51
|
Zhu F, Pan Z, Tang Y, Fu P, Cheng S, Hou W, Zhang Q, Huang H, Sun Y. Machine learning models predict coagulopathy in spontaneous intracerebral hemorrhage patients in ER. CNS Neurosci Ther 2021; 27:92-100. [PMID: 33249760 PMCID: PMC7804781 DOI: 10.1111/cns.13509] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Revised: 10/25/2020] [Accepted: 10/25/2020] [Indexed: 01/01/2023] Open
Abstract
AIMS Coagulation abnormality is one of the primary concerns for patients with spontaneous intracerebral hemorrhage admitted to ER. Conventional laboratory indicators require hours for coagulopathy diagnosis, which brings difficulties for appropriate intervention within the optimal window. This study evaluates the possibility of building efficient coagulopathy prediction models using data mining and machine learning algorithms. METHODS A retrospective cohort enrolled 1668 cases with acute spontaneous intracerebral hemorrhage from three medical centers, excluding those under antithrombotic therapies. Coagulopathy-related clinical parameters were initially screened by univariate analysis. Two machine learning algorithms, the random forest and the support vector machine, were deployed via an approach of four-fold cross-validation to screen out the most important parameters contributing to the occurrence of coagulopathy. Model discrimination was assessed using metrics, including accuracy, precision, recall, and F1 score. RESULTS Albumin/globulin ratio, neutrophil count, lymphocyte percentage, aspartate transaminase, alanine transaminase, hemoglobin, platelet count, white blood cell count, neutrophil percentage, systolic and diastolic pressure were identified as major predictors to the occurrence of acute coagulopathy. Compared to support vector machine, the model based on the random forest algorithm showed better accuracy (93.1%, 95% confidence interval [CI]: 0.913-0.950), precision (92.4%, 95% CI: 0.897-0.951), F1 score (91.5%, 95% CI: 0.889-0.964), and recall score (93.6%, 95% CI: 0.909-0.964), and yielded higher area under the receiver operating characteristic curve (AU-ROC) (0.962, 95% CI: 0.942-0.982). CONCLUSION The constructed models exhibit good prediction accuracy and efficiency. It might be used in clinical practice to facilitate target intervention for acute coagulopathy in patients with spontaneous intracerebral hemorrhage.
Collapse
Affiliation(s)
- Fengping Zhu
- Department of NeurosurgeryHuahsan HospitalFudan UniversityShanghaiChina
- Neurosurgical Institute of Fudan UniversityShanghaiChina
- Shanghai Clinical Medical Center of NeurosurgeryShanghaiChina
- Shanghai Key Laboratory of Brain Function and Restoration and Neural RegenerationShanghaiChina
| | - Zhiguang Pan
- Department of NeurosurgeryHuahsan HospitalFudan UniversityShanghaiChina
- Neurosurgical Institute of Fudan UniversityShanghaiChina
- Shanghai Clinical Medical Center of NeurosurgeryShanghaiChina
- Shanghai Key Laboratory of Brain Function and Restoration and Neural RegenerationShanghaiChina
| | - Ying Tang
- Department of NursingHuahsan HospitalFudan UniversityShanghaiChina
| | - Pengfei Fu
- Department of NeurosurgeryHuahsan HospitalFudan UniversityShanghaiChina
| | - Sijie Cheng
- Information CenterHuahsan HospitalFudan UniversityShanghaiChina
| | - Wenzhong Hou
- Information CenterHuahsan HospitalFudan UniversityShanghaiChina
| | - Qi Zhang
- Information CenterHuahsan HospitalFudan UniversityShanghaiChina
| | - Hong Huang
- Information CenterHuahsan HospitalFudan UniversityShanghaiChina
| | - Yirui Sun
- Department of NeurosurgeryHuahsan HospitalFudan UniversityShanghaiChina
- Neurosurgical Institute of Fudan UniversityShanghaiChina
- Shanghai Clinical Medical Center of NeurosurgeryShanghaiChina
- Shanghai Key Laboratory of Brain Function and Restoration and Neural RegenerationShanghaiChina
| |
Collapse
|
52
|
Garadi Suresh H, Mattiazzi Usaj M. Systematic High-Content Screening of Fluorescently Tagged Yeast Double Mutant Strains. Methods Mol Biol 2021; 2381:57-78. [PMID: 34590270 DOI: 10.1007/978-1-0716-1740-3_3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2023]
Abstract
We describe a protocol for high-content screening in budding yeast that can be used to study genetic interactions from a cell biological perspective. This approach can be used to map genetic interactions by monitoring one or more subcellular fluorescent markers of interest. In this case, changes in the morphology or abundance of a subcellular compartment, pathway or bioprocess are monitored in the background of a systematic array of yeast double mutants. Alternatively, the protocol can be used to monitor proteome-wide abundance and localization changes in a double mutant of interest by screening the yeast ORF-GFP collection. The protocol can be readily adapted for high-content screening of triple mutants, other large-scale yeast collections or expanded to screening of multiple growth conditions.
Collapse
|
53
|
K R, Voigt SP, Kalidindi SR, Basu B. Critical comparison of image analysis workflows for quantitative cell morphological evaluation in assessing cell response to biomaterials. Biomed Mater 2020; 16. [PMID: 33260169 DOI: 10.1088/1748-605x/abcf5e] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Accepted: 12/01/2020] [Indexed: 11/11/2022]
Abstract
Quantitative image analysis is an important tool in understanding cell fate processes through the study of cell morphological changes in terms of size, shape, number, and orientation. In this context, this work explores systematically the main challenges involved in the quantitative analysis of fluorescence microscopy images and also proposes a new protocol while comparing its outcome with the widely used Image J analysis. It is important to mention that fluorescence microscopy is by far most widely used in biocompatibility analysis (observing cell fate changes) of implantable biomaterials. In this study, we employed two different image analyses toolsets: (i) the conventionally employed ImageJ software, and (ii) a recently developed automated digital image analyses framework, called ImageMKS. While ImageJ offers a powerful toolset for image analyses, it requires sophisticated user expertise to design and iteratively refine the analyses workflow. This workflow primarily comprises a sequence of image transformations that typically involve de-noising and labelling of features. On the other hand, ImageMKS automates the image analyses protocol to a large extent, and thereby mitigates the influence of the user bias on the final results. This aspect is addressed using a case study of C2C12 mouse myoblast cells grown on Poly(vinyldiene difluoride) based polymeric substrates in the presence of an external electric field. In particular, we used a number of fluorescence microscopy images of murine myoblasts (muscle precursor cells) grown on Poly (vinylidene difluoride), PVDF based nanobiocomposites under the influence of electric field. It was observed that when compared with the findings obtained from ImageJ, ImageMKS workflows consistently produced more reliable results that correlated better with the prior studies. Furthermore, the MKS workflows required much less user time, because of their automation.
Collapse
Affiliation(s)
- Ravikumar K
- Materials Research Centre, Indian Institute of Science,Bangalore, C V Raman Avenue, Bangalore, Karnataka, 560012, INDIA
| | - Sven P Voigt
- School of Mechanical Engineering, Georgia Institute of Technology, 801 Ferst Drive Georgia Institute of Technology Atlanta, GA, Atlanta, Georgia, 30332, UNITED STATES
| | - Surya R Kalidindi
- School of Mechanical Engineering, Georgia Institute of Technology, 801 Ferst Drive, Atlanta, GA 30332-0405, USA, Atlanta, Georgia, 30332, UNITED STATES
| | - Bikramjit Basu
- Materials Research Centre, Indian Institute of Science,Bangalore, C V Raman Avenue, Bangalore, Karnataka, 560012, INDIA
| |
Collapse
|
54
|
Next-Generation Lineage Tracing and Fate Mapping to Interrogate Development. Dev Cell 2020; 56:7-21. [PMID: 33217333 DOI: 10.1016/j.devcel.2020.10.021] [Citation(s) in RCA: 79] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 09/15/2020] [Accepted: 10/27/2020] [Indexed: 12/20/2022]
Abstract
Lineage tracing and fate mapping, overlapping yet distinct disciplines to follow cells and their progeny, have evolved rapidly over the last century. Lineage tracing aims to identify all progeny arising from an individual cell, placing them within a lineage hierarchy. The recent emergence of genomic technologies, such as single-cell and spatial transcriptomics, has fostered sophisticated new methods to reconstruct lineage relationships at high resolution. In contrast, fate maps, schematics showing which parts of the embryo will develop into which tissue, have remained relatively static since the 1970s. However, fate maps provide spatial information, often lost in lineage reconstruction, that can offer fundamental mechanistic insight into development. Here, we broadly review the origins of fate mapping and lineage tracing approaches. We focus on the most recent developments in lineage tracing, permitted by advances in single-cell genomics. Finally, we explore the current potential to leverage these new technologies to synthesize high-resolution fate maps and discuss their potential for interrogating development at new depths.
Collapse
|
55
|
Sanicola HW, Stewart CE, Mueller M, Ahmadi F, Wang D, Powell SK, Sarkar K, Cutbush K, Woodruff MA, Brafman DA. Guidelines for establishing a 3-D printing biofabrication laboratory. Biotechnol Adv 2020; 45:107652. [PMID: 33122013 DOI: 10.1016/j.biotechadv.2020.107652] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 10/21/2020] [Accepted: 10/23/2020] [Indexed: 12/23/2022]
Abstract
Advanced manufacturing and 3D printing are transformative technologies currently undergoing rapid adoption in healthcare, a traditionally non-manufacturing sector. Recent development in this field, largely enabled by merging different disciplines, has led to important clinical applications from anatomical models to regenerative bioscaffolding and devices. Although much research to-date has focussed on materials, designs, processes, and products, little attention has been given to the design and requirements of facilities for enabling clinically relevant biofabrication solutions. These facilities are critical to overcoming the major hurdles to clinical translation, including solving important issues such as reproducibility, quality control, regulations, and commercialization. To improve process uniformity and ensure consistent development and production, large-scale manufacturing of engineered tissues and organs will require standardized facilities, equipment, qualification processes, automation, and information systems. This review presents current and forward-thinking guidelines to help design biofabrication laboratories engaged in engineering model and tissue constructs for therapeutic and non-therapeutic applications.
Collapse
Affiliation(s)
- Henry W Sanicola
- Faculty of Medicine, The University of Queensland, Brisbane 4006, Australia
| | - Caleb E Stewart
- Department of Neurosurgery, Louisiana State Health Sciences Center, Shreveport, LA 71103, USA.
| | | | - Farzad Ahmadi
- Department of Electrical and Computer Engineering, Youngstown State University, Youngstown, OH 44555, USA
| | - Dadong Wang
- Quantitative Imaging Research Team, Data61, Commonwealth Scientific and Industrial Research Organization, Marsfield, NSW 2122, Australia
| | - Sean K Powell
- Science and Engineering Faculty, Queensland University of Technology, Brisbane 4029, Australia
| | - Korak Sarkar
- M3D Laboratory, Ochsner Health System, New Orleans, LA 70121, USA
| | - Kenneth Cutbush
- Faculty of Medicine, The University of Queensland, Brisbane 4006, Australia
| | - Maria A Woodruff
- Science and Engineering Faculty, Queensland University of Technology, Brisbane 4029, Australia.
| | - David A Brafman
- School of Biological and Health Systems Engineering, Arizona State University, Tempe, AZ 85287, USA.
| |
Collapse
|
56
|
Hall MS, Decker JT, Shea LD. Towards systems tissue engineering: Elucidating the dynamics, spatial coordination, and individual cells driving emergent behaviors. Biomaterials 2020; 255:120189. [PMID: 32569865 PMCID: PMC7396312 DOI: 10.1016/j.biomaterials.2020.120189] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Revised: 04/20/2020] [Accepted: 06/09/2020] [Indexed: 12/11/2022]
Abstract
Biomaterial systems have enabled the in vitro production of complex, emergent tissue behaviors that were not possible with conventional two-dimensional culture systems, allowing for analysis of both normal development and disease processes. We propose that the path towards developing the design parameters for biomaterial systems lies with identifying the molecular drivers of emergent behavior through leveraging technological advances in systems biology, including single cell omics, genetic engineering, and high content imaging. This growing research opportunity at the intersection of the fields of tissue engineering and systems biology - systems tissue engineering - can uniquely interrogate the mechanisms by which complex tissue behaviors emerge with the potential to capture the contribution of i) dynamic regulation of tissue development and dysregulation, ii) single cell heterogeneity and the function of rare cell types, and iii) the spatial distribution and structure of individual cells and cell types within a tissue. By leveraging advances in both biological and materials data science, systems tissue engineering can facilitate the identification of biomaterial design parameters that will accelerate basic science discovery and translation.
Collapse
Affiliation(s)
- Matthew S Hall
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Joseph T Decker
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Lonnie D Shea
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
57
|
Finkbeiner S. Functional genomics, genetic risk profiling and cell phenotypes in neurodegenerative disease. Neurobiol Dis 2020; 146:105088. [PMID: 32977020 PMCID: PMC7686089 DOI: 10.1016/j.nbd.2020.105088] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 09/11/2020] [Accepted: 09/18/2020] [Indexed: 12/03/2022] Open
Abstract
Human genetics provides unbiased insights into the causes of human disease, which can be used to create a foundation for effective ways to more accurately diagnose patients, stratify patients for more successful clinical trials, discover and develop new therapies, and ultimately help patients choose the safest and most promising therapeutic option based on their risk profile. But the process for translating basic observations from human genetics studies into pathogenic disease mechanisms and treatments is laborious and complex, and this challenge has particularly slowed the development of interventions for neurodegenerative disease. In this review, we discuss the many steps in the process, the important considerations at each stage, and some of the latest tools and technologies that are available to help investigators translate insights from human genetics into diagnostic and therapeutic strategies that will lead to the sort of advances in clinical care that make a difference for patients.
Collapse
Affiliation(s)
- Steven Finkbeiner
- Center for Systems and Therapeutics, USA; Taube/Koret Center for Neurodegenerative Disease Research, Gladstone Institutes, San Francisco, CA 94158, USA; Departments of Neurology and Physiology, University of Califorina, San Francisco, CA 94158, USA.
| |
Collapse
|
58
|
Isozaki A, Harmon J, Zhou Y, Li S, Nakagawa Y, Hayashi M, Mikami H, Lei C, Goda K. AI on a chip. LAB ON A CHIP 2020; 20:3074-3090. [PMID: 32644061 DOI: 10.1039/d0lc00521e] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/09/2023]
Abstract
Artificial intelligence (AI) has dramatically changed the landscape of science, industry, defence, and medicine in the last several years. Supported by considerably enhanced computational power and cloud storage, the field of AI has shifted from mostly theoretical studies in the discipline of computer science to diverse real-life applications such as drug design, material discovery, speech recognition, self-driving cars, advertising, finance, medical imaging, and astronomical observation, where AI-produced outcomes have been proven to be comparable or even superior to the performance of human experts. In these applications, what is essentially important for the development of AI is the data needed for machine learning. Despite its prominent importance, the very first process of the AI development, namely data collection and data preparation, is typically the most laborious task and is often a limiting factor of constructing functional AI algorithms. Lab-on-a-chip technology, in particular microfluidics, is a powerful platform for both the construction and implementation of AI in a large-scale, cost-effective, high-throughput, automated, and multiplexed manner, thereby overcoming the above bottleneck. On this platform, high-throughput imaging is a critical tool as it can generate high-content information (e.g., size, shape, structure, composition, interaction) of objects on a large scale. High-throughput imaging can also be paired with sorting and DNA/RNA sequencing to conduct a massive survey of phenotype-genotype relations whose data is too complex to analyze with traditional computational tools, but is analyzable with the power of AI. In addition to its function as a data provider, lab-on-a-chip technology can also be employed to implement the developed AI for accurate identification, characterization, classification, and prediction of objects in mixed, heterogeneous, or unknown samples. In this review article, motivated by the excellent synergy between AI and lab-on-a-chip technology, we outline fundamental elements, recent advances, future challenges, and emerging opportunities of AI with lab-on-a-chip technology or "AI on a chip" for short.
Collapse
Affiliation(s)
- Akihiro Isozaki
- Department of Chemistry, University of Tokyo, Tokyo 113-0033, Japan. and Kanagawa Institute of Industrial Science and Technology, Kanagawa 213-0012, Japan
| | - Jeffrey Harmon
- Department of Chemistry, University of Tokyo, Tokyo 113-0033, Japan.
| | - Yuqi Zhou
- Department of Chemistry, University of Tokyo, Tokyo 113-0033, Japan.
| | - Shuai Li
- Department of Chemistry, University of Tokyo, Tokyo 113-0033, Japan. and The Cambridge Centre for Data-Driven Discovery, Cambridge University, Cambridge CB3 0WA, UK
| | - Yuta Nakagawa
- Department of Chemistry, University of Tokyo, Tokyo 113-0033, Japan.
| | - Mika Hayashi
- Department of Chemistry, University of Tokyo, Tokyo 113-0033, Japan.
| | - Hideharu Mikami
- Department of Chemistry, University of Tokyo, Tokyo 113-0033, Japan.
| | - Cheng Lei
- Department of Chemistry, University of Tokyo, Tokyo 113-0033, Japan. and Institute of Technological Sciences, Wuhan University, Hubei 430072, China
| | - Keisuke Goda
- Department of Chemistry, University of Tokyo, Tokyo 113-0033, Japan. and Institute of Technological Sciences, Wuhan University, Hubei 430072, China and Department of Bioengineering, University of California, Los Angeles, California 90095, USA
| |
Collapse
|
59
|
Mudali D, Jeevanandam J, Danquah MK. Probing the characteristics and biofunctional effects of disease-affected cells and drug response via machine learning applications. Crit Rev Biotechnol 2020; 40:951-977. [PMID: 32633615 DOI: 10.1080/07388551.2020.1789062] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Drug-induced transformations in disease characteristics at the cellular and molecular level offers the opportunity to predict and evaluate the efficacy of pharmaceutical ingredients whilst enabling the optimal design of new and improved drugs with enhanced pharmacokinetics and pharmacodynamics. Machine learning is a promising in-silico tool used to simulate cells with specific disease properties and to determine their response toward drug uptake. Differences in the properties of normal and infected cells, including biophysical, biochemical and physiological characteristics, plays a key role in developing fundamental cellular probing platforms for machine learning applications. Cellular features can be extracted periodically from both the drug treated, infected, and normal cells via image segmentations in order to probe dynamic differences in cell behavior. Cellular segmentation can be evaluated to reflect the levels of drug effect on a distinct cell or group of cells via probability scoring. This article provides an account for the use of machine learning methods to probe differences in the biophysical, biochemical and physiological characteristics of infected cells in response to pharmacokinetics uptake of drug ingredients for application in cancer, diabetes and neurodegenerative disease therapies.
Collapse
Affiliation(s)
- Deborah Mudali
- Department of Computer Science, University of Tennessee, Chattanooga, TN, USA
| | - Jaison Jeevanandam
- Department of Chemical Engineering, Faculty of Engineering and Science, Curtin University, Miri, Malaysia
| | - Michael K Danquah
- Chemical Engineering Department, University of Tennessee, Chattanooga, TN, USA
| |
Collapse
|
60
|
Lin S, Schorpp K, Rothenaigner I, Hadian K. Image-based high-content screening in drug discovery. Drug Discov Today 2020; 25:1348-1361. [PMID: 32561299 DOI: 10.1016/j.drudis.2020.06.001] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Revised: 05/05/2020] [Accepted: 06/08/2020] [Indexed: 12/16/2022]
Abstract
While target-based drug discovery strategies rely on the precise knowledge of the identity and function of the drug targets, phenotypic drug discovery (PDD) approaches allow the identification of novel drugs based on knowledge of a distinct phenotype. Image-based high-content screening (HCS) is a potent PDD strategy that characterizes small-molecule effects through the quantification of features that depict cellular changes among or within cell populations, thereby generating valuable data sets for subsequent data analysis. However, these data can be complex, making image analysis from large HCS campaigns challenging. Technological advances in image acquisition, processing, and analysis as well as machine-learning (ML) approaches for the analysis of multidimensional data sets have rendered HCS as a viable technology for small-molecule drug discovery. Here, we discuss HCS concepts, current workflows as well as opportunities and challenges of image-based phenotypic screening and data analysis.
Collapse
Affiliation(s)
- Sean Lin
- Assay Development and Screening Platform, Institute of Molecular Toxicology and Pharmacology, Helmholtz Zentrum München, Ingolstaedter Landstrasse 1, 85764 Neuherberg, Germany
| | - Kenji Schorpp
- Assay Development and Screening Platform, Institute of Molecular Toxicology and Pharmacology, Helmholtz Zentrum München, Ingolstaedter Landstrasse 1, 85764 Neuherberg, Germany
| | - Ina Rothenaigner
- Assay Development and Screening Platform, Institute of Molecular Toxicology and Pharmacology, Helmholtz Zentrum München, Ingolstaedter Landstrasse 1, 85764 Neuherberg, Germany
| | - Kamyar Hadian
- Assay Development and Screening Platform, Institute of Molecular Toxicology and Pharmacology, Helmholtz Zentrum München, Ingolstaedter Landstrasse 1, 85764 Neuherberg, Germany.
| |
Collapse
|
61
|
Yang KD, Damodaran K, Venkatachalapathy S, Soylemezoglu AC, Shivashankar GV, Uhler C. Predicting cell lineages using autoencoders and optimal transport. PLoS Comput Biol 2020; 16:e1007828. [PMID: 32343706 PMCID: PMC7209334 DOI: 10.1371/journal.pcbi.1007828] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Revised: 05/08/2020] [Accepted: 03/27/2020] [Indexed: 12/17/2022] Open
Abstract
Lineage tracing involves the identification of all ancestors and descendants of a given cell, and is an important tool for studying biological processes such as development and disease progression. However, in many settings, controlled time-course experiments are not feasible, for example when working with tissue samples from patients. Here we present ImageAEOT, a computational pipeline based on autoencoders and optimal transport for predicting the lineages of cells using time-labeled datasets from different stages of a cellular process. Given a single-cell image from one of the stages, ImageAEOT generates an artificial lineage of this cell based on the population characteristics of the other stages. These lineages can be used to connect subpopulations of cells through the different stages and identify image-based features and biomarkers underlying the biological process. To validate our method, we apply ImageAEOT to a benchmark task based on nuclear and chromatin images during the activation of fibroblasts by tumor cells in engineered 3D tissues. We further validate ImageAEOT on chromatin images of various breast cancer cell lines and human tissue samples, thereby linking alterations in chromatin condensation patterns to different stages of tumor progression. Our results demonstrate the promise of computational methods based on autoencoding and optimal transport principles for lineage tracing in settings where existing experimental strategies cannot be used. Many key biological processes, such as development and disease progression, require analyzing lineages of cells backwards as well as forwards in time. However, current single-cell experiments tend to be destructive to cells, so that a single lineage can only be measured at one point in time. In this work, we introduce a computational framework for predicting the lineage of cells from a single snapshot in time based on measurements of other cells at other time points. The method generates these lineages by computing the most plausible path for a population of cells to transition from one time point to the next, assuming that a cell is more likely to transition to similar cells compared to dissimilar cells. We validate our computational method on imaging data of fibroblasts and cancer cells, though our method could also be applied to other modalities of single-cell data such as genomics and transcriptomics as well as multi-modal single-cell datasets.
Collapse
Affiliation(s)
- Karren Dai Yang
- Institute for Data, Systems and Society, Massachusetts Institute of Technology, Cambridge, Massachusetts, United States of America
- Department of Electrical Engineering & Computer Science, Massachusetts Institute of Technology, Cambridge, Massachusetts, United States of America
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts, United States of America
| | - Karthik Damodaran
- Mechanobiology Institute, National University of Singapore, Singapore
| | | | - Ali C. Soylemezoglu
- Institute for Data, Systems and Society, Massachusetts Institute of Technology, Cambridge, Massachusetts, United States of America
| | - G. V. Shivashankar
- Mechanobiology Institute, National University of Singapore, Singapore
- FIRC Institute of Molecular Oncology (IFOM), Milan, Italy
- Department of Health Sciences and Technology, ETH Zurich and Paul Scherrer Institute, Villigen, Switzerland
| | - Caroline Uhler
- Institute for Data, Systems and Society, Massachusetts Institute of Technology, Cambridge, Massachusetts, United States of America
- Department of Electrical Engineering & Computer Science, Massachusetts Institute of Technology, Cambridge, Massachusetts, United States of America
- Department of Biosystems Science and Engineering, ETH Zurich, Zurich, Switzerland
- * E-mail:
| |
Collapse
|
62
|
Multiple particle tracking analysis in isolated nuclei reveals the mechanical phenotype of leukemia cells. Sci Rep 2020; 10:6707. [PMID: 32317728 PMCID: PMC7174401 DOI: 10.1038/s41598-020-63682-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Accepted: 04/02/2020] [Indexed: 12/14/2022] Open
Abstract
The nucleus is fundamentally composed by lamina and nuclear membranes that enclose the chromatin, nucleoskeletal components and suspending nucleoplasm. The functional connections of this network integrate external stimuli into cell signals, including physical forces to mechanical responses of the nucleus. Canonically, the morphological characteristics of the nucleus, as shape and size, have served for pathologists to stratify and diagnose cancer patients; however, novel biophysical techniques must exploit physical parameters to improve cancer diagnosis. By using multiple particle tracking (MPT) technique on chromatin granules, we designed a SURF (Speeded Up Robust Features)-based algorithm to study the mechanical properties of isolated nuclei and in living cells. We have determined the apparent shear stiffness, viscosity and optical density of the nucleus, and how the chromatin structure influences on these biophysical values. Moreover, we used our MPT-SURF analysis to study the apparent mechanical properties of isolated nuclei from patients of acute lymphoblastic leukemia. We found that leukemia cells exhibited mechanical differences compared to normal lymphocytes. Interestingly, isolated nuclei from high-risk leukemia cells showed increased viscosity than their counterparts from normal lymphocytes, whilst nuclei from relapsed-patient's cells presented higher density than those from normal lymphocytes or standard- and high-risk leukemia cells. Taken together, here we presented how MPT-SURF analysis of nuclear chromatin granules defines nuclear mechanical phenotypic features, which might be clinically relevant.
Collapse
|
63
|
Boyd J, Fennell M, Carpenter A. Harnessing the power of microscopy images to accelerate drug discovery: what are the possibilities? Expert Opin Drug Discov 2020; 15:639-642. [PMID: 32200648 DOI: 10.1080/17460441.2020.1743675] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Affiliation(s)
- Justin Boyd
- Internal Medicines Research Unit, Pfizer Inc ., Cambridge, MA, USA
| | - Myles Fennell
- Neuroscience and Platform Biology, Arvinas , New Haven, CT, USA
| | - Anne Carpenter
- Imaging Platform, Broad Institute of MIT and Harvard , Cambridge, MA, USA
| |
Collapse
|
64
|
Hosseini S, Simianer H, Tetens J, Brenig B, Herzog S, Sharifi AR. Efficient phenotypic sex classification of zebrafish using machine learning methods. Ecol Evol 2019; 9:13332-13343. [PMID: 31871648 PMCID: PMC6912926 DOI: 10.1002/ece3.5788] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2019] [Revised: 09/09/2019] [Accepted: 09/17/2019] [Indexed: 12/14/2022] Open
Abstract
Sex determination in zebrafish by manual approaches according to current guidelines relies on human observation. These guidelines for sex recognition have proven to be subjective and highly labor-intensive. To address this problem, we present a methodology to automatically classify the phenotypic sex using two machine learning methods: Deep Convolutional Neural Networks (DCNNs) based on the whole fish appearance and Support Vector Machine (SVM) based on caudal fin coloration. Machine learning techniques in sex classification provide potential efficiency with the advantage of automatization and robustness in the prediction process. Furthermore, since developmental plasticity can be influenced by environmental conditions, we have investigated the impact of elevated water temperature during embryogenesis on sex and sex-related differences in color intensity of adult zebrafish. The estimated color intensity based on SVM was then applied to detect the association between coloration and body weight and length. Phenotypic sex classifications using machine learning methods resulted in a high degree of association with the real sex in nontreated animals. In temperature-induced animals, DCNNs reached a performance of 100%, whereas 20% of males were misclassified using SVM due to a lower color intensity. Furthermore, a positive association between color intensity and body weight and length was observed in males. Our study demonstrates that high ambient temperature leads to a lower color intensity in male animals and a positive association of male caudal fin coloration with body weight and length, which appears to play a significant role in sexual attraction. The software developed for sex classification in this study is readily applicable to other species with sex-linked visible phenotypic differences.
Collapse
Affiliation(s)
- Shahrbanou Hosseini
- Department of Animal SciencesUniversity of GoettingenGoettingenGermany
- Center for Integrated Breeding ResearchUniversity of GoettingenGoettingenGermany
| | - Henner Simianer
- Department of Animal SciencesUniversity of GoettingenGoettingenGermany
- Center for Integrated Breeding ResearchUniversity of GoettingenGoettingenGermany
| | - Jens Tetens
- Department of Animal SciencesUniversity of GoettingenGoettingenGermany
- Center for Integrated Breeding ResearchUniversity of GoettingenGoettingenGermany
| | - Bertram Brenig
- Department of Animal SciencesUniversity of GoettingenGoettingenGermany
- Center for Integrated Breeding ResearchUniversity of GoettingenGoettingenGermany
- Institute of Veterinary MedicineUniversity of GoettingenGoettingenGermany
| | - Sebastian Herzog
- Max Planck Institute for Dynamics and Self‐OrganizationGoettingenGermany
- Department for Computational Neuroscience3rd Physics Institute‐BiophysicsUniversity of GoettingenGoettingenGermany
| | - Ahmad Reza Sharifi
- Department of Animal SciencesUniversity of GoettingenGoettingenGermany
- Center for Integrated Breeding ResearchUniversity of GoettingenGoettingenGermany
| |
Collapse
|
65
|
Big data aggregation in the case of heterogeneity: a feasibility study for digital health. INT J MACH LEARN CYB 2019. [DOI: 10.1007/s13042-018-00904-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
|
66
|
Qualifying antibodies for image-based immune profiling and multiplexed tissue imaging. Nat Protoc 2019; 14:2900-2930. [PMID: 31534232 DOI: 10.1038/s41596-019-0206-y] [Citation(s) in RCA: 84] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2018] [Accepted: 06/03/2019] [Indexed: 12/27/2022]
Abstract
Multiplexed tissue imaging enables precise, spatially resolved enumeration and characterization of cell types and states in human resection specimens. A growing number of methods applicable to formalin-fixed, paraffin-embedded (FFPE) tissue sections have been described, the majority of which rely on antibodies for antigen detection and mapping. This protocol provides step-by-step procedures for confirming the selectivity and specificity of antibodies used in fluorescence-based tissue imaging and for the construction and validation of antibody panels. Although the protocol is implemented using tissue-based cyclic immunofluorescence (t-CyCIF) as an imaging platform, these antibody-testing methods are broadly applicable. We demonstrate assembly of a 16-antibody panel for enumerating and localizing T cells and B cells, macrophages, and cells expressing immune checkpoint regulators. The protocol is accessible to individuals with experience in microscopy and immunofluorescence; some experience in computation is required for data analysis. A typical 30-antibody dataset for 20 FFPE slides can be generated within 2 weeks.
Collapse
|
67
|
Zhang Z, Chen L, Humphries B, Brien R, Wicha MS, Luker KE, Luker GD, Chen YC, Yoon E. Morphology-based prediction of cancer cell migration using an artificial neural network and a random decision forest. Integr Biol (Camb) 2019; 10:758-767. [PMID: 30420987 DOI: 10.1039/c8ib00106e] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Metastasis is the cause of death in most patients of breast cancer and other solid malignancies. Identification of cancer cells with highly migratory capability to metastasize relies on markers for epithelial-to-mesenchymal transition (EMT), a process increasing cell migration and metastasis. Marker-based approaches are limited by inconsistences among patients, types of cancer, and partial EMT states. Alternatively, we analyzed cancer cell migration behavior using computer vision. Using a microfluidic single-cell migration chip and high-content imaging, we extracted morphological features and recorded migratory direction and speed of breast cancer cells. By applying a Random Decision Forest (RDF) and an Artificial Neural Network (ANN), we achieved over 99% accuracy for cell movement direction prediction and 91% for speed prediction. Unprecedentedly, we identified highly motile cells and non-motile cells based on microscope images and a machine learning model, and pinpointed and validated morphological features determining cell migration, including not only known features related to cell polarization but also novel ones that can drive future mechanistic studies. Predicting cell movement by computer vision and machine learning establishes a ground-breaking approach to analyze cell migration and metastasis.
Collapse
Affiliation(s)
- Zhixiong Zhang
- Department of Electrical Engineering and Computer Science, University of Michigan, 1301 Beal Avenue, Ann Arbor, MI 48109-2122, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
68
|
Hériché JK, Alexander S, Ellenberg J. Integrating Imaging and Omics: Computational Methods and Challenges. Annu Rev Biomed Data Sci 2019. [DOI: 10.1146/annurev-biodatasci-080917-013328] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Fluorescence microscopy imaging has long been complementary to DNA sequencing- and mass spectrometry–based omics in biomedical research, but these approaches are now converging. On the one hand, omics methods are moving from in vitro methods that average across large cell populations to in situ molecular characterization tools with single-cell sensitivity. On the other hand, fluorescence microscopy imaging has moved from a morphological description of tissues and cells to quantitative molecular profiling with single-molecule resolution. Recent technological developments underpinned by computational methods have started to blur the lines between imaging and omics and have made their direct correlation and seamless integration an exciting possibility. As this trend continues rapidly, it will allow us to create comprehensive molecular profiles of living systems with spatial and temporal context and subcellular resolution. Key to achieving this ambitious goal will be novel computational methods and successfully dealing with the challenges of data integration and sharing as well as cloud-enabled big data analysis.
Collapse
Affiliation(s)
- Jean-Karim Hériché
- Cell Biology and Biophysics Unit, European Molecular Biology Laboratory (EMBL), 69117 Heidelberg, Germany
| | - Stephanie Alexander
- Cell Biology and Biophysics Unit, European Molecular Biology Laboratory (EMBL), 69117 Heidelberg, Germany
| | - Jan Ellenberg
- Cell Biology and Biophysics Unit, European Molecular Biology Laboratory (EMBL), 69117 Heidelberg, Germany
| |
Collapse
|
69
|
Culver DA, Behr J, Belperio JA, Corte TJ, de Andrade JA, Flaherty KR, Gulati M, Huie TJ, Lancaster LH, Roman J, Ryerson CJ, Kim HJ. Patient Registries in Idiopathic Pulmonary Fibrosis. Am J Respir Crit Care Med 2019; 200:160-167. [PMID: 31034241 PMCID: PMC6635784 DOI: 10.1164/rccm.201902-0431ci] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Accepted: 04/24/2019] [Indexed: 01/06/2023] Open
Abstract
Over the past decade, several large registries of patients with idiopathic pulmonary fibrosis (IPF) have been established. These registries are collecting a wealth of longitudinal data on thousands of patients with this rare disease. The data collected in these registries will be complementary to data collected in clinical trials because the patient populations studied in registries have a broader spectrum of disease severity and comorbidities and can be followed for a longer period of time. Maintaining the quality and completeness of registry databases presents administrative and resourcing challenges, but it is important to ensuring the robustness of the analyses. Data from patient registries have already helped improve understanding of the clinical characteristics of patients with IPF, the impact that the disease has on their quality of life and survival, and current practices in diagnosis and management. In the future, analyses of biospecimens linked to detailed patient profiles will provide the opportunity to identify biomarkers linked to disease progression, facilitating the development of precision medicine approaches for prognosis and therapy in patients with IPF.
Collapse
Affiliation(s)
| | - Jürgen Behr
- Department of Internal Medicine V, Ludwig-Maximilians University of Munich, Munich, Germany
- Asklepios Clinic Gauting, Member of the German Center for Lung Research, Gauting, Germany
| | - John A. Belperio
- David Geffen School of Medicine at UCLA, University of California, Los Angeles, Los Angeles, California
| | - Tamera J. Corte
- Royal Prince Alfred Hospital, Camperdown, New South Wales, Australia
- University of Sydney, Sydney, New South Wales, Australia
| | | | - Kevin R. Flaherty
- Division of Pulmonary and Critical Care Medicine, University of Michigan, Ann Arbor, Michigan
| | - Mridu Gulati
- Yale University School of Medicine, New Haven, Connecticut
| | | | | | - Jesse Roman
- Jane and Leonard Korman Respiratory Institute, Thomas Jefferson University, Philadelphia, Pennsylvania
| | | | - Hyun J. Kim
- University of Minnesota, Minneapolis, Minnesota
| |
Collapse
|
70
|
Isozaki A, Mikami H, Hiramatsu K, Sakuma S, Kasai Y, Iino T, Yamano T, Yasumoto A, Oguchi Y, Suzuki N, Shirasaki Y, Endo T, Ito T, Hiraki K, Yamada M, Matsusaka S, Hayakawa T, Fukuzawa H, Yatomi Y, Arai F, Di Carlo D, Nakagawa A, Hoshino Y, Hosokawa Y, Uemura S, Sugimura T, Ozeki Y, Nitta N, Goda K. A practical guide to intelligent image-activated cell sorting. Nat Protoc 2019; 14:2370-2415. [PMID: 31278398 DOI: 10.1038/s41596-019-0183-1] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Accepted: 04/18/2019] [Indexed: 02/08/2023]
Abstract
Intelligent image-activated cell sorting (iIACS) is a machine-intelligence technology that performs real-time intelligent image-based sorting of single cells with high throughput. iIACS extends beyond the capabilities of fluorescence-activated cell sorting (FACS) from fluorescence intensity profiles of cells to multidimensional images, thereby enabling high-content sorting of cells or cell clusters with unique spatial chemical and morphological traits. Therefore, iIACS serves as an integral part of holistic single-cell analysis by enabling direct links between population-level analysis (flow cytometry), cell-level analysis (microscopy), and gene-level analysis (sequencing). Specifically, iIACS is based on a seamless integration of high-throughput cell microscopy (e.g., multicolor fluorescence imaging, bright-field imaging), cell focusing, cell sorting, and deep learning on a hybrid software-hardware data management infrastructure, enabling real-time automated operation for data acquisition, data processing, intelligent decision making, and actuation. Here, we provide a practical guide to iIACS that describes how to design, build, characterize, and use an iIACS machine. The guide includes the consideration of several important design parameters, such as throughput, sensitivity, dynamic range, image quality, sort purity, and sort yield; the development and integration of optical, microfluidic, electrical, computational, and mechanical components; and the characterization and practical usage of the integrated system. Assuming that all components are readily available, a team of several researchers experienced in optics, electronics, digital signal processing, microfluidics, mechatronics, and flow cytometry can complete this protocol in ~3 months.
Collapse
Affiliation(s)
- Akihiro Isozaki
- Department of Chemistry, The University of Tokyo, Tokyo, Japan
| | - Hideharu Mikami
- Department of Chemistry, The University of Tokyo, Tokyo, Japan
| | | | - Shinya Sakuma
- Department of Micro-Nano Mechanical Science and Engineering, Nagoya University, Nagoya, Japan
| | - Yusuke Kasai
- Department of Micro-Nano Mechanical Science and Engineering, Nagoya University, Nagoya, Japan
| | - Takanori Iino
- Department of Electrical Engineering and Information Systems, The University of Tokyo, Tokyo, Japan
| | - Takashi Yamano
- Laboratory of Applied Molecular Microbiology, Kyoto University, Kyoto, Japan
| | - Atsushi Yasumoto
- Department of Clinical Laboratory Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Yusuke Oguchi
- Department of Biological Sciences, The University of Tokyo, Tokyo, Japan
| | - Nobutake Suzuki
- Department of Biological Sciences, The University of Tokyo, Tokyo, Japan
| | | | | | - Takuro Ito
- Department of Chemistry, The University of Tokyo, Tokyo, Japan.,Japan Science and Technology Agency, Saitama, Japan
| | - Kei Hiraki
- Department of Chemistry, The University of Tokyo, Tokyo, Japan
| | - Makoto Yamada
- Department of Intelligence Science and Technology, Graduate School of Informatics, Kyoto University, Kyoto, Japan
| | - Satoshi Matsusaka
- Clinical Research and Regional Innovation, Faculty of Medicine, University of Tsukuba, Ibaraki, Japan
| | - Takeshi Hayakawa
- Department of Precision Mechanics, Chuo University, Tokyo, Japan
| | - Hideya Fukuzawa
- Laboratory of Applied Molecular Microbiology, Kyoto University, Kyoto, Japan
| | - Yutaka Yatomi
- Department of Clinical Laboratory Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Fumihito Arai
- Department of Micro-Nano Mechanical Science and Engineering, Nagoya University, Nagoya, Japan
| | - Dino Di Carlo
- Department of Chemistry, The University of Tokyo, Tokyo, Japan.,Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA, USA.,Department of Mechanical Engineering, University of California, Los Angeles, Los Angeles, CA, USA.,California NanoSystems Institute, University of California, Los Angeles, Los Angeles, CA, USA
| | - Atsuhiro Nakagawa
- Department of Neurosurgery, Graduate School of Medicine, Tohoku University, Sendai, Japan
| | - Yu Hoshino
- Department of Chemical Engineering, Kyushu University, Fukuoka, Japan
| | - Yoichiroh Hosokawa
- Division of Materials Science, Graduate School of Science and Technology, Nara Institute of Science and Technology, Ikoma, Japan
| | - Sotaro Uemura
- Department of Biological Sciences, The University of Tokyo, Tokyo, Japan
| | - Takeaki Sugimura
- Department of Chemistry, The University of Tokyo, Tokyo, Japan.,Japan Science and Technology Agency, Saitama, Japan
| | - Yasuyuki Ozeki
- Department of Electrical Engineering and Information Systems, The University of Tokyo, Tokyo, Japan
| | - Nao Nitta
- Department of Chemistry, The University of Tokyo, Tokyo, Japan.,Japan Science and Technology Agency, Saitama, Japan
| | - Keisuke Goda
- Department of Chemistry, The University of Tokyo, Tokyo, Japan. .,Japan Science and Technology Agency, Saitama, Japan. .,Department of Electrical Engineering, University of California, Los Angeles, Los Angeles, CA, USA.
| |
Collapse
|
71
|
Robles-Bykbaev Y, Naya S, Díaz-Prado S, Calle-López D, Robles-Bykbaev V, Garzón L, Sanjurjo-Rodríguez C, Tarrío-Saavedra J. An artificial-vision- and statistical-learning-based method for studying the biodegradation of type I collagen scaffolds in bone regeneration systems. PeerJ 2019; 7:e7233. [PMID: 31316873 PMCID: PMC6613533 DOI: 10.7717/peerj.7233] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2018] [Accepted: 05/31/2019] [Indexed: 12/13/2022] Open
Abstract
This work proposes a method based on image analysis and machine and statistical learning to model and estimate osteocyte growth (in type I collagen scaffolds for bone regeneration systems) and the collagen degradation degree due to cellular growth. To achieve these aims, the mass of collagen -subjected to the action of osteocyte growth and differentiation from stem cells- was measured on 3 days during each of 2 months, under conditions simulating a tissue in the human body. In addition, optical microscopy was applied to obtain information about cellular growth, cellular differentiation, and collagen degradation. Our first contribution consists of the application of a supervised classification random forest algorithm to image texture features (the structure tensor and entropy) for estimating the different regions of interest in an image obtained by optical microscopy: the extracellular matrix, collagen, and image background, and nuclei. Then, extracellular-matrix and collagen regions of interest were determined by the extraction of features related to the progression of the cellular growth and collagen degradation (e.g., mean area of objects and the mode of an intensity histogram). Finally, these critical features were statistically modeled depending on time via nonparametric and parametric linear and nonlinear models such as those based on logistic functions. Namely, the parametric logistic mixture models provided a way to identify and model the degradation due to biological activity by estimating the corresponding proportion of mass loss. The relation between osteocyte growth and differentiation from stem cells, on the one hand, and collagen degradation, on the other hand, was determined too and modeled through analysis of image objects' circularity and area, in addition to collagen mass loss. This set of imaging techniques, machine learning procedures, and statistical tools allowed us to characterize and parameterize type I collagen biodegradation when collagen acts as a scaffold in bone regeneration tasks. Namely, the parametric logistic mixture models provided a way to identify and model the degradation due to biological activity and thus to estimate the corresponding proportion of mass loss. Moreover, the proposed methodology can help to estimate the degradation degree of scaffolds from the information obtained by optical microscopy.
Collapse
Affiliation(s)
- Yaroslava Robles-Bykbaev
- Instituto de Investigación Biomédica de A Coruña (INIBIC), Complexo Hospitalario Universitario de A Coruña (CHUAC), SERGAS, Departamento de Medicina, Universidade da Coruña, A Coruña, Spain.,Cátedra UNESCO UPS Tecnologías de apoyo para la Inclusión Educativa, Universidad Politécnica Salesiana, Cuenca, Ecuador.,Grupo de Investigación en Materiales (GiMaT), Universidad Politécnica Salesiana, Cuenca, Ecuador
| | - Salvador Naya
- Grupo MODES, CITIC, ITMATI, Departamento de Matemáticas, Universidade da Coruña, Ferrol, Spain
| | - Silvia Díaz-Prado
- Instituto de Investigación Biomédica de A Coruña (INIBIC), Complexo Hospitalario Universitario de A Coruña (CHUAC), SERGAS, Departamento de Medicina, Universidade da Coruña, A Coruña, Spain
| | - Daniel Calle-López
- Cátedra UNESCO UPS Tecnologías de apoyo para la Inclusión Educativa, Universidad Politécnica Salesiana, Cuenca, Ecuador
| | - Vladimir Robles-Bykbaev
- Cátedra UNESCO UPS Tecnologías de apoyo para la Inclusión Educativa, Universidad Politécnica Salesiana, Cuenca, Ecuador
| | - Luis Garzón
- Grupo de Investigación en Materiales (GiMaT), Universidad Politécnica Salesiana, Cuenca, Ecuador
| | - Clara Sanjurjo-Rodríguez
- Instituto de Investigación Biomédica de A Coruña (INIBIC), Complexo Hospitalario Universitario de A Coruña (CHUAC), SERGAS, Departamento de Medicina, Universidade da Coruña, A Coruña, Spain
| | - Javier Tarrío-Saavedra
- Grupo MODES, CITIC, ITMATI, Departamento de Matemáticas, Universidade da Coruña, Ferrol, Spain
| |
Collapse
|
72
|
Zhang Y, Xie Y, Liu W, Deng W, Peng D, Wang C, Xu H, Ruan C, Deng Y, Guo Y, Lu C, Yi C, Ren J, Xue Y. DeepPhagy: a deep learning framework for quantitatively measuring autophagy activity in Saccharomyces cerevisiae. Autophagy 2019; 16:626-640. [PMID: 31204567 DOI: 10.1080/15548627.2019.1632622] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Seeing is believing. The direct observation of GFP-Atg8 vacuolar delivery under confocal microscopy is one of the most useful end-point measurements for monitoring yeast macroautophagy/autophagy. However, manually labelling individual cells from large-scale sets of images is time-consuming and labor-intensive, which has greatly hampered its extensive use in functional screens. Herein, we conducted a time-course analysis of nitrogen starvation-induced autophagy in wild-type and knockout mutants of 35 AuTophaGy-related (ATG) genes in Saccharomyces cerevisiae and obtained 1,944 confocal images containing > 200,000 cells. We manually labelled 8,078 autophagic and 18,493 non-autophagic cells as a benchmark dataset and developed a new deep learning tool for autophagy (DeepPhagy), which exhibited superior accuracy in recognizing autophagic cells compared to other existing methods, with an area under the curve (AUC) value of 0.9710 from 10-fold cross-validations. We further used DeepPhagy to automatically analyze all the images and quantitatively classified the autophagic phenotypes of the 35 atg knockout mutants into 3 classes. The high consistency in our computational and biochemical results indicated the reliability of DeepPhagy for measuring autophagic activity. Moreover, we used DeepPhagy to analyze 3 additional types of autophagic phenotypes, including the targeting of Atg1-GFP to the vacuole, the vacuolar delivery of GFP-Atg19, and the disintegration of autophagic bodies indicated by GFP-Atg8, all with satisfying accuracies. Taken together, our study not only enables the GFP-Atg8 fluorescence assay to become a quantitative measurement for analyzing autophagic phenotypes in S. cerevisiae but also demonstrates that deep learning-based methods could potentially be applied to different types of autophagy.Abbreviations: Ac: accuracy; ALP: alkaline phosphatase; ALR: autophagic lysosomal reformation; ATG: AuTophaGy-related; AUC: area under the curve; CNN: convolutional neural network; Cvt: cytoplasm-to-vacuole targeting; DeepPhagy: deep learning for autophagy; fc_2: second fully connected; GFP: green fluorescent protein; MAP1LC3/LC3: microtubule-associated protein 1 light chain 3 beta; HAT: histone acetyltransferase; HemI: Heat map Illustrator; JRE: Java Runtime Environment; KO: knockout; LRN: local response normalization; MCC: Mathew Correlation Coefficient; OS: operating system; PAS: phagophore assembly site; PC: principal component; PCA: principal component analysis; PPI: protein-protein interaction; Pr: precision; QPSO: Quantum-behaved Particle Swarm Optimization; ReLU: rectified linear unit; RF: random forest; ROC: receiver operating characteristic; ROI: region of interest; SD: systematic derivation; SGD: stochastic gradient descent; Sn: sensitivity; Sp: specificity; SRG: seeded region growing; t-SNE: t-distributed stochastic neighbor embedding; 2D: 2-dimensional; WT: wild-type.
Collapse
Affiliation(s)
- Ying Zhang
- Department of Bioinformatics and Systems Biology, Key Laboratory of Molecular Biophysics of the Ministry of Education, Hubei Bioinformatics and Molecular Imaging Key Laboratory, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China
| | - Yubin Xie
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Wenzhong Liu
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Wankun Deng
- Department of Bioinformatics and Systems Biology, Key Laboratory of Molecular Biophysics of the Ministry of Education, Hubei Bioinformatics and Molecular Imaging Key Laboratory, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China
| | - Di Peng
- Department of Bioinformatics and Systems Biology, Key Laboratory of Molecular Biophysics of the Ministry of Education, Hubei Bioinformatics and Molecular Imaging Key Laboratory, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China
| | - Chenwei Wang
- Department of Bioinformatics and Systems Biology, Key Laboratory of Molecular Biophysics of the Ministry of Education, Hubei Bioinformatics and Molecular Imaging Key Laboratory, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China
| | - Haodong Xu
- Department of Bioinformatics and Systems Biology, Key Laboratory of Molecular Biophysics of the Ministry of Education, Hubei Bioinformatics and Molecular Imaging Key Laboratory, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China
| | - Chen Ruan
- Department of Bioinformatics and Systems Biology, Key Laboratory of Molecular Biophysics of the Ministry of Education, Hubei Bioinformatics and Molecular Imaging Key Laboratory, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China
| | - Yongjie Deng
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Yaping Guo
- Department of Bioinformatics and Systems Biology, Key Laboratory of Molecular Biophysics of the Ministry of Education, Hubei Bioinformatics and Molecular Imaging Key Laboratory, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China
| | - Chenjun Lu
- Department of Biochemistry and Molecular Biology, Program in Molecular and Cell Biology, Zhejiang University School of Medicine, Hangzhou, China
| | - Cong Yi
- Department of Biochemistry and Molecular Biology, Program in Molecular and Cell Biology, Zhejiang University School of Medicine, Hangzhou, China
| | - Jian Ren
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Yu Xue
- Department of Bioinformatics and Systems Biology, Key Laboratory of Molecular Biophysics of the Ministry of Education, Hubei Bioinformatics and Molecular Imaging Key Laboratory, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
73
|
Patel DS, Xu N, Lu H. Digging deeper: methodologies for high-content phenotyping in Caenorhabditis elegans. Lab Anim (NY) 2019; 48:207-216. [PMID: 31217565 DOI: 10.1038/s41684-019-0326-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2018] [Accepted: 05/17/2019] [Indexed: 11/09/2022]
Abstract
Deep phenotyping is an emerging conceptual paradigm and experimental approach aimed at measuring and linking many aspects of a phenotype to understand its underlying biology. To date, deep phenotyping has been applied mostly in cultured cells and used less in multicellular organisms. However, in the past decade, it has increasingly been recognized that deep phenotyping could lead to a better understanding of how genetics, environment and stochasticity affect the development, physiology and behavior of an organism. The nematode Caenorhabditis elegans is an invaluable model system for studying how genes affect a phenotypic trait, and new technologies have taken advantage of the worm's physical attributes to increase the throughput and informational content of experiments. Coupling of these technical advancements with computational and analytical tools has enabled a boom in deep-phenotyping studies of C. elegans. In this Review, we highlight how these new technologies and tools are digging into the biological origins of complex, multidimensional phenotypes.
Collapse
Affiliation(s)
- Dhaval S Patel
- School of Chemical & Biomolecular Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| | - Nan Xu
- School of Chemical & Biomolecular Engineering, Georgia Institute of Technology, Atlanta, GA, USA.,The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Hang Lu
- School of Chemical & Biomolecular Engineering, Georgia Institute of Technology, Atlanta, GA, USA.
| |
Collapse
|
74
|
González G, Evans CL. Biomedical Image Processing with Containers and Deep Learning: An Automated Analysis Pipeline: Data architecture, artificial intelligence, automated processing, containerization, and clusters orchestration ease the transition from data acquisition to insights in medium-to-large datasets. Bioessays 2019; 41:e1900004. [PMID: 31094000 PMCID: PMC6538271 DOI: 10.1002/bies.201900004] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Revised: 03/18/2019] [Indexed: 12/13/2022]
Abstract
Here, a streamlined, scalable, laboratory approach is discussed that enables medium-to-large dataset analysis. The presented approach combines data management, artificial intelligence, containerization, cluster orchestration, and quality control in a unified analytic pipeline. The unique combination of these individual building blocks creates a new and powerful analysis approach that can readily be applied to medium-to-large datasets by researchers to accelerate the pace of research. The proposed framework is applied to a project that counts the number of plasmonic nanoparticles bound to peripheral blood mononuclear cells in dark-field microscopy images. By using the techniques presented in this article, the images are automatically processed overnight, without user interaction, streamlining the path from experiment to conclusions.
Collapse
Affiliation(s)
- Germán González
- PNP Research Corporation, Drury, MA. 01343
- Sierra Research S.L.U. Avda Costa Blanca 132. Alicante. Spain. 03540
| | - Conor L. Evans
- Wellman Center for Photomedicine, Harvard Medical School, Massachusetts General Hospital, CNY149-3, 13th St, Charlestown, MA 02129
- Ludwig Center at Harvard, Harvard Medical School, Boston, MA
| |
Collapse
|
75
|
Lam VK, Nguyen T, Phan T, Chung BM, Nehmetallah G, Raub CB. Machine Learning with Optical Phase Signatures for Phenotypic Profiling of Cell Lines. Cytometry A 2019; 95:757-768. [PMID: 31008570 DOI: 10.1002/cyto.a.23774] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Revised: 03/22/2019] [Accepted: 04/03/2019] [Indexed: 12/29/2022]
Abstract
Robust and reproducible profiling of cell lines is essential for phenotypic screening assays. The goals of this study were to determine robust and reproducible optical phase signatures of cell lines for classification with machine learning and to correlate optical phase parameters to motile behavior. Digital holographic microscopy (DHM) reconstructed phase maps of cells from two pairs of cancer and non-cancer cell lines. Seventeen image parameters were extracted from each cell's phase map, used for linear support vector machine learning, and correlated to scratch wound closure and Boyden chamber chemotaxis. The classification accuracy was between 90% and 100% for the six pairwise cell line comparisons. Several phase parameters correlated with wound closure rate and chemotaxis across the four cell lines. The level of cell confluence in culture affected phase parameters in all cell lines tested. Results indicate that optical phase features of cell lines are a robust set of quantitative data of potential utility for phenotypic screening and prediction of motile behavior. © 2019 International Society for Advancement of Cytometry.
Collapse
Affiliation(s)
- Van K Lam
- Department of Biomedical Engineering, The Catholic University of America, Washington, DC
| | - Thanh Nguyen
- Department of Electrical Engineering and Computer Science, The Catholic University of America, Washington, DC
| | - Thuc Phan
- Department of Electrical Engineering and Computer Science, The Catholic University of America, Washington, DC
| | - Byung-Min Chung
- Department of Biology, The Catholic University of America, Washington, DC
| | - George Nehmetallah
- Department of Electrical Engineering and Computer Science, The Catholic University of America, Washington, DC
| | - Christopher B Raub
- Department of Biomedical Engineering, The Catholic University of America, Washington, DC
| |
Collapse
|
76
|
Zenger KR, Khatkar MS, Jones DB, Khalilisamani N, Jerry DR, Raadsma HW. Genomic Selection in Aquaculture: Application, Limitations and Opportunities With Special Reference to Marine Shrimp and Pearl Oysters. Front Genet 2019; 9:693. [PMID: 30728827 PMCID: PMC6351666 DOI: 10.3389/fgene.2018.00693] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Accepted: 12/11/2018] [Indexed: 11/20/2022] Open
Abstract
Within aquaculture industries, selection based on genomic information (genomic selection) has the profound potential to change genetic improvement programs and production systems. Genomic selection exploits the use of realized genomic relationships among individuals and information from genome-wide markers in close linkage disequilibrium with genes of biological and economic importance. We discuss the technical advances, practical requirements, and commercial applications that have made genomic selection feasible in a range of aquaculture industries, with a particular focus on molluscs (pearl oysters, Pinctada maxima) and marine shrimp (Litopenaeus vannamei and Penaeus monodon). The use of low-cost genome sequencing has enabled cost-effective genotyping on a large scale and is of particular value for species without a reference genome or access to commercial genotyping arrays. We highlight the pitfalls and offer the solutions to the genotyping by sequencing approach and the building of appropriate genetic resources to undertake genomic selection from first-hand experience. We describe the potential to capture large-scale commercial phenotypes based on image analysis and artificial intelligence through machine learning, as inputs for calculation of genomic breeding values. The application of genomic selection over traditional aquatic breeding programs offers significant advantages through being able to accurately predict complex polygenic traits including disease resistance; increasing rates of genetic gain; minimizing inbreeding; and negating potential limiting effects of genotype by environment interactions. Further practical advantages of genomic selection through the use of large-scale communal mating and rearing systems are highlighted, as well as presenting rate-limiting steps that impact on attaining maximum benefits from adopting genomic selection. Genomic selection is now at the tipping point where commercial applications can be readily adopted and offer significant short- and long-term solutions to sustainable and profitable aquaculture industries.
Collapse
Affiliation(s)
- Kyall R Zenger
- Centre for Sustainable Tropical Fisheries and Aquaculture, College of Science and Engineering, James Cook University, Townsville, QLD, Australia.,ARC Research Hub for Advanced Prawn Breeding, James Cook University, Townsville, QLD, Australia
| | - Mehar S Khatkar
- ARC Research Hub for Advanced Prawn Breeding, James Cook University, Townsville, QLD, Australia.,Sydney School of Veterinary Science, Faculty of Science, The University of Sydney, Camden, NSW, Australia
| | - David B Jones
- Centre for Sustainable Tropical Fisheries and Aquaculture, College of Science and Engineering, James Cook University, Townsville, QLD, Australia
| | - Nima Khalilisamani
- ARC Research Hub for Advanced Prawn Breeding, James Cook University, Townsville, QLD, Australia.,Sydney School of Veterinary Science, Faculty of Science, The University of Sydney, Camden, NSW, Australia
| | - Dean R Jerry
- Centre for Sustainable Tropical Fisheries and Aquaculture, College of Science and Engineering, James Cook University, Townsville, QLD, Australia.,ARC Research Hub for Advanced Prawn Breeding, James Cook University, Townsville, QLD, Australia.,Tropical Futures Institute, James Cook University Singapore, Singapore, Singapore
| | - Herman W Raadsma
- ARC Research Hub for Advanced Prawn Breeding, James Cook University, Townsville, QLD, Australia.,Sydney School of Veterinary Science, Faculty of Science, The University of Sydney, Camden, NSW, Australia
| |
Collapse
|
77
|
Panepucci RA, de Souza Lima IM. Arrayed functional genetic screenings in pluripotency reprogramming and differentiation. Stem Cell Res Ther 2019; 10:24. [PMID: 30635073 PMCID: PMC6330485 DOI: 10.1186/s13287-018-1124-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Thoroughly understanding the molecular mechanisms responsible for the biological properties of pluripotent stem cells, as well as for the processes involved in reprograming, differentiation, and transition between Naïve and Primed pluripotent states, is of great interest in basic and applied research. Although pluripotent cells have been extensively characterized in terms of their transcriptome and miRNome, a comprehensive understanding of how these gene products specifically impact their biology, depends on gain- or loss-of-function experimental approaches capable to systematically interrogate their function. We review all studies carried up to date that used arrayed screening approaches to explore the function of these genetic elements on those biological contexts, using focused or genome-wide genetic libraries. We further discuss the limitations and advantages of approaches based on assays with population-level primary readouts, derived from single-parameter plate readers, or cell-level primary readouts, obtained using multiparametric flow cytometry or quantitative fluorescence microscopy (i.e., high-content screening). Finally, we discuss technical limitation and future perspectives, highlighting how the integration of screening data may lead to major advances in the field of stem cell research and therapy.
Collapse
Affiliation(s)
- Rodrigo Alexandre Panepucci
- Laboratory of Functional Biology (LFBio), Center for Cell-Based Therapy (CTC), Regional Blood Center of Ribeirão Preto, Rua Tenente Catão Roxo, 2501, Ribeirão Preto, SP CEP: 14051-140 Brazil
- Department of Genetics, Ribeirao Preto Medical School, University of São Paulo (FMRP-USP), Ribeirão Preto, SP Brazil
| | - Ildercílio Mota de Souza Lima
- Laboratory of Functional Biology (LFBio), Center for Cell-Based Therapy (CTC), Regional Blood Center of Ribeirão Preto, Rua Tenente Catão Roxo, 2501, Ribeirão Preto, SP CEP: 14051-140 Brazil
- Department of Genetics, Ribeirao Preto Medical School, University of São Paulo (FMRP-USP), Ribeirão Preto, SP Brazil
| |
Collapse
|
78
|
Danter WR. DeepNEU: cellular reprogramming comes of age - a machine learning platform with application to rare diseases research. Orphanet J Rare Dis 2019; 14:13. [PMID: 30630505 PMCID: PMC6327463 DOI: 10.1186/s13023-018-0983-3] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2018] [Accepted: 12/21/2018] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Conversion of human somatic cells into induced pluripotent stem cells (iPSCs) is often an inefficient, time consuming and expensive process. Also, the tendency of iPSCs to revert to their original somatic cell type over time continues to be problematic. A computational model of iPSCs identifying genes/molecules necessary for iPSC generation and maintenance could represent a crucial step forward for improved stem cell research. The combination of substantial genetic relationship data, advanced computing hardware and powerful nonlinear modeling software could make the possibility of artificially-induced pluripotent stem cells (aiPSC) a reality. We have developed an unsupervised deep machine learning technology, called DeepNEU that is based on a fully-connected recurrent neural network architecture with one network processing layer for each input. DeepNEU was used to simulate aiPSC systems using a defined set of reprogramming transcription factors. Genes/proteins that were reported to be essential in human pluripotent stem cells (hPSC) were used for system modelling. RESULTS The Mean Squared Error (MSE) function was used to assess system learning. System convergence was defined at MSE < 0.001. The markers of human iPSC pluripotency (N = 15) were all upregulated in the aiPSC final model. These upregulated/expressed genes in the aiPSC system were entirely consistent with results obtained for iPSCs. CONCLUSION This research introduces and validates the potential use of aiPSCs as computer models of human pluripotent stem cell systems. Disease-specific aiPSCs have the potential to improve disease modeling, prototyping of wet lab experiments, and prediction of genes relevant and necessary for aiPSC production and maintenance for both common and rare diseases in a cost-effective manner.
Collapse
Affiliation(s)
- Wayne R Danter
- 123Genetix, 147 Chesham Ave, London, ON, N6G 3V2, Canada.
| |
Collapse
|
79
|
Abstract
Research on yeast has produced a plethora of tools and resources that have been central to the progress of systems biology. This chapter reviews these resources, explains the innovations that have been made since the first edition of this book, and introduces the constituent chapters of the current edition. The value of these resources not only in building and testing models of the functional networks of the yeast cell, but also in providing a foundation for network studies on the molecular basis of complex human diseases is considered. The gaps in this vast compendium of data, including enzyme kinetic characteristics, biomass composition, transport processes, and cell-cell interactions are discussed, as are the interactions between yeast cells and those of other species. The relevance of these studies to both traditional and advanced biotechnologies and to human medicine is considered, and the opportunities and challenges in using unicellular yeasts to model the systems of multicellular organisms are presented.
Collapse
Affiliation(s)
- Stephen G Oliver
- Department of Biochemistry, University of Cambridge, Cambridge, UK.
- Cambridge Systems Biology Centre, University of Cambridge, Cambridge, UK.
| |
Collapse
|
80
|
Artificial intelligence in drug development: present status and future prospects. Drug Discov Today 2018; 24:773-780. [PMID: 30472429 DOI: 10.1016/j.drudis.2018.11.014] [Citation(s) in RCA: 299] [Impact Index Per Article: 42.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2018] [Revised: 10/14/2018] [Accepted: 11/19/2018] [Indexed: 12/11/2022]
Abstract
Artificial intelligence (AI) uses personified knowledge and learns from the solutions it produces to address not only specific but also complex problems. Remarkable improvements in computational power coupled with advancements in AI technology could be utilised to revolutionise the drug development process. At present, the pharmaceutical industry is facing challenges in sustaining their drug development programmes because of increased R&D costs and reduced efficiency. In this review, we discuss the major causes of attrition rates in new drug approvals, the possible ways that AI can improve the efficiency of the drug development process and collaboration of pharmaceutical industry giants with AI-powered drug discovery firms.
Collapse
|
81
|
Bakker E, Swain PS, Crane MM. Morphologically constrained and data informed cell segmentation of budding yeast. Bioinformatics 2018; 34:88-96. [PMID: 28968663 DOI: 10.1093/bioinformatics/btx550] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2017] [Accepted: 09/03/2017] [Indexed: 01/11/2023] Open
Abstract
Motivation Although high-content image cytometry is becoming increasingly routine, processing the large amount of data acquired during time-lapse experiments remains a challenge. The majority of approaches for automated single-cell segmentation focus on flat, uniform fields of view covered with a single layer of cells. In the increasingly popular microfluidic devices that trap individual cells for long term imaging, these conditions are not met. Consequently, most techniques for segmentation perform poorly. Although potentially constraining the generalizability of software, incorporating information about the microfluidic features, flow of media and the morphology of the cells can substantially improve performance. Results Here we present DISCO (Data Informed Segmentation of Cell Objects), a framework for using the physical constraints imposed by microfluidic traps, the shape based morphological constraints of budding yeast and temporal information about cell growth and motion to allow tracking and segmentation of cells in microfluidic devices. Using manually curated datasets, we demonstrate substantial improvements in both tracking and segmentation when compared with existing software. Availability and implementation The MATLAB code for the algorithm and for measuring performance is available at https://github.com/pswain/segmentation-software and the test images and the curated ground-truth results used for comparing the algorithms are available at http://datashare.is.ed.ac.uk/handle/10283/2002. Contact mcrane2@uw.edu. Supplementary information Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Elco Bakker
- SynthSys-Synthetic and Systems Biology, University of Edinburgh, Edinburgh EH9 3BF, UK.,School of Biological Sciences, University of Edinburgh, Edinburgh EH9 3BF, UK
| | - Peter S Swain
- SynthSys-Synthetic and Systems Biology, University of Edinburgh, Edinburgh EH9 3BF, UK.,School of Biological Sciences, University of Edinburgh, Edinburgh EH9 3BF, UK
| | - Matthew M Crane
- SynthSys-Synthetic and Systems Biology, University of Edinburgh, Edinburgh EH9 3BF, UK.,School of Biological Sciences, University of Edinburgh, Edinburgh EH9 3BF, UK
| |
Collapse
|
82
|
Nitta N, Sugimura T, Isozaki A, Mikami H, Hiraki K, Sakuma S, Iino T, Arai F, Endo T, Fujiwaki Y, Fukuzawa H, Hase M, Hayakawa T, Hiramatsu K, Hoshino Y, Inaba M, Ito T, Karakawa H, Kasai Y, Koizumi K, Lee S, Lei C, Li M, Maeno T, Matsusaka S, Murakami D, Nakagawa A, Oguchi Y, Oikawa M, Ota T, Shiba K, Shintaku H, Shirasaki Y, Suga K, Suzuki Y, Suzuki N, Tanaka Y, Tezuka H, Toyokawa C, Yalikun Y, Yamada M, Yamagishi M, Yamano T, Yasumoto A, Yatomi Y, Yazawa M, Di Carlo D, Hosokawa Y, Uemura S, Ozeki Y, Goda K. Intelligent Image-Activated Cell Sorting. Cell 2018; 175:266-276.e13. [DOI: 10.1016/j.cell.2018.08.028] [Citation(s) in RCA: 298] [Impact Index Per Article: 42.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2018] [Revised: 08/09/2018] [Accepted: 08/15/2018] [Indexed: 11/27/2022]
|
83
|
Scheeder C, Heigwer F, Boutros M. Machine learning and image-based profiling in drug discovery. CURRENT OPINION IN SYSTEMS BIOLOGY 2018; 10:43-52. [PMID: 30159406 PMCID: PMC6109111 DOI: 10.1016/j.coisb.2018.05.004] [Citation(s) in RCA: 94] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The increase in imaging throughput, new analytical frameworks and high-performance computational resources open new avenues for data-rich phenotypic profiling of small molecules in drug discovery. Image-based profiling assays assessing single-cell phenotypes have been used to explore mechanisms of action, target efficacy and toxicity of small molecules. Technological advances to generate large data sets together with new machine learning approaches for the analysis of high-dimensional profiling data create opportunities to improve many steps in drug discovery. In this review, we will discuss how recent studies applied machine learning approaches in functional profiling workflows with a focus on chemical genetics. While their utility in image-based screening and profiling is predictably evident, examples of novel insights beyond the status quo based on the applications of machine learning approaches are just beginning to emerge. To enable discoveries, future studies also need to develop methodologies that lower the entry barriers to high-throughput profiling experiments by streamlining image-based profiling assays and providing applications for advanced learning technologies such as easy to deploy deep neural networks.
Collapse
Affiliation(s)
| | | | - Michael Boutros
- German Cancer Research Center (DKFZ), Division Signaling and Functional Genomics and Heidelberg University, Department of Cell and Molecular Biology, Medical Faculty Mannheim, D-69120 Heidelberg, Germany
| |
Collapse
|
84
|
Verstraelen P, Van Dyck M, Verschuuren M, Kashikar ND, Nuydens R, Timmermans JP, De Vos WH. Image-Based Profiling of Synaptic Connectivity in Primary Neuronal Cell Culture. Front Neurosci 2018; 12:389. [PMID: 29997468 PMCID: PMC6028601 DOI: 10.3389/fnins.2018.00389] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2017] [Accepted: 05/22/2018] [Indexed: 12/04/2022] Open
Abstract
Neurological disorders display a broad spectrum of clinical manifestations. Yet, at the cellular level, virtually all these diseases converge into a common phenotype of dysregulated synaptic connectivity. In dementia, synapse dysfunction precedes neurodegeneration and cognitive impairment by several years, making the synapse a crucial entry point for the development of diagnostic and therapeutic strategies. Whereas high-resolution imaging and biochemical fractionations yield detailed insight into the molecular composition of the synapse, standardized assays are required to quickly gauge synaptic connectivity across large populations of cells under a variety of experimental conditions. Such screening capabilities have now become widely accessible with the advent of high-throughput, high-content microscopy. In this review, we discuss how microscopy-based approaches can be used to extract quantitative information about synaptic connectivity in primary neurons with deep coverage. We elaborate on microscopic readouts that may serve as a proxy for morphofunctional connectivity and we critically analyze their merits and limitations. Finally, we allude to the potential of alternative culture paradigms and integrative approaches to enable comprehensive profiling of synaptic connectivity.
Collapse
Affiliation(s)
- Peter Verstraelen
- Laboratory of Cell Biology and Histology, Department of Veterinary Sciences, University of Antwerp, Antwerp, Belgium
| | - Michiel Van Dyck
- Laboratory of Cell Biology and Histology, Department of Veterinary Sciences, University of Antwerp, Antwerp, Belgium
| | - Marlies Verschuuren
- Laboratory of Cell Biology and Histology, Department of Veterinary Sciences, University of Antwerp, Antwerp, Belgium
| | | | - Rony Nuydens
- Janssen Research and Development, Janssen Pharmaceutica N.V., Beerse, Belgium
| | - Jean-Pierre Timmermans
- Laboratory of Cell Biology and Histology, Department of Veterinary Sciences, University of Antwerp, Antwerp, Belgium
| | - Winnok H. De Vos
- Laboratory of Cell Biology and Histology, Department of Veterinary Sciences, University of Antwerp, Antwerp, Belgium
- Cell Systems and Imaging, Department of Molecular Biotechnology, Ghent University, Ghent, Belgium
| |
Collapse
|
85
|
Ching T, Himmelstein DS, Beaulieu-Jones BK, Kalinin AA, Do BT, Way GP, Ferrero E, Agapow PM, Zietz M, Hoffman MM, Xie W, Rosen GL, Lengerich BJ, Israeli J, Lanchantin J, Woloszynek S, Carpenter AE, Shrikumar A, Xu J, Cofer EM, Lavender CA, Turaga SC, Alexandari AM, Lu Z, Harris DJ, DeCaprio D, Qi Y, Kundaje A, Peng Y, Wiley LK, Segler MHS, Boca SM, Swamidass SJ, Huang A, Gitter A, Greene CS. Opportunities and obstacles for deep learning in biology and medicine. J R Soc Interface 2018; 15:20170387. [PMID: 29618526 PMCID: PMC5938574 DOI: 10.1098/rsif.2017.0387] [Citation(s) in RCA: 900] [Impact Index Per Article: 128.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2017] [Accepted: 03/07/2018] [Indexed: 11/12/2022] Open
Abstract
Deep learning describes a class of machine learning algorithms that are capable of combining raw inputs into layers of intermediate features. These algorithms have recently shown impressive results across a variety of domains. Biology and medicine are data-rich disciplines, but the data are complex and often ill-understood. Hence, deep learning techniques may be particularly well suited to solve problems of these fields. We examine applications of deep learning to a variety of biomedical problems-patient classification, fundamental biological processes and treatment of patients-and discuss whether deep learning will be able to transform these tasks or if the biomedical sphere poses unique challenges. Following from an extensive literature review, we find that deep learning has yet to revolutionize biomedicine or definitively resolve any of the most pressing challenges in the field, but promising advances have been made on the prior state of the art. Even though improvements over previous baselines have been modest in general, the recent progress indicates that deep learning methods will provide valuable means for speeding up or aiding human investigation. Though progress has been made linking a specific neural network's prediction to input features, understanding how users should interpret these models to make testable hypotheses about the system under study remains an open challenge. Furthermore, the limited amount of labelled data for training presents problems in some domains, as do legal and privacy constraints on work with sensitive health records. Nonetheless, we foresee deep learning enabling changes at both bench and bedside with the potential to transform several areas of biology and medicine.
Collapse
Affiliation(s)
- Travers Ching
- Molecular Biosciences and Bioengineering Graduate Program, University of Hawaii at Manoa, Honolulu, HI, USA
| | - Daniel S Himmelstein
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Brett K Beaulieu-Jones
- Genomics and Computational Biology Graduate Group, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Alexandr A Kalinin
- Department of Computational Medicine and Bioinformatics, University of Michigan Medical School, Ann Arbor, MI, USA
| | | | - Gregory P Way
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Enrico Ferrero
- Computational Biology and Stats, Target Sciences, GlaxoSmithKline, Stevenage, UK
| | | | - Michael Zietz
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Michael M Hoffman
- Princess Margaret Cancer Centre, Toronto, Ontario, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
- Department of Computer Science, University of Toronto, Toronto, Ontario, Canada
| | - Wei Xie
- Electrical Engineering and Computer Science, Vanderbilt University, Nashville, TN, USA
| | - Gail L Rosen
- Ecological and Evolutionary Signal-processing and Informatics Laboratory, Department of Electrical and Computer Engineering, Drexel University, Philadelphia, PA, USA
| | - Benjamin J Lengerich
- Computational Biology Department, School of Computer Science, Carnegie Mellon University, Pittsburgh, PA, USA
| | - Johnny Israeli
- Biophysics Program, Stanford University, Stanford, CA, USA
| | - Jack Lanchantin
- Department of Computer Science, University of Virginia, Charlottesville, VA, USA
| | - Stephen Woloszynek
- Ecological and Evolutionary Signal-processing and Informatics Laboratory, Department of Electrical and Computer Engineering, Drexel University, Philadelphia, PA, USA
| | - Anne E Carpenter
- Imaging Platform, Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Avanti Shrikumar
- Department of Computer Science, Stanford University, Stanford, CA, USA
| | - Jinbo Xu
- Toyota Technological Institute at Chicago, Chicago, IL, USA
| | - Evan M Cofer
- Department of Computer Science, Trinity University, San Antonio, TX, USA
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ, USA
| | - Christopher A Lavender
- Integrative Bioinformatics, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC, USA
| | - Srinivas C Turaga
- Howard Hughes Medical Institute, Janelia Research Campus, Ashburn, VA, USA
| | - Amr M Alexandari
- Department of Computer Science, Stanford University, Stanford, CA, USA
| | - Zhiyong Lu
- National Center for Biotechnology Information and National Library of Medicine, National Institutes of Health, Bethesda, MD, USA
| | - David J Harris
- Department of Wildlife Ecology and Conservation, University of Florida, Gainesville, FL, USA
| | | | - Yanjun Qi
- Department of Computer Science, University of Virginia, Charlottesville, VA, USA
| | - Anshul Kundaje
- Department of Computer Science, Stanford University, Stanford, CA, USA
- Department of Genetics, Stanford University, Stanford, CA, USA
| | - Yifan Peng
- National Center for Biotechnology Information and National Library of Medicine, National Institutes of Health, Bethesda, MD, USA
| | - Laura K Wiley
- Division of Biomedical Informatics and Personalized Medicine, University of Colorado School of Medicine, Aurora, CO, USA
| | - Marwin H S Segler
- Institute of Organic Chemistry, Westfälische Wilhelms-Universität Münster, Münster, Germany
| | - Simina M Boca
- Innovation Center for Biomedical Informatics, Georgetown University Medical Center, Washington, DC, USA
| | - S Joshua Swamidass
- Department of Pathology and Immunology, Washington University in Saint Louis, St Louis, MO, USA
| | - Austin Huang
- Department of Medicine, Brown University, Providence, RI, USA
| | - Anthony Gitter
- Department of Biostatistics and Medical Informatics, University of Wisconsin-Madison, Madison, WI, USA
- Morgridge Institute for Research, Madison, WI, USA
| | - Casey S Greene
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
86
|
Uhler C, Shivashankar GV. Nuclear Mechanopathology and Cancer Diagnosis. Trends Cancer 2018; 4:320-331. [PMID: 29606315 DOI: 10.1016/j.trecan.2018.02.009] [Citation(s) in RCA: 84] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Revised: 02/26/2018] [Accepted: 02/27/2018] [Indexed: 11/29/2022]
Abstract
Abnormalities in nuclear and chromatin organization are hallmarks of many diseases including cancer. In this review, we highlight our understanding of how the cellular microenvironment regulates nuclear morphology and, with it, the spatial organization of chromosomes and genes, resulting in cell type-specific genomic programs. We also discuss the molecular basis for maintaining nuclear and genomic integrity and how alterations in nuclear mechanotransduction pathways result in various diseases. Finally, we highlight the importance of digital pathology based on nuclear morphometric features combined with single-cell genomics for early cancer diagnostics.
Collapse
Affiliation(s)
- Caroline Uhler
- Department of Electrical Engineering & Computer Science, Institute for Data, Systems & Society, MIT, Cambridge, MA, USA
| | - G V Shivashankar
- Mechanobiology Institute, National University of Singapore, Singapore; FIRC Institute of Molecular Oncology (IFOM), Milan, Italy.
| |
Collapse
|
87
|
O'Duibhir E, Paris J, Lawson H, Sepulveda C, Shenton DD, Carragher NO, Kranc KR. Machine Learning Enables Live Label-Free Phenotypic Screening in Three Dimensions. Assay Drug Dev Technol 2018; 16:51-63. [PMID: 29345979 DOI: 10.1089/adt.2017.819] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
There is a large amount of information in brightfield images that was previously inaccessible by using traditional microscopy techniques. This information can now be exploited by using machine-learning approaches for both image segmentation and the classification of objects. We have combined these approaches with a label-free assay for growth and differentiation of leukemic colonies, to generate a novel platform for phenotypic drug discovery. Initially, a supervised machine-learning algorithm was used to identify in-focus colonies growing in a three-dimensional (3D) methylcellulose gel. Once identified, unsupervised clustering and principle component analysis of texture-based phenotypic profiles were applied to group similar phenotypes. In a proof-of-concept study, we successfully identified a novel phenotype induced by a compound that is currently in clinical trials for the treatment of leukemia. We believe that our platform will be of great benefit for the utilization of patient-derived 3D cell culture systems for both drug discovery and diagnostic applications.
Collapse
Affiliation(s)
- Eoghan O'Duibhir
- 1 Centre for Regenerative Medicine, University of Edinburgh , Edinburgh, United Kingdom
| | - Jasmin Paris
- 1 Centre for Regenerative Medicine, University of Edinburgh , Edinburgh, United Kingdom
| | - Hannah Lawson
- 1 Centre for Regenerative Medicine, University of Edinburgh , Edinburgh, United Kingdom
| | - Catarina Sepulveda
- 1 Centre for Regenerative Medicine, University of Edinburgh , Edinburgh, United Kingdom
| | - Dahlia Doughty Shenton
- 2 Edinburgh Phenotypic Assay Centre, The Queen's Medical Research Institute, University of Edinburgh , Edinburgh, United Kingdom
| | - Neil O Carragher
- 3 Cancer Research UK Edinburgh Centre, Institute of Genetics and Molecular Medicine, University of Edinburgh , Edinburgh, United Kingdom
| | - Kamil R Kranc
- 1 Centre for Regenerative Medicine, University of Edinburgh , Edinburgh, United Kingdom
- 3 Cancer Research UK Edinburgh Centre, Institute of Genetics and Molecular Medicine, University of Edinburgh , Edinburgh, United Kingdom
| |
Collapse
|
88
|
Shaping the Cell and the Future: Recent Advancements in Biophysical Aspects Relevant to Regenerative Medicine. J Funct Morphol Kinesiol 2017. [DOI: 10.3390/jfmk3010002] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
|
89
|
Radhakrishnan A, Damodaran K, Soylemezoglu AC, Uhler C, Shivashankar GV. Machine Learning for Nuclear Mechano-Morphometric Biomarkers in Cancer Diagnosis. Sci Rep 2017; 7:17946. [PMID: 29263424 PMCID: PMC5738417 DOI: 10.1038/s41598-017-17858-1] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2017] [Accepted: 11/30/2017] [Indexed: 12/04/2022] Open
Abstract
Current cancer diagnosis employs various nuclear morphometric measures. While these have allowed accurate late-stage prognosis, early diagnosis is still a major challenge. Recent evidence highlights the importance of alterations in mechanical properties of single cells and their nuclei as critical drivers for the onset of cancer. We here present a method to detect subtle changes in nuclear morphometrics at single-cell resolution by combining fluorescence imaging and deep learning. This assay includes a convolutional neural net pipeline and allows us to discriminate between normal and human breast cancer cell lines (fibrocystic and metastatic states) as well as normal and cancer cells in tissue slices with high accuracy. Further, we establish the sensitivity of our pipeline by detecting subtle alterations in normal cells when subjected to small mechano-chemical perturbations that mimic tumor microenvironments. In addition, our assay provides interpretable features that could aid pathological inspections. This pipeline opens new avenues for early disease diagnostics and drug discovery.
Collapse
Affiliation(s)
- Adityanarayanan Radhakrishnan
- Department of Electrical Engineering and Computer Science, Laboratory for Information and Decision Systems, Institute for Data, Systems and Society, MIT, Cambridge, MA, USA
| | - Karthik Damodaran
- Mechanobiology Institute and Department of Biological Sciences, National University of Singapore, Singapore, Singapore
| | - Ali C Soylemezoglu
- Department of Electrical Engineering and Computer Science, Laboratory for Information and Decision Systems, Institute for Data, Systems and Society, MIT, Cambridge, MA, USA
| | - Caroline Uhler
- Department of Electrical Engineering and Computer Science, Laboratory for Information and Decision Systems, Institute for Data, Systems and Society, MIT, Cambridge, MA, USA.
| | - G V Shivashankar
- Mechanobiology Institute and Department of Biological Sciences, National University of Singapore, Singapore, Singapore. .,FIRC Institute for Molecular Oncology (IFOM), Milan, Italy.
| |
Collapse
|
90
|
Doan M. Zooming in on adipocytes: High and deep. Cytometry A 2017; 91:1051-1054. [PMID: 29072805 DOI: 10.1002/cyto.a.23269] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2017] [Revised: 09/29/2017] [Accepted: 10/02/2017] [Indexed: 11/11/2022]
Affiliation(s)
- Minh Doan
- Imaging Platform, Broad Institute of MIT and Harvard, Cambridge, Massachusetts 02142
| |
Collapse
|
91
|
Uhler C, Shivashankar GV. Regulation of genome organization and gene expression by nuclear mechanotransduction. Nat Rev Mol Cell Biol 2017; 18:717-727. [PMID: 29044247 DOI: 10.1038/nrm.2017.101] [Citation(s) in RCA: 258] [Impact Index Per Article: 32.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
It is well established that cells sense chemical signals from their local microenvironment and transduce them to the nucleus to regulate gene expression programmes. Although a number of experiments have shown that mechanical cues can also modulate gene expression, the underlying mechanisms are far from clear. Nevertheless, we are now beginning to understand how mechanical cues are transduced to the nucleus and how they influence nuclear mechanics, genome organization and transcription. In particular, recent progress in super-resolution imaging, in genome-wide application of RNA sequencing, chromatin immunoprecipitation and chromosome conformation capture and in theoretical modelling of 3D genome organization enables the exploration of the relationship between cell mechanics, 3D chromatin configurations and transcription, thereby shedding new light on how mechanical forces regulate gene expression.
Collapse
Affiliation(s)
- Caroline Uhler
- Department of Electrical Engineering and Computer Science, Laboratory of Information and Decision Systems, Institute for Data, Systems and Society, Massachusetts Institute of Technology (MIT), Cambridge, Massachusetts 02139, USA
| | - G V Shivashankar
- Mechanobiology Institute, National University of Singapore, 119077 Singapore.,Italian Foundation for Cancer Research (FIRC) Institute of Molecular Oncology (IFOM), Milan 20139, Italy
| |
Collapse
|
92
|
Québatte M, Dehio C. Systems-level interference strategies to decipher host factors involved in bacterial pathogen interaction: from RNAi to CRISPRi. Curr Opin Microbiol 2017; 39:34-41. [DOI: 10.1016/j.mib.2017.08.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2017] [Revised: 08/02/2017] [Accepted: 08/02/2017] [Indexed: 12/16/2022]
|
93
|
Kobayashi H, Lei C, Wu Y, Mao A, Jiang Y, Guo B, Ozeki Y, Goda K. Label-free detection of cellular drug responses by high-throughput bright-field imaging and machine learning. Sci Rep 2017. [PMID: 28963483 DOI: 10.1038/s41598‐017‐12378‐4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
In the last decade, high-content screening based on multivariate single-cell imaging has been proven effective in drug discovery to evaluate drug-induced phenotypic variations. Unfortunately, this method inherently requires fluorescent labeling which has several drawbacks. Here we present a label-free method for evaluating cellular drug responses only by high-throughput bright-field imaging with the aid of machine learning algorithms. Specifically, we performed high-throughput bright-field imaging of numerous drug-treated and -untreated cells (N = ~240,000) by optofluidic time-stretch microscopy with high throughput up to 10,000 cells/s and applied machine learning to the cell images to identify their morphological variations which are too subtle for human eyes to detect. Consequently, we achieved a high accuracy of 92% in distinguishing drug-treated and -untreated cells without the need for labeling. Furthermore, we also demonstrated that dose-dependent, drug-induced morphological change from different experiments can be inferred from the classification accuracy of a single classification model. Our work lays the groundwork for label-free drug screening in pharmaceutical science and industry.
Collapse
Affiliation(s)
| | - Cheng Lei
- Department of Chemistry, University of Tokyo, Tokyo, 113-0033, Japan.
| | - Yi Wu
- Department of Computational Biology, Carnegie Mellon University, Pittsburgh, Pennsylvania, 15213, USA
| | - Ailin Mao
- Department of Chemistry, University of Tokyo, Tokyo, 113-0033, Japan
| | - Yiyue Jiang
- Department of Chemistry, University of Tokyo, Tokyo, 113-0033, Japan
| | - Baoshan Guo
- Department of Chemistry, University of Tokyo, Tokyo, 113-0033, Japan
| | - Yasuyuki Ozeki
- Department of Electrical Engineering and Information Systems, University of Tokyo, Tokyo, 113-8656, Japan
| | - Keisuke Goda
- Department of Chemistry, University of Tokyo, Tokyo, 113-0033, Japan. .,Japan Science and Technology Agency, Kawaguchi, 332-0012, Japan. .,Department of Electrical Engineering, University of California, Los Angeles, California, 90095, USA.
| |
Collapse
|
94
|
Kobayashi H, Lei C, Wu Y, Mao A, Jiang Y, Guo B, Ozeki Y, Goda K. Label-free detection of cellular drug responses by high-throughput bright-field imaging and machine learning. Sci Rep 2017; 7:12454. [PMID: 28963483 PMCID: PMC5622112 DOI: 10.1038/s41598-017-12378-4] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2017] [Accepted: 09/07/2017] [Indexed: 12/25/2022] Open
Abstract
In the last decade, high-content screening based on multivariate single-cell imaging has been proven effective in drug discovery to evaluate drug-induced phenotypic variations. Unfortunately, this method inherently requires fluorescent labeling which has several drawbacks. Here we present a label-free method for evaluating cellular drug responses only by high-throughput bright-field imaging with the aid of machine learning algorithms. Specifically, we performed high-throughput bright-field imaging of numerous drug-treated and -untreated cells (N = ~240,000) by optofluidic time-stretch microscopy with high throughput up to 10,000 cells/s and applied machine learning to the cell images to identify their morphological variations which are too subtle for human eyes to detect. Consequently, we achieved a high accuracy of 92% in distinguishing drug-treated and -untreated cells without the need for labeling. Furthermore, we also demonstrated that dose-dependent, drug-induced morphological change from different experiments can be inferred from the classification accuracy of a single classification model. Our work lays the groundwork for label-free drug screening in pharmaceutical science and industry.
Collapse
Affiliation(s)
| | - Cheng Lei
- Department of Chemistry, University of Tokyo, Tokyo, 113-0033, Japan.
| | - Yi Wu
- Department of Computational Biology, Carnegie Mellon University, Pittsburgh, Pennsylvania, 15213, USA
| | - Ailin Mao
- Department of Chemistry, University of Tokyo, Tokyo, 113-0033, Japan
| | - Yiyue Jiang
- Department of Chemistry, University of Tokyo, Tokyo, 113-0033, Japan
| | - Baoshan Guo
- Department of Chemistry, University of Tokyo, Tokyo, 113-0033, Japan
| | - Yasuyuki Ozeki
- Department of Electrical Engineering and Information Systems, University of Tokyo, Tokyo, 113-8656, Japan
| | - Keisuke Goda
- Department of Chemistry, University of Tokyo, Tokyo, 113-0033, Japan. .,Japan Science and Technology Agency, Kawaguchi, 332-0012, Japan. .,Department of Electrical Engineering, University of California, Los Angeles, California, 90095, USA.
| |
Collapse
|
95
|
Data-analysis strategies for image-based cell profiling. Nat Methods 2017; 14:849-863. [PMID: 28858338 PMCID: PMC6871000 DOI: 10.1038/nmeth.4397] [Citation(s) in RCA: 433] [Impact Index Per Article: 54.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2016] [Accepted: 07/28/2017] [Indexed: 12/16/2022]
Abstract
Image-based cell profiling is a high-throughput strategy for the quantification of phenotypic differences among a variety of cell populations. It paves the way to studying biological systems on a large scale by using chemical and genetic perturbations. The general workflow for this technology involves image acquisition with high-throughput microscopy systems and subsequent image processing and analysis. Here, we introduce the steps required to create high-quality image-based (i.e., morphological) profiles from a collection of microscopy images. We recommend techniques that have proven useful in each stage of the data analysis process, on the basis of the experience of 20 laboratories worldwide that are refining their image-based cell-profiling methodologies in pursuit of biological discovery. The recommended techniques cover alternatives that may suit various biological goals, experimental designs, and laboratories' preferences.
Collapse
|
96
|
Schroeder JA. Application of laboratory and digital techniques for visual enhancement during the ultrastructural assessment of cilia. Ultrastruct Pathol 2017; 41:399-407. [DOI: 10.1080/01913123.2017.1363335] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Affiliation(s)
- Josef A. Schroeder
- Central EM-Lab, Department of Pathology, University Hospital Regensburg, Regensburg, Germany
| |
Collapse
|
97
|
Leggett SE, Khoo AS, Wong IY. Multicellular tumor invasion and plasticity in biomimetic materials. Biomater Sci 2017; 5:1460-1479. [PMID: 28530743 PMCID: PMC5531215 DOI: 10.1039/c7bm00272f] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Cancer cell invasion through the extracellular matrix is associated with metastatic spread and therapeutic resistance. In carcinomas, the detachment and dissemination of individual cells has been associated with an epithelial-mesenchymal transition, but tumors can also invade using collective, multicellular phenotypes. This malignant tumor progression is also associated with alignment and stiffening of the surrounding extracellular matrix. Historically, tumor invasion has been investigated using 2D monolayer culture, small animal models or patient histology. These assays have been complemented by the use of natural biomaterials such as reconstituted basement membrane and collagen I. More recently, engineered materials with well-defined physical, chemical and biomolecular properties have enabled more controlled microenvironments. In this review, we highlight recent developments in multicellular tumor invasion based on microfabricated structures or hydrogels. We emphasize the role of interfacial geometries, biomaterial stiffness, matrix remodeling, and co-culture models. Finally, we discuss future directions for the field, particularly integration with precision measurements of biomaterial properties and single cell heterogeneity, standardization and scale-up of these platforms, as well as integration with patient-derived samples.
Collapse
Affiliation(s)
- Susan E Leggett
- School of Engineering, Center for Biomedical Engineering, Brown University, Providence, RI 02912, USA. and Pathobiology Graduate Program, Brown University, Providence, RI 02912, USA
| | - Amanda S Khoo
- School of Engineering, Center for Biomedical Engineering, Brown University, Providence, RI 02912, USA.
| | - Ian Y Wong
- School of Engineering, Center for Biomedical Engineering, Brown University, Providence, RI 02912, USA. and Pathobiology Graduate Program, Brown University, Providence, RI 02912, USA
| |
Collapse
|
98
|
Kraus OZ, Grys BT, Ba J, Chong Y, Frey BJ, Boone C, Andrews BJ. Automated analysis of high-content microscopy data with deep learning. Mol Syst Biol 2017; 13:924. [PMID: 28420678 PMCID: PMC5408780 DOI: 10.15252/msb.20177551] [Citation(s) in RCA: 152] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
Existing computational pipelines for quantitative analysis of high‐content microscopy data rely on traditional machine learning approaches that fail to accurately classify more than a single dataset without substantial tuning and training, requiring extensive analysis. Here, we demonstrate that the application of deep learning to biological image data can overcome the pitfalls associated with conventional machine learning classifiers. Using a deep convolutional neural network (DeepLoc) to analyze yeast cell images, we show improved performance over traditional approaches in the automated classification of protein subcellular localization. We also demonstrate the ability of DeepLoc to classify highly divergent image sets, including images of pheromone‐arrested cells with abnormal cellular morphology, as well as images generated in different genetic backgrounds and in different laboratories. We offer an open‐source implementation that enables updating DeepLoc on new microscopy datasets. This study highlights deep learning as an important tool for the expedited analysis of high‐content microscopy data.
Collapse
Affiliation(s)
- Oren Z Kraus
- Department of Electrical and Computer Engineering, University of Toronto, Toronto, ON, Canada.,Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, ON, Canada
| | - Ben T Grys
- Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, ON, Canada.,Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
| | - Jimmy Ba
- Department of Electrical and Computer Engineering, University of Toronto, Toronto, ON, Canada
| | - Yolanda Chong
- Cellular Pharmacology, Discovery Sciences, Janssen Pharmaceutical Companies, Johnson & Johnson, Beerse, Belgium
| | - Brendan J Frey
- Department of Electrical and Computer Engineering, University of Toronto, Toronto, ON, Canada.,Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, ON, Canada.,Canadian Institute for Advanced Research, Program on Genetic Networks, Toronto, ON, Canada.,Canadian Institute for Advanced Research, Program on Learning in Machines & Brains, Toronto, ON, Canada
| | - Charles Boone
- Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, ON, Canada .,Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada.,Canadian Institute for Advanced Research, Program on Genetic Networks, Toronto, ON, Canada
| | - Brenda J Andrews
- Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, ON, Canada .,Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada.,Canadian Institute for Advanced Research, Program on Genetic Networks, Toronto, ON, Canada
| |
Collapse
|
99
|
Kan A. Machine learning applications in cell image analysis. Immunol Cell Biol 2017; 95:525-530. [PMID: 28294138 DOI: 10.1038/icb.2017.16] [Citation(s) in RCA: 75] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2016] [Revised: 02/28/2017] [Accepted: 03/08/2017] [Indexed: 02/06/2023]
Abstract
Machine learning (ML) refers to a set of automatic pattern recognition methods that have been successfully applied across various problem domains, including biomedical image analysis. This review focuses on ML applications for image analysis in light microscopy experiments with typical tasks of segmenting and tracking individual cells, and modelling of reconstructed lineage trees. After describing a typical image analysis pipeline and highlighting challenges of automatic analysis (for example, variability in cell morphology, tracking in presence of clutters) this review gives a brief historical outlook of ML, followed by basic concepts and definitions required for understanding examples. This article then presents several example applications at various image processing stages, including the use of supervised learning methods for improving cell segmentation, and the application of active learning for tracking. The review concludes with remarks on parameter setting and future directions.
Collapse
Affiliation(s)
- Andrey Kan
- Division of Immunology, The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia.,Department of Medical Biology, The University of Melbourne, Parkville, Victoria, Australia
| |
Collapse
|