201
|
Saini N, Aamir M, Khan ZA, Singh VK, Sah P, Mona S. Deciphering Toxic Pollutants Breakdown Potential in Microbial Community of Chumathang Hot Spring, Ladakh, India via Shotgun Metagenome Sequencing. Curr Microbiol 2024; 81:430. [PMID: 39467883 DOI: 10.1007/s00284-024-03915-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Accepted: 09/18/2024] [Indexed: 10/30/2024]
Abstract
Persistent Organic Pollutants (POPs) have been in focus of research due to their massive contamination of environment and bio-accumulation. Bioremediation and high-throughput research have gained momentum to curb the harmful effects of POPs. The present research has explored the microbial diversity of Chumathang Hot Spring, Ladakh, India, through Illumina metagenomic HiSeq 4000 sequencing platform and their potential to degrade persistent pollutants, especially xenobiotics. Taxonomic characterization based on raw metagenomic data illuminated the abundance of members of Pseudomonadota and Actinomyceota. The re-construction of the microbial genomes from assembled contigs and scaffolds using de novo assembler metaSPAdes and their further annotation through contig alignment with available reference genomes elucidated the landscape of the hot spring's microbes. The predominantly occupied key genera reported were Pannonibacter and Novosphingobium. Comparative genomic analysis established evolutionary relationships and functional diversities among hot spring microbial communities. The function annotation through MG-RAST has revealed their metabolic versatility of degrading a wide array of xenobiotic compounds, including caprolactam, dioxin, chlorobenzene, benzoate, and. Further, the hydroxylating dioxygenase (Saro_3901) was identified as a pivotal component in the aromatic degradation pathways, showcasing extensive metabolic interconnectivity. Interestingly, protein interaction network analysis identified hub genes like Saro_1233 (protocatechuate 4,5-dioxygenase alpha subunit), while Saro_3057 (amidase) was noted for its critical role in network communication and control. The resilience of thermal ecosystems, evidenced by robust enzymatic activity and degradation capability among organisms with < 95% genetic similarity, underscores their potential for industrial and bioremediation exploration, emphasizing the importance of preserving and studying biodiverse habitats.
Collapse
Affiliation(s)
- Neha Saini
- Department of Environmental Science & Engineering, Guru Jambheshwar University of Science & Technology, Hisar, Haryana, 125001, India
| | - Mohd Aamir
- Division of Plant Pathology, ICAR- Indian Agricultural Research Institute, Pusa, New Delhi, 110012, India
- Department of Life Sciences, Parul Institute of Applied Sciences, Parul University, Vadodara, Gujarat, 391760, India
| | - Zainul Abdeen Khan
- Division of Plant Pathology, Advanced Centre for Plant Virology, ICAR-Indian Agricultural Research Institute, New Delhi, 110012, India
| | - Vinay Kumar Singh
- Centre for Bioinformatics, School of Biotechnology, Banaras Hindu University, Varanasi, 221005, India
| | - Pankaj Sah
- Applied Sciences Department, College of Applied Sciences and Pharmacy, University of Technology and Applied Sciences, Al Khuwair, PO Box 74, Muscat, 133, Sultanate of Oman
| | - Sharma Mona
- Department of Environmental Studies, School of Interdisciplinary and Applied Sciences, Central University of Haryana, Mahendergarh, Haryana, 123031, India.
| |
Collapse
|
202
|
Sreenithya KH, Sugumar S. Protein-protein interaction network study of metallo-beta-lactamase-L1 present in Stenotrophomonas maltophilia and identification of potential drug targets. In Silico Pharmacol 2024; 12:94. [PMID: 39479381 PMCID: PMC11519282 DOI: 10.1007/s40203-024-00270-9] [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: 11/17/2023] [Accepted: 10/11/2024] [Indexed: 11/02/2024] Open
Abstract
Microorganisms are evolving to withstand the effect of antimicrobial agents and thereby pose a global threat known as antimicrobial resistance. Resistance towards multiple drugs due to various intrinsic as well environmental factors leads to an even more dangerous drug resistance property known as multi-drug resistance (MDR). WHO has recognized MDR bacteria as a top global threat as they complicate the treatment and augment mortality and morbidity risks. Gram-negative bacteria produce beta-lactamase enzymes that can hydrolyze beta-lactam antibiotics, impacting drug susceptibility. Stenotrophomonas maltophilia, an opportunistic pathogen, exemplifies MDR due to the production of two types of beta-lactamases. The metallo-beta-lactamase (MBL) L1 produced by the bacteria is a class B1 zinc-dependent MBL that is broadly substrate-specific and is a challenge to the currently available treatment options. This study constructs and analyzes a protein-protein interaction network of L1 beta-lactamase to comprehend its role in the MDR property of the bacteria. The network encompasses 51 proteins including L1 MBL (Smlt2667) and 382 interactions, revealing key players in MDR and potential drug targets. The network analysis aids the discernment of antimicrobial gene impact on cellular function, informing drug discovery strategies. This research addresses the emerging challenge of antibiotic resistance and identifies pathways for therapeutic intervention. Supplementary Information The online version contains supplementary material available at 10.1007/s40203-024-00270-9.
Collapse
Affiliation(s)
- K. H. Sreenithya
- Department of Genetic Engineering, School of Bioengineering, College of Engineering and Technology, SRM Institute of Science and Technology, Kattankulathur, India
| | - Shobana Sugumar
- Department of Genetic Engineering, School of Bioengineering, College of Engineering and Technology, SRM Institute of Science and Technology, Kattankulathur, India
| |
Collapse
|
203
|
Hristov BH, Noble WS, Bertero A. Systematic identification of interchromosomal interaction networks supports the existence of specialized RNA factories. Genome Res 2024; 34:1610-1623. [PMID: 39322282 PMCID: PMC11529845 DOI: 10.1101/gr.278327.123] [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: 08/08/2023] [Accepted: 08/30/2024] [Indexed: 09/27/2024]
Abstract
Most studies of genome organization have focused on intrachromosomal (cis) contacts because they harbor key features such as DNA loops and topologically associating domains. Interchromosomal (trans) contacts have received much less attention, and tools for interrogating potential biologically relevant trans structures are lacking. Here, we develop a computational framework that uses Hi-C data to identify sets of loci that jointly interact in trans This method, trans-C, initiates probabilistic random walks with restarts from a set of seed loci to traverse an input Hi-C contact network, thereby identifying sets of trans-contacting loci. We validate trans-C in three increasingly complex models of established trans contacts: the Plasmodium falciparum var genes, the mouse olfactory receptor "Greek islands," and the human RBM20 cardiac splicing factory. We then apply trans-C to systematically test the hypothesis that genes coregulated by the same trans-acting element (i.e., a transcription or splicing factor) colocalize in three dimensions to form "RNA factories" that maximize the efficiency and accuracy of RNA biogenesis. We find that many loci with multiple binding sites of the same DNA-binding proteins interact with one another in trans, especially those bound by factors with intrinsically disordered domains. Similarly, clustered binding of a subset of RNA-binding proteins correlates with trans interaction of the encoding loci. We observe that these trans-interacting loci are close to nuclear speckles. These findings support the existence of trans- interacting chromatin domains (TIDs) driven by RNA biogenesis. Trans-C provides an efficient computational framework for studying these and other types of trans interactions, empowering studies of a poorly understood aspect of genome architecture.
Collapse
Affiliation(s)
| | - William Stafford Noble
- Department of Genome Sciences, University of Washington, Seattle, Washington 98195, USA
- Paul G. Allen School of Computer Science and Engineering, University of Washington, Seattle, Washington 98195, USA
| | - Alessandro Bertero
- Molecular Biotechnology Center "Guido Tarone," Department of Molecular Biotechnology and Health Sciences, University of Turin, 10126 Torino, Italy
| |
Collapse
|
204
|
Ren J, Wang Y, Zhang Y, Jin H, Cheng J, Tao F, Zhu Y. Placental Transcriptomic Signatures of Prenatal Phthalate Exposure and Identification of Placenta-Brain Genes Associated with the Effects of Phthalate Exposure on Neurodevelopment in Children. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2024; 58:19141-19151. [PMID: 39392919 DOI: 10.1021/acs.est.4c04082] [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: 10/13/2024]
Abstract
Prenatal exposure to phthalates may affect placental function and fetal development, but the underlying mechanisms are unclear. The aim of our study was to explore the alterations in the placental transcriptome associated with prenatal phthalate exposure and to further analyze whether the placental-brain axis (PBA) genes play a mediating role in the association between prenatal phthalate exposure and children's neurodevelopment. We included 172 participants from the Ma'anshan Birth Cohort and collected data on seven phthalate metabolites in urine during pregnancy, placental tissue RNA-seq, and neurodevelopment of offspring. Bioinformatics analysis revealed that aberrant regulation of the placental transcriptome was associated with prenatal phthalate exposure. Exposure to phthalates during pregnancy was found to be associated with neurodevelopmental delay in children aged 6, 18, and 48 months using the multiple linear regression model. Meanwhile, employing mediation analysis, nine PBA genes were identified that mediate the association between exposure to phthalates during pregnancy and the neurodevelopment of children. Our study will provide a basis for potential mechanisms by which prenatal exposure to phthalates affects placental function and children's neurodevelopment.
Collapse
Affiliation(s)
- Jiawen Ren
- Department of Maternal, Child and Adolescent Health, School of Public Health, Anhui Medical University, Hefei 230032, Anhui, China
- MOE Key Laboratory of Population Health Across Life Cycle, Anhui Provincial Key Laboratory of Environment and Population Health across the Life Course, Anhui Medical University, Hefei 230032, Anhui, China
| | - Yifan Wang
- Department of Maternal, Child and Adolescent Health, School of Public Health, Anhui Medical University, Hefei 230032, Anhui, China
- MOE Key Laboratory of Population Health Across Life Cycle, Anhui Provincial Key Laboratory of Environment and Population Health across the Life Course, Anhui Medical University, Hefei 230032, Anhui, China
| | - Yimin Zhang
- Department of Maternal, Child and Adolescent Health, School of Public Health, Anhui Medical University, Hefei 230032, Anhui, China
- MOE Key Laboratory of Population Health Across Life Cycle, Anhui Provincial Key Laboratory of Environment and Population Health across the Life Course, Anhui Medical University, Hefei 230032, Anhui, China
| | - Heyue Jin
- Department of Maternal, Child and Adolescent Health, School of Public Health, Anhui Medical University, Hefei 230032, Anhui, China
- MOE Key Laboratory of Population Health Across Life Cycle, Anhui Provincial Key Laboratory of Environment and Population Health across the Life Course, Anhui Medical University, Hefei 230032, Anhui, China
| | - Jingjing Cheng
- Department of Maternal, Child and Adolescent Health, School of Public Health, Anhui Medical University, Hefei 230032, Anhui, China
- MOE Key Laboratory of Population Health Across Life Cycle, Anhui Provincial Key Laboratory of Environment and Population Health across the Life Course, Anhui Medical University, Hefei 230032, Anhui, China
| | - Fangbiao Tao
- Department of Maternal, Child and Adolescent Health, School of Public Health, Anhui Medical University, Hefei 230032, Anhui, China
- MOE Key Laboratory of Population Health Across Life Cycle, Anhui Provincial Key Laboratory of Environment and Population Health across the Life Course, Anhui Medical University, Hefei 230032, Anhui, China
| | - Yumin Zhu
- Department of Maternal, Child and Adolescent Health, School of Public Health, Anhui Medical University, Hefei 230032, Anhui, China
- MOE Key Laboratory of Population Health Across Life Cycle, Anhui Provincial Key Laboratory of Environment and Population Health across the Life Course, Anhui Medical University, Hefei 230032, Anhui, China
- Medical School, Nanjing University, Nanjing 210093, Jiangsu, China
| |
Collapse
|
205
|
Boyd SS, Robarts DR, Nguyen K, Villar M, Alghusen IM, Kotulkar M, Denson A, Fedosyuk H, Whelan SA, Lee NCY, Hanover J, Dias WB, Tan EP, McGreal SR, Artigues A, Swerdlow RH, Thompson JA, Apte U, Slawson C. Multi-omics after O-GlcNAc alteration identified cellular processes promoting aneuploidy after loss of O-GlcNAc transferase. Mol Metab 2024; 90:102060. [PMID: 39481848 DOI: 10.1016/j.molmet.2024.102060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Revised: 10/15/2024] [Accepted: 10/25/2024] [Indexed: 11/03/2024] Open
Abstract
OBJECTIVE Pharmacologic or genetic manipulation of O-GlcNAcylation, an intracellular, single sugar post-translational modification, are difficult to interpret due to the pleotropic nature of O-GlcNAc and the vast signaling pathways it regulates. METHOD To address the pleotropic nature of O-GlcNAc, we employed either OGT (O-GlcNAc transferase), OGA (O-GlcNAcase) liver knockouts, or pharmacological inhibition of OGA coupled with multi-Omics analysis and bioinformatics. RESULTS We identified numerous genes, proteins, phospho-proteins, or metabolites that were either inversely or equivalently changed between conditions. Moreover, we identified pathways in OGT knockout samples associated with increased aneuploidy. To test and validate these pathways, we induced liver growth in OGT knockouts by partial hepatectomy. OGT knockout livers showed a robust aneuploidy phenotype with disruptions in mitosis, nutrient sensing, protein metabolism/amino acid metabolism, stress response, and HIPPO signaling demonstrating how OGT is essential in controlling aneuploidy pathways. CONCLUSION These data show how a multi-Omics platform can disentangle the pleotropic nature of O-GlcNAc to discern how OGT fine-tunes multiple cellular pathways involved in aneuploidy.
Collapse
Affiliation(s)
- Samuel S Boyd
- Department of Biostatistics and Data Science, University of Kansas Medical Center, Kansas City, KS, USA
| | - Dakota R Robarts
- Department of Pharmacology, Toxicology and Therapeutics, Kansas City, KS, USA
| | - Khue Nguyen
- Department of Biochemistry, University of Kansas Medical Center, Kansas City, KS, USA
| | - Maite Villar
- Department of Biochemistry, University of Kansas Medical Center, Kansas City, KS, USA
| | - Ibtihal M Alghusen
- Department of Biochemistry, University of Kansas Medical Center, Kansas City, KS, USA
| | - Manasi Kotulkar
- Department of Pharmacology, Toxicology and Therapeutics, Kansas City, KS, USA
| | - Aspin Denson
- Department of Biochemistry, University of Kansas Medical Center, Kansas City, KS, USA
| | - Halyna Fedosyuk
- Department of Biochemistry, University of Kansas Medical Center, Kansas City, KS, USA
| | - Stephen A Whelan
- Department of Chemistry, Boston University, Boston, MA, USA; Precision Biomarker Laboratories, Cedars-Sinai Medical Center, Beverly Hills, CA, USA
| | - Norman C Y Lee
- Department of Chemistry, Boston University, Boston, MA, USA; Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - John Hanover
- Laboratory of Cell Biochemistry and Molecular Biology, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Wagner B Dias
- Federal University of Rio De Janeiro, Rio De Janeiro, Brazil
| | - Ee Phie Tan
- Department of Biochemistry, University of Kansas Medical Center, Kansas City, KS, USA; Department of Chemistry, Neurodegeneration New Medicines Center, and the Skaggs Institute of Chemical Biology, The Scripps Research Institute, La Jolla, CA, USA
| | - Steven R McGreal
- Department of Pharmacology, Toxicology and Therapeutics, Kansas City, KS, USA; XenoTech, A BioIVT Company, Kansas City, KS, USA
| | - Antonio Artigues
- Department of Biochemistry, University of Kansas Medical Center, Kansas City, KS, USA
| | - Russell H Swerdlow
- Department of Biochemistry, University of Kansas Medical Center, Kansas City, KS, USA; Department of Neurology, University of Kansas Medical Center, Kansas City, KS, USA; University of Kansas Alzheimer's Disease Research Center, KS, USA
| | - Jeffrey A Thompson
- Department of Biostatistics and Data Science, University of Kansas Medical Center, Kansas City, KS, USA; University of Kansas Cancer Center, Kansas City, KS, USA
| | - Udayan Apte
- Department of Pharmacology, Toxicology and Therapeutics, Kansas City, KS, USA
| | - Chad Slawson
- Department of Biochemistry, University of Kansas Medical Center, Kansas City, KS, USA; University of Kansas Alzheimer's Disease Research Center, KS, USA; University of Kansas Cancer Center, Kansas City, KS, USA.
| |
Collapse
|
206
|
Valentin-Alvarado LE, Shi LD, Appler KE, Crits-Christoph A, De Anda V, Adler BA, Cui ML, Ly L, Leão P, Roberts RJ, Sachdeva R, Baker BJ, Savage DF, Banfield JF. Complete genomes of Asgard archaea reveal diverse integrated and mobile genetic elements. Genome Res 2024; 34:1595-1609. [PMID: 39406503 PMCID: PMC11529989 DOI: 10.1101/gr.279480.124] [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: 04/17/2024] [Accepted: 08/27/2024] [Indexed: 11/01/2024]
Abstract
Asgard archaea are of great interest as the progenitors of Eukaryotes, but little is known about the mobile genetic elements (MGEs) that may shape their ongoing evolution. Here, we describe MGEs that replicate in Atabeyarchaeia, a wetland Asgard archaea lineage represented by two complete genomes. We used soil depth-resolved population metagenomic data sets to track 18 MGEs for which genome structures were defined and precise chromosome integration sites could be identified for confident host linkage. Additionally, we identified a complete 20.67 kbp circular plasmid and two family-level groups of viruses linked to Atabeyarchaeia, via CRISPR spacer targeting. Closely related 40 kbp viruses possess a hypervariable genomic region encoding combinations of specific genes for small cysteine-rich proteins structurally similar to restriction-homing endonucleases. One 10.9 kbp integrative conjugative element (ICE) integrates genomically into the Atabeyarchaeum deiterrae-1 chromosome and has a 2.5 kbp circularizable element integrated within it. The 10.9 kbp ICE encodes an expressed Type IIG restriction-modification system with a sequence specificity matching an active methylation motif identified by Pacific Biosciences (PacBio) high-accuracy long-read (HiFi) metagenomic sequencing. Restriction-modification of Atabeyarchaeia differs from that of another coexisting Asgard archaea, Freyarchaeia, which has few identified MGEs but possesses diverse defense mechanisms, including DISARM and Hachiman, not found in Atabeyarchaeia. Overall, defense systems and methylation mechanisms of Asgard archaea likely modulate their interactions with MGEs, and integration/excision and copy number variation of MGEs in turn enable host genetic versatility.
Collapse
Affiliation(s)
- Luis E Valentin-Alvarado
- Innovative Genomics Institute, University of California, Berkeley, California 94720, USA
- Department of Plant and Microbial Biology, University of California, Berkeley, California 94720, USA
| | - Ling-Dong Shi
- Innovative Genomics Institute, University of California, Berkeley, California 94720, USA
| | - Kathryn E Appler
- Department of Marine Science, University of Texas at Austin, Marine Science Institute, Port Aransas, Texas 78373, USA
| | - Alexander Crits-Christoph
- Innovative Genomics Institute, University of California, Berkeley, California 94720, USA
- Department of Plant and Microbial Biology, University of California, Berkeley, California 94720, USA
| | - Valerie De Anda
- Department of Marine Science, University of Texas at Austin, Marine Science Institute, Port Aransas, Texas 78373, USA
- Department of Integrative Biology, University of Texas at Austin, Austin, Texas 78712, USA
| | - Benjamin A Adler
- Innovative Genomics Institute, University of California, Berkeley, California 94720, USA
| | - Michael L Cui
- Innovative Genomics Institute, University of California, Berkeley, California 94720, USA
| | - Lynn Ly
- Oxford Nanopore Technologies Incorporated, New York, New York 10013, USA
| | - Pedro Leão
- Department of Marine Science, University of Texas at Austin, Marine Science Institute, Port Aransas, Texas 78373, USA
| | | | - Rohan Sachdeva
- Innovative Genomics Institute, University of California, Berkeley, California 94720, USA
| | - Brett J Baker
- Department of Marine Science, University of Texas at Austin, Marine Science Institute, Port Aransas, Texas 78373, USA
- Department of Integrative Biology, University of Texas at Austin, Austin, Texas 78712, USA
| | - David F Savage
- Innovative Genomics Institute, University of California, Berkeley, California 94720, USA
- Howard Hughes Medical Institute, University of California, Berkeley, California 94720, USA
- Department of Molecular and Cell Biology, University of California, Berkeley, California 94720, USA
| | - Jillian F Banfield
- Innovative Genomics Institute, University of California, Berkeley, California 94720, USA;
- Department of Integrative Biology, University of Texas at Austin, Austin, Texas 78712, USA
- Earth and Planetary Science, University of California, Berkeley, California 94720, USA
- Department of Microbiology, Biomedicine Discovery Institute, Monash University, Victoria 3168, Australia
| |
Collapse
|
207
|
Qin F, Luo X, Lu Q, Cai B, Xiao F, Cai G. Spatial pattern and differential expression analysis with spatial transcriptomic data. Nucleic Acids Res 2024:gkae962. [PMID: 39470725 DOI: 10.1093/nar/gkae962] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2024] [Revised: 10/03/2024] [Accepted: 10/11/2024] [Indexed: 10/30/2024] Open
Abstract
The emergence of spatial transcriptomic technologies has opened new avenues for investigating gene activities while preserving the spatial context of tissues. Utilizing data generated by such technologies, the identification of spatially variable (SV) genes is an essential step in exploring tissue landscapes and biological processes. Particularly in typical experimental designs, such as case-control or longitudinal studies, identifying SV genes between groups is crucial for discovering significant biomarkers or developing targeted therapies for diseases. However, current methods available for analyzing spatial transcriptomic data are still in their infancy, and none of the existing methods are capable of identifying SV genes between groups. To overcome this challenge, we developed SPADE for spatial pattern and differential expression analysis to identify SV genes in spatial transcriptomic data. SPADE is based on a machine learning model of Gaussian process regression with a gene-specific Gaussian kernel, enabling the detection of SV genes both within and between groups. Through benchmarking against existing methods in extensive simulations and real data analyses, we demonstrated the preferred performance of SPADE in detecting SV genes within and between groups. The SPADE source code and documentation are publicly available at https://github.com/thecailab/SPADE.
Collapse
Affiliation(s)
- Fei Qin
- Department of Epidemiology and Biostatistics, Arnold School of Public Health, University of South Carolina, 921 Assembly Street, Columbia, SC, 29208, USA
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, 9609 Medical Center Drive, Rockville, MD, 20850, USA
| | - Xizhi Luo
- Department of Epidemiology and Biostatistics, Arnold School of Public Health, University of South Carolina, 921 Assembly Street, Columbia, SC, 29208, USA
- Data and Statistical Sciences, AbbVie Inc., 1 N. Waukegan Road, North Chicago, IL, 60064, USA
| | - Qing Lu
- Department of Biostatistics, College of Public Health and Health Professions and College of Medicine, University of Florida, 2004 Mowry Rd., Gainesville, FL, 32608, USA
| | - Bo Cai
- Department of Epidemiology and Biostatistics, Arnold School of Public Health, University of South Carolina, 921 Assembly Street, Columbia, SC, 29208, USA
| | - Feifei Xiao
- Department of Biostatistics, College of Public Health and Health Professions and College of Medicine, University of Florida, 2004 Mowry Rd., Gainesville, FL, 32608, USA
| | - Guoshuai Cai
- Department of Biostatistics, College of Public Health and Health Professions and College of Medicine, University of Florida, 2004 Mowry Rd., Gainesville, FL, 32608, USA
- Department of Surgery, College of Medicine, University of Florida, 1600 SW Archer Rd., Gainesville, FL, 32610, USA
| |
Collapse
|
208
|
Zhan K, Chen S, Ji L, Xu L, Zhang Y, Zhang Q, Dai Q, Wu S. Network pharmacology to unveil the mechanism of Astragali Radix in the treatment of lupus nephritis via PI3K/AKT/mTOR pathway. Sci Rep 2024; 14:25983. [PMID: 39472740 PMCID: PMC11522298 DOI: 10.1038/s41598-024-77897-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Accepted: 10/25/2024] [Indexed: 11/02/2024] Open
Abstract
We used network pharmacology, molecular docking, and in vitro experiments to explore the mechanisms of Astragali Radix in the treatment of lupus nephritis. We screened compounds and targets of Astragali Radix, as well as related genes of lupus nephritis from databases. We identified 211 common genes and 44 compounds between the herb and the disease, and constructed global, narrowed, hierarchical Compound-Target Interaction networks to illustrate the possible mechanism. We found that the PI3K/AKT/mTOR pathway is a core target gene set identified through enrichment analysis, PPI analysis and MCODE analysis. In vitro experiments showed that freeze-dried Astragali powder inhibits activation of PI3K, AKT1 and mTOR in TGF-β1 stimulated HK-2 cells. Molecular docking demonstrated that (R)-isomucronulatol, 3,9,10-trimethoxypterocarpan and astrapterocarpan exhibited promising binding affinity to PI3K, AKT, and mTOR proteins.
Collapse
Affiliation(s)
- Kuijun Zhan
- The Second School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - Shuo Chen
- The First School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - Lina Ji
- The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, China
| | - Liping Xu
- The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, China
| | - Yan Zhang
- The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, China
| | - Qi Zhang
- The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, China
| | - Qiaoding Dai
- The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, China.
| | - Shan Wu
- The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, China.
| |
Collapse
|
209
|
Adindu CS, Tombrello K, Martz LA, Zeczycki TN, Ellis HR. A distinct co-expressed sulfurtransferase extends the physiological role of mercaptopropionate dioxygenase in Pseudomonas aeruginosa PAO1. BIOCHIMICA ET BIOPHYSICA ACTA. PROTEINS AND PROTEOMICS 2024; 1873:141059. [PMID: 39471965 DOI: 10.1016/j.bbapap.2024.141059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Revised: 09/26/2024] [Accepted: 10/23/2024] [Indexed: 11/01/2024]
Abstract
Oxidation and assimilation of persulfides in bacteria is often catalyzed by a persulfide dioxygenase and sulfurtransferase in consecutive reactions. Enzymes responsible for the oxidation of persulfides have not been clearly defined in Pseudomonas aeruginosa PAO1. The characterized mercaptopropionate dioxygenase (MDO) in P. aeruginosa PAO1 has been proposed to catalyze the oxidation of 3-mercaptopropionate. However, the physiological role of MDO is uncertain given the expression of a sulfurtransferase (ST) enzyme on the same operon as the thiol dioxygenase. The st gene had a co-occurrence frequency with mdo of 0.94 demonstrating the co-expression and physiological link of the two genes. There are four tandem rhodanese domains in the ST enzyme with two of the domains containing potential catalytic Cys residues (Cys191 and Cys435) capable of forming a persulfide. Only Cys435 was accessible in thiol quantification assays, and results from H/D-X MS analyses further established the accessibility of the domain containing Cys435. Both thiosulfate and mercaptopyruvate served as sulfur donors to the ST enzyme, with Cys435 forming the persulfide intermediate. Kinetic investigations of MDO suggested the enzyme had a broader substrate specificity than previously identified, oxidizing both mercaptopropionate and mercaptopyruvate thiol and persulfide substrates. The results obtained from these investigations provide insight into the overall mechanism and physiological role of the mdo operon in sulfide oxidation and assimilation.
Collapse
Affiliation(s)
- Chukwuemeka S Adindu
- Department of Biochemistry and Molecular Biology, Brody School of Medicine at East Carolina University, Greenville, North Carolina 27834, United States
| | - Katie Tombrello
- Takeda Pharmaceuticals, Social Circle, GA 30025, United States
| | - Luke A Martz
- Department of Chemistry and Biochemistry, Auburn University, Auburn, AL 36849, United States
| | - Tonya N Zeczycki
- Department of Biochemistry and Molecular Biology, Brody School of Medicine at East Carolina University, Greenville, North Carolina 27834, United States
| | - Holly R Ellis
- Department of Biochemistry and Molecular Biology, Brody School of Medicine at East Carolina University, Greenville, North Carolina 27834, United States.
| |
Collapse
|
210
|
Mogollón García HD, de Andrade Ferrazza R, Ochoa JC, de Athayde FF, Vidigal PMP, Wiltbank M, Kastelic JP, Sartori R, Ferreira JCP. Landscape transcriptomic analysis of bovine follicular cells during key phases of ovarian follicular development. Biol Res 2024; 57:76. [PMID: 39468655 PMCID: PMC11514973 DOI: 10.1186/s40659-024-00558-2] [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: 05/24/2024] [Accepted: 10/15/2024] [Indexed: 10/30/2024] Open
Abstract
BACKGROUND There are many gaps in our understanding of the mechanisms involved in ovarian follicular development in cattle, particularly regarding follicular deviation, acquisition of ovulatory capacity, and preovulatory changes. Molecular evaluations of ovarian follicular cells during follicular development in cattle, especially serial transcriptomic analyses across key growth phases, have not been reported. This study aims to address this gap by analyzing gene expression using RNA-seq in granulosa and antral cells recovered from ovarian follicular fluid during critical phases of ovarian follicular development in Holstein cows. RESULTS Integrated analysis of gene ontology (GO), gene set enrichment (GSEA), protein-protein interaction (PPI), and gene topology identified that differentially expressed genes (DEGs) in the largest ovarian follicles at deviation (Dev) were primarily involved in FSH-negative feedback, steroidogenesis, cell proliferation, apoptosis, and the prevention of early follicle rupture. In contrast, DEGs in the second largest follicles (DevF2) were mainly related to loss of cell viability, apoptosis, and immune cell invasion. In the dominant (PostDev) and preovulatory (PreOv) follicles, DEGs were associated with vascular changes and inflammatory responses. CONCLUSIONS The transcriptome of ovarian follicular fluid cells had a predominance of granulosa cells in the dominant follicle at deviation, with upregulation of genes involved in cell viability, steroidogenesis, and apoptosis prevention, whereas in the non-selected follicle there was upregulation of cell death-related transcripts. Immune cell transcripts increased significantly after deviation, particularly in preovulatory follicles, indicating strong intrafollicular chemotactic activity. We inferred that immune cell invasion occurred despite an intact basal lamina, contributing to follicular maturation.
Collapse
Affiliation(s)
- Henry David Mogollón García
- Department of Veterinary Surgery and Animal Reproduction, School of Veterinary Medicine and Animal Science, São Paulo State University (UNESP), Rua Prof. Doutor Walter Mauricio Correa, s/n, Botucatu, São Paulo, 18618-681, Brazil
- Department of Genetic, Evolution, Microbiology and Immunology. Biology Institute, Campinas State University, Campinas, São Paulo, Brazil
- Computational Systems Biology Laboratory (CSBL), Institut Pasteur, University of São Paulo (USP), São Paulo, Brazil
| | | | - Julian Camilo Ochoa
- Department of Veterinary Surgery and Animal Reproduction, School of Veterinary Medicine and Animal Science, São Paulo State University (UNESP), Rua Prof. Doutor Walter Mauricio Correa, s/n, Botucatu, São Paulo, 18618-681, Brazil
| | - Flávia Florencio de Athayde
- Department of Animal Production and Health, School of Veterinary Medicine, São Paulo State University (UNESP), Araçatuba, São Paulo, Brazil
| | | | - Milo Wiltbank
- Department of Animal & Dairy Sciences, University of Wisconsin-Madison, Madison, USA
| | | | - Roberto Sartori
- Department of Animal Science, Luiz de Queiroz College of Agriculture (ESALQ), University of São Paulo, Piracicaba, São Paulo, Brazil
| | - João Carlos Pinheiro Ferreira
- Department of Veterinary Surgery and Animal Reproduction, School of Veterinary Medicine and Animal Science, São Paulo State University (UNESP), Rua Prof. Doutor Walter Mauricio Correa, s/n, Botucatu, São Paulo, 18618-681, Brazil.
| |
Collapse
|
211
|
Singha CJ, Krishna R. Molecular dynamics simulations, essential dynamics and MMPBSA to evaluate natural compounds as potential inhibitors for AccD6, a key drug target in the fatty acid biosynthesis pathway in Mycobacterium tuberculosis. J Mol Graph Model 2024; 134:108898. [PMID: 39486266 DOI: 10.1016/j.jmgm.2024.108898] [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: 04/08/2024] [Revised: 10/24/2024] [Accepted: 10/25/2024] [Indexed: 11/04/2024]
Abstract
Antibiotic resistance in Mycobacterium tuberculosis, the primary causative agent of the tuberculosis disease is an ever growing threat especially in developing and underdeveloped countries. Isoniazid is a commonly used first line anti-tuberculosis drug used during the first phase of tuberculosis treatment. However, due to its improper use, many strains of Mycobacterium tuberculosis have acquired resistance to the drug. Advancements in next generation sequencing technologies, such as transcriptomics have paved way for identifying alternative drug targets based on the differential expression pattern of genes. Therefore, this study makes use of RNA-Seq data of Mycobacterium tuberculosis isolates treated with different concentrations of isoniazid to identify genes that can be proposed as drug targets. From the differential expression analysis, it was observed that four genes were significantly upregulated under all the conditions. Among the four genes, accD6 was selected as the drug target for virtual screening and molecular dynamics studies, because of its role in fatty acid elongation and contribution to the synthesis of mycolic acids. The protein-protein interaction network and gene ontology based functional enrichment studies show an enrichment in fatty acid biosynthesis related pathways. Furthermore, virtual screening studies successfully screened the top three natural inhibitor molecules with satisfactory ADME properties and a better glide score than the reference compound, NCI-172033. The trajectory analysis, essential dynamics studies and MMPBSA analysis, concluded that among the hit molecules, NPC41982, a thiazole derivative showed the most promising results and can be considered as a potential drug candidate.
Collapse
Affiliation(s)
| | - Ramadas Krishna
- Department of Bioinformatics, Pondicherry University, Puducherry, 605014, India.
| |
Collapse
|
212
|
Chatatikun M, Pattaranggoon NC, Sama-Ae I, Ranteh O, Poolpirom M, Pantanakong O, Chumworadet P, Kawakami F, Imai M, Tedasen A. Mechanistic exploration of bioactive constituents in Gnetum gnemon for GPCR-related cancer treatment through network pharmacology and molecular docking. Sci Rep 2024; 14:25738. [PMID: 39468096 PMCID: PMC11519448 DOI: 10.1038/s41598-024-75240-4] [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: 07/14/2023] [Accepted: 10/03/2024] [Indexed: 10/30/2024] Open
Abstract
G Protein-Coupled Receptors (GPCRs) are integral membrane proteins that have gained considerable attention as drug targets, particularly in cancer treatment. In this study, we explored the capacity of bioactive compounds derived from Gnetum gnemon (GG) for the development of of pharmaceuticals targeting GPCRs within the context of cancer therapy. Integrated approach combined network pharmacology and molecular docking to identify and validate the underlying pharmacological mechanisms. We retrieved targets for GG-derived compounds and GPCRs-related cancer from databases. Subsequently, we established a protein-protein interaction (PPI) network by mapping the shared targets. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analyses were employed to predict the mechanism of action of these targets. Molecular docking was conducted to validate our findings. We identified a total of 265 targets associated with GG-derived bioactive compounds for the treatment of GPCRs-related cancer. Functional enrichment analysis revealed the promising therapeutic effects of these targets on GPCRs-related cancer pathways. The PPI network analysis identified hub targets, including MAPK3, SRC, EGFR, STAT3, ESR1, MTOR, CCND1, and PPARG, which demonstrate as treatment targets for GPCRs-related cancer using GG-derived compounds. Additionally, molecular docking experiments demonstrated the strong binding affinity of gnetin A, gnetin C, (-)-viniferin, and resveratrol dimer, thus inhibiting MAPK3, SRC, EGFR, and MTOR. Survival analysis established the clinical prognostic relevance of identified hub genes in cancer. This study presents a novel approach for comprehending the therapeutic mechanisms of GG-derived active compounds and thereby paving the way for their prospective clinical applications in the field of cancer treatment.
Collapse
Affiliation(s)
- Moragot Chatatikun
- School of Allied Health Sciences, Walailak University, Nakhon Si Thammarat, 80161, Thailand
- Research Excellence Center for Innovation and Health Products, Walailak University, Nakhon Si Thammarat, 80161, Thailand
| | - Nawanwat C Pattaranggoon
- Faculty of Medical Technology, Rangsit University, Muang Pathumthani, Pathumthani, 12000, Thailand
- Program in Bioinformatics and Computational Biology, Graduate School, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Imran Sama-Ae
- School of Allied Health Sciences, Walailak University, Nakhon Si Thammarat, 80161, Thailand
- Center of Excellence Research for Melioidosis and Microorganisms (CERMM), Walailak University, Nakhon Si Thammarat, 80161, Thailand
| | - Onggan Ranteh
- Department of Community Public Health, School of Public Health, Walailak University, Nakhon Si Thammarat, 80161, Thailand
- Excellent Center for Dengue and Community Public Health (EC for DACH), Walailak University, Nakhon Si Thammarat, 80161, Thailand
| | - Manlika Poolpirom
- School of Allied Health Sciences, Walailak University, Nakhon Si Thammarat, 80161, Thailand
| | - Oranan Pantanakong
- School of Allied Health Sciences, Walailak University, Nakhon Si Thammarat, 80161, Thailand
| | - Pitchaporn Chumworadet
- School of Allied Health Sciences, Walailak University, Nakhon Si Thammarat, 80161, Thailand
| | - Fumitaka Kawakami
- Research Facility of Regenerative Medicine and Cell Design, School of Allied Health Sciences, Kitasato University, Sagamihara, 252-0373, Japan
- Department of Regulatory Biochemistry, Kitasato University Graduate School of Medical Sciences, Sagamihara, 252-0373, Japan
| | - Motoki Imai
- Research Facility of Regenerative Medicine and Cell Design, School of Allied Health Sciences, Kitasato University, Sagamihara, 252-0373, Japan
- Department of Molecular Diagnostics, School of Allied Health Sciences, Kitasato University, Sagamihara, 252-0373, Japan
| | - Aman Tedasen
- School of Allied Health Sciences, Walailak University, Nakhon Si Thammarat, 80161, Thailand.
- Research Excellence Center for Innovation and Health Products, Walailak University, Nakhon Si Thammarat, 80161, Thailand.
| |
Collapse
|
213
|
Di Luca A, Bennato F, Ianni A, Martino C, Henry M, Meleady P, Martino G. Label-free liquid chromatography mass spectrometry analysis of changes in broiler liver proteins under transport stress. PLoS One 2024; 19:e0311539. [PMID: 39466737 PMCID: PMC11515959 DOI: 10.1371/journal.pone.0311539] [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: 02/07/2024] [Accepted: 09/19/2024] [Indexed: 10/30/2024] Open
Abstract
Transportation duration and distance are significant concerns for animal welfare, particularly in the poultry industry. However, limited proteomic studies have investigated the impact of transport duration on poultry welfare. In this study, mass spectrometry based bottom up proteomics was employed to sensitively and impartially profile the liver tissue proteome of chickens, addressing the issue of animal stress and welfare in response to transportation before slaughter. The liver exudates obtained from Ross 508 chickens exposed to either short or long road transportation underwent quantitative label-free LC-MS proteomic profiling. This method identified a total of 1,368 proteins, among which 35 were found to be significantly different (p < 0.05) and capable of distinguishing between short and long road transportation conditions. Specifically, 23 proteins exhibited up-regulation in the non stressed group, while 12 proteins showed up-regulation in the stressed group. The proteins identified in this pilot study encompassed those linked to homeostasis and cellular energetic balance, including heat shock proteins and the 5'-nucleotidase domain-containing family. These results contribute to a deeper understanding of the proteome in broiler liver tissues, shedding light on poultry adaptability to transport stress. Furthermore, the identified proteins present potential as biomarkers, suggesting promising approaches to enhance poultry care and management within the industry.
Collapse
Affiliation(s)
- Alessio Di Luca
- Department of Bioscience and Technology for Food Agro-Food and Environmental Technology, University of Teramo, Teramo, Italy
| | - Francesca Bennato
- Department of Bioscience and Technology for Food Agro-Food and Environmental Technology, University of Teramo, Teramo, Italy
| | - Andrea Ianni
- Department of Bioscience and Technology for Food Agro-Food and Environmental Technology, University of Teramo, Teramo, Italy
| | - Camillo Martino
- Department of Veterinary Medicine, University of Perugia, Perugia, Italy
| | - Michael Henry
- National Institute for Cellular Biotechnology, Dublin City University, Dublin, Ireland
| | - Paula Meleady
- National Institute for Cellular Biotechnology, Dublin City University, Dublin, Ireland
- School of Biotechnology, Dublin City University, Dublin, Ireland
| | - Giuseppe Martino
- Department of Bioscience and Technology for Food Agro-Food and Environmental Technology, University of Teramo, Teramo, Italy
| |
Collapse
|
214
|
Tu Y, Gu H, Li N, Sun D, Yang Z, He L. Identification of Key Genes Related to Immune-Lipid Metabolism in Skin Barrier Damage and Analysis of Immune Infiltration. Inflammation 2024:10.1007/s10753-024-02174-4. [PMID: 39465470 DOI: 10.1007/s10753-024-02174-4] [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: 07/09/2024] [Revised: 09/23/2024] [Accepted: 10/21/2024] [Indexed: 10/29/2024]
Abstract
Several physical and chemical factors regulate skin barrier function. Skin barrier dysfunction causes many inflammatory skin diseases, such as atopic dermatitis and psoriasis. Activation of the immune response may lead to damage to the epidermal barrier. Abnormal lipid metabolism is defined as abnormally high or low values of plasma lipid components such as plasma cholesterol and triglycerides. The mouse skin barrier damage model was used for RNA sequencing. Bioinformatics analysis and validation were performed. Differently expressed genes (DEGs) related to immune and lipid metabolism were screened by differentially expressed gene analysis, and the enriched biological processes and pathways of these genes were identified by GO-KEGG. The interactions between DEGs were confirmed by constructing a PPI network. GSEA, transcription factor regulatory network, and immune infiltration analyses were performed for the 10 genes. Expression validation was performed by public datasets. The expression of key genes in mouse skin tissue was detected by qPCR. The expression of differentially expressed immune cell markers in the skin was detected by immunofluorescence. Based on the trans epidermal water loss (TEWL) score, the expression of key genes was detected by qPCR before skin barrier injury, at 4h and 7d, and at recovery from injury. Il17a, Il6, Tnf, Itgam, and Cxcl1 were immune-related key genes. Pla2g2f, Ptgs2, Plb1, Pla2g3, and Pla2g2d were key genes for lipid metabolism. Database validation and experimental results revealed that the expression trends of these genes were consistent with our analyses. The research value of these genes has been demonstrated through mouse datasets and experimental validation, and future therapeutic approaches may be able to mitigate the disease by targeting these genes to modulate the function of the skin barrier.
Collapse
Affiliation(s)
- Ying Tu
- Department of Dermatology, First Affiliated Hospital of Kunming Medical University, No. 295 XiChang Road, KunMing, 650032, China
| | - Hua Gu
- Department of Dermatology, First Affiliated Hospital of Kunming Medical University, No. 295 XiChang Road, KunMing, 650032, China
| | - Na Li
- Department of Dermatology, First Affiliated Hospital of Kunming Medical University, No. 295 XiChang Road, KunMing, 650032, China
| | - Dongjie Sun
- Department of Dermatology, First Affiliated Hospital of Kunming Medical University, No. 295 XiChang Road, KunMing, 650032, China
| | - Zhenghui Yang
- Department of Dermatology, First Affiliated Hospital of Kunming Medical University, No. 295 XiChang Road, KunMing, 650032, China
| | - Li He
- Department of Dermatology, First Affiliated Hospital of Kunming Medical University, No. 295 XiChang Road, KunMing, 650032, China.
| |
Collapse
|
215
|
Wu B, Sun M, Zhong T, Zhang J, Lei T, Yan Y, Chen X, Nan R, Sun F, Zhang C, Xi Y. Genome-wide identification and expression analysis of two-component system genes in switchgrass (Panicum virgatum L.). BMC PLANT BIOLOGY 2024; 24:1014. [PMID: 39465364 PMCID: PMC11520087 DOI: 10.1186/s12870-024-05687-0] [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: 08/07/2024] [Accepted: 10/09/2024] [Indexed: 10/29/2024]
Abstract
The two-component system (TCS) consists of histidine kinase (HK), histidine phosphate transfer protein (HP), and response regulatory factor (RR). It is one of the most crucial components of signal transduction in plants, playing a significant role in regulating plant growth, development, and responses to various abiotic stresses. Although TCS genes have been extensively identified in a variety of plants, the genome-wide recognition and examination of TCS in switchgrass remain unreported. Accordingly, this study identified a total of 87 TCS members in the genome of switchgrass, comprising 20 HK(L)s, 10 HPs, and 57 RRs. Detailed analyses were also conducted on their gene structures, conserved domains, and phylogenetic relationships. Moreover, this study analysed the gene expression profiles across diverse organs and investigated their response patterns to adverse environmental stresses. Results revealed that 87 TCS genes were distributed across 18 chromosomes, with uneven distribution. Expansion of these genes in switchgrass was achieved through both fragment and tandem duplication. PvTCS members are relatively conservative in the evolutionary process, but the gene structure varies significantly. Various cis-acting elements, varying in types and amounts, are present in the promoter region of PvTCSs, all related to plant growth, development, and abiotic stress, due to the TCS gene structure. Protein-protein interaction and microRNA prediction suggest complex interactions and transcriptional regulation among TCS members. Additionally, most TCS members are expressed in roots and stems, with some genes showing organ-specific expression at different stages of leaf and inflorescence development. Under conditions of abiotic stress such as drought, low temperature, high temperature, and salt stress, as well as exogenous abscisic acid (ABA), the expression of most TCS genes is either stimulated or inhibited. Our systematic analysis could offer insight into the characterization of the TCS genes, and further the growth of functional studies in switchgrass.
Collapse
Affiliation(s)
- Baolin Wu
- College of Agronomy, Northwest A&F University, Yangling, Shaanxi, 712100, China
- State Key Laboratory of Crop Stress Biology for Arid Areas, Yangling, Shaanxi, 712100, China
| | - Mengyu Sun
- College of Agronomy, Northwest A&F University, Yangling, Shaanxi, 712100, China
- State Key Laboratory of Crop Stress Biology for Arid Areas, Yangling, Shaanxi, 712100, China
| | - Tao Zhong
- College of Agronomy, Northwest A&F University, Yangling, Shaanxi, 712100, China
- State Key Laboratory of Crop Stress Biology for Arid Areas, Yangling, Shaanxi, 712100, China
| | - Jiawei Zhang
- College of Agronomy, Northwest A&F University, Yangling, Shaanxi, 712100, China
- State Key Laboratory of Crop Stress Biology for Arid Areas, Yangling, Shaanxi, 712100, China
| | - Tingshu Lei
- College of Agronomy, Northwest A&F University, Yangling, Shaanxi, 712100, China
- State Key Laboratory of Crop Stress Biology for Arid Areas, Yangling, Shaanxi, 712100, China
| | - Yuming Yan
- College of Agronomy, Northwest A&F University, Yangling, Shaanxi, 712100, China
- State Key Laboratory of Crop Stress Biology for Arid Areas, Yangling, Shaanxi, 712100, China
| | - Xiaohong Chen
- College of Agronomy, Northwest A&F University, Yangling, Shaanxi, 712100, China
- State Key Laboratory of Crop Stress Biology for Arid Areas, Yangling, Shaanxi, 712100, China
| | - Rui Nan
- College of Agronomy, Northwest A&F University, Yangling, Shaanxi, 712100, China
- State Key Laboratory of Crop Stress Biology for Arid Areas, Yangling, Shaanxi, 712100, China
| | - Fengli Sun
- College of Agronomy, Northwest A&F University, Yangling, Shaanxi, 712100, China
- State Key Laboratory of Crop Stress Biology for Arid Areas, Yangling, Shaanxi, 712100, China
| | - Chao Zhang
- College of Agronomy, Northwest A&F University, Yangling, Shaanxi, 712100, China
- State Key Laboratory of Crop Stress Biology for Arid Areas, Yangling, Shaanxi, 712100, China
| | - Yajun Xi
- College of Agronomy, Northwest A&F University, Yangling, Shaanxi, 712100, China.
- State Key Laboratory of Crop Stress Biology for Arid Areas, Yangling, Shaanxi, 712100, China.
| |
Collapse
|
216
|
Zwierzchowski G, Zhang G, Tobolski D, Wójcik R, Wishart DS, Ametaj BN. Metabolomic fingerprinting of milk fever cows: Pre- and postpartum metabolite alterations. J Vet Intern Med 2024. [PMID: 39466655 DOI: 10.1111/jvim.17217] [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: 01/27/2024] [Accepted: 09/25/2024] [Indexed: 10/30/2024] Open
Abstract
BACKGROUND Milk fever (MF), a metabolic disorder in dairy cows characterized by low blood calcium concentrations postpartum, is well-recognized clinically. However, comprehensive data on the alteration of metabolites associated with this condition remains sparse. HYPOTHESIS Delineate serum metabolite profiles and metabolic pathways preceding, coinciding with, and after the onset of MF. ANIMALS Twenty-six cows, including 20 healthy cows and 6 cows initially affected by MF. Because of culling, the number of MF-affected cows decreased to 4 at MF week, +4 weeks, and +8 weeks postpartum. METHODS A nested case-control longitudinal study was conducted, with blood samples collected at -8 and -4 weeks prepartum, MF week, and +4 and +8 weeks postpartum. Serum analysis utilized direct injection/liquid chromatography/tandem mass spectrometry (DI/LC/MS/MS) techniques. RESULTS Key findings included the identification of diverse metabolites such as hexose, amino acids, phosphatidylcholines, lysophosphatidylcholines, and sphingomyelin, which varied between studied groups (P < .05). The most marked metabolic alterations were observed 4 weeks prepartum. In total, 42, 56, 38, 29, and 24 metabolites distinguished the MF group at the respective time points (P < .05). Additionally, 33 metabolic pathways, including amino acid, antioxidant metabolism, fatty acid degradation, and carbohydrate processing, were impacted (P < .05). CONCLUSIONS AND CLINICAL IMPORTANCE Metabolic disruptions in dairy cows begin several weeks before the clinical manifestation of MF and persist up to 8 weeks postpartum. These findings emphasize the complexity of MF, extending beyond only hypocalcemia and indicate the necessity for preemptive monitoring in dairy herd management.
Collapse
Affiliation(s)
- Grzegorz Zwierzchowski
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, Alberta, Canada
- Faculty of Biology and Biotechnology, University of Warmia and Mazury, Olsztyn, Poland
| | - Guanshi Zhang
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, Alberta, Canada
| | - Dawid Tobolski
- Department of Large Animal Diseases and Clinic, Institute of Veterinary Medicine, Warsaw University of Life Sciences, Warsaw, Poland
| | - Roman Wójcik
- Department of Microbiology and Clinical Immunology, Faculty of Veterinary Medicine, University of Warmia and Mazury in Olsztyn, Olsztyn, Poland
| | - David S Wishart
- Department of Biological and Computer Sciences, University of Alberta, Edmonton, Alberta, Canada
| | - Burim N Ametaj
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
217
|
Bae M, Le C, Mehta RS, Dong X, Pieper LM, Ramirez L, Alexander M, Kiamehr S, Turnbaugh PJ, Huttenhower C, Chan AT, Balskus EP. Metatranscriptomics-guided discovery and characterization of a polyphenol-metabolizing gut microbial enzyme. Cell Host Microbe 2024:S1931-3128(24)00360-3. [PMID: 39471822 DOI: 10.1016/j.chom.2024.10.002] [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: 06/14/2024] [Revised: 09/18/2024] [Accepted: 10/01/2024] [Indexed: 11/01/2024]
Abstract
Gut microbial catechol dehydroxylases are a largely uncharacterized family of metalloenzymes that potentially impact human health by metabolizing dietary polyphenols. Here, we use metatranscriptomics (MTX) to identify highly transcribed catechol-dehydroxylase-encoding genes in human gut microbiomes. We discover a prevalent, previously uncharacterized catechol dehydroxylase (Gp Hcdh) from Gordonibacter pamelaeae that dehydroxylates hydrocaffeic acid (HCA), an anti-inflammatory gut microbial metabolite derived from plant-based foods. Further analyses suggest that the activity of Gp Hcdh may reduce anti-inflammatory benefits of polyphenol-rich foods. Together, these results show the utility of combining MTX analysis and biochemical characterization for gut microbial enzyme discovery and reveal a potential link between host inflammation and a specific polyphenol-metabolizing gut microbial enzyme.
Collapse
Affiliation(s)
- Minwoo Bae
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA 02138, USA
| | - Chi Le
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA 02138, USA
| | - Raaj S Mehta
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Division of Gastroenterology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA; Clinical and Translational Epidemiology Unit, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Xueyang Dong
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA 02138, USA
| | - Lindsey M Pieper
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Lorenzo Ramirez
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Margaret Alexander
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Sina Kiamehr
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA 02138, USA
| | - Peter J Turnbaugh
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA 94143, USA; Chan Zuckerberg Biohub-San Francisco, San Francisco, CA 94158, USA
| | - Curtis Huttenhower
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA; Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA; Harvard Chan Microbiome in Public Health Center, Harvard T.H. Chan School of Public Health, Harvard University, Boston, MA 02115, USA
| | - Andrew T Chan
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Division of Gastroenterology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA; Clinical and Translational Epidemiology Unit, Massachusetts General Hospital, Boston, MA 02114, USA; Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA; Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Emily P Balskus
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA 02138, USA; Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Howard Hughes Medical Institute, Harvard University, Cambridge, MA 02138, USA.
| |
Collapse
|
218
|
Mokhtari N, Ahmadi N, Moradi S, Farmani S, Kheyrani E, Dolatabadi NF. Experimental and in silico analysis of LINC01279 expression in tumor of patients with breast cancer. J Appl Genet 2024:10.1007/s13353-024-00908-6. [PMID: 39465460 DOI: 10.1007/s13353-024-00908-6] [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: 09/08/2023] [Revised: 08/18/2024] [Accepted: 09/22/2024] [Indexed: 10/29/2024]
Abstract
Breast cancer (BC) is characterized by the increase of malignant cells in the breast. The malignant cells begin in the lining of the breast milk glands or ducts (ductal epithelium). BC is the most frequent cancer in women, but it may also occur in males. Long non-coding RNAs (lncRNA) have been demonstrated to control the development and incidence of cancer. However, some lncRNAs experience potential changes in BC, but their role has not been well studied. LINC01279 is known as a valuable biomarker in gastric cancer but has not yet been studied in BC. Changes in LINC01279 expression levels in BC samples were investigated by microarray. Q-PCR was also used to evaluate the expression of LINC01279 in the tumor and normal adjacent samples of 30 BC patients. The LINC01279 co-expressed gene module was discovered using weighted gene correlation network analysis (WGCNA) on the relevant dataset. The top ten hub genes were determined using gene ontology (GO) functional enrichments on the co-expressed gene module. The results of the bioinformatics study showed an increase in LINC01279 expression levels (log2FC = 3.228749561, adj.P.Val = 1.69E - 12) in tumor samples compared to normal marginal tissue. Q-PCR results also showed a significant increase in LINC01279 expression (P-value = 0.0005) in tumor samples. WGCNA analysis identified that the black module is the LINC01279 co-expressed module, and functional annotation analysis of black module genes enriched in significant cancer-related pathways and processes, including cell growth and/or maintenance, regulation of immune response, regulation of cell proliferation, and epithelial-to-mesenchymal transition (EMT). Regarding the real-time PCR results, the analysis of expression patterns has illuminated a distinct association between the heightened expression levels of LINC01279, and the stages of cancer progression as well as the metastatic potential of tumors. However, intriguingly, our observations have failed to reveal any statistically significant correlations between the relative expression of LINC01279 and tumor grade classification, or the presence of ER, PR, and HER2 biomarkers. The present study could provide a new perspective on the molecular regulatory. Processes associated with BC pathogenic mechanisms are linked to the LINC01279, although further research is needed on the possible role of this lncRNA in BC.
Collapse
Affiliation(s)
- Negar Mokhtari
- Department of Cellular and Molecular Biology, Islamic Azad University, Najafabad Branch, Isfahan, Iran
| | - Najmeh Ahmadi
- Departmant of Medical Laboratory Sciences, School of Paramedical Sciences, Gerash University of Medical Sciences, Gerash, Iran
| | - Sahar Moradi
- Department of Medical Laboratory Sciences, School of Allied Medical Sciences, Isfahan, Iran
| | - Shiva Farmani
- Department of Biology, Faculty of Basic Sciences, Yazd University, Yazd, Iran
| | | | | |
Collapse
|
219
|
Zhao X, Ding A, Chen P, Zhang L, Qu J, Bao B. Celosia cristata L.-an underutilized Chinese medicine: A review of the ethnic applications, phytochemistry, pharmacology, quality control and toxicity. JOURNAL OF ETHNOPHARMACOLOGY 2024; 333:118479. [PMID: 38909823 DOI: 10.1016/j.jep.2024.118479] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Revised: 05/22/2024] [Accepted: 06/20/2024] [Indexed: 06/25/2024]
Affiliation(s)
- Xian Zhao
- Department of Pharmacy, Shaanxi Provincial Hospital of Tuberculosis Prevention and Treatment (The Fifth People's Hospital of Shaanxi Province), Xi'an, 710110, China
| | - Anwei Ding
- Jiangsu Collaborative Innovation Centre of Chinese Medicinal Resources Industrialization, National and Local Collaborative Engineering Centre of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Peidong Chen
- Jiangsu Collaborative Innovation Centre of Chinese Medicinal Resources Industrialization, National and Local Collaborative Engineering Centre of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Li Zhang
- Jiangsu Collaborative Innovation Centre of Chinese Medicinal Resources Industrialization, National and Local Collaborative Engineering Centre of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Jian Qu
- Medical Institute, Qinghai University, Xining, 810016, China
| | - Beihua Bao
- Jiangsu Collaborative Innovation Centre of Chinese Medicinal Resources Industrialization, National and Local Collaborative Engineering Centre of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| |
Collapse
|
220
|
Zhao Y, Chen X, Huang Y, Zhang Z, Wang K, Zou D, Ma T. Transcriptomic Insights into Hub Genes, Immune Infiltration, and Candidate Drugs in Erosive Esophagitis. J Inflamm Res 2024; 17:7745-7760. [PMID: 39494202 PMCID: PMC11529285 DOI: 10.2147/jir.s479032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2024] [Accepted: 09/17/2024] [Indexed: 11/05/2024] Open
Abstract
Purpose This study aimed to investigate gene expression profiles, identify potential hub genes, and predict drugs for patients with erosive esophagitis (EE). Despite its clinical significance, molecular-level exploration of this condition has been limited. Patients and Methods RNA sequencing was performed on clinical biopsy samples from eight EE patients and eight healthy controls. Integrated bioinformatic tools were then utilized to analyze the data, including functional enrichment analysis, protein-protein interaction network analysis, weighted gene co-expression network analysis, immune infiltration analysis, and identification of small-molecule compounds. Additionally, the expressions of the identified hub genes were assessed in clinical samples. Results A total of 2801 genes with differential expression were identified, including four potential hub genes: SOX9, SPP1, TIMP1, and TLR4. Moreover, the overexpression of these hub genes was verified in clinical samples. Analysis of Immune infiltration indicated an imbalance in the distribution of immune cell types in patients with EE. Correlation analysis between immune cells and hub genes unveiled noteworthy relationships. Specifically, SOX9 exhibited a negative correlation with CD8 T cells but a positive correlation with resting memory CD4 T cells. SPP1 displayed a positive correlation with naïve B cells, while TIMP1 exhibited a negative correlation with resting dendritic cells. Furthermore, the study identified ten small-molecule drugs with potential therapeutic effects for EE, including loreclezole and mercaptopurine. Conclusion This study provides valuable insights into the molecular understanding of EE, offering new perspectives on disease mechanisms. The findings may inspire further research leading to the development of novel treatment strategies for EE.
Collapse
Affiliation(s)
- Ye Zhao
- Department of Gastroenterology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People’s Republic of China
| | - Xi Chen
- Department of Gastroenterology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People’s Republic of China
| | - Yuhan Huang
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, People’s Republic of China
| | - Zhihan Zhang
- Department of Pathology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People’s Republic of China
| | - Kui Wang
- Department of Gastroenterology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People’s Republic of China
| | - Duowu Zou
- Department of Gastroenterology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People’s Republic of China
| | - Teng Ma
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, People’s Republic of China
| |
Collapse
|
221
|
Kong X, Wang W, Chen S, Song M, Zhi Y, Cai Y, Zhang H, Shen X. Comparative study of lysine acetylation in Vesicomyidae clam Archivesica marissinica and the manila clam Ruditapes philippinarum: adaptation mechanisms in cold seep environments. BMC Genomics 2024; 25:1006. [PMID: 39465380 PMCID: PMC11514971 DOI: 10.1186/s12864-024-10916-9] [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: 05/31/2024] [Accepted: 10/17/2024] [Indexed: 10/29/2024] Open
Abstract
BACKGROUND The deep-sea cold seep zone is characterized by high pressure, low temperature, darkness, and oligotrophy. Vesicomyidae clams are the dominant species within this environment, often forming symbiotic relationships with chemosynthetic microbes. Understanding the mechanisms by which Vesicomyidae clams adapt to the cold seep environment is significant. Acetylation modification of lysine is known to play a crucial role in various metabolic processes. Consequently, investigating the role of lysine acetylation in the adaptation of Vesicomyidae clams to deep-sea environments is worthwhile. So, a comparative study of lysine acetylation in cold seep clam Archivesica marissinica and shallow water shellfish Ruditapes philippinarum was conducted. RESULTS A total of 539 acetylated proteins were identified with 1634 acetylation sites. Conservative motif enrichment analysis revealed that the motifs -KacR-, -KacT-, and -KacF- were the most conserved. Subsequent gene ontology (GO) and Kyoto encyclopedia of genes and genomes (KEGG) enrichment analyses were conducted on significantly differentially expressed acetylated proteins. The GO enrichment analysis indicated that acetylated proteins are crucial in various biological processes, including cellular response to stimulation, and other cellular processes ( p < 0.05 and false discovery rate (FDR) < 0.25). The results of KEGG enrichment analysis indicated that acetylated proteins are involved in various cellular processes, including tight junction, motor proteins, gap junction, phagosome, cGMP-PKG signaling pathways, endocytosis, glycolysis/gluconeogenesis, among others (p < 0.05 and FDR < 0.25). Notably, a high abundance of lysine acetylation was observed in the glycolysis/glycogenesis pathways, and the acetylation of glyceraldehyde 3-phosphate dehydrogenase might facilitate ATP production. Subsequent investigation into acetylation modifications associated with deep-sea adaptation revealed the specific identification of key acetylated proteins. Among these, the adaptation of cold seep clam hemoglobin and heat shock protein to high hydrostatic pressure and low temperature might involve an increase in acetylation levels. Acetylation of arginine kinase might be related to ATP production and interaction with symbiotic bacteria. Myosin heavy chain (Ama01085) has the most acetylation sites and might improve the actomyosin system stability through acetylation. Further validation is required for the acetylation modification from Vesicomyidae clams. CONCLUSION A novel comparative analysis was undertaken to investigate the acetylation of lysine in Vesicomyidae clams, yielding novel insights into the regulatory role of lysine acetylation in deep-sea organisms. The findings present many potential proteins for further exploration of acetylation functions in cold seep clams and other deep-sea mollusks.
Collapse
Affiliation(s)
- Xue Kong
- School of Marine Science and Fisheries, Jiangsu Ocean University, Lianyungang, 222000, China
- Co-Innovation Center of Jiangsu Marine Bio-Industry Technology, Jiangsu Ocean University, Lianyungang, 222000, China
| | - Wei Wang
- School of Marine Science and Fisheries, Jiangsu Ocean University, Lianyungang, 222000, China
- Co-Innovation Center of Jiangsu Marine Bio-Industry Technology, Jiangsu Ocean University, Lianyungang, 222000, China
| | - Sunan Chen
- School of Marine Science and Fisheries, Jiangsu Ocean University, Lianyungang, 222000, China
- Co-Innovation Center of Jiangsu Marine Bio-Industry Technology, Jiangsu Ocean University, Lianyungang, 222000, China
| | - Manzong Song
- School of Marine Science and Fisheries, Jiangsu Ocean University, Lianyungang, 222000, China
- Co-Innovation Center of Jiangsu Marine Bio-Industry Technology, Jiangsu Ocean University, Lianyungang, 222000, China
| | - Ying Zhi
- School of Marine Science and Fisheries, Jiangsu Ocean University, Lianyungang, 222000, China
| | - Yuefeng Cai
- School of Marine Science and Fisheries, Jiangsu Ocean University, Lianyungang, 222000, China
- Co-Innovation Center of Jiangsu Marine Bio-Industry Technology, Jiangsu Ocean University, Lianyungang, 222000, China
| | - Haibin Zhang
- Institute of Deep-sea Science and Engineering, Chinese Academy of Sciences, Sanya, 572000, China
| | - Xin Shen
- School of Marine Science and Fisheries, Jiangsu Ocean University, Lianyungang, 222000, China.
- Co-Innovation Center of Jiangsu Marine Bio-Industry Technology, Jiangsu Ocean University, Lianyungang, 222000, China.
| |
Collapse
|
222
|
Deng M, Chen S, Wu J, Su L, Xu Z, Jiang C, Sheng L, Yang X, Zeng L, Wang J, Dai W. Exploring the anti-inflammatory and immune regulatory effects of Taohe Chengqi decoction in sepsis-induced lung injury. JOURNAL OF ETHNOPHARMACOLOGY 2024; 333:118404. [PMID: 38824977 DOI: 10.1016/j.jep.2024.118404] [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: 01/14/2024] [Revised: 05/24/2024] [Accepted: 05/27/2024] [Indexed: 06/04/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Sepsis presents complex pathophysiological challenges. Taohe Chengqi Decoction (THCQ), a traditional Chinese medicine, offers potential in managing sepsis-related complications, though its exact mechanisms are not fully understood. AIM OF THE STUDY This research aimed to assess the therapeutic efficacy and underlying mechanisms of THCQ on sepsis-induced lung injury. MATERIALS AND METHODS The study began with validating THCQ's anti-inflammatory effects through in vitro and in vivo experiments. Network pharmacology was employed for mechanistic exploration, incorporating GO, KEGG, and PPI analyses of targets. Hub gene-immune cell correlations were assessed using CIBERSORT, with further scrutiny at clinical and single-cell levels. Molecular docking explored THCQ's drug-gene interactions, culminating in qPCR and WB validations of hub gene expressions in sepsis and post-THCQ treatment scenarios. RESULTS THCQ demonstrated efficacy in modulating inflammatory responses in sepsis, identified through network pharmacology. Key genes like MAPK14, MAPK3, MMP9, STAT3, LYN, AKT1, PTPN11, and HSP90AA1 emerged as central targets. Molecular docking revealed interactions between these genes and THCQ components. qPCR results showed significant modulation of these genes, indicating THCQ's potential in reducing inflammation and regulating immune responses in sepsis. CONCLUSION This study sheds light on THCQ's anti-inflammatory and immune regulatory mechanisms in sepsis, providing a foundation for further research and potential clinical application.
Collapse
Affiliation(s)
- Mingtao Deng
- Shangrao Key Laboratory of Health Hazards and Bioprevention of Heavy Metals, Jiangxi Medical College, No. 399 Zhimin Avenue, Xinzhou District, Shangrao, Jiangxi Province, 334000, People's Republic of China; Department of Medical Technology, Jiangxi Medical College, No. 399 Zhimin Avenue, Xinzhou District, Shangrao, Jiangxi Province, 334000, People's Republic of China
| | - Siqi Chen
- Shangrao Key Laboratory of Health Hazards and Bioprevention of Heavy Metals, Jiangxi Medical College, No. 399 Zhimin Avenue, Xinzhou District, Shangrao, Jiangxi Province, 334000, People's Republic of China; Department of Medical Technology, Jiangxi Medical College, No. 399 Zhimin Avenue, Xinzhou District, Shangrao, Jiangxi Province, 334000, People's Republic of China
| | - Jian Wu
- Department of Medical Technology, Jiangxi Medical College, No. 399 Zhimin Avenue, Xinzhou District, Shangrao, Jiangxi Province, 334000, People's Republic of China
| | - Liling Su
- Shangrao Key Laboratory of Health Hazards and Bioprevention of Heavy Metals, Jiangxi Medical College, No. 399 Zhimin Avenue, Xinzhou District, Shangrao, Jiangxi Province, 334000, People's Republic of China
| | - Zijin Xu
- Shangrao Key Laboratory of Health Hazards and Bioprevention of Heavy Metals, Jiangxi Medical College, No. 399 Zhimin Avenue, Xinzhou District, Shangrao, Jiangxi Province, 334000, People's Republic of China
| | - Changrun Jiang
- Department of Critical Care Medicine, The First Affiliated Hospital of Jiangxi Medical College, No. 31 Qingfeng Road, Xinzhou District, Shangrao, Jiangxi Province, 334000, People's Republic of China
| | - Lei Sheng
- Department of Critical Care Medicine, The First Affiliated Hospital of Jiangxi Medical College, No. 31 Qingfeng Road, Xinzhou District, Shangrao, Jiangxi Province, 334000, People's Republic of China
| | - Xinyi Yang
- Department of Critical Care Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, No. 17 Yongwaizheng Street, Dong Lake District, Nanchang, Jiangxi Province, 330000, People's Republic of China
| | - Long Zeng
- Shangrao Key Laboratory of Health Hazards and Bioprevention of Heavy Metals, Jiangxi Medical College, No. 399 Zhimin Avenue, Xinzhou District, Shangrao, Jiangxi Province, 334000, People's Republic of China
| | - Jingwei Wang
- Shangrao Key Laboratory of Health Hazards and Bioprevention of Heavy Metals, Jiangxi Medical College, No. 399 Zhimin Avenue, Xinzhou District, Shangrao, Jiangxi Province, 334000, People's Republic of China
| | - Wei Dai
- Shangrao Key Laboratory of Health Hazards and Bioprevention of Heavy Metals, Jiangxi Medical College, No. 399 Zhimin Avenue, Xinzhou District, Shangrao, Jiangxi Province, 334000, People's Republic of China; Department of Critical Care Medicine, The First Affiliated Hospital of Jiangxi Medical College, No. 31 Qingfeng Road, Xinzhou District, Shangrao, Jiangxi Province, 334000, People's Republic of China; Department of Clinical Medicine, Jiangxi Medical College, No. 399 Zhimin Avenue, Xinzhou District, Shangrao, Jiangxi Province, 334000, People's Republic of China.
| |
Collapse
|
223
|
Madasu PK, Chandran T. Structural insights into the toxicity of type II ribosome inactivating proteins (RIPs): a molecular dynamics study. J Biomol Struct Dyn 2024:1-12. [PMID: 39466135 DOI: 10.1080/07391102.2024.2419855] [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: 10/19/2023] [Accepted: 05/17/2024] [Indexed: 10/29/2024]
Abstract
Ribosome Inactivating Proteins (RIPs) act by irreversibly depurinating the 28S rRNA ricin-sarcin loop (SRL) of the eukaryotic ribosome resulting in protein synthesis inhibition. In general, they consist of two variants: Type I which is single chained (∼30 kDa), and Type II, a more toxic variant which is a Type I N-glycosidase chain covalently linked to a lectin chain. These proteins are believed to play a pivotal role in defence mechanisms. Intriguingly, non-toxic variants of such toxic proteins do exist in nature. To explore their mode of action, in the present study we have selected three toxic (Ricin, Ebulin and HmRIP) as well as two non-toxic (BGSL and SGSL) RIPs and performed molecular docking and molecular dynamic simulations with the SRL loop. This study throws light on the structural stability and plasticity of the toxic and non-toxic RIP complexes. Furthermore, analysis of the active site cavity volume and binding free energy calculations reveal that the SRL, particularly the specific adenine (A4605), is relatively unstable in the case of non-toxic RIPs which is also supported by the free binding energy calculations, and the pocket size analysis indicates the abnormal increase in active site cavity volume of non-toxic RIPs with time. This first-of-its-kind comprehensive study of toxic and non-toxic RIPs gives insights about the mode of action and the dynamic nature of their interaction with the SRL loop. These observations will be helpful in the development of toxoids against RIPs and also in designing novel therapeutic approaches against human diseases.
Collapse
Affiliation(s)
- Pavan K Madasu
- Biomolecular Structure and Dynamics Group, Department of Biotechnology, National Institute of Technology, Warangal, India
| | - Thyageshwar Chandran
- Biomolecular Structure and Dynamics Group, Department of Biotechnology, National Institute of Technology, Warangal, India
| |
Collapse
|
224
|
Lin R, Li H, Lai L, Yang F, Qiu J, Lin W, Bao X, Pan C, Lin W, Jiang X. Analysis of genetic structure and identification of important genes associated with muscle growth in Fujian Muscovy duck. Poult Sci 2024; 103:104445. [PMID: 39504826 DOI: 10.1016/j.psj.2024.104445] [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: 07/03/2024] [Revised: 10/02/2024] [Accepted: 10/25/2024] [Indexed: 11/08/2024] Open
Abstract
Fujian Muscovy duck is a well-known meat waterfowl in Fujian Province due to its high meat production, superior breeding potential, and strong resistance. To fully explore the genetic characteristics of these advantages, Fujian black Muscovy duck and white Muscovy duck were used for whole-genome re-sequencing and transcriptome analyses. Population structure analysis showed significant differentiation between the two feather strains. Runs of homozygosity analysis indicated a stronger artificial influence on the black-feathered strain, with ROH island genes notably enriched in muscle tissue-related terms and pathways. Selective sweep and transcriptome analysis revealed a significant enrichment of genes linked to muscle tissue and muscle fiber-related terms and pathways. Key candidate genes identified, such as MEF2C, MYOZ2, and METTL21C, are believed to play crucial roles in meat production in Fujian Muscovy duck. This study offers a new perspective on improving meat production in Fujian Muscovy duck, which can benefit breeding strategies and production management.
Collapse
Affiliation(s)
- Ruiyi Lin
- College of Animal Sciences, Fujian Agriculture and Forestry University, Fuzhou, Fujian, China, 350002
| | - Huihuang Li
- College of Animal Sciences, Fujian Agriculture and Forestry University, Fuzhou, Fujian, China, 350002
| | - Lianjie Lai
- College of Animal Sciences, Fujian Agriculture and Forestry University, Fuzhou, Fujian, China, 350002
| | - Fan Yang
- College of Animal Sciences, Fujian Agriculture and Forestry University, Fuzhou, Fujian, China, 350002
| | - Jialing Qiu
- General Animal husbandry Station of Fujian Province, Fuzhou, Fujian, China, 350003
| | - Weilong Lin
- College of Animal Sciences, Fujian Agriculture and Forestry University, Fuzhou, Fujian, China, 350002
| | - Xinguo Bao
- College of Animal Sciences, Fujian Agriculture and Forestry University, Fuzhou, Fujian, China, 350002
| | - Chengfu Pan
- College of Animal Sciences, Fujian Agriculture and Forestry University, Fuzhou, Fujian, China, 350002
| | - Weimin Lin
- College of Animal Sciences, Fujian Agriculture and Forestry University, Fuzhou, Fujian, China, 350002
| | - Xiaobing Jiang
- General Animal husbandry Station of Fujian Province, Fuzhou, Fujian, China, 350003.
| |
Collapse
|
225
|
Zhu R, Gao Z, Wu S, Ma S, Zhu Y, Zhang S, Zhang Y, Zeng H, Ma C, Zhao J, Ye J, Zhang W. Multi-omics and network pharmacology approaches reveal Gui-Ling-Ji alleviates oligoasthenoteratozoospermia by regulating arachidonic acid pathway. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 135:156184. [PMID: 39488872 DOI: 10.1016/j.phymed.2024.156184] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Revised: 10/15/2024] [Accepted: 10/23/2024] [Indexed: 11/05/2024]
Abstract
BACKGROUND Gui-Ling-Ji (GLJ) described in the ancient medical book 'Yunji Qijian' is a traditional Chinese medicine formula used to improve male fertility. It is now available for the treatment of oligoasthenoteratozoospermia (OAT). However, the active ingredients and mechanism of GLJ are not clear. PURPOSE The aim of this study was to clarify the active ingredients and mechanism of GLJ in OAT. METHODS Firstly, the cyclophosphamide-induced OAT rat model was established to evaluate the efficacy of GLJ. Secondly, serum/urine-based metabolomics and lipidomics and tissue-based transcriptomics were performed to discover the differential metabolites and genes in rats. Furthermore, network pharmacology was constructed to explore the associated mechanisms based on the results of multi-omics analysis. Finally, cellular experiment on testicular mesenchymal stromal cells (TM3) was used to validate the active ingredients and the key metabolic pathway. RESULTS Rats were administered GLJ by gavage every day for 3 weeks. Testicular damage and weight loss caused by cyclophosphamide were restored in rats, the sperm count and motility were improved, and levels of luteinizing hormone (LH), follicle-stimulating hormone (FSH) and testosterone (T) secretion were also elevated. Compared to the metabolites of OAT rats, 51 and 37 differential metabolites regulated by GLJ were identified from serum and urine respectively, 54 lipid differential metabolites regulated by GLJ were identified by lipidomics. At the same time, 23 of the 258 differential genes were found to be regulated by OAT rats and then reverse-regulated by GLJ. Network pharmacology has identified 13 pathways (Steroid hormone biosynthesis, Taurine and hypotaurine metabolism, Primary bile acid biosynthesis, Linoleic acid metabolism, Retinol metabolism, Glycerophospholipid metabolism, Ether lipid metabolism, Sphingolipid metabolism, Arachidonic acid metabolism, Glutathione metabolism, Arginine biosynthesis, Arginine and proline metabolism, D-Arginine and D-ornithine metabolism), four metabolites (arachidonic acid, oestrone sulphate, phosphatidylglycerol choline and sphingomyelin) and 15 targets (ABCB11, ALDH18A1, CCL3, CD244, CIITA, CYP2C8, DLL1, ITGA4, ESR1, AR, ABCB1, ABCC1, ALB, PLA2G1B and NOS2). GLJ, psoralen, isopsoralen, liquiritin, isoliquiritin, liquiritigenin, and ginsenoside Ro could significantly promote T secretion from TM3 cells. Additionally, arachidonic acid metabolism particularly the cyclooxygenase pathway, is closely related to the promotion of testosterone secretion by GLJ in TM3. CONCLUSION GLJ has a therapeutic efficacy in cyclophosphamide-induced OAT rats, which can modulate the disorders of lipid metabolism and amino acid metabolism. Arachidonic acid metabolism may be a key pathway, and six prototype compounds are potential key active ingredients for GLJ.
Collapse
Affiliation(s)
- Renwen Zhu
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China..
| | - Ziqing Gao
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Shiyu Wu
- School of Pharmacy, Fujian University of Traditional Chinese Medicine, Fuzhou, 350122, China
| | - Siyi Ma
- School of Pharmacy, East China University of Science and Technology, Shanghai, 200237, China
| | - Yiqing Zhu
- School of Pharmacy, Fujian University of Traditional Chinese Medicine, Fuzhou, 350122, China
| | - Shiyu Zhang
- School of Pharmacy, Henan University, Kaifeng, 475004, China
| | - Yuhao Zhang
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Huawu Zeng
- School of Pharmacy, Naval Medical University, Shanghai 200433, China
| | - Chi Ma
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Jing Zhao
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China..
| | - Ji Ye
- School of Pharmacy, Naval Medical University, Shanghai 200433, China..
| | - Weidong Zhang
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.; School of Pharmacy, Naval Medical University, Shanghai 200433, China.; School of Pharmacy, Fujian University of Traditional Chinese Medicine, Fuzhou, 350122, China.; School of Pharmacy, East China University of Science and Technology, Shanghai, 200237, China..
| |
Collapse
|
226
|
Tran MN, Kim NS, Lee S. Biological network comparison identifies a novel synergistic mechanism of Ginseng Radix-Astragali Radix herb pair in cancer-related fatigue. JOURNAL OF ETHNOPHARMACOLOGY 2024; 333:118447. [PMID: 38885914 DOI: 10.1016/j.jep.2024.118447] [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: 12/21/2023] [Revised: 06/01/2024] [Accepted: 06/06/2024] [Indexed: 06/20/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Ginseng Radix and Astragali Radix are commonly combined to tonify Qi and alleviate fatigue. Previous studies have employed biological networks to investigate the mechanisms of herb pairs in treating different diseases. However, these studies have only elucidated a single network for each herb pair, without emphasizing the superiority of the herb combination over individual herbs. AIM OF THE STUDY This study proposes an approach of comparing biological networks to highlight the synergistic effect of the pair in treating cancer-related fatigue (CRF). METHODS The compounds and targets of Ginseng Radix, Astragali Radix, and CRF diseases were collected and predicted using different databases. Subsequently, the overlapping targets between herbs and disease were imported into the STRING and DAVID tools to build protein-protein interaction (PPI) networks and analyze enriched KEGG pathways. The biological networks of Ginseng Radix and Astragali Radix were compared separately or together using the DyNet application. Molecular docking was used to verify the predicted results. Further, in vitro experiments were conducted to validate the synergistic pathways identified in in silico studies. RESULTS In the PPI network comparison, the combination created 89 new interactions and an increased average degree (11.260) when compared to single herbs (10.296 and 9.394). The new interactions concentrated on HRAS, STAT3, JUN, and IL6. The topological analysis identified 20 core targets of the combination, including three Ginseng Radix-specific targets, three Astragali Radix-specific targets, and 14 shared targets. In KEGG enrichment analysis, the combination regulated additional signaling pathways (152) more than Ginseng Radix (146) and Astragali Radix (134) alone. The targets of the herb pair synergistically regulated cancer pathways, specifically hypoxia-inducible factor 1 (HIF-1) signaling pathway. In vitro experiments including enzyme-linked immunosorbent assay and Western blot demonstrated that two herbs combination could up-regulate HIF-1α signaling pathway at different combined concentrations compared to either single herb alone. CONCLUSION The herb pair increased protein interactions and adjusted metabolic pathways more than single herbs. This study provides insights into the combination of Ginseng Radix and Astragali Radix in clinical practice.
Collapse
Affiliation(s)
- Minh Nhat Tran
- Korean Medicine Data Division, Korea Institute of Oriental Medicine, Daejeon, Republic of Korea; Korean Convergence Medical Science, University of Science and Technology, Daejeon, Republic of Korea; Faculty of Traditional Medicine, Hue University of Medicine and Pharmacy, Hue University, Thua Thien Hue, Viet Nam.
| | - No Soo Kim
- Korean Medicine Convergence Research Division, Korea Institute of Oriental Medicine, Daejeon, Republic of Korea.
| | - Sanghun Lee
- Korean Medicine Data Division, Korea Institute of Oriental Medicine, Daejeon, Republic of Korea; Korean Convergence Medical Science, University of Science and Technology, Daejeon, Republic of Korea.
| |
Collapse
|
227
|
Winardi K, Mach J, McKay MJ, Molloy MP, Mitchell SJ, MacArthur MR, McKenzie C, Le Couteur DG, Hilmer SN. Chronic polypharmacy, monotherapy, and deprescribing: Understanding complex effects on the hepatic proteome of aging mice. Aging Cell 2024:e14357. [PMID: 39462793 DOI: 10.1111/acel.14357] [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: 02/08/2024] [Revised: 08/09/2024] [Accepted: 09/16/2024] [Indexed: 10/29/2024] Open
Abstract
Polypharmacy (use of ≥5 concurrent medications) is highly prevalent among older adults to manage chronic diseases and is linked to adverse geriatric outcomes, including physical and cognitive functional impairments, falls, frailty, hospitalization, and mortality. Deprescribing (withdrawal) is a potential strategy to manage polypharmacy. The broad molecular changes by which polypharmacy causes harm and deprescribing may be beneficial are unknown and unfeasible to study rigorously in tissue from geriatric patients. Therefore, in a randomized controlled trial, we administered therapeutic doses of commonly used chronic medications (oxycodone, oxybutynin, citalopram, simvastatin, or metoprolol) as monotherapy or concurrently (polypharmacy) from middle-age (12 months) to old-age (26 months) to male C57BL/6J (B6) mice and deprescribed (gradually withdrew) treatments in a subset from age 21 months. We compared drug-related hepatic effects by applying proteomics along with transcriptomics and histology. We found that monotherapy effects on hepatic proteomics were limited but significant changes were seen with polypharmacy (93% unique to polypharmacy). Polypharmacy altered the hepatic expression of proteins involved in immunity, and in drug, cholesterol, and amino acid metabolism, accompanied by higher serum drug levels than monotherapies. Deprescribing not only reversed some effects but also caused irreversible and novel changes in the hepatic proteome. Furthermore, our study identified several hepatic protein co-expressed modules that are associated with clinically relevant adverse geriatric outcomes, such as mobility, frailty, and activities of daily living. This study highlights the complex molecular changes following aging, chronic polypharmacy, and deprescribing. Further exploration of these mechanistic pathways may inform management of polypharmacy and deprescribing in older adults.
Collapse
Affiliation(s)
- Kevin Winardi
- Laboratory of Ageing and Pharmacology, Kolling Institute, Faculty of Medicine and Health, The University of Sydney and the Northern Sydney Local Health District, Sydney, New South Wales, Australia
| | - John Mach
- Laboratory of Ageing and Pharmacology, Kolling Institute, Faculty of Medicine and Health, The University of Sydney and the Northern Sydney Local Health District, Sydney, New South Wales, Australia
| | - Matthew J McKay
- Bowel Cancer and Biomarker Laboratory, School of Medical Science, Faculty of Medicine and Health, University of Sydney, Sydney, New South Wales, Australia
| | - Mark P Molloy
- Bowel Cancer and Biomarker Laboratory, School of Medical Science, Faculty of Medicine and Health, University of Sydney, Sydney, New South Wales, Australia
| | - Sarah J Mitchell
- Ludwig Princeton Branch, Princeton University, Princeton, New Jersey, USA
| | | | - Catriona McKenzie
- Department of Tissue Pathology and Diagnostic Oncology, Royal Prince Alfred Hospital, Sydney, New South Wales, Australia
- Sydney Medical School, University of Sydney, Sydney, New South Wales, Australia
| | - David G Le Couteur
- Charles Perkins Centre, University of Sydney, Sydney, New South Wales, Australia
- ANZAC Research Institute, University of Sydney and Concord Hospital, Concord, New South Wales, Australia
- Centre for Education and Research on Ageing, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
| | - Sarah N Hilmer
- Laboratory of Ageing and Pharmacology, Kolling Institute, Faculty of Medicine and Health, The University of Sydney and the Northern Sydney Local Health District, Sydney, New South Wales, Australia
| |
Collapse
|
228
|
Savla SR, Bhatt LK. Exploration of anti-atherosclerotic activity of 1,8-cineole through network pharmacology, molecular docking, and in vivo efficacy studies in high-fat-diet-induced atherosclerosis in hamsters. Mol Divers 2024:10.1007/s11030-024-11015-3. [PMID: 39463214 DOI: 10.1007/s11030-024-11015-3] [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: 05/31/2024] [Accepted: 10/09/2024] [Indexed: 10/29/2024]
Abstract
The anti-atherogenic potential of liver X receptors (LXRs) has been attributed to their inhibitory role in macrophage-mediated inflammation and promotion of reverse cholesterol transport. This study aimed to evaluate the efficacy of an LXR agonist, 1,8-cineole (Eucalyptol), in atherosclerosis through network pharmacology, molecular docking, and in vivo efficacy studies in high-fat-diet-induced atherosclerosis in hamsters. Network pharmacology analysis was performed by identifying potential targets of 1,8-Cineole and atherosclerosis, followed by the construction of component-target-disease and protein-protein interaction networks. Gene Ontology and KEGG pathway enrichment analysis of targets were performed. The top 5 targets were selected for molecular docking studies. Atherosclerosis was induced in male Golden Syrian hamsters, and the results of network pharmacology were verified. Fifty-one overlapped targets were identified for 1,8-cineole and atherosclerosis. In the protein-protein interaction studies, the top 5 ranked proteins were PPARG, FXR, ABCA-1, ABCG1, and LXRΑ. KEGG pathway analysis and molecular docking showed that ABCA-1 and LXRΑ were correlated in atherosclerosis. Animal studies showed amelioration of atherosclerotic lesions in the aorta of animals treated with 1,8-cineole compared to disease control aortas. A dose-dependent attenuation in ABCA-1 levels and inflammatory markers was observed in animals treated with 1,8-cineole, comparable to its levels in normal animals. In conclusion, 1,8-cineole showed anti-atherosclerotic effects in Golden Syrian hamsters via LXRΑ-induced ABCA-1 overexpression.
Collapse
Affiliation(s)
- Shreya R Savla
- Department of Pharmacology, SVKM's Dr. Bhanuben Nanavati College of Pharmacy, Vile Parle (West), Mumbai, 400056, India
| | - Lokesh Kumar Bhatt
- Department of Pharmacology, SVKM's Dr. Bhanuben Nanavati College of Pharmacy, Vile Parle (West), Mumbai, 400056, India.
| |
Collapse
|
229
|
Shukla M, Sarkar RR. Differential cellular communication in tumor immune microenvironment during early and advanced stages of lung adenocarcinoma. Mol Genet Genomics 2024; 299:100. [PMID: 39460829 DOI: 10.1007/s00438-024-02193-8] [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: 04/30/2024] [Accepted: 10/04/2024] [Indexed: 10/28/2024]
Abstract
Heterogeneous behavior of each cell type and their cross-talks in tumor immune microenvironment (TIME) refers to tumor immunological heterogeneity that emerges during tumor progression and represents formidable challenges for effective anti-tumor immune response and promotes drug resistance. To comprehensively elucidate the heterogeneous behavior of individual cell types and their interactions across different stages of tumor development at system level, a computational framework was devised that integrates cell specific data from single-cell RNASeq into networks illustrating interactions among signaling and metabolic response genes within and between cells in TIME. This study identified stage specific novel markers which remodel the cross-talks, thereby facilitating immune stimulation. Particularly, multicellular knockout of metabolic gene APOE (Apolipoprotein E in mast cell, myeloid cell and fibroblast) combined with signaling gene CAV1 (Caveolin1 in endothelial and epithelial cells) resulted in the activation of T-cell mediated signaling pathways. Additionally, this knockout also initiated intervention of cytotoxic gene regulations during tumor immune cell interactions at the early stage of Lung Adenocarcinoma (LUAD). Furthermore, a unique interaction motif from multiple cells emerged significant in regulating the overall immune response at the advanced stage of LUAD. Most significantly, FCER1G (Fc Fragment of IgE Receptor Ig) was identified as the common regulator in activating the anti-tumor immune response at both stages. Predicted markers exhibited significant association with patient overall survival in patient specific dataset. This study uncovers the significance of signaling and metabolic interplay within TIME and discovers important targets to enhance anti-tumor immune response at each stage of tumor development.
Collapse
Affiliation(s)
- Mudita Shukla
- Chemical Engineering and Process Development Division, CSIR-National Chemical Laboratory, Pune, Maharashtra, India
- Academy of Scientific & Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Ram Rup Sarkar
- Chemical Engineering and Process Development Division, CSIR-National Chemical Laboratory, Pune, Maharashtra, India.
- Academy of Scientific & Innovative Research (AcSIR), Ghaziabad, 201002, India.
| |
Collapse
|
230
|
Chen Y, Huang M, You Z, Sa R, Zhao L, Ku C, Wang W, Duan X. Unveiling the genetic link and pathogenesis between psoriasis and IgA nephropathy based on Mendelian randomization and transcriptome data analyses. Arch Dermatol Res 2024; 316:717. [PMID: 39460798 DOI: 10.1007/s00403-024-03465-4] [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: 09/01/2024] [Revised: 10/07/2024] [Accepted: 10/17/2024] [Indexed: 10/28/2024]
Abstract
It has been reported that many people with psoriasis have been diagnosed with secondary IgA nephropathy (IgAN). However, the mechanisms behind the association between psoriasis and IgAN have not been well clarified. The connection between psoriasis and IgAN deserves deeper exploration. Mendelian randomization (MR) analysis would be employed to explore the link of causality between IgAN and psoriasis, psoriasis vulgaris, other and unspecified psoriasis, guttate psoriasis, and arthropathic psoriasis. Transcriptomic analyses were carried out against the Gene Expression Omnibus databases. We identified crosstalk genes through the analysis of Differentially expressed genes and weight gene co-expression network analysis. Functional annotations were enriched for these crosstalk genes. Subsequently, we established a protein-protein interaction network, and candidate genes would be discovered through the utilization of the MCODE and CytoHubba plug-in applications. Lastly, the predictive efficacy of these genes was examined via creating receiver operating characteristic curves. The MR analysis suggested that psoriasis vulgaris patients were at a higher risk for IgAN. [OR = 1.040, 95%CI (1.005,1.076), p = 0.026 < 0.05]. Additionally, arthropathic psoriasis may augment the incidence of IgAN [OR = 1.081, 95%CI (1.040-1.124), p < 0.01] in the European population. Through the analysis of DEGs and WGCNA, we identified 12 significant genes (NETO2, RRM2, SLAMF7, GBP1, KIF20A, CCL4, MMP1, IL1β, NDC80, CXCL9, C15orf48, GSTA3), which may be potential crosstalk genes between the two diseases. Then, the functional annotation results indicated that the crosstalk genes seemed primarily involved in immune and inflammatory responses. By establishing the PPI network, we further discovered that CXCL9, IL1β, CCL4, and MMP1 play a vital part in psoriasis and IgAN, and all have good diagnostic values. Our MR analysis provided evidence that genetic vulnerability to IgAN may be associated with an elevated risk of psoriasis vulgaris and arthropathic psoriasis respectively among Europeans. Doctors should be aware of these associations when patients with psoriasis present with renal dysfunction, especially those with psoriasis vulgaris and arthropathic psoriasis. Chronic inflammation, drug effects, and immunity may contribute to the generation and development of both diseases. IL1β, CXCL9, CCL4, and MMP1 may be core biomarkers for psoriasis and IgAN.
Collapse
Affiliation(s)
- Yingwen Chen
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, 100700, China
- Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Min Huang
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, 100700, China
| | - Ziqing You
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, 100700, China
- Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Rule Sa
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, 100700, China
- Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Lu Zhao
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, 100700, China
- Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Congwen Ku
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, 100700, China
- Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Wenying Wang
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, 100700, China.
| | - Xingwu Duan
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, 100700, China.
| |
Collapse
|
231
|
Gregorich ZR, Larson EJ, Zhang Y, Braz CU, Liu C, Ge Y, Guo W. Integrated proteomics and transcriptomics analysis reveals insights into differences in premature mortality associated with disparate pathogenic RBM20 variants. J Mol Cell Cardiol 2024; 197:S0022-2828(24)00173-1. [PMID: 39490642 DOI: 10.1016/j.yjmcc.2024.10.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Revised: 10/14/2024] [Accepted: 10/23/2024] [Indexed: 11/05/2024]
Abstract
Variants in RNA binding motif protein 20 (RBM20) are causative in a severe form of dilated cardiomyopathy referred to as RBM20 cardiomyopathy, yet the mechanisms are unclear. Moreover, the reason(s) for phenotypic heterogeneity in carriers with different pathogenic variants are similarly opaque. To gain insight, we carried out multi-omics analysis, including the first analysis of gene expression changes at the protein level, of mice carrying two different pathogenic variants in the RBM20 nuclear localization signal (NLS). Direct comparison of the phenotypes confirmed greater premature morality in S639G variant carrying mice compared to mice with the S637A variant despite similar cardiac remodeling and dysfunction. Analysis of differentially spliced genes uncovered alterations in the splicing of both RBM20 target genes and non-target genes, including several genes previously implicated in arrhythmia. Global proteomics analysis found that a greater number of proteins were differentially expressed in the hearts of Rbm20S639G mice relative to WT than in Rbm20S637A versus WT. Gene ontology analysis suggested greater mitochondrial dysfunction in Rbm20S639G mice, although direct comparison of protein expression in the hearts of Rbm20S639G versus Rbm20S637A mice failed to identify any significant differences. Similarly, few differences were found by direct comparison of gene expression at the transcript level in Rbm20S639G and Rbm20S637A despite greater coverage. Our data provide a comprehensive overview of gene splicing and expression differences associated with pathogenic variants in RBM20, as well as insights into the molecular underpinnings of phenotypic heterogeneity associated with different dilated cardiomyopathy-associated variants.
Collapse
Affiliation(s)
- Zachery R Gregorich
- Department of Animal and Dairy Sciences, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Eli J Larson
- Department of Chemistry, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Yanghai Zhang
- Department of Animal and Dairy Sciences, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Camila U Braz
- Department of Animal Sciences, University of Illinois Urbana-Champaign, Urbana, IL 61801, USA
| | - Chunling Liu
- Department of Animal and Dairy Sciences, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Ying Ge
- Department of Chemistry, University of Wisconsin-Madison, Madison, WI 53706, USA; Human Proteomics Program, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Wei Guo
- Department of Animal and Dairy Sciences, University of Wisconsin-Madison, Madison, WI 53706, USA.
| |
Collapse
|
232
|
Chen J, Tan Y, Chen Z, Yang H, Li X, Long X, Han Y, Yang J. Exosomes derived from primary cartilage stem/progenitor cells promote the repair of osteoarthritic chondrocytes by modulating immune responses. Int Immunopharmacol 2024; 143:113397. [PMID: 39461237 DOI: 10.1016/j.intimp.2024.113397] [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: 08/12/2024] [Revised: 10/10/2024] [Accepted: 10/10/2024] [Indexed: 10/29/2024]
Abstract
BACKGROUND Exosomes derived from primary chondrogenic stem/progenitor cells (CSPCs-EXOs) show promise in cartilage repair due to their immunomodulatory and regenerative properties. However, their specific therapeutic potential in osteoarthritis (OA), especially in modulating immune responses and enhancing chondrocyte function, requires further exploration. This study aims to clarify CSPCs-EXOs' effects on OA by investigating their role in chondrocyte proliferation, migration, inflammation inhibition, and cartilage regeneration. METHODS A rat model of osteoarthritis was established using monosodium iodoacetate (MIA). CSPCs-EXOs were isolated and characterized before being administered to the OA rats. Comprehensive transcriptomic analysis was conducted to identify differentially expressed genes (DEGs) and signaling pathways influenced by CSPCs-EXOs. Histopathological evaluation of cartilage tissue, immunohistochemistry, and in vitro assays were performed to assess chondrocyte proliferation, migration, inflammation, and intracellular environmental changes. RESULTS CSPCs-EXOs treatment significantly reduced OA-induced cartilage damage, shown by improved histopathological features, increased chondrocyte proliferation, migration, and enhanced cartilage matrix integrity. CSPCs-EXOs uniquely modulated immune pathways and enhanced cellular repair, setting them apart from traditional treatments. Transcriptomic analysis revealed regulation of immune response, inflammation, oxidative stress, and DNA repair pathways. CSPCs-EXOs downregulated inflammatory cytokines (TNF, IL-17) and upregulated pathways for cellular proliferation, migration, and metabolism. They also altered splicing patterns of DNA repair enzymes, indicating a role in boosting repair mechanisms. CONCLUSIONS CSPCs-EXOs promote cartilage repair in osteoarthritis by modulating immune responses, inhibiting inflammation, and improving the intracellular environment. These findings emphasize their innovative therapeutic potential and offer key insights into their regenerative mechanisms, positioning CSPCs-EXOs as a promising strategy for OA treatment and a foundation for future clinical applications in cartilage tissue engineering and regenerative medicine.
Collapse
Affiliation(s)
- Jing Chen
- Department of Neurosurgery, People's Hospital of Deyang City, Sichuan Clinical Research Center for Neurological Diseases, Deyang; Co-innovation Center of Neuroregeneration, Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education; Department of Orthopedics, Affiliated Hospital of Nantong University, Nantong University, Nantong, China
| | - Ya Tan
- Department of Neurosurgery, People's Hospital of Deyang City, Sichuan Clinical Research Center for Neurological Diseases, Deyang; Co-innovation Center of Neuroregeneration, Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education; Department of Orthopedics, Affiliated Hospital of Nantong University, Nantong University, Nantong, China
| | - Zhifeng Chen
- Department of Neurosurgery, People's Hospital of Deyang City, Sichuan Clinical Research Center for Neurological Diseases, Deyang; Co-innovation Center of Neuroregeneration, Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education; Department of Orthopedics, Affiliated Hospital of Nantong University, Nantong University, Nantong, China
| | - Hongwei Yang
- Affiliated Nantong Hospital 3 of Nantong University, Nantong Third People's Hospital, Nantong University, Nantong, China
| | - Xiaodi Li
- Chinese Medicine Modernization and Big Data Research Center, Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Nanjing, China
| | - Xiaodong Long
- Department of Neurosurgery, People's Hospital of Deyang City, Sichuan Clinical Research Center for Neurological Diseases, Deyang; Co-innovation Center of Neuroregeneration, Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education; Department of Orthopedics, Affiliated Hospital of Nantong University, Nantong University, Nantong, China.
| | - Yangyun Han
- Department of Neurosurgery, People's Hospital of Deyang City, Sichuan Clinical Research Center for Neurological Diseases, Deyang; Co-innovation Center of Neuroregeneration, Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education; Department of Orthopedics, Affiliated Hospital of Nantong University, Nantong University, Nantong, China.
| | - Jian Yang
- Department of Neurosurgery, People's Hospital of Deyang City, Sichuan Clinical Research Center for Neurological Diseases, Deyang; Co-innovation Center of Neuroregeneration, Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education; Department of Orthopedics, Affiliated Hospital of Nantong University, Nantong University, Nantong, China.
| |
Collapse
|
233
|
Chen JY, Li YF, Zhou Z, Jiang XM, Bi X, Yang MF, Zhao B. De novo mutations promote inflammation in children with STAT3 gain-of-function syndrome by affecting IL-1β expression. Int Immunopharmacol 2024; 140:112755. [PMID: 39098225 DOI: 10.1016/j.intimp.2024.112755] [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: 03/26/2024] [Revised: 07/09/2024] [Accepted: 07/22/2024] [Indexed: 08/06/2024]
Abstract
STAT3 gain-of-function syndrome, characterized by early-onset autoimmunity and primary immune regulatory disorder, remains poorly understood in terms of its immunological mechanisms. We employed whole-genome sequencing of familial trios to elucidate the pivotal role of de novo mutations in genetic diseases. We identified 37 high-risk pathogenic loci affecting 23 genes, including a novel STAT3 c.508G>A mutation. We also observed significant down-regulation of pathogenic genes in affected individuals, potentially associated with inflammatory responses regulated by PTPN14 via miR378c. These findings enhance our understanding of the pathogenesis of STAT3 gain-of-function syndrome and suggest potential therapeutic strategies. Notably, combined JAK inhibitors and IL-6R antagonists may offer promising treatment avenues for mitigating the severity of STAT3 gain-of-function syndrome.
Collapse
Affiliation(s)
- Ji-Yu Chen
- Department of Nephrology & Rheumatology, Kunming Children's Hospital, Kunming 650228, Yunnan, China
| | - Yan-Fang Li
- Department of Nephrology & Rheumatology, Kunming Children's Hospital, Kunming 650228, Yunnan, China
| | - Zhu Zhou
- Department of Nephrology, First Affiliated Hospital of Kunming Medical University, Yunnan Clinical Medical Research Center of Chronic Kidney Disease, Kunming 650032, Yunnan, China
| | - Xue-Mei Jiang
- Department of Nephrology & Rheumatology, Kunming Children's Hospital, Kunming 650228, Yunnan, China
| | - Xin Bi
- Department of Nephrology & Rheumatology, Kunming Children's Hospital, Kunming 650228, Yunnan, China
| | - Mi-Feng Yang
- Department of Nephrology & Rheumatology, Kunming Children's Hospital, Kunming 650228, Yunnan, China
| | - Bo Zhao
- Department of Nephrology & Rheumatology, Kunming Children's Hospital, Kunming 650228, Yunnan, China.
| |
Collapse
|
234
|
Wang H, Boeren S, Bakker W, Rietjens IMCM, Saccenti E, Zheng L. An integrated proteomics and metabolomics analysis of methylglyoxal-induced neurotoxicity in a human neuroblastoma cell line. NPJ Sci Food 2024; 8:84. [PMID: 39448607 PMCID: PMC11502746 DOI: 10.1038/s41538-024-00328-0] [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: 07/03/2024] [Accepted: 10/17/2024] [Indexed: 10/26/2024] Open
Abstract
This study aimed to highlight the molecular and biochemical changes induced by methylglyoxal (MGO) exposure in SH-SY5Y human neuroblastoma cells, and to explore how these changes contribute to its neurotoxicity, utilizing an integrated proteomics and metabolomics approach. Using label-free quantitative nanoLC-MS/MS proteomics and targeted LC-TQ-MS/MS-based metabolomics, the results revealed that MGO exposure, particularly at cytotoxic levels, significantly altered the proteome and metabolome of SH-SY5Y cells. Analysis of proteomics data showed significant alterations in cellular functions including protein synthesis, cellular structural integrity, mitochondrial function, and oxidative stress responses. Analysis of metabolomics and integration of metabolomics and proteomics data highlighted significant changes in key metabolic pathways including arginine biosynthesis, glutathione metabolism, cysteine and methionine metabolism, and the tricarboxylic acid cycle. These results suggest that MGO exposure induced both toxic effects and adaptive responses in cells. MGO exposure led to increased endoplasmic reticulum stress, disruptions in cellular adhesion and extracellular matrix integrity, mitochondrial dysfunction, and amino acid metabolism disruption, contributing to cellular toxicity. Conversely, cells exhibited adaptive responses by upregulating protein synthesis, activating the Nrf2 pathway, and reprogramming metabolism to counteract dicarbonyl stress and maintain energy levels. Furthermore, a set of key proteins and metabolites associated with these changes were shown to exhibit a significant concentration-dependent decrease or increase in their expression levels with increasing MGO concentrations, suggesting their potential as biomarkers for MGO exposure. Taken together, these findings provide insight into the molecular mechanisms underlying MGO-induced neurotoxicity and potential targets for therapeutic intervention.
Collapse
Affiliation(s)
- Haomiao Wang
- Division of Toxicology, Wageningen University & Research, Wageningen, The Netherlands
| | - Sjef Boeren
- Laboratory of Biochemistry, Wageningen University & Research, Wageningen, The Netherlands
| | - Wouter Bakker
- Division of Toxicology, Wageningen University & Research, Wageningen, The Netherlands
| | - Ivonne M C M Rietjens
- Division of Toxicology, Wageningen University & Research, Wageningen, The Netherlands
| | - Edoardo Saccenti
- Laboratory of Systems and Synthetic Biology, Wageningen University & Research, Wageningen, The Netherlands.
| | - Liang Zheng
- Division of Toxicology, Wageningen University & Research, Wageningen, The Netherlands.
| |
Collapse
|
235
|
Jia X, Zhao W, Zhang H, Zhang X, Ji Q, Li X, Pan Y, Jiang X, Zhang J, Bai L. Cell-Specific Gene Expressions Underlie Selective White Matter Loss Vulnerability in Mild Traumatic Brain Injury. J Neurotrauma 2024. [PMID: 39453870 DOI: 10.1089/neu.2024.0022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2024] Open
Abstract
Traumatic brain injury (TBI), a risk factor for later-life dementia, leads to salient brain atrophy, particularly in the white matter. It is not clear how white matter atrophy progresses or why some brain regions are damaged while others are spared. We hypothesized that spatial variations of cell-specific gene expression contributed to the selective white matter loss vulnerability following mild TBI (mTBI). Gene expression data were sourced from the publicly available Allen Human Brain Atlas, which comprises microarray data spanning nearly the entire brain, derived from six neurologically normal adult donors. A total of 100 patients with acute stage (within 7 days post-injury) mTBI were enrolled. Of these, 60 patients were followed up at 3 months post-injury and 37 were followed up at 6-12 months post-injury. In addition, 59 healthy controls (HCs), matched for age, gender, and education, were included for comparative analysis. White matter volume changes were analyzed at both the acute stage, 3 months, and 6-12 months follow-up in mTBI patients compared with HCs. Patients with mTBI exhibited significant white matter atrophy in the frontal, parietal, and temporal cortices at 3 months post-injury, which even persisted at 6-12 months follow-up. In addition, mTBI patients with cognitive deficits showed more severe brain atrophy compared with those without cognitive deficits. Crucially, the gene expression marking endothelial cells and S1 pyramidal neurons were associated with increased brain atrophy, whereas the gene expression marking microglia and CA1 pyramidal neurons were associated with decreased brain atrophy in mTBI patients at 3 months post-injury. Microglia and endothelial cells can explain 23.6% of regional variations in the white matter atrophy. These findings suggested that modulating cellular activation, especially by promoting microglial activation at 3 months post-injury, might be a promising approach to prevent white matter atrophy, enhance cognitive outcomes, and reduce the risk of later-life dementia.
Collapse
Affiliation(s)
- Xiaoyan Jia
- The Key Laboratory of Biomedical Information Engineering, Ministry of Education, Department of Biomedical Engineering, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, China
| | - Wenpu Zhao
- The Key Laboratory of Biomedical Information Engineering, Ministry of Education, Department of Biomedical Engineering, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, China
| | - Haonan Zhang
- The Key Laboratory of Biomedical Information Engineering, Ministry of Education, Department of Biomedical Engineering, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, China
| | - Xiang Zhang
- The Key Laboratory of Biomedical Information Engineering, Ministry of Education, Department of Biomedical Engineering, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, China
| | - Qiuyu Ji
- The Key Laboratory of Biomedical Information Engineering, Ministry of Education, Department of Biomedical Engineering, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, China
| | - Xuan Li
- The Key Laboratory of Biomedical Information Engineering, Ministry of Education, Department of Biomedical Engineering, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, China
| | - Yizhen Pan
- The Key Laboratory of Biomedical Information Engineering, Ministry of Education, Department of Biomedical Engineering, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, China
| | - Xiaofan Jiang
- Department of Neurosurgery, Xijing Hospital, Air Force Medical University, Xi'an, China
| | - Jie Zhang
- Department of Radiation Medicine, School of Preventive Medicine, Air Force Medical University, Xi'an, China
| | - Lijun Bai
- The Key Laboratory of Biomedical Information Engineering, Ministry of Education, Department of Biomedical Engineering, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, China
- Department of Medical Imaging, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| |
Collapse
|
236
|
Bohutskyi P, Pomraning KR, Jenkins JP, Kim YM, Poirier BC, Betenbaugh MJ, Magnuson JK. Mixed and membrane-separated culturing of synthetic cyanobacteria-yeast consortia reveals metabolic cross-talk mimicking natural cyanolichens. Sci Rep 2024; 14:25303. [PMID: 39455633 PMCID: PMC11511929 DOI: 10.1038/s41598-024-74743-4] [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: 07/20/2024] [Accepted: 09/30/2024] [Indexed: 10/28/2024] Open
Abstract
Metabolite exchange mediates crucial interactions in microbial communities, significantly impacting global carbon and nitrogen cycling. Understanding these chemically-mediated interactions is essential for elucidating natural community functions and developing engineered synthetic communities. This study investigated membrane-separated bioreactors (mBRs) as a novel tool to identify transient metabolites and their producers/consumers in mixed microbial communities. We compared three co-culture methods (direct mixed, 2-chamber mBR, and 3-chamber mBR) to grow a synthetic binary community of the cyanobacterium Synechococcus elongatus PCC 7942 and the fungus Rhodotorula toruloides NBRC 0880, as well as axenic S. elongatus. Despite not being natural lichen constituents, these organisms exhibited interactions resembling those in cyanolichens. S. elongatus fixed CO2 into sugars as the primary shared metabolite, while R. toruloides secreted various biochemicals, predominantly sugar alcohols, mirroring the metabolite exchange observed in natural lichens. The mBR systems successfully captured metabolite gradients and revealed rapidly consumed compounds, including TCA cycle intermediates and amino acids. Our approach demonstrated that the 2-chamber mBR optimally balanced metabolite exchange and growth dynamics. This study provides insights into cross-species metabolic interactions and presents a valuable tool for investigating and engineering synthetic microbial communities with potential applications in biotechnology and environmental science.
Collapse
Affiliation(s)
- Pavlo Bohutskyi
- Earth and Biological Sciences Directorate, Pacific Northwest National Laboratory, Richland, WA, 99354, USA.
- Department of Biological Systems Engineering, Washington State University, Pullman, WA, 99164, USA.
| | - Kyle R Pomraning
- Energy and Environment Directorate, Pacific Northwest National Laboratory, Richland, WA, 99354, USA
| | - Jackson P Jenkins
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, 21218, USA
| | - Young-Mo Kim
- Earth and Biological Sciences Directorate, Pacific Northwest National Laboratory, Richland, WA, 99354, USA
| | - Brenton C Poirier
- Earth and Biological Sciences Directorate, Pacific Northwest National Laboratory, Richland, WA, 99354, USA
| | - Michael J Betenbaugh
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, 21218, USA
| | - Jon K Magnuson
- Energy and Environment Directorate, Pacific Northwest National Laboratory, Richland, WA, 99354, USA
| |
Collapse
|
237
|
Ren X, Feng N. Unveiling novel prognostic biomarkers and therapeutic targets for HBV-associated hepatocellular carcinoma through integrated bioinformatic analysis. Medicine (Baltimore) 2024; 103:e40134. [PMID: 39470543 PMCID: PMC11521037 DOI: 10.1097/md.0000000000040134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Revised: 06/15/2024] [Accepted: 09/27/2024] [Indexed: 10/30/2024] Open
Abstract
Hepatocellular carcinoma (HCC) is a leading cause of cancer-related deaths globally, with limited treatment options. The goal of this study was to use integrated bioinformatic analysis to find possible biomarkers for prognosis and therapeutic targets for hepatitis B (HBV)-associated HCC. Three microarray datasets (GSE84402, GSE121248, and E-GEOD-19665) from patients with HBV-associated HCC were combined and analyzed. We identified differentially expressed genes (DEGs) and performed pathway enrichment analysis. We constructed protein-protein interaction networks to identify hub genes. We identified a total of 374 DEGs, which included 90 up-regulated and 284 down-regulated genes. Pathway enrichment analysis revealed associations with cell cycle, oocyte meiosis, and the p53 signaling pathway for up-regulated DEGs. Twenty hub genes were identified, and 9 of them (ZWINT, MELK, DLGAP5, BIRC5, AURKA, HMMR, CDK1, TTK, and MAD2L1) were validated using the Cancer Genome Atlas data and Kaplan-Meier survival analysis. These genes were significantly associated with a poor prognosis in HCC patients. Our research shows that ZWINT, MELK, DLGAP5, BIRC5, AURKA, HMMR, CDK1, TTK, and MAD2L1 may be useful for predicting how HBV-associated HCC will progress and for finding new ways to treat it. In addition to these further studies are needed to elucidate the functions of the remaining 11 identified hub genes (RRM2, NUSAP1, PBK, CCNB1, CCNB2, BUB1B, NEK2, CENPF, ASPM, TOP2A, and BUB1) in HCC development and progression.
Collapse
Affiliation(s)
- Xue Ren
- Medical Laboratory Center, Xi’an TCM Hospital of Encephalopathy, Xi’an, China
| | - Niaoniao Feng
- Medical Laboratory Center, Xi’an TCM Hospital of Encephalopathy, Xi’an, China
| |
Collapse
|
238
|
Bakhtina AA, Wippel HH, Chavez JD, Bruce JE. Combining Quantitative Proteomics and Interactomics for a Deeper Insight into Molecular Differences between Human Cell Lines. J Proteome Res 2024. [PMID: 39453897 DOI: 10.1021/acs.jproteome.4c00503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2024]
Abstract
In modern biomedical research, cultivable cell lines are an indispensable tool, and the selection of cell lines that exhibit specific functional profiles is often critical to success. Cellular functional pathways have evolved through the selection of protein intra- and intermolecular interactions collectively referred to as the interactome. In the present work, quantitative in vivo protein cross-linking and mass spectrometry were used to probe large-scale protein interactome differences among three commonly employed human cell lines, namely, HEK293, MCF-7, and HeLa cells. These data illustrated highly reproducible quantitative interactome levels with R2 values larger than 0.8 for all biological replicates. Proteome abundance levels were also measured using data-independent acquisition quantitative proteomics methods. Combining quantitative interactome and proteome information allowed the visualization of cell type-specific interactome changes mediated by proteome level adaptations and independently regulated interactome changes to gain deeper insight into possible drivers of these changes. Among the largest detected alterations in protein interactions and conformations are changes in cytoskeletal proteins, RNA-binding proteins, chromatin remodeling complexes, mitochondrial proteins, and others. Overall, these data demonstrate the utility and reproducibility of quantitative cross-linking to study system-level interactome variations. Moreover, these results illustrate how combined quantitative interactomics and proteomics can provide unique insight into cellular functional landscapes.
Collapse
Affiliation(s)
- Anna A Bakhtina
- Department of Genome Sciences, University of Washington, Seattle, Washington 98195, United States
| | - Helisa H Wippel
- Department of Genome Sciences, University of Washington, Seattle, Washington 98195, United States
| | - Juan D Chavez
- Department of Genome Sciences, University of Washington, Seattle, Washington 98195, United States
| | - James E Bruce
- Department of Genome Sciences, University of Washington, Seattle, Washington 98195, United States
| |
Collapse
|
239
|
Dayama G, Gupta S, Connizzo BK, Labadorf AT, Myers RH, Lau NC. Transposable element small and long RNAs in aging brains and implications in Huntington's and Parkinson's disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.22.619758. [PMID: 39484439 PMCID: PMC11526979 DOI: 10.1101/2024.10.22.619758] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/03/2024]
Abstract
Transposable Elements (TEs) are implicated in aging and neurodegenerative disorders, but the impact of brain TE RNA dynamics on these phenomena is not fully understood. Therefore, we quantified TE RNA changes in aging post-mortem human and mouse brains and in the neurodegenerative disorders Huntington's Disease (HD) and Parkinson's Disease (PD). We tracked TE small RNAs (smRNAs) expression landscape to assess the relationship to the active processing from TE long RNAs (lnRNAs). Human brain transcriptomes from the BrainSpan Atlas displayed a significant shift of TE smRNA patterns at age 20 years, whereas aging mouse brains lacked any such marked change, despite clear shift in aging-associated mRNA levels. Human frontal cortex displayed pronounced sense TE smRNAs during aging with a negative relationship between the TE smRNAs and lnRNAs indicative of age associated regulatory effects. Our analysis revealed TE smRNAs dysregulation in HD, while PD showed a stronger impact on TE lnRNAs, potentially correlating with the early average age of death for HD relative to PD. Furthermore, TE-silencing factor TRIM28 was down-regulated only in aging human brains, possibly explaining the lack of substantial TE RNA changes in aging mouse brains. Our study suggests brain TE RNAs may serve as novel biomarkers of human brain aging and neurodegenerative disorders.
Collapse
|
240
|
Wei H, Ma Y, Chen S, Zou C, Wang L. Multi-omics analysis identifies PTTG1 as a prognostic biomarker associated with immunotherapy and chemotherapy resistance. BMC Cancer 2024; 24:1315. [PMID: 39455949 PMCID: PMC11520140 DOI: 10.1186/s12885-024-13060-5] [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: 08/16/2024] [Accepted: 10/14/2024] [Indexed: 10/28/2024] Open
Abstract
BACKGROUND Pituitary tumor-transforming gene 1 (PTTG1) is an important gene in tumour development. However, the relevance of PTTG1 in tumour prognosis, immunotherapy response, and medication sensitivity in human pan-cancer has to be determined. METHODS TIMER, GEPIA, the human protein atlas, GEPIA, TISCH2, and cBioportal examined the gene expression, protein expression, prognostic value, and genetic modification landscape of PTTG1 in 33 malignancies based on the TCGA cohort. The association between PTTG1 and tumour immunity, tumour microenvironment, immunotherapy response, and anticancer drug sensitivity was investigated using GSCA, TIDE, and CellMiner CDB. Molecular docking was used to validate the possible chemotherapeutic medicines for PTTG1. Additionally, siRNA-mediated knockdown was employed to confirm the probable role of PTTG1 in paclitaxel-resistant cells. RESULTS PTTG1 is overexpressed and associated with poor survival in most tumors. Functional enrichment study revealed that PTTG1 is involved in the cell cycle and DNA replication. A substantial connection between PTTG1 expression and immune cell infiltration points to PTTG1's possible role in the tumour microenvironment. High PTTG1 expression is associated with tumour immunotherapy resistance. The process could be connected to PTTG1, which mediates T cell exhaustion and promotes cytotoxic T lymphocyte malfunction. Furthermore, PTTG1 was found to be substantially linked with sensitivity to several anticancer medications. Suppressing PTTG1 with siRNA reduced clone formation and migration, implying that PTTG1 may play a role in paclitaxel resistance. CONCLUSION PTTG1 shows potential as a cancer diagnostic, prognostic, and chemosensitivity marker. Increased PTTG1 expression is linked to resistance to cancer treatment. The mechanism could be linked to PTTG1's role in promoting cytotoxic T lymphocyte dysfunction and mediating T cell exhaustion. It is feasible to consider PTTG1, which is expressed on Treg and Tprolif cells, as a new therapeutic target for overcoming immunotherapy resistance.
Collapse
Affiliation(s)
- Handong Wei
- Key Laboratory of Longevity and Aging-related Diseases of Chinese Ministry of Education, Center for Translational Medicine and School of Preclinical Medicine, Guangxi Medical University, Nanning, Guangxi, China
| | - Yaxin Ma
- Key Laboratory of Longevity and Aging-related Diseases of Chinese Ministry of Education, Center for Translational Medicine and School of Preclinical Medicine, Guangxi Medical University, Nanning, Guangxi, China
| | - Shuxing Chen
- Key Laboratory of Longevity and Aging-related Diseases of Chinese Ministry of Education, Center for Translational Medicine and School of Preclinical Medicine, Guangxi Medical University, Nanning, Guangxi, China
| | - Chunlin Zou
- Key Laboratory of Longevity and Aging-related Diseases of Chinese Ministry of Education, Center for Translational Medicine and School of Preclinical Medicine, Guangxi Medical University, Nanning, Guangxi, China.
| | - Lihui Wang
- Key Laboratory of Longevity and Aging-related Diseases of Chinese Ministry of Education, Center for Translational Medicine and School of Preclinical Medicine, Guangxi Medical University, Nanning, Guangxi, China.
| |
Collapse
|
241
|
Heng YC, Wong GWJ, Kittelmann S. Expanding the biosynthesis spectrum of hydroxy fatty acids: unleashing the potential of novel bacterial fatty acid hydratases. BIOTECHNOLOGY FOR BIOFUELS AND BIOPRODUCTS 2024; 17:131. [PMID: 39456067 PMCID: PMC11515146 DOI: 10.1186/s13068-024-02578-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Accepted: 10/18/2024] [Indexed: 10/28/2024]
Abstract
BACKGROUND Hydroxy fatty acids represent an emerging class of compounds with promising applications in the chemical, medicinal and functional food sectors. The challenges associated with their chemical synthesis have spurred exploration of biological synthesis as an alternative route, particularly through the use of fatty acid hydratases. Fatty acid hydratases catalyse the regioselective addition of a hydrogen atom and a hydroxyl group from a water molecule to the carbon-carbon cis-double bond of unsaturated fatty acids to form hydroxy fatty acids. Despite having been discovered in the early 1960s, previous research has primarily focused on characterizing single fatty acid hydratase variants with a limited range of substrates. Comprehensive studies that systematically examine and compare the characteristics of multiple variants of fatty acid hydratases are still lacking. RESULTS In this study, we employed an integrated bioinformatics workflow to identify 23 fatty acid hydratases and characterized their activities against nine unsaturated fatty acid substrates using whole-cell biotransformation assays. Additionally, we tested a dual-protein system involving two fatty acid hydratases of distinct regioselectivity and demonstrated its suitability in enhancing the biosynthesis of di-hydroxy fatty acids. CONCLUSIONS Our study demonstrates that fatty acid hydratases can be classified into three subtypes based on their regioselectivity and provides insights into their preferred substrate structures. These understandings pave ways for the design of optimal fatty acid hydratase variants and bioprocesses for the cost-efficient biosynthesis of hydroxy fatty acids.
Collapse
Affiliation(s)
- Yu Chyuan Heng
- Wilmar International Limited, 28 Biopolis Road, Singapore, 138568, Singapore.
| | - Garrett Wei Jie Wong
- Wilmar International Limited, 28 Biopolis Road, Singapore, 138568, Singapore
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, 14 Medical Drive, Singapore, 117599, Singapore
| | - Sandra Kittelmann
- Wilmar International Limited, 28 Biopolis Road, Singapore, 138568, Singapore.
| |
Collapse
|
242
|
Yarbro JM, Han X, Dasgupta A, Yang K, Liu D, Shrestha HK, Zaman M, Wang Z, Yu K, Lee DG, Vanderwall D, Niu M, Sun H, Xie B, Chen PC, Jiao Y, Zhang X, Wu Z, Fu Y, Li Y, Yuan ZF, Wang X, Poudel S, Vagnerova B, He Q, Tang A, Ronaldson PT, Chang R, Yu G, Liu Y, Peng J. Human-mouse proteomics reveals the shared pathways in Alzheimer's disease and delayed protein turnover in the amyloidome. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.25.620263. [PMID: 39484428 PMCID: PMC11527136 DOI: 10.1101/2024.10.25.620263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/03/2024]
Abstract
Murine models of Alzheimer's disease (AD) are crucial for elucidating disease mechanisms but have limitations in fully representing AD molecular complexities. We comprehensively profiled age-dependent brain proteome and phosphoproteome (n > 10,000 for both) across multiple mouse models of amyloidosis. We identified shared pathways by integrating with human metadata, and prioritized novel components by multi-omics analysis. Collectively, two commonly used models (5xFAD and APP-KI) replicate 30% of the human protein alterations; additional genetic incorporation of tau and splicing pathologies increases this similarity to 42%. We dissected the proteome-transcriptome inconsistency in AD and 5xFAD mouse brains, revealing that inconsistent proteins are enriched within amyloid plaque microenvironment (amyloidome). Determining the 5xFAD proteome turnover demonstrates that amyloid formation delays the degradation of amyloidome components, including Aβ-binding proteins and autophagy/lysosomal proteins. Our proteomic strategy defines shared AD pathways, identify potential new targets, and underscores that protein turnover contributes to proteome-transcriptome discrepancies during AD progression.
Collapse
Affiliation(s)
- Jay M Yarbro
- Department of Structural Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
- These authors contributed equally
| | - Xian Han
- Department of Structural Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
- These authors contributed equally
| | - Abhijit Dasgupta
- Department of Structural Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
- Current address: Department of Computer Science and Engineering, SRM University AP, Andhra Pradesh 522240, India
- These authors contributed equally
| | - Ka Yang
- Department of Structural Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
- These authors contributed equally
| | - Danting Liu
- Department of Structural Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Him K Shrestha
- Department of Structural Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Masihuz Zaman
- Department of Structural Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Zhen Wang
- Department of Structural Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Kaiwen Yu
- Center for Proteomics and Metabolomics, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Dong Geun Lee
- Department of Structural Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - David Vanderwall
- Department of Structural Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Mingming Niu
- Department of Structural Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Huan Sun
- Department of Structural Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Boer Xie
- Center for Proteomics and Metabolomics, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Ping-Chung Chen
- Department of Structural Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Yun Jiao
- Department of Structural Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Xue Zhang
- Department of Structural Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Zhiping Wu
- Department of Structural Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Yingxue Fu
- Center for Proteomics and Metabolomics, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Yuxin Li
- Center for Proteomics and Metabolomics, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Zuo-Fei Yuan
- Center for Proteomics and Metabolomics, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Xusheng Wang
- Department of Genetics, Genomics and Informatics, University of Tennessee Health Science Center, Memphis, TN 38103, USA
| | - Suresh Poudel
- Center for Proteomics and Metabolomics, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Barbora Vagnerova
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, AZ 85724, USA
| | - Qianying He
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, AZ 85724, USA
| | - Andrew Tang
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, AZ 85724, USA
| | - Patrick T Ronaldson
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, AZ 85724, USA
| | - Rui Chang
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, AZ 85724, USA
| | - Gang Yu
- Department of Neuroscience, Peter O'Donnell Jr. Brain Institute, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Yansheng Liu
- Department of Pharmacology, Yale University School of Medicine, New Haven, CT 06510, USA
- Yale Cancer Research Institute, Yale University School of Medicine, West Haven, CT, 06516, USA
| | - Junmin Peng
- Department of Structural Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
- Center for Proteomics and Metabolomics, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| |
Collapse
|
243
|
Liang P, Zhu M, Sun X, Wang L, Li B, Ming S, Younis M, Yang J, Wu Y, Huang X. LncRNA-mRNA co-expression analysis reveals aquaporin-9-promoted neutrophil extracellular trap formation and inflammatory activation in sepsis. Int Immunopharmacol 2024; 140:112916. [PMID: 39133961 DOI: 10.1016/j.intimp.2024.112916] [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: 06/03/2024] [Revised: 07/29/2024] [Accepted: 08/06/2024] [Indexed: 09/01/2024]
Abstract
Sepsis is a life-threatening condition caused by an excessive inflammatory response to an infection. However, the precise regulatory mechanism of sepsis remains unclear. Using a strand-specific RNA-sequencing, we identified 115 hub differentially expressed long noncoding RNAs (lncRNAs) and 443 mRNAs in septic patients, primarily participated in crucial pathways including neutrophil extracellular trap (NET) formation and toll-like receptor signaling. Notably, NETs related gene aquaporin-9 (AQP9) and its associated lncRNAs exhibited significant upregulation in septic neutrophils. Functional experiments revealed AQP9 interacts with its lncRNAs to augment the formation of neutrophil NETs. In murine sepsis models, AQP9 inhibition with phloretin reduced proinflammatory cytokine production and lung damage. These findings provide crucial insights into the regulatory role of AQP9 in sepsis, unraveling its interaction with associated lncRNAs in transmitting downstream signals, holding promise in informing the development of novel therapeutic strategies aimed at ameliorating the debilitating effects of sepsis.
Collapse
Affiliation(s)
- Pingping Liang
- Center for Infection and Immunity and Guangdong Provincial Engineering Research Center of Molecular Imaging, the Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, Guangdong 519000, China; Guangdong-Hong Kong-Macao University Joint Laboratory of Interventional Medicine, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong 519000, China; Key Laboratory of Traditional Chinese Medicine for the Prevention and Treatment of Infectious Diseases, Traditional Chinese Medicine Bureau of Guangdong Province, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong 519000, China
| | - Manman Zhu
- Department of Clinical Laboratory, The Fifth Affiliated Hospital of Sun Yat-Sen University, Zhuhai, Guangdong 519000, China
| | - Xingzi Sun
- Center for Infection and Immunity and Guangdong Provincial Engineering Research Center of Molecular Imaging, the Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, Guangdong 519000, China
| | - Li Wang
- Department of Obstetrics and Gynecology, Perinatal Medical Center, the Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, Guangdong 519000, China
| | - Bin Li
- MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, Guangdong 510275, China
| | - Siqi Ming
- Center for Infection and Immunity and Guangdong Provincial Engineering Research Center of Molecular Imaging, the Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, Guangdong 519000, China
| | - Muhammad Younis
- Center for Infection and Immunity and Guangdong Provincial Engineering Research Center of Molecular Imaging, the Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, Guangdong 519000, China
| | - Jianhua Yang
- MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, Guangdong 510275, China
| | - Yongjian Wu
- Center for Infection and Immunity and Guangdong Provincial Engineering Research Center of Molecular Imaging, the Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, Guangdong 519000, China; Guangdong-Hong Kong-Macao University Joint Laboratory of Interventional Medicine, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong 519000, China; Key Laboratory of Traditional Chinese Medicine for the Prevention and Treatment of Infectious Diseases, Traditional Chinese Medicine Bureau of Guangdong Province, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong 519000, China.
| | - Xi Huang
- Center for Infection and Immunity and Guangdong Provincial Engineering Research Center of Molecular Imaging, the Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, Guangdong 519000, China; Guangdong-Hong Kong-Macao University Joint Laboratory of Interventional Medicine, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong 519000, China; Key Laboratory of Traditional Chinese Medicine for the Prevention and Treatment of Infectious Diseases, Traditional Chinese Medicine Bureau of Guangdong Province, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong 519000, China.
| |
Collapse
|
244
|
Rahlff J, Westmeijer G, Weissenbach J, Antson A, Holmfeldt K. Surface microlayer-mediated virome dissemination in the Central Arctic. MICROBIOME 2024; 12:218. [PMID: 39449105 PMCID: PMC11515562 DOI: 10.1186/s40168-024-01902-0] [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: 03/18/2024] [Accepted: 08/06/2024] [Indexed: 10/26/2024]
Abstract
BACKGROUND Aquatic viruses act as key players in shaping microbial communities. In polar environments, they face significant challenges such as limited host availability and harsh conditions. However, due to the restricted accessibility of these ecosystems, our understanding of viral diversity, abundance, adaptations, and host interactions remains limited. RESULTS To fill this knowledge gap, we studied viruses from atmosphere-close aquatic ecosystems in the Central Arctic and Northern Greenland. Aquatic samples for virus-host analysis were collected from ~60 cm depth and the submillimeter surface microlayer (SML) during the Synoptic Arctic Survey 2021 on icebreaker Oden in the Arctic summer. Water was sampled from a melt pond and open water before undergoing size-fractioned filtration, followed by genome-resolved metagenomic and cultivation investigations. The prokaryotic diversity in the melt pond was considerably lower compared to that of open water. The melt pond was dominated by a Flavobacterium sp. and Aquiluna sp., the latter having a relatively small genome size of 1.2 Mb and the metabolic potential to generate ATP using the phosphate acetyltransferase-acetate kinase pathway. Viral diversity on the host fraction (0.2-5 µm) of the melt pond was strikingly limited compared to that of open water. From the 1154 viral operational taxonomic units (vOTUs), of which two-thirds were predicted bacteriophages, 17.2% encoded for auxiliary metabolic genes (AMGs) with metabolic functions. Some AMGs like glycerol-3-phosphate cytidylyltransferase and ice-binding like proteins might serve to provide cryoprotection for the host. Prophages were often associated with SML genomes, and two active prophages of new viral genera from the Arctic SML strain Leeuwenhoekiella aequorea Arc30 were induced. We found evidence that vOTU abundance in the SML compared to that of ~60 cm depth was more positively correlated with the distribution of a vOTU across five different Arctic stations. CONCLUSIONS The results indicate that viruses employ elaborate strategies to endure in extreme, host-limited environments. Moreover, our observations suggest that the immediate air-sea interface serves as a platform for viral distribution in the Central Arctic. Video Abstract.
Collapse
Affiliation(s)
- Janina Rahlff
- Centre for Ecology and Evolution in Microbial Model Systems (EEMiS), Department of Biology and Environmental Science, Linnaeus University, Kalmar, Sweden.
- Aero-Aquatic Virus Research Group, Faculty of Mathematics and Computer Science, Friedrich Schiller University Jena, Jena, Germany.
- Leibniz Institute on Aging-Fritz Lipmann Institute (FLI), Jena, Germany.
| | - George Westmeijer
- Centre for Ecology and Evolution in Microbial Model Systems (EEMiS), Department of Biology and Environmental Science, Linnaeus University, Kalmar, Sweden
| | - Julia Weissenbach
- Centre for Ecology and Evolution in Microbial Model Systems (EEMiS), Department of Biology and Environmental Science, Linnaeus University, Kalmar, Sweden
| | - Alfred Antson
- York Structural Biology Laboratory, Department of Chemistry, University of York, York, UK
| | - Karin Holmfeldt
- Centre for Ecology and Evolution in Microbial Model Systems (EEMiS), Department of Biology and Environmental Science, Linnaeus University, Kalmar, Sweden
| |
Collapse
|
245
|
Balhara R, Verma D, Kaur R, Singh K. MYB transcription factors, their regulation and interactions with non-coding RNAs during drought stress in Brassica juncea. BMC PLANT BIOLOGY 2024; 24:999. [PMID: 39448923 PMCID: PMC11515528 DOI: 10.1186/s12870-024-05736-8] [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: 01/30/2024] [Accepted: 10/21/2024] [Indexed: 10/26/2024]
Abstract
BACKGROUND Brassica juncea (L.) Czern is an important oilseed crop affected by various abiotic stresses like drought, heat, and salt. These stresses have detrimental effects on the crop's overall growth, development and yield. Various Transcription factors (TFs) are involved in regulation of plant stress response by modulating expression of stress-responsive genes. The myeloblastosis (MYB) TFs is one of the largest families of TFs associated with various developmental and biological processes such as plant growth, secondary metabolism, stress response etc. However, MYB TFs and their regulation by non-coding RNAs (ncRNAs) in response to stress have not been studied in B. juncea. Thus, we performed a detailed study on the MYB TF family and their interactions with miRNAs and Long non coding RNAs. RESULTS Computational investigation of genome and proteome data presented a comprehensive picture of the MYB genes and their protein architecture, including intron-exon organisation, conserved motif analysis, R2R3 MYB DNA-binding domains analysis, sub-cellular localization, protein-protein interaction and chromosomal locations. Phylogenetically, BjuMYBs were further classified into different subclades on the basis of topology and classification in Arabidopsis. A total of 751 MYBs were identified in B. juncea corresponding to 297 1R-BjuMYBs, 440 R2R3-BjuMYBs, 12 3R-BjuMYBs, and 2 4R-BjuMYBs types. We validated the transcriptional profiles of nine selected BjuMYBs under drought stress through RT-qPCR. Promoter analysis indicated the presence of drought-responsive cis-regulatory components. Additionally, the miRNA-MYB TF interactions was also studied, and most of the microRNAs (miRNAs) that target BjuMYBs were involved in abiotic stress response and developmental processes. Regulatory network analysis and expression patterns of lncRNA-miRNA-MYB indicated that selected long non-coding RNAs (lncRNAs) acted as strong endogenous target mimics (eTMs) of the miRNAs regulated expression of BjuMYBs under drought stress. CONCLUSIONS The present study has established preliminary groundwork of MYB TFs and their interaction with ncRNAs in B. juncea and it will help in developing drought- tolerant Brassica crops.
Collapse
Affiliation(s)
- Rinku Balhara
- Department of Biotechnology, BMS Block I, Panjab University, Sector 25, Chandigarh, 160014, India
| | - Deepika Verma
- Department of Biotechnology, BMS Block I, Panjab University, Sector 25, Chandigarh, 160014, India
| | - Ravneet Kaur
- Department of Biotechnology, BMS Block I, Panjab University, Sector 25, Chandigarh, 160014, India
| | - Kashmir Singh
- Department of Biotechnology, BMS Block I, Panjab University, Sector 25, Chandigarh, 160014, India.
| |
Collapse
|
246
|
Das P, Alex R, Gowane GR, Vohra V, Paul D, Khan KD, Upadhyay A, De S, Ludri A. Chronic heat stress upregulates pyruvate metabolic process and gluconeogenesis but downregulates immune responses in Sahiwal cattle. INTERNATIONAL JOURNAL OF BIOMETEOROLOGY 2024:10.1007/s00484-024-02804-4. [PMID: 39446186 DOI: 10.1007/s00484-024-02804-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Revised: 07/24/2024] [Accepted: 10/11/2024] [Indexed: 10/25/2024]
Abstract
Climate change and growing population and their strain on animal production are the impending challenges that the developing countries, like India, need to tackle in the coming days. This study aimed to detect and analyze the uncharacterized variation in the gene expression patterns with the change of condition, from thermoneutral to chronic hot-humid, in the Sahiwal cattle, one of the best breeds of milk-producing cattle in India, known for being heat-tolerant. Using RNA-Seq analysis on peripheral blood mononuclear cells (PBMCs), 4021 differentially expressed mRNAs (2772 upregulated, 1249 downregulated) and 1303 differentially expressed long non-coding RNAs (769 upregulated, 534 downregulated) were identified, with the thresholds of false discovery rate < 0.05 and|log2(fold change)| > 2. Significantly (p-adjusted < 0.05) overrepresented Gene Ontology (GO) terms, Kyoto Encyclopedia of Genes and Genomes (KEGG), and Reactome pathways were analyzed, revealing upregulation of processes like pyruvate metabolic process, gluconeogenesis, ion transmembrane transport, neuropeptide signaling pathway, and animal organ development, with genes like SHH, GRK1, CHRM3, CAMK2A, and HSPB7 were upregulated, while translation and immune responses, with genes like RPS3, EEF1A1, TNF, BoLA-DRB3, and UBB were downregulated. Analysis of cis-mRNAs of DE-lncRNAs showed presence of both up- and down-regulated cis-mRNAs for both up- and down-regulated lncRNAs indicating existence of positive and negative regulation of mRNA expression by lncRNAs. Managemental nudges that decrease metabolic heat generation, like betaine and chromium supplementation, and increase heat dissipation, like microenvironment cooling, should be utilized. This study highlights the role of pyruvate metabolism and gluconeogenesis in coping up with heat stress and offers an improved understanding of the heat stress response of Sahiwal cattle along with the genes and pathways responsible for it.
Collapse
Affiliation(s)
- Pradyut Das
- Division of Animal Genetics and Breeding, ICAR-National Dairy Research Institute, Karnal, Haryana, 132001, India
| | - Rani Alex
- Division of Animal Genetics and Breeding, ICAR-National Dairy Research Institute, Karnal, Haryana, 132001, India.
| | - Gopal Ramdasji Gowane
- Division of Animal Genetics and Breeding, ICAR-National Dairy Research Institute, Karnal, Haryana, 132001, India
| | - Vikas Vohra
- Division of Animal Genetics and Breeding, ICAR-National Dairy Research Institute, Karnal, Haryana, 132001, India
| | - Dipankar Paul
- Division of Animal Genetics and Breeding, ICAR-National Dairy Research Institute, Karnal, Haryana, 132001, India
| | - Kashif Dawood Khan
- Division of Animal Genetics and Breeding, ICAR-National Dairy Research Institute, Karnal, Haryana, 132001, India
| | - Amritanshu Upadhyay
- Division of Animal Genetics and Breeding, ICAR-National Dairy Research Institute, Karnal, Haryana, 132001, India
| | - Sachinandan De
- Division of Animal Biotechnology, ICAR-National Dairy Research Institute, Karnal, Haryana, 132001, India
| | - Ashutosh Ludri
- Division of Animal Physiology, ICAR-National Dairy Research Institute, Karnal, Haryana, 132001, India
| |
Collapse
|
247
|
Older EA, Mitchell MK, Campbell A, Lian X, Madden M, Wang Y, van de Wal LE, Zaw T, VanderVeen BN, Tatum R, Murphy EA, Chen YH, Fan D, Ellermann M, Li J. Human gut commensal Alistipes timonensis modulates the host lipidome and delivers anti-inflammatory outer membrane vesicles to suppress colitis in an Il10 -deficient mouse model. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.23.619966. [PMID: 39484420 PMCID: PMC11527014 DOI: 10.1101/2024.10.23.619966] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/03/2024]
Abstract
Correlative studies have linked human gut microbes to specific health conditions. Alistipes is one such microbial genus negatively linked to inflammatory bowel disease (IBD). However, the protective role of Alistipes in IBD has not been studied and the underlying molecular mechanisms also remain unknown. In this study, colonization of Il10 -deficient mice with Alistipes timonensis DSM 27924 delays the development of colitis. Colonization with Alistipes does not significantly alter the gut microbiome composition during colitis development, but instead shifts the host plasma lipidome, increasing phosphatidic acids while decreasing triglycerides. Outer membrane vesicles (OMVs) derived from Alistipes are also detected in the plasma of colonized mice, which carry metabolites with immunomodulatory potential into the host circulatory system. We further demonstrate that fractions of A. timonensis OMVs suppress LPS-induced Il6 , Il1b , and Tnfa expression in vitro in murine macrophages. We detect immunomodulatory sulfonolipids (SoLs) in the active fraction, which are also increased in the blood of A. timonensis -colonized mice; and we identify other putative bioactive lipids in the A. timonensis OMVs. Thus, A. timonensis OMVs represent a potential mechanism for Alistipes -mediated delay of colitis progression in Il10 -deficient mice through the delivery of immunomodulatory lipids, including SoLs, and modulation of the host plasma lipidome.
Collapse
|
248
|
Wang C, Wang C, Liu Y, Yue Y, Lu X, Wang H, Ying Y, Chen J. Targeted discovery of polyketides with antioxidant activity through integrated omics and cocultivation strategies. Appl Environ Microbiol 2024:e0160324. [PMID: 39445804 DOI: 10.1128/aem.01603-24] [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: 08/14/2024] [Accepted: 09/27/2024] [Indexed: 10/25/2024] Open
Abstract
Fungi generate a diverse array of bioactive compounds with significant pharmaceutical applications. However, the chemical diversity of natural products in fungi remains largely unexplored. Here, we present a paradigm for specifically discovering diverse and bioactive compounds from fungi by integrating genome mining with building block molecular network and coculture analysis. Through pangenome and sequence similarity network analysis, we identified a rare type I polyketide enzyme from Penicillium sp. ZJUT-34. Subsequent building block molecular network and coculture strategy led to the identification and isolation of a pair of novel polyketides, (±)-peniphenone E [(±)-1], three known polyketides (2-4), and three precursor compounds (5-7) from a combined culture of Penicillium sp. ZJUT-34 and Penicillium sp. ZJUT23. Their structures were established through extensive spectroscopic analysis, including NMR and HRESIMS. Chiral HPLC separation of compound 1 yielded a pair of enantiomers (+)-1 and (-)-1, with their absolute configurations determined using calculated ECD methods. Compound (±)-1 is notable for its unprecedented structure, featuring a unique 2-methyl-hexenyl-3-one moiety fused with a polyketide clavatol core. We proposed a hypothetical biosynthetic pathway for (±)-1. Furthermore, compounds 2, 5, and 6 exhibited strong antioxidant activity, whereas (-)-1, (+)-1, 3, and four exhibited moderate antioxidant activity compared to the positive control, ascorbic acid. Our research demonstrates a pioneering strategy for uncovering novel polyketides by merging genome mining, metabolomics, and cocultivation methods. This approach addresses the challenge of discovering natural compounds produced by rare biosynthetic enzymes that are often silent under conventional conditions due to gene regulation.IMPORTANCEPolyketides, particularly those with complex structures, are crucial in drug development and synthesis. This study introduces a novel approach to discover new polyketides by integrating genomics, metabolomics, and cocultivation strategies. By combining genome mining, building block molecular networks, and coculturing techniques, we identified and isolated a unique polyketide, (±)-peniphenone E, along with three known polyketides and three precursor compounds from Penicillium sp. ZJUT-34 and Penicillium sp. ZJUT23. This approach highlights the potential of using combined strategies to explore fungal chemical diversity and discover novel bioactive compounds. The successful identification of (±)-peniphenone E, with its distinctive structure, demonstrates the effectiveness of this integrated method in enhancing natural product discovery and underscores the value of innovative approaches in natural product research.
Collapse
Affiliation(s)
- Cancan Wang
- College of Pharmaceutical Science and Key Laboratory for Green Pharmaceutical Technologies and Related Equipment of Ministry of Education and Key Laboratory of Pharmaceutical Engineering of Zhejiang Province, Zhejiang University of Technology, Hangzhou, China
| | - Chenjie Wang
- College of Pharmaceutical Science and Key Laboratory for Green Pharmaceutical Technologies and Related Equipment of Ministry of Education and Key Laboratory of Pharmaceutical Engineering of Zhejiang Province, Zhejiang University of Technology, Hangzhou, China
| | - Yanjun Liu
- College of Pharmaceutical Science and Key Laboratory for Green Pharmaceutical Technologies and Related Equipment of Ministry of Education and Key Laboratory of Pharmaceutical Engineering of Zhejiang Province, Zhejiang University of Technology, Hangzhou, China
| | - Yujie Yue
- College of Pharmaceutical Science and Key Laboratory for Green Pharmaceutical Technologies and Related Equipment of Ministry of Education and Key Laboratory of Pharmaceutical Engineering of Zhejiang Province, Zhejiang University of Technology, Hangzhou, China
| | - Xingyue Lu
- College of Pharmaceutical Science and Key Laboratory for Green Pharmaceutical Technologies and Related Equipment of Ministry of Education and Key Laboratory of Pharmaceutical Engineering of Zhejiang Province, Zhejiang University of Technology, Hangzhou, China
| | - Hong Wang
- College of Pharmaceutical Science and Key Laboratory for Green Pharmaceutical Technologies and Related Equipment of Ministry of Education and Key Laboratory of Pharmaceutical Engineering of Zhejiang Province, Zhejiang University of Technology, Hangzhou, China
| | - Youmin Ying
- College of Pharmaceutical Science and Key Laboratory for Green Pharmaceutical Technologies and Related Equipment of Ministry of Education and Key Laboratory of Pharmaceutical Engineering of Zhejiang Province, Zhejiang University of Technology, Hangzhou, China
| | - Jianwei Chen
- College of Pharmaceutical Science and Key Laboratory for Green Pharmaceutical Technologies and Related Equipment of Ministry of Education and Key Laboratory of Pharmaceutical Engineering of Zhejiang Province, Zhejiang University of Technology, Hangzhou, China
| |
Collapse
|
249
|
Claverie C, Coppolino F, Mazzuoli MV, Guyonnet C, Jacquemet E, Legendre R, Sismeiro O, De Gaetano GV, Teti G, Trieu-Cuot P, Tazi A, Beninati C, Firon A. Constitutive activation of two-component systems reveals regulatory network interactions in Streptococcus agalactiae. Nat Commun 2024; 15:9175. [PMID: 39448655 PMCID: PMC11502775 DOI: 10.1038/s41467-024-53439-3] [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: 04/04/2024] [Accepted: 10/07/2024] [Indexed: 10/26/2024] Open
Abstract
Bacterial two-component systems (TCSs) are signaling modules that control physiology, adaptation, and host interactions. A typical TCS consists of a histidine kinase (HK) that activates a response regulator via phosphorylation in response to environmental signals. Here, we systematically test the effect of inactivating the conserved phosphatase activity of HKs to activate TCS signaling pathways. Transcriptome analyses of 14 HK mutants in Streptococcus agalactiae, the leading cause of neonatal meningitis, validate the conserved HK phosphatase mechanism and its role in the inhibition of TCS activity in vivo. Constitutive TCS activation, independent of environmental signals, enables high-resolution mapping of the regulons for several TCSs (e.g., SaeRS, BceRS, VncRS, DltRS, HK11030, HK02290) and reveals the functional diversity of TCS signaling pathways, ranging from highly specialized to interconnected global regulatory networks. Targeted analysis shows that the SaeRS-regulated PbsP adhesin acts as a signaling molecule to activate CovRS signaling, thereby linking the major regulators of host-pathogen interactions. Furthermore, constitutive BceRS activation reveals drug-independent activity, suggesting a role in cell envelope homeostasis beyond antimicrobial resistance. This study highlights the versatility of constitutive TCS activation, via phosphatase-deficient HKs, to uncover regulatory networks and biological processes.
Collapse
Affiliation(s)
- Cosme Claverie
- Institut Pasteur, Université Paris Cité, Department of Microbiology, Biology of Gram-Positive Pathogens, Paris, France
| | - Francesco Coppolino
- Institut Pasteur, Université Paris Cité, Department of Microbiology, Biology of Gram-Positive Pathogens, Paris, France
- University of Messina, Department of Human Pathology, Messina, Italy
| | - Maria-Vittoria Mazzuoli
- Institut Pasteur, Université Paris Cité, Department of Microbiology, Biology of Gram-Positive Pathogens, Paris, France
| | - Cécile Guyonnet
- Assistance Publique-Hôpitaux de Paris, Hôpital Cochin, Department of Bacteriology, French National Reference Center for Streptococci, Paris, France
- Université Paris Cité, Institut Cochin, Institut National de la Santé et de la Recherche Médicale U1016, Centre National de la Recherche Scientifique UMR8104, Team Bacteria and Perinatality, Paris, France
- Fédération Hospitalo-Universitaire Fighting Prematurity, Paris, France
| | - Elise Jacquemet
- Institut Pasteur, Université Paris Cité, Bioinformatics and Biostatistics Hub, Paris, France
| | - Rachel Legendre
- Institut Pasteur, Université Paris Cité, Bioinformatics and Biostatistics Hub, Paris, France
| | - Odile Sismeiro
- Institut Pasteur, Université Paris Cité, Department of Microbiology, Biology of Gram-Positive Pathogens, Paris, France
| | | | | | - Patrick Trieu-Cuot
- Institut Pasteur, Université Paris Cité, Department of Microbiology, Biology of Gram-Positive Pathogens, Paris, France
| | - Asmaa Tazi
- Assistance Publique-Hôpitaux de Paris, Hôpital Cochin, Department of Bacteriology, French National Reference Center for Streptococci, Paris, France
- Université Paris Cité, Institut Cochin, Institut National de la Santé et de la Recherche Médicale U1016, Centre National de la Recherche Scientifique UMR8104, Team Bacteria and Perinatality, Paris, France
- Fédération Hospitalo-Universitaire Fighting Prematurity, Paris, France
| | - Concetta Beninati
- University of Messina, Department of Human Pathology, Messina, Italy
| | - Arnaud Firon
- Institut Pasteur, Université Paris Cité, Department of Microbiology, Biology of Gram-Positive Pathogens, Paris, France.
| |
Collapse
|
250
|
Eyuboglu S, Alpsoy S, Uversky VN, Coskuner-Weber O. Key genes and pathways in the molecular landscape of pancreatic ductal adenocarcinoma: A bioinformatics and machine learning study. Comput Biol Chem 2024; 113:108268. [PMID: 39467488 DOI: 10.1016/j.compbiolchem.2024.108268] [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: 10/03/2024] [Accepted: 10/20/2024] [Indexed: 10/30/2024]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is recognized for its aggressive nature, dismal prognosis, and a notably low five-year survival rate, underscoring the critical need for early detection methods and more effective therapeutic approaches. This research rigorously investigates the molecular mechanisms underlying PDAC, with a focus on the identification of pivotal genes and pathways that may hold therapeutic relevance and prognostic value. Through the construction of a protein-protein interaction (PPI) network and the examination of differentially expressed genes (DEGs), the study uncovers key hub genes such as CDK1, KIF11, and BUB1, demonstrating their substantial role in the pathogenesis of PDAC. Notably, the dysregulation of these genes is consistent across a spectrum of cancers, positing them as potential targets for wide-ranging cancer therapeutics. This study also brings to the fore significant genes encoding intrinsically disordered proteins, in particular GPRC5A and KRT7, unveiling promising new pathways for therapeutic intervention. Advanced machine learning techniques were harnessed to classify PDAC patients with high accuracy, utilizing the key genetic markers as a dataset. The Support Vector Machine (SVM) model leveraged the hub genes to achieve a sensitivity of 91 % and a specificity of 85 %, while the RandomForest model notched a sensitivity of 91 % and specificity of 92.5 %. Crucially, when the identified genes were cross-referenced with TCGA-PAAD clinical datasets, a tangible correlation with patient survival rates was discovered, reinforcing the potential of these genes as prognostic biomarkers and their viability as targets for therapeutic intervention. This study's findings serve as a potent testament to the value of molecular analysis in enhancing the understanding of PDAC and in advancing the pursuit for more effective diagnostic and treatment strategies.
Collapse
Affiliation(s)
- Sinan Eyuboglu
- Turkish-German University, Molecular Biotechnology, Sahinkaya Caddesi, No. 106, Beykoz, Istanbul 34820, Turkey
| | - Semih Alpsoy
- Turkish-German University, Molecular Biotechnology, Sahinkaya Caddesi, No. 106, Beykoz, Istanbul 34820, Turkey
| | - Vladimir N Uversky
- USF Health Byrd Alzheimer's Research Institute, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA
| | - Orkid Coskuner-Weber
- Turkish-German University, Molecular Biotechnology, Sahinkaya Caddesi, No. 106, Beykoz, Istanbul 34820, Turkey.
| |
Collapse
|