject(s)
Ovalbumin , Animals , Mice , Ovalbumin/chemistry , Ovalbumin/immunology , Biomimetic Materials/chemistry , Biomimetic Materials/pharmacology , Vaccination/methods , Lipid A/chemistry , Lipid A/analogs & derivatives , Vaccines/chemistry , Vaccines/immunology , Ceramides/chemistry , Lipids/chemistry , Nanoparticles/chemistry , Female , Mice, Inbred C57BL
8.
J Med Chem ; 67(12): 9976-9990, 2024 Jun 27.
Article in English | MEDLINE | ID: mdl-38886162

ABSTRACT

This study describes the design and synthesis of five TF-based cancer vaccine candidates using a lipid A mimetic as the carrier and a built-in adjuvant. All synthesized conjugates elicited robust and consistent TF-specific immune responses in mice without external adjuvants. Immunological studies subsequently conducted in wild-type and TLR4 knockout C57BL/6 mice demonstrated that the activation of TLR4 was the main reason that the synthesized lipid A mimetics increased the TF-specific immune responses. All antisera induced by these conjugates can specifically recognize, bind to, and induce the lysis of TF-positive cancer cells. Moreover, representative conjugates 2 and 3 could effectively reduce the growth of tumors and prolong the survival time of mice in vivo, and the efficacies were better than glycoprotein TF-CRM197 with alum adjuvant. Lipid A mimetics could therefore be a promising platform for the development of new carbohydrate-based vaccine carriers with self-adjuvanting properties for the treatment of cancer.


Subject(s)
Adjuvants, Immunologic , Cancer Vaccines , Drug Design , Lipid A , Mice, Inbred C57BL , Animals , Lipid A/analogs & derivatives , Lipid A/chemistry , Lipid A/pharmacology , Cancer Vaccines/immunology , Cancer Vaccines/pharmacology , Cancer Vaccines/chemical synthesis , Adjuvants, Immunologic/pharmacology , Adjuvants, Immunologic/chemical synthesis , Adjuvants, Immunologic/chemistry , Mice , Mice, Knockout , Humans , Female , Toll-Like Receptor 4/metabolism , Cell Line, Tumor
9.
Microbiol Res ; 283: 127712, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38593580

ABSTRACT

Lipid A plays a crucial role in Vibrio parahaemolyticus. Previously we have reported the diversity of secondary acylation of lipid A in V. parahaemolyticus and four V. parahaemolyticus genes VP_RS08405, VP_RS01045, VP_RS12170, and VP_RS00880 exhibiting homology to the secondary acyltransferases in Escherichia coli. In this study, the gene VP_RS12170 was identified as a specific lipid A secondary hydroxy-acyltransferase responsible for transferring a 3-hydroxymyristate to the 2'-position of lipid A. Four E. coli mutant strains WHL00, WHM00, WH300, and WH001 were constructed, and they would synthesize lipid A with different structures due to the absence of genes encoding lipid A secondary acyltransferases or Kdo transferase. Then V. parahaemolyticus VP_RS12170 was overexpressed in W3110, WHL00, WHM00, WH300, and WH001, and lipid A was isolated from these strains and analyzed by using thin-layer chromatography and high-performance liquid chromatography-tandem mass spectrometry. The detailed structural changes of lipid A in these mutant strains with and without VP_RS12170 overexpression were compared and conclude that VP_RS12170 can specifically transfer a 3-hydroxymyristate to the 2'-position of lipid A. This study also demonstrated that the function of VP_RS12170 is Kdo-dependent and its favorite substrate is Kdo-lipid IVA. These findings give us better understanding the biosynthetic pathway and the structural diversity of V. parahaemolyticus lipid A.


Subject(s)
Lipid A , Vibrio parahaemolyticus , Lipid A/chemistry , Lipid A/metabolism , Escherichia coli/genetics , Escherichia coli/metabolism , Vibrio parahaemolyticus/genetics , Vibrio parahaemolyticus/metabolism , Acyltransferases/genetics , Acyltransferases/metabolism , Mass Spectrometry
10.
J Med Chem ; 67(9): 7458-7469, 2024 May 09.
Article in English | MEDLINE | ID: mdl-38634150

ABSTRACT

Adjuvant is an integral part of all vaccine formulations but only a few adjuvants with limited efficacies or application scopes are available. Thus, developing more robust and diverse adjuvants is necessary. To this end, a new class of adjuvants having α- and ß-rhamnose (Rha) attached to the 1- and 6'-positions of monophosphoryl lipid A (MPLA) was designed, synthesized, and immunologically evaluated in mice. The results indicated a synergistic effect of MPLA and Rha, two immunostimulators that function via interacting with toll-like receptor 4 and recruiting endogenous anti-Rha antibodies, respectively. All the tested MPLA-Rha conjugates exhibited potent adjuvant activities to promote antibody production against both protein and carbohydrate antigens. Overall, MPLA-α-Rha exhibited better activities than MPLA-ß-Rha, and 6'-linked conjugates were slightly better than 1-linked ones. Particularly, MPLA-1-α-Rha and MPLA-6'-α-Rha were the most effective adjuvants in promoting IgG antibody responses against protein antigen keyhole limpet hemocyanin and carbohydrate antigen sTn, respectively.


Subject(s)
Lipid A , Rhamnose , Lipid A/analogs & derivatives , Lipid A/chemistry , Lipid A/pharmacology , Lipid A/immunology , Animals , Rhamnose/chemistry , Rhamnose/immunology , Rhamnose/pharmacology , Mice , Adjuvants, Vaccine/chemistry , Adjuvants, Vaccine/pharmacology , Female , Adjuvants, Immunologic/pharmacology , Adjuvants, Immunologic/chemistry , Adjuvants, Immunologic/chemical synthesis , Toll-Like Receptor 4/metabolism , Toll-Like Receptor 4/agonists , Toll-Like Receptor 4/immunology , Immunoglobulin G/immunology , Immunoglobulin G/blood , Mice, Inbred BALB C , Hemocyanins/chemistry , Hemocyanins/immunology
11.
Chemistry ; 30(32): e202400429, 2024 Jun 06.
Article in English | MEDLINE | ID: mdl-38587187

ABSTRACT

Agonists of Toll like receptors (TLRs) have attracted interest as adjuvants and immune modulators. A crystal structure of TLR4/MD2 with E. coli LPS indicates that the fatty acid at C-2 of the lipid A component of LPS induces dimerization of two TLR4-MD2 complexes, which in turn initiates cell signaling leading to the production of (pro)inflammatory cytokines. To probe the importance of the (R)-3-hydroxymyristate at C-2 of lipid A, a range of bis- and mono-phosphoryl lipid A derivatives with different modifications at C-2 were prepared by a strategy in which 2-methylnaphthyl ethers were employed as permanent protecting group that could be readily removed by catalytic hydrogenation. The C-2 amine was protected as 9-fluorenylmethyloxycarbamate, which at a later stage could be removed to give a free amine that was modified by different fatty acids. LPS and the synthetic lipid As induced the same cytokines, however, large differences in activity were observed. A compound having a hexanoyl moiety at C-2 still showed agonistic properties, but further shortening to a butanoyl abolished activity. The modifications had a larger influence on monophosphoryl lipid As. The lipid As having a butanoyl moiety at C-2 could selectively antagonize TRIF associated cytokines induced by LPS or lipid A.


Subject(s)
Cytokines , Lipid A , Lipopolysaccharides , Lipid A/chemistry , Lipid A/pharmacology , Lipid A/analogs & derivatives , Lipid A/chemical synthesis , Cytokines/metabolism , Lipopolysaccharides/pharmacology , Toll-Like Receptor 4/agonists , Toll-Like Receptor 4/metabolism , Toll-Like Receptor 4/chemistry , Humans , Lymphocyte Antigen 96/metabolism , Lymphocyte Antigen 96/chemistry , Drug Design , Structure-Activity Relationship , Signal Transduction/drug effects
12.
Glycoconj J ; 41(2): 119-131, 2024 04.
Article in English | MEDLINE | ID: mdl-38642279

ABSTRACT

Gram-negative bacteria living in marine waters have evolved peculiar adaptation strategies to deal with the numerous stress conditions that characterize aquatic environments. Among the multiple mechanisms for efficient adaptation, these bacteria typically exhibit chemical modifications in the structure of the lipopolysaccharide (LPS), which is a fundamental component of their outer membrane. In particular, the glycolipid anchor to the membrane of marine bacteria LPSs, i.e. the lipid A, frequently shows unusual chemical structures, which are reflected in equally singular immunological properties with potential applications as immune adjuvants or anti-sepsis drugs. In this work, we determined the chemical structure of the lipid A from Cellulophaga pacifica KMM 3664T isolated from the Sea of Japan. This bacterium showed to produce a heterogeneous mixture of lipid A molecules that mainly display five acyl chains and carry a single phosphate and a D-mannose disaccharide on the glucosamine backbone. Furthermore, we proved that C. pacifica KMM 3664T LPS acts as a weaker activator of Toll-like receptor 4 (TLR4) compared to the prototypical enterobacterial Salmonella typhimurium LPS. Our results are relevant to the future development of novel vaccine adjuvants and immunomodulators inspired by marine LPS chemistry.


Subject(s)
Lipid A , Lipid A/chemistry , Toll-Like Receptor 4/metabolism , Toll-Like Receptor 4/chemistry , Bacterial Outer Membrane/metabolism , Bacterial Outer Membrane/chemistry , Animals , Lipopolysaccharides/chemistry , Mice
13.
Angew Chem Int Ed Engl ; 63(24): e202402922, 2024 06 10.
Article in English | MEDLINE | ID: mdl-38581637

ABSTRACT

Lipopolysaccharide (LPS), a cell surface component of Gram-negative bacteria, activates innate immunity. Its active principle is the terminal glycolipid lipid A. Acetobacter pasteurianus is a Gram-negative bacterium used in the fermentation of traditional Japanese black rice vinegar (kurozu). In this study, we focused on A. pasteurianus lipid A, which is a potential immunostimulatory component of kurozu. The active principle structure of A. pasteurianus lipid A has not yet been identified. Herein, we first systematically synthesized three types of A. pasteurianus lipid As containing a common and unique tetrasaccharide backbone. We developed an efficient method for constructing the 2-trehalosamine skeleton utilizing borinic acid-catalyzed glycosylation to afford 1,1'-α,α-glycoside in high yield and stereoselectivity. A common tetrasaccharide intermediate with an orthogonal protecting group pattern was constructed via [2+2] glycosylation. After introducing various fatty acids, all protecting groups were removed to achieve the first chemical synthesis of three distinct types of A. pasteurianus lipid As. After evaluating their immunological function using both human and murine cell lines, we identified the active principles of A. pasteurianus LPS. We also found the unique anomeric structure of A. pasteurianus lipid A contributes to its high chemical stability.


Subject(s)
Acetobacter , Lipid A , Lipid A/chemistry , Lipid A/immunology , Lipid A/chemical synthesis , Humans , Mice , Acetobacter/chemistry , Animals , Oligosaccharides/chemistry , Oligosaccharides/chemical synthesis , Glycosylation
14.
Int J Biol Macromol ; 267(Pt 2): 131327, 2024 May.
Article in English | MEDLINE | ID: mdl-38574903

ABSTRACT

The emergence of multidrug resistance has provided a great challenge to treat nosocomial infections, which have become a major health threat around the globe. Lipid A (an active endotoxin component), the final product of the Raetz lipid A metabolism pathway, is a membrane anchor of lipopolysaccharide (LPS) of the gram-negative bacterial outer membrane. It shields bacterial cells and serves as a protective barrier from antibiotics, thereby eliciting host response and making it difficult to destroy. UDP-2,3-diacylglucosamine pyrophosphate hydrolase (LpxH), a crucial peripheral membrane enzyme of the Raetz pathway, turned out to be the potential target to inhibit the production of Lipid A. This review provides a comprehensive compilation of information regarding the structural and functional aspects of LpxH, as well as its analogous LpxI and LpxG. In addition, apart from by providing a broader understanding of the enzyme-inhibitor mechanism, this review facilitates the development of novel drug candidates that can inhibit the pathogenicity of the lethal bacterium.


Subject(s)
Gram-Negative Bacteria , Gram-Negative Bacteria/enzymology , Gram-Negative Bacteria/drug effects , Pyrophosphatases/metabolism , Pyrophosphatases/chemistry , Lipid A/chemistry , Lipid A/metabolism , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/chemistry , Humans
15.
Int J Mol Sci ; 25(5)2024 Feb 27.
Article in English | MEDLINE | ID: mdl-38474006

ABSTRACT

The lipopolysaccharide (LPS) that resides on the outermost surface and protects Gram-negative bacteria from host defenses is one of the key components leading to Salmonella infection, particularly the endotoxic lipid A domain of LPS. Lipid A modifications have been associated with several genes such as the arnT that encodes 4-amino-4-deoxy-L-arabinose transferase, which can be critical for bacteria to resist cationic antimicrobial peptides and interfere with host immune recognition. However, the association of arnT with virulence is not completely understood. Thus, this study aimed to elucidate the interrelationship of the major lipid A modification gene arnT with Salmonella Typhimurium virulence. We observed that the arnT-deficient S. Typhimurium (JOL2943), compared to the wild type (JOL401), displayed a significant decrease in several virulence phenotypes such as polymyxin B resistance, intracellular survival, swarming, and biofilm and extracellular polymeric substance (EPS) production. Interestingly, the cell-surface hydrophobicity, adhesion, and invasion characteristics remained unaffected. Additionally, LPS isolated from the mutant induced notably lower levels of endotoxicity-related cytokines in RAW and Hela cells and mice, particularly IL-1ß with a nine-fold decrease, than WT. In terms of in vivo colonization, JOL2943 showed diminished presence in internal organs such as the spleen and liver by more than 60%, while ileal infectivity remained similar to JOL401. Overall, the arnT deletion rendered the strain less virulent, with low endotoxicity, maintained gut infectivity, and reduced colonization in internal organs. With these ideal characteristics, it can be further explored as a potential attenuated Salmonella strain for therapeutics or vaccine delivery systems.


Subject(s)
Lipid A , Salmonella typhimurium , Humans , Animals , Mice , Salmonella typhimurium/genetics , Lipid A/chemistry , Lipopolysaccharides/chemistry , Virulence , Extracellular Polymeric Substance Matrix , HeLa Cells , Bacterial Proteins/genetics
16.
Arh Hig Rada Toksikol ; 74(3): 145-166, 2023 Sep 01.
Article in English | MEDLINE | ID: mdl-37791675

ABSTRACT

Polymyxin antibiotics are the last resort for treating patients in intensive care units infected with multiple-resistant Gram-negative bacteria. Due to their polycationic structure, their mode of action is based on an ionic interaction with the negatively charged lipid A portion of the lipopolysaccharide (LPS). The most prevalent polymyxin resistance mechanisms involve covalent modifications of lipid A: addition of the cationic sugar 4-amino-L-arabinose (L-Ara4N) and/or phosphoethanolamine (pEtN). The modified structure of lipid A has a lower net negative charge, leading to the repulsion of polymyxins and bacterial resistance to membrane disruption. Genes encoding the enzymatic systems involved in these modifications can be transferred either through chromosomes or mobile genetic elements. Therefore, new approaches to resistance diagnostics have been developed. On another note, interfering with these enzymatic systems might offer new therapeutic targets for drug discovery. This literature review focuses on diagnostic approaches based on structural changes in lipid A and on the therapeutic potential of molecules interfering with these changes.


Subject(s)
Lipopolysaccharides , Polymyxins , Humans , Polymyxins/therapeutic use , Polymyxins/pharmacology , Lipopolysaccharides/chemistry , Lipid A/chemistry , Drug Resistance, Bacterial/genetics , Anti-Bacterial Agents/therapeutic use , Anti-Bacterial Agents/pharmacology
17.
Carbohydr Polym ; 318: 121094, 2023 Oct 15.
Article in English | MEDLINE | ID: mdl-37479429

ABSTRACT

Lipopolysaccharides (LPS) are major players in bacterial infection through the recognition by Toll-like receptor 4 (TLR4). The LPS chemical structure, including the oligosaccharide core and the lipid A moiety, can be strongly influenced by adaptation and modulated to assure bacteria protection, evade immune surveillance, or reduce host immune responses. Deep structural understanding of TLRs signaling is essential for the modulation of the innate immune system in sepsis control and inflammation, during bacterial infection. To advance this knowledge, we have employed computational techniques to characterize the TLR4 molecular recognition of atypical LPSs from different opportunistic members of α2-Proteobacteria, including Brucella melitensis, Ochrobactrum anthropi, and Ochrobactrum intermedium, with diverse immunostimulatory activities. We contribute to unraveling the role of uncommon lipid A chemical features such as bearing very long-chain fatty acid chains, whose presence has been rarely reported, on modulating the proper heterodimerization of the TLR4 receptor complex. Moreover, we further evaluated the influence of the different oligosaccharide cores, including sugar composition and net charge, on TLR4 activation. Our studies contribute to elucidating, from the molecular and biological perspectives, the impact of the α2-Proteobacteria LPS cores and the chemical structure of the atypical lipid A for immune system evasion in opportunistic bacteria.


Subject(s)
Bacterial Infections , Lipopolysaccharides , Humans , Lipopolysaccharides/chemistry , Toll-Like Receptor 4 , Lipid A/chemistry , Proteobacteria , Immune Evasion , Bacteria , Oligosaccharides
18.
J Am Soc Mass Spectrom ; 34(6): 1047-1055, 2023 Jun 07.
Article in English | MEDLINE | ID: mdl-37184080

ABSTRACT

FLATn is a tandem mass spectrometric technique that can be used to rapidly generate spectral information applicable for structural elucidation of lipids like lipid A from Gram-negative bacterial species from a single bacterial colony. In this study, we extend the scope and capability of FLATn by tandem MS fragmentation of lithium-adducted molecular lipid A anions and fragments (FLATn-Li) that provides additional structural and diagnostic data from FLATn samples allowing for the discrimination of terminal phosphate modifications in a variety of pathogenic and environmental species. Using FLATn-Li, we elucidated the lipid A structure from several bacterial species, including novel structures from arctic bacterioplankton of the Duganella and Massilia genera that favor 4-amino-4-deoxy-l-arabinopyranose (Ara4N) modification at the 1-phosphate position and that demonstrate double glycosylation with Ara4N at the 1 and 4' phosphate positions simultaneously. The structures characterized in this work demonstrate that some environmental psychrophilic species make extensive use of this structural lipid A modification previously characterized as a pathogenic adaptation and the structural basis of resistance to cationic antimicrobial peptides. This observation extends the role of phosphate modification(s) in environmental species adaptation and suggests that Ara4N modification can functionally replace the positive charge of the phosphoethanolamine modification that is more typically found attached to the 1-phosphate position of modified lipid A.


Subject(s)
Lipid A , Lithium , Lipid A/chemistry , Glycosylation , Bacterial Proteins/chemistry , Escherichia coli/metabolism , Phosphates , Ions
19.
FASEB J ; 37(5): e22928, 2023 05.
Article in English | MEDLINE | ID: mdl-37071453

ABSTRACT

Colistin (polymyxin E) is a group of cationic antimicrobial cyclic peptides and is recognized as a last-resort defense against lethal infections with carbapenem-resistant pathogens. In addition to the plasmid-borne mobilized phosphoethanolamine (PEA) transferases, the functional expression of lipid A-modifying enzymes encoded on chromosomes has been attributed to intrinsic bacterial colistin resistance. However, the mechanisms of colistin resistance in Riemerella anatipestifer remain unknown. Herein, the GE296_RS09715 gene-encoded Lipid A PEA transferases (RaEptA) was identified in R. anatipestifer. Genetic and structural analyses revealed that the amino acid sequence of RaEptA shared 26.6%-33.1% similarities with the family of Lipid A PEA transferases (EptA) and MCR-like proteins and have defined 12 residues that contribute to the formation of phosphatidylethanolamine (PE)-recognizable cavities. Comparative analyses of colistin resistance in RA-LZ01 and RA-LZ01ΔRaEptA showed the level of colistin has fallen from 96 µg mL-1 down to 24 ~ 32 µg mL-1 . Site-directed mutagenesis assay of the PE-binding cavity and expression of the mutants reveals that K309-rRaEptA can remodel the surface of Escherichia coli and rendering it resistant to colistin, suggesting this point-mutation of P309K is necessary for EptA-mediated lipid A modification. Moreover, the virulence of RA-LZ01ΔRaEptA was attenuated compared with RA-LZ01 both in vivo and vitro. Taken together, the results represent the RaEptA involved in the colistin resistance and pathogenicity, and the P309K mutation might alter bacterial adaptation and increase the spread of colistin resistance from R. anatipestifer to other gram-negative bacteria. The findings of this study suggest another scenario for the spread of colistin resistance genes and should be considered by a wide audience.


Subject(s)
Anti-Bacterial Agents , Colistin , Colistin/pharmacology , Colistin/chemistry , Anti-Bacterial Agents/pharmacology , Virulence/genetics , Lipid A/chemistry , Drug Resistance, Bacterial/genetics , Escherichia coli/genetics , Escherichia coli/metabolism , Phenotype , Transferases
20.
J Bacteriol ; 205(5): e0006723, 2023 05 25.
Article in English | MEDLINE | ID: mdl-37070977

ABSTRACT

Gram-negative bacteria have a unique cell surface that can be modified to maintain bacterial fitness in diverse environments. A well-defined example is the modification of the lipid A component of lipopolysaccharide (LPS), which promotes resistance to polymyxin antibiotics and antimicrobial peptides. In many organisms, such modifications include the addition of the amine-containing constituents 4-amino-4-deoxy-l-arabinose (l-Ara4N) and phosphoethanolamine (pEtN). Addition of pEtN is catalyzed by EptA, which uses phosphatidylethanolamine (PE) as its substrate donor, resulting in production of diacylglycerol (DAG). DAG is then quickly recycled into glycerophospholipid (GPL) synthesis by the DAG kinase A (DgkA) to produce phosphatidic acid, the major GPL precursor. Previously, we hypothesized that loss of DgkA recycling would be detrimental to the cell when LPS is heavily modified. Instead, we found that DAG accumulation inhibits EptA activity, preventing further degradation of PE, the predominant GPL of the cell. However, DAG inhibition of pEtN addition results in complete loss of polymyxin resistance. Here, we selected for suppressors to find a mechanism of resistance independent of DAG recycling or pEtN modification. Disrupting the gene encoding the adenylate cyclase, cyaA, fully restored antibiotic resistance without restoring DAG recycling or pEtN modification. Supporting this, disruptions of genes that reduce CyaA-derived cAMP formation (e.g., ptsI) or disruption of the cAMP receptor protein, Crp, also restored resistance. We found that loss of the cAMP-CRP regulatory complex was necessary for suppression and that resistance arises from a substantial increase in l-Ara4N-modified LPS, bypassing the need for pEtN modification. IMPORTANCE Gram-negative bacteria can alter the structure of their LPS to promote resistance to cationic antimicrobial peptides, including polymyxin antibiotics. Polymyxins are considered last-resort antibiotics for treatment against multidrug-resistant Gram-negative organisms. Here, we explore how changes in general metabolism and carbon catabolite repression pathways can alter LPS structure and influence polymyxin resistance.


Subject(s)
Lipopolysaccharides , Polymyxin B , Polymyxin B/pharmacology , Lipopolysaccharides/metabolism , Escherichia coli/genetics , Escherichia coli/metabolism , Cyclic AMP Receptor Protein/metabolism , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/metabolism , Polymyxins/pharmacology , Lipid A/chemistry , Drug Resistance, Bacterial/genetics
SELECTION OF CITATIONS
SEARCH DETAIL