Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 657
Filter
1.
Nature ; 633(8030): 662-669, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39261738

ABSTRACT

The ability to sequence single protein molecules in their native, full-length form would enable a more comprehensive understanding of proteomic diversity. Current technologies, however, are limited in achieving this goal1,2. Here, we establish a method for the long-range, single-molecule reading of intact protein strands on a commercial nanopore sensor array. By using the ClpX unfoldase to ratchet proteins through a CsgG nanopore3,4, we provide single-molecule evidence that ClpX translocates substrates in two-residue steps. This mechanism achieves sensitivity to single amino acids on synthetic protein strands hundreds of amino acids in length, enabling the sequencing of combinations of single-amino-acid substitutions and the mapping of post-translational modifications, such as phosphorylation. To enhance classification accuracy further, we demonstrate the ability to reread individual protein molecules multiple times, and we explore the potential for highly accurate protein barcode sequencing. Furthermore, we develop a biophysical model that can simulate raw nanopore signals a priori on the basis of residue volume and charge, enhancing the interpretation of raw signal data. Finally, we apply these methods to examine full-length, folded protein domains for complete end-to-end analysis. These results provide proof of concept for a platform that has the potential to identify and characterize full-length proteoforms at single-molecule resolution.


Subject(s)
Nanopores , Proteins , Sequence Analysis, Protein , Single Molecule Imaging , Amino Acid Substitution , Endopeptidase Clp/chemistry , Endopeptidase Clp/metabolism , Phosphorylation , Protein Domains , Protein Processing, Post-Translational , Proteins/chemistry , Proteins/metabolism , Sequence Analysis, Protein/methods , Single Molecule Imaging/methods
2.
J Med Chem ; 67(17): 15131-15147, 2024 Sep 12.
Article in English | MEDLINE | ID: mdl-39221504

ABSTRACT

A series of arylsulfones and heteroarylsulfones have previously been demonstrated to dysregulate the conserved bacterial ClpP protease, causing the unspecific degradation of essential cellular housekeeping proteins and ultimately resulting in cell death. A cocrystal structure of a 2-ß-sulfonylamide analog, ACP1-06, with Escherichia coli ClpP showed that its 2-pyridyl sulfonyl substituent adopts two orientations in the binding site related through a sulfone bond rotation. From this, a new bis-aryl phosphine oxide scaffold, designated as ACP6, was designed based on a "conformation merging" approach of the dual orientation of the ACP1-06 sulfone. One analog, ACP6-12, exhibited over a 10-fold increase in activity over the parent ACP1-06 compound, and a cocrystal X-ray structure with ClpP confirmed its predicted binding conformation. This allowed for a comparative analysis of how different ligand classes bind to the hydrophobic binding site. The study highlights the successful application of structure-based rational design of novel phosphine oxide-based antibiotics.


Subject(s)
Anti-Bacterial Agents , Drug Design , Endopeptidase Clp , Escherichia coli , Oxides , Phosphines , Phosphines/chemistry , Phosphines/pharmacology , Endopeptidase Clp/metabolism , Endopeptidase Clp/antagonists & inhibitors , Endopeptidase Clp/chemistry , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/chemical synthesis , Oxides/chemistry , Escherichia coli/enzymology , Escherichia coli/drug effects , Structure-Activity Relationship , Escherichia coli Proteins/metabolism , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/antagonists & inhibitors , Crystallography, X-Ray , Models, Molecular , Binding Sites , Molecular Structure
3.
Molecules ; 29(17)2024 Sep 09.
Article in English | MEDLINE | ID: mdl-39275125

ABSTRACT

Acetylation modification has become one of the most popular topics in protein post-translational modification (PTM) research and plays an important role in bacterial virulence. A previous study indicated that the virulence-associated caseinolytic protease proteolytic subunit (ClpP) is acetylated at the K165 site in Vibrio alginolyticus strain HY9901, but its regulation regarding the virulence of V. alginolyticus is still unknown. We further confirmed that ClpP undergoes lysine acetylation (Kace) modification by immunoprecipitation and Western blot analysis and constructed the complementation strain (C-clpP) and site-directed mutagenesis strains including K165Q and K165R. The K165R strain significantly increased biofilm formation at 36 h of incubation, and K165Q significantly decreased biofilm formation at 24 h of incubation. However, the acetylation modification of ClpP did not affect the extracellular protease (ECPase) activity. In addition, we found that the virulence of K165Q was significantly reduced in zebrafish by in vivo injection. To further study the effect of lysine acetylation on the pathogenicity of V. alginolyticus, GS cells were infected with four strains, namely HY9901, C-clpP, K165Q and K165R. This indicated that the effect of the K165Q strain on cytotoxicity was significantly reduced compared with the wild-type strain, while K165R showed similar levels to the wild-type strain. In summary, the results of this study indicate that the Kace of ClpP is involved in the regulation of the virulence of V. alginolyticus.


Subject(s)
Biofilms , Endopeptidase Clp , Lysine , Protein Processing, Post-Translational , Vibrio alginolyticus , Zebrafish , Vibrio alginolyticus/pathogenicity , Vibrio alginolyticus/genetics , Vibrio alginolyticus/metabolism , Acetylation , Lysine/metabolism , Virulence , Endopeptidase Clp/metabolism , Endopeptidase Clp/genetics , Animals , Biofilms/growth & development , Bacterial Proteins/metabolism , Bacterial Proteins/genetics
4.
mSphere ; 9(9): e0036324, 2024 Sep 25.
Article in English | MEDLINE | ID: mdl-39189774

ABSTRACT

ClpXP is a protease complex that plays important roles in protein quality control and cell cycle regulation, but the functions of multiple ClpXs and multiple ClpPs in M. xanthus remain unknown. The genome of Myxococcus xanthus DK1622 contains two clpPs and three clpXs. The clpP1 and clpX1 genes are cotranscribed and are both essential, while the other copies are isolated in the genome and are deletable. The deletion of clpX2 caused the mutant to be deficient in fruiting body development, while the clpX3 gene is involved in resistance to thermal stress. Both ClpPs possess catalytic active sites, but only ClpP1 shows in vitro peptidase activity on the typical substrate Suc-LY-AMC. All of these clpP and clpX genes exhibit strong transcriptional upregulation in the stationary phase, and the transcription of the three clpX genes appears to be coordinated. Our results demonstrated that multiple ClpPs and multiple ClpXs are functionally divergent and may assist in the environmental adaptation and functional diversification of M. xanthus.IMPORTANCEClpXP is an important protease complex of bacteria and is involved in various physiological processes. Myxococcus xanthus DK1622 possesses two ClpPs and three ClpXs with unclear functions. We investigated the functions of these genes and demonstrated the essential roles of clpP1 and clpX1. Only ClpP1 has in vitro peptidase activity on Suc-LY-AMC, and the isolated clpX copies participate in distinct cellular processes. All of these genes exhibited significant transcriptional upregulation in the stationary phase. Divergent functions appear in multiple ClpPs and multiple ClpXs in M. xanthus DK1622.


Subject(s)
Bacterial Proteins , Endopeptidase Clp , Gene Expression Regulation, Bacterial , Myxococcus xanthus , Myxococcus xanthus/genetics , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Endopeptidase Clp/genetics , Endopeptidase Clp/metabolism
5.
ACS Chem Biol ; 19(8): 1794-1802, 2024 Aug 16.
Article in English | MEDLINE | ID: mdl-39096241

ABSTRACT

Protein degradation is a tightly regulated biological process that maintains bacterial proteostasis. ClpPs are a highly conserved family of serine proteases that associate with the AAA + ATPase (an ATPase associated with diverse cellular activities) to degrade protein substrates. Identification and biochemical characterization of protein substrates for the AAA + ATPase-dependent ClpP degradation systems are considered essential for gaining an understanding of the molecular operation of the complex ClpP degradation machinery. Consequently, expanding the repertoire of protein substrates that can be degraded in vitro and within bacterial cells is necessary. Here, we report that AAA + ATPase-ClpP proteolytic complexes promote degradation of the secondary metabolite surfactin synthetases SrfAA, SrfAB, and SrfAC in Bacillus subtilis. On the basis of in vitro and in-cell studies coupled with activity-based protein profiling of nonribosomal peptide synthetases, we showed that SrfAC is targeted to the ClpC-ClpP proteolytic complex, whereas SrfAA is hydrolyzed not only by the ClpC-ClpP proteolytic complex but also by different ClpP proteolytic complexes. Furthermore, SrfAB does not appear to be a substrate for the ClpC-ClpP proteolytic complex, thereby implying that other ClpP proteolytic complexes are involved in the degradation of this surfactin synthetase. Natural product biosynthesis is regulated by the AAA + ATPase-ClpP degradation system, indicating that protein degradation plays a role in the regulatory stages of biosynthesis. However, few studies have examined the regulation of protein degradation levels. Furthermore, SrfAA, SrfAB, and SrfAC were identified as protein substrates for AAA + ATPase-ClpP degradation systems, thereby contributing to a better understanding of the complex ClpP degradation machinery.


Subject(s)
Bacillus subtilis , Bacterial Proteins , Biological Products , Endopeptidase Clp , Proteolysis , Endopeptidase Clp/metabolism , Biological Products/metabolism , Biological Products/chemistry , Bacillus subtilis/enzymology , Bacillus subtilis/metabolism , Bacterial Proteins/metabolism , Peptide Synthases/metabolism , Adenosine Triphosphatases/metabolism
6.
Environ Microbiol ; 26(7): e16677, 2024 Jul.
Article in English | MEDLINE | ID: mdl-39039821

ABSTRACT

Heat stress can lead to protein misfolding and aggregation, potentially causing cell death due to the loss of essential proteins. Bacteria, being particularly exposed to environmental stress, are equipped with disaggregases that rescue these aggregated proteins. The bacterial Hsp70 chaperone DnaK and the ATPase associated with diverse cellular activities protein ClpB form the canonical disaggregase in bacteria. While this combination operates effectively during physiological heat stress, it is ineffective against massive aggregation caused by temperature-based sterilization protocols used in the food industry and clinics. This leaves bacteria unprotected against these thermal processes. However, bacteria that can withstand extreme, man-made stress conditions have emerged. These bacteria possess novel ATPase associated with diverse cellular activities disaggregases, ClpG and ClpL, which are key players in extreme heat resistance. These disaggregases, present in selected Gram-negative or Gram-positive bacteria, respectively, function superiorly by exhibiting increased thermal stability and enhanced threading power compared to DnaK/ClpB. This enables ClpG and ClpL to operate at extreme temperatures and process large and tight protein aggregates, thereby contributing to heat resistance. The genes for ClpG and ClpL are often encoded on mobile genomic islands or conjugative plasmids, allowing for their rapid spread among bacteria via horizontal gene transfer. This threatens the efficiency of sterilization protocols. In this review, we describe the various bacterial disaggregases identified to date, characterizing their commonalities and the specific features that enable these novel disaggregases to provide stress protection against extreme stress conditions.


Subject(s)
Bacterial Proteins , Bacterial Proteins/metabolism , Bacterial Proteins/genetics , HSP70 Heat-Shock Proteins/metabolism , HSP70 Heat-Shock Proteins/genetics , Thermotolerance , Hot Temperature , Bacteria/genetics , Bacteria/metabolism , Endopeptidase Clp/metabolism , Endopeptidase Clp/genetics , Heat-Shock Proteins/metabolism , Heat-Shock Proteins/genetics , Heat-Shock Response
7.
Genes Cells ; 29(9): 695-709, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38965067

ABSTRACT

In cells, proteins are synthesized, function, and degraded (dead). Protein synthesis (spring) is important for the life of proteins. However, how proteins die is equally important for organisms. Proteases are secreted from cells and used as nutrients to break down external proteins. Proteases degrade unwanted and harmful cellular proteins. In eukaryotes, a large enzyme complex called the proteasome is primarily responsible for cellular protein degradation. Prokaryotes, such as bacteria, have similar protein degradation systems. In this review, we describe the structure and function of the ClpXP complex in the degradation system, which is an ATP-dependent protease in bacterial cells, with a particular focus on ClpP.


Subject(s)
Endopeptidase Clp , Proteolysis , Endopeptidase Clp/metabolism , Proteasome Endopeptidase Complex/metabolism , Bacterial Proteins/metabolism , Bacterial Proteins/genetics , Chaperonins/metabolism
8.
J Basic Microbiol ; 64(9): e2400030, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39031597

ABSTRACT

Streptococcus suis is an important zoonotic pathogen, causing cytokine storms of Streptococcal toxic shock-like syndrome amongst humans after a wound infection into the bloodstream. To overcome the challenges of fever and leukocyte recruitment, invasive S. suis must deploy multiple stress responses forming a network and utilize proteases to degrade short-lived regulatory and misfolded proteins induced by adverse stresses, thereby adapting and evading host immune responses. In this study, we found that S. suis encodes multiple ATP-dependent proteases, including single-chain FtsH and double-subunit Clp protease complexes ClpAP, ClpBP, ClpCP, and ClpXP, which were activated as the fever of infected mice in vivo. The expression of genes ftsH, clpA/B/C, and clpP, but not clpX, were significantly upregulated in S. suis in response to heat stress, while were not changed notably under the treatments with several other stresses, including oxidative, acidic, and cold stimulation. FtsH and ClpP were required for S. suis survival within host blood under heat stress in vitro and in vivo. Deletion of ftsH or clpP attenuated the tolerance of S. suis to heat, oxidative and acidic stresses, and significantly impaired the bacterial survival within macrophages. Further analysis identified that repressor CtsR directly binds and controls the clpA/B/C and clpP operons and is relieved by heat stress. In summary, the deployments of multiple ATP-dependent proteases form a flexible heat stress response network that appears to allow S. suis to fine-tune the degradation or refolding of the misfolded proteins to maintain cellular homeostasis and optimal survival during infection.


Subject(s)
Bacterial Proteins , Streptococcal Infections , Streptococcus suis , Streptococcus suis/enzymology , Streptococcus suis/genetics , Streptococcus suis/pathogenicity , Animals , Mice , Bacterial Proteins/metabolism , Bacterial Proteins/genetics , Streptococcal Infections/microbiology , Heat-Shock Response , ATP-Dependent Proteases/metabolism , ATP-Dependent Proteases/genetics , Gene Expression Regulation, Bacterial , Macrophages/microbiology , Macrophages/immunology , Endopeptidase Clp/metabolism , Endopeptidase Clp/genetics , Female
9.
Genes (Basel) ; 15(6)2024 May 27.
Article in English | MEDLINE | ID: mdl-38927630

ABSTRACT

LONP1 is the principal AAA+ unfoldase and bulk protease in the mitochondrial matrix, so its deletion causes embryonic lethality. The AAA+ unfoldase CLPX and the peptidase CLPP also act in the matrix, especially during stress periods, but their substrates are poorly defined. Mammalian CLPP deletion triggers infertility, deafness, growth retardation, and cGAS-STING-activated cytosolic innate immunity. CLPX mutations impair heme biosynthesis and heavy metal homeostasis. CLPP and CLPX are conserved from bacteria to humans, despite their secondary role in proteolysis. Based on recent proteomic-metabolomic evidence from knockout mice and patient cells, we propose that CLPP acts on phase-separated ribonucleoprotein granules and CLPX on multi-enzyme condensates as first-aid systems near the inner mitochondrial membrane. Trimming within assemblies, CLPP rescues stalled processes in mitoribosomes, mitochondrial RNA granules and nucleoids, and the D-foci-mediated degradation of toxic double-stranded mtRNA/mtDNA. Unfolding multi-enzyme condensates, CLPX maximizes PLP-dependent delta-transamination and rescues malformed nascent peptides. Overall, their actions occur in granules with multivalent or hydrophobic interactions, separated from the aqueous phase. Thus, the role of CLPXP in the matrix is compartment-selective, as other mitochondrial peptidases: MPPs at precursor import pores, m-AAA and i-AAA at either IMM face, PARL within the IMM, and OMA1/HTRA2 in the intermembrane space.


Subject(s)
Endopeptidase Clp , Heme , Mice, Knockout , Mitochondria , Mitochondrial Proteins , Endopeptidase Clp/metabolism , Endopeptidase Clp/genetics , Animals , Mice , Mitochondria/metabolism , Mitochondria/genetics , Mitochondrial Proteins/metabolism , Mitochondrial Proteins/genetics , Heme/metabolism , Protein Biosynthesis , Humans , Mitochondrial Membranes/metabolism , Stress, Physiological
10.
Redox Biol ; 73: 103203, 2024 07.
Article in English | MEDLINE | ID: mdl-38823208

ABSTRACT

Vascular smooth muscle cells (VSMCs), known for their remarkable lifelong phenotypic plasticity, play a pivotal role in vascular pathologies through their ability to transition between different phenotypes. Our group discovered that the deficiency of the mitochondrial protein Poldip2 induces VSMC differentiation both in vivo and in vitro. Further comprehensive biochemical investigations revealed Poldip2's specific interaction with the mitochondrial ATPase caseinolytic protease chaperone subunit X (CLPX), which is the regulatory subunit for the caseinolytic protease proteolytic subunit (ClpP) that forms part of the ClpXP complex - a proteasome-like protease evolutionarily conserved from bacteria to humans. This interaction limits the protease's activity, and reduced Poldip2 levels lead to ClpXP complex activation. This finding prompted the hypothesis that ClpXP complex activity within the mitochondria may regulate the VSMC phenotype. Employing gain-of-function and loss-of-function strategies, we demonstrated that ClpXP activity significantly influences the VSMC phenotype. Notably, both genetic and pharmacological activation of ClpXP inhibits VSMC plasticity and fosters a quiescent, differentiated, and anti-inflammatory VSMC phenotype. The pharmacological activation of ClpP using TIC10, currently in phase III clinical trials for cancer, successfully replicates this phenotype both in vitro and in vivo and markedly reduces aneurysm development in a mouse model of elastase-induced aortic aneurysms. Our mechanistic exploration indicates that ClpP activation regulates the VSMC phenotype by modifying the cellular NAD+/NADH ratio and activating Sirtuin 1. Our findings reveal the crucial role of mitochondrial proteostasis in the regulation of the VSMC phenotype and propose the ClpP protease as a novel, actionable target for manipulating the VSMC phenotype.


Subject(s)
Endopeptidase Clp , Mitochondria , Muscle, Smooth, Vascular , Myocytes, Smooth Muscle , Phenotype , Sirtuin 1 , Animals , Humans , Mice , Cell Differentiation , Endopeptidase Clp/metabolism , Endopeptidase Clp/genetics , Mitochondria/metabolism , Mitochondrial Proteins/metabolism , Mitochondrial Proteins/genetics , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/cytology , Myocytes, Smooth Muscle/metabolism , Sirtuin 1/metabolism , Sirtuin 1/genetics
11.
mBio ; 15(7): e0138924, 2024 Jul 17.
Article in English | MEDLINE | ID: mdl-38920392

ABSTRACT

The host protein calprotectin inhibits the growth of a variety of bacterial pathogens through metal sequestration in a process known as "nutritional immunity." Staphylococcus aureus growth is inhibited by calprotectin in vitro, and calprotectin is localized in vivo to staphylococcal abscesses during infection. However, the staphylococcal adaptations that provide defense against nutritional immunity and the role of metal-responsive regulators are not fully characterized. In this work, we define the transcriptional response of S. aureus and the role of the metal-responsive regulators, Zur, Fur, and MntR, in response to metal limitation by calprotectin exposure. Additionally, we identified genes affecting the fitness of S. aureus during metal limitation through a Transposon sequencing (Tn-seq) approach. Loss of function mutations in clpP, which encodes a proteolytic subunit of the ATP-dependent Clp protease, demonstrate reduced fitness of S. aureus to the presence of calprotectin. ClpP contributes to pathogenesis in vivo in a calprotectin-dependent manner. These studies establish a critical role for ClpP to combat metal limitation by calprotectin and reveal the genes required for S. aureus to outcompete the host for metals. IMPORTANCE: Staphylococcus aureus is a leading cause of skin and soft tissue infections, bloodstream infections, and endocarditis. Antibiotic treatment failures during S. aureus infections are increasingly prevalent, highlighting the need for novel antimicrobial agents. Metal chelator-based therapeutics have tremendous potential as antimicrobials due to the strict requirement for nutrient metals exhibited by bacterial pathogens. The high-affinity transition metal-binding properties of calprotectin represents a potential therapeutic strategy that functions through metal chelation. Our studies provide a foundation to define mechanisms by which S. aureus combats nutritional immunity and may be useful for the development of novel therapeutics to counter the ability of S. aureus to survive in a metal-limited environment.


Subject(s)
Leukocyte L1 Antigen Complex , Staphylococcal Infections , Staphylococcus aureus , Leukocyte L1 Antigen Complex/metabolism , Staphylococcus aureus/genetics , Staphylococcus aureus/drug effects , Staphylococcus aureus/metabolism , Staphylococcal Infections/microbiology , Metals/metabolism , Bacterial Proteins/metabolism , Bacterial Proteins/genetics , Animals , Gene Expression Regulation, Bacterial , Endopeptidase Clp/metabolism , Endopeptidase Clp/genetics , Mice , Adaptation, Physiological
12.
Neoplasia ; 55: 101015, 2024 09.
Article in English | MEDLINE | ID: mdl-38944913

ABSTRACT

Hepatocellular carcinoma (HCC) is the most common form of liver cancer, accounting for approximately 90 % of all cases. ONC201, a member of the imipridone drug family, has shown promising therapeutic potential and a good safety profile in both malignant pediatric central nervous system tumors (diffuse midline glioma [DMG]) and hematologic malignancies. ONC206 is a more potent analog of ONC201. However, the ONC206 potential and mechanism of action in HCC remain to be elucidated. We found that ONC206 hindered HCC growth by suppressing cell proliferation and inducing apoptosis. Moreover, ONC206 induced cytoprotective autophagy, and blocking autophagy enhanced the proapoptotic effect of ONC206. Additionally, ONC206 induced mitochondrial swelling, reduced the mitochondrial membrane potential (MMP), and led to the accumulation of mitochondrial ROS in HCC cells, ultimately resulting in mitochondrial dysfunction. The HCC patient samples exhibited notably elevated levels of caseinolytic protease proteolytic subunit (ClpP), which serves as a mediator of ONC206-induced mitochondrial dysfunction and the activation of protective autophagy. knockdown of ClpP reversed the cytotoxic effects of ONC206 on HCC cells. In summary, our results provide the first insight into the mechanism by which ONC206 exerts its anti-HCC effects and induces protective autophagy in HCC cells through ClpP.


Subject(s)
Apoptosis , Autophagy , Carcinoma, Hepatocellular , Endopeptidase Clp , Liver Neoplasms , Mitochondria , Humans , Carcinoma, Hepatocellular/pathology , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/genetics , Liver Neoplasms/pathology , Liver Neoplasms/metabolism , Liver Neoplasms/drug therapy , Liver Neoplasms/genetics , Autophagy/drug effects , Mitochondria/metabolism , Mitochondria/drug effects , Cell Line, Tumor , Endopeptidase Clp/metabolism , Endopeptidase Clp/genetics , Apoptosis/drug effects , Cell Proliferation/drug effects , Animals , Mice , Membrane Potential, Mitochondrial/drug effects , Reactive Oxygen Species/metabolism , Antineoplastic Agents/pharmacology , Xenograft Model Antitumor Assays , Imidazoles/pharmacology , Benzyl Compounds , Heterocyclic Compounds, 3-Ring
13.
Cell Stress Chaperones ; 29(4): 540-551, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38908470

ABSTRACT

Anaplasma phagocytophilum is an intracellular tick-transmitted bacterial pathogen that infects neutrophils in mammals and causes granulocytic anaplasmosis. In this study, we investigated the molecular chaperones ClpB and DnaK from A. phagocytophilum. In Escherichia coli, ClpB cooperates with DnaK and its co-chaperones DnaJ and GrpE in ATP-dependent reactivation of aggregated proteins. Since ClpB is not produced in metazoans, it is a promising target for developing antimicrobial therapies, which generates interest in studies on that chaperone's role in pathogenic bacteria. We found that ClpB and DnaK are transcriptionally upregulated in A. phagocytophilum 3-5 days after infection of human HL-60 and tick ISE6 cells, which suggests an essential role of the chaperones in supporting the pathogen's intracellular life cycle. Multiple sequence alignments show that A. phagocytophilum ClpB and DnaK contain all structural domains that were identified in their previously studied orthologs from other bacteria. Both A. phagocytophilum ClpB and DnaK display ATPase activity, which is consistent with their participation in the ATP-dependent protein disaggregation system. However, despite a significant sequence similarity between the chaperones from A. phagocytophilum and those from E. coli, the former were not as effective as their E. coli orthologs during reactivation of aggregated proteins in vitro and in supporting the survival of E. coli cells under heat stress. We conclude that the A. phagocytophilum chaperones might have evolved with distinct biochemical properties to maintain the integrity of pathogenic proteins under unique stress conditions of an intracellular environment of host cells.


Subject(s)
Anaplasma phagocytophilum , Bacterial Proteins , HSP70 Heat-Shock Proteins , Anaplasma phagocytophilum/metabolism , HSP70 Heat-Shock Proteins/metabolism , Humans , Bacterial Proteins/metabolism , Bacterial Proteins/chemistry , Endopeptidase Clp/metabolism , Escherichia coli/metabolism , Animals , HL-60 Cells , Amino Acid Sequence , Adenosine Triphosphatases/metabolism , Heat-Shock Proteins/metabolism
14.
Biochem Pharmacol ; 226: 116394, 2024 08.
Article in English | MEDLINE | ID: mdl-38942090

ABSTRACT

Concurrent infection in breast cancer patients is the direct cause of the high mortality rate of the disease. However, there is no available method to increase the survival rate until now. To address the problem, we propose one drug with two target strategy to treat the refractory disease. A small chemical, ph-ph+, was attempted to be used in the study to explore the feasibility of the approach in anticancer and antifungus at the same time. The results showed that ph-ph+ could prevent the proliferation and metastasis of breast cancer cells, and kill C. albicans simultaneously. The molecular mechanism was associated with the activation of an evolutionarily conserved protease CLpP in the cancer and C. albicans cells. Also, the signaling pathway mediated by PLAGL2 that highly expressed in cancer cells participated in preventing cell metastasis and inducing apoptosis of ph-ph+. The one drug with dual targets inhibited the growth and metastasis of the cancer cells, and meanwhile eliminated C. albicans in tissues in the experimental animals. The results suggested that ph-ph+ with dual targets of CLpP and PLAGL2 would be a feasible approach to prolong the survival rate in patients with metastatic breast cancer and pathogenic infection.


Subject(s)
Breast Neoplasms , Candida albicans , Candidiasis , Humans , Female , Breast Neoplasms/pathology , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Animals , Candida albicans/drug effects , Candidiasis/drug therapy , Mice , Cell Line, Tumor , Antifungal Agents/pharmacology , Antifungal Agents/therapeutic use , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Endopeptidase Clp/metabolism , Endopeptidase Clp/antagonists & inhibitors , Endopeptidase Clp/genetics , Mice, Inbred BALB C , Neoplasm Metastasis , Mice, Nude , Cell Proliferation/drug effects
15.
Nucleic Acids Res ; 52(9): 5195-5208, 2024 May 22.
Article in English | MEDLINE | ID: mdl-38567730

ABSTRACT

Bacterial defence systems are tightly regulated to avoid autoimmunity. In Type I restriction-modification (R-M) systems, a specific mechanism called restriction alleviation (RA) controls the activity of the restriction module. In the case of the Escherichia coli Type I R-M system EcoKI, RA proceeds through ClpXP-mediated proteolysis of restriction complexes bound to non-methylated sites that appear after replication or reparation of host DNA. Here, we show that RA is also induced in the presence of plasmids carrying EcoKI recognition sites, a phenomenon we refer to as plasmid-induced RA. Further, we show that the anti-restriction behavior of plasmid-borne non-conjugative transposons such as Tn5053, previously attributed to their ardD loci, is due to plasmid-induced RA. Plasmids carrying both EcoKI and Chi sites induce RA in RecA- and RecBCD-dependent manner. However, inactivation of both RecA and RecBCD restores RA, indicating that there exists an alternative, RecA-independent, homologous recombination pathway that is blocked in the presence of RecBCD. Indeed, plasmid-induced RA in a RecBCD-deficient background does not depend on the presence of Chi sites. We propose that processing of random dsDNA breaks in plasmid DNA via homologous recombination generates non-methylated EcoKI sites, which attract EcoKI restriction complexes channeling them for ClpXP-mediated proteolysis.


Subject(s)
Escherichia coli Proteins , Escherichia coli , Plasmids , Rec A Recombinases , Plasmids/genetics , Escherichia coli/genetics , Rec A Recombinases/metabolism , Rec A Recombinases/genetics , Escherichia coli Proteins/metabolism , Escherichia coli Proteins/genetics , Recombination, Genetic , Deoxyribonucleases, Type I Site-Specific/metabolism , Deoxyribonucleases, Type I Site-Specific/genetics , Endopeptidase Clp/metabolism , Endopeptidase Clp/genetics , Exodeoxyribonuclease V/metabolism , Exodeoxyribonuclease V/genetics , DNA, Bacterial/metabolism , DNA Transposable Elements/genetics , DNA Restriction Enzymes , DNA-Binding Proteins
16.
Int J Biol Macromol ; 266(Pt 2): 131371, 2024 May.
Article in English | MEDLINE | ID: mdl-38580013

ABSTRACT

Bacterial caseinolytic protease-chaperone complexes participate in the elimination of misfolded and aggregated protein substrates. The spirochete Leptospira interrogans possess a set of Clp-chaperones (ClpX, ClpA, and ClpC), which may associate functionally with two different isoforms of LinClpP (ClpP1 and ClpP2). The L. interrogans ClpC (LinClpC) belongs to class-I chaperone with two active ATPase domains separated by a middle domain. Using the size exclusion chromatography, ANS dye binding, and dynamic light scattering analysis, the LinClpC is suggested to undergo nucleotide-induced oligomerization. LinClpC associates with either pure LinClpP1 or LinClpP2 isoforms non-preferentially and with equal affinity. Regardless, pure LinClpP isoforms cannot constitute an active protease complex with LinClpC. Interestingly, the heterocomplex LinClpP1P2 in association with LinClpC forms a functional proteolytic machinery and degrade ß-casein or FITC-casein in an energy-independent manner. Adding either ATP or ATPγS further fosters the LinClpCP1P2 complex protease activity by nurturing the functional oligomerization of LinClpC. The antibiotic, acyldepsipeptides (ADEP1) display a higher activatory role on LinClpP1P2 protease activity than LinClpC. Altogether, this work illustrates an in-depth study of hetero-tetradecamer LinClpP1P2 association with its cognate ATPase and unveils a new insight into the structural reorganization of LinClpP1P2 in the presence of chaperone, LinClpC to gain protease activity.


Subject(s)
Bacterial Proteins , Heat-Shock Proteins , Leptospira , Protein Multimerization , Adenosine Triphosphate/metabolism , Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Endopeptidase Clp/metabolism , Endopeptidase Clp/chemistry , Heat-Shock Proteins/chemistry , Heat-Shock Proteins/metabolism , Leptospira/metabolism , Leptospira/enzymology , Leptospira interrogans/enzymology , Leptospira interrogans/metabolism , Molecular Chaperones/metabolism , Molecular Chaperones/chemistry , Protein Binding , Protein Isoforms/metabolism , Protein Isoforms/chemistry , Proteolysis
17.
J Med Chem ; 67(8): 6769-6792, 2024 Apr 25.
Article in English | MEDLINE | ID: mdl-38620134

ABSTRACT

The activation of Homo sapiens Casein lysing protease P (HsClpP) by a chemical or genetic strategy has been proved to be a new potential therapy in acute myeloid leukemia (AML). However, limited efficacy has been achieved with classic agonist imipridone ONC201. Here, a novel class of HsClpP agonists is designed and synthesized using a ring-opening strategy based on the lead compound 1 reported in our previous study. Among these novel scaffold agonists, compound 7k exhibited remarkably enhanced proteolytic activity of HsClpP (EC50 = 0.79 ± 0.03 µM) and antitumor activity in vitro (IC50 = 0.038 ± 0.003 µM). Moreover, the intraperitoneal administration of compound 7k markedly suppressed tumor growth in Mv4-11 xenograft models, achieving a tumor growth inhibition rate of 88%. Concurrently, 7k displayed advantageous pharmacokinetic properties in vivo. This study underscores the promise of compound 7k as a significant HsClpP agonist and an antileukemia drug candidate, warranting further exploration for AML treatment.


Subject(s)
Antineoplastic Agents , Drug Design , Endopeptidase Clp , Leukemia, Myeloid, Acute , Humans , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/therapeutic use , Mice , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/pathology , Endopeptidase Clp/metabolism , Structure-Activity Relationship , Cell Line, Tumor , Xenograft Model Antitumor Assays , Cell Proliferation/drug effects , Mice, Nude , Mice, Inbred BALB C
18.
J Biol Chem ; 300(4): 107165, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38484801

ABSTRACT

ClpG is a novel autonomous disaggregase found in Pseudomonas aeruginosa that confers resistance to lethal heat stress. The mechanism by which ClpG specifically targets protein aggregates for disaggregation is unknown. In their recent work published in JBC, Katikaridis et al. (2023) identify an avidity-based mechanism by which four or more ClpG subunits, through specific N-terminal hydrophobic residues located on an exposed ß-sheet loop, interact with multiple hydrophobic patches on an aggregated protein substrate. This study establishes a model for substrate binding to a prokaryotic disaggregase that should inform further investigations into other autonomous disaggregases.


Subject(s)
Bacterial Proteins , Protein Binding , Pseudomonas aeruginosa , Pseudomonas aeruginosa/metabolism , Pseudomonas aeruginosa/enzymology , Bacterial Proteins/metabolism , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Protein Aggregates , Hydrophobic and Hydrophilic Interactions , Endopeptidase Clp/metabolism , Endopeptidase Clp/genetics , Heat-Shock Proteins/metabolism , Heat-Shock Proteins/chemistry
19.
mBio ; 15(4): e0003124, 2024 Apr 10.
Article in English | MEDLINE | ID: mdl-38501868

ABSTRACT

The Clp protease system is important for maintaining proteostasis in bacteria. It consists of ClpP serine proteases and an AAA+ Clp-ATPase such as ClpC1. The hexameric ATPase ClpC1 utilizes the energy of ATP binding and hydrolysis to engage, unfold, and translocate substrates into the proteolytic chamber of homo- or hetero-tetradecameric ClpP for degradation. The assembly between the hetero-tetradecameric ClpP1P2 chamber and the Clp-ATPases containing tandem ATPase domains from the same species has not been studied in depth. Here, we present cryo-EM structures of the substrate-bound ClpC1:shClpP1P2 from Streptomyces hawaiiensis, and shClpP1P2 in complex with ADEP1, a natural compound produced by S. hawaiiensis and known to cause over-activation and dysregulation of the ClpP proteolytic core chamber. Our structures provide detailed information on the shClpP1-shClpP2, shClpP2-ClpC1, and ADEP1-shClpP1/P2 interactions, reveal conformational transition of ClpC1 during the substrate translocation, and capture a rotational ATP hydrolysis mechanism likely dominated by the D1 ATPase activity of chaperones.IMPORTANCEThe Clp-dependent proteolysis plays an important role in bacterial homeostasis and pathogenesis. The ClpP protease system is an effective drug target for antibacterial therapy. Streptomyces hawaiiensis can produce a class of potent acyldepsipeptide antibiotics such as ADEP1, which could affect the ClpP protease activity. Although S. hawaiiensis hosts one of the most intricate ClpP systems in nature, very little was known about its Clp protease mechanism and the impact of ADEP molecules on ClpP. The significance of our research is in dissecting the functional mechanism of the assembled Clp degradation machinery, as well as the interaction between ADEP1 and the ClpP proteolytic chamber, by solving high-resolution structures of the substrate-bound Clp system in S. hawaiiensis. The findings shed light on our understanding of the Clp-dependent proteolysis in bacteria, which will enhance the development of antimicrobial drugs targeting the Clp protease system, and help fighting against bacterial multidrug resistance.


Subject(s)
Adenosine Triphosphatases , Endopeptidase Clp , Streptomyces , Endopeptidase Clp/genetics , Endopeptidase Clp/metabolism , Proteolysis , Adenosine Triphosphatases/metabolism , ATPases Associated with Diverse Cellular Activities/metabolism , Peptide Hydrolases/metabolism , Adenosine Triphosphate/metabolism
20.
ACS Synth Biol ; 13(2): 669-682, 2024 02 16.
Article in English | MEDLINE | ID: mdl-38317378

ABSTRACT

Protein degron tags have proven to be uniquely useful for the characterization of gene function. Degrons can mediate quick depletion, usually within minutes, of a protein of interest, allowing researchers to characterize cellular responses to the loss of function. To develop a general-purpose degron tool in Escherichia coli, we sought to build upon a previously characterized system of SspB-dependent inducible protein degradation. For this, we created a family of expression vectors containing a destabilized allele of SspB, capable of a rapid and nearly perfect "off-to-on" induction response. Using this system, we demonstrated excellent control over several DNA metabolism enzymes. However, other substrates did not respond to degron tagging in such an ideal manner, indicating the apparent limitations of SspB-dependent systems. Several degron-tagged proteins were degraded too slowly to be completely depleted during active growth, whereas others appeared to be completely refractory to degron-promoted degradation. Thus, only a minority of our, admittedly biased, selection of degron substrates proved to be amenable to efficient SspB-catalyzed degradation. We also uncovered an apparent stalling and/or disengagement of ClpXP from a degron-tagged allele of beta-galactosidase (beta-gal). While a degron-containing fusion peptide attached to the carboxy-terminus of beta-gal was degraded quantitatively, no reductions in beta-gal activity or concentration were detected, demonstrating an apparently novel mechanism of protease resistance. We conclude that substrate-dependent effects of the SspB system present a continued challenge to the widespread adoption of this degron system. For substrates that prove to be degradable, we provide a series of titratable SspB-expression vehicles.


Subject(s)
Escherichia coli Proteins , Escherichia coli , Escherichia coli/genetics , Escherichia coli/metabolism , Escherichia coli Proteins/metabolism , Carrier Proteins/genetics , Proteolysis , Degrons , Adenosine Triphosphatases/metabolism , Endopeptidase Clp/genetics , Endopeptidase Clp/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL