11
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Single-Cell Transcriptomic Analysis of Tumor-Derived Fibroblasts and Normal Tissue-Resident Fibroblasts Reveals Fibroblast Heterogeneity in Breast Cancer

      research-article

      Read this article at

      Bookmark
          There is no author summary for this article yet. Authors can add summaries to their articles on ScienceOpen to make them more accessible to a non-specialist audience.

          Abstract

          Cancer-associated fibroblasts (CAFs) are a prominent stromal cell type in solid tumors and molecules secreted by CAFs play an important role in tumor progression and metastasis. CAFs coexist as heterogeneous populations with potentially different biological functions. Although CAFs are a major component of the breast cancer stroma, molecular and phenotypic heterogeneity of CAFs in breast cancer is poorly understood. In this study, we investigated CAF heterogeneity in triple-negative breast cancer (TNBC) using a syngeneic mouse model, BALB/c-derived 4T1 mammary tumors. Using single-cell RNA sequencing (scRNA-seq), we identified six CAF subpopulations in 4T1 tumors including: 1) myofibroblastic CAFs, enriched for α-smooth muscle actin and several other contractile proteins; 2) ‘inflammatory’ CAFs with elevated expression of inflammatory cytokines; and 3) a CAF subpopulation expressing major histocompatibility complex (MHC) class II proteins that are generally expressed in antigen-presenting cells. Comparison of 4T1-derived CAFs to CAFs from pancreatic cancer revealed that these three CAF subpopulations exist in both tumor types. Interestingly, cells with inflammatory and MHC class II-expressing CAF profiles were also detected in normal breast/pancreas tissue, suggesting that these phenotypes are not tumor microenvironment-induced. This work enhances our understanding of CAF heterogeneity, and specifically targeting these CAF subpopulations could be an effective therapeutic approach for treating highly aggressive TNBCs.

          Related collections

          Most cited references45

          • Record: found
          • Abstract: found
          • Article: not found

          Transforming growth factor-beta regulation of immune responses.

          Transforming growth factor-beta (TGF-beta) is a potent regulatory cytokine with diverse effects on hemopoietic cells. The pivotal function of TGF-beta in the immune system is to maintain tolerance via the regulation of lymphocyte proliferation, differentiation, and survival. In addition, TGF-beta controls the initiation and resolution of inflammatory responses through the regulation of chemotaxis, activation, and survival of lymphocytes, natural killer cells, dendritic cells, macrophages, mast cells, and granulocytes. The regulatory activity of TGF-beta is modulated by the cell differentiation state and by the presence of inflammatory cytokines and costimulatory molecules. Collectively, TGF-beta inhibits the development of immunopathology to self or nonharmful antigens without compromising immune responses to pathogens. This review highlights the findings that have advanced our understanding of TGF-beta in the immune system and in disease.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: found
            Is Open Access

            Cancer-associated fibroblasts: an emerging target of anti-cancer immunotherapy

            Among all the stromal cells that present in the tumor microenvironment, cancer-associated fibroblasts (CAFs) are one of the most abundant and critical components of the tumor mesenchyme, which not only provide physical support for tumor cells but also play a key role in promoting and retarding tumorigenesis in a context-dependent manner. CAFs have also been involved in the modulation of many components of the immune system, and recent studies have revealed their roles in immune evasion and poor responses to cancer immunotherapy. In this review, we describe our current understanding of the tumorigenic significance, origin, and heterogeneity of CAFs, as well as the roles of different CAFs subtypes in distinct immune cell types. More importantly, we highlight potential therapeutic strategies that target CAFs to unleash the immune system against the tumor.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: found
              Is Open Access

              Spatially and functionally distinct subclasses of breast cancer-associated fibroblasts revealed by single cell RNA sequencing

              Cancer-associated fibroblasts (CAFs) are a major constituent of the tumor microenvironment, although their origin and roles in shaping disease initiation, progression and treatment response remain unclear due to significant heterogeneity. Here, following a negative selection strategy combined with single-cell RNA sequencing of 768 transcriptomes of mesenchymal cells from a genetically engineered mouse model of breast cancer, we define three distinct subpopulations of CAFs. Validation at the transcriptional and protein level in several experimental models of cancer and human tumors reveal spatial separation of the CAF subclasses attributable to different origins, including the peri-vascular niche, the mammary fat pad and the transformed epithelium. Gene profiles for each CAF subtype correlate to distinctive functional programs and hold independent prognostic capability in clinical cohorts by association to metastatic disease. In conclusion, the improved resolution of the widely defined CAF population opens the possibility for biomarker-driven development of drugs for precision targeting of CAFs.
                Bookmark

                Author and article information

                Journal
                Cancers (Basel)
                Cancers (Basel)
                cancers
                Cancers
                MDPI
                2072-6694
                21 May 2020
                May 2020
                : 12
                : 5
                : 1307
                Affiliations
                [1 ]Physical and Life Sciences Directorate, Lawrence Livermore National Laboratory, Livermore, CA 94550, USA; sebastian4@ 123456llnl.gov (A.S.); hum3@ 123456llnl.gov (N.R.H.); martin249@ 123456llnl.gov (K.A.M.); gilmore24@ 123456llnl.gov (S.F.G.); coleman16@ 123456llnl.gov (M.A.C.)
                [2 ]School of Natural Sciences, University of California Merced, Merced, CA 95343, USA
                [3 ]Georgetown-Lombardi Comprehensive Cancer Center, Department of Oncology, Georgetown University Medical Center, Washington, DC 20007, USA; ip62@ 123456georgetown.edu (I.P.); byerss@ 123456georgetown.edu (S.W.B.)
                [4 ]Engineering Directorate, Lawrence Livermore National Laboratory, Livermore, CA 94550, USA; wheeler16@ 123456llnl.gov
                [5 ]Department of Biochemistry and Molecular Medicine, University of California Davis, Sacramento, CA 95817, USA
                Author notes
                [* ]Correspondence: loots1@ 123456llnl.gov ; Tel.: +1-925-423-0923
                Author information
                https://orcid.org/0000-0002-7822-7040
                https://orcid.org/0000-0003-1605-3193
                https://orcid.org/0000-0001-7399-7452
                https://orcid.org/0000-0001-9546-5561
                Article
                cancers-12-01307
                10.3390/cancers12051307
                7281266
                32455670
                95942692-dcff-4100-95e2-abcefe0dbeed
                © 2020 by the authors.

                Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license ( http://creativecommons.org/licenses/by/4.0/).

                History
                : 02 April 2020
                : 18 May 2020
                Categories
                Article

                cancer-associated fibroblasts,breast cancer,mammary fat pad,gene expression profiling,scrna-seq,caf heterogeneity,normal fibroblasts,myofibroblasts,inflammatory fibroblasts,pancreatic cancer

                Comments

                Comment on this article