1
|
Herik AI, Sinha S, Arora R, Small C, Dufour A, Biernaskie J, Cobo ER, McKay DM. In silico integrative scRNA analysis of human colonic epithelium indicates four tuft cell subtypes. Am J Physiol Gastrointest Liver Physiol 2025; 328:G96-G109. [PMID: 39589317 DOI: 10.1152/ajpgi.00182.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 11/04/2024] [Accepted: 11/17/2024] [Indexed: 11/27/2024]
Abstract
This study integrated and analyzed human single-cell RNA sequencing data from four publicly available datasets to enhance cellular resolution, unveiling a complex landscape of tuft cell heterogeneity within the human colon. Four tuft subtypes (TC1-TC4) emerged, as defined by unique gene expression profiles, indicating potentially novel biological functions. Tuft cell 1 (TC1) was characterized by an antimicrobial peptide signature; TC2 had an increased transcription machinery gene expression profile consistent with a progenitor-like cell; TC3 expressed genes related to ganglion (neuronal) development; and TC4 expressed genes related to tight junctions. Our analysis of subtype-specific gene expression and pathway enrichment showed variances in tuft cell subtypes between healthy individuals and those with inflammatory bowel disease (IBD). The frequency of TC1 and TC2 differed between healthy controls and IBD. Relative to healthy controls, TC1 and TC2 in IBD tissue showed an upregulation of gene expression, favoring increased metabolism and immune function. These findings provide foundational knowledge about the complexity of the human colon tuft cell population and hint at their potential contributions to gut health. They provide a basis for future studies to explore the specific roles these cells may play in gut function during homeostasis and disease. We demonstrate the value of in silico approaches for hypothesis generation in relation to the putative functions of low-frequency gut cells for subsequent physiological analyses.NEW & NOTEWORTHY This study reveals the nuanced and novel landscape of human colonic tuft cells through integrative scRNA-seq analysis. Four distinct tuft cell subtypes were identified, varying markedly between healthy and individuals with IBD. We uncovered human colonic tuft cell subtypes with unexpected antimicrobial and progenitor-like gene expression signatures. These insights into tuft cell diversity offer new avenues for understanding gut health and disease pathophysiology.
Collapse
Affiliation(s)
- Aydin I Herik
- Faculty of Veterinary Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Sarthak Sinha
- Department of Comparative Biology and Experimental Medicine, Faculty of Veterinary Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Rohit Arora
- Faculty of Veterinary Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Caleb Small
- Faculty of Veterinary Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Antoine Dufour
- McCaig Institute for Bone and Joint Health, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
- Department of Physiology and Pharmacology, Alberta Children's Hospital Research Institute, Calgary, Alberta, Canada
| | - Jeff Biernaskie
- Faculty of Veterinary Medicine, University of Calgary, Calgary, Alberta, Canada
- Department of Physiology and Pharmacology, Alberta Children's Hospital Research Institute, Calgary, Alberta, Canada
| | - Eduardo R Cobo
- Faculty of Veterinary Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Derek M McKay
- Gastrointestinal Research Group and Inflammation Research Network, Department of Physiology and Pharmacology, Calvin, Phoebe, and Joan Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
2
|
Foote AG, Sun X. A Single-Cell Atlas of the Upper Respiratory Epithelium Reveals Heterogeneity in Cell Types and Patterning Strategies. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.16.633456. [PMID: 39896587 PMCID: PMC11785068 DOI: 10.1101/2025.01.16.633456] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/04/2025]
Abstract
The upper respiratory tract, organized along the pharyngolaryngeal-to-tracheobronchial axis, is essential for homeostatic functions such as breathing and vocalization. The upper respiratory epithelium is frequently exposed to pollutants and pathogens, making this an area of first-line defense against respiratory injury and infection. The respiratory epithelium is composed of a rich array of specialized cell types, each with unique capabilities in immune defense and injury repair. However, the precise transcriptomic signature and spatial distribution of these cell populations, as well as potential cell subpopulations, have not been well defined. Here, using single cell RNAseq combined with spatial validation, we present a comprehensive atlas of the mouse upper respiratory epithelium. We systematically analyzed our rich RNAseq dataset of the upper respiratory epithelium to reveal 17 cell types, which we further organized into three spatially distinct compartments: the Tmprss11a + pharyngolaryngeal, the Nkx2-1 + tracheobronchial, and the Dmbt1 + submucosal gland epithelium. We profiled/analyzed the pharyngolaryngeal epithelium, composed of stratified squamous epithelium, and identified distinct regional signatures, including a Keratin gene expression code. In profiling the tracheobronchial epithelium, which is composed of a pseudostratified epithelium-with the exception of the hillock structure-we identified that regional luminal cells, such as club cells and basal cells, show varying gradients of marker expression along the proximal-distal and/or dorsal-ventral axis. Lastly, our analysis of the submucosal gland epithelium, composed of an array of cell types, such as the unique myoepithelial cells, revealed the colorful diversity of between and within cell populations. Our single-cell atlas with spatial validation highlights the distinct transcriptional programs of the upper respiratory epithelium and serves as a valuable resource for future investigations to address how cells behave in homeostasis and pathogenesis. Highlights - Defined three spatially distinct epithelial compartments, Tmprss11a + pharyngolaryngeal, Nkx2-1 + tracheobronchial, and Dmbt1 + submucosal gland, comprising 17 total cell types - Profiled Keratin gene expression code along proximal-distal and basal-luminal axes and highlighted "stress-induced" Keratins KRT6A and KRT17 at homeostasis - Demarcated expression gradients of Scgb1a1 + and Scgb3a2+ club cells along the proximal-distal axes - Specified submucosal gland cell heterogeneity including Nkx3-1+ mucin-producing cells, with ACTA2+ basal myoepithelial cells exhibiting gene profile for neuroimmune mediated signaling.
Collapse
|
3
|
Feng X, Flüchter P, De Tenorio JC, Schneider C. Tuft cells in the intestine, immunity and beyond. Nat Rev Gastroenterol Hepatol 2024; 21:852-868. [PMID: 39327439 DOI: 10.1038/s41575-024-00978-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/02/2024] [Indexed: 09/28/2024]
Abstract
Tuft cells have gained substantial attention over the past 10 years due to numerous reports linking them with type 2 immunity and microorganism-sensing capacity in many mucosal tissues. This heightened interest is fuelled by their unique ability to produce an array of biological effector molecules, including IL-25, allergy-related eicosanoids, and the neurotransmitter acetylcholine, enabling downstream responses in diverse cell types. Operating through G protein-coupled receptor-mediated signalling pathways reminiscent of type II taste cells in oral taste buds, tuft cells emerge as chemosensory sentinels that integrate luminal conditions, eliciting appropriate responses in immune, epithelial and neuronal populations. How tuft cells promote tissue alterations and adaptation to the variety of stimuli at mucosal surfaces has been explored in multiple studies in the past few years. Since the initial recognition of the role of tuft cells, the discovery of diverse tuft cell effector functions and associated feedback loops have also revealed the complexity of tuft cell biology. Although earlier work largely focused on extraintestinal tissues, novel genetic tools and recent mechanistic studies on intestinal tuft cells established fundamental concepts of tuft cell activation and functions. This Review is an overview of intestinal tuft cells, providing insights into their development, signalling and interaction modules in immunity and other states.
Collapse
Affiliation(s)
- Xiaogang Feng
- Department of Physiology, University of Zurich, Zurich, Switzerland
| | - Pascal Flüchter
- Department of Physiology, University of Zurich, Zurich, Switzerland
| | | | | |
Collapse
|
4
|
Wu B, Bennett HM, Ye X, Sridhar A, Eidenschenk C, Everett C, Nazarova EV, Chen HH, Kim IK, Deangelis M, Owen LA, Chen C, Lau J, Shi M, Lund JM, Xavier-Magalhães A, Patel N, Liang Y, Modrusan Z, Darmanis S. Overloading And unpacKing (OAK) - droplet-based combinatorial indexing for ultra-high throughput single-cell multiomic profiling. Nat Commun 2024; 15:9146. [PMID: 39443484 PMCID: PMC11499997 DOI: 10.1038/s41467-024-53227-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Accepted: 10/02/2024] [Indexed: 10/25/2024] Open
Abstract
Multiomic profiling of single cells by sequencing is a powerful technique for investigating cellular diversity. Existing droplet-based microfluidic methods produce many cell-free droplets, underutilizing bead barcodes and reagents. Combinatorial indexing on microplates is more efficient for barcoding but labor-intensive. Here we present Overloading And unpacKing (OAK), which uses a droplet-based barcoding system for initial compartmentalization followed by a second aliquoting round to achieve combinatorial indexing. We demonstrate OAK's versatility with single-cell RNA sequencing as well as paired single-nucleus RNA sequencing and accessible chromatin profiling. We further showcase OAK's performance on complex samples, including differentiated bronchial epithelial cells and primary retinal tissue. Finally, we examine transcriptomic responses of over 400,000 melanoma cells to a RAF inhibitor, belvarafenib, discovering a rare resistant cell population (0.12%). OAK's ultra-high throughput, broad compatibility, high sensitivity, and simplified procedures make it a powerful tool for large-scale molecular analysis, even for rare cells.
Collapse
Affiliation(s)
- Bing Wu
- Department of Proteomic and Genomic Technologies, Genentech, South San Francisco, CA, USA
| | - Hayley M Bennett
- Department of Proteomic and Genomic Technologies, Genentech, South San Francisco, CA, USA
| | - Xin Ye
- Department of Discovery Oncology, Genentech, South San Francisco, CA, USA
| | - Akshayalakshmi Sridhar
- Department of Human Pathobiology & OMNI Reverse Translation, Genentech, South San Francisco, CA, USA
| | - Celine Eidenschenk
- Department of Functional Genomics, Genentech, South San Francisco, CA, USA
| | - Christine Everett
- Department of Functional Genomics, Genentech, South San Francisco, CA, USA
| | | | - Hsu-Hsin Chen
- Department of Human Pathobiology & OMNI Reverse Translation, Genentech, South San Francisco, CA, USA
| | - Ivana K Kim
- Retina Service, Massachusetts Eye & Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
| | - Margaret Deangelis
- Department of Ophthalmology, Ross Eye Institute; Department of Biochemistry; Neuroscience Graduate Program; Genetics, Genomics and Bioinformatics Graduate Program, Jacobs School of Medicine and Biomedical Sciences, State University of New York, University at Buffalo, Buffalo, NY, USA
| | - Leah A Owen
- Department of Ophthalmology and Visual Sciences, University of Utah School of Medicine, The University of Utah, Salt Lake City, UT, USA
| | - Cynthia Chen
- Department of Proteomic and Genomic Technologies, Genentech, South San Francisco, CA, USA
| | - Julia Lau
- Department of Proteomic and Genomic Technologies, Genentech, South San Francisco, CA, USA
| | - Minyi Shi
- Department of Proteomic and Genomic Technologies, Genentech, South San Francisco, CA, USA
| | - Jessica M Lund
- Department of Proteomic and Genomic Technologies, Genentech, South San Francisco, CA, USA
| | - Ana Xavier-Magalhães
- Department of Proteomic and Genomic Technologies, Genentech, South San Francisco, CA, USA
| | - Neha Patel
- Department of Proteomic and Genomic Technologies, Genentech, South San Francisco, CA, USA
| | - Yuxin Liang
- Department of Proteomic and Genomic Technologies, Genentech, South San Francisco, CA, USA
| | - Zora Modrusan
- Department of Proteomic and Genomic Technologies, Genentech, South San Francisco, CA, USA.
| | - Spyros Darmanis
- Department of Proteomic and Genomic Technologies, Genentech, South San Francisco, CA, USA.
| |
Collapse
|
5
|
Lin X, Chen W, Yang G, Zhang J, Wang H, Liu Z, Xi Y, Ren T, Liu B, Sui P. Viral infection induces inflammatory signals that coordinate YAP regulation of dysplastic cells in lung alveoli. J Clin Invest 2024; 134:e176828. [PMID: 39352385 PMCID: PMC11444164 DOI: 10.1172/jci176828] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Accepted: 08/06/2024] [Indexed: 10/03/2024] Open
Abstract
Severe viral pneumonia can induce rapid expansion of KRT5+ basal-like cells in small airways and alveoli; this forms a scar-like structure that persists in the injured alveoli and impedes normal alveolar epithelium regeneration. In this study, we investigated the mechanism by which viral infection induced this remodeling response. Through comparing different lung-injury models, we demonstrated that infection induced strong IFN-γ signal-stimulated dysplastic KRT5+ cell formation. Inactivation of interferon receptor 1 (Ifngr1) reduced dysplastic cell formation, ameliorated lung fibrosis, and improved lung-function recovery. Mechanistically, IFN-γ regulated dysplastic cell formation via the focal adhesion kinase (FAK)/Yes-associated protein 1 (YAP) pathway. Inhibiting FAK/Src diminished IFN-γ-induced YAP nuclear translocation and dysplastic cell formation. Inhibiting YAP during viral infection prevented dysplastic cell formation, whereas inhibiting YAP in persistent KRT5+ cells led to their conversion into distal club cells. Importantly, human dysplastic cells exhibited elevated FAK and YAP activity, and IFN-γ treatment promoted the transformation of human alveolar progenitor cells into dysplastic cells. These findings uncover the role of infection-induced inflammatory response in alveolar remodeling and may provide potential therapeutic avenues for the treatment of alveolar remodeling in patients with severe viral pneumonia.
Collapse
Affiliation(s)
- Xiuyu Lin
- State Key Laboratory of Multi-Cell Systems, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai , University of Chinese Academy of Sciences, China
| | - Weicheng Chen
- Cardiothoracic Surgery Department, Children’s Hospital of Fudan University, Shanghai, China
| | - Guilin Yang
- State Key Laboratory of Multi-Cell Systems, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai , University of Chinese Academy of Sciences, China
| | - Jiazhu Zhang
- State Key Laboratory of Multi-Cell Systems, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai , University of Chinese Academy of Sciences, China
| | - Huilin Wang
- Key Laboratory of Immune Response and Immunotherapy, Shanghai Institute of Immunity and Infection, Chinese Academy of Sciences, Shanghai, China
| | - Zeyu Liu
- Department of Respiratory Medicine, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai, China
| | - Ying Xi
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Tao Ren
- Department of Respiratory Medicine, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai, China
| | - Bo Liu
- Key Laboratory of Immune Response and Immunotherapy, Shanghai Institute of Immunity and Infection, Chinese Academy of Sciences, Shanghai, China
| | - Pengfei Sui
- State Key Laboratory of Multi-Cell Systems, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai , University of Chinese Academy of Sciences, China
| |
Collapse
|
6
|
Rajeev S, Li S, Leon-Coria A, Wang A, Kraemer L, Wang SJ, Boim A, Flannigan K, Shute A, Baggio CH, Callejas BE, MacNaughton WK, Finney CAM, McKay DM. Enteric tuft cells coordinate timely expulsion of the tapeworm Hymenolepis diminuta from the murine host by coordinating local but not systemic immunity. PLoS Pathog 2024; 20:e1012381. [PMID: 39083533 PMCID: PMC11290655 DOI: 10.1371/journal.ppat.1012381] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Accepted: 06/28/2024] [Indexed: 08/02/2024] Open
Abstract
Recognizing that enteric tuft cells can signal the presence of nematode parasites, we investigated whether tuft cells are required for the expulsion of the cestode, Hymenolepis diminuta, from the non-permissive mouse host, and in concomitant anti-helminthic responses. BALB/c and C57BL/6 mice infected with H. diminuta expelled the worms by 11 days post-infection (dpi) and displayed DCLK1+ (doublecortin-like kinase 1) tuft cell hyperplasia in the small intestine (not the colon) at 11 dpi. This tuft cell hyperplasia was dependent on IL-4Rα signalling and adaptive immunity, but not the microbiota. Expulsion of H. diminuta was slowed until at least 14 dpi, but not negated, in tuft cell-deficient Pou2f3-/- mice and was accompanied by delayed goblet cell hyperplasia and slowed small bowel transit. Worm antigen and mitogen evoked production of IL-4 and IL-10 by splenocytes from wild-type and Pou2f3-/- mice was not appreciably different, suggesting similar systemic immune reactivity to infection with H. diminuta. Wild-type and Pou2f3-/- mice infected with H. diminuta displayed partial protection against subsequent infection with the nematode Heligmosomoides bakeri. We speculate that, with respect to H. diminuta, enteric tuft cells are important for local immune events driving the rapidity of H. diminuta expulsion but are not critical in initiating or sustaining systemic Th2 responses that provide concomitant immunity against secondary infection with H. bakeri.
Collapse
Affiliation(s)
- Sruthi Rajeev
- Gastrointestinal Research Group and Inflammation Research Network, Department of Physiology and Pharmacology, Calvin, Phoebe and Joan Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
- Host-Parasite Interactions Program, University of Calgary, Calgary, Alberta, Canada
| | - ShuHua Li
- Gastrointestinal Research Group and Inflammation Research Network, Department of Physiology and Pharmacology, Calvin, Phoebe and Joan Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
- Host-Parasite Interactions Program, University of Calgary, Calgary, Alberta, Canada
| | - Aralia Leon-Coria
- Host-Parasite Interactions Program, University of Calgary, Calgary, Alberta, Canada
- Department of Biology, Faculty of Science, University of Calgary, Calgary, Alberta, Canada
| | - Arthur Wang
- Gastrointestinal Research Group and Inflammation Research Network, Department of Physiology and Pharmacology, Calvin, Phoebe and Joan Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
- Host-Parasite Interactions Program, University of Calgary, Calgary, Alberta, Canada
| | - Lucas Kraemer
- Gastrointestinal Research Group and Inflammation Research Network, Department of Physiology and Pharmacology, Calvin, Phoebe and Joan Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
- Host-Parasite Interactions Program, University of Calgary, Calgary, Alberta, Canada
| | - Susan Joanne Wang
- Gastrointestinal Research Group and Inflammation Research Network, Department of Physiology and Pharmacology, Calvin, Phoebe and Joan Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
- Host-Parasite Interactions Program, University of Calgary, Calgary, Alberta, Canada
| | - Annaliese Boim
- Gastrointestinal Research Group and Inflammation Research Network, Department of Physiology and Pharmacology, Calvin, Phoebe and Joan Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
- Host-Parasite Interactions Program, University of Calgary, Calgary, Alberta, Canada
| | - Kyle Flannigan
- Gastrointestinal Research Group and Inflammation Research Network, Department of Physiology and Pharmacology, Calvin, Phoebe and Joan Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Adam Shute
- Gastrointestinal Research Group and Inflammation Research Network, Department of Physiology and Pharmacology, Calvin, Phoebe and Joan Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
- Host-Parasite Interactions Program, University of Calgary, Calgary, Alberta, Canada
| | - Cristiane H. Baggio
- Gastrointestinal Research Group and Inflammation Research Network, Department of Physiology and Pharmacology, Calvin, Phoebe and Joan Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Blanca E. Callejas
- Gastrointestinal Research Group and Inflammation Research Network, Department of Physiology and Pharmacology, Calvin, Phoebe and Joan Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Wallace K. MacNaughton
- Gastrointestinal Research Group and Inflammation Research Network, Department of Physiology and Pharmacology, Calvin, Phoebe and Joan Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Constance A. M. Finney
- Host-Parasite Interactions Program, University of Calgary, Calgary, Alberta, Canada
- Department of Biology, Faculty of Science, University of Calgary, Calgary, Alberta, Canada
| | - Derek M. McKay
- Gastrointestinal Research Group and Inflammation Research Network, Department of Physiology and Pharmacology, Calvin, Phoebe and Joan Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
- Host-Parasite Interactions Program, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
7
|
Li YH, Yang YS, Xue YB, Lei H, Zhang SS, Qian J, Yao Y, Zhou R, Huang L. G protein subunit G γ13-mediated signaling pathway is critical to the inflammation resolution and functional recovery of severely injured lungs. eLife 2024; 12:RP92956. [PMID: 38836551 DOI: 10.7554/elife.92956] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/06/2024] Open
Abstract
Tuft cells are a group of rare epithelial cells that can detect pathogenic microbes and parasites. Many of these cells express signaling proteins initially found in taste buds. It is, however, not well understood how these taste signaling proteins contribute to the response to the invading pathogens or to the recovery of injured tissues. In this study, we conditionally nullified the signaling G protein subunit Gγ13 and found that the number of ectopic tuft cells in the injured lung was reduced following the infection of the influenza virus H1N1. Furthermore, the infected mutant mice exhibited significantly larger areas of lung injury, increased macrophage infiltration, severer pulmonary epithelial leakage, augmented pyroptosis and cell death, greater bodyweight loss, slower recovery, worsened fibrosis and increased fatality. Our data demonstrate that the Gγ13-mediated signal transduction pathway is critical to tuft cells-mediated inflammation resolution and functional repair of the damaged lungs.To our best knowledge, it is the first report indicating subtype-specific contributions of tuft cells to the resolution and recovery.
Collapse
Affiliation(s)
- Yi-Hong Li
- College of Life Sciences, Zhejiang University, Hangzhou, China
| | - Yi-Sen Yang
- College of Life Sciences, Zhejiang University, Hangzhou, China
| | - Yan-Bo Xue
- College of Life Sciences, Zhejiang University, Hangzhou, China
| | - Hao Lei
- College of Life Sciences, Zhejiang University, Hangzhou, China
| | - Sai-Sai Zhang
- College of Life Sciences, Zhejiang University, Hangzhou, China
| | - Junbin Qian
- Zhejiang Provincial Key Laboratory of Precision Diagnosis and Therapy for Major Gynecological Diseases, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Institute of Genetics, Zhejiang University School of Medicine, Hangzhou, China
- Cancer Center, Zhejiang University, Hangzhou, China
| | - Yushi Yao
- Institute of Immunology and Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Ruhong Zhou
- College of Life Sciences, Zhejiang University, Hangzhou, China
- Zhejiang University Shanghai Institute for Advanced Study, Shanghai, Shanghai, China
| | - Liquan Huang
- College of Life Sciences, Zhejiang University, Hangzhou, China
- Zhejiang University Shanghai Institute for Advanced Study, Shanghai, Shanghai, China
- Monell Chemical Senses Center, Philadelphia, United States
| |
Collapse
|
8
|
Emanuel E, Arifuzzaman M, Artis D. Epithelial-neuronal-immune cell interactions: Implications for immunity, inflammation, and tissue homeostasis at mucosal sites. J Allergy Clin Immunol 2024; 153:1169-1180. [PMID: 38369030 PMCID: PMC11070312 DOI: 10.1016/j.jaci.2024.02.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Revised: 02/13/2024] [Accepted: 02/14/2024] [Indexed: 02/20/2024]
Abstract
The epithelial lining of the respiratory tract and intestine provides a critical physical barrier to protect host tissues against environmental insults, including dietary antigens, allergens, chemicals, and microorganisms. In addition, specialized epithelial cells communicate directly with hematopoietic and neuronal cells. These epithelial-immune and epithelial-neuronal interactions control host immune responses and have important implications for inflammatory conditions associated with defects in the epithelial barrier, including asthma, allergy, and inflammatory bowel diseases. In this review, we discuss emerging research that identifies the mechanisms and impact of epithelial-immune and epithelial-neuronal cross talk in regulating immunity, inflammation, and tissue homeostasis at mucosal barrier surfaces. Understanding the regulation and impact of these pathways could provide new therapeutic targets for inflammatory diseases at mucosal sites.
Collapse
Affiliation(s)
- Elizabeth Emanuel
- Jill Roberts Institute for Research in Inflammatory Bowel Disease, Joan and Sanford I. Weill Department of Medicine, Department of Microbiology and Immunology, Weill Cornell Medicine, New York, NY; Immunology and Microbial Pathogenesis Program, Weill Cornell Medicine, New York, NY
| | - Mohammad Arifuzzaman
- Jill Roberts Institute for Research in Inflammatory Bowel Disease, Joan and Sanford I. Weill Department of Medicine, Department of Microbiology and Immunology, Weill Cornell Medicine, New York, NY
| | - David Artis
- Jill Roberts Institute for Research in Inflammatory Bowel Disease, Joan and Sanford I. Weill Department of Medicine, Department of Microbiology and Immunology, Weill Cornell Medicine, New York, NY; Immunology and Microbial Pathogenesis Program, Weill Cornell Medicine, New York, NY; Friedman Center for Nutrition and Inflammation, Joan and Sanford I. Weill Department of Medicine, Department of Microbiology and Immunology, Weill Cornell Medicine, New York, NY; Allen Discovery Center for Neuroimmune Interactions, New York, NY; Department of Microbiology and Immunology, Weill Cornell Medicine, New York, NY.
| |
Collapse
|
9
|
McCauley KB, Kukreja K, Tovar Walker AE, Jaffe AB, Klein AM. A map of signaling responses in the human airway epithelium. Cell Syst 2024; 15:307-321.e10. [PMID: 38508187 PMCID: PMC11031335 DOI: 10.1016/j.cels.2024.02.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 11/14/2023] [Accepted: 02/28/2024] [Indexed: 03/22/2024]
Abstract
Receptor-mediated signaling plays a central role in tissue regeneration, and it is dysregulated in disease. Here, we build a signaling-response map for a model regenerative human tissue: the airway epithelium. We analyzed the effect of 17 receptor-mediated signaling pathways on organotypic cultures to determine changes in abundance and phenotype of epithelial cell types. This map recapitulates the gamut of known airway epithelial signaling responses to these pathways. It defines convergent states induced by multiple ligands and diverse, ligand-specific responses in basal cell and secretory cell metaplasia. We show that loss of canonical differentiation induced by multiple pathways is associated with cell-cycle arrest, but that arrest is not sufficient to block differentiation. Using the signaling-response map, we show that a TGFB1-mediated response underlies specific aberrant cells found in multiple lung diseases and identify interferon responses in COVID-19 patient samples. Thus, we offer a framework enabling systematic evaluation of tissue signaling responses. A record of this paper's transparent peer review process is included in the supplemental information.
Collapse
Affiliation(s)
- Katherine B McCauley
- Department of Systems Biology, Harvard Medical School, Boston, MA 02115, USA; Respiratory Diseases, Novartis Institutes for BioMedical Research, Cambridge, MA 02139, USA; Disease Area X, Biomedical Research, Novartis, Cambridge, MA 02139, USA
| | - Kalki Kukreja
- Department of Systems Biology, Harvard Medical School, Boston, MA 02115, USA
| | | | - Aron B Jaffe
- Respiratory Diseases, Novartis Institutes for BioMedical Research, Cambridge, MA 02139, USA
| | - Allon M Klein
- Department of Systems Biology, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
10
|
Barr J, Xu L, Li R, Verheyden J, Sun X. Airway Microfold Cells Emerge in the Post-Influenza Lung. Am J Respir Cell Mol Biol 2024; 70:322-325. [PMID: 38127584 PMCID: PMC11478125 DOI: 10.1165/rcmb.2023-0351le] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Accepted: 12/21/2023] [Indexed: 12/23/2023] Open
Affiliation(s)
- Justinn Barr
- University of California, San DiegoLa Jolla, California
| | - Le Xu
- University of California, San DiegoLa Jolla, California
| | - Rongbo Li
- University of California, San DiegoLa Jolla, California
| | | | - Xin Sun
- University of California, San DiegoLa Jolla, California
| |
Collapse
|
11
|
Strine MS, Fagerberg E, Darcy PW, Barrón GM, Filler RB, Alfajaro MM, D'Angelo-Gavrish N, Wang F, Graziano VR, Menasché BL, Damo M, Wang YT, Howitt MR, Lee S, Joshi NS, Mucida D, Wilen CB. Intestinal tuft cell immune privilege enables norovirus persistence. Sci Immunol 2024; 9:eadi7038. [PMID: 38517952 PMCID: PMC11555782 DOI: 10.1126/sciimmunol.adi7038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Accepted: 02/28/2024] [Indexed: 03/24/2024]
Abstract
The persistent murine norovirus strain MNVCR6 is a model for human norovirus and enteric viral persistence. MNVCR6 causes chronic infection by directly infecting intestinal tuft cells, rare chemosensory epithelial cells. Although MNVCR6 induces functional MNV-specific CD8+ T cells, these lymphocytes fail to clear infection. To examine how tuft cells promote immune escape, we interrogated tuft cell interactions with CD8+ T cells by adoptively transferring JEDI (just EGFP death inducing) CD8+ T cells into Gfi1b-GFP tuft cell reporter mice. Unexpectedly, some intestinal tuft cells partially resisted JEDI CD8+ T cell-mediated killing-unlike Lgr5+ intestinal stem cells and extraintestinal tuft cells-despite seemingly normal antigen presentation. When targeting intestinal tuft cells, JEDI CD8+ T cells predominantly adopted a T resident memory phenotype with decreased effector and cytotoxic capacity, enabling tuft cell survival. JEDI CD8+ T cells neither cleared nor prevented MNVCR6 infection in the colon, the site of viral persistence, despite targeting a virus-independent antigen. Ultimately, we show that intestinal tuft cells are relatively resistant to CD8+ T cells independent of norovirus infection, representing an immune-privileged niche that can be leveraged by enteric microbes.
Collapse
Affiliation(s)
- Madison S Strine
- Department of Immunobiology, Yale School of Medicine, New Haven, CT, USA
| | - Eric Fagerberg
- Department of Immunobiology, Yale School of Medicine, New Haven, CT, USA
| | - Patrick W Darcy
- Laboratory of Mucosal Immunology, Rockefeller University, New York, NY, USA
| | - Gabriel M Barrón
- Program in Immunology, Stanford University, Stanford, CA, USA
- Department of Pathology, Stanford University, Stanford, CA, USA
| | - Renata B Filler
- Department of Laboratory Medicine, Yale School of Medicine, New Haven, CT, USA
| | - Mia Madel Alfajaro
- Department of Immunobiology, Yale School of Medicine, New Haven, CT, USA
- Department of Laboratory Medicine, Yale School of Medicine, New Haven, CT, USA
| | | | - Fang Wang
- Department of Immunobiology, Yale School of Medicine, New Haven, CT, USA
| | - Vincent R Graziano
- Department of Immunology, School of Medicine, UConn Health, Farmington, CT, USA
| | - Bridget L Menasché
- Department of Immunobiology, Yale School of Medicine, New Haven, CT, USA
- Department of Laboratory Medicine, Yale School of Medicine, New Haven, CT, USA
| | - Martina Damo
- Department of Immunobiology, Yale School of Medicine, New Haven, CT, USA
| | - Ya-Ting Wang
- SXMU-Tsinghua Collaborative Innovation Center for Frontier Medicine, Tsinghua University School of Medicine, Beijing, China
| | - Michael R Howitt
- Program in Immunology, Stanford University, Stanford, CA, USA
- Department of Pathology, Stanford University, Stanford, CA, USA
- Department of Microbiology and Immunology, Stanford University, Stanford, CA, USA
| | - Sanghyun Lee
- Department of Molecular Microbiology and Immunology, Brown University, Providence, RI, USA
| | - Nikhil S Joshi
- Department of Immunobiology, Yale School of Medicine, New Haven, CT, USA
| | - Daniel Mucida
- Laboratory of Mucosal Immunology, Rockefeller University, New York, NY, USA
- Howard Hughes Medical Institute, Rockefeller University, New York, NY, USA
| | - Craig B Wilen
- Department of Immunobiology, Yale School of Medicine, New Haven, CT, USA
- Department of Laboratory Medicine, Yale School of Medicine, New Haven, CT, USA
- Yale Cancer Center, Yale School of Medicine, New Haven, CT, USA
| |
Collapse
|
12
|
Solta A, Ernhofer B, Boettiger K, Megyesfalvi Z, Heeke S, Hoda MA, Lang C, Aigner C, Hirsch FR, Schelch K, Döme B. Small cells - big issues: biological implications and preclinical advancements in small cell lung cancer. Mol Cancer 2024; 23:41. [PMID: 38395864 PMCID: PMC10893629 DOI: 10.1186/s12943-024-01953-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Accepted: 01/31/2024] [Indexed: 02/25/2024] Open
Abstract
Current treatment guidelines refer to small cell lung cancer (SCLC), one of the deadliest human malignancies, as a homogeneous disease. Accordingly, SCLC therapy comprises chemoradiation with or without immunotherapy. Meanwhile, recent studies have made significant advances in subclassifying SCLC based on the elevated expression of the transcription factors ASCL1, NEUROD1, and POU2F3, as well as on certain inflammatory characteristics. The role of the transcription regulator YAP1 in defining a unique SCLC subset remains to be established. Although preclinical analyses have described numerous subtype-specific characteristics and vulnerabilities, the so far non-existing clinical subtype distinction may be a contributor to negative clinical trial outcomes. This comprehensive review aims to provide a framework for the development of novel personalized therapeutic approaches by compiling the most recent discoveries achieved by preclinical SCLC research. We highlight the challenges faced due to limited access to patient material as well as the advances accomplished by implementing state-of-the-art models and methodologies.
Collapse
Affiliation(s)
- Anna Solta
- Department of Thoracic Surgery, Comprehensive Cancer Center, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria
| | - Büsra Ernhofer
- Department of Thoracic Surgery, Comprehensive Cancer Center, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria
| | - Kristiina Boettiger
- Department of Thoracic Surgery, Comprehensive Cancer Center, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria
| | - Zsolt Megyesfalvi
- Department of Thoracic Surgery, Comprehensive Cancer Center, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria
- Department of Thoracic Surgery, Semmelweis University and National Institute of Oncology, Budapest, Hungary
- National Koranyi Institute of Pulmonology, Budapest, Hungary
| | - Simon Heeke
- Department of Thoracic Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Mir Alireza Hoda
- Department of Thoracic Surgery, Comprehensive Cancer Center, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria
| | - Christian Lang
- Department of Thoracic Surgery, Comprehensive Cancer Center, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria
- Division of Pulmonology, Department of Medicine II, Medical University of Vienna, Vienna, Austria
| | - Clemens Aigner
- Department of Thoracic Surgery, Comprehensive Cancer Center, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria
| | - Fred R Hirsch
- Division of Medical Oncology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA.
- Center for Thoracic Oncology, Mount Sinai Health System, Tisch Cancer Institute, New York, NY, USA.
| | - Karin Schelch
- Department of Thoracic Surgery, Comprehensive Cancer Center, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria
- Center for Cancer Research, Medical University of Vienna, Vienna, Austria
| | - Balazs Döme
- Department of Thoracic Surgery, Comprehensive Cancer Center, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria.
- Department of Thoracic Surgery, Semmelweis University and National Institute of Oncology, Budapest, Hungary.
- National Koranyi Institute of Pulmonology, Budapest, Hungary.
- Department of Translational Medicine, Lund University, Lund, Sweden.
| |
Collapse
|
13
|
Hoki M, Yamada Y, Hiratomo E, Hirata M, Takeuchi Y, Yoshimatsu M, Kikuchi M, Kishimoto Y, Marx A, Haga H. Expression of FOXI1 and POU2F3 varies among different salivary gland neoplasms and is higher in Warthin tumor. Discov Oncol 2024; 15:36. [PMID: 38358561 PMCID: PMC10869675 DOI: 10.1007/s12672-024-00892-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Accepted: 02/11/2024] [Indexed: 02/16/2024] Open
Abstract
PURPOSE Salivary gland tumors are histologically diverse. Ionocytes and tuft cells, rare epithelial cells found in normal salivary glands, might be associated with salivary tumors. Here, we explored the expression of FOXI1 and POU2F3, master regulators of ionocytes and tuft cells, respectively, for common salivary neoplasms using immunohistochemistry. METHODS We analyzed normal salivary tissues and nine salivary gland tumors; Warthin tumors (WT), pleomorphic adenomas (PA), basal cell adenomas, and oncocytomas were benign, whereas mucoepidermoid, adenoid cystic, acinic cell, salivary duct carcinomas, and polymorphous adenocarcinomas were malignant. RESULTS Normal salivary glands contained a few FOXI1- and POU2F3-positive cells in the ducts instead of the acini, consistent with ionocytes and tuft cells, respectively. Among the benign tumors, only WTs and PAs consistently expressed FOXI1 (10/10 and 9/10, respectively). The median H-score of WTs was significantly higher than that of PAs (17.5 vs. 4, P = 0.01). While WTs and PAs harbored POU2F3-positive cells (10/10 and 9/10, respectively), the median H-score was higher in WTs than in PAs (10.5 vs 4, respectively). Furthermore, WTs exhibited a unique staining pattern of FOXI1- and POU2F3-positive cells, which were present in luminal and abluminal locations, respectively. Whereas none of the malignant tumors expressed FOXI1, only adenoid cystic carcinoma consistently expressed POU2F3 (5/5), with a median H-score of 4. CONCLUSION The expression patterns of the characteristic transcription factors found in ionocytes and tuft cells vary among salivary gland tumor types and are higher in WT, which might be relevant for understanding and diagnosing salivary gland neoplasms.
Collapse
Affiliation(s)
- Masahito Hoki
- Department of Diagnostic Pathology, Kyoto University Hospital, 54 Shogoin Kawahara-cho, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Yosuke Yamada
- Department of Diagnostic Pathology, Kyoto University Hospital, 54 Shogoin Kawahara-cho, Sakyo-ku, Kyoto, 606-8507, Japan.
| | - Emi Hiratomo
- Department of Diagnostic Pathology, Kyoto University Hospital, 54 Shogoin Kawahara-cho, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Masahiro Hirata
- Department of Diagnostic Pathology, Kyoto University Hospital, 54 Shogoin Kawahara-cho, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Yasuhide Takeuchi
- Department of Diagnostic Pathology, Kyoto University Hospital, 54 Shogoin Kawahara-cho, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Masayoshi Yoshimatsu
- Department of Otolaryngology, Head and Neck Surgery, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima, Japan
| | - Masahiro Kikuchi
- Department of Otolaryngology-Head & Neck Surgery, Kobe City Medical Center General Hospital, Kobe, Japan
| | - Yo Kishimoto
- Department of Otolaryngology, Head and Neck Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Alexander Marx
- Institute of Pathology, University Medical Center Göttingen, University of Göttingen, Göttingen, Germany
| | - Hironori Haga
- Department of Diagnostic Pathology, Kyoto University Hospital, 54 Shogoin Kawahara-cho, Sakyo-ku, Kyoto, 606-8507, Japan
| |
Collapse
|
14
|
Silverman JB, Vega PN, Tyska MJ, Lau KS. Intestinal Tuft Cells: Morphology, Function, and Implications for Human Health. Annu Rev Physiol 2024; 86:479-504. [PMID: 37863104 PMCID: PMC11193883 DOI: 10.1146/annurev-physiol-042022-030310] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2023]
Abstract
Tuft cells are a rare and morphologically distinct chemosensory cell type found throughout many organs, including the gastrointestinal tract. These cells were identified by their unique morphologies distinguished by large apical protrusions. Ultrastructural data have begun to describe the molecular underpinnings of their cytoskeletal features, and tuft cell-enriched cytoskeletal proteins have been identified, although the connection of tuft cell morphology to tuft cell functionality has not yet been established. Furthermore, tuft cells display variations in function and identity between and within tissues, leading to the delineation of distinct tuft cell populations. As a chemosensory cell type, they display receptors that are responsive to ligands specific for their environment. While many studies have demonstrated the tuft cell response to protists and helminths in the intestine, recent research has highlighted other roles of tuft cells as well as implicated tuft cells in other disease processes including inflammation, cancer, and viral infections. Here, we review the literature on the cytoskeletal structure of tuft cells. Additionally, we focus on new research discussing tuft cell lineage, ligand-receptor interactions, tuft cell tropism, and the role of tuft cells in intestinal disease. Finally, we discuss the implication of tuft cell-targeted therapies in human health and how the morphology of tuft cells may contribute to their functionality.
Collapse
Affiliation(s)
- Jennifer B Silverman
- Epithelial Biology Center and Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, Tennessee, USA; ,
| | - Paige N Vega
- Epithelial Biology Center and Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, Tennessee, USA; ,
| | - Matthew J Tyska
- Epithelial Biology Center and Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, Tennessee, USA; ,
| | - Ken S Lau
- Epithelial Biology Center and Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, Tennessee, USA; ,
| |
Collapse
|
15
|
Ualiyeva S, Lemire E, Wong C, Perniss A, Boyd A, Avilés EC, Minichetti DG, Maxfield A, Roditi R, Matsumoto I, Wang X, Deng W, Barrett NA, Buchheit KM, Laidlaw TM, Boyce JA, Bankova LG, Haber AL. A nasal cell atlas reveals heterogeneity of tuft cells and their role in directing olfactory stem cell proliferation. Sci Immunol 2024; 9:eabq4341. [PMID: 38306414 PMCID: PMC11127180 DOI: 10.1126/sciimmunol.abq4341] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Accepted: 12/08/2023] [Indexed: 02/04/2024]
Abstract
The olfactory neuroepithelium serves as a sensory organ for odors and forms part of the nasal mucosal barrier. Olfactory sensory neurons are surrounded and supported by epithelial cells. Among them, microvillous cells (MVCs) are strategically positioned at the apical surface, but their specific functions are enigmatic, and their relationship to the other specialized epithelial cells is unclear. Here, we establish that the family of MVCs comprises tuft cells and ionocytes in both mice and humans. Integrating analysis of the respiratory and olfactory epithelia, we define the distinct receptor expression of TRPM5+ tuft-MVCs compared with Gɑ-gustducinhigh respiratory tuft cells and characterize a previously undescribed population of glandular DCLK1+ tuft cells. To establish how allergen sensing by tuft-MVCs might direct olfactory mucosal responses, we used an integrated single-cell transcriptional and protein analysis. Inhalation of Alternaria induced mucosal epithelial effector molecules including Chil4 and a distinct pathway leading to proliferation of the quiescent olfactory horizontal basal stem cell (HBC) pool, both triggered in the absence of olfactory apoptosis. Alternaria- and ATP-elicited HBC proliferation was dependent on TRPM5+ tuft-MVCs, identifying these specialized epithelial cells as regulators of olfactory stem cell responses. Together, our data provide high-resolution characterization of nasal tuft cell heterogeneity and identify a function of TRPM5+ tuft-MVCs in directing the olfactory mucosal response to allergens.
Collapse
Affiliation(s)
- Saltanat Ualiyeva
- Division of Allergy and Clinical Immunology, Jeff and Penny Vinik Center for Allergic Disease Research, Brigham & Women’s Hospital and Department of Medicine, Harvard Medical School, Boston, MA
| | - Evan Lemire
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA
| | - Caitlin Wong
- Division of Allergy and Clinical Immunology, Jeff and Penny Vinik Center for Allergic Disease Research, Brigham & Women’s Hospital and Department of Medicine, Harvard Medical School, Boston, MA
| | - Alexander Perniss
- Division of Allergy and Clinical Immunology, Jeff and Penny Vinik Center for Allergic Disease Research, Brigham & Women’s Hospital and Department of Medicine, Harvard Medical School, Boston, MA
| | - Amelia Boyd
- Division of Allergy and Clinical Immunology, Jeff and Penny Vinik Center for Allergic Disease Research, Brigham & Women’s Hospital and Department of Medicine, Harvard Medical School, Boston, MA
| | - Evelyn C. Avilés
- Department of Neurobiology, Harvard Medical School, Boston, MA; currently at Faculty of Biological Sciences, Pontificia Universidad Católica de Chile
| | - Dante G. Minichetti
- Division of Allergy and Clinical Immunology, Jeff and Penny Vinik Center for Allergic Disease Research, Brigham & Women’s Hospital and Department of Medicine, Harvard Medical School, Boston, MA
| | - Alice Maxfield
- Division of Otolaryngology-Head and Neck Surgery, Brigham and Women’s Hospital and Department of Otolaryngology-Head and Neck Surgery, Harvard Medical School, Boston, MA
| | - Rachel Roditi
- Division of Otolaryngology-Head and Neck Surgery, Brigham and Women’s Hospital and Department of Otolaryngology-Head and Neck Surgery, Harvard Medical School, Boston, MA
| | | | - Xin Wang
- Division of Allergy and Clinical Immunology, Jeff and Penny Vinik Center for Allergic Disease Research, Brigham & Women’s Hospital and Department of Medicine, Harvard Medical School, Boston, MA
| | - Wenjiang Deng
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA
| | - Nora A. Barrett
- Division of Allergy and Clinical Immunology, Jeff and Penny Vinik Center for Allergic Disease Research, Brigham & Women’s Hospital and Department of Medicine, Harvard Medical School, Boston, MA
| | - Kathleen M. Buchheit
- Division of Allergy and Clinical Immunology, Jeff and Penny Vinik Center for Allergic Disease Research, Brigham & Women’s Hospital and Department of Medicine, Harvard Medical School, Boston, MA
| | - Tanya M. Laidlaw
- Division of Allergy and Clinical Immunology, Jeff and Penny Vinik Center for Allergic Disease Research, Brigham & Women’s Hospital and Department of Medicine, Harvard Medical School, Boston, MA
| | - Joshua A. Boyce
- Division of Allergy and Clinical Immunology, Jeff and Penny Vinik Center for Allergic Disease Research, Brigham & Women’s Hospital and Department of Medicine, Harvard Medical School, Boston, MA
| | - Lora G. Bankova
- Division of Allergy and Clinical Immunology, Jeff and Penny Vinik Center for Allergic Disease Research, Brigham & Women’s Hospital and Department of Medicine, Harvard Medical School, Boston, MA
| | - Adam L. Haber
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA
| |
Collapse
|
16
|
Li Y, Han M, Singh S, Breckenridge HA, Kreger JE, Stroupe CC, Sawicky DA, Kuo S, Goldsmith AM, Ke F, Shenoy AT, Bentley JK, Matsumoto I, Hershenson MB. Tuft cells are required for a rhinovirus-induced asthma phenotype in immature mice. JCI Insight 2024; 9:e166136. [PMID: 38061015 PMCID: PMC10906234 DOI: 10.1172/jci.insight.166136] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Accepted: 12/05/2023] [Indexed: 01/17/2024] Open
Abstract
Infection of immature mice with rhinovirus (RV) induces an asthma-like phenotype consisting of type 2 inflammation, mucous metaplasia, eosinophilic inflammation, and airway hyperresponsiveness that is dependent on IL-25 and type 2 innate lymphoid cells (ILC2s). Doublecortin-like kinase 1-positive (DCLK1+) tuft cells are a major source of IL-25. We sought to determine the requirement of tuft cells for the RV-induced asthma phenotype in wild-type mice and mice deficient in Pou2f3, a transcription factor required for tuft cell development. C57BL/6J mice infected with RV-A1B on day 6 of life and RV-A2 on day 13 of life showed increased DCLK1+ tuft cells in the large airways. Compared with wild-type mice, RV-infected Pou2f3-/- mice showed reductions in IL-25 mRNA and protein expression, ILC2 expansion, type 2 cytokine expression, mucous metaplasia, lung eosinophils, and airway methacholine responsiveness. We conclude that airway tuft cells are required for the asthma phenotype observed in immature mice undergoing repeated RV infections. Furthermore, RV-induced tuft cell development provides a mechanism by which early-life viral infections could potentiate type 2 inflammatory responses to future infections.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Fang Ke
- Department of Microbiology and Immunology, and
| | - Anukul T. Shenoy
- Department of Microbiology and Immunology, and
- Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | | | | | - Marc B. Hershenson
- Department of Pediatrics
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| |
Collapse
|
17
|
Purev E, Bahmed K, Kosmider B. Alveolar Organoids in Lung Disease Modeling. Biomolecules 2024; 14:115. [PMID: 38254715 PMCID: PMC10813493 DOI: 10.3390/biom14010115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Revised: 01/06/2024] [Accepted: 01/11/2024] [Indexed: 01/24/2024] Open
Abstract
Lung organoids display a tissue-specific functional phenomenon and mimic the features of the original organ. They can reflect the properties of the cells, such as morphology, polarity, proliferation rate, gene expression, and genomic profile. Alveolar type 2 (AT2) cells have a stem cell potential in the adult lung. They produce and secrete pulmonary surfactant and proliferate to restore the epithelium after damage. Therefore, AT2 cells are used to generate alveolar organoids and can recapitulate distal lung structures. Also, AT2 cells in human-induced pluripotent stem cell (iPSC)-derived alveolospheres express surfactant proteins and other factors, indicating their application as suitable models for studying cell-cell interactions. Recently, they have been utilized to define mechanisms of disease development, such as COVID-19, lung cancer, idiopathic pulmonary fibrosis, and chronic obstructive pulmonary disease. In this review, we show lung organoid applications in various pulmonary diseases, drug screening, and personalized medicine. In addition, stem cell-based therapeutics and approaches relevant to lung repair were highlighted. We also described the signaling pathways and epigenetic regulation of lung regeneration. It is critical to identify novel regulators of alveolar organoid generations to promote lung repair in pulmonary diseases.
Collapse
Affiliation(s)
- Enkhee Purev
- Department of Microbiology, Immunology, and Inflammation, Temple University, Philadelphia, PA 19140, USA
- Center for Inflammation and Lung Research, Temple University, Philadelphia, PA 19140, USA
| | - Karim Bahmed
- Department of Microbiology, Immunology, and Inflammation, Temple University, Philadelphia, PA 19140, USA
- Center for Inflammation and Lung Research, Temple University, Philadelphia, PA 19140, USA
- Department of Thoracic Medicine and Surgery, Temple University, Philadelphia, PA 19140, USA
| | - Beata Kosmider
- Department of Microbiology, Immunology, and Inflammation, Temple University, Philadelphia, PA 19140, USA
- Center for Inflammation and Lung Research, Temple University, Philadelphia, PA 19140, USA
- Department of Thoracic Medicine and Surgery, Temple University, Philadelphia, PA 19140, USA
- Department of Cardiovascular Sciences, Temple University, Philadelphia, PA 19140, USA
| |
Collapse
|
18
|
Chen XY, Kao C, Peng SW, Chang JH, Lee YL, Laiman V, Chung KF, Bhavsar PK, Heriyanto DS, Chuang KJ, Chuang HC. Role of DCLK1/Hippo pathway in type II alveolar epithelial cells differentiation in acute respiratory distress syndrome. Mol Med 2023; 29:159. [PMID: 37996782 PMCID: PMC10668445 DOI: 10.1186/s10020-023-00760-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Accepted: 11/16/2023] [Indexed: 11/25/2023] Open
Abstract
BACKGROUND Delay in type II alveolar epithelial cell (AECII) regeneration has been linked to higher mortality in patients with acute respiratory distress syndrome (ARDS). However, the interaction between Doublecortin-like kinase 1 (DCLK1) and the Hippo signaling pathway in ARDS-associated AECII differentiation remains unclear. Therefore, the objective of this study was to understand the role of the DCLK1/Hippo pathway in mediating AECII differentiation in ARDS. MATERIALS AND METHODS AECII MLE-12 cells were exposed to 0, 0.1, or 1 μg/mL of lipopolysaccharide (LPS) for 6 and 12 h. In the mouse model, C57BL/6JNarl mice were intratracheally (i.t.) injected with 0 (control) or 5 mg/kg LPS and were euthanized for lung collection on days 3 and 7. RESULTS We found that LPS induced AECII markers of differentiation by reducing surfactant protein C (SPC) and p53 while increasing T1α (podoplanin) and E-cadherin at 12 h. Concurrently, nuclear YAP dynamic regulation and increased TAZ levels were observed in LPS-exposed AECII within 12 h. Inhibition of YAP consistently decreased cell levels of SPC, claudin 4 (CLDN-4), galectin 3 (LGALS-3), and p53 while increasing transepithelial electrical resistance (TEER) at 6 h. Furthermore, DCLK1 expression was reduced in isolated human AECII of ARDS, consistent with the results in LPS-exposed AECII at 6 h and mouse SPC-positive (SPC+) cells after 3-day LPS exposure. We observed that downregulated DCLK1 increased p-YAP/YAP, while DCLK1 overexpression slightly reduced p-YAP/YAP, indicating an association between DCLK1 and Hippo-YAP pathway. CONCLUSIONS We conclude that DCLK1-mediated Hippo signaling components of YAP/TAZ regulated markers of AECII-to-AECI differentiation in an LPS-induced ARDS model.
Collapse
Affiliation(s)
- Xiao-Yue Chen
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, Taiwan
- School of Respiratory Therapy, College of Medicine, Taipei Medical University, 250 Wuxing Street, Taipei, 11031, Taiwan
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Ching Kao
- School of Respiratory Therapy, College of Medicine, Taipei Medical University, 250 Wuxing Street, Taipei, 11031, Taiwan
| | - Syue-Wei Peng
- School of Respiratory Therapy, College of Medicine, Taipei Medical University, 250 Wuxing Street, Taipei, 11031, Taiwan
| | - Jer-Hwa Chang
- School of Respiratory Therapy, College of Medicine, Taipei Medical University, 250 Wuxing Street, Taipei, 11031, Taiwan.
- Division of Pulmonary Medicine, Department of Internal Medicine, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan.
| | - Yueh-Lun Lee
- Department of Microbiology and Immunology, School of Medicine, College of Medicine, Taipei Medical University, 250 Wuxing Street, Taipei, 11031, Taiwan.
| | - Vincent Laiman
- International Ph.D. Program in Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Department of Anatomical Pathology, Faculty of Medicine, Public Health, and Nursing, Universitas Gadjah Mada, Dr. Sardjito Hospital, Yogyakarta, Indonesia
| | - Kian Fan Chung
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Pankaj K Bhavsar
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Didik Setyo Heriyanto
- Department of Anatomical Pathology, Faculty of Medicine, Public Health, and Nursing, Universitas Gadjah Mada, Dr. Sardjito Hospital, Yogyakarta, Indonesia
| | - Kai-Jen Chuang
- School of Public Health, College of Public Health, Taipei Medical University, Taipei, Taiwan
- Department of Public Health, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Hsiao-Chi Chuang
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, Taiwan.
- School of Respiratory Therapy, College of Medicine, Taipei Medical University, 250 Wuxing Street, Taipei, 11031, Taiwan.
- National Heart and Lung Institute, Imperial College London, London, UK.
- Division of Pulmonary Medicine, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan.
- Cell Physiology and Molecular Image Research Center, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan.
- Inhalation Toxicology Research Lab (ITRL), School of Respiratory Therapy, College of Medicine, Taipei Medical University, 250 Wuxing Street, Taipei, 110, Taiwan.
| |
Collapse
|
19
|
Cheon IS, Son YM, Sun J. Tissue-resident memory T cells and lung immunopathology. Immunol Rev 2023; 316:63-83. [PMID: 37014096 PMCID: PMC10524334 DOI: 10.1111/imr.13201] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Revised: 03/10/2023] [Accepted: 03/21/2023] [Indexed: 04/05/2023]
Abstract
Rapid reaction to microbes invading mucosal tissues is key to protect the host against disease. Respiratory tissue-resident memory T (TRM ) cells provide superior immunity against pathogen infection and/or re-infection, due to their presence at the site of pathogen entry. However, there has been emerging evidence that exuberant TRM -cell responses contribute to the development of various chronic respiratory conditions including pulmonary sequelae post-acute viral infections. In this review, we have described the characteristics of respiratory TRM cells and processes underlying their development and maintenance. We have reviewed TRM -cell protective functions against various respiratory pathogens as well as their pathological activities in chronic lung conditions including post-viral pulmonary sequelae. Furthermore, we have discussed potential mechanisms regulating the pathological activity of TRM cells and proposed therapeutic strategies to alleviate TRM -cell-mediated lung immunopathology. We hope that this review provides insights toward the development of future vaccines or interventions that can harness the superior protective abilities of TRM cells, while minimizing the potential for immunopathology, a particularly important topic in the era of coronavirus disease 2019 (COVID-19) pandemic.
Collapse
Affiliation(s)
- In Su Cheon
- Carter Immunology Center, University of Virginia, Charlottesville, VA 22908, USA
- Division of Infectious Disease and International Health, Department of Medicine, University of Virginia, Charlottesville, VA 22908, USA
| | - Young Min Son
- Department of Systems Biotechnology, Chung-Ang University, Anseong, Gyeonggi-do, Republic of Korea 17546
| | - Jie Sun
- Carter Immunology Center, University of Virginia, Charlottesville, VA 22908, USA
- Division of Infectious Disease and International Health, Department of Medicine, University of Virginia, Charlottesville, VA 22908, USA
| |
Collapse
|
20
|
Cable J, Sun J, Cheon IS, Vaughan AE, Castro IA, Stein SR, López CB, Gostic KM, Openshaw PJM, Ellebedy AH, Wack A, Hutchinson E, Thomas MM, Langlois RA, Lingwood D, Baker SF, Folkins M, Foxman EF, Ward AB, Schwemmle M, Russell AB, Chiu C, Ganti K, Subbarao K, Sheahan TP, Penaloza-MacMaster P, Eddens T. Respiratory viruses: New frontiers-a Keystone Symposia report. Ann N Y Acad Sci 2023; 1522:60-73. [PMID: 36722473 PMCID: PMC10580159 DOI: 10.1111/nyas.14958] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Respiratory viruses are a common cause of morbidity and mortality around the world. Viruses like influenza, RSV, and most recently SARS-CoV-2 can rapidly spread through a population, causing acute infection and, in vulnerable populations, severe or chronic disease. Developing effective treatment and prevention strategies often becomes a race against ever-evolving viruses that develop resistance, leaving therapy efficacy either short-lived or relevant for specific viral strains. On June 29 to July 2, 2022, researchers met for the Keystone symposium "Respiratory Viruses: New Frontiers." Researchers presented new insights into viral biology and virus-host interactions to understand the mechanisms of disease and identify novel treatment and prevention approaches that are effective, durable, and broad.
Collapse
Affiliation(s)
| | - Jie Sun
- Division of Pulmonary and Critical Medicine, Department of Medicine; Department of Immunology; and Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota, USA
- Carter Immunology Center and Division of Infectious Disease and International Health, Department of Medicine, University of Virginia, Charlottesville, Virginia, USA
| | - In Su Cheon
- Division of Pulmonary and Critical Medicine, Department of Medicine; Department of Immunology; and Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota, USA
- Carter Immunology Center and Division of Infectious Disease and International Health, Department of Medicine, University of Virginia, Charlottesville, Virginia, USA
| | - Andrew E Vaughan
- University of Pennsylvania School of Veterinary Medicine, Biomedical Sciences, Philadelphia, Pennsylvania, USA
| | - Italo A Castro
- Virology Research Center, Ribeirao Preto Medical School, University of São Paulo - USP, São Paulo, Brazil
| | - Sydney R Stein
- Emerging Pathogens Section, Critical Care Medicine Department, Clinical Center and Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Carolina B López
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Department of Molecular Microbiology and Center for Women Infectious Disease Research, Washington University School of Medicine, St Louis, Missouri, USA
| | - Katelyn M Gostic
- Department of Ecology and Evolution, University of Chicago, Chicago, Illinois, USA
| | | | - Ali H Ellebedy
- Department of Pathology and Immunology; The Andrew M. and Jane M. Bursky Center for Human Immunology & Immunotherapy Programs; and Center for Vaccines and Immunity to Microbial Pathogens, Washington University School of Medicine, St Louis, Missouri, USA
| | - Andreas Wack
- Immunoregulation Laboratory, The Francis Crick Institute, London, UK
| | | | | | - Ryan A Langlois
- Center for Immunology and Department of Microbiology and Immunology, University of Minnesota, Minneapolis, Minnesota, USA
| | - Daniel Lingwood
- The Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology, and Harvard University, Cambridge, Massachusetts, USA
| | - Steven F Baker
- Lovelace Biomedical Research Institute, Albuquerque, New Mexico, USA
| | - Melanie Folkins
- Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, Alberta, Canada
| | - Ellen F Foxman
- Department of Laboratory Medicine and Department of Immunology, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Andrew B Ward
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, California, USA
| | - Martin Schwemmle
- Institute of Virology, Freiburg University Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Alistair B Russell
- Division of Biological Sciences, University of California, San Diego, La Jolla, California, USA
| | - Christopher Chiu
- Department of Infectious Disease, Imperial College London, London, UK
| | - Ketaki Ganti
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Kanta Subbarao
- Department of Microbiology and Immunology, WHO Collaborating Centre for Reference and Research on Influenza at the Peter Doherty Institute for Infection and Immunity, University of Melbourne at The Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Timothy P Sheahan
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Pablo Penaloza-MacMaster
- Department of Microbiology-Immunology, School of Medicine, Northwestern University Feinberg, Chicago, Illinois, USA
| | - Taylor Eddens
- Pediatric Scientist Development Program, University of Pittsburgh Medical Center (UPMC) Children's Hospital of Pittsburgh, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
21
|
Mccauley KB, Kukreja K, Jaffe AB, Klein AM. A map of signaling responses in the human airway epithelium. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2022:2022.12.21.521460. [PMID: 36597531 PMCID: PMC9810218 DOI: 10.1101/2022.12.21.521460] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Receptor-mediated signaling plays a central role in tissue regeneration, and it is dysregulated in disease. Here, we build a signaling-response map for a model regenerative human tissue: the airway epithelium. We analyzed the effect of 17 receptor-mediated signaling pathways on organotypic cultures to determine changes in abundance and phenotype of all epithelial cell types. This map recapitulates the gamut of known airway epithelial signaling responses to these pathways. It defines convergent states induced by multiple ligands and diverse, ligand-specific responses in basal-cell and secretory-cell metaplasia. We show that loss of canonical differentiation induced by multiple pathways is associated with cell cycle arrest, but that arrest is not sufficient to block differentiation. Using the signaling-response map, we show that a TGFB1-mediated response underlies specific aberrant cells found in multiple lung diseases and identify interferon responses in COVID-19 patient samples. Thus, we offer a framework enabling systematic evaluation of tissue signaling responses.
Collapse
Affiliation(s)
- Katherine B Mccauley
- Department of Systems Biology, Harvard Medical School, Boston, MA, USA
- Disease Area X, Respiratory Therapeutic Area, Novartis Institutes for BioMedical Research, Cambridge, MA, USA
| | - Kalki Kukreja
- Department of Systems Biology, Harvard Medical School, Boston, MA, USA
| | - Aron B Jaffe
- Disease Area X, Respiratory Therapeutic Area, Novartis Institutes for BioMedical Research, Cambridge, MA, USA
- Current address: Chroma Medicine, Boston, MA, USA
| | - Allon M Klein
- Department of Systems Biology, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
22
|
Li L, Ma M, Duan T, Sui X. The critical roles and therapeutic implications of tuft cells in cancer. Front Pharmacol 2022; 13:1047188. [PMID: 36569325 PMCID: PMC9780677 DOI: 10.3389/fphar.2022.1047188] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2022] [Accepted: 11/21/2022] [Indexed: 12/13/2022] Open
Abstract
Tuft cells are solitary chemosensory epithelial cells with microvilli at the top, which are found in hollow organs such as the gastrointestinal tract, pancreas, and lungs. Recently, an increasing number of studies have revealed the chemotactic abilities and immune function of the tuft cells, and numerous efforts have been devoted to uncovering the role of tuft cells in tumors. Notably, accumulating evidence has shown that the specific genes (POU2F3, DCLK1) expressed in tuft cells are involved in vital processes related with carcinogenesis and cancer development. However, the interaction between the tuft cells and cancer remains to be further elucidated. Here, based on an introduction of biological functions and specific markers of the tuft cells, we have summarized the functional roles and potential therapeutic implications of tuft cells in cancers, including pancreatic cancer, lung cancer, gastric cancer, colon cancer, and liver cancer, which is in the hope of inspiring the future research in validating tuft cells as novel strategies for cancer therapies.
Collapse
Affiliation(s)
- Lin Li
- School of Pharmacy, Hangzhou Normal University, Hangzhou, China
| | - Mengmeng Ma
- School of Pharmacy, Hangzhou Normal University, Hangzhou, China
| | - Ting Duan
- School of Pharmacy, Hangzhou Normal University, Hangzhou, China
| | - Xinbing Sui
- School of Pharmacy, Hangzhou Normal University, Hangzhou, China
- Department of Medical Oncology, The Affiliated Hospital of Hangzhou Normal University, Hangzhou Normal University, Hangzhou, China
| |
Collapse
|