1
|
Ishikawa C, Barreyro L, Sampson AM, Hueneman KM, Choi K, Philbrook SY, Choi I, Bolanos LC, Wunderlich M, Volk AG, Watowich SS, Greis KD, Starczynowski DT. Ubiquitin-conjugating enzyme UBE2N modulates proteostasis in immunoproteasome-positive acute myeloid leukemia. J Clin Invest 2025; 135:e184665. [PMID: 40371639 PMCID: PMC12077902 DOI: 10.1172/jci184665] [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: 07/31/2024] [Accepted: 03/06/2025] [Indexed: 05/16/2025] Open
Abstract
Altered protein homeostasis through proteasomal degradation of ubiquitinated proteins is a hallmark of many cancers. Ubiquitination, coordinated by E1, E2, and E3 enzymes, involves up to 40 E2-conjugating enzymes in humans to specify substrates and ubiquitin linkages. In a screen for E2 dependencies in acute myeloid leukemia (AML), ubiquitin conjugating enzyme E2 N (UBE2N) emerged as the top candidate. To investigate UBE2N's role in AML, we characterized an enzymatically defective mouse model of UBE2N, revealing UBE2N's requirement in AML without an impact on normal hematopoiesis. Unlike other E2s, which mediate lysine-48 (K48) polyubiquitination and degradation of proteins, UBE2N primarily synthesizes K63-linked chains, stabilizing or altering protein function. Proteomic analyses and a whole-genome CRISPR-activation screen in pharmacologically and genetically UBE2N-inhibited AML cells unveiled a network of UBE2N-regulated proteins, many of which are implicated in cancer. UBE2N inhibition reduced their protein levels, leading to increased K48-linked ubiquitination and degradation through the immunoproteasome and revealing UBE2N activity is enriched in immunoproteasome-positive AML. Furthermore, an interactome screen identified tripartite motif-containing protein 21 (TRIM21) as the E3 ligase partnering with activated UBE2N in AML to modulate UBE2N-dependent proteostasis. In conclusion, UBE2N maintains proteostasis in AML by stabilizing target proteins through K63-linked ubiquitination and prevention of K48 ubiquitin-mediated degradation by the immunoproteasome. Thus, inhibition of UBE2N catalytic function suppresses leukemic cells through selective degradation of critical proteins in immunoproteasome-positive AML.
Collapse
Affiliation(s)
- Chiharu Ishikawa
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA
- Department of Cancer Biology, University of Cincinnati, Cincinnati, Ohio, USA
| | - Laura Barreyro
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA
| | - Avery M. Sampson
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA
| | - Kathleen M. Hueneman
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA
| | - Kwangmin Choi
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA
| | - Sophia Y. Philbrook
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA
| | - Issac Choi
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA
- Department of Cancer Biology, University of Cincinnati, Cincinnati, Ohio, USA
| | - Lyndsey C. Bolanos
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA
| | - Mark Wunderlich
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA
| | - Andrew G. Volk
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA
- Department of Cancer Biology, University of Cincinnati, Cincinnati, Ohio, USA
| | | | - Kenneth D. Greis
- Department of Cancer Biology, University of Cincinnati, Cincinnati, Ohio, USA
| | - Daniel T. Starczynowski
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA
- Department of Cancer Biology, University of Cincinnati, Cincinnati, Ohio, USA
- Department of Pediatrics, University of Cincinnati, Cincinnati, Ohio, USA
- University of Cincinnati Cancer Center, Cincinnati, USA
| |
Collapse
|
2
|
Shah M, Anum H, Sarfraz A, Aktaruzzaman M, Hasan AR, Khan MU, Fawy KF, Altwaim SA, Alasmari SMN, Ali A, Nishan U, Chen K. Bioinformatics-guided decoding of the Ancylostoma duodenale genome for the identification of potential vaccine targets. BMC Genomics 2025; 26:468. [PMID: 40355819 PMCID: PMC12067957 DOI: 10.1186/s12864-025-11652-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2025] [Accepted: 04/29/2025] [Indexed: 05/15/2025] Open
Abstract
Ancylostoma duodenale, a parasitic nematode worm, is found to be involved in various infections, including intestinal blood loss, protein malnutrition, and anemia. Antimicrobial resistance to the available therapeutics has prompted the search for new drug and vaccine targets against A. duodenale. Despite significant advances in vaccine development against A. duodenale, no commercial and FDA-approved vaccine exists to safeguard humans from infections caused by this pathogen. In this investigation, a stringent bioinformatics analysis identified 36 unique essential and host-interacting proteins. Based on their subcellular localization, 6 proteins located in the extracellular space and outer membrane were categorized as vaccine targets, while the remaining proteins were predicted to act as potential drug candidates. These vaccine candidates were further assessed for antigenicity, allergenicity, and physicochemical analysis to determine their suitability for the designing of a multi-epitope vaccine. Two candidate proteins were chosen as optimal targets in the development of vaccine design. The identified T- and B-cell epitopes from these proteins were then combined with appropriate linkers and adjuvants to design chimeric vaccine constructs aimed at inducing both cellular and humoral immune responses. Molecular docking, molecular dynamic simulations, PCA analysis, DCCM analysis, and binding free energy calculations proved stable interactions of the designed vaccine with human immune cell receptors. Within a bacterial cloning system, the vaccine constructs demonstrated the ability to be cloned and expressed. The immunological stimulation elicited significant immunological responses to the proposed vaccine. Our investigation identified new therapeutic targets and developed a peptide-based multi-epitope vaccine against A. duodenale infection. Additional experimental verification will open up new therapeutic alternatives for this emerging resistant pathogen.
Collapse
Affiliation(s)
- Mohibullah Shah
- Department of Biochemistry, Bahauddin Zakariya University, Multan, Punjab, 66000, Pakistan.
- Department of Animal Science, Federal University of Ceara, Fortaleza, Brazil.
| | - Hira Anum
- Department of Biochemistry, Bahauddin Zakariya University, Multan, Punjab, 66000, Pakistan
| | - Asifa Sarfraz
- Department of Biochemistry, Bahauddin Zakariya University, Multan, Punjab, 66000, Pakistan
| | - Md Aktaruzzaman
- Department of Pharmacy, Faculty of Biological Science and Technology, Jashore University of Science and Technology, Jashore, 7408, Bangladesh
| | - Al Riyad Hasan
- Department of Pharmacy, Faculty of Biological Science and Technology, Jashore University of Science and Technology, Jashore, 7408, Bangladesh
| | - Muhammad Umer Khan
- Institute of Molecular Biology and Biotechnology, The University of Lahore, Lahore, Pakistan
| | - Khaled Fahmi Fawy
- Chemistry Department, Faculty of Science, King Khalid University, P.O. Box 9004, Abha, 61413, Saudi Arabia
- Research Center for Advanced Materials Science (RCAMS), King Khalid University, P.O. Box 960, AlQura'a, Abha, Saudi Arabia
| | - Sarah A Altwaim
- Department of Clinical Microbiology and Immunology, Faculty of Medicine, King Abdulaziz University, Jeddah, 21589, Saudi Arabia
- Special Infectious Agents Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Saeed M N Alasmari
- Department of Biology, Faculty of Science and Arts, Najran University, Najran, 1988, Saudi Arabia
| | - Abid Ali
- Department of Zoology, Abdul Wali Khan University, Mardan, Khyber Pakhtunkhwa, 23200, Pakistan
| | - Umar Nishan
- Department of Chemistry, Kohat University of Science & Technology, Kohat, Pakistan
| | - Ke Chen
- Department of Infectious Diseases, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China.
| |
Collapse
|
3
|
Hasnat S, Rahman MM, Yeasmin F, Jubair M, Helmy YA, Islam T, Hoque MN. Genomic and Computational Analysis Unveils Bacteriocin Based Therapeutics against Clinical Mastitis Pathogens in Dairy Cows. Probiotics Antimicrob Proteins 2025:10.1007/s12602-025-10563-w. [PMID: 40295467 DOI: 10.1007/s12602-025-10563-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/24/2025] [Indexed: 04/30/2025]
Abstract
Clinical mastitis (CM) remains a critical challenge in dairy production, exacerbated by the global rise of antibiotic-resistant pathogens, which threatens herd health and productivity. This study pioneers a dual genomic-computational strategy to develop bacteriocin-based therapeutics-a promising alternative to conventional antibiotics-by targeting conserved virulence mechanisms in CM-causing pathogens. We aimed to (i) identify essential core proteins in CM-causing pathogens of dairy cows using the genomic approach; and (ii) assess the efficacy of bacteriocin peptides (BPs) as novel therapeutic agents targeting the selected core proteins for sustainable management of mastitis. Through pan-genomic analysis of 16 clinically relevant pathogens, including Staphylococcus aureus, S. warneri, Streptococcus agalactiae, S. uberis, Escherichia coli, Klebsiella pneumoniae, Pseudomonas aeruginosa, P. putida, and P. asiatica, we identified 65 evolutionarily conserved core proteins. Prioritization based on essentiality, virulence, and resistance potential revealed Rho (transcription termination factor) and HupB (nucleoid-associated protein) as high-value therapeutic targets due to their critical roles in bacterial survival and pathogenicity. A computational screen of 70 BPs identified 14 candidates with high binding affinity for both Rho and HupB proteins. Molecular dynamics simulations demonstrated that BP8, a novel dual-action bacteriocin, competitively inhibits Rho-mediated transcription termination and disrupts HupB-DNA interactions, effectively crippling bacterial replication and virulence. BP8 exhibited superior structural stability and binding efficacy compared to other candidates, positioning it as a potent broad-spectrum agent against diverse CM pathogens, including multidrug-resistant strains. Our study underscores the untapped potential of bacteriocins in veterinary medicine, offering a sustainable solution to mitigate antibiotic overuse and resistance. The computational validation of BP8 provides a foundational framework for developing targeted therapies, with implications for reducing dairy industry losses and improving animal welfare. Further in vitro and in vivo studies are warranted to translate these insights into practical therapeutics.
Collapse
Affiliation(s)
- Soharth Hasnat
- Molecular Biology and Bioinformatics Laboratory, Department of Gynecology, Obstetrics and Reproductive Health, Gazipur Agricultural University, Gazipur, 1706, Bangladesh
| | - Md Morshedur Rahman
- Molecular Biology and Bioinformatics Laboratory, Department of Gynecology, Obstetrics and Reproductive Health, Gazipur Agricultural University, Gazipur, 1706, Bangladesh
| | - Farzana Yeasmin
- Institute of Biotechnology and Genetic Engineering, Gazipur Agricultural University, Gazipur, 1706, Bangladesh
| | - Mohammad Jubair
- iccdr'b (International Centre for Diarrhoeal Disease Research, Bangladesh), Dhaka, 1212, Bangladesh
| | - Yosra A Helmy
- Department of Veterinary Science, University of Kentucky, 1400 Nicholasville Rd., Lexington, KY, 40546-0099, USA
| | - Tofazzal Islam
- Institute of Biotechnology and Genetic Engineering, Gazipur Agricultural University, Gazipur, 1706, Bangladesh.
| | - M Nazmul Hoque
- Molecular Biology and Bioinformatics Laboratory, Department of Gynecology, Obstetrics and Reproductive Health, Gazipur Agricultural University, Gazipur, 1706, Bangladesh.
| |
Collapse
|
4
|
Arriagada V, Osorio A, Carrera-Naipil C, Villacis-Aguirre CA, Escobar C, Morales N, Villa D, Mardones L, Pérez D, Jara M, Molina RE, Ferrari Í, Azocar S, Gómez LA, Oñate ÁA. In Silico Design and Characterization of a Multiepitope Vaccine Candidate Against Brucella canis Using a Reverse Vaccinology Approach. J Immunol Res 2025; 2025:6348238. [PMID: 40265107 PMCID: PMC12014272 DOI: 10.1155/jimr/6348238] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Accepted: 03/13/2025] [Indexed: 04/24/2025] Open
Abstract
Brucella canis is a Gram-negative bacterium that causes canine brucellosis, a zoonotic disease with serious implications for public health and the global economy. Currently, there is no effective preventive vaccine for B. canis. Control measures include diagnostic testing, isolation, and euthanasia of infected animals. However, these measures face significant limitations, such as diagnostic challenges, ethical concerns, and limited success in preventing transmission. Epidemiologically, canine brucellosis exhibits seroprevalence rates ranging from less than 1% to over 15%, with higher rates reported in stray dogs and regions of low socioeconomic development. This study employed a reverse vaccinology approach to design and characterize a multiepitope vaccine candidate against B. canis, aiming to prevent infection caused by this pathogen. A comprehensive in silico analysis of the complete B. canis proteome was conducted to identify proteins with potential as vaccine targets. Predicted epitopes for B and T cells were analyzed, and those with the highest capacity to elicit a robust immune response were selected. These proteins were classified as plasma membrane proteins, outer membrane proteins (OMPs), or proteins with similarity to virulence factors. Selection criteria emphasized their essential roles in bacterial function, lack of homology with proteins from dogs or mice, and presence of fewer than two transmembrane domains. From this process, four candidate proteins were identified. Epitopes for B and T cells within these proteins were predicted and analyzed, selecting the most immunogenic sequences. The overlap between B- and T-cell epitopes narrowed the selection to six final epitopes. These selected epitopes were then assembled into a multiepitope vaccine construct using flexible linkers to ensure structural integrity and molecular adjuvants to enhance immunogenicity. The physicochemical properties, antigenicity, and toxicity of the designed vaccine were evaluated. Additionally, the secondary and tertiary structure of the vaccine was predicted and refined, followed by a molecular interaction analysis with the Toll-like receptor 4 (TLR4) receptor. The designed vaccine proved to be highly antigenic, nonallergenic, and nontoxic. Validation of its secondary and tertiary structures, along with molecular docking analysis, revealed a high binding affinity to the TLR4 receptor. Molecular dynamics simulations and normal mode analysis further confirmed the vaccine's structural stability and binding capacity. A multiepitope vaccine candidate against B. canis was successfully designed and characterized using a reverse vaccinology approach. This vaccine construct is expected to induce robust humoral and cellular immune responses, potentially conferring protective immunity against B. canis. The results of this study are promising; however, in vitro and in vivo tests are necessary to validate the vaccine's protective efficacy. Furthermore, the described method could serve as a framework for developing vaccines against other pathogens.
Collapse
Affiliation(s)
- Vicente Arriagada
- Laboratory of Molecular Immunology, Department of Microbiology, Faculty of Biological Sciences, University of Concepción, Concepción, Chile
| | | | - Crisleri Carrera-Naipil
- Laboratory of Molecular Immunology, Department of Microbiology, Faculty of Biological Sciences, University of Concepción, Concepción, Chile
| | - Carlos A. Villacis-Aguirre
- Laboratory of Molecular Immunology, Department of Microbiology, Faculty of Biological Sciences, University of Concepción, Concepción, Chile
| | - Cristian Escobar
- Laboratory of Molecular Immunology, Department of Microbiology, Faculty of Biological Sciences, University of Concepción, Concepción, Chile
| | - Nicolás Morales
- Laboratory of Molecular Immunology, Department of Microbiology, Faculty of Biological Sciences, University of Concepción, Concepción, Chile
| | - Danthe Villa
- Laboratory of Molecular Immunology, Department of Microbiology, Faculty of Biological Sciences, University of Concepción, Concepción, Chile
| | - Lien Mardones
- Laboratory of Molecular Immunology, Department of Microbiology, Faculty of Biological Sciences, University of Concepción, Concepción, Chile
| | - Dafne Pérez
- Laboratory of Molecular Immunology, Department of Microbiology, Faculty of Biological Sciences, University of Concepción, Concepción, Chile
| | - Macarena Jara
- Laboratory of Molecular Immunology, Department of Microbiology, Faculty of Biological Sciences, University of Concepción, Concepción, Chile
| | - Raúl E. Molina
- Laboratory of Molecular Immunology, Department of Microbiology, Faculty of Biological Sciences, University of Concepción, Concepción, Chile
| | - Ítalo Ferrari
- Laboratory of Molecular Immunology, Department of Microbiology, Faculty of Biological Sciences, University of Concepción, Concepción, Chile
| | - Sebastián Azocar
- Laboratory of Molecular Immunology, Department of Microbiology, Faculty of Biological Sciences, University of Concepción, Concepción, Chile
| | - Leonardo A. Gómez
- Laboratory of Molecular Immunology, Department of Microbiology, Faculty of Biological Sciences, University of Concepción, Concepción, Chile
| | - Ángel A. Oñate
- Laboratory of Molecular Immunology, Department of Microbiology, Faculty of Biological Sciences, University of Concepción, Concepción, Chile
| |
Collapse
|
5
|
Dingding H, Muhammad S, Manzoor I, Ghaffar SA, Alodaini HA, Moubayed NMS, Hatamleh AA, Songxiao X. Subtractive proteomics and reverse-vaccinology approaches for novel drug targets and designing a chimeric vaccine against Ruminococcus gnavus strain RJX1120. Front Immunol 2025; 16:1555741. [PMID: 40297578 PMCID: PMC12034673 DOI: 10.3389/fimmu.2025.1555741] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2025] [Accepted: 03/25/2025] [Indexed: 04/30/2025] Open
Abstract
Mediterraneibacter gnavus, also known as Ruminococcus gnavus, is a Gram-positive anaerobic bacterium that resides in the human gut microbiota. Notably, this bacterium plays dual roles in health and disease. On one side it supports nutrient metabolism essential for bodily functions and on the other it contributes to the development of Inflammatory Bowel Disease (IBD) and other gastrointestinal disorders. R. gnavus strain RJX1120 is an encapsulated strain and has been linked to develop IBD. Despite the advances made on its role in gut homeostasis, limited information is available on strain-specific virulence factors, metabolic pathways, and regulatory mechanisms. The study of such aspects is crucial to make microbiota-targeted therapy and understand its implications in host health. A multi-epitope vaccine against R. gnavus strain RJX1120 was designed using reverse vaccinology-based subtractive proteomics approach. Among the 3,219 proteins identified in the R. gnavus strain RJX1120, two critical virulent and antigenic proteins, a Single-stranded DNA-binding protein SSB (A0A2N5PT08) and Cell division ATP-binding protein FtsE (A0A2N5NK05) were screened and identified as potential targets. The predicted B-cell and T-cell epitopes from these proteins were screened for essential immunological properties such as antigenicity, allergenicity, solubility, MHC binding affinity, and toxicity. Epitopes chosen were cross-linked using suitable spacers and an adjuvant to develop a multi-epitope vaccine. Structural refinement of the construct revealed that 95.7% of the amino acid residues were located in favored regions, indicating a high-quality structural model. Molecular docking analysis demonstrated a robust interaction between the vaccine construct and the human Toll-like receptor 4 (TLR4), with a binding energy of -1277.0 kcal/mol. The results of molecular dynamics simulations further confirmed the stability of the vaccine-receptor complex under physiological conditions. In silico cloning of the vaccine construct yielded a GC content of 48% and a Codon Adaptation Index (CAI) value of 1.0, indicating optimal expression in the host system. These results indicate the possibility of the designed vaccine construct as a candidate for the prevention of R. gnavus-associated diseases. However, experimental validation is required to confirm its immunogenicity and protective efficacy.
Collapse
Affiliation(s)
- Hou Dingding
- Department of Clinical Laboratory, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, China
- Postgraduate Training Base Alliance of Wenzhou Medical University (Zhejiang Cancer Hospital), Hangzhou, Zhejiang, China
| | - Sher Muhammad
- Faculty of Agriculture and Veterinary Sciences, Superior University Lahore, Lahore, Pakistan
| | - Irfan Manzoor
- Department of Bioinformatics and Biotechnology, Government College University Faisalabad (GCUF), Faisalabad, Pakistan
| | - Sana Abdul Ghaffar
- Department of Bioinformatics and Biotechnology, Government College University Faisalabad (GCUF), Faisalabad, Pakistan
| | | | - Nadine MS. Moubayed
- Department of Botany and Microbiology, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Ashraf Atef Hatamleh
- Department of Botany and Microbiology, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Xu Songxiao
- Department of Clinical Laboratory, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, China
| |
Collapse
|
6
|
Syed R, Aghayeva S, Uddin R, Subramaniyan V, Hassan ESG, Abdelhameed AS, Harshini M, Wadood A. A Computational Quest for Finding Novel Drug Targets against Mycobacterium tuberculosis. Indian J Microbiol 2025. [DOI: 10.1007/s12088-025-01473-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Accepted: 03/21/2025] [Indexed: 05/04/2025] Open
|
7
|
Asim MN, Ibrahim MA, Zaib A, Dengel A. DNA sequence analysis landscape: a comprehensive review of DNA sequence analysis task types, databases, datasets, word embedding methods, and language models. Front Med (Lausanne) 2025; 12:1503229. [PMID: 40265190 PMCID: PMC12011883 DOI: 10.3389/fmed.2025.1503229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2024] [Accepted: 03/10/2025] [Indexed: 04/24/2025] Open
Abstract
Deoxyribonucleic acid (DNA) serves as fundamental genetic blueprint that governs development, functioning, growth, and reproduction of all living organisms. DNA can be altered through germline and somatic mutations. Germline mutations underlie hereditary conditions, while somatic mutations can be induced by various factors including environmental influences, chemicals, lifestyle choices, and errors in DNA replication and repair mechanisms which can lead to cancer. DNA sequence analysis plays a pivotal role in uncovering the intricate information embedded within an organism's genetic blueprint and understanding the factors that can modify it. This analysis helps in early detection of genetic diseases and the design of targeted therapies. Traditional wet-lab experimental DNA sequence analysis through traditional wet-lab experimental methods is costly, time-consuming, and prone to errors. To accelerate large-scale DNA sequence analysis, researchers are developing AI applications that complement wet-lab experimental methods. These AI approaches can help generate hypotheses, prioritize experiments, and interpret results by identifying patterns in large genomic datasets. Effective integration of AI methods with experimental validation requires scientists to understand both fields. Considering the need of a comprehensive literature that bridges the gap between both fields, contributions of this paper are manifold: It presents diverse range of DNA sequence analysis tasks and AI methodologies. It equips AI researchers with essential biological knowledge of 44 distinct DNA sequence analysis tasks and aligns these tasks with 3 distinct AI-paradigms, namely, classification, regression, and clustering. It streamlines the integration of AI into DNA sequence analysis tasks by consolidating information of 36 diverse biological databases that can be used to develop benchmark datasets for 44 different DNA sequence analysis tasks. To ensure performance comparisons between new and existing AI predictors, it provides insights into 140 benchmark datasets related to 44 distinct DNA sequence analysis tasks. It presents word embeddings and language models applications across 44 distinct DNA sequence analysis tasks. It streamlines the development of new predictors by providing a comprehensive survey of 39 word embeddings and 67 language models based predictive pipeline performance values as well as top performing traditional sequence encoding-based predictors and their performances across 44 DNA sequence analysis tasks.
Collapse
Affiliation(s)
- Muhammad Nabeel Asim
- German Research Center for Artificial Intelligence GmbH, Kaiserslautern, Germany
- Intelligentx GmbH (intelligentx.com), Kaiserslautern, Germany
| | - Muhammad Ali Ibrahim
- German Research Center for Artificial Intelligence GmbH, Kaiserslautern, Germany
- Department of Computer Science, Technical University of Kaiserslautern, Kaiserslautern, Germany
| | - Arooj Zaib
- Department of Computer Science, Technical University of Kaiserslautern, Kaiserslautern, Germany
| | - Andreas Dengel
- German Research Center for Artificial Intelligence GmbH, Kaiserslautern, Germany
- Intelligentx GmbH (intelligentx.com), Kaiserslautern, Germany
- Department of Computer Science, Technical University of Kaiserslautern, Kaiserslautern, Germany
| |
Collapse
|
8
|
Moradkasani S, Esmaeili S, Asadi Karam MR, Mostafavi E, Shahbazi B, Salek Farrokhi A, Chiani M, Badmasti F. Development of a multi-epitope vaccine from outer membrane proteins and identification of novel drug targets against Francisella tularensis: an In Silico approach. Front Immunol 2025; 16:1479862. [PMID: 40248715 PMCID: PMC12003292 DOI: 10.3389/fimmu.2025.1479862] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Accepted: 03/17/2025] [Indexed: 04/19/2025] Open
Abstract
Background Francisella tularensis is a category A potential thread agent, making the development of vaccines and countermeasures a high priority. Therefore, identifying new vaccine candidates and novel drug targets is essential for addressing this significant public health concern. Methods This study presents an in silico analysis of two strategies against F. tularensis infection: the development of a multi-epitope vaccine (MEV) and the identification of novel drug targets. Outer membrane proteins (OMPs) were predicted using subcellular localization tools and immunogenicity was evaluated using a reverse vaccinology pipeline. Epitopes from these OMPs were combined to create candidate MEV for prophylactic protection. Concurrently, cytoplasmic proteins were subjected to rigorous analysis to identify potential novel drug targets. Results Of 1,921 proteins, we identified 12 promising protein vaccine candidates from F. tularensis OMPs and proposed a multi-epitope vaccine (MEV) designed using seven immunodominant epitopes derived from four of these OMPs, including two hypothetical proteins (WP_003026145.1 and WP_003029346.1), an OmpA family protein (WP_003020808.1), and PD40 (WP_003021546.1). In addition, we proposed 10 novel drug targets for F. tularensis: Asp-tRNA (Asn)/Glu-tRNA (Gln) amidotransferase subunit GatC (WP_003017413.1), NAD(P)-binding protein (WP_042522581.1), 30S ribosomal protein S16 (WP_003023081.1), Class I SAM-dependent methyltransferase (WP_003022345.1), haloacid dehalogenase (WP_003014157.1), uroporphyrinogen-III synthase (WP_003022220.1), and four hypothetical proteins (WP_003017784.1, WP_003020080.1, WP_003020066.1, and WP_003022350.1). Conclusion This study designed an MEV and proposed novel drug targets to address tularemia, offering broad protection against various F. tularensis strains. MEV, with favorable physicochemical properties, showed strong potential through molecular docking and dynamic simulations. Immune simulations suggest that it may elicit robust responses against pathogens. The identification of novel drug targets can lead to the discovery of new antimicrobial agents. However, further in vitro and in vivo studies are required to validate their efficacy and capability.
Collapse
Affiliation(s)
- Safoura Moradkasani
- WHO Collaborating Centre for Vector-Borne Diseases, Department of Epidemiology and Biostatistics, Research Centre for Emerging and Reemerging Infectious Diseases, Pasteur Institute of Iran, Tehran, Iran
- Student Research Committee, Pasteur Institute of Iran, Tehran, Iran
| | - Saber Esmaeili
- WHO Collaborating Centre for Vector-Borne Diseases, Department of Epidemiology and Biostatistics, Research Centre for Emerging and Reemerging Infectious Diseases, Pasteur Institute of Iran, Tehran, Iran
- Student Research Committee, Pasteur Institute of Iran, Tehran, Iran
- National Reference Laboratory for Plague, Tularemia and Q Fever, Research Centre for Emerging and Reemerging Infectious Diseases, Pasteur Institute of Iran, Akanlu, KabudarAhang, Hamadan, Iran
| | | | - Ehsan Mostafavi
- WHO Collaborating Centre for Vector-Borne Diseases, Department of Epidemiology and Biostatistics, Research Centre for Emerging and Reemerging Infectious Diseases, Pasteur Institute of Iran, Tehran, Iran
- National Reference Laboratory for Plague, Tularemia and Q Fever, Research Centre for Emerging and Reemerging Infectious Diseases, Pasteur Institute of Iran, Akanlu, KabudarAhang, Hamadan, Iran
| | - Behzad Shahbazi
- School of Pharmacy, Semnan University of Medical Sciences, Semnan, Iran
- Nervous System Stem Cells Research Center, Semnan University of Medical Sciences, Semnan, Iran
| | | | - Mohsen Chiani
- Department of Nanobiotechnology, Pasteur Institute of Iran, Tehran, Iran
| | - Farzad Badmasti
- Department of Bacteriology, Pasteur Institute of Iran, Tehran, Iran
| |
Collapse
|
9
|
Alshamrani S, Mashraqi MM, Alzamami A, Alturki NA, Almasoudi HH, Ahmed I, Basharat Z. Leveraging core proteome data of Kingella kingae for multi-epitope vaccine design against TonB dependent receptor (TDR): an in silico approach. J Biomol Struct Dyn 2025:1-18. [PMID: 40105736 DOI: 10.1080/07391102.2025.2480263] [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: 09/19/2023] [Accepted: 04/29/2024] [Indexed: 03/20/2025]
Abstract
Kingella kingae is a Gram-negative bacterium that causes invasive infections in children and older or immunocompromised individuals, making it a significant public health concern. In this study, a pan-proteomic mediated vaccine target mining was attempted to identify potential vaccine targets in K. kingae. Currently, there is no vaccine available against this pathobiont. Therefore, we designed and validated an in silico vaccine construct by targeting the lactoferrin/transferrin-binding TonB-dependent receptor. Antigenic regions of the TonB receptor were mapped, and the predicted epitopes were anticipated to be effective in a broad range of the world population. Using their combinations with linkers and various adjuvants, 12 vaccine constructs were prepared. The best construct (C7) with no allergenicity and high antigenicity was subjected to molecular modeling, docking with important immune receptors of humans, and then molecular dynamics (MD) simulation. After binding validation and stability assessment, it was cloned into a pet-28a + plasmid vector. Immune response was also simulated, and the vaccine was observed to invoke B- and T-cell induction. These findings can help accelerate the development of a new vaccine against K. kingae or other pathogens targeting the homolog of TonB. Nevertheless, we propose additional testing of C7 construct for efficacy and safety in vitro and in vivo.
Collapse
Affiliation(s)
- Saleh Alshamrani
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Najran University, Najran, Saudi Arabia
| | - Mutaib M Mashraqi
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Najran University, Najran, Saudi Arabia
| | - Ahmad Alzamami
- Clinical Laboratory Science Department, College of Applied Medical Science, Shaqra University, AlQuwayiyah, Saudi Arabia
| | - Norah A Alturki
- Clinical Laboratory Science Department, College of Applied Medical Science, King Saud University, Riyadh, Saudi Arabia
| | - Hassan H Almasoudi
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Najran University, Najran, Saudi Arabia
| | - Ibrar Ahmed
- Alpha Genomics Private Limited, Islamabad, Pakistan
- Group for Biometrology, Korea Research Institute of Standards and Science (KRISS), Daejeon, Republic of Korea
| | | |
Collapse
|
10
|
Gohain BB, Mazumder B, Rajkhowa S, Al-Hussain SA, Zaki MEA. Subtractive genomics and drug repurposing strategies for targeting Streptococcus pneumoniae: insights from molecular docking and dynamics simulations. Front Microbiol 2025; 16:1534659. [PMID: 40170924 PMCID: PMC11958985 DOI: 10.3389/fmicb.2025.1534659] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Accepted: 02/28/2025] [Indexed: 04/03/2025] Open
Abstract
Introduction Streptococcus pneumoniae is a Gram-positive bacterium responsible for severe infections such as meningitis and pneumonia. The increasing prevalence of antibiotic resistance necessitates the identification of new therapeutic targets. This study aimed to discover potential drug targets against S. pneumoniae using an in silico subtractive genomics approach. Methods The S. pneumoniae genome was compared to the human genome to identify non-homologous sequences using CD-HIT and BLASTp. Essential genes were identified using the Database of Essential Genes (DEG), with consideration for human gut microflora. Protein-protein interaction analyses were conducted to identify key hub genes, and gene ontology (GO) studies were performed to explore associated pathways. Due to the lack of crystal structure data, a potential target was modeled in silico and subjected to structure-based virtual screening. Results Approximately 2,000 of the 2,027 proteins from the S. pneumoniae genome were identified as non-homologous to humans. The DEG identified 48 essential genes, which was reduced to 21 after considering human gut microflora. Key hub genes included gpi, fba, rpoD, and trpS, associated with 20 pathways. Virtual screening of 2,509 FDA-approved compounds identified Bromfenac as a leading candidate, exhibiting a binding energy of -26.335 ± 29.105 kJ/mol. Discussion Bromfenac, particularly when conjugated with AuAgCu2O nanoparticles, has demonstrated antibacterial and anti-inflammatory properties against Staphylococcus aureus. This suggests that Bromfenac could be repurposed as a potential therapeutic agent against S. pneumoniae, pending further experimental validation. The approach highlights the potential for drug repurposing by targeting proteins essential in pathogens but absent in the host.
Collapse
Affiliation(s)
- Borakha Bura Gohain
- Centre for Biotechnology and Bioinformatics, Dibrugarh University, Dibrugarh, Assam, India
| | - Bhaskar Mazumder
- Department of Pharmaceutical Sciences, Dibrugarh University, Dibrugarh, Assam, India
| | - Sanchaita Rajkhowa
- Centre for Biotechnology and Bioinformatics, Dibrugarh University, Dibrugarh, Assam, India
| | - Sami A. Al-Hussain
- Department of Chemistry, Imam Mohammad Ibn Saud Islamic University (IMSIU), Riyadh, Saudi Arabia
| | - Magdi E. A. Zaki
- Department of Chemistry, Imam Mohammad Ibn Saud Islamic University (IMSIU), Riyadh, Saudi Arabia
| |
Collapse
|
11
|
de Jesus Ramires M, Hummel K, Hatfaludi T, Hess M, Bilic I. Host-specific targets of Histomonas meleagridis antigens revealed by immunoprecipitation. Sci Rep 2025; 15:5800. [PMID: 39962091 PMCID: PMC11832935 DOI: 10.1038/s41598-025-88855-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Accepted: 01/31/2025] [Indexed: 02/20/2025] Open
Abstract
Histomonas meleagridis, a protozoan parasite responsible for histomonosis (syn. Blackhead disease, histomoniasis), presents an increasing challenge for poultry health, particularly with the ban of licensed prophylactic and treatment options. Recent studies have explored H. meleagridis proteome, exoproteome, and surfaceome, linking molecular data to virulence and in vitro attenuation. Nevertheless, proteins involved in interactions with hosts remain largely unknown. In this study, we conducted immunoproteome analyses to identify key antigens involved in the humoral immune response of the parasite's main hosts, turkeys and chickens. Immunogenic proteins were isolated via immunoprecipitation using sera from chickens and turkeys that were vaccinated with a single attenuated strain and challenged with virulent strains of the protozoan, respectively. Mass spectrometry identified 155 putative H. meleagridis immunogenic proteins, of which 43 were recognized by sera from both hosts. In silico antigenicity screening (VaxElan) identified 33 pan-reactive antigens, with VaxiDL further highlighting 10 as potential vaccine candidates. Comparative analysis revealed host-specific immune responses, with 16 differential immunogenic proteins in chickens (6 specific to virulent and 10 to attenuated preparations) and 19 unique proteins in turkeys, all associated with virulent strains. These results enhance our understanding of H. meleagridis immunogenic protein dynamics and host-pathogen specificities, supporting the development of improved diagnostic tools and potential protective measures against the infection.
Collapse
Affiliation(s)
- Marcelo de Jesus Ramires
- Clinic for Poultry and Fish Medicine, Department for Farm Animals and Food System Science, University of Veterinary Medicine Vienna, Veterinärplatz 1, Vienna, A-1210, Austria
| | - Karin Hummel
- VetCore Facility for Research, University of Veterinary Medicine Vienna, Veterinärplatz 1, Vienna, A-1210, Austria
| | - Tamas Hatfaludi
- Clinic for Poultry and Fish Medicine, Department for Farm Animals and Food System Science, University of Veterinary Medicine Vienna, Veterinärplatz 1, Vienna, A-1210, Austria
| | - Michael Hess
- Clinic for Poultry and Fish Medicine, Department for Farm Animals and Food System Science, University of Veterinary Medicine Vienna, Veterinärplatz 1, Vienna, A-1210, Austria
| | - Ivana Bilic
- Clinic for Poultry and Fish Medicine, Department for Farm Animals and Food System Science, University of Veterinary Medicine Vienna, Veterinärplatz 1, Vienna, A-1210, Austria.
| |
Collapse
|
12
|
Mishra V, Singh A, Korzinkin M, Cheng X, Wing C, Sarkisova V, Koppayi AL, Pogorelskaya A, Glushchenko O, Sundaresan M, Thodima V, Carter J, Ito K, Scherle P, Trzcinska A, Ozerov I, Vokes EE, Cole G, Pun FW, Shen L, Miao Y, Pearson AT, Lingen MW, Ruggeri B, Rosenberg AJ, Zhavoronkov A, Agrawal N, Izumchenko E. PRMT5 inhibition has a potent anti-tumor activity against adenoid cystic carcinoma of salivary glands. J Exp Clin Cancer Res 2025; 44:11. [PMID: 39794830 PMCID: PMC11724466 DOI: 10.1186/s13046-024-03270-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2024] [Accepted: 12/30/2024] [Indexed: 01/13/2025] Open
Abstract
BACKGROUND Adenoid cystic carcinoma (ACC) is a rare glandular malignancy, commonly originating in salivary glands of the head and neck. Given its protracted growth, ACC is usually diagnosed in advanced stage. Treatment of ACC is limited to surgery and/or adjuvant radiotherapy, which often fails to prevent disease recurrence, and no FDA-approved targeted therapies are currently available. As such, identification of new therapeutic targets specific to ACC is crucial for improved patients' outcomes. METHODS After thoroughly evaluating the gene expression and signaling patterns characterizing ACC, we applied PandaOmics (an AI-driven software platform for novel therapeutic target discovery) on the unique transcriptomic dataset of 87 primary ACCs. Identifying protein arginine methyl transferase 5 (PRMT5) as a putative candidate with the top-scored druggability, we next determined the applicability of PRMT5 inhibitors (PRT543 and PRT811) using ACC cell lines, organoids, and patient derived xenograft (PDX) models. Molecular changes associated with response to PRMT5 inhibition and anti-proliferative effect of the combination therapy with lenvatinib was then analyzed. RESULTS Using a comprehensive AI-powered engine for target identification, PRMT5 was predicted among potential therapeutic target candidates for ACC. Here we show that monotherapy with selective PRMT5 inhibitors induced a potent anti-tumor activity across several cellular and animal models of ACC, which was paralleled by downregulation of genes associated with ACC tumorigenesis, including MYB and MYC (the recognized drivers of ACC progression). Furthermore, as a subset of genes targeted by lenvatinib is upregulated in ACC, we demonstrate that addition of lenvatinib enhanced the growth inhibitory effect of PRMT5 blockade in vitro, suggesting a potential clinical benefit for patients expressing lenvatinib favorable molecular profile. CONCLUSION Taken together, our study underscores the role of PRMT5 in ACC oncogenesis and provides a strong rationale for the clinical development of PRMT5 inhibitors as a targeted monotherapy or combination therapy for treatment of patients with this rare disease, based on the analysis of their underlying molecular profile.
Collapse
Affiliation(s)
- Vasudha Mishra
- Department of Medicine, Section of Hematology and Oncology, University of Chicago, Chicago, IL, USA
| | - Alka Singh
- Department of Medicine, Section of Hematology and Oncology, University of Chicago, Chicago, IL, USA
| | | | - Xiangying Cheng
- Department of Medicine, Section of Hematology and Oncology, University of Chicago, Chicago, IL, USA
| | - Claudia Wing
- Department of Medicine, Section of Hematology and Oncology, University of Chicago, Chicago, IL, USA
| | | | - Ashwin L Koppayi
- Department of Medicine, Section of Hematology and Oncology, Northwestern University, Chicago, IL, USA
| | | | | | - Manu Sundaresan
- Department of Medicine, Section of Hematology and Oncology, University of Chicago, Chicago, IL, USA
| | | | | | - Koichi Ito
- Prelude Therapeutics, Wilmington, DE, USA
| | | | - Anna Trzcinska
- Department of Pathology, University of Chicago, Chicago, IL, USA
| | | | - Everett E Vokes
- Department of Medicine, Section of Hematology and Oncology, University of Chicago, Chicago, IL, USA
| | - Grayson Cole
- Department of Pathology, University of Chicago, Chicago, IL, USA
| | | | - Le Shen
- Department of Surgery, University of Chicago, Chicago, IL, USA
| | - Yuxuan Miao
- Ben May Department for Cancer Research, University of Chicago, Chicago, IL, USA
| | - Alexander T Pearson
- Department of Medicine, Section of Hematology and Oncology, University of Chicago, Chicago, IL, USA
| | - Mark W Lingen
- Department of Pathology, University of Chicago, Chicago, IL, USA
| | | | - Ari J Rosenberg
- Department of Medicine, Section of Hematology and Oncology, University of Chicago, Chicago, IL, USA
| | | | - Nishant Agrawal
- Department of Surgery, University of Chicago, Chicago, IL, USA.
| | - Evgeny Izumchenko
- Department of Medicine, Section of Hematology and Oncology, University of Chicago, Chicago, IL, USA.
| |
Collapse
|
13
|
Zhang ZY, Fan YE, Huang CB, Du MZ. Human essential gene identification based on feature fusion and feature screening. IET Syst Biol 2024; 18:227-237. [PMID: 39578676 DOI: 10.1049/syb2.12105] [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: 08/24/2024] [Revised: 09/11/2024] [Accepted: 10/25/2024] [Indexed: 11/24/2024] Open
Abstract
Essential genes are necessary to sustain the life of a species under adequate nutritional conditions. These genes have attracted significant attention for their potential as drug targets, especially in developing broad-spectrum antibacterial drugs. However, studying essential genes remains challenging due to their variability in specific environmental conditions. In this study, the authors aim to develop a powerful prediction model for identifying essential genes in humans. The authors first obtained the essential gene data from human cancer cell lines and characterised gene sequences using 7 feature encoding methods such as Kmer, the Composition of K-spaced Nucleic Acid Pairs, and Z-curve. Subsequently, feature fusion and feature optimisation strategies were employed to select the impactful features. Finally, machine learning algorithms were applied to construct the prediction models and evaluate their performance. The single-feature-based model achieved the highest area under the Receiver Operating Characteristic curve (AUC) of 0.830. After fusing and filtering these features, the classical machine learning models achieved the highest AUC at 0.823 while the deep learning model reached 0.860. Results obtained by the authors show that compared to using individual features, feature fusion and feature optimisation strategies significantly improved model performance. Moreover, the study provided an advantageous method for essential gene identification compared to other methods.
Collapse
Affiliation(s)
- Zhao-Yue Zhang
- School of Healthcare Technology, Chengdu Neusoft University, Chengdu, China
- School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Yue-Er Fan
- School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, China
| | - Cheng-Bing Huang
- School of Computer Science and Technology, ABa Teachers University, Chengdu, China
| | - Meng-Ze Du
- School of Healthcare Technology, Chengdu Neusoft University, Chengdu, China
| |
Collapse
|
14
|
Nguyen E, Poli M, Durrant MG, Kang B, Katrekar D, Li DB, Bartie LJ, Thomas AW, King SH, Brixi G, Sullivan J, Ng MY, Lewis A, Lou A, Ermon S, Baccus SA, Hernandez-Boussard T, Ré C, Hsu PD, Hie BL. Sequence modeling and design from molecular to genome scale with Evo. Science 2024; 386:eado9336. [PMID: 39541441 PMCID: PMC12057570 DOI: 10.1126/science.ado9336] [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: 02/27/2024] [Accepted: 09/09/2024] [Indexed: 11/16/2024]
Abstract
The genome is a sequence that encodes the DNA, RNA, and proteins that orchestrate an organism's function. We present Evo, a long-context genomic foundation model with a frontier architecture trained on millions of prokaryotic and phage genomes, and report scaling laws on DNA to complement observations in language and vision. Evo generalizes across DNA, RNA, and proteins, enabling zero-shot function prediction competitive with domain-specific language models and the generation of functional CRISPR-Cas and transposon systems, representing the first examples of protein-RNA and protein-DNA codesign with a language model. Evo also learns how small mutations affect whole-organism fitness and generates megabase-scale sequences with plausible genomic architecture. These prediction and generation capabilities span molecular to genomic scales of complexity, advancing our understanding and control of biology.
Collapse
Affiliation(s)
- Eric Nguyen
- Arc Institute, Palo Alto, CA, USA
- Department of Bioengineering, Stanford University, Stanford, CA, USA
| | - Michael Poli
- Department of Computer Science, Stanford University, Stanford, CA, USA
- TogetherAI, San Francisco, CA, USA
| | | | - Brian Kang
- Arc Institute, Palo Alto, CA, USA
- Department of Bioengineering, Stanford University, Stanford, CA, USA
| | | | - David B. Li
- Arc Institute, Palo Alto, CA, USA
- Department of Bioengineering, Stanford University, Stanford, CA, USA
| | | | - Armin W. Thomas
- Stanford Data Science, Stanford University, Stanford, CA, USA
| | - Samuel H. King
- Arc Institute, Palo Alto, CA, USA
- Department of Bioengineering, Stanford University, Stanford, CA, USA
| | - Garyk Brixi
- Arc Institute, Palo Alto, CA, USA
- Department of Genetics, Stanford University, Stanford, CA, USA
| | | | - Madelena Y. Ng
- Stanford Center for Biomedical Informatics Research, Stanford, CA, USA
| | - Ashley Lewis
- Department of Biomedical Data Science, Stanford University, Stanford, CA, USA
| | - Aaron Lou
- Department of Computer Science, Stanford University, Stanford, CA, USA
| | - Stefano Ermon
- Department of Computer Science, Stanford University, Stanford, CA, USA
- CZ Biohub, San Francisco, CA, USA
| | | | | | - Christopher Ré
- Department of Computer Science, Stanford University, Stanford, CA, USA
| | - Patrick D. Hsu
- Arc Institute, Palo Alto, CA, USA
- Department of Bioengineering and Center for Computational Biology, University of California, Berkeley, Berkeley, CA, USA
| | - Brian L. Hie
- Arc Institute, Palo Alto, CA, USA
- Stanford Data Science, Stanford University, Stanford, CA, USA
- Department of Chemical Engineering, Stanford University, Stanford, CA, USA
| |
Collapse
|
15
|
Ahmad W, Rahman Z, Khan H, Nawab J, Rahman H, Siddiqui MF, Saeed W. Computational proteomics analysis of Taphrina deformans for the identification of antifungal drug targets and validation with commercial fungicides. FRONTIERS IN PLANT SCIENCE 2024; 15:1429890. [PMID: 39574456 PMCID: PMC11578757 DOI: 10.3389/fpls.2024.1429890] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Accepted: 10/10/2024] [Indexed: 11/24/2024]
Abstract
Taphrina deformans is a plant-pathogenic fungus and a responsible agent for causing peach leaf curl disease. Taphrina deformans affects peach fruit production and contributes to global economic losses. Commercial fungicides may provide temporary relief; however, their overuse resulted in adverse environmental consequences as well as led to drug-resistant strains of T. deformans. Therefore, the discovery of novel drug targets for the future synthesis of antifungal drugs against Taphrina deformans is needed. Here we studied Taphrina deformans by computational proteomics approaches. The whole genome and proteome of T. deformans were subjected to subtractive proteomics, high-throughput virtual screening, and molecular dynamic simulations. We employed subtractive proteomics analysis of 4,659 proteins extracted from UniProtKB database; after filtering out homologous and non-essential proteins, we identified 189 essential ones, including nine that participated in the crucial metabolic pathways of the pathogen. These proteins were categorized as nuclear (n = 116), cytoplasmic (n = 37), and membrane (n = 36). Of those essential proteins, glutamate-cysteine ligase (GCL) emerged as one promising target due to its essential function for glutathione biosynthesis process which facilitates T. deformans survival and pathogenicity. To validate GCL as an antifungal target, virtual screening and molecular docking studies with various commercial fungicides were carried out to better characterize GCL as a drug target. The data showed strong binding affinities for polyoxin D, fluoxastrobin, trifloxystrobin, and azoxystrobin within the active site of GCL. Polyoxin D showed a strong affinity when the measured docking score was at -7.34 kcal/mol, while molecular dynamics simulations confirmed stable interactions (three hydrogen bonds, two hydrophobic bonds, and one salt bridge interaction), supporting our findings that GCL represents an excellent target for antifungal drug development efforts. The results showed that GCL, as an innovative target for future fungicide designs to combat T. deformans infections, provides an avenue toward creating more effective peach leaf curl disease treatments while mitigating environmental harm caused by its current use.
Collapse
Affiliation(s)
- Waqar Ahmad
- Department of Microbiology, Abdul Wali Khan University Mardan, Mardan, Khyber Pakhtunkhwa, Pakistan
| | - Ziaur Rahman
- Department of Microbiology, Abdul Wali Khan University Mardan, Mardan, Khyber Pakhtunkhwa, Pakistan
| | - Haji Khan
- Centre of Biotechnology and Microbiology, University of Swat, Swat, Khyber Pakhtunkhwa, Pakistan
| | - Javed Nawab
- Department of Environmental Sciences, Kohat University of Science and Technology, Kohat, Khyber Pakhtunkhwa, Pakistan
| | - Hazir Rahman
- Department of Microbiology, Abdul Wali Khan University Mardan, Mardan, Khyber Pakhtunkhwa, Pakistan
| | | | - Wajeeha Saeed
- Department of Biology, University of Haripur, Haripur, Khyber Pakhtunkhwa, Pakistan
| |
Collapse
|
16
|
Li Y, Farhan MHR, Yang X, Guo Y, Sui Y, Chu J, Huang L, Cheng G. A review on the development of bacterial multi-epitope recombinant protein vaccines via reverse vaccinology. Int J Biol Macromol 2024; 282:136827. [PMID: 39476887 DOI: 10.1016/j.ijbiomac.2024.136827] [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: 07/05/2024] [Revised: 10/04/2024] [Accepted: 10/21/2024] [Indexed: 11/10/2024]
Abstract
Bacterial vaccines play a crucial role in combating bacterial infectious diseases. Apart from the prevention of disease, bacterial vaccines also help to reduce the mortality rates in infected populations. Advancements in vaccine development technologies have addressed the constraints of traditional vaccine design, providing novel approaches for the development of next-generation vaccines. Advancements in reverse vaccinology, bioinformatics, and comparative proteomics have opened horizons in vaccine development. Specifically, the use of protein structural data in crafting multi-epitope vaccines (MEVs) to target pathogens has become an important research focus in vaccinology. In this review, we focused on describing the methodologies and tools for epitope vaccine development, along with recent progress in this field. Moreover, this article also discusses the challenges in epitope vaccine development, providing insights for the future development of bacterial multi-epitope genetically engineered vaccines.
Collapse
Affiliation(s)
- Yuxin Li
- National Reference Laboratory of Veterinary Drug Residues (HZAU), Huazhong Agricultural University, Wuhan, Hubei 430070, PR China
| | - Muhammad Haris Raza Farhan
- National Reference Laboratory of Veterinary Drug Residues (HZAU), Huazhong Agricultural University, Wuhan, Hubei 430070, PR China
| | - Xiaohan Yang
- National Reference Laboratory of Veterinary Drug Residues (HZAU), Huazhong Agricultural University, Wuhan, Hubei 430070, PR China
| | - Ying Guo
- National Reference Laboratory of Veterinary Drug Residues (HZAU), Huazhong Agricultural University, Wuhan, Hubei 430070, PR China
| | - Yuxin Sui
- National Reference Laboratory of Veterinary Drug Residues (HZAU), Huazhong Agricultural University, Wuhan, Hubei 430070, PR China
| | - Jinhua Chu
- National Reference Laboratory of Veterinary Drug Residues (HZAU), Huazhong Agricultural University, Wuhan, Hubei 430070, PR China
| | - Lingli Huang
- National Reference Laboratory of Veterinary Drug Residues (HZAU), Huazhong Agricultural University, Wuhan, Hubei 430070, PR China; MOA Laboratory of Risk Assessment for Quality and Safety of Livestock and Poultry Products, Huazhong Agricultural University, Wuhan, Hubei 430070, PR China
| | - Guyue Cheng
- National Reference Laboratory of Veterinary Drug Residues (HZAU), Huazhong Agricultural University, Wuhan, Hubei 430070, PR China; MOA Laboratory of Risk Assessment for Quality and Safety of Livestock and Poultry Products, Huazhong Agricultural University, Wuhan, Hubei 430070, PR China.
| |
Collapse
|
17
|
Hasan A, Ibrahim M, Alonazi WB, Yu R, Li B. Pangenome analysis of five representative Tropheryma whipplei strains following multiepitope-based vaccine design via immunoinformatic approaches. Mol Genet Genomics 2024; 299:101. [PMID: 39460811 DOI: 10.1007/s00438-024-02189-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Accepted: 09/29/2024] [Indexed: 10/28/2024]
Abstract
Whipple disease caused by Tropheryma whipplei a gram-positive bacterium is a systemic disorder that impacts not only the gastrointestinal tract but also the vascular system, joints, central nervous system, and cardiovascular system. Due to the lack of an approved vaccine, this study aimed to utilize immunoinformatic approaches to design multiepitope -based vaccine by utilizing the proteomes of five representative T. whipplei strains. The genomes initially comprised a total of 4,844 proteins ranging from 956 to 1012 proteins per strain. We collected 829 nonredundant lists of core proteins, that were shared among all the strains. Following subtractive proteomics, one extracellular protein, WP_033800108.1, a WhiB family transcriptional regulator, was selected for the chimeric-based multiepitope vaccine. Five immunodominant epitopes were retrieved from the WhiB family transcriptional regulator protein, indicating MHC-I and MHC-II with a global population coverage of 70.61%. The strong binding affinity, high solubility, nontoxicity, nonallergenic properties and high antigenicity scores make the selected epitopes more appropriate. Integration of the epitopes into a chimeric vaccine was carried out by applying appropriate adjuvant molecules and linkers, leading to the vaccine construct having enhanced immunogenicity and successfully eliciting both innate and adaptive immune responses. Moreover, the abilityof the vaccine to bind TLR4, a core innate immune receptor, was confirmed. Molecular dynamics simulations have also revealed the promising potential stability of the designed vaccine at 400 ns. In summary, we have designed a potential vaccine construct that has the ability not only to induce targeted immunogenicity for one strain but also for global T. whipplei strains. This study proposes a potential universal vaccine, reducing Whipple's disease risk and laying the groundwork for future research on multi-strain pathogens.
Collapse
Affiliation(s)
- Ahmad Hasan
- State Key Laboratory of Rice Biology and Breeding, Ministry of Agriculture Key Laboratory of Molecular Biology of Crop Pathogens and Insects, Zhejiang Key Laboratory of Biology and Ecological Regulation of Crop Pathogens and Insects, Institute of Biotechnology, Zhejiang University, Hangzhou, 310058, China
| | - Muhammad Ibrahim
- State Key Laboratory of Rice Biology and Breeding, Ministry of Agriculture Key Laboratory of Molecular Biology of Crop Pathogens and Insects, Zhejiang Key Laboratory of Biology and Ecological Regulation of Crop Pathogens and Insects, Institute of Biotechnology, Zhejiang University, Hangzhou, 310058, China
| | - Wadi B Alonazi
- Health Administration Department, College of Business Administration, King Saud University, Riyadh, Saudi Arabia
| | - Rongrong Yu
- College of Education, Zhejiang University of Technology, Hangzhou, 310032, China.
| | - Bin Li
- State Key Laboratory of Rice Biology and Breeding, Ministry of Agriculture Key Laboratory of Molecular Biology of Crop Pathogens and Insects, Zhejiang Key Laboratory of Biology and Ecological Regulation of Crop Pathogens and Insects, Institute of Biotechnology, Zhejiang University, Hangzhou, 310058, China.
| |
Collapse
|
18
|
Noori Goodarzi N, Khazani Asforooshani M, Shahbazi B, Rezaie Rahimi N, Badmasti F. Identification of novel drug targets for Helicobacter pylori: structure-based virtual screening of potential inhibitors against DAH7PS protein involved in the shikimate pathway. FRONTIERS IN BIOINFORMATICS 2024; 4:1482338. [PMID: 39493576 PMCID: PMC11527725 DOI: 10.3389/fbinf.2024.1482338] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2024] [Accepted: 10/07/2024] [Indexed: 11/05/2024] Open
Abstract
Background Helicobacter pylori, a bacterium associated with severe gastrointestinal diseases and malignancies, poses a significant challenge because of its increasing antibiotic resistance rates. This study aimed to identify potential drug targets and inhibitors against H. pylori using a structure-based virtual screening (SBVS) approach. Methods Core-proteome analysis of 132 H. pylori genomes was performed using the EDGAR database. Essential genes were identified and human and gut microbiota homolog proteins were excluded. The DAH7PS protein involved in the shikimate pathway was selected for the structure-based virtual screening (SBVS) approach. The tertiary structure of the protein was predicted through homology modeling (based on PDB ID: 5UXM). Molecular docking was performed to identify potential inhibitors of DAH7PS among StreptomeDB compounds using the AutoDock Vina tool. Molecular dynamics (MD) simulations assessed the stability of DAH7PS-ligand complexes. The complexes were further evaluated in terms of their binding affinity, Lipinski's Rule of Five, and ADMET properties. Results A total of 54 novel drug targets with desirable properties were identified. DAH7PS was selected for further investigation, and virtual screening of StreptomeDB compounds yielded 36 high-affinity binding of the ligands. Two small molecules, 6,8-Dihydroxyisocoumarin-3-carboxylic acid and Epicatechin, also showed favorable RO5 and ADMET properties. MD simulations confirmed the stability and reliability of DAH7PS-ligand complexes, indicating their potential as inhibitors. Conclusion This study identified 54 novel drug targets against H. pylori. The DAH7PS protein as a promising drug target was evaluated using a computer-aided drug design. 6,8-Dihydroxyisocoumarin-3-carboxylic acid and Epicatechin demonstrated desirable properties and stable interactions, highlighting their potential to inhibit DAH7PS as an essential protein. Undoubtedly, more experimental validations are needed to advance these findings into practical therapies for treating drug-resistant H. pylori.
Collapse
Affiliation(s)
- Narjes Noori Goodarzi
- Department of Pathobiology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
- Department of Bacteriology, Pasteur Institute of Iran, Tehran, Iran
| | - Mahshid Khazani Asforooshani
- Department of Bacteriology, Pasteur Institute of Iran, Tehran, Iran
- Department of Microbiology, Faculty of Biological Sciences, Alzahra University, Tehran, Iran
| | - Behzad Shahbazi
- School of Pharmacy, Semnan University of Medical Sciences, Semnan, Iran
| | - Nayereh Rezaie Rahimi
- Department of environmental Health Engineering, School of Public Health, Shiraz University of Medical Sciences, Shiraz, Iran
- Student Research Committee, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Farzad Badmasti
- Department of Bacteriology, Pasteur Institute of Iran, Tehran, Iran
| |
Collapse
|
19
|
Wegner SA, Avalos JL. Mevalonate secretion is not mediated by a singular non-essential transporter in Saccharomyces cerevisiae. BIOTECHNOLOGY NOTES (AMSTERDAM, NETHERLANDS) 2024; 5:140-150. [PMID: 39498316 PMCID: PMC11532745 DOI: 10.1016/j.biotno.2024.10.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 10/12/2024] [Accepted: 10/13/2024] [Indexed: 11/07/2024]
Abstract
Isoprenoids are highly valued targets for microbial chemical production, allowing the creation of fragrances, biofuels, and pharmaceuticals from renewable carbon feedstocks. To increase isoprenoid production, previous efforts have manipulated pyruvate dehydrogenase (PDH) bypass pathway flux to increase cytosolic acetyl-coA; however, this results in mevalonate secretion and does not necessarily translate into higher isoprenoid production. Identification and disruption of the transporter mediating mevalonate secretion would allow us to determine whether increasing PDH bypass activity in the absence of secretion improves conversion of mevalonate into downstream isoprenoids. Attempted identification of the mevalonate transporter was accomplished using a pooled CRISPR library targeting all nonessential transporters and two different screening methods. Using a high throughput screen, based on growth of a mevalonate auxotrophic Escherichia coli strain, it was found that ZRT3 disruption largely abolished accumulation of extracellular mevalonate. However, disruption of ZRT3 was found to lower overall mevalonate pathway activity, rather than prevent secretion, indicating a previously unreported interaction between zinc availability and the mevalonate pathway. In a second screen, significant differences in PDR5/15 and QDR1/2 library representation were found between wild-type and mevalonate secreting Saccharomyces cerevisiae strains. However, no single deletion (or selected pair of double deletions) abolishes mevalonate secretion, indicating that this process appears to be mediated through multiple redundant transporters.
Collapse
Affiliation(s)
- Scott A. Wegner
- Department of Molecular Biology, Princeton University, Princeton, NJ, 08544, USA
| | - José L. Avalos
- Department of Molecular Biology, Princeton University, Princeton, NJ, 08544, USA
- Department of Chemical and Biological Engineering, Princeton University, Princeton, NJ, 08544, USA
- The Andlinger Center for Energy and the Environment, Princeton University, Princeton, NJ, 08544, USA
- High Meadows Environmental Institute, Princeton University, Princeton, NJ, 08544, USA
| |
Collapse
|
20
|
Humphreys IR, Zhang J, Baek M, Wang Y, Krishnakumar A, Pei J, Anishchenko I, Tower CA, Jackson BA, Warrier T, Hung DT, Peterson SB, Mougous JD, Cong Q, Baker D. Protein interactions in human pathogens revealed through deep learning. Nat Microbiol 2024; 9:2642-2652. [PMID: 39294458 PMCID: PMC11445079 DOI: 10.1038/s41564-024-01791-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Accepted: 07/23/2024] [Indexed: 09/20/2024]
Abstract
Identification of bacterial protein-protein interactions and predicting the structures of these complexes could aid in the understanding of pathogenicity mechanisms and developing treatments for infectious diseases. Here we developed RoseTTAFold2-Lite, a rapid deep learning model that leverages residue-residue coevolution and protein structure prediction to systematically identify and structurally characterize protein-protein interactions at the proteome-wide scale. Using this pipeline, we searched through 78 million pairs of proteins across 19 human bacterial pathogens and identified 1,923 confidently predicted complexes involving essential genes and 256 involving virulence factors. Many of these complexes were not previously known; we experimentally tested 12 such predictions, and half of them were validated. The predicted interactions span core metabolic and virulence pathways ranging from post-transcriptional modification to acid neutralization to outer-membrane machinery and should contribute to our understanding of the biology of these important pathogens and the design of drugs to combat them.
Collapse
Affiliation(s)
- Ian R Humphreys
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Jing Zhang
- Eugene McDermott Center for Human Growth and Development, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Department of Biophysics, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Minkyung Baek
- Department of Biological Sciences, Seoul National University, Seoul, South Korea.
| | - Yaxi Wang
- Department of Microbiology, University of Washington, Seattle, WA, USA
| | - Aditya Krishnakumar
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Jimin Pei
- Eugene McDermott Center for Human Growth and Development, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Department of Biophysics, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Ivan Anishchenko
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Catherine A Tower
- Department of Microbiology, University of Washington, Seattle, WA, USA
| | - Blake A Jackson
- Department of Microbiology, University of Washington, Seattle, WA, USA
| | - Thulasi Warrier
- Department of Molecular Biology and Center for Computational and Integrative Biology, Massachusetts General Hospital, Boston, MA, USA
- Department of Genetics, Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Deborah T Hung
- Department of Molecular Biology and Center for Computational and Integrative Biology, Massachusetts General Hospital, Boston, MA, USA
- Department of Genetics, Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - S Brook Peterson
- Department of Microbiology, University of Washington, Seattle, WA, USA
| | - Joseph D Mougous
- Department of Microbiology, University of Washington, Seattle, WA, USA
- Howard Hughes Medical Institute, University of Washington, Seattle, WA, USA
- Microbial Interactions and Microbiome Center, University of Washington, Seattle, WA, USA
| | - Qian Cong
- Eugene McDermott Center for Human Growth and Development, University of Texas Southwestern Medical Center, Dallas, TX, USA.
- Department of Biophysics, University of Texas Southwestern Medical Center, Dallas, TX, USA.
- Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX, USA.
| | - David Baker
- Department of Biochemistry, University of Washington, Seattle, WA, USA.
- Institute for Protein Design, University of Washington, Seattle, WA, USA.
- Howard Hughes Medical Institute, University of Washington, Seattle, WA, USA.
| |
Collapse
|
21
|
Bi GW, Wu ZG, Li Y, Wang JB, Yao ZW, Yang XY, Yu YB. Intestinal flora and inflammatory bowel disease: Causal relationships and predictive models. Heliyon 2024; 10:e38101. [PMID: 39381207 PMCID: PMC11458943 DOI: 10.1016/j.heliyon.2024.e38101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Revised: 09/17/2024] [Accepted: 09/17/2024] [Indexed: 10/10/2024] Open
Abstract
BACKGROUND Inflammatory bowel disease (IBD), including Crohn's disease and ulcerative colitis, is significantly influenced by intestinal flora. Understanding the genetic and microbiotic interplay is crucial for IBD prediction and treatment. METHODS We used Mendelian randomization (MR), transcriptomic analysis, and machine learning techniques, integrating data from the MiBioGen Consortium and various GWAS datasets. SNPs associated with intestinal flora were mapped to genes, with LASSO regression refining gene selection. Differentially expressed genes (DEGs) and immune infiltration patterns were identified through transcriptomic analysis. Six machine learning models were used for predictive modeling. FINDINGS MR analysis identified 25 gut microbiota classifications causally related to IBD. SNP mapping and gene expression analysis highlighted 24 significant genes. Drug target MR and colocalization validated these genes' causal relationships with IBD. Key pathways identified included the PI3K-Akt signaling pathway and epithelial-mesenchymal transition. Immune infiltration analysis revealed distinct patterns between high and low LASSO score groups. Machine learning models demonstrated high predictive value, with soft voting enhancing reliability. INTERPRETATION By integrating MR, transcriptomic analysis, and sophisticated machine learning approaches, this study elucidates the causal relationships between intestinal flora and IBD. The application of machine learning not only enhanced predictive modeling but also offered new insights into IBD pathogenesis, highlighted potential therapeutic targets, and established a robust framework for predicting IBD onset.
Collapse
Affiliation(s)
- Guan-Wei Bi
- First Clinical College, Shandong University, Jinan, Shandong Province, PR China
- Department of Gastroenterology, Qilu Hospital, Shandong University, Jinan, Shandong, PR China
| | - Zhen-Guo Wu
- First Clinical College, Shandong University, Jinan, Shandong Province, PR China
- Department of Gastroenterology, Qilu Hospital, Shandong University, Jinan, Shandong, PR China
| | - Yu Li
- First Clinical College, Shandong University, Jinan, Shandong Province, PR China
- Department of Gastroenterology, Qilu Hospital, Shandong University, Jinan, Shandong, PR China
| | - Jin-Bei Wang
- First Clinical College, Shandong University, Jinan, Shandong Province, PR China
- Department of Gastroenterology, Qilu Hospital, Shandong University, Jinan, Shandong, PR China
| | - Zhi-Wen Yao
- First Clinical College, Shandong University, Jinan, Shandong Province, PR China
- Department of Gastroenterology, Qilu Hospital, Shandong University, Jinan, Shandong, PR China
| | - Xiao-Yun Yang
- Department of Gastroenterology, Qilu Hospital, Shandong University, Jinan, Shandong, PR China
| | - Yan-Bo Yu
- Department of Gastroenterology, Qilu Hospital, Shandong University, Jinan, Shandong, PR China
- Shandong Provincial Clinical Research Center for Digestive Disease, Qilu Hospital, Shandong University, Jinan, Shandong Province, PR China
| |
Collapse
|
22
|
Stephen C, Palmer D, Mishanina TV. Opportunities for Riboswitch Inhibition by Targeting Co-Transcriptional RNA Folding Events. Int J Mol Sci 2024; 25:10495. [PMID: 39408823 PMCID: PMC11476745 DOI: 10.3390/ijms251910495] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Revised: 09/27/2024] [Accepted: 09/27/2024] [Indexed: 10/20/2024] Open
Abstract
Antibiotic resistance is a critical global health concern, causing millions of prolonged bacterial infections every year and straining our healthcare systems. Novel antibiotic strategies are essential to combating this health crisis and bacterial non-coding RNAs are promising targets for new antibiotics. In particular, a class of bacterial non-coding RNAs called riboswitches has attracted significant interest as antibiotic targets. Riboswitches reside in the 5'-untranslated region of an mRNA transcript and tune gene expression levels in cis by binding to a small-molecule ligand. Riboswitches often control expression of essential genes for bacterial survival, making riboswitch inhibitors an exciting prospect for new antibacterials. Synthetic ligand mimics have predominated the search for new riboswitch inhibitors, which are designed based on static structures of a riboswitch's ligand-sensing aptamer domain or identified by screening a small-molecule library. However, many small-molecule inhibitors that bind an isolated riboswitch aptamer domain with high affinity in vitro lack potency in vivo. Importantly, riboswitches fold and respond to the ligand during active transcription in vivo. This co-transcriptional folding is often not considered during inhibitor design, and may explain the discrepancy between a low Kd in vitro and poor inhibition in vivo. In this review, we cover advances in riboswitch co-transcriptional folding and illustrate how intermediate structures can be targeted by antisense oligonucleotides-an exciting new strategy for riboswitch inhibitor design.
Collapse
Affiliation(s)
| | | | - Tatiana V. Mishanina
- Department of Chemistry and Biochemistry, University of California San Diego, La Jolla, CA 92093, USA (D.P.)
| |
Collapse
|
23
|
Asad M, Hassan A, Wang W, Alonazi WB, Khan MS, Ogunyemi SO, Ibrahim M, Bin L. An integrated in silico approach for the identification of novel potential drug target and chimeric vaccine against Neisseria meningitides strain 331401 serogroup X by subtractive genomics and reverse vaccinology. Comput Biol Med 2024; 178:108738. [PMID: 38870724 DOI: 10.1016/j.compbiomed.2024.108738] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Revised: 05/15/2024] [Accepted: 06/08/2024] [Indexed: 06/15/2024]
Abstract
Neisseria meningitidis, commonly known as the meningococcus, leads to substantial illness and death among children and young adults globally, revealing as either epidemic or sporadic meningitis and/or septicemia. In this study, we have designed a novel peptide-based chimeric vaccine candidate against the N. meningitidis strain 331,401 serogroup X. Through rigorous analysis of subtractive genomics, two essential cytoplasmic proteins, namely UPI000012E8E0(UDP-3-O-acyl-GlcNAc deacetylase) and UPI0000ECF4A9(UDP-N-acetylglucosamine acyltransferase) emerged as potential drug targets. Additionally, using reverse vaccinology, the outer membrane protein UPI0001F4D537 (Membrane fusion protein MtrC) identified by subcellular localization and recognized for its known indispensable role in bacterial survival was identified as a novel chimeric vaccine target. Following a careful comparison of MHC-I, MHC-II, T-cell, and B-cell epitopes, three epitopes derived from UPI0001F4D537 were linked with three types of linkers-GGGS, EAAAK, and the essential PADRE-for vaccine construction. This resulted in eight distinct vaccine models (V1-V8). Among them V1 model was selected as the final vaccine construct. It exhibits exceptional immunogenicity, safety, and enhanced antigenicity, with 97.7 % of its residues in the Ramachandran plot's most favored region. Subsequently, the vaccine structure was docked with the TLR4/MD2 complex and six different HLA allele receptors using the HADDOCK server. The docking resulted in the lowest HADDOCK score of 39.3 ± 9.0 for TLR/MD2. Immune stimulation showed a strong immune response, including antibodies creation and the activation of B-cells, T Cytotoxic cells, T Helper cells, Natural Killer cells, and interleukins. Furthermore, the vaccine construct was successfully expressed in the Escherichia coli system by reverse transcription, optimization, and ligation in the pET-28a (+) vector for the expression study. The current study proposes V1 construct has the potential to elicit both cellular and humoral responses, crucial for the developing an epitope-based vaccine against N. meningitidis strain 331,401 serogroup X.
Collapse
Affiliation(s)
- Muhammad Asad
- Department of Biosciences, COMSATS University Islamabad, Sahiwal Campus, Pakistan
| | - Ahmad Hassan
- Department of Biosciences, COMSATS University Islamabad, Sahiwal Campus, Pakistan
| | - Weiyu Wang
- State Key Laboratory of Rice Biology and Breeding, Key Laboratory of Molecular Biology of Crop Pathogens and Insects, Institute of Biotechnology, Zhejiang University, Hangzhou, 310058, China
| | - Wadi B Alonazi
- Health Administration Department, College of Business Administration, King Saud University, Riyadh, Saudi Arabia
| | | | - Solabomi Olaitan Ogunyemi
- State Key Laboratory of Rice Biology and Breeding, Key Laboratory of Molecular Biology of Crop Pathogens and Insects, Institute of Biotechnology, Zhejiang University, Hangzhou, 310058, China
| | - Muhammad Ibrahim
- Department of Biosciences, COMSATS University Islamabad, Sahiwal Campus, Pakistan.
| | - Li Bin
- State Key Laboratory of Rice Biology and Breeding, Key Laboratory of Molecular Biology of Crop Pathogens and Insects, Institute of Biotechnology, Zhejiang University, Hangzhou, 310058, China
| |
Collapse
|
24
|
Ahmad V, Jamal A, Khan MI, Alzahrani FA, Albiheyri R, Jamal QMS. Cefoperazone targets D-alanyl-D-alanine carboxypeptidase (DAC) to control Morganella morganii-mediated infection: a subtractive genomic and molecular dynamics approach. J Biomol Struct Dyn 2024; 42:6799-6812. [PMID: 37480259 DOI: 10.1080/07391102.2023.2238088] [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: 03/21/2023] [Accepted: 07/08/2023] [Indexed: 07/23/2023]
Abstract
Morganella morganii is a Gram-negative bacterial pathogen that causes bacteremia, urinary tract infections, intra-abdominal infections, chorioamnionitis, neonatal sepsis, and newborn meningitis. To control this bacterial pathogen a total of 3565 putative proteins targets in Morganella morganii were screened using comparative subtractive analysis of biochemical pathways annotated by the KEGG that did not share any similarities with human proteins. One of the targets, D-alanyl-D-alanine carboxypeptidase DacB [Morganella] was observed to be implicated in the majority of cell wall synthesis pathways, leading to its selection as a novel pharmacological target. The drug that interacted optimally with the identified target was observed to be Cefoperazone (DB01329) with the estimated free energy of binding -8.9 Kcal/mol. During molecular dynamics simulations; it was observed that DB01328-2exb and DB01329-2exb complexes showed similar values as the control FMX-2exb complex near 0.2 nm with better stability. Furthermore, MMPBSA total free energy calculation showed better binding energy than the control complex for DB01329-2exb interaction i.e. -31.50 (±0.93) kcal/mol. Our presented research suggested that D-alanyl-D-alanine carboxypeptidase DacB could be a therapeutic target and cefoperazone could be a promising ligand to inhibit the D-alanyl-D-alanine carboxypeptidase DacB protein of Morganella morganii. To identify prospective therapeutic and vaccine targets in Morganella morganii, this is the first computational and subtractive genomics investigation of various metabolic pathways exploring other therapeutic targets of Morganella morganii. In vitro/in vivo experimental validation of the identified target D-alanyl-D-alanine carboxypeptidase and the design of its inhibitors is suggested to figure out the best dose, the drug's effectiveness, and its toxicity.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Varish Ahmad
- Health Information Technology Department, The Applied College, King Abdulaziz University, Jeddah, Saudi Arabia
- Centre for Artificial Intelligence in Precision Medicines, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Alam Jamal
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Mohammad Imran Khan
- Centre for Artificial Intelligence in Precision Medicines, King Abdulaziz University, Jeddah, Saudi Arabia
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Faisal A Alzahrani
- Department of Biochemistry, Faculty of Science, Embryonic Stem Cell Unit, King Fahad Center for Medical Research, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Raed Albiheyri
- Department of Biological Sciences, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia
- Centre of Excellence in Bionanoscience Research, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Qazi Mohammad Sajid Jamal
- Department of Health Informatics, College of Public Health and Health Informatics, Qassim University, Al Bukayriyah, Saudi Arabia
| |
Collapse
|
25
|
Hasnat S, Hoque MN, Mahbub MM, Sakif TI, Shahinuzzaman A, Islam T. Pantothenate kinase: A promising therapeutic target against pathogenic Clostridium species. Heliyon 2024; 10:e34544. [PMID: 39130480 PMCID: PMC11315101 DOI: 10.1016/j.heliyon.2024.e34544] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Revised: 07/08/2024] [Accepted: 07/11/2024] [Indexed: 08/13/2024] Open
Abstract
Current treatment of clostridial infections includes broad-spectrum antibiotics and antitoxins, yet antitoxins are ineffective against all Clostridiumspecies. Moreover, rising antimicrobial resistance (AMR) threatens treatment effectiveness and public health. This study therefore aimed to discover a common drug target for four pathogenic clostridial species, Clostridium botulinum, C. difficile, C. tetani, and C. perfringens through an in-silico core genomic approach. Using four reference genomes of C. botulinum, C. difficile, C. tetani, and C. perfringens, we identified 1484 core genomic proteins (371/genome) and screened them for potential drug targets. Through a subtractive approach, four core proteins were finally identified as drug targets, represented by type III pantothenate kinase (CoaX) and, selected for further analyses. Interestingly, the CoaX is involved in the phosphorylation of pantothenate (vitamin B5), which is a critical precursor for coenzyme A (CoA) biosynthesis. Investigation of druggability analysis on the identified drug target reinforces CoaX as a promising novel drug target for the selected Clostridium species. During the molecular screening of 1201 compounds, a known agonist drug compound (Vibegron) showed strong inhibitory activity against targeted clostridial CoaX. Additionally, we identified tazobactam, a beta-lactamase inhibitor, as effective against the newly proposed target, CoaX. Therefore, identifying CoaX as a single drug target effective against all four clostridial pathogens presents a valuable opportunity to develop a cost-effective treatment for multispecies clostridial infections.
Collapse
Affiliation(s)
- Soharth Hasnat
- Institute of Biotechnology and Genetic Engineering (IBGE), Bangabandhu Sheikh Mujibur Rahman Agricultural University (BSMRAU), Gazipur, 1706, Bangladesh
- Molecular Biology and Bioinformatics Laboratory (MBBL), Department of Gynecology, Obstetrics and Reproductive Health, BSMRAU, Gazipur, 1706, Bangladesh
- Department of Genetic Engineering and Biotechnology, East West University, Dhaka, 1212, Bangladesh
| | - M. Nazmul Hoque
- Molecular Biology and Bioinformatics Laboratory (MBBL), Department of Gynecology, Obstetrics and Reproductive Health, BSMRAU, Gazipur, 1706, Bangladesh
| | - M Murshida Mahbub
- Department of Genetic Engineering and Biotechnology, East West University, Dhaka, 1212, Bangladesh
| | - Tahsin Islam Sakif
- Lane Department of Computer Science and Electrical Engineering, West Virginia University, Morgantown, WV, WV 26506, USA
| | - A.D.A. Shahinuzzaman
- Pharmaceutical Sciences Research Division, Bangladesh Council of Scientific and Industrial Research (BCSIR), Dhaka, 1205, Bangladesh
| | - Tofazzal Islam
- Institute of Biotechnology and Genetic Engineering (IBGE), Bangabandhu Sheikh Mujibur Rahman Agricultural University (BSMRAU), Gazipur, 1706, Bangladesh
| |
Collapse
|
26
|
Fan C, Basharat Z, Mah K, Wei CR. Computational approach for drug discovery against Gardnerella vaginalis in quest for safer and effective treatments for bacterial vaginosis. Sci Rep 2024; 14:17437. [PMID: 39075099 PMCID: PMC11286753 DOI: 10.1038/s41598-024-68443-2] [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: 05/20/2024] [Accepted: 07/23/2024] [Indexed: 07/31/2024] Open
Abstract
Bacterial vaginosis (BV), primarily attributed to Gardnerella vaginalis, poses significant challenges due to antibiotic resistance and suboptimal treatment outcomes. This study presents an integrated approach to identify potential drug targets and screen compounds against this bacterium by leveraging a computational methodology. Subtractive proteomics of the reference strain ASM286196v1/UMB0386 (assembly accession: GCA_002861965.1) facilitated the prioritization of proteins with essential bacterial functions and pathways as potential drug targets. We selected 3-deoxy-7-phosphoheptulonate synthase (aroG gene product; also known as DAHP synthase) for downstream analysis. Molecular docking was employed in PyRx (AutoDock Vina) to predict binding affinities between aroG inhibitors from the ZINC database and 3-deoxy-7-phosphoheptulonate synthase. Molecular dynamics simulations of 100 ns, using GROMACS, validated the stability of drug-target interactions. Additionally, ADMET profiling aided in the selection of compounds with favorable pharmacokinetic properties and safety profile for human hosts. PBPK profiling showed that ZINC98088375 had the highest bioavailability and efficient systemic circulation. Conversely, ZINC5113880 demonstrated the lowest absorption rate (39.661%). Moreover, cirrhosis, steatosis, and renal impairment appeared to influence blood concentration of the drug, impacting bioavailability. The integrative -omics approach utilized in this study underscores the potential of computer-aided drug design and offers a rational strategy for targeted inhibitor discovery against G. vaginalis. The strategy is an attempt to address the limitations of current BV treatments, including antibiotic resistance, and pave way for the development of safer and more effective therapeutics.
Collapse
Affiliation(s)
- Chenyue Fan
- Department of Research and Development, Shing Huei Group, Taipei, 10617, Taiwan
- College of Pharmacy, University of Arizona, Tuscon, AZ, 85721, USA
| | | | - Karmen Mah
- Department of Research and Development, Shing Huei Group, Taipei, 10617, Taiwan
| | - Calvin R Wei
- Department of Research and Development, Shing Huei Group, Taipei, 10617, Taiwan.
| |
Collapse
|
27
|
Kurmi A, Sen P, Dash M, Ray SK, Satapathy SS. Differentially used codons among essential genes in bacteria identified by machine learning-based analysis. Mol Genet Genomics 2024; 299:72. [PMID: 39060647 DOI: 10.1007/s00438-024-02163-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2024] [Accepted: 07/10/2024] [Indexed: 07/28/2024]
Abstract
Codon usage bias (CUB), the uneven usage of synonymous codons encoding the same amino acid, differs among genes within and across bacteria genomes. CUB is known to be influenced by gene expression and accordingly, CUB differs between the high-expression and low-expression genes in several bacteria. In this article, we have extended codon usage study considering gene essentiality as a feature. Using machine learning (ML) based approaches, we have analysed Relative Synonymous Codon Usage (RSCU) values between essential and non-essential genes in Escherichia coli and thirty-four other bacterial genomes whose gene essentiality features were available in public databases. We observed significant differences in codon usage patterns between essential and non-essential genes for majority of the bacterial genomes and accordingly, ML based classifiers achieved high area under curve (AUC) scores, with a minimum score of 70.0 across twenty-eight organisms. Further, importance of the codons towards classifying genes found to differ among the codons in each genome. Arg codon CGT and Gly codon GGT were observed to be the most preferred codons among essential genes in Escherichia coli. Interestingly, some of the codons like CGT, ATA, GGT and GGG observed to be contributing consistently towards classifying essential genes across thirty-five bacteria genomes studied. In other hand, codons TGY and CAY encoding amino acids Cys and His respectively were among the least contributing codons towards classification among all these bacteria. This study demonstrates the gene essentiality based differences in synonymous codon usage in bacteria genomes and presents a common codon usage pattern across bacteria.
Collapse
Affiliation(s)
- Annushree Kurmi
- Department of Computer Science and Engineering, Tezpur University, Napaam, Assam, 784028, India
- Department of Computer Science and Engineering, The Assam Kaziranga University, Jorhat, Assam, 785006, India
| | - Piyali Sen
- Department of Computer Science and Engineering, Tezpur University, Napaam, Assam, 784028, India
| | - Madhusmita Dash
- Department of Electronics and Communication Engineering, NIT, Jote, Arunachal Pradesh, 791113, India
| | - Suvendra Kumar Ray
- Department of Molecular Biology and Biotechnology, Tezpur University, Napaam, Assam, 784028, India
| | | |
Collapse
|
28
|
Urra G, Valdés-Muñoz E, Suardiaz R, Hernández-Rodríguez EW, Palma JM, Ríos-Rozas SE, Flores-Morales CA, Alegría-Arcos M, Yáñez O, Morales-Quintana L, D’Afonseca V, Bustos D. From Proteome to Potential Drugs: Integration of Subtractive Proteomics and Ensemble Docking for Drug Repurposing against Pseudomonas aeruginosa RND Superfamily Proteins. Int J Mol Sci 2024; 25:8027. [PMID: 39125594 PMCID: PMC11312079 DOI: 10.3390/ijms25158027] [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: 05/27/2024] [Revised: 07/08/2024] [Accepted: 07/17/2024] [Indexed: 08/12/2024] Open
Abstract
Pseudomonas aeruginosa (P. aeruginosa) poses a significant threat as a nosocomial pathogen due to its robust resistance mechanisms and virulence factors. This study integrates subtractive proteomics and ensemble docking to identify and characterize essential proteins in P. aeruginosa, aiming to discover therapeutic targets and repurpose commercial existing drugs. Using subtractive proteomics, we refined the dataset to discard redundant proteins and minimize potential cross-interactions with human proteins and the microbiome proteins. We identified 12 key proteins, including a histidine kinase and members of the RND efflux pump family, known for their roles in antibiotic resistance, virulence, and antigenicity. Predictive modeling of the three-dimensional structures of these RND proteins and subsequent molecular ensemble-docking simulations led to the identification of MK-3207, R-428, and Suramin as promising inhibitor candidates. These compounds demonstrated high binding affinities and effective inhibition across multiple metrics. Further refinement using non-covalent interaction index methods provided deeper insights into the electronic effects in protein-ligand interactions, with Suramin exhibiting superior binding energies, suggesting its broad-spectrum inhibitory potential. Our findings confirm the critical role of RND efflux pumps in antibiotic resistance and suggest that MK-3207, R-428, and Suramin could be effectively repurposed to target these proteins. This approach highlights the potential of drug repurposing as a viable strategy to combat P. aeruginosa infections.
Collapse
Affiliation(s)
- Gabriela Urra
- Laboratorio de Bioinformática y Química Computacional, Departamento de Medicina Traslacional, Facultad de Medicina, Universidad Católica del Maule, Talca 3480094, Chile; (G.U.); (E.W.H.-R.); (S.E.R.-R.)
| | - Elizabeth Valdés-Muñoz
- Doctorado en Biotecnología Traslacional, Facultad de Ciencias Agrarias y Forestales, Universidad Católica del Maule, Talca 3480094, Chile;
| | - Reynier Suardiaz
- Departamento de Química Física, Facultad de Ciencias Químicas, Universidad Complutense de Madrid, 28040 Madrid, Spain;
| | - Erix W. Hernández-Rodríguez
- Laboratorio de Bioinformática y Química Computacional, Departamento de Medicina Traslacional, Facultad de Medicina, Universidad Católica del Maule, Talca 3480094, Chile; (G.U.); (E.W.H.-R.); (S.E.R.-R.)
- Unidad de Bioinformática Clínica, Centro Oncológico, Facultad de Medicina, Universidad Católica del Maule, Talca 3480094, Chile
| | - Jonathan M. Palma
- Facultad de Ingeniería, Universidad de Talca, Curicó 3344158, Chile;
| | - Sofía E. Ríos-Rozas
- Laboratorio de Bioinformática y Química Computacional, Departamento de Medicina Traslacional, Facultad de Medicina, Universidad Católica del Maule, Talca 3480094, Chile; (G.U.); (E.W.H.-R.); (S.E.R.-R.)
| | | | - Melissa Alegría-Arcos
- Núcleo de Investigación en Data Science, Facultad de Ingeniería y Negocios, Universidad de las Américas, Santiago 7500000, Chile; (M.A.-A.); (O.Y.)
| | - Osvaldo Yáñez
- Núcleo de Investigación en Data Science, Facultad de Ingeniería y Negocios, Universidad de las Américas, Santiago 7500000, Chile; (M.A.-A.); (O.Y.)
| | - Luis Morales-Quintana
- Multidisciplinary Agroindustry Research Laboratory, Instituto de Ciencias Biomédicas, Facultad de Ciencias de la Salud, Universidad Autónoma de Chile, Cinco Pte. N° 1670, Talca 3467987, Chile;
| | - Vívian D’Afonseca
- Departamento de Ciencias Preclínicas, Facultad de Medicina, Universidad Católica del Maule, Ave. San Miguel 3605, Talca 3466706, Chile
| | - Daniel Bustos
- Laboratorio de Bioinformática y Química Computacional, Departamento de Medicina Traslacional, Facultad de Medicina, Universidad Católica del Maule, Talca 3480094, Chile; (G.U.); (E.W.H.-R.); (S.E.R.-R.)
| |
Collapse
|
29
|
Bedree JK, Bourgeois J, Balani P, Cen L, Hendrickson EL, Kerns KA, Camilli A, McLean JS, Shi W, He X. Identifying essential genes in Schaalia odontolytica using a highly-saturated transposon library. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.17.604004. [PMID: 39071323 PMCID: PMC11275721 DOI: 10.1101/2024.07.17.604004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/30/2024]
Abstract
The unique epibiotic-parasitic relationship between Nanosynbacter lyticus type strain TM7x, a member of the newly identified Candidate Phyla Radiation, now referred to as Patescibacteria, and its basibiont, Schaalia odontolytica strain XH001 (formerly Actinomyces odontolyticus), require more powerful genetic tools for deeper understanding of the genetic underpinnings that mediate their obligate relationship. Previous studies have mainly characterized the genomic landscape of XH001 during or post TM7x infection through comparative genomic or transcriptomic analyses followed by phenotypic analysis. Comprehensive genetic dissection of the pair is currently cumbersome due to the lack of robust genetic tools in TM7x. However, basic genetic tools are available for XH001 and this study expands the current genetic toolset by developing high-throughput transposon insertion sequencing (Tn-seq). Tn-seq was employed to screen for essential genes in XH001 under laboratory conditions. A highly saturated Tn-seq library was generated with nearly 660,000 unique insertion mutations, averaging one insertion every 2-3 nucleotides. 203 genes, 10.5% of the XH001 genome, were identified as putatively essential.
Collapse
Affiliation(s)
- Joseph K Bedree
- Section of Oral Biology, Division of Oral Biology and Medicine, School of Dentistry, University of California-Los Angeles, Los Angeles, CA, 90095
- Department of Microbiology, The ADA Forsyth Institute; Cambridge, MA, 02142
| | - Jacob Bourgeois
- Department of Molecular Biology and Microbiology, Tufts University School of Medicine, Boston, MA, USA
| | - Pooja Balani
- Department of Microbiology, The ADA Forsyth Institute; Cambridge, MA, 02142
- Department of Oral Medicine, Infection and Immunity, Harvard School of Dental Medicine, Boston, MA 02115
| | - Lujia Cen
- Department of Microbiology, The ADA Forsyth Institute; Cambridge, MA, 02142
| | - Erik L Hendrickson
- Department of Periodontics, School of Dentistry, University of Washington, Seattle, WA, 98195
| | - Kristopher A Kerns
- Department of Periodontics, School of Dentistry, University of Washington, Seattle, WA, 98195
| | - Andrew Camilli
- Department of Molecular Biology and Microbiology, Tufts University School of Medicine, Boston, MA, USA
| | - Jeffrey S McLean
- Department of Periodontics, School of Dentistry, University of Washington, Seattle, WA, 98195
| | - Wenyuan Shi
- Department of Microbiology, The ADA Forsyth Institute; Cambridge, MA, 02142
| | - Xuesong He
- Department of Microbiology, The ADA Forsyth Institute; Cambridge, MA, 02142
| |
Collapse
|
30
|
Bhattacharjee A, Hosen MR, Lamisa AB, Ahammad I, Chowdhury ZM, Jamal TB, Sohag MMH, Hossain MU, Das KC, Keya CA, Salimullah M. An integrated comparative genomics, subtractive proteomics and immunoinformatics framework for the rational design of a Pan-Salmonella multi-epitope vaccine. PLoS One 2024; 19:e0292413. [PMID: 38959229 PMCID: PMC11221655 DOI: 10.1371/journal.pone.0292413] [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: 09/19/2023] [Accepted: 04/23/2024] [Indexed: 07/05/2024] Open
Abstract
Salmonella infections pose a significant global public health concern due to the substantial expenses associated with monitoring, preventing, and treating the infection. In this study, we explored the core proteome of Salmonella to design a multi-epitope vaccine through Subtractive Proteomics and immunoinformatics approaches. A total of 2395 core proteins were curated from 30 different isolates of Salmonella (strain NZ CP014051 was taken as reference). Utilizing the subtractive proteomics approach on the Salmonella core proteome, Curlin major subunit A (CsgA) was selected as the vaccine candidate. csgA is a conserved gene that is related to biofilm formation. Immunodominant B and T cell epitopes from CsgA were predicted using numerous immunoinformatics tools. T lymphocyte epitopes had adequate population coverage and their corresponding MHC alleles showed significant binding scores after peptide-protein based molecular docking. Afterward, a multi-epitope vaccine was constructed with peptide linkers and Human Beta Defensin-2 (as an adjuvant). The vaccine could be highly antigenic, non-toxic, non-allergic, and have suitable physicochemical properties. Additionally, Molecular Dynamics Simulation and Immune Simulation demonstrated that the vaccine can bind with Toll Like Receptor 4 and elicit a robust immune response. Using in vitro, in vivo, and clinical trials, our findings could yield a Pan-Salmonella vaccine that might provide protection against various Salmonella species.
Collapse
Affiliation(s)
- Arittra Bhattacharjee
- Bioinformatics Division, National Institute of Biotechnology, Ganakbari, Ashulia, Savar, Dhaka, Bangladesh
| | - Md. Rakib Hosen
- Department of Genetic Engineering and Biotechnology, Jagannath University, Dhaka, Bangladesh
| | - Anika Bushra Lamisa
- Bioinformatics Division, National Institute of Biotechnology, Ganakbari, Ashulia, Savar, Dhaka, Bangladesh
| | - Ishtiaque Ahammad
- Bioinformatics Division, National Institute of Biotechnology, Ganakbari, Ashulia, Savar, Dhaka, Bangladesh
| | - Zeshan Mahmud Chowdhury
- Bioinformatics Division, National Institute of Biotechnology, Ganakbari, Ashulia, Savar, Dhaka, Bangladesh
| | - Tabassum Binte Jamal
- Bioinformatics Division, National Institute of Biotechnology, Ganakbari, Ashulia, Savar, Dhaka, Bangladesh
| | - Md. Mehadi Hasan Sohag
- Department of Genetic Engineering and Biotechnology, Jagannath University, Dhaka, Bangladesh
| | - Mohammad Uzzal Hossain
- Bioinformatics Division, National Institute of Biotechnology, Ganakbari, Ashulia, Savar, Dhaka, Bangladesh
| | - Keshob Chandra Das
- Molecular Biotechnology Division, National Institute of Biotechnology, Ganakbari, Ashulia, Savar, Dhaka, Bangladesh
| | - Chaman Ara Keya
- Department of Biochemistry and Microbiology, North South University, Bashundhara, Dhaka, Bangladesh
| | - Md Salimullah
- Molecular Biotechnology Division, National Institute of Biotechnology, Ganakbari, Ashulia, Savar, Dhaka, Bangladesh
| |
Collapse
|
31
|
Hasan A, Alonazi WB, Ibrahim M, Bin L. Immunoinformatics and Reverse Vaccinology Approach for the Identification of Potential Vaccine Candidates against Vandammella animalimors. Microorganisms 2024; 12:1270. [PMID: 39065039 PMCID: PMC11278545 DOI: 10.3390/microorganisms12071270] [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: 04/29/2024] [Revised: 06/17/2024] [Accepted: 06/19/2024] [Indexed: 07/28/2024] Open
Abstract
Vandammella animalimorsus is a Gram-negative and non-motile bacterium typically transmitted to humans through direct contact with the saliva of infected animals, primarily through biting, scratches, or licks on fractured skin. The absence of a confirmed post-exposure treatment of V. animalimorsus bacterium highlights the imperative for developing an effective vaccine. We intended to determine potential vaccine candidates and paradigm a chimeric vaccine against V. animalimorsus by accessible public data analysis of the strain by utilizing reverse vaccinology. By subtractive genomics, five outer membranes were prioritized as potential vaccine candidates out of 2590 proteins. Based on the instability index and transmembrane helices, a multidrug transporter protein with locus ID A0A2A2AHJ4 was designated as a potential candidate for vaccine construct. Sixteen immunodominant epitopes were retrieved by utilizing the Immune Epitope Database. The epitope encodes the strong binding affinity, nonallergenic properties, non-toxicity, high antigenicity scores, and high solubility revealing the more appropriate vaccine construct. By utilizing appropriate linkers and adjuvants alongside a suitable adjuvant molecule, the epitopes were integrated into a chimeric vaccine to enhance immunogenicity, successfully eliciting both adaptive and innate immune responses. Moreover, the promising physicochemical features, the binding confirmation of the vaccine to the major innate immune receptor TLR-4, and molecular dynamics simulations of the designed vaccine have revealed the promising potential of the selected candidate. The integration of computational methods and omics data has demonstrated significant advantages in discovering novel vaccine targets and mitigating vaccine failure rates during clinical trials in recent years.
Collapse
Affiliation(s)
- Ahmad Hasan
- State Key Laboratory of Rice Biology and Breeding, Key Laboratory of Molecular Biology of Crop Pathogens and Insects, Institute of Biotechnology, Zhejiang University, Hangzhou 310058, China; (A.H.); (M.I.)
| | - Wadi B. Alonazi
- Health Administration Department, College of Business Administration, King Saud University, Riyadh 11421, Saudi Arabia;
| | - Muhammad Ibrahim
- State Key Laboratory of Rice Biology and Breeding, Key Laboratory of Molecular Biology of Crop Pathogens and Insects, Institute of Biotechnology, Zhejiang University, Hangzhou 310058, China; (A.H.); (M.I.)
| | - Li Bin
- State Key Laboratory of Rice Biology and Breeding, Key Laboratory of Molecular Biology of Crop Pathogens and Insects, Institute of Biotechnology, Zhejiang University, Hangzhou 310058, China; (A.H.); (M.I.)
| |
Collapse
|
32
|
Fronk AD, Manzanares MA, Zheng P, Geier A, Anderson K, Stanton S, Zumrut H, Gera S, Munch R, Frederick V, Dhingra P, Arun G, Akerman M. Development and validation of AI/ML derived splice-switching oligonucleotides. Mol Syst Biol 2024; 20:676-701. [PMID: 38664594 PMCID: PMC11148135 DOI: 10.1038/s44320-024-00034-9] [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: 09/15/2023] [Revised: 04/03/2024] [Accepted: 04/09/2024] [Indexed: 06/05/2024] Open
Abstract
Splice-switching oligonucleotides (SSOs) are antisense compounds that act directly on pre-mRNA to modulate alternative splicing (AS). This study demonstrates the value that artificial intelligence/machine learning (AI/ML) provides for the identification of functional, verifiable, and therapeutic SSOs. We trained XGboost tree models using splicing factor (SF) pre-mRNA binding profiles and spliceosome assembly information to identify modulatory SSO binding sites on pre-mRNA. Using Shapley and out-of-bag analyses we also predicted the identity of specific SFs whose binding to pre-mRNA is blocked by SSOs. This step adds considerable transparency to AI/ML-driven drug discovery and informs biological insights useful in further validation steps. We applied this approach to previously established functional SSOs to retrospectively identify the SFs likely to regulate those events. We then took a prospective validation approach using a novel target in triple negative breast cancer (TNBC), NEDD4L exon 13 (NEDD4Le13). Targeting NEDD4Le13 with an AI/ML-designed SSO decreased the proliferative and migratory behavior of TNBC cells via downregulation of the TGFβ pathway. Overall, this study illustrates the ability of AI/ML to extract actionable insights from RNA-seq data.
Collapse
Affiliation(s)
| | | | - Paulina Zheng
- Envisagenics, Inc., Long Island City, NY, 11101, USA
| | - Adam Geier
- Envisagenics, Inc., Long Island City, NY, 11101, USA
| | | | | | - Hasan Zumrut
- Envisagenics, Inc., Long Island City, NY, 11101, USA
| | - Sakshi Gera
- Envisagenics, Inc., Long Island City, NY, 11101, USA
| | - Robin Munch
- Envisagenics, Inc., Long Island City, NY, 11101, USA
| | | | | | - Gayatri Arun
- Envisagenics, Inc., Long Island City, NY, 11101, USA
| | | |
Collapse
|
33
|
Rebhi S, Basharat Z, Wei CR, Lebbal S, Najjaa H, Sadfi-Zouaoui N, Messaoudi A. Core proteome mediated subtractive approach for the identification of potential therapeutic drug target against the honeybee pathogen Paenibacillus larvae. PeerJ 2024; 12:e17292. [PMID: 38818453 PMCID: PMC11138523 DOI: 10.7717/peerj.17292] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Accepted: 04/02/2024] [Indexed: 06/01/2024] Open
Abstract
Background & Objectives American foulbrood (AFB), caused by the highly virulent, spore-forming bacterium Paenibacillus larvae, poses a significant threat to honey bee brood. The widespread use of antibiotics not only fails to effectively combat the disease but also raises concerns regarding honey safety. The current computational study was attempted to identify a novel therapeutic drug target against P. larvae, a causative agent of American foulbrood disease in honey bee. Methods We investigated effective novel drug targets through a comprehensive in silico pan-proteome and hierarchal subtractive sequence analysis. In total, 14 strains of P. larvae genomes were used to identify core genes. Subsequently, the core proteome was systematically narrowed down to a single protein predicted as the potential drug target. Alphafold software was then employed to predict the 3D structure of the potential drug target. Structural docking was carried out between a library of phytochemicals derived from traditional Chinese flora (n > 36,000) and the potential receptor using Autodock tool 1.5.6. Finally, molecular dynamics (MD) simulation study was conducted using GROMACS to assess the stability of the best-docked ligand. Results Proteome mining led to the identification of Ketoacyl-ACP synthase III as a highly promising therapeutic target, making it a prime candidate for inhibitor screening. The subsequent virtual screening and MD simulation analyses further affirmed the selection of ZINC95910054 as a potent inhibitor, with the lowest binding energy. This finding presents significant promise in the battle against P. larvae. Conclusions Computer aided drug design provides a novel approach for managing American foulbrood in honey bee populations, potentially mitigating its detrimental effects on both bee colonies and the honey industry.
Collapse
Affiliation(s)
- Sawsen Rebhi
- Université de Tunis-El Manar, Laboratoire de Mycologie, Pathologies et Biomarqueurs, Département de Biologie, Tunis, Tunisia
| | | | - Calvin R. Wei
- Department of Research and Development, Shing Huei Group, Taipei, Taiwan
| | - Salim Lebbal
- University of Khenchela, Department of Agricultural Sciences, Faculty of Nature and Life Sciences, Khenchela, Algeria
| | - Hanen Najjaa
- University of Gabes, Laboratory of Pastoral Ecosystem and Valorization of Spontaneous Plants and Associated Microorganisms, Institute of Arid Lands of Medenine, Medenine, Tunisia
| | - Najla Sadfi-Zouaoui
- Université de Tunis-El Manar, Laboratoire de Mycologie, Pathologies et Biomarqueurs, Département de Biologie, Tunis, Tunisia
| | - Abdelmonaem Messaoudi
- Université de Tunis-El Manar, Laboratoire de Mycologie, Pathologies et Biomarqueurs, Département de Biologie, Tunis, Tunisia
- Jendouba University, Higher Institute of Biotechnology of Beja, Beja, Tunisia
| |
Collapse
|
34
|
Ahammad I, Bushra Lamisa A, Sharmin S, Bhattacharjee A, Mahmud Chowdhury Z, Ahamed T, Uzzal Hossain M, Chandra Das K, Salimullah M, Ara Keya C. Subtractive genomics study for the identification of therapeutic targets against Cronobacter sakazakii: A threat to infants. Heliyon 2024; 10:e30332. [PMID: 38707387 PMCID: PMC11066692 DOI: 10.1016/j.heliyon.2024.e30332] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2023] [Revised: 04/23/2024] [Accepted: 04/24/2024] [Indexed: 05/07/2024] Open
Abstract
Cronobacter sakazakii is an opportunistic pathogen that has been associated with severe infection in neonates such as necrotizing enterocolitis (NEC), neonatal meningitis, and bacteremia. This pathogen can survive in a relatively dry environment, especially in powdered infant formula (PIF). Unfortunately, conventional drugs that were once effective against C. sakazakii are gradually losing their efficacy due to rising antibiotic resistance. In this study, a subtractive genomic approach was followed in order to identify potential therapeutic targets in the pathogen. The whole proteome of the pathogen was filtered through a step-by-step process, which involved removing paralogous proteins, human homologs, sequences that are less essential for survival, proteins with shared metabolic pathways, and proteins that are located in cells other than the cytoplasmic membrane. As a result, nine novel drug targets were identified. Further, the analysis also unveiled that the FDA-approved drug Terbinafine can be repurposed against the Glutathione/l-cysteine transport system ATP-binding/permease protein CydC of C. sakazakii. Moreover, molecular docking and dynamics studies of Terbinafine and CydC suggested that this drug can be used to treat C. sakazakii infection in neonates. However, for clinical purposes further in vitro and in vivo studies are necessary.
Collapse
Affiliation(s)
- Ishtiaque Ahammad
- Bioinformatics Division, National Institute of Biotechnology, Ganakbari, Ashulia, Savar, Dhaka, 1349, Bangladesh
| | - Anika Bushra Lamisa
- Bioinformatics Division, National Institute of Biotechnology, Ganakbari, Ashulia, Savar, Dhaka, 1349, Bangladesh
- Department of Biochemistry and Microbiology, North South University, Bashundhara, Dhaka, 1229, Bangladesh
| | - Sadia Sharmin
- Department of Biotechnology & Genetic Engineering, Jahangirnagar University, Savar, Dhaka, 1342, Bangladesh
| | - Arittra Bhattacharjee
- Bioinformatics Division, National Institute of Biotechnology, Ganakbari, Ashulia, Savar, Dhaka, 1349, Bangladesh
| | - Zeshan Mahmud Chowdhury
- Bioinformatics Division, National Institute of Biotechnology, Ganakbari, Ashulia, Savar, Dhaka, 1349, Bangladesh
| | - Tanvir Ahamed
- Department of Biotechnology & Genetic Engineering, Jahangirnagar University, Savar, Dhaka, 1342, Bangladesh
| | - Mohammad Uzzal Hossain
- Bioinformatics Division, National Institute of Biotechnology, Ganakbari, Ashulia, Savar, Dhaka, 1349, Bangladesh
| | - Keshob Chandra Das
- Molecular Biotechnology Division, National Institute of Biotechnology, Ganakbari, Ashulia, Savar, Dhaka, 1349, Bangladesh
| | - Md Salimullah
- Molecular Biotechnology Division, National Institute of Biotechnology, Ganakbari, Ashulia, Savar, Dhaka, 1349, Bangladesh
| | - Chaman Ara Keya
- Department of Biochemistry and Microbiology, North South University, Bashundhara, Dhaka, 1229, Bangladesh
| |
Collapse
|
35
|
Gorityala N, Baidya AS, Sagurthi SR. Genome mining of Mycobacterium tuberculosis: targeting SufD as a novel drug candidate through in silico characterization and inhibitor screening. Front Microbiol 2024; 15:1369645. [PMID: 38686111 PMCID: PMC11057465 DOI: 10.3389/fmicb.2024.1369645] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Accepted: 03/15/2024] [Indexed: 05/02/2024] Open
Abstract
Tuberculosis (TB) stands as the second most fatal infectious disease globally, causing 1.3 million deaths in 2022. The resurgence of TB and the alarming rise of antibiotic resistance demand urgent call to develop novel antituberculosis drugs. Despite concerted efforts to control TB, the disease persists and spreads rapidly on a global scale. Targeting stress response pathways in Mycobacterium tuberculosis (Mtb) has become imperative to achieve complete eradication. This study employs subtractive genomics to identify and prioritize potential drug targets among the hypothetical proteins of Mtb, focusing on indispensable pathways. Amongst 177 essential hypothetical proteins, 152 were nonhomologous to human. These proteins participated in 34 pathways, and a 20-fold enrichment of SUF pathway genes led to its selection as a target pathway. Fe-S clusters are fundamental, widely distributed protein cofactors involved in vital cellular processes. The survival of Mtb in a hypoxic environment relies on the iron-sulfur (Fe-S) cluster biogenesis pathway for the repair of damaged Fe-S clusters. It also protects pathogen against drugs, ensuring controlled iron utilization and contributing to drug resistance. In Mtb, six proteins of Fe-S cluster assembly pathway are encoded by the suf operon. The present study was focused on SufD because of its role in iron acquisition and prevention of Fenton reaction. The research further delves into the in silico characterization of SufD, utilizing bioinformatics tools for sequence and structure based analysis. The protein's structural features, including the identification of conserved regions, motifs, and 3D structure prediction enhanced functional annotation. Target based virtual screening of compounds from the ChEMBL database resulted in 12 inhibitors with best binding affinities. Drug likeness and ADMET profiling of potential inhibitors identified promising compounds with favorable drug-like properties. The study also involved cloning in SUMO-pRSF-Duet1 expression vector, overexpression, and purification of recombinant SufD from E. coli BL21 (DE3) cells. Optimization of expression conditions resulted in soluble production, and subsequent purification highlighting the efficacy of the SUMO fusion system for challenging Mtb proteins in E. coli. These findings provide valuable insights into pharmacological targets for future experimental studies, holding promise for the development of targeted therapy against Mtb.
Collapse
Affiliation(s)
- Neelima Gorityala
- Department of Genetics and Biotechnology, Osmania University, Hyderabad, Telangana, India
| | - Anthony Samit Baidya
- Department of Genetics and Biotechnology, Osmania University, Hyderabad, Telangana, India
| | - Someswar R Sagurthi
- Department of Genetics and Biotechnology, Osmania University, Hyderabad, Telangana, India
| |
Collapse
|
36
|
Humphreys IR, Zhang J, Baek M, Wang Y, Krishnakumar A, Pei J, Anishchenko I, Tower CA, Jackson BA, Warrier T, Hung DT, Peterson SB, Mougous JD, Cong Q, Baker D. Essential and virulence-related protein interactions of pathogens revealed through deep learning. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.12.589144. [PMID: 38645026 PMCID: PMC11030334 DOI: 10.1101/2024.04.12.589144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/23/2024]
Abstract
Identification of bacterial protein-protein interactions and predicting the structures of the complexes could aid in the understanding of pathogenicity mechanisms and developing treatments for infectious diseases. Here, we developed a deep learning-based pipeline that leverages residue-residue coevolution and protein structure prediction to systematically identify and structurally characterize protein-protein interactions at the proteome-wide scale. Using this pipeline, we searched through 78 million pairs of proteins across 19 human bacterial pathogens and identified 1923 confidently predicted complexes involving essential genes and 256 involving virulence factors. Many of these complexes were not previously known; we experimentally tested 12 such predictions, and half of them were validated. The predicted interactions span core metabolic and virulence pathways ranging from post-transcriptional modification to acid neutralization to outer membrane machinery and should contribute to our understanding of the biology of these important pathogens and the design of drugs to combat them.
Collapse
|
37
|
Basharat Z, Murtaza Z, Siddiqa A, Alnasser SM, Meshal A. Therapeutic target mapping from the genome of Kingella negevensis and biophysical inhibition assessment through PNP synthase binding with traditional medicinal compounds. Mol Divers 2024; 28:581-594. [PMID: 36645537 PMCID: PMC9842218 DOI: 10.1007/s11030-023-10604-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Accepted: 01/10/2023] [Indexed: 01/17/2023]
Abstract
Kingella negevensis belongs to the Neisseriaceae family. It is implied that it has significant virulence potential due to RTX toxin production, which can cause hemolysis. It usually colonizes the orophayrynx of pediatric population, along with Kingella kingae but has also been isolated from vagina. Todate no report on its drug targets is present, therefore putative therapeutic targets were identified from its genomic sequence data. Traditional Chinese (n > 36,000) and Indian medicinal compounds (n > 2000) were then screened against its pyridoxine 5'-phosphate synthase, a vital therapeutic target. Prioritized TCM compounds included ZINC02525131, ZINC33833737 and ZINC85486932, and Cadiyenol, 9,11,13-Octadecatrienoic acid and 6-Gingerol from Indian medicinal library. Molecular dynamics simulation of top compounds revealed ZINC02525131 as having best stability for 100 ns, compared to Cadiyenol. ADMET profiling was then done, along with physiologically based pharmacokinetic simulation of these compounds in a population of 200 individuals, for 12 h to see fate of the ingested compound. Additionally, the impact of these compounds in a population with cirrhosis and renal impairment was also simulated. We imply in light of all the studied parameters of safety and bioavailability, etc., that 6-Gingerol from Zingiber officinalis rhizome must be proceeded further for in vitro and in vivo testing for inhibition of K. negevensis.
Collapse
Affiliation(s)
- Zarrin Basharat
- Jamil-ur-Rahman Center for Genome Research, Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, 75270, Pakistan.
| | - Zainab Murtaza
- Department of Zoology, Government College University, Lahore, 54000, Pakistan
| | - Aisha Siddiqa
- Jamil-ur-Rahman Center for Genome Research, Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, 75270, Pakistan
| | - Sulaiman Mohammed Alnasser
- Department of Pharmacology and Toxicology, Unaizah College of Pharmacy, Qassim University, Buraydah, 52571, Saudi Arabia
| | - Alotaibi Meshal
- Department of Pharmacy Practice, College of Pharmacy, University of Hafr Albatin, Hafr Albatin, Saudi Arabia
| |
Collapse
|
38
|
Saha S, Chatterjee P, Basu S, Nasipuri M. EPI-SF: essential protein identification in protein interaction networks using sequence features. PeerJ 2024; 12:e17010. [PMID: 38495766 PMCID: PMC10944162 DOI: 10.7717/peerj.17010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Accepted: 02/05/2024] [Indexed: 03/19/2024] Open
Abstract
Proteins are considered indispensable for facilitating an organism's viability, reproductive capabilities, and other fundamental physiological functions. Conventional biological assays are characterized by prolonged duration, extensive labor requirements, and financial expenses in order to identify essential proteins. Therefore, it is widely accepted that employing computational methods is the most expeditious and effective approach to successfully discerning essential proteins. Despite being a popular choice in machine learning (ML) applications, the deep learning (DL) method is not suggested for this specific research work based on sequence features due to the restricted availability of high-quality training sets of positive and negative samples. However, some DL works on limited availability of data are also executed at recent times which will be our future scope of work. Conventional ML techniques are thus utilized in this work due to their superior performance compared to DL methodologies. In consideration of the aforementioned, a technique called EPI-SF is proposed here, which employs ML to identify essential proteins within the protein-protein interaction network (PPIN). The protein sequence is the primary determinant of protein structure and function. So, initially, relevant protein sequence features are extracted from the proteins within the PPIN. These features are subsequently utilized as input for various machine learning models, including XGB Boost Classifier, AdaBoost Classifier, logistic regression (LR), support vector classification (SVM), Decision Tree model (DT), Random Forest model (RF), and Naïve Bayes model (NB). The objective is to detect the essential proteins within the PPIN. The primary investigation conducted on yeast examined the performance of various ML models for yeast PPIN. Among these models, the RF model technique had the highest level of effectiveness, as indicated by its precision, recall, F1-score, and AUC values of 0.703, 0.720, 0.711, and 0.745, respectively. It is also found to be better in performance when compared to the other state-of-arts based on traditional centrality like betweenness centrality (BC), closeness centrality (CC), etc. and deep learning methods as well like DeepEP, as emphasized in the result section. As a result of its favorable performance, EPI-SF is later employed for the prediction of novel essential proteins inside the human PPIN. Due to the tendency of viruses to selectively target essential proteins involved in the transmission of diseases within human PPIN, investigations are conducted to assess the probable involvement of these proteins in COVID-19 and other related severe diseases.
Collapse
Affiliation(s)
- Sovan Saha
- Department of Computer Science & Engineering (Artificial Intelligence & Machine Learning), Techno Main Salt Lake, Kolkata, West Bengal, India
| | - Piyali Chatterjee
- Department of Computer Science & Engineering, Netaji Subhash Engineering College, Kolkata, West Bengal, India
| | - Subhadip Basu
- Department of Computer Science & Engineering, Jadavpur University, Kolkata, West Bengal, India
| | - Mita Nasipuri
- Department of Computer Science & Engineering, Jadavpur University, Kolkata, West Bengal, India
| |
Collapse
|
39
|
Ahammad I, Jamal TB, Lamisa AB, Bhattacharjee A, Zinan N, Hasan Chowdhury MZ, Naimul Islam SM, Faruque KMO, Mahmud Chowdhury Z, Uzzal Hossain M, Chandra Das K, Ara Keya C, Salimullah M. Subtractive genomics study of Xanthomonas oryzae pv. Oryzae reveals repurposable drug candidate for the treatment of bacterial leaf blight in rice. J Genet Eng Biotechnol 2024; 22:100353. [PMID: 38494267 PMCID: PMC10980872 DOI: 10.1016/j.jgeb.2024.100353] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Accepted: 01/15/2024] [Indexed: 03/19/2024]
Abstract
BACKGROUND Xanthomonas oryzae pv. oryzae is a plant pathogen responsible for causing one of the most severe bacterial diseases in rice, known as bacterial leaf blight that poses a major threat to global rice production. Even though several experimental compounds and chemical agents have been tested against X. oryzae pv. oryzae, still no approved drug is available. In this study, a subtractive genomic approach was used to identify potential therapeutic targets and repurposible drug candidates that could control of bacterial leaf blight in rice plants. RESULTS The entire proteome of the pathogen underwent an extensive filtering process which involved removal of the paralogous proteins, rice homologs, non-essential proteins. Out of the 4382 proteins present in Xoo proteome, five hub proteins such as dnaA, dnaN, recJ, ruvA, and recR were identified for the druggability analysis. This analysis led to the identification of dnaN-encoded Beta sliding clamp protein as a potential therapeutic target and one experimental drug named [(5R)-5-(2,3-dibromo-5-ethoxy-4hydroxybenzyl)-4-oxo-2-thioxo-1,3-thiazolidin-3-yl]acetic acid that can be repurposed against it. Molecular docking and 100 ns long molecular dynamics simulation suggested that the drug can form stable complexes with the target protein over time. CONCLUSION Findings from our study indicated that the proposed drug showed potential effectiveness against bacterial leaf blight in rice caused by X. oryzae pv. oryzae. It is essential to keep in consideration that the procedure for developing novel drugs can be challenging and complicated. Even the most promising results from in silico studies should be validated through further in vitro and in vivo investigation before approval.
Collapse
Affiliation(s)
- Ishtiaque Ahammad
- Bioinformatics Division, National Institute of Biotechnology, Ganakbari, Ashulia, Savar, Dhaka 1349, Bangladesh
| | - Tabassum Binte Jamal
- Bioinformatics Division, National Institute of Biotechnology, Ganakbari, Ashulia, Savar, Dhaka 1349, Bangladesh
| | - Anika Bushra Lamisa
- Bioinformatics Division, National Institute of Biotechnology, Ganakbari, Ashulia, Savar, Dhaka 1349, Bangladesh
| | - Arittra Bhattacharjee
- Bioinformatics Division, National Institute of Biotechnology, Ganakbari, Ashulia, Savar, Dhaka 1349, Bangladesh
| | - Nayeematul Zinan
- Institute of Biotechnology and Genetic Engineering, Bangabandhu Sheikh Mujibur Rahman Agricultural University, Gazipur 1706, Bangladesh
| | - Md Zahid Hasan Chowdhury
- Institute of Biotechnology and Genetic Engineering, Bangabandhu Sheikh Mujibur Rahman Agricultural University, Gazipur 1706, Bangladesh
| | - Shah Mohammad Naimul Islam
- Institute of Biotechnology and Genetic Engineering, Bangabandhu Sheikh Mujibur Rahman Agricultural University, Gazipur 1706, Bangladesh
| | | | - Zeshan Mahmud Chowdhury
- Bioinformatics Division, National Institute of Biotechnology, Ganakbari, Ashulia, Savar, Dhaka 1349, Bangladesh
| | - Mohammad Uzzal Hossain
- Bioinformatics Division, National Institute of Biotechnology, Ganakbari, Ashulia, Savar, Dhaka 1349, Bangladesh
| | - Keshob Chandra Das
- Molecular Biotechnology Division, National Institute of Biotechnology, Ganakbari, Ashulia, Savar, Dhaka 1349, Bangladesh
| | - Chaman Ara Keya
- Department of Biochemistry and Microbiology, North South University, Bashundhara, Dhaka 1229, Bangladesh
| | - Md Salimullah
- Molecular Biotechnology Division, National Institute of Biotechnology, Ganakbari, Ashulia, Savar, Dhaka 1349, Bangladesh.
| |
Collapse
|
40
|
Basharat Z, Sattar S, Bahauddin AA, Al Mouslem AK, Alotaibi G. Screening Marine Microbial Metabolites as Promising Inhibitors of Borrelia garinii: A Structural Docking Approach towards Developing Novel Lyme Disease Treatment. BIOMED RESEARCH INTERNATIONAL 2024; 2024:9997082. [PMID: 38456098 PMCID: PMC10919988 DOI: 10.1155/2024/9997082] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/23/2023] [Revised: 01/26/2024] [Accepted: 02/13/2024] [Indexed: 03/09/2024]
Abstract
Lyme disease caused by the Borrelia species is a growing health concern in many parts of the world. Current treatments for the disease may have side effects, and there is also a need for new therapies that can selectively target the bacteria. Pathogens responsible for Lyme disease include B. burgdorferi, B. afzelii, and B. garinii. In this study, we employed structural docking-based screening to identify potential lead-like inhibitors against the bacterium. We first identified the core essential genome fraction of the bacterium, using 37 strains. Later, we screened a library of lead-like marine microbial metabolites (n = 4730) against the arginine deiminase (ADI) protein of Borrelia garinii. This protein plays a crucial role in the survival of the bacteria, and inhibiting it can kill the bacterium. The prioritized lead compounds demonstrating favorable binding energies and interactions with the active site of ADI were then evaluated for their drug-like and pharmacokinetic parameters to assess their suitability for development as drugs. Results from molecular dynamics simulation (100 ns) and other scoring parameters suggest that the compound CMNPD18759 (common name: aureobasidin; IUPAC name: 2-[(4R,6R)-4,6-dihydroxydecanoyl]oxypropan-2-yl (3S,5R)-3,5-dihydroxydecanoate) holds promise as a potential drug candidate for the treatment of Lyme disease, caused by B. garinii. However, further experimental studies are needed to validate the efficacy and safety of this compound in vivo.
Collapse
Affiliation(s)
| | - Sadia Sattar
- Molecular Virology Labs, Department of Biosciences, COMSATS University Islamabad, Islamabad Campus, Islamabad 45550, Pakistan
| | | | - Abdulaziz K. Al Mouslem
- Department of Pharmaceutical Sciences, College of Clinical Pharmacy, King Faisal University, Al Ahsa 31982, Saudi Arabia
| | - Ghallab Alotaibi
- Department of Pharmacology, College of Pharmacy, Al-Dawadmi Campus, Shaqra University, Shaqra, Saudi Arabia
| |
Collapse
|
41
|
Ahmad F, Ismail S, Azam SS. Discovery of novel inhibitor via molecular dynamics simulations against D-alanyl-D-alanine carboxypeptidase of Enterobacter cloacae. J Biomol Struct Dyn 2024:1-16. [PMID: 38375604 DOI: 10.1080/07391102.2024.2316790] [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: 09/26/2023] [Accepted: 02/01/2024] [Indexed: 02/21/2024]
Abstract
Antibiotics resistance by bacterial pathogens is a major concern to public health worldwide resulting in high health care costs and rising mortality. Subtractive proteomics prioritized D-alanyl-D-alanine carboxypeptidas (DacB) enzyme from Enterobacter cloacae ATCC 13047 as a potential candidate for drugs designing to block pathogen cell wall biosynthesis. Virtual screening of an antibacterial library against the target unraveled a hit compound (2-[(1-methylsulfonylpiperidin-3-yl)methyl]-6-(1H-pyrazol-4-yl) pyrazine) showing high affinity and stability with the target. The N-methyl-N-propyl-methanesulfonamide of the compound is seen as a closed affinity towards domain involving strong hydrogen bonds with Ser41, Lys44, Ser285, and Asn287. The 4-methyl-1H-pyrazole is posed towards the open cavity of domain I and II and formed hydrophobic and hydrophilic contacts. The system is highly stable with average carbon-alpha deviations of 1.69 Å over trajectories of 400-ns. Three vital residues projected: Arg437, Arg438 and Leu400 from enzyme pocket via Radial distribution function (RDF) assay, which actively engaged the inhibitor. Further confirmation is done by estimating binding free energies, which confirms the very low delta energy of -7.24 kcal/mol in Generalized Born (GB) method and -7.4363 kcal/mol in Poisson-Boltzmann (PB) method. WaterSwap calculations were performed that revealed the energies highly converged, an agreement on good system stability. Lastly, three DacB mutants were created to investigate the role of functional active residues and a decline in binding affinity of the residues was noticed. These computational results provide a gateway for experimentalists to further confirm their efficacy both in-vitro and in-vivo.
Collapse
Affiliation(s)
- Faisal Ahmad
- Computational Biology Lab, National Center for Bioinformatics (NCB), Quaid-i-Azam University, Islamabad, Pakistan
| | - Saba Ismail
- Computational Biology Lab, National Center for Bioinformatics (NCB), Quaid-i-Azam University, Islamabad, Pakistan
| | - Syed Sikander Azam
- Computational Biology Lab, National Center for Bioinformatics (NCB), Quaid-i-Azam University, Islamabad, Pakistan
| |
Collapse
|
42
|
Liang Y, Luo H, Lin Y, Gao F. Recent advances in the characterization of essential genes and development of a database of essential genes. IMETA 2024; 3:e157. [PMID: 38868518 PMCID: PMC10989110 DOI: 10.1002/imt2.157] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Accepted: 10/09/2023] [Indexed: 06/14/2024]
Abstract
Over the past few decades, there has been a significant interest in the study of essential genes, which are crucial for the survival of an organism under specific environmental conditions and thus have practical applications in the fields of synthetic biology and medicine. An increasing amount of experimental data on essential genes has been obtained with the continuous development of technological methods. Meanwhile, various computational prediction methods, related databases and web servers have emerged accordingly. To facilitate the study of essential genes, we have established a database of essential genes (DEG), which has become popular with continuous updates to facilitate essential gene feature analysis and prediction, drug and vaccine development, as well as artificial genome design and construction. In this article, we summarized the studies of essential genes, overviewed the relevant databases, and discussed their practical applications. Furthermore, we provided an overview of the main applications of DEG and conducted comprehensive analyses based on its latest version. However, it should be noted that the essential gene is a dynamic concept instead of a binary one, which presents both opportunities and challenges for their future development.
Collapse
Affiliation(s)
| | - Hao Luo
- Department of PhysicsTianjin UniversityTianjinChina
| | - Yan Lin
- Department of PhysicsTianjin UniversityTianjinChina
| | - Feng Gao
- Department of PhysicsTianjin UniversityTianjinChina
- Frontiers Science Center for Synthetic Biology and Key Laboratory of Systems Bioengineering (Ministry of Education)Tianjin UniversityTianjinChina
- SynBio Research PlatformCollaborative Innovation Center of Chemical Science and Engineering (Tianjin)TianjinChina
| |
Collapse
|
43
|
Aarthy M, Pandiyan GN, Paramasivan R, Kumar A, Gupta B. Identification and prioritisation of potential vaccine candidates using subtractive proteomics and designing of a multi-epitope vaccine against Wuchereria bancrofti. Sci Rep 2024; 14:1970. [PMID: 38263422 PMCID: PMC10806236 DOI: 10.1038/s41598-024-52457-x] [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: 06/26/2023] [Accepted: 01/18/2024] [Indexed: 01/25/2024] Open
Abstract
This study employed subtractive proteomics and immunoinformatics to analyze the Wuchereria bancrofti proteome and identify potential therapeutic targets, with a focus on designing a vaccine against the parasite species. A comprehensive bioinformatics analysis of the parasite's proteome identified 51 probable therapeutic targets, among which "Kunitz/bovine pancreatic trypsin inhibitor domain-containing protein" was identified as the most promising vaccine candidate. The candidate protein was used to design a multi-epitope vaccine, incorporating B-cell and T-cell epitopes identified through various tools. The vaccine construct underwent extensive analysis of its antigenic, physical, and chemical features, including the determination of secondary and tertiary structures. Docking and molecular dynamics simulations were performed with HLA alleles, Toll-like receptor 4 (TLR4), and TLR3 to assess its potential to elicit the human immune response. Immune simulation analysis confirmed the predicted vaccine's strong binding affinity with immunoglobulins, indicating its potential efficacy in generating an immune response. However, experimental validation and testing of this multi-epitope vaccine construct would be needed to assess its potential against W. bancrofti and even for a broader range of lymphatic filarial infections given the similarities between W. bancrofti and Brugia.
Collapse
Affiliation(s)
- Murali Aarthy
- ICMR-Vector Control Research Centre (VCRC), Field Station, Madurai, Tamil Nadu, 625002, India
| | - G Navaneetha Pandiyan
- ICMR-Vector Control Research Centre (VCRC), Field Station, Madurai, Tamil Nadu, 625002, India
| | - R Paramasivan
- ICMR-Vector Control Research Centre (VCRC), Field Station, Madurai, Tamil Nadu, 625002, India
| | - Ashwani Kumar
- ICMR-Vector Control Research Centre (VCRC), Puducherry, India
- Saveetha Institute of Medical and Technical Sciences (SIMATS), Saveetha University, Tandhalam, Chennai, Tamil Nadu, 602105, India
| | - Bhavna Gupta
- ICMR-Vector Control Research Centre (VCRC), Field Station, Madurai, Tamil Nadu, 625002, India.
| |
Collapse
|
44
|
Hu W, Li M, Xiao H, Guan L. Essential genes identification model based on sequence feature map and graph convolutional neural network. BMC Genomics 2024; 25:47. [PMID: 38200437 PMCID: PMC10777564 DOI: 10.1186/s12864-024-09958-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2023] [Accepted: 01/01/2024] [Indexed: 01/12/2024] Open
Abstract
BACKGROUND Essential genes encode functions that play a vital role in the life activities of organisms, encompassing growth, development, immune system functioning, and cell structure maintenance. Conventional experimental techniques for identifying essential genes are resource-intensive and time-consuming, and the accuracy of current machine learning models needs further enhancement. Therefore, it is crucial to develop a robust computational model to accurately predict essential genes. RESULTS In this study, we introduce GCNN-SFM, a computational model for identifying essential genes in organisms, based on graph convolutional neural networks (GCNN). GCNN-SFM integrates a graph convolutional layer, a convolutional layer, and a fully connected layer to model and extract features from gene sequences of essential genes. Initially, the gene sequence is transformed into a feature map using coding techniques. Subsequently, a multi-layer GCN is employed to perform graph convolution operations, effectively capturing both local and global features of the gene sequence. Further feature extraction is performed, followed by integrating convolution and fully-connected layers to generate prediction results for essential genes. The gradient descent algorithm is utilized to iteratively update the cross-entropy loss function, thereby enhancing the accuracy of the prediction results. Meanwhile, model parameters are tuned to determine the optimal parameter combination that yields the best prediction performance during training. CONCLUSIONS Experimental evaluation demonstrates that GCNN-SFM surpasses various advanced essential gene prediction models and achieves an average accuracy of 94.53%. This study presents a novel and effective approach for identifying essential genes, which has significant implications for biology and genomics research.
Collapse
Affiliation(s)
- Wenxing Hu
- College of Physics and Electronic Information, Gannan Normal University, Ganzhou, Jiangxi, 341000, China
| | - Mengshan Li
- College of Physics and Electronic Information, Gannan Normal University, Ganzhou, Jiangxi, 341000, China.
| | - Haiyang Xiao
- College of Physics and Electronic Information, Gannan Normal University, Ganzhou, Jiangxi, 341000, China
| | - Lixin Guan
- College of Physics and Electronic Information, Gannan Normal University, Ganzhou, Jiangxi, 341000, China
| |
Collapse
|
45
|
Nazli A, Qiu J, Tang Z, He Y. Recent Advances and Techniques for Identifying Novel Antibacterial Targets. Curr Med Chem 2024; 31:464-501. [PMID: 36734893 DOI: 10.2174/0929867330666230123143458] [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: 05/24/2022] [Revised: 10/30/2022] [Accepted: 11/11/2022] [Indexed: 02/04/2023]
Abstract
BACKGROUND With the emergence of drug-resistant bacteria, the development of new antibiotics is urgently required. Target-based drug discovery is the most frequently employed approach for the drug development process. However, traditional drug target identification techniques are costly and time-consuming. As research continues, innovative approaches for antibacterial target identification have been developed which enabled us to discover drug targets more easily and quickly. METHODS In this review, methods for finding drug targets from omics databases have been discussed in detail including principles, procedures, advantages, and potential limitations. The role of phage-driven and bacterial cytological profiling approaches is also discussed. Moreover, current article demonstrates the advancements being made in the establishment of computational tools, machine learning algorithms, and databases for antibacterial target identification. RESULTS Bacterial drug targets successfully identified by employing these aforementioned techniques are described as well. CONCLUSION The goal of this review is to attract the interest of synthetic chemists, biologists, and computational researchers to discuss and improve these methods for easier and quicker development of new drugs.
Collapse
Affiliation(s)
- Adila Nazli
- Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, School of Pharmaceutical Sciences, Chongqing University, Chongqing, 401331, P. R. China
| | - Jingyi Qiu
- Chongqing Institute of Green and Intelligent Technology, Chinese Academy of Sciences, 266 Fangzheng Avenue, Chongqing, 400714, P. R. China
| | - Ziyi Tang
- Chongqing Institute of Green and Intelligent Technology, Chinese Academy of Sciences, 266 Fangzheng Avenue, Chongqing, 400714, P. R. China
| | - Yun He
- Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, School of Pharmaceutical Sciences, Chongqing University, Chongqing, 401331, P. R. China
| |
Collapse
|
46
|
Carter EW, Peraza OG, Wang N. The protein interactome of the citrus Huanglongbing pathogen Candidatus Liberibacter asiaticus. Nat Commun 2023; 14:7838. [PMID: 38030598 PMCID: PMC10687234 DOI: 10.1038/s41467-023-43648-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Accepted: 11/15/2023] [Indexed: 12/01/2023] Open
Abstract
The bacterium Candidatus Liberibacter asiaticus (CLas) causes citrus Huanglongbing disease. Our understanding of the pathogenicity and biology of this microorganism remains limited because CLas has not yet been cultivated in artificial media. Its genome is relatively small and encodes approximately 1136 proteins, of which 415 have unknown functions. Here, we use a high-throughput yeast-two-hybrid (Y2H) screen to identify interactions between CLas proteins, thus providing insights into their potential functions. We identify 4245 interactions between 542 proteins, after screening 916 bait and 936 prey proteins. The false positive rate of the Y2H assay is estimated to be 2.9%. Pull-down assays for nine protein-protein interactions (PPIs) likely involved in flagellar function support the robustness of the Y2H results. The average number of PPIs per node in the CLas interactome is 15.6, which is higher than the numbers previously reported for interactomes of free-living bacteria, suggesting that CLas genome reduction has been accompanied by increased protein multi-functionality. We propose potential functions for 171 uncharacterized proteins, based on the PPI results, guilt-by-association analyses, and comparison with data from other bacterial species. We identify 40 hub-node proteins, including quinone oxidoreductase and LysR, which are known to protect other bacteria against oxidative stress and might be important for CLas survival in the phloem. We expect our PPI database to facilitate research on CLas biology and pathogenicity mechanisms.
Collapse
Affiliation(s)
- Erica W Carter
- Citrus Research and Education Center, Institute of Food and Agricultural Sciences, University of Florida, Lake Alfred, FL, USA
- Department of Plant Pathology, Institute of Food and Agricultural Sciences, University of Florida, Lake Alfred, FL, USA
| | - Orlene Guerra Peraza
- Citrus Research and Education Center, Institute of Food and Agricultural Sciences, University of Florida, Lake Alfred, FL, USA
| | - Nian Wang
- Citrus Research and Education Center, Institute of Food and Agricultural Sciences, University of Florida, Lake Alfred, FL, USA.
- Department of Microbiology and Cell Science, Institute of Food and Agricultural Sciences, University of Florida, Lake Alfred, FL, US.
| |
Collapse
|
47
|
Chen ES, Ho ES. In-silico study of antisense oligonucleotide antibiotics. PeerJ 2023; 11:e16343. [PMID: 38025700 PMCID: PMC10656905 DOI: 10.7717/peerj.16343] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Accepted: 10/03/2023] [Indexed: 12/01/2023] Open
Abstract
Background The rapid emergence of antibiotic-resistant bacteria directly contributes to a wave of untreatable infections. The lack of new drug development is an important driver of this crisis. Most antibiotics today are small molecules that block vital processes in bacteria. To optimize such effects, the three-dimensional structure of targeted bacterial proteins is imperative, although such a task is time-consuming and tedious, impeding the development of antibiotics. The development of RNA-based therapeutics has catalyzed a new platform of antibiotics-antisense oligonucleotides (ASOs). These molecules hybridize with their target mRNAs with high specificity, knocking down or interfering with protein translation. This study aims to develop a bioinformatics pipeline to identify potent ASO targets in essential bacterial genes. Methods Three bacterial species (P. gingivalis, H. influenzae, and S. aureus) were used to demonstrate the utility of the pipeline. Open reading frames of bacterial essential genes were downloaded from the Database of Essential Genes (DEG). After filtering for specificity and accessibility, ASO candidates were ranked based on their self-hybridization score, predicted melting temperature, and the position on the gene in an operon. Enrichment analysis was conducted on genes associated with putative potent ASOs. Results A total of 45,628 ASOs were generated from 348 unique essential genes in P. gingivalis. A total of 1,117 of them were considered putative. A total of 27,273 ASOs were generated from 191 unique essential genes in H. influenzae. A total of 847 of them were considered putative. A total of 175,606 ASOs were generated from 346 essential genes in S. aureus. A total of 7,061 of them were considered putative. Critical biological processes associated with these genes include translation, regulation of cell shape, cell division, and peptidoglycan biosynthetic process. Putative ASO targets generated for each bacterial species are publicly available here: https://github.com/EricSHo/AOA. The results demonstrate that our bioinformatics pipeline is useful in identifying unique and accessible ASO targets in bacterial species that post major public health issues.
Collapse
Affiliation(s)
- Erica S. Chen
- Biology, Lafayette College, Easton, PA, United States
| | - Eric S. Ho
- Biology, Lafayette College, Easton, PA, United States
| |
Collapse
|
48
|
Chowdhury ZM, Jamal TB, Ahammad I, Bhattacharjee A, Lamisa AB, Jani JM, Israk MF, Hossain MU, Das KC, Keya CA, Salimullah M. Identification of repurposable drug targets in Mycoplasma pneumoniae using subtractive genomics, molecular docking and dynamics simulation. Heliyon 2023; 9:e21466. [PMID: 38034688 PMCID: PMC10682543 DOI: 10.1016/j.heliyon.2023.e21466] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Revised: 10/05/2023] [Accepted: 10/21/2023] [Indexed: 12/02/2023] Open
Abstract
Mycoplasma pneumoniae is a significant causative agent of community-acquired pneumonia, causing acute inflammation in the upper and lower respiratory tract as well as extrapulmonary syndromes. In particular, the elderly and infants are at greater risk of developing severe, life-threatening pneumonia caused by M. pneumoniae. Yet, the global increase in antimicrobial resistance against antibiotics for the treatment of M. pneumoniae infection highlights the urgent need to explore novel drug targets. To this end, bioinformatics approaches, such as subtractive genomics, can be employed to identify specific metabolic pathways and essential proteins unique to the pathogen that could be potential targets for new drugs. In this study, we implemented a subtractive genomics approach to identify 61 metabolic pathways and 42 essential proteins that are unique to M. pneumoniae. A subsequent screening in the DrugBank database revealed three druggable proteins with similarity to FDA-approved small-molecule drugs, and finally, the compound CHEBI:97093 was identified as a promising novel putative drug target. These findings can provide crucial insights for the development of highly effective drugs that selectively inhibit the pathogen-specific metabolic pathways, leading to better management and treatment of M. pneumoniae infections.
Collapse
Affiliation(s)
- Zeshan Mahmud Chowdhury
- Bioinformatics Division, National Institute of Biotechnology, Ganakbari, Ashulia, Savar, Dhaka, 1349, Bangladesh
| | - Tabassum Binte Jamal
- Bioinformatics Division, National Institute of Biotechnology, Ganakbari, Ashulia, Savar, Dhaka, 1349, Bangladesh
| | - Ishtiaque Ahammad
- Bioinformatics Division, National Institute of Biotechnology, Ganakbari, Ashulia, Savar, Dhaka, 1349, Bangladesh
| | - Arittra Bhattacharjee
- Bioinformatics Division, National Institute of Biotechnology, Ganakbari, Ashulia, Savar, Dhaka, 1349, Bangladesh
| | - Anika Bushra Lamisa
- Bioinformatics Division, National Institute of Biotechnology, Ganakbari, Ashulia, Savar, Dhaka, 1349, Bangladesh
| | - Jannatul Maoa Jani
- Department of Biotechnology and Genetic Engineering, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj, 8100, Bangladesh
| | - Md Fahim Israk
- Department of Biotechnology and Genetic Engineering, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj, 8100, Bangladesh
| | - Mohammad Uzzal Hossain
- Bioinformatics Division, National Institute of Biotechnology, Ganakbari, Ashulia, Savar, Dhaka, 1349, Bangladesh
| | - Keshob Chandra Das
- Molecular Biotechnology Division, National Institute of Biotechnology, Ganakbari, Ashulia, Savar, Dhaka, 1349, Bangladesh
| | - Chaman Ara Keya
- Department of Biochemistry and Microbiology, North South University, Bashundhara, Dhaka, 1229, Bangladesh
| | - Md Salimullah
- Molecular Biotechnology Division, National Institute of Biotechnology, Ganakbari, Ashulia, Savar, Dhaka, 1349, Bangladesh
| |
Collapse
|
49
|
Sun J, Pan L, Li B, Wang H, Yang B, Li W. A Construction Method of Dynamic Protein Interaction Networks by Using Relevant Features of Gene Expression Data. IEEE/ACM TRANSACTIONS ON COMPUTATIONAL BIOLOGY AND BIOINFORMATICS 2023; 20:2790-2801. [PMID: 37030714 DOI: 10.1109/tcbb.2023.3264241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/19/2023]
Abstract
Essential proteins play an important role in various life activities and are considered to be a vital part of the organism. Gene expression data are an important dataset to construct dynamic protein-protein interaction networks (DPIN). The existing methods for the construction of DPINs generally utilize all features (or the features in a cycle) of the gene expression data. However, the features observed from successive time points tend to be highly correlated, and thus there are some redundant and irrelevant features in the gene expression data, which will influence the quality of the constructed network and the predictive performance of essential proteins. To address this problem, we propose a construction method of DPINs by using selected relevant features rather than continuous and periodic features. We adopt an improved unsupervised feature selection method based on Laplacian algorithm to remove irrelevant and redundant features from gene expression data, then integrate the chosen relevant features into the static protein-protein interaction network (SPIN) to construct a more concise and effective DPIN (FS-DPIN). To evaluate the effectiveness of the FS-DPIN, we apply 15 network-based centrality methods on the FS-DPIN and compare the results with those on the SPIN and the existing DPINs. Then the predictive performance of the 15 centrality methods is validated in terms of sensitivity, specificity, positive predictive value, negative predictive value, F-measure, accuracy, Jackknife and AUPRC. The experimental results show that the FS-DPIN is superior to the existing DPINs in the identification accuracy of essential proteins.
Collapse
|
50
|
Ali H, Samad A, Ajmal A, Ali A, Ali I, Danial M, Kamal M, Ullah M, Ullah R, Kalim M. Identification of Drug Targets and Their Inhibitors in Yersinia pestis Strain 91001 through Subtractive Genomics, Machine Learning, and MD Simulation Approaches. Pharmaceuticals (Basel) 2023; 16:1124. [PMID: 37631039 PMCID: PMC10459760 DOI: 10.3390/ph16081124] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Revised: 07/20/2023] [Accepted: 07/28/2023] [Indexed: 08/27/2023] Open
Abstract
Yersinia pestis, the causative agent of plague, is a Gram-negative bacterium. If the plague is not properly treated it can cause rapid death of the host. Bubonic, pneumonic, and septicemic are the three types of plague described. Bubonic plague can progress to septicemic plague, if not diagnosed and treated on time. The mortality rate of pneumonic and septicemic plague is quite high. The symptom-defining disease is the bubo, which is a painful lymph node swelling. Almost 50% of bubonic plague leads to sepsis and death if not treated immediately with antibiotics. The host immune response is slow as compared to other bacterial infections. Clinical isolates of Yersinia pestis revealed resistance to many antibiotics such as tetracycline, spectinomycin, kanamycin, streptomycin, minocycline, chloramphenicol, and sulfonamides. Drug discovery is a time-consuming process. It always takes ten to fifteen years to bring a single drug to the market. In this regard, in silico subtractive proteomics is an accurate, rapid, and cost-effective approach for the discovery of drug targets. An ideal drug target must be essential to the pathogen's survival and must be absent in the host. Machine learning approaches are more accurate as compared to traditional virtual screening. In this study, k-nearest neighbor (kNN) and support vector machine (SVM) were used to predict the active hits against the beta-ketoacyl-ACP synthase III drug target predicted by the subtractive genomics approach. Among the 1012 compounds of the South African Natural Products database, 11 hits were predicted as active. Further, the active hits were docked against the active site of beta-ketoacyl-ACP synthase III. Out of the total 11 active hits, the 3 lowest docking score hits that showed strong interaction with the drug target were shortlisted along with the standard drug and were simulated for 100 ns. The MD simulation revealed that all the shortlisted compounds display stable behavior and the compounds formed stable complexes with the drug target. These compounds may have the potential to inhibit the beta-ketoacyl-ACP synthase III drug target and can help to combat Yersinia pestis-related infections. The dataset and the source codes are freely available on GitHub.
Collapse
Affiliation(s)
- Hamid Ali
- Department of Biosciences, COMSATS University Islamabad, Park Road, Tarlai Kalan, Islamabad 44000, Pakistan
| | - Abdus Samad
- Department of Biochemistry, Abdul Wali Khan University, Mardan 23200, Pakistan; (A.S.); (A.A.); (M.D.); (M.K.)
| | - Amar Ajmal
- Department of Biochemistry, Abdul Wali Khan University, Mardan 23200, Pakistan; (A.S.); (A.A.); (M.D.); (M.K.)
| | - Amjad Ali
- Faculty of Biological Sciences, Department of Biochemistry, Quaid-i-Azam University, Islamabad 45320, Pakistan;
| | - Ijaz Ali
- Centre for Applied Mathematics and Bioinformatics (CAMB), Gulf University for Science and Technology, Hawally 32093, Kuwait;
| | - Muhammad Danial
- Department of Biochemistry, Abdul Wali Khan University, Mardan 23200, Pakistan; (A.S.); (A.A.); (M.D.); (M.K.)
| | - Masroor Kamal
- Department of Biochemistry, Abdul Wali Khan University, Mardan 23200, Pakistan; (A.S.); (A.A.); (M.D.); (M.K.)
| | - Midrar Ullah
- Department of Biotechnology, Shaheed Benazir Bhutto University Sheringal, Dir Upper 18050, Pakistan;
| | - Riaz Ullah
- Department of Pharmacognosy, College of Pharmacy King Saud University, Riyadh 11451, Saudi Arabia;
| | - Muhammad Kalim
- Department of Microbiology and Immunology, Wake Forest School of Medicine, Winston-Salem, NC 27101, USA;
- Houston Methodist Cancer Center/Weill Cornel Medicine, Houston, TX 77030, USA
| |
Collapse
|