1
|
Güngör Ö, Solmaz AE, Karaca E, Akalın T, Bolat E, Akın H. A Rare Case of Lhermitte Duclos Disease Associated with Somatic PTEN and Germline SUFU Variants. CEREBELLUM (LONDON, ENGLAND) 2025; 24:85. [PMID: 40208419 DOI: 10.1007/s12311-025-01835-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 03/27/2025] [Indexed: 04/11/2025]
Abstract
Lhermitte-Duclos disease (LDD) is a rare dysplastic gangliocytoma of the cerebellum, typically manifesting as a hamartomatous lesion in the posterior fossa. Currently, LDD has been only linked to PTEN pathogenic variants, with the PI3K/AKT/mTOR pathway acting as the primary signaling cascade responsible for its pathogenesis. We present a case of LDD in which a novel germline heterozygous splice site variant (c.183-2 A > G) in the SUFU gene and a somatic heterozygous missense variant (c.389 G > A) in the PTEN gene, identified from tumor tissue were detected by targeted next-generation sequencing (NGS). SUFU, a tumor suppressor gene, primarily inhibits the hedgehog (Hh) signaling pathway and furthermore influences the AKT/mTOR pathway. Pathogenic variants in SUFU have been linked to medulloblastoma, and their potential role in LDD remains under investigation. Given that both conditions involve granule cell progenitors and are influenced by impaired Hh signaling, they may share a similar developmental path. This is the first research indicating that SUFU may play a role in the etiology of LDD, despite SUFU variants being associated with several central nervous system malignancies. The SUFU variant was shown to disrupt splicing via Sanger sequencing and gel electrophoresis of RNA extracted from blood. Analysis of DNA from tumor tissue using the TWIST Exome 2.0 Panel revealed de novo pathogenic SUFU (c.183-2 A > G) and PTEN (c.389G > A) variants. This paper establishes an initial link between LDD and germline SUFU along with somatic PTEN variants identified from tumor tissue, providing novel insights into the molecular pathogenesis of this rare condition.
Collapse
Affiliation(s)
- Özge Güngör
- EGE University School of Medicine Medical Genetics, Izmir, Turkey
| | - Aslı Ece Solmaz
- EGE University School of Medicine Medical Genetics, Izmir, Turkey.
| | - Emin Karaca
- EGE University School of Medicine Medical Genetics, Izmir, Turkey
| | - Taner Akalın
- EGE University School of Medicine Medical pathology, Izmir, Turkey
| | - Elif Bolat
- School of Medicine Brain and Neurosurgery, EGE University, Izmir, Turkey
| | - Haluk Akın
- EGE University School of Medicine Medical Genetics, Izmir, Turkey
| |
Collapse
|
2
|
Maurya D, Mittal S, Jena MK, Pathak B. Machine Learning-Driven Quantum Sequencing of Natural and Chemically Modified DNA. ACS APPLIED MATERIALS & INTERFACES 2025; 17:20778-20789. [PMID: 40156522 DOI: 10.1021/acsami.4c22809] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/01/2025]
Abstract
Simultaneous identification of natural and chemically modified DNA nucleotides at molecular resolution remains a pivotal challenge in genomic science. Despite significant advances in current sequencing technologies, the ability to identify subtle changes in natural and chemically modified nucleotides is hindered by structural and configurational complexity. Given the critical role of nucleobase modifications in data storage and personalized medicine, we propose a computational approach using a graphene nanopore coupled with machine learning (ML) to simultaneously recognize both natural and chemically modified nucleotides, exploring a wide range of modifications in the nucleobase, sugar, and phosphate moieties while investigating quantum transport mechanisms to uncover distinct molecular signatures and detailed electronic and orbital insights of the nucleotides. Integrating with the best-fitted model, the graphene nanopore achieves a good classification accuracy of up to 96% for each natural, chemically modified, purine, and pyrimidine nucleotide. Our approach offers a rapid and precise solution for real-time DNA sequencing by decoding natural and chemically modified nucleotides on a single platform.
Collapse
Affiliation(s)
- Dipti Maurya
- Department of Chemistry, Indian Institute of Technology (IIT) Indore, Indore, Madhya Pradesh 453552, India
| | - Sneha Mittal
- Department of Chemistry, Indian Institute of Technology (IIT) Indore, Indore, Madhya Pradesh 453552, India
| | - Milan Kumar Jena
- Department of Chemistry, Indian Institute of Technology (IIT) Indore, Indore, Madhya Pradesh 453552, India
| | - Biswarup Pathak
- Department of Chemistry, Indian Institute of Technology (IIT) Indore, Indore, Madhya Pradesh 453552, India
| |
Collapse
|
3
|
Chu AT, Sze SY, Tse DM, Lai CW, Ng CS, Yu CW, Chung PH, Pang FC, Chung BH, Lo SV, Quan J. Experiences of participants with undiagnosed diseases and hereditary cancers during the initial phase of the Hong Kong genome project: a mixed-methods study. Hum Genomics 2025; 19:36. [PMID: 40188098 PMCID: PMC11972539 DOI: 10.1186/s40246-025-00746-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2025] [Accepted: 03/22/2025] [Indexed: 04/07/2025] Open
Abstract
BACKGROUND The Hong Kong Genome Project (HKGP) is the first population-wide whole genome sequencing (WGS) programme in Hong Kong and aimed to integrate genomic medicine into the healthcare system. Implementing genetic counselling is essential to help participants understand the genetic basis of diseases and guide informed decision making. We assessed participant experiences during the initial HKGP pilot phase that enrolled patients with undiagnosed diseases and hereditary cancers. METHODS Participants were recruited from three partnering centres at public hospitals during June-September 2023. Participant surveys covered four domains: (1) overall satisfaction, (2) informed consent process, (3) genetic counselling, and (4) attitude towards HKGP. Associations with demographic and socioeconomic characteristics were assessed with multivariable logistic regression. Qualitative feedback was collected in focus group interviews. RESULTS Among 422 eligible participants, 341 completed the survey (80.8% response) and five focus group interviews were held (21 participants). We found 89.8% [95% CI: 86.1-92.7] were satisfied with their HKGP experience. Almost all felt that HKGP participation could benefit others (86.8% [95% CI: 82.7-90.0]) and advance genomic research in Hong Kong (88.9% [95% CI: 85.0-91.9]). The survey item with the lowest agreement among respondents was feeling that HKGP participation could improve their/child's medical treatment (73.5% [95% CI: 68.5-78.0]). Those with secondary and tertiary education were less likely to agree genetic counselling was helpful (Odds Ratio [OR]: 0.02 [95% CI: 0.001-0.41]; 0.02 [0.001-0.51]), or the appropriate length of time (OR: 0.12 [95% CI: 0.014-0.81]; 0.11 [0.01-0.91]). Focus group participants cited helping scientific advances and shortening the diagnostic odyssey of future patients as key reasons for participation. Participants hoped for a shorter reporting time of WGS results, additional medical follow-up, and allowing referral of relatives. CONCLUSIONS Participants were overall highly satisfied with the HKGP and genetic counselling experience. Satisfaction levels were comparable to overseas genomic programmes and locally provided healthcare services. Participants' major concerns on WGS reporting time could be addressed by strengthening the informed consent process to ensure their expectations align with project implementation. Emphasizing the long-term value of genomic research and its potential for personalized treatments may increase participant engagement.
Collapse
Affiliation(s)
- Annie Tw Chu
- Hong Kong Genome Institute, Hong Kong SAR, China
| | - Samuel Yc Sze
- School of Public Health, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | | | - Cheryl Wy Lai
- School of Public Health, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Carmen S Ng
- School of Public Health, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Coco Ws Yu
- School of Public Health, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Pui-Hong Chung
- School of Public Health, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | | | - Brian Hy Chung
- Hong Kong Genome Institute, Hong Kong SAR, China.
- Department of Paediatrics and Adolescent Medicine, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China.
| | - Su-Vui Lo
- Hong Kong Genome Institute, Hong Kong SAR, China.
| | - Jianchao Quan
- School of Public Health, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China.
- HKU Business School, The University of Hong Kong SAR, Hong Kong SAR, China.
| |
Collapse
|
4
|
Westphalen CB, Boscolo Bielo L, Aftimos P, Beltran H, Benary M, Chakravarty D, Collienne M, Dienstmann R, El Helali A, Gainor J, Horak P, Le Tourneau C, Marchiò C, Massard C, Meric-Bernstam F, Pauli C, Pruneri G, Roitberg F, Russnes HEG, Solit DB, Starling N, Subbiah V, Tamborero D, Tarazona N, Turnbull C, van de Haar J, André F, Mateo J, Curigliano G. ESMO Precision Oncology Working Group recommendations on the structure and quality indicators for molecular tumour boards in clinical practice. Ann Oncol 2025:S0923-7534(25)00080-8. [PMID: 40194904 DOI: 10.1016/j.annonc.2025.02.009] [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: 12/20/2024] [Revised: 02/17/2025] [Accepted: 02/23/2025] [Indexed: 04/09/2025] Open
Abstract
BACKGROUND With an increased uptake of genomic profiling in clinical practice and the evolving complexity of diagnostic modalities, vast amounts of complex data need to be properly interpreted and integrated into an individualised care plan. To address these challenges, molecular tumour boards (MTBs) have been widely established. As of today, no international recommendations regulating the composition and workflows of MTBs have been defined. METHODS ESMO's Precision Oncology Working Group (POWG) established an international expert panel in precision oncology and defined core areas of interest. After several consultations and through an expert consensus process, the group reached a consensus level for each recommendation. RESULTS The group defined five components in the MTB process that are critical to its function and clinical use: (i) the primary task of MTBs consists in providing genomic-informed clinical recommendations, particularly for cases exhibiting complex genomic alterations; (ii) to achieve this, MTBs should encompass interdisciplinary expertise, with key roles for oncologists with genomic expertise, pathologists with molecular training and clinical geneticists; (iii) MTBs' recommendations should be documented in a structured report that includes genomic-informed treatment strategies, management plans for potential tumour-detected germline alterations and guidance for additional genomic testing; (iv) structured follow-up processes should be implemented for monitoring the clinical effectiveness of MTBs recommendations and (v) finally, the panel proposed quality indicators for operating MTBs, including turnaround times for cases discussion and the proportion of cases for which actionable recommendations and clinical trial enrolments were successfully implemented. CONCLUSIONS These ESMO's POWG recommendations can serve as a guidance and help to define quality standards for MTBs to allow for harmonisation and to further expedite the integration of precision oncology into clinical practice.
Collapse
Affiliation(s)
- C B Westphalen
- Comprehensive Cancer Center Munich & Department of Medicine III, University Hospital, LMU Munich, Munich; German Cancer Consortium (DKTK), partner site Munich, German Cancer Research Center (DKFZ), Heidelberg, Germany.
| | - L Boscolo Bielo
- Division of New Drugs and Early Drug Development for Innovative Therapies, European Institute of Oncology, IRCCS, Milan, Italy; Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - P Aftimos
- Clinical Trials Conduct Unit, Institut Jules Bordet, Hôpital Universitaire de Bruxelles (HUB), Brussels, Belgium
| | - H Beltran
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, USA; Harvard Medical School, Boston, USA
| | - M Benary
- Charité Comprehensive Cancer Center, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany; Core Unit Bioinformatics, Berlin Institute of Health at Charité-Universitätsmedizin Berlin, Berlin, Germany; German Cancer Consortium (DKTK), partner site Berlin, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - D Chakravarty
- Kravis Center for Molecular Oncology, Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, USA
| | - M Collienne
- DKFZ-Hector Cancer Institute at the University Medical Center Mannheim, Department of Personalized Oncology, University Hospital Mannheim, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany; Division of Personalized Medical Oncology (A420), German Cancer Research Center (DKFZ), German Center for Lung Research (DZL), Heidelberg, Germany
| | - R Dienstmann
- Vall d'Hebron Institute of Oncology (VHIO), Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain; University of Vic - Central University of Catalonia, Vic, Spain; Oncoclínicas&Co, São Paulo, Brazil
| | - A El Helali
- Department of Clinical Oncology, School of Clinical Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR
| | - J Gainor
- Harvard Medical School, Boston, USA; Massachusetts General Hospital, Boston, USA
| | - P Horak
- Department of Translational Medical Oncology, National Center for Tumor Diseases (NCT) Heidelberg, a partnership between DKFZ and Heidelberg University Hospital, Heidelberg, Germany; German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - C Le Tourneau
- Department of Drug Development & Innovation (D3i), Institut Curie, Paris, France; Faculty of Medicine, Paris-Saclay University, Paris, France
| | - C Marchiò
- Department of Medical Sciences, University of Turin, Turin, Italy; Division of Pathology, Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Italy
| | - C Massard
- DITEP Department, Gustave Roussy, Villejuif, France
| | - F Meric-Bernstam
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, USA
| | - C Pauli
- Department of Pathology and Molecular Pathology, University Hospital Zurich, Zurich, Switzerland; Medical Faculty, University of Zurich, Zurich, Switzerland
| | - G Pruneri
- Department of Advanced Diagnostics, Fondazione IRCCS, Istituto Nazionale Tumori, Milan, Italy; School of Medicine, University of Milan, Milan, Italy
| | - F Roitberg
- Research and Innovation Branch, Ebserh, Empresa Brasileira de Servicos Hospitalares, Brasilia, Brasil
| | - H E G Russnes
- Department of Pathology, Department of Cancer Genetics, Institute for Cancer Research, Oslo University Hospital, Radiumhospitalet, Oslo, Norway; Institute for Clinical Medicine, University of Oslo, Oslo, Norway
| | - D B Solit
- Department of Medicine, Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, USA
| | - N Starling
- Department of Medical Oncology (Gastrointestinal Unit), The Royal Marsden NHS Foundation Trust, London, UK
| | - V Subbiah
- Early-Phase Drug Development, Sarah Cannon Research Institute (SCRI), Nashville, USA
| | - D Tamborero
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| | - N Tarazona
- Department of Medical Oncology, INCLIVA Biomedical Research Institute, University of Valencia, Valencia, Spain; Instituto de Salud Carlos III, CIBERONC, Madrid, Spain
| | - C Turnbull
- Division of Genetics and Epidemiology, The Institute of Cancer Research, London, UK; The Royal Marsden NHS Foundation Trust, London, UK
| | - J van de Haar
- Department of Molecular Oncology & Immunology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - F André
- INSERM U981, Gustave Roussy, Villejuif, France; Department of Cancer Medicine, Gustave Roussy, Villejuif, France; Faculty of Medicine, Université Paris-Saclay, Kremlin Bicêtre, France
| | - J Mateo
- Vall d'Hebron Institute of Oncology (VHIO), Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain
| | - G Curigliano
- Division of New Drugs and Early Drug Development for Innovative Therapies, European Institute of Oncology, IRCCS, Milan, Italy; Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy.
| |
Collapse
|
5
|
Pipitò L, Trizzino M, Mascarella C, Cannella S, Gaudiano R, Ganci I, D'Alessandro G, Romanin B, Santoro MM, Giammanco GM, Cascio A, Bonura C. Next-generation sequencing and drug resistance mutations of HIV-1 subtypes in people living with HIV in Sicily, Italy, 2021-2023. J Glob Antimicrob Resist 2025; 41:68-76. [PMID: 39733833 DOI: 10.1016/j.jgar.2024.12.015] [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: 07/29/2024] [Revised: 12/11/2024] [Accepted: 12/15/2024] [Indexed: 12/31/2024] Open
Abstract
OBJECTIVES HIV-1 infection continues to be a significant public health concern, notwithstanding the expanded utilization of antiretroviral treatment (ART), due to the emergence of drug resistance. The prevalence of transmitted drug resistance remains uncertain, particularly concerning integrase inhibitors. This study aimed to assess the extent of HIV resistance in both ART-naïve and experienced individuals living with HIV (PLHIV) at the University Hospital in Palermo, Italy. METHODS Genotyping and mutation analysis were performed on ART naïve and experienced PLHIV admitted from June 2021 to October 2023 by the NGS method. Mutations were detected by testing different NGS frequency cut-offs: ≥5 %, ≥10 %, and ≥20 %. Demographic, clinical, virological, and immunological data were retrospectively collected. RESULTS Of the PLHIV, 85 (70 %) were ART-naïve, while 36 (30 %) were ART-experienced with virological failure. The main HIV-1 subtype was B (54 %), which was significantly associated with Italy-born (P < 0.001) and experienced PLHIV (P = 0.024). In the remaining cases, A1 (6 %), C (3 %), F1 (7 %), G (2 %), and Circulating Recombinant Forms (28 %) were reported. At least one mutation for a drug class was detected in 39.7 %, 45.4 %, and 53.7 % of cases at HIV-1 NGS thresholds of 20 %, 10 %, and 5 %, respectively. Drug resistance was found in 18.2 %, 25.6 %, and 33.0 %, by NGS cut-off of 20 %, 10 %, and 5 % respectively. The lowering of NGS cut-offs mainly increased the rates of integrase strand transfer inhibitor resistance. For overall resistance, no difference was observed between B and non-B subtypes for any NGS cut-offs.
Collapse
Affiliation(s)
- Luca Pipitò
- Department of Health Promotion, Mother and Child Care, Internal Medicine, and Medical Specialties "G D'Alessandro," University of Palermo, Palermo, Italy; Infectious and Tropical Diseases Unit and Sicilian Regional Reference Center for the fight against AIDS, AOU Policlinico "P. Giaccone," Palermo, Italy; Palermo Fast-Track City, Casa dei Diritti, Palermo, Italy
| | - Marcello Trizzino
- Infectious and Tropical Diseases Unit and Sicilian Regional Reference Center for the fight against AIDS, AOU Policlinico "P. Giaccone," Palermo, Italy; Palermo Fast-Track City, Casa dei Diritti, Palermo, Italy
| | - Chiara Mascarella
- Department of Health Promotion, Mother and Child Care, Internal Medicine, and Medical Specialties "G D'Alessandro," University of Palermo, Palermo, Italy; Microbiology and Virology Unit, AOU Policlinico "P. Giaccone", Palermo, Italy
| | - Sara Cannella
- Department of Health Promotion, Mother and Child Care, Internal Medicine, and Medical Specialties "G D'Alessandro," University of Palermo, Palermo, Italy; Microbiology and Virology Unit, AOU Policlinico "P. Giaccone", Palermo, Italy
| | - Roberta Gaudiano
- Department of Health Promotion, Mother and Child Care, Internal Medicine, and Medical Specialties "G D'Alessandro," University of Palermo, Palermo, Italy; Infectious and Tropical Diseases Unit and Sicilian Regional Reference Center for the fight against AIDS, AOU Policlinico "P. Giaccone," Palermo, Italy; Palermo Fast-Track City, Casa dei Diritti, Palermo, Italy
| | - Irene Ganci
- Department of Health Promotion, Mother and Child Care, Internal Medicine, and Medical Specialties "G D'Alessandro," University of Palermo, Palermo, Italy; Infectious and Tropical Diseases Unit and Sicilian Regional Reference Center for the fight against AIDS, AOU Policlinico "P. Giaccone," Palermo, Italy; Palermo Fast-Track City, Casa dei Diritti, Palermo, Italy
| | - Gaetano D'Alessandro
- Department of Health Promotion, Mother and Child Care, Internal Medicine, and Medical Specialties "G D'Alessandro," University of Palermo, Palermo, Italy; Microbiology and Virology Unit, AOU Policlinico "P. Giaccone", Palermo, Italy
| | - Benedetta Romanin
- Infectious and Tropical Diseases Unit and Sicilian Regional Reference Center for the fight against AIDS, AOU Policlinico "P. Giaccone," Palermo, Italy; Palermo Fast-Track City, Casa dei Diritti, Palermo, Italy
| | | | - Giovanni M Giammanco
- Department of Health Promotion, Mother and Child Care, Internal Medicine, and Medical Specialties "G D'Alessandro," University of Palermo, Palermo, Italy; Microbiology and Virology Unit, AOU Policlinico "P. Giaccone", Palermo, Italy
| | - Antonio Cascio
- Department of Health Promotion, Mother and Child Care, Internal Medicine, and Medical Specialties "G D'Alessandro," University of Palermo, Palermo, Italy; Infectious and Tropical Diseases Unit and Sicilian Regional Reference Center for the fight against AIDS, AOU Policlinico "P. Giaccone," Palermo, Italy; Palermo Fast-Track City, Casa dei Diritti, Palermo, Italy.
| | - Celestino Bonura
- Department of Health Promotion, Mother and Child Care, Internal Medicine, and Medical Specialties "G D'Alessandro," University of Palermo, Palermo, Italy; Microbiology and Virology Unit, AOU Policlinico "P. Giaccone", Palermo, Italy
| |
Collapse
|
6
|
Luo R, Liu J, Guan L, Li M. HybProm: An attention-assisted hybrid CNN-BiLSTM model for the interpretable prediction of DNA promoter. Methods 2025; 235:71-80. [PMID: 39929298 DOI: 10.1016/j.ymeth.2025.02.001] [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: 12/21/2024] [Revised: 01/18/2025] [Accepted: 02/03/2025] [Indexed: 02/13/2025] Open
Abstract
Promoter prediction is essential for analyzing gene structures, understanding regulatory networks, transcription mechanisms, and precisely controlling gene expression. Recently, computational and deep learning methods for promoter prediction have gained attention. However, there is still room to improve their accuracy. To address this, we propose the HybProm model, which uses DNA2Vec to transform DNA sequences into low-dimensional vectors, followed by a CNN-BiLSTM-Attention architecture to extract features and predict promoters across species, including E. coli, humans, mice, and plants. Experiments show that HybProm consistently achieves high accuracy (90%-99%) and offers good interpretability by identifying key sequence patterns and positions that drive predictions.
Collapse
Affiliation(s)
- Rentao Luo
- College of Physics and Electronic Information, Gannan Normal University, Ganzhou 341000 Jiangxi, China
| | - Jiawei Liu
- College of Physics and Electronic Information, Gannan Normal University, Ganzhou 341000 Jiangxi, China
| | - Lixin Guan
- College of Physics and Electronic Information, Gannan Normal University, Ganzhou 341000 Jiangxi, China
| | - Mengshan Li
- College of Physics and Electronic Information, Gannan Normal University, Ganzhou 341000 Jiangxi, China.
| |
Collapse
|
7
|
Backmann L, Umberath KM, Wegmann-Herr P, Weber F, Jürgens A, Scharfenberger-Schmeer M. Putting Laccase Gene Differences on Genomic Level into Context: An Analysis of Botrytis cinerea Strains from Grapes. Microorganisms 2025; 13:483. [PMID: 40142375 PMCID: PMC11945579 DOI: 10.3390/microorganisms13030483] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2025] [Revised: 02/10/2025] [Accepted: 02/14/2025] [Indexed: 03/28/2025] Open
Abstract
One of the most important crop pathogens is Botrytis cinerea. It overcomes plant defenses using laccase, an enzyme which is frequently researched. Yet the differences between strains regarding their laccase activity is poorly understood. The aim of this study was to analyze laccase genes in the context of the regionality, vintage, and laccase activity of the strains. Eight strains were analyzed using whole genome sequencing, and the laccase activity was assessed. The strains were differentiated by SSR-PCR. We looked at all 14 known laccase genome regions as well as the promoter and terminator regions using variant metrics and phylogenetic trees. The laccase genes seem to be correlated with the regionality of the strains rather than the laccase activity, which provides new understanding to the study of pathogen adaption in specific environments. Some of the laccase gene regions showed little to no evolutionary change, while other regions showed a great variety of changes. This research highlights taking different laccase gene regions into context. We provide fundamental information for further research. Further studies, especially on gene expression, could provide insightful information regarding the potential of pathogen infection.
Collapse
Affiliation(s)
- Louis Backmann
- Department of Biology, Chemical Plant Ecology, Technische Universität Darmstadt, Schnittspahnstrasse 4, D-64287 Darmstadt, Germany; (L.B.); (A.J.)
- Institute for Viticulture and Oenology, Dienstleistungszentrum Ländlicher Raum (DLR) Rheinpfalz, Breitenweg 71, D-67435 Neustadt, Germany;
| | - Kim Marie Umberath
- Institute of Nutritional and Food Sciences, Molecular Food Technology, Agricultural Faculty, University of Bonn, Friedrich-Hirzebruch-Allee 7, D-53115 Bonn, Germany;
| | - Pascal Wegmann-Herr
- Institute for Viticulture and Oenology, Dienstleistungszentrum Ländlicher Raum (DLR) Rheinpfalz, Breitenweg 71, D-67435 Neustadt, Germany;
| | - Fabian Weber
- Section of Organic Food Quality, University of Kassel, Nordbahnhofstr. 1a, D-37213 Witzenhausen, Germany;
| | - Andreas Jürgens
- Department of Biology, Chemical Plant Ecology, Technische Universität Darmstadt, Schnittspahnstrasse 4, D-64287 Darmstadt, Germany; (L.B.); (A.J.)
| | - Maren Scharfenberger-Schmeer
- Institute for Viticulture and Oenology, Dienstleistungszentrum Ländlicher Raum (DLR) Rheinpfalz, Breitenweg 71, D-67435 Neustadt, Germany;
- Weincampus Neustadt, Hochschule Kaiserslautern, Breitenweg 71, D-67435 Neustadt, Germany
| |
Collapse
|
8
|
Tsai MH, Chen CH, Chen CL, Lee MH, Wu LC, Hsu YC, Hsiao CY, Lee CT, Pi KL, Su LJ. Areca catechu L. Extract Inhibits Colorectal Cancer Tumor Growth by Modulating Cell Apoptosis and Autophagy. Curr Issues Mol Biol 2025; 47:128. [PMID: 39996849 PMCID: PMC11854706 DOI: 10.3390/cimb47020128] [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: 12/25/2024] [Revised: 02/06/2025] [Accepted: 02/13/2025] [Indexed: 02/26/2025] Open
Abstract
Colorectal cancer (CRC) is a common cancer globally, and chemotherapy often causes severe complications, necessitating effective drugs with minimal side effects. As Areca catechu L. extract (ACE) is a Traditional Chinese Medicine that contains numerous active compounds with anticancer effects, in this study, the Cell Counting Kit-8 (CCK-8) assay was used to determine ACE's effect on CRC cell lines, revealing that it significantly inhibits CoLo320DM and HCT116 cells. In vivo experiments with NU-Foxn1nu mice indicated that ACE inhibits tumor growth, while a flow cytometry assay revealed that higher ACE concentrations increased cell apoptosis and ROS levels. Next-generation sequencing (NGS) showed that ACE increases the fold changes in apoptosis, DNA damage, and autophagy-related genes while inhibiting the fold changes in cell proliferation and Wnt signaling pathway genes. We conducted Western blotting to confirm these findings. Overall, ACE demonstrates potential as a drug candidate by promoting apoptosis and autophagy, and significantly reducing cell viability and tumor growth, thus offering a new approach for effective colorectal cancer treatment with minimal side effects.
Collapse
Affiliation(s)
- Meng-Hsiu Tsai
- Department of Biomedical Science and Engineering, National Central University, Taoyuan 320317, Taiwan; (M.-H.T.)
| | - Chang-Han Chen
- Department of Applied Chemistry, Graduate Institute of Biomedicine and Biomedical Technology, National Chi Nan University, Nantou County 345301, Taiwan
- Department of Medical Research, Taichung Veterans General Hospital, Taichung 407219, Taiwan
| | | | - Mei-Hsien Lee
- Graduated Institute of Pharmacognosy, Taipei Medical University, Taipei 110301, Taiwan
| | - Li-Ching Wu
- Department of Biomedical Science and Engineering, National Central University, Taoyuan 320317, Taiwan; (M.-H.T.)
| | - Yi-Chiung Hsu
- Department of Biomedical Science and Engineering, National Central University, Taoyuan 320317, Taiwan; (M.-H.T.)
| | - Chao-Yang Hsiao
- Department of Biomedical Science and Engineering, National Central University, Taoyuan 320317, Taiwan; (M.-H.T.)
- Division of Rheumatology, Allergy and Immunology, Department of Internal Medicine, Chang Gung Memorial Hospital, Taoyuan 333423, Taiwan
| | - Chang-Ti Lee
- Department of Biomedical Science and Engineering, National Central University, Taoyuan 320317, Taiwan; (M.-H.T.)
- Department of Chinese Medicine, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei 231016, Taiwan
| | - Kuo-Li Pi
- Graduate Institute of History, National Central University, Taoyuan 320317, Taiwan
| | - Li-Jen Su
- Department of Biomedical Science and Engineering, National Central University, Taoyuan 320317, Taiwan; (M.-H.T.)
- IHMED Reproductive Center, Taipei 106028, Taiwan
- Education and Research Center for Technology Assisted Substance Abuse Prevention and Management, National Central University, Taoyuan 320317, Taiwan
- Core Facilities for High Throughput Experimental Analysis, Department of Biomedical Science and Engineering, National Central University, Taoyuan 320317, Taiwan
| |
Collapse
|
9
|
Smart K, Pieper JB, Viall AK, Noxon JO, Berger DJ. Comparison of commercial next-generation sequencing assays to conventional culture methods for bacterial identification and antimicrobial susceptibility of samples obtained from clinical cases of canine superficial bacterial folliculitis. Vet Dermatol 2025; 36:14-23. [PMID: 39323044 PMCID: PMC11696477 DOI: 10.1111/vde.13299] [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: 02/15/2024] [Revised: 04/19/2024] [Accepted: 09/10/2024] [Indexed: 09/27/2024]
Abstract
BACKGROUND Bacterial identification and antimicrobial susceptibility testing is an important step in timely therapeutic decisions for canine superficial bacterial folliculitis (SBF), commonly caused by Staphylococcus pseudintermedius. Next-generation sequencing (NGS) offers the appeal of potentially expedited results with complete detection of bacterial organisms and associated resistance genes compared to culture. Limited studies exist comparing the two methodologies for clinical samples. HYPOTHESIS/OBJECTIVES To compare and contrast genotypic and phenotypic methods for bacterial identification and antimicrobial susceptibility from cases of canine SBF. ANIMALS Twenty-four client-owned dogs with lesions consistent with SBF were enrolled. MATERIALS AND METHODS A sterile culturette swab was used to sample dogs with SBF lesions. The swab was rinsed in 0.9 mL of sterile phosphate-buffered saline and vortexed to create a homogenous solution. Two swabs for NGS laboratories (Labs) and one swab for culture (Culture Lab) were randomly sampled from this solution and submitted for bacterial identification and antimicrobial susceptibility. RESULTS No statistical difference regarding turnaround time for NGS Labs compared to Culture Lab was found. NGS Lab 1 identified more organisms than NGS Lab 2 and Culture Lab, which were both statistically significant. There was no statistical difference in detection frequency for Staphylococcus spp. among all laboratories. There was poor agreement for the presence of meticillin resistance and most antimicrobials among all laboratories. CONCLUSIONS AND CLINICAL RELEVANCE Utilisation of NGS as a replacement for traditional culture when sampling canine SBF lesions is not supported at this time.
Collapse
Affiliation(s)
- Kimberly Smart
- Department of Veterinary Clinical SciencesIowa State UniversityAmesIowaUSA
| | - Jason B. Pieper
- Department of Veterinary Clinical SciencesIowa State UniversityAmesIowaUSA
| | - Austin K. Viall
- Department of Pathology, Microbiology, and ImmunologyUniversity of California DavisDavisCaliforniaUSA
| | - James O. Noxon
- Department of Veterinary Clinical SciencesIowa State UniversityAmesIowaUSA
| | - Darren J. Berger
- Department of Veterinary Clinical SciencesIowa State UniversityAmesIowaUSA
| |
Collapse
|
10
|
Cox M, Vitello DJ, Chawla A. The Current Role of Circulating Tumor DNA in the Management of Pancreatic Cancer. J Gastrointest Cancer 2025; 56:44. [PMID: 39808248 DOI: 10.1007/s12029-024-01129-0] [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] [Accepted: 10/23/2024] [Indexed: 01/16/2025]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is projected to be the second leading cause of cancer-related death by 2030. Early identification is rare, with a 5-year overall survival (OS) of less than 10%. Advances in the understanding of PDAC tumor biology are needed to improve these outcomes. Circulating tumor DNA (ctDNA) represents a promising novel biomarker in the identification and management of PDAC. Drawn from peripheral blood and analyzed using a variety of techniques, the detection of ctDNA in PDAC has been associated with shorter OS, minimal residual disease presence, and shorter recurrence-free survival. The use of ctDNA has also been examined as an indicator of therapeutic resistance, susceptibility to targeted therapy, and therapeutic response. While promising, ctDNA analysis is limited by its low rates of detection in some settings and lack of predictive ability in others. Many studies examining the utility of ctDNA for the management of PDAC have been relatively small retrospective cohort studies. The current findings will need to be validated by incorporation of ctDNA analysis into cancer registries and larger prospective studies. Given the current, rapid evolution in the field, it is possible that with time, ctDNA will be more routinely incorporated into the clinical management of PDAC.
Collapse
Affiliation(s)
- Madison Cox
- Division of Surgical Oncology, Department of Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Northwestern Medicine Cancer Centers, Northwestern Medicine Regional Medical Group, Winfield, IL, USA
| | - Dominic J Vitello
- Division of Surgical Oncology, Department of Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Northwestern Quality Improvement, Research and Education in Surgery, Department of Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Akhil Chawla
- Division of Surgical Oncology, Department of Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL, USA.
- Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, IL, USA.
- Northwestern Medicine Cancer Centers, Northwestern Medicine Regional Medical Group, Winfield, IL, USA.
- Northwestern Quality Improvement, Research and Education in Surgery, Department of Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL, USA.
| |
Collapse
|
11
|
Lanzillotti MB, Brodbelt JS. Progress in Tandem Mass Spectrometry Data Analysis for Nucleic Acids. MASS SPECTROMETRY REVIEWS 2025. [PMID: 39797409 DOI: 10.1002/mas.21923] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Revised: 12/17/2024] [Accepted: 12/20/2024] [Indexed: 01/13/2025]
Abstract
Mass spectrometry (MS) has become a critical tool in the characterization of covalently modified nucleic acids. Well-developed bottom-up approaches, where nucleic acids are digested with an endonuclease and the resulting oligonucleotides are separated before MS and MS/MS analysis, provide substantial insight into modified nucleotides in biological and synthetic nucleic. Top-down MS presents an alternative approach where the entire nucleic acid molecule is introduced to the mass spectrometer intact and then fragmented by MS/MS. Current top-down MS workflows have incorporated automated, on-line HPLC workflows to enable rapid desalting of nucleic acid samples for facile mass analysis without complication from adduction. Furthermore, optimization of MS/MS parameters utilizing collision, electron, or photon-based activation methods have enabled effective bond cleavage throughout the phosphodiester backbone while limiting secondary fragmentation, allowing characterization of progressively larger (~100 nt) nucleic acids and localization of covalent modifications. Development of software applications to perform automated identification of fragment ions has accelerated the broader adoption of mass spectrometry for analysis of nucleic acids. This review focuses on progress in tandem mass spectrometry for characterization of nucleic acids with particular emphasis on the software tools that have proven critical for advancing the field.
Collapse
|
12
|
Li L, Xiao H, Wu X, Tang Z, Khoury JD, Wang J, Wan S. RanBALL: An Ensemble Random Projection Model for Identifying Subtypes of B-Cell Acute Lymphoblastic Leukemia. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.09.24.614777. [PMID: 39386448 PMCID: PMC11463541 DOI: 10.1101/2024.09.24.614777] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/12/2024]
Abstract
As the most common pediatric malignancy, B-cell acute lymphoblastic leukemia (B-ALL) has multiple distinct subtypes characterized by recurrent and sporadic somatic and germline genetic alterations. Identifying B-ALL subtypes can facilitate risk stratification and enable tailored therapeutic design. Existing methods for B-ALL subtyping primarily depend on immunophenotyping, cytogenetic tests and genomic profiling, which would be costly, complicated, and laborious. To overcome these challenges, we present RanBALL (an ensemble Random projection-based model for identifying B-ALL subtypes), an accurate and cost-effective model for B-ALL subtype identification. By leveraging random projection (RP) and ensemble learning, RanBALL can preserve patient-to-patient distances after dimension reduction and yield robustly accurate classification performance for B-ALL subtyping. Benchmarking results based on > 1700 B-ALL patients demonstrated that RanBALL achieved remarkable performance (accuracy: 0.93, F1-score: 0.93, and Matthews correlation coefficient: 0.93), significantly outperforming state-of-the-art methods like ALLSorts in terms of all performance metrics. In addition, RanBALL performs better than tSNE in terms of visualizing B-ALL subtype information. We believe RanBALL will facilitate the discovery of B-ALL subtype-specific marker genes and therapeutic targets to have consequential positive impacts on downstream risk stratification and tailored treatment design. To extend its applicability and impacts, a Python-based RanBALL package is available at https://github.com/wan-mlab/RanBALL.
Collapse
Affiliation(s)
- Lusheng Li
- Department of Genetics, Cell Biology and Anatomy, University of Nebraska Medical Center, Omaha, NE, USA
| | - Hanyu Xiao
- Department of Genetics, Cell Biology and Anatomy, University of Nebraska Medical Center, Omaha, NE, USA
| | - Xinchao Wu
- Department of Genetics, Cell Biology and Anatomy, University of Nebraska Medical Center, Omaha, NE, USA
| | - Zhenya Tang
- Department of Pathology, Microbiology and Immunology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Joseph D. Khoury
- Department of Pathology, Microbiology and Immunology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Jieqiong Wang
- Department of Neurological Sciences, University of Nebraska Medical Center, Omaha, NE, USA
| | - Shibiao Wan
- Department of Genetics, Cell Biology and Anatomy, University of Nebraska Medical Center, Omaha, NE, USA
| |
Collapse
|
13
|
Shrestha S, Addae A, Miller C, Ismail N, Zwerling A. Cost-effectiveness of targeted next-generation sequencing (tNGS) for detection of tuberculosis drug resistance in India, South Africa and Georgia: a modeling analysis. EClinicalMedicine 2025; 79:103003. [PMID: 39810935 PMCID: PMC11732181 DOI: 10.1016/j.eclinm.2024.103003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Revised: 10/23/2024] [Accepted: 11/27/2024] [Indexed: 01/16/2025] Open
Abstract
Background Targeted next-generation sequencing (tNGS) is promising alternative to phenotypic drug susceptibility testing (pDST) for detecting drug-resistant tuberculosis (DRTB). This study explored the potential cost-effectiveness of tNGS for the diagnosis of DR-TB across 3 settings: India, South Africa and Georgia. Methods To inform WHO guideline development group (GDG) on tNGS we developed a stochastic decision analysis model and assessed cost-effectiveness of tNGS for DST among rifampicin resistance individuals. We also assessed tNGS as initial test for TB drug resistance in bacteriologically confirmed TB. Diagnostic accuracy and cost data were sourced from a systematic review conducted for GDG, covering studies published until September 2022. The primary outcome was incremental cost (2021 US$) per disability-adjusted life year (DALY) averted. Findings tNGS when compared with in-country DST, tNGS proved cost-effective in South Africa (ICER: $15,619/DALY averted, WTP: $21,165) but not in Georgia (ICER: $18,375/DALY averted, WTP: $15,069). In India, tNGS dominated in-country DST practice, providing greater health impact at lower cost. When comparing tNGS with universal pDST, tNGS was dominated by pDST in all three countries. In Georgia, using tNGS as initial test for TB drug-resistance compared to Xpert MTB/Rif followed by pDST appeared cost-effective. Scenario with 50% reduction in tNGS test kit costs made tNGS cost-effective across all three countries, while a high Bedaquiline resistance prevalence (30%) led to a worsening cost-effectiveness. Interpretation tNGS may be cost-effective in India, South Africa and Georgia when comprehensive DST is not routinely performed. Thus, existing DST practice and healthcare infrastructure should be considered before implementation and scale-up of tNGS. Funding Global Tuberculosis Program, World Health Organization (2022/1249364-0).
Collapse
Affiliation(s)
- Suvesh Shrestha
- School of Epidemiology and Public Health, University of Ottawa, Ottawa, Canada
| | - Angelina Addae
- School of Epidemiology and Public Health, University of Ottawa, Ottawa, Canada
| | - Cecily Miller
- Global Tuberculosis Program, World Health Organization, Geneva, Switzerland
| | - Nazir Ismail
- Global Tuberculosis Program, World Health Organization, Geneva, Switzerland
- Department of Clinical Microbiology and Infectious Diseases, Faculty of Health Sciences, Wits University, Johannesburg, South Africa
| | - Alice Zwerling
- School of Epidemiology and Public Health, University of Ottawa, Ottawa, Canada
| |
Collapse
|
14
|
Lutz S, D'Angelo A, Hammerl S, Schmutz M, Claus R, Fischer NM, Kramer F, Hammoud Z. Unveiling the Digital Evolution of Molecular Tumor Boards. Target Oncol 2025; 20:27-43. [PMID: 39609355 PMCID: PMC11762447 DOI: 10.1007/s11523-024-01109-1] [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] [Accepted: 10/08/2024] [Indexed: 11/30/2024]
Abstract
Molecular tumor boards (MTB) are interdisciplinary conferences involving various experts discussing patients with advanced tumors, to derive individualized treatment suggestions based on molecular variants. These discussions involve using heterogeneous internal data, such as patient clinical data, but also external resources such as knowledge databases for annotations and search for relevant clinical studies. This imposes a certain level of complexity that requires huge effort to homogenize the data and use it in a speedy manner to reach the needed treatment. For this purpose, most institutions involving an MTB are heading toward automation and digitalization of the process, hence reducing manual work requiring human intervention and subsequently time in deriving personalized treatment suggestions. The tools are also used to better visualize the patient's data, which allows a refined overview for the board members. In this paper, we present the results of our thorough literature research about MTBs, their process, the most common knowledge bases, and tools used to support this decision-making process.
Collapse
Affiliation(s)
- Sebastian Lutz
- IT-Infrastructure for Translational Medical Research, University of Augsburg, Augsburg, Germany.
| | - Alicia D'Angelo
- IT-Infrastructure for Translational Medical Research, University of Augsburg, Augsburg, Germany
| | - Sonja Hammerl
- IT-Infrastructure for Translational Medical Research, University of Augsburg, Augsburg, Germany
| | - Maximilian Schmutz
- Institute of Digital Medicine (IDM), Medical Faculty, University of Augsburg, Augsburg, Germany
- Hematology and Oncology, Faculty of Medicine, University of Augsburg, Augsburg, Germany
- Bavarian Cancer Research Center (BZKF), Augsburg, Germany
| | - Rainer Claus
- Hematology and Oncology, Faculty of Medicine, University of Augsburg, Augsburg, Germany
- Bavarian Cancer Research Center (BZKF), Augsburg, Germany
- Pathology, Faculty of Medicine, University of Augsburg, Augsburg, Germany
- Comprehensive Cancer Center Augsburg (CCCA), Faculty of Medicine, University of Augsburg, Augsburg, Germany
| | - Nina M Fischer
- Bavarian Cancer Research Center (BZKF), Augsburg, Germany
- Comprehensive Cancer Center Augsburg (CCCA), Faculty of Medicine, University of Augsburg, Augsburg, Germany
| | - Frank Kramer
- IT-Infrastructure for Translational Medical Research, University of Augsburg, Augsburg, Germany
| | - Zaynab Hammoud
- IT-Infrastructure for Translational Medical Research, University of Augsburg, Augsburg, Germany
| |
Collapse
|
15
|
Kalaignazhal G, Sejian V, Velayudhan SM, Mishra C, Rebez EB, Chauhan SS, DiGiacomo K, Lacetera N, Dunshea FR. Applications of Next-Generation Sequencing Technologies and Statistical Tools in Identifying Pathways and Biomarkers for Heat Tolerance in Livestock. Vet Sci 2024; 11:616. [PMID: 39728955 DOI: 10.3390/vetsci11120616] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Revised: 11/17/2024] [Accepted: 11/29/2024] [Indexed: 12/28/2024] Open
Abstract
The climate change-associated abnormal weather patterns negatively influences the productivity and performance of farm animals. Heat stress is the major detrimental factor hampering production, causing substantial economic loss to the livestock industry. Therefore, it is important to identify heat-tolerant breeds that can survive and produce optimally in any given environment. To achieve this goal, a clearer understanding of the genetic differences and the underlying molecular mechanisms associated with climate change impacts and heat tolerance are a prerequisite. Adopting next-generation biotechnological and statistical tools like whole transcriptome analysis, whole metagenome sequencing, bisulphite sequencing, genome-wide association studies (GWAS), and selection signatures provides an opportunity to achieve this goal. Through these techniques, it is possible to identify permanent genetic markers for heat tolerance, and by incorporating those markers in marker-assisted breeding selection, it is possible to achieve the target of breeding for heat tolerance in livestock. This review gives an overview of the recent advancements in assessing heat tolerance in livestock using such 'omics' approaches and statistical models. The salient findings from this research highlighted several candidate biomarkers that have the potential to be incorporated into future heat-tolerance studies. Such approaches could revolutionise livestock production in the changing climate scenario and support the food demands of the growing human population.
Collapse
Affiliation(s)
- Gajendirane Kalaignazhal
- Rajiv Gandhi Institute of Veterinary Education and Research, Kurumbapet 605009, Puducherry, India
- Department of Animal Breeding and Genetics, College of Veterinary Science and Animal Husbandry, Odisha University of Agriculture and Technology, Bhubaneshwar 751003, Odisha, India
| | - Veerasamy Sejian
- Rajiv Gandhi Institute of Veterinary Education and Research, Kurumbapet 605009, Puducherry, India
| | | | - Chinmoy Mishra
- Department of Animal Breeding and Genetics, College of Veterinary Science and Animal Husbandry, Odisha University of Agriculture and Technology, Bhubaneshwar 751003, Odisha, India
| | - Ebenezer Binuni Rebez
- Rajiv Gandhi Institute of Veterinary Education and Research, Kurumbapet 605009, Puducherry, India
| | - Surinder Singh Chauhan
- School of Agriculture, Food and Ecosystem Sciences, Faculty of Science, The University of Melbourne, Melbourne, VIC 3010, Australia
| | - Kristy DiGiacomo
- School of Agriculture, Food and Ecosystem Sciences, Faculty of Science, The University of Melbourne, Melbourne, VIC 3010, Australia
| | - Nicola Lacetera
- Department of Agriculture and Forest Sciences, University of Tuscia, 01100 Viterbo, Italy
| | - Frank Rowland Dunshea
- School of Agriculture, Food and Ecosystem Sciences, Faculty of Science, The University of Melbourne, Melbourne, VIC 3010, Australia
- Faculty of Biological Sciences, The University of Leeds, Leeds LS2 9JT, UK
| |
Collapse
|
16
|
Hussein Y, Weisblum‐Neuman H, Ben Zeev B, Stern S. Previously defined variants of uncertain significance may play an important role in epilepsy and interactions between certain variants may become pathogenic. Epilepsia Open 2024; 9:2443-2453. [PMID: 39509559 PMCID: PMC11633689 DOI: 10.1002/epi4.13085] [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: 05/26/2024] [Revised: 09/28/2024] [Accepted: 10/09/2024] [Indexed: 11/15/2024] Open
Abstract
OBJECTIVE Epilepsy is a chronic neurological disorder related to various etiologies, and the prevalence of active epilepsy is estimated to be between 4 and 10 per 1000 individuals having a significant role in genetic mutations. Next-Generation Sequencing (NGS) panels are utilized for genetic testing, but a substantial proportion of the results remain uncertain and are not considered directly causative of epilepsy. This study aimed to reevaluate pediatric patients diagnosed with epilepsy who underwent genetic investigation using NGS panels, focusing on inconclusive variant findings or multiple variants of uncertain significance (VUSs). METHODS A subgroup of pediatric patients aged 0-25 years, diagnosed with epilepsy, who underwent genetic investigation with an NGS epilepsy panel at the Child Neurology Unit, The Edmond and Lily Safra Children's Hospital, Sheba Medical Center, between 2018 and 2022 through Invitae, was reevaluated. Patients with inconclusive variant findings or multiple VUSs in their test results were included. Genetic data were analyzed to identify potentially pathogenic variants and frequent genetic combinations. RESULTS Two unrelated potentially pathogenic variants were identified in the SCN9A and QARS1 genes. A frequent genetic combination, RANBP2&RYR3, was also observed among other combinations. The RANBP2 gene consistently co-occurred with RYR3 variants in uncertain results, suggesting potential pathogenicity. Analysis of unaffected parents' data revealed certain combinations inherited from different parents, suggesting specific gene combinations as possible risk factors for the disease. SIGNIFICANCE This study highlights the importance of reevaluating genetic data from pediatric epilepsy patients with inconclusive variant findings or multiple VUSs. Identification of potentially pathogenic variants and frequent genetic combinations, such as RANBP2&RYR3, could aid in understanding the genetic basis of epilepsy and identifying potential hotspots. PLAIN LANGUAGE SUMMARY We have performed a retrospective analysis on a subpopulation of pediatric patients diagnosed with epilepsy. We found that specific genetic variants were repeatable, indicating their potential pathogenicity to the disease.
Collapse
Affiliation(s)
- Yara Hussein
- Sagol Department of Neurobiology, Faculty of Natural SciencesUniversity of HaifaHaifaIsrael
| | - Hila Weisblum‐Neuman
- Pediatric Neurology Unit, The Edmond and Lily Safra Children's HospitalSheba Medical CenterRamat GanIsrael
| | - Bruria Ben Zeev
- Pediatric Neurology Unit, The Edmond and Lily Safra Children's HospitalSheba Medical CenterRamat GanIsrael
- Faculty of MedicineTel Aviv UniversityTel AvivIsrael
| | - Shani Stern
- Sagol Department of Neurobiology, Faculty of Natural SciencesUniversity of HaifaHaifaIsrael
| |
Collapse
|
17
|
Ren C, D'Amato G, Hornicek FJ, Tao H, Duan Z. Advances in the molecular biology of the solitary fibrous tumor and potential impact on clinical applications. Cancer Metastasis Rev 2024; 43:1337-1352. [PMID: 39120790 PMCID: PMC11554739 DOI: 10.1007/s10555-024-10204-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Accepted: 07/29/2024] [Indexed: 08/10/2024]
Abstract
Solitary fibrous tumor (SFT) is a rare fibroblastic mesenchymal neoplasm. The current classification has merged SFT and hemangiopericytoma (HPC) into the same tumor entity, while the risk stratification models have been developed to compensate for clinical prediction. Typically, slow-growing and asymptomatic, SFT can occur in various anatomical sites, most commonly in the pleura. Histologically, SFT consists of spindle to oval cells with minimal patterned growth, surrounded by stromal collagen and unique vascular patterns. Molecularly, SFT is defined by the fusion of NGFI-A-binding protein 2 (NAB2) and signal transducer and activator of transcription 6 (STAT6) genes as NAB2-STAT6. This fusion transforms NAB2 into a transcriptional activator, activating early growth response 1 (EGR1) and contributing to SFT pathogenesis and development. There are several fusion variants of NAB2-STAT6 in tumor tissues, with the most frequent ones being NAB2ex4-STAT6ex2 and NAB2ex6-STAT6ex16/ex17. Diagnostic methods play a crucial role in SFT clinical practice and basic research, including RT-PCR, next-generation sequencing (NGS), FISH, immunohistochemistry (IHC), and Western blot analysis, each with distinct capabilities and limitations. Traditional treatment strategies of SFT encompass surgical resection, radiation therapy, and chemotherapy, while emerging management regimes include antiangiogenic agents, immunotherapy, RNA-targeting technologies, and potential targeted drugs. This review provides an update on SFT's clinical and molecular aspects, diagnostic methods, and potential therapies.
Collapse
Affiliation(s)
- Chongmin Ren
- Department of Bone Tumor, The Affiliated Hospital of Qingdao University, No.59 Haier Road, Qingdao, 266101, Shandong, China
- Department of Orthopedic Surgery, Sarcoma Biology Laboratory, Sylvester Comprehensive Cancer Center, and the University of Miami Miller School of Medicine, Papanicolaou Cancer Research Building, 1550 NW. 10Th Avenue, Miami, FL, 33136, USA
- The Orthopedic Hospital, The Affiliated Hospital of Qingdao University, No.59 Haier Road, Qingdao, 266101, Shandong, China
| | - Gina D'Amato
- Department of Orthopedic Surgery, Sarcoma Biology Laboratory, Sylvester Comprehensive Cancer Center, and the University of Miami Miller School of Medicine, Papanicolaou Cancer Research Building, 1550 NW. 10Th Avenue, Miami, FL, 33136, USA
| | - Francis J Hornicek
- Department of Orthopedic Surgery, Sarcoma Biology Laboratory, Sylvester Comprehensive Cancer Center, and the University of Miami Miller School of Medicine, Papanicolaou Cancer Research Building, 1550 NW. 10Th Avenue, Miami, FL, 33136, USA
| | - Hao Tao
- The Orthopedic Hospital, The Affiliated Hospital of Qingdao University, No.59 Haier Road, Qingdao, 266101, Shandong, China.
| | - Zhenfeng Duan
- Department of Orthopedic Surgery, Sarcoma Biology Laboratory, Sylvester Comprehensive Cancer Center, and the University of Miami Miller School of Medicine, Papanicolaou Cancer Research Building, 1550 NW. 10Th Avenue, Miami, FL, 33136, USA.
| |
Collapse
|
18
|
Hiley AS, Mongiardino Koch N, Rouse GW. Phylogenetics of Lepidonotopodini (Macellicephalinae, Polynoidae, Annelida) and Comparative Mitogenomics of Shallow-Water vs. Deep-Sea Scaleworms (Aphroditiformia). BIOLOGY 2024; 13:979. [PMID: 39765646 PMCID: PMC11726774 DOI: 10.3390/biology13120979] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/27/2024] [Revised: 11/20/2024] [Accepted: 11/20/2024] [Indexed: 01/15/2025]
Abstract
Within Polynoidae, a diverse aphroditiform family, the subfamily Macellicephalinae comprises anchialine cave-dwelling and deep-sea scaleworms. In this study, Lepidonotopodinae is synonymized with Macellicephalinae, and the tribe Lepidonotopodini is applied to a well-supported clade inhabiting deep-sea chemosynthetic-based ecosystems. Newly sequenced "genome skimming" data for 30 deep-sea polynoids and the comparatively shallow living Eulagisca gigantea is used to bioinformatically assemble their mitogenomes. When analyzed with existing scaleworm mitogenomes, deep-sea scaleworms exhibit increased gene order rearrangement events compared to shallow-water relatives. Additionally, comparative analyses of shallow-water vs. deep-sea polynoid substitution rates in mitochondrial protein-coding genes show an overall relaxed purifying selection and a positive selection of several amino acid sites in deep-sea species, indicating that polynoid mitogenomes have undergone selective pressure to evolve metabolic adaptations suited to deep-sea environments. Furthermore, the inclusion of skimming data for already known Lepidonotopodini species allowed for an increased coverage of DNA data and a representation of the taxa necessary to create a more robust phylogeny using 18 genes, as opposed to the six genes previously used. The phylogenetic results support the erection of Cladopolynoe gen. nov., Mamiwata gen. nov., Photinopolynoe gen. nov., Stratigos gen. nov., and Themis gen. nov., and emended diagnoses for Branchinotogluma, Branchipolynoe, Lepidonotopodium, and Levensteiniella.
Collapse
Affiliation(s)
- Avery S. Hiley
- Scripps Institution of Oceanography, University of California San Diego, La Jolla, CA 92093-0202, USA;
| | | | - Greg W. Rouse
- Scripps Institution of Oceanography, University of California San Diego, La Jolla, CA 92093-0202, USA;
| |
Collapse
|
19
|
Lefèvre C, Irigoyen N. Why is next-generation sequencing essential in modern virology? J Gen Virol 2024; 105. [PMID: 39570665 DOI: 10.1099/jgv.0.002050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2024] Open
Abstract
A contaminated viral stock results in considerable loss of time, resources and financial means and is generally discovered only by chance after years of research. Thus, it is necessary to implement a technique that can detect contamination without prior knowledge or assumptions, such as next-generation sequencing (NGS). Here, we describe the discovery of a cross-contaminated viral stock from a biological repository of an African Zika virus isolate with Toscana virus after performing NGS on infected cells. In addition, we also describe the consequences that we faced using this contaminated stock. These included the economic and time loss to the lab that needed to repeat all previously performed experiments, the generation of biologically flawed results with a subsequent potential retraction and the severe risk of infection of lab members who manipulated the contaminated stock.
Collapse
Affiliation(s)
- Charlotte Lefèvre
- Division of Virology, Department of Pathology, University of Cambridge, Tennis Court Road, Cambridge, UK
| | - Nerea Irigoyen
- Division of Virology, Department of Pathology, University of Cambridge, Tennis Court Road, Cambridge, UK
| |
Collapse
|
20
|
Kang DW, Park SK, Kang S, Lee EK. Cost-effectiveness of next-generation sequencing for advanced EGFR/ALK-negative non-small cell lung cancer. Lung Cancer 2024; 197:107970. [PMID: 39366308 DOI: 10.1016/j.lungcan.2024.107970] [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: 02/02/2024] [Revised: 07/16/2024] [Accepted: 09/27/2024] [Indexed: 10/06/2024]
Abstract
OBJECTIVES This study aimed to evaluate the cost-effectiveness of next-generation sequencing (NGS) versus sequential single-gene testing (SGT), including the long-term costs and survival outcomes of relevant treatments for advanced EGFR/ALK-negative non-small cell lung cancer (NSCLC). MATERIALS AND METHODS We developed a decision tree linked to a partitioned survival model to estimate the clinical outcomes and costs over the five-year analysis period. The decision tree consisted of treatment types based on molecular biomarker (ROS1, BRAF, NTRK, MET, RET, and KRAS alterations) test results. The probability of receiving each targeted therapy was estimated based on 1) the testing rate, 2) the proportion of alterations detected, and 3) the proportion of patients receiving treatment consistent with the testing results. We estimated the long-term overall survival and progression-free survival for each treatment using parametric estimation by reconstructing patient-level data from clinical trials. The costs of testing, drugs, administration, physician visits, monitoring, adverse events, post-progression, and end-of-life care were included. The utility values were obtained from a previous study. The incremental cost-effectiveness ratio (ICER) was used to evaluate the cost-effectiveness of NGS within a threshold of $38,701 (50,000,000 KRW) per quality-adjusted life year (QALY). RESULTS The incremental life-years (LYs) and QALYs for the NGS group versus the SGT group were 0.028 and 0.023, respectively. The total medical cost for the NGS group was $8,375 higher than that for the SGT group. The difference in drug costs accounted for most of the differences in total medical costs. NGS was not cost-effective compared to sequential SGT, with an ICER of $300,233/LY and $359,405/QALY, respectively. CONCLUSIONS NGS is not cost-effective for advanced EGFR/ALK-negative NSCLC, but has a survival benefit over sequential SGT. Our findings provide a basis for decision-making regarding the coverage and clinical utilization of NGS in regions where EGFR alterations are prevalent.
Collapse
Affiliation(s)
- Dong-Won Kang
- School of Pharmacy, Sungkyunkwan University, Suwon, Gyeonggi-do, Republic of Korea
| | - Sun-Kyeong Park
- College of Pharmacy, The Catholic University of Korea, Bucheon, Gyeonggi-do, Republic of Korea
| | - Sokbom Kang
- Center for Gynecologic Cancer, Research Institute and Hospital, National Cancer Center, Goyang, Gyeonggi-do, Republic of Korea; Department of Cancer Control and Policy, Graduate School of Cancer Science and Policy, National Cancer Center, Goyang, Gyeonggi-do, Republic of Korea.
| | - Eui-Kyung Lee
- School of Pharmacy, Sungkyunkwan University, Suwon, Gyeonggi-do, Republic of Korea.
| |
Collapse
|
21
|
Ren C, Liu J, Hornicek FJ, Yue B, Duan Z. Advances of SS18-SSX fusion gene in synovial sarcoma: Emerging novel functions and therapeutic potentials. Biochim Biophys Acta Rev Cancer 2024; 1879:189215. [PMID: 39528099 DOI: 10.1016/j.bbcan.2024.189215] [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] [Revised: 10/31/2024] [Accepted: 11/02/2024] [Indexed: 11/16/2024]
Abstract
Synovial sarcoma is a rare type of soft tissue sarcoma that primarily affects adolescents and young adults, featured by aggressive behavior and a high potential for metastasis. Genetically, synovial sarcoma is defined by the fusion oncogene SS18-SSX arising from the translocation of t(X;18)(p11;q11). SS18-SSX fusion gene is the major driver of the oncogenic event in synovial sarcoma. SS18-SSX fusion protein, while not containing any DNA-binding motifs, binds to the SWI/SNF (BAF) complex, a major epigenetic regulator, leading to the disruption of gene expression which results in tumor initiation and progression. Emerging studies on the molecular mechanisms of SS18-SSX associated signaling pathway hold promise for developments in diagnosis and treatments. Advanced diagnostic methods facilitate early and precise detection of the tumor, enabling disease monitoring and prognostic improvements. Treatment of synovial sarcoma typically comprises local surgery, radiotherapy and chemotherapy, while novel managements such as immunotherapy, targeted therapies and epigenetic modifiers are explored. This review focuses on the recent studies of SS18-SSX fusion gene, epigenetic landscape, signaling pathways, diagnostic techniques, and relevant therapeutic advances, aiming to inhibit the oncogenic processes and improve outcomes for patients with synovial sarcoma.
Collapse
Affiliation(s)
- Chongmin Ren
- Department of Bone Tumor, The Affiliated Hospital of Qingdao University, No.59 Haier Road, Qingdao, Shandong 266101, China; Department of Orthopedic Surgery, Sarcoma Biology Laboratory, Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Papanicolaou Cancer Research Building, 1550 NW. 10th Avenue, Miami, Florida 33136, USA.
| | - Jia Liu
- Department of Pediatric Nephrology, Rheumatology and Immunity, The Affiliated Hospital of Qingdao University, No.16 Jiangsu Road, Qingdao, Shandong 266003, China.
| | - Francis J Hornicek
- Department of Orthopedic Surgery, Sarcoma Biology Laboratory, Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Papanicolaou Cancer Research Building, 1550 NW. 10th Avenue, Miami, Florida 33136, USA.
| | - Bin Yue
- Department of Bone Tumor, The Affiliated Hospital of Qingdao University, No.59 Haier Road, Qingdao, Shandong 266101, China.
| | - Zhenfeng Duan
- Department of Orthopedic Surgery, Sarcoma Biology Laboratory, Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Papanicolaou Cancer Research Building, 1550 NW. 10th Avenue, Miami, Florida 33136, USA.
| |
Collapse
|
22
|
Motlagh Asghari K, Novinbahador T, Mehdizadeh A, Zolfaghari M, Yousefi M. Revolutionized attitude toward recurrent pregnancy loss and recurrent implantation failure based on precision regenerative medicine. Heliyon 2024; 10:e39584. [PMID: 39498089 PMCID: PMC11532865 DOI: 10.1016/j.heliyon.2024.e39584] [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: 07/13/2024] [Revised: 10/08/2024] [Accepted: 10/17/2024] [Indexed: 11/07/2024] Open
Abstract
Traditional treatment strategies for recurrent pregnancy loss (RPL) and recurrent implantation failure (RIF) often result in limited success, placing significant emotional and financial burdens on couples. However, novel approaches such as diagnostic gene profiling, cell therapy, stem cell-derived exosome therapy, and pharmacogenomics offer promising, personalized treatments. Combining traditional treatments with precision and regenerative medicine may enhance the efficacy of these approaches and improve pregnancy outcomes. This review explores how integrating these strategies can potentially transform the lives of couples experiencing repeated pregnancy loss or implantation failure, providing hope for improved treatment success. Precision and regenerative medicine represent a new frontier for managing RPL and RIF, offering promising solutions.
Collapse
Affiliation(s)
| | - Tannaz Novinbahador
- Department of Biology, Faculty of Natural Sciences, University of Tabriz, Tabriz, Iran
- Department of Clinical Biochemistry, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Amir Mehdizadeh
- Hematology and Oncology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Mehdi Yousefi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
23
|
Eom JS, Kim SH, Kim K, Kim A, Ahn HY, Mok J, Cho JS, Lee MK, Song JS, Kim MH. The clinical relevance of surgical specimens for RNA sequencing in lung cancer: a cohort study. Front Oncol 2024; 14:1462519. [PMID: 39493448 PMCID: PMC11527598 DOI: 10.3389/fonc.2024.1462519] [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: 07/10/2024] [Accepted: 10/02/2024] [Indexed: 11/05/2024] Open
Abstract
Background Molecular screening using next-generation sequencing (NGS) in the pathologic evaluation of lung cancer is considered the standard in clinical practice; hence, we evaluated the diagnostic yields of various sampling methods for NGS. Methods NGS data from patients with lung cancer at the Pusan National University Hospital (Busan, South Korea), admitted October, 2020-April, 2023, was obtained. The sampling methods by which NGS data was obtained were divided into surgical and nonsurgical. Surgical methods included thoracoscopic surgery, surgical biopsy from the metastatic site, and lymph node excisional biopsy, whereas nonsurgical methods included bronchoscopy procedures and medical thoracoscopic biopsy. Results In total, we obtained 319 patients' NGS data:150 (47.0%) and 169 (53.0%) was obtained using surgical and nonsurgical methods, respectively. The overall diagnostic yield of NGS analysis was 97.5% for all samples. There were no significant differences in the success rates of deoxyribonucleic acid sequencing between surgical and nonsurgical sampling methods (98.0% vs. 96.4%, p = 0.313). On the other hand, the success rate of ribonucleic acid (RNA) sequencing was significantly lower in the surgical method group (78.0% vs. 92.3%; p < 0.001). Multivariate analysis showed that surgical sampling significantly correlated with RNA sequencing failure (Odd Ratio 4.128, 95% Confidence Interval 1.681-10.133, p = 0.002). Conclusions Small samples obtained using nonsurgical procedures are suitable for NGS analysis in clinical practice. However, surgical sampling showed a relatively lower success rate for RNA sequencing than nonsurgical sampling. This information may help in the development of protocols to reduce RNA degradation during the surgical process.
Collapse
Affiliation(s)
- Jung Seop Eom
- Department of Internal Medicine, Pusan National University School of Medicine, Busan, Republic of Korea
- Department of Internal Medicine, Pusan National University Hospital, Busan, Republic of Korea
- Biomedical Research Institute, Pusan National University Hospital, Busan, Republic of Korea
| | - Soo Han Kim
- Department of Internal Medicine, Pusan National University Hospital, Busan, Republic of Korea
- Biomedical Research Institute, Pusan National University Hospital, Busan, Republic of Korea
| | - Kyungbin Kim
- Department of Pathology, Pusan National University Hospital, Busan, Republic of Korea
| | - Ahrong Kim
- Department of Pathology, Pusan National University Hospital, Busan, Republic of Korea
- Department of Pathology, Pusan National University School of Medicine, Busan, Republic of Korea
| | - Hyo Yeong Ahn
- Department of Thoracic and Cardiovascular Surgery, Pusan National University School of Medicine, Busan, Republic of Korea
| | - Jeongha Mok
- Department of Internal Medicine, Pusan National University School of Medicine, Busan, Republic of Korea
- Department of Internal Medicine, Pusan National University Hospital, Busan, Republic of Korea
- Biomedical Research Institute, Pusan National University Hospital, Busan, Republic of Korea
| | - Jeong Su Cho
- Department of Thoracic and Cardiovascular Surgery, Pusan National University School of Medicine, Busan, Republic of Korea
| | - Min Ki Lee
- Department of Internal Medicine, Pusan National University School of Medicine, Busan, Republic of Korea
- Department of Internal Medicine, Pusan National University Hospital, Busan, Republic of Korea
| | - Ju Sun Song
- Department of Laboratory Medicine, Green Cross (GC) Genome Corporation, Yongin, Republic of Korea
| | - Mi-Hyun Kim
- Department of Internal Medicine, Pusan National University School of Medicine, Busan, Republic of Korea
- Department of Internal Medicine, Pusan National University Hospital, Busan, Republic of Korea
- Biomedical Research Institute, Pusan National University Hospital, Busan, Republic of Korea
| |
Collapse
|
24
|
Liu Z, Zhu X, Zhang S, Li D, Wang D, Wang Y, Tang Y, Tong F, Xu W, Li G, Wei L, Chu M. Comparative study of pathogen detection methods for central nervous system infections: laboratory testing of tuberculous meningitis. BMC Infect Dis 2024; 24:1172. [PMID: 39420282 PMCID: PMC11488244 DOI: 10.1186/s12879-024-10037-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Accepted: 10/01/2024] [Indexed: 10/19/2024] Open
Abstract
BACKGROUND Tuberculous meningitis (TBM) is a severe central nervous system (CNS) infection with a challenging diagnosis due to inadequate detection methods. This study evaluated current clinical detection methods and their applicability. METHODS A cohort of 514 CNS infection patients from 2018 to 2020 was studied. Data on general demographics, Cerebrospinal Fluid (CSF) analysis, epidemiology, and clinical outcomes were collected. TBM patients were identified, and the sensitivities of mmetagenomic next-generation sequencing (NGS), GeneXpert, and microbial culture were compared. Kappa statistic assessed the consistency between methods. RESULTS Among the patients involved, TBM (29%) and neurosyphilis (25%) were the two most prevalent CNS infections. CSF analysis indicated that 76% of patients had leukocytosis, suggesting a potential CNS inflammation. In TBM cases, 92.5% had elevated CSF protein and leukocyte counts. Moreover, the percentage of positive mNGS results was 55.6%. GeneXpert and MTB cultures alone had lower sensitivity, but combined use resulted in a 53.4% positive rate. CONCLUSIONS This study highlights the high sensitivity of mNGS, comparable to GeneXpert and MTB culture. The combined methods are cost-effective and straightforward, and can partially substitute for mNGS, offering valuable alternatives for TBM diagnosis and providing insights into multiple diagnostic strategies in clinical practice.
Collapse
Affiliation(s)
- Zengchen Liu
- Department of Neurosurgery, Shenzhen Third People's Hospital, Shenzhen, 518112, China
- Three Departments of Lung Diseases, Shenzhen Third People's Hospital, Shenzhen, Guangdong, 518112, China
- Pulmonary Diseases Departments (3), Shenzhen Third People's Hospital, Shenzhen, 518112, China
| | - Xujie Zhu
- Central Laboratory, Longgang District Maternity & Child Healthcare Hospital of Shenzhen City (Longgang Maternity and Child Institute of Shantou University Medical College), Shenzhen, China
| | - Shengkun Zhang
- Department of Neurosurgery, Shenzhen Third People's Hospital, Shenzhen, 518112, China
- Three Departments of Lung Diseases, Shenzhen Third People's Hospital, Shenzhen, Guangdong, 518112, China
| | - Dapeng Li
- Three Departments of Lung Diseases, Shenzhen Third People's Hospital, Shenzhen, Guangdong, 518112, China
- The Second Affiliated Hospital, School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong, 518000, China
| | - Dian Wang
- Department of Neurosurgery, Shenzhen Third People's Hospital, Shenzhen, 518112, China
- Three Departments of Lung Diseases, Shenzhen Third People's Hospital, Shenzhen, Guangdong, 518112, China
| | - Yijie Wang
- Department of Neurosurgery, Shenzhen Third People's Hospital, Shenzhen, 518112, China
- Three Departments of Lung Diseases, Shenzhen Third People's Hospital, Shenzhen, Guangdong, 518112, China
| | - Yunyan Tang
- Department of Neurosurgery, Shenzhen Third People's Hospital, Shenzhen, 518112, China
- Three Departments of Lung Diseases, Shenzhen Third People's Hospital, Shenzhen, Guangdong, 518112, China
| | - Fangjia Tong
- Pulmonary Diseases Departments (3), Shenzhen Third People's Hospital, Shenzhen, 518112, China
| | - Wanzhen Xu
- Department of Neurosurgery, Shenzhen Third People's Hospital, Shenzhen, 518112, China
- Three Departments of Lung Diseases, Shenzhen Third People's Hospital, Shenzhen, Guangdong, 518112, China
| | - Guobao Li
- Three Departments of Lung Diseases, Shenzhen Third People's Hospital, Shenzhen, Guangdong, 518112, China.
- Institute for Hepatology, National Clinical Research Center for Infectious Diseases, Shenzhen Third People's Hospital, Shenzhen, Guangdong, 518112, China.
| | - Lanlan Wei
- Pulmonary Diseases Departments (3), Shenzhen Third People's Hospital, Shenzhen, 518112, China.
| | - Ming Chu
- Department of Neurosurgery, Shenzhen Third People's Hospital, Shenzhen, 518112, China.
- Three Departments of Lung Diseases, Shenzhen Third People's Hospital, Shenzhen, Guangdong, 518112, China.
| |
Collapse
|
25
|
Zhu M, Zuber J, Tan Z, Sharma G, Mathews DH. DecoyFinder: Identification of Contaminants in Sets of Homologous RNA Sequences. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.12.618037. [PMID: 39464058 PMCID: PMC11507696 DOI: 10.1101/2024.10.12.618037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/29/2024]
Abstract
Motivation RNA structure is essential for the function of many non-coding RNAs. Using multiple homologous sequences, which share structure and function, secondary structure can be predicted with much higher accuracy than with a single sequence. It can be difficult, however, to establish a set of homologous sequences when their structure is not yet known. We developed a method to identify sequences in a set of putative homologs that are in fact non-homologs. Results Previously, we developed TurboFold to estimate conserved structure using multiple, unaligned RNA homologs. Here, we report that the positive predictive value of TurboFold is significantly reduced by the presence of contamination by non-homologous sequences, although the reduction is less than 1%. We developed a method called DecoyFinder, which applies machine learning trained with features determined by TurboFold, to detect sequences that are not homologous with the other sequences in the set. This method can identify approximately 45% of non-homologous sequences, at a rate of 5% misidentification of true homologous sequences. Availability DecoyFinder and TurboFold are incorporated in RNAstructure, which is provided for free and open source under the GPL V2 license. It can be downloaded at http://rna.urmc.rochester.edu/RNAstructure.html.
Collapse
Affiliation(s)
- Mingyi Zhu
- Center for RNA Biology, University of Rochester Medical Center, Rochester, NY, United States
- Department of Biochemistry and Biophysics, University of Rochester Medical Center, Rochester, NY, United States
| | - Jeffrey Zuber
- Center for RNA Biology, University of Rochester Medical Center, Rochester, NY, United States
- Department of Biochemistry and Biophysics, University of Rochester Medical Center, Rochester, NY, United States
| | - Zhen Tan
- Center for RNA Biology, University of Rochester Medical Center, Rochester, NY, United States
- Department of Biochemistry and Biophysics, University of Rochester Medical Center, Rochester, NY, United States
| | - Gaurav Sharma
- University of Rochester, Department of Electrical and Computer Engineering, Rochester, NY, United States
- University of Rochester, Department of Computer Science, Rochester, NY, United States
| | - David H Mathews
- Center for RNA Biology, University of Rochester Medical Center, Rochester, NY, United States
- Department of Biochemistry and Biophysics, University of Rochester Medical Center, Rochester, NY, United States
| |
Collapse
|
26
|
Gregorova K, Plachy L, Dusatkova P, Maratova K, Neuman V, Kolouskova S, Snajderova M, Obermannova B, Drnkova L, Soucek O, Lebl J, Sumnik Z, Pruhova S. Genetic Testing of Children With Familial Tall Stature: Is it Worth Doing? J Clin Endocrinol Metab 2024; 109:e2009-e2015. [PMID: 38307035 DOI: 10.1210/clinem/dgae067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Revised: 01/25/2024] [Accepted: 02/01/2024] [Indexed: 02/04/2024]
Abstract
CONTEXT Familial tall stature (FTS) is considered to be a benign variant of growth with a presumed polygenic etiology. However, monogenic disorders with possible associated pathological features could also be hidden under the FTS phenotype. OBJECTIVE To elucidate the genetic etiology in families with FTS and to describe their phenotype in detail. METHODS Children with FTS (the life-maximum height in both the child and his/her taller parent > 2 SD for age and sex) referred to the Endocrinology center of Motol University Hospital were enrolled into the study. Their DNA was examined cytogenetically and via a next-generation sequencing panel of 786 genes associated with growth. The genetic results were evaluated by the American College of Molecular Genetics and Genomics guidelines. All of the participants underwent standard endocrinological examination followed by specialized anthropometric evaluation. RESULTS In total, 34 children (19 girls) with FTS were enrolled in the study. Their median height and their taller parent's height were 3.1 SD and 2.5 SD, respectively. The genetic cause of FTS was elucidated in 11/34 (32.4%) children (47,XXX and 47,XYY karyotypes, SHOX duplication, and causative variants in NSD1 [in 2], SUZ12 [in 2], FGFR3, CHD8, GPC3, and PPP2R5D genes). Ten children had absent syndromic signs and 24 had dysmorphic features. CONCLUSION Monogenic (and cytogenetic) etiology of FTS can be found among children with FTS. Genetic examination should be considered in all children with FTS regardless of the presence of dysmorphic features.
Collapse
Affiliation(s)
- Katerina Gregorova
- Department of Pediatrics, Second Faculty of Medicine, Charles University in Prague and University Hospital Motol, Prague 5, 150 06, Czech Republic
| | - Lukas Plachy
- Department of Pediatrics, Second Faculty of Medicine, Charles University in Prague and University Hospital Motol, Prague 5, 150 06, Czech Republic
| | - Petra Dusatkova
- Department of Pediatrics, Second Faculty of Medicine, Charles University in Prague and University Hospital Motol, Prague 5, 150 06, Czech Republic
| | - Klara Maratova
- Department of Pediatrics, Second Faculty of Medicine, Charles University in Prague and University Hospital Motol, Prague 5, 150 06, Czech Republic
| | - Vit Neuman
- Department of Pediatrics, Second Faculty of Medicine, Charles University in Prague and University Hospital Motol, Prague 5, 150 06, Czech Republic
| | - Stanislava Kolouskova
- Department of Pediatrics, Second Faculty of Medicine, Charles University in Prague and University Hospital Motol, Prague 5, 150 06, Czech Republic
| | - Marta Snajderova
- Department of Pediatrics, Second Faculty of Medicine, Charles University in Prague and University Hospital Motol, Prague 5, 150 06, Czech Republic
| | - Barbora Obermannova
- Department of Pediatrics, Second Faculty of Medicine, Charles University in Prague and University Hospital Motol, Prague 5, 150 06, Czech Republic
| | - Lenka Drnkova
- Department of Pediatrics, Second Faculty of Medicine, Charles University in Prague and University Hospital Motol, Prague 5, 150 06, Czech Republic
| | - Ondrej Soucek
- Department of Pediatrics, Second Faculty of Medicine, Charles University in Prague and University Hospital Motol, Prague 5, 150 06, Czech Republic
| | - Jan Lebl
- Department of Pediatrics, Second Faculty of Medicine, Charles University in Prague and University Hospital Motol, Prague 5, 150 06, Czech Republic
| | - Zdenek Sumnik
- Department of Pediatrics, Second Faculty of Medicine, Charles University in Prague and University Hospital Motol, Prague 5, 150 06, Czech Republic
| | - Stepanka Pruhova
- Department of Pediatrics, Second Faculty of Medicine, Charles University in Prague and University Hospital Motol, Prague 5, 150 06, Czech Republic
| |
Collapse
|
27
|
Hu M, Luo R, Yang K, Yu Y, Pan Q, Yuan M, Chen R, Wang H, Qin Q, Ma T, Wang H. Genomic landscape defines peritoneal metastatic pattern and related target of peritoneal metastasis in colorectal cancer. Int J Cancer 2024; 155:1327-1339. [PMID: 38738976 DOI: 10.1002/ijc.35005] [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: 09/17/2023] [Revised: 03/31/2024] [Accepted: 04/17/2024] [Indexed: 05/14/2024]
Abstract
The primary objective of this study is to develop a prediction model for peritoneal metastasis (PM) in colorectal cancer by integrating the genomic features of primary colorectal cancer, along with clinicopathological features. Concurrently, we aim to identify potential target implicated in the peritoneal dissemination of colorectal cancer through bioinformatics exploration and experimental validation. By analyzing the genomic landscape of primary colorectal cancer and clinicopathological features from 363 metastatic colorectal cancer patients, we identified 22 differently distributed variables, which were used for subsequent LASSO regression to construct a PM prediction model. The integrated model established by LASSO regression, which incorporated two clinicopathological variables and seven genomic variables, precisely discriminated PM cases (AUC 0.899; 95% CI 0.860-0.937) with good calibration (Hosmer-Lemeshow test p = .147). Model validation yielded AUCs of 0.898 (95% CI 0.896-0.899) and 0.704 (95% CI 0.622-0.787) internally and externally, respectively. Additionally, the peritoneal metastasis-related genomic signature (PGS), which was composed of the seven genes in the integrated model, has prognostic stratification capability for colorectal cancer. The divergent genomic landscape drives the driver genes of PM. Bioinformatic analysis concerning these driver genes indicated SERINC1 may be associated with PM. Subsequent experiments indicate that knocking down of SERINC1 functionally suppresses peritoneal dissemination, emphasizing its importance in CRCPM. In summary, the genomic landscape of primary cancer in colorectal cancer defines peritoneal metastatic pattern and reveals the potential target of SERINC1 for PM in colorectal cancer.
Collapse
Affiliation(s)
- Minhui Hu
- Department of Gastrointestinal Endoscopy, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Rui Luo
- Department of General Surgery, Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Keli Yang
- Department of General Surgery, Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yang Yu
- Department of General Surgery, Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Qiwen Pan
- Department of Radiation Oncology, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Mingming Yuan
- Geneplus-Beijing, Medical Park Road, Zhongguancun Life Science Park, Beijing, China
| | - Rongrong Chen
- Geneplus-Beijing, Medical Park Road, Zhongguancun Life Science Park, Beijing, China
| | - Hui Wang
- Department of General Surgery, Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Qiyuan Qin
- Department of General Surgery, Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Tenghui Ma
- Department of General Surgery, Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Huaiming Wang
- Department of General Surgery, Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
28
|
Mukhopadhyay S, Dixit P, Khanom N, Sanghera G, McGurk KA. The Genetic Factors Influencing Cardiomyopathies and Heart Failure across the Allele Frequency Spectrum. J Cardiovasc Transl Res 2024; 17:1119-1139. [PMID: 38771459 PMCID: PMC11519107 DOI: 10.1007/s12265-024-10520-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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Accepted: 05/03/2024] [Indexed: 05/22/2024]
Abstract
Heart failure (HF) remains a major cause of mortality and morbidity worldwide. Understanding the genetic basis of HF allows for the development of disease-modifying therapies, more appropriate risk stratification, and personalised management of patients. The advent of next-generation sequencing has enabled genome-wide association studies; moving beyond rare variants identified in a Mendelian fashion and detecting common DNA variants associated with disease. We summarise the latest GWAS and rare variant data on mixed and refined HF aetiologies, and cardiomyopathies. We describe the recent understanding of the functional impact of titin variants and highlight FHOD3 as a novel cardiomyopathy-associated gene. We describe future directions of research in this field and how genetic data can be leveraged to improve the care of patients with HF.
Collapse
Affiliation(s)
- Srinjay Mukhopadhyay
- National Heart and Lung Institute, Imperial College London, LMS Building, Hammersmith Campus, London, UK
- School of Medicine, Cardiff University, Wales, UK
| | - Prithvi Dixit
- National Heart and Lung Institute, Imperial College London, LMS Building, Hammersmith Campus, London, UK
| | - Najiyah Khanom
- National Heart and Lung Institute, Imperial College London, LMS Building, Hammersmith Campus, London, UK
| | - Gianluca Sanghera
- National Heart and Lung Institute, Imperial College London, LMS Building, Hammersmith Campus, London, UK
| | - Kathryn A McGurk
- National Heart and Lung Institute, Imperial College London, LMS Building, Hammersmith Campus, London, UK.
- MRC Laboratory of Medical Sciences, Imperial College London, London, UK.
| |
Collapse
|
29
|
Irankhah L, Khorsand B, Naghibzadeh M, Savadi A. Analyzing the performance of short-read classification tools on metagenomic samples toward proper diagnosis of diseases. J Bioinform Comput Biol 2024:2450012. [PMID: 39287058 DOI: 10.1142/s0219720024500124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/19/2024]
Abstract
Accurate knowledge of the genome, virus and bacteria that have invaded our bodies is crucial for diagnosing many human diseases. The field of bioinformatics encompasses the complex computational methods required for this purpose. Metagenomics employs next-generation sequencing (NGS) technology to study and identify microbial communities in environmental samples. This technique allows for the measurement of the relative abundance of different microbes. Various tools are available for detecting bacterial species in sequenced metagenomic samples. In this study, we focus on well-known taxonomic classification tools such as MetaPhlAn4, Centrifuge, Kraken2, and Bracken, and evaluate their performance at the species level using synthetic and real datasets. The results indicate that MetaPhlAn4 exhibited high precision in identifying species in the simulated dataset, while Kraken2 had the best area under the precision-recall curve (AUPR) performance. Centrifuge, Kraken2, and Bracken showed accurate estimation of species abundances, unlike MetaPhlAn4, which had a higher L2 distance. In the real dataset analysis with samples from an inflammatory bowel disease (IBD) research, MetaPhlAn4, and Kraken2 had faster execution times, with differences in performance at family and species levels among the tools. Enterobacteriaceae and Pasteurellaceae were highlighted as the most abundant families by Centrifuge, Kraken2, and MetaPhlAn4, with variations in abundance among ulcerative colitis (UC), Crohn's disease (CD), and control non-IBD (CN) groups. Escherichia coli (E. coli) has the highest abundance among Enterobacteriaceae species in the CD and UC groups in comparison with the CN group. Bracken overestimated E. coli abundance, emphasizing result interpretation caution. The findings of this research can assist in selecting the appropriate short-read classifier, thereby aiding in the diagnosis of target diseases.
Collapse
Affiliation(s)
- Leili Irankhah
- Computer Engineering Department, Ferdowsi University of Mashhad, Mashhad, Iran
| | - Babak Khorsand
- Department of Neurology, University of California Irvine, CA, USA
| | - Mahmoud Naghibzadeh
- Computer Engineering Department, Ferdowsi University of Mashhad, Mashhad, Iran
| | - Abdorreza Savadi
- Computer Engineering Department, Ferdowsi University of Mashhad, Mashhad, Iran
| |
Collapse
|
30
|
Liborio MP, Harris PNA, Ravi C, Irwin AD. Getting Up to Speed: Rapid Pathogen and Antimicrobial Resistance Diagnostics in Sepsis. Microorganisms 2024; 12:1824. [PMID: 39338498 PMCID: PMC11434042 DOI: 10.3390/microorganisms12091824] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Revised: 08/26/2024] [Accepted: 08/27/2024] [Indexed: 09/30/2024] Open
Abstract
Sepsis is a life-threatening organ dysfunction caused by a dysregulated host response to infection. Time to receive effective therapy is a primary determinant of mortality in patients with sepsis. Blood culture is the reference standard for the microbiological diagnosis of bloodstream infections, despite its low sensitivity and prolonged time to receive a pathogen detection. In recent years, rapid tests for pathogen identification, antimicrobial susceptibility, and sepsis identification have emerged, both culture-based and culture-independent methods. This rapid narrative review presents currently commercially available approved diagnostic molecular technologies in bloodstream infections, including their clinical performance and impact on patient outcome, when available. Peer-reviewed publications relevant to the topic were searched through PubMed, and manufacturer websites of commercially available assays identified were also consulted as further sources of information. We have reviewed data about the following technologies for pathogen identification: fluorescence in situ hybridization with peptide nucleic acid probes (Accelerate PhenoTM), microarray-based assay (Verigene®), multiplex polymerase chain reaction (cobas® eplex, BioFire® FilmArray®, Molecular Mouse, Unyvero BCU SystemTM), matrix-assisted laser desorption-ionization time-of-flight mass spectrometry (Rapid MBT Sepsityper®), T2 magnetic resonance (T2Bacteria Panel), and metagenomics-based assays (Karius©, DISQVER®, Day Zero Diagnostics). Technologies for antimicrobial susceptibility testing included the following: Alfed 60 ASTTM, VITEK® REVEALTM, dRASTTM, ASTar®, Fastinov®, QuickMIC®, ResistellTM, and LifeScale. Characteristics, microbiological performance, and issues of each method are described, as well as their clinical performance, when available.
Collapse
Affiliation(s)
- Mariana P. Liborio
- UQ Centre for Clinical Research, The University of Queensland, Herston, QLD 4029, Australia; (M.P.L.); (C.R.)
| | - Patrick N. A. Harris
- UQ Centre for Clinical Research, The University of Queensland, Herston, QLD 4029, Australia; (M.P.L.); (C.R.)
- Herston Infectious Disease Institute, Metro North, QLD Health, Herston, QLD 4029, Australia
- Central Microbiology, Pathology Queensland, Royal Brisbane and Women’s Hospital, Herston, QLD 4006, Australia
| | - Chitra Ravi
- UQ Centre for Clinical Research, The University of Queensland, Herston, QLD 4029, Australia; (M.P.L.); (C.R.)
| | - Adam D. Irwin
- UQ Centre for Clinical Research, The University of Queensland, Herston, QLD 4029, Australia; (M.P.L.); (C.R.)
- Infection Management and Prevention Service, Queensland Children’s Hospital, Brisbane, QLD 4101, Australia
| |
Collapse
|
31
|
Hoch C, Allen JR, Morningstar J, Materon SR, Scott DJ, Gross CE. Identification and Analysis of the Ankle Microbiome Using Next-Generation DNA Sequencing: An Observational Analysis. J Am Acad Orthop Surg 2024; 32:786-792. [PMID: 37976386 DOI: 10.5435/jaaos-d-23-00387] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Indexed: 11/19/2023] Open
Abstract
INTRODUCTION Next-generation DNA sequencing (NGS) technologies have increased the sensitivity for detecting the bacterial presence and have been used in other areas of orthopaedics to better understand the native microbiome of various joints. This study uses NGS to determine whether (1) a unique microbiome exists in human ankle tissues, (2) if components of the ankle microbiome affect patient outcomes, and (3) whether microbes found on the skin are a normal part of the ankle microbiome. METHODS A prospective study recruited 32 patients undergoing total ankle arthroplasty (n = 23) or ankle arthrodesis (n = 9) via direct anterior approach between November 2020 and October 2021. During surgery, five layers of the ankle were swabbed: skin (n = 32), retinaculum (n = 31), tibialis anterior tendon (n = 31), joint capsule (n = 31), and distal tibia (n = 32). These swabs (N = 157) were sent to MicroGen Diagnostics (Lubbock) for NGS. Demographics, medical comorbidities, surgical indication, postoperative complications, readmission, and revision surgery rates were collected from patient records. RESULTS The mean age was 60.7 (range, 19 to 85) years, and the mean follow-up duration was 10.2 (range, 4.8 to 20.6) months. Of 157 swabs sent for NGS, 19 (12.1%) indicated that bacteria were present (positive), whereas the remaining 138 (87.9%) had no bacteria present (negative). The most common organisms were Cutibacterium acnes in eight ankles (25.0%) and Staphylococcus epidermidis in two ankles (6.25%). Most bacteria were found in the retinaculum (29.6%). Complications, nonunions, infections, 90-day readmission, and revision surgery rates did not differ by NGS profile. DISCUSSION This study found that C acnes and S epidermidis were the most common bacteria in the ankle microbiome, with C acnes being present in 25% of ankles. Complication rates did not differ between patients with or without positive bacterial DNA remnants. Thus, we concluded that a unique ankle microbiome is present in some patients, which is unique from that of the skin of the ankle. LEVEL OF EVIDENCE Level II, Prospective cohort study.
Collapse
Affiliation(s)
- Caroline Hoch
- From the Department of Orthopaedics and Physical, Medical University of South Carolina, Charleston, SC (Allen, Morningstar, Materon, Scott, and Gross), and the University of North Carolina, Gillings School of Global Public Health, Chapel Hill, NC (Hoch)
| | | | | | | | | | | |
Collapse
|
32
|
Feng J, Ma T, Wang C, Wang B, Liu Q, Liu Z, Tao H, Ye Z. Clinical relevance and druggability of sole reciprocal kinase fusions: A large-scale study. Cancer Med 2024; 13:e70191. [PMID: 39254060 PMCID: PMC11386300 DOI: 10.1002/cam4.70191] [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: 02/26/2024] [Revised: 08/07/2024] [Accepted: 08/21/2024] [Indexed: 09/11/2024] Open
Abstract
BACKGROUND Building on our prior work that RNA alternative splicing modulates the druggability of kinase fusions, this study probes the clinical significance of sole reciprocal fusions. These rare genomic arrangements, despite lacking kinase domains at the DNA level, demonstrated potential RNA-level druggability in sporadic cases from our prior research. METHODS Utilizing the large-scale multicenter approach, we performed RNA sequencing and clinical follow-up to evaluate a broad spectrum of kinase fusions, including ALK, ROS1, RET, BRAF, NTRK, MET, NRG1, and EGFR, in 1943 patients. RESULTS Our findings revealed 51 instances (2.57%) of sole reciprocal fusions, predominantly in lung (57%), colorectal (14%), and glioma (10%) cancers. Comparative analysis with an MSKCC cohort confirmed the prevalence in diverse cancer types and identified unique fusion partners and chromosomal locales. Cross-validation through RNA-NGS and FISH authenticated the existence of functional kinase domains in subsets including ALK, ROS1, RET, and BRAF, which correlated with positive clinical responses to targeted kinase inhibitors (KIs). Conversely, fusions involving EGFR, NRG1, and NTRK1/2/3 generated nonfunctional transcripts, suggesting the need for alternative therapeutic interventions. CONCLUSION This inaugural multicenter study introduces a novel algorithm for detecting and treating sole reciprocal fusions in advanced cancers, expanding the patient population potentially amenable to KIs.
Collapse
Affiliation(s)
- Jiao Feng
- General Surgery, Cancer CenterZhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical CollegeZhejiangChina
- School of Pharmacy, Hangzhou Normal UniversityZhejiangChina
| | - Tonghui Ma
- Jichenjunchuang Clinical LaboratoryZhejiangChina
- Genecn‐Biotech Co.LtdZhejiangChina
| | - Chunyang Wang
- Jichenjunchuang Clinical LaboratoryZhejiangChina
- Genecn‐Biotech Co.LtdZhejiangChina
| | - Baoming Wang
- Jichenjunchuang Clinical LaboratoryZhejiangChina
- Genecn‐Biotech Co.LtdZhejiangChina
| | - Qian Liu
- College of Medicine, Zhejiang UniversityZhejiangChina
| | - Zhengchuang Liu
- General Surgery, Cancer CenterZhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical CollegeZhejiangChina
| | - Houquan Tao
- General Surgery, Cancer CenterZhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical CollegeZhejiangChina
| | - Zaiyuan Ye
- General Surgery, Cancer CenterZhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical CollegeZhejiangChina
| |
Collapse
|
33
|
Kumawat RL, Jena MK, Mittal S, Pathak B. Advancement of Next-Generation DNA Sequencing through Ionic Blockade and Transverse Tunneling Current Methods. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2401112. [PMID: 38716623 DOI: 10.1002/smll.202401112] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Revised: 04/05/2024] [Indexed: 10/04/2024]
Abstract
DNA sequencing is transforming the field of medical diagnostics and personalized medicine development by providing a pool of genetic information. Recent advancements have propelled solid-state material-based sequencing into the forefront as a promising next-generation sequencing (NGS) technology, offering amplification-free, cost-effective, and high-throughput DNA analysis. Consequently, a comprehensive framework for diverse sequencing methodologies and a cross-sectional understanding with meticulous documentation of the latest advancements is of timely need. This review explores a broad spectrum of progress and accomplishments in the field of DNA sequencing, focusing mainly on electrical detection methods. The review delves deep into both the theoretical and experimental demonstrations of the ionic blockade and transverse tunneling current methods across a broad range of device architectures, nanopore, nanogap, nanochannel, and hybrid/heterostructures. Additionally, various aspects of each architecture are explored along with their strengths and weaknesses, scrutinizing their potential applications for ultrafast DNA sequencing. Finally, an overview of existing challenges and future directions is provided to expedite the emergence of high-precision and ultrafast DNA sequencing with ionic and transverse current approaches.
Collapse
Affiliation(s)
- Rameshwar L Kumawat
- Department of Chemistry, Indian Institute of Technology (IIT) Indore, Indore, Madhya Pradesh, 453552, India
| | - Milan Kumar Jena
- Department of Chemistry, Indian Institute of Technology (IIT) Indore, Indore, Madhya Pradesh, 453552, India
| | - Sneha Mittal
- Department of Chemistry, Indian Institute of Technology (IIT) Indore, Indore, Madhya Pradesh, 453552, India
| | - Biswarup Pathak
- Department of Chemistry, Indian Institute of Technology (IIT) Indore, Indore, Madhya Pradesh, 453552, India
| |
Collapse
|
34
|
Liu T, Zhou S, Wang T, Teng Y. Cyberbiosecurity: Advancements in DNA-based information security. BIOSAFETY AND HEALTH 2024; 6:251-256. [PMID: 40078663 PMCID: PMC11895033 DOI: 10.1016/j.bsheal.2024.06.002] [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: 01/31/2024] [Revised: 06/14/2024] [Accepted: 06/19/2024] [Indexed: 03/14/2025] Open
Abstract
Synthetic biology is a crucial component of the "cyber-biological revolution" in this new industrial revolution. Owing to breakthroughs in synthetic biology, deoxyribonucleic acid (DNA), the storehouse of hereditary material in biological systems, can now be used as a medium for storage (synthesis) and reading (sequencing) of information. However, integrating synthetic biology with computerization has also caused cyberbiosecurity concerns, encompassing biosecurity and information security issues. Malicious codes intended to attack computer systems can be stored as artificially synthesized DNA fragments, which can be released during DNA sequencing and decoding and attack computer and network systems. As these cyberbiosecurity threats become increasingly realistic, spreading awareness and information about how they can be prevented and controlled is crucial. This review aims to address this need by offering crucial theoretical backing for cyberbiosecurity research and raising awareness of risk mitigation and control measures in information security, biosecurity, and national security.
Collapse
Affiliation(s)
- Tuoyu Liu
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing 100071, China
- School of Life Sciences, Tianjin University, Tianjin 300072, China
| | - Sijie Zhou
- Frontiers Science Center for Synthetic Biology and Key Laboratory of Systems Bioengineering (Ministry of Education), School of Chemical Engineering and Technology, Tianjin University, Tianjin 300072, China
- Frontiers Research Institute for Synthetic Biology, Tianjin University, Tianjin 300072, China
| | - Tao Wang
- School of Life Sciences, Tianjin University, Tianjin 300072, China
| | - Yue Teng
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing 100071, China
| |
Collapse
|
35
|
Singh S, Ahmed AI, Almansoori S, Alameri S, Adlan A, Odivilas G, Chattaway MA, Salem SB, Brudecki G, Elamin W. A narrative review of wastewater surveillance: pathogens of concern, applications, detection methods, and challenges. Front Public Health 2024; 12:1445961. [PMID: 39139672 PMCID: PMC11319304 DOI: 10.3389/fpubh.2024.1445961] [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: 06/08/2024] [Accepted: 07/18/2024] [Indexed: 08/15/2024] Open
Abstract
Introduction The emergence and resurgence of pathogens have led to significant global health challenges. Wastewater surveillance has historically been used to track water-borne or fecal-orally transmitted pathogens, providing a sensitive means of monitoring pathogens within a community. This technique offers a comprehensive, real-time, and cost-effective approach to disease surveillance, especially for diseases that are difficult to monitor through individual clinical screenings. Methods This narrative review examines the current state of knowledge on wastewater surveillance, emphasizing important findings and techniques used to detect potential pathogens from wastewater. It includes a review of literature on the detection methods, the pathogens of concern, and the challenges faced in the surveillance process. Results Wastewater surveillance has proven to be a powerful tool for early warning and timely intervention of infectious diseases. It can detect pathogens shed by asymptomatic and pre-symptomatic individuals, providing an accurate population-level view of disease transmission. The review highlights the applications of wastewater surveillance in tracking key pathogens of concern, such as gastrointestinal pathogens, respiratory pathogens, and viruses like SARS-CoV-2. Discussion The review discusses the benefits of wastewater surveillance in public health, particularly its role in enhancing existing systems for infectious disease surveillance. It also addresses the challenges faced, such as the need for improved detection methods and the management of antimicrobial resistance. The potential for wastewater surveillance to inform public health mitigation strategies and outbreak response protocols is emphasized. Conclusion Wastewater surveillance is a valuable tool in the fight against infectious diseases. It offers a unique perspective on the spread and evolution of pathogens, aiding in the prevention and control of disease epidemics. This review underscores the importance of continued research and development in this field to overcome current challenges and maximize the potential of wastewater surveillance in public health.
Collapse
Affiliation(s)
- Surabhi Singh
- Microbiology Lab, Reference and Surveillance Intelligence Department, Abu Dhabi, United Arab Emirates
| | - Amina Ismail Ahmed
- Microbiology Lab, Reference and Surveillance Intelligence Department, Abu Dhabi, United Arab Emirates
| | - Sumayya Almansoori
- Microbiology Lab, Reference and Surveillance Intelligence Department, Abu Dhabi, United Arab Emirates
| | - Shaikha Alameri
- Microbiology Lab, Reference and Surveillance Intelligence Department, Abu Dhabi, United Arab Emirates
| | - Ashraf Adlan
- Microbiology Lab, Reference and Surveillance Intelligence Department, Abu Dhabi, United Arab Emirates
| | - Giovanni Odivilas
- Microbiology Lab, Reference and Surveillance Intelligence Department, Abu Dhabi, United Arab Emirates
| | - Marie Anne Chattaway
- United Kingdom Health Security Agency, Gastrointestinal Bacteria Reference Laboratory, London, United Kingdom
| | - Samara Bin Salem
- Central Testing Laboratory, Abu Dhabi Quality and Conformity Council, Abu Dhabi, United Arab Emirates
| | - Grzegorz Brudecki
- Microbiology Lab, Reference and Surveillance Intelligence Department, Abu Dhabi, United Arab Emirates
| | - Wael Elamin
- Microbiology Lab, Reference and Surveillance Intelligence Department, Abu Dhabi, United Arab Emirates
| |
Collapse
|
36
|
Arak H, Sahinoğlu B, Kus T. The effect of examining somatic alterations with NGS in patients with solid tumors, on patient management: A single-center experience in Turkey. J Oncol Pharm Pract 2024:10781552241266552. [PMID: 39053146 DOI: 10.1177/10781552241266552] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/27/2024]
Abstract
OBJECTIVE This study aimed to investigate the impact of analyzing somatic alterations using next-generation sequencing (NGS) on treatment management in patients with metastatic solid cancers and their ability to access NGS recommended treatments. METHODS This retrospective study included eligible patients who underwent NGS on somatic tumor tissue. We examined the clinical and pathological characteristics of these patients and the alterations in their treatment following NGS results. RESULTS A total of 101 patients who underwent NGS were included in the study. The most common cancers were non-small cell lung cancer (NSCLC), colorectal, and breast cancers, in that order. The median age was 58 (range 21-82) years, with 60 (59.4%) male participants. The median NGS turnaround time was 23 (range 17-29) days. NGS was performed on tissue from the primary lesion in 89(88%) patients. Predictive, prognostic, actionable, or variants of unknown significance were detected in 62(61.4%) patients. The most frequent variants identified were KRAS, EGFR, TP53, PIK3CA, and other rare mutations. Treatment was altered in 17(16.8%) patients based on NGS results. Of the 30 (29.7%) patients for whom NGS-informed treatment was recommended, only seven (6.9%) received the recommended therapy. There was no significant difference in overall survival (OS) between patients whose treatment was changed based on NGS results and those whose treatment remained unchanged (p = 0.897). There was no difference in OS between patients with and without variants (p = 0.384). CONCLUSIONS NGS analysis of somatic alterations in patients with metastatic cancer may reveal additional variants beyond those identified by baseline tests. However, based on the recommendations of the reimbursement institution in Turkey, only a limited number of patients are able to access treatments recommended by NGS results. Therefore, baseline tests established in Turkey need to be made available in more centers in an appropriate time.
Collapse
Affiliation(s)
- Hacı Arak
- Department of Medical Oncology, Gaziantep City Hospital, Gaziantep, Turkey
| | - Bahtiyar Sahinoğlu
- Genetic Diseases Diagnosis Center, Gaziantep City Hospital, Gaziantep, Turkey
| | - Tulay Kus
- Department of Medical Oncology, Gaziantep University School of Medicine, Gaziantep, Turkey
| |
Collapse
|
37
|
Rocha LGDN, Guimarães PAS, Carvalho MGR, Ruiz JC. Tumor Neoepitope-Based Vaccines: A Scoping Review on Current Predictive Computational Strategies. Vaccines (Basel) 2024; 12:836. [PMID: 39203962 PMCID: PMC11360805 DOI: 10.3390/vaccines12080836] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Revised: 07/09/2024] [Accepted: 07/11/2024] [Indexed: 09/03/2024] Open
Abstract
Therapeutic cancer vaccines have been considered in recent decades as important immunotherapeutic strategies capable of leading to tumor regression. In the development of these vaccines, the identification of neoepitopes plays a critical role, and different computational methods have been proposed and employed to direct and accelerate this process. In this context, this review identified and systematically analyzed the most recent studies published in the literature on the computational prediction of epitopes for the development of therapeutic vaccines, outlining critical steps, along with the associated program's strengths and limitations. A scoping review was conducted following the PRISMA extension (PRISMA-ScR). Searches were performed in databases (Scopus, PubMed, Web of Science, Science Direct) using the keywords: neoepitope, epitope, vaccine, prediction, algorithm, cancer, and tumor. Forty-nine articles published from 2012 to 2024 were synthesized and analyzed. Most of the identified studies focus on the prediction of epitopes with an affinity for MHC I molecules in solid tumors, such as lung carcinoma. Predicting epitopes with class II MHC affinity has been relatively underexplored. Besides neoepitope prediction from high-throughput sequencing data, additional steps were identified, such as the prioritization of neoepitopes and validation. Mutect2 is the most used tool for variant calling, while NetMHCpan is favored for neoepitope prediction. Artificial/convolutional neural networks are the preferred methods for neoepitope prediction. For prioritizing immunogenic epitopes, the random forest algorithm is the most used for classification. The performance values related to the computational models for the prediction and prioritization of neoepitopes are high; however, a large part of the studies still use microbiome databases for training. The in vitro/in vivo validations of the predicted neoepitopes were verified in 55% of the analyzed studies. Clinical trials that led to successful tumor remission were identified, highlighting that this immunotherapeutic approach can benefit these patients. Integrating high-throughput sequencing, sophisticated bioinformatics tools, and rigorous validation methods through in vitro/in vivo assays as well as clinical trials, the tumor neoepitope-based vaccine approach holds promise for developing personalized therapeutic vaccines that target specific tumor cancers.
Collapse
Affiliation(s)
- Luiz Gustavo do Nascimento Rocha
- Biologia Computacional e Sistemas (BCS), Instituto Oswaldo Cruz (IOC), Fundação Oswaldo Cruz, Rio de Janeiro 21040-900, Brazil; (L.G.d.N.R.); (P.A.S.G.)
- Grupo Informática de Biossistemas e Genômica, Instituto René Rachou, Fundação Oswaldo Cruz, Belo Horizonte 30190-002, Brazil
| | - Paul Anderson Souza Guimarães
- Biologia Computacional e Sistemas (BCS), Instituto Oswaldo Cruz (IOC), Fundação Oswaldo Cruz, Rio de Janeiro 21040-900, Brazil; (L.G.d.N.R.); (P.A.S.G.)
- Grupo Informática de Biossistemas e Genômica, Instituto René Rachou, Fundação Oswaldo Cruz, Belo Horizonte 30190-002, Brazil
| | - Maria Gabriela Reis Carvalho
- Biologia Computacional e Sistemas (BCS), Instituto Oswaldo Cruz (IOC), Fundação Oswaldo Cruz, Rio de Janeiro 21040-900, Brazil; (L.G.d.N.R.); (P.A.S.G.)
- Grupo Informática de Biossistemas e Genômica, Instituto René Rachou, Fundação Oswaldo Cruz, Belo Horizonte 30190-002, Brazil
| | - Jeronimo Conceição Ruiz
- Biologia Computacional e Sistemas (BCS), Instituto Oswaldo Cruz (IOC), Fundação Oswaldo Cruz, Rio de Janeiro 21040-900, Brazil; (L.G.d.N.R.); (P.A.S.G.)
- Grupo Informática de Biossistemas e Genômica, Instituto René Rachou, Fundação Oswaldo Cruz, Belo Horizonte 30190-002, Brazil
| |
Collapse
|
38
|
Alagarswamy K, Shi W, Boini A, Messaoudi N, Grasso V, Cattabiani T, Turner B, Croner R, Kahlert UD, Gumbs A. Should AI-Powered Whole-Genome Sequencing Be Used Routinely for Personalized Decision Support in Surgical Oncology—A Scoping Review. BIOMEDINFORMATICS 2024; 4:1757-1772. [DOI: 10.3390/biomedinformatics4030096] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/12/2025]
Abstract
In this scoping review, we delve into the transformative potential of artificial intelligence (AI) in addressing challenges inherent in whole-genome sequencing (WGS) analysis, with a specific focus on its implications in oncology. Unveiling the limitations of existing sequencing technologies, the review illuminates how AI-powered methods emerge as innovative solutions to surmount these obstacles. The evolution of DNA sequencing technologies, progressing from Sanger sequencing to next-generation sequencing, sets the backdrop for AI’s emergence as a potent ally in processing and analyzing the voluminous genomic data generated. Particularly, deep learning methods play a pivotal role in extracting knowledge and discerning patterns from the vast landscape of genomic information. In the context of oncology, AI-powered methods exhibit considerable potential across diverse facets of WGS analysis, including variant calling, structural variation identification, and pharmacogenomic analysis. This review underscores the significance of multimodal approaches in diagnoses and therapies, highlighting the importance of ongoing research and development in AI-powered WGS techniques. Integrating AI into the analytical framework empowers scientists and clinicians to unravel the intricate interplay of genomics within the realm of multi-omics research, paving the way for more successful personalized and targeted treatments.
Collapse
Affiliation(s)
| | - Wenjie Shi
- Department of General-, Visceral-, Vascular and Transplantation Surgery, University of Magdeburg, Haus 60a, Leipziger Str. 44, 39120 Magdeburg, Germany
| | - Aishwarya Boini
- Davao Medical School Foundation, Davao City 8000, Philippines
| | - Nouredin Messaoudi
- Department of Hepatopancreatobiliary Surgery, Vrije Universiteit Brussel (VUB), Universitair Ziekenhuis Brussel (UZ Brussel), Europe Hospitals, 1090 Brussels, Belgium
| | - Vincent Grasso
- Department of Electrical and Computer Engineering, University of New Mexico, Albuquerque, NM 87131, USA
| | | | | | - Roland Croner
- Department of General-, Visceral-, Vascular and Transplantation Surgery, University of Magdeburg, Haus 60a, Leipziger Str. 44, 39120 Magdeburg, Germany
| | - Ulf D. Kahlert
- Department of General-, Visceral-, Vascular and Transplantation Surgery, University of Magdeburg, Haus 60a, Leipziger Str. 44, 39120 Magdeburg, Germany
| | - Andrew Gumbs
- Department of General-, Visceral-, Vascular and Transplantation Surgery, University of Magdeburg, Haus 60a, Leipziger Str. 44, 39120 Magdeburg, Germany
- Talos Surgical, Inc., New Castle, DE 19720, USA
- Department of Surgery, American Hospital of Tbilisi, 0102 Tbilisi, Georgia
| |
Collapse
|
39
|
Muthaffar OY, Alazhary NW, Alyazidi AS, Alsubaie MA, Bahowarth SY, Odeh NB, Bamaga AK. Clinical description and evaluation of 30 pediatric patients with ultra-rare diseases: A multicenter study with real-world data from Saudi Arabia. PLoS One 2024; 19:e0307454. [PMID: 39024300 PMCID: PMC11257271 DOI: 10.1371/journal.pone.0307454] [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: 02/10/2024] [Accepted: 07/04/2024] [Indexed: 07/20/2024] Open
Abstract
BACKGROUND With the advancement of next-generation sequencing, clinicians are now able to detect ultra-rare mutations that are barely encountered by the majority of physicians. Ultra-rare and rare diseases cumulatively acquire a prevalence equivalent to type 2 diabetes with 80% being genetic in origin and more prevalent among high consanguinity communities including Saudi Arabia. The challenge of these diseases is the ability to predict their prevalence and define clear phenotypic features. METHODS This is a non-interventional retrospective multicenter study. We included pediatric patients with a pathogenic variant designated as ultra-rare according to the National Institute for Clinical Excellence's criteria. Demographic, clinical, laboratory, and radiological data of all patients were collected and analyzed using multinomial regression models. RESULTS We included 30 patients. Their mean age of diagnosis was 16.77 months (range 3-96 months) and their current age was 8.83 years (range = 2-15 years). Eleven patients were females and 19 were males. The majority were of Arab ethnicity (96.77%). Twelve patients were West-Saudis and 8 patients were South-Saudis. SCN1A mutation was reported among 19 patients. Other mutations included SZT2, ROGDI, PRF1, ATP1A3, and SHANK3. The heterozygous mutation was reported among 67.86%. Twenty-nine patients experienced seizures with GTC being the most frequently reported semiology. The mean response to ASMs was 45.50% (range 0-100%). CONCLUSION The results suggest that ultra-rare diseases must be viewed as a distinct category from rare diseases with potential demographic and clinical hallmarks. Additional objective and descriptive criteria to detect such cases are needed.
Collapse
Affiliation(s)
- Osama Y Muthaffar
- Department of Pediatric, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Noura W Alazhary
- Department of General Pediatric, Dr. Soliman Fakeeh Hospital, Jeddah, Saudi Arabia
| | - Anas S Alyazidi
- Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
| | | | - Sarah Y Bahowarth
- Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Nour B Odeh
- College of Medicine, Alfaisal University, Riyadh, Saudi Arabia
| | - Ahmed K Bamaga
- Department of Pediatric, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
| |
Collapse
|
40
|
Li L, Sun M, Wang J, Wan S. Multi-omics based artificial intelligence for cancer research. Adv Cancer Res 2024; 163:303-356. [PMID: 39271266 DOI: 10.1016/bs.acr.2024.06.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/15/2024]
Abstract
With significant advancements of next generation sequencing technologies, large amounts of multi-omics data, including genomics, epigenomics, transcriptomics, proteomics, and metabolomics, have been accumulated, offering an unprecedented opportunity to explore the heterogeneity and complexity of cancer across various molecular levels and scales. One of the promising aspects of multi-omics lies in its capacity to offer a holistic view of the biological networks and pathways underpinning cancer, facilitating a deeper understanding of its development, progression, and response to treatment. However, the exponential growth of data generated by multi-omics studies present significant analytical challenges. Processing, analyzing, integrating, and interpreting these multi-omics datasets to extract meaningful insights is an ambitious task that stands at the forefront of current cancer research. The application of artificial intelligence (AI) has emerged as a powerful solution to these challenges, demonstrating exceptional capabilities in deciphering complex patterns and extracting valuable information from large-scale, intricate omics datasets. This review delves into the synergy of AI and multi-omics, highlighting its revolutionary impact on oncology. We dissect how this confluence is reshaping the landscape of cancer research and clinical practice, particularly in the realms of early detection, diagnosis, prognosis, treatment and pathology. Additionally, we elaborate the latest AI methods for multi-omics integration to provide a comprehensive insight of the complex biological mechanisms and inherent heterogeneity of cancer. Finally, we discuss the current challenges of data harmonization, algorithm interpretability, and ethical considerations. Addressing these challenges necessitates a multidisciplinary collaboration, paving the promising way for more precise, personalized, and effective treatments for cancer patients.
Collapse
Affiliation(s)
- Lusheng Li
- Department of Genetics, Cell Biology and Anatomy, University of Nebraska Medical Center, Omaha, NE, United States
| | - Mengtao Sun
- Department of Genetics, Cell Biology and Anatomy, University of Nebraska Medical Center, Omaha, NE, United States
| | - Jieqiong Wang
- Department of Neurological Sciences, University of Nebraska Medical Center, Omaha, NE, United States
| | - Shibiao Wan
- Department of Genetics, Cell Biology and Anatomy, University of Nebraska Medical Center, Omaha, NE, United States.
| |
Collapse
|
41
|
Hayashi N, Mori S, Ohmoto A, Fukada I, Yamazaki M, Hosonaga M, Wang X, Ueki A, Kiyotani K, Tonooka A, Takeuchi K, Takahashi S. Availability of genome-matched therapy based on clinical practice. Int J Clin Oncol 2024; 29:964-971. [PMID: 38668785 PMCID: PMC11196305 DOI: 10.1007/s10147-024-02533-z] [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: 07/11/2023] [Accepted: 04/06/2024] [Indexed: 06/25/2024]
Abstract
BACKGROUND Comprehensive genomic profiling (CGP) provides new opportunities for patients with advanced cancer to receive genome-matched therapies, but the availability rate of these remains low. We reviewed our CGP cases and suggested possible strategies to improve the current status from a clinical perspective. METHODS Druggable genomic alterations and barriers to accessing genome-matched therapies were investigated in 653 patients with 30 various types of cancers who underwent CGP. RESULTS While the availability rate of genome-matched therapies as a whole was 9.5%, CGP was useful in some cancer types. Patients with thyroid cancer and lung cancer harbored druggable genomic alterations at high rates, while sarcoma rarely harbored these alterations (100%, 76%, and 15.2%, respectively). In contrast, the availability rate of genome-matched therapies was highest in patients with sarcoma and head and neck cancer (HNC) (60% and 40%, respectively). One hundred thirteen patients (63.5%) had multiple barriers to accessing genome-matched therapy. Of 178 patients, 21 patients (11.8%) could not be considered for genome-matched therapies solely because of the deterioration of their performance status. CONCLUSION This study demonstrated the usefulness of CGP for patients with sarcoma and HNC in addition to lung cancer in clinical practice. Performing CGP at the front line has the potential to improve the availability of genome-matched therapy.
Collapse
Affiliation(s)
- Naomi Hayashi
- Department of Genomic Medicine, Cancer Institute Hospital of Japanese Foundation for Cancer Research, 3-8-31 Ariake, Koto, Tokyo, 135-8550, Japan.
- Department of Clinical Genetic Oncology, Cancer Institute Hospital of Japanese Foundation for Cancer Research, 3-8-31 Ariake, Koto, Tokyo, 135-8550, Japan.
| | - Seiichi Mori
- Project for Development of Innovative Research On Cancer Therapeutics, Cancer Precision Medicine Center, Japanese Foundation for Cancer Research, 3-8-31 Ariake, Koto, Tokyo, 135-8550, Japan
| | - Akihiro Ohmoto
- Department of Medical Oncology, Cancer Institute Hospital of Japanese Foundation for Cancer Research, 3-8-31 Ariake, Koto, Tokyo, 135-8550, Japan
- Memorial Sloan Kettering Cancer Center, Human Oncology and Pathogenesis Program, 1275 York Avenue, New York, NY, 10065, USA
| | - Ippei Fukada
- Department of Genomic Medicine, Cancer Institute Hospital of Japanese Foundation for Cancer Research, 3-8-31 Ariake, Koto, Tokyo, 135-8550, Japan
- Department of Breast Medical Oncology, Cancer Institute Hospital of Japanese Foundation for Cancer Research, 3-8-31 Ariake, Koto, Tokyo, 135-8550, Japan
| | - Masumi Yamazaki
- Department of Genomic Medicine, Cancer Institute Hospital of Japanese Foundation for Cancer Research, 3-8-31 Ariake, Koto, Tokyo, 135-8550, Japan
- Center for Advanced Medical Development, Cancer Institute Hospital of Japanese Foundation for Cancer Research, 3-8-31 Ariake, Koto, Tokyo, 135-8550, Japan
| | - Mari Hosonaga
- Department of Breast Medical Oncology, Cancer Institute Hospital of Japanese Foundation for Cancer Research, 3-8-31 Ariake, Koto, Tokyo, 135-8550, Japan
| | - Xiaofei Wang
- Department of Medical Oncology, Cancer Institute Hospital of Japanese Foundation for Cancer Research, 3-8-31 Ariake, Koto, Tokyo, 135-8550, Japan
| | - Arisa Ueki
- Department of Clinical Genetic Oncology, Cancer Institute Hospital of Japanese Foundation for Cancer Research, 3-8-31 Ariake, Koto, Tokyo, 135-8550, Japan
| | - Kazuma Kiyotani
- Project for Immunogenomics, Cancer Precision Medicine Center, Japanese Foundation for Cancer Research, 3-8-31 Ariake, Koto, Tokyo, 135-8550, Japan
- Laboratory of Immunogenomics, Center for Intractable Diseases and ImmunoGenomics (CiDIG), Health and Nutrition (NIBIOHN), National Institutes of Biomedical Innovation, 7-6-8, Saito-Asagi, Ibaraki City, Osaka, 567-0085, Japan
| | - Akiko Tonooka
- Division of Pathology, Cancer Institute, Japanese Foundation for Cancer Research, 3-8-31 Ariake, Koto, Tokyo, 135-8550, Japan
- Department of Pathology, Cancer Institute Hospital, Japanese Foundation for Cancer Research, 3-8-31 Ariake, Koto, Tokyo, 135-8550, Japan
| | - Kengo Takeuchi
- Division of Pathology, Cancer Institute, Japanese Foundation for Cancer Research, 3-8-31 Ariake, Koto, Tokyo, 135-8550, Japan
- Department of Pathology, Cancer Institute Hospital, Japanese Foundation for Cancer Research, 3-8-31 Ariake, Koto, Tokyo, 135-8550, Japan
- Pathology Project for Molecular Targets, Cancer Institute, Japanese Foundation for Cancer Research, 3-8-31 Ariake, Koto, Tokyo, 135-8550, Japan
| | - Shunji Takahashi
- Department of Genomic Medicine, Cancer Institute Hospital of Japanese Foundation for Cancer Research, 3-8-31 Ariake, Koto, Tokyo, 135-8550, Japan
- Department of Medical Oncology, Cancer Institute Hospital of Japanese Foundation for Cancer Research, 3-8-31 Ariake, Koto, Tokyo, 135-8550, Japan
- Center for Advanced Medical Development, Cancer Institute Hospital of Japanese Foundation for Cancer Research, 3-8-31 Ariake, Koto, Tokyo, 135-8550, Japan
| |
Collapse
|
42
|
Rolando JC, Melkonian AV, Walt DR. The Present and Future Landscapes of Molecular Diagnostics. ANNUAL REVIEW OF ANALYTICAL CHEMISTRY (PALO ALTO, CALIF.) 2024; 17:459-474. [PMID: 38360553 DOI: 10.1146/annurev-anchem-061622-015112] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/17/2024]
Abstract
Nucleic acid testing is the cornerstone of modern molecular diagnostics. This review describes the current status and future directions of molecular diagnostics, focusing on four major techniques: polymerase chain reaction (PCR), next-generation sequencing (NGS), isothermal amplification methods such as recombinase polymerase amplification (RPA) and loop-mediated isothermal amplification (LAMP), and clustered regularly interspaced short palindromic repeats (CRISPR)-based detection methods. We explore the advantages and limitations of each technique, describe how each overlaps with or complements other techniques, and examine current clinical offerings. This review provides a broad perspective into the landscape of molecular diagnostics and highlights potential future directions in this rapidly evolving field.
Collapse
Affiliation(s)
- Justin C Rolando
- 1Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts, USA;
- 2Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, Massachusetts, USA
- 3Harvard Medical School, Harvard University, Boston, Massachusetts, USA
| | - Arek V Melkonian
- 1Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts, USA;
- 2Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, Massachusetts, USA
- 3Harvard Medical School, Harvard University, Boston, Massachusetts, USA
| | - David R Walt
- 1Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts, USA;
- 2Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, Massachusetts, USA
- 3Harvard Medical School, Harvard University, Boston, Massachusetts, USA
| |
Collapse
|
43
|
Vatopoulos A. Clinical Microbiology: where do we stand? FRONTIERS IN ANTIBIOTICS 2024; 3:1250632. [PMID: 39816265 PMCID: PMC11731818 DOI: 10.3389/frabi.2024.1250632] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Accepted: 04/29/2024] [Indexed: 01/18/2025]
Abstract
Clinical Microbiology has developed during the last 100 years, simultaneous with the discovery of microorganisms as causes of infections. Globalization and One Health determine present needs whereas molecular biology, automation, artificial intelligence, and bioinformatics are new tools that characterize the new developments in the field.
Collapse
Affiliation(s)
- Alkiviadis Vatopoulos
- Department of Public Health Policy, School of Public Health, University of West Attica, Athens, Greece
| |
Collapse
|
44
|
Mikziński P, Kraus K, Widelski J, Paluch E. Modern Microbiological Methods to Detect Biofilm Formation in Orthopedy and Suggestions for Antibiotic Therapy, with Particular Emphasis on Prosthetic Joint Infection (PJI). Microorganisms 2024; 12:1198. [PMID: 38930580 PMCID: PMC11205407 DOI: 10.3390/microorganisms12061198] [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: 05/12/2024] [Revised: 06/05/2024] [Accepted: 06/11/2024] [Indexed: 06/28/2024] Open
Abstract
Biofilm formation is a serious problem that relatively often causes complications in orthopedic surgery. Biofilm-forming pathogens invade implanted foreign bodies and surrounding tissues. Such a condition, if not limited at the appropriate time, often requires reoperation. This can be partially prevented by selecting an appropriate prosthesis material that prevents the development of biofilm. There are many modern techniques available to detect the formed biofilm. By applying them we can identify and visualize biofilm-forming microorganisms. The most common etiological factors associated with biofilms in orthopedics are: Staphylococcus aureus, coagulase-negative Staphylococci (CoNS), and Enterococcus spp., whereas Gram-negative bacilli and Candida spp. also deserve attention. It seems crucial, for therapeutic success, to eradicate the microorganisms able to form biofilm after the implantation of endoprostheses. Planning the effective targeted antimicrobial treatment of postoperative infections requires accurate identification of the microorganism responsible for the complications of the procedure. The modern microbiological testing techniques described in this article show the diagnostic options that can be followed to enable the implementation of effective treatment.
Collapse
Affiliation(s)
- Paweł Mikziński
- Faculty of Medicine, Wroclaw Medical University, Wyb. Pasteura 1, 50-376 Wroclaw, Poland; (P.M.); (K.K.)
| | - Karolina Kraus
- Faculty of Medicine, Wroclaw Medical University, Wyb. Pasteura 1, 50-376 Wroclaw, Poland; (P.M.); (K.K.)
| | - Jarosław Widelski
- Department of Pharmacognosy with Medicinal Plants Garden, Lublin Medical University, 20-093 Lublin, Poland;
| | - Emil Paluch
- Department of Microbiology, Faculty of Medicine, Wroclaw Medical University, Tytusa Chalubinskiego 4, 50-376 Wroclaw, Poland
| |
Collapse
|
45
|
Pace-Loscos T, Gal J, Contu S, Schiappa R, Chamorey E, Culié D. StatiCAL: an interactive tool for statistical analysis of biomedical data and scientific valorization. BMC Bioinformatics 2024; 25:210. [PMID: 38867185 PMCID: PMC11167775 DOI: 10.1186/s12859-024-05829-z] [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: 03/07/2024] [Accepted: 06/10/2024] [Indexed: 06/14/2024] Open
Abstract
BACKGROUND In the realm of biomedical research, the growing volume, diversity and quantity of data has escalated the demand for statistical analysis as it is indispensable for synthesizing, interpreting, and publishing data. Hence the need for accessible analysis tools drastically increased. StatiCAL emerges as a user-friendly solution, enabling researchers to conduct basic analyses without necessitating extensive programming expertise. RESULTS StatiCAL includes divers functionalities: data management, visualization on variables and statistical analysis. Data management functionalities allow the user to freely add or remove variables, to select sub-population and to visualise selected data to better perform the analysis. With this tool, users can freely perform statistical analysis such as descriptive, graphical, univariate, and multivariate analysis. All of this can be performed without the need to learn R coding as the software is a graphical user interface where all the action can be performed by clicking a button. CONCLUSIONS StatiCAL represents a valuable contribution to the field of biomedical research. By being open-access and by providing an intuitive interface with robust features, StatiCAL allow researchers to gain autonomy in conducting their projects.
Collapse
Affiliation(s)
- Tanguy Pace-Loscos
- Département d'Epidémiologie, de Biostatistique et des Données de Santé, Centre Antoine Lacassagne, Université Cote d'Azur, Nice, France.
| | - Jocelyn Gal
- Département d'Epidémiologie, de Biostatistique et des Données de Santé, Centre Antoine Lacassagne, Université Cote d'Azur, Nice, France
| | - Sara Contu
- Département d'Epidémiologie, de Biostatistique et des Données de Santé, Centre Antoine Lacassagne, Université Cote d'Azur, Nice, France
| | - Renaud Schiappa
- Département d'Epidémiologie, de Biostatistique et des Données de Santé, Centre Antoine Lacassagne, Université Cote d'Azur, Nice, France
| | - Emmanuel Chamorey
- Département d'Epidémiologie, de Biostatistique et des Données de Santé, Centre Antoine Lacassagne, Université Cote d'Azur, Nice, France
| | - Dorian Culié
- Institut Universitaire de la Face et du Cou, Centre Antoine Lacassagne, Département de Chirurgie Cervico-Faciale, Université Cote d'Azur, 06103, Nice, France
| |
Collapse
|
46
|
Teixeira M, Silva F, Ferreira RM, Pereira T, Figueiredo C, Oliveira HP. A review of machine learning methods for cancer characterization from microbiome data. NPJ Precis Oncol 2024; 8:123. [PMID: 38816569 PMCID: PMC11139966 DOI: 10.1038/s41698-024-00617-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Accepted: 05/17/2024] [Indexed: 06/01/2024] Open
Abstract
Recent studies have shown that the microbiome can impact cancer development, progression, and response to therapies suggesting microbiome-based approaches for cancer characterization. As cancer-related signatures are complex and implicate many taxa, their discovery often requires Machine Learning approaches. This review discusses Machine Learning methods for cancer characterization from microbiome data. It focuses on the implications of choices undertaken during sample collection, feature selection and pre-processing. It also discusses ML model selection, guiding how to choose an ML model, and model validation. Finally, it enumerates current limitations and how these may be surpassed. Proposed methods, often based on Random Forests, show promising results, however insufficient for widespread clinical usage. Studies often report conflicting results mainly due to ML models with poor generalizability. We expect that evaluating models with expanded, hold-out datasets, removing technical artifacts, exploring representations of the microbiome other than taxonomical profiles, leveraging advances in deep learning, and developing ML models better adapted to the characteristics of microbiome data will improve the performance and generalizability of models and enable their usage in the clinic.
Collapse
Affiliation(s)
- Marco Teixeira
- Institute for Systems and Computer Engineering, Technology and Science, Porto, Portugal.
- Faculty of Engineering, University of Porto, Porto, Portugal.
| | - Francisco Silva
- Institute for Systems and Computer Engineering, Technology and Science, Porto, Portugal
- Faculty of Science, University of Porto, Porto, Portugal
| | - Rui M Ferreira
- Ipatimup - Institute of Molecular Pathology and Immunology of the University of Porto, Porto, Portugal
- Instituto de Investigação e Inovação em Saúde, University of Porto, Porto, Portugal
| | - Tania Pereira
- Institute for Systems and Computer Engineering, Technology and Science, Porto, Portugal
- Faculty of Sciences and Technology, University of Coimbra, Coimbra, Portugal
| | - Ceu Figueiredo
- Ipatimup - Institute of Molecular Pathology and Immunology of the University of Porto, Porto, Portugal
- Instituto de Investigação e Inovação em Saúde, University of Porto, Porto, Portugal
- Faculty of Medicine, University of Porto, Porto, Portugal
| | - Hélder P Oliveira
- Institute for Systems and Computer Engineering, Technology and Science, Porto, Portugal
- Faculty of Science, University of Porto, Porto, Portugal
| |
Collapse
|
47
|
Shokoohi F, Stephens DA, Greenwood CMT. Identifying Differential Methylation in Cancer Epigenetics via a Bayesian Functional Regression Model. Biomolecules 2024; 14:639. [PMID: 38927043 PMCID: PMC11201607 DOI: 10.3390/biom14060639] [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: 04/01/2024] [Revised: 05/20/2024] [Accepted: 05/20/2024] [Indexed: 06/28/2024] Open
Abstract
DNA methylation plays an essential role in regulating gene activity, modulating disease risk, and determining treatment response. We can obtain insight into methylation patterns at a single-nucleotide level via next-generation sequencing technologies. However, complex features inherent in the data obtained via these technologies pose challenges beyond the typical big data problems. Identifying differentially methylated cytosines (dmc) or regions is one such challenge. We have developed DMCFB, an efficient dmc identification method based on Bayesian functional regression, to tackle these challenges. Using simulations, we establish that DMCFB outperforms current methods and results in better smoothing and efficient imputation. We analyzed a dataset of patients with acute promyelocytic leukemia and control samples. With DMCFB, we discovered many new dmcs and, more importantly, exhibited enhanced consistency of differential methylation within islands and their adjacent shores. Additionally, we detected differential methylation at more of the binding sites of the fused gene involved in this cancer.
Collapse
Affiliation(s)
- Farhad Shokoohi
- Department of Mathematical Sciences, University of Nevada Las Vegas, Las Vegas, NV 89154, USA
| | - David A. Stephens
- Department of Mathematics and Statistics, McGill University, Montreal, QC H3A 0B9, Canada;
| | - Celia M. T. Greenwood
- Lady Davis Institute for Medical Research, Montreal, QC H3T 1E2, Canada;
- Gerald Bronfman Department of Oncology, McGill University, Montreal, QC H4A 3T2, Canada
- Department of Epidemiology, Biostatistics & Occupational Health, McGill University, Montreal, QC H3A 1G1, Canada
| |
Collapse
|
48
|
Chen X, Chen X, Lai Y. Development and emerging trends of drug resistance mutations in HIV: a bibliometric analysis based on CiteSpace. Front Microbiol 2024; 15:1374582. [PMID: 38812690 PMCID: PMC11133539 DOI: 10.3389/fmicb.2024.1374582] [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: 01/22/2024] [Accepted: 05/06/2024] [Indexed: 05/31/2024] Open
Abstract
Background Antiretroviral therapy has led to AIDS being a chronic disease. Nevertheless, the presence of constantly emerging drug resistance mutations poses a challenge to clinical treatment. A systematic analysis to summarize the advancements and uncharted territory of drug resistance mutations is urgently needed and may provide new clues for solving this problem. Methods We gathered 3,694 publications on drug resistance mutations from the Web of Science Core Collection with CiteSpace software and performed an analysis to visualize the results and predict future new directions and emerging trends. Betweenness centrality, count, and burst value were taken as standards. Results The number of papers on HIV medication resistance mutations during the last 10 years shows a wave-like trend. In terms of nation, organization, and author, the United States (1449), University of London (193), and Mark A. Wainberg (66) are the most significant contributors. The most frequently cited article is "Drug resistance mutations for surveillance of transmitted HIV-1 drug-resistance: 2009 update." Hot topics in this field include "next-generation sequencing," "tenofovir alafenamide," "children," "regimens," "accumulation," "dolutegravir," "rilpivirine," "sex," "pretreatment drug resistance," and "open label." Research on drug resistance in teenagers, novel mutation detection techniques, and drug development is ongoing, and numerous publications have indicated the presence of mutations related to current medications. Therefore, testing must be performed regularly for patients who have used medications for a long period. Additionally, by choosing medications with a longer half-life, patients can take fewer doses of their prescription, increasing patient compliance. Conclusion This study involved a bibliometric visualization analysis of the literature on drug resistance mutations, providing insight into the field's evolution and emerging patterns and offering academics a resource to better understand HIV drug resistance mutations and contribute to the field's advancement.
Collapse
Affiliation(s)
- Xuannan Chen
- Acupunture and Tuina School, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xi Chen
- School of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yu Lai
- School of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
49
|
Church J. The Natural History of Hereditary Colorectal Cancer Syndromes: From Phenotype to Genotype? Where Do We Stand and What Does the Future Hold? Clin Colon Rectal Surg 2024; 37:127-132. [PMID: 38606050 PMCID: PMC11006442 DOI: 10.1055/s-0043-1770380] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/13/2024]
Abstract
Applying the concept of a "natural history" to hereditary colorectal cancer is an interesting exercise because the way the syndromes are approached has changed so drastically. However, the exercise is instructive as it forces us to think in depth about where we are, where we have been, and, most helpfully, about where we may be going. In this article the diagnosis, along with endoscopic and surgical management of hereditary colorectal cancer are discussed in the context of their history and the changes in genomics and technology that have occurred over the last one hundred years.
Collapse
Affiliation(s)
- James Church
- Division of Colorectal Surgery, Department of Surgery, Columbia University Medical Center, New York, New York
| |
Collapse
|
50
|
Yen PW, Chen YA, Wang W, Mao FS, Chao CT, Chiang CK, Lin SH, Tarng DC, Chiu YW, Wu MJ, Chen YC, Kao JTW, Wu MS, Lin CL, Huang JW, Hung KY. The screening, diagnosis, and management of patients with autosomal dominant polycystic kidney disease: A national consensus statement from Taiwan. Nephrology (Carlton) 2024; 29:245-258. [PMID: 38462235 DOI: 10.1111/nep.14287] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 01/29/2024] [Accepted: 02/25/2024] [Indexed: 03/12/2024]
Abstract
Autosomal dominant polycystic kidney disease (ADPKD) is the most common inherited cause of end-stage kidney disease (ESKD) worldwide. Guidelines for the diagnosis and management of ADPKD in Taiwan remains unavailable. In this consensus statement, we summarize updated information on clinical features of international and domestic patients with ADPKD, followed by suggestions for optimal diagnosis and care in Taiwan. Specifically, counselling for at-risk minors and reproductive issues can be important, including ethical dilemmas surrounding prenatal diagnosis and pre-implantation genetic diagnosis. Studies reveal that ADPKD typically remains asymptomatic until the fourth decade of life, with symptoms resulting from cystic expansion with visceral compression, or rupture. The diagnosis can be made based on a detailed family history, followed by imaging studies (ultrasound, computed tomography, or magnetic resonance imaging). Genetic testing is reserved for atypical cases mostly. Common tools for prognosis prediction include total kidney volume, Mayo classification and PROPKD/genetic score. Screening and management of complications such as hypertension, proteinuria, urological infections, intracranial aneurysms, are also crucial for improving outcome. We suggest that the optimal management strategies of patients with ADPKD include general medical care, dietary recommendations and ADPKD-specific treatments. Key points include rigorous blood pressure control, dietary sodium restriction and Tolvaptan use, whereas the evidence for somatostatin analogues and mammalian target of rapamycin (mTOR) inhibitors remains limited. In summary, we outline an individualized care plan emphasizing careful monitoring of disease progression and highlight the need for shared decision-making among these patients.
Collapse
Affiliation(s)
- Pao-Wen Yen
- Division of Nephrology, Department of Internal Medicine, National Taiwan University Hospital and College of Medicine, Taipei, Taiwan
| | - Yung-An Chen
- Division of Nephrology, Department of Internal Medicine, National Taiwan University Hospital and College of Medicine, Taipei, Taiwan
| | - Wei Wang
- Division of Nephrology, Department of Internal Medicine, National Taiwan University Hospital and College of Medicine, Taipei, Taiwan
| | - Fang-Sheng Mao
- Division of Nephrology, Department of Internal Medicine, National Taiwan University Hospital and College of Medicine, Taipei, Taiwan
| | - Chia-Ter Chao
- Division of Nephrology, Department of Internal Medicine, National Taiwan University Hospital and College of Medicine, Taipei, Taiwan
- Division of Nephrology, Department of Internal Medicine, Min-Sheng General Hospital, Taoyuan City, Taiwan
| | - Chih-Kang Chiang
- Division of Nephrology, Department of Internal Medicine, National Taiwan University Hospital and College of Medicine, Taipei, Taiwan
| | - Shih-Hua Lin
- Division of Nephrology, Department of Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Der-Cherng Tarng
- Division of Nephrology, Department of Internal Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Yi-Wen Chiu
- Division of Nephrology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Ming-Ju Wu
- Division of Nephrology, Department of Internal Medicine, Taichung Veterans General Hospital, Taichung City, Taiwan
| | - Yung-Chang Chen
- Kidney Research Center, Department of Nephrology, Linkou Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Juliana Tze-Wah Kao
- Division of Nephrology, Department of Internal Medicine, Taipei Medical University-Shuang-Ho Hospital, Ministry of Health and Welfare, New Taipei City, Taiwan
- Division of Nephrology, Department of Internal Medicine, Fu-Jen Catholic University Hospital, Fu-Jen Catholic University, New Taipei City, Taiwan
| | - Mai-Szu Wu
- Division of Nephrology, Department of Internal Medicine, Taipei Medical University-Shuang-Ho Hospital, Ministry of Health and Welfare, New Taipei City, Taiwan
- Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Chun-Liang Lin
- Division of Nephrology, Department of Internal Medicine, Chia-Yi Chang Gung Memorial Hospital, Chia-Yi County, Taiwan
| | - Jenq-Wen Huang
- Division of Nephrology, Department of Internal Medicine, National Taiwan University Hospital and College of Medicine, Taipei, Taiwan
| | - Kuan-Yu Hung
- Division of Nephrology, Department of Internal Medicine, National Taiwan University Hospital and College of Medicine, Taipei, Taiwan
- Division of Nephrology, Department of Internal Medicine, Taipei Medical University-Shuang-Ho Hospital, Ministry of Health and Welfare, New Taipei City, Taiwan
| |
Collapse
|