1
|
Xie Y, Wang S, Cha X, Li F, Xu Z, Wu J, Liu H, Ren W. Aging and chronic inflammation: impacts on olfactory dysfunction-a comprehensive review. Cell Mol Life Sci 2025; 82:199. [PMID: 40355677 PMCID: PMC12069206 DOI: 10.1007/s00018-025-05637-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2024] [Revised: 02/04/2025] [Accepted: 02/23/2025] [Indexed: 05/14/2025]
Abstract
Olfactory dysfunction (OD) is a common nasal disease, particularly prevalent among the elderly population, significantly impacting the affected individuals' quality of life. This review focuses on the influence of aging and chronic inflammation on olfactory dysfunction, presenting insights from both the peripheral and central olfactory systems. By exploring the molecular mechanisms and pathological changes underlying the occurrence of olfactory dysfunction in relation to age-related diseases and chronic inflammation conditions, we aim to provide a comprehensive theoretical foundation for further research and offer valuable insights for more effective treatment of olfactory dysfunction.
Collapse
Affiliation(s)
- Yingqi Xie
- Department of Otolaryngology, The Second Affiliated Hospital of the Naval Medical University, Shanghai Changzheng Hospital, Shanghai, China
| | - Shenglei Wang
- Department of Otolaryngology, The Second Affiliated Hospital of the Naval Medical University, Shanghai Changzheng Hospital, Shanghai, China
| | - Xudong Cha
- Department of Otolaryngology, The Second Affiliated Hospital of the Naval Medical University, Shanghai Changzheng Hospital, Shanghai, China
| | - Fengzhen Li
- Department of Otolaryngology, The Second Affiliated Hospital of the Naval Medical University, Shanghai Changzheng Hospital, Shanghai, China
| | - Zengyi Xu
- Department of Otolaryngology, The Second Affiliated Hospital of the Naval Medical University, Shanghai Changzheng Hospital, Shanghai, China
| | - Jian Wu
- Department of Otolaryngology, The Second Affiliated Hospital of the Naval Medical University, Shanghai Changzheng Hospital, Shanghai, China.
| | - Huanhai Liu
- Department of Otolaryngology, The Second Affiliated Hospital of the Naval Medical University, Shanghai Changzheng Hospital, Shanghai, China.
| | - Wenwen Ren
- Department of Otolaryngology, The Second Affiliated Hospital of the Naval Medical University, Shanghai Changzheng Hospital, Shanghai, China.
| |
Collapse
|
2
|
Kobayashi M, Nishida K, Yamakata K, Takeuchi K. Aging inhibits olfactory recovery from traumatic olfactory system injury. Neuroscience 2025; 577:64-70. [PMID: 40324583 DOI: 10.1016/j.neuroscience.2025.05.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2024] [Revised: 04/28/2025] [Accepted: 05/03/2025] [Indexed: 05/07/2025]
Abstract
Although recent advances in treatment of traumatic olfactory dysfunction, which had a low rate of improvement through treatment, have increased the improvement rate in younger patients, it remains low in middle-aged and older patients. Although olfactory function declines with age, its impact on traumatic dysfunction recovery remains unclear. This study was conducted to investigate the effects of aging on olfactory nerve regeneration and olfactory function recovery in an animal model of traumatic olfactory injury. Behavioral experiment using olfactory preference and morphological measurements were performed on OMP-tau-lacZ mice at 3 months, 1.0 year and 1.5 years old, in which the olfactory nerve can be visualized after X-gal staining. In the behavioral experiments, we measured the time required for mice to search for and find a piece of potato chip hidden under the cage bedding. The head trauma model included a bilateral olfactory nerve transection (BNTx) group and a control craniotomy alone without BNTx group. In each age and surgery group, behavioral olfactory tests were performed 6 weeks (42 days) and 1.0 year (365 days) after surgery. The reinnervation of olfactory nerve axons to the olfactory bulb was then assessed in histological tissue samples from the mice. The older BNTx group performed worse in behavioral tests than the younger BNTx group, and reinnervation of olfactory nerve axons to the olfactory bulb was lower in the older BNTx group. These findings suggest that aging leads to poor olfactory nerve axon regeneration and poor recovery of olfactory function after olfactory nerve transection injury.
Collapse
Affiliation(s)
- Masayoshi Kobayashi
- Department of Otorhinolaryngology-Head and Neck Surgery, Mie University Graduate School of Medicine, Japan.
| | - Kohei Nishida
- Department of Otorhinolaryngology-Head and Neck Surgery, Mie University Graduate School of Medicine, Japan
| | - Kanta Yamakata
- Department of Otorhinolaryngology-Head and Neck Surgery, Mie University Graduate School of Medicine, Japan
| | - Kazuhiko Takeuchi
- Department of Otorhinolaryngology-Head and Neck Surgery, Mie University Graduate School of Medicine, Japan
| |
Collapse
|
3
|
Gan S, Qu S, Zhu H, Gong M, Xiang Y, Ye D. Role and Mechanism of Olfactory Stem Cells in the Treatment of Olfactory Disorders. Stem Cells Int 2025; 2025:6631857. [PMID: 40313858 PMCID: PMC12045687 DOI: 10.1155/sci/6631857] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2024] [Accepted: 03/24/2025] [Indexed: 05/03/2025] Open
Abstract
Olfactory dysfunction is one of the most prevalent diseases in otorhinolaryngology, particularly since the coronavirus 2019 (COVID-19) pandemic, with a potential impact on daily life. Several etiological factors can contribute to olfactory dysfunction owing to the complexity and specificity of the olfactory transmission pathway. However, current treatments for olfactory dysfunction are limited and their efficacy is unsatisfactory. Olfactory stem cells are multifunctional stem cells in the olfactory mucosa that comprise both horizontal and global basal stem cells (HBCs and GBCs, respectively). These cells can differentiate into various cell types in response to different stimuli with distinct characteristics. The aim of the study was to discuss the mechanisms and functions of stem cells and their application in the treatment of olfactory dysfunction.
Collapse
Affiliation(s)
- Shengqi Gan
- Department of Otorhinolaryngology-Head and Neck Surgery, The Affiliated Lihuili Hospital, Ningbo University, Ningbo 315040, Zhejiang, China
| | - Siyuan Qu
- Department of Otorhinolaryngology-Head and Neck Surgery, The Affiliated Lihuili Hospital, Ningbo University, Ningbo 315040, Zhejiang, China
| | - Hai Zhu
- Department of Otorhinolaryngology-Head and Neck Surgery, The Affiliated Lihuili Hospital, Ningbo University, Ningbo 315040, Zhejiang, China
| | - Mengdan Gong
- Department of Otorhinolaryngology-Head and Neck Surgery, The Affiliated Lihuili Hospital, Ningbo University, Ningbo 315040, Zhejiang, China
| | - Yizhen Xiang
- Department of Otorhinolaryngology-Head and Neck Surgery, The Affiliated Lihuili Hospital, Ningbo University, Ningbo 315040, Zhejiang, China
| | - Dong Ye
- Department of Otorhinolaryngology-Head and Neck Surgery, The Affiliated Lihuili Hospital, Ningbo University, Ningbo 315040, Zhejiang, China
| |
Collapse
|
4
|
Larionov A, Hammer CM, Fiedler K, Filgueira L. Dynamics of Endothelial Cell Diversity and Plasticity in Health and Disease. Cells 2024; 13:1276. [PMID: 39120307 PMCID: PMC11312403 DOI: 10.3390/cells13151276] [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: 06/27/2024] [Revised: 07/19/2024] [Accepted: 07/19/2024] [Indexed: 08/10/2024] Open
Abstract
Endothelial cells (ECs) are vital structural units of the cardiovascular system possessing two principal distinctive properties: heterogeneity and plasticity. Endothelial heterogeneity is defined by differences in tissue-specific endothelial phenotypes and their high predisposition to modification along the length of the vascular bed. This aspect of heterogeneity is closely associated with plasticity, the ability of ECs to adapt to environmental cues through the mobilization of genetic, molecular, and structural alterations. The specific endothelial cytoarchitectonics facilitate a quick structural cell reorganization and, furthermore, easy adaptation to the extrinsic and intrinsic environmental stimuli, known as the epigenetic landscape. ECs, as universally distributed and ubiquitous cells of the human body, play a role that extends far beyond their structural function in the cardiovascular system. They play a crucial role in terms of barrier function, cell-to-cell communication, and a myriad of physiological and pathologic processes. These include development, ontogenesis, disease initiation, and progression, as well as growth, regeneration, and repair. Despite substantial progress in the understanding of endothelial cell biology, the role of ECs in healthy conditions and pathologies remains a fascinating area of exploration. This review aims to summarize knowledge and concepts in endothelial biology. It focuses on the development and functional characteristics of endothelial cells in health and pathological conditions, with a particular emphasis on endothelial phenotypic and functional heterogeneity.
Collapse
Affiliation(s)
- Alexey Larionov
- Faculty of Science and Medicine, Anatomy, University of Fribourg, Route Albert-Gockel 1, CH-1700 Fribourg, Switzerland; (C.M.H.); (L.F.)
| | - Christian Manfred Hammer
- Faculty of Science and Medicine, Anatomy, University of Fribourg, Route Albert-Gockel 1, CH-1700 Fribourg, Switzerland; (C.M.H.); (L.F.)
| | - Klaus Fiedler
- Independent Researcher, CH-1700 Fribourg, Switzerland;
| | - Luis Filgueira
- Faculty of Science and Medicine, Anatomy, University of Fribourg, Route Albert-Gockel 1, CH-1700 Fribourg, Switzerland; (C.M.H.); (L.F.)
| |
Collapse
|
5
|
Marin C, Alobid I, López-Chacón M, VanStrahlen CR, Mullol J. Type 2 and Non-type 2 Inflammation in the Upper Airways: Cellular and Molecular Alterations in Olfactory Neuroepithelium Cell Populations. Curr Allergy Asthma Rep 2024; 24:211-219. [PMID: 38492160 PMCID: PMC11008081 DOI: 10.1007/s11882-024-01137-x] [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] [Accepted: 02/15/2024] [Indexed: 03/18/2024]
Abstract
PURPOSE OF REVIEW Neurogenesis occurring in the olfactory epithelium is critical to continuously replace olfactory neurons to maintain olfactory function, but is impaired during chronic type 2 and non-type 2 inflammation of the upper airways. In this review, we describe the neurobiology of olfaction and the olfactory alterations in chronic rhinosinusitis with nasal polyps (type 2 inflammation) and post-viral acute rhinosinusitis (non-type 2 inflammation), highlighting the role of immune response attenuating olfactory neurogenesis as a possibly mechanism for the loss of smell in these diseases. RECENT FINDINGS Several studies have provided relevant insights into the role of basal stem cells as direct participants in the progression of chronic inflammation identifying a functional switch away from a neuro-regenerative phenotype to one contributing to immune defense, a process that induces a deficient replacement of olfactory neurons. The interaction between olfactory stem cells and immune system might critically underlie ongoing loss of smell in type 2 and non-type 2 inflammatory upper airway diseases. In this review, we describe the neurobiology of olfaction and the olfactory alterations in type 2 and non-type 2 inflammatory upper airway diseases, highlighting the role of immune response attenuating olfactory neurogenesis, as a possibly mechanism for the lack of loss of smell recovery.
Collapse
Affiliation(s)
- Concepció Marin
- INGENIO, IRCE, Fundació Recerca Clínic Barcelona-Institut d'Investigacions Biomèdiques August Pi i Sunyer (FRCB-IDIBAPS), Barcelona, Catalonia, Spain.
- Centre for Biomedical Research in Respiratory Diseases (CIBERES), Health Institute Carlos III, Madrid, Spain.
| | - Isam Alobid
- INGENIO, IRCE, Fundació Recerca Clínic Barcelona-Institut d'Investigacions Biomèdiques August Pi i Sunyer (FRCB-IDIBAPS), Barcelona, Catalonia, Spain
- Centre for Biomedical Research in Respiratory Diseases (CIBERES), Health Institute Carlos III, Madrid, Spain
- Rhinology Unit and Smell Clinic, ENT Department, Hospital Clínic, Barcelona, Catalonia, Spain
- Universitat de Barcelona, Barcelona, Spain
| | - Mauricio López-Chacón
- INGENIO, IRCE, Fundació Recerca Clínic Barcelona-Institut d'Investigacions Biomèdiques August Pi i Sunyer (FRCB-IDIBAPS), Barcelona, Catalonia, Spain
- Centre for Biomedical Research in Respiratory Diseases (CIBERES), Health Institute Carlos III, Madrid, Spain
- Rhinology Unit and Smell Clinic, ENT Department, Hospital Clínic, Barcelona, Catalonia, Spain
| | - Camilo R VanStrahlen
- INGENIO, IRCE, Fundació Recerca Clínic Barcelona-Institut d'Investigacions Biomèdiques August Pi i Sunyer (FRCB-IDIBAPS), Barcelona, Catalonia, Spain
- Centre for Biomedical Research in Respiratory Diseases (CIBERES), Health Institute Carlos III, Madrid, Spain
- Rhinology Unit and Smell Clinic, ENT Department, Hospital Clínic, Barcelona, Catalonia, Spain
| | - Joaquim Mullol
- INGENIO, IRCE, Fundació Recerca Clínic Barcelona-Institut d'Investigacions Biomèdiques August Pi i Sunyer (FRCB-IDIBAPS), Barcelona, Catalonia, Spain.
- Centre for Biomedical Research in Respiratory Diseases (CIBERES), Health Institute Carlos III, Madrid, Spain.
- Rhinology Unit and Smell Clinic, ENT Department, Hospital Clínic, Barcelona, Catalonia, Spain.
- Universitat de Barcelona, Barcelona, Spain.
| |
Collapse
|
6
|
LeFever NM, Katreddi RR, Dolphin NM, Mathias NA, Forni PE. Following the p63/Keratin5 basal cells in the sensory and non-sensory epithelia of the vomeronasal organ. Genesis 2024; 62:e23596. [PMID: 38665067 PMCID: PMC11141727 DOI: 10.1002/dvg.23596] [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: 11/16/2023] [Revised: 02/27/2024] [Accepted: 03/19/2024] [Indexed: 05/01/2024]
Abstract
The vomeronasal organ (VNO) is a part of the accessory olfactory system, which detects pheromones and chemical factors that trigger a spectrum of sexual and social behaviors. The vomeronasal epithelium (VNE) shares several features with the epithelium of the main olfactory epithelium (MOE). However, it is a distinct neuroepithelium populated by chemosensory neurons that differ from the olfactory sensory neurons in cellular structure, receptor expression, and connectivity. The vomeronasal organ of rodents comprises a sensory epithelium (SE) and a thin non-sensory epithelium (NSE) that morphologically resembles the respiratory epithelium. Sox2-positive cells have been previously identified as the stem cell population that gives rise to neuronal progenitors in MOE and VNE. In addition, the MOE also comprises p63 positive horizontal basal cells, a second pool of quiescent stem cells that become active in response to injury. Immunolabeling against the transcription factor p63, Keratin-5 (Krt5), Krt14, NrCAM, and Krt5Cre tracing experiments highlighted the existence of horizontal basal cells distributed along the basal lamina of SE of the VNO. Single cell sequencing and genetic lineage tracing suggest that the vomeronasal horizontal basal cells arise from basal progenitors at the boundary between the SE and NSE proximal to the marginal zones. Moreover, our experiments revealed that the NSE of rodents is, like the respiratory epithelium, a stratified epithelium where the p63/Krt5+ basal progenitor cells self-replicate and give rise to the apical columnar cells facing the lumen of the VNO.
Collapse
Affiliation(s)
| | | | | | | | - Paolo E. Forni
- Department of Biological Sciences
- The RNA Institute
- The Center for Neuroscience Research, University at Albany, State University of New York, Albany, NY, United States
| |
Collapse
|
7
|
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
|
8
|
Ma Z, Li W, Zhuang L, Wen T, Wang P, Yu H, Liu Y, Yu Y. TMEM59 ablation leads to loss of olfactory sensory neurons and impairs olfactory functions via interaction with inflammation. Brain Behav Immun 2023; 111:151-168. [PMID: 37061103 DOI: 10.1016/j.bbi.2023.04.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 03/23/2023] [Accepted: 04/11/2023] [Indexed: 04/17/2023] Open
Abstract
The olfactory epithelium undergoes constant neurogenesis throughout life in mammals. Several factors including key signaling pathways and inflammatory microenvironment regulate the maintenance and regeneration of the olfactory epithelium. In this study, we identify TMEM59 (also known as DCF1) as a critical regulator to the epithelial maintenance and regeneration. Single-cell RNA-Seq data show downregulation of TMEM59 in multiple epithelial cell lineages with aging. Ablation of TMEM59 leads to apparent alteration at the transcriptional level, including genes associated with olfactory transduction and inflammatory/immune response. These differentially expressed genes are key components belonging to several signaling pathways, such as NF-κB, chemokine, etc. TMEM59 deletion impairs olfactory functions, attenuates proliferation, causes loss of both mature and immature olfactory sensory neurons, and promotes infiltration of inflammatory cells, macrophages, microglia cells and neutrophils into the olfactory epithelium and lamina propria. TMEM59 deletion deteriorates regeneration of the olfactory epithelium after injury, with significant reduction in the number of proliferative cells, immature and mature sensory neurons, accompanied by the increasing number of inflammatory cells and macrophages. Anti-inflammation by dexamethasone recovers neuronal generation and olfactory functions in the TMEM59-KO animals, suggesting the correlation between TMEM59 and inflammation in regulating the epithelial maintenance. Collectively, TMEM59 regulates olfactory functions, as well as neuronal generation in the olfactory epithelium via interaction with inflammation, suggesting a potential role in therapy against olfactory dysfunction associated with inflamm-aging.
Collapse
Affiliation(s)
- Zhenjie Ma
- School of Life Sciences, Shanghai University, Shanghai, People's Republic of China
| | - Weihao Li
- Department of Otolaryngology, Eye, Ear, Nose & Throat Hospital, Fudan University, Shanghai, People's Republic of China; Ear, Nose & Throat Institute, Eye, Ear, Nose & Throat Hospital, Fudan University, Shanghai, People's Republic of China; Clinical and Research Center for Olfactory Disorders, Eye, Ear, Nose & Throat Hospital, Fudan University, Shanghai, People's Republic of China
| | - Liujing Zhuang
- Biosensor National Special Laboratory, Key Laboratory for Biomedical Engineering of Education Ministry, Department of Biomedical Engineering, Zhejiang University, Hangzhou 310027, China; The MOE Frontier Science Center for Brain Science & Brain-machine Integration, Zhejiang University, Hangzhou 310027, China
| | - Tieqiao Wen
- School of Life Sciences, Shanghai University, Shanghai, People's Republic of China
| | - Ping Wang
- Biosensor National Special Laboratory, Key Laboratory for Biomedical Engineering of Education Ministry, Department of Biomedical Engineering, Zhejiang University, Hangzhou 310027, China; The MOE Frontier Science Center for Brain Science & Brain-machine Integration, Zhejiang University, Hangzhou 310027, China.
| | - Hongmeng Yu
- Department of Otolaryngology, Eye, Ear, Nose & Throat Hospital, Fudan University, Shanghai, People's Republic of China; Ear, Nose & Throat Institute, Eye, Ear, Nose & Throat Hospital, Fudan University, Shanghai, People's Republic of China; Clinical and Research Center for Olfactory Disorders, Eye, Ear, Nose & Throat Hospital, Fudan University, Shanghai, People's Republic of China.
| | - Yongliang Liu
- Department of Otolaryngology, Zibo Central Hospital, Zibo 255036, China.
| | - Yiqun Yu
- Department of Otolaryngology, Eye, Ear, Nose & Throat Hospital, Fudan University, Shanghai, People's Republic of China; Ear, Nose & Throat Institute, Eye, Ear, Nose & Throat Hospital, Fudan University, Shanghai, People's Republic of China; Clinical and Research Center for Olfactory Disorders, Eye, Ear, Nose & Throat Hospital, Fudan University, Shanghai, People's Republic of China.
| |
Collapse
|
9
|
Louie JD, Bromberg BH, Zunitch MJ, Schwob JE. Horizontal basal cells self-govern their neurogenic potential during injury-induced regeneration of the olfactory epithelium. Development 2023; 150:dev201552. [PMID: 37260223 PMCID: PMC10323233 DOI: 10.1242/dev.201552] [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: 12/22/2022] [Accepted: 05/18/2023] [Indexed: 06/02/2023]
Abstract
Horizontal basal cells (HBCs) residing within severely damaged olfactory epithelium (OE) mediate OE regeneration by differentiating into odorant-detecting olfactory sensory neurons (OSNs) and other tissue supporting non-neuronal cell types. Depending on both tissue type and integrity, the Notch signaling pathway can either positively or negatively regulate resident stem cell activity. Although Notch1 specifies HBC dormancy in the uninjured OE, little is known about how HBCs are influenced by the Notch pathway following OE injury. Here, we show that HBCs depend on a functional inversion of the Notch pathway to appropriately mediate OE regeneration. At 24 h post-injury, HBCs enhance Notch1-mediated signaling. Moreover, at 3 days post-injury when the regenerating OE is composed of multiple cell layers, HBCs enrich both Notch1 and the Notch ligand, Dll1. Notably, HBC-specific Notch1 knockout increases HBC quiescence and impairs HBC differentiation into neuronal progenitors and OSNs. Interestingly, complete HBC knockout of Dll1 only decreases differentiation of HBC-derived OSNs. These data underscore the context-dependent nature of Notch signaling. Furthermore, they reveal that HBCs regulate their own neurogenic potential after OE injury.
Collapse
Affiliation(s)
- Jonathan D. Louie
- Medical Scientist Training Program, Tufts University School of Medicine, Boston, MA 02111, USA
- Neuroscience Graduate Program, Tufts University Graduate School of Biomedical Sciences, Boston, MA 02111, USA
- Department of Developmental, Molecular and Chemical Biology, Tufts University Graduate School of Biomedical Sciences, Boston, MA 02111, USA
| | - Benjamin H. Bromberg
- Department of Developmental, Molecular and Chemical Biology, Tufts University Graduate School of Biomedical Sciences, Boston, MA 02111, USA
| | - Matthew J. Zunitch
- Medical Scientist Training Program, Tufts University School of Medicine, Boston, MA 02111, USA
- Department of Developmental, Molecular and Chemical Biology, Tufts University Graduate School of Biomedical Sciences, Boston, MA 02111, USA
- Cell, Molecular and Developmental Biology Graduate Program, Tufts University Graduate School of Biomedical Sciences, Boston, MA 02111, USA
| | - James E. Schwob
- Department of Developmental, Molecular and Chemical Biology, Tufts University Graduate School of Biomedical Sciences, Boston, MA 02111, USA
| |
Collapse
|
10
|
Paronett EM, Bryan CA, Maynard TM, LaMantia AS. Identity, lineage and fates of a temporally distinct progenitor population in the embryonic olfactory epithelium. Dev Biol 2023; 495:76-91. [PMID: 36627077 PMCID: PMC9926479 DOI: 10.1016/j.ydbio.2023.01.001] [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: 08/16/2021] [Revised: 12/29/2022] [Accepted: 01/01/2023] [Indexed: 01/09/2023]
Abstract
We defined a temporally and transcriptionally divergent precursor cohort in the medial olfactory epithelium (OE) shortly after it differentiates as a distinct tissue at mid-gestation in the mouse. This temporally distinct population of Ascl1+ cells in the dorsomedial OE is segregated from Meis1+/Pax7+ progenitors in the lateral OE, and does not appear to be generated by Pax7+ lateral OE precursors. The medial Ascl1+ precursors do not yield a substantial number of early-generated ORNs. Instead, they first generate additional proliferative precursors as well as a distinct population of frontonasal mesenchymal cells associated with the migratory mass that surrounds the nascent olfactory nerve. Parallel to these in vivo distinctions, isolated medial versus lateral OE precursors in vitro retain distinct proliferative capacities and modes of division that reflect their in vivo identities. At later fetal stages, these early dorsomedial Ascl1+ precursors cells generate spatially restricted subsets of ORNs as well as other non-neuronal cell classes. Accordingly, the initial compliment of ORNs and other OE cell types is derived from at least two distinct early precursor populations: lateral Meis1/Pax7+ precursors that generate primarily early ORNs, and a temporally, spatially, and transcriptionally distinct subset of medial Ascl1+ precursors that initially generate additional OE progenitors and apparent migratory mass cells before yielding a subset of ORNs and likely supporting cell classes.
Collapse
Affiliation(s)
- Elizabeth M Paronett
- Department of Pharmacology and Physiology, George Washington University School of Medicine, Washington, DC, 20037, USA
| | - Corey A Bryan
- Laboratory of Developmental Disorders and Genetics, The Fralin Biomedical Research Institute, Virginia Tech-Carilion School of Medicine, Roanoke, VA, USA
| | - Thomas M Maynard
- Center for Neurobiology Research, The Fralin Biomedical Research Institute, Virginia Tech-Carilion School of Medicine, Roanoke, VA, USA
| | - Anthony-S LaMantia
- Center for Neurobiology Research, The Fralin Biomedical Research Institute, Virginia Tech-Carilion School of Medicine, Roanoke, VA, USA; Department of Biological Sciences Virginia Tech, Blacksburg, VA, USA.
| |
Collapse
|
11
|
Katreddi RR, Taroc EZM, Hicks SM, Lin JM, Liu S, Xiang M, Forni PE. Notch signaling determines cell-fate specification of the two main types of vomeronasal neurons of rodents. Development 2022; 149:dev200448. [PMID: 35781337 PMCID: PMC9340558 DOI: 10.1242/dev.200448] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Accepted: 06/15/2022] [Indexed: 01/09/2023]
Abstract
The ability of terrestrial vertebrates to find food and mating partners, and to avoid predators, relies on the detection of chemosensory information. Semiochemicals responsible for social and sexual behaviors are detected by chemosensory neurons of the vomeronasal organ (VNO), which transmits information to the accessory olfactory bulb. The vomeronasal sensory epithelium of most mammalian species contains a uniform vomeronasal system; however, rodents and marsupials have developed a more complex binary vomeronasal system, containing vomeronasal sensory neurons (VSNs) expressing receptors of either the V1R or V2R family. In rodents, V1R/apical and V2R/basal VSNs originate from a common pool of progenitors. Using single cell RNA-sequencing, we identified differential expression of Notch1 receptor and Dll4 ligand between the neuronal precursors at the VSN differentiation dichotomy. Our experiments show that Notch signaling is required for effective differentiation of V2R/basal VSNs. In fact, Notch1 loss of function in neuronal progenitors diverts them to the V1R/apical fate, whereas Notch1 gain of function redirects precursors to V2R/basal. Our results indicate that Notch signaling plays a pivotal role in triggering the binary differentiation dichotomy in the VNO of rodents.
Collapse
Affiliation(s)
- Raghu Ram Katreddi
- Department of Biological Sciences, University at Albany, State University of New York, Albany, NY 12222, USA
- The RNA Institute, University at Albany, State University of New York, Albany, NY 12222, USA
- The Center for Neuroscience Research, University at Albany, State University of New York, Albany, NY 12222, USA
| | - Ed Zandro M. Taroc
- Department of Biological Sciences, University at Albany, State University of New York, Albany, NY 12222, USA
- The RNA Institute, University at Albany, State University of New York, Albany, NY 12222, USA
- The Center for Neuroscience Research, University at Albany, State University of New York, Albany, NY 12222, USA
| | - Sawyer M. Hicks
- Department of Biological Sciences, University at Albany, State University of New York, Albany, NY 12222, USA
- The RNA Institute, University at Albany, State University of New York, Albany, NY 12222, USA
| | - Jennifer M. Lin
- Department of Biological Sciences, University at Albany, State University of New York, Albany, NY 12222, USA
- The RNA Institute, University at Albany, State University of New York, Albany, NY 12222, USA
- The Center for Neuroscience Research, University at Albany, State University of New York, Albany, NY 12222, USA
| | - Shuting Liu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou 510060, China
| | - Mengqing Xiang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou 510060, China
| | - Paolo E. Forni
- Department of Biological Sciences, University at Albany, State University of New York, Albany, NY 12222, USA
- The RNA Institute, University at Albany, State University of New York, Albany, NY 12222, USA
- The Center for Neuroscience Research, University at Albany, State University of New York, Albany, NY 12222, USA
| |
Collapse
|
12
|
Wu Q, Xu X, Miao X, Bao X, Li X, Xiang L, Wang W, Du S, Lu Y, Wang X, Yang D, Zhang J, Shen X, Li F, Lu S, Fan Y, Xu S, Chen Z, Wang Y, Teng H, Huang Z. YAP signaling in horizontal basal cells promotes the regeneration of olfactory epithelium after injury. Stem Cell Reports 2022; 17:664-677. [PMID: 35148842 PMCID: PMC9039758 DOI: 10.1016/j.stemcr.2022.01.007] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Revised: 01/07/2022] [Accepted: 01/10/2022] [Indexed: 10/29/2022] Open
Abstract
The horizontal basal cells (HBCs) of olfactory epithelium (OE) serve as reservoirs for stem cells during OE regeneration, through proliferation and differentiation, which is important in recovery of olfactory function. However, the molecular mechanism of regulation of HBC proliferation and differentiation after injury remains unclear. Here, we found that yes-associated protein (YAP) was upregulated and activated in HBCs after OE injury. Deletion of YAP in HBCs led to impairment in OE regeneration and functional recovery of olfaction after injury. Mechanically, YAP was activated by S1P/S1PR2 signaling, thereby promoting the proliferation of HBCs and OE regeneration after injury. Finally, activation of YAP signaling enhanced the proliferation of HBCs and improved functional recovery of olfaction after OE injury or in Alzheimer's disease model mice. Taken together, these results reveal an S1P/S1PR2/YAP pathway in OE regeneration in response to injury, providing a promising therapeutic strategy for OE injury.
Collapse
Affiliation(s)
- Qian Wu
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China; School of Mental Health, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Xingxing Xu
- School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Xuemeng Miao
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China; School of Mental Health, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Xiaomei Bao
- School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Xiuchun Li
- Department of Orthopedics (Spine Surgery), The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Ludan Xiang
- School of Mental Health, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Wei Wang
- School of Mental Health, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Siyu Du
- School of Mental Health, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Yi Lu
- School of Mental Health, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Xiwu Wang
- School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Danlu Yang
- School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Jingjing Zhang
- School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Xiya Shen
- School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Fayi Li
- Department of Orthopedics (Spine Surgery), The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Sheng Lu
- Department of Orthopedics (Spine Surgery), The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Yiren Fan
- School of Mental Health, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Shujie Xu
- School of Mental Health, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Zihao Chen
- School of Mental Health, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Ying Wang
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China; Department of Transfusion Medicine, Zhejiang Provincial People's Hospital of Hangzhou Medical College, Hangzhou, Zhejiang 310053, China.
| | - Honglin Teng
- Department of Orthopedics (Spine Surgery), The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325035, China.
| | - Zhihui Huang
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China; School of Mental Health, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China; Department of Orthopedics (Spine Surgery), The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325035, China.
| |
Collapse
|
13
|
Joseph KB, Awadallah N, Delay ER, Delay RJ. Transient Effects of Cyclophosphamide on Basal Cell Proliferation of Olfactory Epithelia. Chem Senses 2021; 45:549-561. [PMID: 32531016 DOI: 10.1093/chemse/bjaa039] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Cancer is often treated with broad-spectrum cytotoxic drugs that not only eradicate cancerous cells but also have detrimental side effects. One of these side effects, disruption of the olfactory system, impedes a patient's ability to smell, perceive flavor, and ultimately may interfere with their nutritional intake and recovery from cancer. Recent studies reported that the chemotherapy drug, cyclophosphamide (CYP), can damage gustatory epithelia and disrupt cell proliferation in olfactory epithelia. In this study, we asked if CYP altered globose and horizontal basal cell proliferation in the murine main olfactory epithelium (MOE) and vomeronasal organ (VNO). We used antibodies for Ki67, a marker strictly associated with cell proliferation, and Keratin 5, a marker for the cytoskeleton of horizontal basal cells. Our results revealed a significant CYP-induced decrease in the number of proliferative cells in both epithelia, especially globose basal cells in the MOE, within the first 1-2 days postinjection. Recovery of cell renewal was apparent 6 days after injection. The immunohistochemical markers showed significantly higher levels of globose and horizontal basal cell proliferation in CYP-injected mice at 14 and 30 days postinjection compared with control mice. The prolonged proliferative activation of globose and horizontal basal cells suggests that, besides altering proliferation of olfactory epithelia, the epithelial substrate needed for successful cell renewal may be adversely affected by CYP.
Collapse
Affiliation(s)
- Kyle B Joseph
- Department of Biology and Vermont Chemosensory Group, University of Vermont, Burlington, VT, USA.,Department of Bio-Medical Sciences, Philadelphia College of Osteopathic Medicine, Philadelphia, PA, USA
| | - Nora Awadallah
- Department of Biology and Vermont Chemosensory Group, University of Vermont, Burlington, VT, USA.,City University of New York (CUNY) Neuroscience Collaborative, CUNY Graduate Center, New York City, NY, USA.,Department of Molecular, Cellular and Biomedical Sciences, The CUNY School of Medicine, City College, The City University of New York, New York City, NY, USA
| | - Eugene R Delay
- Department of Biology and Vermont Chemosensory Group, University of Vermont, Burlington, VT, USA
| | - Rona J Delay
- Department of Biology and Vermont Chemosensory Group, University of Vermont, Burlington, VT, USA
| |
Collapse
|
14
|
Katreddi RR, Forni PE. Mechanisms underlying pre- and postnatal development of the vomeronasal organ. Cell Mol Life Sci 2021; 78:5069-5082. [PMID: 33871676 PMCID: PMC8254721 DOI: 10.1007/s00018-021-03829-3] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Revised: 03/17/2021] [Accepted: 03/29/2021] [Indexed: 02/06/2023]
Abstract
The vomeronasal organ (VNO) is sensory organ located in the ventral region of the nasal cavity in rodents. The VNO develops from the olfactory placode during the secondary invagination of olfactory pit. The embryonic vomeronasal structure appears as a neurogenic area where migratory neuronal populations like endocrine gonadotropin-releasing hormone-1 (GnRH-1) neurons form. Even though embryonic vomeronasal structures are conserved across most vertebrate species, many species including humans do not have a functional VNO after birth. The vomeronasal epithelium (VNE) of rodents is composed of two major types of vomeronasal sensory neurons (VSNs): (1) VSNs distributed in the apical VNE regions that express vomeronasal type-1 receptors (V1Rs) and the G protein subunit Gαi2, and (2) VSNs in the basal territories of the VNE that express vomeronasal type-2 receptors (V2Rs) and the G subunit Gαo. Recent studies identified a third subclass of Gαi2 and Gαo VSNs that express the formyl peptide receptor family. VSNs expressing V1Rs or V2Rs send their axons to distinct regions of the accessory olfactory bulb (AOB). Together, VNO and AOB form the accessory olfactory system (AOS), an olfactory subsystem that coordinates the social and sexual behaviors of many vertebrate species. In this review, we summarize our current understanding of cellular and molecular mechanisms that underlie VNO development. We also discuss open questions for study, which we suggest will further enhance our understanding of VNO morphogenesis at embryonic and postnatal stages.
Collapse
Affiliation(s)
- Raghu Ram Katreddi
- Department of Biological Sciences, Center for Neuroscience Research, The RNA Institute, University At Albany, State University of New York, Albany, NY, USA
| | - Paolo E Forni
- Department of Biological Sciences, Center for Neuroscience Research, The RNA Institute, University At Albany, State University of New York, Albany, NY, USA.
| |
Collapse
|
15
|
Sarkar A, Saha S, Paul A, Maji A, Roy P, Maity TK. Understanding stem cells and its pivotal role in regenerative medicine. Life Sci 2021; 273:119270. [PMID: 33640402 DOI: 10.1016/j.lfs.2021.119270] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Revised: 02/06/2021] [Accepted: 02/14/2021] [Indexed: 02/07/2023]
Abstract
Stem cells (SCs) are clonogenic cells that develop into the specialized cells which later responsible for making up various types of tissue in the human body. SCs are not only the appropriate source of information for cell division, molecular and cellular processes, and tissue homeostasis but also one of the major putative biological aids to diagnose and cure various degenerative diseases. This study emphasises on various research outputs that occurred in the past two decades. This will give brief information on classification, differentiation, detection, and various isolation techniques of SCs. Here, the various signalling pathways which includes WNT, Sonic hedgehog, Notch, BMI1 and C-met pathways and how does it effect on the regeneration of various classes of SCs and factors that regulates the potency of the SCs are also been discussed. We also focused on the application of SCs in the area of regenerative medicine along with the cellular markers that are useful as salient diagnostic or curative tools or in both, by the process of reprogramming, which includes diabetes, cancer, cardiovascular disorders and neurological disorders. The biomarkers that are mentioned in various literatures and experiments include PDX1, FOXA2, HNF6, and NKX6-1 (for diabetes); CD33, CD24, CD133 (for cancer); c-Kit, SCA-1, Wilm's tumor 1 (for cardiovascular disorders); and OCT4, SOX2, c-MYC, EN1, DAT and VMAT2 (for neurological disorders). In this review, we come to know the advancements and scopes of potential SC-based therapies, its diverse applications in clinical fields that can be helpful in the near future.
Collapse
Affiliation(s)
- Arnab Sarkar
- Department of Pharmaceutical Technology, Jadavpur University, West Bengal, Kolkata 700032, India
| | - Sanjukta Saha
- Department of Pharmaceutical Technology, Jadavpur University, West Bengal, Kolkata 700032, India
| | - Abhik Paul
- Department of Pharmaceutical Technology, Jadavpur University, West Bengal, Kolkata 700032, India
| | - Avik Maji
- Department of Pharmaceutical Technology, Jadavpur University, West Bengal, Kolkata 700032, India
| | - Puspita Roy
- Department of Pharmaceutical Technology, Jadavpur University, West Bengal, Kolkata 700032, India
| | - Tapan Kumar Maity
- Department of Pharmaceutical Technology, Jadavpur University, West Bengal, Kolkata 700032, India.
| |
Collapse
|
16
|
Senf K, Karius J, Stumm R, Neuhaus EM. Chemokine signaling is required for homeostatic and injury-induced neurogenesis in the olfactory epithelium. Stem Cells 2021; 39:617-635. [PMID: 33470495 DOI: 10.1002/stem.3338] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Accepted: 12/10/2020] [Indexed: 12/24/2022]
Abstract
The olfactory epithelium (OE) possesses unique lifelong neuroregenerative capacities and undergoes constitutive neurogenesis throughout mammalian lifespan. Two populations of stem cells, frequently dividing globose basal cells (GBCs) and quiescent horizontal basal cells (HBCs), readily replace olfactory neurons throughout lifetime. Although lineage commitment and neuronal differentiation of stem cells has already been described in terms of transcription factor expression, little is known about external factors balancing between differentiation and self-renewal. We show here that expression of the CXC-motif chemokine receptor 4 (CXCR4) distinguishes both types of stem cells. Extensive colocalization analysis revealed exclusive expression of CXCR4 in proliferating GBCs and their neuronal progenies. Moreover, only neuronal lineage cells were derived from CXCR4-CreER-tdTomato reporter mice in the OE. Furthermore, Cre-tdTomato mice specific for HBCs (Nestin+ and Cytokeratin14+) did not reduce CXCR4 expression when bred to mice bearing floxed CXCR4 alleles, and did not show labeling of the neuronal cells. CXCR4 and its ligand CXCL12 were markedly upregulated upon induction of GBC proliferation during injury-induced regeneration. in vivo overexpression of CXCL12 did downregulate CXCR4 levels, which results in reduced GBC maintenance and neuronal differentiation. We proved that these effects were caused by CXCR4 downregulation rather than over-activation by showing that the phenotypes of CXCL12-overexpressing mice were highly similar to the phenotypes of CXCR4 knockout mice. Our results demonstrate functional CXCR4 signaling in GBCs regulates cell cycle exit and neural differentiation. We propose that CXCR4/CXCL12 signaling is an essential regulator of olfactory neurogenesis and provide new insights into the dynamics of neurogenesis in the OE.
Collapse
Affiliation(s)
- Katja Senf
- Pharmacology and Toxicology, Jena University Hospital, Friedrich Schiller University Jena, Jena, Germany
| | - Julia Karius
- Pharmacology and Toxicology, Jena University Hospital, Friedrich Schiller University Jena, Jena, Germany
| | - Ralf Stumm
- Pharmacology and Toxicology, Jena University Hospital, Friedrich Schiller University Jena, Jena, Germany
| | - Eva M Neuhaus
- Pharmacology and Toxicology, Jena University Hospital, Friedrich Schiller University Jena, Jena, Germany
| |
Collapse
|
17
|
Li X, Tong M, Wang L, Qin Y, Yu H, Yu Y. Age-Dependent Activation and Neuronal Differentiation of Lgr5+ Basal Cells in Injured Olfactory Epithelium via Notch Signaling Pathway. Front Aging Neurosci 2020; 12:602688. [PMID: 33390928 PMCID: PMC7773941 DOI: 10.3389/fnagi.2020.602688] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Accepted: 11/16/2020] [Indexed: 01/15/2023] Open
Abstract
Aging is an important factor affecting function of smell, leading to the degeneration of mature olfactory sensory neurons and inducing the occurrence of smell loss. The mammalian olfactory epithelium (OE) can regenerate when subjected to chemical assaults. However, this capacity is not limitless. Inactivation of globose basal cells and failure to generate sensory neurons are the main obstacles to prevent the OE regeneration. Here, we found the significant attenuation in mature sensory neuronal generation and apparent transcriptional alternation in the OE from aged mice compared with young ones. The recruitment of leucine-rich repeat-containing G-protein coupled receptor 5 (Lgr5)-positive cells in injured OE was weakened in aged mice, and more Lgr5+ cells remained quiescence in aged OE postinjury. Lineage-traced progenies from Lgr5+ cells were significantly fewer in the OE with aging. Moreover, Notch activation enhanced the neuronal regeneration in aged OE, making the regenerative capacity of aged OE comparable with that of young animals after injury. The growth and morphology of three-dimensional (3D)-cultured organoids from the OE of young and aged mice varied and was modulated by small molecules regulating the Notch signaling pathway. Thus, we concluded that activation of Lgr5+ cells in injured OE was age dependent and Notch activation could enhance the capacity of neuronal generation from Lgr5+ cells in aged OE after injury.
Collapse
Affiliation(s)
- Xuewen Li
- School of Life Sciences, Shanghai University, Shanghai, China
| | - Meimei Tong
- Ear, Nose and Throat Department, Yuecheng People's Hospital, Shaoxing, China
| | - Li Wang
- Department of Otolaryngology, Eye, Ear, Nose and Throat Hospital, Shanghai Key Clinical Disciplines of Otorhinolaryngology, Fudan University, Shanghai, China
| | - Yumei Qin
- School of Food Science and Bioengineering, Zhejiang Gongshang University, Hangzhou, China
| | - Hongmeng Yu
- Department of Otolaryngology, Eye, Ear, Nose and Throat Hospital, Shanghai Key Clinical Disciplines of Otorhinolaryngology, Fudan University, Shanghai, China.,Research Units of New Technologies of Endoscopic Surgery in Skull Base Tumor, Chinese Academy of Medical Sciences, Beijing, China
| | - Yiqun Yu
- School of Life Sciences, Shanghai University, Shanghai, China.,Department of Otolaryngology, Eye, Ear, Nose and Throat Hospital, Shanghai Key Clinical Disciplines of Otorhinolaryngology, Fudan University, Shanghai, China
| |
Collapse
|
18
|
Yang D, Wu X, Zhou Y, Wang W, Wang Z. The microRNA/TET3/REST axis is required for olfactory globose basal cell proliferation and male behavior. EMBO Rep 2020; 21:e49431. [PMID: 32677323 DOI: 10.15252/embr.201949431] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Revised: 06/14/2020] [Accepted: 06/18/2020] [Indexed: 12/16/2022] Open
Abstract
In the main olfactory epithelium (MOE), new olfactory sensory neurons (OSNs) are persistently generated to replace lost neurons throughout an organism's lifespan. This process predominantly depends on the proliferation of globose basal cells (GBCs), the actively dividing stem cells in the MOE. Here, by using CRISPR/Cas9 and RNAi coupled with adeno-associated virus (AAV) nose delivery approaches, we demonstrated that knockdown of miR-200b/a in the MOE resulted in supernumerary Mash1-marked GBCs and decreased numbers of differentiated OSNs, accompanied by abrogation of male behaviors. We further showed that in the MOE, miR-200b/a targets the ten-eleven translocation methylcytosine dioxygenase TET3, which cooperates with RE1-silencing transcription factor (REST) to exert their functions. Deficiencies including proliferation, differentiation, and behaviors illustrated in miR-200b/a knockdown mice were rescued by suppressing either TET3 or REST. Our work describes a mechanism of coordination of GBC proliferation and differentiation in the MOE and olfactory male behaviors through miR-200/TET3/REST signaling.
Collapse
Affiliation(s)
- Dong Yang
- College of Life Science, Institute of Life Science and Green Development, Hebei University, Baoding, China
| | - Xiangbo Wu
- College of Life Science, Institute of Life Science and Green Development, Hebei University, Baoding, China
| | - Yanfen Zhou
- College of Life Science, Institute of Life Science and Green Development, Hebei University, Baoding, China
| | - Weina Wang
- College of Life Science, Institute of Life Science and Green Development, Hebei University, Baoding, China
| | - Zhenshan Wang
- College of Life Science, Institute of Life Science and Green Development, Hebei University, Baoding, China
| |
Collapse
|
19
|
Sun L, Zhou T, Wan QH, Fang SG. Transcriptome Comparison Reveals Key Components of Nuptial Plumage Coloration in Crested Ibis. Biomolecules 2020; 10:E905. [PMID: 32549189 PMCID: PMC7356354 DOI: 10.3390/biom10060905] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Revised: 06/02/2020] [Accepted: 06/13/2020] [Indexed: 11/16/2022] Open
Abstract
Nuptial plumage coloration is critical in the mating choice of the crested ibis. This species has a characteristic nuptial plumage that develops from the application of a black sticky substance, secreted by a patch of skin in the throat and neck region. We aimed to identify the genes regulating its coloring, by comparing skin transcriptomes between ibises during the breeding and nonbreeding seasons. In breeding season skins, key eumelanin synthesis genes, TYR, DCT, and TYRP1 were upregulated. Tyrosine metabolism, which is closely related to melanin synthesis, was also upregulated, as were transporter proteins belonging to multiple SLC families, which might act during melanosome transportation to keratinocytes. These results indicate that eumelanin is likely an important component of the black substance. In addition, we observed upregulation in lipid metabolism in breeding season skins. We suggest that the lipids contribute to an oil base, which imbues the black substance with water insolubility and enhances its adhesion to feather surfaces. In nonbreeding season skins, we observed upregulation in cell adhesion molecules, which play critical roles in cell interactions. A number of molecules involved in innervation and angiogenesis were upregulated, indicating an ongoing expansion of nerves and blood vessels in sampled skins. Feather β keratin, a basic component of avian feather filament, was also upregulated. These results are consistent with feather regeneration in the black skin of nonbreeding season ibises. Our results provide the first molecular evidence indicating that eumelanin is the key component of ibis coloration.
Collapse
Affiliation(s)
| | | | | | - Sheng-Guo Fang
- MOE Key Laboratory of Biosystems Homeostasis & Protection, State Conservation Centre for Gene Resources of Endangered Wildlife, College of Life Sciences, Zhejiang University, Hangzhou 310058, China; (L.S.); (T.Z.); (Q.-H.W.)
| |
Collapse
|
20
|
Lund C, Yellapragada V, Vuoristo S, Balboa D, Trova S, Allet C, Eskici N, Pulli K, Giacobini P, Tuuri T, Raivio T. Characterization of the human GnRH neuron developmental transcriptome using a GNRH1-TdTomato reporter line in human pluripotent stem cells. Dis Model Mech 2020; 13:dmm040105. [PMID: 31996360 PMCID: PMC7075073 DOI: 10.1242/dmm.040105] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Accepted: 01/16/2020] [Indexed: 12/21/2022] Open
Abstract
Gonadotropin-releasing hormone (GnRH) neurons provide a fundamental signal for the onset of puberty and subsequent reproductive functions by secretion of gonadotropin-releasing hormone. Their disrupted development or function leads to congenital hypogonadotropic hypogonadism (CHH). To model the development of human GnRH neurons, we generated a stable GNRH1-TdTomato reporter cell line in human pluripotent stem cells (hPSCs) using CRISPR-Cas9 genome editing. RNA-sequencing of the reporter clone, differentiated into GnRH neurons by dual SMAD inhibition and FGF8 treatment, revealed 6461 differentially expressed genes between progenitors and GnRH neurons. Expression of the transcription factor ISL1, one of the top 50 most upregulated genes in the TdTomato-expressing GnRH neurons, was confirmed in 10.5 gestational week-old human fetal GnRH neurons. Among the differentially expressed genes, we detected 15 genes that are implicated in CHH and several genes that are implicated in human puberty timing. Finally, FGF8 treatment in the neuronal progenitor pool led to upregulation of 37 genes expressed both in progenitors and in TdTomato-expressing GnRH neurons, which suggests upstream regulation of these genes by FGF8 signaling during GnRH neuron differentiation. These results illustrate how hPSC-derived human GnRH neuron transcriptomic analysis can be utilized to dissect signaling pathways and gene regulatory networks involved in human GnRH neuron development.This article has an associated First Person interview with the first author of the paper.
Collapse
Affiliation(s)
- Carina Lund
- Stem Cells and Metabolism Research Program, Faculty of Medicine, 00014 University of Helsinki, Helsinki, Finland
- Medicum, Faculty of Medicine, 00014 University of Helsinki, Helsinki, Finland
| | - Venkatram Yellapragada
- Stem Cells and Metabolism Research Program, Faculty of Medicine, 00014 University of Helsinki, Helsinki, Finland
- Medicum, Faculty of Medicine, 00014 University of Helsinki, Helsinki, Finland
| | - Sanna Vuoristo
- Department of Obstetrics and Gynecology, 00029 Helsinki University Hospital, Helsinki, Finland
| | - Diego Balboa
- Centre for Genomic Regulation, The Barcelona Institute of Science and Technology, Dr. Aiguader 88, 08003 Barcelona, Spain
| | - Sara Trova
- Inserm, Laboratory of Development and Plasticity of the Neuroendocrine Brain, Jean-Pierre Aubert Research Center, U1172 Lille, France
| | - Cecile Allet
- Inserm, Laboratory of Development and Plasticity of the Neuroendocrine Brain, Jean-Pierre Aubert Research Center, U1172 Lille, France
| | - Nazli Eskici
- Stem Cells and Metabolism Research Program, Faculty of Medicine, 00014 University of Helsinki, Helsinki, Finland
- Medicum, Faculty of Medicine, 00014 University of Helsinki, Helsinki, Finland
| | - Kristiina Pulli
- Stem Cells and Metabolism Research Program, Faculty of Medicine, 00014 University of Helsinki, Helsinki, Finland
- Medicum, Faculty of Medicine, 00014 University of Helsinki, Helsinki, Finland
| | - Paolo Giacobini
- Inserm, Laboratory of Development and Plasticity of the Neuroendocrine Brain, Jean-Pierre Aubert Research Center, U1172 Lille, France
- University of Lille, FHU 1000 Days for Health, School of Medicine, 59000 Lille, France
| | - Timo Tuuri
- Department of Obstetrics and Gynecology, 00029 Helsinki University Hospital, Helsinki, Finland
| | - Taneli Raivio
- Stem Cells and Metabolism Research Program, Faculty of Medicine, 00014 University of Helsinki, Helsinki, Finland
- Medicum, Faculty of Medicine, 00014 University of Helsinki, Helsinki, Finland
- New Children's Hospital, Pediatric Research Center, 00029 Helsinki University Hospital, Helsinki, Finland
| |
Collapse
|
21
|
Das D, Fletcher RB, Ngai J. Cellular mechanisms of epithelial stem cell self-renewal and differentiation during homeostasis and repair. WILEY INTERDISCIPLINARY REVIEWS-DEVELOPMENTAL BIOLOGY 2019; 9:e361. [PMID: 31468728 DOI: 10.1002/wdev.361] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/27/2019] [Revised: 07/31/2019] [Accepted: 08/02/2019] [Indexed: 12/14/2022]
Abstract
Epithelia in adult mammals exhibit remarkable regenerative capacities owing to the presence of adult stem cells, which self-renew and differentiate to replace cells lost to normal turnover or injury. The mechanisms supporting tissue homeostasis and injury-induced repair often differ from each other as well as from those used in embryonic development. Recent studies have also highlighted the phenomenon of cellular plasticity in adult tissues, in which differentiated cells can change fate and even give rise to new stem cell populations to complement the canonical stem cells in promoting repair following injury. Signaling pathways such as WNT, bone morphogenetic protein, and Sonic Hedgehog play critical roles in stem cell maintenance and cell fate decisions across diverse epithelia and conditions, suggesting that conserved mechanisms underlie the regenerative capacity of adult epithelial structures. This article is categorized under: Gene Expression and Transcriptional Hierarchies > Regulatory Mechanisms Adult Stem Cells, Tissue Renewal, and Regeneration > Tissue Stem Cells and Niches Adult Stem Cells, Tissue Renewal, and Regeneration > Stem Cell Differentiation and Reversion Adult Stem Cells, Tissue Renewal, and Regeneration > Regeneration.
Collapse
Affiliation(s)
- Diya Das
- Department of Molecular and Cell Biology, University of California, Berkeley, California.,Berkeley Institute for Data Science, University of California, Berkeley, California
| | - Russell B Fletcher
- Department of Molecular and Cell Biology, University of California, Berkeley, California
| | - John Ngai
- Department of Molecular and Cell Biology, University of California, Berkeley, California.,Helen Wills Neuroscience Institute, University of California, Berkeley, California.,QB3 Functional Genomics Laboratory, University of California, Berkeley, California
| |
Collapse
|
22
|
Trac D, Maxwell JT, Brown ME, Xu C, Davis ME. Aggregation of Child Cardiac Progenitor Cells Into Spheres Activates Notch Signaling and Improves Treatment of Right Ventricular Heart Failure. Circ Res 2019; 124:526-538. [PMID: 30590978 PMCID: PMC6375764 DOI: 10.1161/circresaha.118.313845] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
RATIONALE Congenital heart disease can lead to life-threatening right ventricular (RV) heart failure. Results from clinical trials support expanding cardiac progenitor cell (CPC) based therapies. However, our recent data show that CPCs lose function as they age, starting as early as 1 year. OBJECTIVE To determine whether the aggregation of child (1-5-year-old) CPCs into scaffold-free spheres can improve differentiation by enhancing Notch signaling, a known regulator of CPC fate. We hypothesized that aggregated (3-dimensional [3D]) CPCs will repair RV heart failure better than monolayer (2-dimensional [2D]) CPCs. METHODS AND RESULTS Spheres were produced with 1500 CPCs each using a microwell array. CPC aggregation significantly increased gene expression of Notch1 compared with 2D CPCs, accompanied by significant upregulation of cardiogenic transcription factors (GATA4, HAND1, MEF2C, NKX2.5, and TBX5) and endothelial markers (CD31, FLK1, FLT1, VWF). Blocking Notch receptor activation with the γ-secretase inhibitor DAPT (N-[N-(3,5-difluorophenacetyl)-L-alanyl]-S-phenylglycine t-butyl ester) diminished these effects. To evaluate the therapeutic improvements of CPC aggregation, RV heart failure was induced in athymic rats by pulmonary artery banding, and cells were implanted into the RV free wall. Echocardiographic measurements 28 days postimplantation showed significantly improved RV function with 3D compared with 2D CPCs. Tracking implanted CPCs via DiR (1,1'-dioctadecyl-3,3,3',3'-tetramethylindotricarbocyanine iodide)-labeling showed improved retention of 3D CPCs. Transducing 3D CPCs with Notch1-shRNA (short hairpin RNA) did not reduce retention, but significantly reduced RV functional improvements. Histological analyses showed 3D treatment reduced RV fibrosis and increased angiogenesis. Although 3D CPCs formed CD31+ vessel-like cells in vivo, these effects are more likely because of improved 3D CPC exosome function compared with 2D CPC exosomes. CONCLUSIONS Spherical aggregation improves child CPC function in a Notch-dependent manner. The strong reparative ability of CPC spheres warrants further investigation as a treatment for pediatric heart failure, especially in older children where reparative ability may be reduced.
Collapse
Affiliation(s)
- David Trac
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology & Emory University School of Medicine, Atlanta, Georgia, 30322, USA
| | - Joshua T. Maxwell
- Division of Pediatric Cardiology, Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia, 30322, USA
| | - Milton E. Brown
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology & Emory University School of Medicine, Atlanta, Georgia, 30322, USA
| | - Chunhui Xu
- Division of Pediatric Cardiology, Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia, 30322, USA;,Children’s Heart Research & Outcomes (HeRO) Center, Children’s Healthcare of Atlanta & Emory University, Atlanta, Georgia, 30322, USA
| | - Michael E. Davis
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology & Emory University School of Medicine, Atlanta, Georgia, 30322, USA;,Division of Pediatric Cardiology, Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia, 30322, USA;,Children’s Heart Research & Outcomes (HeRO) Center, Children’s Healthcare of Atlanta & Emory University, Atlanta, Georgia, 30322, USA
| |
Collapse
|