1
|
Lei X, Miao S, Wang X, Gao Y, Wu H, Cheng P, Song Y, Bi L, Pei G. Microgroove Cues Guiding Fibrogenesis of Stem Cells via Intracellular Force. ACS APPLIED MATERIALS & INTERFACES 2023; 15:16380-16393. [PMID: 36961871 DOI: 10.1021/acsami.2c20903] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
Groove patterns are widely used in material surface modifications. However, the independent role of ditches/ridges in regulating fibrosis of soft tissues is not well-understood, especially the lack of linkage evidence in vitro and in vivo. Herein, two kinds of combinational microgroove chips with the gradient ditch/ridge width were fabricated by photolithography technology, termed R and G groups, respectively. In group R, the ridge width was 1, 5, 10, and 30 μm, with a ditch width of 30 μm; in group G, the groove width was 5, 10, 20, and 30 μm, and the ridge width was 5 μm. The effect of microgrooves on the morphology, proliferation, and expression of fibrous markers of stem cells was systematically investigated in vitro. Moreover, thicknesses of fibrous capsules were evaluated after chips were implanted into the muscular pouches of rats for 5 months. The results show that microgrooves have almost no effect on cell proliferation but significantly modulate the morphology of cells and focal adhesions (FAs) in vitro, as well as fibrosis differentiation. In particular, the differentiation of stem cells is attenuated after the intracellular force caused by stress fibers and FAs is interfered by drugs, such as rotenone and blebbistatin. Histological analysis shows that patterns of high intracellular force can apparently stimulate soft tissue fibrosis in vivo. This study not only reveals the specific rules and mechanisms of ditch/ridge regulating stem cell behaviors but also offers insight into tailoring implant surface patterns to induce controlled soft tissue fibrosis.
Collapse
Affiliation(s)
- Xing Lei
- Department of Orthopedics, Xijing Hospital, Air Force Medical University, Xi'an 710032, China
- Department of Orthopedic Surgery, Linyi People's Hospital, Linyi 276000, China
| | - Sheng Miao
- Department of Orthopedics, Xijing Hospital, Air Force Medical University, Xi'an 710032, China
| | - Xiuli Wang
- State Key Laboratory of Molecular Engineering of Polymers, Department of Macromolecular Science, Fudan University, Shanghai 200438, China
| | - Yi Gao
- Southern University of Science and Technology Hospital, No. 6019 Liuxian Street, Xili Avenue, Nanshan District, Shenzhen 518055, China
- School of Medicine, Southern University of Science and Technology, Shenzhen 518055, China
| | - Hao Wu
- Department of Orthopedics, Xijing Hospital, Air Force Medical University, Xi'an 710032, China
| | - Pengzhen Cheng
- Department of Orthopedics, Xijing Hospital, Air Force Medical University, Xi'an 710032, China
| | - Yue Song
- Department of Orthopedics, Xijing Hospital, Air Force Medical University, Xi'an 710032, China
| | - Long Bi
- Department of Orthopedics, Xijing Hospital, Air Force Medical University, Xi'an 710032, China
| | - Guoxian Pei
- Department of Orthopedics, Xijing Hospital, Air Force Medical University, Xi'an 710032, China
- Southern University of Science and Technology Hospital, No. 6019 Liuxian Street, Xili Avenue, Nanshan District, Shenzhen 518055, China
- School of Medicine, Southern University of Science and Technology, Shenzhen 518055, China
| |
Collapse
|
2
|
Maruszak A, Silajdžić E, Lee H, Murphy T, Liu B, Shi L, de Lucia C, Douiri A, Salta E, Nevado AJ, Teunissen CE, Visser PJ, Price J, Zetterberg H, Lovestone S, Thuret S. Predicting progression to Alzheimer's disease with human hippocampal progenitors exposed to serum. Brain 2023; 146:2045-2058. [PMID: 36703180 PMCID: PMC10151193 DOI: 10.1093/brain/awac472] [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] [Received: 08/02/2022] [Revised: 10/11/2022] [Accepted: 11/10/2022] [Indexed: 01/28/2023] Open
Abstract
Adult hippocampal neurogenesis is important for learning and memory and is altered early in Alzheimer's disease. As hippocampal neurogenesis is modulated by the circulatory systemic environment, evaluating a proxy of how hippocampal neurogenesis is affected by the systemic milieu could serve as an early biomarker for Alzheimer's disease progression. Here, we used an in vitro assay to model the impact of systemic environment on hippocampal neurogenesis. A human hippocampal progenitor cell line was treated with longitudinal serum samples from individuals with mild cognitive impairment, who either progressed to Alzheimer's disease or remained cognitively stable. Mild cognitive impairment to Alzheimer's disease progression was characterized most prominently with decreased proliferation, increased cell death and increased neurogenesis. A subset of 'baseline' cellular readouts together with education level were able to predict Alzheimer's disease progression. The assay could provide a powerful platform for early prognosis, monitoring disease progression and further mechanistic studies.
Collapse
Affiliation(s)
- Aleksandra Maruszak
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry Psychology & Neuroscience, King's College London, London, SE5 9RX, UK
| | - Edina Silajdžić
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry Psychology & Neuroscience, King's College London, London, SE5 9RX, UK
| | - Hyunah Lee
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry Psychology & Neuroscience, King's College London, London, SE5 9RX, UK
| | - Tytus Murphy
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry Psychology & Neuroscience, King's College London, London, SE5 9RX, UK
| | - Benjamine Liu
- Department of Psychiatry, University of Oxford, Oxford, OX3 7JX, UK
| | - Liu Shi
- Department of Psychiatry, University of Oxford, Oxford, OX3 7JX, UK
| | - Chiara de Lucia
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry Psychology & Neuroscience, King's College London, London, SE5 9RX, UK
| | - Abdel Douiri
- Department of Population Health Sciences, King's College London, London, SE1 1UL, UK
| | - Evgenia Salta
- Netherlands Institute for Neuroscience, 1105 BA Amsterdam, The Netherlands.,Neurochemistry Lab and Biobank, Department of Clinical Chemistry, Amsterdam Neuroscience, VU University Medical Center, 1007 MB Amsterdam, The Netherlands
| | - Alejo J Nevado
- Department of Psychiatry, University of Oxford, Oxford, OX3 7JX, UK
| | - Charlotte E Teunissen
- Neurochemistry Lab and Biobank, Department of Clinical Chemistry, Amsterdam Neuroscience, VU University Medical Center, 1007 MB Amsterdam, The Netherlands
| | - Pieter J Visser
- Department of Psychiatry and Neuropsychology, Alzheimer Center Limburg, School for Mental Health and Neuroscience, Maastricht University, 6200 MD Maastricht, The Netherlands.,Department of Neurology, Alzheimer Center, VU University Medical Center, 1081 HZ Amsterdam, The Netherlands
| | - Jack Price
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry Psychology & Neuroscience, King's College London, London, SE5 9RX, UK
| | - Henrik Zetterberg
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, S-431 80 Mölndal, Sweden.,Department of Neurodegenerative Disease, UCL Institute of Neurology, London, WC1N 3BG, UK.,Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, the Sahlgrenska Academy at the University of Gothenburg, S-431 80 Mölndal, Sweden.,UK Dementia Research Institute at UCL, London, WC1E 6BT, UK
| | - Simon Lovestone
- Department of Psychiatry, University of Oxford, Oxford, OX3 7JX, UK.,Janssen Medical UK, B-2340 Beerse, Belgium
| | - Sandrine Thuret
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry Psychology & Neuroscience, King's College London, London, SE5 9RX, UK
| |
Collapse
|
3
|
Janowska A, Iannone M, Fidanzi C, Romanelli M, Filippi L, Del Re M, Martins M, Dini V. The Genetic Basis of Dormancy and Awakening in Cutaneous Metastatic Melanoma. Cancers (Basel) 2022; 14:2104. [PMID: 35565234 PMCID: PMC9102235 DOI: 10.3390/cancers14092104] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Revised: 04/19/2022] [Accepted: 04/21/2022] [Indexed: 01/27/2023] Open
Abstract
Immune dysregulation, in combination with genetic and epigenetic alterations, induces an excessive proliferation of uncontrolled melanoma cells followed by dissemination of the tumor cells to distant sites, invading organs and creating metastasis. Although immunotherapy, checkpoint inhibitors and molecular targeted therapies have been developed as treatment options for advanced melanoma, there are specific mechanisms by which cancer cells can escape treatment. One of the main factors associated with reduced response to therapy is the ability of residual tumor cells to persist in a dormant state, without proliferation. This comprehensive review aimed at understanding the genetic basis of dormancy/awakening phenomenon in metastatic melanoma will help identify the possible therapeutical strategies that might eliminate melanoma circulating tumor cells (CTCs) or keep them in the dormant state forever, thereby repressing tumor relapse and metastatic spread.
Collapse
Affiliation(s)
- Agata Janowska
- Unit of Dermatology, University of Pisa, 56126 Pisa, Italy; (M.I.); (C.F.); (M.R.); (M.M.); (V.D.)
| | - Michela Iannone
- Unit of Dermatology, University of Pisa, 56126 Pisa, Italy; (M.I.); (C.F.); (M.R.); (M.M.); (V.D.)
| | - Cristian Fidanzi
- Unit of Dermatology, University of Pisa, 56126 Pisa, Italy; (M.I.); (C.F.); (M.R.); (M.M.); (V.D.)
| | - Marco Romanelli
- Unit of Dermatology, University of Pisa, 56126 Pisa, Italy; (M.I.); (C.F.); (M.R.); (M.M.); (V.D.)
| | - Luca Filippi
- Unit of Neonatology, University of Pisa, 56126 Pisa, Italy;
| | - Marzia Del Re
- Unit of Clinical Pharmacology and Pharmacogenetics, University of Pisa, 56126 Pisa, Italy;
| | - Manuella Martins
- Unit of Dermatology, University of Pisa, 56126 Pisa, Italy; (M.I.); (C.F.); (M.R.); (M.M.); (V.D.)
| | - Valentina Dini
- Unit of Dermatology, University of Pisa, 56126 Pisa, Italy; (M.I.); (C.F.); (M.R.); (M.M.); (V.D.)
| |
Collapse
|
4
|
Walker NM, Liu J, Young SM, Woode RA, Clarke LL. Goblet cell hyperplasia is not epithelial-autonomous in the Cftr knockout intestine. Am J Physiol Gastrointest Liver Physiol 2022; 322:G282-G293. [PMID: 34878935 PMCID: PMC8793866 DOI: 10.1152/ajpgi.00290.2021] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Revised: 11/22/2021] [Accepted: 12/01/2021] [Indexed: 02/03/2023]
Abstract
Goblet cell hyperplasia is an important manifestation of cystic fibrosis (CF) disease in epithelial-lined organs. Explants of CF airway epithelium show normalization of goblet cell numbers; therefore, we hypothesized that small intestinal enteroids from Cftr knockout (KO) mice would not exhibit goblet cell hyperplasia. Toll-like receptors 2 and 4 (Tlr2 and Tlr4) were investigated as markers of inflammation and influence on goblet cell differentiation. Ex vivo studies found goblet cell hyperplasia in Cftr KO jejunum compared with wild-type (WT) mice. IL-13, SAM pointed domain-containing ETS transcription factor (Spdef), Tlr2, and Tlr4 protein expression were increased in Cftr KO intestine relative to WT. In contrast, WT and Cftr KO enteroids did not exhibit differences in basal or IL-13-stimulated goblet cell numbers, or differences in expression of Tlr2, Tlr4, and Spdef. Ileal goblet cell numbers in Cftr KO/Tlr4 KO and Cftr KO/Tlr2 KO mice were not different from Cftr KO mice, but enumeration was confounded by altered mucosal morphology. Treatment with Tlr4 agonist LPS did not affect goblet cell numbers in WT or Cftr KO enteroids, whereas the Tlr2 agonist Pam3Csk4 stimulated goblet cell hyperplasia in both genotypes. Pam3Csk4 stimulation of goblet cell numbers was associated with suppression of Notch1 and Neurog3 expression and upregulated determinants of goblet cell differentiation. We conclude that goblet cell hyperplasia and inflammation of the Cftr KO small intestine are not exhibited by enteroids, indicating that this manifestation of CF intestinal disease is not epithelial-automatous but secondary to the altered CF intestinal environment.NEW & NOTEWORTHY Studies of small intestinal organoids from cystic fibrosis (CF) mice show that goblet cell hyperplasia and increased Toll-like receptor 2/4 expression are not primary manifestations of the CF intestine. Intestinal goblet cell hyperplasia in the CF mice was not strongly altered by genetic ablation of Tlr2 and Tlr 4, but could be induced in both wild-type and CF intestinal organoids by a Tlr2-dependent suppression of Notch signaling.
Collapse
Affiliation(s)
- Nancy M Walker
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri
| | - Jinghua Liu
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri
| | - Sarah M Young
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri
- Department of Pathobiology, University of Missouri, Columbia, Missouri
| | - Rowena A Woode
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri
- Department of Biomedical Sciences, University of Missouri, Columbia, Missouri
| | - Lane L Clarke
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri
- Department of Biomedical Sciences, University of Missouri, Columbia, Missouri
| |
Collapse
|
5
|
Lieu YK, Liu Z, Ali AM, Wei X, Penson A, Zhang J, An X, Rabadan R, Raza A, Manley JL, Mukherjee S. SF3B1 mutant-induced missplicing of MAP3K7 causes anemia in myelodysplastic syndromes. Proc Natl Acad Sci U S A 2022; 119:e2111703119. [PMID: 34930825 PMCID: PMC8740767 DOI: 10.1073/pnas.2111703119] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Accepted: 11/10/2021] [Indexed: 12/12/2022] Open
Abstract
SF3B1 is the most frequently mutated RNA splicing factor in cancer, including in ∼25% of myelodysplastic syndromes (MDS) patients. SF3B1-mutated MDS, which is strongly associated with ringed sideroblast morphology, is characterized by ineffective erythropoiesis, leading to severe, often fatal anemia. However, functional evidence linking SF3B1 mutations to the anemia described in MDS patients harboring this genetic aberration is weak, and the underlying mechanism is completely unknown. Using isogenic SF3B1 WT and mutant cell lines, normal human CD34 cells, and MDS patient cells, we define a previously unrecognized role of the kinase MAP3K7, encoded by a known mutant SF3B1-targeted transcript, in controlling proper terminal erythroid differentiation, and show how MAP3K7 missplicing leads to the anemia characteristic of SF3B1-mutated MDS, although not to ringed sideroblast formation. We found that p38 MAPK is deactivated in SF3B1 mutant isogenic and patient cells and that MAP3K7 is an upstream positive effector of p38 MAPK. We demonstrate that disruption of this MAP3K7-p38 MAPK pathway leads to premature down-regulation of GATA1, a master regulator of erythroid differentiation, and that this is sufficient to trigger accelerated differentiation, erythroid hyperplasia, and ultimately apoptosis. Our findings thus define the mechanism leading to the severe anemia found in MDS patients harboring SF3B1 mutations.
Collapse
Affiliation(s)
- Yen K Lieu
- Department of Biological Sciences, Columbia University, New York, NY 10027;
- Irving Cancer Research Center, Columbia University, New York, NY 10032
| | - Zhaoqi Liu
- Chinese Academy of Sciences Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences 100101 Beijing, China
- Department of Systems Biology, Columbia University, New York, NY 10032
- Department of Biomedical Informatics, Columbia University, New York, NY 10032
- Program for Mathematical Genomics, Columbia University, New York, NY 10032
| | - Abdullah M Ali
- Division of Hematology and Oncology, Department of Medicine, Columbia University, New York, NY 10032
| | - Xin Wei
- Laboratory of Membrane Biology, New York Blood Center, New York, NY 10065
- Department of Anesthesiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450008, China
| | - Alex Penson
- Department of Systems Biology, Columbia University, New York, NY 10032
- Department of Biomedical Informatics, Columbia University, New York, NY 10032
| | - Jian Zhang
- Department of Biological Sciences, Columbia University, New York, NY 10027
| | - Xiuli An
- Laboratory of Membrane Biology, New York Blood Center, New York, NY 10065
| | - Raul Rabadan
- Department of Systems Biology, Columbia University, New York, NY 10032
- Department of Biomedical Informatics, Columbia University, New York, NY 10032
- Program for Mathematical Genomics, Columbia University, New York, NY 10032
| | - Azra Raza
- Irving Cancer Research Center, Columbia University, New York, NY 10032
- Division of Hematology and Oncology, Department of Medicine, Columbia University, New York, NY 10032
| | - James L Manley
- Department of Biological Sciences, Columbia University, New York, NY 10027;
| | | |
Collapse
|
6
|
Stress Relief Techniques: p38 MAPK Determines the Balance of Cell Cycle and Apoptosis Pathways. Biomolecules 2021; 11:biom11101444. [PMID: 34680077 PMCID: PMC8533283 DOI: 10.3390/biom11101444] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Revised: 09/23/2021] [Accepted: 09/30/2021] [Indexed: 12/18/2022] Open
Abstract
Protein signaling networks are formed from diverse and inter-connected cell signaling pathways converging into webs of function and regulation. These signaling pathways both receive and conduct molecular messages, often by a series of post-translation modifications such as phosphorylation or through protein-protein interactions via intrinsic motifs. The mitogen activated protein kinases (MAPKs) are components of kinase cascades that transmit signals through phosphorylation. There are several MAPK subfamilies, and one subfamily is the stress-activated protein kinases, which in mammals is the p38 family. The p38 enzymes mediate a variety of cellular outcomes including DNA repair, cell survival/cell fate decisions, and cell cycle arrest. The cell cycle is itself a signaling system that precisely controls DNA replication, chromosome segregation, and cellular division. Another indispensable cell function influenced by the p38 stress response is programmed cell death (apoptosis). As the regulators of cell survival, the BCL2 family of proteins and their dynamics are exquisitely sensitive to cell stress. The BCL2 family forms a protein-protein interaction network divided into anti-apoptotic and pro-apoptotic members, and the balance of binding between these two sides determines cell survival. Here, we discuss the intersections among the p38 MAPK, cell cycle, and apoptosis signaling pathways.
Collapse
|
7
|
Canovas B, Nebreda AR. Diversity and versatility of p38 kinase signalling in health and disease. Nat Rev Mol Cell Biol 2021; 22:346-366. [PMID: 33504982 PMCID: PMC7838852 DOI: 10.1038/s41580-020-00322-w] [Citation(s) in RCA: 304] [Impact Index Per Article: 76.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/02/2020] [Indexed: 02/06/2023]
Abstract
The ability of cells to deal with different types of stressful situations in a precise and coordinated manner is key for survival and involves various signalling networks. Over the past 25 years, p38 kinases — in particular, p38α — have been implicated in the cellular response to stress at many levels. These span from environmental and intracellular stresses, such as hyperosmolarity, oxidative stress or DNA damage, to physiological situations that involve important cellular changes such as differentiation. Given that p38α controls a plethora of functions, dysregulation of this pathway has been linked to diseases such as inflammation, immune disorders or cancer, suggesting the possibility that targeting p38α could be of therapeutic interest. In this Review, we discuss the organization of this signalling pathway focusing on the diversity of p38α substrates, their mechanisms and their links to particular cellular functions. We then address how the different cellular responses can be generated depending on the signal received and the cell type, and highlight the roles of this kinase in human physiology and in pathological contexts. p38α — the best-characterized member of the p38 kinase family — is a key mediator of cellular stress responses. p38α is activated by a plethora of signals and functions through a multitude of substrates to regulate different cellular behaviours. Understanding context-dependent p38α signalling provides important insights into p38α roles in physiology and pathology.
Collapse
Affiliation(s)
- Begoña Canovas
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Barcelona, Spain
| | - Angel R Nebreda
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Barcelona, Spain. .,ICREA, Barcelona, Spain.
| |
Collapse
|
8
|
Shafiquzzaman M, Biswas S, Li P, Mishina Y, Li B, Liu H. The noncanonical BMP signaling pathway plays an important role in club cell regeneration. Stem Cells 2019; 38:437-450. [PMID: 31758827 DOI: 10.1002/stem.3125] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Revised: 09/19/2019] [Accepted: 11/05/2019] [Indexed: 12/13/2022]
Abstract
The bronchiole is a major site for the development of several life-threatening disorders, including chronic obstructive pulmonary disease and lung adenocarcinomas. The bronchiolar epithelium is composed of club cells and ciliated epithelial cells, with club cells serving as progenitor cells. Presently, the identity of the cells involved in regeneration of bronchiolar epithelium and the underlying mechanisms remain incompletely understood. Here, we show that Prrx1, a homeobox transcription factor, can mark club cells in adult mice during homeostasis and regeneration. We further show that the noncanonical signaling pathway of BMPs, BMPR1A-Tak1-p38MAPK, plays a critical role in club cell regeneration. Ablation of Bmpr1a, Tak1, or Mapk14 (encoding p38α) in Prrx1+ club cells caused minimal effect on bronchiolar epithelium homeostasis, yet it resulted in severe defects in club cell regeneration and bronchiole repair in adult mice. We further show that this pathway supports proliferation and expansion of the regenerating club cells. Our findings thus identify a marker for club cells and reveal a critical role for the BMP noncanonical pathway in club cell regeneration.
Collapse
Affiliation(s)
- Md Shafiquzzaman
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai, People's Republic of China.,Metabolic Bone Disease and Genetics Research Unit, Department of Osteoporosis and Bone Diseases, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, People's Republic of China.,National Institute of Biotechnology, Ministry of Science and Technology, Dhaka, Bangladesh
| | - Soma Biswas
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai, People's Republic of China
| | - Ping Li
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai, People's Republic of China
| | - Yuji Mishina
- Department of Biologic & Materials Sciences, School of Dentistry, University of Michigan, Ann Arbor, Michigan
| | - Baojie Li
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai, People's Republic of China.,Center for Traditional Chinese Medicine and Stem Cell Research, The Chengdu University of Traditional Chinese Medicine, Sichuan, People's Republic of China
| | - Huijuan Liu
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai, People's Republic of China.,Metabolic Bone Disease and Genetics Research Unit, Department of Osteoporosis and Bone Diseases, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, People's Republic of China
| |
Collapse
|
9
|
Lee H, Heo JW, Kim AR, Kweon M, Nam S, Lim JS, Sung MK, Kim SE, Ryu JH. Z-ajoene from Crushed Garlic Alleviates Cancer-Induced Skeletal Muscle Atrophy. Nutrients 2019; 11:nu11112724. [PMID: 31717643 PMCID: PMC6893518 DOI: 10.3390/nu11112724] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2019] [Revised: 11/05/2019] [Accepted: 11/07/2019] [Indexed: 12/24/2022] Open
Abstract
Skeletal muscle atrophy is one of the major symptoms of cancer cachexia. Garlic (Allium sativum), one of the world's most commonly used and versatile herbs, has been employed for the prevention and treatment of diverse diseases for centuries. In the present study, we found that ajoene, a sulfur compound found in crushed garlic, exhibits protective effects against muscle atrophy. Using CT26 tumor-bearing BALB/c mice, we demonstrate in vivo that ajoene extract alleviated muscle degradation by decreasing not only myokines secretion but also janus kinase/signal transducer and activator of transcription 3 (JAK/STAT3) and SMADs/forkhead box (FoxO) signaling pathways, thereby suppressing muscle-specific E3 ligases. In mouse skeletal myoblasts, Z-ajoene enhanced myogenesis as evidenced by increased expression of myogenic markers via p38 mitogen-activated protein kinase (MAPK) activation. In mature myotubes, Z-ajoene protected against muscle protein degradation induced by conditioned media from CT26 colon carcinoma cells, by suppressing expression of muscle specific E3 ligases and nuclear transcription factor kappa B (NF-κB) phosphorylation which contribute to muscle atrophy. Moreover, Z-ajoene treatment improved myofiber formation via stimulation of muscle protein synthesis. These findings suggest that ajoene extract and Z-ajoene can attenuate skeletal muscle atrophy induced by cancer cachexia through suppressing inflammatory responses and the muscle wasting as well as by promoting muscle protein synthesis.
Collapse
Affiliation(s)
- Hyejin Lee
- Research Institute of Pharmaceutical Sciences, Sookmyung Women’s University, Yongsan-Gu, Seoul 04310, Korea; (H.L.); minson-_-@nate.com (M.K.)
| | - Ji-Won Heo
- Department of Food and Nutrition, Sookmyung Women’s University, Yongsan-Gu, Seoul 04310, Korea; (J.-W.H.); (A.-R.K.); (M.-K.S.)
| | - A-Reum Kim
- Department of Food and Nutrition, Sookmyung Women’s University, Yongsan-Gu, Seoul 04310, Korea; (J.-W.H.); (A.-R.K.); (M.-K.S.)
| | - Minson Kweon
- Research Institute of Pharmaceutical Sciences, Sookmyung Women’s University, Yongsan-Gu, Seoul 04310, Korea; (H.L.); minson-_-@nate.com (M.K.)
| | - Sorim Nam
- Division of Biological Sciences and Cellular Heterogeneity Research Center, Sookmyung Women’s University, Yongsan-Gu, Seoul 04310, Korea; (S.N.); (J.-S.L.)
| | - Jong-Seok Lim
- Division of Biological Sciences and Cellular Heterogeneity Research Center, Sookmyung Women’s University, Yongsan-Gu, Seoul 04310, Korea; (S.N.); (J.-S.L.)
| | - Mi-Kyung Sung
- Department of Food and Nutrition, Sookmyung Women’s University, Yongsan-Gu, Seoul 04310, Korea; (J.-W.H.); (A.-R.K.); (M.-K.S.)
| | - Sung-Eun Kim
- Department of Food and Nutrition, Sookmyung Women’s University, Yongsan-Gu, Seoul 04310, Korea; (J.-W.H.); (A.-R.K.); (M.-K.S.)
- Correspondence: (S.-E.K.); (J.-H.R.); Tel.: +82-2-2077-7722 (S.-E.K.); +82-2-710-9568 (J.-H.R.)
| | - Jae-Ha Ryu
- Research Institute of Pharmaceutical Sciences, Sookmyung Women’s University, Yongsan-Gu, Seoul 04310, Korea; (H.L.); minson-_-@nate.com (M.K.)
- Correspondence: (S.-E.K.); (J.-H.R.); Tel.: +82-2-2077-7722 (S.-E.K.); +82-2-710-9568 (J.-H.R.)
| |
Collapse
|
10
|
A Chalcone from Ashitaba ( Angelica keiskei) Stimulates Myoblast Differentiation and Inhibits Dexamethasone-Induced Muscle Atrophy. Nutrients 2019; 11:nu11102419. [PMID: 31658768 PMCID: PMC6835314 DOI: 10.3390/nu11102419] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2019] [Revised: 09/30/2019] [Accepted: 10/05/2019] [Indexed: 02/07/2023] Open
Abstract
Ashitaba, Angelica keiskei Koidzumi (AK), as a traditional medicine in Korea, Japan, and China, has been known as an elixir of life having therapeutic potential. However, there is no scientific evidence to support that Ashitaba can enhance or maintain muscle strength. To find a new therapeutic agent from the medicinal plant, we evaluated the anti-myopathy effect of chalcones from ethanol extract of AK (EAK) in cellular and animal models of muscle atrophy. To examine anti-myopathy activity, EAK was treated into dexamethasone injected rats and muscle thickness and histopathological images were analyzed. Oral administration of EAK (250 or 500 mg/kg) alleviated muscle atrophic damages and down-regulated the mRNA levels of muscle-specific ubiquitin-E3 ligases. Among ten compounds isolated from EAK, 4-hydroxyderricin was the most effective principle in stimulating myogenesis of C2C12 myoblasts via activation of p38 mitogen-activated protein kinase (MAPK). In three cellular muscle atrophy models with C2C12 myoblasts damaged by dexamethasone or cancer cell-conditioned medium, 4-hydroxyderricin protected the myosin heavy chain (MHC) degradation through suppressing expressions of MAFbx, MuRF-1 and myostatin. These results suggest that the ethanol extract and its active principle, 4-hydroxyderricin from AK, can overcome the muscle atrophy through double mechanisms of decreasing muscle protein degradation and activating myoblast differentiation.
Collapse
|
11
|
Binó L, Veselá I, Papežíková I, Procházková J, Vašíček O, Štefková K, Kučera J, Hanáčková M, Kubala L, Pacherník J. The depletion of p38alpha kinase upregulates NADPH oxidase 2/NOX2/gp91 expression and the production of superoxide in mouse embryonic stem cells. Arch Biochem Biophys 2019; 671:18-26. [PMID: 31176685 DOI: 10.1016/j.abb.2019.06.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2018] [Revised: 05/31/2019] [Accepted: 06/05/2019] [Indexed: 01/04/2023]
Abstract
P38alpha kinase plays an important role in the regulation of both cell stress response and cell fate. In this study, we report that p38alpha kinase-deficient embryonic stem cells exhibit a higher production of reactive oxygen species (ROS) in contrast to their wild-type counterpart. Analysis of the expressions of NADPH oxidases (NOXs) and dual oxidases, crucial enzymes involved in intracellular ROS formation, shows NOX2/gp91phox is over-expressed in p38alpha deficient cells. The particular increase in superoxide formation was confirmed by the specific detection of hydroethidine derivate 2-hydroxyethidium. ROS formation decreased when the level of NOX2 was silenced by siRNA in p38alpha deficient cells. These data suggest the importance of p38alpha kinase in the regulation of ROS metabolism in embryonic stem cells and the significance of the observed phenomena of cancer cell-like phenotypes, which is discussed.
Collapse
Affiliation(s)
- Lucia Binó
- Institute of Experimental Biology, Faculty of Science, Masaryk University, Brno, Czech Republic; Department of Free Radical Pathophysiology, Institute of Biophysics, Academy of Sciences of the Czech Republic, Brno, Czech Republic.
| | - Iva Veselá
- Institute of Experimental Biology, Faculty of Science, Masaryk University, Brno, Czech Republic.
| | - Iva Papežíková
- Department of Free Radical Pathophysiology, Institute of Biophysics, Academy of Sciences of the Czech Republic, Brno, Czech Republic.
| | - Jiřina Procházková
- Department of Cytokinetics, Institute of Biophysics, Academy of Sciences of the Czech Republic, Brno, Czech Republic.
| | - Ondřej Vašíček
- Department of Free Radical Pathophysiology, Institute of Biophysics, Academy of Sciences of the Czech Republic, Brno, Czech Republic.
| | - Kateřina Štefková
- Institute of Experimental Biology, Faculty of Science, Masaryk University, Brno, Czech Republic.
| | - Jan Kučera
- Institute of Experimental Biology, Faculty of Science, Masaryk University, Brno, Czech Republic.
| | - Markéta Hanáčková
- Institute of Experimental Biology, Faculty of Science, Masaryk University, Brno, Czech Republic.
| | - Lukáš Kubala
- Department of Free Radical Pathophysiology, Institute of Biophysics, Academy of Sciences of the Czech Republic, Brno, Czech Republic.
| | - Jiří Pacherník
- Institute of Experimental Biology, Faculty of Science, Masaryk University, Brno, Czech Republic.
| |
Collapse
|
12
|
Kwon MJ, Lee YJ, Jung HS, Shin HM, Kim TN, Lee SH, Rhee BD, Kim MK, Park JH. The direct effect of lobeglitazone, a new thiazolidinedione, on pancreatic beta cells: A comparison with other thiazolidinediones. Diabetes Res Clin Pract 2019; 151:209-223. [PMID: 30954516 DOI: 10.1016/j.diabres.2019.04.006] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Revised: 03/14/2019] [Accepted: 04/01/2019] [Indexed: 12/11/2022]
Abstract
AIMS The direct effects of thiazolidinediones (TZDs) on pancreatic beta cells have been controversial. The aim of this study was to find out whether a novel TZD, lobeglitazone, has beneficial effects on pancreatic beta cells and db/db mice compared to those of other TZDs. METHODS INS-1 cells were incubated at a high-glucose concentration with various concentrations of troglitazone, rosiglitazone, pioglitazone, and lobeglitazone. Apoptosis and proliferation of beta cells, markers for ER stress and glucose-stimulated insulin secretion (GSIS) were assessed. In addition, C57BL/6 db/db mice were treated with pioglitazone or lobeglitazone for 4 weeks, and metabolic parameters and the configuration of pancreatic islets were also examined. RESULTS Lobeglitazone and other TZDs decreased INS-1 cell apoptosis in high-glucose conditions. Lobeglitazone and other TZDs significantly decreased hyperglycemia-induced increases in ER stress markers and increased GSIS. Metabolic parameters showed greater improvement in db/db mice treated with pioglitazone and lobeglitazone than in control mice. Islet size, cell proliferation, and beta cell mass were increased, and collagen surrounding the islets was decreased in treated mice. CONCLUSIONS Lobeglitazone showed beneficial effects on beta cell survival and function against hyperglycemia. The prosurvival and profunction effects of lobeglitazone were comparable to those of other TZDs.
Collapse
Affiliation(s)
- Min Jeong Kwon
- Division of Endocrinology and Metabolism, Department of Internal Medicine, College of Medicine, Inje University, Busan, Republic of Korea; Paik Institute for Clinical Research, Molecular Therapy Lab, Inje University, Busan, Republic of Korea
| | - Yong Jae Lee
- CKD Research Institute, Yongin, Gyeonggi-do, Republic of Korea
| | - Hye Sook Jung
- Paik Institute for Clinical Research, Molecular Therapy Lab, Inje University, Busan, Republic of Korea
| | - Hyun Mi Shin
- Paik Institute for Clinical Research, Molecular Therapy Lab, Inje University, Busan, Republic of Korea
| | - Tae Nyun Kim
- Division of Endocrinology and Metabolism, Department of Internal Medicine, College of Medicine, Inje University, Busan, Republic of Korea
| | - Soon Hee Lee
- Division of Endocrinology and Metabolism, Department of Internal Medicine, College of Medicine, Inje University, Busan, Republic of Korea
| | - Byoung Doo Rhee
- Division of Endocrinology and Metabolism, Department of Internal Medicine, College of Medicine, Inje University, Busan, Republic of Korea
| | - Mi-Kyung Kim
- Division of Endocrinology and Metabolism, Department of Internal Medicine, College of Medicine, Inje University, Busan, Republic of Korea; Paik Institute for Clinical Research, Molecular Therapy Lab, Inje University, Busan, Republic of Korea.
| | - Jeong Hyun Park
- Division of Endocrinology and Metabolism, Department of Internal Medicine, College of Medicine, Inje University, Busan, Republic of Korea; Paik Institute for Clinical Research, Molecular Therapy Lab, Inje University, Busan, Republic of Korea.
| |
Collapse
|
13
|
Rosina M, Langone F, Giuliani G, Cerquone Perpetuini A, Reggio A, Calderone A, Fuoco C, Castagnoli L, Gargioli C, Cesareni G. Osteogenic differentiation of skeletal muscle progenitor cells is activated by the DNA damage response. Sci Rep 2019; 9:5447. [PMID: 30931986 PMCID: PMC6443689 DOI: 10.1038/s41598-019-41926-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Accepted: 03/19/2019] [Indexed: 12/27/2022] Open
Abstract
Heterotopic ossification (HO) is a pathological condition characterized by the deposition of mineralized tissue in ectopic locations such as the skeletal muscle. The precise cellular origin and molecular mechanisms underlying HO are still debated. In our study we focus on the differentiation of mesoangioblasts (MABs), a population of multipotent skeletal muscle precursors. High-content screening for small molecules that perturb MAB differentiation decisions identified Idoxuridine (IdU), an antiviral and radiotherapy adjuvant, as a molecule that promotes MAB osteogenic differentiation while inhibiting myogenesis. IdU-dependent osteogenesis does not rely on the canonical BMP-2/SMADs osteogenic pathway. At pro-osteogenic conditions IdU induces a mild DNA Damage Response (DDR) that activates ATM and p38 eventually promoting the phosphorylation of the osteogenesis master regulator RUNX2. By interfering with this pathway IdU-induced osteogenesis is severely impaired. Overall, our study suggests that induction of the DDR promotes osteogenesis in muscle resident MABs thereby offering a new mechanism that may be involved in the ectopic deposition of mineralized tissue in the muscle.
Collapse
Affiliation(s)
- M Rosina
- Department of Biology, University of Rome "Tor Vergata", Rome, Italy
| | - F Langone
- Department of Biology, University of Rome "Tor Vergata", Rome, Italy
| | - G Giuliani
- Department of Biology, University of Rome "Tor Vergata", Rome, Italy
| | | | - A Reggio
- Department of Biology, University of Rome "Tor Vergata", Rome, Italy
| | - A Calderone
- Department of Biology, University of Rome "Tor Vergata", Rome, Italy
| | - C Fuoco
- Department of Biology, University of Rome "Tor Vergata", Rome, Italy
| | - L Castagnoli
- Department of Biology, University of Rome "Tor Vergata", Rome, Italy
| | - C Gargioli
- Department of Biology, University of Rome "Tor Vergata", Rome, Italy.
| | - G Cesareni
- Department of Biology, University of Rome "Tor Vergata", Rome, Italy.
- Fondazione Santa Lucia Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Rome, Italy.
| |
Collapse
|
14
|
Boothello RS, Patel NJ, Sharon C, Abdelfadiel EI, Morla S, Brophy DF, Lippman HR, Desai UR, Patel BB. A Unique Nonsaccharide Mimetic of Heparin Hexasaccharide Inhibits Colon Cancer Stem Cells via p38 MAP Kinase Activation. Mol Cancer Ther 2019; 18:51-61. [PMID: 30337351 PMCID: PMC6332501 DOI: 10.1158/1535-7163.mct-18-0104] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2018] [Revised: 07/23/2018] [Accepted: 10/12/2018] [Indexed: 12/13/2022]
Abstract
Targeting of cancer stem cells (CSC) is expected to be a paradigm-shifting approach for the treatment of cancers. Cell surface proteoglycans bearing sulfated glycosaminoglycan (GAG) chains are known to play a critical role in the regulation of stem cell fate. Here, we show for the first time that G2.2, a sulfated nonsaccharide GAG mimetic (NSGM) of heparin hexasaccharide, selectively inhibits colonic CSCs in vivo G2.2-reduced CSCs (CD133+/CXCR4+, Dual hi) induced HT-29 and HCT 116 colon xenografts' growth in a dose-dependent fashion. G2.2 also significantly delayed the growth of colon xenograft further enriched in CSCs following oxaliplatin and 5-fluorouracil treatment compared with vehicle-treated xenograft controls. In fact, G2.2 robustly inhibited CSCs' abundance (measured by levels of CSC markers, e.g., CD133, DCMLK1, LGR5, and LRIG1) and self-renewal (quaternary spheroids) in colon cancer xenografts. Intriguingly, G2.2 selectively induced apoptosis in the Dual hi CSCs in vivo eluding to its CSC targeting effects. More importantly, G2.2 displayed none to minimal toxicity as observed through morphologic and biochemical studies of vital organ functions, blood coagulation profile, and ex vivo analyses of normal intestinal (and bone marrow) progenitor cell growth. Through extensive in vitro, in vivo, and ex vivo mechanistic studies, we showed that G2.2's inhibition of CSC self-renewal was mediated through activation of p38α, uncovering important signaling that can be targeted to deplete CSCs selectively while minimizing host toxicity. Hence, G2.2 represents a first-in-class (NSGM) anticancer agent to reduce colorectal CSCs.
Collapse
Affiliation(s)
- Rio S Boothello
- Division of Hematology and Oncology, Department of Internal Medicine, Massey Cancer Center, Virginia Commonwealth University, Richmond, Virginia
- McGuire VA Medical Center, Richmond, Virginia
| | - Nirmita J Patel
- McGuire VA Medical Center, Richmond, Virginia
- Institute for Structural Biology, Drug Discovery and Development, Virginia Commonwealth University, Richmond, Virginia
- Department of Medicinal Chemistry, Virginia Commonwealth University, Richmond, Virginia
| | | | - Elsamani I Abdelfadiel
- Institute for Structural Biology, Drug Discovery and Development, Virginia Commonwealth University, Richmond, Virginia
| | - Shravan Morla
- Institute for Structural Biology, Drug Discovery and Development, Virginia Commonwealth University, Richmond, Virginia
- Department of Medicinal Chemistry, Virginia Commonwealth University, Richmond, Virginia
| | - Donald F Brophy
- Department of Pharmacotherapy and Outcomes Sciences, Virginia Commonwealth University, Richmond, Virginia
| | | | - Umesh R Desai
- Institute for Structural Biology, Drug Discovery and Development, Virginia Commonwealth University, Richmond, Virginia.
- Department of Medicinal Chemistry, Virginia Commonwealth University, Richmond, Virginia
| | - Bhaumik B Patel
- Division of Hematology and Oncology, Department of Internal Medicine, Massey Cancer Center, Virginia Commonwealth University, Richmond, Virginia.
- McGuire VA Medical Center, Richmond, Virginia
| |
Collapse
|
15
|
Sheller-Miller S, Richardson L, Martin L, Jin J, Menon R. Systematic review of p38 mitogen-activated kinase and its functional role in reproductive tissues. Am J Reprod Immunol 2018; 80:e13047. [PMID: 30178469 PMCID: PMC6261682 DOI: 10.1111/aji.13047] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2018] [Revised: 08/13/2018] [Accepted: 08/13/2018] [Indexed: 12/12/2022] Open
Abstract
Oxidative stress (OS) plays a role in uterine tissue remodeling during pregnancy and parturition. While p38 MAPK is an OS-response kinase, a precise functional role is unknown. Therefore, we conducted a systematic review of literature on p38 MAPK expression, activation, and function in reproductive tissues throughout pregnancy and parturition, published between January 1980 and August 2017, using four electronic databases (Web of Science, PubMed, Medline, and CoCHRANE). We identified 418 reports; 108 were selected for full-text evaluation and 74 were included in final review. p38 MAPK was investigated using feto-maternal primary or immortalized cells, tissue explants, and animal models. Western blot was most commonly used to report phosphorylated (active) p38 MAPK. Human placenta (27), chorioamniotic membranes (14), myometrium (13), decidua (8), and cervix (1) were the studied tissues. p38 MAPK's functions were tissue and gestational age dependent. Isoform specificity was hardly reported. p38 MAPK activity was induced by ROS or proinflammatory cytokines to promote cell signaling linked to cell fate, primed uterus, ripened cervix, and proinflammatory cytokine/chemokine production. In 35 years, reports on p38 MAPK's role during pregnancy and parturition are scarce and current literature is insufficient to provide a comprehensive description of p38 MAPK's mechanistic role during pregnancy and parturition.
Collapse
Affiliation(s)
- Samantha Sheller-Miller
- Division of Maternal-Fetal Medicine & Perinatal Research, Department of Obstetrics & Gynecology, The University of Texas Medical Branch at Galveston, Galveston, Texas
| | - Lauren Richardson
- Division of Maternal-Fetal Medicine & Perinatal Research, Department of Obstetrics & Gynecology, The University of Texas Medical Branch at Galveston, Galveston, Texas
- Department of Neuroscience & Cell Biology, The University of Texas Medical Branch at Galveston, Galveston, Texas
| | - Laura Martin
- Department of Pathology, Botucatu Medical School, São Paulo State University (UNESP), Botucatu, Brazil
| | - Jin Jin
- Department of Gynecology and Obstetrics, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Ramkumar Menon
- Division of Maternal-Fetal Medicine & Perinatal Research, Department of Obstetrics & Gynecology, The University of Texas Medical Branch at Galveston, Galveston, Texas
| |
Collapse
|
16
|
Aisenbrey EA, Bryant SJ. The role of chondroitin sulfate in regulating hypertrophy during MSC chondrogenesis in a cartilage mimetic hydrogel under dynamic loading. Biomaterials 2018; 190-191:51-62. [PMID: 30391802 DOI: 10.1016/j.biomaterials.2018.10.028] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2018] [Revised: 10/18/2018] [Accepted: 10/21/2018] [Indexed: 01/29/2023]
Abstract
Mesenchymal stem cells (MSCs) are promising for cartilage regeneration, but readily undergo terminal differentiation. The aim of this study was two-fold: a) investigate physiochemical cues from a cartilage-mimetic hydrogel under dynamic compressive loading on MSC chondrogenesis and hypertrophy and b) identify whether Smad signaling and p38 MAPK signaling mediate hypertrophy during MSC chondrogenesis. Human MSCs were encapsulated in photoclickable poly(ethylene glycol) hydrogels containing chondroitin sulfate and RGD, cultured under dynamic compressive loading or free swelling for three weeks, and evaluated by qPCR and immunohistochemistry. Loading inhibited hypertrophy in the cartilage-mimetic hydrogel indicated by a reduction in pSmad 1/5/8, Runx2, and collagen X proteins, while maintaining chondrogenesis by pSmad 2/3 and collagen II proteins. Inhibiting pSmad 1/5/8 under free swelling culture significantly reduced collagen X protein, similar to the loading condition. Chondroitin sulfate was necessary for load-inhibited hypertrophy and correlated with enhanced S100A4 expression, which is downstream of the osmotic responsive transcription factor NFAT5. Inhibiting p38 MAPK under loading reduced S100A4 expression, and upregulated Runx2 and collagen X protein. Findings from this study indicate that chondroitin sulfate with dynamic loading create physiochemical cues that support MSC chondrogenesis and attenuate hypertrophy through Smad 1/5/8 inhibition and p38 MAPK upregulation.
Collapse
Affiliation(s)
- Elizabeth A Aisenbrey
- Department of Chemical and Biological Engineering, University of Colorado, Boulder, CO 80309-0596, USA
| | - Stephanie J Bryant
- Department of Chemical and Biological Engineering, University of Colorado, Boulder, CO 80309-0596, USA; BioFrontiers Institute, University of Colorado, Boulder, CO 80309-0596, USA; Material Science and Engineering Program, University of Colorado, Boulder, CO 80309-0596, USA.
| |
Collapse
|
17
|
Srivastava A, Singh S, Pandey A, Kumar D, Rajpurohit CS, Khanna VK, Pant AB. Secretome of Differentiated PC12 Cells Enhances Neuronal Differentiation in Human Mesenchymal Stem Cells Via NGF-Like Mechanism. Mol Neurobiol 2018. [PMID: 29527653 DOI: 10.1007/s12035-018-0981-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The secretome-mediated responses over cellular physiology are well documented. Stem cells have been ruling the field of secretomics and its role in regenerative medicine since the past few years. However, the mechanistic aspects of secretome-mediated responses and the role of other cells in this area remain somewhat elusive. Here, we investigate the effects of secretome-enriched conditioned medium (CM) of neuronally differentiated PC12 cells on the neuronal differentiation of human mesenchymal stem cells (hMSCs). The exposure to CM at a ratio of 1:1 (CM: conditioned medium of PC12 cells) led to neuronal induction in hMSCs. This neuronal induction was compared with a parallel group of cells exposed to nerve growth factor (NGF). There was a marked increase in neurite length and expression of neuronal markers (β-III tubulin, neurofilament-M (NF-M), synaptophysin, NeuN in exposed hMSCs). Experimental group co-exposed to NGF and CM showed an additive response via MAPK signaling and directed the cells particularly towards cholinergic lineage. The ability of CM to enhance the neuronal properties of stem cells could aid in their rapid differentiation into neuronal subtypes in case of stem cell transplantation for neuronal injuries, thus broadening the scope of non-stem cell-based applications in the area of secretomics.
Collapse
Affiliation(s)
- A Srivastava
- System Toxicology and Health Risk Assessment Group, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), MG Marg, Lucknow, Uttar Pradesh, -226001, India
| | - S Singh
- System Toxicology and Health Risk Assessment Group, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), MG Marg, Lucknow, Uttar Pradesh, -226001, India.,Academy of Scientific and Innovative Research, CSIR-IITR Campus, Lucknow, India
| | - A Pandey
- System Toxicology and Health Risk Assessment Group, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), MG Marg, Lucknow, Uttar Pradesh, -226001, India
| | - D Kumar
- System Toxicology and Health Risk Assessment Group, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), MG Marg, Lucknow, Uttar Pradesh, -226001, India.,Academy of Scientific and Innovative Research, CSIR-IITR Campus, Lucknow, India
| | - C S Rajpurohit
- System Toxicology and Health Risk Assessment Group, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), MG Marg, Lucknow, Uttar Pradesh, -226001, India.,Academy of Scientific and Innovative Research, CSIR-IITR Campus, Lucknow, India
| | - V K Khanna
- System Toxicology and Health Risk Assessment Group, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), MG Marg, Lucknow, Uttar Pradesh, -226001, India.,Academy of Scientific and Innovative Research, CSIR-IITR Campus, Lucknow, India
| | - A B Pant
- System Toxicology and Health Risk Assessment Group, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), MG Marg, Lucknow, Uttar Pradesh, -226001, India. .,Academy of Scientific and Innovative Research, CSIR-IITR Campus, Lucknow, India.
| |
Collapse
|
18
|
Pei H, Zhai C, Li H, Yan F, Qin J, Yuan H, Zhang R, Wang S, Zhang W, Chang M, Wang Y, Pei X. Connexin 32 and connexin 43 are involved in lineage restriction of hepatic progenitor cells to hepatocytes. Stem Cell Res Ther 2017; 8:252. [PMID: 29116012 PMCID: PMC5678556 DOI: 10.1186/s13287-017-0703-2] [Citation(s) in RCA: 10] [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: 03/06/2017] [Revised: 09/06/2017] [Accepted: 10/19/2017] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Bi-potential hepatic progenitor cells can give rise to both hepatocytes and cholangiocytes, which is the last phase and critical juncture in terms of sequentially hepatic lineage restriction from any kind of stem cells. If their differentiation can be controlled, it might access to functional hepatocytes to develop pharmaceutical and biotechnology industries as well as cell therapies for end-stage liver diseases. METHODS In this study, we investigated the influence of Cx32 and Cx43 on hepatocyte differentiation of WB-F344 cells by in vitro gain and loss of function analyses. An inhibitor of Cx32 was also used to make further clarification. To reveal p38 MAPK pathway is closely related to Cxs, rats with 70% partial hepatectomy were injected intraperitoneally with a p38 inhibitor, SB203580. Besides, the effects of p38 MAPK pathway on differentiation of hepatoblasts isolated from fetal rat livers were evaluated by addition of SB203580 in culture medium. RESULTS In vitro gain and loss of function analyses showed overexpression of Connexin 32 and knockdown of Connexin 43 promoted hepatocytes differentiation from hepatic progenitor cells. In addition, in vitro and ex vivo research revealed inhibition of p38 mitogen-activated protein kinase pathway can improve hepatocytes differentiation correlating with upregulation of Connexin 32 expression and downregulation of Connexin 43 expression. CONCLUSIONS Here we demonstrate that Connexins play crucial roles in facilitating differentiation of hepatic progenitors. Our work further implicates that regulators of Connexins and their related pathways might provide new insights to improve lineage restriction of stem cells to mature hepatocytes.
Collapse
Affiliation(s)
- Haiyun Pei
- Stem Cell and Regenerative Medicine Lab, Beijing Institute of Transfusion Medicine, Beijing, 100850 China
- South China Institute of Biomedicine, Guangzhou, 510005 China
| | - Chao Zhai
- Stem Cell and Regenerative Medicine Lab, Beijing Institute of Transfusion Medicine, Beijing, 100850 China
| | - Huilin Li
- Stem Cell and Regenerative Medicine Lab, Beijing Institute of Transfusion Medicine, Beijing, 100850 China
| | - Fang Yan
- Tissue Engineering Lab, Beijing Institute of Transfusion Medicine, Beijing, 100850 China
| | - Jinhua Qin
- Stem Cell and Regenerative Medicine Lab, Beijing Institute of Transfusion Medicine, Beijing, 100850 China
- Tissue Engineering Lab, Beijing Institute of Transfusion Medicine, Beijing, 100850 China
- South China Institute of Biomedicine, Guangzhou, 510005 China
| | - Hongfeng Yuan
- Stem Cell and Regenerative Medicine Lab, Beijing Institute of Transfusion Medicine, Beijing, 100850 China
| | - Rui Zhang
- Stem Cell and Regenerative Medicine Lab, Beijing Institute of Transfusion Medicine, Beijing, 100850 China
| | - Shuyong Wang
- Stem Cell and Regenerative Medicine Lab, Beijing Institute of Transfusion Medicine, Beijing, 100850 China
- Tissue Engineering Lab, Beijing Institute of Transfusion Medicine, Beijing, 100850 China
- South China Institute of Biomedicine, Guangzhou, 510005 China
| | - Wencheng Zhang
- Stem Cell and Regenerative Medicine Lab, Beijing Institute of Transfusion Medicine, Beijing, 100850 China
| | - Mingyang Chang
- Tissue Engineering Lab, Beijing Institute of Transfusion Medicine, Beijing, 100850 China
| | - Yunfang Wang
- Tissue Engineering Lab, Beijing Institute of Transfusion Medicine, Beijing, 100850 China
| | - Xuetao Pei
- Stem Cell and Regenerative Medicine Lab, Beijing Institute of Transfusion Medicine, Beijing, 100850 China
- South China Institute of Biomedicine, Guangzhou, 510005 China
| |
Collapse
|
19
|
Gao XL, Zhang M, Tang YL, Liang XH. Cancer cell dormancy: mechanisms and implications of cancer recurrence and metastasis. Onco Targets Ther 2017; 10:5219-5228. [PMID: 29138574 PMCID: PMC5667781 DOI: 10.2147/ott.s140854] [Citation(s) in RCA: 81] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
More recently, disease metastasis and relapse in many cancer patients several years (even some decades) after surgical remission are regarded as tumor dormancy. However, the knowledge of this phenomenon is cripplingly limited. Substantial quantities of reviews have summarized three main potential models that can be put forth to explain such process, including angiogenic dormancy, immunologic dormancy, and cellular dormancy. In this review, newly uncovered mechanisms governing cancer cell dormancy are discussed, with an emphasis on the cross talk between dormant cancer cells and their microenvironments. In addition, potential mechanisms of reactivation of these dormant cells in certain anatomic sites including lymph nodes and bone marrow are discussed. Molecular mechanism of cellular dormancy in head and neck cancer is also involved.
Collapse
Affiliation(s)
- Xiao-Lei Gao
- State Key Laboratory of Oral Diseases.,Department of Oral and Maxillofacial Surgery
| | - Mei Zhang
- State Key Laboratory of Oral Diseases.,Department of Oral Pathology, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, People's Republic of China
| | - Ya-Ling Tang
- State Key Laboratory of Oral Diseases.,Department of Oral Pathology, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, People's Republic of China
| | - Xin-Hua Liang
- State Key Laboratory of Oral Diseases.,Department of Oral and Maxillofacial Surgery
| |
Collapse
|
20
|
Cong Q, Jia H, Li P, Qiu S, Yeh J, Wang Y, Zhang ZL, Ao J, Li B, Liu H. p38α MAPK regulates proliferation and differentiation of osteoclast progenitors and bone remodeling in an aging-dependent manner. Sci Rep 2017; 7:45964. [PMID: 28382965 PMCID: PMC5382695 DOI: 10.1038/srep45964] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2016] [Accepted: 03/07/2017] [Indexed: 02/07/2023] Open
Abstract
Bone mass is determined by the balance between bone formation, carried out by mesenchymal stem cell-derived osteoblasts, and bone resorption, carried out by monocyte-derived osteoclasts. Here we investigated the potential roles of p38 MAPKs, which are activated by growth factors and cytokines including RANKL and BMPs, in osteoclastogenesis and bone resorption by ablating p38α MAPK in LysM+monocytes. p38α deficiency promoted monocyte proliferation but regulated monocyte osteoclastic differentiation in a cell-density dependent manner, with proliferating p38α−/− cultures showing increased differentiation. While young mutant mice showed minor increase in bone mass, 6-month-old mutant mice developed osteoporosis, associated with an increase in osteoclastogenesis and bone resorption and an increase in the pool of monocytes. Moreover, monocyte-specific p38α ablation resulted in a decrease in bone formation and the number of bone marrow mesenchymal stem/stromal cells, likely due to decreased expression of PDGF-AA and BMP2. The expression of PDGF-AA and BMP2 was positively regulated by the p38 MAPK-Creb axis in osteoclasts, with the promoters of PDGF-AA and BMP2 having Creb binding sites. These findings uncovered the molecular mechanisms by which p38α MAPK regulates osteoclastogenesis and coordinates osteoclastogenesis and osteoblastogenesis.
Collapse
Affiliation(s)
- Qian Cong
- Dept. of Osteoporosis and Bone Diseases, Shanghai Key Clinical Center for Metabolic Disease, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, China.,Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Hao Jia
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai 200240, China.,Department of Biochemistry and Molecular cellular Biology Shanghai Jiaotong University, School of Medicine, 280 Chongqing Rd, Shanghai, 200025, China
| | - Ping Li
- Dept. of Osteoporosis and Bone Diseases, Shanghai Key Clinical Center for Metabolic Disease, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, China.,Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Shoutao Qiu
- Dept. of Osteoporosis and Bone Diseases, Shanghai Key Clinical Center for Metabolic Disease, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, China.,Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai 200240, China
| | - James Yeh
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Yibin Wang
- Division of Molecular Medicine, Departments of Anesthesiology, Medicine and Physiology, Molecular Biology Institute, Cardiovascular Research Laboratories, David Geffen School of Medicine, Los Angeles, CA90095, USA
| | - Zhen-Lin Zhang
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Junping Ao
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, China
| | - Baojie Li
- Dept. of Osteoporosis and Bone Diseases, Shanghai Key Clinical Center for Metabolic Disease, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, China.,Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Huijuan Liu
- Dept. of Osteoporosis and Bone Diseases, Shanghai Key Clinical Center for Metabolic Disease, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, China.,Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai 200240, China
| |
Collapse
|
21
|
Papaconstantinou J, Wang CZ, Zhang M, Yang S, Deford J, Bulavin DV, Ansari NH. Attenuation of p38α MAPK stress response signaling delays the in vivo aging of skeletal muscle myofibers and progenitor cells. Aging (Albany NY) 2016; 7:718-33. [PMID: 26423835 PMCID: PMC4600628 DOI: 10.18632/aging.100802] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Functional competence and self-renewal of mammalian skeletal muscle myofibers and progenitor cells declines with age. Progression of the muscle aging phenotype involves the decline of juvenile protective factors i.e., proteins whose beneficial functions translate directly to the quality of life, and self-renewal of progenitor cells. These characteristics occur simultaneously with the age-associated increase of p38α stress response signaling. This suggests that the maintenance of low levels of p38α activity of juvenile tissues may delay or attenuate aging. We used the dominant negative haploinsufficient p38α mouse (DN-p38αAF/+) to demonstrate that in vivo attenuation of p38α activity in the gastrocnemius of the aged mutant delays age-associated processes that include: a) the decline of the juvenile protective factors, BubR1, aldehyde dehydrogenase 1A (ALDH1A1), and aldehyde dehydrogenase 2 (ALDH2); b) attenuated expression of p16Ink4a and p19Arf tumor suppressor genes of the Cdkn2a locus; c) decreased levels of hydroxynonenal protein adducts, expression of COX2 and iNOS; d) decline of the senescent progenitor cell pool level and d) the loss of gastrocnemius muscle mass. We propose that elevated P-p38α activity promotes skeletal muscle aging and that the homeostasis of p38α impacts the maintenance of a beneficial healthspan.
Collapse
Affiliation(s)
- John Papaconstantinou
- The Department of Biochemistry and Molecular Biology, The University of Texas Medical Branch, Galveston, TX 77551-06743, USA
| | - Chen Z Wang
- The Department of Biochemistry and Molecular Biology, The University of Texas Medical Branch, Galveston, TX 77551-06743, USA
| | - Min Zhang
- The Department of Biochemistry and Molecular Biology, The University of Texas Medical Branch, Galveston, TX 77551-06743, USA
| | - San Yang
- The Department of Biochemistry and Molecular Biology, The University of Texas Medical Branch, Galveston, TX 77551-06743, USA
| | - James Deford
- The Department of Biochemistry and Molecular Biology, The University of Texas Medical Branch, Galveston, TX 77551-06743, USA
| | - Dmitry V Bulavin
- Institute for Research on Cancer and Ageing of Nice, INSERM, U1081-UMR CNRS 7284, University of Nice - Sophia Antipolis, Centre Antoine Lacassagne, Nice, France
| | - Naseem H Ansari
- The Department of Biochemistry and Molecular Biology, The University of Texas Medical Branch, Galveston, TX 77551-06743, USA
| |
Collapse
|
22
|
Rossitto M, Philibert P, Poulat F, Boizet-Bonhoure B. Molecular events and signalling pathways of male germ cell differentiation in mouse. Semin Cell Dev Biol 2015; 45:84-93. [PMID: 26454096 DOI: 10.1016/j.semcdb.2015.09.014] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2015] [Accepted: 09/22/2015] [Indexed: 12/15/2022]
Abstract
Germ cells, the precursors of gametes, represent a unique cell lineage that is able to differentiate into spermatozoa or oocytes depending on the chromosomal sex of the organism. In the mammalian embryonic gonad, commitment to oogenesis involves pre-meiotic DNA replication and entry into the first meiotic division; whereas, commitment to spermatogenesis involves inhibition of meiotic initiation, suppression of pluripotency, mitotic arrest and expression of specific markers that will control the development of the male germ cells. The crucial decision made by the germ line to commit to either a male or a female fate has been partially explained by genetic and ex vivo studies in mice which have implicated a complex network of regulatory genes, numerous factors and pathways. Besides the reproductive failure that may follow a deregulation of this complex network, the germ cells may, in view of their proliferative and pluripotent nature, act as precursors of potential malignant transformation and as putative targets for exogenous environmental compounds. Our review summarizes and discusses recent developments that have improved our understanding on how germ cell precursors are committed to a male or a female cell fate in the mouse gonad.
Collapse
Affiliation(s)
- Moïra Rossitto
- Genetic and Development Department, Institute of Human Genetics, CNRS UPR1142, Montpellier, France.
| | - Pascal Philibert
- Genetic and Development Department, Institute of Human Genetics, CNRS UPR1142, Montpellier, France.
| | - Francis Poulat
- Genetic and Development Department, Institute of Human Genetics, CNRS UPR1142, Montpellier, France.
| | - Brigitte Boizet-Bonhoure
- Genetic and Development Department, Institute of Human Genetics, CNRS UPR1142, Montpellier, France.
| |
Collapse
|
23
|
Wu MZ, Chen SF, Nieh S, Benner C, Ger LP, Jan CI, Ma L, Chen CH, Hishida T, Chang HT, Lin YS, Montserrat N, Gascon P, Sancho-Martinez I, Izpisua Belmonte JC. Hypoxia Drives Breast Tumor Malignancy through a TET–TNFα–p38–MAPK Signaling Axis. Cancer Res 2015; 75:3912-24. [DOI: 10.1158/0008-5472.can-14-3208] [Citation(s) in RCA: 90] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2014] [Accepted: 05/31/2015] [Indexed: 11/16/2022]
|
24
|
Xu Y, Guan R, Lei H, Gao Z, Li H, Hui Y, Zhou F, Wang L, Lin G, Xin Z. Implications for Differentiation of Endogenous Stem Cells: Therapeutic Effect from Icariside II on a Rat Model of Postprostatectomy Erectile Dysfunction. Stem Cells Dev 2015; 24:747-55. [PMID: 25365340 DOI: 10.1089/scd.2014.0380] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Affiliation(s)
- Yongde Xu
- Molecular Biology Laboratory of Andrology Center, Peking University First Hospital, Peking University, Beijing, China
| | - Ruili Guan
- Molecular Biology Laboratory of Andrology Center, Peking University First Hospital, Peking University, Beijing, China
| | - Hongen Lei
- Molecular Biology Laboratory of Andrology Center, Peking University First Hospital, Peking University, Beijing, China
| | - Zhezhu Gao
- Molecular Biology Laboratory of Andrology Center, Peking University First Hospital, Peking University, Beijing, China
| | - Huixi Li
- Knuppe Molecular Urology Laboratory, Department of Urology, School of Medicine, University of California, San Francisco, California
| | - Yu Hui
- Department of Urology, First Affiliated Hospital of Soochow University, Suzhou, China
| | - Feng Zhou
- Department of Urology, First Affiliated Hospital of Soochow University, Suzhou, China
| | - Lin Wang
- Knuppe Molecular Urology Laboratory, Department of Urology, School of Medicine, University of California, San Francisco, California
| | - Guiting Lin
- Knuppe Molecular Urology Laboratory, Department of Urology, School of Medicine, University of California, San Francisco, California
| | - Zhongcheng Xin
- Molecular Biology Laboratory of Andrology Center, Peking University First Hospital, Peking University, Beijing, China
| |
Collapse
|
25
|
Wang J, Ye L, Jin M, Wang X. Global analyses of Chromosome 17 and 18 genes of lung telocytes compared with mesenchymal stem cells, fibroblasts, alveolar type II cells, airway epithelial cells, and lymphocytes. Biol Direct 2015; 10:9. [PMID: 25888380 PMCID: PMC4355521 DOI: 10.1186/s13062-015-0042-0] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2014] [Accepted: 02/10/2015] [Indexed: 12/15/2022] Open
Abstract
Background Telocytes (TCs) is an interstitial cell with extremely long and thin telopodes (Tps) with thin segments (podomers) and dilations (podoms) to interact with neighboring cells. TCs have been found in different organs, while there is still a lack of TCs-specific biomarkers to distinguish TCs from the other cells. Results We compared gene expression profiles of murine pulmonary TCs on days 5 (TC5) and days 10 (TC10) with mesenchymal stem cells (MSCs), fibroblasts (Fbs), alveolar type II cells (ATII), airway basal cells (ABCs), proximal airway cells (PACs), CD8+ T cells from bronchial lymph nodes (T-BL), and CD8+ T cells from lungs (T-LL). The chromosome 17 and 18 genes were extracted for further analysis. The TCs-specific genes and functional networks were identified and analyzed by bioinformatics tools. 16 and 10 of TCs-specific genes were up-regulated and 68 and 22 were down-regulated in chromosome 17 and 18, as compared with other cells respectively. Of them, Mapk14 and Trem2 were up-regulated to indicate the biological function of TCs in immune regulation, and up-regulated MCFD2 and down-regulated E4F1 and PDCD2 had an association with tissue homeostasis for TCs. Over-expressed Dpysl3 may promote TCs self-proliferation and cell-cell network forming. Conclusions The differential gene expression in chromosomes 17 and 18 clearly revealed that TCs were the distinctive type of interstitial cells. Our data also indicates that TCs may play a dual role in immune surveillance and immune homoeostasis to keep from immune disorder in acute and chronic pulmonary diseases. TCs also participated in proliferation, differentiation and regeneration. Reviewers This article was reviewed by Qing Kay Li and Dragos Cretoiu. Electronic supplementary material The online version of this article (doi:10.1186/s13062-015-0042-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Jian Wang
- Department of Pulmonary Medicine, Zhongshan Hospital, Shanghai Institute of Clinical Bioinformatics, Fudan University Center for Clinical Bioinformatics, Biomedical Research Center, Fudan University Medical School, Shanghai, China.
| | - Ling Ye
- Department of Pulmonary Medicine, Zhongshan Hospital, Shanghai Institute of Clinical Bioinformatics, Fudan University Center for Clinical Bioinformatics, Biomedical Research Center, Fudan University Medical School, Shanghai, China.
| | - Meiling Jin
- Department of Pulmonary Medicine, Zhongshan Hospital, Shanghai Institute of Clinical Bioinformatics, Fudan University Center for Clinical Bioinformatics, Biomedical Research Center, Fudan University Medical School, Shanghai, China.
| | - Xiangdong Wang
- Department of Pulmonary Medicine, Zhongshan Hospital, Shanghai Institute of Clinical Bioinformatics, Fudan University Center for Clinical Bioinformatics, Biomedical Research Center, Fudan University Medical School, Shanghai, China.
| |
Collapse
|
26
|
Bruserud Ø, Reikvam H, Fredly H, Skavland J, Hagen KM, van Hoang TT, Brenner AK, Kadi A, Astori A, Gjertsen BT, Pendino F. Expression of the potential therapeutic target CXXC5 in primary acute myeloid leukemia cells - high expression is associated with adverse prognosis as well as altered intracellular signaling and transcriptional regulation. Oncotarget 2015; 6:2794-811. [PMID: 25605239 PMCID: PMC4413618 DOI: 10.18632/oncotarget.3056] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2014] [Accepted: 12/21/2014] [Indexed: 12/24/2022] Open
Abstract
The CXXC5 gene encodes a transcriptional activator with a zinc-finger domain, and high expression in human acute myeloid leukemia (AML) cells is associated with adverse prognosis. We now characterized the biological context of CXXC5 expression in primary human AML cells. The global gene expression profile of AML cells derived from 48 consecutive patients was analyzed; cells with high and low CXXC5 expression then showed major differences with regard to extracellular communication and intracellular signaling. We observed significant differences in the phosphorylation status of several intracellular signaling mediators (CREB, PDK1, SRC, STAT1, p38, STAT3, rpS6) that are important for PI3K-Akt-mTOR signaling and/or transcriptional regulation. High CXXC5 expression was also associated with high mRNA expression of several stem cell-associated transcriptional regulators, the strongest associations being with WT1, GATA2, RUNX1, LYL1, DNMT3, SPI1, and MYB. Finally, CXXC5 knockdown in human AML cell lines caused significantly increased expression of the potential tumor suppressor gene TSC22 and genes encoding the growth factor receptor KIT, the cytokine Angiopoietin 1 and the selenium-containing glycoprotein Selenoprotein P. Thus, high CXXC5 expression seems to affect several steps in human leukemogenesis, including intracellular events as well as extracellular communication.
Collapse
MESH Headings
- Adult
- Aged
- Aged, 80 and over
- Biomarkers, Tumor/genetics
- Biomarkers, Tumor/metabolism
- Carrier Proteins/genetics
- Carrier Proteins/metabolism
- Cell Line, Tumor
- DNA-Binding Proteins
- Female
- Gene Expression Profiling
- Gene Expression Regulation, Leukemic
- Humans
- Leukemia, Myeloid, Acute/genetics
- Leukemia, Myeloid, Acute/metabolism
- Male
- Middle Aged
- Phosphorylation
- Primary Cell Culture
- Prognosis
- RNA Interference
- RNA, Messenger/metabolism
- Signal Transduction
- Transcription Factors
- Transcription, Genetic
- Transfection
- Tumor Cells, Cultured
- Up-Regulation
Collapse
Affiliation(s)
- Øystein Bruserud
- Section for Hematology, Department of Clinical Science, University of Bergen, Norway
- Department of Medicine, Haukeland University Hospital, Bergen, Norway
| | - Håkon Reikvam
- Section for Hematology, Department of Clinical Science, University of Bergen, Norway
- Department of Medicine, Haukeland University Hospital, Bergen, Norway
| | - Hanne Fredly
- Section for Hematology, Department of Clinical Science, University of Bergen, Norway
- Department of Medicine, Haukeland University Hospital, Bergen, Norway
| | - Jørn Skavland
- Department of Medicine, Haukeland University Hospital, Bergen, Norway
| | - Karen-Marie Hagen
- Section for Hematology, Department of Clinical Science, University of Bergen, Norway
| | - Tuyen Thy van Hoang
- Section for Hematology, Department of Clinical Science, University of Bergen, Norway
| | - Annette K. Brenner
- Section for Hematology, Department of Clinical Science, University of Bergen, Norway
| | - Amir Kadi
- Inserm, U1016, Institut Cochin, F-75014, Paris, France
- CNRS, UMR8104, F-75014, Paris, France
- Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Audrey Astori
- Inserm, U1016, Institut Cochin, F-75014, Paris, France
- CNRS, UMR8104, F-75014, Paris, France
- Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Bjørn Tore Gjertsen
- Section for Hematology, Department of Clinical Science, University of Bergen, Norway
- Department of Medicine, Haukeland University Hospital, Bergen, Norway
| | - Frederic Pendino
- Department of Molecular Biology, University of Bergen, Bergen, Norway
- Inserm, U1016, Institut Cochin, F-75014, Paris, France
- CNRS, UMR8104, F-75014, Paris, France
- Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| |
Collapse
|
27
|
Long Y, Scheres B, Blilou I. The logic of communication: roles for mobile transcription factors in plants. JOURNAL OF EXPERIMENTAL BOTANY 2015; 66:1133-44. [PMID: 25635110 DOI: 10.1093/jxb/eru548] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/03/2023]
Abstract
Mobile transcription factors play many roles in plant development. Here, we compare the use of mobile transcription factors as signals with some canonical signal transduction processes in prokaryotes and eukaryotes. After an initial survey, we focus on the SHORT-ROOT pathway in Arabidopsis roots to show that, despite the simplicity of the concept of mobile transcription factor signalling, many lines of evidence reveal a surprising complexity in control mechanisms linked to this process. We argue that these controls bestow precision, robustness, and versatility on mobile transcription factor signalling.
Collapse
Affiliation(s)
- Yuchen Long
- Plant Developmental Biology, Plant Sciences, Wageningen University and Research Centre, Droevendaalsesteeg 1, Wageningen 6708PB, The Netherlands
| | - Ben Scheres
- Plant Developmental Biology, Plant Sciences, Wageningen University and Research Centre, Droevendaalsesteeg 1, Wageningen 6708PB, The Netherlands
| | - Ikram Blilou
- Plant Developmental Biology, Plant Sciences, Wageningen University and Research Centre, Droevendaalsesteeg 1, Wageningen 6708PB, The Netherlands.
| |
Collapse
|
28
|
Laco F, Low JL, Seow J, Woo TL, Zhong Q, Seayad J, Liu Z, Wei H, Reuveny S, Elliott DA, Chai CLL, Oh SKW. Cardiomyocyte differentiation of pluripotent stem cells with SB203580 analogues correlates with Wnt pathway CK1 inhibition independent of p38 MAPK signaling. J Mol Cell Cardiol 2014; 80:56-70. [PMID: 25528965 DOI: 10.1016/j.yjmcc.2014.12.003] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/12/2014] [Revised: 11/16/2014] [Accepted: 12/03/2014] [Indexed: 01/22/2023]
Abstract
Differentiation of human pluripotent stem cells as embryoid bodies (EBs) has been achieved previously with p38alfa MAPK inhibitors such as SB203580 with moderate efficiency of 10-15%. We synthesized and screened 42 compounds that are 2,4,5-trisubstituted azole analogues of SB203580 for efficient cardiomyocyte differentiation. Our screen identified novel compounds that have similar cardiac differentiation activity as SB203580. However, the cardiac differentiation did not correlate with p38alfa MAPK inhibition, indicating an alternative mechanism in cardiac differentiation. Upon profiling several 2,4,5-trisubstituted azole compounds against a panel of 97 kinases we identified several off targets, among them casein kinases 1 (CK1). The cardiomyogenic activities of SB203580 and its analogues showed a correlation with post mesoderm Wnt/beta-catenin pathway inhibition of CK1 epsilon and delta. These findings united the mechanism of 2,4,5-trisubstituted azole with the current theory of Wnt/beta-catenin regulated pathway of cardiac differentiation. Consequently an efficient cardiomyocyte protocol was developed with Wnt activator CHIR99021 and 2,4,5-trisubstituted azoles to give high yields of 50-70% cardiomyocytes and a 2-fold increase in growth.
Collapse
Affiliation(s)
- Filip Laco
- Bioprocessing Technology Institute, 20 Biopolis Way, Centros #06-01, Singapore 138668, Singapore
| | - Joo-Leng Low
- Institute of Chemical and Engineering Sciences, 8 Biomedical Grove, Neuros #07-01, Singapore 138665, Singapore
| | - Jasmin Seow
- Bioprocessing Technology Institute, 20 Biopolis Way, Centros #06-01, Singapore 138668, Singapore
| | - Tsung Liang Woo
- Bioprocessing Technology Institute, 20 Biopolis Way, Centros #06-01, Singapore 138668, Singapore
| | - Qixing Zhong
- Department of Pharmacy, Faculty of Science, National University of Singapore, 18 Science Drive 4, Singapore 117543, Singapore
| | - Jayasree Seayad
- Institute of Chemical and Engineering Sciences, 8 Biomedical Grove, Neuros #07-01, Singapore 138665, Singapore
| | - Zhenfeng Liu
- National Heart Research Institute Singapore, National Heart Centre Singapore, Singapore; Cardiovascular & Metabolic Disorders Program, DUKE-NUS Graduate Medical School Singapore, Singapore
| | - Heiming Wei
- National Heart Research Institute Singapore, National Heart Centre Singapore, Singapore
| | - Shaul Reuveny
- Bioprocessing Technology Institute, 20 Biopolis Way, Centros #06-01, Singapore 138668, Singapore
| | - David A Elliott
- Murdoch Childrens Research Institute, The Royal Children's Hospital, Flemington Road, Parkville, Melbourne, Australia
| | - Christina L L Chai
- Institute of Chemical and Engineering Sciences, 8 Biomedical Grove, Neuros #07-01, Singapore 138665, Singapore; Department of Pharmacy, Faculty of Science, National University of Singapore, 18 Science Drive 4, Singapore 117543, Singapore
| | - Steve K W Oh
- Bioprocessing Technology Institute, 20 Biopolis Way, Centros #06-01, Singapore 138668, Singapore.
| |
Collapse
|
29
|
Mantovani C, Terenghi G, Magnaghi V. Senescence in adipose-derived stem cells and its implications in nerve regeneration. Neural Regen Res 2014; 9:10-5. [PMID: 25206738 PMCID: PMC4146315 DOI: 10.4103/1673-5374.125324] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/25/2013] [Indexed: 12/11/2022] Open
Abstract
Adult mesenchymal stem cells, specifically adipose-derived stem cells have self-renewal and multiple differentiation potentials and have shown to be the ideal candidate for therapeutic applications in regenerative medicine, particularly in peripheral nerve regeneration. Adipose-derived stem cells are easily harvested, although they may show the effects of aging, hence their potential in nerve repair may be limited by cellular senescence or donor age. Cellular senescence is a complex process whereby stem cells grow old as consequence of intrinsic events (e.g., DNA damage) or environmental cues (e.g., stressful stimuli or diseases), which determine a permanent growth arrest. Several mechanisms are implicated in stem cell senescence, although no one is exclusive of the others. In this review we report some of the most important factors modulating the senescence process, which can influence adipose-derived stem cell morphology and function, and compromise their clinical application for peripheral nerve regenerative cell therapy.
Collapse
Affiliation(s)
- Cristina Mantovani
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milan, Italy
| | - Giorgio Terenghi
- Blond McIndoe Laboratories, Faculty of Medical and Human Sciences, The University of Manchester, Manchester, UK
| | - Valerio Magnaghi
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milan, Italy
| |
Collapse
|
30
|
Vandomme J, Touil Y, Ostyn P, Olejnik C, Flamenco P, El Machhour R, Segard P, Masselot B, Bailliez Y, Formstecher P, Polakowska R. Insulin-like growth factor 1 receptor and p38 mitogen-activated protein kinase signals inversely regulate signal transducer and activator of transcription 3 activity to control human dental pulp stem cell quiescence, propagation, and differentiation. Stem Cells Dev 2014; 23:839-51. [PMID: 24266654 DOI: 10.1089/scd.2013.0400] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Dental pulp stem cells (DPSCs) remain quiescent until activated in response to severe dental pulp damage. Once activated, they exit quiescence and enter regenerative odontogenesis, producing reparative dentin. The factors and signaling molecules that control the quiescence/activation and commitment to differentiation of human DPSCs are not known. In this study, we determined that the inhibition of insulin-like growth factor 1 receptor (IGF-1R) and p38 mitogen-activated protein kinase (p38 MAPK) signaling commonly activates DPSCs and promotes their exit from the G0 phase of the cell cycle as well as from the pyronin Y(low) stem cell compartment. The inhibition of these two pathways, however, inversely determines DPSC fate. In contrast to p38 MAPK inhibitors, IGF-1R inhibitors enhance dental pulp cell sphere-forming capacity and reduce the cells' colony-forming capacity without inducing cell death. The inverse cellular changes initiated by IGF-1R and p38 MAPK inhibitors were accompanied by inverse changes in the levels of active signal transducer and activator of transcription 3 (STAT3) factor, inactive glycogen synthase kinase 3, and matrix extracellular phosphoglycoprotein, a marker of early odontoblast differentiation. Our data suggest that there is cross talk between the IGF-1R and p38 MAPK signaling pathways in DPSCs and that the signals provided by these pathways converge at STAT3 and inversely regulate its activity to maintain quiescence or to promote self-renewal and differentiation of the cells. We propose a working model that explains the possible interactions between IGF-1R and p38 MAPK at the molecular level and describes the cellular consequences of these interactions. This model may inspire further fundamental study and stimulate research on the clinical applications of DPSC in cellular therapy and tissue regeneration.
Collapse
Affiliation(s)
- Jerome Vandomme
- 1 Inserm U837 Jean-Pierre Aubert Research Center, Institut pour la Recherche sur le Cancer de Lille (IRCL) , Lille, France
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Ishida K, Yuge Y, Hanaoka M, Yasukawa M, Minami Y, Ogawa M, Masumoto KH, Shigeyoshi Y, Saito M, Tsuji T. Gadd45gregulates dental epithelial cell proliferation through p38 MAPK-mediatedp21expression. Genes Cells 2013; 18:660-71. [DOI: 10.1111/gtc.12067] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2012] [Accepted: 04/15/2013] [Indexed: 01/09/2023]
Affiliation(s)
- Kentaro Ishida
- Research Institute for Science and Technology; Tokyo University of Science; Chiba; 278-8510; Japan
| | - Yohei Yuge
- Department of Biological Science and Technology; Graduate School of Industrial Science and Technology; Tokyo University of Science; Chiba; 278-8510; Japan
| | - Mai Hanaoka
- Department of Biological Science and Technology; Graduate School of Industrial Science and Technology; Tokyo University of Science; Chiba; 278-8510; Japan
| | - Masato Yasukawa
- Department of Biological Science and Technology; Graduate School of Industrial Science and Technology; Tokyo University of Science; Chiba; 278-8510; Japan
| | - Yoko Minami
- Department of Biological Science and Technology; Graduate School of Industrial Science and Technology; Tokyo University of Science; Chiba; 278-8510; Japan
| | - Miho Ogawa
- Organ Technologies Inc.; Tokyo; 101-0048; Japan
| | - Ko-hei Masumoto
- Department of Anatomy and Neurobiology; Kiniki University Faculty of Medicine; Osaka; 589-8511; Japan
| | - Yasufumi Shigeyoshi
- Department of Anatomy and Neurobiology; Kiniki University Faculty of Medicine; Osaka; 589-8511; Japan
| | | | | |
Collapse
|
32
|
Steering signal transduction pathway towards cardiac lineage from human pluripotent stem cells: A review. Cell Signal 2013; 25:1096-107. [DOI: 10.1016/j.cellsig.2013.01.027] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2012] [Accepted: 01/25/2013] [Indexed: 10/27/2022]
|