1
|
Lee SY, Jeong YW, Choe YH, Oh SJ, Miah R, Lee WJ, Lee SL, Bok EY, Yoo DS, Son YB. Identification of Reference Gene for Quantitative Gene Expression in Early-Term and Late-Term Cultured Canine Fibroblasts Derived from Ear Skin. Animals (Basel) 2024; 14:2722. [PMID: 39335311 PMCID: PMC11429031 DOI: 10.3390/ani14182722] [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: 08/11/2024] [Revised: 09/06/2024] [Accepted: 09/18/2024] [Indexed: 09/30/2024] Open
Abstract
Fibroblasts are cells that reside within the fibrous or loose connective tissues of most mammalian organs. For research purposes, fibroblasts are often subjected to long-term culture under defined conditions, during which their properties can significantly change. It is essential to understand and document these changes to obtain reliable outcomes. For the quantification of specific gene expressions, the most reliable and widely used technique is quantitative real-time polymerase chain reaction (qRT-PCR). Here, we assessed the impact of a reference gene's stability on a qRT-PCR analysis of long-term cultured canine skin fibroblasts. After successfully isolating the fibroblasts from canine skin tissues, they were cultured and evaluated for proliferation and β-galactosidase activity at different passage numbers. With extended culture, the fibroblasts showed a long doubling time and elevated β-galactosidase activity. Using three widely used algorithms, geNorm, Normfinder, and Bestkeeper, we identified HPRT1, YWHAZ, and GUSB as the most stable reference genes for both early- and late-passage fibroblasts. Conventional reference genes such as GAPDH were found to be less stable than those genes. The normalization of Vimentin by the stable genes showed statistical differences, whereas normalization by an unstable gene did not. Collectively, this study indicates that using stable reference genes is essential for accurately and reliably measuring gene expression in both early- and late-passage fibroblasts. These findings provide valuable insights into internal controls for gene expression studies and are expected to be utilized for analyzing gene expression patterns in molecular biology research.
Collapse
Affiliation(s)
- Sang-Yun Lee
- Department of Theriogenology and Biotechnology, College of Veterinary Medicine, Gyeongsang National University, Jinju 52628, Republic of Korea
- Stem Cell Convergence Research Center, Korea Research Institute Bioscience and Biotechnology (KRIBB), Daejeon 34141, Republic of Korea
| | - Yeon-Woo Jeong
- Department of Companion Animal and Animal Resources Science, Joongbu University, Geumsan 32713, Republic of Korea
| | - Yong-Ho Choe
- Department of Theriogenology and Biotechnology, College of Veterinary Medicine, Gyeongsang National University, Jinju 52628, Republic of Korea
| | - Seong-Ju Oh
- Department of Theriogenology and Biotechnology, College of Veterinary Medicine, Gyeongsang National University, Jinju 52628, Republic of Korea
| | - Rubel Miah
- Department of Obstetrics, College of Veterinary Medicine, Chonnam National University, 300 Yonbongdong, Buk-gu, Gwangju 61186, Republic of Korea
| | - Won-Jae Lee
- Department of Obstetrics, College of Veterinary Medicine, Kyungpook National University, Daegu 41566, Republic of Korea
| | - Sung-Lim Lee
- Department of Theriogenology and Biotechnology, College of Veterinary Medicine, Gyeongsang National University, Jinju 52628, Republic of Korea
- Research Institute of Life Sciences, Gyeongsang National University, Jinju 52828, Republic of Korea
| | - Eun-Yeong Bok
- Division of Animal Diseases & Health, National Institute of Animal Science, Rural Development Administration, Wanju 55365, Republic of Korea
| | - Dae-Sung Yoo
- Departement of Veterinary Epidemiology, College of Veterinary Medicine, Chonnam National University, 300 Yonbongdong, Buk-gu, Gwangju 61186, Republic of Korea
| | - Young-Bum Son
- Department of Obstetrics, College of Veterinary Medicine, Chonnam National University, 300 Yonbongdong, Buk-gu, Gwangju 61186, Republic of Korea
| |
Collapse
|
2
|
Stevenson AW, Cadby G, Wallace HJ, Melton PE, Martin LJ, Wood FM, Fear MW. Genetic influence on scar vascularity after burn injury in individuals of European ancestry: A prospective cohort study. Burns 2024; 50:1871-1884. [PMID: 38902133 DOI: 10.1016/j.burns.2024.05.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 03/01/2024] [Accepted: 05/02/2024] [Indexed: 06/22/2024]
Abstract
After burn injury there is considerable variation in scar outcome, partially due to genetic factors. Scar vascularity is one characteristic that varies between individuals, and this study aimed to identify genetic variants contributing to different scar vascularity outcomes. An exome-wide array association study and gene pathway analysis was performed on a prospective cohort of 665 patients of European ancestry treated for burn injury, using their scar vascularity (SV) sub-score, part of the modified Vancouver Scar Scale (mVSS), as an outcome measure. DNA was genotyped using the Infinium HumanCoreExome-24 BeadChip, imputed to the Haplotype Reference Consortium panel. Associations between genetic variants (single nucleotide polymorphisms) and SV were estimated using an additive genetic model adjusting for sex, age, % total body surface area and number of surgical procedures, utilising linear and multinomial logistic regression. No individual genetic variants achieved the cut-off threshold for significance. Gene sets were also analysed using the Functional Mapping and Annotation (FUMA) platform, in which biological processes indirectly related to angiogenesis were significantly represented. This study suggests that SNPs in genes associated with angiogenesis may influence SV, but further studies with larger datasets are essential to validate these findings.
Collapse
Affiliation(s)
- Andrew W Stevenson
- Burn Injury Research Unit, School of Biomedical Sciences, Faculty of Health and Medical Sciences, The University of Western Australia, Crawley, Australia.
| | - Gemma Cadby
- School of Population and Global Health, The University of Western Australia, Perth, Australia
| | - Hilary J Wallace
- School of Population and Global Health, The University of Western Australia, Perth, Australia
| | - Phillip E Melton
- School of Population and Global Health, The University of Western Australia, Perth, Australia; Menzies Research Institute, University of Tasmania, Hobart, Tasmania, Australia
| | - Lisa J Martin
- Burn Injury Research Unit, School of Biomedical Sciences, Faculty of Health and Medical Sciences, The University of Western Australia, Crawley, Australia; Burns Service of Western Australia, Princess Margaret Hospital for Children and Fiona Stanley Hospital, Perth, Australia
| | - Fiona M Wood
- Burn Injury Research Unit, School of Biomedical Sciences, Faculty of Health and Medical Sciences, The University of Western Australia, Crawley, Australia; Burns Service of Western Australia, Princess Margaret Hospital for Children and Fiona Stanley Hospital, Perth, Australia
| | - Mark W Fear
- Burn Injury Research Unit, School of Biomedical Sciences, Faculty of Health and Medical Sciences, The University of Western Australia, Crawley, Australia
| |
Collapse
|
3
|
Bae SG, Yin GN, Ock J, Suh JK, Ryu JK, Park J. Single-cell transcriptome analysis of cavernous tissues reveals the key roles of pericytes in diabetic erectile dysfunction. eLife 2024; 12:RP88942. [PMID: 38856719 PMCID: PMC11164535 DOI: 10.7554/elife.88942] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/11/2024] Open
Abstract
Erectile dysfunction (ED) affects a significant proportion of men aged 40-70 and is caused by cavernous tissue dysfunction. Presently, the most common treatment for ED is phosphodiesterase 5 inhibitors; however, this is less effective in patients with severe vascular disease such as diabetic ED. Therefore, there is a need for development of new treatment, which requires a better understanding of the cavernous microenvironment and cell-cell communications under diabetic condition. Pericytes are vital in penile erection; however, their dysfunction due to diabetes remains unclear. In this study, we performed single-cell RNA sequencing to understand the cellular landscape of cavernous tissues and cell type-specific transcriptional changes in diabetic ED. We found a decreased expression of genes associated with collagen or extracellular matrix organization and angiogenesis in diabetic fibroblasts, chondrocytes, myofibroblasts, valve-related lymphatic endothelial cells, and pericytes. Moreover, the newly identified pericyte-specific marker, Limb Bud-Heart (Lbh), in mouse and human cavernous tissues, clearly distinguishing pericytes from smooth muscle cells. Cell-cell interaction analysis revealed that pericytes are involved in angiogenesis, adhesion, and migration by communicating with other cell types in the corpus cavernosum; however, these interactions were highly reduced under diabetic conditions. Lbh expression is low in diabetic pericytes, and overexpression of LBH prevents erectile function by regulating neurovascular regeneration. Furthermore, the LBH-interacting proteins (Crystallin Alpha B and Vimentin) were identified in mouse cavernous pericytes through LC-MS/MS analysis, indicating that their interactions were critical for maintaining pericyte function. Thus, our study reveals novel targets and insights into the pathogenesis of ED in patients with diabetes.
Collapse
Affiliation(s)
- Seo-Gyeong Bae
- School of Life Sciences, Gwangju Institute of Science and Technology (GIST)GwangjuRepublic of Korea
| | - Guo Nan Yin
- National Research Center for Sexual Medicine and Department of Urolog, Inha University School of MedicineIncheonRepublic of Korea
| | - Jiyeon Ock
- National Research Center for Sexual Medicine and Department of Urolog, Inha University School of MedicineIncheonRepublic of Korea
| | - Jun-Kyu Suh
- National Research Center for Sexual Medicine and Department of Urolog, Inha University School of MedicineIncheonRepublic of Korea
| | - Ji-Kan Ryu
- National Research Center for Sexual Medicine and Department of Urolog, Inha University School of MedicineIncheonRepublic of Korea
- Program in Biomedical Science & Engineering, Inha UniversityIncheonRepublic of Korea
| | - Jihwan Park
- School of Life Sciences, Gwangju Institute of Science and Technology (GIST)GwangjuRepublic of Korea
| |
Collapse
|
4
|
Petrova IM, Chebanova SI, Khatsko SL, Kalinina TA, Zaitsev DV, Glukhareva TV. Spiroconjugated 1,2,3-triazolo[5,1- b]1,3,4-thiadiazine stimulates functional activity of fibroblasts under skin injury regeneration. Res Pharm Sci 2024; 19:267-275. [PMID: 39035820 PMCID: PMC11257193 DOI: 10.4103/rps.rps_74_23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2023] [Revised: 08/13/2023] [Accepted: 11/25/2023] [Indexed: 07/23/2024] Open
Abstract
Background and purpose One of the most important mechanisms of tissue regeneration is the high functional activity of cells, including proliferation. Currently, there are practically no effective skin cell activators on the pharmaceutical market. The purpose of this work was to demonstrate the stimulating effect of spiroconjugated 1,2,3-triazolo[5,1-b]1,3,4-thiadiazine (STT) on the functional activity of fibroblasts. Experimental approach STT containing ointment for dermal application was made. To assess in vivo effect of the STT a linear wound model in rats was tested. A combination of histological techniques and mechanical testing was employed to estimate the stimulating effect of STT on the functional activity of fibroblasts. Findings/Results The STT significantly increased the number of fibroblasts as well as the density and order of produced collagen fibers in the dermis during the wound healing process. As a result, a tissue was formed at the site of damage with the structure corresponding to normal skin. In addition, skin functions were restored, in particular mechanically. Conclusion and implications The results suggested the stimulating effect of the STT on fibroblast activity and demonstrated its potential for skin regeneration.
Collapse
Affiliation(s)
- Irina M Petrova
- Ural Federal University named after the first President of Russia B. N. Yeltsin, Yekaterinburg, 620026, Russia
| | - Sofya Iu Chebanova
- Ural Federal University named after the first President of Russia B. N. Yeltsin, Yekaterinburg, 620026, Russia
| | - Sergey L Khatsko
- Ural Federal University named after the first President of Russia B. N. Yeltsin, Yekaterinburg, 620026, Russia
- Federal State Budgetary Scientific Institution “Ural Federal Agrarian Scientific Research Centre, Ural Branch of Russian Academy of Sciences”, Yekaterinburg, 620142, Russia
| | - Tatyana A Kalinina
- Ural Federal University named after the first President of Russia B. N. Yeltsin, Yekaterinburg, 620026, Russia
| | - Dmitry V Zaitsev
- Ural Federal University named after the first President of Russia B. N. Yeltsin, Yekaterinburg, 620026, Russia
- Ural State Mining University, Yekaterinburg, 620144, Russia
| | - Tatyana V Glukhareva
- Ural Federal University named after the first President of Russia B. N. Yeltsin, Yekaterinburg, 620026, Russia
| |
Collapse
|
5
|
Yi D, An N, Li Q, Liu Q, Shao H, Zhou R, Wang J, Zhang Y, Ma L, Guo F, Li X, Liu Z, Cen S. Interferon-induced MXB protein restricts vimentin-dependent viral infection. Acta Pharm Sin B 2024; 14:2520-2536. [PMID: 38828143 PMCID: PMC11143536 DOI: 10.1016/j.apsb.2024.03.029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Revised: 02/16/2024] [Accepted: 03/14/2024] [Indexed: 06/05/2024] Open
Abstract
Type I interferon (IFN) inhibits a wide spectrum of viruses through stimulating the expression of antiviral proteins. As an IFN-induced protein, myxovirus resistance B (MXB) protein was reported to inhibit multiple highly pathogenic human viruses. It remains to be determined whether MXB employs a common mechanism to restrict different viruses. Here, we find that IFN alters the subcellular localization of hundreds of host proteins, and this IFN effect is partially lost upon MXB depletion. The results of our mechanistic study reveal that MXB recognizes vimentin (VIM) and recruits protein kinase B (AKT) to phosphorylate VIM at amino acid S38, which leads to reorganization of the VIM network and impairment of intracellular trafficking of virus protein complexes, hence causing a restriction of virus infection. These results highlight a new function of MXB in modulating VIM-mediated trafficking, which may lead towards a novel broad-spectrum antiviral strategy to control a large group of viruses that depend on VIM for successful replication.
Collapse
Affiliation(s)
- Dongrong Yi
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Science, Beijing 100050, China
| | - Ni An
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Science, Beijing 100050, China
| | - Quanjie Li
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Science, Beijing 100050, China
| | - Qian Liu
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Science, Beijing 100050, China
| | - Huihan Shao
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Science, Beijing 100050, China
| | - Rui Zhou
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Science, Beijing 100050, China
| | - Jing Wang
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Science, Beijing 100050, China
| | - Yongxin Zhang
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Science, Beijing 100050, China
| | - Ling Ma
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Science, Beijing 100050, China
| | - Fei Guo
- Institute of Pathogen Biology, Chinese Academy of Medical Science, Beijing 100730, China
| | - Xiaoyu Li
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Science, Beijing 100050, China
| | - Zhenlong Liu
- Lady Davis Institute for Medical Research, Jewish General Hospital, Montreal, Quebec H3T 1E2, Canada
| | - Shan Cen
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Science, Beijing 100050, China
| |
Collapse
|
6
|
Xu Y, Chen W, Yang H, Song Z, Wang Y, Su R, Mwacharo JM, Lv X, Sun W. miR-329b-5p Affects Sheep Intestinal Epithelial Cells against Escherichia coli F17 Infection. Vet Sci 2024; 11:206. [PMID: 38787178 PMCID: PMC11126089 DOI: 10.3390/vetsci11050206] [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: 04/10/2024] [Revised: 05/03/2024] [Accepted: 05/06/2024] [Indexed: 05/25/2024] Open
Abstract
Diarrhea is the most common issue in sheep farms, typically due to pathogenic Escherichia coli (E. coli) infections, such as E. coli F17. microRNA, a primary type of non-coding RNA, has been shown to be involved in diarrhea caused by pathogenic E. coli. To elucidate the profound mechanisms of miRNA in E. coli F17 infections, methods such as E. coli F17 adhesion assay, colony counting assay, relative quantification of bacterial E. coli fimbriae gene expression, indirect immune fluorescence (IF), Cell Counting Kit-8 (CCK-8), 5-ethynyl-2'-deoxyuridine (EdU), Western blotting (WB), and scratch assay were conducted to investigate the effect of miR-329b-5p overexpression/knock-down on E. coli F17 susceptibility of sheep intestinal epithelial cells (IECs). The findings indicated that miR-329b-5p enhances the E. coli F17 resistance of sheep IECs to E.coli F17 by promoting adhesion between E. coli F17 and IEC, as well as IEC proliferation and migration. In summary, miR-329b-5p plays a crucial role in the defense of sheep IECs against E. coli F17 infection, providing valuable insights into its mechanism of action.
Collapse
Affiliation(s)
- Yeling Xu
- College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China; (Y.X.); (W.C.)
| | - Weihao Chen
- College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China; (Y.X.); (W.C.)
| | - Huiguo Yang
- Institute of Animal Husbandry, Xinjiang Academy of Animal Sciences, Urumqi 830013, China;
| | - Zhenghai Song
- Dongshan Animal Epidemic Prevention Station, Wuzhong District, Suzhou 215100, China;
| | - Yeqing Wang
- Suzhou Taihu Dongshang Sheep Industry Development Co., Ltd., Suzhou 215000, China;
| | - Rui Su
- Suzhou Stud Farm Co., Ltd., Suzhou 215200, China;
| | - Joram M. Mwacharo
- International Centre for Agricultural Research in the Dry Areas, Addis Ababa 999047, Ethiopia;
| | - Xiaoyang Lv
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of Ministry of Education, Yangzhou University, Yangzhou 225009, China;
- International Joint Research Laboratory in Universities of Jiangsu Province of China for Domestic Animal Germplasm Resources and Genetic Improvement, Yangzhou 225009, China
| | - Wei Sun
- College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China; (Y.X.); (W.C.)
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of Ministry of Education, Yangzhou University, Yangzhou 225009, China;
- International Joint Research Laboratory in Universities of Jiangsu Province of China for Domestic Animal Germplasm Resources and Genetic Improvement, Yangzhou 225009, China
| |
Collapse
|
7
|
Herre C, Nshdejan A, Klopfleisch R, Corte GM, Bahramsoltani M. Knockdown of TPI in human dermal microvascular endothelial cells and its impact on angiogenesis in vitro. PLoS One 2023; 18:e0294933. [PMID: 38117832 PMCID: PMC10732452 DOI: 10.1371/journal.pone.0294933] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Accepted: 11/10/2023] [Indexed: 12/22/2023] Open
Abstract
INTRODUCTION Angiogenic behaviour has been shown as highly versatile among Endothelial cells (ECs) causing problems of in vitro assays of angiogenesis considering their reproducibility. It is indispensable to investigate influencing factors of the angiogenic potency of ECs. OBJECTIVE The present study aimed to analyse the impact of knocking down triosephosphate isomerase (TPI) on in vitro angiogenesis and simultaneously on vimentin (VIM) and adenosylmethionine synthetase isoform type 2 (MAT2A) expression. Furthermore, native expression profiles of TPI, VIM and MAT2A in the course of angiogenesis in vitro were examined. METHODS Two batches of human dermal microvascular ECs were cultivated over 50 days and stimulated to undergo angiogenesis. A shRNA-mediated knockdown of TPI was performed. During cultivation, time-dependant morphological changes were detected and applied for EC-staging as prerequisite for quantifying in vitro angiogenesis. Additionally, mRNA and protein levels of all proteins were monitored. RESULTS Opposed to native cells, knockdown cells were not able to enter late stages of angiogenesis and primarily displayed a downregulation of VIM and an uprise in MAT2A expression. Native cells increased their TPI expression and decreased their VIM expression during the course of angiogenesis in vitro. For MAT2A, highest expression was observed to be in the beginning and at the end of angiogenesis. CONCLUSION Knocking down TPI provoked expressional changes in VIM and MAT2A and a deceleration of in vitro angiogenesis, indicating that TPI represents an angiogenic protein. Native expression profiles lead to the assumption of VIM being predominantly relevant in beginning stages, MAT2A in beginning and late stages and TPI during the whole course of angiogenesis in vitro.
Collapse
Affiliation(s)
- Christina Herre
- Institute of Veterinary Anatomy, School of Veterinary Medicine, Freie Universität Berlin, Berlin, Germany
| | - Arpenik Nshdejan
- Institute of Veterinary Anatomy, School of Veterinary Medicine, Freie Universität Berlin, Berlin, Germany
| | - Robert Klopfleisch
- Institute of Veterinary Pathology, School of Veterinary Medicine, Freie Universität Berlin, Berlin, Germany
| | - Giuliano Mario Corte
- Department of Veterinary Medicine, Institute of Veterinary Anatomy, Universität Zürich, Zurich, Switzerland
| | - Mahtab Bahramsoltani
- Institute of Veterinary Anatomy, School of Veterinary Medicine, Freie Universität Berlin, Berlin, Germany
| |
Collapse
|
8
|
Morales-Guadarrama G, Méndez-Pérez EA, García-Quiroz J, Avila E, Ibarra-Sánchez MJ, Esparza-López J, García-Becerra R, Larrea F, Díaz L. The Inhibition of the FGFR/PI3K/Akt Axis by AZD4547 Disrupts the Proangiogenic Microenvironment and Vasculogenic Mimicry Arising from the Interplay between Endothelial and Triple-Negative Breast Cancer Cells. Int J Mol Sci 2023; 24:13770. [PMID: 37762073 PMCID: PMC10531243 DOI: 10.3390/ijms241813770] [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: 07/15/2023] [Revised: 08/29/2023] [Accepted: 09/04/2023] [Indexed: 09/29/2023] Open
Abstract
Vasculogenic mimicry (VM), a process in which aggressive cancer cells form tube-like structures, plays a crucial role in providing nutrients and escape routes. Highly plastic tumor cells, such as those with the triple-negative breast cancer (TNBC) phenotype, can develop VM. However, little is known about the interplay between the cellular components of the tumor microenvironment and TNBC cells' VM capacity. In this study, we analyzed the ability of endothelial and stromal cells to induce VM when interacting with TNBC cells and analyzed the involvement of the FGFR/PI3K/Akt pathway in this process. VM was corroborated using fluorescently labeled TNBC cells. Only endothelial cells triggered VM formation, suggesting a predominant role of paracrine/juxtacrine factors from an endothelial origin in VM development. Via immunocytochemistry, qPCR, and secretome analyses, we determined an increased expression of proangiogenic factors as well as stemness markers in VM-forming cancer cells. Similarly, endothelial cells primed by TNBC cells showed an upregulation of proangiogenic molecules, including FGF, VEGFA, and several inflammatory cytokines. Endothelium-dependent TNBC-VM formation was prevented by AZD4547 or LY294002, strongly suggesting the involvement of the FGFR/PI3K/Akt axis in this process. Given that VM is associated with poor clinical prognosis, targeting FGFR/PI3K/Akt pharmacologically may hold promise for treating and preventing VM in TNBC tumors.
Collapse
Affiliation(s)
- Gabriela Morales-Guadarrama
- Departamento de Biología de la Reproducción Dr. Carlos Gual Castro, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán (INCMNSZ), Vasco de Quiroga No. 15, Belisario Domínguez Sección XVI, Tlalpan, Ciudad de México 14080, Mexico; (G.M.-G.)
| | - Edgar A. Méndez-Pérez
- Departamento de Biología de la Reproducción Dr. Carlos Gual Castro, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán (INCMNSZ), Vasco de Quiroga No. 15, Belisario Domínguez Sección XVI, Tlalpan, Ciudad de México 14080, Mexico; (G.M.-G.)
| | - Janice García-Quiroz
- Departamento de Biología de la Reproducción Dr. Carlos Gual Castro, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán (INCMNSZ), Vasco de Quiroga No. 15, Belisario Domínguez Sección XVI, Tlalpan, Ciudad de México 14080, Mexico; (G.M.-G.)
| | - Euclides Avila
- Departamento de Biología de la Reproducción Dr. Carlos Gual Castro, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán (INCMNSZ), Vasco de Quiroga No. 15, Belisario Domínguez Sección XVI, Tlalpan, Ciudad de México 14080, Mexico; (G.M.-G.)
| | - María J. Ibarra-Sánchez
- Unidad de Bioquímica Dr. Guillermo Soberón Acevedo, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán (INCMNSZ), Vasco de Quiroga No. 15, Belisario Domínguez Sección XVI, Tlalpan, Ciudad de México 14080, Mexico
| | - José Esparza-López
- Unidad de Bioquímica Dr. Guillermo Soberón Acevedo, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán (INCMNSZ), Vasco de Quiroga No. 15, Belisario Domínguez Sección XVI, Tlalpan, Ciudad de México 14080, Mexico
| | - Rocío García-Becerra
- Departamento de Biología Molecular y Biotecnología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Av. Universidad 3000, Coyoacán, Ciudad de México 04510, Mexico
- Programa de Investigación de Cáncer de Mama, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Av. Universidad 3000, Coyoacán, Ciudad de México 04510, Mexico
| | - Fernando Larrea
- Departamento de Biología de la Reproducción Dr. Carlos Gual Castro, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán (INCMNSZ), Vasco de Quiroga No. 15, Belisario Domínguez Sección XVI, Tlalpan, Ciudad de México 14080, Mexico; (G.M.-G.)
| | - Lorenza Díaz
- Departamento de Biología de la Reproducción Dr. Carlos Gual Castro, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán (INCMNSZ), Vasco de Quiroga No. 15, Belisario Domínguez Sección XVI, Tlalpan, Ciudad de México 14080, Mexico; (G.M.-G.)
| |
Collapse
|
9
|
Dhumale P, Nielsen JV, Hansen ACS, Burton M, Beck HC, Jørgensen MG, Toyserkani NM, Haahr MK, Hansen ST, Lund L, Thomassen M, Sørensen JA, Andersen DC, Jensen CH, Sheikh SP. CD31 defines a subpopulation of human adipose-derived regenerative cells with potent angiogenic effects. Sci Rep 2023; 13:14401. [PMID: 37658225 PMCID: PMC10474028 DOI: 10.1038/s41598-023-41535-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Accepted: 08/28/2023] [Indexed: 09/03/2023] Open
Abstract
Cellular heterogeneity represents a major challenge for regenerative treatment using freshly isolated Adipose Derived Regenerative Cells (ADRCs). Emerging data suggest superior efficacy of ADRCs as compared to the ex vivo expanded and more homogeneous ADRCs (= ASCs) for indications involving (micro)vascular deficiency, however, it remains unknown which ADRC cell subtypes account for the improvement. Surprisingly, we found regarding erectile dysfunction (ED) that the number of injected CD31+ ADRCs correlated positively with erectile function 12 months after one bolus of autologous ADRCs. Comprehensive in vitro and ex vivo analyses confirmed superior pro-angiogenic and paracrine effects of human CD31+ enriched ADRCs compared to the corresponding CD31- and parent ADRCs. When CD31+, CD31- and ADRCs were co-cultured in aortic ring- and corpus cavernous tube formation assays, the CD31+ ADRCs induced significantly higher tube development. This effect was corroborated using conditioned medium (CM), while quantitative mass spectrometric analysis suggested that this is likely explained by secretory pro-angiogenic proteins including DKK3, ANGPT2, ANAX2 and VIM, all enriched in CD31+ ADRC CM. Single-cell RNA sequencing showed that transcripts of the upregulated and secreted proteins were present in 9 endothelial ADRC subsets including endothelial progenitor cells in the heterogenous non-cultured ADRCs. Our data suggest that the vascular benefit of using ADRCs in regenerative medicine is dictated by CD31+ ADRCs.
Collapse
Affiliation(s)
- Pratibha Dhumale
- Department of Clinical Research, University of Southern Denmark (SDU), Odense, Denmark
- Department of Clinical Biochemistry, Odense University Hospital (OUH), Odense, Denmark
| | - Jakob Vennike Nielsen
- Department of Clinical Biochemistry, Odense University Hospital (OUH), Odense, Denmark
| | | | - Mark Burton
- Department of Clinical Genetics, OUH, Odense, Denmark
| | - Hans Christian Beck
- Department of Clinical Research, University of Southern Denmark (SDU), Odense, Denmark
- Department of Clinical Biochemistry, Odense University Hospital (OUH), Odense, Denmark
| | - Mads Gustaf Jørgensen
- Department of Clinical Research, University of Southern Denmark (SDU), Odense, Denmark
- Research Unit for Plastic Surgery, Department of Clinical Research, SDU, Odense, Denmark
| | - Navid Mohamadpour Toyserkani
- Department of Plastic Surgery, OUH, Odense, Denmark
- Research Unit for Plastic Surgery, Department of Clinical Research, SDU, Odense, Denmark
| | | | - Sabrina Toft Hansen
- Department of Clinical Research, University of Southern Denmark (SDU), Odense, Denmark
- Department of Urology, OUH, Odense, Denmark
| | - Lars Lund
- Department of Clinical Research, University of Southern Denmark (SDU), Odense, Denmark
- Department of Urology, OUH, Odense, Denmark
| | - Mads Thomassen
- Department of Clinical Research, University of Southern Denmark (SDU), Odense, Denmark
- Department of Clinical Genetics, OUH, Odense, Denmark
| | - Jens Ahm Sørensen
- Department of Clinical Research, University of Southern Denmark (SDU), Odense, Denmark
- Department of Plastic Surgery, OUH, Odense, Denmark
- Research Unit for Plastic Surgery, Department of Clinical Research, SDU, Odense, Denmark
| | - Ditte Caroline Andersen
- Department of Clinical Research, University of Southern Denmark (SDU), Odense, Denmark
- Department of Clinical Biochemistry, Odense University Hospital (OUH), Odense, Denmark
| | - Charlotte Harken Jensen
- Department of Clinical Research, University of Southern Denmark (SDU), Odense, Denmark
- Department of Clinical Biochemistry, Odense University Hospital (OUH), Odense, Denmark
| | - Søren Paludan Sheikh
- Department of Clinical Research, University of Southern Denmark (SDU), Odense, Denmark.
- Department of Clinical Biochemistry, Odense University Hospital (OUH), Odense, Denmark.
| |
Collapse
|
10
|
Ye L, Fan T, Qin Y, Qiu C, Li L, Dai M, Zhou Y, Chen Y, Jiang Y. MicroRNA-455-3p accelerate malignant progression of tumor by targeting H2AFZ in colorectal cancer. Cell Cycle 2023; 22:777-795. [PMID: 36482739 PMCID: PMC10026930 DOI: 10.1080/15384101.2022.2154549] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Colorectal cancer (CRC) becomes the second leading cause of cancer-related deaths in 2020. Emerging studies have indicated that microRNAs (miRNAs) play a key role in tumorigenesis and progression. The dysfunctions of miR-455-3p are observed in many cancers. However, its biological function in CRC remains to be confirmed. By sequencing serum sample, miR-455-3p was found to be up-regulated in CRC patients. RT-qPCR demonstrated that the miR-455-3p expression was both higher in the serum and tumor tissues of CRC patients. Furthermore, it indicated that miR-455-3p had the ability in promoting cell proliferation, suppressing cell apoptosis, and stimulating cell migration. In vivo experiments also showed that miR-455-3p promoted tumor growth. Additionally, H2AFZ was proved as the direct gene target of miR-455-3p by dual-luciferase assay. Taken together, miR-455-3p functioned as a tumor promoter in CRC development by regulating H2AFZ directly. Thus, it has enormous potential as a biomarker in the diagnosis of CRC.
Collapse
Affiliation(s)
- Lizhen Ye
- State Key Laboratory of Chemical Oncogenomics, Key Laboratory of Chemical Biology, Tsinghua Shenzhen International Graduate School, Shenzhen, Guangdong, China
| | - Tingting Fan
- Department of Chemistry, Southern University of Science and Technology, Shenzhen, China
| | - Ying Qin
- Department of Gastrointestinal Surgery, Shenzhen Second People's Hospital, Shenzhen, Guangdong, China
| | - Cheng Qiu
- National & Local United Engineering Lab for Personalized Anti-tumor Drugs, Shenzhen Kivita Innovative Drug Discovery Institute, Tsinghua Shenzhen International Graduate School, Shenzhen, Guangdong, China
| | - Lulu Li
- State Key Laboratory of Chemical Oncogenomics, Key Laboratory of Chemical Biology, Tsinghua Shenzhen International Graduate School, Shenzhen, Guangdong, China
| | - Mengmeng Dai
- State Key Laboratory of Chemical Oncogenomics, Key Laboratory of Chemical Biology, Tsinghua Shenzhen International Graduate School, Shenzhen, Guangdong, China
| | - Yaoyao Zhou
- State Key Laboratory of Chemical Oncogenomics, Key Laboratory of Chemical Biology, Tsinghua Shenzhen International Graduate School, Shenzhen, Guangdong, China
| | - Yan Chen
- State Key Laboratory of Chemical Oncogenomics, Key Laboratory of Chemical Biology, Tsinghua Shenzhen International Graduate School, Shenzhen, Guangdong, China
| | - Yuyang Jiang
- State Key Laboratory of Chemical Oncogenomics, Key Laboratory of Chemical Biology, Tsinghua Shenzhen International Graduate School, Shenzhen, Guangdong, China
- Department of Chemistry, Southern University of Science and Technology, Shenzhen, China
- School of Pharmaceutical Sciences, Tsinghua University, Beijing, China
| |
Collapse
|
11
|
Nasehi R, Schieren J, Grannemann C, Palkowitz AL, Babendreyer A, Schwarz N, Aveic S, Ludwig A, Leube RE, Fischer H. Bioprinting-associated pulsatile hydrostatic pressure elicits a mild proinflammatory response in epi- and endothelial cells. BIOMATERIALS ADVANCES 2023; 147:213329. [PMID: 36801795 DOI: 10.1016/j.bioadv.2023.213329] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Revised: 01/19/2023] [Accepted: 02/02/2023] [Indexed: 02/10/2023]
Abstract
During nozzle-based bioprinting, like inkjet and microextrusion, cells are subjected to hydrostatic pressure for up to several minutes. The modality of the bioprinting-related hydrostatic pressure is either constant or pulsatile depending on the technique. We hypothesized that the difference in the modality of hydrostatic pressure affects the biological response of the processed cells differently. To test this, we used a custom-made setup to apply either controlled constant or pulsatile hydrostatic pressure on endothelial and epithelial cells. Neither bioprinting procedure visibly altered the distribution of selected cytoskeletal filaments, cell-substrate adhesions, and cell-cell contacts in either cell type. In addition, pulsatile hydrostatic pressure led to an immediate increase of intracellular ATP in both cell types. However, the bioprinting-associated hydrostatic pressure triggered a pro-inflammatory response in only the endothelial cells, with an increase of interleukin 8 (IL-8) and a decrease of thrombomodulin (THBD) transcripts. These findings demonstrate that the settings adopted during nozzle-based bioprinting cause hydrostatic pressure that can trigger a pro-inflammatory response in different barrier-forming cell types. This response is cell-type and pressure-modality dependent. The immediate interaction of the printed cells with native tissue and the immune system in vivo might potentially trigger a cascade of events. Our findings, therefore, are of major relevance in particular for novel intra-operative, multicellular bioprinting approaches.
Collapse
Affiliation(s)
- Ramin Nasehi
- Department of Dental Materials and Biomaterials Research, RWTH Aachen University Hospital, Pauwelsstraße 30, 52074 Aachen, Germany
| | - Jana Schieren
- Institute of Molecular and Cellular Anatomy, RWTH Aachen University, Templergraben 55, 52062 Aachen, Germany
| | - Caroline Grannemann
- Institute of Molecular Pharmacology, RWTH Aachen University Hospital, Pauwelsstraße 30, 52074 Aachen, Germany
| | - Alena L Palkowitz
- Department of Dental Materials and Biomaterials Research, RWTH Aachen University Hospital, Pauwelsstraße 30, 52074 Aachen, Germany
| | - Aaron Babendreyer
- Institute of Molecular Pharmacology, RWTH Aachen University Hospital, Pauwelsstraße 30, 52074 Aachen, Germany
| | - Nicole Schwarz
- Institute of Molecular and Cellular Anatomy, RWTH Aachen University, Templergraben 55, 52062 Aachen, Germany
| | - Sanja Aveic
- Department of Dental Materials and Biomaterials Research, RWTH Aachen University Hospital, Pauwelsstraße 30, 52074 Aachen, Germany
| | - Andreas Ludwig
- Institute of Molecular Pharmacology, RWTH Aachen University Hospital, Pauwelsstraße 30, 52074 Aachen, Germany
| | - Rudolf E Leube
- Institute of Molecular and Cellular Anatomy, RWTH Aachen University, Templergraben 55, 52062 Aachen, Germany
| | - Horst Fischer
- Department of Dental Materials and Biomaterials Research, RWTH Aachen University Hospital, Pauwelsstraße 30, 52074 Aachen, Germany.
| |
Collapse
|
12
|
Kapoor M, Chinnathambi S. TGF-β1 signalling in Alzheimer's pathology and cytoskeletal reorganization: a specialized Tau perspective. J Neuroinflammation 2023; 20:72. [PMID: 36915196 PMCID: PMC10012507 DOI: 10.1186/s12974-023-02751-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2022] [Accepted: 02/23/2023] [Indexed: 03/16/2023] Open
Abstract
Microtubule-associated protein, Tau has been implicated in Alzheimer's disease for its detachment from microtubules and formation of insoluble intracellular aggregates within the neurons. Recent findings have suggested the expulsion of Tau seeds in the extracellular domain and their prion-like propagation between neurons. Transforming Growth Factor-β1 (TGF-β1) is a ubiquitously occurring cytokine reported to carry out immunomodulation and neuroprotection in the brain. TGF-β-mediated regulation occurs at the level of neuronal survival and differentiation, glial activation (astrocyte and microglia), amyloid production-distribution-clearance and neurofibrillary tangle formation, all of which contributes to Alzheimer's pathophysiology. Its role in the reorganization of cytoskeletal architecture and remodelling of extracellular matrix to facilitate cellular migration has been well-documented. Microglia are the resident immune sentinels of the brain responsible for surveying the local microenvironment, migrating towards the beacon of pertinent damage and phagocytosing the cellular debris or patho-protein deposits at the site of insult. Channelizing microglia to target extracellular Tau could be a good strategy to combat the prion-like transmission and seeding problem in Alzheimer's disease. The current review focuses on reaffirming the role of TGF-β1 signalling in Alzheimer's pathology and cytoskeletal reorganization and considers utilizing the approach of TGF-β-triggered microglia-mediated targeting of extracellular patho-protein, Tau, as a possible potential strategy to combat Alzheimer's disease.
Collapse
Affiliation(s)
- Mahima Kapoor
- Neurobiology Group, Division of Biochemical Sciences, CSIR-National Chemical Laboratory (CSIR-NCL), Dr. Homi Bhabha Road, 411008, Pune, India.,Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Subashchandrabose Chinnathambi
- Neurobiology Group, Division of Biochemical Sciences, CSIR-National Chemical Laboratory (CSIR-NCL), Dr. Homi Bhabha Road, 411008, Pune, India. .,Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India. .,Department of Neurochemistry, National Institute of Mental Health and Neuro Sciences (NIMHANS), Institute of National Importance, Hosur Road, Bangalore, 560029, Karnataka, India.
| |
Collapse
|
13
|
Henao‐Restrepo J, López‐Murillo C, Valderrama‐Carmona P, Orozco‐Santa N, Gomez J, Gutiérrez‐Vargas J, Moraga R, Toledo J, Littau JL, Härtel S, Arboleda‐Velásquez JF, Sepulveda‐Falla D, Lopera F, Cardona‐Gómez GP, Villegas A, Posada‐Duque R. Gliovascular alterations in sporadic and familial Alzheimer's disease: APOE3 Christchurch homozygote glioprotection. Brain Pathol 2023; 33:e13119. [PMID: 36130084 PMCID: PMC10041169 DOI: 10.1111/bpa.13119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Accepted: 08/30/2022] [Indexed: 11/28/2022] Open
Abstract
In response to brain insults, astrocytes become reactive, promoting protection and tissue repair. However, astroglial reactivity is typical of brain pathologies, including Alzheimer's disease (AD). Considering the heterogeneity of the reactive response, the role of astrocytes in the course of different forms of AD has been underestimated. Colombia has the largest human group known to have familial AD (FAD). This group carries the autosomal dominant and fully penetrant mutation E280A in PSEN1, which causes early-onset AD. Recently, our group identified an E280A carrier who did not develop FAD. The individual was homozygous for the Christchurch mutation R136S in APOE3 (APOEch). Remarkably, APOE is the main genetic risk factor for developing sporadic AD (SAD) and most of cerebral ApoE is produced by astroglia. Here, we characterized astrocyte properties related to reactivity, glutamate homeostasis, and structural integrity of the gliovascular unit (GVU), as factors that could underlie the pathogenesis or protection of AD. Specifically, through histological and 3D microscopy analyses of postmortem samples, we briefly describe the histopathology and cytoarchitecture of the frontal cortex of SAD, FAD, and APOEch, and demonstrate that, while astrodegeneration and vascular deterioration are prominent in SAD, FAD is characterized by hyperreactive-like glia, and APOEch displays the mildest astrocytic and vascular alterations despite having the highest burden of Aβ. Notably, astroglial, gliovascular, and vascular disturbances, as well as brain cell death, correlate with the specific astrocytic phenotypes identified in each condition. This study provides new insights into the potential relevance of the gliovasculature in the development and protection of AD. To our knowledge, this is the first study assessing the components of the GVU in human samples of SAD, FAD, and APOEch.
Collapse
Affiliation(s)
- Julián Henao‐Restrepo
- Instituto de Biología, Facultad de Ciencias Exactas y NaturalesUniversidad de AntioquiaMedellínColombia
- Área de Neurobiología Celular y Molecular, Grupo de Neurociencias de AntioquiaUniversidad de AntioquiaMedellínColombia
| | - Carolina López‐Murillo
- Instituto de Biología, Facultad de Ciencias Exactas y NaturalesUniversidad de AntioquiaMedellínColombia
- Área de Neurobiología Celular y Molecular, Grupo de Neurociencias de AntioquiaUniversidad de AntioquiaMedellínColombia
| | - Pablo Valderrama‐Carmona
- Instituto de Biología, Facultad de Ciencias Exactas y NaturalesUniversidad de AntioquiaMedellínColombia
- Área de Neurobiología Celular y Molecular, Grupo de Neurociencias de AntioquiaUniversidad de AntioquiaMedellínColombia
| | - Natalia Orozco‐Santa
- Instituto de Biología, Facultad de Ciencias Exactas y NaturalesUniversidad de AntioquiaMedellínColombia
- Área de Neurobiología Celular y Molecular, Grupo de Neurociencias de AntioquiaUniversidad de AntioquiaMedellínColombia
| | - Johana Gomez
- Grupo de Neurociencias de Antioquia, Facultad de MedicinaSIU, Universidad de AntioquiaMedellínColombia
| | - Johanna Gutiérrez‐Vargas
- Instituto de Biología, Facultad de Ciencias Exactas y NaturalesUniversidad de AntioquiaMedellínColombia
- Health Sciences FacultyRemington University CorporationMedellínColombia
| | - Renato Moraga
- Biomedical Neuroscience Institute BNI, Faculty of MedicineUniversity of ChileSantiagoChile
| | - Jorge Toledo
- Biomedical Neuroscience Institute BNI, Faculty of MedicineUniversity of ChileSantiagoChile
| | - Jessica Lisa Littau
- Molecular Neuropathology of Alzheimer's DiseaseInstitute of Neuropathology, University Medical Center Hamburg‐EppendorfHamburgGermany
| | - Steffen Härtel
- Biomedical Neuroscience Institute BNI, Faculty of MedicineUniversity of ChileSantiagoChile
| | - Joseph F. Arboleda‐Velásquez
- Schepens Eye Research Institute of Mass Eye and Ear, Department of OphthalmologyHarvard Medical SchoolBostonMassachusettsUSA
| | - Diego Sepulveda‐Falla
- Molecular Neuropathology of Alzheimer's DiseaseInstitute of Neuropathology, University Medical Center Hamburg‐EppendorfHamburgGermany
| | - Francisco Lopera
- Grupo de Neurociencias de Antioquia, Facultad de MedicinaSIU, Universidad de AntioquiaMedellínColombia
| | - Gloria Patricia Cardona‐Gómez
- Área de Neurobiología Celular y Molecular, Grupo de Neurociencias de AntioquiaUniversidad de AntioquiaMedellínColombia
| | - Andrés Villegas
- Grupo de Neurociencias de Antioquia, Facultad de MedicinaSIU, Universidad de AntioquiaMedellínColombia
| | - Rafael Posada‐Duque
- Instituto de Biología, Facultad de Ciencias Exactas y NaturalesUniversidad de AntioquiaMedellínColombia
- Área de Neurobiología Celular y Molecular, Grupo de Neurociencias de AntioquiaUniversidad de AntioquiaMedellínColombia
| |
Collapse
|
14
|
Lampe JB, Desai PP, Tripathi AK, Sabnis NA, Chen Z, Ranjan AP, Vishwanatha JK. Cabazitaxel-Loaded Nanoparticles Reduce the Invasiveness in Metastatic Prostate Cancer Cells: Beyond the Classical Taxane Function. Pharmaceutics 2023; 15:662. [PMID: 36839985 PMCID: PMC9967362 DOI: 10.3390/pharmaceutics15020662] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 02/09/2023] [Accepted: 02/09/2023] [Indexed: 02/18/2023] Open
Abstract
Bone-metastatic prostate cancer symbolizes the beginning of the later stages of the disease. We designed a cabazitaxel-loaded, poly (lactic-co-glycolic acid) (PLGA) nanoparticle using an emulsion-diffusion-evaporation technique. Bis (sulfosuccinimidyl) suberate (BS3) was non-covalently inserted into the nanoparticle as a linker for the conjugation of a bone-targeting moiety to the outside of the nanoparticle. We hypothesized that the nanoparticles would have the ability to inhibit the epithelial-to-mesenchymal transition (EMT), invasion, and migration in prostate cancer cells. Targeted, cabazitaxel-loaded nanoparticles attenuated the EMT marker, Vimentin, and led to an increased E-cadherin expression. These changes impart epithelial characteristics and inhibit invasive properties in cancer progression. Consequently, progression to distant sites is also mitigated. We observed the reduction of phosphorylated Src at tyrosine 416, along with increased expression of phosphorylated cofilin at serine 3. These changes could affect migration and invasion pathways in cancer cells. Both increased p-120 catenin and inhibition in IL-8 expression were seen in targeted, cabazitaxel-loaded nanoparticles. Overall, our data show that the targeted, cabazitaxel-loaded nanoparticles can act as a promising treatment for metastatic prostate cancer by inhibiting EMT, invasion, and migration, in prostate cancer cells.
Collapse
Affiliation(s)
- Jana B. Lampe
- Department of Microbiology, Immunology, and Genetics, University of North Texas Health Science Center, 3500 Camp Bowie Blvd., Fort Worth, TX 76107, USA
| | - Priyanka P. Desai
- Department of Microbiology, Immunology, and Genetics, University of North Texas Health Science Center, 3500 Camp Bowie Blvd., Fort Worth, TX 76107, USA
| | - Amit K. Tripathi
- Department of Microbiology, Immunology, and Genetics, University of North Texas Health Science Center, 3500 Camp Bowie Blvd., Fort Worth, TX 76107, USA
| | - Nirupama A. Sabnis
- Department of Microbiology, Immunology, and Genetics, University of North Texas Health Science Center, 3500 Camp Bowie Blvd., Fort Worth, TX 76107, USA
| | - Zhe Chen
- Department of Biophysics, University of Texas Southwestern Medical Center, 6001 Forest Park Road, Dallas, TX 75390, USA
| | - Amalendu P. Ranjan
- Department of Microbiology, Immunology, and Genetics, University of North Texas Health Science Center, 3500 Camp Bowie Blvd., Fort Worth, TX 76107, USA
| | - Jamboor K. Vishwanatha
- Department of Microbiology, Immunology, and Genetics, University of North Texas Health Science Center, 3500 Camp Bowie Blvd., Fort Worth, TX 76107, USA
| |
Collapse
|
15
|
Fleischer JR, Schmitt AM, Haas G, Xu X, Zeisberg EM, Bohnenberger H, Küffer S, Teuwen LA, Karras PJ, Beißbarth T, Bleckmann A, Planque M, Fendt SM, Vermeulen P, Ghadimi M, Kalucka J, De Oliveira T, Conradi LC. Molecular differences of angiogenic versus vessel co-opting colorectal cancer liver metastases at single-cell resolution. Mol Cancer 2023; 22:17. [PMID: 36691028 PMCID: PMC9872436 DOI: 10.1186/s12943-023-01713-1] [Citation(s) in RCA: 18] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2022] [Accepted: 12/31/2022] [Indexed: 01/25/2023] Open
Abstract
BACKGROUND Colorectal cancer liver metastases (CRCLM) are associated with a poor prognosis, reflected by a five-year survival rate of 14%. Anti-angiogenic therapy through anti-VEGF antibody administration is one of the limited therapies available. However, only a subgroup of metastases uses sprouting angiogenesis to secure their nutrients and oxygen supply, while others rely on vessel co-option (VCO). The distinct mode of vascularization is reflected by specific histopathological growth patterns (HGPs), which have proven prognostic and predictive significance. Nevertheless, their molecular mechanisms are poorly understood. METHODS We evaluated CRCLM from 225 patients regarding their HGP and clinical data. Moreover, we performed spatial (21,804 spots) and single-cell (22,419 cells) RNA sequencing analyses to explore molecular differences in detail, further validated in vitro through immunohistochemical analysis and patient-derived organoid cultures. RESULTS We detected specific metabolic alterations and a signature of WNT signalling activation in metastatic cancer cells related to the VCO phenotype. Importantly, in the corresponding healthy liver of CRCLM displaying sprouting angiogenesis, we identified a predominantly expressed capillary subtype of endothelial cells, which could be further explored as a possible predictor for HGP relying on sprouting angiogenesis. CONCLUSION These findings may prove to be novel therapeutic targets to the treatment of CRCLM, in special the ones relying on VCO.
Collapse
Affiliation(s)
- Johannes Robert Fleischer
- Department of General, Visceral and Pediatric Surgery, University Medical Center Göttingen, Robert-Koch-Straβe 40, 37075, Göttingen, Germany
| | - Alexandra Maria Schmitt
- Department of General, Visceral and Pediatric Surgery, University Medical Center Göttingen, Robert-Koch-Straβe 40, 37075, Göttingen, Germany
| | - Gwendolyn Haas
- Department of General, Visceral and Pediatric Surgery, University Medical Center Göttingen, Robert-Koch-Straβe 40, 37075, Göttingen, Germany
| | - Xingbo Xu
- Department of Cardiology and Pneumology, University Medical Center Göttingen, Robert-Koch-Straβe 40, 37075, Göttingen, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site, Göttingen, Germany
| | - Elisabeth Maria Zeisberg
- Department of Cardiology and Pneumology, University Medical Center Göttingen, Robert-Koch-Straβe 40, 37075, Göttingen, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site, Göttingen, Germany
| | - Hanibal Bohnenberger
- Institute of Pathology, University Medical Center Göttingen, Robert-Koch-Straβe40, 37075, Göttingen, Germany
| | - Stefan Küffer
- Institute of Pathology, University Medical Center Göttingen, Robert-Koch-Straβe40, 37075, Göttingen, Germany
| | - Laure-Anne Teuwen
- Department of Oncology, Antwerp University Hospital (UZA), Drie Eikenstraat 655, 2650, Edegem, Belgium
| | - Philipp Johannes Karras
- Department of General, Visceral and Pediatric Surgery, University Medical Center Göttingen, Robert-Koch-Straβe 40, 37075, Göttingen, Germany
- Department of General- and Visceral Surgery, Raphaelsklinik Münster, Loerstraße 23, 48143, Münster, Germany
| | - Tim Beißbarth
- Department of Medical Bioinformatics, University Medical Center Göttingen, Goldschmidtstraße 1, 37077, Göttingen, Germany
| | - Annalen Bleckmann
- Department of Medicine A, Hematology, Oncology, and Pneumology, University Hospital Münster, 48149, Münster, Germany
| | - Mélanie Planque
- Laboratory of Cellular Metabolism and Metabolic Regulation, VIB-KU Leuven Center for Cancer Biology, VIB, Leuven, Belgium
- Laboratory of Cellular Metabolism and Metabolic Regulation, Department of Oncology, KU Leuven and Leuven Cancer Institute (LKI), Leuven, Belgium
| | - Sarah-Maria Fendt
- Laboratory of Cellular Metabolism and Metabolic Regulation, VIB-KU Leuven Center for Cancer Biology, VIB, Leuven, Belgium
- Laboratory of Cellular Metabolism and Metabolic Regulation, Department of Oncology, KU Leuven and Leuven Cancer Institute (LKI), Leuven, Belgium
| | - Peter Vermeulen
- Translational Cancer Research Unit, GZA Hospitals, Sint-Augustinus, University of Antwerp, Antwerp, Belgium
| | - Michael Ghadimi
- Department of General, Visceral and Pediatric Surgery, University Medical Center Göttingen, Robert-Koch-Straβe 40, 37075, Göttingen, Germany
| | - Joanna Kalucka
- Department of Biomedicine, Aarhus University, Høegh-Guldbergsgade 10, 8000, Aarhus C, Denmark
- Steno Diabetes Center Aarhus, Aarhus University Hospital, Aarhus, Denmark
| | - Tiago De Oliveira
- Department of General, Visceral and Pediatric Surgery, University Medical Center Göttingen, Robert-Koch-Straβe 40, 37075, Göttingen, Germany
| | - Lena-Christin Conradi
- Department of General, Visceral and Pediatric Surgery, University Medical Center Göttingen, Robert-Koch-Straβe 40, 37075, Göttingen, Germany.
| |
Collapse
|
16
|
Liu S, Ortiz A, Stavrou A, Talusan AR, Costa M. Extracellular Vesicles as Mediators of Nickel-Induced Cancer Progression. Int J Mol Sci 2022; 23:ijms232416111. [PMID: 36555753 PMCID: PMC9785150 DOI: 10.3390/ijms232416111] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 12/07/2022] [Accepted: 12/13/2022] [Indexed: 12/23/2022] Open
Abstract
Emerging evidence suggests that extracellular vesicles (EVs), which represent a crucial mode of intercellular communication, play important roles in cancer progression by transferring oncogenic materials. Nickel (Ni) has been identified as a human group I carcinogen; however, the underlying mechanisms governing Ni-induced carcinogenesis are still being elucidated. Here, we present data demonstrating that Ni exposure generates EVs that contribute to Ni-mediated carcinogenesis and cancer progression. Human bronchial epithelial (BEAS-2B) cells and human embryonic kidney-293 (HEK293) cells were chronically exposed to Ni to generate Ni-treated cells (Ni-6W), Ni-transformed BEAS-2B cells (Ni-3) and Ni-transformed HEK293 cells (HNi-4). The signatures of EVs isolated from Ni-6W, Ni-3, HNi-4, BEAS-2B, and HEK293 were analyzed. Compared to their respective untreated cells, Ni-6W, Ni-3, and HNi-4 released more EVs. This change in EV release coincided with increased transcription of the EV biogenesis markers CD82, CD63, and flotillin-1 (FLOT). Additionally, EVs from Ni-transformed cells had enriched protein and RNA, a phenotype also observed in other studies characterizing EVs from cancer cells. Interestingly, both epithelial cells and human umbilical vein endothelial (HUVEC) cells showed a preference for taking up Ni-altered EVs compared to EVs released from the untreated cells. Moreover, these Ni-altered EVs induced inflammatory responses in both epithelial and endothelial cells and increased the expression of coagulation markers in endothelial cells. Prolonged treatment of Ni-alerted EVs for two weeks induced the epithelial-to-mesenchymal transition (EMT) in BEAS-2B cells. This study is the first to characterize the effect of Ni on EVs and suggests the potential role of EVs in Ni-induced cancer progression.
Collapse
Affiliation(s)
| | | | | | | | - Max Costa
- Correspondence: ; Tel.: +1-646-754-9443
| |
Collapse
|
17
|
Dewing JM, Saunders V, O’Kelly I, Wilson DI. Defining cardiac cell populations and relative cellular composition of the early fetal human heart. PLoS One 2022; 17:e0259477. [PMID: 36449524 PMCID: PMC9710754 DOI: 10.1371/journal.pone.0259477] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Accepted: 11/08/2022] [Indexed: 12/03/2022] Open
Abstract
While the adult human heart is primarily composed of cardiomyocytes, fibroblasts, endothelial and smooth muscle cells, the cellular composition during early development remains largely unknown. Reliable identification of fetal cardiac cell types using protein markers is critical to understand cardiac development and delineate the cellular composition of the developing human heart. This is the first study to use immunohistochemistry (IHC), flow cytometry and RT-PCR analyses to investigate the expression and specificity of commonly used cardiac cell markers in the early human fetal heart (8-12 post-conception weeks). The expression of previously reported protein markers for the detection of cardiomyocytes (Myosin Heavy Chain (MHC) and cardiac troponin I (cTnI), fibroblasts (DDR2, THY1, Vimentin), endothelial cells (CD31) and smooth muscle cells (α-SMA) were assessed. Two distinct populations of cTnI positive cells were identified through flow cytometry, with MHC positive cardiomyocytes showing high cTnI expression (cTnIHigh) while MHC negative non-myocytes showed lower cTnI expression (cTnILow). cTnI expression in non-myocytes was further confirmed by IHC and RT-PCR analyses, suggesting troponins are not cardiomyocyte-specific and may play distinct roles in non-muscle cells during early development. Vimentin (VIM) was expressed in cultured ventricular fibroblast populations and flow cytometry revealed VIMHigh and VIMLow cell populations in the fetal heart. MHC positive cardiomyocytes were VIMLow whilst CD31 positive endothelial cells were VIMHigh. Using markers investigated within this study, we characterised fetal human cardiac populations and estimate that 75-80% of fetal cardiac cells are cardiomyocytes and are MHC+/cTnIHigh/VIMLow, whilst non-myocytes comprise 20-25% of total cells and are MHC-/cTnILow/VIMHigh, with CD31+ endothelial cells comprising ~9% of this population. These findings show distinct differences from those reported for adult heart.
Collapse
Affiliation(s)
- Jennifer M. Dewing
- Institute for Developmental Sciences, School of Human Development and Health, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
- * E-mail:
| | - Vinay Saunders
- Institute for Developmental Sciences, School of Human Development and Health, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - Ita O’Kelly
- Institute for Developmental Sciences, School of Human Development and Health, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
- Immunocore Ltd, Abingdon, Oxford, United Kingdom
| | - David I. Wilson
- Institute for Developmental Sciences, School of Human Development and Health, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| |
Collapse
|
18
|
Li Y, Zhang H, Long W, Gao M, Guo W, Yu L. Inhibition of NLRP3 and Golph3 ameliorates diabetes-induced neuroinflammation in vitro and in vivo. Aging (Albany NY) 2022; 14:8745-8762. [DOI: 10.18632/aging.204363] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Accepted: 10/21/2022] [Indexed: 11/16/2022]
Affiliation(s)
- Yuan Li
- State Key Laboratory for Zoonotic Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine, Department of Endocrinology and Department of Interventional Therapy of First Hospital of Jilin University, Jilin University, Changchun 130000, China
- Innovation Pharmaceutical Research Institute of Shijiazhuang No. 4 Pharmaceutical Co., Ltd., Hebei Guangxiang Pharmaceutical Co., Ltd., Shijiazhuang 050000, China
| | - Haifeng Zhang
- State Key Laboratory for Zoonotic Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine, Department of Endocrinology and Department of Interventional Therapy of First Hospital of Jilin University, Jilin University, Changchun 130000, China
| | - Weihong Long
- State Key Laboratory for Zoonotic Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine, Department of Endocrinology and Department of Interventional Therapy of First Hospital of Jilin University, Jilin University, Changchun 130000, China
| | - Menghan Gao
- State Key Laboratory for Zoonotic Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine, Department of Endocrinology and Department of Interventional Therapy of First Hospital of Jilin University, Jilin University, Changchun 130000, China
| | - Weiying Guo
- State Key Laboratory for Zoonotic Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine, Department of Endocrinology and Department of Interventional Therapy of First Hospital of Jilin University, Jilin University, Changchun 130000, China
| | - Lu Yu
- State Key Laboratory for Zoonotic Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine, Department of Endocrinology and Department of Interventional Therapy of First Hospital of Jilin University, Jilin University, Changchun 130000, China
| |
Collapse
|
19
|
Defining A Liquid Biopsy Profile of Circulating Tumor Cells and Oncosomes in Metastatic Colorectal Cancer for Clinical Utility. Cancers (Basel) 2022; 14:cancers14194891. [PMID: 36230811 PMCID: PMC9563925 DOI: 10.3390/cancers14194891] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2022] [Accepted: 09/30/2022] [Indexed: 11/23/2022] Open
Abstract
Simple Summary Metastatic colorectal cancer (mCRC) is typified by its tumor heterogeneity and changing disease states, suggesting that personalized medicine approaches could be vital to improving clinical practice. As a minimally invasive approach, the liquid biopsy has the potential to be a powerful longitudinal prognostic tool. We investigated mCRC patients’ peripheral blood samples using an enrichment-free single-cell approach to capture the broader rare-event population beyond the conventionally detected epithelial-derived circulating tumor cell (CTC). Our analysis reveals a heterogenous profile of CTCs and oncosomes not commonly found in normal donor samples. We identified select rare cell types based on their distinct immunofluorescence expression and morphology across multiple assays. Lastly, we highlight correlations between enumerations of the blood-based analytes and progression-free survival. This study clinically validates an unbiased rare-event approach in the liquid biopsy, motivating future studies to further investigate these analytes for their prognostic potential. Abstract Metastatic colorectal cancer (mCRC) is characterized by its extensive disease heterogeneity, suggesting that individualized analysis could be vital to improving patient outcomes. As a minimally invasive approach, the liquid biopsy has the potential to longitudinally monitor heterogeneous analytes. Current platforms primarily utilize enrichment-based approaches for epithelial-derived circulating tumor cells (CTC), but this subtype is infrequent in the peripheral blood (PB) of mCRC patients, leading to the liquid biopsy’s relative disuse in this cancer type. In this study, we evaluated 18 PB samples from 10 mCRC patients using the unbiased high-definition single-cell assay (HDSCA). We first employed a rare-event (Landscape) immunofluorescence (IF) protocol, which captured a heterogenous CTC and oncosome population, the likes of which was not observed across 50 normal donor (ND) samples. Subsequent analysis was conducted using a colorectal-targeted IF protocol to assess the frequency of CDX2-expressing CTCs and oncosomes. A multi-assay clustering analysis isolated morphologically distinct subtypes across the two IF stains, demonstrating the value of applying an unbiased single-cell approach to multiple assays in tandem. Rare-event enumerations at a single timepoint and the variation of these events over time correlated with progression-free survival. This study supports the clinical utility of an unbiased approach to interrogating the liquid biopsy in mCRC, representing the heterogeneity within the CTC classification and warranting the further molecular characterization of the rare-event analytes with clinical promise.
Collapse
|
20
|
Di Carmine S, Scott MM, McLean MH, McSorley HJ. The role of interleukin-33 in organ fibrosis. DISCOVERY IMMUNOLOGY 2022; 1:kyac006. [PMID: 38566909 PMCID: PMC10917208 DOI: 10.1093/discim/kyac006] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Revised: 08/06/2022] [Accepted: 09/22/2022] [Indexed: 04/04/2024]
Abstract
Interleukin (IL)-33 is highly expressed in the nucleus of cells present at barrier sites and signals via the ST2 receptor. IL-33 signalling via ST2 is essential for return to tissue homeostasis after acute inflammation, promoting fibrinogenesis and wound healing at injury sites. However, this wound-healing response becomes aberrant during chronic or sustained inflammation, leading to transforming growth factor beta (TGF-β) release, excessive extracellular matrix deposition, and fibrosis. This review addresses the role of the IL-33 pathway in fibrotic diseases of the lung, liver, gastrointestinal tract, skin, kidney and heart. In the lung and liver, IL-33 release leads to the activation of pro-fibrotic TGF-β, and in these sites, IL-33 has clear pro-fibrotic roles. In the gastrointestinal tract, skin, and kidney, the role of IL-33 is more complex, being both pro-fibrotic and tissue protective. Finally, in the heart, IL-33 serves cardioprotective functions by favouring tissue healing and preventing cardiomyocyte death. Altogether, this review indicates the presence of an unclear and delicate balance between resolving and pro-fibrotic capabilities of IL-33, which has a central role in the modulation of type 2 inflammation and fibrosis in response to tissue injury.
Collapse
Affiliation(s)
- Samuele Di Carmine
- Division of Cell Signalling and Immunology, School of Life Sciences, Wellcome Trust Building, University of Dundee, Dundee, UK
| | - Molly M Scott
- Division of Molecular and Clinical Medicine, School of Medicine, University of Dundee, Ninewells Hospital, Dundee, UK
| | - Mairi H McLean
- Division of Molecular and Clinical Medicine, School of Medicine, University of Dundee, Ninewells Hospital, Dundee, UK
| | - Henry J McSorley
- Division of Cell Signalling and Immunology, School of Life Sciences, Wellcome Trust Building, University of Dundee, Dundee, UK
| |
Collapse
|
21
|
Ahmad M, Sun Y, Jia X, Li J, Zhang L, Yang Z, Lin Y, Zhang X, Khan ZA, Qian J, Luo Y. Therapeutic values of chick early amniotic fluid (ceAF) that facilitates wound healing via potentiating a SASP-mediated transient senescence. Genes Dis 2022; 9:1345-1356. [PMID: 35873014 PMCID: PMC9293714 DOI: 10.1016/j.gendis.2021.03.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 02/24/2021] [Accepted: 03/12/2021] [Indexed: 12/17/2022] Open
Abstract
Inflammatory, proliferative and remodeling phases constitute a cutaneous wound healing program. Therapeutic applications and medication are available; however, they commonly are comprised of fortified preservatives that might prolong the healing process. Chick early amniotic fluids (ceAF) contain native therapeutic factors with balanced chemokines, cytokines and growth-related factors; their origins in principle dictate no existence of harmful agents that would otherwise hamper embryo development. Instead, they possess a spectrum of molecules driving expeditious mitotic divisions and possibly exerting other functions. Employing both in vitro and in vivo models, we examined ceAF's therapeutic potentials in wound healing and found intriguing involvement of transient senescence, known to be intimately intermingled with Senescence Associated Secretory Phenotypes (SASP) that function in addition to or in conjunction with ceAF to facilitate wound healing. In our cutaneous wound healing models, a low dose of ceAF exhibited the best efficacies; however, higher doses attenuated the wound healing presumably by inducing p16 expression over a threshold. Our studies thus link an INK4/ARF locus-mediated signaling cascade to cutaneous wound healing, suggesting therapeutic potentials of ceAF exerting functions likely by driving transient senescence, expediting cellular proliferation, migration, and describing a homeostatic and balanced dosage strategy in medical intervention.
Collapse
Affiliation(s)
- Mashaal Ahmad
- Department of Biochemistry and Cancer Institute of the Second Affiliated Hospital, Key Laboratory of Cancer Prevention and Intervention of China National MOE, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, PR China
| | - Yandi Sun
- Department of Biochemistry and Cancer Institute of the Second Affiliated Hospital, Key Laboratory of Cancer Prevention and Intervention of China National MOE, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, PR China
| | - Xueyao Jia
- Department of Biochemistry and Cancer Institute of the Second Affiliated Hospital, Key Laboratory of Cancer Prevention and Intervention of China National MOE, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, PR China
| | - Jingjia Li
- Department of Biochemistry and Cancer Institute of the Second Affiliated Hospital, Key Laboratory of Cancer Prevention and Intervention of China National MOE, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, PR China
| | - Lihong Zhang
- Department of Biochemistry and Cancer Institute of the Second Affiliated Hospital, Key Laboratory of Cancer Prevention and Intervention of China National MOE, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, PR China
| | - Ze Yang
- Department of Biochemistry and Cancer Institute of the Second Affiliated Hospital, Key Laboratory of Cancer Prevention and Intervention of China National MOE, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, PR China
| | - Yindan Lin
- Department of Biochemistry and Cancer Institute of the Second Affiliated Hospital, Key Laboratory of Cancer Prevention and Intervention of China National MOE, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, PR China
| | - Xueyun Zhang
- Department of Biochemistry and Cancer Institute of the Second Affiliated Hospital, Key Laboratory of Cancer Prevention and Intervention of China National MOE, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, PR China
| | - Zara Ahmad Khan
- State Key Laboratory of Oncogenes and Related Genes, Institute for Personalized Medicine, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200240, PR China
| | - Jin Qian
- Zhejiang HygeianCells BioMedical Co. Ltd., Hangzhou, Zhejiang 310000, PR China
| | - Yan Luo
- Department of Biochemistry and Cancer Institute of the Second Affiliated Hospital, Key Laboratory of Cancer Prevention and Intervention of China National MOE, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, PR China
| |
Collapse
|
22
|
Methacrylic Acid-Based Regenerative Biomaterials: Explorations into the MAAgic. REGENERATIVE ENGINEERING AND TRANSLATIONAL MEDICINE 2022. [DOI: 10.1007/s40883-022-00263-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
23
|
Salvador J, Hernandez GE, Ma F, Abrahamson CW, Pellegrini M, Goldman R, Ridge KM, Iruela-Arispe ML. Transcriptional Evaluation of the Ductus Arteriosus at the Single-Cell Level Uncovers a Requirement for Vim (Vimentin) for Complete Closure. Arterioscler Thromb Vasc Biol 2022; 42:732-742. [PMID: 35443793 PMCID: PMC9806842 DOI: 10.1161/atvbaha.121.317172] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
OBJECTIVE Failure to close the ductus arteriosus, patent ductus arteriosus, accounts for 10% of all congenital heart defects. Despite significant advances in patent ductus arteriosus management, including pharmacological treatment targeting the prostaglandin pathway, a proportion of patients fail to respond and must undergo surgical intervention. Thus, further refinement of the cellular and molecular mechanisms that govern vascular remodeling of this vessel is required. METHODS We performed single-cell RNA-sequencing of the ductus arteriosus in mouse embryos at E18.5 (embryonic day 18.5), and P0.5 (postnatal day 0.5), and P5 to identify transcriptional alterations that might be associated with remodeling. We further confirmed our findings using transgenic mouse models coupled with immunohistochemistry analysis. RESULTS The intermediate filament vimentin emerged as a candidate that might contribute to closure of the ductus arteriosus. Indeed, mice with genetic deletion of vimentin fail to complete vascular remodeling of the ductus arteriosus. To seek mechanisms, we turned to the RNA-sequencing data that indicated changes in Jagged1 with similar profile to vimentin and pointed to potential links with Notch. In fact, Notch3 signaling was impaired in vimentin null mice and vimentin null mice phenocopies patent ductus arteriosus in Jagged1 endothelial and smooth muscle deleted mice. CONCLUSIONS Through single-cell RNA-sequencing and by tracking closure of the ductus arteriosus in mice, we uncovered the unexpected contribution of vimentin in driving complete closure of the ductus arteriosus through a mechanism that includes deregulation of the Notch signaling pathway.
Collapse
Affiliation(s)
- Jocelynda Salvador
- Department of Cell and Development Biology (J.S., C.W.A., R.G., K.M.R., M.L.I.-A.), Northwestern University, Chicago
| | - Gloria E Hernandez
- Molecular Biology Institute (G.E.H., F.M.), University of California, Los Angeles
| | - Feiyang Ma
- Molecular Biology Institute (G.E.H., F.M.), University of California, Los Angeles
| | - Cyrus W Abrahamson
- Department of Cell and Development Biology (J.S., C.W.A., R.G., K.M.R., M.L.I.-A.), Northwestern University, Chicago
| | - Matteo Pellegrini
- Department of Molecular, Cell and Development Biology (M.P.), University of California, Los Angeles
| | - Robert Goldman
- Department of Cell and Development Biology (J.S., C.W.A., R.G., K.M.R., M.L.I.-A.), Northwestern University, Chicago
| | - Karen M Ridge
- Department of Cell and Development Biology (J.S., C.W.A., R.G., K.M.R., M.L.I.-A.), Northwestern University, Chicago.,Department of Medicine, Feinberg School of Medicine (K.M.R.), Northwestern University, Chicago
| | - M Luisa Iruela-Arispe
- Department of Cell and Development Biology (J.S., C.W.A., R.G., K.M.R., M.L.I.-A.), Northwestern University, Chicago
| |
Collapse
|
24
|
Photo-Crosslinked Hyaluronic Acid/Carboxymethyl Cellulose Composite Hydrogel as a Dural Substitute to Prevent Post-Surgical Adhesion. Int J Mol Sci 2022; 23:ijms23116177. [PMID: 35682853 PMCID: PMC9181059 DOI: 10.3390/ijms23116177] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Revised: 05/25/2022] [Accepted: 05/28/2022] [Indexed: 02/01/2023] Open
Abstract
A dural substitute is frequently used to repair dura mater during neurosurgical procedures. Although autologous or commercially available dural substitutes matched most of the requirements; difficulties during dural repair, including insufficient space for suturing, insufficient mechanical strength, easy tear and cerebrospinal fluid leakage, represent major challenges. To meet this need, a photo-crosslinked hydrogel was developed as a dural substitute/anti-adhesion barrier in this study, which can show sol-to-gel phase transition in situ upon short-time exposure to visible light. For this purpose, hyaluronic acid (HA) and carboxymethyl cellulose (CMC), materials used in abdominal surgery for anti-adhesion purposes, were reacted separately with glycidyl methacrylate to form hyaluronic acid methacrylate (HAMA) and carboxymethyl cellulose methacrylate (CMCMA). The HA/CMC (HC) hydrogels with different HA compositions could be prepared by photo-crosslinking HAMA and CMCMA with a 400 nm light source using lithium phenyl-2,4,6-trimethylbenzoylphosphinate as a photo-initiator. From studies of physico-chemical and biological properties of HC composite hydrogels, they are bio-compatible, bio-degradable and mechanically robust, to be suitable as a dural substitute. By drastically reducing attachment and penetration of adhesion-forming fibroblasts in vitro, the HC hydrogel can also act as an anti-adhesion barrier to prevent adhesion formation after dural repair. From in vivo study in rabbits, the HC hydrogel can repair dural defects as well as protect the dura from post-operative adhesion, endorsing the possible application of this hydrogel as a novel dural substitute.
Collapse
|
25
|
Miao H, Cui Y, Lu Y, Sun T, Dou J, Ren Y, Wang C, Zhang Y. Serum vimentin predicts mortality in pediatric severe sepsis: A prospective observational study. Int J Infect Dis 2022; 121:141-147. [PMID: 35568360 DOI: 10.1016/j.ijid.2022.05.018] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2022] [Revised: 05/06/2022] [Accepted: 05/06/2022] [Indexed: 11/17/2022] Open
Abstract
OBJECTIVES Vascular hyperpermeability by loss of endothelial barrier integrity is a hallmark of sepsis. Vimentin is involved in the regulation of the endothelial function and inflammatory response. However, the serum level of vimentin and its clinical relevance in pediatric severe sepsis (PSS) remain unknown. METHODS We conducted a prospective study of PSS cases who were admitted to the pediatric intensive care unit (PICU) from January 2018 to December 2020. RESULTS A total of 108 patients with PSS with a median age of 19.5 month were enrolled. The hospital mortality rate was 19.44% (21/108). Comparing with healthy controls, serum vimentin levels on PICU admission were significantly higher in patients with PSS (P < 0.001). The area under the ROC curve for vimentin to predict the hospital mortality was 0.712 (95% CI: 0.578-846) with a sensitivity of 71.43% and a specificity of 70.11%. Moreover, hospital mortality was significantly higher in patients with vimentin level over the cutoff value of 24.53 ng/ml than in patients with vimentin level below 24.53 ng/ml (P < 0.001). CONCLUSIONS Serum vimentin level as an indicator of endothelial injury is associated with the prognosis of PSS, and serum vimentin level ≥24.53 ng/ml on PICU admission predicts high risk for hospital mortality in PSS.
Collapse
Affiliation(s)
- Huijie Miao
- Department of Critical Care Medicine, Shanghai Children's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Institute of Pediatric Critical Care, Shanghai Jiao Tong University, Shanghai, China
| | - Yun Cui
- Department of Critical Care Medicine, Shanghai Children's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Institute of Pediatric Critical Care, Shanghai Jiao Tong University, Shanghai, China
| | - Ye Lu
- Department of Critical Care Medicine, Shanghai Children's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Institute of Pediatric Critical Care, Shanghai Jiao Tong University, Shanghai, China
| | - Ting Sun
- Department of Critical Care Medicine, Shanghai Children's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Institute of Pediatric Critical Care, Shanghai Jiao Tong University, Shanghai, China
| | - Jiaying Dou
- Department of Critical Care Medicine, Shanghai Children's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Institute of Pediatric Critical Care, Shanghai Jiao Tong University, Shanghai, China
| | - Yuqian Ren
- Department of Critical Care Medicine, Shanghai Children's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Institute of Pediatric Critical Care, Shanghai Jiao Tong University, Shanghai, China
| | - Chunxia Wang
- Department of Critical Care Medicine, Shanghai Children's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Institute of Pediatric Critical Care, Shanghai Jiao Tong University, Shanghai, China; Institute of Pediatric Infection, Immunity and Critical Care Medicine, Shanghai Children's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Clinical Research Unit, Shanghai Children's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Yucai Zhang
- Department of Critical Care Medicine, Shanghai Children's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Institute of Pediatric Critical Care, Shanghai Jiao Tong University, Shanghai, China; Institute of Pediatric Infection, Immunity and Critical Care Medicine, Shanghai Children's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
26
|
Herre C, Nshdejan A, Klopfleisch R, Corte GM, Bahramsoltani M. Expression of vimentin, TPI and MAT2A in human dermal microvascular endothelial cells during angiogenesis in vitro. PLoS One 2022; 17:e0266774. [PMID: 35482724 PMCID: PMC9049311 DOI: 10.1371/journal.pone.0266774] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Accepted: 03/27/2022] [Indexed: 11/19/2022] Open
Abstract
Introduction
In vitro assays of angiogenesis face immense problems considering their reproducibility based on the inhomogeneous characters of endothelial cells (ECs). It is necessary to detect influencing factors, which affect the angiogenic potency of ECs.
Objective
This study aimed to analyse expression profiles of vimentin (VIM), triosephosphate isomerase (TPI) and adenosylmethionine synthetase isoform type–2 (MAT2A) during the whole angiogenic cascade in vitro. Furthermore, the impact of knocking down vimentin (VIM) on angiogenesis in vitro was evaluated, while monitoring TPI and MAT2A expression.
Methods
A long–term cultivation and angiogenic stimulation of human dermal microvascular ECs was performed. Cells were characterized via VEGFR–1 and VEGFR–2 expression and a shRNA–mediated knockdown of VIM was performed. The process of angiogenesis in vitro was quantified via morphological staging and mRNA–and protein–levels of all proteins were analysed.
Results
While native cells ran through the angiogenic cascade chronologically, knockdown cells only entered beginning stages of angiogenesis and died eventually. Cell cultures showing a higher VEGFR–1 expression survived exclusively and displayed an upregulation of MAT2A and TPI expression. Native cells highly expressed VIM in early stages, MAT2A mainly in the beginning and TPI during the course of angiogenesis in vitro.
Conclusion
VIM knockdown led to a deceleration of angiogenesis in vitro and knockdown cells displayed expressional changes in TPI and MAT2A. Cell populations with a higher number of stalk cells emerged as being more stable against manipulations and native expression profiles provided an indication of VIM and MAT2A being relevant predominantly in beginning stages and TPI during the whole angiogenic cascade in vitro.
Collapse
Affiliation(s)
- Christina Herre
- Department of Veterinary Medicine, Institute of Veterinary Anatomy, Freie Universität Berlin, Berlin, Germany
- * E-mail:
| | - Arpenik Nshdejan
- Department of Veterinary Medicine, Institute of Veterinary Anatomy, Freie Universität Berlin, Berlin, Germany
| | - Robert Klopfleisch
- Department of Veterinary Medicine, Institute of Veterinary Pathology, Freie Universität Berlin, Berlin, Germany
| | - Giuliano Mario Corte
- Department of Veterinary Medicine, Institute of Veterinary Anatomy, Freie Universität Berlin, Berlin, Germany
| | - Mahtab Bahramsoltani
- Department of Veterinary Medicine, Institute of Veterinary Anatomy, Freie Universität Berlin, Berlin, Germany
| |
Collapse
|
27
|
Wang L, Mohanasundaram P, Lindström M, Asghar MN, Sultana G, Misiorek JO, Jiu Y, Chen H, Chen Z, Toivola DM, Cheng F, Eriksson JE. Vimentin Suppresses Inflammation and Tumorigenesis in the Mouse Intestine. Front Cell Dev Biol 2022; 10:862237. [PMID: 35399505 PMCID: PMC8993042 DOI: 10.3389/fcell.2022.862237] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Accepted: 02/22/2022] [Indexed: 01/03/2023] Open
Abstract
Vimentin has been implicated in wound healing, inflammation, and cancer, but its functional contribution to intestinal diseases is poorly understood. To study how vimentin is involved during tissue injury and repair of simple epithelium, we induced colonic epithelial cell damage in the vimentin null (Vim−/−) mouse model. Vim−/− mice challenged with dextran sodium sulfate (DSS) had worse colitis manifestations than wild-type (WT) mice. Vim−/− colons also produced more reactive oxygen and nitrogen species, possibly contributing to the pathogenesis of gut inflammation and tumorigenesis than in WT mice. We subsequently describe that CD11b+ macrophages served as the mainly cellular source of reactive oxygen species (ROS) production via vimentin-ROS-pSTAT3–interleukin-6 inflammatory pathways. Further, we demonstrated that Vim−/− mice did not develop colitis-associated cancer model upon DSS treatment spontaneously but increased tumor numbers and size in the distal colon in the azoxymethane/DSS model comparing with WT mice. Thus, vimentin has a crucial role in protection from colitis induction and tumorigenesis of the colon.
Collapse
Affiliation(s)
- Linglu Wang
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, China
| | - Ponnuswamy Mohanasundaram
- Cell Biology, Biosciences, Faculty of Science and Engineering, Åbo Akademi University, Turku, Finland
| | - Michelle Lindström
- Cell Biology, Biosciences, Faculty of Science and Engineering, Åbo Akademi University, Turku, Finland
| | - Muhammad Nadeem Asghar
- Cell Biology, Biosciences, Faculty of Science and Engineering, Åbo Akademi University, Turku, Finland
| | - Giulia Sultana
- Cell Biology, Biosciences, Faculty of Science and Engineering, Åbo Akademi University, Turku, Finland
| | - Julia O Misiorek
- Cell Biology, Biosciences, Faculty of Science and Engineering, Åbo Akademi University, Turku, Finland.,Department of Molecular Neurooncology, Institute of Bioorganic Chemistry Polish Academy of Sciences, Poznan, Poland
| | - Yaming Jiu
- Key Laboratory of Molecular Virology and Immunology, The Center for Microbes, Development and Health, Institut Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Hongbo Chen
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, China
| | - Zhi Chen
- Faculty of Biochemistry and Molecular Medicine, University of Oulu, Oulu, Finland
| | - Diana M Toivola
- Cell Biology, Biosciences, Faculty of Science and Engineering, Åbo Akademi University, Turku, Finland.,Turku Center for Disease Modeling, University of Turku, Turku, Finland.,InFLAMES Research Flagship Center, Åbo Akademi University, Turku, Finland
| | - Fang Cheng
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, China
| | - John E Eriksson
- Cell Biology, Biosciences, Faculty of Science and Engineering, Åbo Akademi University, Turku, Finland.,InFLAMES Research Flagship Center, Åbo Akademi University, Turku, Finland
| |
Collapse
|
28
|
Shakhov AS, Alieva IB. The "Third Violin" in the Cytoskeleton Orchestra-The Role of Intermediate Filaments in the Endothelial Cell's Life. Biomedicines 2022; 10:828. [PMID: 35453578 PMCID: PMC9027429 DOI: 10.3390/biomedicines10040828] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Revised: 03/29/2022] [Accepted: 03/30/2022] [Indexed: 02/01/2023] Open
Abstract
The endothelium plays an important role in the transcytosis of lipoproteins. According to one of the theories, endothelial injury is a triggering factor for the development of atherosclerosis, and intracellular structures, including components of the endotheliocyte cytoskeleton (microtubules, actin, and intermediate filaments), are involved in its development. In contrast to the proteins of tubulin-based microtubules and actin microfilaments, intermediate filaments are comprised of various tissue-specific protein members. Vimentin, the main protein of endothelial intermediate filaments, is one of the most well-studied of these and belongs to type-III intermediate filaments, commonly found in cells of mesenchymal origin. Vimentin filaments are linked mechanically or by signaling molecules to microfilaments and microtubules by which coordinated cell polarisation and migration are carried out, as well as control over several endotheliocyte functions. Moreover, the soluble vimentin acts as an indicator of the state of the cardiovascular system, and the involvement of vimentin in the development and course of atherosclerosis has been demonstrated. Here we discuss current concepts of the participation of vimentin filaments in the vital activity and functioning of endothelial cells, as well as the role of vimentin in the development of inflammatory processes and atherosclerosis.
Collapse
Affiliation(s)
| | - Irina B. Alieva
- A.N. Belozersky Institute of Physical and Chemical Biology, Lomonosov Moscow State University, 119992 Moscow, Russia;
| |
Collapse
|
29
|
Abstract
More than 27 yr ago, the vimentin knockout (Vim-/- ) mouse was reported to develop and reproduce without an obvious phenotype, implying that this major cytoskeletal protein was nonessential. Subsequently, comprehensive and careful analyses have revealed numerous phenotypes in Vim-/- mice and their organs, tissues, and cells, frequently reflecting altered responses in the recovery of tissues following various insults or injuries. These findings have been supported by cell-based experiments demonstrating that vimentin intermediate filaments (IFs) play a critical role in regulating cell mechanics and are required to coordinate mechanosensing, transduction, signaling pathways, motility, and inflammatory responses. This review highlights the essential functions of vimentin IFs revealed from studies of Vim-/- mice and cells derived from them.
Collapse
Affiliation(s)
- Karen M Ridge
- Division of Pulmonary and Critical Care Medicine, Northwestern University, Chicago, Illinois 60611, USA
- Department of Cell and Developmental Biology, Northwestern University, Chicago, Illinois 60611, USA
| | - John E Eriksson
- Cell Biology, Faculty of Science and Technology, Åbo Akademi University, FIN-20521 Turku, Finland
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, FIN-20521 Turku, Finland
- Euro-Bioimaging European Research Infrastructure Consortium (ERIC), FIN-20521 Turku, Finland
| | - Milos Pekny
- Laboratory of Astrocyte Biology and CNS Regeneration, Center for Brain Repair, Department of Clinical Neuroscience, Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg, 413 90 Gothenburg, Sweden
- Florey Institute of Neuroscience and Mental Health, Parkville, Victoria 3052, Australia
- University of Newcastle, Newcastle, New South Wales 2300, Australia
| | - Robert D Goldman
- Division of Pulmonary and Critical Care Medicine, Northwestern University, Chicago, Illinois 60611, USA
- Department of Cell and Developmental Biology, Northwestern University, Chicago, Illinois 60611, USA
| |
Collapse
|
30
|
Amraei R, Xia C, Olejnik J, White MR, Napoleon MA, Lotfollahzadeh S, Hauser BM, Schmidt AG, Chitalia V, Mühlberger E, Costello CE, Rahimi N. Extracellular vimentin is an attachment factor that facilitates SARS-CoV-2 entry into human endothelial cells. Proc Natl Acad Sci U S A 2022; 119:2113874119. [PMID: 35078919 PMCID: PMC8833221 DOI: 10.1073/pnas.2113874119] [Citation(s) in RCA: 66] [Impact Index Per Article: 33.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/18/2021] [Indexed: 02/07/2023] Open
Abstract
SARS-CoV-2 entry into host cells is a crucial step for virus tropism, transmission, and pathogenesis. Angiotensin-converting enzyme 2 (ACE2) has been identified as the primary entry receptor for SARS-CoV-2; however, the possible involvement of other cellular components in the viral entry has not yet been fully elucidated. Here we describe the identification of vimentin (VIM), an intermediate filament protein widely expressed in cells of mesenchymal origin, as an important attachment factor for SARS-CoV-2 on human endothelial cells. Using liquid chromatography-tandem mass spectrometry, we identified VIM as a protein that binds to the SARS-CoV-2 spike (S) protein. We showed that the S-protein receptor binding domain (RBD) is sufficient for S-protein interaction with VIM. Further analysis revealed that extracellular VIM binds to SARS-CoV-2 S-protein and facilitates SARS-CoV-2 infection, as determined by entry assays performed with pseudotyped viruses expressing S and with infectious SARS-CoV-2. Coexpression of VIM with ACE2 increased SARS-CoV-2 entry in HEK-293 cells, and shRNA-mediated knockdown of VIM significantly reduced SARS-CoV-2 infection of human endothelial cells. Moreover, incubation of A549 cells expressing ACE2 with purified VIM increased pseudotyped SARS-CoV-2-S entry. CR3022 antibody, which recognizes a distinct epitope on SARS-CoV-2-S-RBD without interfering with the binding of the spike with ACE2, inhibited the binding of VIM with CoV-2 S-RBD, and neutralized viral entry in human endothelial cells, suggesting a key role for VIM in SARS-CoV-2 infection of endothelial cells. This work provides insight into the pathogenesis of COVID-19 linked to the vascular system, with implications for the development of therapeutics and vaccines.
Collapse
Affiliation(s)
- Razie Amraei
- Department of Pathology, Boston University School of Medicine, Boston, MA 02118
| | - Chaoshuang Xia
- Center for Biomedical Mass Spectrometry, Boston University School of Medicine, Boston, MA 02118
| | - Judith Olejnik
- Department of Microbiology, Boston University School of Medicine, Boston, MA 02118
- National Emerging Infectious Diseases Laboratories, Boston University, Boston, MA 02118
| | - Mitchell R White
- Department of Microbiology, Boston University School of Medicine, Boston, MA 02118
- National Emerging Infectious Diseases Laboratories, Boston University, Boston, MA 02118
| | - Marc A Napoleon
- Renal Section, Department of Medicine, Boston University Medical Center, Boston, MA 02118
| | - Saran Lotfollahzadeh
- Renal Section, Department of Medicine, Boston University Medical Center, Boston, MA 02118
| | - Blake M Hauser
- Ragon Institute of Massachusetts General Hospital (MGH), Massachusetts Institute of Technology and Harvard University, Cambridge, MA 02139
- Department of Microbiology, Harvard Medical School, Boston, MA 02115
| | - Aaron G Schmidt
- Ragon Institute of Massachusetts General Hospital (MGH), Massachusetts Institute of Technology and Harvard University, Cambridge, MA 02139
- Department of Microbiology, Harvard Medical School, Boston, MA 02115
| | - Vipul Chitalia
- Renal Section, Department of Medicine, Boston University Medical Center, Boston, MA 02118
- Veterans Affairs Boston Healthcare System, Boston, MA 02118
- Institute of Medical Engineering and Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139
| | - Elke Mühlberger
- Department of Microbiology, Boston University School of Medicine, Boston, MA 02118
- National Emerging Infectious Diseases Laboratories, Boston University, Boston, MA 02118
| | - Catherine E Costello
- Center for Biomedical Mass Spectrometry, Boston University School of Medicine, Boston, MA 02118;
| | - Nader Rahimi
- Department of Pathology, Boston University School of Medicine, Boston, MA 02118;
| |
Collapse
|
31
|
Hammer L, Levin‐Salomon V, Yaeli‐Slonim N, Weiss M, Dekel‐Bird NP, Olender T, Porat Z, Winograd‐Katz S, Savidor A, Levin Y, Bialik S, Geiger B, Kimchi A. A new function for the serine protease HtrA2 in controlling radiation‐induced senescence in cancer cells. Mol Oncol 2022; 16:1365-1383. [PMID: 35122388 PMCID: PMC8936513 DOI: 10.1002/1878-0261.13187] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Revised: 12/08/2021] [Accepted: 02/03/2022] [Indexed: 11/09/2022] Open
Affiliation(s)
- Liat Hammer
- Dept. Molecular Genetics Weizmann Institute of Science Rehovot 7610001 Israel
| | - Vered Levin‐Salomon
- Dept. Molecular Genetics Weizmann Institute of Science Rehovot 7610001 Israel
| | - Naama Yaeli‐Slonim
- Dept. Molecular Genetics Weizmann Institute of Science Rehovot 7610001 Israel
| | - Moria Weiss
- Dept. Molecular Genetics Weizmann Institute of Science Rehovot 7610001 Israel
| | - Naama P. Dekel‐Bird
- Dept. Molecular Genetics Weizmann Institute of Science Rehovot 7610001 Israel
| | - Tsviya Olender
- Dept. Molecular Genetics Weizmann Institute of Science Rehovot 7610001 Israel
| | - Ziv Porat
- Dept. Life Sciences Core Facilities Weizmann Institute of Science Rehovot 7610001 Israel
| | | | - Alon Savidor
- The Nancy and Stephen Grand Israel National Center for Personalized Medicine (G‐INCPM) Weizmann Institute of Science Rehovot 7610001 Israel
| | - Yishai Levin
- The Nancy and Stephen Grand Israel National Center for Personalized Medicine (G‐INCPM) Weizmann Institute of Science Rehovot 7610001 Israel
| | - Shani Bialik
- Dept. Molecular Genetics Weizmann Institute of Science Rehovot 7610001 Israel
| | - Benjamin Geiger
- Dept. Immunology Weizmann Institute of Science Rehovot 7610001 Israel
| | - Adi Kimchi
- Dept. Molecular Genetics Weizmann Institute of Science Rehovot 7610001 Israel
| |
Collapse
|
32
|
de Almeida GP, Lichtner P, Eckstein G, Brinkschmidt T, Chu CF, Sun S, Reinhard J, Mädler SC, Kloeppel M, Verbeek M, Zielinski CE. Human skin-resident host T cells can persist long term after allogeneic stem cell transplantation and maintain recirculation potential. Sci Immunol 2022; 7:eabe2634. [PMID: 35089814 DOI: 10.1126/sciimmunol.abe2634] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Tissue-resident memory T cells (TRM) have recently emerged as crucial cellular players for host defense in a wide variety of tissues and barrier sites. Insights into the maintenance and regulatory checkpoints of human TRM cells remain scarce, especially due to the difficulties associated with tracking T cells through time and space in humans. We therefore sought to identify and characterize skin-resident T cells in humans defined by their long-term in situ lodgment. Allogeneic hematopoietic stem cell transplantation (allo-HSCT) preceded by myeloablative chemotherapy unmasked long-term sequestration of host T cell subsets in human skin despite complete donor T cell chimerism in the blood. Single-cell chimerism analysis paired with single-cell transcriptional profiling comprehensively characterized these bona fide long-term skin-resident T cells and revealed differential tissue maintenance for distinct T cell subsets, specific TRM cell markers such as galectin-3, but also tissue exit potential with retention of the transcriptomic TRM cell identity. Analysis of 26 allo-HSCT patients revealed profound interindividual variation in the tissue maintenance of host skin T cells. The long-term persistence of host skin T cells in a subset of these patients did not correlate with the development of chronic GvHD. Our data exemplify the power of exploiting a clinical situation as a proof of concept for the existence of bona fide human skin TRM cells and reveal long-term persistence of host T cells in a peripheral tissue but not in the circulation or bone marrow in a subset of allo-HSCT patients.
Collapse
Affiliation(s)
- Gustavo P de Almeida
- Institute of Virology, Technical University of Munich, Munich, Germany.,TranslaTUM, Technical University of Munich, Munich, Germany.,German Center for Infection Research partner site, Munich, Germany
| | - Peter Lichtner
- Genome Analysis Center, Helmholtz Zentrum Munich, Munich, Germany
| | - Gertrud Eckstein
- Genome Analysis Center, Helmholtz Zentrum Munich, Munich, Germany
| | - Tonio Brinkschmidt
- Institute of Virology, Technical University of Munich, Munich, Germany.,TranslaTUM, Technical University of Munich, Munich, Germany.,Department of Infection Immunology, Leibniz-Institute for Natural Product Research and Infection Biology, Jena, Germany
| | - Chang-Feng Chu
- TranslaTUM, Technical University of Munich, Munich, Germany.,Department of Infection Immunology, Leibniz-Institute for Natural Product Research and Infection Biology, Jena, Germany.,Institute of Microbiology, Faculty of Biological Sciences, Friedrich Schiller University, Jena, Germany
| | - Shan Sun
- Institute of Virology, Technical University of Munich, Munich, Germany.,TranslaTUM, Technical University of Munich, Munich, Germany.,German Center for Infection Research partner site, Munich, Germany.,Department of Infection Immunology, Leibniz-Institute for Natural Product Research and Infection Biology, Jena, Germany
| | | | | | - Markus Kloeppel
- Klinikum rechts der Isar and Praxisklinik für Ästhetische Chirurgie und Medizin, Munich, Germany
| | - Mareike Verbeek
- Department of Medicine III, Klinikum rechts der Isar, Munich, Germany
| | - Christina E Zielinski
- Institute of Virology, Technical University of Munich, Munich, Germany.,TranslaTUM, Technical University of Munich, Munich, Germany.,German Center for Infection Research partner site, Munich, Germany.,Department of Infection Immunology, Leibniz-Institute for Natural Product Research and Infection Biology, Jena, Germany.,Institute of Microbiology, Faculty of Biological Sciences, Friedrich Schiller University, Jena, Germany
| |
Collapse
|
33
|
Liang Y, Li L, Chen Y, Zhang S, Li Z, Xiao J, Wei D. Research Progress on the Role of Intermediate Filament Vimentin in Atherosclerosis. DNA Cell Biol 2021; 40:1495-1502. [PMID: 34931866 DOI: 10.1089/dna.2021.0623] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
The cytoskeleton is a biopolymer network composed of intermediate filaments, actin, and microtubules, which is the main mechanical structure of cells. Vimentin is an intermediate filament protein that regulates the mechanical and contractile properties of cells, thereby reflecting their mechanical properties. In recent years, the "nonmechanical function" of vimentin inside and outside of cells has attracted extensive attention. The content of vimentin in atherosclerotic plaques is increased, and the serum secretion of vimentin in patients with coronary heart disease is remarkably increased. In this review, the mechanistic and nonmechanistic roles of vimentin in atherosclerosis progression were summarized on the basis of current studies.
Collapse
Affiliation(s)
- Yamin Liang
- Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Institute of Cardiovascular Disease, Hengyang Medical College, University of South China, Hengyang, Hunan, China
| | - Lu Li
- Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Institute of Cardiovascular Disease, Hengyang Medical College, University of South China, Hengyang, Hunan, China
| | - Yanmei Chen
- Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Institute of Cardiovascular Disease, Hengyang Medical College, University of South China, Hengyang, Hunan, China
| | - Shulei Zhang
- Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Institute of Cardiovascular Disease, Hengyang Medical College, University of South China, Hengyang, Hunan, China
| | - Zhaozhi Li
- Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Institute of Cardiovascular Disease, Hengyang Medical College, University of South China, Hengyang, Hunan, China
| | - Jinyan Xiao
- YueYang Maternal-Child Medicine Health Hospital Hunan Province Innovative Training Base for Medical Postgraduates, University of China South China and Yueyang Women and Children's Medical Center, Yueyang, Hunan, China
| | - Dangheng Wei
- Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Institute of Cardiovascular Disease, Hengyang Medical College, University of South China, Hengyang, Hunan, China
| |
Collapse
|
34
|
Vimentin Regulates Chemokine Expression and NOD2 Activation in Brain Endothelium during Group B Streptococcal Infection. Infect Immun 2021; 89:e0034021. [PMID: 34491787 DOI: 10.1128/iai.00340-21] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Streptococcus agalactiae (group B Streptococcus, or GBS) is an opportunistic pathogen capable of causing invasive disease in susceptible individuals, including the newborn. Currently, GBS is the leading cause of meningitis in the neonatal period. We have recently shown that GBS interacts directly with host type III intermediate filament vimentin to gain access to the central nervous system. This results in characteristic meningeal inflammation and disease progression; however, the specific role of vimentin in the inflammatory process is unknown. Here, we investigate the contribution of vimentin to the pathogenesis of GBS meningitis. We show that a CRISPR-targeted deletion of vimentin in human cerebral microvascular endothelial cells (hCMEC) reduced GBS induction of neutrophil attractants interleukin-8 (IL-8) and CXCL-1 as well as NF-κB activation. We further show that inhibition of vimentin localization also prevented similar chemokine activation by GBS. One known chemokine regulator is the nucleotide-binding oligomerization domain containing protein 2 (NOD2), which is known to interact directly with vimentin. Thus, we hypothesized that NOD2 would also promote GBS chemokine induction. We show that GBS infection induced NOD2 transcription in hCMEC comparably to the muramyl dipeptide (MDP) NOD2 agonist, and the chemokine induction was reduced in the presence of a NOD2 inhibitor. Using a mouse model of GBS meningitis, we also observed increased NOD2 transcript and NOD2 activation in brain tissue of infected mice. Lastly, we show that NOD2-mediated IL-8 and CXCL1 induction required vimentin, further indicating the importance of vimentin in mediating inflammatory responses in brain endothelium.
Collapse
|
35
|
Romero-Araya P, Pino V, Nenen A, Cárdenas V, Pavicic F, Ehrenfeld P, Serandour G, Lisoni JG, Moreno-Villoslada I, Flores ME. Combining Materials Obtained by 3D-Printing and Electrospinning from Commercial Polylactide Filament to Produce Biocompatible Composites. Polymers (Basel) 2021; 13:polym13213806. [PMID: 34771361 PMCID: PMC8588263 DOI: 10.3390/polym13213806] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Revised: 10/29/2021] [Accepted: 11/01/2021] [Indexed: 01/29/2023] Open
Abstract
The design of scaffolds to reach similar three-dimensional structures mimicking the natural and fibrous environment of some cells is a challenge for tissue engineering, and 3D-printing and electrospinning highlights from other techniques in the production of scaffolds. The former is a well-known additive manufacturing technique devoted to the production of custom-made structures with mechanical properties similar to tissues and bones found in the human body, but lacks the resolution to produce small and interconnected structures. The latter is a well-studied technique to produce materials possessing a fibrillar structure, having the advantage of producing materials with tuned composition compared with a 3D-print. Taking the advantage that commercial 3D-printers work with polylactide (PLA) based filaments, a biocompatible and biodegradable polymer, in this work we produce PLA-based composites by blending materials obtained by 3D-printing and electrospinning. Porous PLA fibers have been obtained by the electrospinning of recovered PLA from 3D-printer filaments, tuning the mechanical properties by blending PLA with small amounts of polyethylene glycol and hydroxyapatite. A composite has been obtained by blending two layers of 3D-printed pieces with a central mat of PLA fibers. The composite presented a reduced storage modulus as compared with a single 3D-print piece and possessing similar mechanical properties to bone tissues. Furthermore, the biocompatibility of the composites is assessed by a simulated body fluid assay and by culturing composites with 3T3 fibroblasts. We observed that all these composites induce the growing and attaching of fibroblast over the surface of a 3D-printed layer and in the fibrous layer, showing the potential of commercial 3D-printers and filaments to produce scaffolds to be used in bone tissue engineering.
Collapse
Affiliation(s)
- Pablo Romero-Araya
- Laboratorio de Polímeros, Facultad de Ciencias, Instituto de Ciencias Químicas, Universidad Austral de Chile, Valdivia 5090000, Chile; (P.R.-A.); (V.P.); (A.N.); (V.C.); (I.M.-V.)
- Escuela de Odontología, Facultad de Medicina, Universidad Austral de Chile, Valdivia 5090000, Chile
| | - Victor Pino
- Laboratorio de Polímeros, Facultad de Ciencias, Instituto de Ciencias Químicas, Universidad Austral de Chile, Valdivia 5090000, Chile; (P.R.-A.); (V.P.); (A.N.); (V.C.); (I.M.-V.)
- Escuela de Odontología, Facultad de Medicina, Universidad Austral de Chile, Valdivia 5090000, Chile
| | - Ariel Nenen
- Laboratorio de Polímeros, Facultad de Ciencias, Instituto de Ciencias Químicas, Universidad Austral de Chile, Valdivia 5090000, Chile; (P.R.-A.); (V.P.); (A.N.); (V.C.); (I.M.-V.)
| | - Verena Cárdenas
- Laboratorio de Polímeros, Facultad de Ciencias, Instituto de Ciencias Químicas, Universidad Austral de Chile, Valdivia 5090000, Chile; (P.R.-A.); (V.P.); (A.N.); (V.C.); (I.M.-V.)
| | - Francisca Pavicic
- Facultad de Medicina, Instituto de Anatomia, Histologia y Patologia, Universidad Austral de Chile, Valdivia 5090000, Chile; (F.P.); (P.E.)
- Centro de Estudios Interdisciplinarios del Sistema Nervioso (CISNe), Universidad Austral de Chile, Valdivia 5090000, Chile
| | - Pamela Ehrenfeld
- Facultad de Medicina, Instituto de Anatomia, Histologia y Patologia, Universidad Austral de Chile, Valdivia 5090000, Chile; (F.P.); (P.E.)
- Centro de Estudios Interdisciplinarios del Sistema Nervioso (CISNe), Universidad Austral de Chile, Valdivia 5090000, Chile
| | - Guillaume Serandour
- LeufüLAB, Facultad de Ciencias de la Ingeniería, Instituto de Diseño y Métodos Industriales, Universidad Austral de Chile, Valdivia 5090000, Chile;
| | - Judit G. Lisoni
- Facultad de Ciencias, Instituto de Ciencias Físicas y Matemáticas, Universidad Austral de Chile, Valdivia 5090000, Chile;
| | - Ignacio Moreno-Villoslada
- Laboratorio de Polímeros, Facultad de Ciencias, Instituto de Ciencias Químicas, Universidad Austral de Chile, Valdivia 5090000, Chile; (P.R.-A.); (V.P.); (A.N.); (V.C.); (I.M.-V.)
| | - Mario E. Flores
- Laboratorio de Polímeros, Facultad de Ciencias, Instituto de Ciencias Químicas, Universidad Austral de Chile, Valdivia 5090000, Chile; (P.R.-A.); (V.P.); (A.N.); (V.C.); (I.M.-V.)
- Correspondence: ; Tel.: +56-63-2293521
| |
Collapse
|
36
|
Xiao J, Zhang B, Su Z, Liu Y, Shelite TR, Chang Q, Qiu Y, Bei J, Wang P, Bukreyev A, Soong L, Jin Y, Ksiazek T, Gaitas A, Rossi SL, Zhou J, Laposata M, Saito TB, Gong B. Intracellular receptor EPAC regulates von Willebrand factor secretion from endothelial cells in a PI3K-/eNOS-dependent manner during inflammation. J Biol Chem 2021; 297:101315. [PMID: 34678311 PMCID: PMC8526113 DOI: 10.1016/j.jbc.2021.101315] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 10/14/2021] [Accepted: 10/18/2021] [Indexed: 02/06/2023] Open
Abstract
Coagulopathy is associated with both inflammation and infection, including infections with novel severe acute respiratory syndrome coronavirus-2, the causative agent Coagulopathy is associated with both inflammation and infection, including infection with novel severe acute respiratory syndrome coronavirus-2, the causative agent of COVID-19. Clot formation is promoted via cAMP-mediated secretion of von Willebrand factor (vWF), which fine-tunes the process of hemostasis. The exchange protein directly activated by cAMP (EPAC) is a ubiquitously expressed intracellular cAMP receptor that plays a regulatory role in suppressing inflammation. To assess whether EPAC could regulate vWF release during inflammation, we utilized our EPAC1-null mouse model and revealed increased secretion of vWF in endotoxemic mice in the absence of the EPAC1 gene. Pharmacological inhibition of EPAC1 in vitro mimicked the EPAC1-/- phenotype. In addition, EPAC1 regulated tumor necrosis factor-α-triggered vWF secretion from human umbilical vein endothelial cells in a manner dependent upon inflammatory effector molecules PI3K and endothelial nitric oxide synthase. Furthermore, EPAC1 activation reduced inflammation-triggered vWF release, both in vivo and in vitro. Our data delineate a novel regulatory role for EPAC1 in vWF secretion and shed light on the potential development of new strategies to control thrombosis during inflammation.
Collapse
Affiliation(s)
- Jie Xiao
- Department of Pathology, University of Texas Medical Branch, Galveston, Texas, USA
| | - Ben Zhang
- Department of Pathology, University of Texas Medical Branch, Galveston, Texas, USA
| | - Zhengchen Su
- Department of Pathology, University of Texas Medical Branch, Galveston, Texas, USA
| | - Yakun Liu
- Department of Pathology, University of Texas Medical Branch, Galveston, Texas, USA
| | - Thomas R Shelite
- Department of Internal Medicine, Infectious Diseases, University of Texas Medical Branch, Galveston, Texas, USA
| | - Qing Chang
- Department of Pathology, University of Texas Medical Branch, Galveston, Texas, USA
| | - Yuan Qiu
- Department of Pathology, University of Texas Medical Branch, Galveston, Texas, USA
| | - Jiani Bei
- Department of Pathology, University of Texas Medical Branch, Galveston, Texas, USA
| | - Pingyuan Wang
- Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, Texas, USA
| | - Alexander Bukreyev
- Department of Pathology, University of Texas Medical Branch, Galveston, Texas, USA
| | - Lynn Soong
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas, USA
| | - Yang Jin
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Boston University Medical Campus, Boston, Massachusetts, USA
| | - Thomas Ksiazek
- Department of Pathology, University of Texas Medical Branch, Galveston, Texas, USA
| | - Angelo Gaitas
- The Estelle and Daniel Maggin Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Shannan L Rossi
- Department of Pathology, University of Texas Medical Branch, Galveston, Texas, USA
| | - Jia Zhou
- Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, Texas, USA
| | - Michael Laposata
- Department of Pathology, University of Texas Medical Branch, Galveston, Texas, USA
| | - Tais B Saito
- Department of Pathology, University of Texas Medical Branch, Galveston, Texas, USA
| | - Bin Gong
- Department of Pathology, University of Texas Medical Branch, Galveston, Texas, USA.
| |
Collapse
|
37
|
Shin SU, Cho HM, Das R, Gil-Henn H, Ramakrishnan S, Al Bayati A, Carroll SF, Zhang Y, Sankar AP, Elledge C, Pimentel A, Blonska M, Rosenblatt JD. Inhibition of Vasculogenic Mimicry and Angiogenesis by an Anti-EGFR IgG1-Human Endostatin-P125A Fusion Protein Reduces Triple Negative Breast Cancer Metastases. Cells 2021; 10:cells10112904. [PMID: 34831127 PMCID: PMC8616280 DOI: 10.3390/cells10112904] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Revised: 10/08/2021] [Accepted: 10/19/2021] [Indexed: 12/16/2022] Open
Abstract
Triple negative breast cancer (TNBC) is an aggressive breast cancer subtype with limited therapeutic options. Metastasis is the major cause of TNBC mortality. Angiogenesis facilitates TNBC metastases. Many TNBCs also form vascular channels lined by tumor cells rather than endothelial cells, known as ‘vasculogenic mimicry’ (VM). VM has been linked to metastatic TNBC behavior and resistance to anti-angiogenic agents. Epidermal growth factor receptor (EGFR) is frequently expressed on TNBC, but anti-EGFR antibodies have limited efficacy. We synthesized an anti-EGFR antibody–endostatin fusion protein, αEGFR IgG1-huEndo-P125A (αEGFR-E-P125A), designed to deliver a mutant endostatin, huEndo-P125A (E-P125A), to EGFR expressing tumors, and tested its effects on angiogenesis, TNBC VM, and motility in vitro, and on the growth and metastasis of two independent human TNBC xenograft models in vivo. αEGFR-E-P125A completely inhibited the ability of human umbilical vein endothelial cells to form capillary-like structures (CLS) and of TNBC cells to engage in VM and form tubes in vitro. αEGFR-E-P125A treatment reduced endothelial and TNBC motility in vitro more effectively than E-P125A or cetuximab, delivered alone or in combination. Treatment of TNBC with αEGFR-E-P125A was associated with a reduction in cytoplasmic and nuclear β-catenin and reduced phosphorylation of vimentin. αEGFR-E-P125A treatment of TNBC xenografts in vivo inhibited angiogenesis and VM, reduced primary tumor growth and lung metastasis of orthotopically implanted MDA-MB-468 TNBC cells, and markedly decreased lung metastases following intravenous injection of MDA-MB-231-4175 lung-tropic TNBC cells. Combined inhibition of angiogenesis, VM, and TNBC motility mediated by αEGFR-E-P125A is a promising strategy for the prevention of TNBC metastases.
Collapse
Affiliation(s)
- Seung-Uon Shin
- Sylvester Comprehensive Cancer Center, Department of Medicine, Division of Hematology, University of Miami Miller School of Medicine (UMMSOM), Miami, FL 33136, USA; (S.-U.S.); (H.-M.C.); (A.A.B.); (Y.Z.); (M.B.)
| | - Hyun-Mi Cho
- Sylvester Comprehensive Cancer Center, Department of Medicine, Division of Hematology, University of Miami Miller School of Medicine (UMMSOM), Miami, FL 33136, USA; (S.-U.S.); (H.-M.C.); (A.A.B.); (Y.Z.); (M.B.)
| | - Rathin Das
- Synergys Biotherapeutics Inc., Alamo, CA 94507, USA; (R.D.); (S.F.C.)
| | - Hava Gil-Henn
- The Azrieli Faculty of Medicine, Bar-Ilan University, Safed 1311502, Israel;
| | - Sundaram Ramakrishnan
- Department of Surgery, University of Miami Miller School of Medicine (UMMSOM), Miami, FL 33136, USA;
| | - Ahmed Al Bayati
- Sylvester Comprehensive Cancer Center, Department of Medicine, Division of Hematology, University of Miami Miller School of Medicine (UMMSOM), Miami, FL 33136, USA; (S.-U.S.); (H.-M.C.); (A.A.B.); (Y.Z.); (M.B.)
- Kentucky Clinic, University of Kentucky, Lexington, KY 40536, USA
| | | | - Yu Zhang
- Sylvester Comprehensive Cancer Center, Department of Medicine, Division of Hematology, University of Miami Miller School of Medicine (UMMSOM), Miami, FL 33136, USA; (S.-U.S.); (H.-M.C.); (A.A.B.); (Y.Z.); (M.B.)
| | - Ankita P. Sankar
- Sheila and David Fuente Graduate Program in Cancer Biology, Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine (UMMSOM), Miami, FL 33136, USA; (A.P.S.); (C.E.)
| | - Christian Elledge
- Sheila and David Fuente Graduate Program in Cancer Biology, Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine (UMMSOM), Miami, FL 33136, USA; (A.P.S.); (C.E.)
| | - Augustin Pimentel
- Sylvester Comprehensive Cancer Center, Department of Medicine, Division of Medical Oncology, University of Miami Miller School of Medicine (UMMSOM), Miami, FL 33136, USA;
| | - Marzenna Blonska
- Sylvester Comprehensive Cancer Center, Department of Medicine, Division of Hematology, University of Miami Miller School of Medicine (UMMSOM), Miami, FL 33136, USA; (S.-U.S.); (H.-M.C.); (A.A.B.); (Y.Z.); (M.B.)
| | - Joseph D. Rosenblatt
- Sylvester Comprehensive Cancer Center, Department of Medicine, Division of Hematology, University of Miami Miller School of Medicine (UMMSOM), Miami, FL 33136, USA; (S.-U.S.); (H.-M.C.); (A.A.B.); (Y.Z.); (M.B.)
- Correspondence: ; Tel.: +1-305-243-4618; Fax: +1-305-243-9161
| |
Collapse
|
38
|
Kant RJ, Bare CF, Coulombe KL. Tissues with Patterned Vessels or Protein Release Induce Vascular Chemotaxis in an In Vitro Platform. Tissue Eng Part A 2021; 27:1290-1304. [PMID: 33472529 PMCID: PMC8610033 DOI: 10.1089/ten.tea.2020.0269] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Accepted: 01/11/2021] [Indexed: 12/14/2022] Open
Abstract
Engineered tissues designed for translational applications in regenerative medicine require vascular networks to deliver oxygen and nutrients rapidly to the implanted cells. A limiting factor of in vivo translation is the rapid and successful inosculation, or connection, of host and implanted vascular networks and subsequent perfusion of the implant. An approach gaining favor in vascular tissue engineering is to provide instructive cues from the engineered tissue to enhance host vascular penetration and connection with the implant. Here, we use a novel in vitro platform based on the aortic ring assay to evaluate the impact of patterned, endothelialized vessels or growth factor release from engineered constructs on preinosculative vascular cell outgrowth from surrogate host tissue in a controlled, defined environment, and introduce robust tools for evaluating vascular morphogenesis and chemotaxis. We demonstrate the creation of engineered vessels at the arteriole scale, which develop basement membrane, exhibit tight junctions, and actively sprout into the surrounding bulk hydrogel. Vessel-containing constructs are co-cultured adjacent to rodent aortic rings, and the resulting heterocellular outgrowth is quantified. Cells originating from the aortic ring migrate preferentially toward constructs containing engineered vessels with 1.5-fold faster outgrowth kinetics, 2.5-fold increased cellular density, and 1.6-fold greater network formation versus control (no endothelial cells and growth factor-reduced culture medium). Growth factor release from constructs with nonendothelialized channels and in reduced factor medium equivalently stimulates sustained vascular outgrowth distance, cellular density, and network formation, akin to engineered vessels in endothelial growth medium 2 (EGM-2) medium. In conclusion, we show that three-dimensional endothelialized patterned vessels or growth factor release stimulate a robust, host-derived vascular cell chemotactic response at early time points critical for instructive angiogenic cues. Further, we developed robust, unbiased tools to quantify metrics of vascular morphogenesis and preinosculative heterocellular outgrowth from rat aortic rings and demonstrated the utility of our complex, controlled environment, heterocellular in vitro platform. Impact statement Using a novel in vitro platform, we show that engineered constructs with patterned vessels or angiogenic growth factor release, two methods of instructing host revascularization responses, equivalently improve early host-derived vascular outgrowth. Our platform leverages the aortic ring assay in a tissue engineering context to study preinosculative vascular cell chemotaxis from surrogate host vascular cells in response to paracrine cues from co-cultured engineered tissues using robust, open-source quantification tools. Our accessible and flexible platform enables translationally focused studies in revascularization using implantable therapeutics containing prepatterned vessels with greater environmental control than in vivo studies to advance vascular tissue engineering.
Collapse
Affiliation(s)
- Rajeev J. Kant
- Center for Biomedical Engineering, School of Engineering, Brown University, Providence, Rhode Island, USA
| | - Colette F. Bare
- Center for Biomedical Engineering, School of Engineering, Brown University, Providence, Rhode Island, USA
| | - Kareen L.K. Coulombe
- Center for Biomedical Engineering, School of Engineering, Brown University, Providence, Rhode Island, USA
| |
Collapse
|
39
|
Lin PK, Salvador J, Xie J, Aguera KN, Koller GM, Kemp SS, Griffin CT, Davis GE. Selective and Marked Blockade of Endothelial Sprouting Behavior Using Paclitaxel and Related Pharmacologic Agents. THE AMERICAN JOURNAL OF PATHOLOGY 2021; 191:2245-2264. [PMID: 34563512 DOI: 10.1016/j.ajpath.2021.08.017] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Revised: 08/10/2021] [Accepted: 08/26/2021] [Indexed: 12/11/2022]
Abstract
Whether alterations in the microtubule cytoskeleton affect the ability of endothelial cells (ECs) to sprout and form branching networks of tubes was investigated in this study. Bioassays of human EC tubulogenesis, where both sprouting behavior and lumen formation can be rigorously evaluated, were used to demonstrate that addition of the microtubule-stabilizing drugs, paclitaxel, docetaxel, ixabepilone, and epothilone B, completely interferes with EC tip cells and sprouting behavior, while allowing for EC lumen formation. In bioassays mimicking vasculogenesis using single or aggregated ECs, these drugs induce ring-like lumens from single cells or cyst-like spherical lumens from multicellular aggregates with no evidence of EC sprouting behavior. Remarkably, treatment of these cultures with a low dose of the microtubule-destabilizing drug, vinblastine, led to an identical result, with complete blockade of EC sprouting, but allowing for EC lumen formation. Administration of paclitaxel in vivo markedly interfered with angiogenic sprouting behavior in developing mouse retina, providing corroboration. These findings reveal novel biological activities for pharmacologic agents that are widely utilized in multidrug chemotherapeutic regimens for the treatment of human malignant cancers. Overall, this work demonstrates that manipulation of microtubule stability selectively interferes with the ability of ECs to sprout, a necessary step to initiate and form branched capillary tube networks.
Collapse
Affiliation(s)
- Prisca K Lin
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida School of Medicine, Tampa, Florida
| | - Jocelynda Salvador
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida School of Medicine, Tampa, Florida
| | - Jun Xie
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - Kalia N Aguera
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida School of Medicine, Tampa, Florida
| | - Gretchen M Koller
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida School of Medicine, Tampa, Florida
| | - Scott S Kemp
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida School of Medicine, Tampa, Florida
| | - Courtney T Griffin
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma; Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - George E Davis
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida School of Medicine, Tampa, Florida.
| |
Collapse
|
40
|
Vimentin-Rab7a Pathway Mediates the Migration of MSCs and Lead to Therapeutic Effects on ARDS. Stem Cells Int 2021; 2021:9992381. [PMID: 34367295 PMCID: PMC8342148 DOI: 10.1155/2021/9992381] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2021] [Accepted: 07/14/2021] [Indexed: 01/29/2023] Open
Abstract
Acute respiratory distress syndrome (ARDS) is difficult to treat and has a high mortality rate. Mesenchymal stem cells (MSCs) have an important therapeutic effect in ARDS. While the mechanism of MSC migration to the lungs remains unclear, the role of MSCs is of great clinical significance. To this end, we constructed vimentin knockout mice, extracted bone MSCs from the mice, and used them for the treatment of LPS-induced ARDS. H&E staining and Masson staining of mouse lung tissue allowed us to assess the degree of damage and fibrosis of mouse lung tissue. By measuring serum TNF-α, TGF-β, and INF-γ, we were able to monitor the release of inflammatory factors. Finally, through immunoprecipitation and gene knockout experiments, we identified upstream molecules that regulate vimentin and elucidated the mechanism that mediates MSC migration. As a result, we found that MSCs from wild-type mice can significantly alleviate ARDS and reduce lung inflammation, while vimentin gene knockout reduced the therapeutic effect of MSCs in ARDS. Cytological experiments showed that vimentin gene knockout can significantly inhibit the migration of MSCs and showed that it changes the proliferation and differentiation status of MSCs. Further experiments found that vimentin's regulation of MSC migration is mainly mediated by Rab7a. Rab7a knockout blocked the migration of MSCs and weakened the therapeutic effect of MSCs in ARDS. In conclusion, we have shown that the Vimentin-Rab7a pathway mediates migration of MSCs and leads to therapeutic effects in ARDS.
Collapse
|
41
|
Miljanovic N, Hauck SM, van Dijk RM, Di Liberto V, Rezaei A, Potschka H. Proteomic signature of the Dravet syndrome in the genetic Scn1a-A1783V mouse model. Neurobiol Dis 2021; 157:105423. [PMID: 34144125 DOI: 10.1016/j.nbd.2021.105423] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2021] [Revised: 05/14/2021] [Accepted: 06/08/2021] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Dravet syndrome is a rare, severe pediatric epileptic encephalopathy associated with intellectual and motor disabilities. Proteomic profiling in a mouse model of Dravet syndrome can provide information about the molecular consequences of the genetic deficiency and about pathophysiological mechanisms developing during the disease course. METHODS A knock-in mouse model of Dravet syndrome with Scn1a haploinsufficiency was used for whole proteome, seizure, and behavioral analysis. Hippocampal tissue was dissected from two- (prior to epilepsy manifestation) and four- (following epilepsy manifestation) week-old male mice and analyzed using LC-MS/MS with label-free quantification. Proteomic data sets were subjected to bioinformatic analysis including pathway enrichment analysis. The differential expression of selected proteins was confirmed by immunohistochemical staining. RESULTS The findings confirmed an increased susceptibility to hyperthermia-associated seizures, the development of spontaneous seizures, and behavioral alterations in the novel Scn1a-A1873V mouse model of Dravet syndrome. As expected, proteomic analysis demonstrated more pronounced alterations following epilepsy manifestation. In particular, proteins involved in neurotransmitter dynamics, receptor and ion channel function, synaptic plasticity, astrogliosis, neoangiogenesis, and nitric oxide signaling showed a pronounced regulation in Dravet mice. Pathway enrichment analysis identified several significantly regulated pathways at the later time point, with pathways linked to synaptic transmission and glutamatergic signaling dominating the list. CONCLUSION In conclusion, the whole proteome analysis in a mouse model of Dravet syndrome demonstrated complex molecular alterations in the hippocampus. Some of these alterations may have an impact on excitability or may serve a compensatory function, which, however, needs to be further confirmed by future investigations. The proteomic data indicate that, due to the molecular consequences of the genetic deficiency, the pathophysiological mechanisms may become more complex during the course of the disease. As a result, the management of Dravet syndrome may need to consider further molecular and cellular alterations. Ensuing functional follow-up studies, this data set may provide valuable guidance for the future development of novel therapeutic approaches.
Collapse
Affiliation(s)
- Nina Miljanovic
- Institute of Pharmacology, Toxicology & Pharmacy, Ludwig-Maximilians-University (LMU), Munich, Germany; Graduate School of Systemic Neurosciences (GSN), Ludwig-Maximilians-University (LMU), Munich, Germany
| | - Stefanie M Hauck
- Research Unit Protein Science, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Germany
| | - R Maarten van Dijk
- Institute of Pharmacology, Toxicology & Pharmacy, Ludwig-Maximilians-University (LMU), Munich, Germany
| | - Valentina Di Liberto
- Institute of Pharmacology, Toxicology & Pharmacy, Ludwig-Maximilians-University (LMU), Munich, Germany
| | - Ali Rezaei
- Institute of Pharmacology, Toxicology & Pharmacy, Ludwig-Maximilians-University (LMU), Munich, Germany; Graduate School of Systemic Neurosciences (GSN), Ludwig-Maximilians-University (LMU), Munich, Germany
| | - Heidrun Potschka
- Institute of Pharmacology, Toxicology & Pharmacy, Ludwig-Maximilians-University (LMU), Munich, Germany.
| |
Collapse
|
42
|
Wu F, Yang J, Liu J, Wang Y, Mu J, Zeng Q, Deng S, Zhou H. Signaling pathways in cancer-associated fibroblasts and targeted therapy for cancer. Signal Transduct Target Ther 2021; 6:218. [PMID: 34108441 PMCID: PMC8190181 DOI: 10.1038/s41392-021-00641-0] [Citation(s) in RCA: 294] [Impact Index Per Article: 98.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Revised: 04/20/2021] [Accepted: 05/06/2021] [Indexed: 02/05/2023] Open
Abstract
To flourish, cancers greatly depend on their surrounding tumor microenvironment (TME), and cancer-associated fibroblasts (CAFs) in TME are critical for cancer occurrence and progression because of their versatile roles in extracellular matrix remodeling, maintenance of stemness, blood vessel formation, modulation of tumor metabolism, immune response, and promotion of cancer cell proliferation, migration, invasion, and therapeutic resistance. CAFs are highly heterogeneous stromal cells and their crosstalk with cancer cells is mediated by a complex and intricate signaling network consisting of transforming growth factor-beta, phosphoinositide 3-kinase/AKT/mammalian target of rapamycin, mitogen-activated protein kinase, Wnt, Janus kinase/signal transducers and activators of transcription, epidermal growth factor receptor, Hippo, and nuclear factor kappa-light-chain-enhancer of activated B cells, etc., signaling pathways. These signals in CAFs exhibit their own special characteristics during the cancer progression and have the potential to be targeted for anticancer therapy. Therefore, a comprehensive understanding of these signaling cascades in interactions between cancer cells and CAFs is necessary to fully realize the pivotal roles of CAFs in cancers. Herein, in this review, we will summarize the enormous amounts of findings on the signals mediating crosstalk of CAFs with cancer cells and its related targets or trials. Further, we hypothesize three potential targeting strategies, including, namely, epithelial-mesenchymal common targets, sequential target perturbation, and crosstalk-directed signaling targets, paving the way for CAF-directed or host cell-directed antitumor therapy.
Collapse
Affiliation(s)
- Fanglong Wu
- State Key Laboratory of Oral Diseases, National Center of Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, People's Republic of China
| | - Jin Yang
- State Key Laboratory of Oral Diseases, National Center of Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, People's Republic of China
| | - Junjiang Liu
- State Key Laboratory of Oral Diseases, National Center of Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, People's Republic of China
| | - Ye Wang
- State Key Laboratory of Oral Diseases, National Center of Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, People's Republic of China
| | - Jingtian Mu
- State Key Laboratory of Oral Diseases, National Center of Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, People's Republic of China
| | - Qingxiang Zeng
- State Key Laboratory of Oral Diseases, National Center of Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, People's Republic of China
| | - Shuzhi Deng
- State Key Laboratory of Oral Diseases, National Center of Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, People's Republic of China
| | - Hongmei Zhou
- State Key Laboratory of Oral Diseases, National Center of Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, People's Republic of China.
| |
Collapse
|
43
|
Bayir E, Sendemir A. Role of Intermediate Filaments in Blood-Brain Barrier in Health and Disease. Cells 2021; 10:cells10061400. [PMID: 34198868 PMCID: PMC8226756 DOI: 10.3390/cells10061400] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2021] [Revised: 05/31/2021] [Accepted: 06/02/2021] [Indexed: 12/11/2022] Open
Abstract
The blood–brain barrier (BBB) is a highly selective cellular monolayer unique to the microvasculature of the central nervous system (CNS), and it mediates the communication of the CNS with the rest of the body by regulating the passage of molecules into the CNS microenvironment. Limitation of passage of substances through the BBB is mainly due to tight junctions (TJ) and adherens junctions (AJ) between brain microvascular endothelial cells. The importance of actin filaments and microtubules in establishing and maintaining TJs and AJs has been indicated; however, recent studies have shown that intermediate filaments are also important in the formation and function of cell–cell junctions. The most common intermediate filament protein in endothelial cells is vimentin. Vimentin plays a role in blood–brain barrier permeability in both cell–cell and cell–matrix interactions by affecting the actin and microtubule reorganization and by binding directly to VE-cadherin or integrin proteins. The BBB permeability increases due to the formation of stress fibers and the disruption of VE–cadherin interactions between two neighboring cells in various diseases, disrupting the fiber network of intermediate filament vimentin in different ways. Intermediate filaments may be long ignored key targets in regulation of BBB permeability in health and disease.
Collapse
Affiliation(s)
- Ece Bayir
- Ege University Central Research Test and Analysis Laboratory Application and Research Center (EGE-MATAL), Ege University, 35100 Izmir, Turkey;
| | - Aylin Sendemir
- Department of Bioengineering, Faculty of Engineering, Ege University, 35100 Izmir, Turkey
- Department of Biomedical Technologies, Graduate School of Natural and Applied Science, Ege University, 35100 Izmir, Turkey
- Correspondence: ; Tel.: +90-232-3114817
| |
Collapse
|
44
|
Ho Thanh MT, Grella A, Kole D, Ambady S, Wen Q. Vimentin intermediate filaments modulate cell traction force but not cell sensitivity to substrate stiffness. Cytoskeleton (Hoboken) 2021; 78:293-302. [PMID: 33993652 DOI: 10.1002/cm.21675] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Revised: 04/29/2021] [Accepted: 05/05/2021] [Indexed: 12/17/2022]
Abstract
The ability of cells to sense and respond to the mechanical stiffness of the surrounding matrix is important to support normal cell function, wound healing, and development. Central to the process of durosensing is the cytoskeleton composed of three classes of filaments: F-actin, microtubules, and intermediate filaments (IFs). Vimentin is an IF protein that contributes significantly to cell mechanics and cell traction force, which is required to probe extracellular matrix. The role of vimentin in how cells sense and respond to the mechanical rigidity of extracellular matrix is largely unclear. To investigate the role of vimentin in durosensing, we knocked down the vimentin expression level in 3T3 fibroblasts using shRNA transfection and measured cellular responses as functions of substrate stiffness. We quantified durosensitivity by the rates at which cell area and traction force change with substrate stiffness. Our results show that that vimentin plays a role in durosensing by modulating traction force and knocking out vimentin did not significantly affect durosensitivity. These results indicate that vimentin may be a redundant component of the machinery that cells use to sense substrate stiffness.
Collapse
Affiliation(s)
- Minh-Tri Ho Thanh
- Physics Department, Worcester Polytechnic Institute, Worcester, Massachusetts, USA
| | - Alexandra Grella
- Biology & Biotechnology Department, Worcester Polytechnic Institute, Worcester, Massachusetts, USA
| | - Denis Kole
- Biology & Biotechnology Department, Worcester Polytechnic Institute, Worcester, Massachusetts, USA
| | - Sakthikumar Ambady
- Biomedical Engineering Department, Worcester Polytechnic Institute, Worcester, Massachusetts, USA
| | - Qi Wen
- Physics Department, Worcester Polytechnic Institute, Worcester, Massachusetts, USA.,Biomedical Engineering Department, Worcester Polytechnic Institute, Worcester, Massachusetts, USA
| |
Collapse
|
45
|
Zhang P, He W, Huang Y, Xiao K, Tang Y, Huang L, Huang X, Zhang J, Yang W, Liu R, Fu Q, Lu Y, Zhang M. Proteomic and phosphoproteomic profiles of Sertoli cells in buffalo. Theriogenology 2021; 170:1-14. [PMID: 33945957 DOI: 10.1016/j.theriogenology.2021.04.013] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Revised: 03/27/2021] [Accepted: 04/21/2021] [Indexed: 01/12/2023]
Abstract
Sertoli cells provide nutrients and support for germ cell differentiation and maintain a stable microenvironment for spermatogenesis. Comprehensive identification of Sertoli cellular proteins is important in understanding spermatogenesis. In this study, we performed an integrative analysis of the proteome and phosphoproteome to explore the role of Sertoli cells in spermatogenesis. A total of 2912 and 753 proteins were identified from the proteome and phosphoproteome in Sertoli cells, respectively; 438 proteins were common to the proteome and phosphoproteome. Data are available via ProteomeXchange with identifier PXD024984. In the proteome, ACTG1, ACTB, ACTA2, MYH9 were the most abundant proteins. Gene Ontology (GO) analysis indicated that most of the proteins were involved in the processes of localization, biosynthesis, gene expression, and transport. In addition, some of the proteins related to Sertoli cell functions were also enriched. In the phosphoproteome, most of the proteins were involved in gene expression and the RNA metabolic process; the pathways mainly involved the spliceosome, mitogen-activated protein kinase signaling pathway, focal adhesion, and tight junctions. The pleckstrin homology-like domain is the most highly enriched protein domain in phosphoproteins. Cyclin-dependent kinases and protein kinases C were found to be highly active kinases in the kinase-substrate network analysis. Ten proteins most closely related to network stability were found in the analysis of the network interactions of proteins identified jointly in the phosphoproteome and proteome. Through immunohistochemistry and immunofluorescence verification of vimentin, it was found that there were localization differences between phosphorylated and non-phosphorylated vimentin in testicular tissue. This study is the first in-depth proteomic and phosphoproteomic analysis of buffalo testicular Sertoli cells. The results provide insight into the role of Sertoli cells in spermatogenesis and provide clues for further study of male reproduction.
Collapse
Affiliation(s)
- Pengfei Zhang
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, Animal Reproduction Institute, Guangxi University, Nanning, Guangxi, China
| | - Wengtan He
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, Animal Reproduction Institute, Guangxi University, Nanning, Guangxi, China
| | - Yulin Huang
- Department of Cell and Genetics, College of Basic Medicine, Guangxi University of Chinese Medicine, Nanning, Guangxi, China
| | - Kai Xiao
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, Animal Reproduction Institute, Guangxi University, Nanning, Guangxi, China
| | - Yuyan Tang
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, Animal Reproduction Institute, Guangxi University, Nanning, Guangxi, China
| | - Liangfeng Huang
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, Animal Reproduction Institute, Guangxi University, Nanning, Guangxi, China
| | - Xingchen Huang
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, Animal Reproduction Institute, Guangxi University, Nanning, Guangxi, China
| | - Junjun Zhang
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, Animal Reproduction Institute, Guangxi University, Nanning, Guangxi, China
| | - Weihan Yang
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, Animal Reproduction Institute, Guangxi University, Nanning, Guangxi, China
| | - Runfeng Liu
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, Animal Reproduction Institute, Guangxi University, Nanning, Guangxi, China
| | - Qiang Fu
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, Animal Reproduction Institute, Guangxi University, Nanning, Guangxi, China.
| | - Yangqing Lu
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, Animal Reproduction Institute, Guangxi University, Nanning, Guangxi, China.
| | - Ming Zhang
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, Animal Reproduction Institute, Guangxi University, Nanning, Guangxi, China.
| |
Collapse
|
46
|
Chang N, Cui Y, Liang X, Han D, Zheng X, Wu A, Qian L. Long Noncoding RNA LINC00857 Promotes Proliferation, Migration, and Invasion of Colorectal Cancer Cell through miR-1306/Vimentin Axis. COMPUTATIONAL AND MATHEMATICAL METHODS IN MEDICINE 2021; 2021:5525763. [PMID: 33833823 PMCID: PMC8012143 DOI: 10.1155/2021/5525763] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Revised: 02/19/2021] [Accepted: 02/28/2021] [Indexed: 12/13/2022]
Abstract
Colorectal cancer is a commonly diagnosed cancer and the leading cause of cancer-related death which still increasing in many countries. The lack of biomarkers for early detection and clinic treatment results in high morbidity and mortality. The novel role of long noncoding RNA LINC00857 on cell proliferation migration and invasion was explored in this article. The expression level of LINC00857 in colorectal cancer tissue samples and cells was determined notably higher than normal tissue samples and cells. Silence LINC00857 can significantly inhibit colorectal cancer cell viability and metastasis in vitro. Moreover, LINC00857 depletion caused cell accumulation in the G0/G1 phase. In addition, we recognized the novel LINC00857-miR-1306-vimentin axis and demonstrated it by dual-luciferase reporter assay. And this signaling axis could be considered as the target for colorectal cancer treatment. In conclusion, LINC00857 can promote colorectal cancer progress by sponging miR-1306 and upregulate vimentin to accelerate the epithelial-mesenchymal transition process.
Collapse
Affiliation(s)
- Na Chang
- Department of Radiation Oncology, Anhui Provincial Cancer Hospital (West District, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China), Hefei, Anhui 230031, China
| | - Yayun Cui
- Department of Radiation Oncology, Anhui Provincial Cancer Hospital (West District, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China), Hefei, Anhui 230031, China
| | - Xue Liang
- Department of Radiation Oncology, Anhui Provincial Cancer Hospital (West District, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China), Hefei, Anhui 230031, China
| | - Dan Han
- Department of Radiation Oncology, Anhui Provincial Cancer Hospital (West District, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China), Hefei, Anhui 230031, China
| | - Xiaomin Zheng
- Department of Radiation Oncology, Anhui Provincial Cancer Hospital (West District, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China), Hefei, Anhui 230031, China
| | - Ailin Wu
- Department of Radiation Oncology, Anhui Provincial Cancer Hospital (West District, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China), Hefei, Anhui 230031, China
| | - Liting Qian
- Department of Radiation Oncology, Anhui Provincial Cancer Hospital (West District, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China), Hefei, Anhui 230031, China
| |
Collapse
|
47
|
Zeng WR, Doran PM. Interactivity of biochemical and physical stimuli during epigenetic conditioning and cardiomyocytic differentiation of stem and progenitor cells derived from adult hearts. Integr Biol (Camb) 2021; 13:73-85. [PMID: 33704437 DOI: 10.1093/intbio/zyab003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Revised: 12/10/2020] [Accepted: 01/19/2021] [Indexed: 11/13/2022]
Abstract
Mixed populations of cardiosphere-derived stem and progenitor cells containing proliferative and cardiomyogenically committed cells were obtained from adult rat hearts. The cells were cultured in either static 2D monolayers or dynamic 3D scaffold systems with fluid flow. Cardiomyocyte lineage commitment in terms of GATA4 and Nkx2.5 expression was significantly enhanced in the dynamic 3D cultures compared with static 2D conditions. Treatment of the cells with 5-azacytidine (5-aza) produced different responses in the two culture systems, as activity of this chemical epigenetic conditioning agent depended on the cell attachment and hydrodynamic conditions provided during culture. Cell growth was unaffected by 5-aza in the static 2D cultures but was significantly reduced under dynamic 3D conditions relative to untreated controls. Myogenic differentiation measured as Mef2c expression was markedly upregulated by 5-aza in the dynamic 3D cultures but downregulated in the static 2D cultures. The ability of the physical environment to modulate the cellular cardiomyogenic response to 5-aza underscores the interactivity of biochemical and physical stimuli applied for cell differentiation. Accordingly, observations about the efficacy of 5-aza as a cardiomyocyte induction agent may not be applicable across different culture systems. Overall, use of dynamic 3D rather than static 2D culture was more beneficial for cardio-specific myogenesis than 5-aza treatment, which generated a more ambiguous differentiation response.
Collapse
Affiliation(s)
- Wendy R Zeng
- Faculty of Science, Engineering and Technology, Swinburne University of Technology, Melbourne, VIC, Australia
| | - Pauline M Doran
- Faculty of Science, Engineering and Technology, Swinburne University of Technology, Melbourne, VIC, Australia
| |
Collapse
|
48
|
Vašíček J, Baláži A, Bauer M, Svoradová A, Tirpáková M, Tomka M, Chrenek P. Molecular Profiling and Gene Banking of Rabbit EPCs Derived from Two Biological Sources. Genes (Basel) 2021; 12:genes12030366. [PMID: 33806502 PMCID: PMC7998175 DOI: 10.3390/genes12030366] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 02/27/2021] [Accepted: 03/03/2021] [Indexed: 12/20/2022] Open
Abstract
Endothelial progenitor cells (EPCs) have been broadly studied for several years due to their outstanding regenerative potential. Moreover, these cells might be a valuable source of genetic information for the preservation of endangered animal species. However, a controversy regarding their characterization still exists. The aim of this study was to isolate and compare the rabbit peripheral blood- and bone marrow-derived EPCs with human umbilical vein endothelial cells (HUVECs) in terms of their phenotype and morphology that could be affected by the passage number or cryopreservation as well as to assess their possible neuro-differentiation potential. Briefly, cells were isolated and cultured under standard endothelial conditions until passage 3. The morphological changes during the culture were monitored and each passage was analyzed for the typical phenotype using flow cytometry, quantitative real–time polymerase chain reaction (qPCR) and novel digital droplet PCR (ddPCR), and compared to HUVECs. The neurogenic differentiation was induced using a commercial kit. Rabbit cells were also cryopreserved for at least 3 months and then analyzed after thawing. According to the obtained results, both rabbit EPCs exhibit a spindle-shaped morphology and high proliferation rate. The both cell lines possess same stable phenotype: CD14−CD29+CD31−CD34−CD44+CD45−CD49f+CD73+CD90+CD105+CD133−CD146−CD166+VE-cadherin+VEGFR-2+SSEA-4+MSCA-1−vWF+eNOS+AcLDL+ALDH+vimentin+desmin+α-SMA+, slightly different from HUVECs. Moreover, both induced rabbit EPCs exhibit neuron-like morphological changes and expression of neuronal markers ENO2 and MAP2. In addition, cryopreserved rabbit cells maintained high viability (>85%) and endothelial phenotype after thawing. In conclusion, our findings suggest that cells expanded from the rabbit peripheral blood and bone marrow are of the endothelial origin with a stable marker expression and interesting proliferation and differentiation capacity.
Collapse
Affiliation(s)
- Jaromír Vašíček
- NPPC, Research Institute for Animal Production Nitra, Institute of Farm Animal Genetics and Reproduction, Hlohovecká 2, 951 41 Lužianky, Slovakia; (A.B.); (M.B.); (A.S.); (M.T.)
- Department of Biochemistry and Biotechnology, Faculty of Biotechnology and Food Science, Slovak University of Agriculture in Nitra, Tr. A. Hlinku 2, 949 76 Nitra, Slovakia;
- Correspondence: (J.V.); (P.C.); Tel.: +421-37-654-6600 (J.V.); +421-37-641-4274 (P.C.)
| | - Andrej Baláži
- NPPC, Research Institute for Animal Production Nitra, Institute of Farm Animal Genetics and Reproduction, Hlohovecká 2, 951 41 Lužianky, Slovakia; (A.B.); (M.B.); (A.S.); (M.T.)
| | - Miroslav Bauer
- NPPC, Research Institute for Animal Production Nitra, Institute of Farm Animal Genetics and Reproduction, Hlohovecká 2, 951 41 Lužianky, Slovakia; (A.B.); (M.B.); (A.S.); (M.T.)
- Department of Botany and Genetics, Faculty of Natural Sciences, Constantine the Philosopher University in Nitra, Nábrežie mládeže 91, 949 74 Nitra, Slovakia
| | - Andrea Svoradová
- NPPC, Research Institute for Animal Production Nitra, Institute of Farm Animal Genetics and Reproduction, Hlohovecká 2, 951 41 Lužianky, Slovakia; (A.B.); (M.B.); (A.S.); (M.T.)
| | - Mária Tirpáková
- Department of Biochemistry and Biotechnology, Faculty of Biotechnology and Food Science, Slovak University of Agriculture in Nitra, Tr. A. Hlinku 2, 949 76 Nitra, Slovakia;
- AgroBioTech Research Center, Slovak University of Agriculture in Nitra, Tr. A. Hlinku 2, 949 76 Nitra, Slovakia
| | - Marián Tomka
- NPPC, Research Institute for Animal Production Nitra, Institute of Farm Animal Genetics and Reproduction, Hlohovecká 2, 951 41 Lužianky, Slovakia; (A.B.); (M.B.); (A.S.); (M.T.)
| | - Peter Chrenek
- NPPC, Research Institute for Animal Production Nitra, Institute of Farm Animal Genetics and Reproduction, Hlohovecká 2, 951 41 Lužianky, Slovakia; (A.B.); (M.B.); (A.S.); (M.T.)
- Department of Biochemistry and Biotechnology, Faculty of Biotechnology and Food Science, Slovak University of Agriculture in Nitra, Tr. A. Hlinku 2, 949 76 Nitra, Slovakia;
- Correspondence: (J.V.); (P.C.); Tel.: +421-37-654-6600 (J.V.); +421-37-641-4274 (P.C.)
| |
Collapse
|
49
|
Herzog R, Bartosova M, Tarantino S, Wagner A, Unterwurzacher M, Sacnun JM, Lichtenauer AM, Kuster L, Schaefer B, Alper SL, Aufricht C, Schmitt CP, Kratochwill K. Peritoneal Dialysis Fluid Supplementation with Alanyl-Glutamine Attenuates Conventional Dialysis Fluid-Mediated Endothelial Cell Injury by Restoring Perturbed Cytoprotective Responses. Biomolecules 2020; 10:biom10121678. [PMID: 33334074 PMCID: PMC7765520 DOI: 10.3390/biom10121678] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Revised: 12/09/2020] [Accepted: 12/12/2020] [Indexed: 12/18/2022] Open
Abstract
Long-term clinical outcome of peritoneal dialysis (PD) depends on adequate removal of small solutes and water. The peritoneal endothelium represents the key barrier and peritoneal transport dysfunction is associated with vascular changes. Alanyl-glutamine (AlaGln) has been shown to counteract PD-induced deteriorations but the effect on vascular changes has not yet been elucidated. Using multiplexed proteomic and bioinformatic analyses we investigated the molecular mechanisms of vascular pathology in-vitro (primary human umbilical vein endothelial cells, HUVEC) and ex-vivo (arterioles of patients undergoing PD) following exposure to PD-fluid. An overlap of 1813 proteins (40%) of over 3100 proteins was identified in both sample types. PD-fluid treatment significantly altered 378 in endothelial cells and 192 in arterioles. The HUVEC proteome resembles the arteriolar proteome with expected sample specific differences of mainly immune system processes only present in arterioles and extracellular region proteins primarily found in HUVEC. AlaGln-addition to PD-fluid revealed 359 differentially abundant proteins and restored the molecular process landscape altered by PD fluid. This study provides evidence on validity and inherent limitations of studying endothelial pathomechanisms in-vitro compared to vascular ex-vivo findings. AlaGln could reduce PD-associated vasculopathy by reducing endothelial cellular damage, restoring perturbed abundances of pathologically important proteins and enriching protective processes.
Collapse
Affiliation(s)
- Rebecca Herzog
- Division of Pediatric Nephrology and Gastroenterology, Department of Pediatrics and Adolescent Medicine, Comprehensive Center for Pediatrics, Medical University of Vienna, 1090 Vienna, Austria; (R.H.); (S.T.); (J.M.S.); (A.M.L.); (L.K.); (C.A.)
- Christian Doppler Laboratory for Molecular Stress Research in Peritoneal Dialysis, Medical University of Vienna, 1090 Vienna, Austria; (A.W.); (M.U.)
| | - Maria Bartosova
- Center for Pediatric and Adolescent Medicine, University of Heidelberg, 69120 Heidelberg, Germany; (M.B.); (B.S.); (C.P.S.)
| | - Silvia Tarantino
- Division of Pediatric Nephrology and Gastroenterology, Department of Pediatrics and Adolescent Medicine, Comprehensive Center for Pediatrics, Medical University of Vienna, 1090 Vienna, Austria; (R.H.); (S.T.); (J.M.S.); (A.M.L.); (L.K.); (C.A.)
- Zytoprotec GmbH, 1090 Vienna, Austria
| | - Anja Wagner
- Division of Pediatric Nephrology and Gastroenterology, Department of Pediatrics and Adolescent Medicine, Comprehensive Center for Pediatrics, Medical University of Vienna, 1090 Vienna, Austria; (R.H.); (S.T.); (J.M.S.); (A.M.L.); (L.K.); (C.A.)
- Christian Doppler Laboratory for Molecular Stress Research in Peritoneal Dialysis, Medical University of Vienna, 1090 Vienna, Austria; (A.W.); (M.U.)
| | - Markus Unterwurzacher
- Division of Pediatric Nephrology and Gastroenterology, Department of Pediatrics and Adolescent Medicine, Comprehensive Center for Pediatrics, Medical University of Vienna, 1090 Vienna, Austria; (R.H.); (S.T.); (J.M.S.); (A.M.L.); (L.K.); (C.A.)
- Christian Doppler Laboratory for Molecular Stress Research in Peritoneal Dialysis, Medical University of Vienna, 1090 Vienna, Austria; (A.W.); (M.U.)
| | - Juan Manuel Sacnun
- Division of Pediatric Nephrology and Gastroenterology, Department of Pediatrics and Adolescent Medicine, Comprehensive Center for Pediatrics, Medical University of Vienna, 1090 Vienna, Austria; (R.H.); (S.T.); (J.M.S.); (A.M.L.); (L.K.); (C.A.)
- Zytoprotec GmbH, 1090 Vienna, Austria
| | - Anton M. Lichtenauer
- Division of Pediatric Nephrology and Gastroenterology, Department of Pediatrics and Adolescent Medicine, Comprehensive Center for Pediatrics, Medical University of Vienna, 1090 Vienna, Austria; (R.H.); (S.T.); (J.M.S.); (A.M.L.); (L.K.); (C.A.)
| | - Lilian Kuster
- Division of Pediatric Nephrology and Gastroenterology, Department of Pediatrics and Adolescent Medicine, Comprehensive Center for Pediatrics, Medical University of Vienna, 1090 Vienna, Austria; (R.H.); (S.T.); (J.M.S.); (A.M.L.); (L.K.); (C.A.)
| | - Betti Schaefer
- Center for Pediatric and Adolescent Medicine, University of Heidelberg, 69120 Heidelberg, Germany; (M.B.); (B.S.); (C.P.S.)
| | - Seth L. Alper
- Division of Nephrology and Center for Vascular Biology Research, Beth Israel Deaconess Medical Center, Boston, MA 02215, USA;
- Department of Medicine, Harvard Medical School, Boston, MA 02115, USA
| | - Christoph Aufricht
- Division of Pediatric Nephrology and Gastroenterology, Department of Pediatrics and Adolescent Medicine, Comprehensive Center for Pediatrics, Medical University of Vienna, 1090 Vienna, Austria; (R.H.); (S.T.); (J.M.S.); (A.M.L.); (L.K.); (C.A.)
| | - Claus Peter Schmitt
- Center for Pediatric and Adolescent Medicine, University of Heidelberg, 69120 Heidelberg, Germany; (M.B.); (B.S.); (C.P.S.)
| | - Klaus Kratochwill
- Division of Pediatric Nephrology and Gastroenterology, Department of Pediatrics and Adolescent Medicine, Comprehensive Center for Pediatrics, Medical University of Vienna, 1090 Vienna, Austria; (R.H.); (S.T.); (J.M.S.); (A.M.L.); (L.K.); (C.A.)
- Christian Doppler Laboratory for Molecular Stress Research in Peritoneal Dialysis, Medical University of Vienna, 1090 Vienna, Austria; (A.W.); (M.U.)
- Correspondence: ; Tel.: +43-140400-80
| |
Collapse
|
50
|
Regulatory mechanisms and clinical significance of vimentin in breast cancer. Biomed Pharmacother 2020; 133:111068. [PMID: 33378968 DOI: 10.1016/j.biopha.2020.111068] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 11/18/2020] [Accepted: 11/20/2020] [Indexed: 02/05/2023] Open
Abstract
Vimentin, a kind of intermediate filament protein III in mesenchymal cells, has become a highly researched topic around the world in recent years, as it holds complex biological functions and plays an important role in the epithelial-mesenchymal transition in the evolution of various tumors. This article reviews the biological function of vimentin and its relationship with breast cancer in order to provide novel ideas about the clinical diagnosis and targeted therapy of breast cancer.
Collapse
|