151
|
Lambert AW, Weinberg RA. Linking EMT programmes to normal and neoplastic epithelial stem cells. Nat Rev Cancer 2021; 21:325-338. [PMID: 33547455 DOI: 10.1038/s41568-021-00332-6] [Citation(s) in RCA: 329] [Impact Index Per Article: 82.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/06/2021] [Indexed: 02/07/2023]
Abstract
Epithelial stem cells serve critical physiological functions in the generation, maintenance and repair of diverse tissues through their ability to self-renew and spawn more specialized, differentiated cell types. In an analogous fashion, cancer stem cells have been proposed to fuel the growth, progression and recurrence of many carcinomas. Activation of an epithelial-mesenchymal transition (EMT), a latent cell-biological programme involved in development and wound healing, has been linked to the formation of both normal and neoplastic stem cells, but the mechanistic basis underlying this connection remains unclear. In this Perspective, we outline the instances where aspects of an EMT have been implicated in normal and neoplastic epithelial stem cells and consider the involvement of this programme during tissue regeneration and repair. We also discuss emerging concepts and evidence related to the heterogeneous and plastic cell states generated by EMT programmes and how these bear on our understanding of cancer stem cell biology and cancer metastasis. A more comprehensive accounting of the still-elusive links between EMT programmes and the stem cell state will surely advance our understanding of both normal stem cell biology and cancer pathogenesis.
Collapse
Affiliation(s)
| | - Robert A Weinberg
- Whitehead Institute for Biomedical Research, Cambridge, MA, USA.
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA.
- MIT Ludwig Center for Molecular Oncology, Cambridge, MA, USA.
| |
Collapse
|
152
|
Intratumor heterogeneity, microenvironment, and mechanisms of drug resistance in glioma recurrence and evolution. Front Med 2021; 15:551-561. [PMID: 33893983 DOI: 10.1007/s11684-020-0760-2] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Accepted: 02/13/2020] [Indexed: 02/07/2023]
Abstract
Glioma is the most common lethal tumor of the human brain. The median survival of patients with primary World Health Organization grade IV glioma is only 14.6 months. The World Health Organization classification of tumors of the central nervous system categorized gliomas into lower-grade gliomas and glioblastomas. Unlike primary glioblastoma that usually develop de novo in the elderly, secondary glioblastoma enriched with an isocitrate dehydrogenase mutant typically progresses from lower-grade glioma within 5-10 years from the time of diagnosis. Based on various evolutional trajectories brought on by clonal and subclonal alterations, the evolution patterns of glioma vary according to different theories. Some important features distinguish the normal brain from other tissues, e.g., the composition of the microenvironment around the tumor cells, the presence of the blood-brain barrier, and others. The underlying mechanism of glioma recurrence and evolution patterns of glioma are different from those of other types of cancer. Several studies correlated tumor recurrence with tumor heterogeneity and the immune microenvironment. However, the detailed reasons for the progression and recurrence of glioma remain controversial. In this review, we introduce the different mechanisms involved in glioma progression, including tumor heterogeneity, the tumor microenvironment and drug resistance, and their pre-clinical implements in clinical trials. This review aimed to provide new insights into further clinical strategies for the treatment of patients with recurrent and secondary glioma.
Collapse
|
153
|
Kim SG. A Cell-Based Approach to Dental Pulp Regeneration Using Mesenchymal Stem Cells: A Scoping Review. Int J Mol Sci 2021; 22:4357. [PMID: 33921924 PMCID: PMC8122243 DOI: 10.3390/ijms22094357] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 04/18/2021] [Accepted: 04/20/2021] [Indexed: 02/07/2023] Open
Abstract
Despite the recent explosion of investigations on dental pulp regeneration using various tissue engineering strategies, the translation of the findings from such studies into therapeutic applications has not been properly achieved. The purpose of this scoping review was to systematically review the efficacy of mesenchymal stem cell transplantation for dental pulp regeneration. A literature search was conducted using five electronic databases from their inception to January 2021 and supplemented by hand searches. A total of 17 studies, including two clinical trials and 15 animal studies using orthotopic pulp regeneration models, were included for the review. The risk of bias for the individual studies was assessed. This scoping review demonstrated that the regeneration of vascularized pulp-like tissue was achieved using the stem cell transplantation strategy in animal models. Autologous cell transplantation in two clinical studies also successfully regenerated vascularized vital tissue. Dental pulp stem cell subpopulations, such as mobilized dental pulp stem cells, injectable scaffolds such as atelocollagen, and a granulocyte-colony forming factor, were the most commonly used for pulp regeneration. The overall risk of bias was unclear for animal studies and was moderate or judged to raise some concerns for clinical studies. More high-quality clinical studies are needed to further determine the safety and efficacy of the stem cell transplantation strategy for dental pulp regeneration.
Collapse
Affiliation(s)
- Sahng G Kim
- Division of Endodontics, Columbia University College of Dental Medicine, New York, NY 10032, USA
| |
Collapse
|
154
|
Bonnet C, Roberts JS, Deng SX. Limbal stem cell diseases. Exp Eye Res 2021; 205:108437. [PMID: 33571530 PMCID: PMC8044031 DOI: 10.1016/j.exer.2021.108437] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Revised: 12/14/2020] [Accepted: 01/02/2021] [Indexed: 12/13/2022]
Abstract
The function of limbal stem/progenitor cells (LSCs) is critical to maintain corneal epithelial homeostasis. Many external insults and intrinsic defects can be deleterious to LSCs and their niche microenvironment, resulting in limbal stem cell dysfunction or deficiency (LSCD). Ocular comorbidities, frequent in eyes with LSCD, can exacerbate the dysfunction of residual LSCs, and limit the survival of transplanted LSCs. Clinical presentation and disease evolution vary among different etiologies of LSCD. New ocular imaging modalities and molecular markers are now available to standardize the diagnosis criteria and stage the severity of the disease. Medical therapies may be sufficient to reverse the disease if residual LSCs are present. A stepwise approach should be followed to optimize the ocular surface, eliminate the causative factors and treat comorbid conditions, before considering surgical interventions. Furthermore, surgical options are selected depending on the severity and laterality of the disease. The standardized diagnostic criteria to stage the disease is necessary to objectively evaluate and compare the efficacy of the emerging customized therapies.
Collapse
Affiliation(s)
- Clémence Bonnet
- Stein Eye Institute, Department of Ophthalmology, David Geffen School of Medicine, University of California, Los Angeles, CA, 90095, USA; Cornea Department, Paris University, Cochin Hospital, AP-HP, F-75014, Paris, France.
| | - JoAnn S Roberts
- Stein Eye Institute, Department of Ophthalmology, David Geffen School of Medicine, University of California, Los Angeles, CA, 90095, USA.
| | - Sophie X Deng
- Stein Eye Institute, Department of Ophthalmology, David Geffen School of Medicine, University of California, Los Angeles, CA, 90095, USA.
| |
Collapse
|
155
|
Chen P, Zhang F, Fan Z, Shen T, Liu B, Chen R, Qu Q, Wang J, Miao Y, Hu Z. Nanoscale microenvironment engineering for expanding human hair follicle stem cell and revealing their plasticity. J Nanobiotechnology 2021; 19:94. [PMID: 33789665 PMCID: PMC8010974 DOI: 10.1186/s12951-021-00840-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Accepted: 03/23/2021] [Indexed: 11/17/2022] Open
Abstract
Background Periodically regenerated hair follicles provide an excellent research model for studying tissue regeneration and stem cell homeostasis. Periodic activation and differentiation of hair follicle stem cells (HFSCs) fuel cyclical bouts of hair regeneration. HFSCs represent an excellent paradigm for studying tissue regeneration and somatic stem cell homeostasis. However, these crucial studies are hampered by the lack of a culture system able to stably expand human HFSCs and regulate their fate. Results Here, we use layer-by-layer (LbL) self-assembly with gelatin/alginate to construct a nanoscale biomimetic extracellular matrix (ECM) for an HFSC population. The LbL coating provides ECM and mechanical support for individual cells, which helps to maintain the CD200+α6+ HFSC population to a certain extent. Addition of key signal molecules (FGF-7 and VEGF-A) simulates the minimum essential components of the stem cell microenvironment, thereby effectively and stably expanding HFSCs and maintaining the CD200+α6+ HFSC population. Subsequently, BMP2 loaded to the nanocoated layer, as a slow-release signal molecule, activates BMP signaling to regulate HFSCs’ fate in order to obtain a purified CD200+α6+ HFSC population. Conclusion This system can minimize the microenvironment of HFSCs; thus, stably amplifying HFSCs and revealing their plasticity. Our study thus provides a new tool for studies of hair follicle reconstruction and stem cell homeostasis. ![]()
Supplementary Information The online version contains supplementary material available at 10.1186/s12951-021-00840-5.
Collapse
Affiliation(s)
- Peng Chen
- Department of Plastic and Aesthetic Surgery, Nanfang Hospital, Southern Medical University, 1838 North Guangzhou Avenue, Guangzhou, 510515, Guangdong, China
| | - Feifei Zhang
- Department of Plastic and Aesthetic Surgery, Nanfang Hospital, Southern Medical University, 1838 North Guangzhou Avenue, Guangzhou, 510515, Guangdong, China
| | - Zhexiang Fan
- Department of Plastic and Aesthetic Surgery, Nanfang Hospital, Southern Medical University, 1838 North Guangzhou Avenue, Guangzhou, 510515, Guangdong, China
| | - Tianding Shen
- Department of Plastic and Aesthetic Surgery, Nanfang Hospital, Southern Medical University, 1838 North Guangzhou Avenue, Guangzhou, 510515, Guangdong, China
| | - Bingcheng Liu
- Department of Plastic and Aesthetic Surgery, Nanfang Hospital, Southern Medical University, 1838 North Guangzhou Avenue, Guangzhou, 510515, Guangdong, China
| | - Ruosi Chen
- Department of Plastic and Aesthetic Surgery, Nanfang Hospital, Southern Medical University, 1838 North Guangzhou Avenue, Guangzhou, 510515, Guangdong, China
| | - Qian Qu
- Department of Plastic and Aesthetic Surgery, Nanfang Hospital, Southern Medical University, 1838 North Guangzhou Avenue, Guangzhou, 510515, Guangdong, China
| | - Jin Wang
- Department of Plastic and Aesthetic Surgery, Nanfang Hospital, Southern Medical University, 1838 North Guangzhou Avenue, Guangzhou, 510515, Guangdong, China.
| | - Yong Miao
- Department of Plastic and Aesthetic Surgery, Nanfang Hospital, Southern Medical University, 1838 North Guangzhou Avenue, Guangzhou, 510515, Guangdong, China.
| | - Zhiqi Hu
- Department of Plastic and Aesthetic Surgery, Nanfang Hospital, Southern Medical University, 1838 North Guangzhou Avenue, Guangzhou, 510515, Guangdong, China.
| |
Collapse
|
156
|
Oshimori N, Guo Y, Taniguchi S. An emerging role for cellular crosstalk in the cancer stem cell niche. J Pathol 2021; 254:384-394. [DOI: 10.1002/path.5655] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Revised: 02/12/2021] [Accepted: 02/23/2021] [Indexed: 12/17/2022]
Affiliation(s)
- Naoki Oshimori
- Department of Cell, Developmental and Cancer Biology Oregon Health & Science University Portland OR USA
- Department of Dermatology Oregon Health & Science University Portland OR USA
- Department of Otolaryngology – Head & Neck Surgery Oregon Health & Science University Portland OR USA
- Knight Cancer Institute Oregon Health & Science University Portland OR USA
| | - Yifei Guo
- Department of Cell, Developmental and Cancer Biology Oregon Health & Science University Portland OR USA
| | - Sachiko Taniguchi
- Department of Cell, Developmental and Cancer Biology Oregon Health & Science University Portland OR USA
| |
Collapse
|
157
|
Functional States in Tumor-Initiating Cell Differentiation in Human Colorectal Cancer. Cancers (Basel) 2021; 13:cancers13051097. [PMID: 33806447 PMCID: PMC7961698 DOI: 10.3390/cancers13051097] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Accepted: 02/28/2021] [Indexed: 12/19/2022] Open
Abstract
Simple Summary Different types of cells with tumor-initiating cell (TIC) activity contribute to colorectal cancer (CRC) progression and resistance to anti-cancer treatment. In this study, we aimed to understand whether different cell types exist within a patient-derived tumor culture, distinguishable by different patterns of their gene expression. By mRNA sequencing of patient-derived CRC cultures at the single-cell level, we defined expression programs that closely resemble differentiated cell populations of the normal intestine. Here, cell type-associated subpopulations showed differences in functional properties such as cell growth and energy metabolism. Subsequent functional analyses in vitro and in vivo demonstrated that metabolic states are linked to TIC activity in primary CRC cultures. We also show that TIC activity is dependent on oxidative phosphorylation, which may therefore represent a target for novel therapies. Abstract Intra-tumor heterogeneity of tumor-initiating cell (TIC) activity drives colorectal cancer (CRC) progression and therapy resistance. Here, we used single-cell RNA-sequencing of patient-derived CRC models to decipher distinct cell subpopulations based on their transcriptional profiles. Cell type-specific expression modules of stem-like, transit amplifying-like, and differentiated CRC cells resemble differentiation states of normal intestinal epithelial cells. Strikingly, identified subpopulations differ in proliferative activity and metabolic state. In summary, we here show at single-cell resolution that transcriptional heterogeneity identifies functional states during TIC differentiation. Furthermore, identified expression signatures are linked to patient prognosis. Targeting transcriptional states associated to cancer cell differentiation might unravel novel vulnerabilities in human CRC.
Collapse
|
158
|
Tsai SL, Noedl MT, Galloway JL. Bringing tendon biology to heel: Leveraging mechanisms of tendon development, healing, and regeneration to advance therapeutic strategies. Dev Dyn 2021; 250:393-413. [PMID: 33169466 PMCID: PMC8486356 DOI: 10.1002/dvdy.269] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Revised: 10/29/2020] [Accepted: 11/03/2020] [Indexed: 12/11/2022] Open
Abstract
Tendons are specialized matrix-rich connective tissues that transmit forces from muscle to bone and are essential for movement. As tissues that frequently transfer large mechanical loads, tendons are commonly injured in patients of all ages. Following injury, mammalian tendons heal poorly through a slow process that forms disorganized fibrotic scar tissue with inferior biomechanical function. Current treatments are limited and patients can be left with a weaker tendon that is likely to rerupture and an increased chance of developing degenerative conditions. More effective, alternative treatments are needed. However, our current understanding of tendon biology remains limited. Here, we emphasize why expanding our knowledge of tendon development, healing, and regeneration is imperative for advancing tendon regenerative medicine. We provide a comprehensive review of the current mechanisms governing tendon development and healing and further highlight recent work in regenerative tendon models including the neonatal mouse and zebrafish. Importantly, we discuss how present and future discoveries can be applied to both augment current treatments and design novel strategies to treat tendon injuries.
Collapse
Affiliation(s)
- Stephanie L. Tsai
- Center for Regenerative Medicine, Department of Orthopedic Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114
- Harvard Stem Cell Institute, Cambridge, MA 02138
| | - Marie-Therese Noedl
- Center for Regenerative Medicine, Department of Orthopedic Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114
- Harvard Stem Cell Institute, Cambridge, MA 02138
| | - Jenna L. Galloway
- Center for Regenerative Medicine, Department of Orthopedic Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114
- Harvard Stem Cell Institute, Cambridge, MA 02138
| |
Collapse
|
159
|
Ishikawa T, Akiyama N, Akiyama T. In Pursuit of Adult Progenitors of Thymic Epithelial Cells. Front Immunol 2021; 12:621824. [PMID: 33717123 PMCID: PMC7946825 DOI: 10.3389/fimmu.2021.621824] [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: 10/27/2020] [Accepted: 02/08/2021] [Indexed: 12/25/2022] Open
Abstract
Peripheral T cells capable of discriminating between self and non-self antigens are major components of a robust adaptive immune system. The development of self-tolerant T cells is orchestrated by thymic epithelial cells (TECs), which are localized in the thymic cortex (cortical TECs, cTECs) and medulla (medullary TECs, mTECs). cTECs and mTECs are essential for differentiation, proliferation, and positive and negative selection of thymocytes. Recent advances in single-cell RNA-sequencing technology have revealed a previously unknown degree of TEC heterogeneity, but we still lack a clear picture of the identity of TEC progenitors in the adult thymus. In this review, we describe both earlier and recent findings that shed light on features of these elusive adult progenitors in the context of tissue homeostasis, as well as recovery from stress-induced thymic atrophy.
Collapse
Affiliation(s)
- Tatsuya Ishikawa
- Laboratory of Immune Homeostasis, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan.,Graduate School of Medical Life Science, Yokohama City University, Yokohama, Japan
| | - Nobuko Akiyama
- Laboratory for Immunogenetics, RIKEN Center of Integrative Medical Sciences, Yokohama, Japan
| | - Taishin Akiyama
- Laboratory of Immune Homeostasis, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan.,Graduate School of Medical Life Science, Yokohama City University, Yokohama, Japan
| |
Collapse
|
160
|
Tan SH, Ngo ZH, Leavesley D, Liang K. Recent Advances in the Design of Three-Dimensional and Bioprinted Scaffolds for Full-Thickness Wound Healing. TISSUE ENGINEERING PART B-REVIEWS 2021; 28:160-181. [PMID: 33446047 DOI: 10.1089/ten.teb.2020.0339] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Three-dimensional (3D) printed scaffolds have recently emerged as an innovative treatment option for patients with critical-sized skin wounds. Current approaches to managing life-threatening wounds include skin grafting and application of commercially sourced skin substitutes. However, these approaches are not without several challenges. Limited donor tissue and donor site morbidity remain a concern for tissue grafting, while engineered skin substitutes fail to fully recapitulate the complex native environment required for wound healing. The implementation of 3D printed dermal scaffolds offers a potential solution for these shortcomings. Spatial control over scaffold structure, the ability to incorporate multiple materials and bioactive ingredients, enables the creation of conditions specifically optimized for wound healing. Three-dimensional bioprinting, a subset of 3D printing, allows for the replacement of lost cell populations and secreted active compounds that contribute to tissue repair and recovery. The replacement of damaged and lost cells delivers beneficial effects directly, or synergistically, supporting injured tissue to recover its native state. Despite encouraging results, the promise of 3D printed scaffolds has yet to be realized. Further improvements to current material formulations and scaffold designs are required to achieve the goal of clinical adoption. Herein, we provide an overview of 3D printing techniques and discuss several strategies for healing of full-thickness wounds by using 3D printed acellular scaffolds or bioprinted cellular scaffolds, aimed at translating this technology to the clinical management of skin lesions. We identify the challenges associated with designing and optimizing printed tissue replacements, and discuss the future perspectives of this emerging option for managing patients who present with critical-sized life-threatening cutaneous wounds.
Collapse
Affiliation(s)
- Shi Hua Tan
- Skin Research Institute of Singapore (SRIS), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Zong Heng Ngo
- Skin Research Institute of Singapore (SRIS), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - David Leavesley
- Skin Research Institute of Singapore (SRIS), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Kun Liang
- Skin Research Institute of Singapore (SRIS), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| |
Collapse
|
161
|
Cheah E, Wu Z, Thakur SS, O'Carroll SJ, Svirskis D. Externally triggered release of growth factors - A tissue regeneration approach. J Control Release 2021; 332:74-95. [PMID: 33600882 DOI: 10.1016/j.jconrel.2021.02.015] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2020] [Revised: 02/10/2021] [Accepted: 02/11/2021] [Indexed: 12/22/2022]
Abstract
Tissue regeneration aims to achieve functional restoration following injury by creating an environment to enable the body to self-repair. Strategies for regeneration rely on the introduction of biomaterial scaffolding, cells and bioactive molecules into the body, at or near the injury site. Of these bioactive molecules, growth factors (GFs) play a pivotal role in directing regenerative pathways for many cell populations. However, the therapeutic use of GFs has been limited by the complexity of biological injury and repair, and the properties of the GFs themselves, including their short half-life, poor tissue penetration, and off-target side effects. Externally triggered delivery systems have the potential to facilitate the delivery of GFs into the target tissues with considerations of the timing, sequence, amount, and location of GF presentation. This review briefly discusses the challenges facing the therapeutic use of GFs, then, we discuss approaches to externally trigger GF release from delivery systems categorised by stimulation type; ultrasound, temperature, light, magnetic fields and electric fields. Overall, while the use of GFs for tissue regeneration is still in its infancy, externally controlled GF delivery technologies have the potential to achieve robust and effective solutions to present GFs to injured tissues. Future technological developments must occur in conjunction with a comprehensive understanding of the biology at the injury site to ensure translation of promising technologies into real world benefit.
Collapse
Affiliation(s)
- Ernest Cheah
- School of Pharmacy, Faculty of Medical and Health Sciences, University of Auckland, Private Bag 92019, Auckland, New Zealand
| | - Zimei Wu
- School of Pharmacy, Faculty of Medical and Health Sciences, University of Auckland, Private Bag 92019, Auckland, New Zealand
| | - Sachin S Thakur
- School of Pharmacy, Faculty of Medical and Health Sciences, University of Auckland, Private Bag 92019, Auckland, New Zealand
| | - Simon J O'Carroll
- Department of Anatomy and Medical Imaging, School of Medical Sciences, Faculty of Medical and Health Sciences, University of Auckland, Private Bag 92019, Auckland, New Zealand
| | - Darren Svirskis
- School of Pharmacy, Faculty of Medical and Health Sciences, University of Auckland, Private Bag 92019, Auckland, New Zealand.
| |
Collapse
|
162
|
Zhou HM, Zhang JG, Zhang X, Li Q. Targeting cancer stem cells for reversing therapy resistance: mechanism, signaling, and prospective agents. Signal Transduct Target Ther 2021; 6:62. [PMID: 33589595 PMCID: PMC7884707 DOI: 10.1038/s41392-020-00430-1] [Citation(s) in RCA: 247] [Impact Index Per Article: 61.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Revised: 07/26/2020] [Accepted: 10/08/2020] [Indexed: 02/06/2023] Open
Abstract
Cancer stem cells (CSCs) show a self-renewal capacity and differentiation potential that contribute to tumor progression and therapy resistance. However, the underlying processes are still unclear. Elucidation of the key hallmarks and resistance mechanisms of CSCs may help improve patient outcomes and reduce relapse by altering therapeutic regimens. Here, we reviewed the identification of CSCs, the intrinsic and extrinsic mechanisms of therapy resistance in CSCs, the signaling pathways of CSCs that mediate treatment failure, and potential CSC-targeting agents in various tumors from the clinical perspective. Targeting the mechanisms and pathways described here might contribute to further drug discovery and therapy.
Collapse
Affiliation(s)
- He-Ming Zhou
- Department of Clinical Pharmacy, Shanghai General Hospital, Shanghai Jiao Tong University School of medicine, No.100 Haining Road, 200080, Shanghai, People's Republic of China
| | - Ji-Gang Zhang
- Department of Clinical Pharmacy, Shanghai General Hospital, Shanghai Jiao Tong University School of medicine, No.100 Haining Road, 200080, Shanghai, People's Republic of China
| | - Xue Zhang
- Department of Clinical Pharmacy, Shanghai General Hospital, Shanghai Jiao Tong University School of medicine, No.100 Haining Road, 200080, Shanghai, People's Republic of China
| | - Qin Li
- Department of Clinical Pharmacy, Shanghai General Hospital, Shanghai Jiao Tong University School of medicine, No.100 Haining Road, 200080, Shanghai, People's Republic of China.
| |
Collapse
|
163
|
Boecker W, Reusch M, Mielke V, Reusch U, Hallermann C, Loening T, Tiemann M, Buchwalow I. Twenty-Eight Cases of Extraocular Sebaceous Carcinoma: A Correlative Clinicopathological and Immunohistochemical Analysis of Extraocular Sebaceous Carcinomas and Benign Sebaceous Gland Tumors. Am J Dermatopathol 2021; 43:93-102. [PMID: 32568835 DOI: 10.1097/dad.0000000000001667] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
ABSTRACT Extraocular sebaceous carcinoma (ESC) is a rare appendiceal skin tumor. In contrast to ocular sebaceous carcinoma, information about the exact cellular architecture of these lesions is scarce and the histogenesis of ESC is unknown. Here, we extend our previous study and investigate 28 extraocular carcinomas in comparison to 54 benign sebaceous tumors and 8 cases of normal sebaceous glands using a broad spectrum of antibodies against p63, several keratins, adipophilin, EMA, Ki67, androgen receptor, and mismatch repair proteins. This observational study demonstrates that p63- and K5/14-positive basaloid cells are key cells in normal sebaceous gland and in all sebaceous tumors and that these basaloid cells give rise to EMA+, adipophilin+ sebocytes, and K5/14+, K7±, K10± ductal structures. Finally, about half of ESC is associated with superficial in situ neoplasia, which provides evidence that at least part of these carcinomas arises from flat superficial in situ carcinoma. In contrast to the normal sebaceous gland, about half of all sebaceous tumors lack keratin K7. MMR protein IHC-profiles role will be discussed.
Collapse
Affiliation(s)
- Werner Boecker
- Gerhard-Domagk-Institute of Pathology, University of Muenster, Münster, Germany
- Dermatopathology Laboratory, Hamburg, Germany
- Gerhard-Seifert Referenzzentrum, Hamburg, Germany; and
| | | | | | | | | | | | | | - Igor Buchwalow
- Gerhard-Domagk-Institute of Pathology, University of Muenster, Münster, Germany
- Institute for Hematopathology, Hamburg, Germany
| |
Collapse
|
164
|
Yang BA, Westerhof TM, Sabin K, Merajver SD, Aguilar CA. Engineered Tools to Study Intercellular Communication. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2021; 8:2002825. [PMID: 33552865 PMCID: PMC7856891 DOI: 10.1002/advs.202002825] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/25/2020] [Revised: 10/10/2020] [Indexed: 05/08/2023]
Abstract
All multicellular organisms rely on intercellular communication networks to coordinate physiological functions. As members of a dynamic social network, each cell receives, processes, and redistributes biological information to define and maintain tissue homeostasis. Uncovering the molecular programs underlying these processes is critical for prevention of disease and aging and development of therapeutics. The study of intercellular communication requires techniques that reduce the scale and complexity of in vivo biological networks while resolving the molecular heterogeneity in "omic" layers that contribute to cell state and function. Recent advances in microengineering and high-throughput genomics offer unprecedented spatiotemporal control over cellular interactions and the ability to study intercellular communication in a high-throughput and mechanistic manner. Herein, this review discusses how salient engineered approaches and sequencing techniques can be applied to understand collective cell behavior and tissue functions.
Collapse
Affiliation(s)
- Benjamin A. Yang
- Department of Biomedical Engineering and Biointerfaces Institute2800 Plymouth Road, North Campus Research ComplexAnn ArborMIA10‐183USA
| | - Trisha M. Westerhof
- Department of Biomedical Engineering and Biointerfaces Institute2800 Plymouth Road, North Campus Research ComplexAnn ArborMIA10‐183USA
- Department of Internal MedicineDivision of Hematology/Oncology and Rogel Cancer Center1500 East Medical Center Drive, Rogel Cancer CenterAnn ArborMI7314USA
| | - Kaitlyn Sabin
- Department of Biomedical Engineering and Biointerfaces Institute2800 Plymouth Road, North Campus Research ComplexAnn ArborMIA10‐183USA
| | - Sofia D. Merajver
- Department of Internal MedicineDivision of Hematology/Oncology and Rogel Cancer Center1500 East Medical Center Drive, Rogel Cancer CenterAnn ArborMI7314USA
| | - Carlos A. Aguilar
- Department of Biomedical Engineering and Biointerfaces Institute2800 Plymouth Road, North Campus Research ComplexAnn ArborMIA10‐183USA
- Program in Cellular and Molecular Biology2800 Plymouth Road, North Campus Research ComplexAnn ArborMIA10‐183USA
| |
Collapse
|
165
|
Gola A, Fuchs E. Environmental control of lineage plasticity and stem cell memory. Curr Opin Cell Biol 2021; 69:88-95. [PMID: 33535130 DOI: 10.1016/j.ceb.2020.12.015] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Revised: 12/16/2020] [Accepted: 12/22/2020] [Indexed: 12/19/2022]
Abstract
Tissue-resident stem cells (SCs) are critical players in the maintenance of tissue homeostasis. SCs reside in complex and uniquely anatomically organized microenvironments (SC niches), that carefully control SC lineage outputs depending on localized tissue needs. Upon environmental perturbations and tissue stressors, SCs respond and restore the tissue to homeostasis, as well as protect it from secondary assaults. Critical to this function are two key processes, SC lineage plasticity and SC memory. In this review, we delineate the multifactorial determinants and key principles underlining these two remarkable SC behaviors. Understanding lineage plasticity and SC memory will be critical not only to design new regenerative therapies but also to determine how these processes are altered in a multitude of pathologies such as cancer and chronic tissue damage.
Collapse
Affiliation(s)
- Anita Gola
- Robin Chemers Neustein Laboratory of Mammalian Cell Biology and Development, Howard Hughes Medical Institute, The Rockefeller University, New York, 10065, NY, USA
| | - Elaine Fuchs
- Robin Chemers Neustein Laboratory of Mammalian Cell Biology and Development, Howard Hughes Medical Institute, The Rockefeller University, New York, 10065, NY, USA.
| |
Collapse
|
166
|
Molecular Mechanisms of Renal Progenitor Regulation: How Many Pieces in the Puzzle? Cells 2021; 10:cells10010059. [PMID: 33401654 PMCID: PMC7823786 DOI: 10.3390/cells10010059] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 12/26/2020] [Accepted: 12/29/2020] [Indexed: 12/12/2022] Open
Abstract
Kidneys of mice, rats and humans possess progenitors that maintain daily homeostasis and take part in endogenous regenerative processes following injury, owing to their capacity to proliferate and differentiate. In the glomerular and tubular compartments of the nephron, consistent studies demonstrated that well-characterized, distinct populations of progenitor cells, localized in the parietal epithelium of Bowman capsule and scattered in the proximal and distal tubules, could generate segment-specific cells in physiological conditions and following tissue injury. However, defective or abnormal regenerative responses of these progenitors can contribute to pathologic conditions. The molecular characteristics of renal progenitors have been extensively studied, revealing that numerous classical and evolutionarily conserved pathways, such as Notch or Wnt/β-catenin, play a major role in cell regulation. Others, such as retinoic acid, renin-angiotensin-aldosterone system, TLR2 (Toll-like receptor 2) and leptin, are also important in this process. In this review, we summarize the plethora of molecular mechanisms directing renal progenitor responses during homeostasis and following kidney injury. Finally, we will explore how single-cell RNA sequencing could bring the characterization of renal progenitors to the next level, while knowing their molecular signature is gaining relevance in the clinic.
Collapse
|
167
|
Yeola A, Subramanian S, Oliver RA, Lucas CA, Thoms JAI, Yan F, Olivier J, Chacon D, Tursky ML, Srivastava P, Potas JR, Hung T, Power C, Hardy P, Ma DD, Kilian KA, McCarroll J, Kavallaris M, Hesson LB, Beck D, Curtis DJ, Wong JWH, Hardeman EC, Walsh WR, Mobbs R, Chandrakanthan V, Pimanda JE. Induction of muscle-regenerative multipotent stem cells from human adipocytes by PDGF-AB and 5-azacytidine. SCIENCE ADVANCES 2021; 7:7/3/eabd1929. [PMID: 33523875 PMCID: PMC7806226 DOI: 10.1126/sciadv.abd1929] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Accepted: 11/23/2020] [Indexed: 06/12/2023]
Abstract
Terminally differentiated murine osteocytes and adipocytes can be reprogrammed using platelet-derived growth factor-AB and 5-azacytidine into multipotent stem cells with stromal cell characteristics. We have now optimized culture conditions to reprogram human adipocytes into induced multipotent stem (iMS) cells and characterized their molecular and functional properties. Although the basal transcriptomes of adipocyte-derived iMS cells and adipose tissue-derived mesenchymal stem cells were similar, there were changes in histone modifications and CpG methylation at cis-regulatory regions consistent with an epigenetic landscape that was primed for tissue development and differentiation. In a non-specific tissue injury xenograft model, iMS cells contributed directly to muscle, bone, cartilage, and blood vessels, with no evidence of teratogenic potential. In a cardiotoxin muscle injury model, iMS cells contributed specifically to satellite cells and myofibers without ectopic tissue formation. Together, human adipocyte-derived iMS cells regenerate tissues in a context-dependent manner without ectopic or neoplastic growth.
Collapse
Affiliation(s)
- Avani Yeola
- Adult Cancer Program, Lowy Cancer Research Centre, UNSW Sydney, Sydney, NSW 2052, Australia
- School of Medical Sciences, Faculty of Medicine, UNSW Sydney, Sydney, NSW 2052, Australia
| | - Shruthi Subramanian
- Adult Cancer Program, Lowy Cancer Research Centre, UNSW Sydney, Sydney, NSW 2052, Australia
- Prince of Wales Clinical School, Faculty of Medicine, UNSW Sydney, Sydney, NSW 2052, Australia
| | - Rema A Oliver
- Surgical and Orthopaedic Research Laboratories, Prince of Wales Clinical School, UNSW Sydney, Sydney, NSW 2052, Australia
| | - Christine A Lucas
- Cellular and Genetic Medicine Unit, School of Medical Sciences, UNSW Sydney, Sydney, NSW 2052, Australia
| | - Julie A I Thoms
- Adult Cancer Program, Lowy Cancer Research Centre, UNSW Sydney, Sydney, NSW 2052, Australia
- School of Medical Sciences, Faculty of Medicine, UNSW Sydney, Sydney, NSW 2052, Australia
| | - Feng Yan
- Australian Centre for Blood Diseases, Central Clinical School, Monash University, Melbourne, VIC, Australia
| | - Jake Olivier
- School of Mathematics and Statistics, UNSW Sydney, Sydney, NSW 2052, Australia
| | - Diego Chacon
- School of Biomedical Engineering, University of Technology Sydney, Sydney, NSW 2007, Australia
| | - Melinda L Tursky
- St. Vincent's Centre for Applied Medical Research, St Vincent's Hospital Sydney and St Vincent's Clinical School, Faculty of Medicine, UNSW Sydney, Sydney, NSW 2010, Australia
| | - Pallavi Srivastava
- School of Material Sciences and Engineering, School of Chemistry, Australian Centre for Nanomedicine, UNSW Sydney, Sydney, NSW 2052, Australia
| | - Jason R Potas
- Translational Neuroscience Facility, School of Medical Sciences, UNSW Sydney, Sydney, NSW 2052, Australia
| | - Tzongtyng Hung
- Biological Resources Imaging Laboratory, Mark Wainwright Analytical Centre, UNSW Sydney, Sydney, NSW 2052, Australia
| | - Carl Power
- Biological Resources Imaging Laboratory, Mark Wainwright Analytical Centre, UNSW Sydney, Sydney, NSW 2052, Australia
| | | | - David D Ma
- St. Vincent's Centre for Applied Medical Research, St Vincent's Hospital Sydney and St Vincent's Clinical School, Faculty of Medicine, UNSW Sydney, Sydney, NSW 2010, Australia
| | - Kristopher A Kilian
- School of Material Sciences and Engineering, School of Chemistry, Australian Centre for Nanomedicine, UNSW Sydney, Sydney, NSW 2052, Australia
| | - Joshua McCarroll
- Children's Cancer Institute, Lowy Cancer Research Centre, University of New South Wales Sydney, Sydney, NSW, Australia
| | - Maria Kavallaris
- Children's Cancer Institute, Lowy Cancer Research Centre, University of New South Wales Sydney, Sydney, NSW, Australia
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, Australian Centre for Nanomedicine, UNSW Sydney, Sydney, NSW 2052, Australia
- School of Women's and Children's Health, Faculty of Medicine, UNSW Sydney, Sydney, NSW 2052, Australia
| | - Luke B Hesson
- Prince of Wales Clinical School, Faculty of Medicine, UNSW Sydney, Sydney, NSW 2052, Australia
- Kinghorn Centre for Clinical Genomics, Garvan Institute of Medical Research, Darlinghurst, NSW 2010, Australia
| | - Dominik Beck
- School of Biomedical Engineering, University of Technology Sydney, Sydney, NSW 2007, Australia
| | - David J Curtis
- Australian Centre for Blood Diseases, Central Clinical School, Monash University, Melbourne, VIC, Australia
- Department of Clinical Haematology, Alfred Health, Melbourne, VIC, Australia
| | - Jason W H Wong
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region
| | - Edna C Hardeman
- Cellular and Genetic Medicine Unit, School of Medical Sciences, UNSW Sydney, Sydney, NSW 2052, Australia
| | - William R Walsh
- Surgical and Orthopaedic Research Laboratories, Prince of Wales Clinical School, UNSW Sydney, Sydney, NSW 2052, Australia
| | - Ralph Mobbs
- Surgical and Orthopaedic Research Laboratories, Prince of Wales Clinical School, UNSW Sydney, Sydney, NSW 2052, Australia
- Department of Neurosurgery, Prince of Wales Hospital, Randwick, NSW 2031, Australia
| | - Vashe Chandrakanthan
- Adult Cancer Program, Lowy Cancer Research Centre, UNSW Sydney, Sydney, NSW 2052, Australia.
- School of Medical Sciences, Faculty of Medicine, UNSW Sydney, Sydney, NSW 2052, Australia
| | - John E Pimanda
- Adult Cancer Program, Lowy Cancer Research Centre, UNSW Sydney, Sydney, NSW 2052, Australia.
- School of Medical Sciences, Faculty of Medicine, UNSW Sydney, Sydney, NSW 2052, Australia
- Prince of Wales Clinical School, Faculty of Medicine, UNSW Sydney, Sydney, NSW 2052, Australia
- Department of Haematology, Prince of Wales Hospital, Randwick, NSW 2031, Australia
| |
Collapse
|
168
|
Tenorio-Mina A, Cortés D, Esquivel-Estudillo J, López-Ornelas A, Cabrera-Wrooman A, Lara-Rodarte R, Escobedo-Avila I, Vargas-Romero F, Toledo-Hernández D, Estudillo E, Acevedo-Fernández JJ, Tapia JSO, Velasco I. Human Keratinocytes Adopt Neuronal Fates After In Utero Transplantation in the Developing Rat Brain. Cell Transplant 2021; 30:963689720978219. [PMID: 33435710 PMCID: PMC7809298 DOI: 10.1177/0963689720978219] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Revised: 11/02/2020] [Accepted: 11/12/2020] [Indexed: 11/30/2022] Open
Abstract
Human skin contains keratinocytes in the epidermis. Such cells share their ectodermal origin with the central nervous system (CNS). Recent studies have demonstrated that terminally differentiated somatic cells can adopt a pluripotent state, or can directly convert its phenotype to neurons, after ectopic expression of transcription factors. In this article we tested the hypothesis that human keratinocytes can adopt neural fates after culturing them in suspension with a neural medium. Initially, keratinocytes expressed Keratins and Vimentin. After neural induction, transcriptional upregulation of NESTIN, SOX2, VIMENTIN, SOX1, and MUSASHI1 was observed, concomitant with significant increases in NESTIN detected by immunostaining. However, in vitro differentiation did not yield the expression of neuronal or astrocytic markers. We tested the differentiation potential of control and neural-induced keratinocytes by grafting them in the developing CNS of rats, through ultrasound-guided injection. For this purpose, keratinocytes were transduced with lentivirus that contained the coding sequence of green fluorescent protein. Cell sorting was employed to select cells with high fluorescence. Unexpectedly, 4 days after grafting these cells in the ventricles, both control and neural-induced cells expressed green fluorescent protein together with the neuronal proteins βIII-Tubulin and Microtubule-Associated Protein 2. These results support the notion that in vivo environment provides appropriate signals to evaluate the neuronal differentiation potential of keratinocytes or other non-neural cell populations.
Collapse
Affiliation(s)
- Andrea Tenorio-Mina
- Instituto de Fisiología Celular - Neurociencias, Universidad Nacional Autónoma de México (UNAM), Mexico City, Mexico
- Laboratorio de Reprogramación Celular, Instituto Nacional de Neurología y Neurocirugía “Manuel Velasco Suárez”, Mexico City, Mexico
| | - Daniel Cortés
- Instituto de Fisiología Celular - Neurociencias, Universidad Nacional Autónoma de México (UNAM), Mexico City, Mexico
- Laboratorio de Reprogramación Celular, Instituto Nacional de Neurología y Neurocirugía “Manuel Velasco Suárez”, Mexico City, Mexico
| | - Joel Esquivel-Estudillo
- Facultad de Medicina, Universidad Autónoma del Estado de Morelos, Cuernavaca, Morelos, Mexico
- Unidad de Diagnóstico y Medicina Molecular, “Dr. Ruy Pérez Tamayo”, Hospital del Niño Morelense/Facultad de Medicina-UAEM, Zapata, Morelos, Mexico
| | - Adolfo López-Ornelas
- Instituto de Fisiología Celular - Neurociencias, Universidad Nacional Autónoma de México (UNAM), Mexico City, Mexico
- Laboratorio de Reprogramación Celular, Instituto Nacional de Neurología y Neurocirugía “Manuel Velasco Suárez”, Mexico City, Mexico
- División de Investigación, Hospital Juárez de México, Mexico City, Mexico
| | - Alejandro Cabrera-Wrooman
- Instituto de Fisiología Celular - Neurociencias, Universidad Nacional Autónoma de México (UNAM), Mexico City, Mexico
- Unidad de Diagnóstico y Medicina Molecular, “Dr. Ruy Pérez Tamayo”, Hospital del Niño Morelense/Facultad de Medicina-UAEM, Zapata, Morelos, Mexico
- Instituto Nacional de Rehabilitación, Mexico City, Mexico
| | - Rolando Lara-Rodarte
- Instituto de Fisiología Celular - Neurociencias, Universidad Nacional Autónoma de México (UNAM), Mexico City, Mexico
- Laboratorio de Reprogramación Celular, Instituto Nacional de Neurología y Neurocirugía “Manuel Velasco Suárez”, Mexico City, Mexico
| | - Itzel Escobedo-Avila
- Instituto de Fisiología Celular - Neurociencias, Universidad Nacional Autónoma de México (UNAM), Mexico City, Mexico
| | - Fernanda Vargas-Romero
- Instituto de Fisiología Celular - Neurociencias, Universidad Nacional Autónoma de México (UNAM), Mexico City, Mexico
- Laboratorio de Reprogramación Celular, Instituto Nacional de Neurología y Neurocirugía “Manuel Velasco Suárez”, Mexico City, Mexico
| | - Diana Toledo-Hernández
- Unidad de Diagnóstico y Medicina Molecular, “Dr. Ruy Pérez Tamayo”, Hospital del Niño Morelense/Facultad de Medicina-UAEM, Zapata, Morelos, Mexico
- Centro de Investigación en Dinámica Celular, Instituto de Ciencias, Universidad Autónoma del Estado de Morelos, Cuernavaca, Morelos, Mexico
| | - Enrique Estudillo
- Laboratorio de Reprogramación Celular, Instituto Nacional de Neurología y Neurocirugía “Manuel Velasco Suárez”, Mexico City, Mexico
| | | | - Jesús Santa-Olalla Tapia
- Facultad de Medicina, Universidad Autónoma del Estado de Morelos, Cuernavaca, Morelos, Mexico
- Unidad de Diagnóstico y Medicina Molecular, “Dr. Ruy Pérez Tamayo”, Hospital del Niño Morelense/Facultad de Medicina-UAEM, Zapata, Morelos, Mexico
| | - Iván Velasco
- Instituto de Fisiología Celular - Neurociencias, Universidad Nacional Autónoma de México (UNAM), Mexico City, Mexico
- Laboratorio de Reprogramación Celular, Instituto Nacional de Neurología y Neurocirugía “Manuel Velasco Suárez”, Mexico City, Mexico
| |
Collapse
|
169
|
Hammer GD, Basham KJ. Stem cell function and plasticity in the normal physiology of the adrenal cortex. Mol Cell Endocrinol 2021; 519:111043. [PMID: 33058950 PMCID: PMC7736543 DOI: 10.1016/j.mce.2020.111043] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2020] [Revised: 09/07/2020] [Accepted: 09/28/2020] [Indexed: 02/07/2023]
Abstract
The adrenal cortex functions to produce steroid hormones necessary for life. To maintain its functional capacity throughout life, the adrenal cortex must be continually replenished and rapidly repaired following injury. Moreover, the adrenal responds to endocrine-mediated organismal needs, which are highly dynamic and necessitate a precise steroidogenic response. To meet these diverse needs, the adrenal employs multiple cell populations with stem cell function. Here, we discuss the literature on adrenocortical stem cells using hematopoietic stem cells as a benchmark to examine the functional capacity of particular cell populations, including those located in the capsule and peripheral cortex. These populations are coordinately regulated by paracrine and endocrine signaling mechanisms, and display remarkable plasticity to adapt to different physiological and pathological conditions. Some populations also exhibit sex-specific activity, which contributes to highly divergent proliferation rates between sexes. Understanding mechanisms that govern adrenocortical renewal has broad implications for both regenerative medicine and cancer.
Collapse
Affiliation(s)
- Gary D Hammer
- Department of Internal Medicine, Division of Metabolism, Endocrinology, and Diabetes, University of Michigan, Ann Arbor, MI, USA; Endocrine Oncology Program, Comprehensive Cancer Center, University of Michigan, Ann Arbor, MI, USA
| | - Kaitlin J Basham
- Department of Internal Medicine, Division of Metabolism, Endocrinology, and Diabetes, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
170
|
Seldin L, Macara IG. DNA Damage Promotes Epithelial Hyperplasia and Fate Mis-specification via Fibroblast Inflammasome Activation. Dev Cell 2020; 55:558-573.e6. [PMID: 33058780 PMCID: PMC7725994 DOI: 10.1016/j.devcel.2020.09.021] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2020] [Revised: 08/04/2020] [Accepted: 09/21/2020] [Indexed: 12/21/2022]
Abstract
DNA crosslinking agents are commonly used in cancer chemotherapy; however, responses of normal tissues to these agents have not been widely investigated. We reveal in mouse interfollicular epidermal, mammary and hair follicle epithelia that genotoxicity does not promote apoptosis but paradoxically induces hyperplasia and fate specification defects in quiescent stem cells. DNA damage to skin causes epithelial and dermal hyperplasia, tissue expansion, and proliferation-independent formation of abnormal K14/K10 dual-positive suprabasal cells. Unexpectedly, this behavior is epithelial cell non-autonomous and independent of an intact immune system. Instead, dermal fibroblasts are both necessary and sufficient to induce the epithelial response, which is mediated by activation of a fibroblast-specific NLRP3 inflammasome and subsequent IL-1β production. Thus, genotoxic agents that are used chemotherapeutically to promote cancer cell death can have the opposite effect on wild-type epithelia by inducing, via a non-autonomous IL-1β-driven mechanism, both hyperplasia and stem cell lineage defects.
Collapse
Affiliation(s)
- Lindsey Seldin
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN 37240, USA
| | - Ian G Macara
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN 37240, USA.
| |
Collapse
|
171
|
Nikolenko VN, Oganesyan MV, Sankova MV, Bulygin KV, Vovkogon AD, Rizaeva NA, Sinelnikov MY. Paneth cells: Maintaining dynamic microbiome-host homeostasis, protecting against inflammation and cancer. Bioessays 2020; 43:e2000180. [PMID: 33244814 DOI: 10.1002/bies.202000180] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Revised: 10/15/2020] [Accepted: 10/15/2020] [Indexed: 12/30/2022]
Abstract
The human intestines are constantly under the influence of numerous pathological factors: enteropathogenic microorganisms, food antigens, physico-chemical stress associated with digestion and bacterial metabolism, therefore it must be provided with a system of protection against adverse impact. Recent studies have shown that Paneth cells play a crucial role in maintaining homeostasis of the small intestines. Paneth cells perform many vital functions aimed at maintaining a homeostatic balance between normal microbiota, infectious pathogens and the human body, regulate the qualitative composition and number of intestinal microorganisms, prevent the introduction of potentially pathogenic species, and protect stem cells from damage. Paneth cells take part in adaptive and protective-inflammatory reactions. Paneth cells maintain dynamic balance between microbial populations, and the macroorganism, preventing the development of intestinal infections and cancer. They play a crucial role in gastrointestinal homeostasis and may be key factors in the etiopathological progression of intestinal diseases.
Collapse
Affiliation(s)
- Vladimir N Nikolenko
- Department of Human Anatomy, First Moscow State Medical University named after I.M.Sechenov (Sechenov University), Moscow, Russia.,Department of Normal and Topographic Anatomy, Lomonosov Moscow State University, Moscow, Russia
| | - Marine V Oganesyan
- Department of Human Anatomy, First Moscow State Medical University named after I.M.Sechenov (Sechenov University), Moscow, Russia
| | - Maria V Sankova
- Department of Human Anatomy, First Moscow State Medical University named after I.M.Sechenov (Sechenov University), Moscow, Russia
| | - Kirill V Bulygin
- Department of Human Anatomy, First Moscow State Medical University named after I.M.Sechenov (Sechenov University), Moscow, Russia.,Department of Normal and Topographic Anatomy, Lomonosov Moscow State University, Moscow, Russia
| | - Andzhela D Vovkogon
- Department of Human Anatomy, First Moscow State Medical University named after I.M.Sechenov (Sechenov University), Moscow, Russia
| | - Negoriya A Rizaeva
- Department of Human Anatomy, First Moscow State Medical University named after I.M.Sechenov (Sechenov University), Moscow, Russia
| | | |
Collapse
|
172
|
Coelomocyte replenishment in adult Asterias rubens: the possible ways. Cell Tissue Res 2020; 383:1043-1060. [PMID: 33237478 DOI: 10.1007/s00441-020-03337-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Accepted: 11/05/2020] [Indexed: 10/22/2022]
Abstract
The origin of cells involved in regeneration in echinoderms remains an open question. Replenishment of circulatory coelomocytes-cells of the coelomic cavity in starfish-is an example of physiological regeneration. The coelomic epithelium is considered to be the main source of coelomocytes, but many details of this process remain unclear. This study examined the role of coelomocytes outside circulation, named marginal coelomocytes and small undifferentiated cells of the coelomic epithelium in coelomocyte replenishment in Asterias rubens. A qualitative and quantitative comparison of circulatory and marginal coelomocytes, as well as changes of circulatory coelomocyte concentrations in response to injury at different physiological statuses, was analysed. The presence of cells morphologically similar to coelomocytes in the context of coelomic epithelium was evaluated by electron microscopy. The irregular distribution of small cells on the surface and within the coelomic epithelium was demonstrated and the origin of small undifferentiated cells and large agranulocytes from the coelomic epithelium was suggested. Two events have been proposed to mediate the replenishment of coelomocytes in the coelom: migration of mature coelomocytes of the marginal cell pool and migration of small undifferentiated cells of the coelomic epithelium. The proteomic analysis of circulatory coelomocytes, coelomic epithelial cells and a subpopulation of coelomic epithelial cells, enriched in small undifferentiated cells, revealed proteins that were common and specific for each cell pool. Among these molecules were regulatory proteins, potential participants of regenerative processes.
Collapse
|
173
|
Abstract
The cardinal properties of adult tissue stem cells are self-renewal and the ability to generate diverse resident cell types. The daily losses of terminally differentiated intestinal, skin, and blood cells require "professional" stem cells to produce replacements. This occurs by continuous expansion of stem cells and their immediate progeny, followed by coordinated activation of divergent transcriptional programs to generate stable cells with diverse functions. Other tissues turn over slowly, if at all, and vary widely in strategies for facultative stem cell activity or interconversion among mature resident cell types (transdifferentiation). Cell fate potential is programmed in tissue-specific configurations of chromatin, which restrict the complement of available genes and cis-regulatory elements, hence allowing specific cell types to arise. Using as a model the transcriptional and chromatin basis of cell differentiation and dedifferentiation in intestinal crypts, we discuss here how self-renewing and other tissues execute homeostatic and injury-responsive stem cell activity.
Collapse
Affiliation(s)
- Madhurima Saxena
- Department of Medical Oncology and Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, Massachusetts 02215, USA; .,Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Harvard University, Boston, Massachusetts 02215, USA.,Current affiliation: Translational Medicine, Bristol-Myers-Squibb, Cambridge, Massachusetts 02142, USA;
| | - Ramesh A Shivdasani
- Department of Medical Oncology and Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, Massachusetts 02215, USA; .,Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Harvard University, Boston, Massachusetts 02215, USA.,Harvard Stem Cell Institute, Cambridge, Massachusetts 02138, USA
| |
Collapse
|
174
|
Froidure A, Marchal-Duval E, Homps-Legrand M, Ghanem M, Justet A, Crestani B, Mailleux A. Chaotic activation of developmental signalling pathways drives idiopathic pulmonary fibrosis. Eur Respir Rev 2020; 29:29/158/190140. [PMID: 33208483 DOI: 10.1183/16000617.0140-2019] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Accepted: 05/25/2020] [Indexed: 12/28/2022] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is characterised by an important remodelling of lung parenchyma. Current evidence indicates that the disease is triggered by alveolar epithelium activation following chronic lung injury, resulting in alveolar epithelial type 2 cell hyperplasia and bronchiolisation of alveoli. Signals are then delivered to fibroblasts that undergo differentiation into myofibroblasts. These changes in lung architecture require the activation of developmental pathways that are important regulators of cell transformation, growth and migration. Among others, aberrant expression of profibrotic Wnt-β-catenin, transforming growth factor-β and Sonic hedgehog pathways in IPF fibroblasts has been assessed. In the present review, we will discuss the transcriptional integration of these different pathways during IPF as compared with lung early ontogeny. We will challenge the hypothesis that aberrant transcriptional integration of these pathways might be under the control of a chaotic dynamic, meaning that a small change in baseline conditions could be sufficient to trigger fibrosis rather than repair in a chronically injured lung. Finally, we will discuss some potential opportunities for treatment, either by suppressing deleterious mechanisms or by enhancing the expression of pathways involved in lung repair. Whether developmental mechanisms are involved in repair processes induced by stem cell therapy will also be discussed.
Collapse
Affiliation(s)
- Antoine Froidure
- Institut National de la Santé et de la Recherche Médical, UMR1152, Labex Inflamex, DHU FIRE, Université de Paris, Faculté de médecine Xavier Bichat, Paris, France.,Institut de Recherche Expérimentale et Clinique, Pôle de Pneumologie, Université catholique de Louvain, Belgium Service de pneumologie, Cliniques Universitaires Saint-Luc, Brussels, Belgium
| | - Emmeline Marchal-Duval
- Institut National de la Santé et de la Recherche Médical, UMR1152, Labex Inflamex, DHU FIRE, Université de Paris, Faculté de médecine Xavier Bichat, Paris, France
| | - Meline Homps-Legrand
- Institut National de la Santé et de la Recherche Médical, UMR1152, Labex Inflamex, DHU FIRE, Université de Paris, Faculté de médecine Xavier Bichat, Paris, France
| | - Mada Ghanem
- Institut National de la Santé et de la Recherche Médical, UMR1152, Labex Inflamex, DHU FIRE, Université de Paris, Faculté de médecine Xavier Bichat, Paris, France.,Assistance Publique des Hôpitaux de Paris, Hôpital Bichat, Service de Pneumologie A, DHU FIRE, Paris, France
| | - Aurélien Justet
- Institut National de la Santé et de la Recherche Médical, UMR1152, Labex Inflamex, DHU FIRE, Université de Paris, Faculté de médecine Xavier Bichat, Paris, France.,Assistance Publique des Hôpitaux de Paris, Hôpital Bichat, Service de Pneumologie A, DHU FIRE, Paris, France.,Service de pneumologie, CHU de Caen, Caen, France
| | - Bruno Crestani
- Institut National de la Santé et de la Recherche Médical, UMR1152, Labex Inflamex, DHU FIRE, Université de Paris, Faculté de médecine Xavier Bichat, Paris, France.,Assistance Publique des Hôpitaux de Paris, Hôpital Bichat, Service de Pneumologie A, DHU FIRE, Paris, France
| | - Arnaud Mailleux
- Institut National de la Santé et de la Recherche Médical, UMR1152, Labex Inflamex, DHU FIRE, Université de Paris, Faculté de médecine Xavier Bichat, Paris, France
| |
Collapse
|
175
|
Agudo J. Immune privilege of skin stem cells: What do we know and what can we learn? Exp Dermatol 2020; 30:522-528. [PMID: 33103270 DOI: 10.1111/exd.14221] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Revised: 09/25/2020] [Accepted: 10/19/2020] [Indexed: 12/26/2022]
Abstract
The skin forms a barrier that prevents dehydration and keeps us safe from pathogens. To ensure proper function, the skin possesses a myriad of stem cell populations that are essential for maintenance and repair upon damage. In order to protect, the skin is also an active immunological site, with abundant resident immune cells and strong recruitment of even more immune cells during wounding or infection. Such active and strong immunity makes the skin susceptible to a diverse spectrum of autoimmune diseases, such as vitiligo and alopecia areata. Conversely, despite constant immune surveillance, the skin is also a tissue where frequent malignancies occur, which suggests that immune evasion must also take place. Skin stem cells play a crucial role during both regeneration and tumorigenesis. How immune cells, and in particular T cells, interact with skin stem cells and the implications this crosstalk has in skin disease (both autoimmunity and cancer) is not fully understood. Uncovering the mechanisms governing immune-stem cells interactions in the skin is critical for the development of new therapeutic strategies to safeguard susceptible cells during autoimmunity and, conversely, to improve cancer immunotherapy. Here, I will discuss how distinct skin stem cell populations are attacked by, or conversely, cloaked from immune cells, and the implications their differences have in autoimmunity and cancer.
Collapse
Affiliation(s)
- Judith Agudo
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, MA, USA.,Department of Immunology, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
176
|
Tp63-expressing adult epithelial stem cells cross lineages boundaries revealing latent hairy skin competence. Nat Commun 2020; 11:5645. [PMID: 33159086 PMCID: PMC7648065 DOI: 10.1038/s41467-020-19485-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2018] [Accepted: 10/15/2020] [Indexed: 12/30/2022] Open
Abstract
The formation of hair follicles, a landmark of mammals, requires complex mesenchymal–epithelial interactions and it is commonly believed that embryonic epidermal cells are the only cells that can respond to hair follicle morphogenetic signals in vivo. Here, we demonstrate that epithelial stem cells of non-skin origin (e.g. that of cornea, oesophagus, vagina, bladder, prostate) that express the transcription factor Tp63, a master gene for the development of epidermis and its appendages, can respond to skin morphogenetic signals. When exposed to a newborn skin microenvironment, these cells express hair-follicle lineage markers and contribute to hair follicles, sebaceous glands and/or epidermis renewal. Our results demonstrate that lineage restriction is not immutable and support the notion that all Tp63-expressing epithelial stem cells, independently of their embryonic origin, have latent skin competence explaining why aberrant hair follicles or sebaceous glands are sometimes observed in non-skin tissues (e.g. in cornea, vagina or thymus). Adult stem cells are thought to be fate restricted to lineages distinct to their tissue of origin. Here, the authors demonstrate that Tp63 expressing epithelial stem cells from several disparate tissues can respond to skin morphogenetic signals and contribute to hair follicles, sebaceous glands and/or epidermis.
Collapse
|
177
|
Matsushita Y, Ono W, Ono N. Bone regeneration via skeletal cell lineage plasticity: All hands mobilized for emergencies: Quiescent mature skeletal cells can be activated in response to injury and robustly participate in bone regeneration through cellular plasticity. Bioessays 2020; 43:e2000202. [PMID: 33155283 DOI: 10.1002/bies.202000202] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2020] [Revised: 09/16/2020] [Accepted: 09/24/2020] [Indexed: 12/17/2022]
Abstract
An emerging concept is that quiescent mature skeletal cells provide an important cellular source for bone regeneration. It has long been considered that a small number of resident skeletal stem cells are solely responsible for the remarkable regenerative capacity of adult bones. However, recent in vivo lineage-tracing studies suggest that all stages of skeletal lineage cells, including dormant pre-adipocyte-like stromal cells in the marrow, osteoblast precursor cells on the bone surface and other stem and progenitor cells, are concomitantly recruited to the injury site and collectively participate in regeneration of the damaged skeletal structure. Lineage plasticity appears to play an important role in this process, by which mature skeletal cells can transform their identities into skeletal stem cell-like cells in response to injury. These highly malleable, long-living mature skeletal cells, readily available throughout postnatal life, might represent an ideal cellular resource that can be exploited for regenerative medicine.
Collapse
Affiliation(s)
- Yuki Matsushita
- University of Michigan School of Dentistry, Ann Arbor, Michigan, 48109, USA
| | - Wanida Ono
- University of Michigan School of Dentistry, Ann Arbor, Michigan, 48109, USA
| | - Noriaki Ono
- University of Michigan School of Dentistry, Ann Arbor, Michigan, 48109, USA
| |
Collapse
|
178
|
Andreeva NV, Gabbasova RR, Grivennikov SI. Microbiome in cancer progression and therapy. Curr Opin Microbiol 2020; 56:118-126. [PMID: 33147555 DOI: 10.1016/j.mib.2020.09.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Revised: 09/01/2020] [Accepted: 09/03/2020] [Indexed: 02/07/2023]
Abstract
A myriad of microbes living together with the host constitute microbiota, which possesses very diverse functions in regulation of host physiology. Recently, it has been unequivocally demonstrated that microbiota regulates cancer initiation, progression and responses to therapy. Here we review known pro-tumorigenic and anti-tumorigenic function of microbiota and mechanisms how microbes can regulate cancer cells and immune and stromal cells within the tumor microenvironment.
Collapse
Affiliation(s)
- Natalia V Andreeva
- Cancer Prevention and Control Program, Fox Chase Cancer Center, Philadelphia, PA, 19111, USA; Samuel Oschin Comprehensive Cancer Institute and Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA
| | - Railia R Gabbasova
- Cancer Prevention and Control Program, Fox Chase Cancer Center, Philadelphia, PA, 19111, USA; Samuel Oschin Comprehensive Cancer Institute and Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA
| | - Sergei I Grivennikov
- Cancer Prevention and Control Program, Fox Chase Cancer Center, Philadelphia, PA, 19111, USA; Samuel Oschin Comprehensive Cancer Institute and Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA.
| |
Collapse
|
179
|
Ninche N, Kwak M, Ghazizadeh S. Diverse epithelial cell populations contribute to the regeneration of secretory units in injured salivary glands. Development 2020; 147:dev.192807. [PMID: 32994165 DOI: 10.1242/dev.192807] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Accepted: 09/11/2020] [Indexed: 12/15/2022]
Abstract
Salivary glands exert exocrine secretory function to provide saliva for lubrication and protection of the oral cavity. Its epithelium consists of several differentiated cell types, including acinar, ductal and myoepithelial cells, that are maintained in a lineage-restricted manner during homeostasis or after mild injuries. Glandular regeneration following a near complete loss of secretory cells, however, may involve cellular plasticity, although the mechanism and extent of such plasticity remain unclear. Here, by combining lineage-tracing experiments with a model of severe glandular injury in the mouse submandibular gland, we show that de novo formation of acini involves induction of cellular plasticity in multiple non-acinar cell populations. Fate-mapping analysis revealed that, although ductal stem cells marked by cytokeratin K14 and Axin2 undergo a multipotency switch, they do not make a significant contribution to acinar regeneration. Intriguingly, more than 80% of regenerated acini derive from differentiated cells, including myoepithelial and ductal cells, that appear to dedifferentiate to a progenitor-like state before re-differentiation into acinar cells. The potential of diverse cell populations serving as a reserve source for acini widens the therapeutic options for hyposalivation.
Collapse
Affiliation(s)
- Ninche Ninche
- Department of Oral Biology and Pathology, Stony Brook University, Stony Brook, NY 11794, USA
| | - Mingyu Kwak
- Department of Oral Biology and Pathology, Stony Brook University, Stony Brook, NY 11794, USA
| | - Soosan Ghazizadeh
- Department of Oral Biology and Pathology, Stony Brook University, Stony Brook, NY 11794, USA
| |
Collapse
|
180
|
Engelsen AST, Wnuk-Lipinska K, Bougnaud S, Pelissier Vatter FA, Tiron C, Villadsen R, Miyano M, Lotsberg ML, Madeleine N, Panahandeh P, Dhakal S, Tan TZ, Peters SD, Grøndal S, Aziz SM, Nord S, Herfindal L, Stampfer MR, Sørlie T, Brekken RA, Straume O, Halberg N, Gausdal G, Thiery JP, Akslen LA, Petersen OW, LaBarge MA, Lorens JB. AXL Is a Driver of Stemness in Normal Mammary Gland and Breast Cancer. iScience 2020; 23:101649. [PMID: 33103086 PMCID: PMC7578759 DOI: 10.1016/j.isci.2020.101649] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2018] [Revised: 08/03/2020] [Accepted: 10/02/2020] [Indexed: 12/17/2022] Open
Abstract
The receptor tyrosine kinase AXL is associated with epithelial plasticity in several solid tumors including breast cancer and AXL-targeting agents are currently in clinical trials. We hypothesized that AXL is a driver of stemness traits in cancer by co-option of a regulatory function normally reserved for stem cells. AXL-expressing cells in human mammary epithelial ducts co-expressed markers associated with multipotency, and AXL inhibition abolished colony formation and self-maintenance activities while promoting terminal differentiation in vitro. Axl-null mice did not exhibit a strong developmental phenotype, but enrichment of Axl + cells was required for mouse mammary gland reconstitution upon transplantation, and Axl-null mice had reduced incidence of Wnt1-driven mammary tumors. An AXL-dependent gene signature is a feature of transcriptomes in basal breast cancers and reduced patient survival irrespective of subtype. Our interpretation is that AXL regulates access to epithelial plasticity programs in MaSCs and, when co-opted, maintains acquired stemness in breast cancer cells.
Collapse
Affiliation(s)
- Agnete S T Engelsen
- Department of Biomedicine, University of Bergen, 5021 Bergen, Norway.,Centre for Cancer Biomarkers, University of Bergen, 5021 Bergen, Norway.,INSERM UMR 1186, Integrative Tumor Immunology and Genetic Oncology, Gustave Roussy Cancer Campus Grand Paris, 94800 Villejuif, France
| | | | - Sebastien Bougnaud
- Department of Biomedicine, University of Bergen, 5021 Bergen, Norway.,Centre for Cancer Biomarkers, University of Bergen, 5021 Bergen, Norway
| | - Fanny A Pelissier Vatter
- Department of Biomedicine, University of Bergen, 5021 Bergen, Norway.,Centre for Cancer Biomarkers, University of Bergen, 5021 Bergen, Norway
| | - Crina Tiron
- Department of Biomedicine, University of Bergen, 5021 Bergen, Norway
| | - René Villadsen
- Department of Cellular and Molecular Medicine, University of Copenhagen, Copenhagen, Copenhagen N 2200, Denmark
| | - Masaru Miyano
- Biolgical Systems and Engineering Division, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA.,Department of Population Sciences, Beckman Research Institute at City of Hope, Duarte, CA 91910, USA
| | - Maria L Lotsberg
- Department of Biomedicine, University of Bergen, 5021 Bergen, Norway.,Centre for Cancer Biomarkers, University of Bergen, 5021 Bergen, Norway
| | - Noëlly Madeleine
- Department of Biomedicine, University of Bergen, 5021 Bergen, Norway
| | - Pouda Panahandeh
- Department of Biomedicine, University of Bergen, 5021 Bergen, Norway
| | - Sushil Dhakal
- Department of Biomedicine, University of Bergen, 5021 Bergen, Norway
| | - Tuan Zea Tan
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228, Singapore
| | | | - Sturla Grøndal
- Department of Biomedicine, University of Bergen, 5021 Bergen, Norway
| | - Sura M Aziz
- Department of Clinical Science, University of Bergen, 5021 Bergen, Norway.,Centre for Cancer Biomarkers, University of Bergen, 5021 Bergen, Norway.,Department of Pathology, Haukeland University Hospital, 5021 Bergen, Norway
| | - Silje Nord
- Department of Cancer Research, Oslo University Hospital, 0310 Oslo, Norway
| | - Lars Herfindal
- Department of Biomedicine, University of Bergen, 5021 Bergen, Norway
| | - Martha R Stampfer
- Biolgical Systems and Engineering Division, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA
| | - Therese Sørlie
- Department of Cancer Research, Oslo University Hospital, 0310 Oslo, Norway
| | - Rolf A Brekken
- Hamon Center for Therapeutic Oncology Research, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Oddbjørn Straume
- Centre for Cancer Biomarkers, University of Bergen, 5021 Bergen, Norway.,Department of Oncology and Medical Physics, Haukeland University Hospital, 5021 Bergen, Norway
| | - Nils Halberg
- Department of Biomedicine, University of Bergen, 5021 Bergen, Norway
| | - Gro Gausdal
- Department of Biomedicine, University of Bergen, 5021 Bergen, Norway
| | - Jean Paul Thiery
- Centre for Cancer Biomarkers, University of Bergen, 5021 Bergen, Norway.,INSERM UMR 1186, Integrative Tumor Immunology and Genetic Oncology, Gustave Roussy Cancer Campus Grand Paris, 94800 Villejuif, France.,Cancer Science Institute of Singapore, National University of Singapore, Singapore 117599, Singapore.,Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228, Singapore.,Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, A-STAR, Singapore 138673, Singapore.,Bioland Laboratory, Guangzhou Regenerative Medicine and Health, Bio-island, Guangzhou, 510320, China
| | - Lars A Akslen
- Department of Clinical Science, University of Bergen, 5021 Bergen, Norway.,Centre for Cancer Biomarkers, University of Bergen, 5021 Bergen, Norway.,Department of Pathology, Haukeland University Hospital, 5021 Bergen, Norway
| | - Ole W Petersen
- Department of Cellular and Molecular Medicine, University of Copenhagen, Copenhagen, Copenhagen N 2200, Denmark.,Novo Nordisk Foundation Center for Stem Cell Biology, University of Copenhagen, Copenhagen, Copenhagen N 2200, Denmark
| | - Mark A LaBarge
- Centre for Cancer Biomarkers, University of Bergen, 5021 Bergen, Norway.,Biolgical Systems and Engineering Division, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA.,Department of Population Sciences, Beckman Research Institute at City of Hope, Duarte, CA 91910, USA
| | - James B Lorens
- Department of Biomedicine, University of Bergen, 5021 Bergen, Norway.,Centre for Cancer Biomarkers, University of Bergen, 5021 Bergen, Norway
| |
Collapse
|
181
|
Kim CS, Ding X, Allmeroth K, Biggs LC, Kolenc OI, L'Hoest N, Chacón-Martínez CA, Edlich-Muth C, Giavalisco P, Quinn KP, Denzel MS, Eming SA, Wickström SA. Glutamine Metabolism Controls Stem Cell Fate Reversibility and Long-Term Maintenance in the Hair Follicle. Cell Metab 2020; 32:629-642.e8. [PMID: 32905798 DOI: 10.1016/j.cmet.2020.08.011] [Citation(s) in RCA: 63] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Revised: 06/30/2020] [Accepted: 08/13/2020] [Indexed: 12/12/2022]
Abstract
Stem cells reside in specialized niches that are critical for their function. Upon activation, hair follicle stem cells (HFSCs) exit their niche to generate the outer root sheath (ORS), but a subset of ORS progeny returns to the niche to resume an SC state. Mechanisms of this fate reversibility are unclear. We show that the ability of ORS cells to return to the SC state requires suppression of a metabolic switch from glycolysis to oxidative phosphorylation and glutamine metabolism that occurs during early HFSC lineage progression. HFSC fate reversibility and glutamine metabolism are regulated by the mammalian target of rapamycin complex 2 (mTORC2)-Akt signaling axis within the niche. Deletion of mTORC2 results in a failure to re-establish the HFSC niche, defective hair follicle regeneration, and compromised long-term maintenance of HFSCs. These findings highlight the importance of spatiotemporal control of SC metabolic states in organ homeostasis.
Collapse
Affiliation(s)
- Christine S Kim
- Max Planck Institute for Biology of Ageing, Cologne, Germany; Cluster of Excellence Cellular Stress Responses in Ageing-associated Diseases (CECAD), University of Cologne, Cologne, Germany
| | - Xiaolei Ding
- Department of Dermatology, University of Cologne, Cologne, Germany
| | - Kira Allmeroth
- Max Planck Institute for Biology of Ageing, Cologne, Germany; Cluster of Excellence Cellular Stress Responses in Ageing-associated Diseases (CECAD), University of Cologne, Cologne, Germany
| | - Leah C Biggs
- Helsinki Institute of Life Science, Biomedicum Helsinki, University of Helsinki, Helsinki, Finland; Wihuri Research Institute, Biomedicum Helsinki, University of Helsinki, Helsinki, Finland; Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Olivia I Kolenc
- Department of Biomedical Engineering, University of Arkansas, Fayetteville, AR, USA
| | - Nina L'Hoest
- Max Planck Institute for Biology of Ageing, Cologne, Germany; Cluster of Excellence Cellular Stress Responses in Ageing-associated Diseases (CECAD), University of Cologne, Cologne, Germany
| | - Carlos Andrés Chacón-Martínez
- Max Planck Institute for Biology of Ageing, Cologne, Germany; Cluster of Excellence Cellular Stress Responses in Ageing-associated Diseases (CECAD), University of Cologne, Cologne, Germany
| | | | | | - Kyle P Quinn
- Department of Biomedical Engineering, University of Arkansas, Fayetteville, AR, USA
| | - Martin S Denzel
- Max Planck Institute for Biology of Ageing, Cologne, Germany; Cluster of Excellence Cellular Stress Responses in Ageing-associated Diseases (CECAD), University of Cologne, Cologne, Germany
| | - Sabine A Eming
- Cluster of Excellence Cellular Stress Responses in Ageing-associated Diseases (CECAD), University of Cologne, Cologne, Germany; Department of Dermatology, University of Cologne, Cologne, Germany; Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany; Institute of Zoology, Developmental Biology Unit, University of Cologne, Cologne, Germany.
| | - Sara A Wickström
- Max Planck Institute for Biology of Ageing, Cologne, Germany; Cluster of Excellence Cellular Stress Responses in Ageing-associated Diseases (CECAD), University of Cologne, Cologne, Germany; Helsinki Institute of Life Science, Biomedicum Helsinki, University of Helsinki, Helsinki, Finland; Wihuri Research Institute, Biomedicum Helsinki, University of Helsinki, Helsinki, Finland; Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland.
| |
Collapse
|
182
|
Abstract
Interactions between disseminated cancer cells and the microenvironment in secondary organs are essential for the development of metastasis in most malignancies. Metastasis-initiating cells and their progeny can impose changes in the microenvironment leading to the formation of a metastatic niche that supports malignant growth at secondary sites. Our recent findings indicate that stress responses play a crucial role in generation of metastatic niches in breast cancer by modulating the extracellular matrix and promoting interactions with reactive fibroblasts.
Collapse
Affiliation(s)
- Thordur Oskarsson
- Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM gGmbH), Heidelberg, Germany.,Division of Stem Cells and Cancer, German Cancer Research Center (DKFZ), Heidelberg, Germany.,German Cancer Consortium (DKTK), Heidelberg, Germany
| |
Collapse
|
183
|
Jung Y, Kim JK, Lee E, Cackowski FC, Decker AM, Krebsbach PH, Taichman RS. CXCL12γ induces human prostate and mammary gland development. Prostate 2020; 80:1145-1156. [PMID: 32659025 PMCID: PMC7491592 DOI: 10.1002/pros.24043] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Accepted: 06/11/2020] [Indexed: 01/01/2023]
Abstract
BACKGROUND Epithelial stem cells (ESCs) demonstrate a capacity to maintain normal tissues homeostasis and ESCs with a deregulated behavior can contribute to cancer development. The ability to reprogram normal tissue epithelial cells into prostate or mammary stem-like cells holds great promise to help understand cell of origin and lineage plasticity in prostate and breast cancers in addition to understanding normal gland development. We previously showed that an intracellular chemokine, CXCL12γ induced cancer stem cells and neuroendocrine characteristics in both prostate and breast adenocarcinoma cell lines. However, its role in normal prostate or mammary epithelial cell fate and development remains unknown. Therefore, we sought to elucidate the functional role of CXCL12γ in the regulation of ESCs and tissue development. METHODS Prostate epithelial cells (PNT2) or mammary epithelial cells (MCF10A) with overexpressed CXCL12γ was characterized by quantitative real-time polymerase chain reaction, Western blots, and immunofluorescence for lineage marker expression, and fluorescence activated cell sorting analyses and sphere formation assays to examine stem cell surface phenotype and function. Xenotransplantation animal models were used to evaluate gland or acini formation in vivo. RESULTS Overexpression of CXCL12γ promotes the reprogramming of cells with a differentiated luminal phenotype to a nonluminal phenotype in both prostate (PNT2) and mammary (MCF10A) epithelial cells. The CXCL12γ-mediated nonluminal type cells results in an increase of epithelial stem-like phenotype including the subpopulation of EPCAMLo /CD49fHi /CD24Lo /CD44Hi cells capable of sphere formation. Critically, overexpression of CXCL12γ promotes the generation of robust gland-like structures from both prostate and mammary epithelial cells in in vivo xenograft animal models. CONCLUSIONS CXCL12γ supports the reprogramming of epithelial cells into nonluminal cell-derived stem cells, which facilitates gland development.
Collapse
Affiliation(s)
- Younghun Jung
- Department of Periodontics and Oral Medicine, University of Michigan School of Dentistry, Ann Arbor, MI 48109, USA
- Department of Biologic and Materials Sciences, University of Michigan School of Dentistry, Ann Arbor, MI 48109, USA
- Co-senior authors
| | - Jin Koo Kim
- Section of Periodontics, University of California Los Angeles School of Dentistry, Los Angeles, CA 90095, USA
| | - Eunsohl Lee
- Department of Periodontics and Oral Medicine, University of Michigan School of Dentistry, Ann Arbor, MI 48109, USA
| | - Frank C. Cackowski
- Department of Periodontics and Oral Medicine, University of Michigan School of Dentistry, Ann Arbor, MI 48109, USA
- Department of Internal Medicine, Division of Hematology and Oncology, University of Michigan School of Medicine, Ann Arbor, MI 48109, USA
- Department of Oncology, Wayne State University and Karmanos Cancer Institute, Detroit, MI 48201, USA
| | - Ann M. Decker
- Department of Periodontics and Oral Medicine, University of Michigan School of Dentistry, Ann Arbor, MI 48109, USA
| | - Paul H. Krebsbach
- Section of Periodontics, University of California Los Angeles School of Dentistry, Los Angeles, CA 90095, USA
| | - Russell S. Taichman
- Department of Periodontics and Oral Medicine, University of Michigan School of Dentistry, Ann Arbor, MI 48109, USA
- Department of Periodontics, University of Alabama at Birmingham, Birmingham, AL 35294, USA
- Co-senior authors
- Corresponding Author Russell S. Taichman D.M.D., D.M.Sc., School of Dentistry, The University of Alabama at Birmingham, 1720 2nd Avenue South, Birmingham, AL 35294-0007, Phone: 205-934-4720,
| |
Collapse
|
184
|
Xie C, Feng H, Zhong L, Shi Y, Wei Z, Hua Y, Ji N, Li J, Tang Z, Chen Q. Proliferative ability and accumulation of cancer stem cells in oral submucous fibrosis epithelium. Oral Dis 2020; 26:1255-1264. [PMID: 32267986 DOI: 10.1111/odi.13347] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Revised: 03/25/2020] [Accepted: 03/27/2020] [Indexed: 02/05/2023]
Abstract
OBJECTIVES The driving force of the malignant transformation of epithelial cells during oral submucous fibrosis (OSF) is an unsettled debate. We hypothesized that the expression and accumulation of cancer stem cells (CSCs) are accompanied by epithelial atrophy in OSF. MATERIALS AND METHODS The expression levels of Ki67 (proliferation marker), SOX2, and Bmi1 (CSC marker) in the epithelium during the early, middle, and late stages of OSF were measured by immunohistochemistry. At the same time, we focused on the expression of three proteins in OSF patients with benign hyperkeratosis and epithelial dysplasia. RESULTS The clinical cohort study showed upregulated expression of the proliferation-associated protein Ki67 in atrophic epithelium in patients with OSF. The expression levels of SOX2 and Bmi1 showed an increasing trend in the progression of OSF. Ki67, SOX2, and Bmi1 were highly expressed in OSF tissues with dysplasia. Moreover, the three proteins were located at the epithelial and mesenchymal junctions, and their expression showed a positive correlation with each other. CONCLUSION The results suggest that CSC accumulation could be accompanied by epithelial atrophy during OSF, which may be responsible for the driving forces for OSF carcinogenesis.
Collapse
Affiliation(s)
- Changqing Xie
- Department of Oral and Maxillofacial Surgery, Xiangya Stomalogical Hospital, Central South University, Changsha, China
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Hui Feng
- Department of Oral and Maxillofacial Surgery, Xiangya Stomalogical Hospital, Central South University, Changsha, China
| | - Liang Zhong
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Yujie Shi
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Zihao Wei
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Yufei Hua
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Ning Ji
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Jing Li
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Zhangui Tang
- Department of Oral and Maxillofacial Surgery, Xiangya Stomalogical Hospital, Central South University, Changsha, China
| | - Qianming Chen
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| |
Collapse
|
185
|
Huang Y, Seitz D, Chevalier Y, Müller PE, Jansson V, Klar RM. Synergistic interaction of hTGF-β 3 with hBMP-6 promotes articular cartilage formation in chitosan scaffolds with hADSCs: implications for regenerative medicine. BMC Biotechnol 2020; 20:48. [PMID: 32854680 PMCID: PMC7457281 DOI: 10.1186/s12896-020-00641-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Accepted: 08/20/2020] [Indexed: 12/31/2022] Open
Abstract
Background Human TGF-β3 has been used in many studies to induce genes coding for typical cartilage matrix components and accelerate chondrogenic differentiation, making it the standard constituent in most cultivation media used for the assessment of chondrogenesis associated with various stem cell types on carrier matrices. However, in vivo data suggests that TGF-β3 and its other isoforms also induce endochondral and intramembranous osteogenesis in non-primate species to other mammals. Based on previously demonstrated improved articular cartilage induction by a using hTGF-β3 and hBMP-6 together on hADSC cultures and the interaction of TGF- β with matrix in vivo, the present study investigates the interaction of a chitosan scaffold as polyanionic polysaccharide with both growth factors. The study analyzes the difference between chondrogenic differentiation that leads to stable hyaline cartilage and the endochondral ossification route that ends in hypertrophy by extending the usual panel of investigated gene expression and stringent employment of quantitative PCR. Results By assessing the viability, proliferation, matrix formation and gene expression patterns it is shown that hTGF-β3 + hBMP-6 promotes improved hyaline articular cartilage formation in a chitosan scaffold in which ACAN with Col2A1 and not Col1A1 nor Col10A1 where highly expressed both at a transcriptional and translational level. Inversely, hTGF-β3 alone tended towards endochondral bone formation showing according protein and gene expression patterns. Conclusion These findings demonstrate that clinical therapies should consider using hTGF-β3 + hBMP-6 in articular cartilage regeneration therapies as the synergistic interaction of these morphogens seems to ensure and maintain proper hyaline articular cartilage matrix formation counteracting degeneration to fibrous tissue or ossification. These effects are produced by interaction of the growth factors with the polysaccharide matrix.
Collapse
Affiliation(s)
- Yijiang Huang
- Department of Orthopaedics, Physical Medicine and Rehabilitation, University Hospital of Munich, 81377, Munich, Germany
| | - Daniel Seitz
- BioMed Center Innovation gGmbh, 95448, Bayreuth, Germany
| | - Yan Chevalier
- Department of Orthopaedics, Physical Medicine and Rehabilitation, University Hospital of Munich, 81377, Munich, Germany
| | - Peter E Müller
- Department of Orthopaedics, Physical Medicine and Rehabilitation, University Hospital of Munich, 81377, Munich, Germany
| | - Volkmar Jansson
- Department of Orthopaedics, Physical Medicine and Rehabilitation, University Hospital of Munich, 81377, Munich, Germany
| | - Roland M Klar
- Department of Orthopaedics, Physical Medicine and Rehabilitation, University Hospital of Munich, 81377, Munich, Germany.
| |
Collapse
|
186
|
Lynch CJ, Bernad R, Calvo I, Serrano M. Manipulating the Mediator complex to induce naïve pluripotency. Exp Cell Res 2020; 395:112215. [PMID: 32771524 PMCID: PMC7584500 DOI: 10.1016/j.yexcr.2020.112215] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Revised: 07/08/2020] [Accepted: 08/01/2020] [Indexed: 12/26/2022]
Abstract
Human naïve pluripotent stem cells (PSCs) represent an optimal homogenous starting point for molecular interventions and differentiation strategies. This is in contrast to the standard primed PSCs which fluctuate in identity and are transcriptionally heterogeneous. However, despite many efforts, the maintenance and expansion of human naïve PSCs remains a challenge. Here, we discuss our recent strategy for the stabilization of human PSC in the naïve state based on the use of a single chemical inhibitor of the related kinases CDK8 and CDK19. These kinases phosphorylate and negatively regulate the multiprotein Mediator complex, which is critical for enhancer-driven recruitment of RNA Pol II. The net effect of CDK8/19 inhibition is a global stimulation of enhancers, which in turn reinforces transcriptional programs including those related to cellular identity. In the case of pluripotent cells, the presence of CDK8/19i efficiently stabilizes the naïve state. Importantly, in contrast to previous chemical methods to induced the naïve state based on the inhibition of the FGF-MEK-ERK pathway, CDK8/19i-naïve human PSCs are chromosomally stable and retain developmental potential after long-term expansion. We suggest this could be related to the fact that CDK8/19 inhibition does not induce DNA demethylation. These principles may apply to other fate decisions.
Collapse
Affiliation(s)
- Cian J Lynch
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology (BIST), Barcelona, Spain.
| | - Raquel Bernad
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
| | - Isabel Calvo
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
| | - Manuel Serrano
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology (BIST), Barcelona, Spain; Catalan Institution for Research and Advanced Studies (ICREA), Barcelona, 08010, Spain.
| |
Collapse
|
187
|
Forkosh E, Kenig A, Ilan Y. Introducing variability in targeting the microtubules: Review of current mechanisms and future directions in colchicine therapy. Pharmacol Res Perspect 2020; 8:e00616. [PMID: 32608157 PMCID: PMC7327382 DOI: 10.1002/prp2.616] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2020] [Accepted: 05/25/2020] [Indexed: 12/14/2022] Open
Abstract
Microtubules (MTs) are highly dynamic polymers that constitute the cellular cytoskeleton and play a role in multiple cellular functions. Variability characterizes biological systems and is considered a part of the normal function of cells and organs. Variability contributes to cell plasticity and is a mechanism for overcoming errors in cellular level assembly and function, and potentially the whole organ level. Dynamic instability is a feature of biological variability that characterizes the function of MTs. The dynamic behavior of MTs constitutes the basis for multiple biological processes that contribute to cellular plasticity and the timing of cell signaling. Colchicine is a MT-modifying drug that exerts anti-inflammatory and anti-cancer effects. This review discusses some of the functions of colchicine and presents a platform for introducing variability while targeting MTs in intestinal cells, the microbiome, the gut, and the systemic immune system. This platform can be used for implementing novel therapies, improving response to chronic MT-based therapies, overcoming drug resistance, exerting gut-based systemic immune responses, and generating patient-tailored dynamic therapeutic regimens.
Collapse
Affiliation(s)
- Esther Forkosh
- Department of MedicineHebrew University‐Hadassah Medical CentreJerusalemIsrael
| | - Ariel Kenig
- Department of MedicineHebrew University‐Hadassah Medical CentreJerusalemIsrael
| | - Yaron Ilan
- Department of MedicineHebrew University‐Hadassah Medical CentreJerusalemIsrael
| |
Collapse
|
188
|
Regan JL, Smalley MJ. Integrating single-cell RNA-sequencing and functional assays to decipher mammary cell states and lineage hierarchies. NPJ Breast Cancer 2020; 6:32. [PMID: 32793804 PMCID: PMC7391676 DOI: 10.1038/s41523-020-00175-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Accepted: 07/02/2020] [Indexed: 12/13/2022] Open
Abstract
The identification and molecular characterization of cellular hierarchies in complex tissues is key to understanding both normal cellular homeostasis and tumorigenesis. The mammary epithelium is a heterogeneous tissue consisting of two main cellular compartments, an outer basal layer containing myoepithelial cells and an inner luminal layer consisting of estrogen receptor-negative (ER−) ductal cells and secretory alveolar cells (in the fully functional differentiated tissue) and hormone-responsive estrogen receptor-positive (ER+) cells. Recent publications have used single-cell RNA-sequencing (scRNA-seq) analysis to decipher epithelial cell differentiation hierarchies in human and murine mammary glands, and reported the identification of new cell types and states based on the expression of the luminal progenitor cell marker KIT (c-Kit). These studies allow for comprehensive and unbiased analysis of the different cell types that constitute a heterogeneous tissue. Here we discuss scRNA-seq studies in the context of previous research in which mammary epithelial cell populations were molecularly and functionally characterized, and identified c-Kit+ progenitors and cell states analogous to those reported in the recent scRNA-seq studies.
Collapse
Affiliation(s)
- Joseph L Regan
- Charité Comprehensive Cancer Centre, Charité - Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany
| | - Matthew J Smalley
- European Cancer Stem Cell Research Institute, School of Biosciences, Cardiff University, Hadyn Ellis Building, Wales, CF24 4HQ UK
| |
Collapse
|
189
|
Aragona M, Sifrim A, Malfait M, Song Y, Van Herck J, Dekoninck S, Gargouri S, Lapouge G, Swedlund B, Dubois C, Baatsen P, Vints K, Han S, Tissir F, Voet T, Simons BD, Blanpain C. Mechanisms of stretch-mediated skin expansion at single-cell resolution. Nature 2020; 584:268-273. [PMID: 32728211 DOI: 10.1038/s41586-020-2555-7] [Citation(s) in RCA: 114] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Accepted: 06/19/2020] [Indexed: 02/06/2023]
Abstract
The ability of the skin to grow in response to stretching has been exploited in reconstructive surgery1. Although the response of epidermal cells to stretching has been studied in vitro2,3, it remains unclear how mechanical forces affect their behaviour in vivo. Here we develop a mouse model in which the consequences of stretching on skin epidermis can be studied at single-cell resolution. Using a multidisciplinary approach that combines clonal analysis with quantitative modelling and single-cell RNA sequencing, we show that stretching induces skin expansion by creating a transient bias in the renewal activity of epidermal stem cells, while a second subpopulation of basal progenitors remains committed to differentiation. Transcriptional and chromatin profiling identifies how cell states and gene-regulatory networks are modulated by stretching. Using pharmacological inhibitors and mouse mutants, we define the step-by-step mechanisms that control stretch-mediated tissue expansion at single-cell resolution in vivo.
Collapse
Affiliation(s)
- Mariaceleste Aragona
- Laboratory of Stem Cells and Cancer, Université Libre de Bruxelles, Brussels, Belgium
| | - Alejandro Sifrim
- Department of Human Genetics, University of Leuven, KU Leuven, Leuven, Belgium.,Sanger Institute-EBI Single-Cell Genomics Centre, Wellcome Trust Sanger Institute, Hinxton, UK
| | - Milan Malfait
- Laboratory of Stem Cells and Cancer, Université Libre de Bruxelles, Brussels, Belgium
| | - Yura Song
- Laboratory of Stem Cells and Cancer, Université Libre de Bruxelles, Brussels, Belgium
| | - Jens Van Herck
- Department of Human Genetics, University of Leuven, KU Leuven, Leuven, Belgium.,Sanger Institute-EBI Single-Cell Genomics Centre, Wellcome Trust Sanger Institute, Hinxton, UK
| | - Sophie Dekoninck
- Laboratory of Stem Cells and Cancer, Université Libre de Bruxelles, Brussels, Belgium
| | - Souhir Gargouri
- Laboratory of Stem Cells and Cancer, Université Libre de Bruxelles, Brussels, Belgium
| | - Gaëlle Lapouge
- Laboratory of Stem Cells and Cancer, Université Libre de Bruxelles, Brussels, Belgium
| | - Benjamin Swedlund
- Laboratory of Stem Cells and Cancer, Université Libre de Bruxelles, Brussels, Belgium
| | - Christine Dubois
- Laboratory of Stem Cells and Cancer, Université Libre de Bruxelles, Brussels, Belgium
| | - Pieter Baatsen
- Electron Microscopy Platform of VIB Bio Imaging Core, Leuven, Belgium
| | - Katlijn Vints
- Electron Microscopy Platform of VIB Bio Imaging Core, Leuven, Belgium
| | - Seungmin Han
- The Wellcome Trust-Cancer Research UK Gurdon Institute, University of Cambridge, Cambridge, UK.,Wellcome Trust-Medical Research Council Stem Cell Institute, University of Cambridge, Cambridge, UK
| | - Fadel Tissir
- Université Catholique de Louvain, Institute of Neuroscience, Developmental Neurobiology, Brussels, Belgium
| | - Thierry Voet
- Department of Human Genetics, University of Leuven, KU Leuven, Leuven, Belgium.,Sanger Institute-EBI Single-Cell Genomics Centre, Wellcome Trust Sanger Institute, Hinxton, UK
| | - Benjamin D Simons
- The Wellcome Trust-Cancer Research UK Gurdon Institute, University of Cambridge, Cambridge, UK. .,Wellcome Trust-Medical Research Council Stem Cell Institute, University of Cambridge, Cambridge, UK. .,Department of Applied Mathematics and Theoretical Physics, Centre for Mathematical Sciences, University of Cambridge, Cambridge, UK.
| | - Cédric Blanpain
- Laboratory of Stem Cells and Cancer, Université Libre de Bruxelles, Brussels, Belgium. .,WELBIO, Université Libre de Bruxelles, Brussels, Belgium.
| |
Collapse
|
190
|
Src Family Tyrosine Kinases in Intestinal Homeostasis, Regeneration and Tumorigenesis. Cancers (Basel) 2020; 12:cancers12082014. [PMID: 32717909 PMCID: PMC7464719 DOI: 10.3390/cancers12082014] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 07/18/2020] [Accepted: 07/19/2020] [Indexed: 01/11/2023] Open
Abstract
Src, originally identified as an oncogene, is a membrane-anchored tyrosine kinase and the Src family kinase (SFK) prototype. SFKs regulate the signalling induced by a wide range of cell surface receptors leading to epithelial cell growth and adhesion. In the intestine, the SFK members Src, Fyn and Yes regulate epithelial cell proliferation and migration during tissue regeneration and transformation, thus implicating conserved and specific functions. In patients with colon cancer, SFK activity is a marker of poor clinical prognosis and a potent driver of metastasis formation. These tumorigenic activities are linked to SFK capacity to promote the dissemination and tumour-initiating capacities of epithelial tumour cells. However, it is unclear how SFKs promote colon tumour formation and metastatic progression because SFK-encoding genes are unfrequently mutated in human cancer. Here, we review recent findings on SFK signalling during intestinal homeostasis, regeneration and tumorigenesis. We also describe the key nongenetic mechanisms underlying SFK tumour activities in colorectal cancer, and discuss how these mechanisms could be exploited in therapeutic strategies to target SFK signalling in metastatic colon cancer.
Collapse
|
191
|
Parigini C, Greulich P. Universality of clonal dynamics poses fundamental limits to identify stem cell self-renewal strategies. eLife 2020; 9:56532. [PMID: 32687057 PMCID: PMC7444910 DOI: 10.7554/elife.56532] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Accepted: 07/03/2020] [Indexed: 12/21/2022] Open
Abstract
How adult stem cells maintain self-renewing tissues is commonly assessed by analysing clonal data from in vivo cell lineage-tracing assays. To identify strategies of stem cell self-renewal requires that different models of stem cell fate choice predict sufficiently different clonal statistics. Here, we show that models of cell fate choice can, in homeostatic tissues, be categorized by exactly two 'universality classes', whereby models of the same class predict, under asymptotic conditions, the same clonal statistics. Those classes relate to generalizations of the canonical asymmetric vs. symmetric stem cell self-renewal strategies and are distinguished by a conservation law. This poses both challenges and opportunities to identify stem cell self-renewal strategies: while under asymptotic conditions, self-renewal models of the same universality class cannot be distinguished by clonal data only, models of different classes can be distinguished by simple means.
Collapse
Affiliation(s)
- Cristina Parigini
- School of Mathematical Science, University of Southampton, Southampton, United Kingdom.,Institute for Life Sciences, University of Southampton, Southampton, United Kingdom
| | - Philip Greulich
- School of Mathematical Science, University of Southampton, Southampton, United Kingdom.,Institute for Life Sciences, University of Southampton, Southampton, United Kingdom
| |
Collapse
|
192
|
Abstract
Understanding to what extent stem cell potential is a cell-intrinsic property or an emergent behavior coming from global tissue dynamics and geometry is a key outstanding question of systems and stem cell biology. Here, we propose a theory of stem cell dynamics as a stochastic competition for access to a spatially localized niche, giving rise to a stochastic conveyor-belt model. Cell divisions produce a steady cellular stream which advects cells away from the niche, while random rearrangements enable cells away from the niche to be favorably repositioned. Importantly, even when assuming that all cells in a tissue are molecularly equivalent, we predict a common ("universal") functional dependence of the long-term clonal survival probability on distance from the niche, as well as the emergence of a well-defined number of functional stem cells, dependent only on the rate of random movements vs. mitosis-driven advection. We test the predictions of this theory on datasets of pubertal mammary gland tips and embryonic kidney tips, as well as homeostatic intestinal crypts. Importantly, we find good agreement for the predicted functional dependency of the competition as a function of position, and thus functional stem cell number in each organ. This argues for a key role of positional fluctuations in dictating stem cell number and dynamics, and we discuss the applicability of this theory to other settings.
Collapse
|
193
|
Sweat gland regeneration: Current strategies and future opportunities. Biomaterials 2020; 255:120201. [PMID: 32592872 DOI: 10.1016/j.biomaterials.2020.120201] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Revised: 04/21/2020] [Accepted: 06/09/2020] [Indexed: 12/13/2022]
Abstract
For patients with extensive skin defects, loss of sweat glands (SwGs) greatly decreases their quality of life. Indeed, difficulties in thermoregulation, ion reabsorption, and maintaining fluid balance might render them susceptible to hyperthermia, heatstroke, or even death. Despite extensive studies on the stem cell biology of the skin in recent years, in-situ regeneration of SwGs with both structural and functional fidelity is still challenging because of the limited regenerative capacity and cell fate control of resident progenitors. To overcome these challenges, one must consider both the intrinsic factors relevant to genetic and epigenetic regulation and cues from the cellular microenvironment. Here, we describe recent progress in molecular biology, developmental pathways, and cellular evolution associated with SwGdevelopment and maturation. This is followed by a summary of the current strategies used for cell-fate modulation, transmembrane drug delivery, and scaffold design associated with SwGregeneration. Finally, we offer perspectives for creating more sophisticated systems to accelerate patients' innate healing capacity and developing engineered skin constructs to treat or replace damaged tissues structurally and functionally.
Collapse
|
194
|
Aggor FEY, Break TJ, Trevejo-Nuñez G, Whibley N, Coleman BM, Bailey RD, Kaplan DH, Naglik JR, Shan W, Shetty AC, McCracken C, Durum SK, Biswas PS, Bruno VM, Kolls JK, Lionakis MS, Gaffen SL. Oral epithelial IL-22/STAT3 signaling licenses IL-17-mediated immunity to oral mucosal candidiasis. Sci Immunol 2020; 5:eaba0570. [PMID: 32503875 PMCID: PMC7340112 DOI: 10.1126/sciimmunol.aba0570] [Citation(s) in RCA: 67] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Accepted: 05/07/2020] [Indexed: 12/29/2022]
Abstract
Oropharyngeal candidiasis (OPC; thrush) is an opportunistic infection caused by the commensal fungus Candida albicans Interleukin-17 (IL-17) and IL-22 are cytokines produced by type 17 lymphocytes. Both cytokines mediate antifungal immunity yet activate quite distinct downstream signaling pathways. While much is now understood about how IL-17 promotes immunity in OPC, the activities of IL-22 are far less well delineated. We show that, despite having similar requirements for induction from type 17 cells, IL-22 and IL-17 function nonredundantly during OPC. We find that the IL-22 and IL-17 receptors are required in anatomically distinct locations within the oral mucosa; loss of IL-22RA1 or signal transducer and activator of transcription 3 (STAT3) in the oral basal epithelial layer (BEL) causes susceptibility to OPC, whereas IL-17RA is needed in the suprabasal epithelial layer (SEL). Transcriptional profiling of the tongue linked IL-22/STAT3 not only to oral epithelial cell proliferation and survival but also, unexpectedly, to driving an IL-17-specific gene signature. We show that IL-22 mediates regenerative signals on the BEL that replenish the IL-17RA-expressing SEL, thereby restoring the ability of the oral epithelium to respond to IL-17 and thus to mediate antifungal events. Consequently, IL-22 signaling in BEL "licenses" IL-17 signaling in the oral mucosa, revealing spatially distinct yet cooperative activities of IL-22 and IL-17 in oral candidiasis.
Collapse
Affiliation(s)
- Felix E Y Aggor
- Division of Rheumatology and Clinical Immunology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Timothy J Break
- Fungal Pathogenesis Section, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD, USA
| | | | - Natasha Whibley
- Division of Rheumatology and Clinical Immunology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Bianca M Coleman
- Division of Rheumatology and Clinical Immunology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Rachel D Bailey
- Division of Rheumatology and Clinical Immunology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Daniel H Kaplan
- Department of Dermatology, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Immunology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Julian R Naglik
- Centre for Host-Microbiome Interactions, Faculty of Dentistry, Oral and Craniofacial Sciences, King's College London, London, UK
| | - Wei Shan
- Cytokines and Immunity Section, Cancer and Inflammation Program, National Cancer Institute, NIH, Frederick, MD, USA
| | - Amol C Shetty
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Carrie McCracken
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Scott K Durum
- Cytokines and Immunity Section, Cancer and Inflammation Program, National Cancer Institute, NIH, Frederick, MD, USA
| | - Partha S Biswas
- Division of Rheumatology and Clinical Immunology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Vincent M Bruno
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Jay K Kolls
- Center for Translational Research in Infection and Inflammation, Tulane University, New Orleans, LA, USA
| | - Michail S Lionakis
- Fungal Pathogenesis Section, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD, USA
| | - Sarah L Gaffen
- Division of Rheumatology and Clinical Immunology, University of Pittsburgh, Pittsburgh, PA, USA.
| |
Collapse
|
195
|
Liu Y, Xiong X, Chen YG. Dedifferentiation: the return road to repair the intestinal epithelium. CELL REGENERATION (LONDON, ENGLAND) 2020; 9:2. [PMID: 32588148 PMCID: PMC7306829 DOI: 10.1186/s13619-020-00048-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
In the March 5 issue of Cell Stem Cell, (Murata K et al. Cell Stem Cell. 26(377–390):e376 2020) reported that intestinal stem cell recovery after injury is principally through Ascl2-dependent dedifferentiation of absorptive and secretory precursors in mice. This study provides evidence for robust regenerative capability of the intestinal epithelium via dedifferentiation of absorptive and secretory progenitors in the crypt.
Collapse
Affiliation(s)
- Yuan Liu
- The State Key Laboratory of Membrane Biology, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Xiaochen Xiong
- Institute of Stem Cell Biology & Regenerative Medicine, School of Medicine, Stanford University, Stanford, CA, 94305, USA
| | - Ye-Guang Chen
- The State Key Laboratory of Membrane Biology, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, 100084, China.
| |
Collapse
|
196
|
Alda-1, an Aldehyde Dehydrogenase 2 Agonist, Improves Cutaneous Wound Healing by Activating Epidermal Keratinocytes via Akt/GSK-3β/β-Catenin Pathway. Aesthetic Plast Surg 2020; 44:993-1005. [PMID: 31953581 DOI: 10.1007/s00266-020-01614-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Accepted: 01/05/2020] [Indexed: 02/07/2023]
Abstract
BACKGROUND The cutaneous wound healing process mainly comprises re-epithelialization, fibrosis, and neovascularization. Impaired wound healing is common but tricky in plastic surgery. Aldehyde dehydrogenase 2 (ALDH2), the most effective subset of the ALDH enzyme family, is known to exert a major role in detoxification of aldehydes. Activation of ALDH2 by Alda-1 (a specific agonist) has been found to protect against cardiovascular diseases. However, no research has paid attention to the potential of ALDH2 activation in regulating wound healing. The previous studies suggested a high expression of ALDH2 in normal skin tissue. The aim of this study was to investigate if Alda-1 may ameliorate wound healing. METHODS A full-thickness excisional wound model was established in vivo. Adult male C57BL/6 mice were randomly divided into DMSO and Alda-1 groups. Mice received an intraperitoneal injection of DMSO or 10 mg/mL Alda-1 (10 mg/kg body weight, dissolved in DMSO) for 7 days. The wound healing rate was measured at 0, 3, 5, and 7 days. Distribution of ALDH2 in wound tissue was showed. ALDH2 enzymatic activity was examined at 3, 5, and 7 days. The elongation of epithelial tongue was detected by hematoxylin-eosin staining, and collagen deposition was analyzed by Masson's trichrome staining at 7 days. Expressions of alpha-smooth muscle actin (alpha-SMA), transforming growth factor beta (TGF-beta), CD31, collagen 1, collagen 3, and elastin were stained by immunohistochemistry at 5 and 7 days. The HaCaT cell line was applied in vitro. Proliferation and migration were tested using CCK8 and wound healing assay separately. The level of TGF-β was examined by ELISA. Protein levels of the Akt/glycogen synthase kinase-3 beta (GSK-3 beta)/beta-catenin pathway were determined by western blotting. RESULTS Alda-1 accelerated wound healing rates. ALDH2 activity in wound sites was restored. Alda-1 promoted the length of the epithelial tongue, collagen deposition, as well as expressions of alpha-SMA, TGF-beta, collagen 1/3, elastin, but did not affect CD31. Proliferation, migration, and TGF-β secretion were promoted by Alda-1 and deregulated by CVT-10216 (an ALDH2 inhibitor). Protein variations of the Akt/GSK-3β/β-catenin pathway were found to accord with ALDH2 changes. CONCLUSIONS Alda-1, an ALDH2 agonist, improves cutaneous wound healing in a full-thickness excisional wound model. Alda-1 activates proliferation, migration, and TGF-β secretion of HaCaT (epidermal keratinocytes) by regulating the Akt/GSK-3β/β-catenin pathway. NO LEVEL ASSIGNED This journal requires that authors assign a level of evidence to each submission to which Evidence-Based Medicine rankings are applicable. This excludes Review Articles, Book Reviews, and manuscripts that concern Basic Science, Animal Studies, Cadaver Studies, and Experimental Studies. For a full description of these Evidence-Based Medicine ratings, please refer to the Table of Contents or the online Instructions to Authors www.springer.com/00266.
Collapse
|
197
|
Zhang P, Ma XY, Huang DT, Yang XY. The capacity of goat epidermal adult stem cells to reconstruct the damaged ocular surface of total LSCD and activate corneal genetic programs. J Mol Histol 2020; 51:277-286. [PMID: 32388840 DOI: 10.1007/s10735-020-09879-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Accepted: 05/04/2020] [Indexed: 11/29/2022]
Abstract
Epidermal adult stem cells (EpiASCs) have the potential for unlimited proliferation and differentiation, however, the ability of these stem cells to activate corneal genetic programs in response to corneal stroma stimulation needs to be further validated. Herein, a feasible strategy was developed to reconstruct the damaged corneal surface in a goat model with total limbal stem cell deficiency (LSCD) by transplanting EpiASCs, which had been explanted and cultured from the skin of an adult ram goat and were then purified by selecting single cell-derived clones and cultivating them on a denuded human amniotic membrane (HAM). These artificial tissues were then successfully transplanted into ewe goats with total LSCD. Binding of EpiASCs to the base membrane of an EpiASCs-HAM-Sheet (EHS) indicated their proliferating status. After transplantation, the EpiASCs could survive in the host tissue and they reconstructed the damaged ocular surface of total LSCD. The crystal reconstructed corneal epithelium expressed CK3 and Pax-6 similar to normal corneal epithelium and expressed the Sry gene after transplantation. These results demonstrated that EpiASCs could be induced to differentiate into corneal epithelial cell types in a corneal microenvironment and had the ability to activate corneal genetic programs. This work offer a foundation for promoting tissue-engineered cornea into clinical application.
Collapse
Affiliation(s)
- Pei Zhang
- Department of Life Science, Luoyang Normal University, Luoyang, 471934, Henan, China
| | - Xi-Ya Ma
- Department of Life Science, Luoyang Normal University, Luoyang, 471934, Henan, China
| | - Dian-Tong Huang
- Department of Life Science, Luoyang Normal University, Luoyang, 471934, Henan, China
| | - Xue-Yi Yang
- Department of Life Science, Luoyang Normal University, Luoyang, 471934, Henan, China.
| |
Collapse
|
198
|
Li S, Hu M, Lorenz HP. Treatment of Full-Thickness Skin Wounds with Blood-Derived CD34 + Precursor Cells Enhances Healing with Hair Follicle Regeneration. Adv Wound Care (New Rochelle) 2020; 9:264-276. [PMID: 32226650 DOI: 10.1089/wound.2019.0974] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Accepted: 04/23/2019] [Indexed: 10/26/2022] Open
Abstract
Objective: Epidermal CD34+ stem cells located in the hair follicle (HF) bulge area are capable of inducing HF neogenesis and enhancing wound healing after transplantation. In this study, we observed CD34+ cells derived from blood directly participate in dermal regeneration during full-thickness excisional wound healing. Approach: We isolated and in vitro expanded a subset of hematopoietic stem cell (HSC)-like precursor cells from the peripheral blood of adult mice with the surface markers: CD34+, leucine rich repeat containing G protein-coupled receptor 5 (LGR5)+, CD44+, c-kit+, lineage negative (lin-), and E-cadherin-. These blood-derived precursor cells (BDPCs), can be further differentiated into epithelial-like cells (eBDPCs) and secret fibroblast growth factor 9 (Fgf9) protein. Result: When transplanted into full-thickness skin wounds, eBDPC treatment produced accelerated healing and enhanced skin structure regeneration with less dermal scar formation. Also, HF neogenesis (HFN) was observed with incorporation of labeled BDPCs in the wound area. Innovation:Nondermal-derived CD34+ cells (BDPCs) from the adult unmobilized peripheral blood are capable of in vitro expansion and differentiation.Successful establishment of an in vitro technical platform for BDPCs expansion and differentiation.The in vitro expanded and differentiated epithelial-like cells (eBDPCs) enhance wound healing and directly contribute to skin regeneration and HFN. Conclusion: BDPCs isolated and expanded from adult peripheral blood may provide a possible new cell-based treatment strategy for HF neogenesis and skin wound regeneration.
Collapse
Affiliation(s)
- Shaowei Li
- Division of Plastic Surgery, Department of Surgery, School of Medicine, Stanford University, Stanford, California
- APstem Therapeutics, Inc., Fremont, California
| | - Min Hu
- Division of Plastic Surgery, Department of Surgery, School of Medicine, Stanford University, Stanford, California
- APstem Therapeutics, Inc., Fremont, California
| | - H. Peter Lorenz
- Division of Plastic Surgery, Department of Surgery, School of Medicine, Stanford University, Stanford, California
| |
Collapse
|
199
|
Frumento G, Verma K, Croft W, White A, Zuo J, Nagy Z, Kissane S, Anderson G, Moss P, Chen FE. Homeostatic Cytokines Drive Epigenetic Reprogramming of Activated T Cells into a "Naive-Memory" Phenotype. iScience 2020; 23:100989. [PMID: 32240954 PMCID: PMC7115140 DOI: 10.1016/j.isci.2020.100989] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Revised: 12/09/2019] [Accepted: 03/11/2020] [Indexed: 11/25/2022] Open
Abstract
Primary stimulation of T cells is believed to trigger unidirectional differentiation from naive to effector and memory subsets. Here we demonstrate that IL-7 can drive the phenotypic reversion of recently differentiated human central and effector memory CD8+ T cells into a naive-like phenotype. These "naive-revertant" cells display a phenotype similar to that of previously reported stem cell memory populations and undergo rapid differentiation and functional response following secondary challenge. The chromatin landscape of reverted cells undergoes substantial epigenetic reorganization with increased accessibility for cytokine-induced mediators such as STAT and closure of BATF-dependent sites that drive terminal differentiation. Phenotypic reversion may at least partly explain the generation of "stem cell memory" CD8+ T cells and reveals cells within the phenotypically naive CD8+ T cell pool that are epigenetically primed for secondary stimulation. This information provides insight into mechanisms that support maintenance of T cell memory and may guide therapeutic manipulation of T cell differentiation.
Collapse
Affiliation(s)
- Guido Frumento
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, UK; NHS Blood and Transplant, Birmingham, UK
| | - Kriti Verma
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, UK
| | - Wayne Croft
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, UK; Centre for Computational Biology, University of Birmingham, Birmingham, UK
| | - Andrea White
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, UK
| | - Jianmin Zuo
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, UK
| | - Zsuzsanna Nagy
- Institute of Inflammation and Ageing, University of Birmingham, Birmingham, UK
| | | | - Graham Anderson
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, UK
| | - Paul Moss
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, UK; Centre for Clinical Haematology, University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK.
| | - Frederick E Chen
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, UK; NHS Blood and Transplant, Birmingham, UK; Clinical Haematology, Barts Health NHS Trust, London, UK; Blizard Institute, Queen Mary University London, London, UK.
| |
Collapse
|
200
|
He N, Su R, Wang Z, Zhang Y, Li J. Exploring differentially expressed genes between anagen and telogen secondary hair follicle stem cells from the Cashmere goat (Capra hircus) by RNA-Seq. PLoS One 2020; 15:e0231376. [PMID: 32298297 PMCID: PMC7162518 DOI: 10.1371/journal.pone.0231376] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2019] [Accepted: 03/23/2020] [Indexed: 12/17/2022] Open
Abstract
Hair follicle stem cells (HFSCs) have been shown to be essential in the development and regeneration of hair follicles (HFs). The Inner Mongolia Cashmere goat (Capra hircus) has two types of HFs, primary and secondary, with cashmere being produced from the secondary hair follicle. To identify the genes associated with cashmere growth, transcriptome profiling of anagen and telogen secondary HFSCs was performed by RNA-Seq. The RNA-Seq analysis generated over 58 million clean reads from each group, with 2717 differentially expressed genes (DEGs) detected between anagen and telogen, including 1500 upregulated and 1217 downregulated DEGs. A large number of DEGs were predominantly associated with cell part, cellular process, binding, biological regulation and organelle. In addition, the PI3K-Akt, MAPK, Ras and Rap1 signaling pathways may be involved in the growth of HFSCs cultured in vitro. The RNA-Seq results showed that the well-defined HFSC signature genes and cell cycle-associated genes showed no significant differences between anagen and telogen HFSCs, indicating a relatively quiescent cellular state of the HFSCs cultured in vitro. These results are useful for future studies of complex molecular mechanisms of hair follicle cycling in cashmere goats.
Collapse
Affiliation(s)
- Nimantana He
- College of Animal Science, Inner Mongolia Agricultural University, Hohhot, Inner Mongolia, China
- Agriculture Research Center, Chifeng University, Chifeng, Inner Mongolia, China
| | - Rui Su
- College of Animal Science, Inner Mongolia Agricultural University, Hohhot, Inner Mongolia, China
- Key Laboratory of Animal Genetics, Breeding and Reproduction, Hohhot, Inner Mongolia Autonomous Region, China
- Key Laboratory of Mutton Sheep Genetics and Breeding, Ministry of Agriculture, Hohhot, China
- Engineering Research Center for Goat Genetics and Breeding, Hohhot, Inner Mongolia Autonomous Region, China
| | - Zhiying Wang
- College of Animal Science, Inner Mongolia Agricultural University, Hohhot, Inner Mongolia, China
- Engineering Research Center for Goat Genetics and Breeding, Hohhot, Inner Mongolia Autonomous Region, China
| | - Yanjun Zhang
- College of Animal Science, Inner Mongolia Agricultural University, Hohhot, Inner Mongolia, China
- Engineering Research Center for Goat Genetics and Breeding, Hohhot, Inner Mongolia Autonomous Region, China
| | - Jinquan Li
- College of Animal Science, Inner Mongolia Agricultural University, Hohhot, Inner Mongolia, China
- Key Laboratory of Animal Genetics, Breeding and Reproduction, Hohhot, Inner Mongolia Autonomous Region, China
- Key Laboratory of Mutton Sheep Genetics and Breeding, Ministry of Agriculture, Hohhot, China
- Engineering Research Center for Goat Genetics and Breeding, Hohhot, Inner Mongolia Autonomous Region, China
- * E-mail:
| |
Collapse
|