1
|
Yang Y, Li Z, Zhang J, Qi H. Engineering thermostable friend mouse leukemia virus reverse transcriptase through mutational combination. Biochem Biophys Res Commun 2025; 760:151716. [PMID: 40164014 DOI: 10.1016/j.bbrc.2025.151716] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Revised: 03/11/2025] [Accepted: 03/26/2025] [Indexed: 04/02/2025]
Abstract
Reverse transcriptase (RTs) is an essential tool in molecular biology and medical research; however, its typical lack of thermostability poses significant limitations. In this study, we engineered thermostable RTs derived from Friend mouse leukemia virus reverse transcriptase (FrMLV RT) through a mutational combination. The thermostable FrM5 variant (D178C/E280R/T284R/W291F/L581W) is obtained through iterative rounds of mutational combination and rapid cell-free RTs activity assays. The FrM5 variant exhibited robust RTs activity across a broad temperature range (35-50 °C) with the template-primer (T/P). Notably, the half-life of the FrM5 variant at 50 °C was approximately 20 min, in contrast to less than 2 min for the wild-type (FrWT) in the presence of T/P. Furthermore, the melting temperature difference between the FrWT and FrM5 variants was less than 2 °C, regardless of the presence or absence of T/P. Finally, we demonstrated that FrM5 exhibits tighter binding to T/P, which likely protects against heat inactivation. This advancement could substantially improve the efficiency and accuracy of molecular biology and medical research applications.
Collapse
Affiliation(s)
- Youhui Yang
- School of Chemical Engineering and Technology, Tianjin University, Tianjin, 300072, PR China; Key Laboratory of Systems Bioengineering (Ministry of Education), Tianjin University, Tianjin, 300072, PR China
| | - Zhong Li
- School of Chemical Engineering and Technology, Tianjin University, Tianjin, 300072, PR China; Key Laboratory of Systems Bioengineering (Ministry of Education), Tianjin University, Tianjin, 300072, PR China
| | - Jie Zhang
- School of Chemical Engineering and Technology, Tianjin University, Tianjin, 300072, PR China; Key Laboratory of Systems Bioengineering (Ministry of Education), Tianjin University, Tianjin, 300072, PR China
| | - Hao Qi
- School of Chemical Engineering and Technology, Tianjin University, Tianjin, 300072, PR China; Key Laboratory of Systems Bioengineering (Ministry of Education), Tianjin University, Tianjin, 300072, PR China.
| |
Collapse
|
2
|
Liu J, Liu X, Song Z, Cao W, Li Y, Xia M, Wang D. A novel α-amylase from Streptococcus thermophilus 17140 with β-glycosidic bond hydrolysis capability for the transformation of rare ginsenosides. Int J Biol Macromol 2025; 306:141621. [PMID: 40024403 DOI: 10.1016/j.ijbiomac.2025.141621] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Revised: 02/17/2025] [Accepted: 02/27/2025] [Indexed: 03/04/2025]
Abstract
Alpha-amylases typically act on starch and oligosaccharides that contain α-1,4 glycosidic linkages, while ginsenosides primarily consist of β-1,2 and β-1,6 glycosidic bonds, which cannot be hydrolyzed by α-amylases. However, for the first time, we have successfully isolated Streptococcus thermophilus 17140 (St17140), capable of converting ginsenoside Rb1 into rare ginsenosides. St17140 expresses a novel α-amylase (StAMY), which hydrolyzes ginsenoside Rb1 to produce Rd, gypenoside XVII, F2, and Rh2. With Rb1 as the substrate, the optimal reaction temperature is 50 °C and the optimal pH is 5.5. When Rb1, Rd, or gypenoside XVII are used as substrates, the Km are 0.135 mM, 0.0871 mM, and 0.260 mM respectively, the Kcat are 0.621 min-1, 0.397 min-1, and 0.297 min-1 respectively, and the Kcat/Km are 4.59 min-1·mM-1, 4.56 min-1·mM-1and 1.14 min-1·mM-1 respectively. Utilizing molecular docking and site-directed mutagenesis techniques, the mechanism of ginsenoside hydrolysis by StAMY was further elucidated. A107, F201, A337 and A268 are key amino acid residues that anchor the saponin in the active pocket, and D234 and D331 as catalytic amino acid residues to hydrolyze ginsenosides into rare ginsenosides. It is the first time to discover a novel α-amylase with the ability to hydrolyze ginsenosides β-1,2 and β-1,6 glycosidic linkage, and the first time to discover the ability of S. thermophilus to transform rare ginsenosides. These not only enrich the resources of glycosidases and offer novel insights for the rational modification of glycosidase, also are important for further elucidating the mechanism of probiotic transformation of rare ginsenosides and developing engineering bacteria with high production of rare ginsenosides.
Collapse
Affiliation(s)
- Jinxia Liu
- School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Benxi 117004, China
| | - Xinyi Liu
- School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Benxi 117004, China
| | - Zhaoqing Song
- School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Benxi 117004, China
| | - Wenying Cao
- School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Benxi 117004, China
| | - Yue Li
- School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Benxi 117004, China
| | - Mingyu Xia
- School of Life Science and Biological Pharmacy, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Dong Wang
- School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Benxi 117004, China.
| |
Collapse
|
3
|
Ma C, Zhao R, Li SW, Zhao J, Jia Z, Tang L, Song Y, Wang RJ, Yang J, Peng YL. Glutamate dehydrogenase MoGDH2 modulates the environmental and host pH to enhance adaptation and virulence of the rice blast fungus Pyricularia oryzae. Int J Biol Macromol 2025; 308:142465. [PMID: 40139586 DOI: 10.1016/j.ijbiomac.2025.142465] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Revised: 03/09/2025] [Accepted: 03/22/2025] [Indexed: 03/29/2025]
Abstract
pH adaptation and modulation are essential for the survival and infection of fungal pathogens. Pyricularia oryzae is a hemi-biotrophic fungal pathogen causes devastating blast disease on rice. How P. oryzae achieves host pH alkalization during the biotrophic-infection stage is unclear. Here, we characterized the NAD+-glutamate dehydrogenase encoding gene MoGDH2 in P. oryzae. The Δmogdh2 mutant failed to utilize glutamate to release NH3 and alkalize the environmental pH. MoGDH2 mediated pH homeostasis under acidic conditions but not alkaline environments. During glutamate utilization and fungal infection, MoGDH2 exhibited high expression levels, and modulated host pH at biotrophic stage. The apoplastic pH of host cells infected by wild-type strain P131 was sharply acidified at 24 h post inoculation (hpi), and the cytoplasmic pH gradually increased from 24 to 36 hpi. In comparison, the pH change patterns disappeared in cells infected by Δmogdh2. Furthermore, MoGDH2 is critical for reactive oxygen species tolerance and virulence, which is regulated via phosphorylation at the T47 site. Protein kinase MoDbf2 directly interacted with and phosphorylated MoGDH2. This study sheds new light on the function of MoGDH2 in pH modulation and infection.
Collapse
Affiliation(s)
- Chang Ma
- MARA Key Laboratory of Pest Monitoring and Green Management, Department of Plant Pathology, China Agricultural University, Beijing, China.
| | - Rui Zhao
- MARA Key Laboratory of Pest Monitoring and Green Management, Department of Plant Pathology, China Agricultural University, Beijing, China.
| | - Shi-Wang Li
- MARA Key Laboratory of Pest Monitoring and Green Management, Department of Plant Pathology, China Agricultural University, Beijing, China; MARA Key Laboratory of Surveillance and Management for Plant Quarantine Pests, Department of Plant Biosecurity, China Agricultural University, Beijing, China.
| | - Jianhui Zhao
- MARA Key Laboratory of Pest Monitoring and Green Management, Department of Plant Pathology, China Agricultural University, Beijing, China; MARA Key Laboratory of Surveillance and Management for Plant Quarantine Pests, Department of Plant Biosecurity, China Agricultural University, Beijing, China.
| | - Zhishuo Jia
- MARA Key Laboratory of Pest Monitoring and Green Management, Department of Plant Pathology, China Agricultural University, Beijing, China.
| | - Liu Tang
- MARA Key Laboratory of Pest Monitoring and Green Management, Department of Plant Pathology, China Agricultural University, Beijing, China.
| | - Yue Song
- MARA Key Laboratory of Pest Monitoring and Green Management, Department of Plant Pathology, China Agricultural University, Beijing, China.
| | - Rui-Jin Wang
- MARA Key Laboratory of Pest Monitoring and Green Management, Department of Plant Pathology, China Agricultural University, Beijing, China; MARA Key Laboratory of Surveillance and Management for Plant Quarantine Pests, Department of Plant Biosecurity, China Agricultural University, Beijing, China.
| | - Jun Yang
- MARA Key Laboratory of Pest Monitoring and Green Management, Department of Plant Pathology, China Agricultural University, Beijing, China; MARA Key Laboratory of Surveillance and Management for Plant Quarantine Pests, Department of Plant Biosecurity, China Agricultural University, Beijing, China.
| | - You-Liang Peng
- MARA Key Laboratory of Pest Monitoring and Green Management, Department of Plant Pathology, China Agricultural University, Beijing, China.
| |
Collapse
|
4
|
Trasviña-Arenas CH, Dissanayake UC, Tamayo N, Hashemian M, Lin WJ, Demir M, Hoyos-Gonzalez N, Fisher AJ, Cisneros GA, Horvath MP, David SS. Structure of human MUTYH and functional profiling of cancer-associated variants reveal an allosteric network between its [4Fe-4S] cluster cofactor and active site required for DNA repair. Nat Commun 2025; 16:3596. [PMID: 40234396 PMCID: PMC12000561 DOI: 10.1038/s41467-025-58361-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Accepted: 03/20/2025] [Indexed: 04/17/2025] Open
Abstract
MUTYH is a clinically important DNA glycosylase that thwarts mutations by initiating base-excision repair at 8-oxoguanine (OG):A lesions. The roles for its [4Fe-4S] cofactor in DNA repair remain enigmatic. Functional profiling of cancer-associated variants near the [4Fe-4S] cofactor reveals that most variations abrogate both retention of the cofactor and enzyme activity. Surprisingly, R241Q and N238S retained the metal cluster and bound substrate DNA tightly, but were completely inactive. We determine the crystal structure of human MUTYH bound to a transition state mimic and this shows that Arg241 and Asn238 build an H-bond network connecting the [4Fe-4S] cluster to the catalytic Asp236 that mediates base excision. The structure of the bacterial MutY variant R149Q, along with molecular dynamics simulations of the human enzyme, support a model in which the cofactor functions to position and activate the catalytic Asp. These results suggest that allosteric cross-talk between the DNA binding [4Fe-4S] cofactor and the base excision site of MUTYH regulate its DNA repair function.
Collapse
Affiliation(s)
- Carlos H Trasviña-Arenas
- Department of Chemistry, University of California, Davis, CA, USA
- Research Center on Aging, Center for Research and Advanced Studies (CINVESTAV), Mexico City, Mexico
| | - Upeksha C Dissanayake
- Department of Chemistry and Biochemistry, University of Texas at Dallas, Richardson, TX, USA
| | - Nikole Tamayo
- Department of Chemistry, University of California, Davis, CA, USA
- Chemistry and Chemical Biology Graduate Program, University of California, Davis, CA, USA
| | - Mohammad Hashemian
- Department of Chemistry, University of California, Davis, CA, USA
- Chemistry and Chemical Biology Graduate Program, University of California, Davis, CA, USA
| | - Wen-Jen Lin
- Department of Chemistry, University of California, Davis, CA, USA
- Chemistry and Chemical Biology Graduate Program, University of California, Davis, CA, USA
| | - Merve Demir
- Department of Chemistry, University of California, Davis, CA, USA
- Chemistry and Chemical Biology Graduate Program, University of California, Davis, CA, USA
| | | | - Andrew J Fisher
- Department of Chemistry, University of California, Davis, CA, USA
- Chemistry and Chemical Biology Graduate Program, University of California, Davis, CA, USA
- Department of Molecular and Cellular Biology, University of California, Davis, CA, USA
| | - G Andrés Cisneros
- Department of Chemistry and Biochemistry, University of Texas at Dallas, Richardson, TX, USA.
- Department of Physics, University of Texas at Dallas, Richardson, TX, USA.
| | - Martin P Horvath
- School of Biological Sciences, University of Utah, Salt Lake City, UT, USA.
| | - Sheila S David
- Department of Chemistry, University of California, Davis, CA, USA.
- Chemistry and Chemical Biology Graduate Program, University of California, Davis, CA, USA.
| |
Collapse
|
5
|
Xiao X, Yang X, Xu K, Huang F, Zhang Y, Jiang Y, Shi Y, Zhou Q, Wang L, Lu J, Gao Z, Lou Y. DNA Fragment Fusion and Nucleic Acid Detection by Fusion Recombinase-Aided Amplification. Anal Chem 2025; 97:6538-6547. [PMID: 40106763 DOI: 10.1021/acs.analchem.4c05991] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/22/2025]
Abstract
Constructing fusion DNA fragments is frequently used for genetic engineering purposes. To date, fusion PCR is one of the most popular approaches for generating fusion DNA fragments. Here, we describe a novel method for DNA fusion based on the isothermal DNA amplification technique, recombinase-aided amplification (RAA). We demonstrate that this method, termed "fusion RAA", can assemble two to three DNA fragments to generate a fusion fragment of up to ∼1 kb in a one-pot reaction within 40 min at 37 °C. We further demonstrate that fusion RAA can realize fragment insertion, deletion, and base mutation. Moreover, we show that fusion RAA can be harnessed to facilitate pathogen detection by simultaneously targeting two genes in one RAA assay, as demonstrated by the rapid and simplified detection of methicillin-resistant Staphylococcus aureus (MRSA). Based on fusion RAA, we establish two novel pathogen detection platforms, FREAC (Fusion REcombinase-aided Amplification combined with CRISPR/Cas13a) and FREAL (Fusion REcombinase-aided Amplification combined with Lateral flow assay). Using these two platforms, we can detect clinical MRSA strains within 55 min with high specificity and a limit of detection of 150 copies/μL of genomic DNA, highlighting their potential as user-friendly platforms for nucleic acid detection.
Collapse
Affiliation(s)
- Xingxing Xiao
- Wenzhou Key Laboratory of Sanitary Microbiology, Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou 325035, China
| | - Xi Yang
- Wenzhou Key Laboratory of Sanitary Microbiology, Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou 325035, China
| | - Kexin Xu
- Wenzhou Key Laboratory of Sanitary Microbiology, Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou 325035, China
| | - Fuyuan Huang
- Wenzhou Key Laboratory of Sanitary Microbiology, Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou 325035, China
| | - Yan Zhang
- Wenzhou Key Laboratory of Sanitary Microbiology, Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou 325035, China
| | - Yelin Jiang
- Wenzhou Key Laboratory of Sanitary Microbiology, Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou 325035, China
| | - Yangbin Shi
- Wenzhou Key Laboratory of Sanitary Microbiology, Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou 325035, China
| | - Qinghong Zhou
- Wenzhou Key Laboratory of Sanitary Microbiology, Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou 325035, China
| | - Luying Wang
- Wenzhou Key Laboratory of Sanitary Microbiology, Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou 325035, China
| | - Jiahai Lu
- Wenzhou Key Laboratory of Sanitary Microbiology, Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou 325035, China
| | - Zongliang Gao
- GMU-GIBH Joint School of Life Sciences, The Guangdong-Hong Kong-Macao Joint Laboratory for Cell Fate Regulation and Diseases, Guangzhou Laboratory, Guangzhou Medical University, Guangzhou 511436, China
| | - Yongliang Lou
- Wenzhou Key Laboratory of Sanitary Microbiology, Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou 325035, China
| |
Collapse
|
6
|
Duan K, Qin S, Cui F, Zhao L, Huang Y, Xu JR, Wang G. MeJA inhibits fungal growth and DON toxin production by interfering with the cAMP-PKA signaling pathway in the wheat scab fungus Fusarium graminearum. mBio 2025; 16:e0315124. [PMID: 39902906 PMCID: PMC11898702 DOI: 10.1128/mbio.03151-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Accepted: 01/03/2025] [Indexed: 02/06/2025] Open
Abstract
Deoxynivalenol (DON), a mycotoxin primarily produced by Fusarium species, is commonly found in cereal grains and poses risks to human and animal health, as well as global grain trade. This study demonstrates that methyl jasmonate (MeJA), a natural plant hormone, inhibits the growth and conidiation of Fusarium graminearum. Importantly, MeJA significantly reduces DON production by suppressing TRI gene expression and toxisome formation. To explore the molecular mechanism, we identified MeJA-tolerant mutants, including a transcription factor MRT1 and cAMP-PKA pathway-related genes (FgGPA1 and FgSNT1). MeJA treatment reduced PKA activity and intracellular cAMP levels in F. graminearum, suggesting it targets the cAMP-PKA pathway. Notably, the MeJA-resistant mutant FgGPA1R178H enhanced fungal growth, DON production, and cAMP levels in the presence of MeJA. Exogenous cAMP alleviated MeJA's inhibitory effects on DON production, further supporting this pathway's involvement. Interestingly, MeJA had no effect on all three MAP kinase pathways (Mgv1, Gpmk1, and FgHog1). Truncated and phospho-mimicking mutations in Mrt1 or FgSnt1 conferred MeJA resistance, suggesting they may act downstream of the cAMP-PKA pathway. In conclusion, MeJA presents a promising approach to control F. graminearum growth and DON production.IMPORTANCEDeoxynivalenol (DON) poses significant risks to both human and animal health and severely disrupts the global grain trade due to its prevalence as a common contaminant in wheat grains. With rising public concern over food safety, finding effective and sustainable methods to reduce DON contamination becomes increasingly urgent. In our study, we found that methyl jasmonate (MeJA), a natural plant hormone, can effectively inhibit the vegetative growth of F. graminearum and significantly reduce its DON toxin production. To explore the underlying molecular mechanism, we identified the mutations in MeJA-tolerant mutants and revealed that MeJA effectively exerts its antifungal activities by inhibiting the cAMP-PKA signaling pathway in F. graminearum. Our work provides a promising natural solution to reduce DON toxin contamination in cereal grains, enhancing food safety while decreasing the reliance on chemical fungicides and their associated environmental impact.
Collapse
Affiliation(s)
- Kaili Duan
- State Key Laboratory for Crop Stress Resistance and High-Efficiency Production, College of Plant Protection, Northwest A&F University, Yangling, Shaanxi, China
| | - Shaozhe Qin
- State Key Laboratory for Crop Stress Resistance and High-Efficiency Production, College of Plant Protection, Northwest A&F University, Yangling, Shaanxi, China
| | - Fangling Cui
- State Key Laboratory for Crop Stress Resistance and High-Efficiency Production, College of Plant Protection, Northwest A&F University, Yangling, Shaanxi, China
| | - Liangyuan Zhao
- State Key Laboratory for Crop Stress Resistance and High-Efficiency Production, College of Plant Protection, Northwest A&F University, Yangling, Shaanxi, China
| | - Yongqing Huang
- State Key Laboratory for Crop Stress Resistance and High-Efficiency Production, College of Plant Protection, Northwest A&F University, Yangling, Shaanxi, China
| | - Jin-Rong Xu
- Department of Botany and Plant Pathology, Purdue University, West Lafayette, Indiana, USA
| | - Guanghui Wang
- State Key Laboratory for Crop Stress Resistance and High-Efficiency Production, College of Plant Protection, Northwest A&F University, Yangling, Shaanxi, China
| |
Collapse
|
7
|
Hsu TW, Wang WY, Chen HA, Wang TH, Su CM, Liao PH, Chen A, Tsai KY, Kokotos G, Kuo CC, Chiu CF, Su YH. FOXO3a/miR-4259-driven LDHA expression as a key mechanism of gemcitabine sensitivity in pancreatic ductal adenocarcinoma. Cancer Metab 2025; 13:7. [PMID: 39930542 PMCID: PMC11809001 DOI: 10.1186/s40170-025-00377-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Accepted: 01/31/2025] [Indexed: 02/13/2025] Open
Abstract
BACKGROUND Lactate dehydrogenase A (LDHA) can regulate tumorigenesis and cancer progression. Nevertheless, whether the regulation of LDHA is involved in the development of gemcitabine resistance in PDAC has not yet been fully elucidated. Increasing studies have shown that cancer acquired drug resistance led to treatment failure is highly attributed to the cancer stem cell (CSC) properties. Therefore, we aim to demonstrate the functions and regulatory mechanisms of LDHA on cancer stem cell (CSC) properties and gemcitabine resistance in PDAC. METHODS We investigate the metabolite profiles by liquid chromatography-mass spectrometry between gemcitabine-resistant PDAC and parental PDAC cells. Additionally, gain-of-function and loss-of-function experiments were conducted to examine the roles of LDHA on CSC properties and gemcitabine resistance in the gemcitabine-resistant PDAC and parental PDAC cells. To investigate regulators involved in LDHA-mediated gemcitabine resistance and CSC of pancreatic cancer cells, we further used a combination of the miRNA microarray results and software predictions and confirmed that miR-4259 is a direct target of LDHA by luciferase assay. Furthermore, we constructed serial miR-4259 promoter reporters and searched for response elements using the TESS 2.0/TFSEARCH software to find the transcription factor binding site in the promoter region of miR-4259. RESULTS We observed that elevated LDHA expression significantly correlates with recurrent pancreatic cancer patients following gemcitabine treatment and with CSC properties. We further identify that FOXO3a-induced miR-4259 directly targets the 3'untranslated region of LDHA and reduced LDHA expression, leading to decreased gemcitabine resistance and a reduction in the CSC phenotypes of pancreatic cancer. CONCLUSION Our results demonstrated that LDHA plays a critical role in cancer stemness and gemcitabine resistance of pancreatic cancer, and indicate that targeting the FOXO3a/miR-4259/LDHA pathway might serve as a new treatment for pancreatic cancer patients with a poor response to gemcitabine chemotherapy.
Collapse
Affiliation(s)
- Tung-Wei Hsu
- Department of Surgery, Division of General Surgery, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan
| | - Wan-Yu Wang
- Department of Surgery, Division of General Surgery, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan
| | - Hsin-An Chen
- Department of Surgery, Division of General Surgery, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan
- Department of Surgery, Division of General Surgery, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- TMU Research Center of Cancer Translational Medicine, Taipei Medical University, Taipei, Taiwan
- TMU Research Center for Digestive Medicine, Taipei Medical University, Taipei, Taiwan
- Department of Surgery, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Tzu-Hsuan Wang
- Department of Surgery, Division of General Surgery, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan
| | - Chih-Ming Su
- Department of Surgery, Division of General Surgery, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan
- Department of Surgery, Division of General Surgery, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Po-Hsiang Liao
- Cardiovascular and Mitochondrial Related Disease Research Center, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan
| | - Alvin Chen
- Department of Surgery, Division of General Surgery, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan
| | - Kuei-Yen Tsai
- Department of Surgery, Division of General Surgery, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan
- Department of Surgery, Division of General Surgery, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - George Kokotos
- Department of Chemistry, National and Kapodistrian University of Athens, Panepistimiopolis, Athens, Greece
| | - Cheng-Chin Kuo
- Institute of Cellular and System Medicine, National Health Research Institutes, Zhunan, Taiwan
- Department of Bioscience Technology, Chung Yuan Christian University, Taoyuan, Taiwan
| | - Ching-Feng Chiu
- Department of Surgery, Division of General Surgery, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan
| | - Yen-Hao Su
- Department of Surgery, Division of General Surgery, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan.
- Department of Surgery, Division of General Surgery, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.
- TMU Research Center of Cancer Translational Medicine, Taipei Medical University, Taipei, Taiwan.
- TMU Research Center for Digestive Medicine, Taipei Medical University, Taipei, Taiwan.
- Department of Surgery, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.
- Metabolic and Weight Management Center, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan.
| |
Collapse
|
8
|
Bezkorovayna V, Hayes BK, Gillett FN, Wright A, Roper DI, Harper M, McGowan S, Boyce JD. Delivery determinants of an Acinetobacter baumannii type VI secretion system bifunctional peptidoglycan hydrolase. mBio 2025; 16:e0262724. [PMID: 39745415 PMCID: PMC11796386 DOI: 10.1128/mbio.02627-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Accepted: 12/09/2024] [Indexed: 02/06/2025] Open
Abstract
Acinetobacter baumannii is a Gram-negative opportunistic pathogen and is a common cause of nosocomial infections. The increasing development of antibiotic resistance in this organism is a global health concern. The A. baumannii clinical isolate AB307-0294 produces a type VI secretion system (T6SS) that delivers three antibacterial effector proteins that give this strain a competitive advantage against other bacteria in polymicrobial environments. Each effector, Tse15, Tde16, and Tae17, is delivered via a non-covalent interaction with a specific T6SS VgrG protein (VgrG15, VgrG16, and VgrG17, respectively). Here we define the regions of interaction between Tae17 and its cognate delivery protein VgrG17 and identify that amino acids G1069 and W1075 in VgrG17 are essential for Tae17 delivery via the T6SS, the first time such specific delivery determinants of T6SS cargo effectors have been defined. Furthermore, we determine that the Tae17 effector is a multidomain, bifunctional, peptidoglycan-degrading enzyme that has both amidase activity, which targets the sugar-peptide bonds, and lytic transglycosylase activity, which targets the peptidoglycan sugar backbone. Moreover, we show that the Tae17 transglycosylase activity is more important than amidase activity for the killing of Escherichia coli. This study provides molecular insight into how the T6SS allows A. baumannii strains to gain dominance in polymicrobial communities and thus improve their chances of survival and transmission.IMPORTANCEWe have shown that the Acinetobacter baumannii T6SS effector Tae17 is a modular, bifunctional, peptidoglycan-degrading enzyme that has both lytic transglycosylase and amidase activities. Both activities contribute to the ability to degrade peptidoglycan, but the transglycosylase activity was more important for the killing of Escherichia coli. We have defined the specific regions of Tae17 and its cognate delivery protein VgrG17 that are necessary for the non-covalent interactions and, for the first time, identified specific amino acids essential for T6SS cargo effector delivery. This work contributes to our molecular understanding of bacterial competition strategies in polymicrobial environments and may provide a window to design new therapeutic approaches for combating infection by A. baumannii.
Collapse
Affiliation(s)
- Valeriya Bezkorovayna
- Infection Program, Department of Microbiology, Monash University, Biomedicine Discovery Institute, Melbourne, Victoria, Australia
| | - Brooke K. Hayes
- Infection Program, Department of Microbiology, Monash University, Biomedicine Discovery Institute, Melbourne, Victoria, Australia
- Centre to Impact AMR, Monash University, Melbourne, Australia
| | | | - Amy Wright
- Infection Program, Department of Microbiology, Monash University, Biomedicine Discovery Institute, Melbourne, Victoria, Australia
- Centre to Impact AMR, Monash University, Melbourne, Australia
| | - David I. Roper
- School of Life Sciences, University of Warwick, Coventry, United Kingdom
| | - Marina Harper
- Infection Program, Department of Microbiology, Monash University, Biomedicine Discovery Institute, Melbourne, Victoria, Australia
- Centre to Impact AMR, Monash University, Melbourne, Australia
| | - Sheena McGowan
- Infection Program, Department of Microbiology, Monash University, Biomedicine Discovery Institute, Melbourne, Victoria, Australia
- Centre to Impact AMR, Monash University, Melbourne, Australia
| | - John D. Boyce
- Infection Program, Department of Microbiology, Monash University, Biomedicine Discovery Institute, Melbourne, Victoria, Australia
- Centre to Impact AMR, Monash University, Melbourne, Australia
| |
Collapse
|
9
|
Teng Y, Luo C, Qiu X, Mu J, Sriwastva MK, Xu Q, Liu M, Hu X, Xu F, Zhang L, Park JW, Hwang JY, Kong M, Liu Z, Zhang X, Xu R, Yan J, Merchant ML, McClain CJ, Zhang HG. Plant-nanoparticles enhance anti-PD-L1 efficacy by shaping human commensal microbiota metabolites. Nat Commun 2025; 16:1295. [PMID: 39900923 PMCID: PMC11790884 DOI: 10.1038/s41467-025-56498-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Accepted: 01/21/2025] [Indexed: 02/05/2025] Open
Abstract
Diet has emerged as a key impact factor for gut microbiota function. However, the complexity of dietary components makes it difficult to predict specific outcomes. Here we investigate the impact of plant-derived nanoparticles (PNP) on gut microbiota and metabolites in context of cancer immunotherapy with the humanized gnotobiotic mouse model. Specifically, we show that ginger-derived exosome-like nanoparticle (GELN) preferentially taken up by Lachnospiraceae and Lactobacillaceae mediated by digalactosyldiacylglycerol (DGDG) and glycine, respectively. We further demonstrate that GELN aly-miR159a-3p enhances anti-PD-L1 therapy in melanoma by inhibiting the expression of recipient bacterial phospholipase C (PLC) and increases the accumulation of docosahexaenoic acid (DHA). An increased level of circulating DHA inhibits PD-L1 expression in tumor cells by binding the PD-L1 promoter and subsequently prevents c-myc-initiated transcription of PD-L1. Colonization of germ-free male mice with gut bacteria from anti-PD-L1 non-responding patients supplemented with DHA enhances the efficacy of anti-PD-L1 therapy compared to controls. Our findings reveal a previously unknown mechanistic impact of PNP on human tumor immunotherapy by modulating gut bacterial metabolic pathways.
Collapse
Affiliation(s)
- Yun Teng
- Brown Cancer Center, University of Louisville School of Medicine, Louisville, USA.
| | - Chao Luo
- Brown Cancer Center, University of Louisville School of Medicine, Louisville, USA
- Department of Central Laboratory, The affiliated Huai'an First People's Hospital of Nanjing Medical University, Huai'an, Jiangsu, China
| | - Xiaolan Qiu
- Brown Cancer Center, University of Louisville School of Medicine, Louisville, USA
- Department of Breast and Thyroid Surgery, The affiliated Huai'an First People's Hospital of Nanjing Medical University, Huai'an, Jiangsu, China
| | - Jingyao Mu
- Brown Cancer Center, University of Louisville School of Medicine, Louisville, USA
| | - Mukesh K Sriwastva
- Brown Cancer Center, University of Louisville School of Medicine, Louisville, USA
| | - Qingbo Xu
- Department of Microbiology and Immunology, University of Louisville, Louisville, KY, USA
| | - Minmin Liu
- Brown Cancer Center, University of Louisville School of Medicine, Louisville, USA
- Department of Breast and Thyroid Surgery, The affiliated Huai'an First People's Hospital of Nanjing Medical University, Huai'an, Jiangsu, China
| | - Xin Hu
- Department of Genomic Medicine, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Fangyi Xu
- Brown Cancer Center, University of Louisville School of Medicine, Louisville, USA
| | - Lifeng Zhang
- Brown Cancer Center, University of Louisville School of Medicine, Louisville, USA
| | - Juw Won Park
- Brown Cancer Center, University of Louisville School of Medicine, Louisville, USA
- Department of Bioinformatics and Biostatistics, SPHIS, University of Louisville, Louisville, KY, USA
| | - Jae Yeon Hwang
- Brown Cancer Center, University of Louisville School of Medicine, Louisville, USA
| | - Maiying Kong
- Brown Cancer Center, University of Louisville School of Medicine, Louisville, USA
- Department of Bioinformatics and Biostatistics, SPHIS, University of Louisville, Louisville, KY, USA
| | - Zhanxu Liu
- Brown Cancer Center, University of Louisville School of Medicine, Louisville, USA
| | - Xiang Zhang
- Department of Pharmacology and Toxicology, University of Louisville, Louisville, KY, USA
| | - Raobo Xu
- Department of Pharmacology and Toxicology, University of Louisville, Louisville, KY, USA
| | - Jun Yan
- Brown Cancer Center, University of Louisville School of Medicine, Louisville, USA
| | - Michael L Merchant
- Kidney Disease Program and Clinical Proteomics Center, University of Louisville, Louisville, KY, USA
| | - Craig J McClain
- Department of Medicine, Division of Gastroenterology, Hepatology and Nutrition, University of Louisville School of Medicine, Louisville, KY, USA
| | - Huang-Ge Zhang
- Brown Cancer Center, University of Louisville School of Medicine, Louisville, USA.
- Department of Microbiology and Immunology, University of Louisville, Louisville, KY, USA.
- Robley Rex Veterans Affairs Medical Center, Louisville, KY, USA.
| |
Collapse
|
10
|
Lavergne M, Schaerer R, De Grandis S, Bouheraoua S, Adenuga O, Muralt T, Schaerer T, Chèvre L, Failla A, Matthey P, Stumpe M, Kressler D, Mantel PY, Walch M. Executioner caspases degrade essential mediators of pathogen-host interactions to inhibit growth of intracellular Listeria monocytogenes. Cell Death Dis 2025; 16:55. [PMID: 39885151 PMCID: PMC11782612 DOI: 10.1038/s41419-025-07365-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Revised: 12/20/2024] [Accepted: 01/16/2025] [Indexed: 02/01/2025]
Abstract
Cell death mediated by executioner caspases is essential during organ development and for organismal homeostasis. The mechanistic role of activated executioner caspases in antibacterial defense during infections with intracellular bacteria, such as Listeria monocytogenes, remains elusive. Cell death upon intracellular bacterial infections is considered altruistic to deprive the pathogens of their protective niche. To establish infections in a human host, Listeria monocytogenes deploy virulence mediators, including membranolytic listeriolysin O (LLO) and the invasion associated protein p60 (Iap), allowing phagosomal escape, intracellular replication and cell-to-cell spread. Here, by means of chemical and genetical modifications, we show that the executioner caspases-3 and -7 efficiently inhibit growth of intracellular Listeria monocytogenes in host cells. Comprehensive proteomics revealed multiple caspase-3 substrates in the Listeria secretome, including LLO, Iap and various other proteins crucially involved in pathogen-host interactions. Listeria secreting caspase-uncleavable LLO or Iap gained significant growth advantage in epithelial cells. With that, we uncovered an underappreciated defense barrier and a non-canonical role of executioner caspases to degrade virulence mediators, thus impairing intracellular Listeria growth.
Collapse
Affiliation(s)
- Marilyne Lavergne
- Faculty of Science and Medicine, Department of Oncology, Microbiology and Immunology, Anatomy unit, University of Fribourg, CH-1700, Fribourg, Switzerland
| | - Raffael Schaerer
- Faculty of Science and Medicine, Department of Oncology, Microbiology and Immunology, Anatomy unit, University of Fribourg, CH-1700, Fribourg, Switzerland
| | - Sara De Grandis
- Faculty of Science and Medicine, Department of Oncology, Microbiology and Immunology, Anatomy unit, University of Fribourg, CH-1700, Fribourg, Switzerland
| | - Safaa Bouheraoua
- Faculty of Science and Medicine, Department of Oncology, Microbiology and Immunology, Anatomy unit, University of Fribourg, CH-1700, Fribourg, Switzerland
| | - Oluwadamilola Adenuga
- Faculty of Science and Medicine, Department of Oncology, Microbiology and Immunology, Anatomy unit, University of Fribourg, CH-1700, Fribourg, Switzerland
| | - Tanja Muralt
- Faculty of Science and Medicine, Department of Oncology, Microbiology and Immunology, Anatomy unit, University of Fribourg, CH-1700, Fribourg, Switzerland
| | - Tiffany Schaerer
- Faculty of Science and Medicine, Department of Oncology, Microbiology and Immunology, Anatomy unit, University of Fribourg, CH-1700, Fribourg, Switzerland
| | - Léa Chèvre
- Faculty of Science and Medicine, Department of Oncology, Microbiology and Immunology, Anatomy unit, University of Fribourg, CH-1700, Fribourg, Switzerland
| | - Alessandro Failla
- Faculty of Science and Medicine, Department of Oncology, Microbiology and Immunology, Anatomy unit, University of Fribourg, CH-1700, Fribourg, Switzerland
| | - Patricia Matthey
- Faculty of Science and Medicine, Department of Oncology, Microbiology and Immunology, Anatomy unit, University of Fribourg, CH-1700, Fribourg, Switzerland
| | - Michael Stumpe
- Faculty of Science and Medicine, Department of Biology, Metabolomics and Proteomics Platform, University of Fribourg, CH-1700, Fribourg, Switzerland
| | - Dieter Kressler
- Faculty of Science and Medicine, Department of Biology, Metabolomics and Proteomics Platform, University of Fribourg, CH-1700, Fribourg, Switzerland
| | - Pierre-Yves Mantel
- Faculty of Science and Medicine, Department of Oncology, Microbiology and Immunology, Anatomy unit, University of Fribourg, CH-1700, Fribourg, Switzerland
- Christine Kühne - Center for Allergy Research and Education (CK-CARE), CH-7265, Davos Wolfgang, Switzerland
| | - Michael Walch
- Faculty of Science and Medicine, Department of Oncology, Microbiology and Immunology, Anatomy unit, University of Fribourg, CH-1700, Fribourg, Switzerland.
| |
Collapse
|
11
|
Wu N, Jiang W, Xiang Z, Asghar R, Akkaya MS. Assessment of Self-Activation and Inhibition of Wheat Coiled-Coil Domain Containing NLR Immune Receptor Yr10 CG. PLANTS (BASEL, SWITZERLAND) 2025; 14:278. [PMID: 39861631 PMCID: PMC11768854 DOI: 10.3390/plants14020278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Revised: 12/27/2024] [Accepted: 01/16/2025] [Indexed: 01/27/2025]
Abstract
Plant immunity is largely governed by nucleotide-binding leucine-rich repeat receptor (NLR). Here, we examine the molecular activation and inhibition mechanisms of the wheat CC-type NLR Yr10CG, a previously proposed candidate for the Yr10 resistance gene. Though recent studies have identified YrNAM as the true Yr10 gene, Yr10CG remains an important NLR in understanding NLR-mediated immunity in wheat. In this study, we found that the overexpression of either the full-length Yr10CG or its CC domain in Nicotiana benthamiana did not trigger cell death, suggesting a robust autoinhibitory mechanism within Yr10CG. However, we observed that mutations in the conserved MHD motif, specifically D502G, activated Yr10CG and induced cell death. Structural modeling indicated that this mutation disrupted key interactions within the MHD motif, promoting local flexibility and activation. We further explored the effector recognition potential of Yr10CG by creating chimeric proteins with Sr50 domains, revealing that both the NB-ARC and LRR domains are necessary for effector recognition, while the CC domain likely functions in downstream immune signaling. Additionally, disrupting membrane localization through an L11E mutation abolished Yr10CG self-activation, suggesting a requirement for membrane association in immune activation. Our findings contribute to the understanding of CC-NLR activation and autoinhibition mechanisms, highlighting the potential of Yr10CG in NLR engineering for crop resistance improvement.
Collapse
Affiliation(s)
- Nan Wu
- School of Bioengineering, Dalian University of Technology, No. 2 Linggong Road, Dalian 116024, China; (N.W.); (Z.X.); (R.A.)
| | - Wanqing Jiang
- National Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, Hubei Key Laboratory of Plant Pathology, College of Plant Science and Technology, Huazhong Agricultural University, Wuhan 430070, China;
| | - Zhaoxia Xiang
- School of Bioengineering, Dalian University of Technology, No. 2 Linggong Road, Dalian 116024, China; (N.W.); (Z.X.); (R.A.)
| | - Raheel Asghar
- School of Bioengineering, Dalian University of Technology, No. 2 Linggong Road, Dalian 116024, China; (N.W.); (Z.X.); (R.A.)
| | - Mahinur S. Akkaya
- School of Bioengineering, Dalian University of Technology, No. 2 Linggong Road, Dalian 116024, China; (N.W.); (Z.X.); (R.A.)
| |
Collapse
|
12
|
Trasviña-Arenas C, Hashemian M, Malek M, Merrill S, Fisher AJ, David SS. Crystal structure of MutYX: A novel clusterless adenine DNA glycosylase with a distinct C-terminal domain and 8-Oxoguanine recognition sphere. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.03.631205. [PMID: 39803464 PMCID: PMC11722440 DOI: 10.1101/2025.01.03.631205] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/23/2025]
Abstract
The [4Fe-4S] cluster is an important cofactor of the base excision repair (BER) adenine DNA glycosylase MutY to prevent mutations associated with 8-oxoguanine (OG). Several MutYs lacking the [4Fe-4S] cofactor have been identified. Phylogenetic analysis shows that clusterless MutYs are distributed in two clades suggesting cofactor loss in two independent evolutionary events. Herein, we determined the first crystal structure of a clusterless MutY complexed with DNA. On the basis of the dramatic structural divergence from canonical MutYs, we refer to this as representative of a clusterless MutY subgroup "MutYX". Interestingly, MutYX compensates for the missing [4Fe-4S] cofactor to maintain positioning of catalytic residues by expanding a pre-existing α-helix and acquisition of the new α-helix. Surprisingly, MutYX also acquired a new C-terminal domain that uniquely recognizes OG using residue Gln201 and Arg209. Adenine glycosylase assays and binding affinity measurements indicate that Arg209 is the primary residue responsible to specificity for OG:A lesions, while Gln201 bridges OG and Arg209. Surprisingly, replacement of Arg209 and Gln201 with Ala increases activity toward G:A mismatches. The MutYX structure serves as an example of devolution, capturing structural features required to retain function in the absence of a metal cofactor considered indispensable.
Collapse
Affiliation(s)
- C.H. Trasviña-Arenas
- Department of Chemistry & Graduate Program in Chemistry and Chemical Biology, University of California, Davis, California, 95616, United States
- Current address; Centro de Investigación Sobre el Envejecimiento, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CIE-Cinvestav), Mexico City, 14330, Mexico
| | - Mohammad Hashemian
- Department of Chemistry & Graduate Program in Chemistry and Chemical Biology, University of California, Davis, California, 95616, United States
| | - Melody Malek
- Department of Chemistry & Graduate Program in Chemistry and Chemical Biology, University of California, Davis, California, 95616, United States
| | - Steven Merrill
- Department of Chemistry & Graduate Program in Chemistry and Chemical Biology, University of California, Davis, California, 95616, United States
| | - Andrew J. Fisher
- Department of Chemistry & Graduate Program in Chemistry and Chemical Biology, University of California, Davis, California, 95616, United States
- Department of Molecular and Cellular Biology, University of California, Davis, California 95616, United States
| | - Sheila S. David
- Department of Chemistry & Graduate Program in Chemistry and Chemical Biology, University of California, Davis, California, 95616, United States
| |
Collapse
|
13
|
Tanabe T, Miyamoto K, Nagaoka K, Tsujibo H, Funahashi T. Effect of (p)ppGpp on the Expression of the Vibrioferrin-Mediated Iron Acquisition System in Vibrio parahaemolyticus. Biol Pharm Bull 2025; 48:188-194. [PMID: 40024720 DOI: 10.1248/bpb.b24-00624] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/04/2025]
Abstract
Bacteria have a stringent response system mediated by guanosine pentaphosphate and tetraphosphate ((p)ppGpp), which suppresses the expression of genes involved in cell growth and promotes the expression of genes involved in nutrient uptake and metabolism under nutrient-limited stress. In environments with limited availability of iron, an essential trace element, bacteria generally produce and secrete siderophores to efficiently utilize water-insoluble ferric iron (Fe3+) in the environment. In Vibrio parahaemolyticus, Fur (iron-responsive repressor) and RyhB (Fur-regulated small RNA) regulate the expression of genes involved in the utilization of vibrioferrin (VF), a siderophore produced by this bacterium. In this study, we examined whether (p)ppGpp is also involved in regulating the expression of genes related to the VF utilization system. Results of the chrome azurol S plate assay revealed that the strain in which 3 (p)ppGpp synthetases were deleted (∆relA∆spoT∆relV) produced less VF than the parental strain. Growth test results showed that the growth rate of ∆relA∆spoT∆relV in an iron-limited medium was suppressed compared with that of the parental strain but was restored with the addition of VF. Furthermore, RT-quantitative (q)PCR results showed that the expression levels of pvsA (VF biosynthesis gene) and pvuA2 (ferric VF receptor gene) in ∆relA∆spoT∆relV under iron limitation were significantly reduced compared with those in the parental strain. Western blot results demonstrated that the expression level of PvuA2 in ∆relA∆spoT∆relV was lower than that in the parental strain. These results suggest that (p)ppGpp promotes the expression of genes related to VF biosynthesis and the ferric VF uptake system under iron limitation.
Collapse
Affiliation(s)
- Tomotaka Tanabe
- Laboratory of Hygienic Chemistry, College of Pharmaceutical Sciences, Matsuyama University, 4-2 Bunkyo-cho, Matsuyama, Ehime 790-8578, Japan
| | - Katsushiro Miyamoto
- Department of Microbiology and Infection Control, Faculty of Pharmacy, Osaka Medical and Pharmaceutical University, 4-20-1 Nasahara, Takatsuki, Osaka 569-1094, Japan
| | - Kenjiro Nagaoka
- Laboratory of Hygienic Chemistry, College of Pharmaceutical Sciences, Matsuyama University, 4-2 Bunkyo-cho, Matsuyama, Ehime 790-8578, Japan
| | - Hiroshi Tsujibo
- Department of Microbiology and Infection Control, Faculty of Pharmacy, Osaka Medical and Pharmaceutical University, 4-20-1 Nasahara, Takatsuki, Osaka 569-1094, Japan
| | - Tatsuya Funahashi
- Laboratory of Hygienic Chemistry, College of Pharmaceutical Sciences, Matsuyama University, 4-2 Bunkyo-cho, Matsuyama, Ehime 790-8578, Japan
| |
Collapse
|
14
|
Park JM, Kim JH, Choi KS, Kwon HJ. Deleterious Effects of Histidine Tagging to the SH3b Cell Wall-Binding Domain on Recombinant Endolysin Activity. J Microbiol Biotechnol 2024; 34:2331-2337. [PMID: 39467703 PMCID: PMC11637818 DOI: 10.4014/jmb.2408.08003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Revised: 09/27/2024] [Accepted: 09/28/2024] [Indexed: 10/30/2024]
Abstract
Natural and artificial endolysins exhibit bactericidal effects by destroying peptidoglycans in the cell wall of gram-positive bacteria and are usually composed of an N-terminal catalytic domain (CTD) and a C-terminal cell wall-binding domain (CBD). The structures and receptors of CBDs are variable, but bacterial Src homology 3 (SH3b) CBDs are prevalent among the natural endolysins of Staphylococcus aureus. Moreover, although recombinant endolysins with a C-terminal 6x histidine tag (His-tag) are often produced and convenient to purify, the deleterious effects of His-tags on antibacterial activity have not been evaluated thoroughly. Recently, we reported that the antibacterial activity of a commercial lysostaphin without a His-tag differed from that of cell-free lysostaphin with a C-terminal His-tag, and lysostaphin also contains a C-terminal SH3b CBD. In this study, we directly compared the effects of His-tags on the antibacterial activities of lysostaphin and several chimeric lysins possessing different SH3b CBDs. We confirmed that antibacterial activity decreased 16.0-32.0-fold after a His-tag was added to the SH3b CBD.
Collapse
Affiliation(s)
- Jin-Mi Park
- Laboratory of Avian Diseases, Department of Farm Animal Medicine, College of Veterinary Medicine and BK21 PLUS for Veterinary Science, Seoul National University, Seoul 08826, Republic of Korea
- Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul 08826, Republic of Korea
- GeNiner Inc., Seoul 08826, Republic of Korea
| | - Jun-Hyun Kim
- Laboratory of Avian Diseases, Department of Farm Animal Medicine, College of Veterinary Medicine and BK21 PLUS for Veterinary Science, Seoul National University, Seoul 08826, Republic of Korea
- Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul 08826, Republic of Korea
- GeNiner Inc., Seoul 08826, Republic of Korea
| | - Kang-Seuk Choi
- Laboratory of Avian Diseases, Department of Farm Animal Medicine, College of Veterinary Medicine and BK21 PLUS for Veterinary Science, Seoul National University, Seoul 08826, Republic of Korea
- Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul 08826, Republic of Korea
| | - Hyuk-Joon Kwon
- Laboratory of Poultry Medicine, Department of Farm Animal Medicine, College of Veterinary Medicine and BK21 PLUS for Veterinary Science, Seoul National University, Seoul 08826, Republic of Korea
- Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul 08826, Republic of Korea
- Farm Animal Clinical Training and Research Center (FACTRC), GBST, Seoul National University, Gangwon-do 25354, Republic of Korea
- GeNiner Inc., Seoul 08826, Republic of Korea
| |
Collapse
|
15
|
Zhao J, Niu D, Liu J, Jin Z, Mchunu NP, Singh S, Wang Z. Enhancing β-Galactosidase Performance for Galactooligosaccharides Preparation via Strategic Glucose Re-Tunneling. Int J Mol Sci 2024; 25:12316. [PMID: 39596386 PMCID: PMC11594752 DOI: 10.3390/ijms252212316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Revised: 11/12/2024] [Accepted: 11/13/2024] [Indexed: 11/28/2024] Open
Abstract
This study focuses on the characterization and re-engineering of glucose transport in β-galactosidase (BglD) to enhance its catalytic efficiency. Computational prediction methods were employed to identify key residues constituting access tunnels for lactose and glucose, revealing distinct pockets for both substrates. In silico simulated saturation mutagenesis of residues T215 and T473 led to the identification of eight mutant variants exhibiting potential enhancements in glucose transport. Site-directed mutagenesis at T215 and T473 resulted in mutants with consistently enhanced specific activities, turnover rates, and catalytic efficiencies. These mutants also demonstrated improved galactooligosaccharide (GOS) synthesis, yielding an 8.1-10.6% enhancement over wild-type BglD yield. Structural analysis revealed that the mutants exhibited transformed configurations and localizations of glucose conduits, facilitating expedited glucose release. This study's findings suggest that the re-engineered mutants offer promising avenues for enhancing BglD's catalytic efficiency and glucose translocation, thereby improving GOS synthesis. By-product (glucose) re-tunneling is a viable approach for enzyme tunnel engineering and holds significant promise for the molecular evolution of enzymes.
Collapse
Affiliation(s)
- Jihua Zhao
- Department of Biological Chemical Engineering, College of Chemical Engineering and Materials Science, Tianjin University of Science and Technology, Tianjin 300457, China; (J.Z.); (J.L.); (Z.J.); (N.P.M.)
| | - Dandan Niu
- Department of Biological Chemical Engineering, College of Chemical Engineering and Materials Science, Tianjin University of Science and Technology, Tianjin 300457, China; (J.Z.); (J.L.); (Z.J.); (N.P.M.)
| | - Jiaqi Liu
- Department of Biological Chemical Engineering, College of Chemical Engineering and Materials Science, Tianjin University of Science and Technology, Tianjin 300457, China; (J.Z.); (J.L.); (Z.J.); (N.P.M.)
| | - Zhuolin Jin
- Department of Biological Chemical Engineering, College of Chemical Engineering and Materials Science, Tianjin University of Science and Technology, Tianjin 300457, China; (J.Z.); (J.L.); (Z.J.); (N.P.M.)
| | - Nokuthula Peace Mchunu
- Department of Biological Chemical Engineering, College of Chemical Engineering and Materials Science, Tianjin University of Science and Technology, Tianjin 300457, China; (J.Z.); (J.L.); (Z.J.); (N.P.M.)
- National Research Foundation, Pretoria 0001, South Africa
- School of Life Science, University of KwaZulu Natal, Durban 4000, South Africa
| | - Suren Singh
- Department of Biotechnology and Food Science, Faculty of Applied Sciences, Durban University of Technology, Durban 4001, South Africa;
| | - Zhengxiang Wang
- Department of Biological Chemical Engineering, College of Chemical Engineering and Materials Science, Tianjin University of Science and Technology, Tianjin 300457, China; (J.Z.); (J.L.); (Z.J.); (N.P.M.)
- Tianjin Key Laboratory of Industrial Microbiology, College of Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, China
| |
Collapse
|
16
|
Li Y, Sun J, Fu Z, He Y, Chen X, Wang S, Zhang L, Jian J, Yang W, Liu C, Liu X, Yang Y, Bai Z. Engineering the L-tryptophan metabolism for efficient de novo biosynthesis of tryptophol in Saccharomyces cerevisiae. BIOTECHNOLOGY FOR BIOFUELS AND BIOPRODUCTS 2024; 17:130. [PMID: 39415302 PMCID: PMC11481463 DOI: 10.1186/s13068-024-02576-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/04/2024] [Accepted: 10/05/2024] [Indexed: 10/18/2024]
Abstract
Tryptophol (IET) is a metabolite derived from L-tryptophan that can be isolated from plants, bacteria, and fungi and has a wide range of biological activities in living systems. Despite the fact that IET biosynthesis pathways exist naturally in living organisms, industrial-scale production of IET and its derivatives is solely based on environmentally unfriendly chemical conversion. With diminishing petroleum reserves and a significant increase in global demand in all major commercial segments, it becomes essential to develop new technologies to produce chemicals from renewable resources and under mild conditions, such as microbial fermentation. Here we characterized and engineered the less-studied L-tryptophan pathway and IET biosynthesis in the baker's yeast Saccharomyces cerevisiae, with the goal of investigating microbial fermentation as an alternative/green strategy to produce IET. In detail, we divided the aromatic amino acids (AAAs) metabolism related to IET synthesis into the shikimate pathway, the L-tryptophan pathway, the competing L-tyrosine/L-phenylalanine pathways, and the Ehrlich pathway based on a modular engineering concept. Through stepwise engineering of these modules, we obtained a yeast mutant capable of producing IET up to 1.04 g/L through fed-batch fermentation, a ~ 650-fold improvement over the wild-type strain. Besides, our engineering process also revealed many insights about the regulation of AAAs metabolism in S. cerevisiae. Finally, during our engineering process, we also discovered yeast mutants that accumulate anthranilate and L-tryptophan, both of which are precursors of various valuable secondary metabolites from fungi and plants. These strains could be developed to the chassis for natural product biosynthesis upon introducing heterologous pathways.
Collapse
Affiliation(s)
- Ye Li
- Key Laboratory of Industrial Biotechnology of Ministry of Education and School of Biotechnology, Jiangnan University, Wuxi, 214122, China.
| | - Jingzhen Sun
- Key Laboratory of Industrial Biotechnology of Ministry of Education and School of Biotechnology, Jiangnan University, Wuxi, 214122, China
| | - Zhenhao Fu
- Key Laboratory of Industrial Biotechnology of Ministry of Education and School of Biotechnology, Jiangnan University, Wuxi, 214122, China
| | - Yubing He
- Key Laboratory of Industrial Biotechnology of Ministry of Education and School of Biotechnology, Jiangnan University, Wuxi, 214122, China
| | - Xiaorui Chen
- Key Laboratory of Industrial Biotechnology of Ministry of Education and School of Biotechnology, Jiangnan University, Wuxi, 214122, China
| | - Shijie Wang
- Key Laboratory of Industrial Biotechnology of Ministry of Education and School of Biotechnology, Jiangnan University, Wuxi, 214122, China
| | - Lele Zhang
- Key Laboratory of Industrial Biotechnology of Ministry of Education and School of Biotechnology, Jiangnan University, Wuxi, 214122, China
- Wuxi Tmaxtree Biotechnology Co. Ltd., Wuxi, 214072, China
| | - Jiansheng Jian
- Wuxi Tmaxtree Biotechnology Co. Ltd., Wuxi, 214072, China
| | - Weihua Yang
- Changxing Pharmaceutical Co. Ltd., Huzhou, 313100, China
| | - Chunli Liu
- Key Laboratory of Industrial Biotechnology of Ministry of Education and School of Biotechnology, Jiangnan University, Wuxi, 214122, China
| | - Xiuxia Liu
- Key Laboratory of Industrial Biotechnology of Ministry of Education and School of Biotechnology, Jiangnan University, Wuxi, 214122, China
| | - Yankun Yang
- Key Laboratory of Industrial Biotechnology of Ministry of Education and School of Biotechnology, Jiangnan University, Wuxi, 214122, China
| | - Zhonghu Bai
- Key Laboratory of Industrial Biotechnology of Ministry of Education and School of Biotechnology, Jiangnan University, Wuxi, 214122, China.
| |
Collapse
|
17
|
Dong Q, Chang Y, Goodwin PH, Liu Q, Xu W, Xia M, Zhang J, Sun R, Xu S, Wu C, Wu K, Yang L. Double-Wing Motif Protein is a Novel Biofilm Regulatory Factor of the Plant Disease Biocontrol Agent, Bacillus subtilis. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:20273-20285. [PMID: 39226040 DOI: 10.1021/acs.jafc.4c02192] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
Transposon mutagenesis screening of Bacillus subtilis YB-1471, a novel rhizosphere biocontrol agent of Fusarium crown rot (FCR) of wheat, resulted in the identification of orf04391, linked to reduced biofilm formation. The gene encodes a protein possessing a putative tertiary structure of a "double-wing" DNA-binding domain. Expression of orf04391 increased during biofilm development in stationary cultures and during rapid growth in shaking cultures. An orf04391 deletion strain showed reduced biofilm production related to lower levels of the extracellular matrix, and the mutant also had reduced sporulation, adhesion, root colonization, and FCR biocontrol efficiency. Transcriptome analysis of YB-1471 and Δorf04391 in stationary culture showed that the loss of orf04391 resulted in altered expression of numerous genes, including sinI, an initiator of biofilm formation. DNA binding was shown with his-tagged Orf04391 binding to the sinIR operon in vivo and in vitro. Orf04391 appears to be a transcriptional regulator of biofilm formation in B. subtilis through the Spo0A-SinI/SinR pathway.
Collapse
Affiliation(s)
- Qianqian Dong
- Henan Biopesticide Engineering Research Center, Henan Agricultural Microbiology Innovation Center, Institute of Plant Protection Research, Henan Academy of Agricultural Sciences, Zhengzhou 450002, China
| | - Yinghang Chang
- Henan Biopesticide Engineering Research Center, Henan Agricultural Microbiology Innovation Center, Institute of Plant Protection Research, Henan Academy of Agricultural Sciences, Zhengzhou 450002, China
- College of Life Sciences, Henan Agricultural University, Zhengzhou 450002, China
| | - Paul H Goodwin
- School of Environmental Sciences, University of Guelph, Guelph, Ontario N1G 2W1, Canada
| | - Qingxiang Liu
- Henan Biopesticide Engineering Research Center, Henan Agricultural Microbiology Innovation Center, Institute of Plant Protection Research, Henan Academy of Agricultural Sciences, Zhengzhou 450002, China
- College of Life Sciences, Henan Agricultural University, Zhengzhou 450002, China
| | - Wen Xu
- Henan Biopesticide Engineering Research Center, Henan Agricultural Microbiology Innovation Center, Institute of Plant Protection Research, Henan Academy of Agricultural Sciences, Zhengzhou 450002, China
| | - Mingcong Xia
- Henan Biopesticide Engineering Research Center, Henan Agricultural Microbiology Innovation Center, Institute of Plant Protection Research, Henan Academy of Agricultural Sciences, Zhengzhou 450002, China
| | - Jie Zhang
- Henan Biopesticide Engineering Research Center, Henan Agricultural Microbiology Innovation Center, Institute of Plant Protection Research, Henan Academy of Agricultural Sciences, Zhengzhou 450002, China
| | - Runhong Sun
- Henan Biopesticide Engineering Research Center, Henan Agricultural Microbiology Innovation Center, Institute of Plant Protection Research, Henan Academy of Agricultural Sciences, Zhengzhou 450002, China
| | - Shuxia Xu
- College of Life Sciences, Henan Agricultural University, Zhengzhou 450002, China
| | - Chao Wu
- Henan Biopesticide Engineering Research Center, Henan Agricultural Microbiology Innovation Center, Institute of Plant Protection Research, Henan Academy of Agricultural Sciences, Zhengzhou 450002, China
| | - Kun Wu
- College of Life Sciences, Henan Agricultural University, Zhengzhou 450002, China
| | - Lirong Yang
- Henan Biopesticide Engineering Research Center, Henan Agricultural Microbiology Innovation Center, Institute of Plant Protection Research, Henan Academy of Agricultural Sciences, Zhengzhou 450002, China
| |
Collapse
|
18
|
Dierick E, Callens C, Bloch Y, Savvides SN, Hark S, Pelzer S, Ducatelle R, Van Immerseel F, Goossens E. Clostridium perfringens chitinases, key enzymes during early stages of necrotic enteritis in broiler chickens. PLoS Pathog 2024; 20:e1012560. [PMID: 39283899 PMCID: PMC11426533 DOI: 10.1371/journal.ppat.1012560] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Revised: 09/26/2024] [Accepted: 09/03/2024] [Indexed: 09/27/2024] Open
Abstract
The interaction between bacteria and the intestinal mucus is crucial during the early pathogenesis of many enteric diseases in mammals. A critical step in this process employed by both commensal and pathogenic bacteria focuses on the breakdown of the protective layer presented by the intestinal mucus by mucolytic enzymes. C. perfringens type G, the causative agent of necrotic enteritis in broilers, produces two glycosyl hydrolase family 18 chitinases, ChiA and ChiB, which display distinct substrate preferences. Whereas ChiB preferentially processes linear substrates such as chitin, ChiA prefers larger and more branched substrates, such as carbohydrates presented by the chicken intestinal mucus. Here, we show via crystal structures of ChiA and ChiB in the apo and ligand-bound forms that the two enzymes display structural features that explain their substrate preferences providing a structural blueprint for further interrogation of their function and inhibition. This research focusses on the roles of ChiA and ChiB in bacterial proliferation and mucosal attachment, two processes leading to colonization and invasion of the gut. ChiA and ChiB, either supplemented or produced by the bacteria, led to a significant increase in C. perfringens growth. In addition to nutrient acquisition, the importance of chitinases in bacterial attachment to the mucus layer was shown using an in vitro binding assay of C. perfringens to chicken intestinal mucus. Both an in vivo colonization trial and a necrotic enteritis trial were conducted, demonstrating that a ChiA chitinase mutant strain was less capable to colonize the intestine and was hampered in its disease-causing ability as compared to the wild-type strain. Our findings reveal that the pathogen-specific chitinases produced by C. perfringens type G strains play a fundamental role during colonization, suggesting their potential as vaccine targets.
Collapse
Affiliation(s)
- Evelien Dierick
- Livestock Gut Health Team (LiGHT) Ghent, Department of Pathobiology, Pharmacology and Zoological Medicine, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| | - Chana Callens
- Livestock Gut Health Team (LiGHT) Ghent, Department of Pathobiology, Pharmacology and Zoological Medicine, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| | - Yehudi Bloch
- Unit for Structural Biology, Department of Biochemistry and Microbiology, Ghent University, Ghent, Belgium
- Unit for Structural Biology, VIB-UGent Center for Inflammation Research, Ghent, Belgium
- Current address: European Molecular Biology Laboratory, EMBL Hamburg, c/o DESY, Hamburg, Germany
| | - Savvas N. Savvides
- Unit for Structural Biology, Department of Biochemistry and Microbiology, Ghent University, Ghent, Belgium
- Unit for Structural Biology, VIB-UGent Center for Inflammation Research, Ghent, Belgium
| | - Sarah Hark
- Evonik Operations GmbH, Nutrition & Care, Halle, Westfalen, Germany
| | - Stefan Pelzer
- Evonik Operations GmbH, Nutrition & Care, Halle, Westfalen, Germany
| | - Richard Ducatelle
- Livestock Gut Health Team (LiGHT) Ghent, Department of Pathobiology, Pharmacology and Zoological Medicine, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| | - Filip Van Immerseel
- Livestock Gut Health Team (LiGHT) Ghent, Department of Pathobiology, Pharmacology and Zoological Medicine, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| | - Evy Goossens
- Livestock Gut Health Team (LiGHT) Ghent, Department of Pathobiology, Pharmacology and Zoological Medicine, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| |
Collapse
|
19
|
Meesapyodsuk D, Sun K, Qiu X. Structural and functional analysis of plant ELO-like elongase for fatty acid elongation. PLANT MOLECULAR BIOLOGY 2024; 114:90. [PMID: 39172265 DOI: 10.1007/s11103-024-01490-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Accepted: 07/16/2024] [Indexed: 08/23/2024]
Abstract
ELO-like elongase is a condensing enzyme elongating long chain fatty acids in eukaryotes. Eranthis hyemalis ELO-like elongase (EhELO1) is the first higher plant ELO-type elongase that is highly active in elongating a wide range of polyunsaturated fatty acids (PUFAs) and some monounsaturated fatty acids (MUFAs). This study attempted using domain swapping and site-directed mutagenesis of EhELO1 and EhELO2, a close homologue of EhELO1 but with no apparent elongase activity, to elucidate the structural determinants critical for catalytic activity and substrate specificity. Domain swapping analysis of the two showed that subdomain B in the C-terminal half of EhELO1 is essential for MUFA elongation while subdomain C in the C-terminal half of EhELO1 is essential for both PUFA and MUFA elongations, implying these regions are critical in defining the architecture of the substrate tunnel for substrate specificity. Site-directed mutagenesis showed that the glycine at position 220 in the subdomain C plays a key role in differentiating the function of the two elongases. In addition, valine at 161 and cysteine at 165 in subdomain A also play critical roles in defining the architecture of the deep substrate tunnel, thereby contributing significantly to the acceptance of, and interaction with primer substrates.
Collapse
Affiliation(s)
| | - Kaiwen Sun
- Department of Food & Bioproduct Sciences, University of Saskatchewan, Saskatoon, Canada
| | - Xiao Qiu
- National Research Council Canada, Saskatoon, Canada.
- Department of Food & Bioproduct Sciences, University of Saskatchewan, Saskatoon, Canada.
| |
Collapse
|
20
|
Lawong A, Gahalawat S, Ray S, Ho N, Han Y, Ward KE, Deng X, Chen Z, Kumar A, Xing C, Hosangadi V, Fairhurst KJ, Tashiro K, Liszczak G, Shackleford DM, Katneni K, Chen G, Saunders J, Crighton E, Casas A, Robinson JJ, Imlay LS, Zhang X, Lemoff A, Zhao Z, Angulo-Barturen I, Jiménez-Díaz MB, Wittlin S, Campbell SF, Fidock DA, Laleu B, Charman SA, Ready JM, Phillips MA. Identification of potent and reversible piperidine carboxamides that are species-selective orally active proteasome inhibitors to treat malaria. Cell Chem Biol 2024; 31:1503-1517.e19. [PMID: 39084225 PMCID: PMC11531662 DOI: 10.1016/j.chembiol.2024.07.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Revised: 05/08/2024] [Accepted: 07/03/2024] [Indexed: 08/02/2024]
Abstract
Malaria remains a global health concern as drug resistance threatens treatment programs. We identified a piperidine carboxamide (SW042) with anti-malarial activity by phenotypic screening. Selection of SW042-resistant Plasmodium falciparum (Pf) parasites revealed point mutations in the Pf_proteasome β5 active-site (Pfβ5). A potent analog (SW584) showed efficacy in a mouse model of human malaria after oral dosing. SW584 had a low propensity to generate resistance (minimum inoculum for resistance [MIR] >109) and was synergistic with dihydroartemisinin. Pf_proteasome purification was facilitated by His8-tag introduction onto β7. Inhibition of Pfβ5 correlated with parasite killing, without inhibiting human proteasome isoforms or showing cytotoxicity. The Pf_proteasome_SW584 cryoelectron microscopy (cryo-EM) structure showed that SW584 bound non-covalently distal from the catalytic threonine, in an unexplored pocket at the β5/β6/β3 subunit interface that has species differences between Pf and human proteasomes. Identification of a reversible, species selective, orally active series with low resistance propensity provides a path for drugging this essential target.
Collapse
Affiliation(s)
- Aloysus Lawong
- Department of Biochemistry, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, TX 75390, USA
| | - Suraksha Gahalawat
- Department of Biochemistry, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, TX 75390, USA
| | - Sneha Ray
- Department of Biochemistry, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, TX 75390, USA
| | - Nhi Ho
- Department of Biochemistry, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, TX 75390, USA
| | - Yan Han
- Department of Biophysics, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, TX 75390, USA
| | - Kurt E Ward
- Department of Microbiology and Immunology, and Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Xiaoyi Deng
- Department of Biochemistry, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, TX 75390, USA
| | - Zhe Chen
- Department of Biophysics, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, TX 75390, USA
| | - Ashwani Kumar
- Department of Eugene McDermott Center for Human Growth and Development, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, TX 75390, USA
| | - Chao Xing
- Department of Eugene McDermott Center for Human Growth and Development, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, TX 75390, USA; Department of Bioinformatics, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, TX 75390, USA
| | - Varun Hosangadi
- Department of Microbiology and Immunology, and Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Kate J Fairhurst
- Department of Microbiology and Immunology, and Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Kyuto Tashiro
- Department of Biochemistry, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, TX 75390, USA
| | - Glen Liszczak
- Department of Biochemistry, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, TX 75390, USA
| | - David M Shackleford
- Centre for Drug Candidate Optimisation, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC 3052, Australia
| | - Kasiram Katneni
- Centre for Drug Candidate Optimisation, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC 3052, Australia
| | - Gong Chen
- Centre for Drug Candidate Optimisation, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC 3052, Australia
| | - Jessica Saunders
- Centre for Drug Candidate Optimisation, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC 3052, Australia
| | - Elly Crighton
- Centre for Drug Candidate Optimisation, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC 3052, Australia
| | - Arturo Casas
- Department of Biochemistry, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, TX 75390, USA
| | - Joshua J Robinson
- Department of Biochemistry, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, TX 75390, USA
| | - Leah S Imlay
- Department of Biochemistry, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, TX 75390, USA
| | - Xiaoyu Zhang
- Department of Biochemistry, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, TX 75390, USA
| | - Andrew Lemoff
- Department of Biochemistry, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, TX 75390, USA
| | - Zhiyu Zhao
- Children's Medical Center Research Institute, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Iñigo Angulo-Barturen
- The Art of Discovery, Biscay Science and Technology Park, Astrondo Bidea, BIC Bizkaia Bd 612, Derio, 48160 Bizkaia, Basque Country, Spain
| | - María Belén Jiménez-Díaz
- The Art of Discovery, Biscay Science and Technology Park, Astrondo Bidea, BIC Bizkaia Bd 612, Derio, 48160 Bizkaia, Basque Country, Spain
| | - Sergio Wittlin
- Swiss Tropical and Public Health Institute, 4123 Allschwil, Switzerland; University of Basel Kreuzstrasse 2, 4123 Allschwil, Switzerland
| | | | - David A Fidock
- Department of Microbiology and Immunology, and Columbia University Irving Medical Center, New York, NY 10032, USA; Center for Malaria Therapeutics and Antimicrobial Resistance, Division of Infectious Diseases, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Benoît Laleu
- Medicines for Malaria Venture, 1215 Geneva, Switzerland
| | - Susan A Charman
- Centre for Drug Candidate Optimisation, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC 3052, Australia
| | - Joseph M Ready
- Department of Biochemistry, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, TX 75390, USA.
| | - Margaret A Phillips
- Department of Biochemistry, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, TX 75390, USA.
| |
Collapse
|
21
|
McGowan EC, Wu PC, Hellberg Å, Lopez GH, Hyland CA, Olsson ML. A Bioinformatically Initiated Approach to Evaluate GATA1 Regulatory Regions in Samples with Weak D, Del, or D- Phenotypes Despite Normal RHD Exons. Transfus Med Hemother 2024; 51:252-264. [PMID: 39021419 PMCID: PMC11250534 DOI: 10.1159/000538469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2023] [Accepted: 03/19/2024] [Indexed: 07/20/2024] Open
Abstract
Introduction With over 360 blood group antigens in systems recognized, there are antigens, such as RhD, which demonstrate a quantitative reduction in antigen expression due to nucleotide variants in the non-coding region of the gene that result in aberrant splicing or a regulatory mechanism. This study aimed to evaluate bioinformatically predicted GATA1-binding regulatory motifs in the RHD gene for samples presenting with weak or apparently negative RhD antigen expression but showing normal RHD exons. Methods Publicly available open chromatin region data were overlayed with GATA1 motif candidates in RHD. Genomic DNA from weak D, Del or D- samples with normal RHD exons (n = 13) was used to confirm RHD zygosity by quantitative PCR. Then, RHD promoter, intron 1, and intron 2 regions were amplified for Sanger sequencing to detect potential disruptions in the GATA1 motif candidates. Electrophoretic mobility shift assay (EMSA) was performed to assess GATA1-binding. Luciferase assays were used to assess transcriptional activity. Results Bioinformatic analysis identified five of six GATA1 motif candidates in the promoter, intron 1 and intron 2 for investigation in the samples. Luciferase assays showed an enhancement in transcription for GATA1 motifs in intron 1 and for intron 2 only when the R 2 haplotype variant (rs675072G>A) was present. GATA1 motifs were intact in 12 of 13 samples. For one sample with a Del phenotype, a novel RHD c.1-110A>C variant disrupted the GATA1 motif in the promoter which was supported by a lack of a GATA1 supershift in the EMSA and 73% transcriptional activity in the luciferase assay. Two samples were D+/D- chimeras. Conclusion The bioinformatic predictions enabled the identification of a novel DEL allele, RHD c.1-110A>C, which disrupted the GATA1 motif in the proximal promoter. Although the majority of the samples investigated here remain unexplained, we provide GATA1 targets which may benefit future RHD regulatory investigations.
Collapse
Affiliation(s)
- Eunike C. McGowan
- Division of Hematology and Transfusion Medicine, Department of Laboratory Medicine, Lund University, Lund, Sweden
| | - Ping Chun Wu
- Division of Hematology and Transfusion Medicine, Department of Laboratory Medicine, Lund University, Lund, Sweden
| | - Åsa Hellberg
- Department of Clinical Immunology and Transfusion Medicine, Office for Medical Services, Region Skåne, Lund, Sweden
| | - Genghis H. Lopez
- Research and Development, Australian Red Cross Lifeblood, Brisbane, QLD, Australia
- School of Health, University of the Sunshine Coast, Sippy Downs, QLD, Australia
| | - Catherine A. Hyland
- Research and Development, Australian Red Cross Lifeblood, Brisbane, QLD, Australia
- Faculty of Health, Queensland University of Technology, Brisbane, QLD, Australia
| | - Martin L. Olsson
- Division of Hematology and Transfusion Medicine, Department of Laboratory Medicine, Lund University, Lund, Sweden
- Department of Clinical Immunology and Transfusion Medicine, Office for Medical Services, Region Skåne, Lund, Sweden
| |
Collapse
|
22
|
Xu Y, Cohen E, Johnson CN, Parent CA, Coulombe PA. Repeated stress to the skin amplifies neutrophil infiltration in a keratin 17- and PKCα-dependent manner. PLoS Biol 2024; 22:e3002779. [PMID: 39159283 PMCID: PMC11361748 DOI: 10.1371/journal.pbio.3002779] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 08/29/2024] [Accepted: 07/31/2024] [Indexed: 08/21/2024] Open
Abstract
Neutrophils are the first immune cells to reach inflamed sites and contribute to the pathogenesis of chronic inflammatory skin diseases. Yet, little is known about the pattern of neutrophil infiltration in inflamed skin in vivo and the mechanisms mediating their recruitment. Here, we provide insight into the dynamics of neutrophil infiltration in skin in response to acute or repeated inflammatory stress, highlighting a novel keratinocyte- and keratin 17 (K17)-dependent mechanism that regulates neutrophil recruitment to inflamed skin. We used the phorbol ester TPA and UVB, alone or in combination, to induce sterile inflammation in mouse skin. A single TPA treatment results in a neutrophil influx in the dermis that peaks at 12 h and resolves within 24 h. A subsequent TPA treatment or a UVB challenge, when applied 24 h but not 48 h later, accelerates, amplifies, and prolongs neutrophil infiltration. This transient amplification response (TAR) is mediated by local signals in inflamed skin, can be recapitulated in ex vivo culture, and involves the K17-dependent sustainment of protein kinase Cα (PKCα) activity and release of chemoattractants by stressed keratinocytes. K17 binds RACK1, a scaffold protein essential for PKCα activity. The N-terminal head domain of K17 is crucial for its association with RACK1 and regulation of PKCα activity. Analysis of RNAseq data reveals a signature consistent with TAR and PKCα activation in inflammatory skin diseases. These findings uncover a novel, keratin-dependent mechanism that amplifies neutrophil recruitment in skin under stress, with direct implications for inflammatory skin disorders.
Collapse
Affiliation(s)
- Yang Xu
- Graduate Program in Pharmacology and Department of Pharmacology, University of Michigan Medical School, Ann Arbor, Michigan, United States of America
| | - Erez Cohen
- Department of Cell & Developmental Biology, University of Michigan Medical School, Ann Arbor, Michigan, United States of America
| | - Craig N. Johnson
- Department of Cell & Developmental Biology, University of Michigan Medical School, Ann Arbor, Michigan, United States of America
| | - Carole A. Parent
- Graduate Program in Pharmacology and Department of Pharmacology, University of Michigan Medical School, Ann Arbor, Michigan, United States of America
- Department of Cell & Developmental Biology, University of Michigan Medical School, Ann Arbor, Michigan, United States of America
- Life Science Institute, University of Michigan, Ann Arbor, Michigan, United States of America
- Rogel Cancer Center, University of Michigan Medical School, Ann Arbor, Michigan, United States of America
| | - Pierre A. Coulombe
- Department of Cell & Developmental Biology, University of Michigan Medical School, Ann Arbor, Michigan, United States of America
- Rogel Cancer Center, University of Michigan Medical School, Ann Arbor, Michigan, United States of America
- Department of Dermatology, University of Michigan Medical School, Ann Arbor, Michigan, United States of America
| |
Collapse
|
23
|
Charoenwongpaiboon T, Charoenwongphaibun C, Wangpaiboon K, Panpetch P, Wanichacheva N, Pichyangkura R. Endo- and exo-levanases from Bacillus subtilis HM7: Catalytic components, synergistic cooperation, and application in fructooligosaccharide synthesis. Int J Biol Macromol 2024; 271:132508. [PMID: 38782321 DOI: 10.1016/j.ijbiomac.2024.132508] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2024] [Revised: 04/28/2024] [Accepted: 05/17/2024] [Indexed: 05/25/2024]
Abstract
Levan-type fructooligosaccharides (LFOS) exhibit significant biological activities and selectively promote the growth of certain beneficial bacteria. Levanase is an important enzyme for LFOS production. In this study, two isoforms of levanases, exo- and endo-type depolymerizing enzymes, from Bacillus subtilis HM7 isolated from Dynastes hercules larvae excrement were cloned, expressed, and characterized. The synergistic effect on the levan hydrolysis and kinetic properties of both isoforms were evaluated, indicating their cooperation in levan metabolism, where the endo-levanase catalyzes a rate-limiting step. In addition, homology models and molecular dynamics simulations revealed the key amino residues of the enzymes for levan binding and catalysis. It was found that both isoforms possessed distinct binding residues in the active sites, suggesting the importance of the specificity of the enzymes. Finally, we demonstrated the potential of endo-type levanase in LFOS synthesis using a one-pot reaction with levansucrase. Overall, this study fills the knowledge gap in understanding levanase's mechanism, making an important contribution to the fields of food science and biotechnology.
Collapse
Affiliation(s)
| | - Chonnipha Charoenwongphaibun
- Division of Chemistry, Department of Physical and Material Sciences, Faculty of Liberal Arts and Science, Kasetsart University, Kamphaeng Saen Campus, Kamphaeng Sean, Nakhon Pathom 73140, Thailand
| | - Karan Wangpaiboon
- Center of Excellence in Structural and Computational Biology, Department of Biochemistry, Faculty of Science, Chulalongkorn University, Bangkok 10330, Thailand
| | - Pawinee Panpetch
- Center of Excellence in Structural and Computational Biology, Department of Biochemistry, Faculty of Science, Chulalongkorn University, Bangkok 10330, Thailand
| | - Nantanit Wanichacheva
- Department of Chemistry, Faculty of Science, Silpakorn University, Nakhon Pathom 73000, Thailand
| | - Rath Pichyangkura
- Department of Biochemistry, Faculty of Science, Chulalongkorn University, Bangkok 10330, Thailand
| |
Collapse
|
24
|
Wu PC, McGowan EC, Lee YQ, Ghosh S, Hansson J, Olsson ML. Epigenetic dissection of human blood group genes reveals regulatory elements and detailed characteristics of KEL and four other loci. Transfusion 2024; 64:1083-1096. [PMID: 38644556 DOI: 10.1111/trf.17840] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2023] [Revised: 03/23/2024] [Accepted: 04/08/2024] [Indexed: 04/23/2024]
Abstract
BACKGROUND Blood typing is essential for safe transfusions and is performed serologically or genetically. Genotyping predominantly focuses on coding regions, but non-coding variants may affect gene regulation, as demonstrated in the ABO, FY and XG systems. To uncover regulatory loci, we expanded a recently developed bioinformatics pipeline for discovery of non-coding variants by including additional epigenetic datasets. METHODS Multiple datasets including ChIP-seq with erythroid transcription factors (TFs), histone modifications (H3K27ac, H3K4me1), and chromatin accessibility (ATAC-seq) were analyzed. Candidate regulatory regions were investigated for activity (luciferase assays) and TF binding (electrophoretic mobility shift assay, EMSA, and mass spectrometry, MS). RESULTS In total, 814 potential regulatory sites in 47 blood-group-related genes were identified where one or more erythroid TFs bound. Enhancer candidates in CR1, EMP3, ABCB6, and ABCC4 indicated by ATAC-seq, histone markers, and co-occupancy of 4 TFs (GATA1/KLF1/RUNX1/NFE2) were investigated but only CR1 and ABCC4 showed increased transcription. Co-occupancy of GATA1 and KLF1 was observed in the KEL promoter, previously reported to contain GATA1 and Sp1 sites. TF binding energy scores decreased when three naturally occurring variants were introduced into GATA1 and KLF1 motifs. Two of three GATA1 sites and the KLF1 site were confirmed functionally. EMSA and MS demonstrated increased GATA1 and KLF1 binding to the wild-type compared to variant motifs. DISCUSSION This combined bioinformatics and experimental approach revealed multiple candidate regulatory regions and predicted TF co-occupancy sites. The KEL promoter was characterized in detail, indicating that two adjacent GATA1 and KLF1 motifs are most crucial for transcription.
Collapse
Affiliation(s)
- Ping Chun Wu
- Division of Hematology and Transfusion Medicine, Department of Laboratory Medicine and the Lund Stem Cell Center, Lund University, Lund, Sweden
| | - Eunike C McGowan
- Division of Hematology and Transfusion Medicine, Department of Laboratory Medicine and the Lund Stem Cell Center, Lund University, Lund, Sweden
| | - Yan Quan Lee
- Division of Hematology and Transfusion Medicine, Department of Laboratory Medicine and the Lund Stem Cell Center, Lund University, Lund, Sweden
| | - Sudip Ghosh
- Department of Experimental Medical Science and Lund Stem Cell Center, Lund University, Lund, Sweden
| | - Jenny Hansson
- Department of Experimental Medical Science and Lund Stem Cell Center, Lund University, Lund, Sweden
| | - Martin L Olsson
- Division of Hematology and Transfusion Medicine, Department of Laboratory Medicine and the Lund Stem Cell Center, Lund University, Lund, Sweden
- Department of Clinical Immunology and Transfusion Medicine, Office for Medical Services, Region Skåne, Sweden
| |
Collapse
|
25
|
Deyev SM, Oroujeni M, Garousi J, Gräslund T, Li R, Rosly AHB, Orlova A, Konovalova E, Schulga A, Vorobyeva A, Tolmachev V. Preclinical Evaluation of HER2-Targeting DARPin G3: Impact of Albumin-Binding Domain (ABD) Fusion. Int J Mol Sci 2024; 25:4246. [PMID: 38673831 PMCID: PMC11050402 DOI: 10.3390/ijms25084246] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Revised: 04/07/2024] [Accepted: 04/09/2024] [Indexed: 04/28/2024] Open
Abstract
Designed ankyrin repeat protein (DARPin) G3 is an engineered scaffold protein. This small (14.5 kDa) targeting protein binds with high affinity to human epidermal growth factor receptor 2 (HER2). HER2 is overexpressed in several cancers. The use of the DARPin G3 for radionuclide therapy is complicated by its high renal reabsorption after clearance via the glomeruli. We tested the hypothesis that a fusion of the DARPin G3 with an albumin-binding domain (ABD) would prevent rapid renal excretion and high renal reabsorption resulting in better tumour targeting. Two fusion proteins were produced, one with the ABD at the C-terminus (G3-ABD) and another at the N-terminus (ABD-G3). Both variants were labelled with 177Lu. The binding properties of the novel constructs were evaluated in vitro and their biodistribution was compared in mice with implanted human HER2-expressing tumours. Fusion with the ABD increased the retention time of both constructs in blood compared with the non-ABD-fused control. The effect of fusion with the ABD depended strongly on the order of the domains in the constructs, resulting in appreciably better targeting properties of [177Lu]Lu-G3-ABD. Our data suggest that the order of domains is critical for the design of targeting constructs based on scaffold proteins.
Collapse
Affiliation(s)
- Sergey M. Deyev
- Molecular Immunology Laboratory, Shemyakin & Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 117997 Moscow, Russia; (S.M.D.); (E.K.); (A.S.)
- Research Centrum for Oncotheranostics, Research School of Chemistry and Applied Biomedical Sciences, Tomsk Polytechnic University, 634050 Tomsk, Russia
| | - Maryam Oroujeni
- Department of Immunology, Genetics and Pathology, Uppsala University, 751 85 Uppsala, Sweden; (M.O.); (J.G.); (A.H.B.R.); (A.V.)
- Affibody AB, 171 65 Solna, Sweden
| | - Javad Garousi
- Department of Immunology, Genetics and Pathology, Uppsala University, 751 85 Uppsala, Sweden; (M.O.); (J.G.); (A.H.B.R.); (A.V.)
- Department of Protein Science, KTH—Royal Institute of Technology, 106 91 Stockholm, Sweden; (T.G.); (R.L.)
| | - Torbjörn Gräslund
- Department of Protein Science, KTH—Royal Institute of Technology, 106 91 Stockholm, Sweden; (T.G.); (R.L.)
| | - Ruonan Li
- Department of Protein Science, KTH—Royal Institute of Technology, 106 91 Stockholm, Sweden; (T.G.); (R.L.)
| | - Alia Hani Binti Rosly
- Department of Immunology, Genetics and Pathology, Uppsala University, 751 85 Uppsala, Sweden; (M.O.); (J.G.); (A.H.B.R.); (A.V.)
| | - Anna Orlova
- Department of Medicinal Chemistry, Uppsala University, 751 83 Uppsala, Sweden;
| | - Elena Konovalova
- Molecular Immunology Laboratory, Shemyakin & Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 117997 Moscow, Russia; (S.M.D.); (E.K.); (A.S.)
| | - Alexey Schulga
- Molecular Immunology Laboratory, Shemyakin & Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 117997 Moscow, Russia; (S.M.D.); (E.K.); (A.S.)
- Research Centrum for Oncotheranostics, Research School of Chemistry and Applied Biomedical Sciences, Tomsk Polytechnic University, 634050 Tomsk, Russia
| | - Anzhelika Vorobyeva
- Department of Immunology, Genetics and Pathology, Uppsala University, 751 85 Uppsala, Sweden; (M.O.); (J.G.); (A.H.B.R.); (A.V.)
| | - Vladimir Tolmachev
- Department of Immunology, Genetics and Pathology, Uppsala University, 751 85 Uppsala, Sweden; (M.O.); (J.G.); (A.H.B.R.); (A.V.)
| |
Collapse
|
26
|
Hsu HC, Wang M, Kovach A, Darwin AJ, Li H. P. aeruginosa CtpA protease adopts a novel activation mechanism to initiate the proteolytic process. EMBO J 2024; 43:1634-1652. [PMID: 38467832 PMCID: PMC11021448 DOI: 10.1038/s44318-024-00069-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 02/19/2024] [Accepted: 02/26/2024] [Indexed: 03/13/2024] Open
Abstract
During bacterial cell growth, hydrolases cleave peptide cross-links between strands of the peptidoglycan sacculus to allow new strand insertion. The Pseudomonas aeruginosa carboxyl-terminal processing protease (CTP) CtpA regulates some of these hydrolases by degrading them. CtpA assembles as an inactive hexamer composed of a trimer-of-dimers, but its lipoprotein binding partner LbcA activates CtpA by an unknown mechanism. Here, we report the cryo-EM structures of the CtpA-LbcA complex. LbcA has an N-terminal adaptor domain that binds to CtpA, and a C-terminal superhelical tetratricopeptide repeat domain. One LbcA molecule attaches to each of the three vertices of a CtpA hexamer. LbcA triggers relocation of the CtpA PDZ domain, remodeling of the substrate binding pocket, and realignment of the catalytic residues. Surprisingly, only one CtpA molecule in a CtpA dimer is activated upon LbcA binding. Also, a long loop from one CtpA dimer inserts into a neighboring dimer to facilitate the proteolytic activity. This work has revealed an activation mechanism for a bacterial CTP that is strikingly different from other CTPs that have been characterized structurally.
Collapse
Affiliation(s)
- Hao-Chi Hsu
- Department of Structural Biology, Van Andel Institute, Grand Rapids, MI, USA
| | - Michelle Wang
- Department of Microbiology, New York University Grossman School of Medicine, New York, NY, USA
| | - Amanda Kovach
- Department of Structural Biology, Van Andel Institute, Grand Rapids, MI, USA
| | - Andrew J Darwin
- Department of Microbiology, New York University Grossman School of Medicine, New York, NY, USA.
| | - Huilin Li
- Department of Structural Biology, Van Andel Institute, Grand Rapids, MI, USA.
| |
Collapse
|
27
|
Wu YC, Yu CW, Chiu JY, Chiang YH, Mitsuda N, Yen XC, Huang TP, Chang TF, Yen CJ, Guo WJ. The AT-hook protein AHL29 promotes Bacillus subtilis colonization by suppressing SWEET2-mediated sugar retrieval in Arabidopsis roots. PLANT, CELL & ENVIRONMENT 2024; 47:1084-1098. [PMID: 38037476 DOI: 10.1111/pce.14779] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Revised: 11/03/2023] [Accepted: 11/23/2023] [Indexed: 12/02/2023]
Abstract
Beneficial Bacillus subtilis (BS) symbiosis could combat root pathogenesis, but it relies on root-secreted sugars. Understanding the molecular control of sugar flux during colonization would benefit biocontrol applications. The SWEET (Sugar Will Eventually Be Exported Transporter) uniporter regulates microbe-induced sugar secretion from roots; thus, its homologs may modulate sugar distribution upon BS colonization. Quantitative polymerase chain reaction revealed that gene transcripts of SWEET2, but not SWEET16 and 17, were significantly induced in seedling roots after 12 h of BS inoculation. Particularly, SWEET2-β-glucuronidase fusion proteins accumulated in the apical mature zone where BS abundantly colonized. Yet, enhanced BS colonization in sweet2 mutant roots suggested a specific role for SWEET2 to constrain BS propagation, probably by limiting hexose secretion. By employing yeast one-hybrid screening and ectopic expression in Arabidopsis protoplasts, the transcription factor AHL29 was identified to function as a repressor of SWEET2 expression through the AT-hook motif. Repression occurred despite immunity signals. Additionally, enhanced SWEET2 expression and reduced colonies were specifically detected in roots of BS-colonized ahl29 mutant. Taken together, we propose that BS colonization may activate repression of AHL29 on SWEET2 transcription that would be enhanced by immunity signals, thereby maintaining adequate sugar secretion for a beneficial Bacillus association.
Collapse
Affiliation(s)
- Yun-Chien Wu
- Department of Biotechnology and Bioindustry Sciences, National Cheng Kung University, Tainan, Taiwan ROC
| | - Chien-Wen Yu
- Department of Biotechnology and Bioindustry Sciences, National Cheng Kung University, Tainan, Taiwan ROC
| | - Jo-Yu Chiu
- Department of Biotechnology and Bioindustry Sciences, National Cheng Kung University, Tainan, Taiwan ROC
| | - Yu-Hsuan Chiang
- Department of Biotechnology and Bioindustry Sciences, National Cheng Kung University, Tainan, Taiwan ROC
| | - Nobutaka Mitsuda
- Bioproduction Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), Tsukuba, Ibaraki, Japan
| | - Xu-Chen Yen
- Department of Plant Pathology, National Chung Hsing University, Taichung, Taiwan ROC
| | - Tzu-Pi Huang
- Department of Plant Pathology, National Chung Hsing University, Taichung, Taiwan ROC
- Innovation and Development Center of Sustainable Agriculture, National Chung Hsing University, Taichung, Taiwan ROC
- Master and Doctoral Degree Program in Plant Health Care, Academy of Circular Economy, National Chung Hsing University, Nantou, Taiwan ROC
| | - Tzu-Fang Chang
- Department of Biotechnology and Bioindustry Sciences, National Cheng Kung University, Tainan, Taiwan ROC
| | - Cen-Jie Yen
- Department of Biotechnology and Bioindustry Sciences, National Cheng Kung University, Tainan, Taiwan ROC
| | - Woei-Jiun Guo
- Department of Biotechnology and Bioindustry Sciences, National Cheng Kung University, Tainan, Taiwan ROC
| |
Collapse
|
28
|
Álvarez-Guerra I, Block E, Broeskamp F, Gabrijelčič S, Infant T, de Ory A, Habernig L, Andréasson C, Levine TP, Höög JL, Büttner S. LDO proteins and Vac8 form a vacuole-lipid droplet contact site to enable starvation-induced lipophagy in yeast. Dev Cell 2024; 59:759-775.e5. [PMID: 38354739 DOI: 10.1016/j.devcel.2024.01.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Revised: 11/15/2023] [Accepted: 01/18/2024] [Indexed: 02/16/2024]
Abstract
Lipid droplets (LDs) are fat storage organelles critical for energy and lipid metabolism. Upon nutrient exhaustion, cells consume LDs via gradual lipolysis or via lipophagy, the en bloc uptake of LDs into the vacuole. Here, we show that LDs dock to the vacuolar membrane via a contact site that is required for lipophagy in yeast. The LD-localized LDO proteins carry an intrinsically disordered region that directly binds vacuolar Vac8 to form vCLIP, the vacuolar-LD contact site. Nutrient limitation drives vCLIP formation, and its inactivation blocks lipophagy, resulting in impaired caloric restriction-induced longevity. We establish a functional link between lipophagy and microautophagy of the nucleus, both requiring Vac8 to form respective contact sites upon metabolic stress. In sum, we identify the tethering machinery of vCLIP and find that Vac8 provides a platform for multiple and competing contact sites associated with autophagy.
Collapse
Affiliation(s)
- Irene Álvarez-Guerra
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, 10691 Stockholm, Sweden
| | - Emma Block
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, 10691 Stockholm, Sweden
| | - Filomena Broeskamp
- Department of Chemistry and Molecular Biology, University of Gothenburg, 40530 Gothenburg, Sweden
| | - Sonja Gabrijelčič
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, 10691 Stockholm, Sweden
| | - Terence Infant
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, 10691 Stockholm, Sweden
| | - Ana de Ory
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, 10691 Stockholm, Sweden
| | - Lukas Habernig
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, 10691 Stockholm, Sweden
| | - Claes Andréasson
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, 10691 Stockholm, Sweden
| | - Tim P Levine
- UCL Institute of Ophthalmology, Bath Street, London EC1V 9EL, UK
| | - Johanna L Höög
- Department of Chemistry and Molecular Biology, University of Gothenburg, 40530 Gothenburg, Sweden
| | - Sabrina Büttner
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, 10691 Stockholm, Sweden.
| |
Collapse
|
29
|
Vélez-González F, Marcos-Vilchis A, Vega-Baray B, Dreyfus G, Poggio S, Camarena L. Rotation of the Fla2 flagella of Cereibacter sphaeroides requires the periplasmic proteins MotK and MotE that interact with the flagellar stator protein MotB2. PLoS One 2024; 19:e0298028. [PMID: 38507361 PMCID: PMC10954123 DOI: 10.1371/journal.pone.0298028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Accepted: 01/16/2024] [Indexed: 03/22/2024] Open
Abstract
The bacterial flagellum is a complex structure formed by more than 25 different proteins, this appendage comprises three conserved structures: the basal body, the hook and filament. The basal body, embedded in the cell envelope, is the most complex structure and houses the export apparatus and the motor. In situ images of the flagellar motor in different species have revealed a huge diversity of structures that surround the well-conserved periplasmic components of the basal body. The identity of the proteins that form these novel structures in many cases has been elucidated genetically and biochemically, but in others they remain to be identified or characterized. In this work, we report that in the alpha proteobacteria Cereibacter sphaeroides the novel protein MotK along with MotE are essential for flagellar rotation. We show evidence that these periplasmic proteins interact with each other and with MotB2. Moreover, these proteins localize to the flagellated pole and MotK localization is dependent on MotB2 and MotA2. These results together suggest that the role of MotK and MotE is to activate or recruit the flagellar stators to the flagellar structure.
Collapse
Affiliation(s)
- Fernanda Vélez-González
- Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Arely Marcos-Vilchis
- Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Benjamín Vega-Baray
- Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Georges Dreyfus
- Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Sebastian Poggio
- Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Laura Camarena
- Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City, Mexico
| |
Collapse
|
30
|
Lee JH, Lee U, Yoo JH, Lee TS, Jung JH, Kim HS. AraDQ: an automated digital phenotyping software for quantifying disease symptoms of flood-inoculated Arabidopsis seedlings. PLANT METHODS 2024; 20:44. [PMID: 38493119 PMCID: PMC10943777 DOI: 10.1186/s13007-024-01171-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Accepted: 03/09/2024] [Indexed: 03/18/2024]
Abstract
BACKGROUND Plant scientists have largely relied on pathogen growth assays and/or transcript analysis of stress-responsive genes for quantification of disease severity and susceptibility. These methods are destructive to plants, labor-intensive, and time-consuming, thereby limiting their application in real-time, large-scale studies. Image-based plant phenotyping is an alternative approach that enables automated measurement of various symptoms. However, most of the currently available plant image analysis tools require specific hardware platform and vendor specific software packages, and thus, are not suited for researchers who are not primarily focused on plant phenotyping. In this study, we aimed to develop a digital phenotyping tool to enhance the speed, accuracy, and reliability of disease quantification in Arabidopsis. RESULTS Here, we present the Arabidopsis Disease Quantification (AraDQ) image analysis tool for examination of flood-inoculated Arabidopsis seedlings grown on plates containing plant growth media. It is a cross-platform application program with a user-friendly graphical interface that contains highly accurate deep neural networks for object detection and segmentation. The only prerequisite is that the input image should contain a fixed-sized 24-color balance card placed next to the objects of interest on a white background to ensure reliable and reproducible results, regardless of the image acquisition method. The image processing pipeline automatically calculates 10 different colors and morphological parameters for individual seedlings in the given image, and disease-associated phenotypic changes can be easily assessed by comparing plant images captured before and after infection. We conducted two case studies involving bacterial and plant mutants with reduced virulence and disease resistance capabilities, respectively, and thereby demonstrated that AraDQ can capture subtle changes in plant color and morphology with a high level of sensitivity. CONCLUSIONS AraDQ offers a simple, fast, and accurate approach for image-based quantification of plant disease symptoms using various parameters. Its fully automated pipeline neither requires prior image processing nor costly hardware setups, allowing easy implementation of the software by researchers interested in digital phenotyping of diseased plants.
Collapse
Grants
- Grant No. 2022R1C1C1012137 The National Research Foundation of Korea
- Grant No. 421002-04) The Korea Institute of Planning and Evaluation for Technology in Food, Agriculture, and Forestry (IPET) and Korea Smart Farm R&D (KosFarm) through the Smart Farm Innovation Technology Development Program, funded by the Ministry of Agriculture, Food and Rural Affairs (MAFRA) and Ministry of Science and ICT (MSIT), Rural Development Administration (RDA)
- The Korea Institute of Planning and Evaluation for Technology in Food, Agriculture, and Forestry (IPET) and Korea Smart Farm R&D (KosFarm) through the Smart Farm Innovation Technology Development Program, funded by the Ministry of Agriculture, Food and Rural Affairs (MAFRA) and Ministry of Science and ICT (MSIT), Rural Development Administration (RDA)
Collapse
Affiliation(s)
- Jae Hoon Lee
- Department of Agricultural Biotechnology, Seoul National University, Seoul, 08826, Republic of Korea
- Research Institute of Agriculture and Life Sciences, Seoul National University, Seoul, 08826, Republic of Korea
| | - Unseok Lee
- Smart Farm Research Center, Korea Institute of Science and Technology, Gangneung, 25451, Republic of Korea
| | - Ji Hye Yoo
- Smart Farm Research Center, Korea Institute of Science and Technology, Gangneung, 25451, Republic of Korea
| | - Taek Sung Lee
- Smart Farm Research Center, Korea Institute of Science and Technology, Gangneung, 25451, Republic of Korea
| | - Je Hyeong Jung
- Smart Farm Research Center, Korea Institute of Science and Technology, Gangneung, 25451, Republic of Korea
| | - Hyoung Seok Kim
- Smart Farm Research Center, Korea Institute of Science and Technology, Gangneung, 25451, Republic of Korea.
| |
Collapse
|
31
|
Clark ZS, O'Connor M. Suppressor analysis links trans-translation and ribosomal protein uS7 to RluD function in Escherichia coli. Biochem Biophys Res Commun 2024; 700:149584. [PMID: 38295647 PMCID: PMC10878134 DOI: 10.1016/j.bbrc.2024.149584] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Accepted: 01/25/2024] [Indexed: 02/17/2024]
Abstract
The pseudouridine (ψ) synthase, RluD is responsible for three ψ modifications in the helix 69 (H69) of bacterial 23S rRNA. While normally dispensable, rluD becomes critical for rapid cell growth in bacteria that are defective in translation-termination. In slow-growing rluD- bacteria, suppressors affecting termination factors RF2 and RF3 arise frequently and restore normal termination and rapid cell growth. Here we describe two weaker suppressors, affecting rpsG, encoding ribosomal protein uS7 and ssrA, encoding tmRNA. In K-12 strains of E. coli, rpsG terminates at a TGA codon. In the suppressor strain, alteration of an upstream CAG to a TAG stop codon results in a shortened uS7 and partial alleviation of slow growth, likely by replacing an inefficient TGA stop codon with the more efficient TAG. Inefficient termination events, such as occurs in some rluD- strains, are targeted by trans-translation. Inactivation of the ssrA gene in slow-growing, termination-defective mutants lacking RluD and RF3, also partially restores robust growth, most probably by preventing destruction of completed polypeptides on ribosomes at slow-terminating stop codons. Finally, an additional role for RluD has been proposed, independent of its pseudouridine synthase activity. This is based on the observation that plasmids expressing catalytically dead (D139N or D139T) RluD proteins could nonetheless restore robust growth to an E. coli K-12 rluD- mutant. However, newly constructed D139N and D139T rluD plasmids do not have any growth-restoring activity and the original observations were likely due to the appearance of suppressors.
Collapse
Affiliation(s)
- Zachary S Clark
- Division of Biology and Biomedical Systems, School of Science and Engineering, 306 Spencer Hall, University of Missouri-Kansas City, 5007 Rockhill Rd., Kansas City, MO, 64110, USA
| | - Michael O'Connor
- Division of Biology and Biomedical Systems, School of Science and Engineering, 306 Spencer Hall, University of Missouri-Kansas City, 5007 Rockhill Rd., Kansas City, MO, 64110, USA.
| |
Collapse
|
32
|
Ibusuki R, Iwama E, Shimauchi A, Tsutsumi H, Yoneshima Y, Tanaka K, Okamoto I. TP53 gain-of-function mutations promote osimertinib resistance via TNF-α-NF-κB signaling in EGFR-mutated lung cancer. NPJ Precis Oncol 2024; 8:60. [PMID: 38431700 PMCID: PMC10908812 DOI: 10.1038/s41698-024-00557-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Accepted: 02/20/2024] [Indexed: 03/05/2024] Open
Abstract
EGFR tyrosine kinase inhibitors (TKIs) are effective against EGFR-mutated lung cancer, but tumors eventually develop resistance to these drugs. Although TP53 gain-of-function (GOF) mutations promote carcinogenesis, their effect on EGFR-TKI efficacy has remained unclear. We here established EGFR-mutated lung cancer cell lines that express wild-type (WT) or various mutant p53 proteins with CRISPR-Cas9 technology and found that TP53-GOF mutations promote early development of resistance to the EGFR-TKI osimertinib associated with sustained activation of ERK and expression of c-Myc. Gene expression analysis revealed that osimertinib activates TNF-α-NF-κB signaling specifically in TP53-GOF mutant cells. In such cells, osimertinib promoted interaction of p53 with the NF-κB subunit p65, translocation of the resulting complex to the nucleus and its binding to the TNF promoter, and TNF-α production. Concurrent treatment of TP53-GOF mutant cells with the TNF-α inhibitor infliximab suppressed acquisition of osimertinib resistance as well as restored osimertinib sensitivity in resistant cells in association with attenuation of ERK activation and c-Myc expression. Our findings indicate that induction of TNF-α expression by osimertinib in TP53-GOF mutant cells contributes to the early development of osimertinib resistance, and that TNF-α inhibition may therefore be an effective strategy to overcome such resistance in EGFR-mutant lung cancer with TP53-GOF mutations.
Collapse
Affiliation(s)
- Ritsu Ibusuki
- Department of Respiratory Medicine, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Eiji Iwama
- Department of Respiratory Medicine, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan.
| | - Atsushi Shimauchi
- Department of Respiratory Medicine, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Hirono Tsutsumi
- Department of Respiratory Medicine, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Yasuto Yoneshima
- Department of Respiratory Medicine, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Kentaro Tanaka
- Department of Respiratory Medicine, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Isamu Okamoto
- Department of Respiratory Medicine, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| |
Collapse
|
33
|
Coyle CW, Knight KA, Brown HC, George SN, Denning G, Branella GM, Childers KC, Spiegel PC, Spencer HT, Doering CB. Humanization and functional characterization of enhanced coagulation factor IX variants identified through ancestral sequence reconstruction. J Thromb Haemost 2024; 22:633-644. [PMID: 38016519 PMCID: PMC10922771 DOI: 10.1016/j.jtha.2023.11.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Revised: 10/17/2023] [Accepted: 11/06/2023] [Indexed: 11/30/2023]
Abstract
BACKGROUND Laboratory resurrection of ancient coagulation factor (F) IX variants generated through ancestral sequence reconstruction led to the discovery of a FIX variant, designated An96, which possesses enhanced specific activity independent of and additive to that provided by human p.Arg384Lys, referred to as FIX-Padua. OBJECTIVES The goal of the current study was to identify the amino acid substitution(s) responsible for the enhanced activity of An96 and create a humanized An96 FIX transgene for gene therapy application. METHODS Reductionist screening approaches, including domain swapping and scanning residue substitution, were used and guided by one-stage FIX activity assays. In vitro characterization of top candidates included recombinant high-purity preparation, specific activity determination, and enzyme kinetic analysis. Final candidates were packaged into adeno-associated viral (AAV) vectors and delivered to hemophilia B mice. RESULTS Five of 42 total amino acid substitutions in An96 appear sufficient to retain the enhanced activity of An96 in an otherwise human FIX variant. Additional substitution of the Padua variant further increased the specific activity 5-fold. This candidate, designated ET9, demonstrated 51-fold greater specific activity than hFIX. AAV2/8-ET9 treated hemophilia B mice produced plasma FIX activities equivalent to those observed previously for AAV2/8-An96-Padua, which were 10-fold higher than AAV2/8-hFIX-Padua. CONCLUSION Starting from computationally inferred ancient FIX sequences, novel amino acid substitutions conferring activity enhancement were identified and translated into an AAV-FIX gene therapy cassette demonstrating high potency. This ancestral sequence reconstruction discovery and sequence mapping refinement approach represents a promising platform for broader protein drug and gene therapy candidate optimization.
Collapse
Affiliation(s)
- Christopher W Coyle
- Molecular and Systems Pharmacology Graduate Program, Graduate Division of Biological and Biomedical Sciences, Laney Graduate School, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Kristopher A Knight
- Molecular and Systems Pharmacology Graduate Program, Graduate Division of Biological and Biomedical Sciences, Laney Graduate School, Emory University School of Medicine, Atlanta, Georgia, USA
| | | | | | | | - Gianna M Branella
- Cancer Biology Graduate Program, Graduate Division of Biological and Biomedical Sciences, Laney Graduate School, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Kenneth C Childers
- Chemistry Department, Western Washington University, Bellingham, Washington, USA
| | - P Clint Spiegel
- Chemistry Department, Western Washington University, Bellingham, Washington, USA
| | - H Trent Spencer
- Cell and Gene Therapy Program, Department of Pediatrics, Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta and Emory University, Atlanta, Georgia, USA
| | - Christopher B Doering
- Cell and Gene Therapy Program, Department of Pediatrics, Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta and Emory University, Atlanta, Georgia, USA.
| |
Collapse
|
34
|
Patel PH, Taylor VL, Zhang C, Getz LJ, Fitzpatrick AD, Davidson AR, Maxwell KL. Anti-phage defence through inhibition of virion assembly. Nat Commun 2024; 15:1644. [PMID: 38388474 PMCID: PMC10884400 DOI: 10.1038/s41467-024-45892-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Accepted: 02/06/2024] [Indexed: 02/24/2024] Open
Abstract
Bacteria have evolved diverse antiviral defence mechanisms to protect themselves against phage infection. Phages integrated into bacterial chromosomes, known as prophages, also encode defences that protect the bacterial hosts in which they reside. Here, we identify a type of anti-phage defence that interferes with the virion assembly pathway of invading phages. The protein that mediates this defence, which we call Tab (for 'Tail assembly blocker'), is constitutively expressed from a Pseudomonas aeruginosa prophage. Tab allows the invading phage replication cycle to proceed, but blocks assembly of the phage tail, thus preventing formation of infectious virions. While the infected cell dies through the activity of the replicating phage lysis proteins, there is no release of infectious phage progeny, and the bacterial community is thereby protected from a phage epidemic. Prophages expressing Tab are not inhibited during their own lytic cycle because they express a counter-defence protein that interferes with Tab function. Thus, our work reveals an anti-phage defence that operates by blocking virion assembly, thereby both preventing formation of phage progeny and allowing destruction of the infected cell due to expression of phage lysis genes.
Collapse
Affiliation(s)
| | | | - Chi Zhang
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
| | - Landon J Getz
- Department of Biochemistry, University of Toronto, Toronto, ON, Canada
| | | | - Alan R Davidson
- Department of Biochemistry, University of Toronto, Toronto, ON, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
| | - Karen L Maxwell
- Department of Biochemistry, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
35
|
Han F, Li H, Lyu E, Zhang Q, Gai H, Xu Y, Bai X, He X, Khan AQ, Li X, Xie F, Li F, Fang X, Wei M. Soybean-mediated suppression of BjaI/BjaR 1 quorum sensing in Bradyrhizobium diazoefficiens impacts symbiotic nitrogen fixation. Appl Environ Microbiol 2024; 90:e0137423. [PMID: 38251894 PMCID: PMC10880635 DOI: 10.1128/aem.01374-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Accepted: 11/23/2023] [Indexed: 01/23/2024] Open
Abstract
The acyl-homoserine lactones (AHLs)-mediated LuxI/LuxR quorum sensing (QS) system orchestrates diverse bacterial behaviors in response to changes in population density. The role of the BjaI/BjaR1 QS system in Bradyrhizobium diazoefficiens USDA 110, which shares homology with LuxI/LuxR, remains elusive during symbiotic interaction with soybean. Here this genetic system in wild-type (WT) bacteria residing inside nodules exhibited significantly reduced activity compared to free-living cells, potentially attributed to soybean-mediated suppression. The deletion mutant strain ΔbjaR1 showed significantly enhanced nodulation induction and nitrogen fixation ability. Nevertheless, its ultimate symbiotic outcome (plant dry weight) in soybeans was compromised. Furthermore, comparative analysis of the transcriptome, proteome, and promoter activity revealed that the inactivation of BjaR1 systematically activated and inhibited genomic modules associated with nodulation and nitrogen metabolism. The former appeared to be linked to a significant decrease in the expression of NodD2, a key cell-density-dependent repressor of nodulation genes, while the latter conferred bacterial growth and nitrogen fixation insensitivity to environmental nitrogen. In addition, BjaR1 exerted a positive influence on the transcription of multiple genes involved in a so-called central intermediate metabolism within the nodule. In conclusion, our findings highlight the crucial role of the BjaI/BjaR1 QS circuit in positively regulating bacterial nitrogen metabolism and emphasize the significance of the soybean-mediated suppression of this genetic system for promoting efficient symbiotic nitrogen fixation by B. diazoefficiens.IMPORTANCEThe present study demonstrates, for the first time, that the BjaI/BjaR1 QS system of Bradyrhizobium diazoefficiens has a significant impact on its nodulation and nitrogen fixation capability in soybean by positively regulating NodD2 expression and bacterial nitrogen metabolism. Moreover, it provides novel insights into the importance of suppressing the activity of this QS circuit by the soybean host plant in establishing an efficient mutual relationship between the two symbiotic partners. This research expands our understanding of legumes' role in modulating symbiotic nitrogen fixation through rhizobial QS-mediated metabolic functioning, thereby deepening our comprehension of symbiotic coevolution theory. In addition, these findings may hold great promise for developing quorum quenching technology in agriculture.
Collapse
Affiliation(s)
- Fang Han
- State Key Laboratory of Herbage Improvement and Grassland Agro-ecosystems, College of Ecology, Lanzhou University, Lanzhou, China
| | - Huiquan Li
- State Key Laboratory of Herbage Improvement and Grassland Agro-ecosystems, College of Ecology, Lanzhou University, Lanzhou, China
| | - Ermeng Lyu
- State Key Laboratory of Herbage Improvement and Grassland Agro-ecosystems, College of Ecology, Lanzhou University, Lanzhou, China
| | - Qianqian Zhang
- State Key Laboratory of Herbage Improvement and Grassland Agro-ecosystems, College of Ecology, Lanzhou University, Lanzhou, China
| | - Haoyu Gai
- State Key Laboratory of Herbage Improvement and Grassland Agro-ecosystems, College of Ecology, Lanzhou University, Lanzhou, China
| | - Yunfang Xu
- State Key Laboratory of Herbage Improvement and Grassland Agro-ecosystems, College of Ecology, Lanzhou University, Lanzhou, China
| | - Xuemei Bai
- State Key Laboratory of Herbage Improvement and Grassland Agro-ecosystems, College of Ecology, Lanzhou University, Lanzhou, China
| | - Xueqian He
- State Key Laboratory of Herbage Improvement and Grassland Agro-ecosystems, College of Ecology, Lanzhou University, Lanzhou, China
| | - Abdul Qadir Khan
- State Key Laboratory of Herbage Improvement and Grassland Agro-ecosystems, College of Ecology, Lanzhou University, Lanzhou, China
| | - Xiaolin Li
- National Key Laboratory of Plant Molecular Genetics, CAS Center for Excellence in Molecular Plant Sciences, Institute of Plant Physiology and Ecology, Chinese Academy of Sciences, Shanghai, China
- University of the Chinese Academy of Sciences, Beijing, China
| | - Fang Xie
- National Key Laboratory of Plant Molecular Genetics, CAS Center for Excellence in Molecular Plant Sciences, Institute of Plant Physiology and Ecology, Chinese Academy of Sciences, Shanghai, China
| | - Fengmin Li
- State Key Laboratory of Herbage Improvement and Grassland Agro-ecosystems, College of Ecology, Lanzhou University, Lanzhou, China
| | - Xiangwen Fang
- State Key Laboratory of Herbage Improvement and Grassland Agro-ecosystems, College of Ecology, Lanzhou University, Lanzhou, China
| | - Min Wei
- State Key Laboratory of Herbage Improvement and Grassland Agro-ecosystems, College of Ecology, Lanzhou University, Lanzhou, China
| |
Collapse
|
36
|
Park JM, Kim JH, Kim G, Sim HJ, Ahn SM, Choi KS, Kwon HJ. Rapid Antibacterial Activity Assessment of Chimeric Lysins. Int J Mol Sci 2024; 25:2430. [PMID: 38397110 PMCID: PMC10888538 DOI: 10.3390/ijms25042430] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Revised: 02/16/2024] [Accepted: 02/18/2024] [Indexed: 02/25/2024] Open
Abstract
Various chimeric lysins have been developed as efficacious antibiotics against multidrug-resistant bacteria, but direct comparisons of their antibacterial activities have been difficult due to the preparation of multiple recombinant chimeric lysins. Previously, we reported an Escherichia coli cell-free expression method to better screen chimeric lysins against Staphylococcus aureus, but we still needed to increase the amounts of expressed proteins enough to be able to detect them non-isotopically for quantity comparisons. In this study, we improved the previous cell-free expression system by adding a previously reported artificial T7 terminator and reversing the different nucleotides between the T7 promoter and start codon to those of the T7 phage. The new method increased the expressed amount of chimeric lysins enough for us to detect them using Western blotting. Therefore, the qualitative comparison of activity between different chimeric lysins has become possible via the adjustment of the number of variables between samples without protein purification. We applied this method to select more active chimeric lysins derived from our previously reported chimeric lysin (ALS2). Finally, we compared the antibacterial activities of our selected chimeric lysins with reported chimeric lysins (ClyC and ClyO) and lysostaphin and determined the rank orders of antibacterial activities on different Staphylococcus aureus strains in our experimental conditions.
Collapse
Affiliation(s)
- Jin-Mi Park
- Laboratory of Poultry Medicine, College of Veterinary Medicine, Seoul National University, Seoul 08826, Republic of Korea; (J.-M.P.); (J.-H.K.); (S.-M.A.)
- College of Veterinary Medicine and BK21 for Veterinary Science, Seoul National University, Seoul 08826, Republic of Korea; (G.K.); (H.-J.S.)
- GeNiner Inc., Seoul 08826, Republic of Korea
| | - Jun-Hyun Kim
- Laboratory of Poultry Medicine, College of Veterinary Medicine, Seoul National University, Seoul 08826, Republic of Korea; (J.-M.P.); (J.-H.K.); (S.-M.A.)
- College of Veterinary Medicine and BK21 for Veterinary Science, Seoul National University, Seoul 08826, Republic of Korea; (G.K.); (H.-J.S.)
- GeNiner Inc., Seoul 08826, Republic of Korea
| | - Gun Kim
- College of Veterinary Medicine and BK21 for Veterinary Science, Seoul National University, Seoul 08826, Republic of Korea; (G.K.); (H.-J.S.)
- Laboratory of Pharmacology, Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul 08826, Republic of Korea
| | - Hun-Ju Sim
- College of Veterinary Medicine and BK21 for Veterinary Science, Seoul National University, Seoul 08826, Republic of Korea; (G.K.); (H.-J.S.)
- Laboratory of Pharmacology, Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul 08826, Republic of Korea
| | - Sun-Min Ahn
- Laboratory of Poultry Medicine, College of Veterinary Medicine, Seoul National University, Seoul 08826, Republic of Korea; (J.-M.P.); (J.-H.K.); (S.-M.A.)
| | - Kang-Seuk Choi
- College of Veterinary Medicine and BK21 for Veterinary Science, Seoul National University, Seoul 08826, Republic of Korea; (G.K.); (H.-J.S.)
- Laboratory of Avian Diseases, College of Veterinary Medicine, Seoul National University, Seoul 08826, Republic of Korea
| | - Hyuk-Joon Kwon
- Laboratory of Poultry Medicine, College of Veterinary Medicine, Seoul National University, Seoul 08826, Republic of Korea; (J.-M.P.); (J.-H.K.); (S.-M.A.)
- College of Veterinary Medicine and BK21 for Veterinary Science, Seoul National University, Seoul 08826, Republic of Korea; (G.K.); (H.-J.S.)
- GeNiner Inc., Seoul 08826, Republic of Korea
| |
Collapse
|
37
|
Klaewkla M, Wangpaiboon K, Pichyangkura R, Charoenwongpaiboon T. Unraveling the role of flexible coil near calcium binding site of levansucrase on thermostability and product profile via proline substitution and molecular dynamics simulations. Proteins 2024; 92:170-178. [PMID: 37753539 DOI: 10.1002/prot.26592] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2023] [Revised: 08/18/2023] [Accepted: 09/07/2023] [Indexed: 09/28/2023]
Abstract
Due to its bioactivity and versatile applications, levan has appeared as a promising biomaterial. Levansucrase is responsible for the conversion of sucrose into levan. With the goal of enhancing levan production, the strategy for enhancing the stability of levansucrase is being intensively studied. To make proteins more stable under high temperatures, proline, the most rigid residue, can be introduced into previously flexible regions. Herein, G249, D250, N251, and H252 on the flexible coil close to the calcium binding site of Bacillus licheniformis levansucrase were replaced with proline. Mutations at G249P greatly enhance both the enzyme's thermodynamic and kinetic stability, while those at H252P improve solely the enzyme's kinetic stability. GPC analysis revealed that G249P synthesize more levan, but H252P generate primarily oligosaccharides. Molecular dynamics simulations (MD) and MM/GBSA analysis revealed that G249P mutation increased not only the stability of levansucrase, but also affinity toward fructan.
Collapse
Affiliation(s)
| | - Karan Wangpaiboon
- Department of Biochemistry, Faculty of Science, Chulalongkorn University, Bangkok, Thailand
| | - Rath Pichyangkura
- Department of Biochemistry, Faculty of Science, Chulalongkorn University, Bangkok, Thailand
| | | |
Collapse
|
38
|
Clavé C, Dheur S, Ament-Velásquez SL, Granger-Farbos A, Saupe SJ. het-B allorecognition in Podospora anserina is determined by pseudo-allelic interaction of genes encoding a HET and lectin fold domain protein and a PII-like protein. PLoS Genet 2024; 20:e1011114. [PMID: 38346076 PMCID: PMC10890737 DOI: 10.1371/journal.pgen.1011114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Revised: 02/23/2024] [Accepted: 01/29/2024] [Indexed: 02/25/2024] Open
Abstract
Filamentous fungi display allorecognition genes that trigger regulated cell death (RCD) when strains of unlike genotype fuse. Podospora anserina is one of several model species for the study of this allorecognition process termed heterokaryon or vegetative incompatibility. Incompatibility restricts transmission of mycoviruses between isolates. In P. anserina, genetic analyses have identified nine incompatibility loci, termed het loci. Here we set out to clone the genes controlling het-B incompatibility. het-B displays two incompatible alleles, het-B1 and het-B2. We find that the het-B locus encompasses two adjacent genes, Bh and Bp that exist as highly divergent allelic variants (Bh1/Bh2 and Bp1/Bp2) in the incompatible haplotypes. Bh encodes a protein with an N-terminal HET domain, a cell death inducing domain bearing homology to Toll/interleukin-1 receptor (TIR) domains and a C-terminal domain with a predicted lectin fold. The Bp product is homologous to PII-like proteins, a family of small trimeric proteins acting as sensors of adenine nucleotides in bacteria. We show that although the het-B system appears genetically allelic, incompatibility is in fact determined by the non-allelic Bh1/Bp2 interaction while the reciprocal Bh2/Bp1 interaction plays no role in incompatibility. The highly divergent C-terminal lectin fold domain of BH determines recognition specificity. Population studies and genome analyses indicate that het-B is under balancing selection with trans-species polymorphism, highlighting the evolutionary significance of the two incompatible haplotypes. In addition to emphasizing anew the central role of TIR-like HET domains in fungal RCD, this study identifies novel players in fungal allorecognition and completes the characterization of the entire het gene set in that species.
Collapse
Affiliation(s)
- Corinne Clavé
- IBGC, UMR 5095, CNRS-Université de Bordeaux, Bordeaux, France
| | - Sonia Dheur
- IBGC, UMR 5095, CNRS-Université de Bordeaux, Bordeaux, France
| | | | | | - Sven J. Saupe
- IBGC, UMR 5095, CNRS-Université de Bordeaux, Bordeaux, France
| |
Collapse
|
39
|
Ahuja JS, Sandhu R, Huang L, Klein F, Börner GV. Temporal and Functional Relationship between Synaptonemal Complex Morphogenesis and Recombination during Meiosis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.11.575218. [PMID: 38260343 PMCID: PMC10802607 DOI: 10.1101/2024.01.11.575218] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2024]
Abstract
During prophase of meiosis I, programmed double strand breaks (DSBs) are processed into crossovers, a critical requirement for segregation of homologous chromosomes (homologs) and genome haploidization in sexually reproducing organisms. Crossovers form via homologous recombination in close temporospatial association with morphogenesis of the synaptonemal complex (SC), a proteinaceous structure that connects paired homologs along their length during the pachytene stage. Synapsis and recombination are a paradigm for the interplay between higher order chromosome structure and DNA metabolism, yet their temporal and functional relationship remains poorly understood. Probing linkage between these processes in budding yeast, we show that SC assembly is associated with a distinct threshold number of unstable D-loops. The transition from bona fide paranemic D-loops to plectonemic DSB single end invasions (SEIs) is completed during midpachynema, when the SC is fully assembled. Double Holliday junctions (dHJs) form at the time of desynapsis and are resolved into crossovers during diplonema. The SC central element component Zip1 shepherds recombination through three transitions, including DSB first end strand exchange and second end capture, as well as dHJ resolution. Zip1 mediates SEI formation independent of its polymerization whereas precocious Zip1 assembly interferes with double Holliday junction resolution. Together, our findings indicate that the synaptonemal complex controls recombination while assembled but also beyond its disassembly, possibly by establishing spatial constraints at recombination sites.
Collapse
|
40
|
Charoenwongpaiboon T, Sommanat N, Wangpaiboon K, Puangpathanachai M, Pongsawasdi P, Pichyangkura R. Improving the thermostability and modulating the inulin profile of inulosucrase through rational glycine-to-proline substitution. RSC Adv 2024; 14:2346-2353. [PMID: 38213970 PMCID: PMC10782431 DOI: 10.1039/d3ra06896j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Accepted: 12/21/2023] [Indexed: 01/13/2024] Open
Abstract
The flexibility of protein structure plays a crucial role in enzyme stability and catalysis. Among the amino acids, glycine is particularly important in conferring flexibility to proteins. In this study, the effects of flexible glycine residues in Lactobacillus reuteri 121 inulosucrase (LrInu) on stability and inulin profile were investigated through glycine-to-proline substitutions. Molecular dynamics (MD) simulations were employed to discover the flexible glycine residues, and eight glycine residues, including Gly217, Gly298, Gly330, Gly416, Gly450, Gly624, Gly627, Gly629, were selected for site-directed mutagenesis. The results demonstrated significant changes in both thermostability and inulin profiles of the variants. Particularly, the G624P and G627P variants showed reduced production of long-chain oligosaccharides compared to the WT. This can be ascribed to the increased rigidity of the active site, which is crucial for the induction-fit mechanism. Overall, this study provides valuable insights into the role of flexible glycine residues in the activity, stability, and inulin synthesis of LrInu.
Collapse
Affiliation(s)
| | - Nawapat Sommanat
- Department of Chemistry, Faculty of Science, Silpakorn University Nakhon Pathom 73000 Thailand
| | - Karan Wangpaiboon
- Department of Biochemistry, Faculty of Science, Chulalongkorn University Bangkok 10330 Thailand
- Center of Excellence in Structural and Computational Biology, Department of Biochemistry, Faculty of Science, Chulalongkorn University Bangkok Thailand
| | | | - Piamsook Pongsawasdi
- Department of Biochemistry, Faculty of Science, Chulalongkorn University Bangkok 10330 Thailand
| | - Rath Pichyangkura
- Department of Biochemistry, Faculty of Science, Chulalongkorn University Bangkok 10330 Thailand
| |
Collapse
|
41
|
Tanabe T, Tsukamoto M, Shioda M, Nagaoka K, Funahashi T. Expression regulation of type III secretion system 2 in Vibrio parahaemolyticus by catabolite activator protein. FEMS Microbiol Lett 2024; 371:fnae054. [PMID: 39054297 DOI: 10.1093/femsle/fnae054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Revised: 06/23/2024] [Accepted: 07/24/2024] [Indexed: 07/27/2024] Open
Abstract
Vibrio parahaemolyticus has two sets of type III secretion systems that are major pathogenic factors: T3SS1 (cytotoxicity) and T3SS2 (enterotoxicity). V. parahaemolyticus mainly colonizes the distal small intestine after oral infection and may be exposed to carbon-limiting stress due to the lack of readily available carbohydrates in this environment. Catabolite activator protein (CAP), a transcription factor involved in carbon-limiting metabolism in many Gram-negative bacteria, is well known to be involved in the regulation of the expression of many virulence factors. In this study, we determined the effects of CAP on the expression of T3SSs in this bacterium. Based on a lactate dehydrogenase-based cytotoxicity assay, CAP was found to have a greater contribution to the expression of T3SS2-dependent cytotoxicity than to that of T3SS1. Reverse transcription quantitative PCR revealed decreased expression of many T3SS2-related genes, including vpa1348, in the cap gene deletion mutant compared to the parent strain. CAP was demonstrated to bind near the T-rich elements within the vpa1348 promoter region in an electrophoretic mobility shift assay and DNase I footprinting. CAP also enhanced the expression of vpa1348 in a β-galactosidase reporter assay. Collectively, these results suggest that CAP is involved in T3SS2-mediated virulence by regulating the expression of vpa1348 in V. parahaemolyticus.
Collapse
Affiliation(s)
- Tomotaka Tanabe
- Laboratory of Hygienic Chemistry, College of Pharmaceutical Sciences, Matsuyama University, Matsuyama, Ehime 790-8578, Japan
| | - Mitsuki Tsukamoto
- Laboratory of Hygienic Chemistry, College of Pharmaceutical Sciences, Matsuyama University, Matsuyama, Ehime 790-8578, Japan
| | - Mahiro Shioda
- Laboratory of Hygienic Chemistry, College of Pharmaceutical Sciences, Matsuyama University, Matsuyama, Ehime 790-8578, Japan
| | - Kenjiro Nagaoka
- Laboratory of Hygienic Chemistry, College of Pharmaceutical Sciences, Matsuyama University, Matsuyama, Ehime 790-8578, Japan
| | - Tatsuya Funahashi
- Laboratory of Hygienic Chemistry, College of Pharmaceutical Sciences, Matsuyama University, Matsuyama, Ehime 790-8578, Japan
| |
Collapse
|
42
|
Hamilton CD, Zaricor B, Dye CJ, Dresserl E, Michaels R, Allen C. Ralstonia solanacearum pandemic lineage strain UW551 overcomes inhibitory xylem chemistry to break tomato bacterial wilt resistance. MOLECULAR PLANT PATHOLOGY 2024; 25:e13395. [PMID: 37846613 PMCID: PMC10782650 DOI: 10.1111/mpp.13395] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Revised: 08/01/2023] [Accepted: 09/15/2023] [Indexed: 10/18/2023]
Abstract
Plant-pathogenic Ralstonia strains cause bacterial wilt disease by colonizing xylem vessels of many crops, including tomato. Host resistance is the best control for bacterial wilt, but resistance mechanisms of the widely used Hawaii 7996 tomato breeding line (H7996) are unknown. Using growth in ex vivo xylem sap as a proxy for host xylem, we found that Ralstonia strain GMI1000 grows in sap from both healthy plants and Ralstonia-infected susceptible plants. However, sap from Ralstonia-infected H7996 plants inhibited Ralstonia growth, suggesting that in response to Ralstonia infection, resistant plants increase inhibitors in their xylem sap. Consistent with this, reciprocal grafting and defence gene expression experiments indicated that H7996 wilt resistance acts in both above- and belowground plant parts. Concerningly, H7996 resistance is broken by Ralstonia strain UW551 of the pandemic lineage that threatens highland tropical agriculture. Unlike other Ralstonia, UW551 grew well in sap from Ralstonia-infected H7996 plants. Moreover, other Ralstonia strains could grow in sap from H7996 plants previously infected by UW551. Thus, UW551 overcomes H7996 resistance in part by detoxifying inhibitors in xylem sap. Testing a panel of xylem sap compounds identified by metabolomics revealed that no single chemical differentially inhibits Ralstonia strains that cannot infect H7996. However, sap from Ralstonia-infected H7996 contained more phenolic compounds, which are known to be involved in plant antimicrobial defence. Culturing UW551 in this sap reduced total phenolic levels, indicating that the resistance-breaking Ralstonia strain degrades these chemical defences. Together, these results suggest that H7996 tomato wilt resistance depends in part on inducible phenolic compounds in xylem sap.
Collapse
Affiliation(s)
- Corri D. Hamilton
- Department of Plant PathologyUniversity of Wisconsin MadisonMadisonWisconsinUSA
- Department of Microbiology and ImmunologyUniversity of British ColumbiaVancouverBritish ColumbiaCanada
| | - Beatriz Zaricor
- Department of Plant PathologyUniversity of Wisconsin MadisonMadisonWisconsinUSA
| | - Carolyn Jean Dye
- Department of Plant PathologyUniversity of Wisconsin MadisonMadisonWisconsinUSA
| | - Emma Dresserl
- Department of Plant PathologyUniversity of Wisconsin MadisonMadisonWisconsinUSA
| | - Renee Michaels
- Department of Plant PathologyUniversity of Wisconsin MadisonMadisonWisconsinUSA
| | - Caitilyn Allen
- Department of Plant PathologyUniversity of Wisconsin MadisonMadisonWisconsinUSA
| |
Collapse
|
43
|
Mishra V, Sharma K, Bose A, Maisonneuve P, Visweswariah SS. The evolutionary divergence of receptor guanylyl cyclase C has implications for preclinical models for receptor-directed therapeutics. J Biol Chem 2024; 300:105505. [PMID: 38029963 PMCID: PMC7615481 DOI: 10.1016/j.jbc.2023.105505] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Revised: 11/16/2023] [Accepted: 11/20/2023] [Indexed: 12/01/2023] Open
Abstract
Mutations in receptor guanylyl cyclase C (GC-C) cause severe gastrointestinal disease, including meconium ileus, early onset acute diarrhea, and pediatric inflammatory bowel disease that continues into adulthood. Agonists of GC-C are US Food and Drug Administration-approved drugs for the treatment of constipation and irritable bowel syndrome. Therapeutic strategies targeting GC-C are tested in preclinical mouse models, assuming that murine GC-C mimics human GC-C in its biochemical properties and downstream signaling events. Here, we reveal important differences in ligand-binding affinity and GC activity between mouse GC-C and human GC-C. We generated a series of chimeric constructs of various domains of human and mouse GC-C to show that the extracellular domain of mouse GC-C contributed to log-orders lower affinity of mouse GC-C for ligands than human GC-C. Further, the Vmax of the murine GC domain was lower than that of human GC-C, and allosteric regulation of the receptor by ATP binding to the intracellular kinase-homology domain also differed. These altered properties are reflected in the high concentrations of ligands required to elicit signaling responses in the mouse gut in preclinical models and the specificity of a GC inhibitor towards human GC-C. Therefore, our studies identify considerations in using the murine model to test molecules for therapeutic purposes that work as either agonists or antagonists of GC-C, and vaccines for the bacterial heat-stable enterotoxin that causes watery diarrhea in humans.
Collapse
Affiliation(s)
- Vishwas Mishra
- Department of Developmental Biology and Genetics, Indian Institute of Science, Bengaluru, India
| | - Kritica Sharma
- Department of Developmental Biology and Genetics, Indian Institute of Science, Bengaluru, India
| | - Avipsa Bose
- Department of Developmental Biology and Genetics, Indian Institute of Science, Bengaluru, India
| | - Pierre Maisonneuve
- UMR 5248 - Chemistry & Biology of Membranes and Nano-Objects, CNRS - Université de Bordeaux, Institut Européen de Chimie et Biologie, Pessac, France
| | - Sandhya S Visweswariah
- Department of Developmental Biology and Genetics, Indian Institute of Science, Bengaluru, India.
| |
Collapse
|
44
|
Charoenwongpaiboon T, Wangpaiboon K, Puangpathanachai M, Pongsawasdi P, Pichyangkura R. Energy- and evolution-based design of inulosucrase for enhanced thermostability and inulin production. Appl Microbiol Biotechnol 2023; 107:6831-6843. [PMID: 37688600 DOI: 10.1007/s00253-023-12759-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Revised: 07/31/2023] [Accepted: 08/30/2023] [Indexed: 09/11/2023]
Abstract
Inulosucrase from Lactobacillus reuteri 121 (LrInu) exhibits promise in the synthesis of prebiotic inulin and fructooligosaccharides. However, for its use in industry, LrInu's thermostability is a crucial consideration. In this study, the computational program FireProt was used to predict the thermostable variants of LrInu. Using rational criteria, nine variants were selected for protein expression and characterization. The G237P variant was determined to be the greatest designed candidate due to its greatly enhanced stability and activity in comparison to the wild-type enzyme. The optimum temperature of G237P increased from 50 to 60°C, with an over 5-fold increase in the half-life. Spectroscopy studies revealed that the G237P mutation could prevent the structural change in LrInu caused by heat or urea treatment. Molecular dynamics (MD) simulations showed that the enhanced thermostability of the G237P variant resulted from an increase in structural rigidity and the number of native contacts within the protein molecule. In addition, G237P variant synthesizes inulin with greater efficiency than WT. KEY POINTS: • Thermostable inulosucrase variant(s) were designed by Fireprot server. • G237P variant showed significantly improved thermostability compared to the wild type. • Inulin is synthesized more efficiently by G237P variant.
Collapse
Affiliation(s)
| | - Karan Wangpaiboon
- Department of Biochemistry, Faculty of Science, Chulalongkorn University, Bangkok, 10330, Thailand
| | | | - Piamsook Pongsawasdi
- Department of Biochemistry, Faculty of Science, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Rath Pichyangkura
- Department of Biochemistry, Faculty of Science, Chulalongkorn University, Bangkok, 10330, Thailand
| |
Collapse
|
45
|
Hong X, Guo T, Xu X, Lin J. Multiplex metabolic pathway engineering of Monascus pilosus enhances lovastatin production. Appl Microbiol Biotechnol 2023; 107:6541-6552. [PMID: 37672068 DOI: 10.1007/s00253-023-12747-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Revised: 07/30/2023] [Accepted: 08/11/2023] [Indexed: 09/07/2023]
Abstract
Monascus sp. is an important food microbial resource with the production of cholesterol-lowering agent lovastatin and other healthy metabolites. However, the mycotoxin citrinin naturally produced by Monascus sp. and the insufficient productivity of lovastatin limit its large-scale use in food industry. The aim of this paper is to modify a lovastatin-producing strain Monascus pilosus GN-01 through metabolic engineering to obtain a citrinin-free M. pilosus strain with higher yield of lovastatin. The citrinin synthesis regulator gene ctnR was firstly disrupted to obtain GN-02 without citrinin production. Based on that, the lovastatin biosynthesis genes (mokC, mokD, mokE, mokF, mokH, mokI, and LaeA) were, respectively, overexpressed, and pigment-regulatory gene (pigR) was knocked out to improve lovastatin production. The results indicated ctnR inactivation effectively disrupted the citrinin release by M. pilosus GN-01. The overexpression of lovastatin biosynthesis genes and pigR knockout could lead higher contents of lovastatin, of which pigR knockout strain achieved 76.60% increase in the yield of lovastatin compared to GN-02. These studies suggest that such multiplex metabolic pathway engineering in M. pilosus GN-01 is promising for high lovastatin production by a safe strain for application in Monascus-related food. KEY POINTS: • Disruption of the regulator gene ctnR inhibited citrinin production of M. pilosus. • Synchronous overexpression of biosynthesis gene enhanced lovastatin production. • pigR knockout enhanced lovastatin of ΔctnR strain of M. pilosus.
Collapse
Affiliation(s)
- Xiaokun Hong
- College of Biological Science and Engineering, Fuzhou University, Fuzhou, 350116, Fujian, China
| | - Tianlong Guo
- College of Biological Science and Engineering, Fuzhou University, Fuzhou, 350116, Fujian, China
| | - Xinqi Xu
- College of Biological Science and Engineering, Fuzhou University, Fuzhou, 350116, Fujian, China.
| | - Juan Lin
- College of Biological Science and Engineering, Fuzhou University, Fuzhou, 350116, Fujian, China.
| |
Collapse
|
46
|
Bukhari M, Patel N, Fontana R, Santiago-Medina M, Jiang Y, Li D, Pestonjamasp K, Christiansen VJ, Jackson KW, McKee PA, Yang J. Fibroblast activation protein drives tumor metastasis via a protease-independent role in invadopodia stabilization. Cell Rep 2023; 42:113302. [PMID: 37862167 PMCID: PMC10742343 DOI: 10.1016/j.celrep.2023.113302] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 08/09/2023] [Accepted: 10/03/2023] [Indexed: 10/22/2023] Open
Abstract
During metastasis, tumor cells invade through the basement membrane and intravasate into blood vessels and then extravasate into distant organs to establish metastases. Here, we report a critical role of a transmembrane serine protease fibroblast activation protein (FAP) in tumor metastasis. Expression of FAP and TWIST1, a metastasis driver, is significantly correlated in several types of human carcinomas, and FAP is required for TWIST1-induced breast cancer metastasis to the lung. Mechanistically, FAP is localized at invadopodia and required for invadopodia-mediated extracellular matrix degradation independent of its proteolytic activity. Live cell imaging shows that association of invadopodia precursors with FAP at the cell membrane promotes the stabilization and growth of invadopodia precursors into mature invadopodia. Together, our study identified FAP as a functional target of TWIST1 in driving tumor metastasis via promoting invadopodia-mediated matrix degradation and uncovered a proteolytic activity-independent role of FAP in stabilizing invadopodia precursors for maturation.
Collapse
Affiliation(s)
- Maurish Bukhari
- Department of Pharmacology, University of California, San Diego, School of Medicine, La Jolla, CA 92093, USA; Moores Cancer Center, University of California, San Diego, School of Medicine, La Jolla, CA 92093, USA
| | - Navneeta Patel
- Department of Pharmacology, University of California, San Diego, School of Medicine, La Jolla, CA 92093, USA; Moores Cancer Center, University of California, San Diego, School of Medicine, La Jolla, CA 92093, USA
| | - Rosa Fontana
- Department of Pharmacology, University of California, San Diego, School of Medicine, La Jolla, CA 92093, USA; Moores Cancer Center, University of California, San Diego, School of Medicine, La Jolla, CA 92093, USA
| | - Miguel Santiago-Medina
- Department of Pharmacology, University of California, San Diego, School of Medicine, La Jolla, CA 92093, USA; Moores Cancer Center, University of California, San Diego, School of Medicine, La Jolla, CA 92093, USA
| | - Yike Jiang
- Department of Pharmacology, University of California, San Diego, School of Medicine, La Jolla, CA 92093, USA; Moores Cancer Center, University of California, San Diego, School of Medicine, La Jolla, CA 92093, USA
| | - Dongmei Li
- Department of Pharmacology, University of California, San Diego, School of Medicine, La Jolla, CA 92093, USA; Moores Cancer Center, University of California, San Diego, School of Medicine, La Jolla, CA 92093, USA
| | - Kersi Pestonjamasp
- Moores Cancer Center, University of California, San Diego, School of Medicine, La Jolla, CA 92093, USA
| | - Victoria J Christiansen
- William K. Warren Medical Research Center, Department of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Kenneth W Jackson
- William K. Warren Medical Research Center, Department of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Patrick A McKee
- William K. Warren Medical Research Center, Department of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Jing Yang
- Department of Pharmacology, University of California, San Diego, School of Medicine, La Jolla, CA 92093, USA; Moores Cancer Center, University of California, San Diego, School of Medicine, La Jolla, CA 92093, USA; Department of Pediatrics, University of California, San Diego, School of Medicine, La Jolla, CA 92093, USA.
| |
Collapse
|
47
|
Xu W, Ren Y, Xia Y, Liu L, Meng X, Chen G, Zhang W, Liu W. A novel transcriptional repressor specifically regulates xylanase gene 1 in Trichoderma reesei. BIOTECHNOLOGY FOR BIOFUELS AND BIOPRODUCTS 2023; 16:161. [PMID: 37891680 PMCID: PMC10612264 DOI: 10.1186/s13068-023-02417-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/19/2023] [Accepted: 10/20/2023] [Indexed: 10/29/2023]
Abstract
BACKGROUND The well-known industrial fungus Trichoderma reesei has an excellent capability of secreting a large amount of cellulases and xylanases. The induced expression of cellulase and xylanase genes is tightly controlled at the transcriptional level. However, compared to the intensive studies on the intricate regulatory mechanism of cellulase genes, efforts to understand how xylanase genes are regulated are relatively limited, which impedes the further improvement of xylanase production by T. reesei via rational strain engineering. RESULTS To identify transcription factors involved in regulating xylanase gene expression in T. reesei, yeast one-hybrid screen was performed based on the promoters of two major extracellular xylanase genes xyn1 and xyn2. A putative transcription factor named XTR1 showing significant binding capability to the xyn1 promoter but not that of xyn2, was successfully isolated. Deletion of xtr1 significantly increased the transcriptional level of xyn1, but only exerted a minor promoting effect on that of xyn2. The xylanase activity was increased by ~ 50% with XTR1 elimination but the cellulase activity was hardly affected. Subcellular localization analysis of XTR1 fused to a green fluorescence protein demonstrated that XTR1 is a nuclear protein. Further analyses revealed the precise binding site of XTR1 and nucleotides critical for the binding within the xyn1 promoter. Moreover, competitive EMSAs indicated that XTR1 competes with the essential transactivator XYR1 for binding to the xyn1 promoter. CONCLUSIONS XTR1 represents a new transcriptional repressor specific for controlling xylanase gene expression. Isolation and functional characterization of this new factor not only contribute to further understanding the stringent regulatory network of xylanase genes, but also provide important clues for boosting xylanase biosynthesis in T. reesei.
Collapse
Affiliation(s)
- Wenqiang Xu
- State Key Laboratory of Microbial Technology, Shandong University, No.72 Binhai Road, Qingdao, 266237, People's Republic of China
- Shandong Lishan Biotechnology Co., LTD, Jinan, China
| | - Yajing Ren
- State Key Laboratory of Microbial Technology, Shandong University, No.72 Binhai Road, Qingdao, 266237, People's Republic of China
| | - Yuxiao Xia
- State Key Laboratory of Microbial Technology, Shandong University, No.72 Binhai Road, Qingdao, 266237, People's Republic of China
| | - Lin Liu
- State Key Laboratory of Microbial Technology, Shandong University, No.72 Binhai Road, Qingdao, 266237, People's Republic of China
| | - Xiangfeng Meng
- State Key Laboratory of Microbial Technology, Shandong University, No.72 Binhai Road, Qingdao, 266237, People's Republic of China
| | - Guanjun Chen
- State Key Laboratory of Microbial Technology, Shandong University, No.72 Binhai Road, Qingdao, 266237, People's Republic of China
| | - Weixin Zhang
- State Key Laboratory of Microbial Technology, Shandong University, No.72 Binhai Road, Qingdao, 266237, People's Republic of China.
| | - Weifeng Liu
- State Key Laboratory of Microbial Technology, Shandong University, No.72 Binhai Road, Qingdao, 266237, People's Republic of China
| |
Collapse
|
48
|
Xu Y, Cohen E, Johnson CN, Parent CA, Coulombe PA. Keratin 17- and PKCα-dependent transient amplification of neutrophil influx after repeated stress to the skin. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.11.561954. [PMID: 37873256 PMCID: PMC10592713 DOI: 10.1101/2023.10.11.561954] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/25/2023]
Abstract
Neutrophils contribute to the pathogenesis of chronic inflammatory skin diseases. Little is known about the source and identity of the signals mediating their recruitment in inflamed skin. We used the phorbol ester TPA and UVB, alone or in combination, to induce sterile inflammation in mouse skin and assess whether keratinocyte-derived signals impact neutrophil recruitment. A single TPA treatment results in a neutrophil influx in the dermis that peaks at 12h and resolves within 24h. A second TPA treatment or a UVB challenge, when applied at 24h but not 48h later, accelerates, amplifies, and prolongs neutrophil infiltration. This transient amplification response (TAR) is mediated by local signals in inflamed skin, can be recapitulated in ex vivo culture, and involves the K17-dependent sustainment of protein kinase Cα (PKCα) activity and release of neutrophil chemoattractants by stressed keratinocytes. We show that K17 binds RACK1, a scaffold essential for PKCα activity. Finally, analyses of RNAseq data reveal the presence of a transcriptomic signature consistent with TAR and PKCα activation in chronic inflammatory skin diseases. These findings uncover a novel, transient, and keratin-dependent mechanism that amplifies neutrophil recruitment to the skin under stress, with direct implications for inflammatory skin disorders.
Collapse
|
49
|
Sammon D, Krueger A, Busse-Wicher M, Morgan RM, Haslam SM, Schumann B, Briggs DC, Hohenester E. Molecular mechanism of decision-making in glycosaminoglycan biosynthesis. Nat Commun 2023; 14:6425. [PMID: 37828045 PMCID: PMC10570366 DOI: 10.1038/s41467-023-42236-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Accepted: 09/27/2023] [Indexed: 10/14/2023] Open
Abstract
Two major glycosaminoglycan types, heparan sulfate (HS) and chondroitin sulfate (CS), control many aspects of development and physiology in a type-specific manner. HS and CS are attached to core proteins via a common linker tetrasaccharide, but differ in their polymer backbones. How core proteins are specifically modified with HS or CS has been an enduring mystery. By reconstituting glycosaminoglycan biosynthesis in vitro, we establish that the CS-initiating N-acetylgalactosaminyltransferase CSGALNACT2 modifies all glycopeptide substrates equally, whereas the HS-initiating N-acetylglucosaminyltransferase EXTL3 is selective. Structure-function analysis reveals that acidic residues in the glycopeptide substrate and a basic exosite in EXTL3 are critical for specifying HS biosynthesis. Linker phosphorylation by the xylose kinase FAM20B accelerates linker synthesis and initiation of both HS and CS, but has no effect on the subsequent polymerisation of the backbone. Our results demonstrate that modification with CS occurs by default and must be overridden by EXTL3 to produce HS.
Collapse
Affiliation(s)
- Douglas Sammon
- Department of Life Sciences, Imperial College London, London, SW7 2AZ, UK
| | - Anja Krueger
- Department of Life Sciences, Imperial College London, London, SW7 2AZ, UK
| | - Marta Busse-Wicher
- Department of Life Sciences, Imperial College London, London, SW7 2AZ, UK
- Abzena, Babraham Research Campus, Cambridge, CB22 3AT, UK
| | - Rhodri Marc Morgan
- Department of Life Sciences, Imperial College London, London, SW7 2AZ, UK
- ZoBio, 2333 CH, Leiden, Netherlands
| | - Stuart M Haslam
- Department of Life Sciences, Imperial College London, London, SW7 2AZ, UK
| | - Benjamin Schumann
- Department of Chemistry, Imperial College London, London, W12 0BZ, UK
- Chemical Glycobiology Laboratory, The Francis Crick Institute, London, NW1 1AT, UK
| | - David C Briggs
- Department of Life Sciences, Imperial College London, London, SW7 2AZ, UK.
- Signalling and Structural Biology Laboratory, The Francis Crick Institute, London, NW1 1AT, UK.
| | - Erhard Hohenester
- Department of Life Sciences, Imperial College London, London, SW7 2AZ, UK.
| |
Collapse
|
50
|
St-Jacques AD, Rodriguez JM, Eason MG, Foster SM, Khan ST, Damry AM, Goto NK, Thompson MC, Chica RA. Computational remodeling of an enzyme conformational landscape for altered substrate selectivity. Nat Commun 2023; 14:6058. [PMID: 37770431 PMCID: PMC10539519 DOI: 10.1038/s41467-023-41762-0] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2023] [Accepted: 09/13/2023] [Indexed: 09/30/2023] Open
Abstract
Structural plasticity of enzymes dictates their function. Yet, our ability to rationally remodel enzyme conformational landscapes to tailor catalytic properties remains limited. Here, we report a computational procedure for tuning conformational landscapes that is based on multistate design of hinge-mediated domain motions. Using this method, we redesign the conformational landscape of a natural aminotransferase to preferentially stabilize a less populated but reactive conformation and thereby increase catalytic efficiency with a non-native substrate, resulting in altered substrate selectivity. Steady-state kinetics of designed variants reveals activity increases with the non-native substrate of approximately 100-fold and selectivity switches of up to 1900-fold. Structural analyses by room-temperature X-ray crystallography and multitemperature nuclear magnetic resonance spectroscopy confirm that conformational equilibria favor the target conformation. Our computational approach opens the door to targeted alterations of conformational states and equilibria, which should facilitate the design of biocatalysts with customized activity and selectivity.
Collapse
Affiliation(s)
- Antony D St-Jacques
- Department of Chemistry and Biomolecular Sciences, University of Ottawa, Ottawa, ON, K1N 6N5, Canada
- Center for Catalysis Research and Innovation, University of Ottawa, Ottawa, ON, K1N 6N5, Canada
| | - Joshua M Rodriguez
- Department of Chemistry and Biochemistry, University of California, Merced, Merced, CA, 95343, USA
| | - Matthew G Eason
- Department of Chemistry and Biomolecular Sciences, University of Ottawa, Ottawa, ON, K1N 6N5, Canada
- Center for Catalysis Research and Innovation, University of Ottawa, Ottawa, ON, K1N 6N5, Canada
| | - Scott M Foster
- Department of Chemistry and Biomolecular Sciences, University of Ottawa, Ottawa, ON, K1N 6N5, Canada
- Center for Catalysis Research and Innovation, University of Ottawa, Ottawa, ON, K1N 6N5, Canada
| | - Safwat T Khan
- Department of Chemistry and Biomolecular Sciences, University of Ottawa, Ottawa, ON, K1N 6N5, Canada
- Center for Catalysis Research and Innovation, University of Ottawa, Ottawa, ON, K1N 6N5, Canada
| | - Adam M Damry
- Department of Chemistry and Biomolecular Sciences, University of Ottawa, Ottawa, ON, K1N 6N5, Canada
- Center for Catalysis Research and Innovation, University of Ottawa, Ottawa, ON, K1N 6N5, Canada
| | - Natalie K Goto
- Department of Chemistry and Biomolecular Sciences, University of Ottawa, Ottawa, ON, K1N 6N5, Canada
- Center for Catalysis Research and Innovation, University of Ottawa, Ottawa, ON, K1N 6N5, Canada
| | - Michael C Thompson
- Department of Chemistry and Biochemistry, University of California, Merced, Merced, CA, 95343, USA
| | - Roberto A Chica
- Department of Chemistry and Biomolecular Sciences, University of Ottawa, Ottawa, ON, K1N 6N5, Canada.
- Center for Catalysis Research and Innovation, University of Ottawa, Ottawa, ON, K1N 6N5, Canada.
| |
Collapse
|