1
|
Harris DE, Kim JJ, Stern SR, Vicars HM, Matias NR, Gallicchio L, Baker CC, Fuller MT. An RNA binding regulatory cascade controls the switch from proliferation to differentiation in the Drosophila male germ line stem cell lineage. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.06.611673. [PMID: 39282418 PMCID: PMC11398533 DOI: 10.1101/2024.09.06.611673] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 09/21/2024]
Abstract
The switch from precursor cell proliferation to onset of differentiation in adult stem cell lineages must be carefully regulated to produce sufficient progeny to maintain and repair tissues, yet prevent overproliferation that may enable oncogenesis. In the Drosophila male germ cell lineage, spermatogonia produced by germ line stem cells undergo a limited number of transit amplifying mitotic divisions before switching to the spermatocyte program that sets up meiosis and eventual spermatid differentiation. The number of transit amplifying divisions is set by accumulation of the bag-of-marbles (Bam) protein to a critical threshold. In bam mutants, spermatogonia proliferate through several extra rounds of mitosis then die without becoming spermatocytes. Here we show that the key role of Bam for the mitosis to differentiation switch is repressing expression of Held Out Wings (how), homolog of mammalian Quaking. Knock down of how in germ cells was sufficient to allow spermatogonia mutant for bam or its partner benign gonial cell neoplasm (bgcn) to differentiate, while forced expression of nuclear-targeted How protein in spermatogonia wild-type for bam resulted in continued proliferation at the expense of differentiation. Our findings suggest that Bam targets how RNA for degradation by acting as an adapter to recruit the CCR4-NOT deadenylation complex via binding its subunit, Caf40. As How is itself an RNA binding protein with roles in RNA processing, our findings reveal that the switch from proliferation to meiosis and differentiation in the Drosophila male germ line adult stem cell lineage is regulated by a cascade of RNA-binding proteins.
Collapse
Affiliation(s)
- Devon E. Harris
- Department of Developmental Biology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Jongmin J. Kim
- Department of Chemical and Systems Biology, Stanford University School of Medicine, Stanford, CA 94305, USA
- Current address: Department of Biomedical Sciences, Cornell University, Ithaca NY, 14853, USA
| | - Sarah R. Stern
- Department of Developmental Biology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Hannah M. Vicars
- Department of Developmental Biology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Neuza R. Matias
- Department of Developmental Biology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Lorenzo Gallicchio
- Department of Developmental Biology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Catherine C. Baker
- Department of Developmental Biology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Margaret T. Fuller
- Department of Developmental Biology, Stanford University School of Medicine, Stanford, CA 94305, USA
- Department of Genetics, Stanford University School of Medicine, Stanford, CA 94305, USA
| |
Collapse
|
2
|
Ge L, Chen W, Wei F. Annexin A1 protects epidermal stem cells against ultraviolet-B irradiation-induced mitochondrial dysfunction. Arch Dermatol Res 2024; 316:385. [PMID: 38874830 DOI: 10.1007/s00403-024-02875-8] [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: 02/28/2024] [Revised: 02/28/2024] [Accepted: 04/07/2024] [Indexed: 06/15/2024]
Abstract
Ultraviolet-B (UV-B) radiation overexposure causes function impairment of epidermal stem cells (ESCs). We explored the mechanism of Annexin A1 (ANXA1) ameliorating UV-B-induced ESC mitochondrial dysfunction/cell injury. ESCs were cultured in vitro and irradiated with different doses of UV-B. Cell viability/ANXA1 protein level were assessed. After oe-ANXA1 transfection, ESCs were treated with oe-ANXA1/UV-B irradiation/CCCP/CCG-1423/3-methyladenine for 12 h. Cell viability/death, and adenosine triphosphate (ATP)/reactive oxygen species (ROS) levels were determined. Mitochondrial membrane potential (MMP) changes/DNA (mtDNA) content/oxygen consumption and RhoA activation were assessed. ROCK1/p-MYPT1/MYPT1/(LC3BII/I)/Beclin-1/p62 protein levels were determined. Mitochondrial morphology was observed. Mito-Tracker Green (MTG) and LC3B levels were determined. UV-B irradiation decreased cell viability/ANXA1 expression in a dose-dependent manner. UV-B-treated ESCs exhibited reduced cell viability/ATP content/MMP level/mitochondrial respiratory control ratio/mtDNA number/RhoA activity/MYPT1 phosphorylation/MTG+LC3B+ cells/(LC3BII/I) and Beclin-1 proteins, increased cell death/ROS/p62/IL-1β/IL-6/TNF-α expression, contracted mitochondrial, disappeared mitochondrial cristae, and increased vacuolar mitochondria, which were averted by ANXA1 overexpression, suggesting that UV-B induced ESC mitochondrial dysfunction/cell injury/inflammation by repressing mitophagy, but ANXA1 promoted mitophagy by activating the RhoA/ROCK1 pathway, thus repressing UV-B's effects. Mitophagy activation ameliorated UV-B-caused ESC mitochondrial dysfunction/cell injury/inflammation. Mitophagy inhibition partly diminished ANXA1-ameliorated UV-B's effects. Conjointly, ANXA1 promoted mitophagy by activating the RhoA/ROCK1 pathway, thereby improving UV-B-induced ESC mitochondrial dysfunction/cell injury.
Collapse
Affiliation(s)
- Lingzhi Ge
- Department of Dermatology and Venereology, The Second Affiliated Hospital of Shandong First Medical University, No. 366 Mount Tai Street, Taian, 271000, Shandong Province, China
| | - Wenfang Chen
- Department of Dermatology and Venereology, The Second Affiliated Hospital of Shandong First Medical University, No. 366 Mount Tai Street, Taian, 271000, Shandong Province, China
| | - Fangli Wei
- Department of Dermatology and Venereology, The Second Affiliated Hospital of Shandong First Medical University, No. 366 Mount Tai Street, Taian, 271000, Shandong Province, China.
| |
Collapse
|
3
|
Klotskova H, Kidess E, Nadal AL, Brugman S. The role of interleukin-22 in mammalian intestinal homeostasis: Friend and foe. Immun Inflamm Dis 2024; 12:e1144. [PMID: 38363052 PMCID: PMC10870696 DOI: 10.1002/iid3.1144] [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: 09/26/2023] [Revised: 12/07/2023] [Accepted: 12/26/2023] [Indexed: 02/17/2024] Open
Abstract
Interleukin-22 (IL-22) is an important cytokine in the intestinal environment. IL-22 is mainly produced by immune cells and targeted at nonimmune cells such as epithelial and stromal cells in a broad array of tissues such as -but not restricted to- the liver and adipose tissue. IL-22 therefore connects immune functions with metabolic functions of the host, and since it is induced by the microbiota, connects host functioning to the outside environment. IL-22 induces epithelial cell proliferation aiding in rapid epithelium regeneration and wound healing. Additionally, IL-22 activates antiapoptotic genes and DNA damage response pathways, enhancing epithelial cell survival. Recently, it has also been shown that IL-22 induces Paneth cell differentiation in humans. However, IL-22 can also contribute to intestinal epithelium damage and reduces microbial diversity in the intestine directly or indirectly by inducing excessive antimicrobial peptide production by epithelial cells. Moreover, IL-22 enhances angiogenesis and may therefore support tumorigenesis in the intestine. In conclusion, it appears that whether IL-22 has a beneficial or harmful effect in the mammalian intestine largely depends on its regulation. This review aims to provide a comprehensive overview of the current literature and emphasizes that IL-22 signaling outcome depends on the timing and duration of IL-22 production, the presence of it regulators such as IL-22BP, and the specific location of the cytokine production in the gastrointestinal tract.
Collapse
Affiliation(s)
- Hedi‐Britt Klotskova
- Host Microbe Interactomics, Animal Sciences GroupWageningen University and ResearchWageningenThe Netherlands
| | - Evelien Kidess
- Host Microbe Interactomics, Animal Sciences GroupWageningen University and ResearchWageningenThe Netherlands
| | - Adria L. Nadal
- Host Microbe Interactomics, Animal Sciences GroupWageningen University and ResearchWageningenThe Netherlands
| | - Sylvia Brugman
- Host Microbe Interactomics, Animal Sciences GroupWageningen University and ResearchWageningenThe Netherlands
| |
Collapse
|
4
|
Liu J, Liu K, Wang Y, Shi Z, Xu R, Zhang Y, Li J, Liu C, Xue B. Death receptor 5 is required for intestinal stem cell activity during intestinal epithelial renewal at homoeostasis. Cell Death Dis 2024; 15:27. [PMID: 38199990 PMCID: PMC10782029 DOI: 10.1038/s41419-023-06409-4] [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: 06/20/2023] [Revised: 12/14/2023] [Accepted: 12/21/2023] [Indexed: 01/12/2024]
Abstract
Intestinal epithelial renewal, which depends on the proliferation and differentiation of intestinal stem cells (ISCs), is essential for epithelial homoeostasis. Understanding the mechanism controlling ISC activity is important. We found that death receptor 5 (DR5) gene deletion (DR5-/-) mice had impaired epithelial absorption and barrier function, resulting in delayed weight gain, which might be related to the general reduction of differentiated epithelial cells. In DR5-/- mice, the expression of ISC marker genes, the number of Olfm4+ ISCs, and the number of Ki67+ and BrdU+ cells in crypt were reduced. Furthermore, DR5 deletion inhibited the expression of lineage differentiation genes driving ISC differentiation into enterocytes, goblet cells, enteroendocrine cells, and Paneth cells. Therefore, DR5 gene loss may inhibit the intestinal epithelial renewal by dampening ISC activity. The ability of crypts from DR5-/- mice to form organoids decreased, and selective DR5 activation by Bioymifi promoted organoid growth and the expression of ISC and intestinal epithelial cell marker genes. Silencing of endogenous DR5 ligand TRAIL in organoids down-regulated the expression of ISC and intestinal epithelial cell marker genes. So, DR5 expressed in intestinal crypts was involved in the regulation of ISC activity. DR5 deletion in vivo or activation in organoids inhibited or enhanced the activity of Wnt, Notch, and BMP signalling through regulating the production of Paneth cell-derived ISC niche factors. DR5 gene deletion caused apoptosis and DNA damage in transit amplifying cells by inhibiting ERK1/2 activity in intestinal crypts. Inhibition of ERK1/2 with PD0325901 dampened the ISC activity and epithelial regeneration. In organoids, when Bioymifi's effect in activating ERK1/2 activity was completely blocked by PD0325901, its role in stimulating ISC activity and promoting epithelial regeneration was also eliminated. In summary, DR5 in intestinal crypts is essential for ISC activity during epithelial renewal under homoeostasis.
Collapse
Affiliation(s)
- Jianbo Liu
- Department of Physiology and Pathophysiology, School of basic medical science, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Kaixuan Liu
- Department of Physiology and Pathophysiology, School of basic medical science, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Ying Wang
- Department of Physiology and Pathophysiology, School of basic medical science, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Ziru Shi
- Department of Physiology and Pathophysiology, School of basic medical science, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Runze Xu
- Department of Physiology and Pathophysiology, School of basic medical science, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Yundi Zhang
- Department of Physiology and Pathophysiology, School of basic medical science, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Jingxin Li
- Department of Physiology and Pathophysiology, School of basic medical science, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Chuanyong Liu
- Department of Physiology and Pathophysiology, School of basic medical science, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Bing Xue
- Department of Physiology and Pathophysiology, School of basic medical science, Cheeloo College of Medicine, Shandong University, Jinan, China.
| |
Collapse
|
5
|
Jeong H, Park J, Kang JH, Sabaté del Río J, Kong S, Park T. Organoid-Based Human Stomach Micro-Physiological System to Recapitulate the Dynamic Mucosal Defense Mechanism. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2300164. [PMID: 37525340 PMCID: PMC10520631 DOI: 10.1002/advs.202300164] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/08/2023] [Revised: 07/11/2023] [Indexed: 08/02/2023]
Abstract
Several stomach diseases are attributed to the dysregulation of physiological function of gastric mucosal barrier by pathogens. Gastric organoids are a promising tool to develop treatment strategies for gastric infections. However, their functional features of in vivo gastric mucosal barrier and host-microbe interactions are limited due to the lack of physiological stimuli. Herein, a human stomach micro-physiological system (hsMPS) with physiologically relevant gastric mucosal defense system is described based on the combination of organoid and MPS technology. A fluid flow enhanced epithelial-mesenchymal interaction in the hsMPS enables functional maturation of gastric epithelial cells, which allows for the recreation of mesh-like mucus layer containing high level of mucus protective peptides and well-developed epithelial junctional complexes. Furthermore, gastroprotection mechanisms against Helicobacter pylori (H. pylori) are successfully demonstrated in this system. Therefore, hsMPS represents a new in vitro tool for research where gastric mucosal defense mechanism is pivotal for developing therapeutic strategies.
Collapse
Affiliation(s)
- Hye‐Jin Jeong
- Department of Biomedical EngineeringUlsan National Institute of Science and TechnologyUlsan44919Republic of Korea
| | - Ji‐Hyeon Park
- Department of SurgerySeoul National University HospitalSeoul National University College of MedicineSeoul03080Republic of Korea
- Department of SurgeryGachon University Gil Medical CenterIncheon21565Republic of Korea
| | - Joo H. Kang
- Department of Biomedical EngineeringUlsan National Institute of Science and TechnologyUlsan44919Republic of Korea
| | - Jonathan Sabaté del Río
- Center for Soft and Living MatterInstitute for Basic Science (IBS)Ulsan44919Republic of Korea
| | - Seong‐Ho Kong
- Department of SurgerySeoul National University HospitalSeoul National University College of MedicineSeoul03080Republic of Korea
| | - Tae‐Eun Park
- Department of Biomedical EngineeringUlsan National Institute of Science and TechnologyUlsan44919Republic of Korea
| |
Collapse
|
6
|
Montes-Olivas S, Legge D, Lund A, Fletcher AG, Williams AC, Marucci L, Homer M. In-silico and in-vitro morphometric analysis of intestinal organoids. PLoS Comput Biol 2023; 19:e1011386. [PMID: 37578984 PMCID: PMC10473498 DOI: 10.1371/journal.pcbi.1011386] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Revised: 09/01/2023] [Accepted: 07/25/2023] [Indexed: 08/16/2023] Open
Abstract
Organoids offer a powerful model to study cellular self-organisation, the growth of specific tissue morphologies in-vitro, and to assess potential medical therapies. However, the intrinsic mechanisms of these systems are not entirely understood yet, which can result in variability of organoids due to differences in culture conditions and basement membrane extracts used. Improving the standardisation of organoid cultures is essential for their implementation in clinical protocols. Developing tools to assess and predict the behaviour of these systems may produce a more robust and standardised biological model to perform accurate clinical studies. Here, we developed an algorithm to automate crypt-like structure counting on intestinal organoids in both in-vitro and in-silico images. In addition, we modified an existing two-dimensional agent-based mathematical model of intestinal organoids to better describe the system physiology, and evaluated its ability to replicate budding structures compared to new experimental data we generated. The crypt-counting algorithm proved useful in approximating the average number of budding structures found in our in-vitro intestinal organoid culture images on days 3 and 7 after seeding. Our changes to the in-silico model maintain the potential to produce simulations that replicate the number of budding structures found on days 5 and 7 of in-vitro data. The present study aims to aid in quantifying key morphological structures and provide a method to compare both in-vitro and in-silico experiments. Our results could be extended later to 3D in-silico models.
Collapse
Affiliation(s)
- Sandra Montes-Olivas
- Department of Engineering Mathematics, University of Bristol, Bristol, United Kingdom
| | - Danny Legge
- Colorectal Tumour Biology Group, School of Cellular and Molecular Medicine, Faculty of Life Sciences, University of Bristol, Bristol, United Kingdom
| | - Abbie Lund
- Department of Engineering Mathematics, University of Bristol, Bristol, United Kingdom
| | - Alexander G. Fletcher
- School of Mathematics and Statistics, University of Sheffield, Sheffield, United Kingdom
- Bateson Centre, University of Sheffield, Sheffield, United Kingdom
| | - Ann C. Williams
- Colorectal Tumour Biology Group, School of Cellular and Molecular Medicine, Faculty of Life Sciences, University of Bristol, Bristol, United Kingdom
| | - Lucia Marucci
- Department of Engineering Mathematics, University of Bristol, Bristol, United Kingdom
- School of Cellular and Molecular Medicine, University of Bristol, Bristol, United Kingdom
- BrisSynBio, Bristol, United Kingdom
| | - Martin Homer
- Department of Engineering Mathematics, University of Bristol, Bristol, United Kingdom
| |
Collapse
|
7
|
Kulkarni P, Salgia R, Rangarajan G. Intrinsically disordered proteins and conformational noise: The hypothesis a decade later. iScience 2023; 26:107109. [PMID: 37408690 PMCID: PMC10319216 DOI: 10.1016/j.isci.2023.107109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/07/2023] Open
Abstract
Phenotypic plasticity is the ability of individual genotypes to produce different phenotypes in response to environmental perturbations. We previously postulated how conformational noise emanating from conformational dynamics of intrinsically disordered proteins (IDPs) which is distinct from transcriptional noise, can contribute to phenotypic switching by rewiring the cellular protein interaction network. Since most transcription factors are IDPs, we posited that conformational noise is an integral component of transcriptional noise implying that IDPs may amplify total noise in the system either stochastically or in response to environmental changes. Here, we review progress in elucidating the details of the hypothesis. We highlight empirical evidence supporting the hypothesis, discuss conceptual advances that underscore its fundamental importance and implications, and identify areas for future investigations.
Collapse
Affiliation(s)
- Prakash Kulkarni
- Department of Medical Oncology and Therapeutics Research, City of Hope National Medical Center, Duarte, CA, USA
- Department of Systems Biology, City of Hope National Medical Center, Duarte, CA, USA
| | - Ravi Salgia
- Department of Medical Oncology and Therapeutics Research, City of Hope National Medical Center, Duarte, CA, USA
| | - Govindan Rangarajan
- Department of Mathematics, Indian Institute of Science, Bangalore 560012, India
- Center for Neuroscience, Indian Institute of Science, Bangalore 560012, India
| |
Collapse
|
8
|
Yin L, Gao M, Xu L, Qi Y, Han L, Peng J. Single-cell analysis of cellular heterogeneity and interactions in the ischemia-reperfusion injured mouse intestine. J Pharm Anal 2023; 13:760-775. [PMID: 37577387 PMCID: PMC10422115 DOI: 10.1016/j.jpha.2023.02.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Revised: 01/12/2023] [Accepted: 02/01/2023] [Indexed: 02/10/2023] Open
Abstract
Nine major cell populations among 46,716 cells were identified in mouse intestinal ischemia‒reperfusion (II/R) injury by single-cell RNA sequencing. For enterocyte cells, 11 subclusters were found, in which enterocyte cluster 1 (EC1), enterocyte cluster 3 (EC3), and enterocyte cluster 8 (EC8) were newly discovered cells in ischemia 45 min/reperfusion 720 min (I 45 min/R 720 min) group. EC1 and EC3 played roles in digestion and absorption, and EC8 played a role in cell junctions. For TA cells, after ischemia 45 min/reperfusion 90 min (I 45 min/R 90 min), many TA cells at the stage of proliferation were identified. For Paneth cells, Paneth cluster 3 was observed in the resting state of normal jejunum. After I 45 min/R 90 min, three new subsets were found, in which Paneth cluster 1 had good antigen presentation activity. The main functions of goblet cells were to synthesize and secrete mucus, and a novel subcluster (goblet cluster 5) with highly proliferative ability was discovered in I 45 min/R 90 min group. As a major part of immune system, the changes in T cells with important roles were clarified. Notably, enterocyte cells secreted Guca2b to interact with Gucy2c receptor on the membranes of stem cells, TA cells, Paneth cells, and goblet cells to elicit intercellular communication. One marker known as glutathione S-transferase mu 3 (GSTM3) affected intestinal mucosal barrier function by adjusting mitogen-activated protein kinases (MAPK) signaling during II/R injury. The data on the heterogeneity of intestinal cells, cellular communication and the mechanism of GSTM3 provide a cellular basis for treating II/R injury.
Collapse
Affiliation(s)
- Lianhong Yin
- College of Pharmacy, Dalian Medical University, Dalian, Liaoning, 116044, China
| | - Meng Gao
- College of Pharmacy, Dalian Medical University, Dalian, Liaoning, 116044, China
| | - Lina Xu
- College of Pharmacy, Dalian Medical University, Dalian, Liaoning, 116044, China
| | - Yan Qi
- College of Pharmacy, Dalian Medical University, Dalian, Liaoning, 116044, China
| | - Lan Han
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, 230012, China
| | - Jinyong Peng
- College of Pharmacy, Dalian Medical University, Dalian, Liaoning, 116044, China
| |
Collapse
|
9
|
Bormann D, Gugerell A, Ankersmit HJ, Mildner M. Therapeutic Application of Cell Secretomes in Cutaneous Wound Healing. J Invest Dermatol 2023; 143:893-912. [PMID: 37211377 DOI: 10.1016/j.jid.2023.02.019] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 02/02/2023] [Accepted: 02/10/2023] [Indexed: 05/23/2023]
Abstract
Although the application of stem cells to chronic wounds emerged as a candidate therapy in the previous century, the mechanism of action remains unclear. Recent evidence has implicated secreted paracrine factors in the regenerative properties of cell-based therapies. In the last two decades, considerable research advances involving the therapeutic potential of stem cell secretomes have expanded the scope of secretome-based therapies beyond stem cell populations. In this study, we review the modes of action of cell secretomes in wound healing, important preconditioning strategies for enhancing their therapeutic efficacy, and clinical trials on secretome-based wound healing.
Collapse
Affiliation(s)
- Daniel Bormann
- Laboratory for Cardiac and Thoracic Diagnosis and Regeneration, Department of Thoracic Surgery, Medical University of Vienna, Vienna, Austria
| | - Alfred Gugerell
- Laboratory for Cardiac and Thoracic Diagnosis and Regeneration, Department of Thoracic Surgery, Medical University of Vienna, Vienna, Austria
| | - Hendrik Jan Ankersmit
- Laboratory for Cardiac and Thoracic Diagnosis and Regeneration, Department of Thoracic Surgery, Medical University of Vienna, Vienna, Austria
| | - Michael Mildner
- Department of Dermatology, Medical University of Vienna, Vienna, Austria.
| |
Collapse
|
10
|
Lotti R, Palazzo E, Quadri M, Dumas M, Schnebert S, Biondini D, Bianchini MA, Nizard C, Pincelli C, Marconi A. Isolation of an "Early" Transit Amplifying Keratinocyte Population in Human Epidermis: A Role for the Low Affinity Neurotrophin Receptor CD271. Stem Cells 2022; 40:1149-1161. [PMID: 36037263 PMCID: PMC9806768 DOI: 10.1093/stmcls/sxac060] [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: 04/28/2021] [Accepted: 08/16/2022] [Indexed: 01/12/2023]
Abstract
In the interfollicular epidermis (IFE), stem cells (KSC) generate transit amplifying (TA) cells that, after symmetric divisions, produce differentiating daughters. Here, we isolated and characterized the highly proliferative interfollicular epidermal basal cell population "early" TA (ETA) cells, based on their capacity to adhere to type IV collagen. Proliferation and colony-forming efficiency in ETA cells are lower than in KSC but higher than in "late" TA (LTA). Stemness, proliferation, and differentiation markers confirmed that ETA cells display a unique phenotype. Skin reconstructs derived from ETA cells present different features (epidermal thickness, Ki67, and Survivin expression), as compared to skin equivalents generated from either KSC or LTA cells. The low-affinity neurotrophin receptor CD271, which regulates the KSC to TA cell transition in the human epidermis through an on/off switch control mechanism, is predominantly expressed in ETA cells. Skin equivalents generated from siRNA CD271 ETA cells display a more proliferative and less differentiated phenotype, as compared to mock-derived reconstructs. Consistently, CD271 overexpression in LTA cells generates a more proliferative skin equivalent than mock LTA cells. Finally, the CD271 level declines with cellular senescence, while it induces a delay in p16INK4 expression. We conclude that ETA cells represent the first KSC progenitor with exclusive features. CD271 identifies and modulates ETA cells, thus participating in the early differentiation and regenerative capacity of the human epidermis.
Collapse
Affiliation(s)
- Roberta Lotti
- DermoLab, Department of Surgical, Medical, Dental and Morphological Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Elisabetta Palazzo
- DermoLab, Department of Surgical, Medical, Dental and Morphological Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Marika Quadri
- DermoLab, Department of Surgical, Medical, Dental and Morphological Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Marc Dumas
- LVMH Recherche, Life Sciences Department, Saint Jean de Braye, France
| | | | - Diego Biondini
- Pediatric Surgery Unit, Department of Pediatric Surgery, University of Modena and Reggio Emilia, Modena, Italy
| | - Maria Anastasia Bianchini
- Pediatric Surgery Unit, Department of Pediatric Surgery, University of Modena and Reggio Emilia, Modena, Italy
| | - Carine Nizard
- LVMH Recherche, Life Sciences Department, Saint Jean de Braye, France
| | - Carlo Pincelli
- DermoLab, Department of Surgical, Medical, Dental and Morphological Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Alessandra Marconi
- Corresponding author: Alessandra Marconi, MSc in Biology, Specialist in Clinical Pathology, DermoLab, Department of Surgical, Medical, Dental and Morphological Sciences, University of Modena and Reggio Emilia, Via Del Pozzo 71, 41124 Modena, Italy. Tel: +39 059 4222812; Fax: +39 059 4224271;
| |
Collapse
|
11
|
Wong YL, Okubo T, Uno E, Suda K, Ishii T. Role of CD99 in regulating homeostasis and differentiation in normal human epidermal keratinocytes. Biochem Biophys Res Commun 2022; 606:108-113. [PMID: 35339749 DOI: 10.1016/j.bbrc.2022.03.087] [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: 02/03/2022] [Revised: 03/07/2022] [Accepted: 03/16/2022] [Indexed: 11/02/2022]
Abstract
CD99 is a glycoprotein primarily expressed in immune cells. Physiologically, it is involved in the adhesion, migration, and development of immune cells. The presence of CD99 in the skin was first reported in 2016 and its function is yet to be determined. In this study, we aimed to understand the role of CD99 in the skin using normal human epidermal keratinocytes (NHEK). CD99 expression increased with the confluency of NHEK, while the CD99-high expressing NHEK lost their stem cell properties and played a role in barrier function. We characterized CD99-expressing NHEK as cells committed to early differentiation because they expressed early differentiation markers. However, the deficiency of CD99 in NHEK disrupted homeostasis and caused aberrant differentiation, as evidenced by larger cells with lesser Ki67 staining and higher expression of terminal differentiation markers. Hence, we propose that CD99 is involved in maintaining homeostasis and initiating early differentiation in the skin.
Collapse
Affiliation(s)
- Yi Li Wong
- Basic Research Development Division, Rohto Pharmaceutical Co., Ltd., 6-5-4 Kunimidai, Kizugawa, Kyoto, 619-0216, Japan
| | - Toru Okubo
- Basic Research Development Division, Rohto Pharmaceutical Co., Ltd., 6-5-4 Kunimidai, Kizugawa, Kyoto, 619-0216, Japan.
| | - Eiko Uno
- Basic Research Development Division, Rohto Pharmaceutical Co., Ltd., 6-5-4 Kunimidai, Kizugawa, Kyoto, 619-0216, Japan
| | - Kazuma Suda
- Basic Research Development Division, Rohto Pharmaceutical Co., Ltd., 6-5-4 Kunimidai, Kizugawa, Kyoto, 619-0216, Japan
| | - Tsuyoshi Ishii
- Basic Research Development Division, Rohto Pharmaceutical Co., Ltd., 6-5-4 Kunimidai, Kizugawa, Kyoto, 619-0216, Japan
| |
Collapse
|
12
|
Transit Amplifying Progenitors in the Cerebellum: Similarities to and Differences from Transit Amplifying Cells in Other Brain Regions and between Species. Cells 2022; 11:cells11040726. [PMID: 35203375 PMCID: PMC8870322 DOI: 10.3390/cells11040726] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Revised: 02/07/2022] [Accepted: 02/15/2022] [Indexed: 12/15/2022] Open
Abstract
Transit amplification of neural progenitors/precursors is widely used in the development of the central nervous system and for tissue homeostasis. In most cases, stem cells, which are relatively less proliferative, first differentiate into transit amplifying cells, which are more proliferative, losing their stemness. Subsequently, transit amplifying cells undergo a limited number of mitoses and differentiation to expand the progeny of differentiated cells. This step-by-step proliferation is considered an efficient system for increasing the number of differentiated cells while maintaining the stem cells. Recently, we reported that cerebellar granule cell progenitors also undergo transit amplification in mice. In this review, we summarize our and others’ recent findings and the prospective contribution of transit amplification to neural development and evolution, as well as the molecular mechanisms regulating transit amplification.
Collapse
|
13
|
Xie G, Peng Z, Liang J, Larabee SM, Drachenberg CB, Yfantis H, Raufman JP. Zinc finger protein 277 is an intestinal transit-amplifying cell marker and colon cancer oncogene. JCI Insight 2022; 7:150894. [PMID: 35015732 PMCID: PMC8876557 DOI: 10.1172/jci.insight.150894] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Accepted: 01/05/2022] [Indexed: 01/10/2023] Open
Abstract
Sustained proliferative signaling and resisting cell death are hallmarks of cancer. Zinc finger protein 277 (ZNF277; murine Zfp277), a transcription factor regulating cellular senescence, is overexpressed in colon cancer, but its actions in intestinal homeostasis and neoplasia are unclear. Using human and murine intestine, human colon cancer cells, and ApcMin/+ mice with dysregulated β-catenin signaling and exuberant intestinal neoplasia, we explored the actions of ZNF277/Zfp277 and defined the underlying mechanisms. In normal human and murine intestine, ZNF277/Zfp277 was expressed uniquely in early stem cell progenitors, undifferentiated transit-amplifying cells (TACs). Zfp277 was overexpressed in the ApcMin/+ mouse colon, implicating ZNF277/Zfp277 as a transcriptional target of β-catenin signaling. We confirmed this by showing β-catenin knockdown reduced ZNF277 expression and, using chromatin IP, identified 2 β-catenin binding sites in the ZNF277 promoter. Zfp277 deficiency attenuated intestinal epithelial cell proliferation and tumor formation, and it strikingly prolonged ApcMin/+ mouse survival. RNA-Seq and PCR analyses revealed that Zfp277 modulates expression of genes in key cancer pathways, including β-catenin signaling, the HOXD family that regulates development, and p21WAF1, a cell cycle inhibitor and tumor suppressor. In both human colon cancer cells and the murine colon, ZNF277/Zfp277 deficiency induced p21WAF1 expression and promoted senescence. Our findings identify ZNF277/Zfp277 as both a TAC marker and colon cancer oncogene that regulates cellular proliferation and senescence, in part by repressing p21WAF1 expression.
Collapse
Affiliation(s)
- Guofeng Xie
- University of Maryland School of Medicine, Baltimore, United States of America
| | - Zhongsheng Peng
- Department of Medicine, University of Maryland School of Medicine, Baltimore, United States of America
| | - Jinqing Liang
- Department of Medicine, University of Maryland School of Medicine, Baltimore, United States of America
| | - Shannon M Larabee
- Department of Surgery, University of Maryland School of Medicine, Baltimore, United States of America
| | - Cinthia B Drachenberg
- Department of Pathology, University of Maryland School of Medicine, Baltimore, United States of America
| | - Harris Yfantis
- Department of Pathology and Laboratory Medicine, Baltimore Veterans Affairs Medical Center, Baltimore, United States of America
| | - Jean-Pierre Raufman
- Department of Medicine, University of Maryland School of Medicine, Baltimore, United States of America
| |
Collapse
|
14
|
Leemans B, Bromfield EG, Stout TAE, Vos M, Van Der Ham H, Van Beek R, Van Soom A, Gadella BM, Henning H. Developing a reproducible protocol for culturing functional confluent monolayers of differentiated equine oviduct epithelial cells. Biol Reprod 2021; 106:710-729. [PMID: 34962550 PMCID: PMC9040661 DOI: 10.1093/biolre/ioab243] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Revised: 07/14/2021] [Accepted: 12/24/2021] [Indexed: 11/13/2022] Open
Abstract
We describe the development of two methods for obtaining confluent monolayers of polarized, differentiated equine oviduct epithelial cells (EOEC) in Transwell inserts and microfluidic chips. EOECs from the ampulla were isolated post-mortem and seeded either (1) directly onto a microporous membrane as differentiated EOECs (direct seeding protocol) or (2) first cultured to a confluent de-differentiated monolayer in conventional wells, then trypsinized and seeded onto a microporous membrane (re-differentiation protocol). Maintenance or induction of EOEC differentiation in these systems was achieved by air-liquid interface introduction. Monolayers cultured via both protocols were characterized by columnar, cytokeratin 19-positive EOECs in Transwell inserts. However, only the re-differentiation protocol could be transferred successfully to the microfluidic chips. Integrity of the monolayers was confirmed by transepithelial resistance measurements, tracer flux and the demonstration of an intimate network of tight junctions. Using the direct protocol, 28% of EOECs showed secondary cilia at the apical surface in a diffuse pattern. In contrast, re-differentiated polarized EOECs rarely showed secondary cilia in either culture system (>90% of the monolayers showed <1% ciliated EOECs). Occasionally (5-10%), re-differentiated monolayers with 11-27% EOECs with secondary cilia in a diffuse pattern were obtained. Additionally, nuclear progesterone receptor expression was found to be inhibited by simulated luteal phase hormone concentrations, and sperm binding to cilia was higher for re-differentiated EOEC monolayers exposed to estrogen-progesterone concentrations mimicking the follicular rather than luteal phase. Overall, a functional equine oviduct model was established with close morphological resemblance to in vivo oviduct epithelium.
Collapse
Affiliation(s)
- Bart Leemans
- Department of Reproduction, Obstetrics and Herd Health, Faculty of Veterinary Medicine, Ghent University, Belgium.,Departments of Clinical Sciences, Utrecht University, The Netherlands
| | - Elizabeth G Bromfield
- Biomolecular Health Sciences, Utrecht University, The Netherlands.,Priority Research Centre for Reproductive Science, Faculty of Science, University of Newcastle, Australia
| | - Tom A E Stout
- Departments of Clinical Sciences, Utrecht University, The Netherlands
| | - Mabel Vos
- Departments of Clinical Sciences, Utrecht University, The Netherlands
| | - Hanna Van Der Ham
- Departments of Clinical Sciences, Utrecht University, The Netherlands
| | - Ramada Van Beek
- Departments of Clinical Sciences, Utrecht University, The Netherlands
| | - Ann Van Soom
- Department of Reproduction, Obstetrics and Herd Health, Faculty of Veterinary Medicine, Ghent University, Belgium
| | - Bart M Gadella
- Department of Reproduction, Obstetrics and Herd Health, Faculty of Veterinary Medicine, Ghent University, Belgium.,Biomolecular Health Sciences, Utrecht University, The Netherlands.,Population Health Sciences, Faculty of Veterinary Medicine, Utrecht University, The Netherlands
| | - Heiko Henning
- Population Health Sciences, Faculty of Veterinary Medicine, Utrecht University, The Netherlands
| |
Collapse
|
15
|
Knyazev E, Maltseva D, Raygorodskaya M, Shkurnikov M. HIF-Dependent NFATC1 Activation Upregulates ITGA5 and PLAUR in Intestinal Epithelium in Inflammatory Bowel Disease. Front Genet 2021; 12:791640. [PMID: 34858489 PMCID: PMC8632048 DOI: 10.3389/fgene.2021.791640] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Accepted: 10/27/2021] [Indexed: 12/11/2022] Open
Abstract
Intestinal epithelial cells exist in physiological hypoxia, leading to hypoxia-inducible factor (HIF) activation and supporting barrier function and cell metabolism of the intestinal epithelium. In contrast, pathological hypoxia is a common feature of some chronic disorders, including inflammatory bowel disease (IBD). This work was aimed at studying HIF-associated changes in the intestinal epithelium in IBD. In the first step, a list of genes responding to chemical activation of hypoxia was obtained in an in vitro intestinal cell model with RNA sequencing. Cobalt (II) chloride and oxyquinoline treatment of both undifferentiated and differentiated Caco-2 cells activate the HIF-signaling pathway according to gene set enrichment analysis. The core gene set responding to chemical hypoxia stimulation in the intestinal model included 115 upregulated and 69 downregulated genes. Of this set, protein product was detected for 32 genes, and fold changes in proteome and RNA sequencing significantly correlate. Analysis of publicly available RNA sequencing set of the intestinal epithelial cells of patients with IBD confirmed HIF-1 signaling pathway activation in sigmoid colon of patients with ulcerative colitis and terminal ileum of patients with Crohn's disease. Of the core gene set from the gut hypoxia model, expression activation of ITGA5 and PLAUR genes encoding integrin α5 and urokinase-type plasminogen activator receptor (uPAR) was detected in IBD specimens. The interaction of these molecules can activate cell migration and regenerative processes in the epithelium. Transcription factor analysis with the previously developed miRGTF tool revealed the possible role of HIF1A and NFATC1 in the regulation of ITGA5 and PLAUR gene expression. Detected genes can serve as markers of IBD progression and intestinal hypoxia.
Collapse
Affiliation(s)
- Evgeny Knyazev
- Laboratory of Microfluidic Technologies for Biomedicine, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, Moscow, Russia
| | - Diana Maltseva
- Faculty of Biology and Biotechnology, National Research University Higher School of Economics (HSE), Moscow, Russia
| | - Maria Raygorodskaya
- Laboratory of Microfluidic Technologies for Biomedicine, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, Moscow, Russia
| | - Maxim Shkurnikov
- Laboratory of Microfluidic Technologies for Biomedicine, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, Moscow, Russia.,Faculty of Biology and Biotechnology, National Research University Higher School of Economics (HSE), Moscow, Russia.,National Center of Medical Radiological Research, P. Hertsen Moscow Oncology Research Institute, Moscow, Russia
| |
Collapse
|
16
|
Kossard S. Eruptive Necrotizing Infundibular Crystalline Folliculitis: An Expression of an Abortive Sebaceous Follicular Repair Pathway Linked to Committed Infundibular Stem Cells? Am J Dermatopathol 2021; 43:867-870. [PMID: 34735106 PMCID: PMC8601669 DOI: 10.1097/dad.0000000000002022] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
ABSTRACT Necrotizing infundibular crystalline folliculitis is a rare entity, which is a distinctive clinical and histopathological entity. Eruptive yellow waxy umbilicated folliculocentric plugs clinically correspond to pale crystalline filaments embedded in an amorphous sebum-rich material. Remarkably, only the superficial infundibular ostia remain, and the distended cavity is devoid of a follicular or sebaceous gland remnant. The pathogenesis of this enigmatic event remains to be established. The emergence of necrotizing infundibular crystalline folliculitis (NICF) as a paradoxical side effect of antitumor inhibitors epidermal growth factor receptor vascular endothelial growth factor and more recently programmed death-1 represents the expression of altered molecular pathways that underpin the pathogenesis of NICF. To explore these pathways, it is necessary to explore the hierarchy of follicular stem cells, particularly the potential role of committed infundibular stem cells that play a key role in wound healing. Committed infundibular stem cells are closely linked to the sebaceous gland stem cell axis, and this has relevance in the process of homeostatic repair of sebaceous follicles in the wake of folliculitis. The unscheduled modulation of this infundibular homeostatic sebaceous repair axis by epidermal growth factor receptor vascular endothelial growth factor, and programmed death-1 may lead to an aberrant outcome with metaplasia of infundibular keratinocytes to sebocytes. In the absence of sebaceous gland differentiation, these metaplastic infundibular sebocyte cells would lead to the consumption and loss of the infundibulum as a result of holocrine sebum production. This conceptual pathogenic pathway for NICF is constructed by incorporating recent advances in the fields of follicular stem cells, wound repair, follicular homeostasis, regulatory T cells, and molecular pathways linked to the biologicals inducing NICF.
Collapse
Affiliation(s)
- Steven Kossard
- Laverty Pathology, Kossard Dermatopatholgists, Sydney, New South Wales, Australia
| |
Collapse
|
17
|
Low E, Alimohammadiha G, Smith LA, Costello LF, Przyborski SA, von Zglinicki T, Miwa S. How good is the evidence that cellular senescence causes skin ageing? Ageing Res Rev 2021; 71:101456. [PMID: 34487917 PMCID: PMC8524668 DOI: 10.1016/j.arr.2021.101456] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Revised: 08/25/2021] [Accepted: 08/31/2021] [Indexed: 12/11/2022]
Abstract
Skin is the largest organ of the body with important protective functions, which become compromised with time due to both intrinsic and extrinsic ageing processes. Cellular senescence is the primary ageing process at cell level, associated with loss of proliferative capacity, mitochondrial dysfunction and significantly altered patterns of expression and secretion of bioactive molecules. Intervention experiments have proven cell senescence as a relevant cause of ageing in many organs. In case of skin, accumulation of senescence in all major compartments with ageing is well documented and might be responsible for most, if not all, the molecular changes observed during ageing. Incorporation of senescent cells into in-vitro skin models (specifically 3D full thickness models) recapitulates changes typically associated with skin ageing. However, crucial evidence is still missing. A beneficial effect of senescent cell ablation on skin ageing has so far only been shown following rather unspecific interventions or in transgenic mouse models. We conclude that evidence for cellular senescence as a relevant cause of intrinsic skin ageing is highly suggestive but not yet completely conclusive.
Collapse
Affiliation(s)
- Evon Low
- Ageing Biology Laboratories, Newcastle University Biosciences Institute, Newcastle University, Newcastle upon Tyne NE4 5PL, UK
| | - Ghazaleh Alimohammadiha
- Ageing Biology Laboratories, Newcastle University Biosciences Institute, Newcastle University, Newcastle upon Tyne NE4 5PL, UK
| | - Lucy A Smith
- Department of Biosciences, Durham University, South Road, Durham DH1 3LE, UK
| | - Lydia F Costello
- Department of Biosciences, Durham University, South Road, Durham DH1 3LE, UK
| | - Stefan A Przyborski
- Department of Biosciences, Durham University, South Road, Durham DH1 3LE, UK
| | - Thomas von Zglinicki
- Ageing Biology Laboratories, Newcastle University Biosciences Institute, Newcastle University, Newcastle upon Tyne NE4 5PL, UK.
| | - Satomi Miwa
- Ageing Biology Laboratories, Newcastle University Biosciences Institute, Newcastle University, Newcastle upon Tyne NE4 5PL, UK
| |
Collapse
|
18
|
Li JM, Kim S, Zhang Y, Bian F, Hu J, Lu R, Pflugfelder SC, Chen R, Li DQ. Single-Cell Transcriptomics Identifies a Unique Entity and Signature Markers of Transit-Amplifying Cells in Human Corneal Limbus. Invest Ophthalmol Vis Sci 2021; 62:36. [PMID: 34297801 PMCID: PMC8300054 DOI: 10.1167/iovs.62.9.36] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Purpose Differentiated from adult stem cells (ASCs), transit-amplifying cells (TACs) play an important role in tissue homeostasis, development, and regeneration. This study aimed to characterize the gene expression profile of a candidate TAC population in limbal basal epithelial cells using single-cell RNA sequencing (scRNA-seq). Methods Single cells isolated from the basal corneal limbus were subjected to scRNA-seq using the 10x Genomics platform. Cell types were clustered by graph-based visualization methods and unbiased computational analysis. BrdU proliferation assays, immunofluorescent staining, and real-time reverse transcription quantitative polymerase chain reaction were performed using multiple culture models of primary human limbal epithelial cells to characterize the TAC pool. Results Single-cell transcriptomics of 16,360 limbal basal cells revealed 12 cell clusters. A unique cluster (3.21% of total cells) was identified as a TAC entity, based on its less differentiated progenitor status and enriched exclusive proliferation marker genes, with 98.1% cells in S and G2/M phases. The cell cycle-dependent genes were revealed to be largely enriched by the TAC population. The top genes were characterized morphologically and functionally at protein and mRNA levels. The specific expression patterns of RRM2, TK1, CENPF, NUSAP1, UBE2C, and CDC20 were well correlated in a time- and cycle-dependent manner with proliferation stages in the cell growth and regeneration models. Conclusions For the first time, to the best of our knowledge, we have identified a unique TAC entity and uncovered a group of cell cycle-dependent genes that serve as TAC signature markers. The findings provide insight into ASCs and TACs and lay the foundation for understanding corneal homeostasis and diseases.
Collapse
Affiliation(s)
- Jin-Miao Li
- Ocular Surface Center, Cullen Eye Institute, Department of Ophthalmology, Baylor College of Medicine, Houston, Texas, United States.,State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Sangbae Kim
- Human Genome Sequencing Center, Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, United States
| | - Yun Zhang
- Ocular Surface Center, Cullen Eye Institute, Department of Ophthalmology, Baylor College of Medicine, Houston, Texas, United States
| | - Fang Bian
- Ocular Surface Center, Cullen Eye Institute, Department of Ophthalmology, Baylor College of Medicine, Houston, Texas, United States
| | - Jiaoyue Hu
- Ocular Surface Center, Cullen Eye Institute, Department of Ophthalmology, Baylor College of Medicine, Houston, Texas, United States
| | - Rong Lu
- Ocular Surface Center, Cullen Eye Institute, Department of Ophthalmology, Baylor College of Medicine, Houston, Texas, United States.,State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Stephen C Pflugfelder
- Ocular Surface Center, Cullen Eye Institute, Department of Ophthalmology, Baylor College of Medicine, Houston, Texas, United States
| | - Rui Chen
- Human Genome Sequencing Center, Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, United States
| | - De-Quan Li
- Ocular Surface Center, Cullen Eye Institute, Department of Ophthalmology, Baylor College of Medicine, Houston, Texas, United States
| |
Collapse
|
19
|
Zullo KM, Douglas B, Maloney NM, Ji Y, Wei Y, Herbine K, Cohen R, Pastore C, Cramer Z, Wang X, Wei W, Somsouk M, Hung LY, Lengner C, Kohanski MH, Cohen NA, Herbert DR. LINGO3 regulates mucosal tissue regeneration and promotes TFF2 dependent recovery from colitis. Scand J Gastroenterol 2021; 56:791-805. [PMID: 33941035 PMCID: PMC8647134 DOI: 10.1080/00365521.2021.1917650] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Aim: Recovery of damaged mucosal surfaces following inflammatory insult requires diverse regenerative mechanisms that remain poorly defined. Previously, we demonstrated that the reparative actions of Trefoil Factor 3 (TFF3) depend upon the enigmatic receptor, leucine rich repeat and immunoglobulin-like domain containing nogo receptor 2 (LINGO2). This study examined the related orphan receptor LINGO3 in the context of intestinal tissue damage to determine whether LINGO family members are generally important for mucosal wound healing and maintenance of the intestinal stem cell (ISC) compartment needed for turnover of mucosal epithelium.Methods and Results: We find that LINGO3 is broadly expressed on human enterocytes and sparsely on discrete cells within the crypt niche, that contains ISCs. Loss of function studies indicate that LINGO3 is involved in recovery of normal intestinal architecture following dextran sodium sulfate (DSS)-induced colitis, and that LINGO3 is needed for therapeutic action of the long acting TFF2 fusion protein (TFF2-Fc), including a number of signaling pathways critical for cell proliferation and wound repair. LINGO3-TFF2 protein-protein interactions were relatively weak however and LINGO3 was only partially responsible for TFF2 induced MAPK signaling suggesting additional un-identified components of a receptor complex. However, deficiency in either TFF2 or LINGO3 abrogated budding/growth of intestinal organoids and reduced expression of the intestinal ISC gene leucine-rich repeat-containing G-protein coupled receptor 5 (LGR5), indicating homologous roles for these proteins in tissue regeneration, possibly via regulation of ISCs in the crypt niche.Conclusion: We propose that LINGO3 serves a previously unappreciated role in promoting mucosal wound healing.
Collapse
Affiliation(s)
- Kelly M. Zullo
- Department of Pathobiology, University of Pennsylvania School of Veterinary Medicine, Philadelphia, PA 19104
| | - Bonnie Douglas
- Department of Pathobiology, University of Pennsylvania School of Veterinary Medicine, Philadelphia, PA 19104
| | - Nicole M. Maloney
- Department of Pathobiology, University of Pennsylvania School of Veterinary Medicine, Philadelphia, PA 19104
| | - Yingbiao Ji
- Department of Pathobiology, University of Pennsylvania School of Veterinary Medicine, Philadelphia, PA 19104
| | - Yun Wei
- Department of Medicine, Division of Experimental Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Karl Herbine
- Department of Pathobiology, University of Pennsylvania School of Veterinary Medicine, Philadelphia, PA 19104
| | - Rachel Cohen
- Department of Pathobiology, University of Pennsylvania School of Veterinary Medicine, Philadelphia, PA 19104
| | - Christopher Pastore
- Department of Pathobiology, University of Pennsylvania School of Veterinary Medicine, Philadelphia, PA 19104
| | - Zvi Cramer
- Department of Biomedical Sciences, University of Pennsylvania School of Veterinary Medicine, Philadelphia, PA 19104
| | - Xin Wang
- Department of Biomedical Sciences, University of Pennsylvania School of Veterinary Medicine, Philadelphia, PA 19104
| | - Wenjie Wei
- Department of Ophthalmology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19147
| | - Ma Somsouk
- Department of Pathology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Li Yin Hung
- Department of Pathobiology, University of Pennsylvania School of Veterinary Medicine, Philadelphia, PA 19104,Department of Medicine, Division of Experimental Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Christopher Lengner
- Department of Biomedical Sciences, University of Pennsylvania School of Veterinary Medicine, Philadelphia, PA 19104
| | - Michael H. Kohanski
- Department of Otorhinolaryngology—Head and Neck Surgery, Perelman School of Medicine at The University of Pennsylvania, Philadelphia, PA 19104,The Corporal Michael J. Crescenz VA Medical Center Surgical Service, Philadelphia, PA 19104
| | - Noam A. Cohen
- Department of Otorhinolaryngology—Head and Neck Surgery, Perelman School of Medicine at The University of Pennsylvania, Philadelphia, PA 19104,The Corporal Michael J. Crescenz VA Medical Center Surgical Service, Philadelphia, PA 19104,Monell Chemical Senses Center, Philadelphia, PA 19104
| | - De’Broski R. Herbert
- Department of Pathobiology, University of Pennsylvania School of Veterinary Medicine, Philadelphia, PA 19104,Department of Medicine, Division of Experimental Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| |
Collapse
|
20
|
Muhr J, Hagey DW. The cell cycle and differentiation as integrated processes: Cyclins and CDKs reciprocally regulate Sox and Notch to balance stem cell maintenance. Bioessays 2021; 43:e2000285. [PMID: 34008221 DOI: 10.1002/bies.202000285] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Revised: 04/15/2021] [Accepted: 04/21/2021] [Indexed: 12/18/2022]
Abstract
Development and maintenance of diverse organ systems require context-specific regulation of stem cell behaviour. We hypothesize that this is achieved via reciprocal regulation between the cell cycle machinery and differentiation factors. This idea is supported by the parallel evolutionary emergence of differentiation pathways, cell cycle components and complex multicellularity. In addition, the activities of different cell cycle phases have been found to bias cells towards stem cell maintenance or differentiation. Finally, several direct mechanistic links between these two processes have been established. Here, we focus on interactions between cyclin-CDK complexes and differentiation regulators of the Notch pathway and Sox family of transcription factors within the context of pluripotent and neural stem cells. Thus, this hypothesis formalizes the links between these two processes as an integrated network. Since such factors are common to all stem cells, better understanding their interconnections will help to explain their behaviour in health and disease.
Collapse
Affiliation(s)
- Jonas Muhr
- Department of Cell and Molecular Biology, Karolinska Institute, Stockholm, Sweden
| | - Daniel W Hagey
- Department of Laboratory Medicine, Karolinska Institute, Stockholm, Sweden
| |
Collapse
|
21
|
Du J, Jing J, Chen S, Yuan Y, Feng J, Ho TV, Sehgal P, Xu J, Jiang X, Chai Y. Arid1a regulates cell cycle exit of transit-amplifying cells by inhibiting the Aurka-Cdk1 axis in mouse incisor. Development 2021; 148:dev198838. [PMID: 33766930 PMCID: PMC8077510 DOI: 10.1242/dev.198838] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Accepted: 03/18/2021] [Indexed: 12/14/2022]
Abstract
Stem cells self-renew or give rise to transit-amplifying cells (TACs) that differentiate into specific functional cell types. The fate determination of stem cells to TACs and their transition to fully differentiated progeny is precisely regulated to maintain tissue homeostasis. Arid1a, a core component of the switch/sucrose nonfermentable complex, performs epigenetic regulation of stage- and tissue-specific genes that is indispensable for stem cell homeostasis and differentiation. However, the functional mechanism of Arid1a in the fate commitment of mesenchymal stem cells (MSCs) and their progeny is not clear. Using the continuously growing adult mouse incisor model, we show that Arid1a maintains tissue homeostasis through limiting proliferation, promoting cell cycle exit and differentiation of TACs by inhibiting the Aurka-Cdk1 axis. Loss of Arid1a overactivates the Aurka-Cdk1 axis, leading to expansion of the mitotic TAC population but compromising their differentiation ability. Furthermore, the defective homeostasis after loss of Arid1a ultimately leads to reduction of the MSC population. These findings reveal the functional significance of Arid1a in regulating the fate of TACs and their interaction with MSCs to maintain tissue homeostasis.
Collapse
Affiliation(s)
- Jiahui Du
- Center for Craniofacial Molecular Biology, University of Southern California, Los Angeles, CA, 90033, USA
- Department of Prosthodontics, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Junjun Jing
- Center for Craniofacial Molecular Biology, University of Southern California, Los Angeles, CA, 90033, USA
| | - Shuo Chen
- Center for Craniofacial Molecular Biology, University of Southern California, Los Angeles, CA, 90033, USA
| | - Yuan Yuan
- Center for Craniofacial Molecular Biology, University of Southern California, Los Angeles, CA, 90033, USA
| | - Jifan Feng
- Center for Craniofacial Molecular Biology, University of Southern California, Los Angeles, CA, 90033, USA
| | - Thach-Vu Ho
- Center for Craniofacial Molecular Biology, University of Southern California, Los Angeles, CA, 90033, USA
| | - Prerna Sehgal
- Center for Craniofacial Molecular Biology, University of Southern California, Los Angeles, CA, 90033, USA
| | - Jian Xu
- Center for Craniofacial Molecular Biology, University of Southern California, Los Angeles, CA, 90033, USA
| | - Xinquan Jiang
- Department of Prosthodontics, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Yang Chai
- Center for Craniofacial Molecular Biology, University of Southern California, Los Angeles, CA, 90033, USA
| |
Collapse
|
22
|
Malijauskaite S, Connolly S, Newport D, McGourty K. Gradients in the in vivo intestinal stem cell compartment and their in vitro recapitulation in mimetic platforms. Cytokine Growth Factor Rev 2021; 60:76-88. [PMID: 33858768 DOI: 10.1016/j.cytogfr.2021.03.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Accepted: 03/05/2021] [Indexed: 02/07/2023]
Abstract
Intestinal tissue, and specifically its mucosal layer, is a complex and gradient-rich environment. Gradients of soluble factor (BMP, Noggin, Notch, Hedgehog, and Wnt), insoluble extracellular matrix proteins (laminins, collagens, fibronectin, and their cognate receptors), stromal stiffness, oxygenation, and sheer stress induced by luminal fluid flow at the crypt-villus axis controls and supports healthy intestinal tissue homeostasis. However, due to current technological challenges, very few of these features have so far been included in in vitro intestinal tissue mimetic platforms. In this review, the tightly defined and dynamic microenvironment of the intestinal tissue is presented in detail. Additionally, the authors introduce the current state-of-the-art intestinal tissue mimetic platforms, as well as the design drawbacks and challenges they face while attempting to capture the complexity of the intestinal tissue's physiology. Finally, the compositions of an "idealized" mimetic system is presented to guide future developmental efforts.
Collapse
Affiliation(s)
- Sigita Malijauskaite
- Dept. of Chemical Sciences, University of Limerick, Limerick, Ireland; Bernal Institute, University of Limerick, Limerick, Ireland.
| | - Sinead Connolly
- Bernal Institute, University of Limerick, Limerick, Ireland; School of Engineering, University of Limerick, Limerick, Ireland.
| | - David Newport
- Bernal Institute, University of Limerick, Limerick, Ireland; Health Research Institute (HRI), University of Limerick, Limerick, Ireland; School of Engineering, University of Limerick, Limerick, Ireland.
| | - Kieran McGourty
- Dept. of Chemical Sciences, University of Limerick, Limerick, Ireland; Bernal Institute, University of Limerick, Limerick, Ireland; Health Research Institute (HRI), University of Limerick, Limerick, Ireland.
| |
Collapse
|
23
|
Zhang L, Cen Y, Huang Q, Li H, Mo X, Meng W, Chen J. Computational flow cytometric analysis to detect epidermal subpopulations in human skin. Biomed Eng Online 2021; 20:22. [PMID: 33596908 PMCID: PMC7891025 DOI: 10.1186/s12938-021-00858-8] [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: 10/29/2020] [Accepted: 02/05/2021] [Indexed: 02/08/2023] Open
Abstract
Background The detection and dissection of epidermal subgroups could lead to an improved understanding of skin homeostasis and wound healing. Flow cytometric analysis provides an effective method to detect the surface markers of epidermal cells while producing high-dimensional data files. Methods A 9-color flow cytometric panel was optimized to reveal the heterogeneous subgroups in the epidermis of human skin. The subsets of epidermal cells were characterized using automated methods based on dimensional reduction approaches (viSNE) and clustering with Spanning-tree Progression Analysis of Density-normalized Events (SPADE). Results The manual analysis revealed differences in epidermal distribution between body sites based on a series biaxial gating starting with the expression of CD49f and CD29. The computational analysis divided the whole epidermal cell population into 25 clusters according to the surface marker phenotype with SPADE. This automatic analysis delineated the differences between body sites. The consistency of the results was confirmed with PhenoGraph. Conclusion A multicolor flow cytometry panel with a streamlined computational analysis pipeline is a feasible approach to delineate the heterogeneity of the epidermis in human skin.
Collapse
Affiliation(s)
- Lidan Zhang
- Department of Burn and Plastic Surgery, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Ying Cen
- Department of Burn and Plastic Surgery, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Qiaorong Huang
- Laboratory of Stem Cell Biology, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Huifang Li
- Laboratory of Stem Cell Biology, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Xianming Mo
- Laboratory of Stem Cell Biology, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Wentong Meng
- Laboratory of Stem Cell Biology, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China.
| | - Junjie Chen
- Department of Burn and Plastic Surgery, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China.
| |
Collapse
|
24
|
Mathew JG, Bowman AS, Saab J, Busam KJ, Nehal K, Pulitzer M. Next Generation Sequencing analysis suggests varied multistep mutational pathogenesis for Endocrine Mucin Producing Sweat Gland Carcinoma with comments on INSM1 and MUC2 suggesting a conjunctival origin. J Am Acad Dermatol 2021; 86:1072-1079. [PMID: 33515627 PMCID: PMC9627720 DOI: 10.1016/j.jaad.2020.11.073] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Revised: 10/14/2020] [Accepted: 11/15/2020] [Indexed: 10/22/2022]
Abstract
Endocrine mucin-producing sweat gland carcinoma (EMPSGC) is a low-grade eyelid tumor. Small biopsies and insensitive immunohistochemistry predispose to misdiagnosis. We aimed to identify clarifying immunohistochemical and/or molecular markers. Clinicopathologic data (22 cases) was reviewed. Immunohistochemistry (Insulinoma-associated protein-1(INSM1), BCL-2, MUC2, MUC4, androgen-receptor, Beta-catenin, MCPyV) and next generation sequencing (MSK-IMPACT, 468 genes) was performed (3 cases). Female (n=15) and male (n=7) patients, mean-age 71.8 years (53-88), had eyelid/periorbital tumors (>90%) with mucin-containing solid/cystic neuroendocrine pathology. Immunohistochemistry (INSM1, BCL2, androgen-receptor, RB1, Beta-catenin) was diffusely-positive (5/5), MUC2 partial, MUC4 focal, and MCPyV negative. MSK-IMPACT identified 12 single-nucleotide-variants and one in-frame deletion in 3 cases, each with DNA damage response/repair (BRD4, PPP4R2, RTEL1) and tumor-suppressor pathway (BRD4, TP53, TSC1, LATS2) mutations. Microsatellite instability, copy number alterations, and structural alterations were absent. INSM1 and MUC2 are positive in EMPSGC. MUC2 positivity suggests conjunctival origin. Multistep pathogenesis involving DNA damage repair and tumor-suppressor pathways may be implicated.
Collapse
Affiliation(s)
- Joseph G Mathew
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, ON, Canada
| | - Anita S Bowman
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Jad Saab
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Klaus J Busam
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Kishwer Nehal
- Department of Medicine, Dermatology Division, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Melissa Pulitzer
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA. https://twitter.com/MPulitzerMD
| |
Collapse
|
25
|
Spatio-temporal regulation of gene expression defines subpopulations of epidermal stem cells. Biochem Soc Trans 2020; 48:2839-2850. [PMID: 33170265 DOI: 10.1042/bst20200740] [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] [Received: 08/09/2020] [Revised: 10/14/2020] [Accepted: 10/16/2020] [Indexed: 02/06/2023]
Abstract
The search for epidermal stem cells has gained the momentum as they possess unique biological characteristics and a potential in regeneration therapies. Several transcription factors and miRNAs have been identified as epidermal stem cell markers. However, the separation of epidermal stem cells from their progeny remains challenging. The introduction of single-cell transcriptomics pointed to the high degree of heterogeneity in epidermal stem cells imbedded within subpopulations of keratinocytes. Pseudotime inference, RNA velocity, and cellular entropy further enhanced our knowledge of stem cells, allowing for the discovery of the epidermal stem cell plasticity. We explore the main findings that lead to the discovery of the plastic trait within the epidermal stem cells and the implications of cell plasticity in regenerative medicine.
Collapse
|
26
|
Wells KL, Miller CN, Gschwind AR, Wei W, Phipps JD, Anderson MS, Steinmetz LM. Combined transient ablation and single-cell RNA-sequencing reveals the development of medullary thymic epithelial cells. eLife 2020; 9:60188. [PMID: 33226342 PMCID: PMC7771965 DOI: 10.7554/elife.60188] [Citation(s) in RCA: 59] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Accepted: 11/21/2020] [Indexed: 12/14/2022] Open
Abstract
Medullary thymic epithelial cells (mTECs) play a critical role in central immune tolerance by mediating negative selection of autoreactive T cells through the collective expression of the peripheral self-antigen compartment, including tissue-specific antigens (TSAs). Recent work has shown that gene-expression patterns within the mTEC compartment are heterogenous and include multiple differentiated cell states. To further define mTEC development and medullary epithelial lineage relationships, we combined lineage tracing and recovery from transient in vivo mTEC ablation with single-cell RNA-sequencing in Mus musculus. The combination of bioinformatic and experimental approaches revealed a non-stem transit-amplifying population of cycling mTECs that preceded Aire expression. We propose a branching model of mTEC development wherein a heterogeneous pool of transit-amplifying cells gives rise to Aire- and Ccl21a-expressing mTEC subsets. We further use experimental techniques to show that within the Aire-expressing developmental branch, TSA expression peaked as Aire expression decreased, implying Aire expression must be established before TSA expression can occur. Collectively, these data provide a roadmap of mTEC development and demonstrate the power of combinatorial approaches leveraging both in vivo models and high-dimensional datasets. Specialized cells in the immune system known as T cells protect the body from infection by destroying disease-causing microbes, such as bacteria or viruses. T cells use proteins on their surface called receptors to stick to infectious microbes and remove them from the body. Some newly developed T-cells, however, contain receptors that recognize and bind to cells that belong in the body. If these faulty T cells are released, they can attack healthy tissues and cause an autoimmune disease. After a new T cell is developed, it gets carried to a gland in the chest known as the thymus. Cells in the thymus called mTECs screen T cells for receptors that may bind to the body’s tissues. mTECs do this by presenting T cells with proteins that are commonly found on the surface of healthy cells in the body. If a T cell recognizes any of these ‘tissue specific proteins’, it is destroyed or given a new role in the body. Some faulty T cells, however, still manage to evade detection. One way to uncover why this might happen is to investigate how mTECs develop. Previous work showed that mTECs transition through various stages before reaching their final form. However, the order in which these events occur remained unclear. To gain a better understanding of these developmental steps, Wells, Miller et al. extracted mTECs from the thymus of mice and analyzed the genetic make-up of individual cells. This uncovered a missing link in mTEC development: a new type of cell that is the immediate predecessor of the final mTEC. These ‘predecessor’ cells were actively growing, highlighting that mTECs can be constantly generated in the body. By probing the genes that generate tissue-specific proteins in mTECs, Wells, Miller et al. revealed that these proteins were only produced for short periods and in the late stages of mTEC development. These findings contribute to our understanding of how mTECs develop to screen T cells. Mapping these developmental stages will make it easier to identify when faulty T cells are able to evade mTECs. This will lead to earlier detection of autoimmune diseases which could result in better treatments.
Collapse
Affiliation(s)
- Kristen L Wells
- Department of Genetics, Stanford University School of Medicine, Stanford, United States
| | - Corey N Miller
- Diabetes Center, University of California, San Francisco, San Francisco, United States.,Department of Medicine, University of California San Francisco, San Francisco, United States
| | - Andreas R Gschwind
- Department of Genetics, Stanford University School of Medicine, Stanford, United States
| | - Wu Wei
- Stanford Genome Technology Center, Stanford University, Palo Alto, United States
| | - Jonah D Phipps
- Diabetes Center, University of California, San Francisco, San Francisco, United States.,Department of Medicine, University of California San Francisco, San Francisco, United States
| | - Mark S Anderson
- Diabetes Center, University of California, San Francisco, San Francisco, United States.,Department of Medicine, University of California San Francisco, San Francisco, United States
| | - Lars M Steinmetz
- Department of Genetics, Stanford University School of Medicine, Stanford, United States.,Stanford Genome Technology Center, Stanford University, Palo Alto, United States.,Genome Biology Unit, European Molecular Biology Laboratory (EMBL), Heidelberg, Germany
| |
Collapse
|
27
|
Fontana E, Nyamundanda G, Cunningham D, Tu D, Cheang MC, Jonker DJ, Siu LL, Sclafani F, Eason K, Ragulan C, Bali MA, Hulkki-Wilson S, Loree JM, Waring PM, Giordano M, Lawrence P, Rodrigues DN, Begum R, Shapiro JD, Price TJ, Cremolini C, Starling N, Pietrantonio F, Trusolino L, O’Callaghan CJ, Sadanandam A. Intratumoral Transcriptome Heterogeneity Is Associated With Patient Prognosis and Sidedness in Patients With Colorectal Cancer Treated With Anti-EGFR Therapy From the CO.20 Trial. JCO Precis Oncol 2020; 4:PO.20.00050. [PMID: 33015526 PMCID: PMC7529528 DOI: 10.1200/po.20.00050] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/06/2020] [Indexed: 01/03/2023] Open
Abstract
PURPOSE Metastatic colorectal cancers (mCRCs) assigned to the transit-amplifying (TA) CRCAssigner subtype are more sensitive to anti-epidermal growth factor receptor (EGFR) therapy. We evaluated the association between the intratumoral presence of TA signature (TA-high/TA-low, dubbed as TA-ness classification) and outcomes in CRCs treated with anti-EGFR therapy. PATIENTS AND METHODS The TA-ness classes were defined in a discovery cohort (n = 84) and independently validated in a clinical trial (CO.20; cetuximab monotherapy arm; n = 121) and other samples using an established NanoString-based gene expression assay. Progression-free survival (PFS), overall survival (OS), and disease control rate (DCR) according to TA-ness classification were assessed by univariate and multivariate analyses. RESULTS The TA-ness was measured in 772 samples from 712 patients. Patients (treated with anti-EGFR therapy) with TA-high tumors had significantly longer PFS (discovery hazard ratio [HR], 0.40; 95% CI, 0.25 to 0.64; P < .001; validation HR, 0.65; 95% CI, 0.45 to 0.93; P = .018), longer OS (discovery HR, 0.48; 95% CI, 0.29 to 0.78; P = .003; validation HR, 0.67; 95% CI, 0.46 to 0.98; P = .04), and higher DCR (discovery odds ratio [OR]; 14.8; 95% CI, 4.30 to 59.54; P < .001; validation OR, 4.35; 95% CI, 2.00 to 9.09; P < .001). TA-ness classification and its association with anti-EGFR therapy outcomes were further confirmed using publicly available data (n = 80) from metastatic samples (PFS P < .001) and patient-derived xenografts (P = .042). In an exploratory analysis of 55 patients with RAS/BRAF wild-type and left-sided tumors, TA-high class was significantly associated with longer PFS and trend toward higher response rate (PFS HR, 0.53; 95% CI, 0.28 to 1.00; P = .049; OR, 5.88; 95% CI, 0.71 to 4.55; P = .09; response rate 33% in TA-high and 7.7% in TA-low). CONCLUSION TA-ness classification is associated with prognosis in patients with mCRC treated with anti-EGFR therapy and may further help understanding the value of sidedness in patients with RAS/BRAF wild-type tumors.
Collapse
Affiliation(s)
- Elisa Fontana
- Division of Molecular Pathology, The Institute of Cancer Research, London, United Kingdom
- The Royal Marsden Hospital, London, United Kingdom
| | - Gift Nyamundanda
- Division of Molecular Pathology, The Institute of Cancer Research, London, United Kingdom
- The Royal Marsden Hospital, London, United Kingdom
| | - David Cunningham
- GI Cancer Unit, The Royal Marsden Hospital, London, United Kingdom
| | - Dongsheng Tu
- Canadian Clinical Trial Group, Kingston, Ontario, Canada
| | - Maggie C.U. Cheang
- Clinical Trials and Statistics Unit, The Institute of Cancer Research, London, United Kingdom
- Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London, United Kingdom
| | | | - Lillian L. Siu
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Francesco Sclafani
- GI Cancer Unit, The Royal Marsden Hospital, London, United Kingdom
- GI Cancer Unit, Institut Jules Bordet, Brussels, Belgium
| | - Katherine Eason
- Division of Molecular Pathology, The Institute of Cancer Research, London, United Kingdom
| | - Chanthirika Ragulan
- Division of Molecular Pathology, The Institute of Cancer Research, London, United Kingdom
- The Royal Marsden Hospital, London, United Kingdom
| | - Maria Antonietta Bali
- Radiology Department, The Royal Marsden Hospital, London, United Kingdom
- Radiology Department, Jules Bordet, Brussels, Belgium
| | - Sanna Hulkki-Wilson
- Division of Molecular Pathology, The Institute of Cancer Research, London, United Kingdom
| | | | - Paul M. Waring
- Department of Pathology, University of Melbourne, Melbourne, Victoria, Australia
| | - Mirella Giordano
- Department of Surgical, Medical, Molecular Pathology, and Critical Area, University of Pisa, Pisa, Italy
| | - Patrick Lawrence
- Division of Molecular Pathology, The Institute of Cancer Research, London, United Kingdom
- The Royal Marsden Hospital, London, United Kingdom
| | | | - Ruwaida Begum
- GI Cancer Unit, The Royal Marsden Hospital, London, United Kingdom
| | - Jeremy D. Shapiro
- Cabrini Health, Department of Medical Oncology, Malvern, Victoria, Australia
| | | | - Chiara Cremolini
- Medical Oncology Unit, Azienda Ospedaliero‐Universitaria Pisana, Pisa, Italy
- Department of Translational Research and New Technologies in Medicine, University of Pisa, Pisa, Italy
| | - Naureen Starling
- GI Cancer Unit, The Royal Marsden Hospital, London, United Kingdom
| | - Filippo Pietrantonio
- Medical Oncology Department, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico Istituto Nazionale dei Tumori, Milan, Italy
- Oncology and Hemato-Oncology Department, Milan University, Milan, Italy
| | - Livio Trusolino
- Department of Oncology, University of Torino Medical School, Candiolo, Torino, Italy
- Translational Cancer Medicine, Candiolo Cancer Institute, Istituto di Ricovero e Cura a Carattere Scientifico, Candiolo, Torino, Italy
| | | | - Anguraj Sadanandam
- Division of Molecular Pathology, The Institute of Cancer Research, London, United Kingdom
- The Royal Marsden Hospital, London, United Kingdom
| |
Collapse
|
28
|
Rangel-Huerta E, Guzman A, Maldonado E. The dynamics of epidermal stratification during post-larval development in zebrafish. Dev Dyn 2020; 250:175-190. [PMID: 32877571 DOI: 10.1002/dvdy.249] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Revised: 08/08/2020] [Accepted: 08/22/2020] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND The epidermis, as a defensive barrier, is a consistent trait throughout animal evolution. During post-larval development, the zebrafish epidermis thickens by stratification or addition of new cell layers. Epidermal basal stem cells, expressing the transcription factor p63, are known to be involved in this process. Zebrafish post-larval epidermal stratification is a tractable system to study how stem cells participate in organ growth. METHODS We used immunohistochemistry, in combination with EdU cell proliferation detection, to study zebrafish epidermal stratification. For this procedure, we selected a window of post-larval stages (5-8 mm of standard length or SL, which normalizes age by size). Simultaneously, we used markers for asymmetric cell division and the Notch signaling pathway. RESULTS We found that epidermal stratification is the consequence of several events, including changes in cell shape, active cell proliferation and asymmetrical cell divisions. We identified a subset of highly proliferative epidermal cells with reduced levels of p63, which differed from the basal stem cells with high levels of p63. Additionally, we described different mechanisms that participate in the stratification process, including the phosphorylation of p63, asymmetric cell division regulated by the Par3 and LGN proteins, and expression of Notch genes.
Collapse
Affiliation(s)
- Emma Rangel-Huerta
- EvoDevo Research Group, Unidad de Sistemas Arrecifales, Instituto de Ciencias del Mar y Limnología, Universidad Nacional Autónoma de México (UNAM), Puerto Morelos, Quintana Roo, Mexico.,Posgrado en Ciencias Biomédicas, Universidad Nacional Autónoma de México, UNAM, Puerto Morelos, Quintana Roo, Mexico
| | - Aida Guzman
- EvoDevo Research Group, Unidad de Sistemas Arrecifales, Instituto de Ciencias del Mar y Limnología, Universidad Nacional Autónoma de México (UNAM), Puerto Morelos, Quintana Roo, Mexico.,Estudio Técnico Especializado en Histopatología, Escuela Nacional Preparatoria, ENP, Universidad Nacional Autónoma de México, UNAM, Ciudad de México, Mexico
| | - Ernesto Maldonado
- EvoDevo Research Group, Unidad de Sistemas Arrecifales, Instituto de Ciencias del Mar y Limnología, Universidad Nacional Autónoma de México (UNAM), Puerto Morelos, Quintana Roo, Mexico
| |
Collapse
|
29
|
Gordon K. Recent Advances in the Genetic, Anatomical, and Environmental Regulation of the C. elegans Germ Line Progenitor Zone. J Dev Biol 2020; 8:E14. [PMID: 32707774 PMCID: PMC7559772 DOI: 10.3390/jdb8030014] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2020] [Revised: 07/13/2020] [Accepted: 07/15/2020] [Indexed: 12/16/2022] Open
Abstract
The C. elegans germ line and its gonadal support cells are well studied from a developmental genetics standpoint and have revealed many foundational principles of stem cell niche biology. Among these are the observations that a niche-like cell supports a self-renewing stem cell population with multipotential, differentiating daughter cells. While genetic features that distinguish stem-like cells from their differentiating progeny have been defined, the mechanisms that structure these populations in the germ line have yet to be explained. The spatial restriction of Notch activation has emerged as an important genetic principle acting in the distal germ line. Synthesizing recent findings, I present a model in which the germ stem cell population of the C. elegans adult hermaphrodite can be recognized as two distinct anatomical and genetic populations. This review describes the recent progress that has been made in characterizing the undifferentiated germ cells and gonad anatomy, and presents open questions in the field and new directions for research to pursue.
Collapse
Affiliation(s)
- Kacy Gordon
- Department of Biology, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| |
Collapse
|
30
|
Gadre P, Chatterjee S, Varshney B, Ray K. Cyclin E and Cdk1 regulate the termination of germline transit-amplification process in Drosophila testis. Cell Cycle 2020; 19:1786-1803. [PMID: 32573329 DOI: 10.1080/15384101.2020.1780381] [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: 12/17/2022] Open
Abstract
An extension of the G1 is correlated with stem cell differentiation. The role of cell cycle regulation during the subsequent transit amplification (TA) divisions is, however, unclear. Here, we report for the first time that in the Drosophila male germline lineage, the transit amplification divisions accelerate after the second TA division. The cell cycle phases, marked by Cyclin E and Cyclin B, are progressively altered during the TA. Antagonistic functions of the bag-of-marbles and the Transforming-Growth-Factor-β signaling regulate the cell division rates after the second TA division and the extent of the Cyclin E phase during the fourth TA division. Furthermore, loss of Cyclin E during the fourth TA cycle retards the cell division and induces premature meiosis in some cases. A similar reduction of Cdk1 activity during this stage arrests the penultimate division and subsequent differentiation, whereas enhancement of the Cdk1 activity prolongs the TA by one extra round. Altogether, the results suggest that modification of the cell cycle structure and the rates of cell division after the second TA division determine the extent of amplification. Also, the regulation of the Cyclin E and CDK1 functions during the penultimate TA division determines the induction of meiosis and subsequent differentiation.
Collapse
Affiliation(s)
- Purna Gadre
- Department of Biological Sciences, Tata Institute of Fundamental Research , Mumbai, India
| | - Shambhabi Chatterjee
- Department of Biological Sciences, Tata Institute of Fundamental Research , Mumbai, India
| | - Bhavna Varshney
- Department of Biological Sciences, Tata Institute of Fundamental Research , Mumbai, India
| | - Krishanu Ray
- Department of Biological Sciences, Tata Institute of Fundamental Research , Mumbai, India
| |
Collapse
|
31
|
Singhal R, Shah YM. Oxygen battle in the gut: Hypoxia and hypoxia-inducible factors in metabolic and inflammatory responses in the intestine. J Biol Chem 2020; 295:10493-10505. [PMID: 32503843 DOI: 10.1074/jbc.rev120.011188] [Citation(s) in RCA: 180] [Impact Index Per Article: 45.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Revised: 06/04/2020] [Indexed: 12/13/2022] Open
Abstract
The gastrointestinal tract is a highly proliferative and regenerative tissue. The intestine also harbors a large and diverse microbial population collectively called the gut microbiome (microbiota). The microbiome-intestine cross-talk includes a dynamic exchange of gaseous signaling mediators generated by bacterial and intestinal metabolisms. Moreover, the microbiome initiates and maintains the hypoxic environment of the intestine that is critical for nutrient absorption, intestinal barrier function, and innate and adaptive immune responses in the mucosal cells of the intestine. The response to hypoxia is mediated by hypoxia-inducible factors (HIFs). In hypoxic conditions, the HIF activation regulates the expression of a cohort of genes that promote adaptation to hypoxia. Physiologically, HIF-dependent genes contribute to the aforementioned maintenance of epithelial barrier function, nutrient absorption, and immune regulation. However, chronic HIF activation exacerbates disease conditions, leading to intestinal injury, inflammation, and colorectal cancer. In this review, we aim to outline the major roles of physiological and pathological hypoxic conditions in the maintenance of intestinal homeostasis and in the onset and progression of disease with a major focus on understanding the complex pathophysiology of the intestine.
Collapse
Affiliation(s)
- Rashi Singhal
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Yatrik M Shah
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, Michigan, USA .,Division of Gastroenterology, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, Michigan, USA.,Rogel Cancer Center, University of Michigan Medical School, Ann Arbor, Michigan, USA
| |
Collapse
|
32
|
Xie L, Hu WY, Hu DP, Shi G, Li Y, Yang J, Prins GS. Effects of Inorganic Arsenic on Human Prostate Stem-Progenitor Cell Transformation, Autophagic Flux Blockade, and NRF2 Pathway Activation. ENVIRONMENTAL HEALTH PERSPECTIVES 2020; 128:67008. [PMID: 32525701 PMCID: PMC7289393 DOI: 10.1289/ehp6471] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Revised: 03/18/2020] [Accepted: 05/06/2020] [Indexed: 05/26/2023]
Abstract
BACKGROUND Inorganic arsenic (iAs) is an environmental toxicant associated with an increased risk of prostate cancer in chronically exposed populations worldwide. However, the biological mechanisms underlying iAs-induced prostate carcinogenesis remain unclear. OBJECTIVES We studied how iAs affects normal human prostate stem-progenitor cells (PrSPCs) and drives transformation and interrogated the molecular mechanisms involved. METHODS PrSPCs were enriched by spheroid culture from normal human primary or immortalized prostate epithelial cells, and their differentiation capability was evaluated by organoid culture. Microarray analysis was conducted to identify iAs-dysregulated genes, and lentiviral infection was used for stable manipulation of identified genes. Soft agar colony growth assays were applied to examine iAs-induced transformation. For in vivo study, PrSPCs mixed with rat urogenital sinus mesenchyme were grafted under the renal capsule of nude mice to generate prostatelike tissues, and mice were exposed to 5 ppm (∼65μM) iAs in drinking water for 3 months. RESULTS Low-dose iAs (1μM) disturbed PrSPC homeostasis in vitro, leading to increased self-renewal and suppressed differentiation. Transcriptomic analysis indicated that iAs activated oncogenic pathways in PrSPCs, including the KEAP1-NRF2 pathway. Further, iAs-exposed proliferative progenitor cells exhibited NRF2 pathway activation that was sustained in their progeny cells. Knockdown of NRF2 inhibited spheroid formation by driving PrSPC differentiation, whereas its activation enhanced spheroid growth. Importantly, iAs-induced transformation was suppressed by NRF2 knockdown. Mechanistically, iAs suppressed Vacuolar ATPase subunit VMA5 expression, impairing lysosome acidification and inhibiting autophagic protein degradation including p62, which further activated NRF2. In vivo, chronic iAs exposure activated NRF2 in both epithelial and stroma cells of chimeric human prostate grafts and induced premalignant events. CONCLUSIONS Low-dose iAs increased self-renewal and decreased differentiation of human PrSPCs by activating the p62-NRF2 axis, resulting in epithelial cell transformation. NRF2 is activated by iAs through specific autophagic flux blockade in progenitor cells, which may have potential therapeutic implications. https://doi.org/10.1289/EHP6471.
Collapse
Affiliation(s)
- Lishi Xie
- Department of Urology, College of Medicine, University of Illinois at Chicago, Chicago, Illinois, USA
- Chicago Center for Health and Environment, Chicago, Illinois, USA
| | - Wen-Yang Hu
- Department of Urology, College of Medicine, University of Illinois at Chicago, Chicago, Illinois, USA
- Chicago Center for Health and Environment, Chicago, Illinois, USA
| | - Dan-Ping Hu
- Department of Urology, College of Medicine, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Guangbin Shi
- Division of Cardiothoracic Surgery, The Warren Alpert Medical School of Brown University, Rhode Island Hospital, Providence, Rhode Island, USA
| | - Ye Li
- Department of Urology, College of Medicine, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Jianfu Yang
- Department of Urology, The Third Xiangya Hospital of Central South University, Changsha, Hunan, People’s Republic of China
| | - Gail S. Prins
- Department of Urology, College of Medicine, University of Illinois at Chicago, Chicago, Illinois, USA
- Chicago Center for Health and Environment, Chicago, Illinois, USA
- Departments of Physiology & Biophysics and Pathology, College of Medicine; Division of Epidemiology & Biostatistics, School of Public Health, University of Illinois at Chicago, Chicago, Illinois, USA
- University of Illinois Cancer Center, Chicago, Illinois, USA
| |
Collapse
|
33
|
Deniz AAH, Abdik EA, Abdik H, Aydın S, Şahin F, Taşlı PN. Zooming in across the Skin: A Macro-to-Molecular Panorama. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1247:157-200. [DOI: 10.1007/5584_2019_442] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
34
|
Bandopadhayay P, Piccioni F, O'Rourke R, Ho P, Gonzalez EM, Buchan G, Qian K, Gionet G, Girard E, Coxon M, Rees MG, Brenan L, Dubois F, Shapira O, Greenwald NF, Pages M, Balboni Iniguez A, Paolella BR, Meng A, Sinai C, Roti G, Dharia NV, Creech A, Tanenbaum B, Khadka P, Tracy A, Tiv HL, Hong AL, Coy S, Rashid R, Lin JR, Cowley GS, Lam FC, Goodale A, Lee Y, Schoolcraft K, Vazquez F, Hahn WC, Tsherniak A, Bradner JE, Yaffe MB, Milde T, Pfister SM, Qi J, Schenone M, Carr SA, Ligon KL, Kieran MW, Santagata S, Olson JM, Gokhale PC, Jaffe JD, Root DE, Stegmaier K, Johannessen CM, Beroukhim R. Neuronal differentiation and cell-cycle programs mediate response to BET-bromodomain inhibition in MYC-driven medulloblastoma. Nat Commun 2019; 10:2400. [PMID: 31160565 PMCID: PMC6546744 DOI: 10.1038/s41467-019-10307-9] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2018] [Accepted: 04/25/2019] [Indexed: 12/26/2022] Open
Abstract
BET-bromodomain inhibition (BETi) has shown pre-clinical promise for MYC-amplified medulloblastoma. However, the mechanisms for its action, and ultimately for resistance, have not been fully defined. Here, using a combination of expression profiling, genome-scale CRISPR/Cas9-mediated loss of function and ORF/cDNA driven rescue screens, and cell-based models of spontaneous resistance, we identify bHLH/homeobox transcription factors and cell-cycle regulators as key genes mediating BETi's response and resistance. Cells that acquire drug tolerance exhibit a more neuronally differentiated cell-state and expression of lineage-specific bHLH/homeobox transcription factors. However, they do not terminally differentiate, maintain expression of CCND2, and continue to cycle through S-phase. Moreover, CDK4/CDK6 inhibition delays acquisition of resistance. Therefore, our data provide insights about the mechanisms underlying BETi effects and the appearance of resistance and support the therapeutic use of combined cell-cycle inhibitors with BETi in MYC-amplified medulloblastoma.
Collapse
Affiliation(s)
- Pratiti Bandopadhayay
- Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Boston, USA
- Broad Institute of MIT and Harvard, Cambridge, USA
- Department of Pediatrics, Harvard Medical School, Boston, USA
| | | | - Ryan O'Rourke
- Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Boston, USA
- Broad Institute of MIT and Harvard, Cambridge, USA
| | - Patricia Ho
- Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Boston, USA
- Broad Institute of MIT and Harvard, Cambridge, USA
| | - Elizabeth M Gonzalez
- Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Boston, USA
- Broad Institute of MIT and Harvard, Cambridge, USA
| | - Graham Buchan
- Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Boston, USA
- Broad Institute of MIT and Harvard, Cambridge, USA
| | - Kenin Qian
- Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Boston, USA
- Broad Institute of MIT and Harvard, Cambridge, USA
| | - Gabrielle Gionet
- Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Boston, USA
- Broad Institute of MIT and Harvard, Cambridge, USA
| | - Emily Girard
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, USA
| | - Margo Coxon
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, USA
| | | | - Lisa Brenan
- Broad Institute of MIT and Harvard, Cambridge, USA
| | - Frank Dubois
- Broad Institute of MIT and Harvard, Cambridge, USA
- Division of Cancer Biology, Dana-Farber Cancer Institute, Boston, USA
| | - Ofer Shapira
- Broad Institute of MIT and Harvard, Cambridge, USA
- Division of Cancer Biology, Dana-Farber Cancer Institute, Boston, USA
| | - Noah F Greenwald
- Broad Institute of MIT and Harvard, Cambridge, USA
- Division of Cancer Biology, Dana-Farber Cancer Institute, Boston, USA
- Department of Neurosurgery, Brigham and Women's Hospital, Boston, USA
| | - Melanie Pages
- Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Boston, USA
- Broad Institute of MIT and Harvard, Cambridge, USA
| | - Amanda Balboni Iniguez
- Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Boston, USA
- Broad Institute of MIT and Harvard, Cambridge, USA
| | - Brenton R Paolella
- Broad Institute of MIT and Harvard, Cambridge, USA
- Division of Cancer Biology, Dana-Farber Cancer Institute, Boston, USA
| | - Alice Meng
- Division of Medical Oncology, Dana-Farber Cancer Institute, Boston, USA
| | - Claire Sinai
- Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Boston, USA
- Division of Medical Oncology, Dana-Farber Cancer Institute, Boston, USA
| | - Giovanni Roti
- Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Boston, USA
- Broad Institute of MIT and Harvard, Cambridge, USA
- Department of Medicine and Surgery, Hematology and BMT, University of Parma, Parma, Italy
| | - Neekesh V Dharia
- Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Boston, USA
- Broad Institute of MIT and Harvard, Cambridge, USA
- Department of Pediatrics, Harvard Medical School, Boston, USA
| | | | | | - Prasidda Khadka
- Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Boston, USA
- Broad Institute of MIT and Harvard, Cambridge, USA
- Department of Pediatrics, Harvard Medical School, Boston, USA
| | - Adam Tracy
- Broad Institute of MIT and Harvard, Cambridge, USA
| | - Hong L Tiv
- Experimental Therapeutics Core and Belfer Center for Applied Cancer Science, Boston, USA
| | - Andrew L Hong
- Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Boston, USA
- Broad Institute of MIT and Harvard, Cambridge, USA
- Department of Pediatrics, Harvard Medical School, Boston, USA
| | - Shannon Coy
- Department of Pathology, Brigham and Women's Hospital, Boston, USA
| | - Rumana Rashid
- Department of Pathology, Brigham and Women's Hospital, Boston, USA
- Department of Biomedical Informatics, Harvard Medical School, Boston, USA
| | - Jia-Ren Lin
- Laboratory of Systems Pharmacology, Harvard Medical School, Boston, USA
- Ludwig Center for Cancer Research at Harvard, Harvard Medical School, Boston, USA
| | - Glenn S Cowley
- Broad Institute of MIT and Harvard, Cambridge, USA
- Discovery Science, Janssen Research and Development (Johnson & Johnson), Spring House, PA, USA
| | - Fred C Lam
- Koch Institute for Integrative Cancer Research, MIT, Cambridge, USA
| | - Amy Goodale
- Broad Institute of MIT and Harvard, Cambridge, USA
| | - Yenarae Lee
- Broad Institute of MIT and Harvard, Cambridge, USA
| | | | | | - William C Hahn
- Broad Institute of MIT and Harvard, Cambridge, USA
- Division of Medical Oncology, Dana-Farber Cancer Institute, Boston, USA
- Department of Medicine, Harvard Medical School, Boston, USA
| | | | - James E Bradner
- Broad Institute of MIT and Harvard, Cambridge, USA
- Division of Medical Oncology, Dana-Farber Cancer Institute, Boston, USA
- Department of Medicine, Harvard Medical School, Boston, USA
- Novartis Institutes for Biomedical Research, Basel, Switzerland
| | - Michael B Yaffe
- Broad Institute of MIT and Harvard, Cambridge, USA
- Koch Institute for Integrative Cancer Research, MIT, Cambridge, USA
| | - Till Milde
- Hopp Children's Cancer Center Heidelberg (KiTZ), Heidelberg, Germany
- CCU Pediatric Oncology, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Pediatric Oncology, Hematology, and Immunology, Center for Child and Adolescent Medicine, Heidelberg University Hospital, Heidelberg, Germany
| | - Stefan M Pfister
- Hopp Children's Cancer Center Heidelberg (KiTZ), Heidelberg, Germany
- Division of Pediatric Neuro-Oncology, German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Pediatric Hematology and Oncology, Heidelberg University Hospital, Heidelberg, Germany
| | - Jun Qi
- Division of Cancer Biology, Dana-Farber Cancer Institute, Boston, USA
| | | | | | - Keith L Ligon
- Broad Institute of MIT and Harvard, Cambridge, USA
- Department of Pathology, Brigham and Women's Hospital, Boston, USA
- Department of Medicine, Harvard Medical School, Boston, USA
- Department of Oncologic Pathology, Dana-Farber Cancer Institute, Boston, USA
- Department of Pathology, Boston Children's Hospital, Boston, USA
| | - Mark W Kieran
- Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Boston, USA
- Department of Pediatrics, Harvard Medical School, Boston, USA
| | - Sandro Santagata
- Division of Medical Oncology, Dana-Farber Cancer Institute, Boston, USA
- Department of Pathology, Brigham and Women's Hospital, Boston, USA
| | - James M Olson
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, USA
| | - Prafulla C Gokhale
- Experimental Therapeutics Core and Belfer Center for Applied Cancer Science, Boston, USA
| | | | - David E Root
- Broad Institute of MIT and Harvard, Cambridge, USA
| | - Kimberly Stegmaier
- Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Boston, USA
- Broad Institute of MIT and Harvard, Cambridge, USA
- Department of Pediatrics, Harvard Medical School, Boston, USA
| | | | - Rameen Beroukhim
- Broad Institute of MIT and Harvard, Cambridge, USA.
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, USA.
- Division of Medical Oncology, Dana-Farber Cancer Institute, Boston, USA.
- Department of Medicine, Harvard Medical School, Boston, USA.
| |
Collapse
|
35
|
Xue Y, Barker N, Hoon S, He P, Thakur T, Abdeen SR, Maruthappan P, Ghadessy FJ, Lane DP. Bortezomib Stabilizes and Activates p53 in Proliferative Compartments of Both Normal and Tumor Tissues In Vivo. Cancer Res 2019; 79:3595-3607. [DOI: 10.1158/0008-5472.can-18-3744] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2018] [Revised: 04/08/2019] [Accepted: 05/22/2019] [Indexed: 11/16/2022]
|
36
|
Kruglikov IL, Zhang Z, Scherer PE. The Role of Immature and Mature Adipocytes in Hair Cycling. Trends Endocrinol Metab 2019; 30:93-105. [PMID: 30558832 PMCID: PMC6348020 DOI: 10.1016/j.tem.2018.11.004] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2018] [Revised: 10/29/2018] [Accepted: 11/20/2018] [Indexed: 02/06/2023]
Abstract
Hair follicles (HFs) strongly interact with adipocytes within the dermal white adipose tissue (dWAT), suggesting a strong physiological dependence on the content of immature and mature adipocytes in this layer. This content is regulated by the proliferation and differentiation of adipocyte precursors, as well as by dedifferentiation of mature existing adipocytes. Spatially, long-range interactions between HFs and dWAT involve the exchange of extracellular vesicles which are differentially released by precursors, preadipocytes, and mature adipocytes. Different exogenous factors, including light irradiation, are likely to modify the release of adipocyte-derived exosomes in dWAT, which can lead to aberrations of the HF cycle. Consequently, dWAT should be considered as a potential target for the modulation of hair growth.
Collapse
Affiliation(s)
| | - Zhuzhen Zhang
- Touchstone Diabetes Center, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390-8549, USA
| | - Philipp E Scherer
- Touchstone Diabetes Center, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390-8549, USA.
| |
Collapse
|
37
|
|
38
|
Camacho Leal MDP, Costamagna A, Tassone B, Saoncella S, Simoni M, Natalini D, Dadone A, Sciortino M, Turco E, Defilippi P, Calautti E, Cabodi S. Conditional ablation of p130Cas/BCAR1 adaptor protein impairs epidermal homeostasis by altering cell adhesion and differentiation. Cell Commun Signal 2018; 16:73. [PMID: 30390666 PMCID: PMC6215608 DOI: 10.1186/s12964-018-0289-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2018] [Accepted: 10/25/2018] [Indexed: 12/20/2022] Open
Abstract
Background p130 Crk-associated substrate (p130CAS; also known as BCAR1) is a scaffold protein that modulates many essential cellular processes such as cell adhesion, proliferation, survival, cell migration, and intracellular signaling. p130Cas has been shown to be highly expressed in a variety of human cancers of epithelial origin. However, few data are available regarding the role of p130Cas during normal epithelial development and homeostasis. Methods To this end, we have generated a genetically modified mouse in which p130Cas protein was specifically ablated in the epidermal tissue. Results By using this murine model, we show that p130Cas loss results in increased cell proliferation and reduction of cell adhesion to extracellular matrix. In addition, epidermal deletion of p130Cas protein leads to premature expression of “late” epidermal differentiation markers, altered membrane E-cadherin/catenin proteins localization and aberrant tyrosine phosphorylation of E-cadherin/catenin complexes. Interestingly, these alterations in adhesive properties in absence of p130Cas correlate with abnormalities in progenitor cells balance resulting in the amplification of a more committed cell population. Conclusion Altogether, these results provide evidence that p130Cas is an important regulator of epidermal cell fate and homeostasis. Electronic supplementary material The online version of this article (10.1186/s12964-018-0289-z) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Maria Del Pilar Camacho Leal
- Department of Biotechnology and Health Science, Molecular Biotechnology Center, Università di Torino, Via Nizza 52, Torino, Italy
| | - Andrea Costamagna
- Department of Biotechnology and Health Science, Molecular Biotechnology Center, Università di Torino, Via Nizza 52, Torino, Italy
| | - Beatrice Tassone
- Department of Biotechnology and Health Science, Molecular Biotechnology Center, Università di Torino, Via Nizza 52, Torino, Italy
| | - Stefania Saoncella
- Department of Biotechnology and Health Science, Molecular Biotechnology Center, Università di Torino, Via Nizza 52, Torino, Italy
| | - Matilde Simoni
- Department of Biotechnology and Health Science, Molecular Biotechnology Center, Università di Torino, Via Nizza 52, Torino, Italy
| | - Dora Natalini
- Department of Biotechnology and Health Science, Molecular Biotechnology Center, Università di Torino, Via Nizza 52, Torino, Italy
| | - Aurora Dadone
- Department of Biotechnology and Health Science, Molecular Biotechnology Center, Università di Torino, Via Nizza 52, Torino, Italy
| | - Marianna Sciortino
- Department of Biotechnology and Health Science, Molecular Biotechnology Center, Università di Torino, Via Nizza 52, Torino, Italy
| | - Emilia Turco
- Department of Biotechnology and Health Science, Molecular Biotechnology Center, Università di Torino, Via Nizza 52, Torino, Italy
| | - Paola Defilippi
- Department of Biotechnology and Health Science, Molecular Biotechnology Center, Università di Torino, Via Nizza 52, Torino, Italy
| | - Enzo Calautti
- Department of Biotechnology and Health Science, Molecular Biotechnology Center, Università di Torino, Via Nizza 52, Torino, Italy
| | - Sara Cabodi
- Department of Biotechnology and Health Science, Molecular Biotechnology Center, Università di Torino, Via Nizza 52, Torino, Italy.
| |
Collapse
|