1
|
Jiang H, Zhang Y, Hu J, Wang Z, Li G, Lu Y. An alternative spliced UPF2 transcript in pancreatic inflammatory myofibroblastic tumors. Biochem Biophys Res Commun 2024; 691:149306. [PMID: 38056247 DOI: 10.1016/j.bbrc.2023.149306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Revised: 11/17/2023] [Accepted: 11/20/2023] [Indexed: 12/08/2023]
Abstract
BACKGROUND Inflammatory myofibroblastic tumors (IMTs) are characterized by myofibroblast proliferation and an inflammatory cell infiltrate. Our previous study on IMTs reveals that disrupt NMD pathway causes to lower the threshold for triggering the immune cell infiltration, thereby resulting in inappropriate immune activation. However, myofibroblast differentiation and proliferation is not yet known. METHODS RT-PCR, RT-qPCR, DNA sequence, western bolt, 5'race analysis and site-specific mutagenesis were used in this study. RESULTS Here, an alternative spliced (ALS) UPF2 mRNA skipping exon 2 and 3 and corresponding to the truncated UPF2 protein were found in 2 pancreatic IMTs. We showed that the uORF present in the 5'UTR of UPF2 mRNA is responsible for the translation inhibition, whiles ALS UPF2 is more facilitated to be translated into the truncated UPF2 protein. Several mRNA targets of the NMD were upregulated in IMT samples, indicating that the truncated UPF2 function is strongly perturbed, resulted in disrupted NMD pathway in IMTs. These upregulated NMD targets included cdkn1a expression and the generation of high levels of p21 (waf1/cip1), which may contribute to triggering IMTs. CONCLUSION The disrupt UPFs/NMD pathway may link to molecular alteration associated with differentiation and proliferation for IMTs.
Collapse
Affiliation(s)
- Hui Jiang
- Department of Pathology, First Affiliated Hospital, Changhai Hospital, Naval Medical University, 168 Changhai Road, Shanghai, 200433, China
| | - Yunshuo Zhang
- Department of Pathology, First Affiliated Hospital, Changhai Hospital, Naval Medical University, 168 Changhai Road, Shanghai, 200433, China
| | - Jiayang Hu
- Department of Hepatopancreatobiliary Surgery, First Affiliated Hospital, Changhai Hospital, Naval Medical University, 168 Changhai Road, Shanghai, 200433, China
| | - Zhen Wang
- Department of Hepatopancreatobiliary Surgery, First Affiliated Hospital, Changhai Hospital, Naval Medical University, 168 Changhai Road, Shanghai, 200433, China
| | - Gang Li
- Department of Hepatopancreatobiliary Surgery, First Affiliated Hospital, Changhai Hospital, Naval Medical University, 168 Changhai Road, Shanghai, 200433, China.
| | - Yanjun Lu
- Department of Hepatopancreatobiliary Surgery, First Affiliated Hospital, Changhai Hospital, Naval Medical University, 168 Changhai Road, Shanghai, 200433, China.
| |
Collapse
|
2
|
Qin C, Wang Y, Zhang Y, Zhu Y, Wang Y, Cao F. Transcriptome-wide analysis reveals the molecular mechanisms of cannabinoid type II receptor agonists in cardiac injury induced by chronic psychological stress. Front Genet 2023; 13:1095428. [PMID: 36704356 PMCID: PMC9871316 DOI: 10.3389/fgene.2022.1095428] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Accepted: 12/27/2022] [Indexed: 01/12/2023] Open
Abstract
Background: Growing evidence has supported that chronic psychological stress would cause heart damage, However the mechanisms involved are not clear and effective interventions are insufficient. Cannabinoid type 2 receptor (CB2R) can be a potential treatment for cardiac injury. This study is aimed to investigate the protective mechanism of CB2R agonist against chronic psychological stress-induced cardiac injury. Methods: A mouse chronic psychological stress model was constructed based on a chronic unpredictable stress pattern. Mice were performed a three-week psychological stress procedure, and cardiac tissues of them were collected for whole-transcriptome sequencing. Overlap analysis was performed on differentially expressed mRNAs (DE-mRNAs) and ER stress-related genes (ERSRGs), and bioinformatic methods were used to predict the ceRNA networks and conduct pathway analysis. The expressions of the DE-ERSRGs were validated by RT-qPCR. Results: In the comparison of DE mRNA in Case group, Control group and Treatment group, three groups of ceRNA networks and ceRNA (circ) networks were constructed. The DE-mRNAs were mainly enriched in chromatid-relevant terms and Hematopoietic cell lineage pathway. Additionally, 13 DE-ERSRGs were obtained by the overlap analysis, which were utilized to establish a ceRNA network with 15 nodes and 14 edges and a ceRNA (circ) network with 23 nodes and 28 edges. Furthermore, four DE-ERSRGs (Cdkn1a, Atf3, Fkbp5, Gabarapl1) in the networks were key, which were mainly enriched in response to extracellular stimulus, response to nutrient levels, cellular response to external stimulus, and FoxO signaling pathway. Finally, the RT-qPCR results showed almost consistent expression patterns of 13 DE-ERSRGs between the transcriptome and tissue samples. Conclusion: The findings of this study provide novel insights into the molecular mechanisms of chronic psychological stress-induced cardiac diseases and reveal novel targets for the cardioprotective effects of CB2R agonists.
Collapse
Affiliation(s)
- Cheng Qin
- Department of Cardiology, National Clinical Research Center for Geriatric Diseases and Second Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Yujia Wang
- Department of Cardiology, National Clinical Research Center for Geriatric Diseases and Second Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Yang Zhang
- Department of Cardiology, National Clinical Research Center for Geriatric Diseases and Second Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Yan Zhu
- Nankai University School of Medicine, Nankai University, Tianjin, China
| | - Yabin Wang
- Department of Cardiology, National Clinical Research Center for Geriatric Diseases and Second Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Feng Cao
- Department of Cardiology, National Clinical Research Center for Geriatric Diseases and Second Medical Center of Chinese PLA General Hospital, Beijing, China,Beijing Key Laboratory of Research on Aging and Related Diseases, Beijing, China,*Correspondence: Feng Cao,
| |
Collapse
|
3
|
Ring NAR, Valdivieso K, Grillari J, Redl H, Ogrodnik M. The role of senescence in cellular plasticity: Lessons from regeneration and development and implications for age-related diseases. Dev Cell 2022; 57:1083-1101. [PMID: 35472291 DOI: 10.1016/j.devcel.2022.04.005] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 02/15/2022] [Accepted: 04/01/2022] [Indexed: 12/14/2022]
Abstract
Senescence is a cellular state which involves cell cycle arrest and a proinflammatory phenotype, and it has traditionally been associated with cellular and organismal aging. However, increasing evidence suggests key roles in tissue growth and regrowth, especially during development and regeneration. Conversely, cellular plasticity-the capacity of cells to undergo identity change, including differentiation and dedifferentiation-is associated with development and regeneration but is now being investigated in the context of age-related diseases such as Alzheimer disease. Here, we discuss the paradox of the role for cellular senescence in cellular plasticity: senescence can act as a cell-autonomous barrier and a paracrine driver of plasticity. We provide a conceptual framework for integrating recent data and use the interplay between cellular senescence and plasticity to provide insight into age-related diseases. Finally, we argue that age-related diseases can be better deciphered when senescence is recognized as a core mechanism of regeneration and development.
Collapse
Affiliation(s)
- Nadja Anneliese Ruth Ring
- Ludwig Boltzmann Research Group Senescence and Healing of Wounds, Vienna, Austria; Ludwig Boltzmann Institute for Traumatology, The Research Center in Cooperation with AUVA, Vienna, Austria; Austrian Cluster for Tissue Regeneration, Vienna, Austria
| | - Karla Valdivieso
- Ludwig Boltzmann Research Group Senescence and Healing of Wounds, Vienna, Austria; Ludwig Boltzmann Institute for Traumatology, The Research Center in Cooperation with AUVA, Vienna, Austria; Austrian Cluster for Tissue Regeneration, Vienna, Austria; Institute of Molecular Biotechnology, Department of Biotechnology, University of Natural Resources and Life Sciences, Vienna, Austria
| | - Johannes Grillari
- Ludwig Boltzmann Institute for Traumatology, The Research Center in Cooperation with AUVA, Vienna, Austria; Austrian Cluster for Tissue Regeneration, Vienna, Austria; Institute of Molecular Biotechnology, Department of Biotechnology, University of Natural Resources and Life Sciences, Vienna, Austria
| | - Heinz Redl
- Ludwig Boltzmann Research Group Senescence and Healing of Wounds, Vienna, Austria; Ludwig Boltzmann Institute for Traumatology, The Research Center in Cooperation with AUVA, Vienna, Austria; Austrian Cluster for Tissue Regeneration, Vienna, Austria
| | - Mikolaj Ogrodnik
- Ludwig Boltzmann Research Group Senescence and Healing of Wounds, Vienna, Austria; Ludwig Boltzmann Institute for Traumatology, The Research Center in Cooperation with AUVA, Vienna, Austria; Austrian Cluster for Tissue Regeneration, Vienna, Austria.
| |
Collapse
|
4
|
Mo C, Guo J, Qin J, Zhang X, Sun Y, Wei H, Cao D, Zhang Y, Zhao C, Xiong Y, Zhang Y, Sun Y, Shen L, Yue R. Single-cell transcriptomics of LepR-positive skeletal cells reveals heterogeneous stress-dependent stem and progenitor pools. EMBO J 2022; 41:e108415. [PMID: 34957577 PMCID: PMC8844986 DOI: 10.15252/embj.2021108415] [Citation(s) in RCA: 52] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 11/17/2021] [Accepted: 11/18/2021] [Indexed: 12/31/2022] Open
Abstract
Leptin receptor (LepR)-positive cells are key components of the bone marrow hematopoietic microenvironment, and highly enrich skeletal stem and progenitor cells that maintain homeostasis of the adult skeleton. However, the heterogeneity and lineage hierarchy within this population has been elusive. Using genetic lineage tracing and single-cell RNA sequencing, we found that Lepr-Cre labels most bone marrow stromal cells and osteogenic lineage cells in adult long bones. Integrated analysis of Lepr-Cre-traced cells under homeostatic and stress conditions revealed dynamic changes of the adipogenic, osteogenic, and periosteal lineages. Importantly, we discovered a Notch3+ bone marrow sub-population that is slow-cycling and closely associated with the vasculatures, as well as key transcriptional networks promoting osteo-chondrogenic differentiation. We also identified a Sca-1+ periosteal sub-population with high clonogenic activity but limited osteo-chondrogenic potential. Together, we mapped the transcriptomic landscape of adult LepR+ stem and progenitor cells and uncovered cellular and molecular mechanisms underlying their maintenance and lineage specification.
Collapse
Affiliation(s)
- Chunyang Mo
- Institute for Regenerative MedicineShanghai East HospitalFrontier Science Center for Stem Cell ResearchShanghai Key Laboratory of Signaling and Disease ResearchSchool of Life Sciences and TechnologyTongji UniversityShanghaiChina
| | - Jingxin Guo
- MOE Key Laboratory of Biosystems Homeostasis & Protection and Zhejiang Provincial Key Laboratory for Cancer Molecular Cell Biology, Life Sciences InstituteZhejiang UniversityHangzhouChina
- Department of Orthopedics Surgery2nd Affiliated HospitalSchool of MedicineZhejiang UniversityHangzhouChina
| | - Jiachen Qin
- Institute for Regenerative MedicineShanghai East HospitalFrontier Science Center for Stem Cell ResearchShanghai Key Laboratory of Signaling and Disease ResearchSchool of Life Sciences and TechnologyTongji UniversityShanghaiChina
| | - Xiaoying Zhang
- Institute for Regenerative MedicineShanghai East HospitalFrontier Science Center for Stem Cell ResearchShanghai Key Laboratory of Signaling and Disease ResearchSchool of Life Sciences and TechnologyTongji UniversityShanghaiChina
| | - Yuxi Sun
- Department of CardiologyShanghai Tenth People's HospitalTongji University School of MedicineShanghaiChina
| | - Hanjing Wei
- Institute for Regenerative MedicineShanghai East HospitalFrontier Science Center for Stem Cell ResearchShanghai Key Laboratory of Signaling and Disease ResearchSchool of Life Sciences and TechnologyTongji UniversityShanghaiChina
| | - Dandan Cao
- Institute for Regenerative MedicineShanghai East HospitalFrontier Science Center for Stem Cell ResearchShanghai Key Laboratory of Signaling and Disease ResearchSchool of Life Sciences and TechnologyTongji UniversityShanghaiChina
| | - Yiying Zhang
- Institute for Regenerative MedicineShanghai East HospitalFrontier Science Center for Stem Cell ResearchShanghai Key Laboratory of Signaling and Disease ResearchSchool of Life Sciences and TechnologyTongji UniversityShanghaiChina
| | - Chengchen Zhao
- Institute for Regenerative MedicineShanghai East HospitalFrontier Science Center for Stem Cell ResearchShanghai Key Laboratory of Signaling and Disease ResearchSchool of Life Sciences and TechnologyTongji UniversityShanghaiChina
| | - Yanhong Xiong
- Institute for Regenerative MedicineShanghai East HospitalFrontier Science Center for Stem Cell ResearchShanghai Key Laboratory of Signaling and Disease ResearchSchool of Life Sciences and TechnologyTongji UniversityShanghaiChina
| | - Yong Zhang
- Institute for Regenerative MedicineShanghai East HospitalFrontier Science Center for Stem Cell ResearchShanghai Key Laboratory of Signaling and Disease ResearchSchool of Life Sciences and TechnologyTongji UniversityShanghaiChina
| | - Yao Sun
- Department of ImplantologySchool & Hospital of StomatologyShanghai Engineering Research Center of Tooth Restoration and RegenerationTongji UniversityShanghaiChina
| | - Li Shen
- MOE Key Laboratory of Biosystems Homeostasis & Protection and Zhejiang Provincial Key Laboratory for Cancer Molecular Cell Biology, Life Sciences InstituteZhejiang UniversityHangzhouChina
- Department of Orthopedics Surgery2nd Affiliated HospitalSchool of MedicineZhejiang UniversityHangzhouChina
- Hangzhou Innovation CenterZhejiang UniversityHangzhouChina
| | - Rui Yue
- Institute for Regenerative MedicineShanghai East HospitalFrontier Science Center for Stem Cell ResearchShanghai Key Laboratory of Signaling and Disease ResearchSchool of Life Sciences and TechnologyTongji UniversityShanghaiChina
- Shanghai Institute of Stem Cell Research and Clinical TranslationShanghaiChina
| |
Collapse
|
5
|
Czibik G, Mezdari Z, Murat Altintas D, Bréhat J, Pini M, d'Humières T, Delmont T, Radu C, Breau M, Liang H, Martel C, Abatan A, Sarwar R, Marion O, Naushad S, Zhang Y, Halfaoui M, Suffee N, Morin D, Adnot S, Hatem S, Yavari A, Sawaki D, Derumeaux G. Dysregulated Phenylalanine Catabolism Plays a Key Role in the Trajectory of Cardiac Aging. Circulation 2021; 144:559-574. [PMID: 34162223 DOI: 10.1161/circulationaha.121.054204] [Citation(s) in RCA: 62] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Aging myocardium undergoes progressive cardiac hypertrophy and interstitial fibrosis with diastolic and systolic dysfunction. Recent metabolomics studies shed light on amino acids in aging. The present study aimed to dissect how aging leads to elevated plasma levels of the essential amino acid phenylalanine and how it may promote age-related cardiac dysfunction. METHODS We studied cardiac structure and function, together with phenylalanine catabolism in wild-type (WT) and p21-/- mice (male; 2-24 months), with the latter known to be protected from cellular senescence. To explore phenylalanine's effects on cellular senescence and ectopic phenylalanine catabolism, we treated cardiomyocytes (primary adult rat or human AC-16) with phenylalanine. To establish a role for phenylalanine in driving cardiac aging, WT male mice were treated twice a day with phenylalanine (200 mg/kg) for a month. We also treated aged WT mice with tetrahydrobiopterin (10 mg/kg), the essential cofactor for the phenylalanine-degrading enzyme PAH (phenylalanine hydroxylase), or restricted dietary phenylalanine intake. The impact of senescence on hepatic phenylalanine catabolism was explored in vitro in AML12 hepatocytes treated with Nutlin3a (a p53 activator), with or without p21-targeting small interfering RNA or tetrahydrobiopterin, with quantification of PAH and tyrosine levels. RESULTS Natural aging is associated with a progressive increase in plasma phenylalanine levels concomitant with cardiac dysfunction, whereas p21 deletion delayed these changes. Phenylalanine treatment induced premature cardiac deterioration in young WT mice, strikingly akin to that occurring with aging, while triggering cellular senescence, redox, and epigenetic changes. Pharmacological restoration of phenylalanine catabolism with tetrahydrobiopterin administration or dietary phenylalanine restriction abrogated the rise in plasma phenylalanine and reversed cardiac senescent alterations in aged WT mice. Observations from aged mice and human samples implicated age-related decline in hepatic phenylalanine catabolism as a key driver of elevated plasma phenylalanine levels and showed increased myocardial PAH-mediated phenylalanine catabolism, a novel signature of cardiac aging. CONCLUSIONS Our findings establish a pathogenic role for increased phenylalanine levels in cardiac aging, linking plasma phenylalanine levels to cardiac senescence via dysregulated phenylalanine catabolism along a hepatic-cardiac axis. They highlight phenylalanine/PAH modulation as a potential therapeutic strategy for age-associated cardiac impairment.
Collapse
Affiliation(s)
- Gabor Czibik
- INSERM (L'Institut National de la Santé et de la Recherche Médicale) U955 (G.C., Z.M., D.M.A., J.B., M.P., T.d'H., T.D., C.R., M.B., H.L., A.A., O.M., S.N., Y.Z., M.H., D.M., S.A., D.S., G.D.), Université Paris-Est Créteil, France
- Department of Physiology (G.C., T.d'H., S.A., G.D.), AP-HP, Henri Mondor Hospital, FHU-SENEC, Créteil, France
| | - Zaineb Mezdari
- INSERM (L'Institut National de la Santé et de la Recherche Médicale) U955 (G.C., Z.M., D.M.A., J.B., M.P., T.d'H., T.D., C.R., M.B., H.L., A.A., O.M., S.N., Y.Z., M.H., D.M., S.A., D.S., G.D.), Université Paris-Est Créteil, France
| | - Dogus Murat Altintas
- INSERM (L'Institut National de la Santé et de la Recherche Médicale) U955 (G.C., Z.M., D.M.A., J.B., M.P., T.d'H., T.D., C.R., M.B., H.L., A.A., O.M., S.N., Y.Z., M.H., D.M., S.A., D.S., G.D.), Université Paris-Est Créteil, France
| | - Juliette Bréhat
- INSERM (L'Institut National de la Santé et de la Recherche Médicale) U955 (G.C., Z.M., D.M.A., J.B., M.P., T.d'H., T.D., C.R., M.B., H.L., A.A., O.M., S.N., Y.Z., M.H., D.M., S.A., D.S., G.D.), Université Paris-Est Créteil, France
| | - Maria Pini
- INSERM (L'Institut National de la Santé et de la Recherche Médicale) U955 (G.C., Z.M., D.M.A., J.B., M.P., T.d'H., T.D., C.R., M.B., H.L., A.A., O.M., S.N., Y.Z., M.H., D.M., S.A., D.S., G.D.), Université Paris-Est Créteil, France
| | - Thomas d'Humières
- INSERM (L'Institut National de la Santé et de la Recherche Médicale) U955 (G.C., Z.M., D.M.A., J.B., M.P., T.d'H., T.D., C.R., M.B., H.L., A.A., O.M., S.N., Y.Z., M.H., D.M., S.A., D.S., G.D.), Université Paris-Est Créteil, France
- Department of Physiology (G.C., T.d'H., S.A., G.D.), AP-HP, Henri Mondor Hospital, FHU-SENEC, Créteil, France
| | - Thaïs Delmont
- INSERM (L'Institut National de la Santé et de la Recherche Médicale) U955 (G.C., Z.M., D.M.A., J.B., M.P., T.d'H., T.D., C.R., M.B., H.L., A.A., O.M., S.N., Y.Z., M.H., D.M., S.A., D.S., G.D.), Université Paris-Est Créteil, France
| | - Costin Radu
- INSERM (L'Institut National de la Santé et de la Recherche Médicale) U955 (G.C., Z.M., D.M.A., J.B., M.P., T.d'H., T.D., C.R., M.B., H.L., A.A., O.M., S.N., Y.Z., M.H., D.M., S.A., D.S., G.D.), Université Paris-Est Créteil, France
- Department of Cardiac Surgery (C.R.), AP-HP, Henri Mondor Hospital, FHU-SENEC, Créteil, France
| | - Marielle Breau
- INSERM (L'Institut National de la Santé et de la Recherche Médicale) U955 (G.C., Z.M., D.M.A., J.B., M.P., T.d'H., T.D., C.R., M.B., H.L., A.A., O.M., S.N., Y.Z., M.H., D.M., S.A., D.S., G.D.), Université Paris-Est Créteil, France
| | - Hao Liang
- INSERM (L'Institut National de la Santé et de la Recherche Médicale) U955 (G.C., Z.M., D.M.A., J.B., M.P., T.d'H., T.D., C.R., M.B., H.L., A.A., O.M., S.N., Y.Z., M.H., D.M., S.A., D.S., G.D.), Université Paris-Est Créteil, France
| | - Cecile Martel
- Mitologics SAS (C.M.), Université Paris-Est Créteil, France
| | - Azania Abatan
- INSERM (L'Institut National de la Santé et de la Recherche Médicale) U955 (G.C., Z.M., D.M.A., J.B., M.P., T.d'H., T.D., C.R., M.B., H.L., A.A., O.M., S.N., Y.Z., M.H., D.M., S.A., D.S., G.D.), Université Paris-Est Créteil, France
| | - Rizwan Sarwar
- Experimental Therapeutics, Radcliffe Department of Medicine (R.S., A.Y.), University of Oxford, United Kingdom
| | - Ophélie Marion
- INSERM (L'Institut National de la Santé et de la Recherche Médicale) U955 (G.C., Z.M., D.M.A., J.B., M.P., T.d'H., T.D., C.R., M.B., H.L., A.A., O.M., S.N., Y.Z., M.H., D.M., S.A., D.S., G.D.), Université Paris-Est Créteil, France
| | - Suzain Naushad
- INSERM (L'Institut National de la Santé et de la Recherche Médicale) U955 (G.C., Z.M., D.M.A., J.B., M.P., T.d'H., T.D., C.R., M.B., H.L., A.A., O.M., S.N., Y.Z., M.H., D.M., S.A., D.S., G.D.), Université Paris-Est Créteil, France
| | - Yanyan Zhang
- INSERM (L'Institut National de la Santé et de la Recherche Médicale) U955 (G.C., Z.M., D.M.A., J.B., M.P., T.d'H., T.D., C.R., M.B., H.L., A.A., O.M., S.N., Y.Z., M.H., D.M., S.A., D.S., G.D.), Université Paris-Est Créteil, France
| | - Maissa Halfaoui
- INSERM (L'Institut National de la Santé et de la Recherche Médicale) U955 (G.C., Z.M., D.M.A., J.B., M.P., T.d'H., T.D., C.R., M.B., H.L., A.A., O.M., S.N., Y.Z., M.H., D.M., S.A., D.S., G.D.), Université Paris-Est Créteil, France
| | - Nadine Suffee
- Sorbonne Universités, INSERM UMR_S1166, Faculté de Médecine UPMC, Paris, France (N.S., S.H.)
- Institute of Cardiometabolism and Nutrition, ICAN, Paris, France (N.S., S.H.)
| | - Didier Morin
- INSERM (L'Institut National de la Santé et de la Recherche Médicale) U955 (G.C., Z.M., D.M.A., J.B., M.P., T.d'H., T.D., C.R., M.B., H.L., A.A., O.M., S.N., Y.Z., M.H., D.M., S.A., D.S., G.D.), Université Paris-Est Créteil, France
| | - Serge Adnot
- INSERM (L'Institut National de la Santé et de la Recherche Médicale) U955 (G.C., Z.M., D.M.A., J.B., M.P., T.d'H., T.D., C.R., M.B., H.L., A.A., O.M., S.N., Y.Z., M.H., D.M., S.A., D.S., G.D.), Université Paris-Est Créteil, France
- Department of Physiology (G.C., T.d'H., S.A., G.D.), AP-HP, Henri Mondor Hospital, FHU-SENEC, Créteil, France
| | - Stéphane Hatem
- Sorbonne Universités, INSERM UMR_S1166, Faculté de Médecine UPMC, Paris, France (N.S., S.H.)
- Institute of Cardiometabolism and Nutrition, ICAN, Paris, France (N.S., S.H.)
| | - Arash Yavari
- Experimental Therapeutics, Radcliffe Department of Medicine (R.S., A.Y.), University of Oxford, United Kingdom
- Wellcome Centre for Human Genetics (A.Y.), University of Oxford, United Kingdom
| | - Daigo Sawaki
- INSERM (L'Institut National de la Santé et de la Recherche Médicale) U955 (G.C., Z.M., D.M.A., J.B., M.P., T.d'H., T.D., C.R., M.B., H.L., A.A., O.M., S.N., Y.Z., M.H., D.M., S.A., D.S., G.D.), Université Paris-Est Créteil, France
| | - Geneviève Derumeaux
- INSERM (L'Institut National de la Santé et de la Recherche Médicale) U955 (G.C., Z.M., D.M.A., J.B., M.P., T.d'H., T.D., C.R., M.B., H.L., A.A., O.M., S.N., Y.Z., M.H., D.M., S.A., D.S., G.D.), Université Paris-Est Créteil, France
- Department of Physiology (G.C., T.d'H., S.A., G.D.), AP-HP, Henri Mondor Hospital, FHU-SENEC, Créteil, France
| |
Collapse
|
6
|
Jiang D, de Vries JC, Muschhammer J, Schatz S, Ye H, Hein T, Fidan M, Romanov VS, Rinkevich Y, Scharffetter-Kochanek K. Local and transient inhibition of p21 expression ameliorates age-related delayed wound healing. Wound Repair Regen 2019; 28:49-60. [PMID: 31571377 DOI: 10.1111/wrr.12763] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Revised: 08/27/2019] [Accepted: 09/13/2019] [Indexed: 01/09/2023]
Abstract
Nonhealing chronic wounds in the constantly growing elderly population represent a major public health problem with high socioeconomic burden. Yet, the underlying mechanism of age-related impairment of wound healing remains elusive. Here, we show that the number of dermal cells expressing cyclin-dependent kinase inhibitor p21 was elevated upon skin injury, particularly in aged population, in both man and mouse. The nuclear expression of p21 in activated wound fibroblasts delayed the onset of the proliferation phase of wound healing in a p53-independent manner. Further, the local and transient inhibition of p21 expression by in vivo delivered p21-targeting siRNA ameliorated the delayed wound healing in aged mice. Our results suggest that the increased number of p21+ wound fibroblasts enforces the age-related compromised healing, and targeting p21 creates potential clinical avenues to promote wound healing in aged population.
Collapse
Affiliation(s)
- Dongsheng Jiang
- Department of Dermatology and Allergic Diseases, University of Ulm, Ulm, Germany.,Comprehensive Pneumology Center, Institute of Lung Biology and Disease, Helmholtz Zentrum München, Munich, Germany
| | - Juliane C de Vries
- Department of Dermatology and Allergic Diseases, University of Ulm, Ulm, Germany
| | - Jana Muschhammer
- Department of Dermatology and Allergic Diseases, University of Ulm, Ulm, Germany
| | - Susanne Schatz
- Department of Dermatology and Allergic Diseases, University of Ulm, Ulm, Germany
| | - Haifeng Ye
- Comprehensive Pneumology Center, Institute of Lung Biology and Disease, Helmholtz Zentrum München, Munich, Germany
| | - Tabea Hein
- Department of Dermatology and Allergic Diseases, University of Ulm, Ulm, Germany
| | - Miray Fidan
- Department of Dermatology and Allergic Diseases, University of Ulm, Ulm, Germany
| | - Vasily S Romanov
- Leibniz Institute on Aging, Fritz Lipmann Institute (FLI), Jena, Germany
| | - Yuval Rinkevich
- Comprehensive Pneumology Center, Institute of Lung Biology and Disease, Helmholtz Zentrum München, Munich, Germany
| | | |
Collapse
|
7
|
Wang S, He F, Li Z, Hu Y, Huangfu N, Chen X. YB1 protects cardiac myocytes against H2O2‑induced injury via suppression of PIAS3 mRNA and phosphorylation of STAT3. Mol Med Rep 2019; 19:4579-4588. [PMID: 30942400 PMCID: PMC6522804 DOI: 10.3892/mmr.2019.10119] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2018] [Accepted: 03/06/2019] [Indexed: 12/11/2022] Open
Abstract
Oxidative stress serves important roles in cardiac injury during the process of ischemia/reperfusion (I/R). Y-box protein 1 (YB1), a member of the highly conserved Y-box protein family, is closely associated with inflammation and stress responses by regulating gene transcription, RNA splicing and mRNA translation. However, the roles of YB1 in oxidative stress-induced myocardial-I/R (M-I/R) injury are unknown. The aim of the present study was to examine the effects of YB1 on H2O2-induced cardiomyocyte injury and its underlying mechanism. The results demonstrated that YB1 expression was upregulated during H2O2-induced myocardial injury. YB1 knockdown through transfection of small interfering RNA significantly aggravated cardiac cell apoptosis. Furthermore, YB1 knockdown significantly reversed the H2O2-mediated increase in phosphorylated signal transducer and activator of transcription (STAT)3, but did not affect the phosphorylation of P38, extracellular signal-regulated kinases 1/2, c-Jun N-terminal kinases, P65, Janus kinase 1 and 2 or STAT1. Moreover, protein co-immunoprecipitation and RNA-binding protein immunoprecipitation assays revealed that YB1 interacted with protein inhibitor of activated STAT 3 (PIAS3) mRNA but not its translated protein. YB1 overexpression may have promoted PIAS3 mRNA decay, decreasing PIAS3 protein levels, and therefore increased the levels of phosphorylated STAT3. Finally, YB1 knockdown, mediated by a lentivirus carrying YB1 targeted short hairpin RNA, significantly decreased left ventricle percentage fractional shortening and ejection fraction values, while increasing the infarct sizes in a rat model of M-I/R injury. These results demonstrated for the first time (to the best of our knowledge) that YB1 may protect cardiac myocytes against H2O2 or M-I/R-induced injury by binding to PIAS3 mRNA and resulting in the phosphorylation of STAT3.
Collapse
Affiliation(s)
- Shiqi Wang
- Department of Cardiology, The Affiliated Hospital Ningbo No. 1 Hospital, Zhejiang University, Ningbo, Zhejiang 315000, P.R. China
| | - Fuwei He
- Department of Cardiology, The Affiliated Hospital Ningbo No. 1 Hospital, Zhejiang University, Ningbo, Zhejiang 315000, P.R. China
| | - Zhenwei Li
- Department of Cardiology, The Affiliated Hospital Ningbo No. 1 Hospital, Zhejiang University, Ningbo, Zhejiang 315000, P.R. China
| | - Yewen Hu
- Department of Cardiology, The Affiliated Hospital Ningbo No. 1 Hospital, Zhejiang University, Ningbo, Zhejiang 315000, P.R. China
| | - Ning Huangfu
- Department of Cardiology, The Affiliated Hospital Ningbo No. 1 Hospital, Zhejiang University, Ningbo, Zhejiang 315000, P.R. China
| | - Xiaomin Chen
- Department of Cardiology, The Affiliated Hospital Ningbo No. 1 Hospital, Zhejiang University, Ningbo, Zhejiang 315000, P.R. China
| |
Collapse
|
8
|
Dusane DH, Lochab V, Jones T, Peters CW, Sindeldecker D, Das A, Roy S, Sen CK, Subramaniam VV, Wozniak DJ, Prakash S, Stoodley P. Electroceutical Treatment of Pseudomonas aeruginosa Biofilms. Sci Rep 2019; 9:2008. [PMID: 30765750 PMCID: PMC6375951 DOI: 10.1038/s41598-018-37891-y] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2018] [Accepted: 11/30/2018] [Indexed: 02/08/2023] Open
Abstract
Electroceutical wound dressings, especially those involving current flow with silver based electrodes, show promise for treating biofilm infections. However, their mechanism of action is poorly understood. We have developed an in vitro agar based model using a bioluminescent strain of Pseudomonas aeruginosa to measure loss of activity and killing when direct current was applied. Silver electrodes were overlaid with agar and lawn biofilms grown for 24 h. A 6 V battery with 1 kΩ ballast resistor was used to treat the biofilms for 1 h or 24 h. Loss of bioluminescence and a 4-log reduction in viable cells was achieved over the anode. Scanning electron microscopy showed damaged cells and disrupted biofilm architecture. The antimicrobial activity continued to spread from the anode for at least 2 days, even after turning off the current. Based on possible electrochemical ractions of silver electrodes in chlorine containing medium; pH measurements of the medium post treatment; the time delay between initiation of treatment and observed bactericidal effects; and the presence of chlorotyrosine in the cell lysates, hypochlorous acid is hypothesized to be the chemical agent responsible for the observed (destruction/killing/eradication) of these biofilm forming bacteria. Similar killing was obtained with gels containing only bovine synovial fluid or human serum. These results suggest that our in vitro model could serve as a platform for fundamental studies to explore the effects of electrochemical treatment on biofilms, complementing clinical studies with electroceutical dressings.
Collapse
Affiliation(s)
- Devendra H Dusane
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus, Ohio, 43210, USA
| | - Varun Lochab
- Department of Mechanical and Aerospace Engineering, The Ohio State University, Columbus, Ohio, 43210, USA
| | - Travis Jones
- Department of Mechanical and Aerospace Engineering, The Ohio State University, Columbus, Ohio, 43210, USA
| | - Casey W Peters
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus, Ohio, 43210, USA
| | - Devin Sindeldecker
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus, Ohio, 43210, USA
| | - Amitava Das
- Department of Surgery, IU Health Comprehensive Wound Center, Indiana Center for Regenerative Medicine and Engineering, Indiana University School of Medicine, Indianapolis, IN, 46202, USA.,Comprehensive Wound Center and Department of Surgery, The Ohio State University Wexner Medical Center, Columbus, Ohio, 43210, USA
| | - Sashwati Roy
- Department of Surgery, IU Health Comprehensive Wound Center, Indiana Center for Regenerative Medicine and Engineering, Indiana University School of Medicine, Indianapolis, IN, 46202, USA.,Comprehensive Wound Center and Department of Surgery, The Ohio State University Wexner Medical Center, Columbus, Ohio, 43210, USA
| | - Chandan K Sen
- Department of Surgery, IU Health Comprehensive Wound Center, Indiana Center for Regenerative Medicine and Engineering, Indiana University School of Medicine, Indianapolis, IN, 46202, USA.,Comprehensive Wound Center and Department of Surgery, The Ohio State University Wexner Medical Center, Columbus, Ohio, 43210, USA
| | - Vish V Subramaniam
- Department of Mechanical and Aerospace Engineering, The Ohio State University, Columbus, Ohio, 43210, USA
| | - Daniel J Wozniak
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus, Ohio, 43210, USA
| | - Shaurya Prakash
- Department of Mechanical and Aerospace Engineering, The Ohio State University, Columbus, Ohio, 43210, USA.
| | - Paul Stoodley
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus, Ohio, 43210, USA. .,Department of Orthopaedics, The Ohio State University, Columbus, Ohio, 43210, USA. .,National Centre for Advanced Tribology, Mechanical Engineering, University of Southampton, Southampton, UK.
| |
Collapse
|
9
|
He Y, Ling S, Sun Y, Sheng Z, Chen Z, Pan X, Ma G. DNA methylation regulates α-smooth muscle actin expression during cardiac fibroblast differentiation. J Cell Physiol 2018; 234:7174-7185. [PMID: 30362530 DOI: 10.1002/jcp.27471] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2018] [Accepted: 08/30/2018] [Indexed: 12/12/2022]
Abstract
Cardiac fibroblast (CF) differentiation to myofibroblasts expressing α-smooth muscle actin (α-SMA) plays a key role in cardiac fibrosis. Therefore, a study of the mechanism regulating α-SMA expression is a means to understanding the mechanism of fibroblast differentiation and cardiac fibrosis. Previous studies have shown that DNA methylation is associated with gene expression and is related to the development of tissue fibrosis. However, the mechanisms by which CF differentiation is regulated by DNA methylation remain unclear. Here, we explored the epigenetic regulation of α-SMA expression and its relevance in CF differentiation. In this study, we demonstrated that α-SMA was overexpressed and DNMT1 expression was downregulated in the infarct area after myocardial infarction. Treatment of CFs with transforming growth factor-β1 (TGF-β1 ) in vitro upregulated α-SMA expression via epigenetic modifications. TGF-β1 also inhibited DNMT1 expression and activity during CF differentiation. In addition, α-SMA expression was regulated by DNMT1. Conversely, increasing DNMT1 expression levels rescued the TGF-β1 -induced upregulation of α-SMA expression. Finally, TGF-β1 regulated α-SMA expression by inhibiting the DNMT1-mediated DNA methylation of the α-SMA promoter. Taken together, our research showed that inhibition of the DNMT1-mediated DNA methylation of the α-SMA promoter plays an essential role in CF differentiation. In addition, DNMT1 may be a new target for the prevention and treatment of myocardial fibrosis.
Collapse
Affiliation(s)
- Yanru He
- Department of Cardiology, Zhongda Hospital Affiliated to Southeast University, Nanjing, China.,Department of Cardiology, School of Medicine, Southeast University, Nanjing, China
| | - Sunkai Ling
- Department of Cardiology, School of Medicine, Southeast University, Nanjing, China
| | - Yuning Sun
- Department of Cardiology, Zhongda Hospital Affiliated to Southeast University, Nanjing, China.,Department of Cardiology, School of Medicine, Southeast University, Nanjing, China
| | - Zulong Sheng
- Department of Cardiology, Zhongda Hospital Affiliated to Southeast University, Nanjing, China
| | - Zhongpu Chen
- Department of Cardiology, Zhongda Hospital Affiliated to Southeast University, Nanjing, China
| | - Xiaodong Pan
- Department of Cardiology, Zhongda Hospital Affiliated to Southeast University, Nanjing, China
| | - Genshan Ma
- Department of Cardiology, Zhongda Hospital Affiliated to Southeast University, Nanjing, China
| |
Collapse
|
10
|
Scoparone attenuates angiotensin II-induced extracellular matrix remodeling in cardiac fibroblasts. J Pharmacol Sci 2018; 137:110-115. [DOI: 10.1016/j.jphs.2018.05.006] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2018] [Revised: 04/15/2018] [Accepted: 05/07/2018] [Indexed: 02/08/2023] Open
|
11
|
Zhou HT, Yu XF, Zhou GM. Diosgenin inhibits angiotensin II-induced extracellular matrix remodeling in cardiac fibroblasts through regulating the TGF-β1/Smad3 signaling pathway. Mol Med Rep 2017; 15:2823-2828. [DOI: 10.3892/mmr.2017.6280] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2015] [Accepted: 11/15/2016] [Indexed: 11/05/2022] Open
|
12
|
Das A, Ghatak S, Sinha M, Chaffee S, Ahmed NS, Parinandi NL, Wohleb ES, Sheridan JF, Sen CK, Roy S. Correction of MFG-E8 Resolves Inflammation and Promotes Cutaneous Wound Healing in Diabetes. THE JOURNAL OF IMMUNOLOGY 2016; 196:5089-100. [PMID: 27194784 DOI: 10.4049/jimmunol.1502270] [Citation(s) in RCA: 79] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/23/2015] [Accepted: 04/18/2016] [Indexed: 12/15/2022]
Abstract
Milk fat globule epidermal growth factor-factor 8 (MFG-E8) is a peripheral glycoprotein that acts as a bridging molecule between the macrophage and apoptotic cells, thus executing a pivotal role in the scavenging of apoptotic cells from affected tissue. We have previously reported that apoptotic cell clearance activity or efferocytosis is compromised in diabetic wound macrophages. In this work, we test the hypothesis that MFG-E8 helps resolve inflammation, supports angiogenesis, and accelerates wound closure. MFG-E8(-/-) mice displayed impaired efferocytosis associated with exaggerated inflammatory response, poor angiogenesis, and wound closure. Wound macrophage-derived MFG-E8 was recognized as a critical driver of wound angiogenesis. Transplantation of MFG-E8(-/-) bone marrow to MFG-E8(+/+) mice resulted in impaired wound closure and compromised wound vascularization. In contrast, MFG-E8(-/-) mice that received wild-type bone marrow showed improved wound closure and improved wound vascularization. Hyperglycemia and exposure to advanced glycated end products inactivated MFG-E8, recognizing a key mechanism that complicates diabetic wound healing. Diabetic db/db mice suffered from impaired efferocytosis accompanied with persistent inflammation and slow wound closure. Topical recombinant MFG-E8 induced resolution of wound inflammation, improvements in angiogenesis, and acceleration of closure, upholding the potential of MFG-E8-directed therapeutics in diabetic wound care.
Collapse
Affiliation(s)
- Amitava Das
- Department of Surgery, Ohio State University Wexner Medical Center, Columbus, OH 43210; Davis Heart and Lung Research Institute, Ohio State University Wexner Medical Center, Columbus, OH 43210; Comprehensive Wound Center, Center for Regenerative Medicine and Cell Based Therapies, Ohio State University Wexner Medical Center, Columbus, OH 43210
| | - Subhadip Ghatak
- Department of Surgery, Ohio State University Wexner Medical Center, Columbus, OH 43210; Davis Heart and Lung Research Institute, Ohio State University Wexner Medical Center, Columbus, OH 43210; Comprehensive Wound Center, Center for Regenerative Medicine and Cell Based Therapies, Ohio State University Wexner Medical Center, Columbus, OH 43210
| | - Mithun Sinha
- Department of Surgery, Ohio State University Wexner Medical Center, Columbus, OH 43210; Davis Heart and Lung Research Institute, Ohio State University Wexner Medical Center, Columbus, OH 43210; Comprehensive Wound Center, Center for Regenerative Medicine and Cell Based Therapies, Ohio State University Wexner Medical Center, Columbus, OH 43210
| | - Scott Chaffee
- Department of Surgery, Ohio State University Wexner Medical Center, Columbus, OH 43210; Davis Heart and Lung Research Institute, Ohio State University Wexner Medical Center, Columbus, OH 43210; Comprehensive Wound Center, Center for Regenerative Medicine and Cell Based Therapies, Ohio State University Wexner Medical Center, Columbus, OH 43210
| | - Noha S Ahmed
- Department of Surgery, Ohio State University Wexner Medical Center, Columbus, OH 43210; Davis Heart and Lung Research Institute, Ohio State University Wexner Medical Center, Columbus, OH 43210; Comprehensive Wound Center, Center for Regenerative Medicine and Cell Based Therapies, Ohio State University Wexner Medical Center, Columbus, OH 43210
| | - Narasimham L Parinandi
- Davis Heart and Lung Research Institute, Ohio State University Wexner Medical Center, Columbus, OH 43210; Department of Internal Medicine, Ohio State University Wexner Medical Center, Columbus, OH 43210; and
| | - Eric S Wohleb
- Division of Biosciences, The Ohio State University, Columbus, OH 43210
| | - John F Sheridan
- Division of Biosciences, The Ohio State University, Columbus, OH 43210
| | - Chandan K Sen
- Department of Surgery, Ohio State University Wexner Medical Center, Columbus, OH 43210; Davis Heart and Lung Research Institute, Ohio State University Wexner Medical Center, Columbus, OH 43210; Comprehensive Wound Center, Center for Regenerative Medicine and Cell Based Therapies, Ohio State University Wexner Medical Center, Columbus, OH 43210
| | - Sashwati Roy
- Department of Surgery, Ohio State University Wexner Medical Center, Columbus, OH 43210; Davis Heart and Lung Research Institute, Ohio State University Wexner Medical Center, Columbus, OH 43210; Comprehensive Wound Center, Center for Regenerative Medicine and Cell Based Therapies, Ohio State University Wexner Medical Center, Columbus, OH 43210;
| |
Collapse
|
13
|
Vivar R, Humeres C, Muñoz C, Boza P, Bolivar S, Tapia F, Lavandero S, Chiong M, Diaz-Araya G. FoxO1 mediates TGF-beta1-dependent cardiac myofibroblast differentiation. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2016; 1863:128-38. [DOI: 10.1016/j.bbamcr.2015.10.019] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/08/2015] [Revised: 10/02/2015] [Accepted: 10/26/2015] [Indexed: 12/31/2022]
|
14
|
Dmitrieva RI, Revittser AV, Klukina MA, Sviryaev YV, Korostovtseva LS, Kostareva AA, Zaritskey AY, Shlyakhto EV. Functional properties of bone marrow derived multipotent mesenchymal stromal cells are altered in heart failure patients, and could be corrected by adjustment of expansion strategies. Aging (Albany NY) 2015; 7:14-25. [PMID: 25606985 PMCID: PMC4350322 DOI: 10.18632/aging.100716] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
BACKGROUND Bone marrow multipotent mesenchymal stromal cells (BM-MMSC) considered as a prospective substrate for cell therapy applications, however adult stem cells could be affected by donor-specific factors: age, gender, medical history. Our aim was to investigate how HF affects the functional properties of BM-MMSC. MATERIALS AND METHODS BM-MMSC from 10 healthy donors (HD), and 16 donors with chronic HF were evaluated for proliferative activity, ability to differentiate, replicative senescence, expression of genes that affect regeneration and fibrosis. The effect of culturing conditions on efficiency of BM-MMSC expansion was determined. RESULTS HF-derived BM-MMSC demonstrated early decrease of proliferative activity and upregulation of genes that control both, regeneration and fibrosis: Tgf-β pathway, synthesis of ECM, remodeling enzymes, adhesion molecules. We assume that these effects were related to increase of frequency of myofibroblast-like CD146+/SMAα+ CFU-F in HF samples; (ii) low seeding density and hypoxia resulted in predominant purification and expansion of CD146+/SMAα- CFU-Fs. (iii) the activity of NPs system was downregulated in HF BM-MMSC; CONCLUSIONS downregulation of NP signaling in combination with upregulation of Tgf-β pathway in BM-MMSC would result in pro-fibrotic phenotype and make these cells non-effective for therapeutic applications; the corrections in culturing strategy resulted in 2(3)-2(7) increase of expansion efficiency.
Collapse
Affiliation(s)
| | - Alla V Revittser
- Federal Almazov Medical Research Centre, St. Petersburg, Russia.,St. Petersburg State Polytechnical University, Branch of Medical Physics and Bioengineering, Russia
| | - Maria A Klukina
- Federal Almazov Medical Research Centre, St. Petersburg, Russia
| | - Yuri V Sviryaev
- Federal Almazov Medical Research Centre, St. Petersburg, Russia
| | | | | | | | | |
Collapse
|
15
|
Pereg D, Cohen K, Mosseri M, Berlin T, Steinberg DM, Ellis M, Ashur-Fabian O. Incidence and Expression of Circulating Cell Free p53-Related Genes in Acute Myocardial Infarction Patients. J Atheroscler Thromb 2015; 22:981-98. [PMID: 25958931 DOI: 10.5551/jat.29223] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
AIM The circulating RNA levels are predictive markers in several diseases. We determined the levels of circulating p53-related genes in patients with acute ST-segment elevation myocardial infarction (STEMI), indicating major heart muscle damage. METHODS Plasma RNA was extracted from the patients (n=45) upon their arrival to the hospital (STEMI 0h) and at four hours post-catheterization (STEMI 4h) as well as from controls (n=34). RESULTS Of 18 circulating p53-related genes, nine genes were detectable. A significantly lower incidence of circulating p21 (p < 0.0001), Notch1 (p=0.042) and BTG2 (p < 0.0001) was observed in the STEMI 0h samples in comparison to the STEMI 4h and control samples. Lower expression levels (2.1-fold) of circulating BNIP3L (p=0.011), p21 (3.4-fold, p=0.005) and BTG2 (6.3-fold, p=0.0001) were observed in the STEMI 0h samples in comparison to the STEMI 4h samples, with a 7.4-fold lower BTG2 expression (p < 0.001) and 2.6-fold lower p21 expression (p=0.034) compared to the control samples. Moreover, the BNIP3L expression (borderline significance, p=0.0655) predicted the level of peak troponin, a marker of myocardial infarction. In addition, the BNIP3L levels on admission (p=0.0025), at post-catheterization (p=0.020) and the change between the groups (p=0.0079) were inversely associated with troponin. The BNIP3L (p=0.0139) and p21 levels (p=0.0447) were also associated with a longer time to catheterization. CONCLUSIONS Our results suggest that circulating downstream targets of p53 are inhibited during severe AMI and subsequently re-expressed after catheterization, uncovering possible novel death-or-survival decisions regarding the fate of p53 in the heart and the potential use of its target genes as prognostic biomarkers for oxygenation normalization.
Collapse
|
16
|
Das A, Ganesh K, Khanna S, Sen CK, Roy S. Engulfment of apoptotic cells by macrophages: a role of microRNA-21 in the resolution of wound inflammation. THE JOURNAL OF IMMUNOLOGY 2014; 192:1120-9. [PMID: 24391209 DOI: 10.4049/jimmunol.1300613] [Citation(s) in RCA: 246] [Impact Index Per Article: 22.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
At an injury site, efficient clearance of apoptotic cells by wound macrophages or efferocytosis is a prerequisite for the timely resolution of inflammation. Emerging evidence indicates that microRNA-21 (miR-21) may regulate the inflammatory response. In this work, we sought to elucidate the significance of miR-21 in the regulation of efferocytosis-mediated suppression of innate immune response, a key process implicated in resolving inflammation following injury. An increased expression of inducible miR-21 was noted in postefferocytotic peripheral blood monocyte-derived macrophages. Such induction of miR-21 was associated with silencing of its target genes PTEN and PDCD4. Successful efferocytosis of apoptotic cells by monocyte-derived macrophages resulted in the suppression of LPS-induced NF-κB activation and TNF-α expression. Interestingly, bolstering of miR-21 levels alone, using miR mimic, resulted in significant suppression of LPS-induced TNF-α expression and NF-κB activation. We report that efferocytosis-induced miR-21, by silencing PTEN and GSK3β, tempers the LPS-induced inflammatory response. Macrophage efferocytosis is known to trigger the release of anti-inflammatory cytokine IL-10. This study demonstrates that following successful efferocytosis, miR-21 induction in macrophages silences PDCD4, favoring c-Jun-AP-1 activity, which in turn results in elevated production of anti-inflammatory IL-10. In summary, this work provides direct evidence implicating miRNA in the process of turning on an anti-inflammatory phenotype in the postefferocytotic macrophage. Elevated macrophage miR-21 promotes efferocytosis and silences target genes PTEN and PDCD4, which in turn accounts for a net anti-inflammatory phenotype. Findings of this study highlight the significance of miRs in the resolution of wound inflammation.
Collapse
Affiliation(s)
- Amitava Das
- Department of Surgery, Davis Heart and Lung Research Institute, Center for Regenerative Medicine and Cell Based Therapies and Comprehensive Wound Center, The Ohio State University Wexner Medical Center, Columbus, OH 43210
| | | | | | | | | |
Collapse
|
17
|
Suppression of Induced microRNA-15b Prevents Rapid Loss of Cardiac Function in a Dicer Depleted Model of Cardiac Dysfunction. PLoS One 2013; 8:e66789. [PMID: 23840532 PMCID: PMC3686742 DOI: 10.1371/journal.pone.0066789] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2012] [Accepted: 05/13/2013] [Indexed: 12/22/2022] Open
Abstract
Background Dicer endonuclease, critical for maturation of miRNAs, is depleted in certain forms of cardiomyopathy which results in differential expression of certain microRNAs. We sought to elucidate the mechanisms underlying the rapid loss of cardiac function following cardiac-specific Dicer depletion in adult mice. Results Conditional Dicer deletion in the adult murine myocardium demonstrated compromised heart function, mitochondrial dysfunction and oxidant stress. Elevated miR-15b was observed as an early response to Dicer depletion and was found to silence Pim-1 kinase, a protein responsible for maintaining mitochondrial integrity and function. Anti-miRNA based suppression of induced miRNA-15b rescued the function of Dicer-depleted adult heart and attenuated hypertrophy. Conclusions Anti-miRNA based suppression of inducible miRNA-15b can prevent rapid loss of cardiac function in a Dicer-depleted adult heart and can be a key approach worthy of therapeutic consideration.
Collapse
|
18
|
Li Y, Cai M, Sun Q, Liu Z, Cardounel AJ, Swartz HM, He G. Hyperoxia and transforming growth factor β1 signaling in the post-ischemic mouse heart. Life Sci 2013; 92:547-54. [PMID: 23352974 DOI: 10.1016/j.lfs.2013.01.018] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2012] [Revised: 12/18/2012] [Accepted: 01/08/2013] [Indexed: 11/24/2022]
Abstract
AIMS Following ischemic injury, myocardial healing and remodeling occur with characteristic myofibroblast trans-differentiation and scar formation. The current study tests the hypothesis that hyperoxia and nitric oxide (NO) regulate TGF-β1 signaling in the post-ischemic myocardium. MAIN METHODS C57BL/6 wild-type (WT), endothelial and inducible nitric oxide synthase knockout (eNOS(-/-) and iNOS(-/-)) mice were subjected to 30-min left anterior descending coronary artery occlusion followed by reperfusion. Myocardial tissue oxygenation was monitored with electron paramagnetic resonance oximetry. Protein expressions of TGF-β1, receptor-activated small mothers against decapentaplegic homolog (Smad), p21 and α-smooth muscle actin (α-SMA) were measured with enzyme-linked immunosorbent assay (ELISA), Western immunoblotting, and immunohistochemical staining. KEY FINDINGS There was a hyperoxic state in the post-ischemic myocardial tissue. Protein expressions of total and active TGF-β1, p-Smad2/3 over t-Smad2/3 ratio, p21, and α-SMA were significantly increased in WT mice compared to Sham control. Knockout of eNOS or iNOS further increased protein expression of these signals. The expression of α-SMA was more abundant in the infarct of eNOS(-/-) and iNOS(-/-) mice than WT mice. A protein band indicating nitration of TGF-β type-II receptor (TGFβRII) was observed from WT heart. Carbogen (95% O2 plus 5% CO2) treatment increased the ratio of p-Smad2/t-Smad2, which was inhibited by 10006329 EUK (EUK134) and sodium nitroprusside (SNP). In conclusion, hyperoxia up-regulated and NO/ONOO(-) inhibited cardiac TGF-β1 signaling and myofibroblast trans-differentiation. SIGNIFICANCE These findings may provide new insights in myocardial infarct healing and repair.
Collapse
Affiliation(s)
- Yuanjing Li
- Davis Heart and Lung Research Institute and Division of Cardiovascular Medicine, Department of Internal Medicine, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| | | | | | | | | | | | | |
Collapse
|
19
|
Koga K, Kenessey A, Ojamaa K. Macrophage migration inhibitory factor antagonizes pressure overload-induced cardiac hypertrophy. Am J Physiol Heart Circ Physiol 2012; 304:H282-93. [PMID: 23144312 DOI: 10.1152/ajpheart.00595.2012] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Macrophage migration inhibitory factor (MIF) functions as a proinflammatory cytokine when secreted from the cell, but it also exhibits antioxidant properties by virtue of its intrinsic oxidoreductase activity. Since increased production of ROS is implicated in the development of left ventricular hypertrophy, we hypothesized that the redox activity of MIF protects the myocardium when exposed to hemodynamic stress. In a mouse model of myocardial hypertrophy induced by transverse aortic coarctation (TAC) for 10 days, we showed that growth of the MIF-deficient heart was significantly greater by 32% compared with wild-type (WT) TAC hearts and that fibrosis was increased by fourfold (2.62 ± 0.2% vs. 0.6 ± 0.1%). Circulating MIF was increased in TAC animals, and expression of MIF receptor, CD74, was increased in the hypertrophic myocardium. Gene expression analysis showed a 10-fold increase (P < 0.01) in ROS-generating mitochondrial NADPH oxidase and 2- to 3-fold reductions (P < 0.01) in mitochondrial SOD2 and mitochondrial aconitase activities, indicating enhanced oxidative injury in the hypertrophied MIF-deficient ventricle. Hypertrophic signaling pathways showed that phosphorylation of cytosolic glycogen synthase kinase-3α was greater (P < 0.05) at baseline in MIF-deficient hearts than in WT hearts and remained elevated after 10-day TAC. In the hemodynamically stressed MIF-deficient heart, nuclear p21(CIP1) increased sevenfold (P < 0.01), and the cytosolic increase of phospho-p21(CIP1) was significantly greater than in WT TAC hearts. We conclude that MIF antagonizes myocardial hypertrophy and fibrosis in response to hemodynamic stress by maintaining a redox homeostatic phenotype and attenuating stress-induced activation of hypertrophic signaling pathways.
Collapse
Affiliation(s)
- Kiyokazu Koga
- Center for Heart and Lung Research, The Feinstein Institute for Medical Research, Manhasset, NY 11030, USA
| | | | | |
Collapse
|
20
|
Dickerson R, Deshpande B, Gnyawali U, Lynch D, Gordillo GM, Schuster D, Osei K, Roy S. Correction of aberrant NADPH oxidase activity in blood-derived mononuclear cells from type II diabetes mellitus patients by a naturally fermented papaya preparation. Antioxid Redox Signal 2012; 17:485-91. [PMID: 22369197 PMCID: PMC3365356 DOI: 10.1089/ars.2011.4292] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2011] [Accepted: 02/08/2012] [Indexed: 01/01/2023]
Abstract
UNLABELLED Supplementation of standardized fermented papaya preparation (FPP) to adult diabetic mice improves dermal wound healing outcomes. Peripheral blood mononuclear cells (PBMC) from type II diabetes mellitus (T2DM) patients elicit a compromised respiratory burst activity resulting in increased risk of infections for the diabetic patients. AIMS The objectives of the current study were to determine the effect of FPP supplementation on human diabetic PBMC respiratory burst activity and to understand underlying mechanisms of such action of FPP. RESULTS When stimulated with phorbol 12-myristate 13-acetate, the production of reactive oxygen species by T2DM PBMC was markedly compromised compared to that of the PBMC from non-DM donors. FPP treated ex vivo improved respiratory burst outcomes in T2DM PBMC. FPP treatment significantly increased phosphorylation of the p47phox subunit of NADPH oxidase. In addition, the protein and mRNA expression of Rac2 was potently upregulated after FPP supplemention. The proximal human Rac2 gene promoter is G-C rich and contains consensus binding sites for Sp1 and AP-1. While FPP had no significant effect on the AP-1 DNA binding activity, the Sp1 DNA binding activity was significantly upregulated in PBMC after treatment of the cells with FPP. INNOVATION This work provided first evidence that compromised respiratory burst performance of T2DM PBMC may be corrected by a nutritional supplement. CONCLUSION FPP can correct respiratory burst performance of T2DM PBMC via an Sp-1-dependant pathway. Studies testing the outcome of FPP supplementation in diabetic patients are warranted.
Collapse
Affiliation(s)
- Ryan Dickerson
- Department of Surgery, Comprehensive Wound Center, Davis Heart and Lung Research Institute, The Ohio State University Medical Center, Columbus, Ohio
| | - Bhakthi Deshpande
- Department of Surgery, Comprehensive Wound Center, Davis Heart and Lung Research Institute, The Ohio State University Medical Center, Columbus, Ohio
| | - Urmila Gnyawali
- Department of Surgery, Comprehensive Wound Center, Davis Heart and Lung Research Institute, The Ohio State University Medical Center, Columbus, Ohio
| | - Debbie Lynch
- Department of Surgery, Comprehensive Wound Center, Davis Heart and Lung Research Institute, The Ohio State University Medical Center, Columbus, Ohio
| | - Gayle M. Gordillo
- Department of Surgery, Comprehensive Wound Center, Davis Heart and Lung Research Institute, The Ohio State University Medical Center, Columbus, Ohio
| | - Dara Schuster
- Endocrinology, Diabetes, and Metabolism, Department of Internal Medicine, The Ohio State University Medical Center, Columbus, Ohio
| | - Kwame Osei
- Endocrinology, Diabetes, and Metabolism, Department of Internal Medicine, The Ohio State University Medical Center, Columbus, Ohio
| | - Sashwati Roy
- Department of Surgery, Comprehensive Wound Center, Davis Heart and Lung Research Institute, The Ohio State University Medical Center, Columbus, Ohio
- Endocrinology, Diabetes, and Metabolism, Department of Internal Medicine, The Ohio State University Medical Center, Columbus, Ohio
| |
Collapse
|
21
|
David JJ, Subramanian SV, Zhang A, Willis WL, Kelm RJ, Leier CV, Strauch AR. Y-box binding protein-1 implicated in translational control of fetal myocardial gene expression after cardiac transplant. Exp Biol Med (Maywood) 2012; 237:593-607. [PMID: 22619371 DOI: 10.1258/ebm.2012.011137] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Peri-transplant surgical trauma and ischemia/reperfusion injury in accepted murine heterotopic heart grafts has been associated with myofibroblast differentiation, cardiac fibrosis and biomechanical-stress activation of the fetal myocardial smooth muscle α-actin (SMαA) gene. The wound-healing agonists, transforming growth factor β1 and thrombin, are known to coordinate SMαA mRNA transcription and translation in activated myofibroblasts by altering the subcellular localization and mRNA-binding affinity of the Y-box binding protein-1 (YB-1) cold-shock domain (CSD) protein that governs a variety of cellular responses to metabolic stress. YB-1 accumulated in polyribosome-enriched regions of the sarcoplasm proximal to cardiac intercalated discs in accepted heart grafts. YB-1 binding to a purine-rich motif in exon 3 of SMαA mRNA that regulates translational efficiency increased substantially in perfusion-isolated, rod-shaped adult rat cardiomyocytes during phenotypic de-differentiation in the presence of serum-derived growth factors. Cardiomyocyte de-differentiation was accompanied by the loss of a 60 kDa YB-1 variant that was highly expressed in both adult myocardium and freshly isolated myocytes and replacement with the 50 kDa form of YB-1 (p50) typically expressed in myofibroblasts that demonstrated sequence-specific interaction with SMαA mRNA. Accumulation of p50 YB-1 in reprogrammed, de-differentiated myocytes was associated with a 10-fold increase in SMαA protein expression. Endomyocardial biopsies collected from patients up to 14 years after heart transplant showed variable yet coordinately elevated expression of SMαA and p50 YB-1 protein and demonstrable p50 YB-1:SMαA mRNA interaction. The p60 YB-1 variant in human heart graft samples, but neither mouse p60 nor mouse or human p50, reacted with an antibody specific for the phosphoserine 102 modification in the YB-1 CSD. Modulation of YB-1 subcellular compartmentalization and mRNA-binding activity may be linked with reprogramming of contractile protein gene expression in ventricular cardiomyocytes that could contribute to maladaptive remodeling in accepted, long-term heart grafts.
Collapse
Affiliation(s)
- Jason J David
- Department of Physiology & Cell Biology, Dorothy M. Davis Heart & Lung Research Institute, College of Medicine, The Ohio State University, Columbus, OH 43210, USA
| | | | | | | | | | | | | |
Collapse
|
22
|
Secchiero P, Rimondi E, di Iasio MG, Voltan R, Gonelli A, Zauli G. Activation of the p53 pathway induces α-smooth muscle actin expression in both myeloid leukemic cells and normal macrophages. J Cell Physiol 2012; 227:1829-37. [PMID: 21732354 DOI: 10.1002/jcp.22910] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
A range of cell types of mesenchymal origin express α-smooth muscle actin (α-SMA), a protein that plays a key role in controlling cell motility and differentiation along the fibrocyte and myofibroblast lineages. Although α-SMA is often expressed in stromal cells associated to a variety of cancers including hematological malignancies, up to now the role of anti-cancer drugs on α-SMA has not been deeply investigated. In this study, we demonstrated that Nutlin-3, the small molecule inhibitor of the MDM2/p53 interactions, significantly up-regulated the mRNA and protein levels of α-SMA in normal macrophages as well as in p53(wild-type) but not in p53(mutated/null) myeloid leukemic cells. The p53-dependence of α-SMA up-regulation induced by Nutlin-3 was demonstrated in experiments performed with siRNA for p53. Of note, Nutlin-3 mediated up-regulation of α-SMA in OCI leukemic cells was accompanied by cell adhesion to plastic substrate and by reduced cell migratory response in transwell assays. Notably, the role of α-SMA induction in the modulation of myeloid cell migration was clearly documented in α-SMA gene knockdown experiments. In addition, Nutlin-3 significantly up-regulated α-SMA expression in primary endothelial cells, but not in fibroblasts and mesenchymal stem cells (MSC). Conversely, transforming growth factor-β1 up-regulated α-SMA in fibroblasts and MSC, but not in macrophages and endothelial cells. Taken together, these data indicate that Nutlin-3 is a potent inducer of α-SMA in both normal and leukemic myeloid cells as well as in endothelial cells.
Collapse
Affiliation(s)
- Paola Secchiero
- Department of Morphology and Embryology and LTTA Centre, University of Ferrara, Ferrara, Italy.
| | | | | | | | | | | |
Collapse
|
23
|
Takahashi K, Sakamoto K, Kimura J. Hypoxic stress induces transient receptor potential melastatin 2 (TRPM2) channel expression in adult rat cardiac fibroblasts. J Pharmacol Sci 2012; 118:186-97. [PMID: 22293297 DOI: 10.1254/jphs.11128fp] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/14/2022] Open
Abstract
When cardiac tissue is exposed to hypoxia, myocytes are damaged, while fibroblasts are activated. However, it is unknown what changes are induced by hypoxia in cardiac fibroblasts. In this study, using the whole cell patch-clamp technique, we investigated the effect of hypoxia on membrane currents in fibroblasts primarily cultured from adult rat hearts. Cardiac fibroblasts were incubated for 24 h under normoxic or hypoxic conditions using Anaeropack. Hypoxia increased a current which reversed at around -20 mV in the cardiac fibroblasts. This current was inhibited by clotrimazole, which is an inhibitor of transient receptor potential melastatin 2 (TRPM2) channel and intermediate-conductance Ca(2+)-activated K(+) channel (KCa3.1). ADP ribose in the pipette solution enhanced this current. Quantitative RT-PCR revealed that mRNA of TRPM2, but not that of KCa3.1, was increased by hypoxia. RNA interference of TRPM2 prevented the development of the hypoxia-induced current. H(2)O(2), an activator of TRPM2 channel, induced a higher [Ca(2+)](i) elevation in hypoxia-exposed cardiac fibroblasts than that in normoxia-exposed cells. We conclude that hypoxia induces TRPM2 channel expression in adult rat cardiac fibroblasts.
Collapse
Affiliation(s)
- Kenji Takahashi
- Department of Pharmacology, Fukushima Medical University, School of Medicine, Japan.
| | | | | |
Collapse
|
24
|
Terrand J, Xu B, Morrissy S, Dinh TN, Williams S, Chen QM. p21(WAF1/Cip1/Sdi1) knockout mice respond to doxorubicin with reduced cardiotoxicity. Toxicol Appl Pharmacol 2011; 257:102-10. [PMID: 21920376 DOI: 10.1016/j.taap.2011.08.024] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2011] [Revised: 08/21/2011] [Accepted: 08/26/2011] [Indexed: 12/31/2022]
Abstract
Doxorubicin (Dox) is an antineoplastic agent that can cause cardiomyopathy in humans and experimental animals. As an inducer of reactive oxygen species and a DNA damaging agent, Dox causes elevated expression of p21(WAF1/Cip1/Sdi1) (p21) gene. Elevated levels of p21 mRNA and p21 protein have been detected in the myocardium of mice following Dox treatment. With chronic treatment of Dox, wild type (WT) animals develop cardiomyopathy evidenced by elongated nuclei, mitochondrial swelling, myofilamental disarray, reduced cardiac output, reduced ejection fraction, reduced left ventricular contractility, and elevated expression of ANF gene. In contrast, p21 knockout (p21KO) mice did not show significant changes in the same parameters in response to Dox treatment. In an effort to understand the mechanism of the resistance against Dox induced cardiomyopathy, we measured levels of antioxidant enzymes and found that p21KO mice did not contain elevated basal or inducible levels of glutathione peroxidase and catalase. Measurements of 6 circulating cytokines indicated elevation of IL-6, IL-12, IFNγ and TNFα in Dox treated WT mice but not p21KO mice. Dox induced elevation of IL-6 mRNA was detected in the myocardium of WT mice but not p21KO mice. While the mechanism of the resistance against Dox induced cardiomyopathy remains unclear, lack of inflammatory response may contribute to the observed cardiac protection in p21KO mice.
Collapse
Affiliation(s)
- Jerome Terrand
- Department of Pharmacology,College of Medicine, University of Arizona, 1501 N. Campbell Ave, Tucson, AZ 85724, USA
| | | | | | | | | | | |
Collapse
|
25
|
p21/waf1 and smooth-muscle actin α expression in stromal fibroblasts of oral cancers. Cell Oncol (Dordr) 2011; 34:483-8. [PMID: 21559927 DOI: 10.1007/s13402-011-0044-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/20/2010] [Indexed: 10/18/2022] Open
Abstract
BACKGROUND Concerted alterations between stromal fibroblasts and neoplastic cells underline the carcinogenic process. Activation of alpha-smooth muscle actin (SMA) expression, a cytoskeleton protein normally expressed only in myoepithelial cells, is considered a landmark for the activation of stromal fibroblasts with little however being known regarding the mechanism governing the expression of SMA in the stroma. METHODS We have evaluated by immunohistochemistry the expression of SMA in the stroma of oral malignant and pre-malignant lesions, in association with the expression of p53 and p21 tumor suppressors that were shown previously to be deregulated and/or mutated in stromal fibroblasts of various cancers. The effects of p21 knockdown in SMA expression and cell migration and the mRNA levels of endogenous p21 in fibroblasts co-cultured with cancer cells were also assessed. RESULTS We found that both p21 and SMA expression was elevated in the stroma, but not the epithelium, of malignant as compared to pre-malignant lesions. We also noted that the expression of both was positively correlated, implying that SMA expression may be regulated by p21. Consistently with this notion we found that siRNA-mediated p21 suppression resulted in the reduction of SMA levels and also inhibited cell migration. CONCLUSION Our results show that p21 deregulation is associated with the activation of stromal fibroblasts of oral cancers by a mechanism that involves the stimulation of SMA expression.
Collapse
|
26
|
Roy S, Dickerson R, Khanna S, Collard E, Gnyawali U, Gordillo GM, Sen CK. Particulate β-glucan induces TNF-α production in wound macrophages via a redox-sensitive NF-κβ-dependent pathway. Wound Repair Regen 2011; 19:411-9. [PMID: 21518092 DOI: 10.1111/j.1524-475x.2011.00688.x] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Glucans are known to promote wound repair. Noncellulosic β-glucans are recognized as potent immunological activators. β-Glucans are generally safe and are known to attenuate the rate of postoperative infection. Glyc101 is a particulate β-glucan isolated from Saccharomyces cerevisiae. In this study, the hypothesis that Glyc101 regulates wound macrophage function was tested. Glyc101 induced tumor necrosis factor (TNF) α transcription in macrophages isolated from murine wound site. Multiplex assay identified interleukin (IL)-10 and TNFα as two cytokines that are induced by Glyc101 in human blood monocyte-derived macrophages. Glyc101-induced TNFα production was observed to be mediated via the TLR-2 and dectin-1 receptors, receptor tyrosine kinases and NFκB activation. In murine wound macrophages, Glyc101 potentiated phorbol 12-myristate 13-acetate-induced respiratory burst. In vivo, implantation of Glyc101-enriched polyvinyl alcohol-sponges at the wound-site induced TNFα expression in macrophages. Consistently, Glyc101 induced TNFα expression in wound-site macrophages isolated from two patients with chronic wounds. These observations establish the translational significance of the net findings of this study. Activation of wound macrophages by Glyc101 represents one of the potential mechanisms by which this β-glucan may benefit chronic wounds where inefficient inflammatory response is one of the underlying causes of impaired healing.
Collapse
Affiliation(s)
- Sashwati Roy
- Comprehensive Wound Center, Department of Surgery, Davis Heart and Lung Research Institute, The Ohio State University Medical Center, Columbus, Ohio 43210, USA.
| | | | | | | | | | | | | |
Collapse
|
27
|
Sen CK, Roy S. Oxygenation state as a driver of myofibroblast differentiation and wound contraction: hypoxia impairs wound closure. J Invest Dermatol 2010; 130:2701-3. [PMID: 21068734 PMCID: PMC3815591 DOI: 10.1038/jid.2010.316] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Myofibroblasts are ubiquitous in the human body and may form from the differentiation of fibroblasts, epithelial cells, endothelial cells, and mononuclear cells, among others. Their clinical significance could be substantial, depending on biomedical context. Myofibroblasts help contract open skin wounds, but they could also be key drivers of fibrosis across numerous tissue systems and support tumor invasiveness. Understanding the molecular events underlying myofibroblast formation is significant for many human diseases. In this issue, Modarressi et al. address the significance of wound tissue hypoxia in impairing wound contraction by compromising myofibroblast formation. They present compelling evidence indicating tissue hypoxia conflicts with wound closure. We are reminded that correcting wound tissue hypoxia is critical for the tissue's response to other therapeutic interventions.
Collapse
Affiliation(s)
- Chandan K Sen
- Comprehensive Wound Center, Davis Heart & Lung Research Institute, Department of Surgery, The Ohio State University Medical Center, Columbus, Ohio 43210, USA.
| | | |
Collapse
|
28
|
Roy S, Khanna S, Azad A, Schnitt R, He G, Weigert C, Ichijo H, Sen CK. Fra-2 mediates oxygen-sensitive induction of transforming growth factor beta in cardiac fibroblasts. Cardiovasc Res 2010; 87:647-55. [PMID: 20427335 DOI: 10.1093/cvr/cvq123] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
AIMS In the ischaemia-reperfused heart, transforming growth factor beta (TGFbeta) proteins trigger the differentiation of cardiac fibroblasts (CFs) contributing to fibrosis. Reoxygenation of the heart, in addition to being a trigger for reperfusion injury, induces tissue remodelling by hyperoxia-sensitive signalling processes involving TGFbeta. Here, we sought to characterize the molecular mechanisms responsible for the O(2)-sensitive transcriptional induction of TGFbeta in murine CF and to test the significance of such findings in the infarcted myocardium in vivo using laser capture microdissection. METHODS AND RESULTS All three isoforms of TGFbeta were induced in the CF-rich peri-infarct tissue as well as in CF exposed to hyperoxic challenge. Reporter studies demonstrated that TGFbeta transcription is hyperoxia inducible. Deletion of any one or both of the activating protein-1 (AP-1) binding sites in the TGFbeta reporter construct resulted in loss of O(2) sensitivity, demonstrating that AP-1 confers O(2) sensitivity to TGFbeta transcription. Fos-related AP-1 transcription factor (Fra-2) and Ask-1 (apoptosis signal-regulating kinase-1) were identified as key mediators of AP-1-dependent O(2)-sensitive TGFbeta transcription. Knockdown of Fra-2 significantly blunted O(2)-induced expression of TGFbeta1 as well as TGFbeta3 in CF. Knockdown of Ask-1 blunted hyperoxia-induced Fra-2 gene expression and nuclear localization in CF. Collectively, these observations point towards a central role of Ask-1 and Fra-2 in O(2)-inducible AP-1 activation and induction of TGFbeta. CONCLUSION Taken together with the observation that Fra-2-regulated genes are implicated in fibrosis, identification of Fra-2 as an O(2)-sensitive transcriptional regulator of inducible TGFbeta expression positions Fra-2 as an important player in reoxygenation-induced fibrosis.
Collapse
Affiliation(s)
- Sashwati Roy
- Department of Surgery, The Ohio State University Medical Center, 513 Davis Heart and Lung Research Institute, 473 West 12th Avenue, Columbus, OH 43210, USA
| | | | | | | | | | | | | | | |
Collapse
|
29
|
Khanna S, Biswas S, Shang Y, Collard E, Azad A, Kauh C, Bhasker V, Gordillo GM, Sen CK, Roy S. Macrophage dysfunction impairs resolution of inflammation in the wounds of diabetic mice. PLoS One 2010; 5:e9539. [PMID: 20209061 PMCID: PMC2832020 DOI: 10.1371/journal.pone.0009539] [Citation(s) in RCA: 454] [Impact Index Per Article: 30.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2009] [Accepted: 02/11/2010] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Chronic inflammation is a characteristic feature of diabetic cutaneous wounds. We sought to delineate novel mechanisms involved in the impairment of resolution of inflammation in diabetic cutaneous wounds. At the wound-site, efficient dead cell clearance (efferocytosis) is a pre-requisite for the timely resolution of inflammation and successful healing. METHODOLOGY/PRINCIPAL FINDINGS Macrophages isolated from wounds of diabetic mice showed significant impairment in efferocytosis. Impaired efferocytosis was associated with significantly higher burden of apoptotic cells in wound tissue as well as higher expression of pro-inflammatory and lower expression of anti-inflammatory cytokines. Observations related to apoptotic cell load at the wound site in mice were validated in the wound tissue of diabetic and non-diabetic patients. Forced Fas ligand driven elevation of apoptotic cell burden at the wound site augmented pro-inflammatory and attenuated anti-inflammatory cytokine response. Furthermore, successful efferocytosis switched wound macrophages from pro-inflammatory to an anti-inflammatory mode. CONCLUSIONS/SIGNIFICANCE Taken together, this study presents first evidence demonstrating that diabetic wounds suffer from dysfunctional macrophage efferocytosis resulting in increased apoptotic cell burden at the wound site. This burden, in turn, prolongs the inflammatory phase and complicates wound healing.
Collapse
Affiliation(s)
- Savita Khanna
- Comprehensive Wound Center, Department of Surgery, Davis Heart and Lung Research Institute, The Ohio State University Medical Center, Columbus, Ohio, United States of America
| | - Sabyasachi Biswas
- Comprehensive Wound Center, Department of Surgery, Davis Heart and Lung Research Institute, The Ohio State University Medical Center, Columbus, Ohio, United States of America
| | - Yingli Shang
- Comprehensive Wound Center, Department of Surgery, Davis Heart and Lung Research Institute, The Ohio State University Medical Center, Columbus, Ohio, United States of America
| | - Eric Collard
- Comprehensive Wound Center, Department of Surgery, Davis Heart and Lung Research Institute, The Ohio State University Medical Center, Columbus, Ohio, United States of America
| | - Ali Azad
- Comprehensive Wound Center, Department of Surgery, Davis Heart and Lung Research Institute, The Ohio State University Medical Center, Columbus, Ohio, United States of America
| | - Courtney Kauh
- Comprehensive Wound Center, Department of Surgery, Davis Heart and Lung Research Institute, The Ohio State University Medical Center, Columbus, Ohio, United States of America
| | - Vineet Bhasker
- Comprehensive Wound Center, Department of Surgery, Davis Heart and Lung Research Institute, The Ohio State University Medical Center, Columbus, Ohio, United States of America
| | - Gayle M. Gordillo
- Comprehensive Wound Center, Department of Surgery, Davis Heart and Lung Research Institute, The Ohio State University Medical Center, Columbus, Ohio, United States of America
| | - Chandan K. Sen
- Comprehensive Wound Center, Department of Surgery, Davis Heart and Lung Research Institute, The Ohio State University Medical Center, Columbus, Ohio, United States of America
| | - Sashwati Roy
- Comprehensive Wound Center, Department of Surgery, Davis Heart and Lung Research Institute, The Ohio State University Medical Center, Columbus, Ohio, United States of America
| |
Collapse
|
30
|
Gnyawali SC, Roy S, McCoy M, Biswas S, Sen CK. Remodeling of the ischemia-reperfused murine heart: 11.7-T cardiac magnetic resonance imaging of contrast-enhanced infarct patches and transmurality. Antioxid Redox Signal 2009; 11:1829-39. [PMID: 19450139 PMCID: PMC2872241 DOI: 10.1089/ars.2009.2635] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Our laboratory has published the first evidence obtained from fast low-angle-shot cine magnetic resonance imaging (11.7 T) studies demonstrating secondary myocyte death after ischemia/reperfusion (IR) of the murine heart. This work provides the first evidence from 11.7-T magnet-assisted pixel-level analysis of the post-IR murine myocardial infarct patches. Changes in function of the remodeling heart were examined in tandem. IR compromised cardiac function and induced LV hypertrophy. During recovery, the IR-induced increase in LV mass was partly offset. IR-induced wall thinning was noted in the anterior aspect of LV and at the diametrically opposite end. Infarct size was observed to be largest on post-IR days 3 and 7. With time (day 28), however, the infarct size was significantly reduced. IR-induced absolute signal-intensity enhancement was highest on post-IR days 3 and 7. As a function of post-IR time, signal-intensity enhancement was attenuated. The threshold of hyperenhanced tissue resulted in delineation of contours that identified necrotic (bona fide infarct) and reversibly injured infarct patches. The study of infarct transmurality indicated that whereas the permanently injured tissue volume remained unchanged, part of the reversibly injured infarct patch recovered in 4 weeks after IR. The approach validated in the current study is powerful in noninvasively monitoring remodeling of the post-IR beating murine myocardium.
Collapse
Affiliation(s)
- Surya C Gnyawali
- Davis Heart & Lung Research Institute, Department of Surgery The Ohio State University Medical Center, Columbus, Ohio 43210, USA
| | | | | | | | | |
Collapse
|
31
|
Cardiac fibroblasts: at the heart of myocardial remodeling. Pharmacol Ther 2009; 123:255-78. [PMID: 19460403 DOI: 10.1016/j.pharmthera.2009.05.002] [Citation(s) in RCA: 779] [Impact Index Per Article: 48.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2009] [Accepted: 05/05/2009] [Indexed: 12/24/2022]
Abstract
Cardiac fibroblasts are the most prevalent cell type in the heart and play a key role in regulating normal myocardial function and in the adverse myocardial remodeling that occurs with hypertension, myocardial infarction and heart failure. Many of the functional effects of cardiac fibroblasts are mediated through differentiation to a myofibroblast phenotype that expresses contractile proteins and exhibits increased migratory, proliferative and secretory properties. Cardiac myofibroblasts respond to proinflammatory cytokines (e.g. TNFalpha, IL-1, IL-6, TGF-beta), vasoactive peptides (e.g. angiotensin II, endothelin-1, natriuretic peptides) and hormones (e.g. noradrenaline), the levels of which are increased in the remodeling heart. Their function is also modulated by mechanical stretch and changes in oxygen availability (e.g. ischaemia-reperfusion). Myofibroblast responses to such stimuli include changes in cell proliferation, cell migration, extracellular matrix metabolism and secretion of various bioactive molecules including cytokines, vasoactive peptides and growth factors. Several classes of commonly prescribed therapeutic agents for cardiovascular disease also exert pleiotropic effects on cardiac fibroblasts that may explain some of their beneficial outcomes on the remodeling heart. These include drugs for reducing hypertension (ACE inhibitors, angiotensin receptor blockers, beta-blockers), cholesterol levels (statins, fibrates) and insulin resistance (thiazolidinediones). In this review, we provide insight into the properties of cardiac fibroblasts that underscores their importance in the remodeling heart, including their origin, electrophysiological properties, role in matrix metabolism, functional responses to environmental stimuli and ability to secrete bioactive molecules. We also review the evidence suggesting that certain cardiovascular drugs can reduce myocardial remodeling specifically via modulatory effects on cardiac fibroblasts.
Collapse
|
32
|
Liu X, Kelm RJ, Strauch AR. Transforming growth factor beta1-mediated activation of the smooth muscle alpha-actin gene in human pulmonary myofibroblasts is inhibited by tumor necrosis factor-alpha via mitogen-activated protein kinase kinase 1-dependent induction of the Egr-1 transcriptional repressor. Mol Biol Cell 2009; 20:2174-85. [PMID: 19261809 DOI: 10.1091/mbc.e08-10-0994] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Transforming growth factor (TGF) beta1 is a mediator of myofibroblast differentiation in healing wounds in which it activates transcription of the smooth muscle alpha-actin (SMalphaA) gene via dynamic interplay of nuclear activators and repressors. Targeting components of TGFbeta1 signaling may be an effective strategy for controlling myofibroblasts in chronic fibrotic diseases. We examined the ability of proinflammatory tumor necrosis factor (TNF)-alpha to antagonize TGFbeta1-mediated human pulmonary myofibroblast differentiation. TNF-alpha abrogated TGFbeta1-induced SMalphaA gene expression at the level of transcription without disrupting phosphorylation of regulatory Smads. Intact mitogen-activated protein kinase kinase (Mek)-extracellular signal-regulated kinase (Erk) kinase signaling was required for myofibroblast repression by TNF-alpha via induction of the early growth response factor-1 (Egr-1) DNA-binding protein. Egr-1 bound to the GC-rich SPUR activation element in the SMalphaA promoter and potently suppressed Smad3- and TGFbeta1-mediated transcription. Reduction in Smad binding to the SMalphaA promoter in TNF-alpha-treated myofibroblasts was accompanied by an increase in Egr-1 and YB-1 repressor binding, suggesting that the molecular mechanism underlying repression may involve competitive interplay between Egr-1, YB-1, and Smads. The ability of TNF-alpha to attenuate myofibroblast differentiation via modulation of a Mek1/Erk/Egr-1 regulatory axis may be useful in designing new therapeutic targets to offset destructive tissue remodeling in chronic fibrotic disease.
Collapse
Affiliation(s)
- Xiaoying Liu
- Department of Physiology and Cell Biology and the Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, 43210, USA
| | | | | |
Collapse
|
33
|
Roy S, Khanna S, Hussain SRA, Biswas S, Azad A, Rink C, Gnyawali S, Shilo S, Nuovo GJ, Sen CK. MicroRNA expression in response to murine myocardial infarction: miR-21 regulates fibroblast metalloprotease-2 via phosphatase and tensin homologue. Cardiovasc Res 2009; 82:21-9. [PMID: 19147652 DOI: 10.1093/cvr/cvp015] [Citation(s) in RCA: 490] [Impact Index Per Article: 30.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIMS MicroRNAs (miRNAs) are small non-coding RNAs that regulate gene expression at the post-transcriptional level by either degradation or translational repression of a target mRNA. Encoded in the genome of most eukaryotes, miRNAs have been proposed to regulate specifically up to 90% of human genes through a process known as miRNA-guided RNA silencing. For the first time, we sought to test how myocardial ischaemia-reperfusion (IR) changes miR expression. METHODS AND RESULTS Following 2 and 7 h of IR or sham operation, myocardial tissue was collected and subjected to miRNA expression profiling and quantification using a Bioarray system that screens for human-, mice-, rat-, and Ambi-miR. Data mining and differential analyses resulted in 13 miRs that were up-regulated on day 2, 9 miRs that were up-regulated on day 7, and 6 miRs that were down-regulated on day 7 post-IR. Results randomly selected from expression profiling were validated using real-time PCR. Tissue elements laser-captured from the infarct site showed marked induction of miR-21. In situ hybridization studies using locked nucleic acid miR-21-specific probe identified that IR-inducible miR-21 was specifically localized in the infarct region of the IR heart. Immunohistochemistry data show that cardiac fibroblasts (CFs) are the major cell type in the infarct zone. Studies with isolated CFs demonstrated that phosphatase and tensin homologue (PTEN) is a direct target of miR-21. Modulation of miR-21 regulated expression of matrix metalloprotease-2 (MMP-2) via a PTEN pathway. Finally, we noted a marked decrease in PTEN expression in the infarct zone. This decrease was associated with increased MMP-2 expression in the infarct area. CONCLUSION This work constitutes the first report describing changes in miR expression in response to IR in the mouse heart, showing that miR-21 regulates MMP-2 expression in CFs of the infarct zone via a PTEN pathway.
Collapse
Affiliation(s)
- Sashwati Roy
- Department of Surgery, Davis Heart and Lung Research Institute, The Ohio State University Medical Center, 473 West 12th Avenue, Columbus, OH 43210, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Cx43 contributes to TGF-beta signaling to regulate differentiation of cardiac fibroblasts into myofibroblasts. Exp Cell Res 2009; 315:1190-9. [PMID: 19162006 DOI: 10.1016/j.yexcr.2008.12.021] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2008] [Revised: 12/02/2008] [Accepted: 12/20/2008] [Indexed: 01/09/2023]
Abstract
Differentiation and activation of fibroblasts into myofibroblasts which express alpha-smooth muscle actin (alpha-SMA) are essential for wound healing and tissue repair. Change in fibroblast properties is initiated by transforming growth factor beta (TGF-beta). Here, we sought to investigate whether connexin43 (Cx43), a gap-junctional protein, contributes to differentiation of cardiac fibroblasts to myofibroblasts. In cultured neonatal rat cardiac fibroblasts, we found that expression of alpha-SMA increases in parallel with Cx43 by using immunocytochemistry, and that knockdown of the endogenous Cx43 activity with antisense oligodeoxynucleotides (AS) inhibits alpha-SMA expression significantly, while overexpression of Cx43 increases alpha-SMA expression remarkably. These findings demonstrate that Cx43 contributes to TGF-beta signaling to regulate alpha-SMA expression. Thus, we propose a novel physiologic function of Cx43, which plays a critical role in the pathological activation of cardiac fibroblasts in the myocardial fibrosis associated with heart failure.
Collapse
|
35
|
Novotny NM, Ray R, Markel TA, Crisostomo PR, Wang M, Wang Y, Meldrum DR. Stem cell therapy in myocardial repair and remodeling. J Am Coll Surg 2008; 207:423-34. [PMID: 18722949 DOI: 10.1016/j.jamcollsurg.2008.04.013] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2008] [Revised: 04/04/2008] [Accepted: 04/07/2008] [Indexed: 01/01/2023]
Affiliation(s)
- Nathan M Novotny
- Department of Surgery, Indiana University School of Medicine, Indianapolis, IN, USA
| | | | | | | | | | | | | |
Collapse
|
36
|
Dudas M, Wysocki A, Gelpi B, Tuan TL. Memory encoded throughout our bodies: molecular and cellular basis of tissue regeneration. Pediatr Res 2008; 63:502-12. [PMID: 18427295 DOI: 10.1203/pdr.0b013e31816a7453] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
When a sheep loses its tail, it cannot regenerate it in the manner of lizards. On the other hand, it is possible to clone mammals from somatic cells, showing that a complete developmental program is intact in a wounded sheep's tail the same way it is in a lizard. Thus, there is a requirement for more than only the presence of the entire genetic code in somatic cells for regenerative abilities. Thoughts like this have motivated us to assemble more than just a factographic synopsis on tissue regeneration. As a model, we review skin wound healing in chronological order, and when possible, we use that overview as a framework to point out possible mechanisms of how damaged tissues can restore their original structure. This article postulates the existence of tissue structural memory as a complex distributed homeostatic mechanism. We support such an idea by referring to an extremely fragmented literature base, trying to synthesize a broad picture of important principles of how tissues and organs may store information about their own structure for the purposes of regeneration. Selected developmental, surgical, and tissue engineering aspects are presented and discussed in the light of recent findings in the field. When a sheep loses its tail, it cannot regenerate it in the manner of lizards. On the other hand, it is possible to clone mammals from somatic cells, showing that a complete developmental program is intact in a wounded sheep's tail the same way it is in a lizard. Thus, there is a requirement for more than only the presence of the entire genetic code in somatic cells for regenerative abilities. Thoughts like this have motivated us to assemble more than just a factographic synopsis on tissue regeneration. As a model, we review skin wound healing in chronological order, and when possible, we use that overview as a framework to point out possible mechanisms of how damaged tissues can restore their original structure. This article postulates the existence of tissue structural memory as a complex distributed homeostatic mechanism. We support such an idea by referring to an extremely fragmented literature base, trying to synthesize a broad picture of important principles of how tissues and organs may store information about their own structure for the purposes of regeneration. Selected developmental, surgical, and tissue engineering aspects are presented and discussed in the light of recent findings in the field.
Collapse
Affiliation(s)
- Marek Dudas
- Developmental Biology Program, The Saban Research Institute, Children's Hospital Los Angeles, Los Angeles, CA 90027, USA
| | | | | | | |
Collapse
|