Change search
Link to record
Permanent link

Direct link
Mattsson Langseth, ChristofferORCID iD iconorcid.org/0000-0003-2230-8594
Publications (10 of 17) Show all publications
Mattsson Langseth, C. (2025). Cells in Time and Space: Beyond comprehensive cellular atlases towards a deeper understanding of disease. (Doctoral dissertation). Stockholm: Department of Biochemistry and Biophysics, Stockholm University
Open this publication in new window or tab >>Cells in Time and Space: Beyond comprehensive cellular atlases towards a deeper understanding of disease
2025 (English)Doctoral thesis, comprehensive summary (Other academic)
Abstract [en]

Recent advancements in single-cell RNA sequencing have transformed the way we classify cells. These innovations have provided an unprecedented view into the complexities of cell types within the human body and enabled the creation of comprehensive cellular atlases for various tissues and organs. While these techniques have revolutionized biology, they are limited in that they lack the necessary information to examine the architecture of complex tissues. This limitation has led to the development of spatially resolved transcriptomic (SRT) techniques, giving rise to the field of spatial biology. The rapid growth of spatial biology has largely been driven by academic research, though commercial entities have recently begun to emerge in the market.

In situ sequencing (ISS) is one such SRT technique that enables the mapping of hundreds of transcripts directly in tissue samples. ISS utilizes padlock probes, which hybridize to a target. Ligation of the probes allows for their circularization. Once circularized, the probes are enzymatically amplified, and their identity can be decoded using fluorescent probes and a microscope. 

In Paper I, we build upon the first iteration of ISS to create a hybridization-based sequencing approach  that allows for more genes to be targeted. Additionally, the improved signal-to-noise ratio enabled more robust signal detection, facilitating the application to aged human brain tissue, which is challenging to analyze due to autofluorescence.

In Paper II, we use the technology developed in Paper I to map out 75 transcriptomically defined cell types in the human cortex. In total, we pinpointed the location of 59,816 cells in their native positions and abundances. We looked at both the within- and across-layer distribution of these cell types.  

In Paper III, we profiled the development of oligodendrocyte lineage cells in mouse spinal cord and brain. Over different time points, we could map out the location of cell types and model the development of these cellular lineages, uncovering the neighboring preferences of these cells. The data revealed spatial heterogeneity of oligodendrocyte lineage progression in the brain and spinal cord. 

In Paper IV, we modeled the development of neuroinflammatory lesions in the multiple sclerosis model of experimental autoimmune encephalomyelitis (EAE). We built a comprehensive atlas of the spatio-temporal dynamics of the disease development by collecting tissues from different time points and different regions of the CNS. We uncovered the dynamic nature of disease-associated glial subtypes, being induced globally at peak and then reverting back to their homeostatic gene expression signature at the reduced inflammatory state. Moreover, the temporal analysis of the lesion development allowed for uncovering the intricate structure of these lesions and how they propagate over time. Human tissue sections confirmed the cell-driven approach to lesion identification and the induction of disease-associated glial subtypes. 

Taken together, the work presented in this thesis serves to showcase how one can use ISS to create comprehensive atlases, but more importantly, move beyond cellular atlases towards understanding disease. 

 

Place, publisher, year, edition, pages
Stockholm: Department of Biochemistry and Biophysics, Stockholm University, 2025. p. 84
Keywords
in situ sequencing, padlock probes, rolling circle amplification, spatially resolved transcriptomics, spatial transcriptomics, molecular atlases, cell atlases, cell type definition, human neuroanatomy, neuroinflammation, multiple sclerosis
National Category
Biochemistry Molecular Biology Bioinformatics and Computational Biology
Research subject
Biochemistry towards Bioinformatics
Identifiers
urn:nbn:se:su:diva-237615 (URN)978-91-8107-086-6 (ISBN)978-91-8107-087-3 (ISBN)
Public defence
2025-03-14, Air & Fire, Gamma 2, SciLifeLab, Tomtebodavägen 23a, Solna, 14:00 (English)
Opponent
Supervisors
Available from: 2025-02-19 Created: 2025-01-21 Last updated: 2025-02-20Bibliographically approved
Marco Salas, S., Kuemmerle, L. B., Mattsson Langseth, C., Tismeyer, S., Avenel, C., Hu, T., . . . Nilsson, M. (2025). Optimizing Xenium In Situ data utility by quality assessment and best-practice analysis workflows. Nature Methods, Article ID aaa6090.
Open this publication in new window or tab >>Optimizing Xenium In Situ data utility by quality assessment and best-practice analysis workflows
Show others...
2025 (English)In: Nature Methods, ISSN 1548-7091, E-ISSN 1548-7105, article id aaa6090Article in journal (Refereed) Epub ahead of print
Abstract [en]

The Xenium In Situ platform is a new spatial transcriptomics product commercialized by 10x Genomics, capable of mapping hundreds of genes in situ at subcellular resolution. Given the multitude of commercially available spatial transcriptomics technologies, recommendations in choice of platform and analysis guidelines are increasingly important. Herein, we explore 25 Xenium datasets generated from multiple tissues and species, comparing scalability, resolution, data quality, capacities and limitations with eight other spatially resolved transcriptomics technologies and commercial platforms. In addition, we benchmark the performance of multiple open-source computational tools, when applied to Xenium datasets, in tasks including preprocessing, cell segmentation, selection of spatially variable features and domain identification. This study serves as an independent analysis of the performance of Xenium, and provides best practices and recommendations for analysis of such datasets.

National Category
Bioinformatics and Computational Biology
Identifiers
urn:nbn:se:su:diva-242429 (URN)10.1038/s41592-025-02617-2 (DOI)001444358900001 ()40082609 (PubMedID)2-s2.0-105000286295 (Scopus ID)
Available from: 2025-04-23 Created: 2025-04-23 Last updated: 2025-04-23
De Jonghe, J., Opzoomer, J. W., Vilas-Zornoza, A., Crane, P., Nilges, B. S., Vicari, M., . . . Taylor-King, J. P. (2024). A community effort to track commercial single-cell and spatial ’omic technologies and business trends [Letter to the editor]. Nature Biotechnology, 42(7), 1017-1023
Open this publication in new window or tab >>A community effort to track commercial single-cell and spatial ’omic technologies and business trends
Show others...
2024 (English)In: Nature Biotechnology, ISSN 1087-0156, E-ISSN 1546-1696, Vol. 42, no 7, p. 1017-1023Article in journal, Letter (Refereed) Published
Abstract [en]

There is an ever-growing choice of single-cell and spatial ’omics platforms for industry and academia. The scTrends Consortium provides a brief historical overview of the established platforms and companies, revealing market trends and presenting possible angles for how technologies may differentiate themselves.

National Category
Computer and Information Sciences
Identifiers
urn:nbn:se:su:diva-238472 (URN)10.1038/s41587-024-02305-0 (DOI)001271920600007 ()39020213 (PubMedID)2-s2.0-85199126484 (Scopus ID)
Available from: 2025-01-27 Created: 2025-01-27 Last updated: 2025-01-27Bibliographically approved
Kukanja, P., Mattsson Langseth, C., Rodríguez-Kirby, L. A. R., Agirre, E., Zheng, C., Raman, A., . . . Castelo-Branco, G. (2024). Cellular architecture of evolving neuroinflammatory lesions and multiple sclerosis pathology. Cell, 187(8), 1990-2009
Open this publication in new window or tab >>Cellular architecture of evolving neuroinflammatory lesions and multiple sclerosis pathology
Show others...
2024 (English)In: Cell, ISSN 0092-8674, E-ISSN 1097-4172, Vol. 187, no 8, p. 1990-2009Article in journal (Refereed) Published
Abstract [en]

Multiple sclerosis (MS) is a neurological disease characterized by multifocal lesions and smoldering pathology. Although single-cell analyses provided insights into cytopathology, evolving cellular processes underlying MS remain poorly understood. We investigated the cellular dynamics of MS by modeling temporal and regional rates of disease progression in mouse experimental autoimmune encephalomyelitis (EAE). By performing single-cell spatial expression profiling using in situ sequencing (ISS), we annotated disease neighborhoods and found centrifugal evolution of active lesions. We demonstrated that disease-associated (DA)-glia arise independently of lesions and are dynamically induced and resolved over the disease course. Single-cell spatial mapping of human archival MS spinal cords confirmed the differential distribution of homeostatic and DA-glia, enabled deconvolution of active and inactive lesions into sub-compartments, and identified new lesion areas. By establishing a spatial resource of mouse and human MS neuropathology at a single-cell resolution, our study unveils the intricate cellular dynamics underlying MS.

National Category
Cell Biology Cell and Molecular Biology
Identifiers
urn:nbn:se:su:diva-231552 (URN)10.1016/j.cell.2024.02.030 (DOI)001229191700001 ()38513664 (PubMedID)2-s2.0-85189500592 (Scopus ID)
Available from: 2024-06-25 Created: 2024-06-25 Last updated: 2025-01-21Bibliographically approved
Wernersson, E., Gelali, E., Girelli, G., Wang, S., Castillo, D., Mattsson Langseth, C., . . . Bienko, M. (2024). Deconwolf enables high-performance deconvolution of widefield fluorescence microscopy images. Nature Methods, 21(7), 1245-1256
Open this publication in new window or tab >>Deconwolf enables high-performance deconvolution of widefield fluorescence microscopy images
Show others...
2024 (English)In: Nature Methods, ISSN 1548-7091, E-ISSN 1548-7105, Vol. 21, no 7, p. 1245-1256Article in journal (Refereed) Published
Abstract [en]

Microscopy-based spatially resolved omic methods are transforming the life sciences. However, these methods rely on high numerical aperture objectives and cannot resolve crowded molecular targets, limiting the amount of extractable biological information. To overcome these limitations, here we develop Deconwolf, an open-source, user-friendly software for high-performance deconvolution of widefield fluorescence microscopy images, which efficiently runs on laptop computers. Deconwolf enables accurate quantification of crowded diffraction limited fluorescence dots in DNA and RNA fluorescence in situ hybridization images and allows robust detection of individual transcripts in tissue sections imaged with ×20 air objectives. Deconvolution of in situ spatial transcriptomics images with Deconwolf increased the number of transcripts identified more than threefold, while the application of Deconwolf to images obtained by fluorescence in situ sequencing of barcoded Oligopaint probes drastically improved chromosome tracing. Deconwolf greatly facilitates the use of deconvolution in many bioimaging applications.

National Category
Other Biological Topics
Identifiers
urn:nbn:se:su:diva-231208 (URN)10.1038/s41592-024-02294-7 (DOI)001242316400003 ()38844629 (PubMedID)2-s2.0-85195378441 (Scopus ID)
Available from: 2024-06-18 Created: 2024-06-18 Last updated: 2024-09-04Bibliographically approved
Lee, H. Z., Mattsson Langseth, C., Marco Salas, S., Sariyar, S., Metousis, A., Rueda-Alaña, E., . . . Nilsson, M. (2024). Open-source, high-throughput targeted in situ transcriptomics for developmental and tissue biology. Development, 151(16), Article ID dev202448.
Open this publication in new window or tab >>Open-source, high-throughput targeted in situ transcriptomics for developmental and tissue biology
Show others...
2024 (English)In: Development, ISSN 0950-1991, E-ISSN 1477-9129, Vol. 151, no 16, article id dev202448Article in journal (Refereed) Published
Abstract [en]

Multiplexed spatial profiling of mRNAs has recently gained traction as a tool to explore the cellular diversity and the architecture of tissues. We propose a sensitive, open-source, simple and flexible method for the generation of in situ expression maps of hundreds of genes. We use direct ligation of padlock probes on mRNAs, coupled with rolling circle amplification and hybridization-based in situ combinatorial barcoding, to achieve high detection efficiency, high-throughput and large multiplexing. We validate the method across a number of species and show its use in combination with orthogonal methods such as antibody staining, highlighting its potential value for developmental and tissue biology studies. Finally, we provide an end-to-end computational workflow that covers the steps of probe design, image processing, data extraction, cell segmentation, clustering and annotation of cell types. By enabling easier access to high-throughput spatially resolved transcriptomics, we hope to encourage a diversity of applications and the exploration of a wide range of biological questions.

Keywords
Spatial transcriptomics, In situ hybridization, Multiplex imaging, Multi-omics, Open source, Padlock probes
National Category
Genetics and Genomics Developmental Biology
Research subject
Biochemistry
Identifiers
urn:nbn:se:su:diva-231347 (URN)10.1242/dev.202448 (DOI)001301313300002 ()39099456 (PubMedID)2-s2.0-85202906678 (Scopus ID)
Available from: 2024-06-19 Created: 2024-06-19 Last updated: 2025-01-21Bibliographically approved
De Jonghe, J., Opzoomer, J. W., Vilas-Zornoza, A., Nilges, B. S., Crane, P., Vicari, M., . . . Taylor-King, J. P. (2024). scTrends: A living review of commercial single-cell and spatial 'omic technologies. Cell Genomics, 4(12), Article ID 100723.
Open this publication in new window or tab >>scTrends: A living review of commercial single-cell and spatial 'omic technologies
Show others...
2024 (English)In: Cell Genomics, E-ISSN 2666-979X, Vol. 4, no 12, article id 100723Article, review/survey (Refereed) Published
Abstract [en]

Understanding the rapidly evolving landscape of single-cell and spatial omic technologies is crucial for advancing biomedical research and drug development. We provide a living review of both mature and emerging commercial platforms, highlighting key methodologies and trends shaping the field. This review spans from foundational single-cell technologies such as microfluidics and plate-based methods to newer approaches like combinatorial indexing; on the spatial side, we consider next-generation sequencing and imaging-based spatial transcriptomics. Finally, we highlight emerging methodologies that may fundamentally expand the scope for data generation within pharmaceutical research, creating opportunities to discover and validate novel drug mechanisms. Overall, this review serves as a critical resource for navigating the commercialization and application of single-cell and spatial omic technologies in pharmaceutical and academic research.

National Category
Genetics and Genomics Cell Biology
Identifiers
urn:nbn:se:su:diva-240557 (URN)10.1016/j.xgen.2024.100723 (DOI)001409711500001 ()2-s2.0-85211348738 (Scopus ID)
Available from: 2025-03-10 Created: 2025-03-10 Last updated: 2025-03-10Bibliographically approved
Li, X., Andrusivova, Z., Czarnewski, P., Mattsson Langseth, C., Andersson, A., Liu, Y., . . . Sundström, E. (2023). Profiling spatiotemporal gene expression of the developing human spinal cord and implications for ependymoma origin. Nature Neuroscience, 26(5), 891-901
Open this publication in new window or tab >>Profiling spatiotemporal gene expression of the developing human spinal cord and implications for ependymoma origin
Show others...
2023 (English)In: Nature Neuroscience, ISSN 1097-6256, E-ISSN 1546-1726, Vol. 26, no 5, p. 891-901Article in journal (Refereed) Published
Abstract [en]

The spatiotemporal regulation of cell fate specification in the human developing spinal cord remains largely unknown. In this study, by performing integrated analysis of single-cell and spatial multi-omics data, we used 16 prenatal human samples to create a comprehensive developmental cell atlas of the spinal cord during post-conceptional weeks 5–12. This revealed how the cell fate commitment of neural progenitor cells and their spatial positioning are spatiotemporally regulated by specific gene sets. We identified unique events in human spinal cord development relative to rodents, including earlier quiescence of active neural stem cells, differential regulation of cell differentiation and distinct spatiotemporal genetic regulation of cell fate choices. In addition, by integrating our atlas with pediatric ependymomas data, we identified specific molecular signatures and lineage-specific genes of cancer stem cells during progression. Thus, we delineate spatiotemporal genetic regulation of human spinal cord development and leverage these data to gain disease insight.

National Category
Neurosciences
Identifiers
urn:nbn:se:su:diva-228932 (URN)10.1038/s41593-023-01312-9 (DOI)000975560000004 ()37095395 (PubMedID)2-s2.0-85153355240 (Scopus ID)
Available from: 2024-05-06 Created: 2024-05-06 Last updated: 2024-06-10Bibliographically approved
Zhang, Y., Miller, J. A., Park, J., Lelieveldt, B. P., Long, B., Abdelaal, T., . . . Scheuermann, R. H. (2023). Reference-based cell type matching of in situ image-based spatial transcriptomics data on primary visual cortex of mouse brain. Scientific Reports, 13, Article ID 9567.
Open this publication in new window or tab >>Reference-based cell type matching of in situ image-based spatial transcriptomics data on primary visual cortex of mouse brain
Show others...
2023 (English)In: Scientific Reports, E-ISSN 2045-2322, Vol. 13, article id 9567Article in journal (Refereed) Published
Abstract [en]

With the advent of multiplex fluorescence in situ hybridization (FISH) and in situ RNA sequencing technologies, spatial transcriptomics analysis is advancing rapidly, providing spatial location and gene expression information about cells in tissue sections at single cell resolution. Cell type classification of these spatially-resolved cells can be inferred by matching the spatial transcriptomics data to reference atlases derived from single cell RNA-sequencing (scRNA-seq) in which cell types are defined by differences in their gene expression profiles. However, robust cell type matching of the spatially-resolved cells to reference scRNA-seq atlases is challenging due to the intrinsic differences in resolution between the spatial and scRNA-seq data. In this study, we systematically evaluated six computational algorithms for cell type matching across four image-based spatial transcriptomics experimental protocols (MERFISH, smFISH, BaristaSeq, and ExSeq) conducted on the same mouse primary visual cortex (VISp) brain region. We find that many cells are assigned as the same type by multiple cell type matching algorithms and are present in spatial patterns previously reported from scRNA-seq studies in VISp. Furthermore, by combining the results of individual matching strategies into consensus cell type assignments, we see even greater alignment with biological expectations. We present two ensemble meta-analysis strategies used in this study and share the consensus cell type matching results in the Cytosplore Viewer (https://viewer.cytosplore.org) for interactive visualization and data exploration. The consensus matching can also guide spatial data analysis using SSAM, allowing segmentation-free cell type assignment.

National Category
Bioinformatics and Computational Biology
Identifiers
urn:nbn:se:su:diva-230560 (URN)10.1038/s41598-023-36638-8 (DOI)001011884700012 ()37311768 (PubMedID)2-s2.0-85161813840 (Scopus ID)
Available from: 2024-06-11 Created: 2024-06-11 Last updated: 2025-02-07Bibliographically approved
van Bruggen, D., Pohl, F., Mattsson Langseth, C., Kukanja, P., Lee, H., Albiach, A. M., . . . Castelo-Branco, G. (2022). Developmental landscape of human forebrain at a single-cell level identifies early waves of oligodendrogenesis. Developmental Cell, 57(11), 1421-1436, 1421-1436.e1-e5
Open this publication in new window or tab >>Developmental landscape of human forebrain at a single-cell level identifies early waves of oligodendrogenesis
Show others...
2022 (English)In: Developmental Cell, ISSN 1534-5807, E-ISSN 1878-1551, Vol. 57, no 11, p. 1421-1436, 1421-1436.e1-e5Article in journal (Refereed) Published
Abstract [en]

Oligodendrogenesis in the human central nervous system has been observed mainly at the second trimester of gestation, a much later developmental stage compared to oligodendrogenesis in mice. Here, we characterize the transcriptomic neural diversity in the human forebrain at post-conception weeks (PCW) 8–10. Using single-cell RNA sequencing, we find evidence of the emergence of a first wave of oligodendrocyte lineage cells as early as PCW 8, which we also confirm at the epigenomic level through the use of single-cell ATAC-seq. Using regulatory network inference, we predict key transcriptional events leading to the specification of oligodendrocyte precursor cells (OPCs). Moreover, by profiling the spatial expression of 50 key genes through the use of in situ sequencing (ISS), we identify regions in the human ventral fetal forebrain where oligodendrogenesis first occurs. Our results indicate evolutionary conservation of the first wave of oligodendrogenesis between mice and humans and describe regulatory mechanisms involved in human OPC specification.

National Category
Biological Sciences
Identifiers
urn:nbn:se:su:diva-208413 (URN)10.1016/j.devcel.2022.04.016 (DOI)000822525500003 ()35523173 (PubMedID)2-s2.0-85131771145 (Scopus ID)
Available from: 2022-08-29 Created: 2022-08-29 Last updated: 2022-08-29Bibliographically approved
Organisations
Identifiers
ORCID iD: ORCID iD iconorcid.org/0000-0003-2230-8594

Search in DiVA

Show all publications