1
|
Tan CT, Lim CY, Lay K. Modelling Human Hair Follicles-Lessons from Animal Models and Beyond. BIOLOGY 2024; 13:312. [PMID: 38785794 PMCID: PMC11117913 DOI: 10.3390/biology13050312] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Revised: 04/26/2024] [Accepted: 04/27/2024] [Indexed: 05/25/2024]
Abstract
The hair follicle is a specialized appendage of the skin that is critical for multiple functions, including thermoregulation, immune surveillance, and sebum production. Mammals are born with a fixed number of hair follicles that develop embryonically. Postnatally, these hair follicles undergo regenerative cycles of regression and growth that recapitulate many of the embryonic signaling pathways. Furthermore, hair cycles have a direct impact on skin regeneration in homeostasis, cutaneous wound healing, and disease conditions such as alopecia. Here, we review the current knowledge of hair follicle formation during embryonic development and the post-natal hair cycle, with an emphasis on the molecular signaling pathways underlying these processes. We then discuss efforts to capitalize on the field's understanding of in vivo mechanisms to bioengineer hair follicles or hair-bearing skin in vitro and how such models may be further improved to develop strategies for hair regeneration.
Collapse
Affiliation(s)
- Chew Teng Tan
- A*STAR Skin Research Labs (A*SRL), Agency for Science, Technology and Research (A*STAR), 8A Biomedical Grove, #06-06 Immunos, Singapore 138648, Singapore
| | - Chin Yan Lim
- A*STAR Skin Research Labs (A*SRL), Agency for Science, Technology and Research (A*STAR), 8A Biomedical Grove, #06-06 Immunos, Singapore 138648, Singapore
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117596, Singapore
| | - Kenneth Lay
- A*STAR Skin Research Labs (A*SRL), Agency for Science, Technology and Research (A*STAR), 8A Biomedical Grove, #06-06 Immunos, Singapore 138648, Singapore
| |
Collapse
|
2
|
Qi WH, Liu T, Zheng CL, Zhao Q, Zhou N, Zhao GJ. Identification of Potential miRNA-mRNA Regulatory Network Associated with Growth and Development of Hair Follicles in Forest Musk Deer. Animals (Basel) 2023; 13:3869. [PMID: 38136906 PMCID: PMC10740511 DOI: 10.3390/ani13243869] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Revised: 12/05/2023] [Accepted: 12/13/2023] [Indexed: 12/24/2023] Open
Abstract
In this study, sRNA libraries and mRNA libraries of HFs of FMD were constructed and sequenced using an Illumina HiSeq 2500, and the expression profiles of miRNAs and genes in the HFs of FMD were obtained at the anagen and catagen stages. In total, 565 differentially expressed unigenes (DEGs) were identified, 90 of which were upregulated and 475 of which were downregulated. In the BP category of GO enrichment, the DEGs were enriched in the processes related to HF development and differentiation, including the hair cycle regulation and processes, HF development, skin epidermis development, regulation of HF development, skin development, the Wnt signaling pathway, and the BMP signaling pathway. Through KEGG analysis it was found that DEGs were significantly enriched in pathways associated with HF development and growth. A total of 186 differentially expressed miRNAs (DEmiRNAs) were screened (p < 0.05) in the HFs of FMD at the anagen stage vs. the catagen stage, 33 of which were upregulated and 153 of which were downregulated. Through DEmiRNA-mRNA association analysis, we found DEmiRNAs and target genes that mainly play regulatory roles in HF development and growth. The enrichment analysis of DEmiRNA target genes revealed similarities with the enrichment results of DEGs associated with HF development. Notably, both sets of genes were enriched in key pathways such as the Notch signaling pathway, melanogenesis, the cAMP signaling pathway, and cGMP-PKG. To validate our findings, we selected 11 DEGs and 11 DEmiRNAs for experimental verification using RT-qPCR. The results of the experimental validation were consistent with the RNA-Seq results.
Collapse
Affiliation(s)
- Wen-Hua Qi
- College of Biological and Food Engineering, Chongqing Three Gorges University, Chongqing 404100, China; (W.-H.Q.); (T.L.); (Q.Z.)
| | - Ting Liu
- College of Biological and Food Engineering, Chongqing Three Gorges University, Chongqing 404100, China; (W.-H.Q.); (T.L.); (Q.Z.)
| | - Cheng-Li Zheng
- Sichuan Institute of Musk Deer Breeding, Chengdu 611830, China;
| | - Qi Zhao
- College of Biological and Food Engineering, Chongqing Three Gorges University, Chongqing 404100, China; (W.-H.Q.); (T.L.); (Q.Z.)
| | - Nong Zhou
- College of Biological and Food Engineering, Chongqing Three Gorges University, Chongqing 404100, China; (W.-H.Q.); (T.L.); (Q.Z.)
| | - Gui-Jun Zhao
- Chongqing Institute of Medicinal Plant Cultivation, Chongqing 408435, China
| |
Collapse
|
3
|
Jeon J, Lee H, Jeon MS, Kim SJ, Choi C, Kim KW, Yang DJ, Lee S, Bae YS, Choi WI, Jung J, Eyun SI, Yang S. Blockade of Activin Receptor IIB Protects Arthritis Pathogenesis by Non-Amplification of Activin A-ACVR2B-NOX4 Axis Pathway. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2205161. [PMID: 36950748 DOI: 10.1002/advs.202205161] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Revised: 02/10/2023] [Indexed: 05/18/2023]
Abstract
Although activin receptor IIB (ACVR2B) is emerging as a novel pathogenic receptor, its ligand and assembled components (or assembly) are totally unknown in the context of osteoarthritis (OA) pathogenesis. The present results suggest that upregulation of ACVR2B and its assembly could affect osteoarthritic cartilage destruction. It is shown that the ACVR2B ligand, activin A, regulates catabolic factor expression through ACVR2B in OA development. Activin A Tg mice (Col2a1-Inhba) exhibit enhanced cartilage destruction, whereas heterozygous activin A KO mice (Inhba+/- ) show protection from cartilage destruction. In silico analysis suggests that the Activin A-ACVR2B axis is involved in Nox4-dependent ROS production. Activin A Tg:Nox4 KO (Col2a1-Inhba:Nox4-/- ) mice show inhibition of experimental OA pathogenesis. NOX4 directly binds to the C-terminal binding site on ACVR2B-ACVR1B and amplifies the pathogenic signal for cartilage destruction through SMAD2/3 signaling. Together, the findings reveal that the ACVR2B assembly, which comprises Activin A, ACVR2B, ACVR1B, Nox4, and AP-1-induced HIF-2α, accelerates OA development. Furthermore, it is shown that shRNA-mediated ACVR2B knockdown or trapping ligands of ACVR2B abrogate OA development by competitively disrupting the ACVR2B-Activin A interaction. These results suggest that the ACVR2B assembly is required to amplify osteoarthritic cartilage destruction and could be a potential therapeutic target in efforts to treat OA.
Collapse
Affiliation(s)
- Jimin Jeon
- Department of Biological Sciences, Sungkyunkwan University, Suwon, 16419, Republic of Korea
- CIRNO, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Hyemi Lee
- Department of Biological Sciences, Sungkyunkwan University, Suwon, 16419, Republic of Korea
- CIRNO, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Min-Seung Jeon
- Department of Life Science, Chung-Ang University, Seoul, 06974, Republic of Korea
| | - Seok-Jung Kim
- Department of Orthopaedic Surgery, Uijeongbu St. Mary's Hospital, The Catholic University of Korea College of Medicine, Uijeongbu, 11765, Republic of Korea
| | - Cham Choi
- MicroCT Applications, 3rd floor, 11, Sumyeong-ro 1-gil, Gangseo-gu, Seoul, 07644, Republic of Korea
| | - Ki Woo Kim
- Department of Oral Biology, Yonsei University College of Dentistry, Seoul, 03722, Republic of Korea
- Department of Applied Biological Science, BK21 FOUR, Yonsei University College of Dentistry, Seoul, 03722, Republic of Korea
| | - Dong Joo Yang
- Department of Oral Biology, Yonsei University College of Dentistry, Seoul, 03722, Republic of Korea
- Department of Applied Biological Science, BK21 FOUR, Yonsei University College of Dentistry, Seoul, 03722, Republic of Korea
| | - Sangho Lee
- Department of Biological Sciences, Sungkyunkwan University, Suwon, 16419, Republic of Korea
- CIRNO, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Yong-Soo Bae
- Department of Biological Sciences, Sungkyunkwan University, Suwon, 16419, Republic of Korea
- CIRNO, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Won Il Choi
- Center for Bio-Healthcare Materials, Bio-Convergence Materials R&D Division, Korea Institute of Ceramic Engineering and Technology, Cheongju, Chungbuk, 28160, Republic of Korea
| | - Juyeon Jung
- Bionanotechnology Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 34141, Republic of Korea
| | - Seong-Il Eyun
- Department of Life Science, Chung-Ang University, Seoul, 06974, Republic of Korea
| | - Siyoung Yang
- Department of Biological Sciences, Sungkyunkwan University, Suwon, 16419, Republic of Korea
- CIRNO, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| |
Collapse
|
4
|
Saeki K, Qiu W, Friedman RA, Pan S, Lu J, Ichimiya S, Chio IIC, Shawber CJ, Kitajewski J, Hu J, Su GH. Inactivation of Notch4 Attenuated Pancreatic Tumorigenesis in Mice. CANCER RESEARCH COMMUNICATIONS 2022; 2:1601-1616. [PMID: 36970723 PMCID: PMC10035463 DOI: 10.1158/2767-9764.crc-22-0106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Revised: 09/17/2022] [Accepted: 11/10/2022] [Indexed: 11/16/2022]
Abstract
Expression of the Notch family of receptors is often upregulated in pancreatic ductal adenocarcinoma (PDAC). In this study, we focused on Notch4, which had not been investigated in PDAC. We generated KC (LSL-KrasG12D;p48-Cre), N4 - / - KC (Notch4- / -;LSL-KrasG12D;p48-Cre), PKC (p16fl/fl;LSL-KrasG12D;p48-Cre), and N4 - / - PKC (Notch4-/ -; p16fl/f l;LSL-KrasG12D;p48-Cre) genetically engineered mouse models (GEMM). We performed caerulein treatment in both KC and N4 - / - KC mice, and the development of acinar-to-ductal metaplasia (ADM) and pancreatic intraepithelial neoplasia (PanIN) lesions were significantly diminished in the N4 - / - KC than in the KC GEMM (P = 0.01). This in vivo result was validated by in vitro ADM induction of the explant cultures of pancreatic acinar cells from the N4 - / - KC and KC mice (P < 0.001), confirming that Notch4 is an important contributor to early pancreatic tumorigenesis. To evaluate the role of Notch4 in the later stage of pancreatic tumorigenesis, we compared the PKC and N4 - / - PKC mice. The N4 - / - PKC mice had better overall survival (P = 0.012) and significantly reduced tumor burden (PanIN: P = 0.018 at 2 months, PDAC: P = 0.039 at 5 months) compared with the PKC GEMM. RNA-sequencing analysis of pancreatic tumor cell lines derived from the PKC and N4 - / - PKC GEMMs revealed that 408 genes were differentially expressed (FDR < 0.05) and Pcsk5 is a potential downstream effector of the Notch4 signaling pathway (P < 0.001). Low expression of Pcsk5 positively correlates with good survival in patients with PDAC (P = 0.028). We have identified a novel role for Notch4 signaling with tumor-promoting function in pancreatic tumorigenesis. Our study also uncovered a novel association between Pcsk5 and Notch4 signaling in PDAC. Significance We demonstrated that global inactivation of Notch4 significantly improved the survival of an aggressive mouse model for PDAC and provided preclinical evidence that Notch4 and Pcsk5 are novel targets for PDAC therapies.
Collapse
Affiliation(s)
- Kiyoshi Saeki
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, New York
- Department of Pathology & Cell Biology, Columbia University Irving Medical Center, New York, New York
| | - Wanglong Qiu
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, New York
- Department of Pathology & Cell Biology, Columbia University Irving Medical Center, New York, New York
| | - Richard A. Friedman
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, New York
- Department of Biomedical Informatics, Columbia University Irving Medical Center, New York, New York
| | - Samuel Pan
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, New York
- Department of Biostatistics, Columbia University Irving Medical Center, New York, New York
| | - Jordan Lu
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, New York
- Institute for Cancer Genetics, Columbia University Irving Medical Center, New York, New York
| | - Shu Ichimiya
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, New York
- Institute for Cancer Genetics, Columbia University Irving Medical Center, New York, New York
| | - Iok In Christine Chio
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, New York
- Institute for Cancer Genetics, Columbia University Irving Medical Center, New York, New York
| | - Carrie J. Shawber
- Deparments of Obstetrics and Gynecology and Surgery, Columbia University Irving Medical Center, New York, New York
| | - Jan Kitajewski
- Department of Physiology and Biophysics, University of Illinois Cancer Center, University of Illinois Chicago, Chicago, Illinois
| | - Jianhua Hu
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, New York
- Department of Biostatistics, Columbia University Irving Medical Center, New York, New York
| | - Gloria H. Su
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, New York
- Department of Pathology & Cell Biology, Columbia University Irving Medical Center, New York, New York
| |
Collapse
|
5
|
Lyu Y, Ge Y. Toward Elucidating Epigenetic and Metabolic Regulation of Stem Cell Lineage Plasticity in Skin Aging. Front Cell Dev Biol 2022; 10:903904. [PMID: 35663405 PMCID: PMC9160930 DOI: 10.3389/fcell.2022.903904] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Accepted: 04/21/2022] [Indexed: 11/13/2022] Open
Abstract
Skin is the largest organ in human body, harboring a plethora of cell types and serving as the organismal barrier. Skin aging such as wrinkling and hair graying is graphically pronounced, and the molecular mechanisms behind these phenotypic manifestations are beginning to unfold. As in many other organs and tissues, epigenetic and metabolic deregulations have emerged as key aging drivers. Particularly in the context of the skin epithelium, the epigenome and metabolome coordinately shape lineage plasticity and orchestrate stem cell function during aging. Our review discusses recent studies that proposed molecular mechanisms that drive the degeneration of hair follicles, a major appendage of the skin. By focusing on skin while comparing it to model organisms and adult stem cells of other tissues, we summarize literature on genotoxic stress, nutritional sensing, metabolic rewiring, mitochondrial activity, and epigenetic regulations of stem cell plasticity. Finally, we speculate about the rejuvenation potential of rate-limiting upstream signals during aging and the dominant role of the tissue microenvironment in dictating aged epithelial stem cell function.
Collapse
Affiliation(s)
| | - Yejing Ge
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| |
Collapse
|
6
|
Lv X, Chen W, Wang S, Cao X, Yuan Z, Getachew T, Mwacharo JM, Haile A, Sun W. Integrated Hair Follicle Profiles of microRNAs and mRNAs to Reveal the Pattern Formation of Hu Sheep Lambskin. Genes (Basel) 2022; 13:genes13020342. [PMID: 35205386 PMCID: PMC8872417 DOI: 10.3390/genes13020342] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Revised: 02/05/2022] [Accepted: 02/10/2022] [Indexed: 02/05/2023] Open
Abstract
Hair follicle development is closely associated with wool curvature. Current studies reveal the crucial role of microRNAs (miRNAs) in hair follicle growth and development. However, few studies are known regarding their role in wool curvature. To reveal the potential roles of miRNAs in Hu sheep lambskin with different patterns, a total of 37 differentially expressed (DE) miRNAs were identified in hair follicles between small waves (SM) and straight wool (ST) groups using RNA-seq. Through functional enrichment and miRNA-mRNA co-expression analysis, some key miRNAs (oar-miR-143, oar-miR-200b, oar-miR-10a, oar-miR-181a, oar-miR-10b, oar-miR-125b, etc.) and miRNA-mRNA pairs (miR-125b target CD34, miR-181a target FGF12, LMO3, miR-200b target ZNF536, etc.) were identified. Though direct or indirect ways affecting hair follicle development, these miRNAs and mRNAs may have possible effects on wool curvature, and this study thus provides valuable insight on potential pattern formation.
Collapse
Affiliation(s)
- Xiaoyang Lv
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of Ministry of Education of China, Yangzhou University, Yangzhou 225009, China; (X.L.); (X.C.); (Z.Y.)
| | - Weihao Chen
- College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China; (W.C.); (S.W.)
| | - Shanhe Wang
- College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China; (W.C.); (S.W.)
| | - Xiukai Cao
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of Ministry of Education of China, Yangzhou University, Yangzhou 225009, China; (X.L.); (X.C.); (Z.Y.)
| | - Zehu Yuan
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of Ministry of Education of China, Yangzhou University, Yangzhou 225009, China; (X.L.); (X.C.); (Z.Y.)
| | - Tesfaye Getachew
- International Centre for Agricultural Research in the Dry Areas, Addis Ababa 999047, Ethiopia; (T.G.); (J.M.M.); (A.H.)
| | - Joram M. Mwacharo
- International Centre for Agricultural Research in the Dry Areas, Addis Ababa 999047, Ethiopia; (T.G.); (J.M.M.); (A.H.)
| | - Aynalem Haile
- International Centre for Agricultural Research in the Dry Areas, Addis Ababa 999047, Ethiopia; (T.G.); (J.M.M.); (A.H.)
| | - Wei Sun
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of Ministry of Education of China, Yangzhou University, Yangzhou 225009, China; (X.L.); (X.C.); (Z.Y.)
- College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China; (W.C.); (S.W.)
- Correspondence: ; Tel.: +86-139-5275-0912
| |
Collapse
|
7
|
Zandonadi FS, Yokoo S, Granato DC, Rivera C, Macedo CCS, Soares CD, Carnielli CM, Domingues RR, Pauletti BA, Consonni SR, Colleta RD, Paes Leme AF. Follistatin-related protein 1 interacting partner of Syndecan-1 promotes an aggressive phenotype on Oral Squamous cell carcinoma (OSCC) models. J Proteomics 2022; 254:104474. [PMID: 34990821 DOI: 10.1016/j.jprot.2021.104474] [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: 07/11/2021] [Revised: 12/16/2021] [Accepted: 12/29/2021] [Indexed: 12/24/2022]
Abstract
Syndecans belong to the family of transmembrane heparan sulfate proteoglycans and are associated with many physiopathological processes, including oral cancer. As previously shown soluble syndecan-1 (SDC1) fragments and synthetic SDC1 peptide were able to induce cell migration in oral cancer cell lines. In order to explore the role of SDC1 in oral cancer, we have investigated SDC1 interacting partners and its functional role in oral cancer models. Here we have shown that SDC1 interacts with follistatin-related protein 1 (FSTL1) by its ectodomain (ectoSDC1) and extracellular juxtamembrane peptide (pepSDC1) and that their transcript levels can affect tumor events. Using orthotopic mouse model we identified that the knock-down for FSTL1 (shFSTL1) or for both FSTL1 and SDC1 (sh2KD) produced less aggressive and infiltrative tumors, with lower keratinization deposition, but with increased levels of epithelial-mesenchymal transition and proliferation compared to control and SDC1 knock-down. Based on cell culture assays, we suggest that the shFSTL1 effect on tumor tissues might be from significant increase of mRNA levels of Activin A (ActA) and its resceptors. This study shows for the first time two different complexes, SDC1 and FSTL1; pepSDC1 and FSTL1, exhibiting a close relationship in cell signaling events, as FSTL1 promotes a more aggressive phenotype. SIGNIFICANCE: This work contributes to the understanding of new SDC1 functions, based on the investigation of protein-protein complex formation in Oral Squamous cell carcinoma (OSCC) models. The FSTL1 identification, as an interacting partner of SDC1 ectodomain and of its derived peptide promotes molecular events that favors cancer development and progression, as highlighted by Activin A (ActA) and Epithelial-mesenchymal transition (EMT) gene expression and by changes in the phenotype of orthotopic OSCC mouse tumor tissues when SDC1-FSTL1 expression is modulated.
Collapse
Affiliation(s)
- Flávia S Zandonadi
- Laboratório de Espectrometria de Massas, Laboratório Nacional de Biociências, LNBio, CNPEM, Campinas, SP, Brazil
| | - Sami Yokoo
- Laboratório de Espectrometria de Massas, Laboratório Nacional de Biociências, LNBio, CNPEM, Campinas, SP, Brazil
| | - Daniela Campos Granato
- Laboratório de Espectrometria de Massas, Laboratório Nacional de Biociências, LNBio, CNPEM, Campinas, SP, Brazil
| | - César Rivera
- Laboratório de Espectrometria de Massas, Laboratório Nacional de Biociências, LNBio, CNPEM, Campinas, SP, Brazil
| | - Carolina Carneiro Soares Macedo
- Laboratório de Espectrometria de Massas, Laboratório Nacional de Biociências, LNBio, CNPEM, Campinas, SP, Brazil; Faculdade de Odontologia de Piracicaba, Universidade Estadual de Campinas, UNICAMP, Piracicaba, SP, Brazil
| | - Ciro Dantas Soares
- Faculdade de Odontologia de Piracicaba, Universidade Estadual de Campinas, UNICAMP, Piracicaba, SP, Brazil
| | - Carolina Moretto Carnielli
- Laboratório de Espectrometria de Massas, Laboratório Nacional de Biociências, LNBio, CNPEM, Campinas, SP, Brazil
| | - Romênia Ramos Domingues
- Laboratório de Espectrometria de Massas, Laboratório Nacional de Biociências, LNBio, CNPEM, Campinas, SP, Brazil
| | - Bianca A Pauletti
- Laboratório de Espectrometria de Massas, Laboratório Nacional de Biociências, LNBio, CNPEM, Campinas, SP, Brazil
| | - Sílvio Roberto Consonni
- Laboratório de Espectrometria de Massas, Laboratório Nacional de Biociências, LNBio, CNPEM, Campinas, SP, Brazil
| | - Ricardo D Colleta
- Faculdade de Odontologia de Piracicaba, Universidade Estadual de Campinas, UNICAMP, Piracicaba, SP, Brazil
| | - Adriana F Paes Leme
- Laboratório de Espectrometria de Massas, Laboratório Nacional de Biociências, LNBio, CNPEM, Campinas, SP, Brazil.
| |
Collapse
|
8
|
Qiu W, Kuo CY, Tian Y, Su GH. Dual Roles of the Activin Signaling Pathway in Pancreatic Cancer. Biomedicines 2021; 9:biomedicines9070821. [PMID: 34356885 PMCID: PMC8301451 DOI: 10.3390/biomedicines9070821] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Revised: 06/29/2021] [Accepted: 07/08/2021] [Indexed: 12/12/2022] Open
Abstract
Activin, a member of the TGF-β superfamily, is involved in many physiological processes, such as embryonic development and follicle development, as well as in multiple human diseases including cancer. Genetic mutations in the activin signaling pathway have been reported in many cancer types, indicating that activin signaling plays a critical role in tumorigenesis. Recent evidence reveals that activin signaling may function as a tumor-suppressor in tumor initiation, and a promoter in the later progression and metastasis of tumors. This article reviews many aspects of activin, including the signaling cascade of activin, activin-related proteins, and its role in tumorigenesis, particularly in pancreatic cancer development. The mechanisms regulating its dual roles in tumorigenesis remain to be elucidated. Further understanding of the activin signaling pathway may identify potential therapeutic targets for human cancers and other diseases.
Collapse
Affiliation(s)
- Wanglong Qiu
- The Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY 10032, USA; (W.Q.); (C.K.); (Y.T.)
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Chia-Yu Kuo
- The Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY 10032, USA; (W.Q.); (C.K.); (Y.T.)
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Yu Tian
- The Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY 10032, USA; (W.Q.); (C.K.); (Y.T.)
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Gloria H. Su
- The Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY 10032, USA; (W.Q.); (C.K.); (Y.T.)
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, NY 10032, USA
- Department of Otolaryngology and Head and Neck Surgery, Columbia University Irving Medical Center, New York, NY 10032, USA
- Correspondence:
| |
Collapse
|
9
|
de Groot SC, Ulrich MMW, Gho CG, Huisman MA. Back to the Future: From Appendage Development Toward Future Human Hair Follicle Neogenesis. Front Cell Dev Biol 2021; 9:661787. [PMID: 33912569 PMCID: PMC8075059 DOI: 10.3389/fcell.2021.661787] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Accepted: 03/18/2021] [Indexed: 11/13/2022] Open
Abstract
Hair disorders such as alopecia and hirsutism often impact the social and psychological well-being of an individual. This also holds true for patients with severe burns who have lost their hair follicles (HFs). HFs stimulate proper wound healing and prevent scar formation; thus, HF research can benefit numerous patients. Although hair development and hair disorders are intensively studied, human HF development has not been fully elucidated. Research on human fetal material is often subject to restrictions, and thus development, disease, and wound healing studies remain largely dependent on time-consuming and costly animal studies. Although animal experiments have yielded considerable and useful information, it is increasingly recognized that significant differences exist between animal and human skin and that it is important to obtain meaningful human models. Human disease specific models could therefore play a key role in future therapy. To this end, hair organoids or hair-bearing skin-on-chip created from the patient’s own cells can be used. To create such a complex 3D structure, knowledge of hair genesis, i.e., the early developmental process, is indispensable. Thus, uncovering the mechanisms underlying how HF progenitor cells within human fetal skin form hair buds and subsequently HFs is of interest. Organoid studies have shown that nearly all organs can be recapitulated as mini-organs by mimicking embryonic conditions and utilizing the relevant morphogens and extracellular matrix (ECM) proteins. Therefore, knowledge of the cellular and ECM proteins in the skin of human fetuses is critical to understand the evolution of epithelial tissues, including skin appendages. This review aims to provide an overview of our current understanding of the cellular changes occurring during human skin and HF development. We further discuss the potential implementation of this knowledge in establishing a human in vitro model of a full skin substitute containing hair follicles and the subsequent translation to clinical use.
Collapse
Affiliation(s)
- Simon C de Groot
- Association of Dutch Burn Centres, Beverwijk, Netherlands.,Hair Science Institute, Maastricht, Netherlands
| | | | - Coen G Gho
- Hair Science Institute, Maastricht, Netherlands
| | - Margriet A Huisman
- Department of Otorhinolaryngology and Head & Neck Surgery, Leiden University Medical Center, Leiden, Netherlands
| |
Collapse
|
10
|
Saxena N, Mok KW, Rendl M. An updated classification of hair follicle morphogenesis. Exp Dermatol 2020; 28:332-344. [PMID: 30887615 DOI: 10.1111/exd.13913] [Citation(s) in RCA: 90] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Accepted: 03/13/2019] [Indexed: 12/12/2022]
Abstract
Hair follicle (HF) formation in developing embryonic skin requires stepwise signalling between the epithelial epidermis and mesenchymal dermis, and their specialized derivatives, the placode/germ/peg and dermal condensate/papilla, respectively. Classically, distinct stages of HF morphogenesis have been defined, in the mouse model, based on (a) changes in cell morphology and aggregation; (b) expression of few known molecular markers; (c) the extent of follicle downgrowth; and (d) the presence of differentiating cell types. Refined genetic strategies and recent emerging technologies, such as live imaging and transcriptome analyses of isolated cell populations or single cells, have enabled a closer dissection of the signalling requirements at different stages of HF formation, particularly early on. They have also led to the discovery of precursor cells for placode, dermal condensate and future bulge stem cells that, combined with molecular insights into their fate specification and subsequent formation, serve as novel landmarks for early HF morphogenetic events and studies of the signalling networks mediating these processes. In this review, we integrate the emergence of HF precursor cell states and novel molecular markers of fate and formation to update the widely used 20-year-old seminal classification guide of HF morphogenetic stages by Paus et al. We then temporally describe the latest insights into the early cellular and molecular events and signalling requirements for HF morphogenesis in relation to one another in a holistic manner.
Collapse
Affiliation(s)
- Nivedita Saxena
- Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, New York.,Department of Cell, Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, New York.,Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Ka-Wai Mok
- Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, New York.,Department of Cell, Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Michael Rendl
- Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, New York.,Department of Cell, Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, New York.,Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, New York.,Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York, New York
| |
Collapse
|
11
|
Oyelami FO, Zhao Q, Xu Z, Zhang Z, Sun H, Zhang Z, Ma P, Wang Q, Pan Y. Haplotype Block Analysis Reveals Candidate Genes and QTLs for Meat Quality and Disease Resistance in Chinese Jiangquhai Pig Breed. Front Genet 2020; 11:752. [PMID: 33101353 PMCID: PMC7498712 DOI: 10.3389/fgene.2020.00752] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2019] [Accepted: 06/23/2020] [Indexed: 11/19/2022] Open
Abstract
The Jiangquhai (JQ) pig breed is one of the most widely recognized pig populations in China due to its unique and dominant characteristics. In this study, we examined the extent of Linkage disequilibrium (LD) and haplotype block structure of the JQ pig breed, and scanned the blocks for possible genes underlying important QTLs that could either be responsible for some adaptive features in these pigs or might have undergone some selection pressure. We compared some of our results with other Chinese and Western pig breeds. The results show that the JQ breed had the highest total block length (349.73 Mb ≈ 15% of its genome), and the coverage rate of blocks in most of its chromosomes was larger than those of other breeds except for Sus scrofa chromosome 4 (SSC4), SSC6, SSC7, SSC8, SSC10, SSC12, SSC13, SSC14, SSC17, SSC18, and SSCX. Moreover, the JQ breed had more SNPs that were clustered into haplotype blocks than the other breeds examined in this study. Our shared and unique haplotype block analysis revealed that the Hongdenglong (HD) breed had the lowest percentage of shared haplotype blocks while the Shanzhu (SZ) breed had the highest. We found that the JQ breed had an average r2 > 0.2 at SNPs distances 10–20 kb and concluded that about 120,000–240,000 SNPs would be needed for a successful GWAS in the breed. Finally, we detected a total of 88 genes harbored by selected haplotype blocks in the JQ breed, of which only 4 were significantly enriched (p-value ≤ 0.05). These genes were significantly enriched in 2 GO terms (p-value < 0.01), and 2 KEGG pathways (p-value < 0.02). Most of these enriched genes were related to health. Also, most of the overlapping QTLs detected in the haplotype blocks were related to meat and carcass quality, as well as health, with a few of them relating to reproduction and production. These results provide insights into the genetic architecture of some adaptive and meat quality traits observed in the JQ pig breed and also revealed the pattern of LD in the genome of the pig. Our result provides significant guidance for improving the statistical power of GWAS and optimizing the conservation strategy for this JQ pig breed.
Collapse
Affiliation(s)
- Favour Oluwapelumi Oyelami
- Department of Animal Science, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai, China
| | - Qingbo Zhao
- Department of Animal Science, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai, China
| | - Zhong Xu
- Department of Animal Science, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai, China
| | - Zhe Zhang
- Department of Animal Breeding and Reproduction, College of Animal Science, Zhejiang University, Hangzhou, China
| | - Hao Sun
- Department of Animal Science, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai, China
| | - Zhenyang Zhang
- Department of Animal Science, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai, China
| | - Peipei Ma
- Department of Animal Science, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai, China
| | - Qishan Wang
- Department of Animal Science, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai, China.,Department of Animal Breeding and Reproduction, College of Animal Science, Zhejiang University, Hangzhou, China
| | - Yuchun Pan
- Department of Animal Science, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai, China.,Department of Animal Breeding and Reproduction, College of Animal Science, Zhejiang University, Hangzhou, China
| |
Collapse
|
12
|
Expression profile analysis to identify circular RNA expression signatures in hair follicle of Hu sheep lambskin. Genomics 2020; 112:4454-4462. [PMID: 32768426 DOI: 10.1016/j.ygeno.2020.07.046] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 07/01/2020] [Accepted: 07/29/2020] [Indexed: 11/24/2022]
Abstract
CircRNAs are involved in the regulation of various cellular and biological processes, but none of the studies have focused on hair follicle in sheep. In this study, the expression profile of circRNAs between small waves (SM) and straight wool (ST) groups was investigated using RNA-Seq. The results showed that a total of 5,527 circRNAs were identified and 114 of them were differentially expressed between two groups. Enrichment analysis revealed that the host genes with DE circRNAs were mainly enriched in TGF-beta pathway, Notch pathway. Miranda software was used to found that 129 miRNAs might be binding to 114 DE circRNAs, including miR-10a, miR-143, miR-let-7a, miR-199a-3p, miR-200a, which also had important influence on hair follicle morphogenesis. Furthermore, the coding potential of circRNAs was predicted, and 11 circRNAs were simultaneously identified with coding potential. In summary, circRNAs have important effects on hair follicle growth and development, and these results will provide a basis for molecular mechanism of pattern formation.
Collapse
|
13
|
Lin JB, Chen JF, Lai FC, Li X, Xie JB, Tu YR, Kang MQ. Involvement of activin a receptor type 1 (ACVR1) in the pathogenesis of primary focal hyperhidrosis. Biochem Biophys Res Commun 2020; 528:299-304. [PMID: 32473755 DOI: 10.1016/j.bbrc.2020.05.052] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Accepted: 05/07/2020] [Indexed: 12/29/2022]
Abstract
The pathogenesis of primary focal hyperhidrosis (PFH) is still not clear. PFH is thought to be a genetic disease. Whether activin A receptor type 1 (ACVR1) is involved in the pathogenesis of PFH is unknown. In this study, the expression of ACVR1 in sweat glands of patients with PAH was detected by western blot and immunofluorescence. The primary sweat gland cells obtained from primary axillary hyperhidrosis (PAH) patients were transfected with acvr1 vector. Cell proliferation, apoptosis and cell cycling of gland cells were measured after transfection with acvr1 vector. The mRNA and protein expression of aquaporin 5 (AQP5) and Na:K:2Cl Cotransporter 1 (NKCC1/SLC12A2) were detected. Our data showed that ACVR1 expression in axillary sweat gland tissue of PAH patients was significantly higher than that of normal control group. The function of ACVR1 was further investigated in the gland cells obtained from PAH patients. Compared with NC group, ACVR1 overexpression significantly promoted the proliferation of sweat gland cells and inhibited the apoptosis of sweat gland cells. Meanwhile, ACVR1 overexpression significantly reduced the percentage of cells in G0/G1 and G2/M phases, and increased the percentage of cells in S phase. In addition, ACVR1 overexpression significantly promoted the expression of AQP5 and NKCC1 at both mRNA and protein levels. Together, ACVR1 expression is related to PFH and ACVR1 overexpression can promote the proliferation of sweat gland cells and inhibit apoptosis by promoting the expression of AQP5 and NKCC1.
Collapse
Affiliation(s)
- Jian-Bo Lin
- Department of Thoracic Surgery, Fujian Medical University Union Hospital, No. 29 Xinquan Road, Gulou District, Fuzhou, People's Republic of China; Department of Thoracic Surgery, First Affiliated Hospital of Fujian Medical University, No. 20 Chazhong Road, Fuzhou, People's Republic of China
| | - Jian-Feng Chen
- Department of Thoracic Surgery, First Affiliated Hospital of Fujian Medical University, No. 20 Chazhong Road, Fuzhou, People's Republic of China
| | - Fan-Cai Lai
- Department of Thoracic Surgery, First Affiliated Hospital of Fujian Medical University, No. 20 Chazhong Road, Fuzhou, People's Republic of China
| | - Xu Li
- Department of Thoracic Surgery, First Affiliated Hospital of Fujian Medical University, No. 20 Chazhong Road, Fuzhou, People's Republic of China
| | - Jin-Bao Xie
- Department of Thoracic Surgery, First Affiliated Hospital of Fujian Medical University, No. 20 Chazhong Road, Fuzhou, People's Republic of China
| | - Yuan-Rong Tu
- Department of Thoracic Surgery, First Affiliated Hospital of Fujian Medical University, No. 20 Chazhong Road, Fuzhou, People's Republic of China
| | - Ming-Qiang Kang
- Department of Thoracic Surgery, Fujian Medical University Union Hospital, No. 29 Xinquan Road, Gulou District, Fuzhou, People's Republic of China.
| |
Collapse
|
14
|
Bta-miR-24-3p Controls the Myogenic Differentiation and Proliferation of Fetal, Bovine, Skeletal Muscle-Derived Progenitor Cells by Targeting ACVR1B. Animals (Basel) 2019; 9:ani9110859. [PMID: 31652908 PMCID: PMC6912306 DOI: 10.3390/ani9110859] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2019] [Revised: 10/16/2019] [Accepted: 10/18/2019] [Indexed: 12/23/2022] Open
Abstract
Simple Summary MicroRNAs play pivotal roles in skeletal muscle development, but the molecular basis of their functions in fetal bovine skeletal muscle development is largely unknown. Here, we report a mechanistic study of bta-miR-24-3p, a key miRNA regulator of the myogenic differentiation of fetal bovine platelet-derived growth factor receptor alpha negative (PDGFRα-) progenitor cells. We isolated progenitor cells from the bovine fetal longissimus dorsi muscle and purified them with PDGFRα antibodies to remove fibro-adipogenic progenitors. We observed elevated bta-miR-24-3p expression during differentiation, and bta-miR-24-3p overexpression led to promoted myogenic differentiation but suppressed proliferation. Moreover, activin receptor type 1B (ACVR1B) was identified as a direct target of bta-miR-24-3p, and ACVR1B-silencing cells exhibited similar phenotypes to bta-miR-24-3p-overexpressing bovine PDGFRα- progenitor cells. These results extended our understanding on the roles of miRNA in fetal muscle development. The method of removing fibro-adipogenic progenitors in our study will also provide useful information for other investigators. Abstract MicroRNAs modulate a variety of cellular events, including skeletal muscle development, but the molecular basis of their functions in fetal bovine skeletal muscle development is poorly understood. In this study, we report that bta-miR-24-3p promotes the myogenic differentiation of fetal bovine PDGFRα- progenitor cells. The expression of bta-miR-24-3p increased during myogenic differentiation. Overexpression of bta-miR-24-3p significantly promoted myogenic differentiation, but inhibited proliferation. A dual-luciferase assay identified ACVR1B as a direct target of bta-miR-24-3p. Similarly, knocking down ACVR1B by RNA interference also significantly inhibited proliferation and promoted the differentiation of bovine PDGFRα- progenitor cells. Thus, our study provides a mechanism in which bta-miR-24-3p regulates myogenesis by inhibiting ACVR1B expression.
Collapse
|
15
|
Bloise E, Ciarmela P, Dela Cruz C, Luisi S, Petraglia F, Reis FM. Activin A in Mammalian Physiology. Physiol Rev 2019; 99:739-780. [DOI: 10.1152/physrev.00002.2018] [Citation(s) in RCA: 78] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Activins are dimeric glycoproteins belonging to the transforming growth factor beta superfamily and resulting from the assembly of two beta subunits, which may also be combined with alpha subunits to form inhibins. Activins were discovered in 1986 following the isolation of inhibins from porcine follicular fluid, and were characterized as ovarian hormones that stimulate follicle stimulating hormone (FSH) release by the pituitary gland. In particular, activin A was shown to be the isoform of greater physiological importance in humans. The current understanding of activin A surpasses the reproductive system and allows its classification as a hormone, a growth factor, and a cytokine. In more than 30 yr of intense research, activin A was localized in female and male reproductive organs but also in other organs and systems as diverse as the brain, liver, lung, bone, and gut. Moreover, its roles include embryonic differentiation, trophoblast invasion of the uterine wall in early pregnancy, and fetal/neonate brain protection in hypoxic conditions. It is now recognized that activin A overexpression may be either cytostatic or mitogenic, depending on the cell type, with important implications for tumor biology. Activin A also regulates bone formation and regeneration, enhances joint inflammation in rheumatoid arthritis, and triggers pathogenic mechanisms in the respiratory system. In this 30-yr review, we analyze the evidence for physiological roles of activin A and the potential use of activin agonists and antagonists as therapeutic agents.
Collapse
Affiliation(s)
- Enrrico Bloise
- Department of Morphology, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil; Department of Experimental and Clinical Medicine, Università Politecnica delle Marche, Ancona, Italy; Department of Obstetrics and Gynecology, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil; Department of Molecular and Developmental Medicine, Obstetrics and Gynecological Clinic, University of Siena, Siena, Italy; and Department of Biomedical, Experimental and Clinical Sciences, Division of Obstetrics and
| | - Pasquapina Ciarmela
- Department of Morphology, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil; Department of Experimental and Clinical Medicine, Università Politecnica delle Marche, Ancona, Italy; Department of Obstetrics and Gynecology, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil; Department of Molecular and Developmental Medicine, Obstetrics and Gynecological Clinic, University of Siena, Siena, Italy; and Department of Biomedical, Experimental and Clinical Sciences, Division of Obstetrics and
| | - Cynthia Dela Cruz
- Department of Morphology, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil; Department of Experimental and Clinical Medicine, Università Politecnica delle Marche, Ancona, Italy; Department of Obstetrics and Gynecology, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil; Department of Molecular and Developmental Medicine, Obstetrics and Gynecological Clinic, University of Siena, Siena, Italy; and Department of Biomedical, Experimental and Clinical Sciences, Division of Obstetrics and
| | - Stefano Luisi
- Department of Morphology, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil; Department of Experimental and Clinical Medicine, Università Politecnica delle Marche, Ancona, Italy; Department of Obstetrics and Gynecology, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil; Department of Molecular and Developmental Medicine, Obstetrics and Gynecological Clinic, University of Siena, Siena, Italy; and Department of Biomedical, Experimental and Clinical Sciences, Division of Obstetrics and
| | - Felice Petraglia
- Department of Morphology, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil; Department of Experimental and Clinical Medicine, Università Politecnica delle Marche, Ancona, Italy; Department of Obstetrics and Gynecology, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil; Department of Molecular and Developmental Medicine, Obstetrics and Gynecological Clinic, University of Siena, Siena, Italy; and Department of Biomedical, Experimental and Clinical Sciences, Division of Obstetrics and
| | - Fernando M. Reis
- Department of Morphology, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil; Department of Experimental and Clinical Medicine, Università Politecnica delle Marche, Ancona, Italy; Department of Obstetrics and Gynecology, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil; Department of Molecular and Developmental Medicine, Obstetrics and Gynecological Clinic, University of Siena, Siena, Italy; and Department of Biomedical, Experimental and Clinical Sciences, Division of Obstetrics and
| |
Collapse
|
16
|
Wang Q, Chen Y, Zhang D, Li C, Chen X, Hou J, Fei Y, Wang Y, Li Y. Activin Receptor-Like Kinase 4 Haplodeficiency Mitigates Arrhythmogenic Atrial Remodeling and Vulnerability to Atrial Fibrillation in Cardiac Pathological Hypertrophy. J Am Heart Assoc 2018; 7:e008842. [PMID: 30369314 PMCID: PMC6201394 DOI: 10.1161/jaha.118.008842] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2018] [Accepted: 06/12/2018] [Indexed: 12/19/2022]
Abstract
Background Activin receptor-like kinase 4 ( ALK 4) is highly expressed in mammal heart. Atrial fibrillation ( AF ) is closely related to ventricular pressure overload. Because pressure overload increases atrial pressure and leads to atrial remodeling, it would be informative to know whether ALK 4 exerts potential effects on atrial remodeling and AF vulnerability in a pressure-overload model. Methods and Results Wild-type littermates and ALK 4+/- mice were subjected to abdominal aortic constriction or a sham operation. After 4 or 8 weeks, echocardiographic and hemodynamic measurements were performed, and inducibility of AF was tested. The hearts were divided into atria and ventricles and then were fixed in formalin for staining, or they were weighted and snap-frozen for quantitative real-time polymerase chain reaction and Western blot analysis. Compared with wild-type littermates, ALK 4+/- mice demonstrated a similar extent of atrial hypertrophy but significantly suppressed atrial fibrosis at 8 weeks post-abdominal aortic constriction. ALK 4 haplodeficiency partially blocked abdominal aortic constriction-induced upregulation of monocyte chemotactic protein 1 and interleukin-6, and the increased chemotaxin of macrophages. ALK 4 haplodeficiency also blunted a reduction of connexin 40 and redistribution of connexin 43 from the intercalated disk to the lateral membranes, thereby improving localized conduction abnormalities. Meanwhile, ALK 4 haplodeficiency inhibited abdominal aortic constriction-induced decreased INa, ICa-L and IK1 densities as well as the accompanying action potential duration shortening. Mechanistically, ALK 4 haploinsufficiency resulted in the suppression of Smad2/3 activity in this model. Conclusions Our results demonstrate that ALK 4 haplodeficiency ameliorates atrial remodeling and vulnerability to AF in a pressure-overload model through inactivation of the Smad2/3 pathway, suggesting that ALK 4 might be a potential therapeutic target in combating pressure overload-induced AF .
Collapse
Affiliation(s)
- Qian Wang
- Department of CardiologyXinhua HospitalSchool of MedicineShanghai Jiao Tong UniversityShanghaiChina
| | - Yihe Chen
- Department of CardiologyXinhua HospitalSchool of MedicineShanghai Jiao Tong UniversityShanghaiChina
| | - Daoliang Zhang
- Department of CardiologyShanghai Chest HospitalShanghai Jiao Tong UniversityShanghaiChina
| | - Changyi Li
- Department of CardiologyXinhua HospitalSchool of MedicineShanghai Jiao Tong UniversityShanghaiChina
| | - Xiaoqing Chen
- Department of CardiologyShanghai General HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Jianwen Hou
- Department of CardiologyXinhua HospitalSchool of MedicineShanghai Jiao Tong UniversityShanghaiChina
| | - Yudong Fei
- Department of CardiologyXinhua HospitalSchool of MedicineShanghai Jiao Tong UniversityShanghaiChina
| | - Yuepeng Wang
- Department of CardiologyXinhua HospitalSchool of MedicineShanghai Jiao Tong UniversityShanghaiChina
| | - Yigang Li
- Department of CardiologyXinhua HospitalSchool of MedicineShanghai Jiao Tong UniversityShanghaiChina
| |
Collapse
|
17
|
Dillenburg A, Ireland G, Holloway RK, Davies CL, Evans FL, Swire M, Bechler ME, Soong D, Yuen TJ, Su GH, Becher JC, Smith C, Williams A, Miron VE. Activin receptors regulate the oligodendrocyte lineage in health and disease. Acta Neuropathol 2018; 135:887-906. [PMID: 29397421 PMCID: PMC5954071 DOI: 10.1007/s00401-018-1813-3] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2017] [Revised: 01/16/2018] [Accepted: 01/29/2018] [Indexed: 12/12/2022]
Abstract
The most prevalent neurological disorders of myelin include perinatal brain injury leading to cerebral palsy in infants and multiple sclerosis in adults. Although these disorders have distinct etiologies, they share a common neuropathological feature of failed progenitor differentiation into myelin-producing oligodendrocytes and lack of myelin, for which there is an unmet clinical need. Here, we reveal that a molecular pathology common to both disorders is dysregulation of activin receptors and that activin receptor signaling is required for the majority of myelin generation in development and following injury. Using a constitutive conditional knockout of all activin receptor signaling in oligodendrocyte lineage cells, we discovered this signaling to be required for myelination via regulation of oligodendrocyte differentiation and myelin compaction. These processes were found to be dependent on the activin receptor subtype Acvr2a, which is expressed during oligodendrocyte differentiation and axonal ensheathment in development and following myelin injury. During efficient myelin regeneration, Acvr2a upregulation was seen to coincide with downregulation of Acvr2b, a receptor subtype with relatively higher ligand affinity; Acvr2b was shown to be dispensable for activin receptor-driven oligodendrocyte differentiation and its overexpression was sufficient to impair the abovementioned ligand-driven responses. In actively myelinating or remyelinating areas of human perinatal brain injury and multiple sclerosis tissue, respectively, oligodendrocyte lineage cells expressing Acvr2a outnumbered those expressing Acvr2b, whereas in non-repairing lesions Acvr2b+ cells were increased. Thus, we propose that following human white matter injury, this increase in Acvr2b expression would sequester ligand and consequently impair Acvr2a-driven oligodendrocyte differentiation and myelin formation. Our results demonstrate dysregulated activin receptor signaling in common myelin disorders and reveal Acvr2a as a novel therapeutic target for myelin generation following injury across the lifespan.
Collapse
|
18
|
Qiu W, Remotti HE, Tang SM, Wang E, Dobberteen L, Lee Youssof A, Lee JH, Cheung EC, Su GH. Pancreatic DCLK1 + cells originate distinctly from PDX1 + progenitors and contribute to the initiation of intraductal papillary mucinous neoplasm in mice. Cancer Lett 2018. [PMID: 29526803 DOI: 10.1016/j.canlet.2018.03.009] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
PanINs and IPMNs are the two most common precursor lesions that can progress to invasive pancreatic ductal adenocarcinoma (PDA). DCLK1 has been identified as a biomarker of progenitor cells in PDA progressed from PanINs. To explore the potential role of DCLK1-expressing cells in the genesis of IPMNs, we compared the incidence of DCLK1-positive cells in pancreatic tissue samples from genetically-engineered mouse models (GEMMs) for IPMNs, PanINs, and acinar to ductal metaplasia by immunohistochemistry and immunofluorescence. Mouse lineage tracing experiments in the IPMN GEMM showed that DCLK1+ cells originated from a cell lineage distinct from PDX1+ progenitors. The DCLK1+ cells shared the features of tuft cells but were devoid of IPMN tumor biomarkers. The DCLK1+ cells were detected in the earliest proliferative acinar clusters prior to the formation of metaplastic ductal cells, and were enriched in the "IPMN niches". In summary, DCLK1 labels a unique pancreatic cellular lineage in the IPMN GEMM. The clustering of DCLK1+ cells is an early event in Kras-induced pancreatic tumorigenesis and may contribute to IPMN initiation.
Collapse
Affiliation(s)
- Wanglong Qiu
- The Department of Pathology & Cell Biology, Columbia University Medical Center, New York, NY 10032, USA; Herbert Irving Comprehensive Cancer Center, Columbia University Medical Center, New York, NY 10032, USA
| | - Helen E Remotti
- The Department of Pathology & Cell Biology, Columbia University Medical Center, New York, NY 10032, USA
| | - Sophia M Tang
- Herbert Irving Comprehensive Cancer Center, Columbia University Medical Center, New York, NY 10032, USA
| | - Elizabeth Wang
- Herbert Irving Comprehensive Cancer Center, Columbia University Medical Center, New York, NY 10032, USA
| | - Lily Dobberteen
- Herbert Irving Comprehensive Cancer Center, Columbia University Medical Center, New York, NY 10032, USA
| | - Ayman Lee Youssof
- The Department of Pathology & Cell Biology, Columbia University Medical Center, New York, NY 10032, USA; Herbert Irving Comprehensive Cancer Center, Columbia University Medical Center, New York, NY 10032, USA
| | - Joo Hee Lee
- Herbert Irving Comprehensive Cancer Center, Columbia University Medical Center, New York, NY 10032, USA
| | - Edwin C Cheung
- Herbert Irving Comprehensive Cancer Center, Columbia University Medical Center, New York, NY 10032, USA
| | - Gloria H Su
- The Department of Pathology & Cell Biology, Columbia University Medical Center, New York, NY 10032, USA; Herbert Irving Comprehensive Cancer Center, Columbia University Medical Center, New York, NY 10032, USA; Department of Otolaryngology and Head and Neck Surgery, Columbia University Medical Center, New York, NY 10032, USA.
| |
Collapse
|
19
|
Loomans HA, Arnold SA, Hebron K, Taylor CJ, Zijlstra A, Andl CD. Loss of ACVRIB leads to increased squamous cell carcinoma aggressiveness through alterations in cell-cell and cell-matrix adhesion proteins. Am J Cancer Res 2017; 7:2422-2437. [PMID: 29312797 PMCID: PMC5752684] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2017] [Accepted: 09/07/2017] [Indexed: 02/24/2023] Open
Abstract
Squamous cell carcinomas of the head and neck (HNSCC) and esophagus (ESCC) pose a global public health issue due to high mortality rates. Unfortunately, little progress has been made in improving patient outcomes. This is partially a result of a lack of understanding the mechanisms that drive SCC progression. Recently, Activin A signaling has been implicated in a number of cancers, yet the role of this pathway in SCC remains poorly understood. We have previously discovered that the Activin A ligand acts as a tumor suppressor when epithelial Activin receptor type IB (ACVRIB) is intact; however, this effect is lost upon ACVRIB downregulation. In the present study, we investigated the function of ACVRIB in the regulation of SCC. Using CRISPR/Cas9-mediated ACVRIB-knockout and knockdown using siRNA, we found an increased capacity to proliferate, migrate, and invade upon ACRIB loss, as ACVRIB-KO cells exhibited an altered cytoskeleton and aberrant expression of E-cadherin and integrins. Based on chemical inhibitor studies, our data suggests that these effects are mediated through ACVRIB-independent signaling via downstream activation of Smad1/5/8 and MEK/ERK. Overall, we present a novel mechanism of SCC progression upon ACVRIB loss by showing that Activin A can transduce a signal in the absence of ACVRIB.
Collapse
Affiliation(s)
- Holli A Loomans
- Department of Cancer Biology, Vanderbilt UniversityNashville, TN, USA
- Cancer Prevention Fellowship Program, Division of Cancer Prevention, National Cancer InstituteBethesda, MD, USA
| | - Shanna A Arnold
- Department of Veterans Affairs, Tennessee Valley Healthcare SystemNashville, TN, USA
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical CenterNashville, TN, USA
| | - Kate Hebron
- Department of Cancer Biology, Vanderbilt UniversityNashville, TN, USA
| | - Chase J Taylor
- Department of Veterans Affairs, Tennessee Valley Healthcare SystemNashville, TN, USA
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical CenterNashville, TN, USA
| | - Andries Zijlstra
- Department of Cancer Biology, Vanderbilt UniversityNashville, TN, USA
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical CenterNashville, TN, USA
| | - Claudia D Andl
- Department of Cancer Biology, Vanderbilt UniversityNashville, TN, USA
- Burnett School of Biomedical Sciences, College of Medicine, University of Central FloridaOrlando, FL, USA
| |
Collapse
|
20
|
Takahashi T, Asano Y, Sugawara K, Yamashita T, Nakamura K, Saigusa R, Ichimura Y, Toyama T, Taniguchi T, Akamata K, Noda S, Yoshizaki A, Tsuruta D, Trojanowska M, Sato S. Epithelial Fli1 deficiency drives systemic autoimmunity and fibrosis: Possible roles in scleroderma. J Exp Med 2017; 214:1129-1151. [PMID: 28232470 PMCID: PMC5379967 DOI: 10.1084/jem.20160247] [Citation(s) in RCA: 65] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2016] [Revised: 11/08/2016] [Accepted: 01/17/2017] [Indexed: 01/06/2023] Open
Abstract
Systemic sclerosis (SSc), or scleroderma, is a multisystem autoimmune disorder characterized by vasculopathy and fibrosis in the skin and internal organs, most frequently in the esophagus and lungs. Hitherto, studies on SSc pathogenesis centered on immune cells, vascular cells, and fibroblasts. Although dysregulated keratinocytes in SSc have been recently reported, the contribution of epithelial cells to pathogenesis remains unexplored. In this study, we demonstrated the induction of SSc-like molecular phenotype in keratinocytes by gene silencing of transcription factor Friend leukemia virus integration 1 (Fli1), the deficiency of which is implicated in SSc pathogenesis. Keratin 14-expressing epithelial cell-specific Fli1 knockout mice spontaneously developed dermal and esophageal fibrosis with epithelial activation. Furthermore, they developed remarkable autoimmunity with interstitial lung disease derived from thymic defects with down-regulation of autoimmune regulator (Aire). Importantly, Fli1 directly regulated Aire expression in epithelial cells. Collectively, epithelial Fli1 deficiency might be involved in the systemic autoimmunity and selective organ fibrosis in SSc. This study uncovers unidentified roles of dysregulated epithelial cells in SSc pathogenesis.
Collapse
Affiliation(s)
- Takehiro Takahashi
- Department of Dermatology, University of Tokyo Graduate School of Medicine, Bunkyo-ku, Tokyo 113-8655, Japan
| | - Yoshihide Asano
- Department of Dermatology, University of Tokyo Graduate School of Medicine, Bunkyo-ku, Tokyo 113-8655, Japan
| | - Koji Sugawara
- Department of Dermatology, Osaka City University Graduate School of Medicine, Abeno-ku, Osaka 545-8585, Japan
| | - Takashi Yamashita
- Department of Dermatology, University of Tokyo Graduate School of Medicine, Bunkyo-ku, Tokyo 113-8655, Japan
| | - Kouki Nakamura
- Department of Dermatology, University of Tokyo Graduate School of Medicine, Bunkyo-ku, Tokyo 113-8655, Japan
| | - Ryosuke Saigusa
- Department of Dermatology, University of Tokyo Graduate School of Medicine, Bunkyo-ku, Tokyo 113-8655, Japan
| | - Yohei Ichimura
- Department of Dermatology, University of Tokyo Graduate School of Medicine, Bunkyo-ku, Tokyo 113-8655, Japan
| | - Tetsuo Toyama
- Department of Dermatology, University of Tokyo Graduate School of Medicine, Bunkyo-ku, Tokyo 113-8655, Japan
| | - Takashi Taniguchi
- Department of Dermatology, University of Tokyo Graduate School of Medicine, Bunkyo-ku, Tokyo 113-8655, Japan
| | - Kaname Akamata
- Department of Dermatology, University of Tokyo Graduate School of Medicine, Bunkyo-ku, Tokyo 113-8655, Japan
| | - Shinji Noda
- Department of Dermatology, University of Tokyo Graduate School of Medicine, Bunkyo-ku, Tokyo 113-8655, Japan
| | - Ayumi Yoshizaki
- Department of Dermatology, University of Tokyo Graduate School of Medicine, Bunkyo-ku, Tokyo 113-8655, Japan
| | - Daisuke Tsuruta
- Department of Dermatology, Osaka City University Graduate School of Medicine, Abeno-ku, Osaka 545-8585, Japan
| | - Maria Trojanowska
- Arthritis Center, Rheumatology, Boston University School of Medicine, Boston, MA 02118
| | - Shinichi Sato
- Department of Dermatology, University of Tokyo Graduate School of Medicine, Bunkyo-ku, Tokyo 113-8655, Japan
| |
Collapse
|
21
|
Seo CH, Kwack MH, Kim MK, Kim JC, Sung YK. Activin A-induced signalling controls hair follicle neogenesis. Exp Dermatol 2017; 26:108-115. [DOI: 10.1111/exd.13234] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/26/2016] [Indexed: 12/20/2022]
Affiliation(s)
- Chang H. Seo
- Department of Immunology; School of Medicine; Kyungpook National University; Daegu Korea
| | - Mi H. Kwack
- Department of Immunology; School of Medicine; Kyungpook National University; Daegu Korea
| | - Moon K. Kim
- Department of Immunology; School of Medicine; Kyungpook National University; Daegu Korea
| | - Jung C. Kim
- Department of Immunology; School of Medicine; Kyungpook National University; Daegu Korea
| | - Young K. Sung
- Department of Immunology; School of Medicine; Kyungpook National University; Daegu Korea
| |
Collapse
|
22
|
Ehrmann C, Schneider MR. Genetically modified laboratory mice with sebaceous glands abnormalities. Cell Mol Life Sci 2016; 73:4623-4642. [PMID: 27457558 PMCID: PMC11108334 DOI: 10.1007/s00018-016-2312-0] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2016] [Revised: 07/12/2016] [Accepted: 07/19/2016] [Indexed: 12/19/2022]
Abstract
Sebaceous glands (SG) are exocrine glands that release their product by holocrine secretion, meaning that the whole cell becomes a secretion following disruption of the membrane. SG may be found in association with a hair follicle, forming the pilosebaceous unit, or as modified SG at different body sites such as the eyelids (Meibomian glands) or the preputial glands. Depending on their location, SG fulfill a number of functions, including protection of the skin and fur, thermoregulation, formation of the tear lipid film, and pheromone-based communication. Accordingly, SG abnormalities are associated with several diseases such as acne, cicatricial alopecia, and dry eye disease. An increasing number of genetically modified laboratory mouse lines develop SG abnormalities, and their study may provide important clues regarding the molecular pathways regulating SG development, physiology, and pathology. Here, we summarize in tabulated form the available mouse lines with SG abnormalities and, focusing on selected examples, discuss the insights they provide into SG biology and pathology. We hope this survey will become a helpful information source for researchers with a primary interest in SG but also as for researchers from unrelated fields that are unexpectedly confronted with a SG phenotype in newly generated mouse lines.
Collapse
Affiliation(s)
- Carmen Ehrmann
- Institute of Molecular Animal Breeding and Biotechnology, Gene Center, LMU Munich, Feodor-Lynen-Str. 25, 81377, Munich, Germany
| | - Marlon R Schneider
- Institute of Molecular Animal Breeding and Biotechnology, Gene Center, LMU Munich, Feodor-Lynen-Str. 25, 81377, Munich, Germany.
| |
Collapse
|
23
|
Loomans HA, Andl CD. Activin receptor-like kinases: a diverse family playing an important role in cancer. Am J Cancer Res 2016; 6:2431-2447. [PMID: 27904762 PMCID: PMC5126264] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2016] [Accepted: 10/12/2016] [Indexed: 06/06/2023] Open
Abstract
The role and function of the members of the TGFβ superfamily has been a substantial area of research focus for the last several decades. During that time, it has become apparent that aberrations in TGFβ family signaling, whether through the BMP, Activin, or TGFβ arms of the pathway, can result in tumorigenesis or contribute to its progression. Downstream signaling regulates cellular growth under normal physiological conditions yet induces diverse processes during carcinogenesis, ranging from epithelial- to-mesenchymal transition to cell migration and invasion to angiogenesis. Due to these observations, the question has been raised how to utilize and target components of these signaling pathways in cancer therapy. Given that these cascades include both ligands and receptors, there are multiple levels at which to interfere. Activin receptor-like kinases (ALKs) are a group of seven type I receptors responsible for TGFβ family signal transduction and are utilized by many ligands within the superfamily. The challenge lies in specifically targeting the often-overlapping functional effects of BMP, Activin, or TGFβ signaling during cancer progression. This review focuses on the characteristic function of the individual receptors within each subfamily and their recognized roles in cancer. We next explore the clinical utility of therapeutically targeting ALKs as some have shown partial responses in Phase I clinical trials but disappointing outcomes when used in Phase II studies. Finally, we discuss the challenges and future directions of this body of work.
Collapse
Affiliation(s)
- Holli A Loomans
- Department of Cancer Biology, Vanderbilt UniversityNashville, TN, USA
| | - Claudia D Andl
- Burnett School of Biomedical Sciences, College of Medicine, University of Central FloridaOrlando, FL, USA
| |
Collapse
|
24
|
Namwanje M, Brown CW. Activins and Inhibins: Roles in Development, Physiology, and Disease. Cold Spring Harb Perspect Biol 2016; 8:cshperspect.a021881. [PMID: 27328872 DOI: 10.1101/cshperspect.a021881] [Citation(s) in RCA: 155] [Impact Index Per Article: 19.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Since their original discovery as regulators of follicle-stimulating hormone (FSH) secretion and erythropoiesis, the TGF-β family members activin and inhibin have been shown to participate in a variety of biological processes, from the earliest stages of embryonic development to highly specialized functions in terminally differentiated cells and tissues. Herein, we present the history, structures, signaling mechanisms, regulation, and biological processes in which activins and inhibins participate, including several recently discovered biological activities and functional antagonists. The potential therapeutic relevance of these advances is also discussed.
Collapse
Affiliation(s)
- Maria Namwanje
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas 77030
| | - Chester W Brown
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas 77030 Department of Pediatrics, Baylor College of Medicine, Houston, Texas 77030 Texas Children's Hospital, Houston, Texas 77030
| |
Collapse
|
25
|
Fang C, Li L, Li J. Conditional Knockout in Mice Reveals the Critical Roles of Ppp2ca in Epidermis Development. Int J Mol Sci 2016; 17:ijms17050756. [PMID: 27213341 PMCID: PMC4881577 DOI: 10.3390/ijms17050756] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2016] [Revised: 05/09/2016] [Accepted: 05/11/2016] [Indexed: 12/12/2022] Open
Abstract
The epidermis is an important tissue in Homo sapines and other animals, and an abnormal epidermis will cause many diseases. Phosphatase 2A (PP2A) is an important serine and threonine phosphatase. The α isoform of the PP2A catalytic subunit (Ppp2ca gene encoding PP2Acα) is critical for cell proliferation, growth, metabolism and tumorigenesis. However, to date, no study has revealed its roles in epidermis development. To specifically investigate the roles of PP2Acα in epidermis development, we first generated Ppp2caflox/flox transgenic mice, and conditionally knocked out Ppp2ca in the epidermis driven by Krt14-Cre. Our study showed that Ppp2caflox/flox; Krt14-Cre mice had significant hair loss. In addition, histological analyses showed that the morphogenesis and hair regeneration cycle of hair follicles were disrupted in these mice. Moreover, Ppp2caflox/flox; Krt14-Cre mice had smaller size, melanin deposition and hyperproliferation at the base of the claws. Accordingly, our study demonstrates that PP2Acα plays important roles in both hair follicle and epidermis development. Additionally, the Ppp2caflox/flox mice generated in this study can serve as a useful transgene model to study the roles of PP2Acα in other developmental processes and diseases.
Collapse
Affiliation(s)
- Chao Fang
- Department of Pharmacology, School of Basic Medical Sciences, Nanjing Medical University, Nanjing 210029, China.
| | - Lei Li
- Translational Medicine Center, Yancheng No. 1 People's Hospital, Yancheng 224000, China.
| | - Jianmin Li
- Department of Pharmacology, School of Basic Medical Sciences, Nanjing Medical University, Nanjing 210029, China.
- Model Animal Research Center of Nanjing Medical University, Nanjing 210029, China.
- Key Laboratory of National Reproductive Medicine Department of Cell Biology and Medical Genetics, Nanjing Medical University, Nanjing 210029, China.
- Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University Nanjing, Nanjing 210029, China.
- Department of cell biology, School of Basic Medical Sciences, Nanjing Medical University, Nanjing 210029, China.
| |
Collapse
|
26
|
Qiu W, Tang SM, Lee S, Turk AT, Sireci AN, Qiu A, Rose C, Xie C, Kitajewski J, Wen HJ, Crawford HC, Sims PA, Hruban RH, Remotti HE, Su GH. Loss of Activin Receptor Type 1B Accelerates Development of Intraductal Papillary Mucinous Neoplasms in Mice With Activated KRAS. Gastroenterology 2016; 150:218-228.e12. [PMID: 26408346 PMCID: PMC4860725 DOI: 10.1053/j.gastro.2015.09.013] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2015] [Revised: 08/27/2015] [Accepted: 09/16/2015] [Indexed: 12/29/2022]
Abstract
BACKGROUND & AIMS Activin, a member of the transforming growth factor-β (TGFB) family, might be involved in pancreatic tumorigenesis, similar to other members of the TGFB family. Human pancreatic ductal adenocarcinomas contain somatic mutations in the activin A receptor type IB (ACVR1B) gene, indicating that ACVR1B could be a suppressor of pancreatic tumorigenesis. METHODS We disrupted Acvr1b specifically in pancreata of mice (Acvr1b(flox/flox);Pdx1-Cre mice) and crossed them with LSL-KRAS(G12D) mice, which express an activated form of KRAS and develop spontaneous pancreatic tumors. The resulting Acvr1b(flox/flox);LSL-KRAS(G12D);Pdx1-Cre mice were monitored; pancreatic tissues were collected and analyzed by histology and immunohistochemical analyses. We also analyzed p16(flox/flox);LSL-Kras(G12D);Pdx1-Cre mice and Cre-negative littermates (controls). Genomic DNA, total RNA, and protein were isolated from mouse tissues and primary pancreatic tumor cell lines and analyzed by reverse-transcription polymerase chain reaction, sequencing, and immunoblot analyses. Human intraductal papillary mucinous neoplasm (IPMN) specimens were analyzed by immunohistochemistry. RESULTS Loss of ACVR1B from pancreata of mice increased the proliferation of pancreatic epithelial cells, led to formation of acinar to ductal metaplasia, and induced focal inflammatory changes compared with control mice. Disruption of Acvr1b in LSL-KRAS(G12D);Pdx1-Cre mice accelerated the growth of pancreatic IPMNs compared with LSL-KRAS(G12D);Pdx1-Cre mice, but did not alter growth of pancreatic intraepithelial neoplasias. We associated perinuclear localization of the activated NOTCH4 intracellular domain to the apical cytoplasm of neoplastic cells with the expansion of IPMN lesions in Acvr1b(flox/flox);LSL-KRAS(G12D);Pdx1-Cre mice. Loss of the gene that encodes p16 (Cdkn2a) was required for progression of IPMNs to pancreatic ductal adenocarcinomas in Acvr1b(flox/flox);LSL-Kras(G12D);Pdx1-Cre mice. We also observed progressive loss of p16 in human IPMNs of increasing grades. CONCLUSIONS Loss of ACVR1B accelerates growth of mutant KRAS-induced pancreatic IPMNs in mice; this process appears to involve NOTCH4 and loss of p16. ACVR1B suppresses early stages of pancreatic tumorigenesis; the activin signaling pathway therefore might be a therapeutic target for pancreatic cancer.
Collapse
Affiliation(s)
- Wanglong Qiu
- The Department of Pathology, Columbia University Medical Center, New York, NY 10032,Herbert Irving Comprehensive Cancer Center, Columbia University Medical Center, New York, NY 10032
| | - Sophia M. Tang
- Herbert Irving Comprehensive Cancer Center, Columbia University Medical Center, New York, NY 10032
| | - Sohyae Lee
- Herbert Irving Comprehensive Cancer Center, Columbia University Medical Center, New York, NY 10032
| | - Andrew T. Turk
- The Department of Pathology, Columbia University Medical Center, New York, NY 10032
| | - Anthony N. Sireci
- The Department of Pathology, Columbia University Medical Center, New York, NY 10032
| | - Anne Qiu
- Herbert Irving Comprehensive Cancer Center, Columbia University Medical Center, New York, NY 10032
| | | | - Chuangao Xie
- Herbert Irving Comprehensive Cancer Center, Columbia University Medical Center, New York, NY 10032
| | - Jan Kitajewski
- Department of Obstetrics and Gynecology, Columbia University Medical Center, New York, NY 10032
| | - Hui-Ju Wen
- Department of Cancer Biology, Mayo Clinic Cancer Center, Jacksonville, FL
| | - Howard C. Crawford
- Department of Cancer Biology, Mayo Clinic Cancer Center, Jacksonville, FL
| | - Peter A. Sims
- Department of Systems Biology, Columbia University Medical Center, New York, NY 10032
| | - Ralph H. Hruban
- The Sol Goldman Pancreatic Cancer Research Center, Department of Pathology, The Johns Hopkins Medical Institutions, Baltimore, MD 21205
| | - Helen E. Remotti
- The Department of Pathology, Columbia University Medical Center, New York, NY 10032
| | - Gloria H. Su
- The Department of Pathology, Columbia University Medical Center, New York, NY 10032,Herbert Irving Comprehensive Cancer Center, Columbia University Medical Center, New York, NY 10032,Department of Otolaryngology and Head and Neck Surgery, Columbia University Medical Center, New York, NY 10032
| |
Collapse
|
27
|
Peng J, Fullerton PT, Monsivais D, Clementi C, Su GH, Matzuk MM. Uterine Activin-Like Kinase 4 Regulates Trophoblast Development During Mouse Placentation. Mol Endocrinol 2015; 29:1684-93. [PMID: 26484579 DOI: 10.1210/me.2015-1048] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
The placenta is the first organ to develop after fertilization. It forms an interface between the maternal uterus and growing fetus to allow nutrient uptake, waste elimination, and gas exchange for a successful pregnancy in both mice and humans. In the past 2 decades, in vivo and in vitro approaches have been used to show that several members of the TGF-β superfamily regulate embryo implantation and placental development. Nodal, a TGF-β superfamily ligand, is essential for mesendoderm formation and left-right axis patterning during embryogenesis, and Nodal null mutants exhibit abnormal placental organization with expansion of trophoblast giant cells and a decrease of spongiotrophoblast and labyrinth. To better understand the importance of Nodal signaling in the uterus, we established a mouse model to conditionally ablate activin-like kinase 4 (ALK4; the Nodal type 1 receptor) using Cre recombinase driven by the progesterone receptor promoter sequences (Pgr-Cre). Alk4 conditional knockout females are subfertile due to placental abnormalities and fetal loss in pregnancy, with a placental disorganization phenotype similar to what is observed in Nodal null mice. Thus, Nodal likely functions as an indirect regulator of placental development by binding to type 1 and type 2 receptors on maternal decidual cells to stimulate expression of unknown regulators of placental development. Our findings not only describe the generation of a mouse model that enables study of Nodal signaling in placentation but also provides insights into the pathogenesis of pregnancy complications in humans, including spontaneous abortion, preeclampsia, intrauterine growth restriction, and preterm birth.
Collapse
Affiliation(s)
- Jia Peng
- Departments of Pathology and Immunology (J.P., P.T.F., D.M., M.M.M.), Molecular and Human Genetics (J.P., P.T.F., M.M.M.), Molecular and Cellular Biology (M.M.M.), and Pharmacology (M.M.M.), Centers for Reproductive Medicine (M.M.M.) and Drug Discovery (P.T.F., D.M., M.M.M.), Baylor College of Medicine, Houston, Texas 77030; Department of Pathology (C.C.), New York University School of Medicine, New York, New York 10016; and Departments of Pathology (G.H.S.) and Otolaryngology/Head and Neck Surgery (G.H.S.), Columbia University College of Physicians and Surgeons, New York, New York 10032
| | - Paul T Fullerton
- Departments of Pathology and Immunology (J.P., P.T.F., D.M., M.M.M.), Molecular and Human Genetics (J.P., P.T.F., M.M.M.), Molecular and Cellular Biology (M.M.M.), and Pharmacology (M.M.M.), Centers for Reproductive Medicine (M.M.M.) and Drug Discovery (P.T.F., D.M., M.M.M.), Baylor College of Medicine, Houston, Texas 77030; Department of Pathology (C.C.), New York University School of Medicine, New York, New York 10016; and Departments of Pathology (G.H.S.) and Otolaryngology/Head and Neck Surgery (G.H.S.), Columbia University College of Physicians and Surgeons, New York, New York 10032
| | - Diana Monsivais
- Departments of Pathology and Immunology (J.P., P.T.F., D.M., M.M.M.), Molecular and Human Genetics (J.P., P.T.F., M.M.M.), Molecular and Cellular Biology (M.M.M.), and Pharmacology (M.M.M.), Centers for Reproductive Medicine (M.M.M.) and Drug Discovery (P.T.F., D.M., M.M.M.), Baylor College of Medicine, Houston, Texas 77030; Department of Pathology (C.C.), New York University School of Medicine, New York, New York 10016; and Departments of Pathology (G.H.S.) and Otolaryngology/Head and Neck Surgery (G.H.S.), Columbia University College of Physicians and Surgeons, New York, New York 10032
| | - Caterina Clementi
- Departments of Pathology and Immunology (J.P., P.T.F., D.M., M.M.M.), Molecular and Human Genetics (J.P., P.T.F., M.M.M.), Molecular and Cellular Biology (M.M.M.), and Pharmacology (M.M.M.), Centers for Reproductive Medicine (M.M.M.) and Drug Discovery (P.T.F., D.M., M.M.M.), Baylor College of Medicine, Houston, Texas 77030; Department of Pathology (C.C.), New York University School of Medicine, New York, New York 10016; and Departments of Pathology (G.H.S.) and Otolaryngology/Head and Neck Surgery (G.H.S.), Columbia University College of Physicians and Surgeons, New York, New York 10032
| | - Gloria H Su
- Departments of Pathology and Immunology (J.P., P.T.F., D.M., M.M.M.), Molecular and Human Genetics (J.P., P.T.F., M.M.M.), Molecular and Cellular Biology (M.M.M.), and Pharmacology (M.M.M.), Centers for Reproductive Medicine (M.M.M.) and Drug Discovery (P.T.F., D.M., M.M.M.), Baylor College of Medicine, Houston, Texas 77030; Department of Pathology (C.C.), New York University School of Medicine, New York, New York 10016; and Departments of Pathology (G.H.S.) and Otolaryngology/Head and Neck Surgery (G.H.S.), Columbia University College of Physicians and Surgeons, New York, New York 10032
| | - Martin M Matzuk
- Departments of Pathology and Immunology (J.P., P.T.F., D.M., M.M.M.), Molecular and Human Genetics (J.P., P.T.F., M.M.M.), Molecular and Cellular Biology (M.M.M.), and Pharmacology (M.M.M.), Centers for Reproductive Medicine (M.M.M.) and Drug Discovery (P.T.F., D.M., M.M.M.), Baylor College of Medicine, Houston, Texas 77030; Department of Pathology (C.C.), New York University School of Medicine, New York, New York 10016; and Departments of Pathology (G.H.S.) and Otolaryngology/Head and Neck Surgery (G.H.S.), Columbia University College of Physicians and Surgeons, New York, New York 10032
| |
Collapse
|
28
|
Abstract
Activin/Nodal growth factors control a broad range of biological processes, including early cell fate decisions, organogenesis and adult tissue homeostasis. Here, we provide an overview of the mechanisms by which the Activin/Nodal signalling pathway governs stem cell function in these different stages of development. We describe recent findings that associate Activin/Nodal signalling to pathological conditions, focusing on cancer stem cells in tumorigenesis and its potential as a target for therapies. Moreover, we will discuss future directions and questions that currently remain unanswered on the role of Activin/Nodal signalling in stem cell self-renewal, differentiation and proliferation.
Collapse
Affiliation(s)
- Siim Pauklin
- Anne McLaren Laboratory For Regenerative Medicine, Wellcome Trust-Medical Research Council Cambridge Stem Cell Institute, West Forvie Building, Robinson Way, University of Cambridge, Cambridge CB2 0SZ, UK
| | - Ludovic Vallier
- Anne McLaren Laboratory For Regenerative Medicine, Wellcome Trust-Medical Research Council Cambridge Stem Cell Institute, West Forvie Building, Robinson Way, University of Cambridge, Cambridge CB2 0SZ, UK
| |
Collapse
|
29
|
Chen CC, Murray PJ, Jiang TX, Plikus MV, Chang YT, Lee OK, Widelitz RB, Chuong CM. Regenerative hair waves in aging mice and extra-follicular modulators follistatin, dkk1, and sfrp4. J Invest Dermatol 2014; 134:2086-2096. [PMID: 24618599 PMCID: PMC4102635 DOI: 10.1038/jid.2014.139] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2013] [Revised: 02/12/2014] [Accepted: 02/20/2014] [Indexed: 01/03/2023]
Abstract
Hair cycling is modulated by factors both intrinsic and extrinsic to hair follicles. Cycling defects lead to conditions such as aging-associated alopecia. Recently, we demonstrated that mouse skin exhibits regenerative hair waves, reflecting a coordinated regenerative behavior in follicle populations. Here, we use this model to explore the regenerative behavior of aging mouse skin. Old mice (>18 months) tracked over several months show that with progressing age, hair waves slow down, wave propagation becomes restricted, and hair cycle domains fragment into smaller domains. Transplanting aged donor mouse skin to a young host can restore donor cycling within a 3 mm range of the interface, suggesting that changes are due to extracellular factors. Therefore, hair stem cells in aged skin can be reactivated. Molecular studies show that extra-follicular modulators Bmp2, Dkk1, and Sfrp4 increase in early anagen. Further, we identify follistatin as an extra-follicular modulator, which is highly expressed in late telogen and early anagen. Indeed, follistatin induces hair wave propagation and its level decreases in aging mice. We present an excitable medium model to simulate the cycling behavior in aging mice and illustrate how the interorgan macroenvironment can regulate the aging process by integrating both "activator" and "inhibitor" signals.
Collapse
Affiliation(s)
- Chih-Chiang Chen
- Department of Pathology, Keck School of Medicine, University of Southern California, Los Angeles, California, USA; Institute of Clinical Medicine and Department of Dermatology, National Yang-Ming University, Taipei, Taiwan; Department of Dermatology, Taipei Veterans General Hospital, Taipei, Taiwan
| | | | - Ting Xin Jiang
- Department of Pathology, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Maksim V Plikus
- Department of Developmental and Cell Biology, Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, Irvine, California, USA
| | - Yun-Ting Chang
- Institute of Clinical Medicine and Department of Dermatology, National Yang-Ming University, Taipei, Taiwan
| | - Oscar K Lee
- Institute of Clinical Medicine and Department of Dermatology, National Yang-Ming University, Taipei, Taiwan; Department of Orthopaedics and Traumatology, Taipei Veterans General Hospital, Taipei and Center for Stem Cell Research, National Yang-Ming University and Veterans General Hospital, Taipei, Taiwan
| | - Randall B Widelitz
- Department of Pathology, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Cheng-Ming Chuong
- Department of Pathology, Keck School of Medicine, University of Southern California, Los Angeles, California, USA; Institute of Clinical Medicine and Department of Dermatology, National Yang-Ming University, Taipei, Taiwan.
| |
Collapse
|
30
|
Kadaja M, Keyes BE, Lin M, Pasolli HA, Genander M, Polak L, Stokes N, Zheng D, Fuchs E. SOX9: a stem cell transcriptional regulator of secreted niche signaling factors. Genes Dev 2014; 28:328-41. [PMID: 24532713 PMCID: PMC3937512 DOI: 10.1101/gad.233247.113] [Citation(s) in RCA: 157] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
Hair follicles (HFs) undergo cyclical periods of growth, which are fueled by stem cells (SCs) at the base of the resting follicle. HF-SC formation occurs during HF development and requires transcription factor SOX9. Whether and how SOX9 functions in HF-SC maintenance remain unknown. By conditionally targeting Sox9 in adult HF-SCs, we show that SOX9 is essential for maintaining them. SOX9-deficient HF-SCs still transition from quiescence to proliferation and launch the subsequent hair cycle. However, once activated, bulge HF-SCs begin to differentiate into epidermal cells, which naturally lack SOX9. In addition, as HF-SC numbers dwindle, outer root sheath production is not sustained, and HF downgrowth arrests prematurely. Probing the mechanism, we used RNA sequencing (RNA-seq) to identify SOX9-dependent transcriptional changes and chromatin immunoprecipitation (ChIP) and deep sequencing (ChIP-seq) to identify SOX9-bound genes in HF-SCs. Intriguingly, a large cohort of SOX9-sensitive targets encode extracellular factors, most notably enhancers of Activin/pSMAD2 signaling. Moreover, compromising Activin signaling recapitulates SOX9-dependent defects, and Activin partially rescues them. Overall, our findings reveal roles for SOX9 in regulating adult HF-SC maintenance and suppressing epidermal differentiation in the niche. In addition, our studies expose a role for SCs in coordinating their own behavior in part through non-cell-autonomous signaling within the niche.
Collapse
Affiliation(s)
- Meelis Kadaja
- Howard Hughes Medical Institute, Laboratory of Mammalian Cell Biology and Development, The Rockefeller University, New York, New York 10065, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Scharfenberger L, Hennerici T, Király G, Kitzmüller S, Vernooij M, Zielinski JG. Transgenic Mouse Technology in Skin Biology: Generation of Complete or Tissue-Specific Knockout Mice. J Invest Dermatol 2014; 134:1-5. [DOI: 10.1038/jid.2013.457] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
32
|
Tsai SY, Sennett R, Rezza A, Clavel C, Grisanti L, Zemla R, Najam S, Rendl M. Wnt/β-catenin signaling in dermal condensates is required for hair follicle formation. Dev Biol 2013; 385:179-88. [PMID: 24309208 DOI: 10.1016/j.ydbio.2013.11.023] [Citation(s) in RCA: 101] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2013] [Revised: 11/14/2013] [Accepted: 11/25/2013] [Indexed: 12/17/2022]
Abstract
Broad dermal Wnt signaling is required for patterned induction of hair follicle placodes and subsequent Wnt signaling in placode stem cells is essential for induction of dermal condensates, cell clusters of precursors for the hair follicle dermal papilla (DP). Progression of hair follicle formation then requires coordinated signal exchange between dermal condensates and placode stem cells. However, it remains unknown whether continued Wnt signaling in DP precursor cells plays a role in this process, largely due to the long-standing inability to specifically target dermal condensates for gene ablation. Here we use the Tbx18(Cre) knockin mouse line to ablate the Wnt-responsive transcription factor β-catenin specifically in these cells at E14.5 during the first wave of guard hair follicle formation. In the absence of β-catenin, canonical Wnt signaling is effectively abolished in these cells. Sox2(+) dermal condensates initiate normally; however by E16.5 guard hair follicle numbers are strongly reduced and by E18.5 most whiskers and guard hair follicles are absent, suggesting that active Wnt signaling in dermal condensates is important for hair follicle formation to proceed after induction. To explore the molecular mechanisms by which Wnt signaling in dermal condensates regulates hair follicle formation, we analyze genome-wide the gene expression changes in embryonic β-catenin null DP precursor cells. We find altered expression of several signaling pathway genes, including Fgfs and Activin, both previously implicated in hair follicle formation. In summary, these data reveal a functional role of Wnt signaling in DP precursors for embryonic hair follicle formation and identify Fgf and Activin signaling as potential effectors of Wnt signaling-regulated events.
Collapse
Affiliation(s)
- Su-Yi Tsai
- Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, NY 10029, USA; Department of Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, NY 10029, USA
| | - Rachel Sennett
- Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, NY 10029, USA; Department of Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, NY 10029, USA
| | - Amélie Rezza
- Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, NY 10029, USA; Department of Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, NY 10029, USA
| | - Carlos Clavel
- Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, NY 10029, USA; Department of Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, NY 10029, USA
| | - Laura Grisanti
- Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, NY 10029, USA; Department of Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, NY 10029, USA
| | - Roland Zemla
- Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, NY 10029, USA; Department of Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, NY 10029, USA
| | - Sara Najam
- Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, NY 10029, USA; Department of Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, NY 10029, USA
| | - Michael Rendl
- Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, NY 10029, USA; Department of Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, NY 10029, USA; Department of Dermatology, Icahn School of Medicine at Mount Sinai, NY 10029, USA; Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, NY 10029, USA.
| |
Collapse
|
33
|
Abstract
Pancreatic cancer is critical for developed countries, where its rate of diagnosis has been increasing steadily annually. In the past decade, the advances of pancreatic cancer research have not contributed to the decline in mortality rates from pancreatic cancer-the overall 5-year survival rate remains about 5% low. This number only underscores an obvious urgency for us to better understand the biological features of pancreatic carcinogenesis, to develop early detection methods, and to improve novel therapeutic treatments. To achieve these goals, animal modeling that faithfully recapitulates the whole process of human pancreatic cancer is central to making the advancements. In this review, we summarize the currently available animal models for pancreatic cancer and the advances in pancreatic cancer animal modeling. We compare and contrast the advantages and disadvantages of three major categories of these models: (1) carcinogen-induced; (2) xenograft and allograft; and (3) genetically engineered mouse models. We focus more on the genetically engineered mouse models, a category which has been rapidly expanded recently for their capacities to mimic human pancreatic cancer and metastasis, and highlight the combinations of these models with various newly developed strategies and cell-lineage labeling systems.
Collapse
Affiliation(s)
- Wanglong Qiu
- Department of Otolaryngology and Head and Neck Surgery, Columbia University Medical Center, 1130 St. Nicholas Ave, ICRC 10-04, New York, NY 10032, USA
- Herbert Irving Comprehensive Cancer Center, Columbia University Medical Center, New York, NY 10032, USA
| | - Gloria H. Su
- Department of Otolaryngology and Head and Neck Surgery, Columbia University Medical Center, 1130 St. Nicholas Ave, ICRC 10-04, New York, NY 10032, USA
- Department of Pathology, Columbia University Medical Center, New York, NY 10032, USA
- Herbert Irving Comprehensive Cancer Center, Columbia University Medical Center, New York, NY 10032, USA
| |
Collapse
|
34
|
Shirokova V, Jussila M, Hytönen MK, Perälä N, Drögemüller C, Leeb T, Lohi H, Sainio K, Thesleff I, Mikkola ML. Expression of Foxi3 is regulated by ectodysplasin in skin appendage placodes. Dev Dyn 2013; 242:593-603. [PMID: 23441037 DOI: 10.1002/dvdy.23952] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2012] [Revised: 02/12/2013] [Accepted: 02/14/2013] [Indexed: 11/09/2022] Open
Abstract
BACKGROUND Foxi3 is a member of the large forkhead box family of transcriptional regulators, which have a wide range of biological activities including manifold developmental processes. Heterozygous mutation in Foxi3 was identified in several hairless dog breeds characterized by sparse fur coat and missing teeth. A related phenotype called hypohidrotic ectodermal dysplasia (HED) is caused by mutations in the ectodysplasin (Eda) pathway genes. RESULTS Expression of Foxi3 was strictly confined to the epithelium in developing ectodermal appendages in mouse embryos, but no expression was detected in the epidermis. Foxi3 was expressed in teeth and hair follicles throughout embryogenesis, but in mammary glands only during the earliest stages of development. Foxi3 expression was decreased and increased in Eda loss- and gain-of-function embryos, respectively, and was highly induced by Eda protein in embryonic skin explants. Also activin A treatment up-regulated Foxi3 mRNA levels in vitro. CONCLUSIONS Eda and activin A were identified as upstream regulators of Foxi3. Foxi3 is a likely transcriptional target of Eda in ectodermal appendage placodes suggesting that HED phenotype may in part be produced by compromised Foxi3 activity. In addition to hair and teeth, Foxi3 may have a role in nail, eye, and mammary, sweat, and salivary gland development.
Collapse
Affiliation(s)
- Vera Shirokova
- Developmental Biology Program, Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Bernard DJ, Tran S. Mechanisms of activin-stimulated FSH synthesis: the story of a pig and a FOX. Biol Reprod 2013; 88:78. [PMID: 23426431 DOI: 10.1095/biolreprod.113.107797] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Activins were discovered and, in fact, named more than a quarter century ago based on their abilities to stimulate pituitary follicle-stimulating hormone (FSH) synthesis and secretion. However, it is only in the last decade that we have finally come to understand their underlying mechanisms of action in gonadotroph cells. In this minireview, we chronicle the research that led to the recent discovery of forkhead box L2 (FOXL2) as an essential mediator of activin-regulated FSH beta subunit (Fshb) transcription in vitro and in vivo.
Collapse
Affiliation(s)
- Daniel J Bernard
- Department of Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada.
| | | |
Collapse
|
36
|
Glick AB. The Role of TGFβ Signaling in Squamous Cell Cancer: Lessons from Mouse Models. J Skin Cancer 2012; 2012:249063. [PMID: 23326666 PMCID: PMC3541634 DOI: 10.1155/2012/249063] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2012] [Accepted: 11/16/2012] [Indexed: 12/31/2022] Open
Abstract
TGFβ1 is a member of a large growth factor family including activins/inhibins and bone morphogenic proteins (BMPs) that have a potent growth regulatory and immunomodulatory functions in normal skin homeostasis, regulation of epidermal stem cells, extracellular matrix production, angiogenesis, and inflammation. TGFβ signaling is tightly regulated in normal tissues and becomes deregulated during cancer development in cutaneous SCC and many other solid tumors. Because of these diverse biological processes regulated by TGFβ1, this cytokine and its signaling pathway appear to function at multiple points during carcinogenesis with distinct effects. The mouse skin carcinogenesis model has been a useful tool to dissect the function of this pathway in cancer pathogenesis, with transgenic and null mice as well as small molecule inhibitors to alter the function of the TGFβ1 pathway and assess the effects on cancer development. This paper will review data on changes in TGFβ1 signaling in human SCC primarily HNSCC and cutaneous SCC and different mouse models that have been generated to investigate the relevance of these changes to cancer. A better understanding of the mechanisms underlying the duality of TGFβ1 action in carcinogenesis will inform potential use of this signaling pathway for targeted therapies.
Collapse
Affiliation(s)
- Adam B. Glick
- Department of Veterinary and Biomedical Sciences, The Pennsylvania State University, University Park, PA 16802, USA
| |
Collapse
|
37
|
Ripoche D, Gout J, Pommier RM, Jaafar R, Zhang CX, Bartholin L, Bertolino P. Generation of a conditional mouse model to target Acvr1b disruption in adult tissues. Genesis 2012; 51:120-7. [PMID: 23109354 DOI: 10.1002/dvg.22352] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2012] [Revised: 10/11/2012] [Accepted: 10/13/2012] [Indexed: 11/06/2022]
Abstract
Alk4 is a type I receptor that belongs to the transforming growth factor-beta (TGF-β) family. It takes part in the signaling of TGF-β ligands such as Activins, Gdfs, and Nodal that had been demonstrated to participate in numerous mechanisms ranging from early embryonic development to adult-tissue homeostasis. Evidences indicate that Alk4 is a key regulator of many embryonic processes, but little is known about its signaling in adult tissues and in pathological conditions where Alk4 mutations had been reported. Conventional deletion of Alk4 gene (Acvr1b) results in early embryonic lethality prior gastrulation, which has precluded study of Alk4 functions in postnatal and adult mice. To circumvent this problem, we have generated a conditional Acvr1b floxed-allele by flanking the fifth and sixth exons of the Acvr1b gene with loxP sites. Cre-mediated deletion of the floxed allele generates a deleted allele, which behaves as an Acvr1b null allele leading to embryonic lethality in homozygous mutant animals. A tamoxifen-inducible approach to target disruption of Acvr1b specifically in adult tissues was used and proved to be efficient for studying Alk4 functions in various organs. We report, therefore, a novel conditional model allowing investigation of biological role played by Alk4 in a variety of tissue-specific contexts.
Collapse
Affiliation(s)
- Doriane Ripoche
- Inserm U1052, Centre de Recherche en Cancérologie de Lyon, F-69000 Lyon, France
| | | | | | | | | | | | | |
Collapse
|
38
|
Kisby G, Palmer V, Lasarev M, Fry R, Iordanov M, Magun E, Samson L, Spencer P. Does the cycad genotoxin MAM implicated in Guam ALS-PDC induce disease-relevant changes in mouse brain that includes olfaction? Commun Integr Biol 2012; 4:731-4. [PMID: 22446540 DOI: 10.4161/cib.17603] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Western Pacific amyotrophic lateral sclerosis (ALS) and parkinsonism-dementia complex (PDC), a prototypical neurodegenerative disease (tauopathy) affecting distinct genetic groups with common exposure to neurotoxic chemicals in cycad seed, has many features of Parkinson's and Alzheimer's diseases (AD), including early olfactory dysfunction. Guam ALS-PDC incidence correlates with cycad flour content of cycasin and its aglycone methylazoxymethanol (MAM), which produces persistent DNA damage (O(6)-methylguanine) in the brains of mice lacking O(6)-methylguanine methyltransferase (Mgmt(-/-)). We described in Mgmt(-/-)mice up to 7 days post-MAM treatment that brain DNA damage was linked to brain gene expression changes found in human neurological disease, cancer, and skin and hair development. This addendum reports 6 months post-MAM treatment- related brain transcriptional changes as well as elevated mitogen activated protein kinases and increased caspase-3 activity, both of which are involved in tau aggregation and neurofibrillary tangle formation typical of ALS-PDC and AD, plus transcriptional changes in olfactory receptors. Does cycasin act as a "slow (geno)toxin" in ALS-PDC?
Collapse
|
39
|
Sennett R, Rendl M. Mesenchymal-epithelial interactions during hair follicle morphogenesis and cycling. Semin Cell Dev Biol 2012; 23:917-27. [PMID: 22960356 DOI: 10.1016/j.semcdb.2012.08.011] [Citation(s) in RCA: 270] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2012] [Revised: 07/27/2012] [Accepted: 08/24/2012] [Indexed: 12/17/2022]
Abstract
Embryonic hair follicle induction and formation are regulated by mesenchymal-epithelial interactions between specialized dermal cells and epidermal stem cells that switch to a hair fate. Similarly, during postnatal hair growth, communication between mesenchymal dermal papilla cells and surrounding epithelial matrix cells coordinates hair shaft production. Adult hair follicle regeneration in the hair cycle again is thought to be controlled by activating signals originating from the mesenchymal compartment and acting on hair follicle stem cells. Although many signaling pathways are implicated in hair follicle formation and growth, the precise nature, timing, and intersection of these inductive and regulatory signals remains elusive. The goal of this review is to summarize our current understanding and to discuss recent new insights into mesenchymal-epithelial interactions during hair follicle morphogenesis and cycling.
Collapse
Affiliation(s)
- Rachel Sennett
- Black Family Stem Cell Institute, Mount Sinai School of Medicine, New York, NY 10029, USA
| | | |
Collapse
|
40
|
Han R, Beppu H, Lee YK, Georgopoulos K, Larue L, Li E, Weiner L, Brissette JL. A pair of transmembrane receptors essential for the retention and pigmentation of hair. Genesis 2012; 50:783-800. [PMID: 22611050 DOI: 10.1002/dvg.22039] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2011] [Revised: 05/03/2011] [Accepted: 05/12/2011] [Indexed: 01/07/2023]
Abstract
Hair follicles are simple, accessible models for many developmental processes. Here, using mutant mice, we show that Bmpr2, a known receptor for bone morphogenetic proteins (Bmps), and Acvr2a, a known receptor for Bmps and activins, are individually redundant but together essential for multiple follicular traits. When Bmpr2/Acvr2a function is reduced in cutaneous epithelium, hair follicles undergo rapid cycles of hair generation and loss. Alopecia results from a failure to terminate hair development properly, as hair clubs never form, and follicular retraction is slowed. Hair regeneration is rapid due to premature activation of new hair-production programs. Hair shafts differentiate aberrantly due to impaired arrest of medullary-cell proliferation. When Bmpr2/Acvr2a function is reduced in melanocytes, gray hair develops, as melanosomes differentiate but fail to grow, resulting in organelle miniaturization. We conclude that Bmpr2 and Acvr2a normally play cell-type-specific, necessary roles in organelle biogenesis and the shutdown of developmental programs and cell division.
Collapse
Affiliation(s)
- Rong Han
- Department of Cell Biology, State University of New York Downstate Medical Center, Brooklyn, NY 11203, USA
| | | | | | | | | | | | | | | |
Collapse
|
41
|
Lien WH, Guo X, Polak L, Lawton LN, Young RA, Zheng D, Fuchs E. Genome-wide maps of histone modifications unwind in vivo chromatin states of the hair follicle lineage. Cell Stem Cell 2011; 9:219-32. [PMID: 21885018 DOI: 10.1016/j.stem.2011.07.015] [Citation(s) in RCA: 167] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2011] [Revised: 07/04/2011] [Accepted: 07/28/2011] [Indexed: 12/17/2022]
Abstract
Using mouse skin, where bountiful reservoirs of synchronized hair follicle stem cells (HF-SCs) fuel cycles of regeneration, we explore how adult SCs remodel chromatin in response to activating cues. By profiling global mRNA and chromatin changes in quiescent and activated HF-SCs and their committed, transit-amplifying (TA) progeny, we show that polycomb-group (PcG)-mediated H3K27-trimethylation features prominently in HF-lineage progression by mechanisms distinct from embryonic-SCs. In HF-SCs, PcG represses nonskin lineages and HF differentiation. In TA progeny, nonskin regulators remain PcG-repressed, HF-SC regulators acquire H3K27me3-marks, and HF-lineage regulators lose them. Interestingly, genes poised in embryonic stem cells, active in HF-SCs, and PcG-repressed in TA progeny encode not only key transcription factors, but also signaling regulators. We document their importance in balancing HF-SC quiescence, underscoring the power of chromatin mapping in dissecting SC behavior. Our findings explain how HF-SCs cycle through quiescent and activated states without losing stemness and define roles for PcG-mediated repression in governing a fate switch irreversibly.
Collapse
Affiliation(s)
- Wen-Hui Lien
- Howard Hughes Medical Institute, Laboratory of Mammalian Cell Biology & Development, The Rockefeller University, New York, NY 10065, USA
| | | | | | | | | | | | | |
Collapse
|