1
|
Lou Q, Jiang K, Wang X, Pan Y, Qiu G, Wu B, Yuan L, Xie S, Chen J, Xu Q, Zhao M, Jiang L. IGF1R signaling in perinatal mesenchymal stem cells determines definitive hematopoiesis in bone marrow. Blood 2024; 144:2773-2787. [PMID: 39437540 DOI: 10.1182/blood.2024024258] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Revised: 09/06/2024] [Accepted: 09/21/2024] [Indexed: 10/25/2024] Open
Abstract
ABTRACT During the transition from embryonic to adult life, the sites of hematopoiesis undergo dynamic shifts across various tissues. In adults, although bone marrow (BM) becomes the primary site for definitive hematopoiesis, the establishment of the BM niche for accommodating hematopoietic stem cells (HSCs) remains incompletely understood. Here, we reveal that perinatal BM mesenchymal stem cells (BMSCs) exhibit highly activated insulin-like growth factor 1 receptor (IGF1R) signaling compared with adult BMSCs (aBMSCs). Deletion of Igf1r in perinatal BMSCs (pBMSCs) hinders the transition of HSCs from the fetal liver to the BM in perinatal mice and disrupts hematopoiesis in adult individuals. Conversely, the deletion of Igf1r in aBMSCs, adipocytes, osteoblasts, or endothelial cells does not affect HSCs in the BM. Mechanistically, IGF1R signaling activates the transcription factor nuclear factor of activated T cells c1 in pBMSCs, which upregulates CXCL12 and other niche factors for HSC retention. Overall, IGF1R signaling in pBMSCs regulates the development of the BM niche for hematopoiesis.
Collapse
Affiliation(s)
- Qi Lou
- RNA Biomedical Institute, Sun Yat-sen Memorial Hospital, State Key Laboratory of Oncology in South China, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
- Advanced Medical Technology Center, The First Affiliated Hospital, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
- Key Laboratory of Stem Cells and Tissue Engineering (Ministry of Education), Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Kaizheng Jiang
- RNA Biomedical Institute, Sun Yat-sen Memorial Hospital, State Key Laboratory of Oncology in South China, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
- Advanced Medical Technology Center, The First Affiliated Hospital, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
- Key Laboratory of Stem Cells and Tissue Engineering (Ministry of Education), Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Xiaoqi Wang
- Medical Center of Hematology, Xinqiao Hospital of Army Medical University, Chongqing, China
| | - Yuan Pan
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - Guo Qiu
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Binghuo Wu
- RNA Biomedical Institute, Sun Yat-sen Memorial Hospital, State Key Laboratory of Oncology in South China, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
- Advanced Medical Technology Center, The First Affiliated Hospital, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
- Key Laboratory of Stem Cells and Tissue Engineering (Ministry of Education), Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Lisha Yuan
- RNA Biomedical Institute, Sun Yat-sen Memorial Hospital, State Key Laboratory of Oncology in South China, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
- Advanced Medical Technology Center, The First Affiliated Hospital, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
- Key Laboratory of Stem Cells and Tissue Engineering (Ministry of Education), Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Siyu Xie
- RNA Biomedical Institute, Sun Yat-sen Memorial Hospital, State Key Laboratory of Oncology in South China, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
- Advanced Medical Technology Center, The First Affiliated Hospital, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
- Key Laboratory of Stem Cells and Tissue Engineering (Ministry of Education), Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Jian Chen
- RNA Biomedical Institute, Sun Yat-sen Memorial Hospital, State Key Laboratory of Oncology in South China, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
- Advanced Medical Technology Center, The First Affiliated Hospital, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
- Key Laboratory of Stem Cells and Tissue Engineering (Ministry of Education), Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Quanhui Xu
- RNA Biomedical Institute, Sun Yat-sen Memorial Hospital, State Key Laboratory of Oncology in South China, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
- Advanced Medical Technology Center, The First Affiliated Hospital, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
- Key Laboratory of Stem Cells and Tissue Engineering (Ministry of Education), Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Meng Zhao
- RNA Biomedical Institute, Sun Yat-sen Memorial Hospital, State Key Laboratory of Oncology in South China, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
- Advanced Medical Technology Center, The First Affiliated Hospital, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
- Key Laboratory of Stem Cells and Tissue Engineering (Ministry of Education), Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Linjia Jiang
- RNA Biomedical Institute, Sun Yat-sen Memorial Hospital, State Key Laboratory of Oncology in South China, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
2
|
Efraim Y, Chen FYT, Niknezhad SV, Pham D, Cheong KN, An L, Sinada H, McNamara NA, Knox SM. Rebuilding the autoimmune-damaged corneal stroma through topical lubrication. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.29.626078. [PMID: 39677756 PMCID: PMC11642755 DOI: 10.1101/2024.11.29.626078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2024]
Abstract
Corneal lubrication is the most common treatment for relieving the signs and symptoms of dry eye and is considered to be largely palliative with no regenerative functions. Here we challenge this notion by demonstrating that wetting the desiccated cornea of an aqueous-deficient mouse model with the simplest form of lubrication, a saline-based solution, is sufficient to rescue the severely disrupted collagen-rich architecture of the stroma, the largest corneal compartment that is essential to transparency and vision. At the single cell level we show that stromal keratocytes responsible for maintaining stromal integrity are converted from an inflammatory state into unique reparative cell states by lubrication alone, thus revealing the extensive plasticity of these cells and the regenerative function of lubricating the surface. We further show that the generation of a reparative phenotype is due, in part, to disruption of an IL1β autocrine amplification loop promoting chronic inflammation. Thus, our study uncovers the regenerative potential of topical lubrication in dry eye and represents a paradigm shift in our understanding of its therapeutic impact.
Collapse
Affiliation(s)
- Yael Efraim
- Program in Craniofacial Biology, Department of Cell & Tissue Biology, University of California San Francisco; San Francisco, CA 94143, USA
| | - Feeling Yu Ting Chen
- Program in Craniofacial Biology, Department of Cell & Tissue Biology, University of California San Francisco; San Francisco, CA 94143, USA
| | - Seyyed Vahid Niknezhad
- Program in Craniofacial Biology, Department of Cell & Tissue Biology, University of California San Francisco; San Francisco, CA 94143, USA
| | - Dylan Pham
- Program in Craniofacial Biology, Department of Cell & Tissue Biology, University of California San Francisco; San Francisco, CA 94143, USA
| | - Ka Neng Cheong
- Program in Craniofacial Biology, Department of Cell & Tissue Biology, University of California San Francisco; San Francisco, CA 94143, USA
| | - Luye An
- Program in Craniofacial Biology, Department of Cell & Tissue Biology, University of California San Francisco; San Francisco, CA 94143, USA
| | - Hanan Sinada
- Program in Craniofacial Biology, Department of Cell & Tissue Biology, University of California San Francisco; San Francisco, CA 94143, USA
| | - Nancy A. McNamara
- School of Optometry and Vision Science Graduate Program, University of California, Berkeley; Oakland, CA 94720, USA
- Department of Anatomy, University of California, San Francisco; San Francisco, CA 94143, USA
| | - Sarah M. Knox
- Program in Craniofacial Biology, Department of Cell & Tissue Biology, University of California San Francisco; San Francisco, CA 94143, USA
| |
Collapse
|
3
|
Abbasian H, Noruzinia M, Garshasbi M. Investigation the role of SIRT3, SIRT7, NFATC1, and PDL-1 genes in androgenetic alopecia. BMC Res Notes 2024; 17:343. [PMID: 39574094 PMCID: PMC11583656 DOI: 10.1186/s13104-024-06980-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Accepted: 10/15/2024] [Indexed: 11/24/2024] Open
Abstract
BACKGROUND Androgenetic alopecia (AGA) stands as the most prevalent form of hair loss, affecting the hair follicles (HFs). Aging emerges as a prominent contributor in this condition. In this study, our aim is to elucidate the expression patterns of candidate genes-SIRT3, SIRT7, NFATC1, and PDL-1-known to exhibit differential expression levels during HF aging, and to underscore the role of aging in AGA. MATERIAL AND METHODS Mesenchymal stem cells (MSCs) were isolated from the vertex and occipital regions of six men affected by AGA. The aim was to assess the expression levels of SIRT3, SIRT7, NFATC1, and PDL-1 genes. RNA extraction was performed followed by cDNA synthesis, and gene expression levels were quantified using real-time PCR. To validate the experimental findings, two different RNA-seq datasets relevant to the study were analyzed using R software. RESULTS In the present study, experimental tests revealed that the expression levels of SIRT3 and SIRT7, known to decrease during HF aging, were diminished in AGA-affected samples as well. Conversely, the mean value of NFATC1 and PDL-1 expression level, which are known to increase during HF aging, were found to be elevated in AGA-affected samples. Moreover, bioinformatic analyses provide additional support for the role of SIRT3, SIRT7 and NFATC1in AGA pathogenesis. CONCLUSION While SIRT3 and SIRT7 may play critical roles in AGA development, further research is needed to elucidate the significance of NFATC1 and PDL-1 in this context and to explore their potential as therapeutic targets for AGA treatment.
Collapse
Affiliation(s)
- Hadis Abbasian
- Department of Medical Genetic, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Mehrdad Noruzinia
- Department of Medical Genetic, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Masoud Garshasbi
- Department of Medical Genetic, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran.
| |
Collapse
|
4
|
Panesso-Gómez S, Cole AJ, Wield A, Anyaeche VI, Shah J, Jiang Q, Ebai T, Sharrow AC, Tseng G, Yoon E, Brown DD, Clark AM, Larsen SD, Eder I, Gau D, Roy P, Dahl KN, Tran L, Jiang H, McAuliffe PF, Lee AV, Buckanovich RJ. Identification of the MRTFA/SRF pathway as a critical regulator of quiescence in cancer. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.15.623825. [PMID: 39605642 PMCID: PMC11601311 DOI: 10.1101/2024.11.15.623825] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
Chemoresistance is a major driver of cancer deaths. One understudied mechanism of chemoresistance is quiescence. We used single cell culture to identify, retrieve, and RNA-Seq profile primary quiescent ovarian cancer cells (qOvCa). We found that many qOvCa differentially expressed genes are transcriptional targets of the Myocardin Related Transcription Factor/Serum Response Factor (MRTF/SRF) pathway. We also found that genetic disruption of MRTF-SRF interaction, or an MRTF/SRF inhibitor (CCG257081) impact qOvCa gene expression and induce a quiescent state in cancer cells. Suggesting a broad role for this pathway in quiescence, CCG257081 treatment induced quiescence in breast, lung, colon, pancreatic and ovarian cancer cells. Furthermore, CCG081 (i) maintained a quiescent state in patient derived breast cancer organoids and, (ii) induced tumor growth arrest in ovarian cancer xenografts. Together, these data suggest that MRTF/SRF pathway is a critical regulator of quiescence in cancer and a possible therapeutic target.
Collapse
Affiliation(s)
- Santiago Panesso-Gómez
- Department of Internal Medicine and Magee-Womens Research Institute, University of Pittsburgh, Pittsburgh, PA, USA
| | - Alexander J Cole
- Department of Internal Medicine and Magee-Womens Research Institute, University of Pittsburgh, Pittsburgh, PA, USA
| | - Alyssa Wield
- Department of Internal Medicine and Magee-Womens Research Institute, University of Pittsburgh, Pittsburgh, PA, USA
| | - Vivian I Anyaeche
- Department of Internal Medicine and Magee-Womens Research Institute, University of Pittsburgh, Pittsburgh, PA, USA
| | - Jaynish Shah
- Australian Centre for Blood Diseases, Central Clinical School, Monash University and Alfred Health, Melbourne, VIC, Australia
| | - Qi Jiang
- Department of Internal Medicine and Magee-Womens Research Institute, University of Pittsburgh, Pittsburgh, PA, USA
| | - Tonge Ebai
- Department of Internal Medicine and Magee-Womens Research Institute, University of Pittsburgh, Pittsburgh, PA, USA
| | - Allison C Sharrow
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA, USA
| | - George Tseng
- Department of Biostatistics, University of Pittsburgh, Pittsburgh, PA, USA
| | - Euisik Yoon
- Department of Electrical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Daniel D Brown
- Women's Cancer Research Center, Magee-Women's Research Institute, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Amanda M Clark
- Department of Pathology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Scott D Larsen
- Department of Medicinal Chemistry, University of Michigan, Ann Arbor, Michigan, USA
| | - Ian Eder
- Department of Bioengineering, University of Pittsburgh, PA, USA
| | - David Gau
- Department of Bioengineering, University of Pittsburgh, PA, USA
| | - Partha Roy
- Department of Bioengineering, University of Pittsburgh, PA, USA
| | - Kris N Dahl
- Department of Chemical Engineering, Carnegie Mellon University, Pittsburgh, PA, USA
| | - Lam Tran
- Department of Biostatistics, University of Michigan, Ann Arbor, MI, USA
| | - Hui Jiang
- Department of Biostatistics, University of Michigan, Ann Arbor, MI, USA
| | | | - Adrian V Lee
- Women's Cancer Research Center, Magee-Women's Research Institute, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Ronald J Buckanovich
- Department of Internal Medicine and Magee-Womens Research Institute, University of Pittsburgh, Pittsburgh, PA, USA
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA, USA
| |
Collapse
|
5
|
Flora P, Li MY, Zhou Y, Mercédes M, Zheng XY, Galbo PM, Zheng D, Ezhkova E. H2AK119ub dynamics controls hair follicle stem cell quiescence. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.10.617646. [PMID: 39416158 PMCID: PMC11482967 DOI: 10.1101/2024.10.10.617646] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/19/2024]
Abstract
The transition of stem cells from a quiescent state to an active state is a finely tuned process that requires the dismantling of the quiescence program and the establishment of a cell cycle-promoting transcriptional landscape. Whether epigenetic processes control stem cell states to promote the regeneration of adult tissues remains elusive. In this study, we show that a repressive histone modification, H2AK119ub, is dynamic between quiescent and active hair follicle stem cells (HFSCs) in the adult murine skin. Ablation of H2AK119ub in HFSCs leads to impaired quiescence leading to premature activation and an eventual exhaustion of HFSC pool. Transcriptional and chromatin studies revealed that H2AK119ub directly represses a proliferation promoting transcriptional program in the HFSCs to preserve quiescence. Lastly, we identify that the inhibitory FGF signaling produced by the hair follicle niche keratinocytes maintains H2AK119ub in quiescent HFSCs. Together, these findings reveal that a repressive histone mark, H2AK119ub, is under the dynamic regulation of inhibitory niche signaling to prevent the untimely establishment of an activated state to preserve SC function and longevity.
Collapse
|
6
|
Leung CWB, Wall J, Esashi F. From rest to repair: Safeguarding genomic integrity in quiescent cells. DNA Repair (Amst) 2024; 142:103752. [PMID: 39167890 DOI: 10.1016/j.dnarep.2024.103752] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Revised: 08/12/2024] [Accepted: 08/12/2024] [Indexed: 08/23/2024]
Abstract
Quiescence is an important non-pathological state in which cells pause cell cycle progression temporarily, sometimes for decades, until they receive appropriate proliferative stimuli. Quiescent cells make up a significant proportion of the body, and maintaining genomic integrity during quiescence is crucial for tissue structure and function. While cells in quiescence are spared from DNA damage associated with DNA replication or mitosis, they are still exposed to various sources of endogenous DNA damage, including those induced by normal transcription and metabolism. As such, it is vital that cells retain their capacity to effectively repair lesions that may occur and return to the cell cycle without losing their cellular properties. Notably, while DNA repair pathways are often found to be downregulated in quiescent cells, emerging evidence suggests the presence of active or differentially regulated repair mechanisms. This review aims to provide a current understanding of DNA repair processes during quiescence in mammalian systems and sheds light on the potential pathological consequences of inefficient or inaccurate repair in quiescent cells.
Collapse
Affiliation(s)
| | - Jacob Wall
- Sir William Dunn School of Pathology, South Parks Road, Oxford, UK
| | - Fumiko Esashi
- Sir William Dunn School of Pathology, South Parks Road, Oxford, UK.
| |
Collapse
|
7
|
Olczak A, Pieczonka TD, Ławicki S, Łukaszyk K, Pulawska-Czub A, Cambier L, Kobielak K. The overexpression of R-spondin 3 affects hair morphogenesis and hair development along with the formation and maturation of the hair follicle stem cells. Front Physiol 2024; 15:1424077. [PMID: 39351282 PMCID: PMC11439821 DOI: 10.3389/fphys.2024.1424077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2024] [Accepted: 08/29/2024] [Indexed: 10/04/2024] Open
Abstract
Mice hair follicles (HFs) are a valuable model for studying various aspects of hair biology, including morphogenesis, development, and regeneration due to their easily observable phenotype and genetic manipulability. The initiation and progression of hair follicle morphogenesis, as well as the hair follicle cycle, are regulated by various signaling pathways, of which the main role is played by the Wingless-type MMTV integration site family (Wnt) and the Bone Morphogenic Protein (BMP). During the hair follicle cycle, the BMP pathway maintains hair follicle stem cells (HFSCs) in a dormant state while the Wnt pathway activates them for hair growth. Given the pivotal role of the Wnt pathway in hair biology and HFSCs regulation, we investigated the influence of the Wnt modulator - R-spondin 3 (Rspo3), in these processes. For this purpose, we developed a transgenic mice model with the overexpression of Rspo3 (Rspo3GOF) in the whole ectoderm and its derivatives, starting from early morphogenesis. Rspo3GOF mice exhibited a distinct phenotype with sparse hair and visible bald areas, caused by reduced proliferation and increased apoptosis of hair matrix progenitor cells, which resulted in a premature anagen-to-catagen transition with a shortened growth phase and decreased overall length of all hair types. In addition, Rspo3GOF promoted induction of auchene and awl, canonical Wnt-dependent hair type during morphogenesis, but the overall hair amount remained reduced. We also discovered a delay in the pre-bulge formation during morphogenesis and prolonged immaturity of the HFSC population in the bulge region postnatally, which further impaired proper hair regeneration throughout the mice's lifespan. Our data supported that Rspo3 function observed in our model works in HFSCs' formation of pre-bulge during morphogenesis via enhancing activation of the canonical Wnt pathway, whereas in contrast, in the postnatal immature bulge, activation of canonical Wnt signaling was attenuated. In vitro studies on keratinocytes revealed changes in proliferation, migration, and colony formation, highlighting the inhibitory effect of constitutive overexpression of Rspo3 on these cellular processes. Our research provides novel insights into the role of Rspo3 in the regulation of hair morphogenesis and development, along with the formation and maturation of the HFSCs, which affect hair regeneration.
Collapse
Affiliation(s)
- Alicja Olczak
- Centre of New Technologies (CeNT), University of Warsaw (UW), Warsaw, Poland
| | - Tomasz D. Pieczonka
- Centre of New Technologies (CeNT), University of Warsaw (UW), Warsaw, Poland
| | - Szymon Ławicki
- Centre of New Technologies (CeNT), University of Warsaw (UW), Warsaw, Poland
| | - Konrad Łukaszyk
- Centre of New Technologies (CeNT), University of Warsaw (UW), Warsaw, Poland
| | - Anna Pulawska-Czub
- Centre of New Technologies (CeNT), University of Warsaw (UW), Warsaw, Poland
| | - Linda Cambier
- The Vision Center and The Saban Research Institute, Children’s Hospital Los Angeles, Los Angeles, CA, United States
| | - Krzysztof Kobielak
- Centre of New Technologies (CeNT), University of Warsaw (UW), Warsaw, Poland
| |
Collapse
|
8
|
Wang Y, Wang Y, Xu P. Cyclosporine-induced alopecia:a case report, FDA adverse event reporting system analysis and literature assessment. Front Pharmacol 2024; 15:1453034. [PMID: 39263573 PMCID: PMC11387167 DOI: 10.3389/fphar.2024.1453034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2024] [Accepted: 08/12/2024] [Indexed: 09/13/2024] Open
Abstract
Cyclosporine is a potent immunosuppressive drug for various immune-mediated diseases in children. Cyclosporine's expected therapeutic effect also carries a wide range of side effects. One of the most common and intriguing dermatological side effects is hypertrichosis. However, recent reports have recognized alopecia as a potential adverse effect of cyclosporine. Here, we report a case of a 29-month-old boy diagnosed with aplastic anemia. During cyclosporine therapy, the patient presented with hair loss on the scalp, which and subsequently spread to the eyebrows and eyelashes. The alopecic symptoms were not relieved following topical minoxidil liniment interventions. When the cyclosporine was discontinued, a remarkable improvement was observed in the scalp, with complete hair regrowth. Data concerning cyclosporine from the FDA Adverse Event Reporting System (FAERS) database were extracted from January 2004 to January 2023. Within FAERS, our post-marketing pharmacovigilance analysis detected the reporting association of cyclosporine and alopecia. In monotherapy, cyclosporine-induced alopecia was observed in 118 cases, and tacrolimus-induced alopecia signals were detected in 197 cases. Although the potential mechanism of medication-induced hair loss is unclear, we identified a potential correlation between alopecia and cyclosporine, and it is still necessary to adequately recognize and clinically monitor this paradoxical reaction.
Collapse
Affiliation(s)
- Ying Wang
- Department of Pharmacy, the Second Xiangya Hospital of Central South University, Changsha, China
- Institute of Clinical Pharmacy, Central South University, Changsha, China
| | - Youhong Wang
- Department of Pharmacy, Xiamen Children's Hospital (Children's Hospital of Fudan University at Xiamen), Xiamen, China
- Fujian Key Laboratory of Neonatal Diseases, Xiamen Key Laboratory of Neonatal Diseases, Xiamen Children's Hospital (Children's Hospital of Fudan University at Xiamen), Xiamen, China
| | - Ping Xu
- Department of Pharmacy, the Second Xiangya Hospital of Central South University, Changsha, China
- Institute of Clinical Pharmacy, Central South University, Changsha, China
| |
Collapse
|
9
|
Dermitzakis I, Kampitsi DD, Manthou ME, Evangelidis P, Vakirlis E, Meditskou S, Theotokis P. Ontogeny of Skin Stem Cells and Molecular Underpinnings. Curr Issues Mol Biol 2024; 46:8118-8147. [PMID: 39194698 DOI: 10.3390/cimb46080481] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Revised: 07/21/2024] [Accepted: 07/25/2024] [Indexed: 08/29/2024] Open
Abstract
Skin stem cells (SCs) play a pivotal role in supporting tissue homeostasis. Several types of SCs are responsible for maintaining and regenerating skin tissue. These include bulge SCs and others residing in the interfollicular epidermis, infundibulum, isthmus, sebaceous glands, and sweat glands. The emergence of skin SCs commences during embryogenesis, where multipotent SCs arise from various precursor populations. These early events set the foundation for the diverse pool of SCs that will reside in the adult skin, ready to respond to tissue repair and regeneration demands. A network of molecular cues regulates skin SC behavior, balancing quiescence, self-renewal, and differentiation. The disruption of this delicate equilibrium can lead to SC exhaustion, impaired wound healing, and pathological conditions such as skin cancer. The present review explores the intricate mechanisms governing the development, activation, and differentiation of skin SCs, shedding light on the molecular signaling pathways that drive their fate decisions and skin homeostasis. Unraveling the complexities of these molecular drivers not only enhances our fundamental knowledge of skin biology but also holds promise for developing novel strategies to modulate skin SC fate for regenerative medicine applications, ultimately benefiting patients with skin disorders and injuries.
Collapse
Affiliation(s)
- Iasonas Dermitzakis
- Department of Histology-Embryology, School of Medicine, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece
| | - Despoina Dimitria Kampitsi
- Department of Histology-Embryology, School of Medicine, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece
| | - Maria Eleni Manthou
- Department of Histology-Embryology, School of Medicine, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece
| | - Paschalis Evangelidis
- Hematology Unit-Hemophilia Centre, 2nd Propedeutic Department of Internal Medicine, Hippocration Hospital, Aristotle University of Thessaloniki, 54642 Thessaloniki, Greece
| | - Efstratios Vakirlis
- First Department of Dermatology and Venereology, School of Medicine, Aristotle University of Thessaloniki, 54643 Thessaloniki, Greece
| | - Soultana Meditskou
- Department of Histology-Embryology, School of Medicine, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece
| | - Paschalis Theotokis
- Department of Histology-Embryology, School of Medicine, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece
| |
Collapse
|
10
|
Roy S, Mehta D, Paradkar A, Chovatiya G, Waghmare SK. Dab2 (Disabled-2), an adaptor protein, regulates self-renewal of hair follicle stem cells. Commun Biol 2024; 7:525. [PMID: 38702433 PMCID: PMC11068889 DOI: 10.1038/s42003-024-06047-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Accepted: 03/13/2024] [Indexed: 05/06/2024] Open
Abstract
Disabled 2 (Dab2), an adaptor protein, is up regulated in the hair follicle stem cells (HFSCs); however, its role in any tissue stem cells has not been studied. In the present study, we have reported that Dab2 conditional knockout (Dab2-cKO) mice exhibited a delay in the HF cycle due to perturbed activation of HFSCs. Further, Dab2-cKO mice showed a reduction in the number of HFSCs and reduced colony forming ability of HFSCs. Dab2-cKO mice showed extended quiescence of HFSCs concomitant with an increased expression of Nfatc1. Dab2-cKO mice showed a decreased expression of anti-aging genes such as Col17a1, decorin, Sirt2 and Sirt7. Dab2-cKO mice did not show full hair coat recovery in aged mice thereby suggesting an accelerated aging process. Overall, we unveil for the first time, the role of Dab2 that regulate activation and self-renewal of HFSCs.
Collapse
Affiliation(s)
- Sayoni Roy
- Stem Cell Biology Group, Waghmare Lab, Cancer Research Institute, Advanced Centre for Treatment Research and Education in Cancer (ACTREC), Tata Memorial Centre, Kharghar, Navi Mumbai, 410210, Maharashtra, India
- Homi Bhabha National Institute, Training School Complex, Anushakti Nagar, Mumbai, 400085, India
| | - Darshan Mehta
- Stem Cell Biology Group, Waghmare Lab, Cancer Research Institute, Advanced Centre for Treatment Research and Education in Cancer (ACTREC), Tata Memorial Centre, Kharghar, Navi Mumbai, 410210, Maharashtra, India
- Homi Bhabha National Institute, Training School Complex, Anushakti Nagar, Mumbai, 400085, India
| | - Akshay Paradkar
- Stem Cell Biology Group, Waghmare Lab, Cancer Research Institute, Advanced Centre for Treatment Research and Education in Cancer (ACTREC), Tata Memorial Centre, Kharghar, Navi Mumbai, 410210, Maharashtra, India
- Homi Bhabha National Institute, Training School Complex, Anushakti Nagar, Mumbai, 400085, India
| | - Gopal Chovatiya
- Stem Cell Biology Group, Waghmare Lab, Cancer Research Institute, Advanced Centre for Treatment Research and Education in Cancer (ACTREC), Tata Memorial Centre, Kharghar, Navi Mumbai, 410210, Maharashtra, India
- Homi Bhabha National Institute, Training School Complex, Anushakti Nagar, Mumbai, 400085, India
| | - Sanjeev K Waghmare
- Stem Cell Biology Group, Waghmare Lab, Cancer Research Institute, Advanced Centre for Treatment Research and Education in Cancer (ACTREC), Tata Memorial Centre, Kharghar, Navi Mumbai, 410210, Maharashtra, India.
- Homi Bhabha National Institute, Training School Complex, Anushakti Nagar, Mumbai, 400085, India.
| |
Collapse
|
11
|
Tierney MT, Polak L, Yang Y, Abdusselamoglu MD, Baek I, Stewart KS, Fuchs E. Vitamin A resolves lineage plasticity to orchestrate stem cell lineage choices. Science 2024; 383:eadi7342. [PMID: 38452090 PMCID: PMC11177320 DOI: 10.1126/science.adi7342] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Accepted: 02/07/2024] [Indexed: 03/09/2024]
Abstract
Lineage plasticity-a state of dual fate expression-is required to release stem cells from their niche constraints and redirect them to tissue compartments where they are most needed. In this work, we found that without resolving lineage plasticity, skin stem cells cannot effectively generate each lineage in vitro nor regrow hair and repair wounded epidermis in vivo. A small-molecule screen unearthed retinoic acid as a critical regulator. Combining high-throughput approaches, cell culture, and in vivo mouse genetics, we dissected its roles in tissue regeneration. We found that retinoic acid is made locally in hair follicle stem cell niches, where its levels determine identity and usage. Our findings have therapeutic implications for hair growth as well as chronic wounds and cancers, where lineage plasticity is unresolved.
Collapse
Affiliation(s)
- Matthew T Tierney
- Howard Hughes Medical Institute, Robin Chemers Neustein Laboratory of Mammalian Cell Biology and Development, The Rockefeller University; New York, NY 10065, USA
| | - Lisa Polak
- Howard Hughes Medical Institute, Robin Chemers Neustein Laboratory of Mammalian Cell Biology and Development, The Rockefeller University; New York, NY 10065, USA
| | | | - Merve Deniz Abdusselamoglu
- Howard Hughes Medical Institute, Robin Chemers Neustein Laboratory of Mammalian Cell Biology and Development, The Rockefeller University; New York, NY 10065, USA
| | | | - Katherine S Stewart
- Howard Hughes Medical Institute, Robin Chemers Neustein Laboratory of Mammalian Cell Biology and Development, The Rockefeller University; New York, NY 10065, USA
| | - Elaine Fuchs
- Howard Hughes Medical Institute, Robin Chemers Neustein Laboratory of Mammalian Cell Biology and Development, The Rockefeller University; New York, NY 10065, USA
| |
Collapse
|
12
|
Xing YZ, Guo HY, Xiang F, Li YH. Recent progress in hair follicle stem cell markers and their regulatory roles. World J Stem Cells 2024; 16:126-136. [PMID: 38455104 PMCID: PMC10915958 DOI: 10.4252/wjsc.v16.i2.126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Revised: 12/19/2023] [Accepted: 01/16/2024] [Indexed: 02/26/2024] Open
Abstract
Hair follicle stem cells (HFSCs) in the bulge are a multipotent adult stem cell population. They can periodically give rise to new HFs and even regenerate the epidermis and sebaceous glands during wound healing. An increasing number of biomarkers have been used to isolate, label, and trace HFSCs in recent years. Considering more detailed data from single-cell transcriptomics technology, we mainly focus on the important HFSC molecular markers and their regulatory roles in this review.
Collapse
Affiliation(s)
- Yi-Zhan Xing
- Department of Cell Biology, Army Medical University, Chongqing 400038, China
| | - Hai-Ying Guo
- Department of Cell Biology, Army Medical University, Chongqing 400038, China
| | - Fei Xiang
- Institute of Burn Research, Southwest Hospital, Army Medical University, Chongqing 400038, China
| | - Yu-Hong Li
- Department of Cell Biology, Army Medical University, Chongqing 400038, China.
| |
Collapse
|
13
|
Lee HC, Oliveira NMM, Hastings C, Baillie-Benson P, Moverley AA, Lu HC, Zheng Y, Wilby EL, Weil TT, Page KM, Fu J, Moris N, Stern CD. Regulation of long-range BMP gradients and embryonic polarity by propagation of local calcium-firing activity. Nat Commun 2024; 15:1463. [PMID: 38368410 PMCID: PMC10874436 DOI: 10.1038/s41467-024-45772-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Accepted: 02/02/2024] [Indexed: 02/19/2024] Open
Abstract
Many amniote vertebrate species including humans can form identical twins from a single embryo, but this only occurs rarely. It has been suggested that the primitive-streak-forming embryonic region emits signals that inhibit streak formation elsewhere but the signals involved, how they are transmitted and how they act has not been elucidated. Here we show that short tracks of calcium firing activity propagate through extraembryonic tissue via gap junctions and prevent ectopic primitive streak formation in chick embryos. Cross-regulation of calcium activity and an inhibitor of primitive streak formation (Bone Morphogenetic Protein, BMP) via NF-κB and NFAT establishes a long-range BMP gradient spanning the embryo. This mechanism explains how embryos of widely different sizes can maintain positional information that determines embryo polarity. We provide evidence for similar mechanisms in two different human embryo models and in Drosophila, suggesting an ancient evolutionary origin.
Collapse
Affiliation(s)
- Hyung Chul Lee
- Department of Cell and Developmental Biology, University College London, Gower Street, London, WC1E 6BT, UK.
- School of Biological Sciences and Technology, College of Natural Sciences, Chonnam National University, 77 Yongbong-ro, Gwangju, 61186, Korea.
| | - Nidia M M Oliveira
- Department of Cell and Developmental Biology, University College London, Gower Street, London, WC1E 6BT, UK
- College of Professional Services, Murdoch University, 90 South St, Murdoch, WA, 6150, Australia
| | - Cato Hastings
- Department of Cell and Developmental Biology, University College London, Gower Street, London, WC1E 6BT, UK
| | | | - Adam A Moverley
- Department of Cell and Developmental Biology, University College London, Gower Street, London, WC1E 6BT, UK
- Department of Cell and Developmental Biology, Institute for Regenerative Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Hui-Chun Lu
- Department of Cell and Developmental Biology, University College London, Gower Street, London, WC1E 6BT, UK
- Centre for Craniofacial & Regenerative Biology, Faculty of Dentistry, Oral and Craniofacial Sciences, King's College London, Guy's Tower, London, SE1 9RT, UK
| | - Yi Zheng
- Departments of Mechanical Engineering, Biomedical Engineering, and Cell & Developmental Biology, University of Michigan, Ann Arbor, MI, USA
- Department of Biomedical and Chemical Engineering, Syracuse University, Syracuse, NY, USA
- BioInspired Syracuse Institute for Materials and Living Systems, Syracuse University, Syracuse, NY, USA
| | - Elise L Wilby
- Department of Zoology, University of Cambridge, Downing Street, Cambridge, CB2 3EJ, UK
| | - Timothy T Weil
- Department of Zoology, University of Cambridge, Downing Street, Cambridge, CB2 3EJ, UK
| | - Karen M Page
- Department of Mathematics, University College London, Gower Street, London, WC1E 6BT, UK
| | - Jianping Fu
- Departments of Mechanical Engineering, Biomedical Engineering, and Cell & Developmental Biology, University of Michigan, Ann Arbor, MI, USA
| | - Naomi Moris
- The Francis Crick Institute, 1 Midland Road, London, NW1 1AT, UK
| | - Claudio D Stern
- Department of Cell and Developmental Biology, University College London, Gower Street, London, WC1E 6BT, UK.
| |
Collapse
|
14
|
Zhang B, Chen T. Local and systemic mechanisms that control the hair follicle stem cell niche. Nat Rev Mol Cell Biol 2024; 25:87-100. [PMID: 37903969 DOI: 10.1038/s41580-023-00662-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/25/2023] [Indexed: 11/01/2023]
Abstract
Hair follicles are essential appendages of the mammalian skin, as hair performs vital functions of protection, thermoregulation and sensation. Hair follicles harbour exceptional regenerative abilities as they contain multiple somatic stem cell populations such as hair follicle stem cells (HFSCs) and melanocyte stem cells. Surrounding the stem cells and their progeny, diverse groups of cells and extracellular matrix proteins are organized to form a microenvironment (called 'niche') that serves to promote and maintain the optimal functioning of these stem cell populations. Recent studies have shed light on the intricate nature of the HFSC niche and its crucial role in regulating hair follicle regeneration. In this Review, we describe how the niche serves as a signalling hub, communicating, deciphering and integrating both local signals within the skin and systemic inputs from the body and environment to modulate HFSC activity. We delve into the recent advancements in identifying the cellular and molecular nature of the niche, providing a holistic perspective on its essential functions in hair follicle morphogenesis, regeneration and ageing.
Collapse
Affiliation(s)
- Bing Zhang
- School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China.
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China.
| | - Ting Chen
- National Institute of Biological Sciences, Beijing, China.
- Tsinghua Institute of Multidisciplinary Biomedical Research, Tsinghua University, Beijing, China.
| |
Collapse
|
15
|
Lee JH, Choi S. Deciphering the molecular mechanisms of stem cell dynamics in hair follicle regeneration. Exp Mol Med 2024; 56:110-117. [PMID: 38182654 PMCID: PMC10834421 DOI: 10.1038/s12276-023-01151-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Revised: 10/24/2023] [Accepted: 11/01/2023] [Indexed: 01/07/2024] Open
Abstract
Hair follicles, which are connected to sebaceous glands in the skin, undergo cyclic periods of regeneration, degeneration, and rest throughout adult life in mammals. The crucial function of hair follicle stem cells is to maintain these hair growth cycles. Another vital aspect is the activity of melanocyte stem cells, which differentiate into melanin-producing melanocytes, contributing to skin and hair pigmentation. Sebaceous gland stem cells also have a pivotal role in maintaining the skin barrier by regenerating mature sebocytes. These stem cells are maintained in a specialized microenvironment or niche and are regulated by internal and external signals, determining their dynamic behaviors in homeostasis and hair follicle regeneration. The activity of these stem cells is tightly controlled by various factors secreted by the niche components around the hair follicles, as well as immune-mediated damage signals, aging, metabolic status, and stress. In this study, we review these diverse stem cell regulatory and related molecular mechanisms of hair regeneration and disease conditions. Molecular insights would provide new perspectives on the disease mechanisms as well as hair and skin disorder treatment.
Collapse
Affiliation(s)
- Jung Hyun Lee
- Department of Dermatology, School of Medicine, University of Washington, Seattle, WA, 98109, USA
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA, 98109, USA
| | - Sekyu Choi
- Department of Life Sciences, Pohang University of Science and Technology (POSTECH), Pohang, 37673, Republic of Korea.
- Medical Science and Engineering, School of Convergence Science and Technology, Pohang University of Science and Technology (POSTECH), Pohang, 37673, Republic of Korea.
- School of Interdisciplinary Bioscience and Bioengineering, Pohang University of Science and Technology (POSTECH), Pohang, 37673, Republic of Korea.
- Institute for Convergence Research and Education in Advanced Technology (I_CREATE), Yonsei University, Incheon, 21983, Republic of Korea.
| |
Collapse
|
16
|
Xing Y, Xiang F, Guo H, Gong H, Li Y. Reversibly immortalization establishes a hair follicle stem cell line with hair follicle reconstruction ability. Exp Dermatol 2024; 33:e14999. [PMID: 38284187 DOI: 10.1111/exd.14999] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Revised: 11/01/2023] [Accepted: 12/13/2023] [Indexed: 01/30/2024]
Abstract
Hair follicle stem cells (HFSCs) play critical roles in the periodic regeneration of hair follicles. HFSCs are also a good model for stem cell biology research. However, no stable mouse HFSC cell line has been reported, which restricts the research and application of HFSCs. We isolated HFSCs from mouse hair follicles and immortalized them by inducing a reversible SV40 large T antigen. Through monoclonal screening, we identified a reversibly immortalized cell line, immortalized HFSC (iHFSC2). RNA sequencing, fluorescence-activated cell sorting, western blotting and immunofluorescence experiments revealed that the expression patterns of iHFSC2 and HFSC were similar at the protein and mRNA levels. After that, iHFSC2s were passaged and morphologically monitored for up to 40 times to detect their long-term culture potential. The long-term cultured iHFSC2 could regenerate hair follicles with complete hair follicle structure and HFSCs in the bulge area. This work successfully established an HFSC cell line with the ability of hair follicle reconstruction.
Collapse
Affiliation(s)
- Yizhan Xing
- Department of Cell Biology, Army Medical University, Chongqing, PR China
| | - Fei Xiang
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Burn Research, Southwest Hospital, Army Medical University, Chongqing, PR China
| | - Haiying Guo
- Department of Cell Biology, Army Medical University, Chongqing, PR China
| | - Hao Gong
- Department of Cell Biology, Army Medical University, Chongqing, PR China
| | - Yuhong Li
- Department of Cell Biology, Army Medical University, Chongqing, PR China
| |
Collapse
|
17
|
Hariton WV, Schulze K, Rahimi S, Shojaeian T, Feldmeyer L, Schwob R, Overmiller AM, Sayar BS, Borradori L, Mahoney MG, Galichet A, Müller EJ. A desmosomal cadherin controls multipotent hair follicle stem cell quiescence and orchestrates regeneration through adhesion signaling. iScience 2023; 26:108568. [PMID: 38162019 PMCID: PMC10755723 DOI: 10.1016/j.isci.2023.108568] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Revised: 11/03/2023] [Accepted: 11/21/2023] [Indexed: 01/03/2024] Open
Abstract
Stem cells (SCs) are critical to maintain tissue homeostasis. However, it is currently not known whether signaling through cell junctions protects quiescent epithelial SC reservoirs from depletion during disease-inflicted damage. Using the autoimmune model disease pemphigus vulgaris (PV), this study reveals an unprecedented role for a desmosomal cadherin in governing SC quiescence and regeneration through adhesion signaling in the multipotent mouse hair follicle compartment known as the bulge. Autoantibody-mediated, mechanical uncoupling of desmoglein (Dsg) 3 transadhesion activates quiescent bulge SC which lose their multipotency and stemness, become actively cycling, and finally delaminate from their epithelial niche. This then initiates a self-organized regenerative program which restores Dsg3 function and bulge morphology including SC quiescence and multipotency. These profound changes are triggered by the sole loss of functional Dsg3, resemble major signaling events in Dsg3-/- mice, and are driven by SC-relevant EGFR activation and Wnt modulation requiring longitudinal repression of Hedgehog signaling.
Collapse
Affiliation(s)
- William V.J. Hariton
- Department of Dermatology, Inselspital, Bern University Hospital, University of Bern, 3010 Bern, Switzerland
- Department for BioMedical Research, Molecular Dermatology and Stem Cell Research, University of Bern, 3008 Bern, Switzerland
- DermFocus, Vetsuisse Faculty, University of Bern, 3008 Bern, Switzerland
- Institute of Animal Pathology, Vetsuisse Faculty, University of Bern, 3012 Bern, Switzerland
| | - Katja Schulze
- DermFocus, Vetsuisse Faculty, University of Bern, 3008 Bern, Switzerland
- Institute of Animal Pathology, Vetsuisse Faculty, University of Bern, 3012 Bern, Switzerland
| | - Siavash Rahimi
- Department of Dermatology, Inselspital, Bern University Hospital, University of Bern, 3010 Bern, Switzerland
- Department for BioMedical Research, Molecular Dermatology and Stem Cell Research, University of Bern, 3008 Bern, Switzerland
- DermFocus, Vetsuisse Faculty, University of Bern, 3008 Bern, Switzerland
| | - Taravat Shojaeian
- Department of Dermatology, Inselspital, Bern University Hospital, University of Bern, 3010 Bern, Switzerland
- Department for BioMedical Research, Molecular Dermatology and Stem Cell Research, University of Bern, 3008 Bern, Switzerland
- DermFocus, Vetsuisse Faculty, University of Bern, 3008 Bern, Switzerland
| | - Laurence Feldmeyer
- Department of Dermatology, Inselspital, Bern University Hospital, University of Bern, 3010 Bern, Switzerland
| | - Roman Schwob
- DermFocus, Vetsuisse Faculty, University of Bern, 3008 Bern, Switzerland
- Institute of Animal Pathology, Vetsuisse Faculty, University of Bern, 3012 Bern, Switzerland
| | - Andrew M. Overmiller
- Department of Dermatology and Cutaneous Biology, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Beyza S. Sayar
- Department of Dermatology, Inselspital, Bern University Hospital, University of Bern, 3010 Bern, Switzerland
- Department for BioMedical Research, Molecular Dermatology and Stem Cell Research, University of Bern, 3008 Bern, Switzerland
- DermFocus, Vetsuisse Faculty, University of Bern, 3008 Bern, Switzerland
| | - Luca Borradori
- Department of Dermatology, Inselspital, Bern University Hospital, University of Bern, 3010 Bern, Switzerland
- DermFocus, Vetsuisse Faculty, University of Bern, 3008 Bern, Switzerland
| | - Mỹ G. Mahoney
- Department of Dermatology and Cutaneous Biology, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Arnaud Galichet
- Department of Dermatology, Inselspital, Bern University Hospital, University of Bern, 3010 Bern, Switzerland
- Department for BioMedical Research, Molecular Dermatology and Stem Cell Research, University of Bern, 3008 Bern, Switzerland
- DermFocus, Vetsuisse Faculty, University of Bern, 3008 Bern, Switzerland
- Institute of Animal Pathology, Vetsuisse Faculty, University of Bern, 3012 Bern, Switzerland
| | - Eliane J. Müller
- Department of Dermatology, Inselspital, Bern University Hospital, University of Bern, 3010 Bern, Switzerland
- Department for BioMedical Research, Molecular Dermatology and Stem Cell Research, University of Bern, 3008 Bern, Switzerland
- DermFocus, Vetsuisse Faculty, University of Bern, 3008 Bern, Switzerland
- Institute of Animal Pathology, Vetsuisse Faculty, University of Bern, 3012 Bern, Switzerland
| |
Collapse
|
18
|
Abstract
Diseases affecting the hair follicle are common in domestic animals, but despite the importance of an intact skin barrier and a fully functional hair coat, knowledge about the detailed morphological features and the diversity of these complex mini-organs are often limited, although mandatory to evaluate skin biopsies with a history of alopecia. The factors that regulate the innate hair follicle formation and the postnatal hair cycle are still not completely understood in rodents, only rudimentarily known in humans, and are poorly understood in our companion animals. This review aims to summarize the current knowledge about hair follicle and hair shaft anatomy, the arrangement of hair follicles, hair follicle morphogenesis in the embryo, and the lifelong regeneration during the postnatal hair cycle in domestic animals. The role of follicular stem cells and the need for a multitude of interacting signaling events during hair follicle morphogenesis and regeneration is unquestioned. Because of the lack of state of the art methods that can be applied in rodents but are not feasible in companion animals, most of the information in this review is based on rodent studies. However, the few data from domestic animals that are available will be discussed, and it can be assumed that at least the principal molecular mechanisms are similar in rodents and other species.
Collapse
|
19
|
Sharma A, Mohapatra H, Arora K, Babbar R, Arora R, Arora P, Kumar P, Algın Yapar E, Rani K, Meenu M, Babu MA, Kaur M, Sindhu RK. Bioactive Compound-Loaded Nanocarriers for Hair Growth Promotion: Current Status and Future Perspectives. PLANTS (BASEL, SWITZERLAND) 2023; 12:3739. [PMID: 37960095 PMCID: PMC10649697 DOI: 10.3390/plants12213739] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Revised: 10/01/2023] [Accepted: 10/21/2023] [Indexed: 11/15/2023]
Abstract
Hair loss (alopecia) has a multitude of causes, and the problem is still poorly defined. For curing alopecia, therapies are available in both natural and synthetic forms; however, natural remedies are gaining popularity due to the multiple effects of complex phytoconstituents on the scalp with fewer side effects. Evidence-based hair growth promotion by some plants has been reported for both traditional and advanced treatment approaches. Nanoarchitectonics may have the ability to evolve in the field of hair- and scalp-altering products and treatments, giving new qualities to hair that can be an effective protective layer or a technique to recover lost hair. This review will provide insights into several plant and herbal formulations that have been reported for the prevention of hair loss and stimulation of new hair growth. This review also focuses on the molecular mechanisms of hair growth/loss, several isolated phytoconstituents with hair growth-promoting properties, patents, in vivo evaluation of hair growth-promoting activity, and recent nanoarchitectonic technologies that have been explored for hair growth.
Collapse
Affiliation(s)
- Arvind Sharma
- School of Pharmaceutical and Health Sciences, Bhoranj (Tikker–Kharwarian), Hamirpur 176041, India;
| | - Harapriya Mohapatra
- Chitkara College of Pharmacy, Chitkara University, Rajpura 140401, India; (H.M.); (K.A.); (R.B.); (R.A.); (P.A.); (K.R.)
| | - Kanika Arora
- Chitkara College of Pharmacy, Chitkara University, Rajpura 140401, India; (H.M.); (K.A.); (R.B.); (R.A.); (P.A.); (K.R.)
| | - Ritchu Babbar
- Chitkara College of Pharmacy, Chitkara University, Rajpura 140401, India; (H.M.); (K.A.); (R.B.); (R.A.); (P.A.); (K.R.)
| | - Rashmi Arora
- Chitkara College of Pharmacy, Chitkara University, Rajpura 140401, India; (H.M.); (K.A.); (R.B.); (R.A.); (P.A.); (K.R.)
| | - Poonam Arora
- Chitkara College of Pharmacy, Chitkara University, Rajpura 140401, India; (H.M.); (K.A.); (R.B.); (R.A.); (P.A.); (K.R.)
| | - Pradeep Kumar
- Wits Advanced Drug Delivery Platform Research Unit, Department of Pharmacy and Pharmacology, School of Therapeutic Sciences, Faculty of Health Sciences, 7 York Road, Parktown, Johannesburg 2193, South Africa;
| | - Evren Algın Yapar
- Faculty of Pharmacy, Sivas Cumhuriyet University, Sivas 58140, Türkiye;
| | - Kailash Rani
- Chitkara College of Pharmacy, Chitkara University, Rajpura 140401, India; (H.M.); (K.A.); (R.B.); (R.A.); (P.A.); (K.R.)
| | - Maninder Meenu
- Department of Agri-Biotechnology, National Agri-Food Biotechnology Institute, Mohali 143005, India;
| | | | - Maninderjit Kaur
- Department of Pharmaceutical Sciences, Lovely Professional University, Phagwara 144411, India;
| | - Rakesh K. Sindhu
- School of Pharmacy, Sharda University, Greater Noida 201306, India
| |
Collapse
|
20
|
Kuburich NA, Sabapathy T, Demestichas BR, Maddela JJ, den Hollander P, Mani SA. Proactive and reactive roles of TGF-β in cancer. Semin Cancer Biol 2023; 95:120-139. [PMID: 37572731 PMCID: PMC10530624 DOI: 10.1016/j.semcancer.2023.08.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 08/04/2023] [Accepted: 08/05/2023] [Indexed: 08/14/2023]
Abstract
Cancer cells adapt to varying stress conditions to survive through plasticity. Stem cells exhibit a high degree of plasticity, allowing them to generate more stem cells or differentiate them into specialized cell types to contribute to tissue development, growth, and repair. Cancer cells can also exhibit plasticity and acquire properties that enhance their survival. TGF-β is an unrivaled growth factor exploited by cancer cells to gain plasticity. TGF-β-mediated signaling enables carcinoma cells to alter their epithelial and mesenchymal properties through epithelial-mesenchymal plasticity (EMP). However, TGF-β is a multifunctional cytokine; thus, the signaling by TGF-β can be detrimental or beneficial to cancer cells depending on the cellular context. Those cells that overcome the anti-tumor effect of TGF-β can induce epithelial-mesenchymal transition (EMT) to gain EMP benefits. EMP allows cancer cells to alter their cell properties and the tumor immune microenvironment (TIME), facilitating their survival. Due to the significant roles of TGF-β and EMP in carcinoma progression, it is essential to understand how TGF-β enables EMP and how cancer cells exploit this plasticity. This understanding will guide the development of effective TGF-β-targeting therapies that eliminate cancer cell plasticity.
Collapse
Affiliation(s)
- Nick A Kuburich
- Legorreta Cancer Center, The Warren Alpert Medical School, Brown University, Providence, RI 02912, USA; Department of Pathology and Lab Medicine, The Warren Alpert Medical School, Brown University, Providence, RI 02912, USA
| | - Thiru Sabapathy
- Legorreta Cancer Center, The Warren Alpert Medical School, Brown University, Providence, RI 02912, USA; Department of Pathology and Lab Medicine, The Warren Alpert Medical School, Brown University, Providence, RI 02912, USA
| | - Breanna R Demestichas
- Legorreta Cancer Center, The Warren Alpert Medical School, Brown University, Providence, RI 02912, USA; Department of Pathology and Lab Medicine, The Warren Alpert Medical School, Brown University, Providence, RI 02912, USA
| | - Joanna Joyce Maddela
- Legorreta Cancer Center, The Warren Alpert Medical School, Brown University, Providence, RI 02912, USA; Department of Pathology and Lab Medicine, The Warren Alpert Medical School, Brown University, Providence, RI 02912, USA
| | - Petra den Hollander
- Legorreta Cancer Center, The Warren Alpert Medical School, Brown University, Providence, RI 02912, USA; Department of Pathology and Lab Medicine, The Warren Alpert Medical School, Brown University, Providence, RI 02912, USA
| | - Sendurai A Mani
- Legorreta Cancer Center, The Warren Alpert Medical School, Brown University, Providence, RI 02912, USA; Department of Pathology and Lab Medicine, The Warren Alpert Medical School, Brown University, Providence, RI 02912, USA.
| |
Collapse
|
21
|
Rönkkö J, Rodriguez Y, Rasila T, Torregrosa-Muñumer R, Pennonen J, Kvist J, Kuuluvainen E, Bosch LVD, Hietakangas V, Bultynck G, Tyynismaa H, Ylikallio E. Human IP 3 receptor triple knockout stem cells remain pluripotent despite altered mitochondrial metabolism. Cell Calcium 2023; 114:102782. [PMID: 37481871 DOI: 10.1016/j.ceca.2023.102782] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Revised: 06/14/2023] [Accepted: 07/13/2023] [Indexed: 07/25/2023]
Abstract
Inositol 1,4,5-trisphosphate receptors (IP3Rs) are ER Ca2+-release channels that control a broad set of cellular processes. Animal models lacking IP3Rs in different combinations display severe developmental phenotypes. Given the importance of IP3Rs in human diseases, we investigated their role in human induced pluripotent stem cells (hiPSC) by developing single IP3R and triple IP3R knockouts (TKO). Genome edited TKO-hiPSC lacking all three IP3R isoforms, IP3R1, IP3R2, IP3R3, failed to generate Ca2+ signals in response to agonists activating GPCRs, but retained stemness and pluripotency. Steady state metabolite profiling and flux analysis of TKO-hiPSC indicated distinct alterations in tricarboxylic acid cycle metabolites consistent with a deficiency in their pyruvate utilization via pyruvate dehydrogenase, shifting towards pyruvate carboxylase pathway. These results demonstrate that IP3Rs are not essential for hiPSC identity and pluripotency but regulate mitochondrial metabolism. This set of knockout hiPSC is a valuable resource for investigating IP3Rs in human cell types of interest.
Collapse
Affiliation(s)
- Julius Rönkkö
- Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, Helsinki, 00290, Finland
| | - Yago Rodriguez
- Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, Helsinki, 00290, Finland
| | - Tiina Rasila
- Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, Helsinki, 00290, Finland
| | - Rubén Torregrosa-Muñumer
- Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, Helsinki, 00290, Finland
| | - Jana Pennonen
- Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, Helsinki, 00290, Finland
| | - Jouni Kvist
- Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, Helsinki, 00290, Finland
| | - Emilia Kuuluvainen
- Molecular and Integrative Bioscience Research Programme, Faculty of Biological and Environmental Sciences, University of Helsinki, Helsinki, 00790, Finland; Institute of Biotechnology, HiLIFE, University of Helsinki, Helsinki, 00790, Finland
| | - Ludo Van Den Bosch
- Department of Neurosciences, Experimental Neurology and Leuven Brain Institute, KU Leuven - University of Leuven, 3000, Leuven, Belgium; VIB Center for Brain & Disease Research, Laboratory of Neurobiology, 3000, Leuven, Belgium
| | - Ville Hietakangas
- Molecular and Integrative Bioscience Research Programme, Faculty of Biological and Environmental Sciences, University of Helsinki, Helsinki, 00790, Finland; Institute of Biotechnology, HiLIFE, University of Helsinki, Helsinki, 00790, Finland
| | - Geert Bultynck
- KU Leuven, Laboratory of Molecular and Cellular Signaling, Department of Cellular and Molecular Medicine & Leuven Kanker Instituut, Leuven, 3000, Belgium
| | - Henna Tyynismaa
- Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, Helsinki, 00290, Finland
| | - Emil Ylikallio
- Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, Helsinki, 00290, Finland; Clinical Neurosciences, Neurology, University of Helsinki and Helsinki University Hospital, Helsinki, 00290, Finland.
| |
Collapse
|
22
|
Sawaya AP, Vecin NM, Burgess JL, Ojeh N, DiBartolomeo G, Stone RC, Pastar I, Tomic-Canic M. Calreticulin: a multifunctional protein with potential therapeutic applications for chronic wounds. Front Med (Lausanne) 2023; 10:1207538. [PMID: 37692787 PMCID: PMC10484228 DOI: 10.3389/fmed.2023.1207538] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Accepted: 08/07/2023] [Indexed: 09/12/2023] Open
Abstract
Calreticulin is recognized as a multifunctional protein that serves an essential role in diverse biological processes that include wound healing, modification and folding of proteins, regulation of the secretory pathway, cell motility, cellular metabolism, protein synthesis, regulation of gene expression, cell cycle regulation and apoptosis. Although the role of calreticulin as an endoplasmic reticulum-chaperone protein has been well described, several studies have demonstrated calreticulin to be a highly versatile protein with an essential role during wound healing. These features make it an ideal molecule for treating a complex, multifactorial diseases that require fine tuning, such as chronic wounds. Indeed, topical application of recombinant calreticulin to wounds in multiple models of wound healing has demonstrated remarkable pro-healing effects. Among them include enhanced keratinocyte and fibroblast migration and proliferation, induction of extracellular matrix proteins, recruitment of macrophages along with increased granulation tissue formation, all of which are important functions in promoting wound healing that are deregulated in chronic wounds. Given the high degree of diverse functions and pro-healing effects, application of exogenous calreticulin warrants further investigation as a potential novel therapeutic option for chronic wound patients. Here, we review and highlight the significant effects of topical application of calreticulin on enhancing wound healing and its potential as a novel therapeutic option to shift chronic wounds into healing, acute-like wounds.
Collapse
Affiliation(s)
- Andrew P. Sawaya
- Wound Healing and Regenerative Medicine Research Program, Dr Phillip Frost Department of Dermatology and Cutaneous Surgery, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Nicole M. Vecin
- Wound Healing and Regenerative Medicine Research Program, Dr Phillip Frost Department of Dermatology and Cutaneous Surgery, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Jamie L. Burgess
- Wound Healing and Regenerative Medicine Research Program, Dr Phillip Frost Department of Dermatology and Cutaneous Surgery, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Nkemcho Ojeh
- Wound Healing and Regenerative Medicine Research Program, Dr Phillip Frost Department of Dermatology and Cutaneous Surgery, University of Miami Miller School of Medicine, Miami, FL, United States
- Faculty of Medical Sciences, The University of the West Indies, Bridgetown, Barbados
| | - Gabrielle DiBartolomeo
- Wound Healing and Regenerative Medicine Research Program, Dr Phillip Frost Department of Dermatology and Cutaneous Surgery, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Rivka C. Stone
- Wound Healing and Regenerative Medicine Research Program, Dr Phillip Frost Department of Dermatology and Cutaneous Surgery, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Irena Pastar
- Wound Healing and Regenerative Medicine Research Program, Dr Phillip Frost Department of Dermatology and Cutaneous Surgery, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Marjana Tomic-Canic
- Wound Healing and Regenerative Medicine Research Program, Dr Phillip Frost Department of Dermatology and Cutaneous Surgery, University of Miami Miller School of Medicine, Miami, FL, United States
| |
Collapse
|
23
|
Ober-Reynolds B, Wang C, Ko JM, Rios EJ, Aasi SZ, Davis MM, Oro AE, Greenleaf WJ. Integrated single-cell chromatin and transcriptomic analyses of human scalp identify gene-regulatory programs and critical cell types for hair and skin diseases. Nat Genet 2023; 55:1288-1300. [PMID: 37500727 PMCID: PMC11190942 DOI: 10.1038/s41588-023-01445-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Accepted: 06/17/2023] [Indexed: 07/29/2023]
Abstract
Genome-wide association studies have identified many loci associated with hair and skin disease, but identification of causal variants requires deciphering of gene-regulatory networks in relevant cell types. We generated matched single-cell chromatin profiles and transcriptomes from scalp tissue from healthy controls and patients with alopecia areata, identifying diverse cell types of the hair follicle niche. By interrogating these datasets at multiple levels of cellular resolution, we infer 50-100% more enhancer-gene links than previous approaches and show that aggregate enhancer accessibility for highly regulated genes predicts expression. We use these gene-regulatory maps to prioritize cell types, genes and causal variants implicated in the pathobiology of androgenetic alopecia (AGA), eczema and other complex traits. AGA genome-wide association studies signals are enriched in dermal papilla regulatory regions, supporting the role of these cells as drivers of AGA pathogenesis. Finally, we train machine learning models to nominate single-nucleotide polymorphisms that affect gene expression through disruption of transcription factor binding, predicting candidate functional single-nucleotide polymorphism for AGA and eczema.
Collapse
Affiliation(s)
| | - Chen Wang
- Department of Dermatology, School of Medicine, Stanford University, Stanford, CA, USA
- Division of Dermatology, Department of Medicine, Santa Clara Valley Medical Center, San Jose, CA, USA
- Institute of Immunity, Transplantation and Infection, School of Medicine, Stanford University, Stanford, CA, USA
| | - Justin M Ko
- Department of Dermatology, School of Medicine, Stanford University, Stanford, CA, USA
| | - Eon J Rios
- Department of Dermatology, School of Medicine, Stanford University, Stanford, CA, USA
- Division of Dermatology, Department of Medicine, Santa Clara Valley Medical Center, San Jose, CA, USA
| | - Sumaira Z Aasi
- Department of Dermatology, School of Medicine, Stanford University, Stanford, CA, USA
| | - Mark M Davis
- Institute of Immunity, Transplantation and Infection, School of Medicine, Stanford University, Stanford, CA, USA
- Department of Microbiology and Immunology, School of Medicine, Stanford University, Stanford, CA, USA
- Howard Hughes Medical Institute, School of Medicine, Stanford University, Stanford, CA, USA
| | - Anthony E Oro
- Department of Dermatology, School of Medicine, Stanford University, Stanford, CA, USA
- Program in Epithelial Biology, Stanford University School of Medicine, Stanford, CA, USA
| | - William J Greenleaf
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA.
- Department of Applied Physics, Stanford University, Stanford, CA, USA.
- Chan Zuckerberg Biohub, San Francisco, CA, USA.
| |
Collapse
|
24
|
Zhao Q, Zheng Y, Zhao D, Zhao L, Geng L, Ma S, Cai Y, Liu C, Yan Y, Belmonte JCI, Wang S, Zhang W, Liu GH, Qu J. Single-cell profiling reveals a potent role of quercetin in promoting hair regeneration. Protein Cell 2023; 14:398-415. [PMID: 37285263 PMCID: PMC10246722 DOI: 10.1093/procel/pwac062] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2022] [Accepted: 10/16/2022] [Indexed: 07/21/2023] Open
Abstract
Hair loss affects millions of people at some time in their life, and safe and efficient treatments for hair loss are a significant unmet medical need. We report that topical delivery of quercetin (Que) stimulates resting hair follicles to grow with rapid follicular keratinocyte proliferation and replenishes perifollicular microvasculature in mice. We construct dynamic single-cell transcriptome landscape over the course of hair regrowth and find that Que treatment stimulates the differentiation trajectory in the hair follicles and induces an angiogenic signature in dermal endothelial cells by activating HIF-1α in endothelial cells. Skin administration of a HIF-1α agonist partially recapitulates the pro-angiogenesis and hair-growing effects of Que. Together, these findings provide a molecular understanding for the efficacy of Que in hair regrowth, which underscores the translational potential of targeting the hair follicle niche as a strategy for regenerative medicine, and suggest a route of pharmacological intervention that may promote hair regrowth.
Collapse
Affiliation(s)
| | | | | | - Liyun Zhao
- Advanced Innovation Center for Human Brain Protection and National Clinical Research Center for Geriatric Disorders, Xuanwu Hospital Capital Medical University, Beijing 100053, China
- Aging Translational Medicine Center, International Center for Aging and Cancer, Beijing Municipal Geriatric Medical Research Center, Xuan Wu Hospital, Capital Medical University, Beijing 100053, China
| | - Lingling Geng
- Advanced Innovation Center for Human Brain Protection and National Clinical Research Center for Geriatric Disorders, Xuanwu Hospital Capital Medical University, Beijing 100053, China
- Aging Translational Medicine Center, International Center for Aging and Cancer, Beijing Municipal Geriatric Medical Research Center, Xuan Wu Hospital, Capital Medical University, Beijing 100053, China
| | - Shuai Ma
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China
| | - Yusheng Cai
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China
| | - Chengyu Liu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yupeng Yan
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China
| | | | | | | | | | | |
Collapse
|
25
|
Ma S, Long L, Huang X, Tian K, Tian Y, Wu C, Zhao Z. Transcriptome analysis reveals genes associated with wool fineness in merinos. PeerJ 2023; 11:e15327. [PMID: 37250719 PMCID: PMC10215774 DOI: 10.7717/peerj.15327] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Accepted: 04/10/2023] [Indexed: 05/31/2023] Open
Abstract
Hair/wool usually plays an important role in maintaining animal physiological activities, and the economic value of wool cannot be ignored. At present, people set higher demands on wool fineness. Hence, improving wool fineness is the concern of fine wool sheep breeding. Using RNA-Seq to screen the potential candidate genes that associate with wool fineness can provide theoretical references for fine-wool sheep breeding, and also provide us new ideas for further understand the molecular regulation mechanism of hair growth. In this study, we compared the expression pattern difference of genome-wide genes between the skin transcriptomes of Subo and Chinese Merinos. The results showed that, 16 candidate differentially expressed genes (DEGs) (Included: CACNA1S, GP5, LOC101102392, HSF5, SLITRK2, LOC101104661, CREB3L4, COL1A1, PTPRR, SFRP4, LOC443220, COL6A6, COL6A5, LAMA1, LOC114115342 and LOC101116863 genes) that may associate with wool fineness were screened, and these genes were located in signaling pathways that regulate hair follicle development, cycle or hair growth. It is worth noting that, among the 16 DEGs, COL1A1 gene has the highest expression level in Merino skins, and the fold change of LOC101116863 gene is the highest, and the structures of these two genes are both highly conserved in different species. In conclusion, we speculate that these two genes may play a key role in regulating wool fineness and respectively have similar and conserved functions in different species.
Collapse
Affiliation(s)
- Shengchao Ma
- College of Animal Science, Xinjiang Agricultural University, Urumqi, China
| | - Li Long
- College of Animal Science, Xinjiang Agricultural University, Urumqi, China
| | - Xixia Huang
- College of Animal Science, Xinjiang Agricultural University, Urumqi, China
| | - Kechuan Tian
- Institute of Animal Science and Veterinary Medicine, Shandong Academy of Agricultural Sciences, Jinan, China
| | - Yuezhen Tian
- Key Laboratory of Genetics Breeding and Reproduction of Xinjiang Wool Sheep and Cashmere-Goat, Institute of Animal Science, Xinjiang Academy of Animal Sciences, Urumqi, China
| | - Cuiling Wu
- College of Animal Science, Xinjiang Agricultural University, Urumqi, China
- Institute of Animal Science and Veterinary Medicine, Shandong Academy of Agricultural Sciences, Jinan, China
- Key Laboratory of Genetics Breeding and Reproduction of Xinjiang Wool Sheep and Cashmere-Goat, Institute of Animal Science, Xinjiang Academy of Animal Sciences, Urumqi, China
| | - Zhiwen Zhao
- College of Animal Science, Xinjiang Agricultural University, Urumqi, China
| |
Collapse
|
26
|
Li F, Chen H, Lu X. The Role of NFATC4 Gene in Human Cutaneous Squamous Cell Carcinoma. Indian J Dermatol 2023; 68:156-160. [PMID: 37275813 PMCID: PMC10238996 DOI: 10.4103/ijd.ijd_837_22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/07/2023] Open
Abstract
Nuclear factor of activated T-cells, cytoplasmic 4 (NFATC4) has been implicated in keratinocyte development and several types of cancer. A well-defined role for NFATC4 in cutaneous squamous cell carcinoma (CSCC) has not yet been established. In this study, NFATC4 gene function in CSCC development was examined. Quantitative reverse transcription polymerase chain reaction (RT-qPCR) was used to measure the mRNA expression of NFATC4 in CSCC tissues and controls. A431 and Colo16 cell proliferation, invasion, and apoptosis were measured by CCK-8 assay, transwell invasion, and flow cytometry, respectively, after an NFATC4 expression lentivirus infection. Animal models were applied to validate the function of the NFATC4 gene. (1) CSCC tissues showed a significant decrease in NFATC4 expression compared to controls. (2) Overexpression of NFATc4 suppresses A431 and Colo16 cell proliferation and invasion but promotes cell apoptosis. (3) Mouse models overexpressing NFATC4 showed reduced tumourigenesis. It was suggested that NFATC4 might be a tumour suppressor gene in CSCC.
Collapse
Affiliation(s)
- Fengjuan Li
- From the Department of Dermatology, The Affiliated Hospital of Qingdao University, Qingdao Shandong, China
- Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Nanjing, Jiangsu, China
| | - Hongquan Chen
- From the Department of Dermatology, The Affiliated Hospital of Qingdao University, Qingdao Shandong, China
| | - Xiaoou Lu
- From the Department of Dermatology, The Affiliated Hospital of Qingdao University, Qingdao Shandong, China
| |
Collapse
|
27
|
Melatonin Promotes the Development of Secondary Hair Follicles in Adult Cashmere Goats by Activating the Keap1-Nrf2 Signaling Pathway and Inhibiting the Inflammatory Transcription Factors NFκB and AP-1. Int J Mol Sci 2023; 24:ijms24043403. [PMID: 36834812 PMCID: PMC9964152 DOI: 10.3390/ijms24043403] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2022] [Revised: 01/21/2023] [Accepted: 01/25/2023] [Indexed: 02/11/2023] Open
Abstract
Exogenous melatonin (MT) has been used to promote the growth of secondary hair follicles and improve cashmere fiber quality, but the specific cellular-level mechanisms involved are unclear. This study was carried out to investigate the effect of MT on the development of secondary hair follicles and on cashmere fiber quality in cashmere goats. The results showed that MT improved secondary follicle numbers and function as well as enhanced cashmere fiber quality and yield. The MT-treated goat groups had high secondary-to-primary ratios (S:P) for hair follicles, greater in the elderly group (p < 0.05). Antioxidant capacities of secondary hair follicles improved fiber quality and yield in comparison with control groups (p < 0.05/0.01). Levels of reactive oxygen and nitrogen species (ROS, RNS) and malondialdehyde (MDA) were lowered (p < 0.05/0.01) by MT. There was significant upregulation of antioxidant genes (for SOD-3; GPX-1; NFE2L2) and the protein of nuclear factor (Nrf2), and downregulation of the Keap1 protein. There were significant differences in the expression of genes for secretory senescence-associated phenotype (SASP) cytokines (IL-1β, IL-6, MMP-9, MMP-27, CCL-21, CXCL-12, CXCL-14, TIMP-1,2,3) plus their protein of key transcription factors, nuclear factor kappa B (NFκB) and activator protein-1 (AP-1), in comparison with the controls. We concluded that MT could enhance antioxidant capacity and reduce ROS and RNS levels of secondary hair follicles through the Keap1-Nrf2 pathway in adult cashmere goats. Furthermore, MT reduced the expression of the SASP cytokines genes by inhibiting the protein of NFκB and AP-1 in the secondary hair follicles in older cashmere goats, thus delaying skin aging, improving follicle survival, and increasing the number of secondary hair follicles. Collectively, these effects of exogenous MT enhanced the quality and yield of cashmere fibers, especially at 5-7 years old.
Collapse
|
28
|
Saghatelyan A. Calcium signaling as an integrator and decoder of niche factors to control somatic stem cell quiescence and activation. FEBS J 2023; 290:677-683. [PMID: 34797958 DOI: 10.1111/febs.16289] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 10/26/2021] [Accepted: 11/18/2021] [Indexed: 02/04/2023]
Abstract
Somatic stem cells (SSCs) play a major role in tissue homeostasis and respond to a panoply of micro-environmental cues by adjusting their quiescence and activation profiles. How these cells integrate and decode multiple niche signals remains elusive. In recent years, Ca2+ signaling has emerged as one of the key intracellular pathways that allow stem cells to dynamically adjust their fate and either to remain quiescent for future needs or to become activated to generate new progeny. Interestingly, not only distinct Ca2+ signatures are associated with the quiescence and activation states of stem cells, but also various extracellular cues impinge on Ca2+ pathways to dynamically regulate the responses of stem cells to different niche signals. This Viewpoint article deals with how Ca2+ signaling may be used to decode and integrate different niche factors and how Ca2+ fluctuations of distinct amplitudes, frequencies, and overall intracellular levels may trigger the differential gene transcription program. Knowledge about mechanisms that allow SSCs to translate the complexity of extracellular niche signaling into intrinsic states of cell quiescence and activation is crucial for understanding life-long tissue homeostasis and regeneration.
Collapse
Affiliation(s)
- Armen Saghatelyan
- CERVO Brain Research Center, Quebec City, Quebec, Canada.,Université Laval, Quebec City, Quebec, Canada
| |
Collapse
|
29
|
Oak ASW, Cotsarelis G. Wound-Induced Hair Neogenesis: A Portal to the Development of New Therapies for Hair Loss and Wound Regeneration. Cold Spring Harb Perspect Biol 2023; 15:cshperspect.a041239. [PMID: 36123030 PMCID: PMC9899649 DOI: 10.1101/cshperspect.a041239] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Adult mammals retain the remarkable ability to regenerate hair follicles after wounding. Wound-induced hair neogenesis (WIHN) in many ways recapitulates embryogenesis. The origin of the stem cells that give rise to a nascent hair follicle after wounding and the role of mesenchymal cells and signaling pathways responsible for this regenerative phenomenon are slowly being elucidated. WIHN provides a potential therapeutic window for manipulating cell fate by the introduction of factors during the wound healing process to enhance hair follicle formation.
Collapse
Affiliation(s)
- Allen S W Oak
- Department of Dermatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | - George Cotsarelis
- Department of Dermatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| |
Collapse
|
30
|
Jang H, Jo Y, Lee JH, Choi S. Aging of hair follicle stem cells and their niches. BMB Rep 2023; 56:2-9. [PMID: 36379515 PMCID: PMC9887102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Indexed: 01/28/2023] Open
Abstract
Hair follicles in the skin undergo cyclic rounds of regeneration, degeneration, and rest throughout life. Stem cells residing in hair follicles play a pivotal role in maintaining tissue homeostasis and hair growth cycles. Research on hair follicle aging and age-related hair loss has demonstrated that a decline in hair follicle stem cell (HFSC) activity with aging can decrease the regeneration capacity of hair follicles. This review summarizes our understanding of how age-associated HFSC intrinsic and extrinsic mechanisms can induce HFSC aging and hair loss. In addition, we discuss approaches developed to attenuate ageassociated changes in HFSCs and their niches, thereby promoting hair regrowth. [BMB Reports 2023; 56(1): 2-9].
Collapse
Affiliation(s)
- Hansaem Jang
- Department of Life Sciences, Pohang University of Science and Technology (POSTECH), Pohang 37673, Korea, Incheon 21983, Korea
| | - Yemin Jo
- Department of Life Sciences, Pohang University of Science and Technology (POSTECH), Pohang 37673, Korea, Incheon 21983, Korea
| | - Jung Hyun Lee
- Division of Dermatology, Department of Medicine, University of Washington, Seattle, WA 98109, Korea,Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA 98109, USA, Incheon 21983, Korea
| | - Sekyu Choi
- Department of Life Sciences, Pohang University of Science and Technology (POSTECH), Pohang 37673, Korea, Incheon 21983, Korea,School of Medical Science and Engineering, Pohang University of Science and Technology (POSTECH), Pohang 37673, Korea,School of Interdisciplinary Bioscience and Bioengineering, Pohang University of Science and Technology (POSTECH), Pohang 37673, Korea,Institute for Convergence Research and Education in Advanced Technology (I_CREATE), Yonsei University, Incheon 21983, Korea,Corresponding author. Tel: +82-54-279-2359; Fax: +82-54-279-2199; E-mail:
| |
Collapse
|
31
|
Jang H, Jo Y, Lee JH, Choi S. Aging of hair follicle stem cells and their niches. BMB Rep 2023; 56:2-9. [PMID: 36379515 PMCID: PMC9887102 DOI: 10.5483/bmbrep.2022-0183] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Revised: 11/14/2022] [Accepted: 11/14/2022] [Indexed: 09/06/2023] Open
Abstract
Hair follicles in the skin undergo cyclic rounds of regeneration, degeneration, and rest throughout life. Stem cells residing in hair follicles play a pivotal role in maintaining tissue homeostasis and hair growth cycles. Research on hair follicle aging and age-related hair loss has demonstrated that a decline in hair follicle stem cell (HFSC) activity with aging can decrease the regeneration capacity of hair follicles. This review summarizes our understanding of how age-associated HFSC intrinsic and extrinsic mechanisms can induce HFSC aging and hair loss. In addition, we discuss approaches developed to attenuate ageassociated changes in HFSCs and their niches, thereby promoting hair regrowth. [BMB Reports 2023; 56(1): 2-9].
Collapse
Affiliation(s)
- Hansaem Jang
- Department of Life Sciences, Pohang University of Science and Technology (POSTECH), Pohang 37673, Korea, Incheon 21983, Korea
| | - Yemin Jo
- Department of Life Sciences, Pohang University of Science and Technology (POSTECH), Pohang 37673, Korea, Incheon 21983, Korea
| | - Jung Hyun Lee
- Division of Dermatology, Department of Medicine, University of Washington, Seattle, WA 98109, Korea
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA 98109, USA, Incheon 21983, Korea
| | - Sekyu Choi
- Department of Life Sciences, Pohang University of Science and Technology (POSTECH), Pohang 37673, Korea, Incheon 21983, Korea
- School of Medical Science and Engineering, Pohang University of Science and Technology (POSTECH), Pohang 37673, Korea
- School of Interdisciplinary Bioscience and Bioengineering, Pohang University of Science and Technology (POSTECH), Pohang 37673, Korea
- Institute for Convergence Research and Education in Advanced Technology (I_CREATE), Yonsei University, Incheon 21983, Korea
| |
Collapse
|
32
|
Sun B, Sherrin M, Roy R. Unscheduled epigenetic modifications cause genome instability and sterility through aberrant R-loops following starvation. Nucleic Acids Res 2022; 51:84-98. [PMID: 36504323 PMCID: PMC9841415 DOI: 10.1093/nar/gkac1155] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Accepted: 12/09/2022] [Indexed: 12/14/2022] Open
Abstract
During starvation, organisms modify both gene expression and metabolism to adjust to the energy stress. We previously reported that Caenorhabditis elegans lacing AMP-activated protein kinase (AMPK) exhibit transgenerational reproductive defects associated with abnormally elevated trimethylated histone H3 at lysine 4 (H3K4me3) levels in the germ line following recovery from acute starvation. Here, we show that these H3K4me3 marks are significantly increased at promoters, driving aberrant transcription elongation resulting in the accumulation of R-loops in starved AMPK mutants. DNA-RNA immunoprecipitation followed by high-throughput sequencing (DRIP-seq) analysis demonstrated that a significant proportion of the genome was affected by R-loop formation. This was most pronounced in the promoter-transcription start site regions of genes, in which the chromatin was modified by H3K4me3. Like H3K4me3, the R-loops were also found to be heritable, likely contributing to the transgenerational reproductive defects typical of these mutants following starvation. Strikingly, AMPK mutant germ lines show considerably more RAD-51 (the RecA recombinase) foci at sites of R-loop formation, potentially sequestering them from their roles at meiotic breaks or at sites of induced DNA damage. Our study reveals a previously unforeseen role of AMPK in maintaining genome stability following starvation. The downstream effects of R-loops on DNA damage sensitivity and germline stem cell integrity may account for inappropriate epigenetic modification that occurs in numerous human disorders, including various cancers.
Collapse
Affiliation(s)
- Bing Sun
- To whom correspondence should be addressed.
| | - McLean Sherrin
- Department of Biology, McGill University, Montreal, Quebec H3A 1B1, Canada
| | - Richard Roy
- Correspondence may also be addressed to Richard Roy. Tel: +1 514 398 6437;
| |
Collapse
|
33
|
Sonam S, Bangru S, Perry KJ, Chembazhi UV, Kalsotra A, Henry JJ. Cellular and molecular profiles of larval and adult Xenopus corneal epithelia resolved at the single-cell level. Dev Biol 2022; 491:13-30. [PMID: 36049533 PMCID: PMC10241109 DOI: 10.1016/j.ydbio.2022.08.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Revised: 08/22/2022] [Accepted: 08/23/2022] [Indexed: 11/24/2022]
Abstract
Corneal Epithelial Stem Cells (CESCs) and their proliferative progeny, the Transit Amplifying Cells (TACs), are responsible for homeostasis and maintaining corneal transparency. Owing to our limited knowledge of cell fates and gene activity within the cornea, the search for unique markers to identify and isolate these cells remains crucial for ocular surface reconstruction. We performed single-cell RNA sequencing of corneal cells from larval and adult stages of Xenopus. Our results indicate that as the cornea develops and matures, there is an increase in cellular diversity, which is accompanied by a substantial shift in transcriptional profile, gene regulatory network and cell-cell communication dynamics. Our data also reveals several novel genes expressed in corneal cells and changes in gene expression during corneal differentiation at both developmental time-points. Importantly, we identify specific basal cell clusters in both the larval and adult cornea that comprise a relatively undifferentiated cell type and express distinct stem cell markers, which we propose are the putative larval and adult CESCs, respectively. This study offers a detailed atlas of single-cell transcriptomes in the frog cornea. In the future, this work will be useful to elucidate the function of novel genes in corneal epithelial homeostasis, wound healing and regeneration.
Collapse
Affiliation(s)
- Surabhi Sonam
- Department of Cell and Developmental Biology, University of Illinois, Urbana-Champaign, IL, USA
| | - Sushant Bangru
- Department of Biochemistry, University of Illinois, Urbana-Champaign, IL, USA; Cancer Center@Illinois, University of Illinois, Urbana-Champaign, IL, USA
| | - Kimberly J Perry
- Department of Cell and Developmental Biology, University of Illinois, Urbana-Champaign, IL, USA
| | - Ullas V Chembazhi
- Department of Biochemistry, University of Illinois, Urbana-Champaign, IL, USA
| | - Auinash Kalsotra
- Department of Biochemistry, University of Illinois, Urbana-Champaign, IL, USA; Carl R. Woese Institute for Genomic Biology, University of Illinois, Urbana-Champaign, IL, USA; Cancer Center@Illinois, University of Illinois, Urbana-Champaign, IL, USA.
| | - Jonathan J Henry
- Department of Cell and Developmental Biology, University of Illinois, Urbana-Champaign, IL, USA.
| |
Collapse
|
34
|
Wang ZH, Liu LP, Zheng YW. Melanocyte stem cells in skin diseases and their potential in cell-based therapy. Histol Histopathol 2022; 37:937-953. [PMID: 35553404 DOI: 10.14670/hh-18-470] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Melanocytes have a complex function and play an important role in a variety of regulatory mechanisms in the human system. Melanocyte stem cells (MelSCs) serve as a reservoir to replenish the melanocytes by regenerating new ones, and they are capable of self-renewal and differentiation to maintain their homeostasis, repair, and regeneration in tissues. The numerical decrease and functional impairment of MelSCs may be closely related to the development and treatment response of many skin diseases. However, the current knowledge about MelSCs mainly comes from studies in mice, and little is known about human MelSC markers; especially, their markers are still unclear or lack consensus. This leads to uncertainty in clinical findings, which further limits our comprehensive understanding of pigmentary disorders and also hinders the progress of new treatments. Thus, in this review article, combined with our previous and current work, we summarize and update the recent advances in MelSC research, including the molecular markers of human MelSCs and their niche, as well as the association of MelSCs with skin diseases, including vitiligo, hair greying, and melanoma. Due to the limited tools available to explore the identified characteristics of human MelSCs, pluripotent stem cells can provide a new research model for further study, especially combined with CRISPR/Cas9 technology. The visualization of human MelSCs' development and differentiation can help to identify their molecular characteristics and understand their cellular fate dynamically, which will allow us not only to further explore their roles in associated diseases, but also to achieve MelSC-based cellular therapy.
Collapse
Affiliation(s)
- Zi-Han Wang
- Institute of Regenerative Medicine, and Department of Dermatology, Affiliated Hospital of Jiangsu University, Jiangsu University, Zhenjiang, Jiangsu, China
| | - Li-Ping Liu
- Institute of Regenerative Medicine, and Department of Dermatology, Affiliated Hospital of Jiangsu University, Jiangsu University, Zhenjiang, Jiangsu, China.
| | - Yun-Wen Zheng
- Institute of Regenerative Medicine, and Department of Dermatology, Affiliated Hospital of Jiangsu University, Jiangsu University, Zhenjiang, Jiangsu, China
- Guangdong Provincial Key Laboratory of Large Animal Models for Biomedicine, and School of Biotechnology and Health Sciences, Wuyi University, Jiangmen, Guangdong, China
- School of Medicine, Yokohama City University, Yokohama, Kanagawa, Japan
- Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
- Department of Medical and Life Sciences, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Noda, Japan.
- Center for Stem Cell Biology and Regenerative Medicine, Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
35
|
Koren E, Feldman A, Yusupova M, Kadosh A, Sedov E, Ankawa R, Yosefzon Y, Nasser W, Gerstberger S, Kimel LB, Priselac N, Brown S, Sharma S, Gorenc T, Shalom-Feuerstein R, Steller H, Shemesh T, Fuchs Y. Thy1 marks a distinct population of slow-cycling stem cells in the mouse epidermis. Nat Commun 2022; 13:4628. [PMID: 35941116 PMCID: PMC9360001 DOI: 10.1038/s41467-022-31629-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Accepted: 06/27/2022] [Indexed: 12/17/2022] Open
Abstract
The presence of distinct stem cells that maintain the interfollicular epidermis is highly debated. Here, we report a population of keratinocytes, marked by Thy1, in the basal layer of the interfollicular epidermis. We find that epidermal cells expressing differential levels of Thy1 display distinct transcriptional signatures. Thy1+ keratinocytes do not express T cell markers, express a unique transcriptional profile, cycle significantly slower than basal epidermal progenitors and display significant expansion potential in vitro. Multicolor lineage tracing analyses and mathematical modeling reveal that Thy1+ basal keratinocytes do not compete neutrally alike interfollicular progenitors and contribute long-term to both epidermal replenishment and wound repair. Importantly, ablation of Thy1+ cells strongly impairs these processes, thus indicating the non-redundant function of Thy1+ stem cells in the epidermis. Collectively, these results reveal a distinct stem cell population that plays a critical role in epidermal homeostasis and repair.
Collapse
Affiliation(s)
- Elle Koren
- Laboratory of Stem Cell Biology and Regenerative Medicine, Department of Biology, Technion Israel Institute of Technology, Haifa, Israel
- Lorry Lokey Interdisciplinary Center for Life Sciences & Engineering, Technion Israel Institute of Technology, Haifa, Israel
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Palo Alto, CA, USA
| | - Alona Feldman
- Laboratory of Stem Cell Biology and Regenerative Medicine, Department of Biology, Technion Israel Institute of Technology, Haifa, Israel
- Lorry Lokey Interdisciplinary Center for Life Sciences & Engineering, Technion Israel Institute of Technology, Haifa, Israel
| | - Marianna Yusupova
- Laboratory of Stem Cell Biology and Regenerative Medicine, Department of Biology, Technion Israel Institute of Technology, Haifa, Israel
- Lorry Lokey Interdisciplinary Center for Life Sciences & Engineering, Technion Israel Institute of Technology, Haifa, Israel
| | - Avihay Kadosh
- Laboratory of Biophysics, Department of Biology, Technion Israel Institute of Technology, Haifa, Israel
| | - Egor Sedov
- Laboratory of Stem Cell Biology and Regenerative Medicine, Department of Biology, Technion Israel Institute of Technology, Haifa, Israel
- Lorry Lokey Interdisciplinary Center for Life Sciences & Engineering, Technion Israel Institute of Technology, Haifa, Israel
| | - Roi Ankawa
- Laboratory of Stem Cell Biology and Regenerative Medicine, Department of Biology, Technion Israel Institute of Technology, Haifa, Israel
- Lorry Lokey Interdisciplinary Center for Life Sciences & Engineering, Technion Israel Institute of Technology, Haifa, Israel
| | - Yahav Yosefzon
- Laboratory of Stem Cell Biology and Regenerative Medicine, Department of Biology, Technion Israel Institute of Technology, Haifa, Israel
- Lorry Lokey Interdisciplinary Center for Life Sciences & Engineering, Technion Israel Institute of Technology, Haifa, Israel
| | - Waseem Nasser
- Department of Genetics and Developmental Biology, The Rappaport Faculty of Medicine and Research Institute, Technion Israel Institute of Technology, Haifa, Israel
| | | | - Liam B Kimel
- Laboratory of Biophysics, Department of Biology, Technion Israel Institute of Technology, Haifa, Israel
| | - Noa Priselac
- Laboratory of Stem Cell Biology and Regenerative Medicine, Department of Biology, Technion Israel Institute of Technology, Haifa, Israel
- Lorry Lokey Interdisciplinary Center for Life Sciences & Engineering, Technion Israel Institute of Technology, Haifa, Israel
| | - Samara Brown
- Strang Laboratory of Apoptosis and Cancer Biology, The Rockefeller University, New York, New York, 10065, USA
| | - Sam Sharma
- Strang Laboratory of Apoptosis and Cancer Biology, The Rockefeller University, New York, New York, 10065, USA
| | - Travis Gorenc
- Strang Laboratory of Apoptosis and Cancer Biology, The Rockefeller University, New York, New York, 10065, USA
| | - Ruby Shalom-Feuerstein
- Department of Genetics and Developmental Biology, The Rappaport Faculty of Medicine and Research Institute, Technion Israel Institute of Technology, Haifa, Israel
| | - Hermann Steller
- Strang Laboratory of Apoptosis and Cancer Biology, The Rockefeller University, New York, New York, 10065, USA
| | - Tom Shemesh
- Laboratory of Biophysics, Department of Biology, Technion Israel Institute of Technology, Haifa, Israel.
| | - Yaron Fuchs
- Laboratory of Stem Cell Biology and Regenerative Medicine, Department of Biology, Technion Israel Institute of Technology, Haifa, Israel.
- Lorry Lokey Interdisciplinary Center for Life Sciences & Engineering, Technion Israel Institute of Technology, Haifa, Israel.
| |
Collapse
|
36
|
Liu Y, Guerrero-Juarez CF, Xiao F, Shettigar NU, Ramos R, Kuan CH, Lin YC, de Jesus Martinez Lomeli L, Park JM, Oh JW, Liu R, Lin SJ, Tartaglia M, Yang RB, Yu Z, Nie Q, Li J, Plikus MV. Hedgehog signaling reprograms hair follicle niche fibroblasts to a hyper-activated state. Dev Cell 2022; 57:1758-1775.e7. [PMID: 35777353 PMCID: PMC9344965 DOI: 10.1016/j.devcel.2022.06.005] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Revised: 03/10/2022] [Accepted: 06/08/2022] [Indexed: 02/06/2023]
Abstract
Hair follicle stem cells are regulated by dermal papilla fibroblasts, their principal signaling niche. Overactivation of Hedgehog signaling in the niche dramatically accelerates hair growth and induces follicle multiplication in mice. On single-cell RNA sequencing, dermal papilla fibroblasts increase heterogeneity to include new Wnt5ahigh states. Transcriptionally, mutant fibroblasts activate regulatory networks for Gli1, Alx3, Ebf1, Hoxc8, Sox18, and Zfp239. These networks jointly upregulate secreted factors for multiple hair morphogenesis and hair-growth-related pathways. Among these is non-conventional TGF-β ligand Scube3. We show that in normal mouse skin, Scube3 is expressed only in dermal papillae of growing, but not in resting follicles. SCUBE3 protein microinjection is sufficient to induce new hair growth, and pharmacological TGF-β inhibition rescues mutant hair hyper-activation phenotype. Moreover, dermal-papilla-enriched expression of SCUBE3 and its growth-activating effect are partially conserved in human scalp hair follicles. Thus, Hedgehog regulates mesenchymal niche function in the hair follicle via SCUBE3/TGF-β mechanism.
Collapse
Affiliation(s)
- Yingzi Liu
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China; Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA 92697, USA; Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, Irvine, CA 92697, USA
| | - Christian F Guerrero-Juarez
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA 92697, USA; Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, Irvine, CA 92697, USA; Department of Mathematics, University of California, Irvine, Irvine, CA 92697, USA; Center for Complex Biological Systems, University of California, Irvine, Irvine, CA 92697, USA; NSF-Simons Center for Multiscale Cell Fate Research, University of California, Irvine, Irvine, CA 92697, USA
| | - Fei Xiao
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA 92697, USA; Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, Irvine, CA 92697, USA
| | - Nitish Udupi Shettigar
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA 92697, USA; Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, Irvine, CA 92697, USA; Amplifica Holdings Group, Inc., San Diego, CA 92128, USA
| | - Raul Ramos
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA 92697, USA; Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, Irvine, CA 92697, USA; Center for Complex Biological Systems, University of California, Irvine, Irvine, CA 92697, USA; NSF-Simons Center for Multiscale Cell Fate Research, University of California, Irvine, Irvine, CA 92697, USA
| | - Chen-Hsiang Kuan
- Division of Plastic Surgery, Department of Surgery, National Taiwan University Hospital, Taipei, Taiwan; Research Center for Developmental Biology and Regenerative Medicine, National Taiwan University, Taipei, Taiwan
| | - Yuh-Charn Lin
- Department of Physiology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan; Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | | | - Jung Min Park
- Department of Anatomy, Yonsei University College of Medicine, Seoul, Korea; Department of Anatomy, School of Medicine, Kyungpook National University, Daegu, Korea
| | - Ji Won Oh
- Department of Anatomy, Yonsei University College of Medicine, Seoul, Korea; Department of Anatomy, School of Medicine, Kyungpook National University, Daegu, Korea; Hair Transplantation Center, Kyungpook National University Hospital, Daegu, Korea
| | - Ruiqi Liu
- State Key Laboratories for Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing 100193, China
| | - Sung-Jan Lin
- Research Center for Developmental Biology and Regenerative Medicine, National Taiwan University, Taipei, Taiwan; Institute of Biomedical Engineering and Department of Dermatology, National Taiwan University, Taipei, Taiwan
| | - Marco Tartaglia
- Genetics and Rare Diseases Research Division, Ospedale Pediatrico Bambino Gesù, IRCCS, Rome 00146, Italy
| | - Ruey-Bing Yang
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Zhengquan Yu
- State Key Laboratories for Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing 100193, China
| | - Qing Nie
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA 92697, USA; Department of Mathematics, University of California, Irvine, Irvine, CA 92697, USA; Center for Complex Biological Systems, University of California, Irvine, Irvine, CA 92697, USA; NSF-Simons Center for Multiscale Cell Fate Research, University of California, Irvine, Irvine, CA 92697, USA
| | - Ji Li
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China; Hunan Key Laboratory of Aging Biology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China.
| | - Maksim V Plikus
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA 92697, USA; Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, Irvine, CA 92697, USA; Center for Complex Biological Systems, University of California, Irvine, Irvine, CA 92697, USA; NSF-Simons Center for Multiscale Cell Fate Research, University of California, Irvine, Irvine, CA 92697, USA.
| |
Collapse
|
37
|
3D Spheroid Human Dermal Papilla Cell as an Effective Model for the Screening of Hair Growth Promoting Compounds: Examples of Minoxidil and 3,4,5-Tri-O-caffeoylquinic acid (TCQA). Cells 2022; 11:cells11132093. [PMID: 35805177 PMCID: PMC9265566 DOI: 10.3390/cells11132093] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Revised: 06/24/2022] [Accepted: 06/28/2022] [Indexed: 11/29/2022] Open
Abstract
Dermal papilla cells (DPCs) are an important element of the hair follicle (HF) niche, widely used as an in vitro model to study hair growth-related research. These cells are usually grown in 2D culture, but this system did not show efficient therapeutic effects on HF regeneration and growth, and key differences were observed between cell activity in vitro and in vivo. Recent studies have showed that DPCs grown in 3D hanging spheroids are more morphologically akin to an intact DP microenvironment. In this current study, global gene molecular analysis showed that the 3D model highly affected cell adhesion molecules and hair growth-related pathways. Furthermore, we compared the expression of signalling molecules and metabolism-associated proteins of DPCs treated with minoxidil (an FDA-approved drug for hair loss treatment) and 3,4,5-tri-O-caffeoylquinic acid (TCQA) (recently found to induce hair growth in vitro and in vivo) in 3D spheroid hanging drops and a 2D monolayer using DNA microarray analysis. Further validations by determining the gene and protein expressions of key signature molecules showed the suitability of this 3D system for enhancing the DPC activity of the hair growth-promoting agents minoxidil and TCQA.
Collapse
|
38
|
Yang Q, Zhang J, Bao Q, Zhong J, Wang X, Tao Y, Xu X, Lv K, Wang Y, Li B, He L, Guo X, Ma G. Foxp1 and Foxp4 deletion causes the loss of follicle stem cell niche and cyclic hair shedding by inducing inner bulge cell apoptosis. Stem Cells 2022; 40:843-856. [PMID: 35759955 DOI: 10.1093/stmcls/sxac045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2021] [Accepted: 06/15/2022] [Indexed: 11/14/2022]
Abstract
Quiescent hair follicle stem cells (HFSCs) reside in specialized bulge niche where they undergo activation and differentiation upon sensing niche-dependent signals during hair follicle (HF) homeostasis and wound repair. The underlying mechanism of HFSCs and bulge niche maintenance is poorly understood. Our previous study has reported that a transcription factor, forkhead box P1 (Foxp1), functions to maintain the quiescence of HFSCs. Here, we further discovered that forkhead box P4 (Foxp4), a close family member of Foxp1, had similar expression profiles in various components of HFs and formed a complex with Foxp1 in vitro and in vivo. The HF-specific deficiency of Foxp4 resulted in the precocious activation of HFSCs during hair cycles. In contrast to single Foxp1 or Foxp4 conditional knockout (cKO) mice, Foxp1/4 double cKO exerted an additive effect in the spectrum and severity of phenotypes in HFSC activation, hair cycling acceleration and hair loss, coupled with remarkable downregulation of fibroblast growth factor 18 (Fgf18) and bone morphogenetic protein 6 (Bmp6) expression in bulge cells. In addition, the double KO of Foxp1/4 induced the apoptosis of K6-positive (K6+) inner bulge cells, a well-established stem cell (SC) niche, thus resulting in the destruction of the bulge SC niche and recurrent hair loss. Our investigation reveals the synergistic role of Foxp1/4 in sustaining K6+ niche cells for the quiescence of HFSCs.
Collapse
Affiliation(s)
- Qingchun Yang
- Bio-X-Renji Hospital Research Center, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, P.R. China.,Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Bio-X Institutes, Shanghai Jiao Tong University, Shanghai, P.R. China
| | - Jie Zhang
- Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Bio-X Institutes, Shanghai Jiao Tong University, Shanghai, P.R. China
| | - Qianyi Bao
- Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Bio-X Institutes, Shanghai Jiao Tong University, Shanghai, P.R. China
| | - Jialin Zhong
- Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Bio-X Institutes, Shanghai Jiao Tong University, Shanghai, P.R. China
| | - Xiaoqing Wang
- Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Bio-X Institutes, Shanghai Jiao Tong University, Shanghai, P.R. China
| | - Yixin Tao
- Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Bio-X Institutes, Shanghai Jiao Tong University, Shanghai, P.R. China
| | - Xuegang Xu
- Department of Dermatology, The First Hospital of China Medical University, Shenyang, P.R. China
| | - Kaiyang Lv
- Department of Plastic Surgery, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, P.R. China
| | - Yushu Wang
- Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Bio-X Institutes, Shanghai Jiao Tong University, Shanghai, P.R. China
| | - Baojie Li
- Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Bio-X Institutes, Shanghai Jiao Tong University, Shanghai, P.R. China
| | - Lin He
- Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Bio-X Institutes, Shanghai Jiao Tong University, Shanghai, P.R. China
| | - Xizhi Guo
- Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Bio-X Institutes, Shanghai Jiao Tong University, Shanghai, P.R. China
| | - Gang Ma
- Bio-X-Renji Hospital Research Center, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, P.R. China.,Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Bio-X Institutes, Shanghai Jiao Tong University, Shanghai, P.R. China
| |
Collapse
|
39
|
Hematopoietic Stem and Progenitor Cell Maintenance and Multiple Lineage Differentiation Is an Integral Function of NFATc1. Cells 2022; 11:cells11132012. [PMID: 35805096 PMCID: PMC9265824 DOI: 10.3390/cells11132012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Accepted: 06/21/2022] [Indexed: 12/10/2022] Open
Abstract
Hematopoietic stem and progenitor cell (HSPC) maintenance and the differentiation of various lineages is a highly complex but precisely regulated process. Multiple signaling pathways and an array of transcription factors influence HSPC maintenance and the differentiation of individual lineages to constitute a functional hematopoietic system. Nuclear factor of activated T cell (NFAT) family transcription factors have been studied in the context of development and function of multiple mature hematopoietic lineage cells. However, until now their contribution in HSPC physiology and HSPC differentiation to multiple hematopoietic lineages has remained poorly understood. Here, we show that NFAT proteins, specifically NFATc1, play an indispensable role in the maintenance of HSPCs. In the absence of NFATc1, very few HSPCs develop in the bone marrow, which are functionally defective. In addition to HSPC maintenance, NFATc1 also critically regulates differentiation of lymphoid, myeloid, and erythroid lineage cells from HSPCs. Deficiency of NFATc1 strongly impaired, while enhanced NFATc1 activity augmented, the differentiation of these lineages, which further attested to the vital involvement of NFATc1 in regulating hematopoiesis. Hematopoietic defects due to lack of NFATc1 activity can lead to severe pathologies such as lymphopenia, myelopenia, and a drastically reduced lifespan underlining the critical role NFATc1 plays in HSPC maintenance and in the differentaion of various lineages. Our findings suggest that NFATc1 is a critical component of the myriad signaling and transcriptional regulators that are essential to maintain normal hematopoiesis.
Collapse
|
40
|
Choi K, Park SH, Park SY, Yoon SK. The stem cell quiescence and niche signaling is disturbed in the hair follicle of the hairpoor mouse, an MUHH model mouse. Stem Cell Res Ther 2022; 13:211. [PMID: 35619120 PMCID: PMC9137081 DOI: 10.1186/s13287-022-02898-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Accepted: 12/16/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Hair follicle stem cells (HFSC) play an essential role in the maintenance of hair homeostasis; during the hair cycle, HFSC remain quiescent for most of its duration. The hairpoor mouse (+ /HrHp), an animal model of Marie-Unna hypotrichosis (MUHH), overexpresses hairless in the bulge, inner root sheath, and outer root sheath of HF and shows the same phenotype as in MUHH patients manifesting sparse hair with progression to alopecia with age. The aim of this study was to gain an understanding of the hair cycle and the status of HFSC during the hair cycle of the hairpoor mouse in order to delineate the pathogenesis of MUHH. METHODS H&E staining was performed in order to define the state of the hair follicle. FACS analysis and immunostaining were performed at the 1st and 2nd telogen stages for observation of the HFSC. A label retaining assay was performed to determine the quiescent state of hair follicles. qRT-PCR was performed to determine expression of factors involved in niche signaling and Wnt signaling. RESULTS We observed a drastic decrease in the number of hair follicles after the 1st telogen, followed by an intensified disturbance in the hair cycle with shorter anagen as well as 2nd telogen in the hairpoor mouse. A dramatic reduction in the number of CD34 expressing bulges as well as cells was observed at the telogen of the HFs, with prominent high proliferation of bulge cells, suggesting the loss of HFSC quiescence in the hairpoor mouse. The increased cell proliferation in HF was reiterated following the synchronization of the hair cycle, leading to acceleration of HF cycling. Reduced expression of Fgf18 and Bmp6, the factors involved in HFSC quiescence, was observed in the HFSC niche of the hairpoor mouse. In addition, disturbed expression of Wnt signaling molecules including Wnt7b, Wnt10b, and Sfrp1 was observed, which induced the telogen-to-anagen transition of HFs in the hairpoor mouse. CONCLUSIONS These results indicate that the quiescent state of HFSC is not properly maintained in the hairpoor mouse, consequently leading HFs to the completely disarrayed hair cycle. These findings may provide an understanding of an underlying mechanism for development of alopecia with age in MUHH patients.
Collapse
Affiliation(s)
- Keonwoo Choi
- Department of Biomedicine & Health Sciences, The Catholic University of Korea, Seoul, Republic of Korea.,Department of Medical Life Sciences, The Catholic University of Korea, 222 Banpo-daero, Seocho-ku, Seoul, 065-591, Republic of Korea
| | - Sang-Hee Park
- Department of Biomedicine & Health Sciences, The Catholic University of Korea, Seoul, Republic of Korea.,Department of Medical Life Sciences, The Catholic University of Korea, 222 Banpo-daero, Seocho-ku, Seoul, 065-591, Republic of Korea
| | - Seo-Yeon Park
- Department of Biomedicine & Health Sciences, The Catholic University of Korea, Seoul, Republic of Korea
| | - Sungjoo Kim Yoon
- Department of Biomedicine & Health Sciences, The Catholic University of Korea, Seoul, Republic of Korea. .,Department of Medical Life Sciences, The Catholic University of Korea, 222 Banpo-daero, Seocho-ku, Seoul, 065-591, Republic of Korea.
| |
Collapse
|
41
|
Liu Y, Yang S, Zeng Y, Tang Z, Zong X, Li X, Yang C, Liu L, Tong X, Zhou L, Wang D. Dysregulated behaviour of hair follicle stem cells triggers alopecia and provides potential therapeutic targets. Exp Dermatol 2022; 31:986-992. [PMID: 35524394 DOI: 10.1111/exd.14600] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2022] [Revised: 04/10/2022] [Accepted: 05/03/2022] [Indexed: 11/26/2022]
Abstract
Due to a steady increase in the number of individuals suffering from alopecia, this condition has recently received increasing attention. Alopecia can be caused by various pathological, environmental or psychological factors, eventually resulting in abnormalities in hair follicle (HF) structures or HF regeneration disorders, especially dysregulated hair follicle stem cell (HFSC) behaviour. HFSC behaviour includes activation, proliferation and differentiation. Appropriate HFSC behaviour sustains a persistent hair cycle (HC). HFSC behaviour is mainly influenced by HFSC metabolism, ageing, and the microenvironment. In this review, we summarize recent findings on how HFSC metabolism, ageing and the microenvironment give rise to hair growth disorders, as well as related genes and signalling pathways. Recent research on the application of stem cell-based hair tissue engineering and regenerative medicine to treat alopecia is also summarized. Determining how dysregulated HFSC behaviour underlies alopecia would be helpful in identifying potential therapeutic targets.
Collapse
Affiliation(s)
- Yuanhong Liu
- Department of Dermatology, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Shengbo Yang
- Department of Dermatology, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Yilan Zeng
- Department of Dermatology, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Ziting Tang
- Department of Dermatology, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Xiule Zong
- Department of Dermatology, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Xuemei Li
- Department of Dermatology, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Caifeng Yang
- Department of Dermatology, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Lulu Liu
- Department of Dermatology, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Xiaoliang Tong
- Department of Dermatology, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Lu Zhou
- Department of Dermatology, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Dan Wang
- Department of Dermatology, The Third Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
42
|
Wang J, Shang L, Zhang J, Yu Z, Huang J, Jin J, Lv C. Nfatc1 + colonic stem cells contribute to regeneration upon colitis. J Gastroenterol Hepatol 2022; 37:734-740. [PMID: 35102609 DOI: 10.1111/jgh.15790] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Revised: 12/14/2021] [Accepted: 01/03/2022] [Indexed: 01/21/2023]
Abstract
BACKGROUND AND AIM Colonic stem cells play important roles in both normal epithelial turnover and injury repair. Lgr5+ colonic stem cells are highly susceptible to DSS-induced damage. However, it is still unclear how colonic stem cells regenerate injured epithelium during colitis. Here, we explored the functions of a new population of NFATc1+ colonic stem cells in experimental colitis. METHODS Nfatc1+ colonic stem cells were labeled using Nfatc1CreERT2 ;R26mTmG reporter mice. Immunostaining assays were used to detect Goblet cells, enteroendocrine cells, and intestinal stem/progenitor cells. We performed lineage tracing assay to investigate whether Nfatc1+ cells are real colonic stem cells using Nfatc1CreERT2 ;R26mTmG mice. The contribution of Nfatc1+ stem cells on epithelial regeneration was detected in experimental colitis induced by DSS. RESULTS Nfatc1-reporter marked cells are enriched for +3 to +5 position in colonic crypts, and they are overlapped with Sox9+ cells and Hopx+ cells that have been identified as stem cells in small intestine. However, Nfatc1-reporter marked cells are not overlapped with Lgr5+ colonic stem cells, as well as differentiated goblet cells and enteroendocrine cells. Furthermore, Nfatc1-reporter marked cells are able to give rise to all lineages of the colonic epithelium, and they preferentially contribute to the regeneration of colonic epithelium in DSS-induced experimental colitis. CONCLUSION Nfatc1+ cells were identified as a novel population of colonic stem cells that are primarily located at +3 to +5 position and contribute to epithelial regeneration during colitis.
Collapse
Affiliation(s)
- Jianyang Wang
- Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Linkuo Shang
- Key Laboratory of Precision Nutrition and Food Quality, Ministry of Education, Department of Nutrition and Health, China Agricultural University, Beijing, China
| | - Jiahui Zhang
- Key Laboratory of Precision Nutrition and Food Quality, Ministry of Education, Department of Nutrition and Health, China Agricultural University, Beijing, China
| | - Zhengquan Yu
- State Key Laboratories for Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, China
| | - Jiaqiang Huang
- Key Laboratory of Precision Nutrition and Food Quality, Ministry of Education, Department of Nutrition and Health, China Agricultural University, Beijing, China
| | - Jing Jin
- National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital and Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen, China
| | - Cong Lv
- Key Laboratory of Precision Nutrition and Food Quality, Ministry of Education, Department of Nutrition and Health, China Agricultural University, Beijing, China
| |
Collapse
|
43
|
Liu R, Hu H, McNeil M, Xu J, Bi X, Lou P, Guerrero-Juarez CF, Dai X, Plikus MV, Shuai J, Yu Z, Lv C. Dormant Nfatc1 reporter-marked basal stem/progenitor cells contribute to mammary lobuloalveoli formation. iScience 2022; 25:103982. [PMID: 35310332 PMCID: PMC8924625 DOI: 10.1016/j.isci.2022.103982] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 12/21/2021] [Accepted: 02/22/2022] [Indexed: 11/13/2022] Open
Abstract
The Mammary gland undergoes complicated epithelial remodeling to form lobuloalveoli during pregnancy, in which basal epithelial cells remarkably increase to form a basket-like architecture. However, it remains largely unknown how dormant mammary basal stem/progenitor cells involve in lobuloalveolar development. Here, we show that Nfatc1 expression marks a rare population of mammary epithelial cells with the majority being basal epithelial cells. Nfatc1 reporter-marked basal epithelial cells are relatively dormant mammary stem/progenitor cells. Although Nfatc1 reporter-marked basal epithelial cells have limited contribution to the homeostasis of mammary epithelium, they divide rapidly during pregnancy and contribute to lobuloalveolar development. Furthermore, Nfatc1 reporter-marked basal epithelial cells are preferentially used for multiple pregnancies. Using single-cell RNA-seq analysis, we identify multiple functionally distinct clusters within the Nfatc1 reporter-marked cell-derived progeny cells during pregnancy. Taken together, our findings underscore Nfatc1 reporter-marked basal cells as dormant stem/progenitor cells that contribute to mammary lobuloalveolar development during pregnancy.
Collapse
Affiliation(s)
- Ruiqi Liu
- State Key Laboratories for Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing 100193, China
| | - Huan Hu
- Department of Physics, and National Institute for Data Science in Health and Medicine, Xiamen University, Xiamen 361005, China
| | - Melissa McNeil
- Departments of Biological Chemistry and Dermatology, School of Medicine, Center for Multiscale Cell Fate Research, University of California, Irvine, Irvine, CA 92697, USA
| | - Jiuzhi Xu
- State Key Laboratories for Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing 100193, China
| | - Xueyun Bi
- State Key Laboratories for Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing 100193, China
| | - Pengbo Lou
- State Key Laboratories for Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing 100193, China
| | - Christian F. Guerrero-Juarez
- Department of Developmental and Cell Biology, NSF-Simons Center for Multiscale Cell Fate Research, Sue and Bill Gross Stem Cell Research, Center for Complex Biological Systems, University of California, Irvine, Irvine, CA 92697, USA
| | - Xing Dai
- Departments of Biological Chemistry and Dermatology, School of Medicine, Center for Multiscale Cell Fate Research, University of California, Irvine, Irvine, CA 92697, USA
| | - Maksim V. Plikus
- Department of Developmental and Cell Biology, NSF-Simons Center for Multiscale Cell Fate Research, Sue and Bill Gross Stem Cell Research, Center for Complex Biological Systems, University of California, Irvine, Irvine, CA 92697, USA
| | - Jianwei Shuai
- Department of Physics, and National Institute for Data Science in Health and Medicine, Xiamen University, Xiamen 361005, China
- Wenzhou Institute, University of Chinese Academy of Sciences, and Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Wenzhou, Zhejiang 325001, China
| | - Zhengquan Yu
- State Key Laboratories for Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing 100193, China
| | - Cong Lv
- Key Laboratory of Precision Nutrition and Food Quality, Ministry of Education, Department of Nutrition and Health, China Agricultural University, Beijing 100193, China
| |
Collapse
|
44
|
Lewuillon C, Laguillaumie MO, Quesnel B, Idziorek T, Touil Y, Lemonnier L. Put in a “Ca2+ll” to Acute Myeloid Leukemia. Cells 2022; 11:cells11030543. [PMID: 35159351 PMCID: PMC8834247 DOI: 10.3390/cells11030543] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 01/28/2022] [Accepted: 02/01/2022] [Indexed: 02/05/2023] Open
Abstract
Acute myeloid leukemia (AML) is a clonal disorder characterized by genetic aberrations in myeloid primitive cells (blasts) which lead to their defective maturation/function and their proliferation in the bone marrow (BM) and blood of affected individuals. Current intensive chemotherapy protocols result in complete remission in 50% to 80% of AML patients depending on their age and the AML type involved. While alterations in calcium signaling have been extensively studied in solid tumors, little is known about the role of calcium in most hematologic malignancies, including AML. Our purpose with this review is to raise awareness about this issue and to present (i) the role of calcium signaling in AML cell proliferation and differentiation and in the quiescence of hematopoietic stem cells; (ii) the interplay between mitochondria, metabolism, and oxidative stress; (iii) the effect of the BM microenvironment on AML cell fate; and finally (iv) the mechanism by which chemotherapeutic treatments modify calcium homeostasis in AML cells.
Collapse
Affiliation(s)
- Clara Lewuillon
- Univ. Lille, CNRS, Inserm, CHU Lille, UMR9020-U1277—CANTHER—Cancer Heterogeneity Plasticity and Resistance to Therapies, F-59000 Lille, France; (C.L.); (M.-O.L.); (B.Q.); (T.I.); (Y.T.)
| | - Marie-Océane Laguillaumie
- Univ. Lille, CNRS, Inserm, CHU Lille, UMR9020-U1277—CANTHER—Cancer Heterogeneity Plasticity and Resistance to Therapies, F-59000 Lille, France; (C.L.); (M.-O.L.); (B.Q.); (T.I.); (Y.T.)
| | - Bruno Quesnel
- Univ. Lille, CNRS, Inserm, CHU Lille, UMR9020-U1277—CANTHER—Cancer Heterogeneity Plasticity and Resistance to Therapies, F-59000 Lille, France; (C.L.); (M.-O.L.); (B.Q.); (T.I.); (Y.T.)
| | - Thierry Idziorek
- Univ. Lille, CNRS, Inserm, CHU Lille, UMR9020-U1277—CANTHER—Cancer Heterogeneity Plasticity and Resistance to Therapies, F-59000 Lille, France; (C.L.); (M.-O.L.); (B.Q.); (T.I.); (Y.T.)
| | - Yasmine Touil
- Univ. Lille, CNRS, Inserm, CHU Lille, UMR9020-U1277—CANTHER—Cancer Heterogeneity Plasticity and Resistance to Therapies, F-59000 Lille, France; (C.L.); (M.-O.L.); (B.Q.); (T.I.); (Y.T.)
| | - Loïc Lemonnier
- Univ. Lille, Inserm, U1003—PHYCEL—Physiologie Cellulaire, F-59000 Lille, France
- Laboratory of Excellence, Ion Channels Science and Therapeutics, F-59655 Villeneuve d’Ascq, France
- Correspondence:
| |
Collapse
|
45
|
Abstract
A definite identification of epidermal stem cells is not known and the mechanism of epidermal differentiation is not fully understood. Toward both of these quests, considerable information is available from the research on lineage tracing and clonal growth analysis in the basal layer of the epidermis, on the hair follicle and the interfollicular epidermal stem cells, and on Wnt signaling along with its role in the developmental patterning and cell differentiation. In this paper, literature on the aforementioned research has been collated and analyzed. In addition, models of the basal layer cellular composition and the epidermal differentiation have been presented. Graphical Abstract.
Collapse
Affiliation(s)
- Raghvendra Singh
- Department of Chemical Engineering, Indian Institute of Technology Kanpur, Kanpur, 208016, India.
| |
Collapse
|
46
|
Kim SH, Rodriguez LRL, Macias E, Rodriguez-Puebla ML. Cyclin-Dependent Kinase 4 expression alters the number of keratinocyte stem cells in the mouse hair follicle. Cell Biol Int 2022; 46:737-746. [PMID: 35032143 PMCID: PMC9035071 DOI: 10.1002/cbin.11765] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Revised: 12/21/2021] [Accepted: 01/08/2022] [Indexed: 11/25/2022]
Abstract
Hair follicles regenerate periodically by spontaneously undergoing cycles of growth, regression, and relative quiescence. During the hair cycle, follicle stem cells residing in a specialized niche remain quiescent, and they are stimulated to proliferate throughout the growth phase of the hair follicle. Although cell cycle regulators play a prominent role during the activation of hair follicle stem cells, the identity and the role of these regulators have not been confirmed. Herein, we reported that stem cells located in the bulge region of the HF (BuSCs) express high levels of cyclin‐dependent kinase 4 (CDK4) through the quiescent phase of the hair cycle. Using gain‐ and loss‐of‐function studies, we have determined that the CDK4 protein level affects the number of BuSCs. Transgenic expression of CDK4 in the bulge region of the hair follicles reduces the number of BuSCs, whereas CDK4 ablation resulted in an increasing number of BuSCs. These results suggest that deregulation of CDK4 protein levels contributes to distorting the self‐renewal/proliferation balance and, in turn, altering the number of BuSCs.
Collapse
Affiliation(s)
- Sun Hye Kim
- Department of Molecular Biomedical Sciences, the Center for Human Health and the Environment, and the Comparative Medicine Institute, North Carolina State University, Raleigh, North Carolina
| | | | - Everardo Macias
- Department of Pathology, School of Medicine, Duke University, Durham, North Carolina
| | - Marcelo L Rodriguez-Puebla
- Department of Molecular Biomedical Sciences, the Center for Human Health and the Environment, and the Comparative Medicine Institute, North Carolina State University, Raleigh, North Carolina
| |
Collapse
|
47
|
Hutchison DM, Duffens A, Yale K, Park A, Cardenas K, Mesinkovska NA. Eyelash trichomegaly: a systematic review of acquired and congenital aetiologies of lengthened lashes. J Eur Acad Dermatol Venereol 2021; 36:536-546. [PMID: 34919300 DOI: 10.1111/jdv.17877] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Revised: 10/05/2021] [Accepted: 11/10/2021] [Indexed: 02/01/2023]
Abstract
Long eyelashes have been popularized and many commercially available products exist to achieve eyelash growth as a desired cosmetic effect. Eyelash trichomegaly may be induced by medications, procedures, or be related to medical conditions; however, the exact mechanisms that govern eyelash growth are not well elucidated. This study aims to identify and summarize aetiologies associated with eyelash trichomegaly. We report a systematic review of 148 clinical trials, prospective and retrospective studies, and case reports describing all evidence-based potential aetiologies of eyelash trichomegaly obtained from the Medline/PubMed and Cochrane Library through January 2021. Inclusion criteria were defined as (i) human studies involving congenital and acquired diseases in which eyelash trichomegaly is a characteristic or (ii) assessment of trichomegaly as an adverse or desired effect of a medication or procedure. Exclusion criteria included: animal studies, articles not available in English, outcomes unrelated to eyelash trichomegaly, and secondary review articles. Pharmacologic agents associated with eyelash trichomegaly included prostaglandin analogues (15-keto fluprostenol isopropyl ester, bimatoprost, latanoprost, and travoprost), epidermal growth factor receptor inhibitors (cetuximab, erlotinib, and panitumumab), interferon-alpha, and calcineurin inhibitors (tacrolimus and cyclosporine). Surgical procedures of the eyelid, as well as allergic rhinitis, atopic dermatitis, HIV, ichthyosis vulgaris (IV), uveitis, and vernal keratoconjunctivitis were also associated with increased eyelash growth. Congenital disorders associated with lengthened eyelashes included Cantú syndrome, CHOPS syndrome, Coffin-Siris syndrome, congenital heart disease, Cornelia de Lange syndrome, Costello syndrome, familial trichomegaly, Floating Harbor syndrome, Hermansky-Pudlak syndrome, Kabuki-Makeup syndrome, KBG syndrome, Oliver-McFarlane syndrome, Rubinstein-Taybi syndrome, and Smith-Magenis syndrome. While the most common cause of eyelash trichomegaly is topical bimatoprost use, better understanding of pathways implicated in eyelash trichomegaly may lead to the discovery of additional medications to stimulate eyelash growth and create avenues for future therapeutic interventions.
Collapse
Affiliation(s)
- D M Hutchison
- Department of Dermatology, University of California Irvine, Irvine, CA, USA.,Beckman Laser Institute, University of California Irvine, Irvine, CA, USA
| | - A Duffens
- Department of Dermatology, University of California Irvine, Irvine, CA, USA
| | - K Yale
- Department of Dermatology, University of California Irvine, Irvine, CA, USA
| | - A Park
- Beckman Laser Institute, University of California Irvine, Irvine, CA, USA
| | - K Cardenas
- Department of Dermatology, University of California Irvine, Irvine, CA, USA
| | - N A Mesinkovska
- Department of Dermatology, University of California Irvine, Irvine, CA, USA.,Beckman Laser Institute, University of California Irvine, Irvine, CA, USA
| |
Collapse
|
48
|
Stepaniak MD, Square TA, Miller CT. Evolved Bmp6 enhancer alleles drive spatial shifts in gene expression during tooth development in sticklebacks. Genetics 2021; 219:6374454. [PMID: 34849839 PMCID: PMC8664583 DOI: 10.1093/genetics/iyab151] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Accepted: 08/31/2021] [Indexed: 11/13/2022] Open
Abstract
Mutations in enhancers have been shown to often underlie natural variation but the evolved differences in enhancer activity can be difficult to identify in vivo. Threespine sticklebacks (Gasterosteus aculeatus) are a robust system for studying enhancer evolution due to abundant natural genetic variation, a diversity of evolved phenotypes between ancestral marine and derived freshwater forms, and the tractability of transgenic techniques. Previous work identified a series of polymorphisms within an intronic enhancer of the Bone morphogenetic protein 6 (Bmp6) gene that are associated with evolved tooth gain, a derived increase in freshwater tooth number that arises late in development. Here, we use a bicistronic reporter construct containing a genetic insulator and a pair of reciprocal two-color transgenic reporter lines to compare enhancer activity of marine and freshwater alleles of this enhancer. In older fish, the two alleles drive partially overlapping expression in both mesenchyme and epithelium of developing teeth, but the freshwater enhancer drives a reduced mesenchymal domain and a larger epithelial domain relative to the marine enhancer. In younger fish, these spatial shifts in enhancer activity are less pronounced. Comparing Bmp6 expression by in situ hybridization in developing teeth of marine and freshwater fish reveals similar evolved spatial shifts in gene expression. Together, these data support a model in which the polymorphisms within this enhancer underlie evolved tooth gain by shifting the spatial expression of Bmp6 during tooth development, and provide a general strategy to identify spatial differences in enhancer activity in vivo.
Collapse
Affiliation(s)
- Mark D Stepaniak
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Tyler A Square
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Craig T Miller
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA
| |
Collapse
|
49
|
McNeil M, Han Y, Sun P, Watanabe K, Jiang J, Chen N, Yu Z, Zhou B, Dai X. Nfatc1's Role in Mammary Epithelial Morphogenesis and Basal Stem/progenitor Cell Self-renewal. J Mammary Gland Biol Neoplasia 2021; 26:357-365. [PMID: 34932179 PMCID: PMC8858291 DOI: 10.1007/s10911-021-09502-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Accepted: 11/05/2021] [Indexed: 11/26/2022] Open
Abstract
Mammary gland is an outstanding system to study the regulatory mechanisms governing adult epithelial stem cell activity. Stem cells in the basal layer of the mammary gland fuel the morphogenesis and regeneration of a complex epithelial network during development and upon transplantation. The self-renewal of basal stem/progenitor cells is subjected to regulation by both cell-intrinsic and extrinsic mechanisms. Nfatc1 is a transcription factor that regulates breast tumorigenesis and metastasis, but its role in mammary epithelial development and stem cell function has not been investigated. Here we show that Nfatc1 is expressed in a small subset of mammary basal epithelial cells and its epithelial-specific deletion results in mild defects in side branching and basal-luminal cell balance. Moreover, Nfatc1-deficient basal cells exhibit reduced colony forming ability in vitro and somewhat compromised regenerative potential upon transplantation. Thus, our study provides evidence for a detectable yet non-essential role of Nfatc1 in mammary epithelial morphogenesis and basal stem/progenitor cell self-renewal.
Collapse
Affiliation(s)
- Melissa McNeil
- Department of Biological Chemistry, School of Medicine, University of California, Irvine, CA, 92697, USA
| | - Yingying Han
- Department of Biological Chemistry, School of Medicine, University of California, Irvine, CA, 92697, USA
| | - Peng Sun
- Department of Biological Chemistry, School of Medicine, University of California, Irvine, CA, 92697, USA
| | - Kazuhide Watanabe
- Department of Biological Chemistry, School of Medicine, University of California, Irvine, CA, 92697, USA
| | - Jun Jiang
- Department of Biological Chemistry, School of Medicine, University of California, Irvine, CA, 92697, USA
| | - Natasha Chen
- Department of Biological Chemistry, School of Medicine, University of California, Irvine, CA, 92697, USA
| | - Zhengquan Yu
- State Key Laboratories for Agrobiotechnology, Department of Nutrition and Health, College of Biological Sciences, China Agricultural University, Beijing, 100193, China
| | - Bin Zhou
- Departments of Genetics, Pediatrics, and Medicine (Cardiology), The Wilf Cardiovascular Research Institute, The Institute for Aging Research, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
| | - Xing Dai
- Department of Biological Chemistry, School of Medicine, University of California, Irvine, CA, 92697, USA.
| |
Collapse
|
50
|
Shen T, Yue C, Wang X, Wang Z, Wu Y, Zhao C, Chang P, Sun X, Wang W. NFATc1 promotes epithelial-mesenchymal transition and facilitates colorectal cancer metastasis by targeting SNAI1. Exp Cell Res 2021; 408:112854. [PMID: 34597678 DOI: 10.1016/j.yexcr.2021.112854] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2021] [Revised: 09/23/2021] [Accepted: 09/24/2021] [Indexed: 01/16/2023]
Abstract
Metastatic recurrence remains a major cause of colorectal cancer (CRC) mortality. In this study, we investigated the mechanistic role of nuclear factor of activated T cells 1 (NFATc1) in CRC metastasis. First, we explored the potential role of NFATc1 in CRC using bioinformatics and hypothesized that NFATc1 might play different roles at different stages of CRC development. Then, we examined the relative expression of NFATc1 in 25 CRC tissues and adjacent normal tissues, and further analyzed the correlation between NFATc1 expression levels and clinical stages in 120 CRC patients. The role of NFATc1 in CRC metastasis and the molecular mechanisms were investigated in both in vitro and in vivo models. Our results showed that the expression of NFATc1 was increased in metastatic CRC tissues and positively associated with clinical stages (stage I vs. stage II, III or IV) of CRC. Overexpression of NFATc1 promoted CRC cell migration, invasion, and epithelial-mesenchymal transition (EMT). Moreover, SNAI1 was verified as the direct transcriptional target of NFATc1 and interacted with SLUG to promote EMT. Remarkably, our lung and liver metastasis mouse model demonstrated that NFATc1 overexpression accelerated CRC metastasis, and treatment with FK506, a calcineurin-NFAT pathway inhibitor, could suppress CRC metastasis in vivo. Taken together, our findings suggest that NFATc1 could transcriptionally activate SNAI1, which in turn interacts with SLUG to mediate EMT to promote CRC metastasis. Thus, making NFATc1 a promising therapeutic target in the treatment of metastatic CRC.
Collapse
Affiliation(s)
- Tianli Shen
- Department of General Surgery, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi Province, China
| | - Chenyang Yue
- Department of Biology, York University, Toronto, Ontario, Canada
| | - Xingjie Wang
- Department of General Surgery, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi Province, China
| | - Zijun Wang
- Department of General Surgery, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi Province, China
| | - Yunhua Wu
- Department of General Surgery, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi Province, China
| | - Chenye Zhao
- Department of General Surgery, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi Province, China
| | - Pengkang Chang
- Department of Hepatobiliary Surgery, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi Province, China
| | - Xuejun Sun
- Department of General Surgery, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi Province, China.
| | - Wei Wang
- Department of General Surgery, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi Province, China.
| |
Collapse
|