701
|
Lyons J, Herring CA, Banerjee A, Simmons AJ, Lau KS. Multiscale analysis of the murine intestine for modeling human diseases. Integr Biol (Camb) 2016; 7:740-57. [PMID: 26040649 DOI: 10.1039/c5ib00030k] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
When functioning properly, the intestine is one of the key interfaces between the human body and its environment. It is responsible for extracting nutrients from our food and excreting our waste products. It provides an environment for a host of healthful microbes and serves as a first defense against pathogenic ones. These processes require tight homeostatic controls, which are provided by the interactions of a complex mix of epithelial, stromal, neural and immune cells, as well as the resident microflora. This homeostasis can be disrupted by invasive microbes, genetic lesions, and carcinogens, resulting in diseases such Clostridium difficile infection, inflammatory bowel disease (IBD) and cancer. Enormous strides have been made in understanding how this important organ functions in health and disease using everything from cell culture systems to animal models to human tissue samples. This has resulted in better therapies for all of these diseases, but there is still significant room for improvement. In the United States alone, 14,000 people per year die of C. difficile, up to 1.6 million people suffer from IBD, and more than 50,000 people die every year from colon cancer. Because these and other intestinal diseases arise from complex interactions between the different components of the gut ecosystem, we propose that systems approaches that address this complexity in an integrative manner may eventually lead to improved therapeutics that deliver lasting cures. This review will discuss the use of systems biology for studying intestinal diseases in vivo with particular emphasis on mouse models. Additionally, it will focus on established experimental techniques that have been used to drive this systems-level analysis, and emerging techniques that will push this field forward in the future.
Collapse
Affiliation(s)
- Jesse Lyons
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | | | | | | | | |
Collapse
|
702
|
Janes KA. Single-cell states versus single-cell atlases - two classes of heterogeneity that differ in meaning and method. Curr Opin Biotechnol 2016; 39:120-125. [PMID: 27042975 DOI: 10.1016/j.copbio.2016.03.015] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2015] [Revised: 03/04/2016] [Accepted: 03/20/2016] [Indexed: 12/30/2022]
Abstract
Recent advances have created new opportunities to dissect cellular heterogeneity at the omics level. The enthusiasm for deep single-cell profiling has obscured a discussion of different types of heterogeneity and the most-appropriate techniques for studying each type. Here, I distinguish heterogeneity in regulation from heterogeneity in lineage. Snapshots of lineage heterogeneity provide a cell atlas that catalogs cellular diversity within complex tissues. Profiles of regulatory heterogeneity seek to interrogate one lineage deeply to capture an ensemble of single-cell states. Single-cell atlases require molecular signatures from many cells at a throughput afforded by mass cytometry-based, microfluidic-based, and microencapsulation-based methods. Single-cell states are more dependent on time, microenvironment, and low-abundance transcripts, emphasizing in situ methods that stress depth of profiling and quantitative accuracy.
Collapse
Affiliation(s)
- Kevin A Janes
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA 22908 USA.
| |
Collapse
|
703
|
Lavin Y, Mortha A, Rahman A, Merad M. Regulation of macrophage development and function in peripheral tissues. Nat Rev Immunol 2016; 15:731-44. [PMID: 26603899 DOI: 10.1038/nri3920] [Citation(s) in RCA: 428] [Impact Index Per Article: 53.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Macrophages are immune cells of haematopoietic origin that provide crucial innate immune defence and have tissue-specific functions in the regulation and maintenance of organ homeostasis. Recent studies of macrophage ontogeny, as well as transcriptional and epigenetic identity, have started to reveal the decisive role of the tissue stroma in the regulation of macrophage function. These findings suggest that most macrophages seed the tissues during embryonic development and functionally specialize in response to cytokines and metabolites that are released by the stroma and drive the expression of unique transcription factors. In this Review, we discuss how recent insights into macrophage ontogeny and macrophage-stroma interactions contribute to our understanding of the crosstalk that shapes macrophage function and the maintenance of organ integrity.
Collapse
Affiliation(s)
- Yonit Lavin
- Department of Oncological Sciences, Tisch Cancer Institute and the Immunology Institute, Icahn School of Medicine at Mount Sinai, New York City, New York 10029, USA
| | - Arthur Mortha
- Department of Oncological Sciences, Tisch Cancer Institute and the Immunology Institute, Icahn School of Medicine at Mount Sinai, New York City, New York 10029, USA
| | - Adeeb Rahman
- Department of Oncological Sciences, Tisch Cancer Institute and the Immunology Institute, Icahn School of Medicine at Mount Sinai, New York City, New York 10029, USA
| | - Miriam Merad
- Department of Oncological Sciences, Tisch Cancer Institute and the Immunology Institute, Icahn School of Medicine at Mount Sinai, New York City, New York 10029, USA
| |
Collapse
|
704
|
Kaznatcheev A, Scott JG, Basanta D. Edge effects in game-theoretic dynamics of spatially structured tumours. J R Soc Interface 2016; 12:20150154. [PMID: 26040596 DOI: 10.1098/rsif.2015.0154] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Cancer dynamics are an evolutionary game between cellular phenotypes. A typical assumption in this modelling paradigm is that the probability of a given phenotypic strategy interacting with another depends exclusively on the abundance of those strategies without regard for local neighbourhood structure. We address this limitation by using the Ohtsuki-Nowak transform to introduce spatial structure to the go versus grow game. We show that spatial structure can promote the invasive (go) strategy. By considering the change in neighbourhood size at a static boundary--such as a blood vessel, organ capsule or basement membrane--we show an edge effect that allows a tumour without invasive phenotypes in the bulk to have a polyclonal boundary with invasive cells. We present an example of this promotion of invasive (epithelial-mesenchymal transition-positive) cells in a metastatic colony of prostate adenocarcinoma in bone marrow. Our results caution that pathologic analyses that do not distinguish between cells in the bulk and cells at a static edge of a tumour can underestimate the number of invasive cells. Although we concentrate on applications in mathematical oncology, we expect our approach to extend to other evolutionary game models where interaction neighbourhoods change at fixed system boundaries.
Collapse
Affiliation(s)
- Artem Kaznatcheev
- School of Computer Science and Department of Psychology, McGill University, Montreal, Quebec, Canada Integrated Mathematical Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Jacob G Scott
- Integrated Mathematical Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA Wolfson Centre for Mathematical Biology, Mathematical Institute, University of Oxford, Oxford, UK
| | - David Basanta
- Integrated Mathematical Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| |
Collapse
|
705
|
Abstract
Imaging reveals complex structures and dynamic interactive processes, located deep inside the body, that are otherwise difficult to decipher. Numerous imaging modalities harness every last inch of the energy spectrum. Clinical modalities include magnetic resonance imaging (MRI), X-ray computed tomography (CT), ultrasound, and light-based methods [endoscopy and optical coherence tomography (OCT)]. Research modalities include various light microscopy techniques (confocal, multiphoton, total internal reflection, superresolution fluorescence microscopy), electron microscopy, mass spectrometry imaging, fluorescence tomography, bioluminescence, variations of OCT, and optoacoustic imaging, among a few others. Although clinical imaging and research microscopy are often isolated from one another, we argue that their combination and integration is not only informative but also essential to discovering new biology and interpreting clinical datasets in which signals invariably originate from hundreds to thousands of cells per voxel.
Collapse
|
706
|
Ahmed Raza SE, Langenkämper D, Sirinukunwattana K, Epstein D, Nattkemper TW, Rajpoot NM. Robust normalization protocols for multiplexed fluorescence bioimage analysis. BioData Min 2016; 9:11. [PMID: 26949415 PMCID: PMC4779207 DOI: 10.1186/s13040-016-0088-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2015] [Accepted: 02/02/2016] [Indexed: 12/18/2022] Open
Abstract
study of mapping and interaction of co-localized proteins at a sub-cellular level is important for understanding complex biological phenomena. One of the recent techniques to map co-localized proteins is to use the standard immuno-fluorescence microscopy in a cyclic manner (Nat Biotechnol 24:1270–8, 2006; Proc Natl Acad Sci 110:11982–7, 2013). Unfortunately, these techniques suffer from variability in intensity and positioning of signals from protein markers within a run and across different runs. Therefore, it is necessary to standardize protocols for preprocessing of the multiplexed bioimaging (MBI) data from multiple runs to a comparable scale before any further analysis can be performed on the data. In this paper, we compare various normalization protocols and propose on the basis of the obtained results, a robust normalization technique that produces consistent results on the MBI data collected from different runs using the Toponome Imaging System (TIS). Normalization results produced by the proposed method on a sample TIS data set for colorectal cancer patients were ranked favorably by two pathologists and two biologists. We show that the proposed method produces higher between class Kullback-Leibler (KL) divergence and lower within class KL divergence on a distribution of cell phenotypes from colorectal cancer and histologically normal samples.
Collapse
Affiliation(s)
- Shan E Ahmed Raza
- Department of Computer Science, University of Warwick, Coventry, CV4 7AL UK
| | | | | | - David Epstein
- Mathematics Institute, University of Warwick, Coventry, CV4 7AL UK
| | | | - Nasir M Rajpoot
- Department of Computer Science, University of Warwick, Coventry, CV4 7AL UK ; Department of Computer Science and Engineering, Qatar University, Doha, Qatar
| |
Collapse
|
707
|
Heath JR, Ribas A, Mischel PS. Single-cell analysis tools for drug discovery and development. Nat Rev Drug Discov 2016; 15:204-16. [PMID: 26669673 PMCID: PMC4883669 DOI: 10.1038/nrd.2015.16] [Citation(s) in RCA: 336] [Impact Index Per Article: 42.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The genetic, functional or compositional heterogeneity of healthy and diseased tissues presents major challenges in drug discovery and development. Such heterogeneity hinders the design of accurate disease models and can confound the interpretation of biomarker levels and of patient responses to specific therapies. The complex nature of virtually all tissues has motivated the development of tools for single-cell genomic, transcriptomic and multiplex proteomic analyses. Here, we review these tools and assess their advantages and limitations. Emerging applications of single cell analysis tools in drug discovery and development, particularly in the field of oncology, are discussed.
Collapse
Affiliation(s)
- James R Heath
- California Institute of Technology Division of Chemistry and Chemical Engineering, MC 127-72, 1200 East California Boulevard, Pasadena, California 91125, USA
| | - Antoni Ribas
- Department of Medicine, University of California, Los Angeles, 10833 Le Conte Avenue, Los Angeles, California 90095, USA
| | - Paul S Mischel
- Ludwig Institute for Cancer Research San Diego, Department of Pathology and Moores Cancer Center, University of California, San Diego, 9500 Gilman Drive, La Jolla, California 92093, USA
| |
Collapse
|
708
|
Li J, Zrazhevskiy P, Gao X. Eliminating Size-Associated Diffusion Constraints for Rapid On-Surface Bioassays with Nanoparticle Probes. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2016; 12:1035-1043. [PMID: 26749053 PMCID: PMC4815929 DOI: 10.1002/smll.201503101] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/13/2015] [Revised: 11/30/2015] [Indexed: 05/21/2023]
Abstract
Nanoparticle probes enable implementation of advanced on-surface assay formats, but impose often underappreciated size-associated constraints, in particular on assay kinetics and sensitivity. The present study highlights substantially slower diffusion-limited assay kinetics due to the rapid development of a nanoprobe depletion layer next to the surface, which static incubation and mixing of bulk solution employed in conventional assay setups often fail to disrupt. In contrast, cyclic solution draining and replenishing yields reaction-limited assay kinetics irrespective of the probe size. Using common surface bioassays, enzyme-linked immunosorbent assays and immunofluorescence, this study shows that this conceptually distinct approach effectively "erases" size-dependent diffusion constraints, providing a straightforward route to rapid on-surface bioassays employing bulky probes and procedures involving multiple labeling cycles, such as multicycle single-cell molecular profiling. For proof-of-concept, the study demonstrates that the assay time can be shortened from hours to minutes with the same probe concentration and, at a typical incubation time, comparable target labeling can be achieved with up to eight times lower nanoprobe concentration. The findings are expected to enable realization of novel assay formats and stimulate development of rapid on-surface bioassays with nanoparticle probes.
Collapse
Affiliation(s)
- Junwei Li
- Department of Bioengineering, University of Washington, Seattle, WA, 98195, USA
| | - Pavel Zrazhevskiy
- Department of Bioengineering, University of Washington, Seattle, WA, 98195, USA
| | - Xiaohu Gao
- Department of Bioengineering, University of Washington, Seattle, WA, 98195, USA
| |
Collapse
|
709
|
Kabatas S, Vreja IC, Saka SK, Höschen C, Kröhnert K, Opazo F, Rizzoli SO, Diederichsen U. A contamination-insensitive probe for imaging specific biomolecules by secondary ion mass spectrometry. Chem Commun (Camb) 2016. [PMID: 26195041 DOI: 10.1039/c5cc03895b] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Imaging techniques should differentiate between specific signals, from the biomolecules of interest, and non-specific signals, from the background. We present a probe containing (15)N and (14)N isotopes in approximately equal proportion, for secondary ion mass spectrometry imaging. This probe designed for a precise biomolecule analysis is insensitive to background signals.
Collapse
Affiliation(s)
- Selda Kabatas
- Institute of Organic and Biomolecular Chemistry, University of Göttingen, Tammannstr. 2, D-37077 Göttingen, Germany.
| | | | | | | | | | | | | | | |
Collapse
|
710
|
Stack EC, Foukas PG, Lee PP. Multiplexed tissue biomarker imaging. J Immunother Cancer 2016; 4:9. [PMID: 26885371 PMCID: PMC4754920 DOI: 10.1186/s40425-016-0115-3] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2016] [Accepted: 02/02/2016] [Indexed: 01/23/2023] Open
Affiliation(s)
- Edward C Stack
- Department of Life science and Technology, PerkinElmer, Hopkinton, MA USA
| | - Periklis G Foukas
- Center of Experimental Therapeutics and Ludwig Institute of Cancer Research, University Hospital of Lausanne, Lausanne, Switzerland ; Department of Pathology, University of Athens Medical School, Attikon University Hospital, Haidari, Greece
| | - Peter P Lee
- Department of Immuno-Oncology, City of Hope, Duarte, CA USA
| |
Collapse
|
711
|
Highly multiplexed simultaneous detection of RNAs and proteins in single cells. Nat Methods 2016; 13:269-75. [PMID: 26808670 PMCID: PMC4767631 DOI: 10.1038/nmeth.3742] [Citation(s) in RCA: 247] [Impact Index Per Article: 30.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2015] [Accepted: 12/16/2015] [Indexed: 12/20/2022]
Abstract
Precise gene expression measurement has been fundamental to developing an advanced understanding of the roles of biological networks in health and disease. To enable detection of expression signatures specific to individual cells we developed PLAYR (Proximity Ligation Assay for RNA). PLAYR enables highly multiplexed quantification of transcripts in single cells by flow- and mass-cytometry and is compatible with standard antibody staining of proteins. With mass cytometry, this currently enables simultaneous quantification of more than 40 different mRNAs and proteins. The technology was demonstrated in primary cells to be capable of quantifying multiple gene expression transcripts while the identity and the functional state of each analyzed cell was defined based on the expression of other transcripts or proteins. PLAYR now enables high throughput deep phenotyping of cells to readily expand beyond protein epitopes to include RNA expression, thereby opening a new venue on the characterization of cellular metabolism.
Collapse
|
712
|
Yuan J, Hegde PS, Clynes R, Foukas PG, Harari A, Kleen TO, Kvistborg P, Maccalli C, Maecker HT, Page DB, Robins H, Song W, Stack EC, Wang E, Whiteside TL, Zhao Y, Zwierzina H, Butterfield LH, Fox BA. Novel technologies and emerging biomarkers for personalized cancer immunotherapy. J Immunother Cancer 2016. [PMID: 26788324 DOI: 10.1186/s40425-016-0107-3.] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
The culmination of over a century's work to understand the role of the immune system in tumor control has led to the recent advances in cancer immunotherapies that have resulted in durable clinical responses in patients with a variety of malignancies. Cancer immunotherapies are rapidly changing traditional treatment paradigms and expanding the therapeutic landscape for cancer patients. However, despite the current success of these therapies, not all patients respond to immunotherapy and even those that do often experience toxicities. Thus, there is a growing need to identify predictive and prognostic biomarkers that enhance our understanding of the mechanisms underlying the complex interactions between the immune system and cancer. Therefore, the Society for Immunotherapy of Cancer (SITC) reconvened an Immune Biomarkers Task Force to review state of the art technologies, identify current hurdlers, and make recommendations for the field. As a product of this task force, Working Group 2 (WG2), consisting of international experts from academia and industry, assembled to identify and discuss promising technologies for biomarker discovery and validation. Thus, this WG2 consensus paper will focus on the current status of emerging biomarkers for immune checkpoint blockade therapy and discuss novel technologies as well as high dimensional data analysis platforms that will be pivotal for future biomarker research. In addition, this paper will include a brief overview of the current challenges with recommendations for future biomarker discovery.
Collapse
Affiliation(s)
- Jianda Yuan
- Memorial Sloan-Kettering Cancer Center, 1275 New York Ave Box 386, New York, NY 10065 USA
| | - Priti S Hegde
- Genentech, Inc., 1 DNA Way South, San Francisco, CA 94080 USA
| | - Raphael Clynes
- Bristol-Myers Squibb, 3551 Lawrenceville Road, Princeton, NJ 08648 USA
| | - Periklis G Foukas
- Center of Experimental Therapeutics and Ludwig Institute of Cancer Research, University Hospital of Lausanne, Rue du Bugnon 21, 1011 Lausanne, Switzerland ; Department of Pathology, University of Athens Medical School, "Attikon" University Hospital, 1st Rimini St, 12462 Haidari, Greece
| | - Alexandre Harari
- Center of Experimental Therapeutics and Ludwig Institute of Cancer Research, University Hospital of Lausanne, Rue du Bugnon 21, 1011 Lausanne, Switzerland
| | - Thomas O Kleen
- Epiontis GmbH, Rudower Chaussee 29, 12489 Berlin, Germany
| | - Pia Kvistborg
- Netherlands Cancer Institute, Postbus 90203, 1006 BE Amsterdam, Netherlands
| | - Cristina Maccalli
- Italian Network for Biotherapy of Tumors (NIBIT)-Laboratory, c/o Medical Oncology and Immunotherapy, University Hospital of Siena, V.le Bracci,16, Siena, 53100 Italy
| | - Holden T Maecker
- Stanford University Medical Center, 299 Campus Drive, Stanford, CA 94303 USA
| | - David B Page
- Earle A. Chiles Research Institute, Providence Cancer Center, 4805 NE Glisan Street, Portland, OR 97213 USA
| | - Harlan Robins
- Adaptive Technologies, Inc., 1551 Eastlake Avenue East Suite 200, Seattle, WA 98102 USA
| | - Wenru Song
- AstraZeneca, One MedImmune Way, Gaithersburg, MD 20878 USA
| | | | - Ena Wang
- Sidra Medical and Research Center, PO Box 26999, Doha, Qatar
| | - Theresa L Whiteside
- University of Pittsburgh Cancer Institute, 5117 Centre Ave, Suite 1.27, Pittsburgh, PA 15213 USA
| | - Yingdong Zhao
- National Cancer Institute, 9609 Medical Center Drive, Rockville, MD 20850 USA
| | - Heinz Zwierzina
- Innsbruck Medical University, Medizinische Klinik, Anichstrasse 35, Innsbruck, A-6020 Austria
| | - Lisa H Butterfield
- Department of Medicine, Surgery and Immunology, University of Pittsburgh Cancer Institute, 5117 Centre Avenue, Pittsburgh, PA 15213 USA
| | - Bernard A Fox
- Earle A. Chiles Research Institute, Providence Cancer Center, 4805 NE Glisan Street, Portland, OR 97213 USA
| |
Collapse
|
713
|
Yuan J, Hegde PS, Clynes R, Foukas PG, Harari A, Kleen TO, Kvistborg P, Maccalli C, Maecker HT, Page DB, Robins H, Song W, Stack EC, Wang E, Whiteside TL, Zhao Y, Zwierzina H, Butterfield LH, Fox BA. Novel technologies and emerging biomarkers for personalized cancer immunotherapy. J Immunother Cancer 2016; 4:3. [PMID: 26788324 PMCID: PMC4717548 DOI: 10.1186/s40425-016-0107-3] [Citation(s) in RCA: 155] [Impact Index Per Article: 19.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2015] [Accepted: 01/05/2016] [Indexed: 12/13/2022] Open
Abstract
The culmination of over a century’s work to understand the role of the immune system in tumor control has led to the recent advances in cancer immunotherapies that have resulted in durable clinical responses in patients with a variety of malignancies. Cancer immunotherapies are rapidly changing traditional treatment paradigms and expanding the therapeutic landscape for cancer patients. However, despite the current success of these therapies, not all patients respond to immunotherapy and even those that do often experience toxicities. Thus, there is a growing need to identify predictive and prognostic biomarkers that enhance our understanding of the mechanisms underlying the complex interactions between the immune system and cancer. Therefore, the Society for Immunotherapy of Cancer (SITC) reconvened an Immune Biomarkers Task Force to review state of the art technologies, identify current hurdlers, and make recommendations for the field. As a product of this task force, Working Group 2 (WG2), consisting of international experts from academia and industry, assembled to identify and discuss promising technologies for biomarker discovery and validation. Thus, this WG2 consensus paper will focus on the current status of emerging biomarkers for immune checkpoint blockade therapy and discuss novel technologies as well as high dimensional data analysis platforms that will be pivotal for future biomarker research. In addition, this paper will include a brief overview of the current challenges with recommendations for future biomarker discovery.
Collapse
Affiliation(s)
- Jianda Yuan
- Memorial Sloan-Kettering Cancer Center, 1275 New York Ave Box 386, New York, NY 10065 USA
| | - Priti S Hegde
- Genentech, Inc., 1 DNA Way South, San Francisco, CA 94080 USA
| | - Raphael Clynes
- Bristol-Myers Squibb, 3551 Lawrenceville Road, Princeton, NJ 08648 USA
| | - Periklis G Foukas
- Center of Experimental Therapeutics and Ludwig Institute of Cancer Research, University Hospital of Lausanne, Rue du Bugnon 21, 1011 Lausanne, Switzerland ; Department of Pathology, University of Athens Medical School, "Attikon" University Hospital, 1st Rimini St, 12462 Haidari, Greece
| | - Alexandre Harari
- Center of Experimental Therapeutics and Ludwig Institute of Cancer Research, University Hospital of Lausanne, Rue du Bugnon 21, 1011 Lausanne, Switzerland
| | - Thomas O Kleen
- Epiontis GmbH, Rudower Chaussee 29, 12489 Berlin, Germany
| | - Pia Kvistborg
- Netherlands Cancer Institute, Postbus 90203, 1006 BE Amsterdam, Netherlands
| | - Cristina Maccalli
- Italian Network for Biotherapy of Tumors (NIBIT)-Laboratory, c/o Medical Oncology and Immunotherapy, University Hospital of Siena, V.le Bracci,16, Siena, 53100 Italy
| | - Holden T Maecker
- Stanford University Medical Center, 299 Campus Drive, Stanford, CA 94303 USA
| | - David B Page
- Earle A. Chiles Research Institute, Providence Cancer Center, 4805 NE Glisan Street, Portland, OR 97213 USA
| | - Harlan Robins
- Adaptive Technologies, Inc., 1551 Eastlake Avenue East Suite 200, Seattle, WA 98102 USA
| | - Wenru Song
- AstraZeneca, One MedImmune Way, Gaithersburg, MD 20878 USA
| | | | - Ena Wang
- Sidra Medical and Research Center, PO Box 26999, Doha, Qatar
| | - Theresa L Whiteside
- University of Pittsburgh Cancer Institute, 5117 Centre Ave, Suite 1.27, Pittsburgh, PA 15213 USA
| | - Yingdong Zhao
- National Cancer Institute, 9609 Medical Center Drive, Rockville, MD 20850 USA
| | - Heinz Zwierzina
- Innsbruck Medical University, Medizinische Klinik, Anichstrasse 35, Innsbruck, A-6020 Austria
| | - Lisa H Butterfield
- Department of Medicine, Surgery and Immunology, University of Pittsburgh Cancer Institute, 5117 Centre Avenue, Pittsburgh, PA 15213 USA
| | - Bernard A Fox
- Earle A. Chiles Research Institute, Providence Cancer Center, 4805 NE Glisan Street, Portland, OR 97213 USA
| |
Collapse
|
714
|
Kurebayashi Y, Emoto K, Hayashi Y, Kamiyama I, Ohtsuka T, Asamura H, Sakamoto M. Comprehensive Immune Profiling of Lung Adenocarcinomas Reveals Four Immunosubtypes with Plasma Cell Subtype a Negative Indicator. Cancer Immunol Res 2016; 4:234-47. [PMID: 26787825 DOI: 10.1158/2326-6066.cir-15-0214] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2015] [Accepted: 12/30/2015] [Indexed: 01/15/2023]
Abstract
Neoplastic cancer cells and cancer stroma (including infiltrating immune cells) determine the biology and prognosis of cancer. Various types of adaptive and innate immune cells are known to infiltrate the cancer stroma. However, the patterns and spatial distribution of immune cell infiltration as well as its association with tumor histology remain poorly understood. To address these issues, we comprehensively analyzed the infiltrating immune cells present in lung adenocarcinoma. The principal types of both adaptive and innate infiltrating immune cells were immunohistochemically evaluated in the predominant histologic components of 111 lung adenocarcinomas. The same analysis was also carried out on 143 samples of histologic subtypes making up more than 20% of tumors. As a result, plasma cells and B cells with interfollicular distribution were almost exclusively observed in invasive histologic subtypes, while an increased number of mast cells were observed in noninvasive histologic subtypes. Cluster analysis revealed four distinct immunosubtypes (CD8, mast cell, macrophage/dendritic cell, and plasma cell subtypes) based on the infiltrating immune cell profiles. These immunosubtypes correlated with histologic subtypes, and univariate and multivariate analyses identified the plasma cell subtype as an independent negative prognostic factor. These plasma cells may be one of the major producers of the immunosuppressive cytokine IL35 in cancer stroma.
Collapse
Affiliation(s)
- Yutaka Kurebayashi
- Department of Pathology, Keio University School of Medicine, Tokyo, Japan
| | - Katsura Emoto
- Department of Pathology, Keio University School of Medicine, Tokyo, Japan. Department of Diagnostic Pathology, Saiseikai Utsunomiya Hospital, Tochigi, Japan
| | - Yuichiro Hayashi
- Department of Pathology, Keio University School of Medicine, Tokyo, Japan. Department of Diagnostic Pathology, Keio University School of Medicine, Tokyo, Japan
| | - Ikuo Kamiyama
- Department of Surgery, Division of Thoracic Surgery, Keio University School of Medicine, Tokyo, Japan
| | - Takashi Ohtsuka
- Department of Surgery, Division of Thoracic Surgery, Keio University School of Medicine, Tokyo, Japan
| | - Hisao Asamura
- Department of Surgery, Division of Thoracic Surgery, Keio University School of Medicine, Tokyo, Japan
| | - Michiie Sakamoto
- Department of Pathology, Keio University School of Medicine, Tokyo, Japan.
| |
Collapse
|
715
|
Donati G. The niche in single-cell technologies. Immunol Cell Biol 2015; 94:250-5. [PMID: 26620629 DOI: 10.1038/icb.2015.107] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2015] [Revised: 11/11/2015] [Accepted: 11/11/2015] [Indexed: 12/16/2022]
Abstract
The niche is the microenvironment in which each cell exists and is able to keep its own peculiar characteristics. The importance of the niche has been intensively studied especially in the context of stem cells, as it is responsible for both the maintenance of stemness and activation of differentiation. In the past few years, a variety of single-cell technologies have shed light on the extraordinary variability that characterizes different stem cell populations both in vitro and in vivo, but in most cases positional information is lost. Recent developments of new technologies aim to integrate both the transcriptomic profiling of cells and their spatial location. In this review I will discuss the state of the art of these technologies and the integration with others approaches that will be important in the study of stem cell populations.
Collapse
Affiliation(s)
- Giacomo Donati
- Centre for Stem Cells and Regenerative Medicine, King's College London, London, UK
| |
Collapse
|
716
|
Huang L, Tang X, Zhang W, Jiang R, Zhong H. Laser Activated Electron Tunneling Based Mass Spectrometric Imaging of Molecular Architectures of Mouse Brain Revealing Regional Specific Lipids. Anal Chem 2015; 88:732-9. [DOI: 10.1021/acs.analchem.5b02871] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Affiliation(s)
- Lulu Huang
- Mass Spectrometry Center
for Structural Identification of Biological Molecules and Precision
Medicine, Key Laboratory of Pesticides and Chemical Biology, Ministry
of Education, College of Chemistry, Central China Normal University, Wuhan, Hubei 430079, People’s Republic of China
| | - Xuemei Tang
- Mass Spectrometry Center
for Structural Identification of Biological Molecules and Precision
Medicine, Key Laboratory of Pesticides and Chemical Biology, Ministry
of Education, College of Chemistry, Central China Normal University, Wuhan, Hubei 430079, People’s Republic of China
| | - Wenyang Zhang
- Mass Spectrometry Center
for Structural Identification of Biological Molecules and Precision
Medicine, Key Laboratory of Pesticides and Chemical Biology, Ministry
of Education, College of Chemistry, Central China Normal University, Wuhan, Hubei 430079, People’s Republic of China
| | - Ruowei Jiang
- Mass Spectrometry Center
for Structural Identification of Biological Molecules and Precision
Medicine, Key Laboratory of Pesticides and Chemical Biology, Ministry
of Education, College of Chemistry, Central China Normal University, Wuhan, Hubei 430079, People’s Republic of China
| | - Hongying Zhong
- Mass Spectrometry Center
for Structural Identification of Biological Molecules and Precision
Medicine, Key Laboratory of Pesticides and Chemical Biology, Ministry
of Education, College of Chemistry, Central China Normal University, Wuhan, Hubei 430079, People’s Republic of China
| |
Collapse
|
717
|
Fredolini C, Byström S, Pin E, Edfors F, Tamburro D, Iglesias MJ, Häggmark A, Hong MG, Uhlen M, Nilsson P, Schwenk JM. Immunocapture strategies in translational proteomics. Expert Rev Proteomics 2015; 13:83-98. [PMID: 26558424 PMCID: PMC4732419 DOI: 10.1586/14789450.2016.1111141] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Aiming at clinical studies of human diseases, antibody-assisted assays have been applied to biomarker discovery and toward a streamlined translation from patient profiling to assays supporting personalized treatments. In recent years, integrated strategies to couple and combine antibodies with mass spectrometry-based proteomic efforts have emerged, allowing for novel possibilities in basic and clinical research. Described in this review are some of the field's current and emerging immunocapture approaches from an affinity proteomics perspective. Discussed are some of their advantages, pitfalls and opportunities for the next phase in clinical and translational proteomics.
Collapse
Affiliation(s)
- Claudia Fredolini
- Affinity Proteomics, SciLifeLab, School of Biotechnology, KTH - Royal Institute of Technology, Solna, Sweden
| | - Sanna Byström
- Affinity Proteomics, SciLifeLab, School of Biotechnology, KTH - Royal Institute of Technology, Solna, Sweden
| | - Elisa Pin
- Affinity Proteomics, SciLifeLab, School of Biotechnology, KTH - Royal Institute of Technology, Solna, Sweden
| | - Fredrik Edfors
- Affinity Proteomics, SciLifeLab, School of Biotechnology, KTH - Royal Institute of Technology, Solna, Sweden
| | - Davide Tamburro
- Department of Oncology-Pathology, Clinical Proteomics Mass Spectrometry, SciLifeLab, Karolinska Institutet, Solna, Sweden
| | - Maria Jesus Iglesias
- Affinity Proteomics, SciLifeLab, School of Biotechnology, KTH - Royal Institute of Technology, Solna, Sweden
| | - Anna Häggmark
- Affinity Proteomics, SciLifeLab, School of Biotechnology, KTH - Royal Institute of Technology, Solna, Sweden
| | - Mun-Gwan Hong
- Affinity Proteomics, SciLifeLab, School of Biotechnology, KTH - Royal Institute of Technology, Solna, Sweden
| | - Mathias Uhlen
- Affinity Proteomics, SciLifeLab, School of Biotechnology, KTH - Royal Institute of Technology, Solna, Sweden
| | - Peter Nilsson
- Affinity Proteomics, SciLifeLab, School of Biotechnology, KTH - Royal Institute of Technology, Solna, Sweden
| | - Jochen M Schwenk
- Affinity Proteomics, SciLifeLab, School of Biotechnology, KTH - Royal Institute of Technology, Solna, Sweden
| |
Collapse
|
718
|
Bowtell DD, Böhm S, Ahmed AA, Aspuria PJ, Bast RC, Beral V, Berek JS, Birrer MJ, Blagden S, Bookman MA, Brenton JD, Chiappinelli KB, Martins FC, Coukos G, Drapkin R, Edmondson R, Fotopoulou C, Gabra H, Galon J, Gourley C, Heong V, Huntsman DG, Iwanicki M, Karlan BY, Kaye A, Lengyel E, Levine DA, Lu KH, McNeish IA, Menon U, Narod SA, Nelson BH, Nephew KP, Pharoah P, Powell DJ, Ramos P, Romero IL, Scott CL, Sood AK, Stronach EA, Balkwill FR. Rethinking ovarian cancer II: reducing mortality from high-grade serous ovarian cancer. Nat Rev Cancer 2015; 15:668-79. [PMID: 26493647 PMCID: PMC4892184 DOI: 10.1038/nrc4019] [Citation(s) in RCA: 803] [Impact Index Per Article: 89.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
High-grade serous ovarian cancer (HGSOC) accounts for 70-80% of ovarian cancer deaths, and overall survival has not changed significantly for several decades. In this Opinion article, we outline a set of research priorities that we believe will reduce incidence and improve outcomes for women with this disease. This 'roadmap' for HGSOC was determined after extensive discussions at an Ovarian Cancer Action meeting in January 2015.
Collapse
Affiliation(s)
- David D Bowtell
- Cancer Genomics and Genetics Program, Peter MacCallum Cancer Centre, Melbourne, Victoria 8006, Australia; and the Kinghorn Cancer Centre, Garvan Institute for Medical Research, Darlinghurst, Sydney, 2010 New South Wales, Australia
| | - Steffen Böhm
- Barts Cancer Institute, Queen Mary University of London, Charterhouse Square, London EC1M6BQ, UK
| | - Ahmed A Ahmed
- Nuffield Department of Obstetrics and Gynaecology and the Weatherall Institute of Molecular Medicine, John Radcliffe Hospital, Headington, Oxford OX3 9DS, UK
| | - Paul-Joseph Aspuria
- Cedars-Sinai Medical Center, 8700 Beverly Boulevard, Los Angeles, California 90048, USA
| | - Robert C Bast
- MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, Texas 77030-4009, USA
| | - Valerie Beral
- University of Oxford, Headington, Oxford, OX3 7LF, UK
| | | | | | - Sarah Blagden
- Ovarian Cancer Action Research Centre, Imperial College London, Hammersmith Campus, London W12 0NN, UK
| | | | - James D Brenton
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Cambridge CB2 0RE, UK
| | | | - Filipe Correia Martins
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Cambridge CB2 0RE, UK
| | - George Coukos
- University Hospital of Lausanne, Lausanne, Switzerland
| | - Ronny Drapkin
- University of Pennsylvania, Penn Ovarian Cancer Research Center, Philadelphia, Pennsylvania 19104, USA
| | | | - Christina Fotopoulou
- Ovarian Cancer Action Research Centre, Imperial College London, Hammersmith Campus, London W12 0NN, UK
| | - Hani Gabra
- Ovarian Cancer Action Research Centre, Imperial College London, Hammersmith Campus, London W12 0NN, UK
| | - Jérôme Galon
- Institut National de la Santé et de la Recherche Médicale, UMRS1138, Laboratory of Integrative Cancer Immunology, Cordeliers Research Center, Université Paris Descartes, Sorbonne Paris Cité, Sorbonne Universités, UPMC Univ Paris 06, 75006 Paris, France
| | - Charlie Gourley
- Cancer Research Centre, University of Edinburgh, Edinburgh EH4 2XR, UK
| | - Valerie Heong
- Walter and Eliza Hall Institute, Parkville, Victoria 3052, Australia
| | - David G Huntsman
- University of British Columbia, Departments of Pathology and Laboratory Medicine and Obstetrics and Gynecology, Faculty of Medicine, Vancouver, British Columbia V6T 2B5, Canada
| | | | - Beth Y Karlan
- Cedars-Sinai Medical Center, 8700 Beverly Boulevard, Los Angeles, California 90048, USA
| | | | | | - Douglas A Levine
- Memorial Sloan Kettering Cancer Center, New York, New York 10065, USA
| | - Karen H Lu
- MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, Texas 77030-4009, USA
| | | | - Usha Menon
- Women's Cancer, Institute for Women's Health, University College London, London WC1E 6BT, UK
| | - Steven A Narod
- Women's College Research Institute, Toronto, Ontario M5G 1N8, Canada
| | - Brad H Nelson
- British Columbia Cancer Agency, Victoria, British Columbia V8R 6V5, Canada
| | - Kenneth P Nephew
- Indiana University School of Medicine &Simon Cancer Center, Bloomington, IN 47405-4401, USA
| | - Paul Pharoah
- University of Cambridge, Strangeways Research Laboratory, Cambridge CB1 8RN, UK
| | - Daniel J Powell
- University of Pennsylvania, Philadelphia, PA 19104-5156, USA
| | - Pilar Ramos
- Translational Genomics Research Institute (Tgen), Phoenix, Arizona 85004, USA
| | | | - Clare L Scott
- Walter and Eliza Hall Institute, Parkville, Victoria 3052, Australia
| | - Anil K Sood
- MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, Texas 77030-4009, USA
| | - Euan A Stronach
- Ovarian Cancer Action Research Centre, Imperial College London, Hammersmith Campus, London W12 0NN, UK
| | - Frances R Balkwill
- Barts Cancer Institute, Queen Mary University of London, Charterhouse Square, London EC1M6BQ, UK
| |
Collapse
|
719
|
Wirtz T, Philipp P, Audinot JN, Dowsett D, Eswara S. High-resolution high-sensitivity elemental imaging by secondary ion mass spectrometry: from traditional 2D and 3D imaging to correlative microscopy. NANOTECHNOLOGY 2015; 26:434001. [PMID: 26436905 DOI: 10.1088/0957-4484/26/43/434001] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/26/2023]
Abstract
Secondary ion mass spectrometry (SIMS) constitutes an extremely sensitive technique for imaging surfaces in 2D and 3D. Apart from its excellent sensitivity and high lateral resolution (50 nm on state-of-the-art SIMS instruments), advantages of SIMS include high dynamic range and the ability to differentiate between isotopes. This paper first reviews the underlying principles of SIMS as well as the performance and applications of 2D and 3D SIMS elemental imaging. The prospects for further improving the capabilities of SIMS imaging are discussed. The lateral resolution in SIMS imaging when using the microprobe mode is limited by (i) the ion probe size, which is dependent on the brightness of the primary ion source, the quality of the optics of the primary ion column and the electric fields in the near sample region used to extract secondary ions; (ii) the sensitivity of the analysis as a reasonable secondary ion signal, which must be detected from very tiny voxel sizes and thus from a very limited number of sputtered atoms; and (iii) the physical dimensions of the collision cascade determining the origin of the sputtered ions with respect to the impact site of the incident primary ion probe. One interesting prospect is the use of SIMS-based correlative microscopy. In this approach SIMS is combined with various high-resolution microscopy techniques, so that elemental/chemical information at the highest sensitivity can be obtained with SIMS, while excellent spatial resolution is provided by overlaying the SIMS images with high-resolution images obtained by these microscopy techniques. Examples of this approach are given by presenting in situ combinations of SIMS with transmission electron microscopy (TEM), helium ion microscopy (HIM) and scanning probe microscopy (SPM).
Collapse
Affiliation(s)
- T Wirtz
- Advanced Instrumentation for Ion Nano-Analytics (AINA), MRT Department, Luxembourg Institute of Science and Technology (LIST), 41 rue du Brill, L-4422 Belvaux, Luxembourg
| | | | | | | | | |
Collapse
|
720
|
Simmons AJ, Banerjee A, McKinley ET, Scurrah CR, Herring CA, Gewin LS, Masuzaki R, Karp SJ, Franklin JL, Gerdes MJ, Irish JM, Coffey RJ, Lau KS. Cytometry-based single-cell analysis of intact epithelial signaling reveals MAPK activation divergent from TNF-α-induced apoptosis in vivo. Mol Syst Biol 2015; 11:835. [PMID: 26519361 PMCID: PMC4631206 DOI: 10.15252/msb.20156282] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Understanding heterogeneous cellular behaviors in a complex tissue requires the evaluation of signaling networks at single-cell resolution. However, probing signaling in epithelial tissues using cytometry-based single-cell analysis has been confounded by the necessity of single-cell dissociation, where disrupting cell-to-cell connections inherently perturbs native cell signaling states. Here, we demonstrate a novel strategy (Disaggregation for Intracellular Signaling in Single Epithelial Cells from Tissue-DISSECT) that preserves native signaling for Cytometry Time-of-Flight (CyTOF) and fluorescent flow cytometry applications. A 21-plex CyTOF analysis encompassing core signaling and cell-identity markers was performed on the small intestinal epithelium after systemic tumor necrosis factor-alpha (TNF-α) stimulation. Unsupervised and supervised analyses robustly selected signaling features that identify a unique subset of epithelial cells that are sensitized to TNF-α-induced apoptosis in the seemingly homogeneous enterocyte population. Specifically, p-ERK and apoptosis are divergently regulated in neighboring enterocytes within the epithelium, suggesting a mechanism of contact-dependent survival. Our novel single-cell approach can broadly be applied, using both CyTOF and multi-parameter flow cytometry, for investigating normal and diseased cell states in a wide range of epithelial tissues.
Collapse
Affiliation(s)
- Alan J Simmons
- Epithelial Biology Center, Vanderbilt University Medical Center, Nashville, TN, USA Department of Cell and Developmental Biology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Amrita Banerjee
- Epithelial Biology Center, Vanderbilt University Medical Center, Nashville, TN, USA Department of Cell and Developmental Biology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Eliot T McKinley
- Epithelial Biology Center, Vanderbilt University Medical Center, Nashville, TN, USA Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Cherie' R Scurrah
- Epithelial Biology Center, Vanderbilt University Medical Center, Nashville, TN, USA Department of Cell and Developmental Biology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Charles A Herring
- Epithelial Biology Center, Vanderbilt University Medical Center, Nashville, TN, USA Department of Chemical and Physical Biology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Leslie S Gewin
- Department of Cell and Developmental Biology, Vanderbilt University Medical Center, Nashville, TN, USA Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA Veterans Affairs Medical Center, Tennessee Valley Healthcare System, Nashville, TN, USA
| | - Ryota Masuzaki
- The Transplant Center and Department of Surgery, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Seth J Karp
- The Transplant Center and Department of Surgery, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Jeffrey L Franklin
- Epithelial Biology Center, Vanderbilt University Medical Center, Nashville, TN, USA Department of Cell and Developmental Biology, Vanderbilt University Medical Center, Nashville, TN, USA
| | | | - Jonathan M Irish
- Departments of Cancer Biology, and Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Robert J Coffey
- Epithelial Biology Center, Vanderbilt University Medical Center, Nashville, TN, USA Department of Cell and Developmental Biology, Vanderbilt University Medical Center, Nashville, TN, USA Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA Veterans Affairs Medical Center, Tennessee Valley Healthcare System, Nashville, TN, USA
| | - Ken S Lau
- Epithelial Biology Center, Vanderbilt University Medical Center, Nashville, TN, USA Department of Cell and Developmental Biology, Vanderbilt University Medical Center, Nashville, TN, USA Department of Chemical and Physical Biology, Vanderbilt University Medical Center, Nashville, TN, USA
| |
Collapse
|
721
|
Rethinking ovarian cancer II: reducing mortality from high-grade serous ovarian cancer. NATURE REVIEWS. CANCER 2015. [PMID: 26493647 DOI: 10.1038/nrc4019]+[] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
High-grade serous ovarian cancer (HGSOC) accounts for 70-80% of ovarian cancer deaths, and overall survival has not changed significantly for several decades. In this Opinion article, we outline a set of research priorities that we believe will reduce incidence and improve outcomes for women with this disease. This 'roadmap' for HGSOC was determined after extensive discussions at an Ovarian Cancer Action meeting in January 2015.
Collapse
|
722
|
Bowtell DD, Böhm S, Ahmed AA, Aspuria PJ, Bast RC, Beral V, Berek JS, Birrer MJ, Blagden S, Bookman MA, Brenton JD, Chiappinelli KB, Martins FC, Coukos G, Drapkin R, Edmondson R, Fotopoulou C, Gabra H, Galon J, Gourley C, Heong V, Huntsman DG, Iwanicki M, Karlan BY, Kaye A, Lengyel E, Levine DA, Lu KH, McNeish IA, Menon U, Narod SA, Nelson BH, Nephew KP, Pharoah P, Powell DJ, Ramos P, Romero IL, Scott CL, Sood AK, Stronach EA, Balkwill FR. Rethinking ovarian cancer II: reducing mortality from high-grade serous ovarian cancer. NATURE REVIEWS. CANCER 2015. [PMID: 26493647 DOI: 10.1038/nrc4019] [] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
High-grade serous ovarian cancer (HGSOC) accounts for 70-80% of ovarian cancer deaths, and overall survival has not changed significantly for several decades. In this Opinion article, we outline a set of research priorities that we believe will reduce incidence and improve outcomes for women with this disease. This 'roadmap' for HGSOC was determined after extensive discussions at an Ovarian Cancer Action meeting in January 2015.
Collapse
Affiliation(s)
- David D Bowtell
- Cancer Genomics and Genetics Program, Peter MacCallum Cancer Centre, Melbourne, Victoria 8006, Australia; and the Kinghorn Cancer Centre, Garvan Institute for Medical Research, Darlinghurst, Sydney, 2010 New South Wales, Australia
| | - Steffen Böhm
- Barts Cancer Institute, Queen Mary University of London, Charterhouse Square, London EC1M6BQ, UK
| | - Ahmed A Ahmed
- Nuffield Department of Obstetrics and Gynaecology and the Weatherall Institute of Molecular Medicine, John Radcliffe Hospital, Headington, Oxford OX3 9DS, UK
| | - Paul-Joseph Aspuria
- Cedars-Sinai Medical Center, 8700 Beverly Boulevard, Los Angeles, California 90048, USA
| | - Robert C Bast
- MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, Texas 77030-4009, USA
| | - Valerie Beral
- University of Oxford, Headington, Oxford, OX3 7LF, UK
| | | | | | - Sarah Blagden
- Ovarian Cancer Action Research Centre, Imperial College London, Hammersmith Campus, London W12 0NN, UK
| | | | - James D Brenton
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Cambridge CB2 0RE, UK
| | | | - Filipe Correia Martins
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Cambridge CB2 0RE, UK
| | - George Coukos
- University Hospital of Lausanne, Lausanne, Switzerland
| | - Ronny Drapkin
- University of Pennsylvania, Penn Ovarian Cancer Research Center, Philadelphia, Pennsylvania 19104, USA
| | | | - Christina Fotopoulou
- Ovarian Cancer Action Research Centre, Imperial College London, Hammersmith Campus, London W12 0NN, UK
| | - Hani Gabra
- Ovarian Cancer Action Research Centre, Imperial College London, Hammersmith Campus, London W12 0NN, UK
| | - Jérôme Galon
- Institut National de la Santé et de la Recherche Médicale, UMRS1138, Laboratory of Integrative Cancer Immunology, Cordeliers Research Center, Université Paris Descartes, Sorbonne Paris Cité, Sorbonne Universités, UPMC Univ Paris 06, 75006 Paris, France
| | - Charlie Gourley
- Cancer Research Centre, University of Edinburgh, Edinburgh EH4 2XR, UK
| | - Valerie Heong
- Walter and Eliza Hall Institute, Parkville, Victoria 3052, Australia
| | - David G Huntsman
- University of British Columbia, Departments of Pathology and Laboratory Medicine and Obstetrics and Gynecology, Faculty of Medicine, Vancouver, British Columbia V6T 2B5, Canada
| | | | - Beth Y Karlan
- Cedars-Sinai Medical Center, 8700 Beverly Boulevard, Los Angeles, California 90048, USA
| | | | | | - Douglas A Levine
- Memorial Sloan Kettering Cancer Center, New York, New York 10065, USA
| | - Karen H Lu
- MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, Texas 77030-4009, USA
| | | | - Usha Menon
- Women's Cancer, Institute for Women's Health, University College London, London WC1E 6BT, UK
| | - Steven A Narod
- Women's College Research Institute, Toronto, Ontario M5G 1N8, Canada
| | - Brad H Nelson
- British Columbia Cancer Agency, Victoria, British Columbia V8R 6V5, Canada
| | - Kenneth P Nephew
- Indiana University School of Medicine &Simon Cancer Center, Bloomington, IN 47405-4401, USA
| | - Paul Pharoah
- University of Cambridge, Strangeways Research Laboratory, Cambridge CB1 8RN, UK
| | - Daniel J Powell
- University of Pennsylvania, Philadelphia, PA 19104-5156, USA
| | - Pilar Ramos
- Translational Genomics Research Institute (Tgen), Phoenix, Arizona 85004, USA
| | | | - Clare L Scott
- Walter and Eliza Hall Institute, Parkville, Victoria 3052, Australia
| | - Anil K Sood
- MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, Texas 77030-4009, USA
| | - Euan A Stronach
- Ovarian Cancer Action Research Centre, Imperial College London, Hammersmith Campus, London W12 0NN, UK
| | - Frances R Balkwill
- Barts Cancer Institute, Queen Mary University of London, Charterhouse Square, London EC1M6BQ, UK
| |
Collapse
|
723
|
Ménoret A, Crocker SJ, Rodriguez A, Rathinam VA, Clark RB, Vella AT. Transition from identity to bioactivity-guided proteomics for biomarker discovery with focus on the PF2D platform. Proteomics Clin Appl 2015. [PMID: 26201056 DOI: 10.1002/prca.201500029] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Proteomic strategies provide a valuable tool kit to identify proteins involved in diseases. With recent progress in MS technology, high throughput proteomics has accelerated protein identification for potential biomarkers. Numerous biomarker candidates have been identified in several diseases, and many are common among pathologies. An overall strategy that could complement and strengthen the search for biomarkers is combining protein identity with biological outcomes. This review describes an emerging framework of bridging bioactivity to protein identity, exploring the possibility that some biomarkers will have a mechanistic role in the disease process. A review of pulmonary, cardiovascular, and CNS biomarkers will be discussed to demonstrate the utility of combining bioactivity with identification as a means to not only find meaningful biomarkers, but also to uncover functional mediators of disease.
Collapse
Affiliation(s)
- Antoine Ménoret
- Department of Immunology, University of Connecticut Health Center, Farmington, CT, USA
| | - Stephen J Crocker
- Department of Neuroscience, University of Connecticut Health Center, Farmington, CT, USA
| | - Annabelle Rodriguez
- Department of Cell Biology, University of Connecticut Health Center, Farmington, CT, USA
| | - Vijay A Rathinam
- Department of Immunology, University of Connecticut Health Center, Farmington, CT, USA
| | - Robert B Clark
- Department of Immunology, University of Connecticut Health Center, Farmington, CT, USA
| | - Anthony T Vella
- Department of Immunology, University of Connecticut Health Center, Farmington, CT, USA
| |
Collapse
|
724
|
|
725
|
|
726
|
Torlakovic EE, Nielsen S, Vyberg M, Taylor CR. Getting controls under control: the time is now for immunohistochemistry. J Clin Pathol 2015; 68:879-82. [PMID: 26286753 PMCID: PMC4680144 DOI: 10.1136/jclinpath-2014-202705] [Citation(s) in RCA: 56] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2015] [Accepted: 07/27/2015] [Indexed: 12/02/2022]
Abstract
For several decades, immunohistochemistry (IHC), more specifically diagnostic IHC (dIHC), has been considered an art rather than a laboratory test. There was no clarity about what test performance characteristics are relevant to dIHC, test performance characteristics were not fully defined for dIHC and partly as a consequence of that, there were no standardised controls or reference standards. Herein, we discuss the role of standardisation of external controls for test performance characteristics and the role of standardised controls and reference standards for overall standardisation of IHC.
Collapse
Affiliation(s)
- Emina Emilia Torlakovic
- Department of Laboratory Hematology, Toronto General Hospital, University Health Network, Toronto, Ontario, Canada Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada Canadian Immunohistochemistry Quality Control (CIQC), Vancouver, BC, Canada
| | - Søren Nielsen
- Department of Clinical Medicine, Institute of Pathology, Aalborg University Hospital, Aalborg, Denmark Nordic Immunohistochemical Quality Control (NordiQC), Aalborg, Denmark
| | - Mogens Vyberg
- Department of Clinical Medicine, Institute of Pathology, Aalborg University Hospital, Aalborg, Denmark Nordic Immunohistochemical Quality Control (NordiQC), Aalborg, Denmark
| | - Clive R Taylor
- Department of Pathology, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| |
Collapse
|
727
|
De Vlieghere E, Verset L, Demetter P, Bracke M, De Wever O. Cancer-associated fibroblasts as target and tool in cancer therapeutics and diagnostics. Virchows Arch 2015; 467:367-82. [PMID: 26259962 DOI: 10.1007/s00428-015-1818-4] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2015] [Revised: 07/21/2015] [Accepted: 07/27/2015] [Indexed: 12/11/2022]
Abstract
Cancer-associated fibroblasts (CAFs) are drivers of tumour progression and are considered as a target and a tool in cancer diagnostic and therapeutic applications. An increased abundance of CAFs or CAF signatures are recognized as a bad prognostic marker in several cancer types. Tumour-environment biomimetics strongly improve our understanding of the communication between CAFs, cancer cells and other host cells. Several experimental drugs targeting CAFs are in clinical trials for multiple tumour entities; alternatively, CAFs can be exploited as a tool to characterize the functionality of circulating tumour cells or to capture them as a tool to prevent metastasis. The continuous interaction between tissue engineers, biomaterial experts and cancer researchers creates the possibility to biomimic the tumour-environment and provides new opportunities in cancer diagnostics and management.
Collapse
Affiliation(s)
- Elly De Vlieghere
- Laboratory of Experimental Cancer Research, Department of Radiation Oncology and Experimental Cancer Research, Ghent University Hospital, De Pintelaan 185, 9000, Ghent, Belgium
| | - Laurine Verset
- Departments of Pathology, Erasme University Hospital, Université Libre de Bruxelles, Brussels, Belgium
| | - Pieter Demetter
- Departments of Pathology, Erasme University Hospital, Université Libre de Bruxelles, Brussels, Belgium
| | - Marc Bracke
- Laboratory of Experimental Cancer Research, Department of Radiation Oncology and Experimental Cancer Research, Ghent University Hospital, De Pintelaan 185, 9000, Ghent, Belgium
| | - Olivier De Wever
- Laboratory of Experimental Cancer Research, Department of Radiation Oncology and Experimental Cancer Research, Ghent University Hospital, De Pintelaan 185, 9000, Ghent, Belgium.
| |
Collapse
|
728
|
Computational and experimental single cell biology techniques for the definition of cell type heterogeneity, interplay and intracellular dynamics. Curr Opin Biotechnol 2015; 34:9-15. [DOI: 10.1016/j.copbio.2014.10.010] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2014] [Revised: 10/21/2014] [Accepted: 10/22/2014] [Indexed: 12/31/2022]
|
729
|
Alizadeh AA, Aranda V, Bardelli A, Blanpain C, Bock C, Borowski C, Caldas C, Califano A, Doherty M, Elsner M, Esteller M, Fitzgerald R, Korbel JO, Lichter P, Mason CE, Navin N, Pe'er D, Polyak K, Roberts CWM, Siu L, Snyder A, Stower H, Swanton C, Verhaak RGW, Zenklusen JC, Zuber J, Zucman-Rossi J. Toward understanding and exploiting tumor heterogeneity. Nat Med 2015; 21:846-53. [PMID: 26248267 PMCID: PMC4785013 DOI: 10.1038/nm.3915] [Citation(s) in RCA: 516] [Impact Index Per Article: 57.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2015] [Accepted: 07/01/2015] [Indexed: 11/09/2022]
Abstract
The extent of tumor heterogeneity is an emerging theme that researchers are only beginning to understand. How genetic and epigenetic heterogeneity affects tumor evolution and clinical progression is unknown. The precise nature of the environmental factors that influence this heterogeneity is also yet to be characterized. Nature Medicine, Nature Biotechnology and the Volkswagen Foundation organized a meeting focused on identifying the obstacles that need to be overcome to advance translational research in and tumor heterogeneity. Once these key questions were established, the attendees devised potential solutions. Their ideas are presented here.
Collapse
Affiliation(s)
- Ash A Alizadeh
- 1] Division of Oncology, Department of Medicine, Stanford University School of Medicine, Stanford, California, USA. [2] Division of Hematology, Department of Medicine, Stanford University School of Medicine, Stanford, California, USA. [3] Cancer Institute, Department of Medicine, Stanford University School of Medicine, Stanford, California, USA
| | | | - Alberto Bardelli
- 1] Department of Oncology, University of Torino, Candiolo, Torino, Italy. [2] Candiolo Cancer Institute-Fondazione del Piemonte per l'Oncologia (FPO), IRCCS, Candiolo, Torino, Italy
| | | | - Christoph Bock
- 1] CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria. [2] Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| | | | - Carlos Caldas
- Department of Oncology, University of Cambridge, Cambridge, UK
| | - Andrea Califano
- 1] Department of Systems Biology, Columbia University, New York, New York, USA. [2] Department of Biochemistry and Molecular Biophysics, Columbia University, New York, New York, USA. [3] Department of Biomedical Informatics, Columbia University, New York, New York, USA
| | | | | | - Manel Esteller
- Cancer Epigenetics and Biology Program, Bellvitge Biomedical Research Institute, Barcelona, Catalonia, Spain
| | - Rebecca Fitzgerald
- MRC Cancer Unit, Hutchison-MRC Research Centre, University of Cambridge, Cambridge, UK
| | - Jan O Korbel
- Genome Biology Unit, European Molecular Biology Laboratory, Heidelberg, Germany
| | - Peter Lichter
- German Cancer Research Center, DKFZ, Heidelberg, Germany
| | | | - Nicholas Navin
- 1] Department of Genetics, MD Anderson Cancer Center, Houston, Texas, USA. [2] Department of Bioinformatics and Computational Biology, MD Anderson Cancer Center, Houston, Texas, USA
| | - Dana Pe'er
- 1] Department of Systems Biology, Columbia University, New York, New York, USA. [2] Department of Biological Sciences, Columbia University, New York, New York, USA
| | - Kornelia Polyak
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA
| | - Charles W M Roberts
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA
| | - Lillian Siu
- Princess Margaret Cancer Centre, Toronto, Ontario, Canada
| | - Alexandra Snyder
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | | | - Charles Swanton
- 1] University College London Cancer Institute, London, UK. [2] University College London Hospitals NHS Foundation Trust, London, UK. [3] The Francis Crick Institute, London, UK
| | - Roel G W Verhaak
- 1] Department of Bioinformatics and Computational Biology, MD Anderson Cancer Center, Houston, Texas, USA. [2] Department of Genomic Medicine, MD Anderson Cancer Center, Houston, Texas, USA
| | - Jean C Zenklusen
- The Cancer Genome Atlas, Center for Cancer Genomics, National Cancer Institute, Bethesda, Maryland, USA
| | - Johannes Zuber
- Research Institute of Molecular Pathology (IMP), Vienna Biocenter (VBC), Vienna, Austria
| | - Jessica Zucman-Rossi
- Inserm, UMR-1162, Génomique fonctionnelle des tumeurs solides, Institut Universitaire d'Hématologie (IUH), Paris, France
| |
Collapse
|
730
|
Paul B, Hare DJ, Bishop DP, Paton C, Nguyen VT, Cole N, Niedwiecki MM, Andreozzi E, Vais A, Billings JL, Bray L, Bush AI, McColl G, Roberts BR, Adlard PA, Finkelstein DI, Hellstrom J, Hergt JM, Woodhead JD, Doble PA. Visualising mouse neuroanatomy and function by metal distribution using laser ablation-inductively coupled plasma-mass spectrometry imaging. Chem Sci 2015; 6:5383-5393. [PMID: 29449912 PMCID: PMC5669312 DOI: 10.1039/c5sc02231b] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2015] [Accepted: 07/27/2015] [Indexed: 11/23/2022] Open
Abstract
Studying the neuroanatomy of the mouse brain using imaging mass spectrometry and chemometric analysis.
Metals have a number of important roles within the brain. We used laser ablation-inductively coupled plasma-mass spectrometry (LA-ICP-MS) to map the three-dimensional concentrations and distributions of transition metals, in particular iron (Fe), copper (Cu) and zinc (Zn) within the murine brain. LA-ICP-MS is one of the leading analytical tools for measuring metals in tissue samples. Here, we present a complete data reduction protocol for measuring metals in biological samples, including the application of a pyramidal voxel registration technique to reproducibly align tissue sections. We used gold (Au) nanoparticle and ytterbium (Yb)-tagged tyrosine hydroxylase antibodies to assess the co-localisation of Fe and dopamine throughout the entire mouse brain. We also examined the natural clustering of metal concentrations within the murine brain to elucidate areas of similar composition. This clustering technique uses a mathematical approach to identify multiple ‘elemental clusters’, avoiding user bias and showing that metal composition follows a hierarchical organisation of neuroanatomical structures. This work provides new insight into the distinct compartmentalisation of metals in the brain, and presents new avenues of exploration with regard to region-specific, metal-associated neurodegeneration observed in several chronic neurodegenerative diseases.
Collapse
Affiliation(s)
- Bence Paul
- School of Earth Sciences , The University of Melbourne , Parkville , Victoria 3052 , Australia.,The Florey Institute of Neuroscience and Mental Health , The University of Melbourne , Parkville , 3052 , Victoria , Australia
| | - Dominic J Hare
- The Florey Institute of Neuroscience and Mental Health , The University of Melbourne , Parkville , 3052 , Victoria , Australia.,Elemental Bio-imaging Facility , University of Technology Sydney , Broadway , 2007 , New South Wales , Australia . ; ; Tel: +61 2 9512 1792.,Senator Frank R. Lautenberg Environmental Health Sciences Laboratory , Department of Preventive Medicine , Icahn School of Medicine at Mount Sinai , New York , 10029 , New York , USA
| | - David P Bishop
- Elemental Bio-imaging Facility , University of Technology Sydney , Broadway , 2007 , New South Wales , Australia . ; ; Tel: +61 2 9512 1792
| | - Chad Paton
- Centre for Star and Planet Formation , Geological Museum , University of Copenhagen , Øster Voldgade 5-7 , DK-1350 Copenhagen , Denmark
| | - Van Tran Nguyen
- Elemental Bio-imaging Facility , University of Technology Sydney , Broadway , 2007 , New South Wales , Australia . ; ; Tel: +61 2 9512 1792
| | - Nerida Cole
- Elemental Bio-imaging Facility , University of Technology Sydney , Broadway , 2007 , New South Wales , Australia . ; ; Tel: +61 2 9512 1792
| | - Megan M Niedwiecki
- Senator Frank R. Lautenberg Environmental Health Sciences Laboratory , Department of Preventive Medicine , Icahn School of Medicine at Mount Sinai , New York , 10029 , New York , USA
| | - Erica Andreozzi
- The Florey Institute of Neuroscience and Mental Health , The University of Melbourne , Parkville , 3052 , Victoria , Australia
| | - Angela Vais
- The Florey Institute of Neuroscience and Mental Health , The University of Melbourne , Parkville , 3052 , Victoria , Australia
| | - Jessica L Billings
- The Florey Institute of Neuroscience and Mental Health , The University of Melbourne , Parkville , 3052 , Victoria , Australia
| | - Lisa Bray
- The Florey Institute of Neuroscience and Mental Health , The University of Melbourne , Parkville , 3052 , Victoria , Australia
| | - Ashley I Bush
- The Florey Institute of Neuroscience and Mental Health , The University of Melbourne , Parkville , 3052 , Victoria , Australia
| | - Gawain McColl
- The Florey Institute of Neuroscience and Mental Health , The University of Melbourne , Parkville , 3052 , Victoria , Australia
| | - Blaine R Roberts
- The Florey Institute of Neuroscience and Mental Health , The University of Melbourne , Parkville , 3052 , Victoria , Australia
| | - Paul A Adlard
- The Florey Institute of Neuroscience and Mental Health , The University of Melbourne , Parkville , 3052 , Victoria , Australia
| | - David I Finkelstein
- The Florey Institute of Neuroscience and Mental Health , The University of Melbourne , Parkville , 3052 , Victoria , Australia
| | - John Hellstrom
- School of Earth Sciences , The University of Melbourne , Parkville , Victoria 3052 , Australia
| | - Janet M Hergt
- School of Earth Sciences , The University of Melbourne , Parkville , Victoria 3052 , Australia
| | - Jon D Woodhead
- School of Earth Sciences , The University of Melbourne , Parkville , Victoria 3052 , Australia
| | - Philip A Doble
- Elemental Bio-imaging Facility , University of Technology Sydney , Broadway , 2007 , New South Wales , Australia . ; ; Tel: +61 2 9512 1792
| |
Collapse
|
731
|
Montgomery RR, Shaw AC. Paradoxical changes in innate immunity in aging: recent progress and new directions. J Leukoc Biol 2015; 98:937-43. [PMID: 26188078 DOI: 10.1189/jlb.5mr0315-104r] [Citation(s) in RCA: 94] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2015] [Accepted: 06/23/2015] [Indexed: 12/29/2022] Open
Abstract
Immunosenescence, describing alterations, including decline of immune responses with age, is comprised of inappropriate elevations, decreases, and dysregulated immune responses, leading to more severe consequences of bacterial and viral infections and reduced responses to vaccination. In adaptive immunity, these changes include increased proportions of antigen-experienced B and T cells at the cost of naïve cell populations. Innate immune changes in aging are complex in spanning multiple cell types, activation states, and tissue context. Innate immune responses are dampened in aging, yet there is also a paradoxical increase in certain signaling pathways and cytokine levels. Here, we review recent progress and highlight novel directions for expected advances that can lead the aging field to a new era of discovery that will embrace the complexity of aging in human populations.
Collapse
Affiliation(s)
- Ruth R Montgomery
- Sections of *Rheumatology and Infectious Diseases, Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Albert C Shaw
- Sections of *Rheumatology and Infectious Diseases, Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| |
Collapse
|
732
|
Automated Analysis and Classification of Histological Tissue Features by Multi-Dimensional Microscopic Molecular Profiling. PLoS One 2015; 10:e0128975. [PMID: 26176839 PMCID: PMC4503351 DOI: 10.1371/journal.pone.0128975] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2014] [Accepted: 05/01/2015] [Indexed: 01/02/2023] Open
Abstract
Characterization of the molecular attributes and spatial arrangements of cells and features within complex human tissues provides a critical basis for understanding processes involved in development and disease. Moreover, the ability to automate steps in the analysis and interpretation of histological images that currently require manual inspection by pathologists could revolutionize medical diagnostics. Toward this end, we developed a new imaging approach called multidimensional microscopic molecular profiling (MMMP) that can measure several independent molecular properties in situ at subcellular resolution for the same tissue specimen. MMMP involves repeated cycles of antibody or histochemical staining, imaging, and signal removal, which ultimately can generate information analogous to a multidimensional flow cytometry analysis on intact tissue sections. We performed a MMMP analysis on a tissue microarray containing a diverse set of 102 human tissues using a panel of 15 informative antibody and 5 histochemical stains plus DAPI. Large-scale unsupervised analysis of MMMP data, and visualization of the resulting classifications, identified molecular profiles that were associated with functional tissue features. We then directly annotated H&E images from this MMMP series such that canonical histological features of interest (e.g. blood vessels, epithelium, red blood cells) were individually labeled. By integrating image annotation data, we identified molecular signatures that were associated with specific histological annotations and we developed statistical models for automatically classifying these features. The classification accuracy for automated histology labeling was objectively evaluated using a cross-validation strategy, and significant accuracy (with a median per-pixel rate of 77% per feature from 15 annotated samples) for de novo feature prediction was obtained. These results suggest that high-dimensional profiling may advance the development of computer-based systems for automatically parsing relevant histological and cellular features from molecular imaging data of arbitrary human tissue samples, and can provide a framework and resource to spur the optimization of these technologies.
Collapse
|
733
|
Mori H, Soonsawad P, Schuetter L, Chen Q, Hubbard NE, Cardiff RD, Borowsky AD. Introduction of Zinc-salt Fixation for Effective Detection of Immune Cell-related Markers by Immunohistochemistry. Toxicol Pathol 2015; 43:883-9. [PMID: 26157038 DOI: 10.1177/0192623315587593] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Tissue localization of immune cells is critical to the study of disease processes in mouse models of human diseases. However, immunohistochemistry (IHC) for immune cell phenotyping in mouse tissue sections presents specific technical challenges. For example, CD4 and CD8 have been difficult to detect using IHC on formalin-fixed and paraffin-embedded mouse tissue, prompting alternative methods. We investigated the use of formalin-free zinc-salt fixation (ZN) and optimized IHC protocols for detecting a panel of immune cell-related markers (CD3, CD4, CD8, Foxp3, B220, F4/80, CD68, and major histocompatibility complex [MHC] class-I, MHC class-II, and Gr-1). The IHC results for these markers were compared on mouse spleen tissue treated with neutral buffered formalin (NBF) or ZN with or ZN without antigen retrieval (AR). Whereas CD4 and CD8 were not detected in NBF-treated tissue, all markers were detected in ZN-treated tissue without AR. Thus, the use of ZN treatment for IHC staining can be a good tool for studying immunoreactive lesions in tissues.
Collapse
Affiliation(s)
- Hidetoshi Mori
- Center of Comparative Medicine, University of California, Davis, California, USA
| | - Pan Soonsawad
- Center of Comparative Medicine, University of California, Davis, California, USA
| | - Louis Schuetter
- Center of Comparative Medicine, University of California, Davis, California, USA
| | - Qian Chen
- Center of Comparative Medicine, University of California, Davis, California, USA
| | - Neil E Hubbard
- Center of Comparative Medicine, University of California, Davis, California, USA
| | - Robert D Cardiff
- Center of Comparative Medicine, University of California, Davis, California, USA Department of Pathology and Laboratory Medicine, School of Medicine, University of California, Davis, California, USA
| | - Alexander D Borowsky
- Center of Comparative Medicine, University of California, Davis, California, USA Department of Pathology and Laboratory Medicine, School of Medicine, University of California, Davis, California, USA
| |
Collapse
|
734
|
Schüffler PJ, Schapiro D, Giesen C, Wang HAO, Bodenmiller B, Buhmann JM. Automatic single cell segmentation on highly multiplexed tissue images. Cytometry A 2015; 87:936-42. [DOI: 10.1002/cyto.a.22702] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2014] [Revised: 04/14/2015] [Accepted: 05/14/2015] [Indexed: 11/12/2022]
Affiliation(s)
- Peter J. Schüffler
- Department of Computer Science; ETH Zurich; Zurich 8092 Switzerland
- Life Science Zurich Graduate School, ETH Zurich and University of Zurich; Zurich Switzerland
| | - Denis Schapiro
- Life Science Zurich Graduate School, ETH Zurich and University of Zurich; Zurich Switzerland
- Institute of Molecular Life Sciences, University of Zurich; Zurich 8057 Switzerland
| | - Charlotte Giesen
- Institute of Molecular Life Sciences, University of Zurich; Zurich 8057 Switzerland
| | - Hao A. O. Wang
- Department of Chemistry; ETH Zurich; Zurich 8093 Switzerland
| | - Bernd Bodenmiller
- Institute of Molecular Life Sciences, University of Zurich; Zurich 8057 Switzerland
| | | |
Collapse
|
735
|
Oh EY, Christensen SM, Ghanta S, Jeong JC, Bucur O, Glass B, Montaser-Kouhsari L, Knoblauch NW, Bertos N, Saleh SM, Haibe-Kains B, Park M, Beck AH. Extensive rewiring of epithelial-stromal co-expression networks in breast cancer. Genome Biol 2015; 16:128. [PMID: 26087699 PMCID: PMC4471934 DOI: 10.1186/s13059-015-0675-4] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2015] [Accepted: 05/13/2015] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Epithelial-stromal crosstalk plays a critical role in invasive breast cancer pathogenesis; however, little is known on a systems level about how epithelial-stromal interactions evolve during carcinogenesis. RESULTS We develop a framework for building genome-wide epithelial-stromal co-expression networks composed of pairwise co-expression relationships between mRNA levels of genes expressed in the epithelium and stroma across a population of patients. We apply this method to laser capture micro-dissection expression profiling datasets in the setting of breast carcinogenesis. Our analysis shows that epithelial-stromal co-expression networks undergo extensive rewiring during carcinogenesis, with the emergence of distinct network hubs in normal breast, and estrogen receptor-positive and estrogen receptor-negative invasive breast cancer, and the emergence of distinct patterns of functional network enrichment. In contrast to normal breast, the strongest epithelial-stromal co-expression relationships in invasive breast cancer mostly represent self-loops, in which the same gene is co-expressed in epithelial and stromal regions. We validate this observation using an independent laser capture micro-dissection dataset and confirm that self-loop interactions are significantly increased in cancer by performing computational image analysis of epithelial and stromal protein expression using images from the Human Protein Atlas. CONCLUSIONS Epithelial-stromal co-expression network analysis represents a new approach for systems-level analyses of spatially localized transcriptomic data. The analysis provides new biological insights into the rewiring of epithelial-stromal co-expression networks and the emergence of epithelial-stromal co-expression self-loops in breast cancer. The approach may facilitate the development of new diagnostics and therapeutics targeting epithelial-stromal interactions in cancer.
Collapse
Affiliation(s)
- Eun-Yeong Oh
- Cancer Research Institute, Beth Israel Deaconess Cancer Center, Boston, MA, 02215, USA. .,Department of Pathology, Beth Israel Deaconess Medical Center, Boston, MA, 02215, USA. .,Harvard Medical School, Boston, MA, 02215, USA.
| | - Stephen M Christensen
- Cancer Research Institute, Beth Israel Deaconess Cancer Center, Boston, MA, 02215, USA. .,Department of Pathology, Beth Israel Deaconess Medical Center, Boston, MA, 02215, USA. .,Harvard Medical School, Boston, MA, 02215, USA.
| | - Sindhu Ghanta
- Cancer Research Institute, Beth Israel Deaconess Cancer Center, Boston, MA, 02215, USA. .,Department of Pathology, Beth Israel Deaconess Medical Center, Boston, MA, 02215, USA. .,Harvard Medical School, Boston, MA, 02215, USA.
| | - Jong Cheol Jeong
- Cancer Research Institute, Beth Israel Deaconess Cancer Center, Boston, MA, 02215, USA. .,Department of Pathology, Beth Israel Deaconess Medical Center, Boston, MA, 02215, USA. .,Harvard Medical School, Boston, MA, 02215, USA.
| | - Octavian Bucur
- Cancer Research Institute, Beth Israel Deaconess Cancer Center, Boston, MA, 02215, USA. .,Department of Pathology, Beth Israel Deaconess Medical Center, Boston, MA, 02215, USA. .,Harvard Medical School, Boston, MA, 02215, USA.
| | - Benjamin Glass
- Cancer Research Institute, Beth Israel Deaconess Cancer Center, Boston, MA, 02215, USA. .,Department of Pathology, Beth Israel Deaconess Medical Center, Boston, MA, 02215, USA. .,Harvard Medical School, Boston, MA, 02215, USA.
| | - Laleh Montaser-Kouhsari
- Cancer Research Institute, Beth Israel Deaconess Cancer Center, Boston, MA, 02215, USA. .,Department of Pathology, Beth Israel Deaconess Medical Center, Boston, MA, 02215, USA. .,Harvard Medical School, Boston, MA, 02215, USA.
| | - Nicholas W Knoblauch
- Cancer Research Institute, Beth Israel Deaconess Cancer Center, Boston, MA, 02215, USA. .,Department of Pathology, Beth Israel Deaconess Medical Center, Boston, MA, 02215, USA. .,Harvard Medical School, Boston, MA, 02215, USA.
| | - Nicholas Bertos
- Goodman Cancer Research Centre, McGill University, Montreal, QC, Canada.
| | - Sadiq Mi Saleh
- Goodman Cancer Research Centre, McGill University, Montreal, QC, Canada.
| | - Benjamin Haibe-Kains
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, M5G 1L7, Canada. .,Department of Medical Biophysics, University of Toronto, Toronto, Ontario, M5G 1L7, Canada.
| | - Morag Park
- Goodman Cancer Research Centre, McGill University, Montreal, QC, Canada.
| | - Andrew H Beck
- Cancer Research Institute, Beth Israel Deaconess Cancer Center, Boston, MA, 02215, USA. .,Department of Pathology, Beth Israel Deaconess Medical Center, Boston, MA, 02215, USA. .,Harvard Medical School, Boston, MA, 02215, USA.
| |
Collapse
|
736
|
Davies KM, Hare DJ, Bohic S, James SA, Billings JL, Finkelstein DI, Doble PA, Double KL. Comparative Study of Metal Quantification in Neurological Tissue Using Laser Ablation-Inductively Coupled Plasma-Mass Spectrometry Imaging and X-ray Fluorescence Microscopy. Anal Chem 2015; 87:6639-45. [DOI: 10.1021/acs.analchem.5b01454] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Affiliation(s)
- Katherine M. Davies
- Neuroscience Research Australia, Randwick, New South Wales, Australia
- School
of Medical Sciences, Faculty of Medicine, University of New South Wales, Kensington, New South Wales, Australia
| | - Dominic J. Hare
- Elemental
Bio-imaging Facility, University of Technology Sydney, Broadway, New South Wales, Australia
- The
Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, Victoria, Australia
- Senator
Frank R. Lautenberg Environmental Health Sciences Laboratory, Department
of Preventive Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, United States
| | - Sylvain Bohic
- Inserm,
U836,
Team 6, Rayonnement Synchrotron et Recherche Médicales, Grenoble Institut des Neurosciences, Grenoble, France
- European Synchrotron Radiation Facility, BP220, Grenoble, France
- Université Joseph Fourier 1, Grenoble Institut des
Neurosciences, Grenoble, France
| | - Simon A. James
- Australian Synchrotron, Clayton, Victoria, Australia
- Materials
Science and Engineering, Commonwealth Scientific and Industrial Research Organisation, Clayton, Victoria, Australia
| | - Jessica L. Billings
- The
Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, Victoria, Australia
| | - David I. Finkelstein
- The
Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, Victoria, Australia
| | - Philip A. Doble
- Elemental
Bio-imaging Facility, University of Technology Sydney, Broadway, New South Wales, Australia
| | - Kay L. Double
- Neuroscience Research Australia, Randwick, New South Wales, Australia
- Brain
and Mind Research Institute, The University of Sydney, 94-100 Mallett
Street,Camperdown, New South
Wales, Australia
| |
Collapse
|
737
|
Leifer Z. The use of virtual microscopy and a wiki in pathology education: Tracking student use, involvement, and response. J Pathol Inform 2015; 6:30. [PMID: 26110097 PMCID: PMC4470013 DOI: 10.4103/2153-3539.158063] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2015] [Accepted: 03/19/2015] [Indexed: 11/23/2022] Open
Abstract
Background: The pathology laboratory course at the New York College of Podiatric Medicine involves the use of Virtual Microscopy. The students can scan the whole slide, section by section, and zoom in or out. Methods: Using the advantages of digital pathology, the students can, in addition, access the slide collections from other medical schools and put up normal histology (control) slides side-by-side with the pathology. They can cut and paste and preserve the region of interest that they find. They can edit and annotate their slides. A wiki was created (http://pathlab2014.wikifoundry.com)for the Class of 2014. The students saved, edited and uploaded their slides. In the wiki format, other students could comment, further edit, and even delete the slides. Results: The students studied Basic Mechanisms and System Pathology. During this time, they saved, edited, shared, and uploaded their slides to the wiki. These were available in one full presentation and were also grouped into 16 albums. They were available to all. Student access was followed by Google analytics. At the end of the course, a questionnaire was distributed, assessing their impression of the wiki format and soliciting strengths and weaknesses. Conclusions: The use of a wiki has a number of important advantages in pathology education. It trains the students in the more sophisticated skills that they will use as professional pathologists or as clinicians: (1) Telepathology-it enables them to share slides and discuss observations. (2) Archiving and retrieval - It models the challenge faced by hospitals, diagnostic labs and physicians in maintaining a collection of slides in a form that is easily accessible. (3) Image analysis-familiarity with the wiki format allows them to jump easily to capturing and storing images found in the literature or in a pathologist's report. Experience with the use of a wiki in pathology education has been quite satisfactory from both the faculty and the student's point of view.
Collapse
Affiliation(s)
- Zev Leifer
- Department of Pre-clinical Sciences, New York College of Podiatric Medicine, New York, NY 10035, USA
| |
Collapse
|
738
|
Koos B, Kamali-Moghaddam M, David L, Sobrinho-Simões M, Dimberg A, Nilsson M, Wählby C, Söderberg O. Next-Generation Pathology—Surveillance of Tumor Microecology. J Mol Biol 2015; 427:2013-22. [DOI: 10.1016/j.jmb.2015.02.017] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2014] [Revised: 02/18/2015] [Accepted: 02/18/2015] [Indexed: 11/30/2022]
|
739
|
Kidd BA, Readhead BP, Eden C, Parekh S, Dudley JT. Integrative network modeling approaches to personalized cancer medicine. Per Med 2015; 12:245-257. [PMID: 27019658 DOI: 10.2217/pme.14.87] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The ability to collect millions of molecular measurements from patients is a now a reality for clinical medicine. This reality has created the challenge of how to integrate these vast amounts of data into models that accurately predict complex pathophysiology and can translate this complexity into clinically actionable outputs. Integrative informatics and data-driven approaches provide a framework for analyzing large-scale datasets and combining them into multiscale models that can be used to determine the key drivers of disease and identify optimal therapies for treating tumors. In this perspective we discuss how an integrative modeling approach is being used to inform individual treatment decisions, highlighting a recent case report that illustrates the challenges and opportunities for personalized oncology.
Collapse
Affiliation(s)
- Brian A Kidd
- Department of Genetics & Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Icahn Institute for Genomics & Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Ben P Readhead
- Department of Genetics & Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Icahn Institute for Genomics & Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Caroline Eden
- Department of Medicine Hematology & Medical Oncology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Samir Parekh
- Department of Medicine Hematology & Medical Oncology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Joel T Dudley
- Department of Genetics & Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Icahn Institute for Genomics & Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Department of Population Health Science and Policy, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| |
Collapse
|
740
|
Nair N, Mei HE, Chen SY, Hale M, Nolan GP, Maecker HT, Genovese M, Fathman CG, Whiting CC. Mass cytometry as a platform for the discovery of cellular biomarkers to guide effective rheumatic disease therapy. Arthritis Res Ther 2015; 17:127. [PMID: 25981462 PMCID: PMC4436107 DOI: 10.1186/s13075-015-0644-z] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
The development of biomarkers for autoimmune diseases has been hampered by a lack of understanding of disease etiopathogenesis and of the mechanisms underlying the induction and maintenance of inflammation, which involves complex activation dynamics of diverse cell types. The heterogeneous nature and suboptimal clinical response to treatment observed in many autoimmune syndromes highlight the need to develop improved strategies to predict patient outcome to therapy and personalize patient care. Mass cytometry, using CyTOF®, is an advanced technology that facilitates multiparametric, phenotypic analysis of immune cells at single-cell resolution. In this review, we outline the capabilities of mass cytometry and illustrate the potential of this technology to enhance the discovery of cellular biomarkers for rheumatoid arthritis, a prototypical autoimmune disease.
Collapse
Affiliation(s)
- Nitya Nair
- Department of Microbiology and Immunology, Stanford University, Stanford, CA, 94305, USA. .,Division of Immune Monitoring and Biomarker Development, Aduro BioTech, Inc., Berkeley, CA, 94710, USA. .,Department of Medicine, Stanford University, Stanford, CA, 94305, USA.
| | - Henrik E Mei
- Institute for Immunity, Transplantation and Infection, Stanford University, Stanford, CA, 94305, USA.
| | - Shih-Yu Chen
- Department of Microbiology and Immunology, Stanford University, Stanford, CA, 94305, USA.
| | - Matthew Hale
- Department of Microbiology and Immunology, Stanford University, Stanford, CA, 94305, USA.
| | - Garry P Nolan
- Department of Microbiology and Immunology, Stanford University, Stanford, CA, 94305, USA.
| | - Holden T Maecker
- Institute for Immunity, Transplantation and Infection, Stanford University, Stanford, CA, 94305, USA.
| | - Mark Genovese
- Department of Medicine, Stanford University, Stanford, CA, 94305, USA.
| | | | - Chan C Whiting
- Division of Immune Monitoring and Biomarker Development, Aduro BioTech, Inc., Berkeley, CA, 94710, USA. .,Department of Medicine, Stanford University, Stanford, CA, 94305, USA.
| |
Collapse
|
741
|
Vreja IC, Kabatas S, Saka SK, Kröhnert K, Höschen C, Opazo F, Diederichsen U, Rizzoli SO. Secondary-ion mass spectrometry of genetically encoded targets. Angew Chem Int Ed Engl 2015; 54:5784-8. [PMID: 25783034 PMCID: PMC4471591 DOI: 10.1002/anie.201411692] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2014] [Indexed: 11/10/2022]
Abstract
Secondary ion mass spectrometry (SIMS) is generally used in imaging the isotopic composition of various materials. It is becoming increasingly popular in biology, especially for investigations of cellular metabolism. However, individual proteins are difficult to identify in SIMS, which limits the ability of this technology to study individual compartments or protein complexes. We present a method for specific protein isotopic and fluorescence labeling (SPILL), based on a novel click reaction with isotopic probes. Using this method, we added (19) F-enriched labels to different proteins, and visualized them by NanoSIMS and fluorescence microscopy. The (19) F signal allowed the precise visualization of the protein of interest, with minimal background, and enabled correlative studies of protein distribution and cellular metabolism or composition. SPILL can be applied to biological systems suitable for click chemistry, which include most cell-culture systems, as well as small model organisms.
Collapse
Affiliation(s)
- Ingrid C Vreja
- Department of Neuro- and Sensory Physiology, University Medical Center GöttingenHumboldtallee 23, 37073 Göttingen (Germany)
- Center for Nanoscale Microscopy and Molecular Physiology of the Brain (CNMPB)Göttingen (Germany)
- International Max Planck Research School Molecular BiologyGöttingen (Germany)
| | - Selda Kabatas
- Institute for Organic and Biomolecular Chemistry, University of GöttingenTammannstrasse 2, 37077 Göttingen (Germany)
- Center for Nanoscale Microscopy and Molecular Physiology of the Brain (CNMPB)Göttingen (Germany)
| | - Sinem K Saka
- Department of Neuro- and Sensory Physiology, University Medical Center GöttingenHumboldtallee 23, 37073 Göttingen (Germany)
- Center for Nanoscale Microscopy and Molecular Physiology of the Brain (CNMPB)Göttingen (Germany)
| | - Katharina Kröhnert
- Department of Neuro- and Sensory Physiology, University Medical Center GöttingenHumboldtallee 23, 37073 Göttingen (Germany)
- Center for Nanoscale Microscopy and Molecular Physiology of the Brain (CNMPB)Göttingen (Germany)
| | - Carmen Höschen
- Department of Ecology and Ecosystem Management, Center of Life and Food Sciences Weihenstephan, Technische Universität MünchenFreising-Weihenstephan (Germany)
| | - Felipe Opazo
- Department of Neuro- and Sensory Physiology, University Medical Center GöttingenHumboldtallee 23, 37073 Göttingen (Germany)
- Center for Nanoscale Microscopy and Molecular Physiology of the Brain (CNMPB)Göttingen (Germany)
| | - Ulf Diederichsen
- Institute for Organic and Biomolecular Chemistry, University of GöttingenTammannstrasse 2, 37077 Göttingen (Germany)
- Center for Nanoscale Microscopy and Molecular Physiology of the Brain (CNMPB)Göttingen (Germany)
| | - Silvio O Rizzoli
- Department of Neuro- and Sensory Physiology, University Medical Center GöttingenHumboldtallee 23, 37073 Göttingen (Germany)
- Center for Nanoscale Microscopy and Molecular Physiology of the Brain (CNMPB)Göttingen (Germany)
| |
Collapse
|
742
|
Cantor DI, Nice EC, Baker MS. Recent findings from the human proteome project: opening the mass spectrometry toolbox to advance cancer diagnosis, surveillance and treatment. Expert Rev Proteomics 2015; 12:279-93. [DOI: 10.1586/14789450.2015.1040770] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
|
743
|
Carvajal-Hausdorf D, Schalper KA, Neumeister V, Rimm DL. Quantitative measurement of cancer tissue biomarkers in the lab and in the clinic. J Transl Med 2015; 95:385-96. [PMID: 25502176 PMCID: PMC4383674 DOI: 10.1038/labinvest.2014.157] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2014] [Accepted: 11/18/2014] [Indexed: 02/06/2023] Open
Abstract
Detection of biomolecules in tissues provides contextual information and the possibility to assess the interaction of different cell types and markers. Routine qualitative assessment of immune- and oligonucleotide-based methods in research and the clinic has been associated with assay variability because of lack of stringent validation and subjective interpretation of results. As a result, the vast majority of in situ assays in clinical usage are nonquantitative and, although useful, often of questionable scientific validity. Here, we revisit the reporters and methods used for single- and multiplexed in situ visualization of protein and RNA. Then we examine methods for the use of quantitative platforms for in situ measurement of protein and mRNA levels. Finally, we discuss the challenges of the transition of these methods to the clinic and their potential role as tools for development of companion diagnostic tests.
Collapse
Affiliation(s)
| | - Kurt A. Schalper
- Department of Pathology, Yale University School of Medicine, New Haven, CT
| | | | - David L. Rimm
- Department of Pathology, Yale University School of Medicine, New Haven, CT
| |
Collapse
|
744
|
Levenson RM, Borowsky AD, Angelo M. Immunohistochemistry and mass spectrometry for highly multiplexed cellular molecular imaging. J Transl Med 2015; 95:397-405. [PMID: 25730370 PMCID: PMC7062454 DOI: 10.1038/labinvest.2015.2] [Citation(s) in RCA: 81] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2014] [Revised: 12/23/2014] [Accepted: 12/24/2014] [Indexed: 01/02/2023] Open
Abstract
The role of immunohistochemistry (IHC) in the management of cancer has expanded to provide improved diagnostic classification, as well as guidance on disease prognosis, therapy, and relapse. These new tasks require evaluation of an increasing number of protein targets; however, conventional multiplexing, usually achieved using serial tissue sections stained for a single analyte per slide, can exhaust small biopsy specimens, complicate slide-to-slide protein expression correlation, and leave insufficient material for additional molecular assays. A new approach, mass spectrometry immunohistochemistry (MSIHC), compatible with high levels of target multiplexing and suitable for use on formalin-fixed, paraffin-embedded samples can circumvent many of these issues. The strategy employs antibodies that are labeled with elemental mass tags, such as isotopically pure lanthanides not typically found in biological specimens, rather than with typical fluorophores or chromogens. The metal-labeled antibodies are then detected in tissue using lasers or ion beams to liberate the tags for subsequent mass spectrometry detection. Within a given multiplexed IHC panel, the metal labels are selected so that their respective masses do not overlap. More than 30 antibodies have been imaged simultaneously, and up to 100 antibodies could potentially be detected at once if the full available mass spectrum is deployed. MSIHC has a number of advantages over conventional IHC techniques. Background due to autofluorescence is absent and the dynamic range is 10(5), exceeding immunofluorescence and chromogenic IHC by 100-fold and 1000-fold, respectively. Detection of labeled primary antibodies improves assay linearity over both chromogenic and fluorescent IHC. Multiplexed mass-tagged antibodies incubated simultaneously with tissue do not appear to cross-interfere, and because the mass tags do not degrade, samples are stable indefinitely. The imaging resolution of multiplexed ion-beam imaging can be better than light microscopy. With appropriate instrumentation, MSIHC has the potential to transform research and clinical pathology practice.
Collapse
Affiliation(s)
- Richard M Levenson
- Department of Pathology and Laboratory Medicine, UC Davis Medical Center, Sacramento, CA, USA
| | - Alexander D Borowsky
- Department of Pathology and Laboratory Medicine, UC Davis Medical Center, Sacramento, CA, USA
| | - Michael Angelo
- Department of Pathology, Stanford University, Palo Alto, CA, USA
| |
Collapse
|
745
|
Vreja IC, Kabatas S, Saka SK, Kröhnert K, Höschen C, Opazo F, Diederichsen U, Rizzoli SO. Sekundärionen-Massenspektrometrie von genetisch kodierten Zielproteinen. Angew Chem Int Ed Engl 2015. [DOI: 10.1002/ange.201411692] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
|
746
|
|
747
|
Beck AH. Open access to large scale datasets is needed to translate knowledge of cancer heterogeneity into better patient outcomes. PLoS Med 2015; 12:e1001794. [PMID: 25710538 PMCID: PMC4339838 DOI: 10.1371/journal.pmed.1001794] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
In this guest editorial, Andrew Beck discusses the importance of open access to big data for translating knowledge of cancer heterogeneity into better outcomes for cancer patients.
Collapse
Affiliation(s)
- Andrew H. Beck
- Cancer Research Institute, Beth Israel Deaconess Cancer Center, Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, United States of America
- * E-mail:
| |
Collapse
|
748
|
Hann S, Dernovics M, Koellensperger G. Elemental analysis in biotechnology. Curr Opin Biotechnol 2015; 31:93-100. [DOI: 10.1016/j.copbio.2014.08.008] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2014] [Accepted: 08/23/2014] [Indexed: 01/25/2023]
|
749
|
Nilsson A, Goodwin RJA, Shariatgorji M, Vallianatou T, Webborn PJH, Andrén PE. Mass Spectrometry Imaging in Drug Development. Anal Chem 2015; 87:1437-55. [DOI: 10.1021/ac504734s] [Citation(s) in RCA: 133] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Anna Nilsson
- Biomolecular
Imaging and Proteomics, National Center for Mass Spectrometry Imaging,
Department of Pharmaceutical Biosciences, Uppsala University, P.O. Box 591 BMC, 75124 Uppsala, Sweden
| | - Richard J. A. Goodwin
- Drug Safety & Metabolism, Innovative Medicines, AstraZeneca, Darwin Building 310, Cambridge Science Park, Milton Road, Cambridge, Cambridgeshire CB4 OWG, U.K
| | - Mohammadreza Shariatgorji
- Biomolecular
Imaging and Proteomics, National Center for Mass Spectrometry Imaging,
Department of Pharmaceutical Biosciences, Uppsala University, P.O. Box 591 BMC, 75124 Uppsala, Sweden
| | - Theodosia Vallianatou
- Biomolecular
Imaging and Proteomics, National Center for Mass Spectrometry Imaging,
Department of Pharmaceutical Biosciences, Uppsala University, P.O. Box 591 BMC, 75124 Uppsala, Sweden
| | - Peter J. H. Webborn
- Drug Safety & Metabolism, Innovative Medicines, AstraZeneca, Darwin Building 310, Cambridge Science Park, Milton Road, Cambridge, Cambridgeshire CB4 OWG, U.K
| | - Per E. Andrén
- Biomolecular
Imaging and Proteomics, National Center for Mass Spectrometry Imaging,
Department of Pharmaceutical Biosciences, Uppsala University, P.O. Box 591 BMC, 75124 Uppsala, Sweden
| |
Collapse
|
750
|
Fornasiero EF, Opazo F. Super-resolution imaging for cell biologists: concepts, applications, current challenges and developments. Bioessays 2015; 37:436-51. [PMID: 25581819 DOI: 10.1002/bies.201400170] [Citation(s) in RCA: 93] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
The recent 2014 Nobel Prize in chemistry honored an era of discoveries and technical advancements in the field of super-resolution microscopy. However, the applications of diffraction-unlimited imaging in biology have a long road ahead and persistently engage scientists with new challenges. Some of the bottlenecks that restrain the dissemination of super-resolution techniques are tangible, and include the limited performance of affinity probes and the yet not capillary diffusion of imaging setups. Likewise, super-resolution microscopy has introduced new paradigms in the design of projects that require imaging with nanometer-resolution and in the interpretation of biological images. Besides structural or morphological characterization, super-resolution imaging is quickly expanding towards interaction mapping, multiple target detection and live imaging. Here we review the recent progress of biologists employing super-resolution imaging, some pitfalls, implications and new trends, with the purpose of animating the field and spurring future developments.
Collapse
Affiliation(s)
- Eugenio F Fornasiero
- STED Microscopy Group, European Neuroscience Institute, Göttingen, Germany; Department of Neuro- and Sensory-physiology, University of Göttingen, Göttingen, Germany
| | | |
Collapse
|