1
|
Zardab M, Grose RP, Kocher HM. AHNAK2: a potential diagnostic biomarker for pancreatic cancer related to cellular motility. Sci Rep 2025; 15:2934. [PMID: 39849106 PMCID: PMC11757713 DOI: 10.1038/s41598-025-87337-5] [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: 09/16/2024] [Accepted: 01/16/2025] [Indexed: 01/25/2025] Open
Abstract
Pancreatic ductal adenocarcinoma lacks suitable biomarkers for early diagnosis of disease. In gene panels developed for early diagnosis of pancreatic cancer, high AHNAK2 mRNA expression was one possible biomarker. In silico analysis of published human sample datasets (n = 177) and ex vivo analysis of human plasma samples (n = 30 PDAC with matched 30 healthy control) suggested AHNAK2 could be a diagnostic biomarker. At a plasma level of 421.47 ng/ml, AHNAK2 could potentially diagnose PDAC with a specificity and sensitivity of 83.33% and 86.67%. In vitro analysis suggests that in cell lines with diffuse cytoplasmic distribution of AHNAK2, there was colocalization of AHNAK2 with Cortactin in filipodia. This colocalization increased when cells were cultured on substrates such as Fibronectin and Collagen, as well as in hypoxia, and resulted in an augmented invasion of cancer cells. However, in cell lines with a vesicular AHNAK2 staining, such changes were not observed. Our study posits AHNAK2 as a valuable diagnostic biomarker in PDAC, now demanding prospective validation. Determination of mechanisms regulating AHNAK2 subcellular localisation may help explain its biological role.
Collapse
Affiliation(s)
- Mohamed Zardab
- Centre for Tumour Biology, Barts Cancer Institute, Queen Mary University of London, Charterhouse Square, London, EC1M 6BQ, UK
- Barts and the London HPB Centre, The Royal London Hospital, Barts Health NHS Trust, London, UK
| | - Richard P Grose
- Centre for Tumour Biology, Barts Cancer Institute, Queen Mary University of London, Charterhouse Square, London, EC1M 6BQ, UK
| | - Hemant M Kocher
- Centre for Tumour Biology, Barts Cancer Institute, Queen Mary University of London, Charterhouse Square, London, EC1M 6BQ, UK.
- Barts and the London HPB Centre, The Royal London Hospital, Barts Health NHS Trust, London, UK.
| |
Collapse
|
2
|
Kadela K, Oleś H, Kosowska K, Liberda-Matyja D, Wiatrowska W, Cader K, Grabowska N, Piana K, Tott S, Roman M, Kozioł-Bohatkiewicz P, Wróbel TP. High-resolution infrared imaging performance of new high numerical aperture Schwarzschild objectives. SPECTROCHIMICA ACTA. PART A, MOLECULAR AND BIOMOLECULAR SPECTROSCOPY 2025; 331:125769. [PMID: 39893848 DOI: 10.1016/j.saa.2025.125769] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Revised: 01/15/2025] [Accepted: 01/18/2025] [Indexed: 02/04/2025]
Abstract
Mid-Infrared (IR) microspectroscopy is an advanced technique that enables the examination of samples providing micrometer-level spatial resolution and generating detailed chemical imaging of the samples. However, despite its advantages, spatial resolution in far-field optical microscopy and image resolution is limited by diffraction and there is a constant desire to push this limitation further. High numerical aperture (NA) objectives have been available for decades but primarily in the form of refractive lenses, however, these lack broadband capability, as they introduce chromatic aberrations. A reflective type objectives avoid this problem at the expense of lower NA and recently reflective objectives with an improved design and NA ranging from 0.7 to 0.8 were released. This study characterized the spatial resolution and signal to noise ratio (SNR) of new high numerical aperture infrared reflective objectives of NA = 0.7 (20x) and NA = 0.78 (40x) and compared their performance to the commonly used 15x (NA = 0.4) and 36x (NA = 0.52) objectives on a set of polymer and tissue samples using a benchtop IR microscope. It was found that the spatial resolution reaches values below 3 µm for the highest NA objective and approximately 6 µm for the lowest NA objective for shortest wavelengths. At the same time, the high-NA objectives provided better SNR compared to a similar magnification objective in both transmission and transflection geometry. Finally, a 3 µm PMMA sphere was successfully imaged using a conventional IR source (not a synchrotron or a laser), giving an impressive performance of the objectives.
Collapse
Affiliation(s)
- Karolina Kadela
- Solaris National Synchrotron Radiation Centre, Jagiellonian University, 98 Czerwone Maki Str. 30-392 Krakow, Poland; Łukasiewicz Research Network - Krakow Institute of Technology, 73 Zakopiańska Str. 30-418 Krakow, Poland; Doctoral School of Exact and Natural Sciences, 11 Łojasiewicza Str. 30-348 Krakow, Poland
| | - Honorata Oleś
- Solaris National Synchrotron Radiation Centre, Jagiellonian University, 98 Czerwone Maki Str. 30-392 Krakow, Poland
| | - Karolina Kosowska
- Solaris National Synchrotron Radiation Centre, Jagiellonian University, 98 Czerwone Maki Str. 30-392 Krakow, Poland
| | - Danuta Liberda-Matyja
- Solaris National Synchrotron Radiation Centre, Jagiellonian University, 98 Czerwone Maki Str. 30-392 Krakow, Poland; Doctoral School of Exact and Natural Sciences, 11 Łojasiewicza Str. 30-348 Krakow, Poland
| | - Wiktoria Wiatrowska
- Solaris National Synchrotron Radiation Centre, Jagiellonian University, 98 Czerwone Maki Str. 30-392 Krakow, Poland
| | - Kinga Cader
- Solaris National Synchrotron Radiation Centre, Jagiellonian University, 98 Czerwone Maki Str. 30-392 Krakow, Poland
| | - Natalia Grabowska
- Solaris National Synchrotron Radiation Centre, Jagiellonian University, 98 Czerwone Maki Str. 30-392 Krakow, Poland
| | - Kaja Piana
- Solaris National Synchrotron Radiation Centre, Jagiellonian University, 98 Czerwone Maki Str. 30-392 Krakow, Poland
| | - Szymon Tott
- Solaris National Synchrotron Radiation Centre, Jagiellonian University, 98 Czerwone Maki Str. 30-392 Krakow, Poland
| | - Maciej Roman
- Solaris National Synchrotron Radiation Centre, Jagiellonian University, 98 Czerwone Maki Str. 30-392 Krakow, Poland
| | - Paulina Kozioł-Bohatkiewicz
- Solaris National Synchrotron Radiation Centre, Jagiellonian University, 98 Czerwone Maki Str. 30-392 Krakow, Poland; Institute of Physics, Jagiellonian University, 11 Łojasiewicza Str. 30-348 Krakow, Poland
| | - Tomasz P Wróbel
- Solaris National Synchrotron Radiation Centre, Jagiellonian University, 98 Czerwone Maki Str. 30-392 Krakow, Poland.
| |
Collapse
|
3
|
Wu M, Tao H, Xu T, Zheng X, Wen C, Wang G, Peng Y, Dai Y. Spatial proteomics: unveiling the multidimensional landscape of protein localization in human diseases. Proteome Sci 2024; 22:7. [PMID: 39304896 DOI: 10.1186/s12953-024-00231-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Accepted: 09/01/2024] [Indexed: 09/22/2024] Open
Abstract
Spatial proteomics is a multidimensional technique that studies the spatial distribution and function of proteins within cells or tissues across both spatial and temporal dimensions. This field multidimensionally reveals the complex structure of the human proteome, including the characteristics of protein spatial distribution, dynamic protein translocation, and protein interaction networks. Recently, as a crucial method for studying protein spatial localization, spatial proteomics has been applied in the clinical investigation of various diseases. This review summarizes the fundamental concepts and characteristics of tissue-level spatial proteomics, its research progress in common human diseases such as cancer, neurological disorders, cardiovascular diseases, autoimmune diseases, and anticipates its future development trends. The aim is to highlight the significant impact of spatial proteomics on understanding disease pathogenesis, advancing diagnostic methods, and developing potential therapeutic targets in clinical research.
Collapse
Affiliation(s)
- Mengyao Wu
- School of Medicine, Anhui University of Science & Technology, Huainan, China
| | - Huihui Tao
- School of Medicine, Anhui University of Science & Technology, Huainan, China.
- Key Laboratory of Industrial Dust Deep Reduction and Occupational Health and Safety of Anhui Higher Education Institutes, Huainan, China.
- Anhui Province Engineering Laboratory of Occupational Health and Safety, Huainan, China.
| | - Tiantian Xu
- School of Medicine, Anhui University of Science & Technology, Huainan, China
| | - Xuejia Zheng
- The First Hospital of Anhui University of Science and Technology, Huainan, China
| | - Chunmei Wen
- School of Medicine, Anhui University of Science & Technology, Huainan, China
| | - Guoying Wang
- School of Medicine, Anhui University of Science & Technology, Huainan, China
| | - Yali Peng
- School of Medicine, Anhui University of Science & Technology, Huainan, China
| | - Yong Dai
- School of Medicine, Anhui University of Science & Technology, Huainan, China
- The First Hospital of Anhui University of Science and Technology, Huainan, China
- Joint Research Center for Occupational Medicine and Health of IHM, Anhui University of Science and Technology, Huainan, China
| |
Collapse
|
4
|
Dressler FF, Diedrichs F, Sabtan D, Hinrichs S, Krisp C, Gemoll T, Hennig M, Mackedanz P, Schlotfeldt M, Voß H, Offermann A, Kirfel J, Roesch MC, Struck JP, Kramer MW, Merseburger AS, Gratzke C, Schoeb DS, Miernik A, Schlüter H, Wetterauer U, Zubarev R, Perner S, Wolf P, Végvári Á. Proteomic analysis of the urothelial cancer landscape. Nat Commun 2024; 15:4513. [PMID: 38802361 PMCID: PMC11130393 DOI: 10.1038/s41467-024-48096-5] [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: 09/02/2023] [Accepted: 04/22/2024] [Indexed: 05/29/2024] Open
Abstract
Urothelial bladder cancer (UC) has a wide tumor biological spectrum with challenging prognostic stratification and relevant therapy-associated morbidity. Most molecular classifications relate only indirectly to the therapeutically relevant protein level. We improve the pre-analytics of clinical samples for proteome analyses and characterize a cohort of 434 samples with 242 tumors and 192 paired normal mucosae covering the full range of UC. We evaluate sample-wise tumor specificity and rank biomarkers by target relevance. We identify robust proteomic subtypes with prognostic information independent from histopathological groups. In silico drug prediction suggests efficacy of several compounds hitherto not in clinical use. Both in silico and in vitro data indicate predictive value of the proteomic clusters for these drugs. We underline that proteomics is relevant for personalized oncology and provide abundance and tumor specificity data for a large part of the UC proteome ( www.cancerproteins.org ).
Collapse
Affiliation(s)
- Franz F Dressler
- Institute of Pathology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany.
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany.
- Institute of Pathology, University Medical Center Schleswig-Holstein, Campus Lübeck, Lübeck, Germany.
| | - Falk Diedrichs
- Institute of Pathology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Deema Sabtan
- Institute of Pathology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Sofie Hinrichs
- Institute of Pathology, University Medical Center Schleswig-Holstein, Campus Lübeck, Lübeck, Germany
| | - Christoph Krisp
- Section Mass Spectrometry and Proteomics, Campus Forschung N27 00.008, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Timo Gemoll
- Section for Translational Surgical Oncology and Biobanking, Department of Surgery, University Medical Center Schleswig-Holstein, Campus Lübeck, Lübeck, Germany
| | - Martin Hennig
- Department of Urology, University Hospital Schleswig-Holstein, Campus Lübeck, Lübeck, Germany
| | - Paulina Mackedanz
- Institute of Pathology, University Medical Center Schleswig-Holstein, Campus Lübeck, Lübeck, Germany
| | - Mareile Schlotfeldt
- Institute of Pathology, University Medical Center Schleswig-Holstein, Campus Lübeck, Lübeck, Germany
| | - Hannah Voß
- Section Mass Spectrometry and Proteomics, Campus Forschung N27 00.008, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Anne Offermann
- Institute of Pathology, University Medical Center Schleswig-Holstein, Campus Lübeck, Lübeck, Germany
| | - Jutta Kirfel
- Institute of Pathology, University Medical Center Schleswig-Holstein, Campus Lübeck, Lübeck, Germany
| | - Marie C Roesch
- Department of Urology, University Hospital Schleswig-Holstein, Campus Lübeck, Lübeck, Germany
| | - Julian P Struck
- Department of Urology, University Hospital Schleswig-Holstein, Campus Lübeck, Lübeck, Germany
- Department of Urology, Faculty of Health Sciences Brandenburg, Brandenburg Medical School Theodor Fontane, Brandenburg, Germany
| | - Mario W Kramer
- Department of Urology, University Hospital Schleswig-Holstein, Campus Lübeck, Lübeck, Germany
| | - Axel S Merseburger
- Department of Urology, University Hospital Schleswig-Holstein, Campus Lübeck, Lübeck, Germany
| | - Christian Gratzke
- Department of Urology, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Dominik S Schoeb
- Department of Urology, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Arkadiusz Miernik
- Department of Urology, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Hartmut Schlüter
- Section Mass Spectrometry and Proteomics, Campus Forschung N27 00.008, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Ulrich Wetterauer
- Department of Urology, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Department of Medicine, Faculty of Medicine and Dentistry, Danube Private University, 3500, Krems, Austria
| | - Roman Zubarev
- Division of Chemistry I, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
- The National Medical Research Center for Endocrinology, Moscow, Russia
- Department of Pharmacological & Technological Chemistry, I.M. Sechenov First Moscow State Medical University, Moscow, Russia
| | - Sven Perner
- Institute of Pathology, University Medical Center Schleswig-Holstein, Campus Lübeck, Lübeck, Germany
- Institute of Pathology, Research Center Borstel, Leibniz Lung Center, Borstel, Germany
- Center for Precision Oncology, Tuebingen, Germany
| | - Philipp Wolf
- Department of Urology, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Ákos Végvári
- Division of Chemistry I, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
5
|
Pateras IS, Igea A, Nikas IP, Leventakou D, Koufopoulos NI, Ieronimaki AI, Bergonzini A, Ryu HS, Chatzigeorgiou A, Frisan T, Kittas C, Panayiotides IG. Diagnostic Challenges during Inflammation and Cancer: Current Biomarkers and Future Perspectives in Navigating through the Minefield of Reactive versus Dysplastic and Cancerous Lesions in the Digestive System. Int J Mol Sci 2024; 25:1251. [PMID: 38279253 PMCID: PMC10816510 DOI: 10.3390/ijms25021251] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 01/12/2024] [Accepted: 01/16/2024] [Indexed: 01/28/2024] Open
Abstract
In the setting of pronounced inflammation, changes in the epithelium may overlap with neoplasia, often rendering it impossible to establish a diagnosis with certainty in daily clinical practice. Here, we discuss the underlying molecular mechanisms driving tissue response during persistent inflammatory signaling along with the potential association with cancer in the gastrointestinal tract, pancreas, extrahepatic bile ducts, and liver. We highlight the histopathological challenges encountered in the diagnosis of chronic inflammation in routine practice and pinpoint tissue-based biomarkers that could complement morphology to differentiate reactive from dysplastic or cancerous lesions. We refer to the advantages and limitations of existing biomarkers employing immunohistochemistry and point to promising new markers, including the generation of novel antibodies targeting mutant proteins, miRNAs, and array assays. Advancements in experimental models, including mouse and 3D models, have improved our understanding of tissue response. The integration of digital pathology along with artificial intelligence may also complement routine visual inspections. Navigating through tissue responses in various chronic inflammatory contexts will help us develop novel and reliable biomarkers that will improve diagnostic decisions and ultimately patient treatment.
Collapse
Affiliation(s)
- Ioannis S. Pateras
- 2nd Department of Pathology, “Attikon” University Hospital, Medical School, National and Kapodistrian University of Athens, 124 62 Athens, Greece; (D.L.); (N.I.K.); (A.I.I.); (I.G.P.)
| | - Ana Igea
- Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS), 15706 Santiago de Compostela, Spain;
- Mobile Genomes, Centre for Research in Molecular Medicine and Chronic Diseases (CiMUS), University of Santiago de Compostela (USC), 15706 Santiago de Compostela, Spain
| | - Ilias P. Nikas
- Medical School, University of Cyprus, 2029 Nicosia, Cyprus
| | - Danai Leventakou
- 2nd Department of Pathology, “Attikon” University Hospital, Medical School, National and Kapodistrian University of Athens, 124 62 Athens, Greece; (D.L.); (N.I.K.); (A.I.I.); (I.G.P.)
| | - Nektarios I. Koufopoulos
- 2nd Department of Pathology, “Attikon” University Hospital, Medical School, National and Kapodistrian University of Athens, 124 62 Athens, Greece; (D.L.); (N.I.K.); (A.I.I.); (I.G.P.)
| | - Argyro Ioanna Ieronimaki
- 2nd Department of Pathology, “Attikon” University Hospital, Medical School, National and Kapodistrian University of Athens, 124 62 Athens, Greece; (D.L.); (N.I.K.); (A.I.I.); (I.G.P.)
| | - Anna Bergonzini
- Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Karolinska University Hospital, Alfred Nobels Allé 8, 141 52 Stockholm, Sweden;
- Department of Molecular Biology and Umeå Centre for Microbial Research (UCMR), Umeå University, 901 87 Umeå, Sweden;
| | - Han Suk Ryu
- Department of Pathology, Seoul National University Hospital, Seoul 03080, Republic of Korea;
| | - Antonios Chatzigeorgiou
- Department of Physiology, Medical School, National and Kapodistrian University of Athens, 115 27 Athens, Greece;
| | - Teresa Frisan
- Department of Molecular Biology and Umeå Centre for Microbial Research (UCMR), Umeå University, 901 87 Umeå, Sweden;
| | - Christos Kittas
- Department of Histopathology, Biomedicine Group of Health Company, 156 26 Athens, Greece;
| | - Ioannis G. Panayiotides
- 2nd Department of Pathology, “Attikon” University Hospital, Medical School, National and Kapodistrian University of Athens, 124 62 Athens, Greece; (D.L.); (N.I.K.); (A.I.I.); (I.G.P.)
| |
Collapse
|
6
|
Zhang S, Cai Z, Li H. AHNAKs roles in physiology and malignant tumors. Front Oncol 2023; 13:1258951. [PMID: 38033502 PMCID: PMC10682155 DOI: 10.3389/fonc.2023.1258951] [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: 07/14/2023] [Accepted: 10/30/2023] [Indexed: 12/02/2023] Open
Abstract
The AHNAK family currently consists of two members, namely AHNAK and AHNAK2, both of which have a molecular weight exceeding 600 kDa. Homologous sequences account for approximately 90% of their composition, indicating a certain degree of similarity in terms of molecular structure and biological functions. AHNAK family members are involved in the regulation of various biological functions, such as calcium channel modulation and membrane repair. Furthermore, with advancements in biological and bioinformatics technologies, research on the relationship between the AHNAK family and tumors has rapidly increased in recent years, and its regulatory role in tumor progression has gradually been discovered. This article briefly describes the physiological functions of the AHNAK family, and reviews and analyzes the expression and molecular regulatory mechanisms of the AHNAK family in malignant tumors using Pubmed and TCGA databases. In summary, AHNAK participates in various physiological and pathological processes in the human body. In multiple types of cancers, abnormal expression of AHNAK and AHNAK2 is associated with prognosis, and they play a key regulatory role in tumor progression by activating signaling pathways such as ERK, MAPK, Wnt, and MEK, as well as promoting epithelial-mesenchymal transition.
Collapse
Affiliation(s)
- Shusen Zhang
- Hebei Province Xingtai People’s Hospital Postdoctoral Workstation, Xingtai, China
- Postdoctoral Mobile Station, Hebei Medical University, Shijiazhuang, China
- Department of Pulmonary and Critical Care Medicine, Affiliated Xing Tai People Hospital of Hebei Medical University, Xingtai, China
- The First Department of Pulmonary and Critical Care Medicine, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Zhigang Cai
- Postdoctoral Mobile Station, Hebei Medical University, Shijiazhuang, China
- The First Department of Pulmonary and Critical Care Medicine, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Hui Li
- Department of surgery, Affiliated Xing Tai People Hospital of Hebei Medical University, Xingtai, China
| |
Collapse
|
7
|
Chang S, Giannico GA, Haugen E, Jardaneh A, Baba J, Mahadevan-Jansen A, Chang SS, Bowden AK. Multiparameter interferometric polarization-enhanced imaging differentiates carcinoma in situ from inflammation of the bladder: an ex vivo study. JOURNAL OF BIOMEDICAL OPTICS 2023; 28:102907. [PMID: 37576611 PMCID: PMC10415042 DOI: 10.1117/1.jbo.28.10.102907] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Revised: 07/28/2023] [Accepted: 07/31/2023] [Indexed: 08/15/2023]
Abstract
Significance Successful differentiation of carcinoma in situ (CIS) from inflammation in the bladder is key to preventing unnecessary biopsies and enabling accurate therapeutic decisions. Current standard-of-care diagnostic imaging techniques lack the specificity needed to differentiate these states, leading to false positives. Aim We introduce multiparameter interferometric polarization-enhanced (MultiPIPE) imaging as a promising technology to improve the specificity of detection for better biopsy guidance and clinical outcomes. Approach In this ex vivo study, we extract tissue attenuation-coefficient-based and birefringence-based parameters from MultiPIPE imaging data, collected with a bench-top system, to develop a classifier for the differentiation of benign and CIS tissues. We also analyze morphological features from second harmonic generation imaging and histology slides and perform imaging-to-morphology correlation analysis. Results MultiPIPE enhances specificity to differentiate CIS from benign tissues by nearly 20% and reduces the false-positive rate by more than four-fold over clinical standards. We also show that the MultiPIPE measurements correlate well with changes in morphological features in histological assessments. Conclusions The results of our study show the promise of MultiPIPE imaging to be used for better differentiation of bladder inflammation from flat tumors, leading to a fewer number of unnecessary procedures and shorter operating room (OR) time.
Collapse
Affiliation(s)
- Shuang Chang
- Vannderbilt University, Vanderbilt Biophotonics Center, Department of Biomedical Engineering, Nashville, Tennessee, United States
| | - Giovanna A. Giannico
- Vanderbilt University Medical Center, Department of Pathology, Microbiology, and Immunology, Nashville, Tennessee, United States
| | - Ezekiel Haugen
- Vannderbilt University, Vanderbilt Biophotonics Center, Department of Biomedical Engineering, Nashville, Tennessee, United States
| | - Ali Jardaneh
- Vanderbilt University Medical Center, Department of Urology, Nashville, Tennessee, United States
| | - Justin Baba
- Vannderbilt University, Vanderbilt Biophotonics Center, Department of Biomedical Engineering, Nashville, Tennessee, United States
| | - Anita Mahadevan-Jansen
- Vannderbilt University, Vanderbilt Biophotonics Center, Department of Biomedical Engineering, Nashville, Tennessee, United States
| | - Sam S. Chang
- Vanderbilt University Medical Center, Department of Urology, Nashville, Tennessee, United States
| | - Audrey K. Bowden
- Vannderbilt University, Vanderbilt Biophotonics Center, Department of Biomedical Engineering, Nashville, Tennessee, United States
- Vanderbilt University, Department of Electrical and Computer Engineering, Nashville, Tennessee, United States
| |
Collapse
|
8
|
Gerwert K, Schörner S, Großerueschkamp F, Kraeft AL, Schuhmacher D, Sternemann C, Feder IS, Wisser S, Lugnier C, Arnold D, Teschendorf C, Mueller L, Timmesfeld N, Mosig A, Reinacher-Schick A, Tannapfel A. Fast and label-free automated detection of microsatellite status in early colon cancer using artificial intelligence integrated infrared imaging. Eur J Cancer 2023; 182:122-131. [PMID: 36773401 DOI: 10.1016/j.ejca.2022.12.026] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Revised: 12/14/2022] [Accepted: 12/23/2022] [Indexed: 01/11/2023]
Abstract
PURPOSE Microsatellite instability (MSI) due to mismatch repair (MMR) defects accounts for 15-20% of colon cancers (CC). MSI testing is currently standard of care in CC with immunohistochemistry of the four MMR proteins representing the gold standard. Instead, label-free quantum cascade laser (QCL) based infrared (IR) imaging combined with artificial intelligence (AI) may classify MSI/microsatellite stability (MSS) in unstained tissue sections user-independently and tissue preserving. METHODS Paraffin-embedded unstained tissue sections of early CC from patients participating in the multicentre AIO ColoPredict Plus (CPP) 2.0 registry were analysed after dividing into three groups (training, test, and validation). IR images of tissue sections using QCL-IR microscopes were classified by AI (convolutional neural networks [CNN]) using a two-step approach. The first CNN (modified U-Net) detected areas of cancer while the second CNN (VGG-Net) classified MSI/MSS. End-points were area under receiver operating characteristic (AUROC) and area under precision recall curve (AUPRC). RESULTS The cancer detection in the first step was based on 629 patients (train n = 273, test n = 138, and validation n = 218). Resulting classification AUROC was 1.0 for the validation dataset. The second step classifying MSI/MSS was performed on 547 patients (train n = 331, test n = 69, and validation n = 147) reaching AUROC and AUPRC of 0.9 and 0.74, respectively, for the validation cohort. CONCLUSION Our novel label-free digital pathology approach accurately and rapidly classifies MSI vs. MSS. The tissue sections analysed were not processed leaving the sample unmodified for subsequent analyses. Our approach demonstrates an AI-based decision support tool potentially driving improved patient stratification and precision oncology in the future.
Collapse
Affiliation(s)
- Klaus Gerwert
- Center for Protein Diagnostics (PRODI), Deptartment of Biophysics, Ruhr University Bochum, Bochum, Germany
| | - Stephanie Schörner
- Center for Protein Diagnostics (PRODI), Deptartment of Biophysics, Ruhr University Bochum, Bochum, Germany
| | - Frederik Großerueschkamp
- Center for Protein Diagnostics (PRODI), Deptartment of Biophysics, Ruhr University Bochum, Bochum, Germany
| | - Anna-Lena Kraeft
- Deptartment of Haematology, Oncology and Palliative Care, St. Josef-Hospital, Ruhr University Bochum, Bochum, Germany
| | - David Schuhmacher
- Center for Protein Diagnostics (PRODI), Dept. of Bioinformatics, Ruhr University Bochum, Bochum, Germany
| | - Carlo Sternemann
- Institut für Pathologie, Ruhr-Universität Bochum, Bochum, Germany
| | - Inke S Feder
- Institut für Pathologie, Ruhr-Universität Bochum, Bochum, Germany
| | - Sarah Wisser
- Institut für Pathologie, Ruhr-Universität Bochum, Bochum, Germany
| | - Celine Lugnier
- Deptartment of Haematology, Oncology and Palliative Care, St. Josef-Hospital, Ruhr University Bochum, Bochum, Germany
| | - Dirk Arnold
- Oncology, Haematology, Palliative Care Deptartment Asklepios Tumorzentrum Hamburg AK Altona, Hamburg, Germany
| | | | - Lothar Mueller
- Onkologie UnterEms Leer Emden Papenburg, Onkologische Schwerpunktpraxis Leer-Emden, Leer, Germany
| | - Nina Timmesfeld
- Medical Informatics, Biometry and Epidemiology, Ruhr University Bochum, Bochum, Germany
| | - Axel Mosig
- Center for Protein Diagnostics (PRODI), Dept. of Bioinformatics, Ruhr University Bochum, Bochum, Germany
| | - Anke Reinacher-Schick
- Deptartment of Haematology, Oncology and Palliative Care, St. Josef-Hospital, Ruhr University Bochum, Bochum, Germany
| | - Andrea Tannapfel
- Institut für Pathologie, Ruhr-Universität Bochum, Bochum, Germany.
| |
Collapse
|
9
|
Kujdowicz M, Perez-Guaita D, Chlosta P, Okon K, Malek K. Evaluation of grade and invasiveness of bladder urothelial carcinoma using infrared imaging and machine learning. Analyst 2023; 148:278-285. [PMID: 36525038 DOI: 10.1039/d2an01583h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Urothelial bladder carcinoma (BC) is primarily diagnosed with a subjective examination of biopsies by histopathologists, but accurate diagnosis remains time-consuming and of low diagnostic accuracy, especially for low grade non-invasive BC. We propose a novel approach for high-throughput BC evaluation by combining infrared (IR) microscopy of bladder sections with machine learning (partial least squares-discriminant analysis) to provide an automated prediction of the presence of cancer, invasiveness and grade. Cystoscopic biopsies from 50 patients with clinical suspicion of BC were histologically examined to assign grades and stages. Adjacent tissue cross-sections were IR imaged to provide hyperspectral datasets and cluster analysis segregated IR images to extract the average spectra of epithelial and subepithelial tissues. Discriminant models, which were validated using repeated random sampling double cross-validation, showed sensitivities (AUROC) ca. 85% (0.85) for the identification of cancer in epithelium and subepithelium. The diagnosis of non-invasive and invasive cases showed sensitivity values around 80% (0.84-0.85) and 76% (0.73-0.80), respectively, while the identification of low and high grade BC showed higher sensitivity values 87-88% (0.91-0.92). Finally, models for the discrimination between cancers with different invasiveness and grades showed more modest AUROC values (0.67-0.72). This proves the high potential of IR imaging in the development of ancillary platforms to screen bladder biopsies.
Collapse
Affiliation(s)
- Monika Kujdowicz
- Department of Pathomorphology, Faculty of Medicine, Jagiellonian University Medical College, Krakow, Grzegorzecka 16, 31-531, Poland.,Faculty of Chemistry, Jagiellonian University in Krakow, Krakow, Gronostajowa 2, 30-387, Poland.
| | - David Perez-Guaita
- Department of Analytical Chemistry, University of Valencia, 50 Dr. Moliner Street, Research Building, 46100 Burjassot, Valencia, Spain.
| | - Piotr Chlosta
- Department of Urology, Medical Faculty, Jagiellonian University Medical College, Krakow, Jakubowskiego 2, 30-688, Poland
| | - Krzysztof Okon
- Department of Pathomorphology, Faculty of Medicine, Jagiellonian University Medical College, Krakow, Grzegorzecka 16, 31-531, Poland
| | - Kamilla Malek
- Faculty of Chemistry, Jagiellonian University in Krakow, Krakow, Gronostajowa 2, 30-387, Poland.
| |
Collapse
|
10
|
Zhang K, Liu R, Tuo Y, Ma K, Zhang D, Wang Z, Huang P. Distinguishing Asphyxia from Sudden Cardiac Death as the Cause of Death from the Lung Tissues of Rats and Humans Using Fourier Transform Infrared Spectroscopy. ACS OMEGA 2022; 7:46859-46869. [PMID: 36570197 PMCID: PMC9773813 DOI: 10.1021/acsomega.2c05968] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Accepted: 11/23/2022] [Indexed: 06/17/2023]
Abstract
The ability to determine asphyxia as a cause of death is important in forensic practice and helps us to judge whether a case is criminal. However, in some cases where the deceased has underlying heart disease, death by asphyxia cannot be determined by traditional autopsy and morphological observation under a microscope because there are no specific morphological features for either asphyxia or sudden cardiac death (SCD). Here, Fourier transform infrared (FTIR) spectroscopy was employed to distinguish asphyxia from SCD. A total of 40 lung tissues (collected at 0 h and 24 h postmortem) from 20 rats (10 died from asphyxia and 10 died from SCD) and 16 human lung tissues from 16 real cases were used for spectral data acquisition. After data preprocessing, 2675 spectra from rat lung tissues and 1526 spectra from human lung tissues were obtained for subsequent analysis. First, we found that there were biochemical differences in the rat lung tissues between the two causes of death by principal component analysis and partial least-squares discriminant analysis (PLS-DA), which were related to alterations in lipids, proteins, and nucleic acids. In addition, a PLS-DA classification model can be built to distinguish asphyxia from SCD. Second, based on the spectral data obtained from lung tissues allowed to decompose for 24 h, we could still distinguish asphyxia from SCD even when decomposition occurred in animal models. Nine important spectral features that contributed to the discrimination in the animal experiment were selected and further analyzed. Third, 7 of the 9 differential spectral features were also found to be significantly different in human lung tissues from 16 real cases. A support vector machine model was finally built by using the seven variables to distinguish asphyxia from SCD in the human samples. Compared with the linear PLS-DA model, its accuracy was significantly improved to 0.798, and the correct rate of determining the cause of death was 100%. This study shows the application potential of FTIR spectroscopy for exploring the subtle biochemical differences resulting from different death processes and determining the cause of death even after decomposition.
Collapse
Affiliation(s)
- Kai Zhang
- Department
of Forensic Pathology, College of Forensic Medicine, Xi’an Jiaotong University, Xi’an 710061, People’s
Republic of China
| | - Ruina Liu
- Department
of Forensic Pathology, College of Forensic Medicine, Xi’an Jiaotong University, Xi’an 710061, People’s
Republic of China
| | - Ya Tuo
- Department
of Biochemistry and Physiology, Shanghai
University of Medicine and Health Sciences, Shanghai 201318, People’s Republic of China
| | - Kaijun Ma
- Shanghai
Key Laboratory of Crime Scene Evidence, Institute of Criminal Science
and Technology, Shanghai Municipal Public
Security Bureau, Shanghai 200042, People’s Republic
of China
| | - Dongchuan Zhang
- Shanghai
Key Laboratory of Crime Scene Evidence, Institute of Criminal Science
and Technology, Shanghai Municipal Public
Security Bureau, Shanghai 200042, People’s Republic
of China
| | - Zhenyuan Wang
- Department
of Forensic Pathology, College of Forensic Medicine, Xi’an Jiaotong University, Xi’an 710061, People’s
Republic of China
| | - Ping Huang
- Shanghai
Key Laboratory of Forensic Medicine, Shanghai Forensic Service Platform, Academy of Forensic Science, Shanghai 200063, People’s Republic of China
| |
Collapse
|
11
|
Bracht T, Kleefisch D, Schork K, Witzke KE, Chen W, Bayer M, Hovanec J, Johnen G, Meier S, Ko YD, Behrens T, Brüning T, Fassunke J, Buettner R, Uszkoreit J, Adamzik M, Eisenacher M, Sitek B. Plasma Proteomics Enable Differentiation of Lung Adenocarcinoma from Chronic Obstructive Pulmonary Disease (COPD). Int J Mol Sci 2022; 23:ijms231911242. [PMID: 36232544 PMCID: PMC9569607 DOI: 10.3390/ijms231911242] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 09/19/2022] [Accepted: 09/20/2022] [Indexed: 11/18/2022] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is a major risk factor for the development of lung adenocarcinoma (AC). AC often develops on underlying COPD; thus, the differentiation of both entities by biomarker is challenging. Although survival of AC patients strongly depends on early diagnosis, a biomarker panel for AC detection and differentiation from COPD is still missing. Plasma samples from 176 patients with AC with or without underlying COPD, COPD patients, and hospital controls were analyzed using mass-spectrometry-based proteomics. We performed univariate statistics and additionally evaluated machine learning algorithms regarding the differentiation of AC vs. COPD and AC with COPD vs. COPD. Univariate statistics revealed significantly regulated proteins that were significantly regulated between the patient groups. Furthermore, random forest classification yielded the best performance for differentiation of AC vs. COPD (area under the curve (AUC) 0.935) and AC with COPD vs. COPD (AUC 0.916). The most influential proteins were identified by permutation feature importance and compared to those identified by univariate testing. We demonstrate the great potential of machine learning for differentiation of highly similar disease entities and present a panel of biomarker candidates that should be considered for the development of a future biomarker panel.
Collapse
Affiliation(s)
- Thilo Bracht
- Clinic for Anesthesiology, Intensive Care and Pain Therapy, University Medical Center Knappschaftskrankenhaus Bochum, 44892 Bochum, Germany
- Medizinisches Proteom-Center, Ruhr-University Bochum, 44801 Bochum, Germany
- Correspondence: (T.B.); (B.S.); Tel.: +49-234-32-29985 (T.B.); +49-234-32-24362 (B.S.)
| | - Daniel Kleefisch
- Medizinisches Proteom-Center, Ruhr-University Bochum, 44801 Bochum, Germany
- Center for Protein Diagnostics (PRODI), Medical Proteome Analysis, Ruhr-University Bochum, 44801 Bochum, Germany
| | - Karin Schork
- Medizinisches Proteom-Center, Ruhr-University Bochum, 44801 Bochum, Germany
- Center for Protein Diagnostics (PRODI), Medical Proteome Analysis, Ruhr-University Bochum, 44801 Bochum, Germany
| | - Kathrin E. Witzke
- Medizinisches Proteom-Center, Ruhr-University Bochum, 44801 Bochum, Germany
- Center for Protein Diagnostics (PRODI), Medical Proteome Analysis, Ruhr-University Bochum, 44801 Bochum, Germany
| | - Weiqiang Chen
- Medizinisches Proteom-Center, Ruhr-University Bochum, 44801 Bochum, Germany
| | - Malte Bayer
- Clinic for Anesthesiology, Intensive Care and Pain Therapy, University Medical Center Knappschaftskrankenhaus Bochum, 44892 Bochum, Germany
- Medizinisches Proteom-Center, Ruhr-University Bochum, 44801 Bochum, Germany
| | - Jan Hovanec
- Institute for Prevention and Occupational Medicine of the German Social Accident Insurance, Institute of the Ruhr University Bochum (IPA), 44789 Bochum, Germany
| | - Georg Johnen
- Institute for Prevention and Occupational Medicine of the German Social Accident Insurance, Institute of the Ruhr University Bochum (IPA), 44789 Bochum, Germany
| | - Swetlana Meier
- Institute for Prevention and Occupational Medicine of the German Social Accident Insurance, Institute of the Ruhr University Bochum (IPA), 44789 Bochum, Germany
| | - Yon-Dschun Ko
- Department of Internal Medicine, Johanniter-Kliniken Bonn GmbH, Johanniter Krankenhaus, 53113 Bonn, Germany
| | - Thomas Behrens
- Institute for Prevention and Occupational Medicine of the German Social Accident Insurance, Institute of the Ruhr University Bochum (IPA), 44789 Bochum, Germany
| | - Thomas Brüning
- Institute for Prevention and Occupational Medicine of the German Social Accident Insurance, Institute of the Ruhr University Bochum (IPA), 44789 Bochum, Germany
| | - Jana Fassunke
- Institute of Pathology, Medical Faculty and Center for Molecular Medicine (CMMC), University of Cologne, 50924 Cologne, Germany
| | - Reinhard Buettner
- Institute of Pathology, Medical Faculty and Center for Molecular Medicine (CMMC), University of Cologne, 50924 Cologne, Germany
| | - Julian Uszkoreit
- Medizinisches Proteom-Center, Ruhr-University Bochum, 44801 Bochum, Germany
- Center for Protein Diagnostics (PRODI), Medical Proteome Analysis, Ruhr-University Bochum, 44801 Bochum, Germany
| | - Michael Adamzik
- Clinic for Anesthesiology, Intensive Care and Pain Therapy, University Medical Center Knappschaftskrankenhaus Bochum, 44892 Bochum, Germany
| | - Martin Eisenacher
- Medizinisches Proteom-Center, Ruhr-University Bochum, 44801 Bochum, Germany
- Center for Protein Diagnostics (PRODI), Medical Proteome Analysis, Ruhr-University Bochum, 44801 Bochum, Germany
| | - Barbara Sitek
- Clinic for Anesthesiology, Intensive Care and Pain Therapy, University Medical Center Knappschaftskrankenhaus Bochum, 44892 Bochum, Germany
- Medizinisches Proteom-Center, Ruhr-University Bochum, 44801 Bochum, Germany
- Correspondence: (T.B.); (B.S.); Tel.: +49-234-32-29985 (T.B.); +49-234-32-24362 (B.S.)
| |
Collapse
|
12
|
Ai F, Wang W, Liu S, Zhang D, Yang Z, Liu F. Integrative Proteo-Genomic Analysis for Recurrent Survival Prognosis in Colon Adenocarcinoma. Front Oncol 2022; 12:871568. [PMID: 35847888 PMCID: PMC9281446 DOI: 10.3389/fonc.2022.871568] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Accepted: 05/09/2022] [Indexed: 12/09/2022] Open
Abstract
Background The survival prognosis is the hallmark of cancer progression. Here, we aimed to develop a recurrence-related gene signature to predict the prognosis of colon adenocarcinoma (COAD). Methods The proteomic data from the Clinical Proteomic Tumor Analysis Consortium (CPTAC) and genomic data from the cancer genomic maps [The Cancer Genome Atlas (TCGA)] dataset were analyzed to identify co-differentially expressed genes (cDEGs) between recurrence samples and non-recurrence samples in COAD using limma package. Functional enrichment analysis, including Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway was conducted. Univariate and multivariate Cox regressions were applied to identify the independent prognostic feature cDEGs and establish the signature whose performance was evaluated by Kaplan–Meier curve, receiver operating characteristic (ROC), Harrell’s concordance index (C-index), and calibration curve. The area under the receiver operating characteristic (ROC) curve (AUROC) and a nomogram were calculated to assess the predictive accuracy. GSE17538 and GSE39582 were used for external validation. Quantitative real-time PCR and Western blot analysis were carried out to validate our findings. Results We identified 86 cDEGs in recurrence samples compared with non-recurrence samples. These genes were primarily enriched in the regulation of carbon metabolic process, fructose and mannose metabolism, and extracellular exosome. Then, an eight-gene-based signature (CA12, HBB, NCF1, KBTBD11, MMAA, DMBT1, AHNAK2, and FBLN2) was developed to separate patients into high- and low-risk groups. Patients in the low-risk group had significantly better prognosis than those in the high-risk group. Four prognostic clinical features, including pathological M, N, T, and RS model status, were screened for building the nomogram survival model. The PCR and Western blot analysis results suggested that CA12 and AHNAK2 were significantly upregulated, while MMAA and DMBT1 were downregulated in the tumor sample compared with adjacent tissues, and in non-recurrent samples compared with non-recurrent samples in COAD. Conclusion These identified recurrence-related gene signatures might provide an effective prognostic predictor and promising therapeutic targets for COAD patients.
Collapse
Affiliation(s)
- FeiYan Ai
- Department of Gastroenterology, The Third Xiangya Hospital of Central South University, Changsha, China
- Hunan Key Laboratory of Non-Resolving Inflammation and Cancer The Third Xiangya Hospital, Central South University, Changsha, China
| | - Wenhao Wang
- Department of Breast and Thyroid Surgery, The First Affiliated Hospital of University of South China, Hengyang, China
| | - Shaojun Liu
- Department of Gastroenterology, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Decai Zhang
- Department of Gastroenterology, The Third Xiangya Hospital of Central South University, Changsha, China
- Hunan Key Laboratory of Non-Resolving Inflammation and Cancer The Third Xiangya Hospital, Central South University, Changsha, China
| | - Zhenyu Yang
- Department of Gastroenterology, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Fen Liu
- Department of Gastroenterology, The Third Xiangya Hospital of Central South University, Changsha, China
- Hunan Key Laboratory of Non-Resolving Inflammation and Cancer The Third Xiangya Hospital, Central South University, Changsha, China
- *Correspondence: Fen Liu,
| |
Collapse
|
13
|
Zacharovas E, Velička M, Platkevičius G, Čekauskas A, Želvys A, Niaura G, Šablinskas V. Toward a SERS Diagnostic Tool for Discrimination between Cancerous and Normal Bladder Tissues via Analysis of the Extracellular Fluid. ACS OMEGA 2022; 7:10539-10549. [PMID: 35382275 PMCID: PMC8973049 DOI: 10.1021/acsomega.2c00058] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Accepted: 03/03/2022] [Indexed: 05/09/2023]
Abstract
Vibrational spectroscopy provides the possibility for sensitive and precise detection of chemical changes in biomolecules due to development of cancers. In this work, label-free near-infrared surface enhanced Raman spectroscopy (SERS) was applied for the differentiation between cancerous and normal human bladder tissues via analysis of the extracellular fluid of the tissue. Specific cancer-related SERS marker bands were identified by using a 1064 nm excitation wavelength. The prominent spectral marker band was found to be located near 1052 cm-1 and was assigned to the C-C, C-O, and C-N stretching vibrations of lactic acid and/or cysteine molecules. The correct identification of 80% of samples is achieved with even limited data set and could be further improved. The further development of such a detection method could be implemented in clinical practice for the aid of surgeons in determining of boundaries of malignant tumors during the surgery.
Collapse
Affiliation(s)
- Edvinas Zacharovas
- Institute
of Chemical Physics, Faculty of Physics, Vilnius University, Saulėtekis Avenue 3, LT-10257 Vilnius, Lithuania
| | - Martynas Velička
- Institute
of Chemical Physics, Faculty of Physics, Vilnius University, Saulėtekis Avenue 3, LT-10257 Vilnius, Lithuania
| | - Gediminas Platkevičius
- Clinic
of Gastroenterology, Nephrourology, and Surgery, Institute of Clinical
Medicine, Faculty of Medicine, Vilnius University, M.K. Čiurlionio st. 21/27, LT-03101 Vilnius, Lithuania
| | - Albertas Čekauskas
- Clinic
of Gastroenterology, Nephrourology, and Surgery, Institute of Clinical
Medicine, Faculty of Medicine, Vilnius University, M.K. Čiurlionio st. 21/27, LT-03101 Vilnius, Lithuania
| | - Aru̅nas Želvys
- Clinic
of Gastroenterology, Nephrourology, and Surgery, Institute of Clinical
Medicine, Faculty of Medicine, Vilnius University, M.K. Čiurlionio st. 21/27, LT-03101 Vilnius, Lithuania
| | - Gediminas Niaura
- Institute
of Chemical Physics, Faculty of Physics, Vilnius University, Saulėtekis Avenue 3, LT-10257 Vilnius, Lithuania
- Department
of Organic Chemistry, Center for Physical
Sciences and Technology (FTMC), Saulėtekis Avenue 3, LT 10257, Vilnius, Lithuania
| | - Valdas Šablinskas
- Institute
of Chemical Physics, Faculty of Physics, Vilnius University, Saulėtekis Avenue 3, LT-10257 Vilnius, Lithuania
| |
Collapse
|
14
|
Czétány P, Gitta S, Balló A, Sulc A, Máté G, Szántó Á, Márk L. Application of Mass Spectrometry Imaging in Uro-Oncology: Discovering Potential Biomarkers. Life (Basel) 2022; 12:life12030366. [PMID: 35330118 PMCID: PMC8954359 DOI: 10.3390/life12030366] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Revised: 02/25/2022] [Accepted: 02/25/2022] [Indexed: 11/17/2022] Open
Abstract
A growing need is emerging worldwide for new molecular markers which could enhance the accuracy of diagnostic and therapeutic methods for detecting urogenital cancers. Mass spectrometry imaging (MSI) is a very promising tool in this regard. In this review, we attempt to provide a subjective summary of the latest publications on potential biomarkers of renal, bladder, prostate, and testicular malignancies detected with MSI through the eyes of a clinical urologist.
Collapse
Affiliation(s)
- Péter Czétány
- National Human Reproduction Laboratory, 7624 Pécs, Hungary; (P.C.); (S.G.); (A.B.); (A.S.); (G.M.); (Á.S.)
- Urology Clinic, University of Pécs Clinical Centre, 7621 Pécs, Hungary
| | - Stefánia Gitta
- National Human Reproduction Laboratory, 7624 Pécs, Hungary; (P.C.); (S.G.); (A.B.); (A.S.); (G.M.); (Á.S.)
- Department of Biochemistry and Medical Chemistry, University of Pécs Medical School, 7624 Pécs, Hungary
| | - András Balló
- National Human Reproduction Laboratory, 7624 Pécs, Hungary; (P.C.); (S.G.); (A.B.); (A.S.); (G.M.); (Á.S.)
- Urology Clinic, University of Pécs Clinical Centre, 7621 Pécs, Hungary
- Pannon Reproduction Institute, 8300 Tapolca, Hungary
| | - Alexandra Sulc
- National Human Reproduction Laboratory, 7624 Pécs, Hungary; (P.C.); (S.G.); (A.B.); (A.S.); (G.M.); (Á.S.)
- Department of Biochemistry and Medical Chemistry, University of Pécs Medical School, 7624 Pécs, Hungary
| | - Gábor Máté
- National Human Reproduction Laboratory, 7624 Pécs, Hungary; (P.C.); (S.G.); (A.B.); (A.S.); (G.M.); (Á.S.)
- Pannon Reproduction Institute, 8300 Tapolca, Hungary
| | - Árpád Szántó
- National Human Reproduction Laboratory, 7624 Pécs, Hungary; (P.C.); (S.G.); (A.B.); (A.S.); (G.M.); (Á.S.)
- Urology Clinic, University of Pécs Clinical Centre, 7621 Pécs, Hungary
| | - László Márk
- National Human Reproduction Laboratory, 7624 Pécs, Hungary; (P.C.); (S.G.); (A.B.); (A.S.); (G.M.); (Á.S.)
- Department of Biochemistry and Medical Chemistry, University of Pécs Medical School, 7624 Pécs, Hungary
- MTA-PTE Human Reproduction Research Group, 7624 Pécs, Hungary
- Correspondence: ; Tel.: +36-304-734-714
| |
Collapse
|
15
|
Zardab M, Stasinos K, Grose RP, Kocher HM. The Obscure Potential of AHNAK2. Cancers (Basel) 2022; 14:cancers14030528. [PMID: 35158796 PMCID: PMC8833689 DOI: 10.3390/cancers14030528] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Revised: 01/17/2022] [Accepted: 01/18/2022] [Indexed: 12/17/2022] Open
Abstract
Simple Summary AHNAK2 is a relatively newly discovered protein. It can interact with many other proteins. This protein is increased in cells of variety of different cancers. AHNAK2 may play a vital role in cancer formation. AHNAK2 may have a role in early detection of cancer. This obscure potential of AHNAK2 is being studied. Abstract AHNAK2 is a protein discovered in 2004, with a strong association with oncogenesis in various epithelial cancers. It has a large 616 kDa tripartite structure and is thought to take part in the formation of large multi-protein complexes. High expression is found in clear cell renal carcinoma, pancreatic ductal adenocarcinoma, uveal melanoma, and lung adenocarcinoma, with a relation to poor prognosis. Little work has been done in exploring the function and relation AHNAK2 has with cancer, with early studies showing promising potential as a future biomarker and therapeutic target.
Collapse
|
16
|
Großerueschkamp F, Jütte H, Gerwert K, Tannapfel A. Advances in Digital Pathology: From Artificial Intelligence to Label-Free Imaging. Visc Med 2021; 37:482-490. [PMID: 35087898 DOI: 10.1159/000518494] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Accepted: 07/14/2021] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Digital pathology, in its primary meaning, describes the utilization of computer screens to view scanned histology slides. Digitized tissue sections can be easily shared for a second opinion. In addition, it allows tissue image analysis using specialized software to identify and measure events previously observed by a human observer. These tissue-based readouts were highly reproducible and precise. Digital pathology has developed over the years through new technologies. Currently, the most discussed development is the application of artificial intelligence to automatically analyze tissue images. However, even new label-free imaging technologies are being developed to allow imaging of tissues by means of their molecular composition. SUMMARY This review provides a summary of the current state-of-the-art and future digital pathologies. Developments in the last few years have been presented and discussed. In particular, the review provides an outlook on interesting new technologies (e.g., infrared imaging), which would allow for deeper understanding and analysis of tissue thin sections beyond conventional histopathology. KEY MESSAGES In digital pathology, mathematical methods are used to analyze images and draw conclusions about diseases and their progression. New innovative methods and techniques (e.g., label-free infrared imaging) will bring significant changes in the field in the coming years.
Collapse
Affiliation(s)
- Frederik Großerueschkamp
- Center for Protein Diagnostics (PRODI), Biospectroscopy, Ruhr University Bochum, Bochum, Germany.,Department of Biophysics, Faculty of Biology and Biotechnology, Ruhr University Bochum, Bochum, Germany
| | - Hendrik Jütte
- Center for Protein Diagnostics (PRODI), Biospectroscopy, Ruhr University Bochum, Bochum, Germany.,Institute of Pathology, Ruhr University Bochum, Bochum, Germany
| | - Klaus Gerwert
- Center for Protein Diagnostics (PRODI), Biospectroscopy, Ruhr University Bochum, Bochum, Germany.,Department of Biophysics, Faculty of Biology and Biotechnology, Ruhr University Bochum, Bochum, Germany
| | - Andrea Tannapfel
- Center for Protein Diagnostics (PRODI), Biospectroscopy, Ruhr University Bochum, Bochum, Germany.,Institute of Pathology, Ruhr University Bochum, Bochum, Germany
| |
Collapse
|
17
|
López-Cortés R, Vázquez-Estévez S, Fernández JÁ, Núñez C. Proteomics as a Complementary Technique to Characterize Bladder Cancer. Cancers (Basel) 2021; 13:cancers13215537. [PMID: 34771699 PMCID: PMC8582709 DOI: 10.3390/cancers13215537] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Revised: 10/21/2021] [Accepted: 10/21/2021] [Indexed: 12/11/2022] Open
Abstract
Simple Summary Although immunohistochemistry is a routine technique in clinics, and genomics has been rapidly incorporated, proteomics is a step behind. This general situation is also the norm in bladder cancer research. This review shows the contributions of proteomics to the molecular classification of bladder cancer, and to the study of histopathology due to tissue insults caused by tumors. Furthermore, the importance of proteomics for understanding the cellular and molecular changes as a consequence of the therapy of bladder cancer cannot be neglected. Abstract Bladder cancer (BC) is the most common tumor of the urinary tract and is conventionally classified as either non-muscle invasive or muscle invasive. In addition, histological variants exist, as organized by the WHO-2016 classification. However, innovations in next-generation sequencing have led to molecular classifications of BC. These innovations have also allowed for the tracing of major tumorigenic pathways and, therefore, are positioned as strong supporters of precision medicine. In parallel, immunohistochemistry is still the clinical reference to discriminate histological layers and to stage BC. Key contributions have been made to enlarge the panel of protein immunomarkers. Moreover, the analysis of proteins in liquid biopsy has also provided potential markers. Notwithstanding, their clinical adoption is still low, with very few approved tests. In this context, mass spectrometry-based proteomics has remained a step behind; hence, we aimed to develop them in the community. Herein, the authors introduce the epidemiology and the conventional classifications to review the molecular classification of BC, highlighting the contributions of proteomics. Then, the advances in mass spectrometry techniques focusing on maintaining the integrity of the biological structures are presented, a milestone for the emergence of histoproteomics. Within this field, the review then discusses selected proteins for the comprehension of the pathophysiological mechanisms of BC. Finally, because there is still insufficient knowledge, this review considers proteomics as an important source for the development of BC therapies.
Collapse
Affiliation(s)
- Rubén López-Cortés
- Research Unit, Hospital Universitario Lucus Augusti (HULA), Servizo Galego de Saúde (SERGAS), 27002 Lugo, Spain;
| | - Sergio Vázquez-Estévez
- Oncology Division, Hospital Universitario Lucus Augusti (HULA), Servizo Galego de Saúde (SERGAS), 27002 Lugo, Spain; (S.V.-E.); (J.Á.F.)
| | - Javier Álvarez Fernández
- Oncology Division, Hospital Universitario Lucus Augusti (HULA), Servizo Galego de Saúde (SERGAS), 27002 Lugo, Spain; (S.V.-E.); (J.Á.F.)
| | - Cristina Núñez
- Research Unit, Hospital Universitario Lucus Augusti (HULA), Servizo Galego de Saúde (SERGAS), 27002 Lugo, Spain;
- Correspondence:
| |
Collapse
|
18
|
Proteomic and bioinformatic profiling of neutrophils in CLL reveals functional defects that predispose to bacterial infections. Blood Adv 2021; 5:1259-1272. [PMID: 33651101 DOI: 10.1182/bloodadvances.2020002949] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Accepted: 01/19/2021] [Indexed: 12/15/2022] Open
Abstract
Patients with chronic lymphocytic leukemia (CLL) typically suffer from frequent and severe bacterial infections. Although it is well known that neutrophils are critical innate immune cells facilitating the early defense, the underlying phenotypical and functional changes in neutrophils during CLL remain largely elusive. Using a murine adoptive transfer model of CLL, we demonstrate aggravated bacterial burden in CLL-bearing mice upon a urinary tract infection with uropathogenic Escherichia coli. Bioinformatic analyses of the neutrophil proteome revealed increased expression of proteins associated with interferon signaling and decreased protein expression associated with granule composition and neutrophil migration. Functional experiments validated these findings by showing reduced levels of myeloperoxidase and acidification of neutrophil granules after ex vivo phagocytosis of bacteria. Pathway enrichment analysis indicated decreased expression of molecules critical for neutrophil recruitment, and migration of neutrophils into the infected urinary bladder was significantly reduced. These altered migratory properties of neutrophils were also associated with reduced expression of CD62L and CXCR4 and correlated with an increased incidence of infections in patients with CLL. In conclusion, this study describes a molecular signature of neutrophils through proteomic, bioinformatic, and functional analyses that are linked to a reduced migratory ability, potentially leading to increased bacterial infections in patients with CLL.
Collapse
|
19
|
Goertzen N, Pappesch R, Fassunke J, Brüning T, Ko YD, Schmidt J, Großerueschkamp F, Buettner R, Gerwert K. Quantum Cascade Laser-Based Infrared Imaging as a Label-Free and Automated Approach to Determine Mutations in Lung Adenocarcinoma. THE AMERICAN JOURNAL OF PATHOLOGY 2021; 191:1269-1280. [PMID: 34004158 DOI: 10.1016/j.ajpath.2021.04.013] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 04/09/2021] [Accepted: 04/22/2021] [Indexed: 12/28/2022]
Abstract
Therapeutic decisions in lung cancer critically depend on the determination of histologic types and oncogene mutations. Therefore, tumor samples are subjected to standard histologic and immunohistochemical analyses and examined for relevant mutations using comprehensive molecular diagnostics. In this study, an alternative diagnostic approach for automatic and label-free detection of mutations in lung adenocarcinoma tissue using quantum cascade laser-based infrared imaging is presented. For this purpose, a five-step supervised classification algorithm was developed, which was not only able to detect tissue types and tumor lesions, but also the tumor type and mutation status of adenocarcinomas. Tumor detection was verified on a data set of 214 patient samples with a specificity of 97% and a sensitivity of 95%. Furthermore, histology typing was verified on samples from 203 of the 214 patients with a specificity of 97% and a sensitivity of 94% for adenocarcinoma. The most frequently occurring mutations in adenocarcinoma (KRAS, EGFR, and TP53) were differentiated by this technique. Detection of mutations was verified in 60 patient samples from the data set with a sensitivity and specificity of 95% for each mutation. This demonstrates that quantum cascade laser infrared imaging can be used to analyze morphologic differences as well as molecular changes. Therefore, this single, one-step measurement provides comprehensive diagnostics of lung cancer histology types and most frequent mutations.
Collapse
Affiliation(s)
- Nina Goertzen
- Center for Protein Diagnostics, Biospectroscopy, Germany; Department of Biophysics, Faculty of Biology and Biotechnology, Ruhr University Bochum, Bochum, Germany
| | | | - Jana Fassunke
- Institut für Pathologie, Universitätsklinikum Köln, Germany
| | - Thomas Brüning
- Institute for Prevention and Occupational Medicine of the German Social Accident Insurance, Institute of the Ruhr University Bochum, Bochum, Germany
| | - Yon-Dschun Ko
- Department of Internal Medicine, Johanniter-Kliniken Bonn GmbH, Johanniter Krankenhaus, Bonn, Germany
| | - Joachim Schmidt
- Lung Cancer Center Bonn, Department of Thoracic Surgery, Helios Klinikum Bonn/Rhein-Sieg and Department of Surgery, Division of Thoracic Surgery, Universitätsklinikum Bonn, Germany
| | - Frederik Großerueschkamp
- Center for Protein Diagnostics, Biospectroscopy, Germany; Department of Biophysics, Faculty of Biology and Biotechnology, Ruhr University Bochum, Bochum, Germany
| | | | - Klaus Gerwert
- Center for Protein Diagnostics, Biospectroscopy, Germany; Department of Biophysics, Faculty of Biology and Biotechnology, Ruhr University Bochum, Bochum, Germany.
| |
Collapse
|
20
|
Koguchi D, Matsumoto K, Shimizu Y, Kobayashi M, Hirano S, Ikeda M, Sato Y, Iwamura M. Prognostic Impact of AHNAK2 Expression in Patients Treated with Radical Cystectomy. Cancers (Basel) 2021; 13:cancers13081748. [PMID: 33918555 PMCID: PMC8069489 DOI: 10.3390/cancers13081748] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2021] [Revised: 04/01/2021] [Accepted: 04/06/2021] [Indexed: 12/20/2022] Open
Abstract
Data regarding expression levels of AHNAK2 in bladder cancer (BCa) have been very scarce. We retrospectively reviewed clinical data including clinicopathological features in 120 patients who underwent radical cystectomy (RC) for BCa. The expression levels of AHNAK2 in the specimens obtained by RC were classified as low expression (LE) or high expression (HE) by immunohistochemical staining. Statistical analyses were performed to compare associations between the two AHNAK2 expression patterns and the prognoses in terms of recurrence-free survival (RFS) and cancer-specific survival (CSS). A Kaplan-Meier analysis showed that patients with HE had a significantly worse RFS and CSS than those with LE (hazard ratio [HR]: 1.78, 95% confidence interval [CI]: 1.02-2.98, p = 0.027 and HR: 1.91, 95% CI: 1.08-3.38, p = 0.023, respectively). In a multivariate analysis, independent risk factors for worse RFS and CSS were shown as HE (HR: 1.96, 95% CI: 1.08-3.53, p = 0.026 and HR: 2.22, 95% CI: 1.14-4.31, p = 0.019, respectively) and lymph node metastasis (HR: 2.04, 95% CI: 1.09-3.84, p = 0.026 and HR: 1.19, 95% CI: 1.25-4.97, p = 0.009, respectively). The present study showed that AHNAK2 acts as a novel prognostic biomarker in patients with RC for BCa.
Collapse
|
21
|
Proteomic comparison between different tissue preservation methods for identification of promising biomarkers of urothelial bladder cancer. Sci Rep 2021; 11:7595. [PMID: 33828141 PMCID: PMC8027873 DOI: 10.1038/s41598-021-87003-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Accepted: 03/22/2021] [Indexed: 11/08/2022] Open
Abstract
Samples in biobanks are generally preserved by formalin-fixation and paraffin-embedding (FFPE) and/or optimal cutting temperature compound (OCT)-embedding and subsequently frozen. Mass spectrometry (MS)-based analysis of these samples is now available via developed protocols, however, the differences in results with respect to preservation methods needs further investigation. Here we use bladder urothelial carcinoma tissue of two different tumor stages (Ta/T1-non-muscle invasive bladder cancer (NMIBC), and T2/T3-muscle invasive bladder cancer (MIBC)) which, upon sampling, were divided and preserved by FFPE and OCT. Samples were parallel processed from the two methods and proteins were analyzed with label-free quantitative MS. Over 700 and 1200 proteins were quantified in FFPE and OCT samples, respectively. Multivariate analysis indicates that the preservation method is the main source of variation, but also tumors of different stages could be differentiated. Proteins involved in mitochondrial function were overrepresented in OCT data but missing in the FFPE data, indicating that these proteins are not well preserved by FFPE. Concordant results for proteins such as HMGCS2 (uniquely quantified in Ta/T1 tumors), and LGALS1, ANXA5 and plastin (upregulated in T2/T3 tumors) were observed in both FFPE and OCT data, which supports the use of MS technology for biobank samples and encourages the further evaluation of these proteins as biomarkers.
Collapse
|
22
|
Raulf AP, Butke J, Menzen L, Küpper C, Großerueschkamp F, Gerwert K, Mosig A. A representation learning approach for recovering scatter-corrected spectra from Fourier-transform infrared spectra of tissue samples. JOURNAL OF BIOPHOTONICS 2021; 14:e202000385. [PMID: 33295130 DOI: 10.1002/jbio.202000385] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Revised: 11/23/2020] [Accepted: 11/29/2020] [Indexed: 06/12/2023]
Abstract
Infrared spectra obtained from cell or tissue specimen have commonly been observed to involve a significant degree of scattering effects, often Mie scattering, which probably overshadows biochemically relevant spectral information by a nonlinear, nonadditive spectral component in Fourier transform infrared (FTIR) spectroscopic measurements. Correspondingly, many successful machine learning approaches for FTIR spectra have relied on preprocessing procedures that computationally remove the scattering components from an infrared spectrum. We propose an approach to approximate this complex preprocessing function using deep neural networks. As we demonstrate, the resulting model is not just several orders of magnitudes faster, which is important for real-time clinical applications, but also generalizes strongly across different tissue types. Using Bayesian machine learning approaches, our approach unveils model uncertainty that coincides with a band shift in the amide I region that occurs when scattering is removed computationally based on an established physical model. Furthermore, our proposed method overcomes the trade-off between computation time and the corrected spectrum being biased towards an artificial reference spectrum.
Collapse
Affiliation(s)
- Arne P Raulf
- Center for Protein Diagnostics, Ruhr-University Bochum, Gesundheitscampus 4, Bochum, Germany
- Bioinformatics Group, Department for Biology and Biotechnology, Ruhr-University Bochum, Gesundheitscampus 4, Bochum, Germany
| | - Joshua Butke
- Center for Protein Diagnostics, Ruhr-University Bochum, Gesundheitscampus 4, Bochum, Germany
- Bioinformatics Group, Department for Biology and Biotechnology, Ruhr-University Bochum, Gesundheitscampus 4, Bochum, Germany
| | - Lukas Menzen
- Center for Protein Diagnostics, Ruhr-University Bochum, Gesundheitscampus 4, Bochum, Germany
| | - Claus Küpper
- Center for Protein Diagnostics, Ruhr-University Bochum, Gesundheitscampus 4, Bochum, Germany
- Chair of Biophysics, Department for Biology and Biotechnology, Ruhr-University Bochum, Gesundheitscampus 4, Bochum, Germany
| | - Frederik Großerueschkamp
- Center for Protein Diagnostics, Ruhr-University Bochum, Gesundheitscampus 4, Bochum, Germany
- Chair of Biophysics, Department for Biology and Biotechnology, Ruhr-University Bochum, Gesundheitscampus 4, Bochum, Germany
| | - Klaus Gerwert
- Center for Protein Diagnostics, Ruhr-University Bochum, Gesundheitscampus 4, Bochum, Germany
- Chair of Biophysics, Department for Biology and Biotechnology, Ruhr-University Bochum, Gesundheitscampus 4, Bochum, Germany
| | - Axel Mosig
- Center for Protein Diagnostics, Ruhr-University Bochum, Gesundheitscampus 4, Bochum, Germany
- Bioinformatics Group, Department for Biology and Biotechnology, Ruhr-University Bochum, Gesundheitscampus 4, Bochum, Germany
| |
Collapse
|
23
|
Li H, Zhu Q, Li K, Wu Z, Tang Z, Wang Z. Investigation of urinary components in rat model of ketamine-induced bladder fibrosis based on metabolomics. Transl Androl Urol 2021; 10:830-840. [PMID: 33718084 PMCID: PMC7947432 DOI: 10.21037/tau-20-1202] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Background Ketamine abuse has been linked to the system's damage, presenting with lower urinary tract symptoms (LUTS). While the pathogenesis of ketamine-induced urinary damage is not fully understood, fibrosis is believed to be a potential mechanism. A metabolomic investigation of the urinary metabolites in ketamine abuse was conducted to gain insights into its pathogenesis. Methods A rat model of ketamine induced bladder fibrosis was established through tail vein injection of ketamine hydrochloride and control group was established through tail vein injection of the equivalent normal saline. Hematoxylin and eosin (H&E) staining and Masson trichrome staining were performed to evaluated bladder pathology. Urinary components were detected based on a metabolomic approach using ultra-high performance liquid tandem chromatography quadrupole time of flight mass spectrometry (UHPLC-QTOFMS platform). Orthogonal projections analyzed the data to latent structures discriminant analysis (OPLS-DA) and bioinformatics analysis. Results The rat model of ketamine induced bladder fibrosis was confirmed through H&E and Masson trichrome staining. There were marked differences in the urinary metabolites between the experimental group and the control group. Compared to the control group, 16 kinds of differential metabolites were up-regulated and 102 differential metabolites were down-regulated in the urine samples of the ketamine group. Bioinformatics analysis revealed the related metabolic pathways. Conclusions Using a ketamine-induced bladder fibrosis rat model, this study identified the differential urinary metabolites expressed following ketamine treatment. These results provide vital clues for exploring the pathogenesis of ketamine-induced LUTS and may further contribute to the disease's diagnosis and treatment.
Collapse
Affiliation(s)
- Haozhen Li
- Department of Urology, Xiangya Hospital, Central South University, Changsha, China
| | - Quan Zhu
- Department of Urology, Xiangya Hospital, Central South University, Changsha, China
| | - Kaixuan Li
- Department of Urology, Xiangya Hospital, Central South University, Changsha, China
| | - Ziqiang Wu
- Department of Urology, Xiangya Hospital, Central South University, Changsha, China
| | - Zhengyan Tang
- Department of Urology, Xiangya Hospital, Central South University, Changsha, China.,Hunan Provincial Engineering Laboratory for Diagnosis and Treatment of Genitourinary System Disease, Changsha, China
| | - Zhao Wang
- Department of Urology, Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
24
|
Zheng L, Li S, Zheng X, Guo R, Qu W. AHNAK2 is a novel prognostic marker and correlates with immune infiltration in papillary thyroid cancer: Evidence from integrated analysis. Int Immunopharmacol 2020; 90:107185. [PMID: 33218938 DOI: 10.1016/j.intimp.2020.107185] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Revised: 11/02/2020] [Accepted: 11/03/2020] [Indexed: 12/20/2022]
Abstract
Papillary thyroid cancer (PTC) is the most prevalent endocrine tumor, and its incidence is still increasing. The mechanisms of PTC dedifferentiation and malignant progression remain unclear. In this study, we identified AHNAK2 as a key gene in PTC by differential expression analysis among four GEO datasets and validated its overexpression profile by data from the Oncomine, TCGA, and HPA databases and IHC staining analysis. AHNAK2 upregulation significantly correlated with advanced grades, stages, and lymph node events. Survival analysis suggested that AHNAK2 overexpression was coupled with poor overall survival. The immune infiltration analysis by TIMER and CIBERSORT indicated that AHNAK2 expression tightly correlated with the infiltration of diverse immune cell types, especially T cell subtypes. In addition, AHNAK2 is correlated with the expression of other conventional key genes of TC, such as PIK3CA, MAPK1, CTNNB1, and SLC5A5. AHNAK2 may be a novel prognostic marker for PTC.
Collapse
Affiliation(s)
- Long Zheng
- Department of Nuclear Medicine, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004, Shaanxi, China
| | - Shanshan Li
- Department of Epidemiology and Health Statistics, Health Science Center, Xi'an Jiaotong University, Xi'an 710061, Shaanxi, China
| | - Xianghong Zheng
- Department of Nuclear Medicine, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004, Shaanxi, China
| | - Rong Guo
- Department of Nuclear Medicine, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004, Shaanxi, China
| | - Wei Qu
- Department of Nuclear Medicine, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004, Shaanxi, China.
| |
Collapse
|
25
|
Xie Z, Lun Y, Li X, He Y, Wu S, Wang S, Sun J, He Y, Zhang J. Bioinformatics analysis of the clinical value and potential mechanisms of AHNAK2 in papillary thyroid carcinoma. Aging (Albany NY) 2020; 12:18163-18180. [PMID: 32966238 PMCID: PMC7585101 DOI: 10.18632/aging.103645] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Accepted: 06/22/2020] [Indexed: 01/24/2023]
Abstract
BACKGROUND AHNAK2 has been recently reported as a biomarker in many cancers. However, a systematic investigation of AHNAK2 in papillary thyroid carcinoma (PTC) has not been conducted. RESULTS AHNAK2 is overexpressed in PTC tissues and could be an independent prognostic factor. AHNAK2 expression was significantly high in patients with advanced stage, advanced T classification, lymph node metastasis, increased BRAF mutations and decreased RAS mutations. Cell adhesion-, cell junction-, and immune-related pathways were the most frequently noted in gene set enrichment analysis. AHNAK2 expression in PTC was positively correlated with immune infiltration and negatively correlated with AHNAK2 methylation. AHNAK2 expression was significantly positively correlated with tumor progression and poor overall survival (OS) in pan-cancer patients. CONCLUSIONS AHNAK2 is a good biomarker for the diagnosis and prognosis of PTC. AHNAK2 may promote thyroid cancer progression through cell adhesion-, cell junction-, and immune-related pathways. Methylation may act as an upstream regulator to inhibit the expression and biological function of AHNAK2. Additionally, AHNAK2 has broad prognostic value in pan-cancer. METHODS Based on The Cancer Genome Atlas (TCGA) data, we screened AHNAK2-related genes through weighted gene coexpression network analysis and explored the clinical value and the potential mechanism of AHNAK2 in PTC by multiomics analysis.
Collapse
Affiliation(s)
- Zhenyu Xie
- Department of Vascular and Thyroid Surgery, The First Hospital, China Medical University, Shenyang, China
| | - Yu Lun
- Department of Vascular and Thyroid Surgery, The First Hospital, China Medical University, Shenyang, China
| | - Xin Li
- Department of Vascular and Thyroid Surgery, The First Hospital, China Medical University, Shenyang, China
| | - Yuzhen He
- Department of Vascular and Thyroid Surgery, The First Hospital, China Medical University, Shenyang, China
| | - Song Wu
- Department of Vascular and Thyroid Surgery, The First Hospital, China Medical University, Shenyang, China
| | - Shiyue Wang
- Department of Vascular and Thyroid Surgery, The First Hospital, China Medical University, Shenyang, China
| | - Jianjian Sun
- Department of Vascular and Thyroid Surgery, The First Hospital, China Medical University, Shenyang, China
| | - Yuchen He
- Department of Vascular and Thyroid Surgery, The First Hospital, China Medical University, Shenyang, China
| | - Jian Zhang
- Department of Vascular and Thyroid Surgery, The First Hospital, China Medical University, Shenyang, China
| |
Collapse
|
26
|
Zhang S, Lu Y, Qi L, Wang H, Wang Z, Cai Z. AHNAK2 Is Associated with Poor Prognosis and Cell Migration in Lung Adenocarcinoma. BIOMED RESEARCH INTERNATIONAL 2020; 2020:8571932. [PMID: 32904605 PMCID: PMC7456490 DOI: 10.1155/2020/8571932] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Revised: 06/30/2020] [Accepted: 08/03/2020] [Indexed: 02/07/2023]
Abstract
BACKGROUND Lung adenocarcinoma (LUAD), as the main subtype of lung cancer, is one of the common causes of cancer-related deaths worldwide. The AHNAK family is correlated with cell structure and migration, cardiac calcium channel signaling, and tumor metastasis. Previous studies showed AHNAK2 could promote tumor progression and cell migration in melanoma and renal clear cell carcinoma. However, the role of AHNAK2 in LUAD remains unknown. METHODS We examined the levels of AHNAK2 in pathological specimens and the database of Clinical Proteomic Tumor Analysis Consortium-Lung adenocarcinoma (CPTAC-LUAD), The Cancer Genome Atlas-Lung Adenocarcinoma (TCGA-LUAD), Gene Expression Omnibus dataset (GSE72094, GSE26939), and The Genotype-Tissue Expression (GTEx) of lung tissue samples. Univariate Cox regression, multivariate Cox regression, and Kaplan-Meier survival analysis were performed to reveal the relationship between AHNAK2 and prognosis. A nomogram was constructed to predict 2- or 3-year overall survival and validated via calibration curves, receiver operating characteristic (ROC) analysis, and decision curve analysis (DCA). Furthermore, Gene Ontology (GO) analysis and Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis were used to explore the functional role of AHNAK2 in lung adenocarcinoma. Finally, by transfecting siRNA, we examined the regulatory effect of AHNAK2 on cell migration. RESULTS The expression of AHNAK2 was upregulated in tumor samples and correlated with poor prognosis in LUAD patients. Nomogram with AHNAK2 and clinical parameters showed a good prediction in overall survival (OS), especially the 2-year OS. In addition, functional analyses and wound healing assay suggested that AHNAK2 might be involved in the regulation of migration in LUAD. CONCLUSION In summary, our study showed that AHNAK2 might be a novel biomarker in LUAD and revealed the potential mechanism of AHNAK2 in LUAD progression which could provide new insights for target therapy.
Collapse
Affiliation(s)
- Shusen Zhang
- Department of Respiratory and Critical Care Medicine, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
- Department of Respiratory and Critical Care Medicine, Affiliated Xing Tai People Hospital of Hebei Medical University, Xingtai, Hebei, China
| | - Yuanyuan Lu
- Department of Anesthesiology, Affiliated Xing Tai People Hospital of Hebei Medical University, Xingtai, Hebei, China
| | - Lei Qi
- Department of Pathology, Affiliated Xing Tai People Hospital of Hebei Medical University, Xingtai, Hebei, China
| | - Hongyan Wang
- Department of Thoracic Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Zhihua Wang
- Department of Respiratory and Critical Care Medicine, Affiliated Xing Tai People Hospital of Hebei Medical University, Xingtai, Hebei, China
| | - Zhigang Cai
- Department of Respiratory and Critical Care Medicine, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| |
Collapse
|
27
|
Trukhan S, Tafintseva V, Tøndel K, Großerueschkamp F, Mosig A, Kovalev V, Gerwert K, Kohler A. Grayscale representation of infrared microscopy images by extended multiplicative signal correction for registration with histological images. JOURNAL OF BIOPHOTONICS 2020; 13:e201960223. [PMID: 32352634 DOI: 10.1002/jbio.201960223] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Revised: 04/20/2020] [Accepted: 04/21/2020] [Indexed: 06/11/2023]
Abstract
Fourier-transform infrared (FTIR) microspectroscopy is rounding the corner to become a label-free routine method for cancer diagnosis. In order to build infrared-spectral based classifiers, infrared images need to be registered with Hematoxylin and Eosin (H&E) stained histological images. While FTIR images have a deep spectral domain with thousands of channels carrying chemical and scatter information, the H&E images have only three color channels for each pixel and carry mainly morphological information. Therefore, image representations of infrared images are needed that match the morphological information in H&E images. In this paper, we propose a novel approach for representation of FTIR images based on extended multiplicative signal correction highlighting morphological features that showed to correlate well with morphological information in H&E images. Based on the obtained representations, we developed a strategy for global-to-local image registration for FTIR images and H&E stained histological images of parallel tissue sections.
Collapse
Affiliation(s)
- Stanislau Trukhan
- Faculty of Science and Technology, Norwegian University of Life Sciences, Ås, Norway
- Department of Biomedical Image Analysis, United Institute of Informatics Problems, Minsk, Belarus
| | - Valeria Tafintseva
- Faculty of Science and Technology, Norwegian University of Life Sciences, Ås, Norway
| | - Kristin Tøndel
- Faculty of Science and Technology, Norwegian University of Life Sciences, Ås, Norway
| | - Frederik Großerueschkamp
- Departament of Biophysics, Ruhr University Bochum, Bochum, Germany
- Center for Protein Diagnostics (ProDi), Ruhr University Bochum, Bochum, Germany
| | - Axel Mosig
- Departament of Biophysics, Ruhr University Bochum, Bochum, Germany
- Center for Protein Diagnostics (ProDi), Ruhr University Bochum, Bochum, Germany
| | - Vassili Kovalev
- Department of Biomedical Image Analysis, United Institute of Informatics Problems, Minsk, Belarus
| | - Klaus Gerwert
- Departament of Biophysics, Ruhr University Bochum, Bochum, Germany
- Center for Protein Diagnostics (ProDi), Ruhr University Bochum, Bochum, Germany
| | - Achim Kohler
- Faculty of Science and Technology, Norwegian University of Life Sciences, Ås, Norway
| |
Collapse
|
28
|
Eggeling F, Hoffmann F. Microdissection—An Essential Prerequisite for Spatial Cancer Omics. Proteomics 2020; 20:e2000077. [DOI: 10.1002/pmic.202000077] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Revised: 06/12/2020] [Indexed: 12/12/2022]
Affiliation(s)
- Ferdinand Eggeling
- Department of OtorhinolaryngologyMALDI Imaging and Core Unit Proteome AnalysisDFG Core Unit Jena Biophotonic and Imaging Laboratory (JBIL)Jena University Hospital Am Klinikum 1 Jena 07747 Germany
| | - Franziska Hoffmann
- Department of OtorhinolaryngologyMALDI Imaging and Core Unit Proteome AnalysisDFG Core Unit Jena Biophotonic and Imaging Laboratory (JBIL)Jena University Hospital Am Klinikum 1 Jena 07747 Germany
| |
Collapse
|
29
|
Kallenbach-Thieltges A, Großerueschkamp F, Jütte H, Kuepper C, Reinacher-Schick A, Tannapfel A, Gerwert K. Label-free, automated classification of microsatellite status in colorectal cancer by infrared imaging. Sci Rep 2020; 10:10161. [PMID: 32576892 PMCID: PMC7311536 DOI: 10.1038/s41598-020-67052-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Accepted: 05/21/2020] [Indexed: 12/12/2022] Open
Abstract
Challenging histopathological diagnostics in cancer include microsatellite instability-high (MSI-H) colorectal cancer (CRC), which occurs in 15% of early-stage CRC and is caused by a deficiency in the mismatch repair system. The diagnosis of MSI-H cannot be reliably achieved by visual inspection of a hematoxylin and eosin stained thin section alone, but additionally requires subsequent molecular analysis. Time- and sample-intensive immunohistochemistry with subsequent fragment length analysis is used. The aim of the presented feasibility study is to test the ability of quantum cascade laser (QCL)-based infrared (IR) imaging as an alternative diagnostic tool for MSI-H in CRC. We analyzed samples from 100 patients with sporadic CRC UICC stage II and III. Forty samples were used to develop the random forest classifier and 60 samples to verify the results on an independent blinded dataset. Specifically, 100% sensitivity and 93% specificity were achieved based on the independent 30 MSI-H- and 30 microsatellite stable (MSS)-patient validation cohort. This showed that QCL-based IR imaging is able to distinguish between MSI-H and MSS for sporadic CRC - a question that goes beyond morphological features - based on the use of spatially resolved infrared spectra used as biomolecular fingerprints.
Collapse
Affiliation(s)
- Angela Kallenbach-Thieltges
- Ruhr University Bochum, Center for Protein Diagnostics (ProDi), Biospectroscopy, Bochum, Germany.,Ruhr University Bochum, Faculty of Biology and Biotechnology, Department of Biophysics, Bochum, Germany
| | - Frederik Großerueschkamp
- Ruhr University Bochum, Center for Protein Diagnostics (ProDi), Biospectroscopy, Bochum, Germany.,Ruhr University Bochum, Faculty of Biology and Biotechnology, Department of Biophysics, Bochum, Germany
| | - Hendrik Jütte
- Institute of Pathology, Ruhr University Bochum, Bochum, Germany
| | - Claus Kuepper
- Ruhr University Bochum, Center for Protein Diagnostics (ProDi), Biospectroscopy, Bochum, Germany.,Ruhr University Bochum, Faculty of Biology and Biotechnology, Department of Biophysics, Bochum, Germany
| | - Anke Reinacher-Schick
- Department of Hematology, Oncology and Palliative Care, St. Josef Hospital, Ruhr University Bochum, Bochum, Germany
| | | | - Klaus Gerwert
- Ruhr University Bochum, Center for Protein Diagnostics (ProDi), Biospectroscopy, Bochum, Germany. .,Ruhr University Bochum, Faculty of Biology and Biotechnology, Department of Biophysics, Bochum, Germany.
| |
Collapse
|
30
|
Criscitiello MF, Kraev I, Petersen LH, Lange S. Deimination Protein Profiles in Alligator mississippiensis Reveal Plasma and Extracellular Vesicle-Specific Signatures Relating to Immunity, Metabolic Function, and Gene Regulation. Front Immunol 2020; 11:651. [PMID: 32411128 PMCID: PMC7198796 DOI: 10.3389/fimmu.2020.00651] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Accepted: 03/23/2020] [Indexed: 12/13/2022] Open
Abstract
Alligators are crocodilians and among few species that endured the Cretaceous-Paleogene extinction event. With long life spans, low metabolic rates, unusual immunological characteristics, including strong antibacterial and antiviral ability, and cancer resistance, crocodilians may hold information for molecular pathways underlying such physiological traits. Peptidylarginine deiminases (PADs) are a group of calcium-activated enzymes that cause posttranslational protein deimination/citrullination in a range of target proteins contributing to protein moonlighting functions in health and disease. PADs are phylogenetically conserved and are also a key regulator of extracellular vesicle (EV) release, a critical part of cellular communication. As little is known about PAD-mediated mechanisms in reptile immunology, this study was aimed at profiling EVs and protein deimination in Alligator mississippiensis. Alligator plasma EVs were found to be polydispersed in a 50-400-nm size range. Key immune, metabolic, and gene regulatory proteins were identified to be posttranslationally deiminated in plasma and plasma EVs, with some overlapping hits, while some were unique to either plasma or plasma EVs. In whole plasma, 112 target proteins were identified to be deiminated, while 77 proteins were found as deiminated protein hits in plasma EVs, whereof 31 were specific for EVs only, including proteins specific for gene regulatory functions (e.g., histones). Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analysis revealed KEGG pathways specific to deiminated proteins in whole plasma related to adipocytokine signaling, while KEGG pathways of deiminated proteins specific to EVs included ribosome, biosynthesis of amino acids, and glycolysis/gluconeogenesis pathways as well as core histones. This highlights roles for EV-mediated export of deiminated protein cargo with roles in metabolism and gene regulation, also related to cancer. The identification of posttranslational deimination and EV-mediated communication in alligator plasma revealed here contributes to current understanding of protein moonlighting functions and EV-mediated communication in these ancient reptiles, providing novel insight into their unusual immune systems and physiological traits. In addition, our findings may shed light on pathways underlying cancer resistance, antibacterial and antiviral resistance, with translatable value to human pathologies.
Collapse
Affiliation(s)
- Michael F. Criscitiello
- Comparative Immunogenetics Laboratory, Department of Veterinary Pathobiology, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX, United States
- Department of Microbial Pathogenesis and Immunology, College of Medicine, Texas A&M Health Science Center, Texas A&M University, College Station, TX, United States
| | - Igor Kraev
- Electron Microscopy Suite, Faculty of Science, Technology, Engineering and Mathematics, Open University, Milton Keynes, United Kingdom
| | - Lene H. Petersen
- Department of Marine Biology, Texas A&M University at Galvestone, Galveston, TX, United States
| | - Sigrun Lange
- Tissue Architecture and Regeneration Research Group, School of Life Sciences, University of Westminster, London, United Kingdom
| |
Collapse
|
31
|
Voith von Voithenberg L, Fomitcheva Khartchenko A, Huber D, Schraml P, Kaigala GV. Spatially multiplexed RNA in situ hybridization to reveal tumor heterogeneity. Nucleic Acids Res 2020; 48:e17. [PMID: 31853536 PMCID: PMC7026647 DOI: 10.1093/nar/gkz1151] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Revised: 11/20/2019] [Accepted: 12/03/2019] [Indexed: 02/07/2023] Open
Abstract
Multiplexed RNA in situ hybridization for the analysis of gene expression patterns plays an important role in investigating development and disease. Here, we present a method for multiplexed RNA-ISH to detect spatial tumor heterogeneity in tissue sections. We made use of a microfluidic chip to deliver ISH-probes locally to regions of a few hundred micrometers over time periods of tens of minutes. This spatial multiplexing method can be combined with ISH-approaches based on signal amplification, with bright field detection and with the commonly used format of formalin-fixed paraffin-embedded tissue sections. By using this method, we analyzed the expression of HER2 with internal positive and negative controls (ActB, dapB) as well as predictive biomarker panels (ER, PgR, HER2) in a spatially multiplexed manner on single mammary carcinoma sections. We further demonstrated the applicability of the technique for subtype differentiation in breast cancer. Local analysis of HER2 revealed medium to high spatial heterogeneity of gene expression (Cohen effect size r = 0.4) in equivocally tested tumor tissues. Thereby, we exemplify the importance of using such a complementary approach for the analysis of spatial heterogeneity, in particular for equivocally tested tumor samples. As the method is compatible with a range of ISH approaches and tissue samples, it has the potential to find broad applicability in the context of molecular analysis of human diseases.
Collapse
Affiliation(s)
| | | | - Deborah Huber
- IBM Research Zürich, Säumerstrasse 4, CH-8803 Rüschlikon, Switzerland
| | - Peter Schraml
- University Hospital Zurich, Department of Pathology and Molecular Pathology, Schmelzbergstr. 12, CH-8091 Zurich, Switzerland
| | - Govind V Kaigala
- IBM Research Zürich, Säumerstrasse 4, CH-8803 Rüschlikon, Switzerland
| |
Collapse
|
32
|
Zhu S, Yu W, Yang X, Wu C, Cheng F. Traditional Classification and Novel Subtyping Systems for Bladder Cancer. Front Oncol 2020; 10:102. [PMID: 32117752 PMCID: PMC7025453 DOI: 10.3389/fonc.2020.00102] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2019] [Accepted: 01/20/2020] [Indexed: 12/31/2022] Open
Abstract
Bladder cancer is the most common tumor in the urinary system, with approximately 420,000 new cases and 160,000 deaths per year. The European Organization for Research and Treatment of Cancer (EOTRC) classifies non-muscular invasive bladder cancer (NMIBC) into low-risk, medium-risk and high-risk groups based on a comprehensive analysis of NMIBC pathological parameters and the risk of recurrence and progression to muscular invasive bladder cancer (MIBC). Traditional classification systems are based on pathologic grading, staging systems, and clinical prognosis. However, the pathological parameters of the tumor cannot fully reflect the “intrinsic characteristics” of bladder cancer, and tumors with a similar pathology exhibit different biological behaviors. Furthermore, although the traditional classification system cannot accurately predict the risk of recurrence or the progression of bladder cancer patients (BCs) individually, this method is widely used in clinical practice because of its convenient operation. With the development of sequencing and other technologies, the genetics-based molecular subtyping of bladder cancer has become increasingly studied. Compared with the traditional classification system, it provides more abundant tumor biological information and is expected to assist or even replace the traditional typing system in the future.
Collapse
Affiliation(s)
- Shaoming Zhu
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Weimin Yu
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Xiao Yang
- Department of Gynaecology and Obstetrics, Renmin Hospital of Peking University, Beijing, China
| | - Cheng Wu
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Fan Cheng
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
33
|
Almeida PP, Cardoso CP, de Freitas LM. PDAC-ANN: an artificial neural network to predict pancreatic ductal adenocarcinoma based on gene expression. BMC Cancer 2020; 20:82. [PMID: 32005189 PMCID: PMC6995241 DOI: 10.1186/s12885-020-6533-0] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2019] [Accepted: 01/13/2020] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Although the pancreatic ductal adenocarcinoma (PDAC) presents high mortality and metastatic potential, there is a lack of effective therapies and a low survival rate for this disease. This PDAC scenario urges new strategies for diagnosis, drug targets, and treatment. METHODS We performed a gene expression microarray meta-analysis of the tumor against normal tissues in order to identify differentially expressed genes (DEG) shared among all datasets, named core-genes (CG). We confirmed the CG protein expression in pancreatic tissue through The Human Protein Atlas. It was selected five genes with the highest area under the curve (AUC) among these proteins with expression confirmed in the tumor group to train an artificial neural network (ANN) to classify samples. RESULTS This microarray included 461 tumor and 187 normal samples. We identified a CG composed of 40 genes, 39 upregulated, and one downregulated. The upregulated CG included proteins and extracellular matrix receptors linked to actin cytoskeleton reorganization. With the Human Protein Atlas, we verified that fourteen genes of the CG are translated, with high or medium expression in most of the pancreatic tumor samples. To train our ANN, we selected the best genes (AHNAK2, KRT19, LAMB3, LAMC2, and S100P) to classify the samples based on AUC using mRNA expression. The network classified tumor samples with an f1-score of 0.83 for the normal samples and 0.88 for the PDAC samples, with an average of 0.86. The PDAC-ANN could classify the test samples with a sensitivity of 87.6 and specificity of 83.1. CONCLUSION The gene expression meta-analysis and confirmation of the protein expression allow us to select five genes highly expressed PDAC samples. We could build a python script to classify the samples based on RNA expression. This software can be useful in the PDAC diagnosis.
Collapse
Affiliation(s)
- Palloma Porto Almeida
- Núcleo de Biointegração, Instituto Multidisciplinar em Saúde, Universidade Federal da Bahia, Vitória da Conquista, Brazil
| | - Cristina Padre Cardoso
- Núcleo de Biointegração, Instituto Multidisciplinar em Saúde, Universidade Federal da Bahia, Vitória da Conquista, Brazil
- Faculdade Santo Agostinho, Vitória da Conquista, Brazil
| | - Leandro Martins de Freitas
- Núcleo de Biointegração, Instituto Multidisciplinar em Saúde, Universidade Federal da Bahia, Vitória da Conquista, Brazil.
| |
Collapse
|
34
|
Yu X, Wang R, Han C, Wang Z, Jin X. A Panel of Urinary Long Non-coding RNAs Differentiate Bladder Cancer from Urocystitis. J Cancer 2020; 11:781-787. [PMID: 31949480 PMCID: PMC6959012 DOI: 10.7150/jca.37006] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2019] [Accepted: 09/27/2019] [Indexed: 12/16/2022] Open
Abstract
Liquid biopsy is becoming a promising method for non-invasive cancer detection. In several proof-of-concept studies, long non-coding RNAs (lncRNAs) were found to be potential biomarkers for bladder cancer detection. The objective of this study was to discover a panel of cell-free, urinary lncRNAs as liquid biopsy biomarkers to non-invasively differentiate bladder cancer from chronic urocystitis. To this end, we collected urine samples from both bladder cancer patients and urocystitis patients. These samples were divided into discovery group and validation group. In the discovery group, the expression levels of 16 cell-free urinary lncRNAs were measured by qPCR to discover candidate biomarkers. The diagnostic performance of the candidate lncRNA biomarkers was then evaluated, which led to a panel of lncRNA biomarkers for bladder cancer detection. The performance of this panel of biomarkers was further evaluated in the validation group to see if these lncRNA biomarkers could discriminate the bladder cancer patients from urocystitis patients. We found that all of the 16 lncRNAs evaluated in this study demonstrated significant difference (p<0.05) of expression between bladder cancer patients and urocystitis patients. Nine lncRNAs provided decent diagnostic performance with area under the receiver operating characteristic (ROC) curve (AUC) reaching 0.70 or higher. We then selected the top four lncRNAs, namely UCA1-201, HOTAIR, HYMA1 and MALAT1, to form a panel of urinary biomarkers. Using this panel, bladder cancer patients could be discriminated from urocystitis patients, with sensitivity and specificity reaching 95.7% and 94.3%, respectively. Finally, we confirmed the applicability of the four-lncRNA panel in an independent validation study that included 60 bladder cancer patients and 60 urocystitis patients. Our study paves the way for further studies aimed at large-scale clinical tests of developing lncRNA biomarkers in urine for bladder cancer diagnostics.
Collapse
Affiliation(s)
- Xiao Yu
- Department of Urology, Shandong Provincial Hospital Affiliated to Shandong University, No. 324 Jingwu Road, Huaiyin District, Jinan, Shandong, 250021, China
| | - Ruiwei Wang
- Department of Anesthesiology, Shandong Provincial Hospital Affiliated to Shandong University, No. 324 Jingwu Road, Huaiyin District, Jinan, Shandong, 250021, China
| | - Chenglin Han
- Department of Urology, Shandong Provincial Hospital Affiliated to Shandong University, No. 324 Jingwu Road, Huaiyin District, Jinan, Shandong, 250021, China
| | - Zilong Wang
- Department of Urology, Shandong Provincial Hospital Affiliated to Shandong University, No. 324 Jingwu Road, Huaiyin District, Jinan, Shandong, 250021, China
| | - Xunbo Jin
- Department of Urology, Shandong Provincial Hospital Affiliated to Shandong University, No. 324 Jingwu Road, Huaiyin District, Jinan, Shandong, 250021, China
| |
Collapse
|
35
|
Lang K, Kahveci S, Bonberg N, Wichert K, Behrens T, Hovanec J, Roghmann F, Noldus J, Tam YC, Tannapfel A, Käfferlein HU, Brüning T. TGFBI Protein Is Increased in the Urine of Patients with High-Grade Urothelial Carcinomas, and Promotes Cell Proliferation and Migration. Int J Mol Sci 2019; 20:ijms20184483. [PMID: 31514337 PMCID: PMC6770034 DOI: 10.3390/ijms20184483] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2019] [Revised: 09/05/2019] [Accepted: 09/06/2019] [Indexed: 01/03/2023] Open
Abstract
Here, we discovered TGFBI as a new urinary biomarker for muscle invasive and high-grade urothelial carcinoma (UC). After biomarker identification using antibody arrays, results were verified in urine samples from a study population consisting of 303 patients with UC, and 128 urological and 58 population controls. The analyses of possible modifying factors (age, sex, smoking status, urinary leukocytes and erythrocytes, and history of UC) were calculated by multiple logistic regression. Additionally, we performed knockdown experiments with TGFBI siRNA in bladder cancer cells and investigated the effects on proliferation and migration by wound closure assays and BrdU cell cycle analysis. TGFBI concentrations in urine are generally increased in patients with UC when compared to urological and population controls (1321.0 versus 701.3 and 475.6 pg/mg creatinine, respectively). However, significantly increased TGFBI was predominantly found in muscle invasive (14,411.7 pg/mg creatinine), high-grade (8190.7 pg/mg) and de novo UC (1856.7 pg/mg; all p < 0.0001). Knockdown experiments in vitro led to a significant decline of cell proliferation and migration. In summary, our results suggest a critical role of TGFBI in UC tumorigenesis and particularly in high-risk UC patients with poor prognosis and an elevated risk of progression on the molecular level.
Collapse
Affiliation(s)
- Kerstin Lang
- Institute for Prevention and Occupational Medicine of the German Social Accident Insurance, Institute of the Ruhr-University Bochum (IPA), Bürkle-de-la-Camp Platz 1, 44789 Bochum, Germany.
| | - Selcan Kahveci
- Institute for Prevention and Occupational Medicine of the German Social Accident Insurance, Institute of the Ruhr-University Bochum (IPA), Bürkle-de-la-Camp Platz 1, 44789 Bochum, Germany.
| | - Nadine Bonberg
- Institute for Prevention and Occupational Medicine of the German Social Accident Insurance, Institute of the Ruhr-University Bochum (IPA), Bürkle-de-la-Camp Platz 1, 44789 Bochum, Germany.
| | - Katharina Wichert
- Institute for Prevention and Occupational Medicine of the German Social Accident Insurance, Institute of the Ruhr-University Bochum (IPA), Bürkle-de-la-Camp Platz 1, 44789 Bochum, Germany.
| | - Thomas Behrens
- Institute for Prevention and Occupational Medicine of the German Social Accident Insurance, Institute of the Ruhr-University Bochum (IPA), Bürkle-de-la-Camp Platz 1, 44789 Bochum, Germany.
| | - Jan Hovanec
- Institute for Prevention and Occupational Medicine of the German Social Accident Insurance, Institute of the Ruhr-University Bochum (IPA), Bürkle-de-la-Camp Platz 1, 44789 Bochum, Germany.
| | - Florian Roghmann
- Department of Urology, Marien Hospital Herne, University Hospital of the Ruhr University Bochum, Hölkeskampring 40, 44625 Herne, Germany.
| | - Joachim Noldus
- Department of Urology, Marien Hospital Herne, University Hospital of the Ruhr University Bochum, Hölkeskampring 40, 44625 Herne, Germany.
| | - Yu Chun Tam
- Institute of Pathology, Georgius Agricola Stiftung Ruhr, Ruhr University Bochum, Bürkle-de-la-Camp Platz 1, 44789 Bochum, Germany.
| | - Andrea Tannapfel
- Institute of Pathology, Georgius Agricola Stiftung Ruhr, Ruhr University Bochum, Bürkle-de-la-Camp Platz 1, 44789 Bochum, Germany.
| | - Heiko U Käfferlein
- Institute for Prevention and Occupational Medicine of the German Social Accident Insurance, Institute of the Ruhr-University Bochum (IPA), Bürkle-de-la-Camp Platz 1, 44789 Bochum, Germany.
| | - Thomas Brüning
- Institute for Prevention and Occupational Medicine of the German Social Accident Insurance, Institute of the Ruhr-University Bochum (IPA), Bürkle-de-la-Camp Platz 1, 44789 Bochum, Germany.
| |
Collapse
|