151
|
DePianto DJ, Heiden JAV, Morshead KB, Sun KH, Modrusan Z, Teng G, Wolters PJ, Arron JR. Molecular mapping of interstitial lung disease reveals a phenotypically distinct senescent basal epithelial cell population. JCI Insight 2021; 6:143626. [PMID: 33705361 PMCID: PMC8119199 DOI: 10.1172/jci.insight.143626] [Citation(s) in RCA: 56] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Accepted: 03/05/2021] [Indexed: 12/14/2022] Open
Abstract
Compromised regenerative capacity of lung epithelial cells can lead to cellular senescence, which may precipitate fibrosis. While increased markers of senescence have been reported in idiopathic pulmonary fibrosis (IPF), the origin and identity of these senescent cells remain unclear, and tools to characterize context-specific cellular senescence in human lung are lacking. We observed that the senescent marker p16 is predominantly localized to bronchiolized epithelial structures in scarred regions of IPF and systemic sclerosis-associated interstitial lung disease (SSc-ILD) lung tissue, overlapping with the basal epithelial markers Keratin 5 and Keratin 17. Using in vitro models, we derived transcriptional signatures of senescence programming specific to different types of lung epithelial cells and interrogated these signatures in a single-cell RNA-Seq data set derived from control, IPF, and SSc-ILD lung tissue. We identified a population of basal epithelial cells defined by, and enriched for, markers of cellular senescence and identified candidate markers specific to senescent basal epithelial cells in ILD that can enable future functional studies. Notably, gene expression of these cells significantly overlaps with terminally differentiating cells in stratified epithelia, where it is driven by p53 activation as part of the senescence program.
Collapse
Affiliation(s)
| | | | | | - Kai-Hui Sun
- Department of Molecular Biology, Genentech Inc., San Francisco, California, USA
| | - Zora Modrusan
- Department of Molecular Biology, Genentech Inc., San Francisco, California, USA
| | | | - Paul J. Wolters
- Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, Department of Medicine, University of California, San Francisco, California, USA
| | | |
Collapse
|
152
|
Helbling-Leclerc A, Garcin C, Rosselli F. Beyond DNA repair and chromosome instability-Fanconi anaemia as a cellular senescence-associated syndrome. Cell Death Differ 2021; 28:1159-1173. [PMID: 33723374 PMCID: PMC8026967 DOI: 10.1038/s41418-021-00764-5] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Revised: 02/17/2021] [Accepted: 02/24/2021] [Indexed: 02/07/2023] Open
Abstract
Fanconi anaemia (FA) is the most frequent inherited bone marrow failure syndrome, due to mutations in genes encoding proteins involved in replication fork protection, DNA interstrand crosslink repair and replication rescue through inducing double-strand break repair and homologous recombination. Clinically, FA is characterised by aplastic anaemia, congenital defects and cancer predisposition. In in vitro studies, FA cells presented hallmarks defining senescent cells, including p53-p21 axis activation, altered telomere length, mitochondrial dysfunction, chromatin alterations, and a pro-inflammatory status. Senescence is a programme leading to proliferation arrest that is involved in different physiological contexts, such as embryogenesis, tissue remodelling and repair and guarantees tumour suppression activity. However, senescence can become a driving force for developmental abnormalities, aging and cancer. Herein, we summarise the current knowledge in the field to highlight the mutual relationships between FA and senescence that lead us to consider FA not only as a DNA repair and chromosome fragility syndrome but also as a "senescence syndrome".
Collapse
Affiliation(s)
- Anne Helbling-Leclerc
- grid.14925.3b0000 0001 2284 9388UMR9019-CNRS, Gustave Roussy, Villejuif, Cedex France ,grid.460789.40000 0004 4910 6535Université Paris-Saclay, Orsay, France ,Equipe labellisée “La Ligue Contre le Cancer”, Villejuif, France
| | - Cécile Garcin
- grid.14925.3b0000 0001 2284 9388UMR9019-CNRS, Gustave Roussy, Villejuif, Cedex France ,grid.460789.40000 0004 4910 6535Université Paris-Saclay, Orsay, France ,Equipe labellisée “La Ligue Contre le Cancer”, Villejuif, France
| | - Filippo Rosselli
- grid.14925.3b0000 0001 2284 9388UMR9019-CNRS, Gustave Roussy, Villejuif, Cedex France ,grid.460789.40000 0004 4910 6535Université Paris-Saclay, Orsay, France ,Equipe labellisée “La Ligue Contre le Cancer”, Villejuif, France
| |
Collapse
|
153
|
Toorani T, Mackie PM, Mastromonaco GF. Investigating Markers of Reprogramming Potential in Somatic Cell Lines Derived from Matched Donors. Cell Reprogram 2021; 23:73-88. [PMID: 33861640 DOI: 10.1089/cell.2020.0075] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Somatic cell biobanking and related technologies, somatic cell nuclear transfer (SCNT), and induction of pluripotent stem cells offer significant promise for wildlife conservation, but have yet to achieve optimal success. Inefficiency and variability in outcome have been linked to incomplete nuclear reprogramming, highlighting the importance of donor cell contribution. Studies show significant differences in SCNT outcome in donor cell lines within and between individuals, highlighting the necessity for a standardized characterization method to evaluate cell line reprogramming potential. Stringently standardized bovine fibroblast cell lines were generated and assessed for inter- and intraindividual variability on cellular (morphology, chromosome number, apoptotic incidence; Experiment 1) and molecular (pluripotency and epigenetic-related gene expression; Experiment 2) levels encompassing putative biomarkers of reprogramming potential. Cellular parameters were similar across cell lines. While some statistically significant differences were observed in DNMT1, DNMT3B, and HAT1, but not HDAC1, their biological relevance could not be determined with the information at hand. This study lays the foundation for understanding cellular characteristics in cultured cell lines; however, further studies are required to determine any correlation with reprogramming potential.
Collapse
Affiliation(s)
- Tahmineh Toorani
- Reproductive Sciences, Toronto Zoo, Scarborough, Canada.,Department of Biomedical Sciences, Ontario Veterinary College, University of Guelph, Guelph, Canada
| | | | - Gabriela F Mastromonaco
- Reproductive Sciences, Toronto Zoo, Scarborough, Canada.,Department of Biomedical Sciences, Ontario Veterinary College, University of Guelph, Guelph, Canada
| |
Collapse
|
154
|
Fitsiou E, Soto-Gamez A, Demaria M. Biological functions of therapy-induced senescence in cancer. Semin Cancer Biol 2021; 81:5-13. [PMID: 33775830 DOI: 10.1016/j.semcancer.2021.03.021] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 03/03/2021] [Accepted: 03/22/2021] [Indexed: 01/10/2023]
Abstract
Therapy-induced cellular senescence is a state of stable growth arrest induced by common cancer treatments such as chemotherapy and radiation. In an oncogenic context, therapy-induced senescence can have different consequences. By blocking cellular proliferation and by facilitating immune cell infiltration, it functions as tumor suppressive mechanism. By fueling the proliferation of bystander cells and facilitating metastasis, it acts as a tumor promoting factor. This dual role is mainly attributed to the differential expression and secretion of a set of pro-inflammatory cytokines and tissue remodeling factors, collectively known as the Senescence-Associated Secretory Phenotype (SASP). Here, we describe cell-autonomous and non-cell-autonomous mechanisms that senescent cells activate in response to chemotherapy and radiation leading to tumor suppression and tumor promotion. We present the current state of knowledge on the stimuli that affect the activation of these opposing mechanisms and the effect of senescent cells on their micro-environment eg. by regulating the functions of immune cells in tumor clearance as well as strategies to eliminate senescent tumor cells before exerting their deleterious side-effects.
Collapse
Affiliation(s)
- Eleni Fitsiou
- European Research Institute for the Biology of Ageing, University Medical Center Groningen, 9713AV, Groningen, The Netherlands
| | - Abel Soto-Gamez
- European Research Institute for the Biology of Ageing, University Medical Center Groningen, 9713AV, Groningen, The Netherlands; University of Groningen, Groningen Research Institute of Pharmacy, Chemical and Pharmaceutical Biology, Groningen, The Netherlands
| | - Marco Demaria
- European Research Institute for the Biology of Ageing, University Medical Center Groningen, 9713AV, Groningen, The Netherlands.
| |
Collapse
|
155
|
Therapeutic Potential of RTA 404 in Human Brain Malignant Glioma Cell Lines via Cell Cycle Arrest via p21/AKT Signaling. BIOMED RESEARCH INTERNATIONAL 2021; 2021:5552226. [PMID: 33763472 PMCID: PMC7963900 DOI: 10.1155/2021/5552226] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Revised: 02/17/2021] [Accepted: 02/27/2021] [Indexed: 01/11/2023]
Abstract
Background Glioblastoma multiforme (GBM) is the most common malignant brain tumor in the world. Despite advances in surgical resection, radiotherapy, and chemotherapy, GBM continues to have a poor overall survival. CDDO (2-cyano-3,12-dioxoolean-1,9-dien-28-oic acid), a synthetic triterpenoid, is an Nrf2 activator used to inhibit proliferation and induce differentiation and apoptosis in various cancer cells. One new trifluoroethylamide derivative of CDDO, RTA 404, has been found to have increased ability to cross the blood-brain barrier. However, it is not clear what effect it may have on tumorigenesis in GBM. Methods This in vitro study evaluated the effects of RTA 404 on GBM cells. To do this, we treated GBM840 and U87 MG cell lines with RTA 404 and assessed apoptosis, cell cycle, cell locomotion, and senescence. DNA content and induction of apoptosis were analyzed by flow cytometry and protein expression by Western blot analysis. Results RTA 404 significantly inhibited the proliferation of tumor cells at concentrations higher than 100 nM (p < 0.05) and reduced their locomotion ability. In addition, treatment with RTA 404 led to an accumulation of RTA 404-treated G2/M phase cells and apoptosis. An analysis of the p21/AKT expression suggested that RTA 404 may not only help prevent brain cancer but it may also exert antitumor activities in established GBM cells. Conclusion RTA404 can inhibit proliferation, cell locomotion, cell cycle progression, and induce apoptosis in GBM cells in vitro, possibly through its inhibition of N-cadherin and E-cadherin expression via its inhibition of the AKT pathway.
Collapse
|
156
|
Peredo AP, Gullbrand SE, Mauck RL, Smith HE. A challenging playing field: Identifying the endogenous impediments to annulus fibrosus repair. JOR Spine 2021; 4:e1133. [PMID: 33778407 PMCID: PMC7984000 DOI: 10.1002/jsp2.1133] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Revised: 11/10/2020] [Accepted: 11/11/2020] [Indexed: 12/31/2022] Open
Abstract
Intervertebral disc (IVD) herniations, caused by annulus fibrosus (AF) tears that enable disc tissue extrusion beyond the disc space, are very prevalent, especially among adults in the third to fifth decade of life. Symptomatic herniations, in which the extruded tissue compresses surrounding nerves, are characterized by back pain, numbness, and tingling and can cause extreme physical disability. Patients whose symptoms persist after nonoperative intervention may undergo surgical removal of the herniated tissue via microdiscectomy surgery. The AF, however, which has a poor endogenous healing ability, is left unrepaired increasing the risk for re-herniation and pre-disposing the IVD to degenerative disc disease. The lack of understanding of the mechanisms involved in native AF repair limits the design of repair systems that overcome the impediments to successful AF restoration. Moreover, the complexity of the AF structure and the challenging anatomy of the repair environment represents a significant challenge for the design of new repair devices. While progress has been made towards the development of an effective AF repair technique, these methods have yet to demonstrate long-term repair and recovery of IVD biomechanics. In this review, the limitations of endogenous AF healing are discussed and key cellular events and factors involved are highlighted to identify potential therapeutic targets that can be integrated into AF repair methods. Clinical repair strategies and their limitations are described to further guide the design of repair approaches that effectively restore native tissue structure and function.
Collapse
Affiliation(s)
- Ana P. Peredo
- Department of BioengineeringSchool of Engineering and Applied Science, University of PennsylvaniaPhiladelphiaPennsylvaniaUSA
- McKay Orthopaedic Research Laboratory, Department of Orthopaedic SurgeryPerelman School of Medicine, University of PennsylvaniaPhiladelphiaPennsylvaniaUSA
- Translational Musculoskeletal Research CenterCorporal Michael J. Crescenz Veterans Affairs Medical CenterPhiladelphiaPennsylvaniaUSA
| | - Sarah E. Gullbrand
- McKay Orthopaedic Research Laboratory, Department of Orthopaedic SurgeryPerelman School of Medicine, University of PennsylvaniaPhiladelphiaPennsylvaniaUSA
- Translational Musculoskeletal Research CenterCorporal Michael J. Crescenz Veterans Affairs Medical CenterPhiladelphiaPennsylvaniaUSA
| | - Robert L. Mauck
- Department of BioengineeringSchool of Engineering and Applied Science, University of PennsylvaniaPhiladelphiaPennsylvaniaUSA
- McKay Orthopaedic Research Laboratory, Department of Orthopaedic SurgeryPerelman School of Medicine, University of PennsylvaniaPhiladelphiaPennsylvaniaUSA
- Translational Musculoskeletal Research CenterCorporal Michael J. Crescenz Veterans Affairs Medical CenterPhiladelphiaPennsylvaniaUSA
| | - Harvey E. Smith
- McKay Orthopaedic Research Laboratory, Department of Orthopaedic SurgeryPerelman School of Medicine, University of PennsylvaniaPhiladelphiaPennsylvaniaUSA
- Translational Musculoskeletal Research CenterCorporal Michael J. Crescenz Veterans Affairs Medical CenterPhiladelphiaPennsylvaniaUSA
| |
Collapse
|
157
|
Csete ME. Basic Science of Frailty-Biological Mechanisms of Age-Related Sarcopenia. Anesth Analg 2021; 132:293-304. [PMID: 32769382 DOI: 10.1213/ane.0000000000005096] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Aging is associated with loss of function across organ systems, contributing to systemic frailty. Loss of skeletal muscle mass and function, in particular, is a major source of frailty in older adults, severely impacting quality of life. Some loss of muscle mass and strength with aging is inevitable, and sarcopenia, the severe loss of muscle mass with aging, is common. Sarcopenia is determined in part by genetics but can be modified by lifestyle choices. The pathophysiologic underpinnings of sarcopenia are complex and multifactorial. In this review, the causes of sarcopenia are surveyed at the systems, cell, subcellular, and molecular levels with emphasis on the interplay between these various causes of this degenerative disease process.
Collapse
Affiliation(s)
- Marie E Csete
- From the Department of Anesthesiology, Keck School of Medicine, University of Southern California, Los Angeles, California
| |
Collapse
|
158
|
Abstract
Significance: Senescence is a cellular state induced by internal or external stimuli, which result in cell cycle arrest, morphological changes, and dysfunctions in mitochondrial and lysosomal functionality as well as the senescence-associated secretory phenotype. Senescent cells accumulate in tissues in physiological and pathological conditions such as development, tissue repair, aging, and cancer. Recent Advances: Growing evidences indicate that senescent cells in vivo are a heterogeneous cell population due to different cell-autonomous activated pathways and distinct microenvironmental contexts. Critical Issues: In this review, we discuss the different contexts where senescence assumes a key role with beneficial or harmful outcomes. The heterogeneous nature of senescence pushes toward resolution of the specific molecular profile and secretome to typify senescent cells in physiological and pathological contexts. Future Directions: Future research will enable exploring the heterogeneity of the senescent population to precisely map the progression of cells through senescent trajectories and study the impact of the therapeutic advantage of senolytic drugs for translational strategies toward supporting the health span. Antioxid. Redox Signal. 34, 294-307.
Collapse
Affiliation(s)
- Alessandra Sacco
- Development, Aging and Regeneration Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California, USA
| | - Laura Belloni
- Department of Internal, Anesthesiological and Cardiovascular Clinical Sciences, Sapienza University of Rome, Rome, Italy
| | - Lucia Latella
- Epigenetics and Regenerative Medicine, IRCCS Fondazione Santa Lucia, Rome, Italy.,Institute of Translational Pharmacology, National Research Council of Italy, Rome, Italy
| |
Collapse
|
159
|
de Mera-Rodríguez JA, Álvarez-Hernán G, Gañán Y, Martín-Partido G, Rodríguez-León J, Francisco-Morcillo J. Is Senescence-Associated β-Galactosidase a Reliable in vivo Marker of Cellular Senescence During Embryonic Development? Front Cell Dev Biol 2021; 9:623175. [PMID: 33585480 PMCID: PMC7876289 DOI: 10.3389/fcell.2021.623175] [Citation(s) in RCA: 74] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Accepted: 01/05/2021] [Indexed: 01/10/2023] Open
Abstract
During vertebrate embryonic development, cellular senescence occurs at multiple locations. To date, it has been accepted that when there has been induction of senescence in an embryonic tissue, β-galactosidase activity is detectable at a pH as high as 6.0, and this has been extensively used as a marker of cellular senescence in vivo in both whole-mount and cryosections. Such senescence-associated β-galactosidase (SA-β-GAL) labeling appears enhanced in degenerating regions of the vertebrate embryo that are also affected by programmed cell death. In this sense, there is a strong SA-β-GAL signal which overlaps with the pattern of cell death in the interdigital tissue of the developing limbs, and indeed, many of the labeled cells detected go on to subsequently undergo apoptosis. However, it has been reported that β-GAL activity at pH 6.0 is also enhanced in healthy neurons, and some retinal neurons are strongly labeled with this histochemical technique when they begin to differentiate during early embryonic development. These labeled early post-mitotic neurons also express other senescence markers such as p21. Therefore, the reliability of this histochemical technique in studying senescence in cells such as neurons that undergo prolonged and irreversible cell-cycle arrest is questionable because it is also expressed in healthy post-mitotic cells. The identification of new biomarkers of cellular senescence would, in combination with established markers, increase the specificity and efficiency of detecting cellular senescence in embryonic and healthy mature tissues.
Collapse
Affiliation(s)
- José Antonio de Mera-Rodríguez
- Área de Biología Celular, Departamento de Anatomía, Biología Celular y Zoología, Facultad de Ciencias, Universidad de Extremadura, Badajoz, Spain
| | - Guadalupe Álvarez-Hernán
- Área de Biología Celular, Departamento de Anatomía, Biología Celular y Zoología, Facultad de Ciencias, Universidad de Extremadura, Badajoz, Spain
| | - Yolanda Gañán
- Área de Anatomía y Embriología Humana, Departamento de Anatomía, Biología Celular y Zoología, Facultad de Medicina, Universidad de Extremadura, Badajoz, Spain
| | - Gervasio Martín-Partido
- Área de Biología Celular, Departamento de Anatomía, Biología Celular y Zoología, Facultad de Ciencias, Universidad de Extremadura, Badajoz, Spain
| | - Joaquín Rodríguez-León
- Área de Anatomía y Embriología Humana, Departamento de Anatomía, Biología Celular y Zoología, Facultad de Medicina, Universidad de Extremadura, Badajoz, Spain
| | - Javier Francisco-Morcillo
- Área de Biología Celular, Departamento de Anatomía, Biología Celular y Zoología, Facultad de Ciencias, Universidad de Extremadura, Badajoz, Spain
| |
Collapse
|
160
|
Sati N, Boyne DJ, Cheung WY, Cash SB, Arora P. Factors Modifying the Associations of Single or Combination Programmed Cell Death 1 and Programmed Cell Death Ligand 1 Inhibitor Therapies With Survival Outcomes in Patients With Metastatic Clear Cell Renal Cell Carcinoma: A Systematic Review and Meta-analysis. JAMA Netw Open 2021; 4:e2034201. [PMID: 33496794 PMCID: PMC7838936 DOI: 10.1001/jamanetworkopen.2020.34201] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
IMPORTANCE Programmed cell death 1/programmed cell death ligand 1 (PD-1/PD-L1) inhibitors are immune checkpoint inhibitors widely used in the treatment of metastatic clear cell renal cell carcinoma (ccRCC) and other cancers. There is a lack of understanding regarding which factors are associated with therapeutic response. OBJECTIVES To conduct a systematic literature review of trials reporting on factors associated with differential response to PD-1/PD-L1 inhibitors among patients diagnosed with metastatic ccRCC and quantitatively synthesize the magnitude to which each factor modified the response to PD-1/PD-L1 inhibitors. DATA SOURCES The MEDLINE and Cochrane Register of Trials databases were searched for studies published in English from 2006 onward. Searches were last run on September 3, 2019. STUDY SELECTION This systematic review and meta-analysis assessed 662 phase 2/3 randomized clinical trials that provided subgroup analyses of any baseline characteristics regarding the treatment response to PD-1/PD-L1 inhibitors, alone or as part of a combination therapy, with respect to overall survival (OS) or progression-free survival (PFS) among patients with metastatic ccRCC. DATA EXTRACTION AND SYNTHESIS A novel quantitative approach was used to synthesize subgroup findings across trials. The ratio of the subgroup-specific hazard ratios (HRs) from each study were pooled using a random-effects meta-analysis whereby ratios of 1.00 would indicate that the subgroup-specific HRs were equal in magnitude. MAIN OUTCOMES AND MEASURES Main outcomes were OS and PFS. RESULTS From an initial 662 reports, 7 trials were considered eligible for inclusion. Meta-analyses suggested the treatment response to PD-1/PD-L1 inhibitors in patients with metastatic ccRCC was significantly associated with age (OS: ratio of HR for age ≥75 years to HR for age <65 years, 1.51; 95% CI, 1.01-2.26), PD-L1 expression (PFS: ratio of HR for PD-L1 < 1% to HR for PD-L1 ≥ 10%, 2.21; 95% CI, 1.14-4.27; ratio of HR for PD-L1 < 1% to HR for PD-L1 ≥ 1%, 1.36; 95% CI, 1.10-1.68), Memorial Sloan Kettering Cancer Center risk score (PFS: ratio of HR for immediate risk score to HR for poor risk score, 1.62; 95% CI, 1.14-2.29; ratio of HR for favorable risk score to HR for poor risk score, 1.53; 95% CI, 1.00-2.34; ratio of HR for favorable risk score to HR for intermediate risk score, 0.96; 95% CI, 0.70-1.30), and sarcomatoid tumor presence (PFS: ratio of HR for no sarcomatoid differentiation to HR for sarcomatoid differentiation, 1.54; 95% CI, 1.07-2.21). CONCLUSIONS AND RELEVANCE This analysis suggests that older age, low levels of PD-L1 expression, and the absence of sarcomatoid tumor differentiation are associated with a diminished response to anti-PD-1/PD-L1 immunotherapies with respect to survival outcomes among patients with metastatic ccRCC.
Collapse
Affiliation(s)
- Neha Sati
- Department of Biology, McMaster University, Hamilton, Ontario, Canada
- Cytel Inc, Toronto, Ontario, Canada
| | - Devon J. Boyne
- Cytel Inc, Toronto, Ontario, Canada
- Department of Oncology, University of Calgary, Calgary, Alberta, Canada
| | - Winson Y. Cheung
- Department of Oncology, University of Calgary, Calgary, Alberta, Canada
| | - Sarah B. Cash
- Department of Public Health, University of Toronto, Toronto, Ontario, Canada
| | - Paul Arora
- Cytel Inc, Toronto, Ontario, Canada
- Department of Public Health, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
161
|
Meiners S, Lehmann M. Senescent Cells in IPF: Locked in Repair? Front Med (Lausanne) 2020; 7:606330. [PMID: 33392228 PMCID: PMC7775527 DOI: 10.3389/fmed.2020.606330] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Accepted: 11/30/2020] [Indexed: 02/06/2023] Open
Affiliation(s)
- Silke Meiners
- Helmholtz Zentrum München, Comprehensive Pneumology Center (CPC), Member of the German Center for Lung Research (DZL), University Hospital, Ludwig-Maximilians University, Munich, Germany
| | - Mareike Lehmann
- Research Unit Lung Repair and Regeneration, Helmholtz Zentrum München, Member of the German Center for Lung Research (DZL), Munich, Germany
| |
Collapse
|
162
|
Ramos-Ibeas P, Gimeno I, Cañón-Beltrán K, Gutiérrez-Adán A, Rizos D, Gómez E. Senescence and Apoptosis During in vitro Embryo Development in a Bovine Model. Front Cell Dev Biol 2020; 8:619902. [PMID: 33392207 PMCID: PMC7775420 DOI: 10.3389/fcell.2020.619902] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Accepted: 12/01/2020] [Indexed: 12/15/2022] Open
Abstract
According to the World Health Organization, infertility affects up to 14% of couples under reproductive age, leading to an exponential rise in the use of assisted reproduction as a route for conceiving a baby. In the same way, thousands of embryos are produced in cattle and other farm animals annually, leading to increased numbers of individuals born. All reproductive manipulations entail deviations of natural phenotypes and genotypes, with in vitro embryo technologies perhaps showing the biggest effects, although these alterations are still emerging. Most of these indications have been provided by animal models, in particular the bovine species, due to its similarities to human early embryo development. Oocytes and embryos are highly sensitive to environmental stress in vivo and in vitro. Thus, during in vitro culture, a number of stressful conditions affect embryonic quality and viability, inducing subfertility and/or long-term consequences that may reach the offspring. A high proportion of the embryos produced in vitro are arrested at a species-specific stage of development during the first cell divisions. These arrested embryos do not show signs of programmed cell death during early cleavage stages. Instead, defective in vitro produced embryos would enter a permanent cell cycle arrest compatible with cellular senescence, in which they show active metabolism and high reactive oxygen species levels. Later in development, mainly during the morula and blastocyst stages, apoptosis would mediate the elimination of certain cells, accomplishing both a physiological role in to balancing cell proliferation and death, and a pathological role preventing the transmission of damaged cells with an altered genome. The latter would acquire relevant importance in in vitro produced embryos that are submitted to stressful environmental stimuli. In this article, we review the mechanisms mediating apoptosis and senescence during early embryo development, with a focus on in vitro produced bovine embryos. Additionally, we shed light on the protective role of senescence and apoptosis to ensure that unhealthy cells and early embryos do not progress in development, avoiding long-term detrimental effects.
Collapse
Affiliation(s)
- Priscila Ramos-Ibeas
- Department of Animal Reproduction, National Institute for Agriculture and Food Research and Technology (INIA), Madrid, Spain
| | - Isabel Gimeno
- Servicio Regional de Investigación y Desarrollo Agroalimentario (SERIDA), Gijón, Spain
| | - Karina Cañón-Beltrán
- Department of Animal Reproduction, National Institute for Agriculture and Food Research and Technology (INIA), Madrid, Spain
| | - Alfonso Gutiérrez-Adán
- Department of Animal Reproduction, National Institute for Agriculture and Food Research and Technology (INIA), Madrid, Spain
| | - Dimitrios Rizos
- Department of Animal Reproduction, National Institute for Agriculture and Food Research and Technology (INIA), Madrid, Spain
| | - Enrique Gómez
- Servicio Regional de Investigación y Desarrollo Agroalimentario (SERIDA), Gijón, Spain
| |
Collapse
|
163
|
Khan A, Fahad TM, Akther T, Zaman T, Hasan MF, Islam Khan MR, Islam MS, Kishi S. Carbofuran accelerates the cellular senescence and declines the life span of spns1 mutant zebrafish. J Cell Mol Med 2020; 25:1048-1059. [PMID: 33277797 PMCID: PMC7812278 DOI: 10.1111/jcmm.16171] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Revised: 11/03/2020] [Accepted: 11/17/2020] [Indexed: 12/16/2022] Open
Abstract
Carbofuran is a carbamate pesticide, widely used in agricultural practices to increase crop productivity. In mammals, carbofuran is known to cause several untoward effects, such as apoptosis in the hippocampal neuron, oxidative stress, loss of memory and chromosomal anomalies. Most of these effects are implicated with cellular senescence. Therefore, the present study aimed to determine the effect of carbofuran on cellular senescence and biological ageing. Spinster homolog 1 (Spns1) is a transmembrane transporter, regulates autolysosomal biogenesis and plays a role in cellular senescence and survival. Using senescence‐associated β‐galactosidase staining, we found that carbofuran accelerates the cellular senescence in spns1 mutant zebrafish. The yolk opaqueness, a premature ageing phenotype in zebrafish embryos, was accelerated by carbofuran treatment. In the survival study, carbofuran shortened the life span of spns1 mutant zebrafish. Autophagy is the cellular lysosomal degradation, usually up‐regulated in the senescent cells. To know the impact of carbofuran exposure on autophagy progress, we established a double‐transgenic zebrafish line, harbouring EGFP‐tagged LC3‐II and mCherry‐tagged Lamp1 on spns1 mutant background, whereas we found, carbofuran exposure synergistically accelerates autolysosome formation with insufficient lysosome‐mediated degradation. Our data collectively suggest that carbofuran exposure synergistically accelerates the cellular senescence and affects biological ageing in spns1 defective animals.
Collapse
Affiliation(s)
- Alam Khan
- Department of Pharmacy, University of Rajshahi, Rajshahi, Bangladesh.,Department of Molecular Medicine, The Scripps Research Institute, Jupiter, FL, USA
| | | | - Tanjima Akther
- Department of Pharmacy, University of Rajshahi, Rajshahi, Bangladesh
| | - Tanjeena Zaman
- Department of Fisheries, University of Rajshahi, Rajshahi, Bangladesh.,Department of Biology, University of Hail, Hail, Saudi Arabia
| | - Md Faruk Hasan
- Department of Genetic Engineering and Biotechnology, University of Rajshahi, Rajshahi, Bangladesh
| | | | | | - Shuji Kishi
- S&J Kishi Research Corporation, Jupiter, FL, USA
| |
Collapse
|
164
|
Kowald A, Passos JF, Kirkwood TBL. On the evolution of cellular senescence. Aging Cell 2020; 19:e13270. [PMID: 33166065 PMCID: PMC7744960 DOI: 10.1111/acel.13270] [Citation(s) in RCA: 103] [Impact Index Per Article: 20.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Revised: 09/11/2020] [Accepted: 10/09/2020] [Indexed: 01/10/2023] Open
Abstract
The idea that senescent cells are causally involved in aging has gained strong support from findings that the removal of such cells alleviates many age‐related diseases and extends the life span of mice. While efforts proceed to make therapeutic use of such discoveries, it is important to ask what evolutionary forces might have been behind the emergence of cellular senescence, in order better to understand the biology that we might seek to alter. Cellular senescence is often regarded as an anti‐cancer mechanism, since it limits the division potential of cells. However, many studies have shown that senescent cells often also have carcinogenic properties. This is difficult to reconcile with the simple idea of an anti‐cancer mechanism. Furthermore, other studies have shown that cellular senescence is involved in wound healing and tissue repair. Here, we bring these findings and ideas together and discuss the possibility that these functions might be the main reason for the evolution of cellular senescence. Furthermore, we discuss the idea that senescent cells might accumulate with age because the immune system had to strike a balance between false negatives (overlooking some senescent cells) and false positives (destroying healthy body cells).
Collapse
Affiliation(s)
- Axel Kowald
- Campus for Ageing and Vitality Newcastle University Institute for Ageing Newcastle upon Tyne UK
- Rostock University Medical Center Institute for Biostatistics and Informatics in Medicine and Aging Research (IBIMA) Rostock Germany
| | - João F. Passos
- Department of Physiology and Biomedical Engineering, Robert and Arlene Kogod Center on Aging Mayo Clinic Rochester Minnesota USA
| | - Thomas B. L. Kirkwood
- Campus for Ageing and Vitality Newcastle University Institute for Ageing Newcastle upon Tyne UK
- Center for Healthy Aging Department of Cellular and Molecular Medicine University of Copenhagen Copenhagen Denmark
| |
Collapse
|
165
|
Resnik SR, Egger A, Abdo Abujamra B, Jozic I. Clinical Implications of Cellular Senescence on Wound Healing. CURRENT DERMATOLOGY REPORTS 2020. [DOI: 10.1007/s13671-020-00320-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
166
|
Roy AL, Sierra F, Howcroft K, Singer DS, Sharpless N, Hodes RJ, Wilder EL, Anderson JM. A Blueprint for Characterizing Senescence. Cell 2020; 183:1143-1146. [PMID: 33128870 DOI: 10.1016/j.cell.2020.10.032] [Citation(s) in RCA: 63] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Given the heterogeneity of senescent cells, our knowledge of both the drivers and consequences of cellular senescence in tissues and organs remains limited, as is our understanding of how this process could be harnessed for human health. Here we identified five broad areas that would help propel the field forward.
Collapse
Affiliation(s)
- Ananda L Roy
- Office of Strategic Coordination, National Institutes of Health, Bethesda, MD 20892, USA; Division of Program Coordination, Planning, and Strategic Initiative, National Institutes of Health, Bethesda, MD 20892, USA.
| | - Felipe Sierra
- National Institute on Aging, National Institutes of Health, Bethesda, MD 20892, USA
| | - Kevin Howcroft
- National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Dinah S Singer
- National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Norman Sharpless
- National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Richard J Hodes
- National Institute on Aging, National Institutes of Health, Bethesda, MD 20892, USA
| | - Elizabeth L Wilder
- Office of Strategic Coordination, National Institutes of Health, Bethesda, MD 20892, USA; Division of Program Coordination, Planning, and Strategic Initiative, National Institutes of Health, Bethesda, MD 20892, USA
| | - James M Anderson
- Division of Program Coordination, Planning, and Strategic Initiative, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
167
|
Shi HZ, Zeng JC, Shi SH, Giannakopoulos H, Zhang QZ, Le AD. Extracellular Vesicles of GMSCs Alleviate Aging-Related Cell Senescence. J Dent Res 2020; 100:283-292. [PMID: 33073684 DOI: 10.1177/0022034520962463] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Healthy aging is a complex biological process with progressive accumulation of senescent cells characterized by stable cell cycle arrest, resulting in impaired homeostasis, regenerative potential, and gradual functional decline in multiple tissues and organs, whereby the aberrant activation of mammalian target of rapamycin (mTOR) signaling networks plays a central role. Herein, we explored the effects of extracellular vesicles (EVs) released by gingiva-derived mesenchymal stem cells (GMSC-EVs) on oxidative stress-induced cellular senescence in human endothelial cells and skin fibroblasts and their antiaging potentials. Our results showed that GMSC-EVs robustly abrogated oxidative stress-induced upregulation in the expression of cellular senescence-related genes, such as β-galactosidase, p21, p53, and γH2AX, and mTOR/pS6 signaling pathway, in human umbilical vein endothelial cells (HUVECs) and skin fibroblasts. Meanwhile, GMSC-EVs restored oxidative stress-induced impairment in proliferation and tube formation by HUVECs. Systemic administration of GMSC-EVs attenuated aging-associated elevation in the expression levels of p21, mTOR/pS6, interleukin 6, and tumor necrosis factor α in skin and heart tissues of aged mice. These findings suggest that GMSC-EVs could be a potential alternative source of cell-free product for attenuation of aging-related skin and vascular dysfunctions due to their potent inhibitory effects on oxidative stress-induced cellular senescence in endothelial cells and skin fibroblasts.
Collapse
Affiliation(s)
- H Z Shi
- Department of Oral and Maxillofacial Surgery and Pharmacology, University of Pennsylvania School of Dental Medicine, Philadelphia, PA, USA
- Health Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | - J C Zeng
- Department of Oral and Maxillofacial Surgery and Pharmacology, University of Pennsylvania School of Dental Medicine, Philadelphia, PA, USA
- Dongguan Key Laboratory of Medical Bioactive Molecular Developmental and Translational Research, Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Guangdong Medical University, Dongguan, China
| | - S H Shi
- Department of Oral and Maxillofacial Surgery and Pharmacology, University of Pennsylvania School of Dental Medicine, Philadelphia, PA, USA
| | - H Giannakopoulos
- Department of Oral and Maxillofacial Surgery and Pharmacology, University of Pennsylvania School of Dental Medicine, Philadelphia, PA, USA
- Department of Oral & Maxillofacial Surgery, Penn Medicine Hospital of the University of Pennsylvania, Perelman Center for Advanced Medicine, Philadelphia, PA, USA
| | - Q Z Zhang
- Department of Oral and Maxillofacial Surgery and Pharmacology, University of Pennsylvania School of Dental Medicine, Philadelphia, PA, USA
| | - A D Le
- Department of Oral and Maxillofacial Surgery and Pharmacology, University of Pennsylvania School of Dental Medicine, Philadelphia, PA, USA
- Department of Oral & Maxillofacial Surgery, Penn Medicine Hospital of the University of Pennsylvania, Perelman Center for Advanced Medicine, Philadelphia, PA, USA
| |
Collapse
|
168
|
Spatio-temporal correlates of gene expression and cortical morphology across lifespan and aging. Neuroimage 2020; 224:117426. [PMID: 33035668 DOI: 10.1016/j.neuroimage.2020.117426] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Revised: 09/29/2020] [Accepted: 09/29/2020] [Indexed: 11/23/2022] Open
Abstract
Evidence from neuroimaging and genetic studies supports the concept that brain aging mirrors development. However, it is unclear whether mechanisms linking brain development and aging provide new insights to delay aging and potentially reverse it. This study determined biological mechanisms and phenotypic traits underpinning brain alterations across the lifespan and in aging by examining spatio-temporal correlations between gene expression and cortical volumes using datasets d with the age range from 2 to 82 years. We revealed that a large proportion of genes whose expression was associated with cortical volumes across the lifespan were in astrocytes. These genes, which showed up-regulation during development and down-regulation during aging, contributed to fundamental homeostatic functions of astrocytes. Included among these genes were those encoding components of cAMP, Ras, and retrograde endocannabinoid signaling pathways. Genes associated with cortical volumes in the same data aged above 55 years were also enriched for the sphingolipid, renin-angiotensin system (RAS), proteasome, and TGF-β signaling pathway, which is linked to senescence-associated secretory phenotypes. Neuroticism, drinking, and smoking were the common phenotypic traits in the lifespan and aging, while memory was the unique phenotype associated with aging. These findings provide biological mechanisms mirroring development and aging as well as unique to aging.
Collapse
|
169
|
Feng M, Peng H, Yao R, Zhang Z, Mao G, Yu H, Qiu Y. Inhibition of cellular communication network factor 1 (CCN1)-driven senescence slows down cartilage inflammaging and osteoarthritis. Bone 2020; 139:115522. [PMID: 32622876 DOI: 10.1016/j.bone.2020.115522] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Revised: 06/16/2020] [Accepted: 06/29/2020] [Indexed: 12/23/2022]
Abstract
OBJECTIVE To explore the role of cellular communication network factor 1 (CCN1) in cartilage inflammaging and osteoarthritis (OA) pathogenesis in the isolated primary human chondrocytes in vitro, cartilage explants ex vivo, and a pre-clinical mice model. METHODS Recombinant human CCN1 stimulation and small interfering RNA inhibition were conducted in human chondrocytes. The RNA was extracted to quantify catabolic targets and pro-inflammatory genes and the proteins were probed with specific antibodies. IL-1β and IL-6 were monitored by ELISA. IHC was performed to evaluate important hypertrophic hallmarks and catabolic markers. The effects of Tanshinone IIA on chondrocytes were investigated in both time-dependent and dose-dependent processes. Cartilage explants were cultured in growth medium and further treated with Tanshinone IIA. The intra-articular injection was performed in 13 months old C57BL/6J mice. Safranin O and fast green staining were performed to evaluate the histological change of cartilage followed by a semi-quantitative analysis using the OARSI scoring system. RESULTS RNA and protein levels of CCN1 increased in an age-dependent manner compared to young donors. Increased CCN1 expression was also found in the damaged area compared to the non-lesion area which correlated with the advanced pathological change in human OA. The overexpression of CCN1 promoted chondrocytes senescence, while the down-regulation of CCN1 by small interfering RNA reduced CCN1 production and limited inflammation secretion suggesting that CCN1 was a possible novel target to intervene OA. Inhibition of CCN1 by using Tanshinone IIA could reduce SASP components in a dose- and time-dependent manner. Additionally, our data showed that Tanshinone IIA was able to preserve articular cartilage integrity, suppress CCN1 production, and inhibit SASP factors in human cartilage explants and in aged mice model. CONCLUSION This study showed that CCN1 signaling aggravated cartilage inflammaing and matrix degradation. Collectively, our findings showed new insight into repurposing Tanshinone IIA for slowing down OA advancement in human and mice by inhibiting the CCN1 axis.
Collapse
Affiliation(s)
- Meng Feng
- Department of Orthopedics, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, People's Republic of China.
| | - Hang Peng
- Department of Orthopedics, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, People's Republic of China
| | - Ricky Yao
- Department of Orthopedics, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, People's Republic of China; Department of Orthopedics, University of Virginia, Charlottesville 22908, USA
| | - Zhifeng Zhang
- Department of Orthopedics, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, People's Republic of China; Department of Joint Surgery, the Second Affiliated Hospital of Inner Mongolia Medical University, 010030 Hohhot, People's Republic of China
| | - Genwen Mao
- Department of Orthopedics, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, People's Republic of China
| | - Haiquan Yu
- Department of Orthopedics, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, People's Republic of China
| | - Yusheng Qiu
- Department of Orthopedics, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, People's Republic of China.
| |
Collapse
|
170
|
Madreiter-Sokolowski CT, Thomas C, Ristow M. Interrelation between ROS and Ca 2+ in aging and age-related diseases. Redox Biol 2020; 36:101678. [PMID: 32810740 PMCID: PMC7451758 DOI: 10.1016/j.redox.2020.101678] [Citation(s) in RCA: 219] [Impact Index Per Article: 43.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Revised: 07/26/2020] [Accepted: 08/05/2020] [Indexed: 02/07/2023] Open
Abstract
Calcium (Ca2+) and reactive oxygen species (ROS) are versatile signaling molecules coordinating physiological and pathophysiological processes. While channels and pumps shuttle Ca2+ ions between extracellular space, cytosol and cellular compartments, short-lived and highly reactive ROS are constantly generated by various production sites within the cell. Ca2+ controls membrane potential, modulates mitochondrial adenosine triphosphate (ATP) production and affects proteins like calcineurin (CaN) or calmodulin (CaM), which, in turn, have a wide area of action. Overwhelming Ca2+ levels within mitochondria efficiently induce and trigger cell death. In contrast, ROS comprise a diverse group of relatively unstable molecules with an odd number of electrons that abstract electrons from other molecules to gain stability. Depending on the type and produced amount, ROS act either as signaling molecules by affecting target proteins or as harmful oxidative stressors by damaging cellular components. Due to their wide range of actions, it is little wonder that Ca2+ and ROS signaling pathways overlap and impact one another. Growing evidence suggests a crucial implication of this mutual interplay on the development and enhancement of age-related disorders, including cardiovascular and neurodegenerative diseases as well as cancer.
Collapse
Affiliation(s)
- Corina T Madreiter-Sokolowski
- Energy Metabolism Laboratory, Institute of Translational Medicine, Department of Health Sciences and Technology, Swiss Federal Institute of Technology (ETH), Zurich, Switzerland; Holder of an Erwin Schroedinger Abroad Fellowship, Austrian Science Fund (FWF), Austria.
| | - Carolin Thomas
- Energy Metabolism Laboratory, Institute of Translational Medicine, Department of Health Sciences and Technology, Swiss Federal Institute of Technology (ETH), Zurich, Switzerland
| | - Michael Ristow
- Energy Metabolism Laboratory, Institute of Translational Medicine, Department of Health Sciences and Technology, Swiss Federal Institute of Technology (ETH), Zurich, Switzerland
| |
Collapse
|
171
|
Magariños M, Barajas-Azpeleta R, Varela-Nieto I, R Aburto M. Otic Neurogenesis Is Regulated by TGFβ in a Senescence-Independent Manner. Front Cell Neurosci 2020; 14:217. [PMID: 32973450 PMCID: PMC7461926 DOI: 10.3389/fncel.2020.00217] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Accepted: 06/18/2020] [Indexed: 01/10/2023] Open
Abstract
Cellular senescence has classically been associated with aging. Intriguingly, recent studies have also unraveled key roles for senescence in embryonic development, regeneration, and reprogramming. Developmental senescence has been reported during embryonic development in different organisms and structures, such as the endolymphatic duct during inner ear development of mammals and birds. However, there is no study addressing the possible role of senescence on otic neurogenesis. TGFβ/SMAD is the best-known pathway linked to the induction of developmentally programmed cell senescence. Here, we studied if TGFβ2 induces cellular senescence during acoustic-vestibular-ganglion (AVG) formation. Using organotypic cultures of AVG, and characterizing different stages of otic neurogenesis in the presence of TGFβ2 and a selective TGF-β receptor type-I inhibitor, we show that TGFβ2 exerts a powerful action in inner ear neurogenesis but, contrary to what we recently observed during endolymphatic duct development, these actions are independent of cellular senescence. We show that TGFβ2 reduces proliferation, and induces differentiation and neuritogenesis of neuroblasts, without altering cell death. Our studies highlight the roles of TGFβ2 and cellular senescence in the precise regulation of cell fate within the developing inner ear and its different cell types, being their mechanisms of action highly cell-type dependent.
Collapse
Affiliation(s)
- Marta Magariños
- Institute for Biomedical Research "Alberto Sols" (IIBM), Spanish National Research Council-Autonomous University of Madrid (CSIC-UAM), Madrid, Spain.,Centre for Biomedical Network Research (CIBER) on Rare Diseases (CIBERER), Institute of Health Carlos III, Madrid, Spain.,Department of Biology, Universidad Autónoma de Madrid, Madrid, Spain
| | - Raquel Barajas-Azpeleta
- Institute for Biomedical Research "Alberto Sols" (IIBM), Spanish National Research Council-Autonomous University of Madrid (CSIC-UAM), Madrid, Spain.,Department of Biology, Universidad Autónoma de Madrid, Madrid, Spain
| | - Isabel Varela-Nieto
- Institute for Biomedical Research "Alberto Sols" (IIBM), Spanish National Research Council-Autonomous University of Madrid (CSIC-UAM), Madrid, Spain.,Centre for Biomedical Network Research (CIBER) on Rare Diseases (CIBERER), Institute of Health Carlos III, Madrid, Spain.,Hospital La Paz Institute for Health Research (IdiPAZ), Madrid, Spain
| | - Maria R Aburto
- Institute for Biomedical Research "Alberto Sols" (IIBM), Spanish National Research Council-Autonomous University of Madrid (CSIC-UAM), Madrid, Spain.,APC Microbiome Ireland, University College Cork, Cork, Ireland
| |
Collapse
|
172
|
Wilkinson HN, Hardman MJ. Senescence in Wound Repair: Emerging Strategies to Target Chronic Healing Wounds. Front Cell Dev Biol 2020; 8:773. [PMID: 32850866 PMCID: PMC7431694 DOI: 10.3389/fcell.2020.00773] [Citation(s) in RCA: 102] [Impact Index Per Article: 20.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Accepted: 07/22/2020] [Indexed: 01/10/2023] Open
Abstract
Cellular senescence is a fundamental stress response that restrains tumour formation. Yet, senescence cells are also present in non-cancerous states, accumulating exponentially with chronological age and contributing to age- and diabetes-related cellular dysfunction. The identification of hypersecretory and phagocytic behaviours in cells that were once believed to be non-functional has led to a recent explosion of senescence research. Here we discuss the profound, and often opposing, roles identified for short-lived vs. chronic tissue senescence. Transiently induced senescence is required for development, regeneration and acute wound repair, while chronic senescence is widely implicated in tissue pathology. We recently demonstrated that sustained senescence contributes to impaired diabetic healing via the CXCR2 receptor, which when blocked promotes repair. Further studies have highlighted the beneficial effects of targeting a range of senescence-linked processes to fight disease. Collectively, these findings hold promise for developing clinically viable strategies to tackle senescence in chronic wounds and other cutaneous pathologies.
Collapse
Affiliation(s)
- Holly N Wilkinson
- Centre for Atherothrombosis and Metabolic Disease, Hull York Medical School, University of Hull, Hull, United Kingdom
| | - Matthew J Hardman
- Centre for Atherothrombosis and Metabolic Disease, Hull York Medical School, University of Hull, Hull, United Kingdom
| |
Collapse
|
173
|
DNA Damage Response and Metabolic Reprogramming in Health and Disease. Trends Genet 2020; 36:777-791. [PMID: 32684438 DOI: 10.1016/j.tig.2020.06.018] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Revised: 06/29/2020] [Accepted: 06/30/2020] [Indexed: 01/08/2023]
Abstract
Nuclear DNA damage contributes to cellular malfunction and the premature onset of age-related diseases, including cancer. Until recently, the canonical DNA damage response (DDR) was thought to represent a collection of nuclear processes that detect, signal and repair damaged DNA. However, recent evidence suggests that beyond nuclear events, the DDR rewires an intricate network of metabolic circuits, fine-tunes protein synthesis, trafficking, and secretion as well as balances growth with defense strategies in response to genotoxic insults. In this review, we discuss how the active DDR signaling mobilizes extranuclear and systemic responses to promote cellular homeostasis and organismal survival in health and disease.
Collapse
|
174
|
Tammaro A, Kers J, Scantlebery AML, Florquin S. Metabolic Flexibility and Innate Immunity in Renal Ischemia Reperfusion Injury: The Fine Balance Between Adaptive Repair and Tissue Degeneration. Front Immunol 2020; 11:1346. [PMID: 32733450 PMCID: PMC7358591 DOI: 10.3389/fimmu.2020.01346] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2019] [Accepted: 05/27/2020] [Indexed: 01/10/2023] Open
Abstract
Renal ischemia reperfusion injury (IRI), a common event after renal transplantation, causes acute kidney injury (AKI), increases the risk of delayed graft function (DGF), primes the donor kidney for rejection, and contributes to the long-term risk of graft loss. In the last decade, epidemiological studies have linked even mild episodes of AKI to chronic kidney disease (CKD) progression, and innate immunity seems to play a crucial role. The ischemic insult triggers an acute inflammatory reaction that is elicited by Pattern Recognition Receptors (PRRs), expressed on both infiltrating immune cells as well as tubular epithelial cells (TECs). Among the PRRs, Toll-like receptors (TLRs), their synergistic receptors, Nod-like receptors (NLRs), and the inflammasomes, play a pivotal role in shaping inflammation and TEC repair, in response to renal IRI. These receptors represent promising targets to modulate the extent of inflammation, but also function as gatekeepers of tissue repair, protecting against AKI-to-CKD progression. Despite the important considerations on timely use of therapeutics, in the context of IRI, treatment options are limited by a lack of understanding of the intra- and intercellular mechanisms associated with the activation of innate immune receptors and their impact on adaptive tubular repair. Accumulating evidence suggests that TEC-associated innate immunity shapes the tubular response to stress through the regulation of immunometabolism. Engagement of innate immune receptors provides TECs with the metabolic flexibility necessary for their plasticity during injury and repair. This could significantly affect pathogenic processes within TECs, such as cell death, mitochondrial damage, senescence, and pro-fibrotic cytokine secretion, well-known to exacerbate inflammation and fibrosis. This article provides an overview of the past 5 years of research on the role of innate immunity in experimental and human IRI, with a focus on the cascade of events activated by hypoxic damage in TECs: from programmed cell death (PCD) and mitochondrial dysfunction-mediated metabolic rewiring of TECs to maladaptive repair and progression to fibrosis. Finally, we will discuss the important crosstalk between metabolism and innate immunity observed in TECs and their therapeutic potential in both experimental and clinical research.
Collapse
Affiliation(s)
- Alessandra Tammaro
- Department of Pathology, Amsterdam UMC, Amsterdam Infection & Immunity Institute, University of Amsterdam, Amsterdam, Netherlands
| | - Jesper Kers
- Department of Pathology, Amsterdam UMC, Amsterdam Infection & Immunity Institute, University of Amsterdam, Amsterdam, Netherlands.,Department of Pathology, Leiden University Medical Center, Leiden, Netherlands.,Biomolecular Systems Analytics, Van 't Hoff Institute for Molecular Sciences (HIMS), University of Amsterdam, Amsterdam, Netherlands
| | - Angelique M L Scantlebery
- Department of Pathology, Amsterdam UMC, Amsterdam Infection & Immunity Institute, University of Amsterdam, Amsterdam, Netherlands
| | - Sandrine Florquin
- Department of Pathology, Amsterdam UMC, Amsterdam Infection & Immunity Institute, University of Amsterdam, Amsterdam, Netherlands.,Department of Pathology, Leiden University Medical Center, Leiden, Netherlands
| |
Collapse
|
175
|
Cianflone E, Torella M, Biamonte F, De Angelis A, Urbanek K, Costanzo FS, Rota M, Ellison-Hughes GM, Torella D. Targeting Cardiac Stem Cell Senescence to Treat Cardiac Aging and Disease. Cells 2020; 9:E1558. [PMID: 32604861 PMCID: PMC7349658 DOI: 10.3390/cells9061558] [Citation(s) in RCA: 80] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 06/19/2020] [Accepted: 06/25/2020] [Indexed: 12/13/2022] Open
Abstract
Adult stem/progenitor are a small population of cells that reside in tissue-specific niches and possess the potential to differentiate in all cell types of the organ in which they operate. Adult stem cells are implicated with the homeostasis, regeneration, and aging of all tissues. Tissue-specific adult stem cell senescence has emerged as an attractive theory for the decline in mammalian tissue and organ function during aging. Cardiac aging, in particular, manifests as functional tissue degeneration that leads to heart failure. Adult cardiac stem/progenitor cell (CSC) senescence has been accordingly associated with physiological and pathological processes encompassing both non-age and age-related decline in cardiac tissue repair and organ dysfunction and disease. Senescence is a highly active and dynamic cell process with a first classical hallmark represented by its replicative limit, which is the establishment of a stable growth arrest over time that is mainly secondary to DNA damage and reactive oxygen species (ROS) accumulation elicited by different intrinsic stimuli (like metabolism), as well as external stimuli and age. Replicative senescence is mainly executed by telomere shortening, the activation of the p53/p16INK4/Rb molecular pathways, and chromatin remodeling. In addition, senescent cells produce and secrete a complex mixture of molecules, commonly known as the senescence-associated secretory phenotype (SASP), that regulate most of their non-cell-autonomous effects. In this review, we discuss the molecular and cellular mechanisms regulating different characteristics of the senescence phenotype and their consequences for adult CSCs in particular. Because senescent cells contribute to the outcome of a variety of cardiac diseases, including age-related and unrelated cardiac diseases like diabetic cardiomyopathy and anthracycline cardiotoxicity, therapies that target senescent cell clearance are actively being explored. Moreover, the further understanding of the reversibility of the senescence phenotype will help to develop novel rational therapeutic strategies.
Collapse
Affiliation(s)
- Eleonora Cianflone
- Department of Medical and Surgical Sciences, Magna Graecia University, 88100 Catanzaro, Italy;
| | - Michele Torella
- Department of Translational Medical Sciences, AORN dei Colli/Monaldi Hospital, University of Campania “L. Vanvitelli”, Via Leonardo Bianchi, 80131 Naples, Italy;
| | - Flavia Biamonte
- Department of Experimental and Clinical Medicine and Interdepartmental Centre of Services (CIS), Magna Graecia University, 88100 Catanzaro, Italy; (F.B.); (F.S.C.)
| | - Antonella De Angelis
- Department of Experimental Medicine, Section of Pharmacology, University of Campania “L.Vanvitelli”, 80121 Naples, Italy;
| | - Konrad Urbanek
- Molecular and Cellular Cardiology, Department of Experimental and Clinical Medicine, Magna Graecia University, 88100 Catanzaro, Italy
| | - Francesco S. Costanzo
- Department of Experimental and Clinical Medicine and Interdepartmental Centre of Services (CIS), Magna Graecia University, 88100 Catanzaro, Italy; (F.B.); (F.S.C.)
| | - Marcello Rota
- Department of Physiology, New York Medical College, Valhalla, NY 10595, USA;
| | - Georgina M. Ellison-Hughes
- Centre for Human and Applied Physiological Sciences and Centre for Stem Cells and Regenerative Medicine, School of Basic and Medical Biosciences, Faculty of Life Sciences & Medicine, King’s College London, Guys Campus-Great Maze Pond rd, London SE1 1UL, UK;
| | - Daniele Torella
- Molecular and Cellular Cardiology, Department of Experimental and Clinical Medicine, Magna Graecia University, 88100 Catanzaro, Italy
| |
Collapse
|
176
|
|
177
|
Docherty MH, Baird DP, Hughes J, Ferenbach DA. Cellular Senescence and Senotherapies in the Kidney: Current Evidence and Future Directions. Front Pharmacol 2020; 11:755. [PMID: 32528288 PMCID: PMC7264097 DOI: 10.3389/fphar.2020.00755] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Accepted: 05/06/2020] [Indexed: 01/10/2023] Open
Abstract
Cellular senescence refers to a cellular phenotype characterized by an altered transcriptome, pro-inflammatory secretome, and generally irreversible growth arrest. Acutely senescent cells are widely recognized as performing key physiological functions in vivo promoting normal organogenesis, successful wound repair, and cancer defense. In contrast, the accumulation of chronically senescent cells in response to aging, cell stress, genotoxic damage, and other injurious stimuli is increasingly recognized as an important contributor to organ dysfunction, tissue fibrosis, and the more generalized aging phenotype. In this review, we summarize our current knowledge of the role of senescent cells in promoting progressive fibrosis and dysfunction with a particular focus on the kidney and reference to other organ systems. Specific differences between healthy and senescent cells are reviewed along with a summary of several experimental pharmacological approaches to deplete or manipulate senescent cells to preserve organ integrity and function with aging and after injury. Finally, key questions for future research and clinical translation are discussed.
Collapse
Affiliation(s)
- Marie Helena Docherty
- Department of Renal Medicine, Royal Infirmary of Edinburgh, Edinburgh, United Kingdom.,Centre for Inflammation Research, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
| | - David P Baird
- Department of Renal Medicine, Royal Infirmary of Edinburgh, Edinburgh, United Kingdom.,Centre for Inflammation Research, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
| | - Jeremy Hughes
- Department of Renal Medicine, Royal Infirmary of Edinburgh, Edinburgh, United Kingdom.,Centre for Inflammation Research, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
| | - David A Ferenbach
- Department of Renal Medicine, Royal Infirmary of Edinburgh, Edinburgh, United Kingdom.,Centre for Inflammation Research, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
178
|
Rosas-González VC, Téllez-Bañuelos MC, Hernández-Flores G, Bravo-Cuellar A, Aguilar-Lemarroy A, Jave-Suárez LF, Haramati J, Solorzano-Ibarra F, Ortiz-Lazareno PC. Differential effects of alliin and allicin on apoptosis and senescence in luminal A and triple-negative breast cancer: Caspase, ΔΨm, and pro-apoptotic gene involvement. Fundam Clin Pharmacol 2020; 34:671-686. [PMID: 32286702 DOI: 10.1111/fcp.12559] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Revised: 04/04/2020] [Accepted: 04/07/2020] [Indexed: 12/12/2022]
Abstract
Breast cancer is the most frequent cancer in women worldwide, and drug resistance is common in all breast cancer types. The combination of natural products with chemotherapies has attracted attention, as it was found that natural compounds enhance the effects of standard cancer chemotherapeutic drugs and protect from side effects. Into the different natural products, garlic has been recognized for its antitumor properties. It is suggested that its anticancer effects are associated with its organo-sulfur compounds, especially alliin and allicin. Here, we evaluated the effects of both molecules on cell death, senescence, and their senolytic potential in luminal A and triple-negative breast cancer cells. MCF-7 (luminal A) and HCC-70 (triple-negative) cells were cultured and treated with different concentrations of alliin or allicin. Then, cell viability was determined using the WST-1 reagent. Apoptosis and caspase activity were evaluated by flow cytometry; ΔΨm was assessed using a JC-10 fluorometric assay kit. Apoptosis-related genes were evaluated by RT-PCR. Proliferation was measured using bromodeoxyuridine incorporation. We also evaluated clonogenicity, senescence (β-Galactosidase Staining), and the senolytic effect of the compounds. Our results showed that allicin has antiproliferative, anticlonogenic, and senolytic effects. In addition, allicin decreased cell viability and induced apoptosis by loss of ΔΨm, caspase-3, caspase-8, and caspase-9 activation, upregulation of NOXA, P21, and BAK, as well as downregulation of BCL-XL expression. Contrary to allicin, alliin promoted clonogenicity, induced senescence, and did not exhibit pro-apoptotic effects in breast cancer cells.
Collapse
Affiliation(s)
- Vida Celeste Rosas-González
- División de Inmunología, Centro de Investigación Biomédica de Occidente (CIBO), Instituto Mexicano del Seguro Social (IMSS), Guadalajara, 44340, Jalisco, México.,Posgrado en Ciencias Biomédicas Orientación en Inmunología, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara, 44340, Jalico, México
| | - Martha Cecilia Téllez-Bañuelos
- Departamento de Biología Celular y Molecular, Centro Universitario de Ciencias Biológicas y Agropecuarias, Universidad de Guadalajara, Las Agujas, 45220, Jalisco, México
| | - Georgina Hernández-Flores
- División de Inmunología, Centro de Investigación Biomédica de Occidente (CIBO), Instituto Mexicano del Seguro Social (IMSS), Guadalajara, 44340, Jalisco, México
| | - Alejandro Bravo-Cuellar
- División de Inmunología, Centro de Investigación Biomédica de Occidente (CIBO), Instituto Mexicano del Seguro Social (IMSS), Guadalajara, 44340, Jalisco, México.,Centro Universitario de los Altos, Universidad de Guadalajara, Tepatitlán de Morelos, 47620, Jalisco, México
| | - Adriana Aguilar-Lemarroy
- División de Inmunología, Centro de Investigación Biomédica de Occidente (CIBO), Instituto Mexicano del Seguro Social (IMSS), Guadalajara, 44340, Jalisco, México
| | - Luis Felipe Jave-Suárez
- División de Inmunología, Centro de Investigación Biomédica de Occidente (CIBO), Instituto Mexicano del Seguro Social (IMSS), Guadalajara, 44340, Jalisco, México
| | - Jesse Haramati
- Departamento de Biología Celular y Molecular, Centro Universitario de Ciencias Biológicas y Agropecuarias, Universidad de Guadalajara, Las Agujas, 45220, Jalisco, México
| | - Fabiola Solorzano-Ibarra
- Posgrado en Ciencias Biomédicas Orientación en Inmunología, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara, 44340, Jalico, México
| | - Pablo Cesar Ortiz-Lazareno
- División de Inmunología, Centro de Investigación Biomédica de Occidente (CIBO), Instituto Mexicano del Seguro Social (IMSS), Guadalajara, 44340, Jalisco, México
| |
Collapse
|
179
|
Functional alterations and transcriptomic changes during zebrafish cardiac aging. Biogerontology 2020; 21:637-652. [PMID: 32372324 DOI: 10.1007/s10522-020-09881-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Accepted: 04/25/2020] [Indexed: 12/22/2022]
Abstract
Aging dramatically increases the risk of cardiovascular diseases in human. Animal models are of great value to study cardiac aging, and zebrafish have become a popular model for aging study recently. However, there is limited knowledge about the progression and regulation of cardiac aging in zebrafish. In this study we first validated the effectiveness of a panel of aging-related markers and revealed their spatial-temporal specificity. Using these markers, we discovered that cardiac aging in zebrafish initiated at mid-age around 24 months, followed by a gradual progression marked with increased DNA damage, inflammatory response and reduced mitochondrial function. Furthermore, we showed aging-related expression profile change in zebrafish hearts was similar to that in rat hearts. Overall, our results provide a deeper insight into the cardiac aging process in zebrafish, which will set up foundation for generating novel cardiac aging models suitable for large scale screening of pharmaceutical targets.
Collapse
|
180
|
Ullah K, Chen S, Lu J, Wang X, Liu Q, Zhang Y, Long Y, Hu Z, Xu G. The E3 ubiquitin ligase STUB1 attenuates cell senescence by promoting the ubiquitination and degradation of the core circadian regulator BMAL1. J Biol Chem 2020; 295:4696-4708. [PMID: 32041778 PMCID: PMC7135990 DOI: 10.1074/jbc.ra119.011280] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2019] [Revised: 02/06/2020] [Indexed: 01/10/2023] Open
Abstract
Cell senescence is one of the most important processes determining cell fate and is involved in many pathophysiological conditions, including cancer, neurodegenerative diseases, and other aging-associated diseases. It has recently been discovered that the E3 ubiquitin ligase STIP1 homology and U-box-containing protein 1 (STUB1 or CHIP) is up-regulated during the senescence of human fibroblasts and modulates cell senescence. However, the molecular mechanism underlying STUB1-controlled senescence is not clear. Here, using affinity purification and MS-based analysis, we discovered that STUB1 binds to brain and muscle ARNT-like 1 (BMAL1, also called aryl hydrocarbon receptor nuclear translocator-like protein 1 (ARNTL)). Through biochemical experiments, we confirmed the STUB1-BMAL1 interaction, identified their interaction domains, and revealed that STUB1 overexpression down-regulates BMAL1 protein levels through STUB1's enzymatic activity and that STUB1 knockdown increases BMAL1 levels. Further experiments disclosed that STUB1 enhances BMAL1 degradation, which is abolished upon proteasome inhibition. Moreover, we found that STUB1 promotes the formation of Lys-48-linked polyubiquitin chains on BMAL1, facilitating its proteasomal degradation. Interestingly, we also discovered that oxidative stress promotes STUB1 nuclear translocation and enhances its co-localization with BMAL1. STUB1 expression attenuates hydrogen peroxide-induced cell senescence, indicated by a reduced signal in senescence-associated β-gal staining and decreased protein levels of two cell senescence markers, p53 and p21. BMAL1 knockdown diminishes this effect, and BMAL1 overexpression abolishes STUB1's effect on cell senescence. In summary, the results of our work reveal that the E3 ubiquitin ligase STUB1 ubiquitinates and degrades its substrate BMAL1 and thereby alleviates hydrogen peroxide-induced cell senescence.
Collapse
Affiliation(s)
- Kifayat Ullah
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and College of Pharmaceutical Sciences, Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Soochow University, 199 Ren'ai Road, Suzhou, Jiangsu 215123, China
| | - Suping Chen
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and College of Pharmaceutical Sciences, Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Soochow University, 199 Ren'ai Road, Suzhou, Jiangsu 215123, China
| | - Jiaqi Lu
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and College of Pharmaceutical Sciences, Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Soochow University, 199 Ren'ai Road, Suzhou, Jiangsu 215123, China
| | - Xiaohui Wang
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and College of Pharmaceutical Sciences, Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Soochow University, 199 Ren'ai Road, Suzhou, Jiangsu 215123, China
| | - Qing Liu
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and College of Pharmaceutical Sciences, Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Soochow University, 199 Ren'ai Road, Suzhou, Jiangsu 215123, China
| | - Yang Zhang
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and College of Pharmaceutical Sciences, Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Soochow University, 199 Ren'ai Road, Suzhou, Jiangsu 215123, China
| | - Yaqiu Long
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and College of Pharmaceutical Sciences, Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Soochow University, 199 Ren'ai Road, Suzhou, Jiangsu 215123, China
| | - Zhanhong Hu
- Department of Pharmacy, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215004, China
| | - Guoqiang Xu
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and College of Pharmaceutical Sciences, Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Soochow University, 199 Ren'ai Road, Suzhou, Jiangsu 215123, China
| |
Collapse
|
181
|
Liu RM, Liu G. Cell senescence and fibrotic lung diseases. Exp Gerontol 2020; 132:110836. [PMID: 31958492 PMCID: PMC7036279 DOI: 10.1016/j.exger.2020.110836] [Citation(s) in RCA: 69] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Revised: 01/08/2020] [Accepted: 01/09/2020] [Indexed: 12/14/2022]
Abstract
Idiopathic pulmonary fibrosis (IPF) is a progressive fatal lung disorder with an unknown etiology and very limited therapeutic options. The incidence and severity of IPF increase with advanced age, suggesting that aging is a major risk factor for IPF. The mechanism underlying the aging-related susceptibility to IPF, however, remains unclear. Cellular senescence, a permanent arrest of cell growth, has been increasingly recognized as an important contributor to aging and aging-related diseases, including IPF. Senescent cells have been identified in IPF lungs and in experimental lung fibrosis models. Removal of senescent cells pharmacologically or genetically improves lung function and reverses pulmonary fibrosis induced by different stimuli in experimental fibrosis models. Treatment with senolytic drugs also improves clinical symptoms in IPF patients. These intriguing findings suggest that cellular senescence contributes importantly to the pathogenesis of fibrotic lung diseases and targeting senescent cells may represent a novel approach for the treatment of fibrotic lung disorders. In this mini review, we summarize the recent advance in the field regarding the role of cellular senescence in fibrotic lung diseases, with a focus on IPF.
Collapse
Affiliation(s)
- Rui-Ming Liu
- Division of Pulmonary, Allergy, and Critical Care, Department of Medicine, School of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA.
| | - Gang Liu
- Division of Pulmonary, Allergy, and Critical Care, Department of Medicine, School of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| |
Collapse
|
182
|
Herrmann M, Engelke K, Ebert R, Müller-Deubert S, Rudert M, Ziouti F, Jundt F, Felsenberg D, Jakob F. Interactions between Muscle and Bone-Where Physics Meets Biology. Biomolecules 2020; 10:biom10030432. [PMID: 32164381 PMCID: PMC7175139 DOI: 10.3390/biom10030432] [Citation(s) in RCA: 103] [Impact Index Per Article: 20.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Revised: 02/27/2020] [Accepted: 03/05/2020] [Indexed: 02/06/2023] Open
Abstract
Muscle and bone interact via physical forces and secreted osteokines and myokines. Physical forces are generated through gravity, locomotion, exercise, and external devices. Cells sense mechanical strain via adhesion molecules and translate it into biochemical responses, modulating the basic mechanisms of cellular biology such as lineage commitment, tissue formation, and maturation. This may result in the initiation of bone formation, muscle hypertrophy, and the enhanced production of extracellular matrix constituents, adhesion molecules, and cytoskeletal elements. Bone and muscle mass, resistance to strain, and the stiffness of matrix, cells, and tissues are enhanced, influencing fracture resistance and muscle power. This propagates a dynamic and continuous reciprocity of physicochemical interaction. Secreted growth and differentiation factors are important effectors of mutual interaction. The acute effects of exercise induce the secretion of exosomes with cargo molecules that are capable of mediating the endocrine effects between muscle, bone, and the organism. Long-term changes induce adaptations of the respective tissue secretome that maintain adequate homeostatic conditions. Lessons from unloading, microgravity, and disuse teach us that gratuitous tissue is removed or reorganized while immobility and inflammation trigger muscle and bone marrow fatty infiltration and propagate degenerative diseases such as sarcopenia and osteoporosis. Ongoing research will certainly find new therapeutic targets for prevention and treatment.
Collapse
Affiliation(s)
- Marietta Herrmann
- Orthopedic Department, Bernhard-Heine-Center for Locomotion Research, IZKF Research Group Tissue regeneration in musculoskeletal diseases, University Hospital Würzburg, University of Wuerzburg, 97070 Würzburg, Germany;
| | - Klaus Engelke
- Department of Medicine 3, FAU University Erlangen-Nürnberg and Universitätsklinikum Erlangen, 91054 Erlangen, Germany;
| | - Regina Ebert
- Orthopedic Department, Bernhard-Heine-Center for Locomotion Research, University of Würzburg, IGZ, 97076 Würzburg, Germany; (R.E.)
| | - Sigrid Müller-Deubert
- Orthopedic Department, Bernhard-Heine-Center for Locomotion Research, University of Würzburg, IGZ, 97076 Würzburg, Germany; (R.E.)
| | - Maximilian Rudert
- Orthopedic Department, Bernhard-Heine-Center for Locomotion Research, University of Würzburg, 97074 Würzburg, Germany;
| | - Fani Ziouti
- Department of Internal Medicine II, University Hospital Würzburg, 97080 Würzburg, Germany; (F.Z.); (F.J.)
| | - Franziska Jundt
- Department of Internal Medicine II, University Hospital Würzburg, 97080 Würzburg, Germany; (F.Z.); (F.J.)
| | - Dieter Felsenberg
- Privatpraxis für Muskel- und Knochenkrankheiten, 12163 Berlin Germany;
| | - Franz Jakob
- Orthopedic Department, Bernhard-Heine-Center for Locomotion Research, University of Würzburg, IGZ, 97076 Würzburg, Germany; (R.E.)
- Orthopedic Department, Bernhard-Heine-Center for Locomotion Research, University of Würzburg, 97074 Würzburg, Germany;
- Correspondence:
| |
Collapse
|
183
|
The Emerging Role of Senescence in Ocular Disease. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:2583601. [PMID: 32215170 PMCID: PMC7085400 DOI: 10.1155/2020/2583601] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Accepted: 02/14/2020] [Indexed: 02/07/2023]
Abstract
Cellular senescence is a state of irreversible cell cycle arrest in response to an array of cellular stresses. An important role for senescence has been shown for a number of pathophysiological conditions that include cardiovascular disease, pulmonary fibrosis, and diseases of the skin. However, whether senescence contributes to the progression of age-related macular degeneration (AMD) has not been studied in detail so far and the present review describes the recent research on this topic. We present an overview of the types of senescence, pathways of senescence, senescence-associated secretory phenotype (SASP), the role of mitochondria, and their functional implications along with antisenescent therapies. As a central mechanism, senescent cells can impact the surrounding tissue microenvironment via the secretion of a pool of bioactive molecules, termed the SASP. An updated summary of a number of new members of the ever-growing SASP family is presented. Further, we introduce the significance of mechanisms by which mitochondria may participate in the development of cellular senescence. Emerging evidence shows that extracellular vesicles (EVs) are important mediators of the effects of senescent cells on their microenvironment. Based on recent studies, there is reasonable evidence that senescence could be a modifiable factor, and hence, it may be possible to delay age-related diseases by modulating basic aging mechanisms using SASP inhibitors/senolytic drugs. Thus, antisenescent therapies in aging and age-related diseases appear to have a promising potential.
Collapse
|
184
|
Wang Y, Kumaishi K, Suzuki T, Ichihashi Y, Yamaguchi N, Shirakawa M, Ito T. Morphological and Physiological Framework Underlying Plant Longevity in Arabidopsis thaliana. FRONTIERS IN PLANT SCIENCE 2020; 11:600726. [PMID: 33224176 PMCID: PMC7674609 DOI: 10.3389/fpls.2020.600726] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Accepted: 10/09/2020] [Indexed: 05/09/2023]
Abstract
Monocarpic plants have a single reproductive phase, in which their longevity is developmentally programmed by molecular networks. In the reproductive phase of Arabidopsis thaliana, the inflorescence meristem (IM) maintains a central pool of stem cells and produces a limited number of flower primordia, which result in seed formation and the death of the whole plant. In this study, we observed morphological changes in the IM at cellular and intracellular resolutions until the end of the plant life cycle. We observed four biological events during the periods from 1 week after bolting (WAB) till the death of stem cells: (1) the gradual reduction in the size of the IM, (2) the dynamic vacuolation of IM cells, (3) the loss of the expression of the stem cell determinant WUSCHEL (WUS), and (4) the upregulation of the programmed cell death marker BIFUNCTIONAL NUCLEASE1 (BFN1) in association with the death of stem cells. These results indicate that the stem cell population gradually decreases in IM during plant aging and eventually is fully terminated. We further show that the expression of WUS became undetectable in IM at 3 WAB prior to the loss of CLAVATA3 (CLV3) expression at 5 WAB; CLV3 is a negative regulator of WUS. Moreover, clv3 plants showed delayed loss of WUS and lived 6 weeks longer compared with wild-type plants. These results indicated that the prolonged expression of CLV3 at 4-5 WAB may be a safeguard that inhibits the reactivation of WUS and promotes plant death. Finally, through transcriptome analysis, we determined that reactive oxygen species (ROS) are involved in the control of plant longevity. Our work presents a morphological and physiological framework for the regulation of plant longevity in Arabidopsis.
Collapse
Affiliation(s)
- Yukun Wang
- Division of Biological Science, Graduate School of Science and Technology, Nara Institute of Science and Technology, Ikoma, Japan
| | - Kie Kumaishi
- RIKEN BioResource Research Center, Tsukuba, Japan
| | - Takamasa Suzuki
- Department of Biological Chemistry, College of Bioscience and Biotechnology, Chubu University, Kasugai, Japan
| | - Yasunori Ichihashi
- RIKEN BioResource Research Center, Tsukuba, Japan
- Precursory Research for Embryonic Science and Technology, Japan Science and Technology Agency, Kawaguchi, Japan
| | - Nobutoshi Yamaguchi
- Division of Biological Science, Graduate School of Science and Technology, Nara Institute of Science and Technology, Ikoma, Japan
- Precursory Research for Embryonic Science and Technology, Japan Science and Technology Agency, Kawaguchi, Japan
| | - Makoto Shirakawa
- Division of Biological Science, Graduate School of Science and Technology, Nara Institute of Science and Technology, Ikoma, Japan
- *Correspondence: Makoto Shirakawa,
| | - Toshiro Ito
- Division of Biological Science, Graduate School of Science and Technology, Nara Institute of Science and Technology, Ikoma, Japan
- Toshiro Ito,
| |
Collapse
|
185
|
Fu Y, Dong T, Tan L, Yin D, Zhang M, Zhao G, Ye M, Wu R. Identification of Shoot Differentiation-Related Genes in Populus euphratica Oliv. Genes (Basel) 2019; 10:genes10121034. [PMID: 31835855 PMCID: PMC6947848 DOI: 10.3390/genes10121034] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Revised: 11/25/2019] [Accepted: 12/09/2019] [Indexed: 12/30/2022] Open
Abstract
De novo shoot regeneration is one of the important manifestations of cell totipotency in organogenesis, which reflects a survival strategy organism evolved when facing natural selection. Compared with tissue regeneration, and somatic embryogenesis, de novo shoot regeneration denotes a shoot regeneration process directly from detatched or injured tissues of plant. Studies on plant shoot regeneration had identified key genes mediating shoot regeneration. However, knowledge was derived from Arabidopsis; the regeneration capacity is hugely distinct among species. To achieve a comprehensive understanding of the shoot regeneration mechanism from tree species, we select four genetic lines of Populus euphratica from a natural population to be sequenced at transcriptome level. On the basis of the large difference of differentiation capacity, between the highly differentiated (HD) and low differentiated (LD) groups, the analysis of differential expression identified 4920 differentially expressed genes (DEGs), which were revealed in five groups of expression patterns by clustering analysis. Enrichment showed crucial pathways involved in regulation of regeneration difference, including “plant hormone signal transduction”, “cell differentiation”, "cellular response to auxin stimulus", and “auxin-activated signaling pathway”. The expression of nine genes reported to be associated with shoot regeneration was validated using quantitative real-time PCR (qRT-PCR). For the specificity of regeneration mechanism with P. euphratica, large amount of DEGs involved in "plant-pathogen interaction", ubiquitin-26S proteosome mediated proteolysis pathway, stress-responsive DEGs, and senescence-associated DEGs were summarized to possibly account for the differentiation difference with distinct genotypes of P. euphratica. The result in this study helps screening of key regulators in mediating the shoot differentiation. The transcriptomic characteristic in P. euphratica further enhances our understanding of key processes affecting the regeneration capacity of de novo shoots among distinct species.
Collapse
Affiliation(s)
- Yaru Fu
- Center for Computational Biology, College of Biological Sciences and Technology, Beijing Forestry University, Beijing 100083, China; (Y.F.); (T.D.); (L.T.); (D.Y.); (M.Z.); (G.Z.); (R.W.)
| | - Tianyu Dong
- Center for Computational Biology, College of Biological Sciences and Technology, Beijing Forestry University, Beijing 100083, China; (Y.F.); (T.D.); (L.T.); (D.Y.); (M.Z.); (G.Z.); (R.W.)
| | - Lizhi Tan
- Center for Computational Biology, College of Biological Sciences and Technology, Beijing Forestry University, Beijing 100083, China; (Y.F.); (T.D.); (L.T.); (D.Y.); (M.Z.); (G.Z.); (R.W.)
| | - Danni Yin
- Center for Computational Biology, College of Biological Sciences and Technology, Beijing Forestry University, Beijing 100083, China; (Y.F.); (T.D.); (L.T.); (D.Y.); (M.Z.); (G.Z.); (R.W.)
| | - Miaomiao Zhang
- Center for Computational Biology, College of Biological Sciences and Technology, Beijing Forestry University, Beijing 100083, China; (Y.F.); (T.D.); (L.T.); (D.Y.); (M.Z.); (G.Z.); (R.W.)
| | - Guomiao Zhao
- Center for Computational Biology, College of Biological Sciences and Technology, Beijing Forestry University, Beijing 100083, China; (Y.F.); (T.D.); (L.T.); (D.Y.); (M.Z.); (G.Z.); (R.W.)
| | - Meixia Ye
- Center for Computational Biology, College of Biological Sciences and Technology, Beijing Forestry University, Beijing 100083, China; (Y.F.); (T.D.); (L.T.); (D.Y.); (M.Z.); (G.Z.); (R.W.)
- Correspondence: ; Tel.: +86-10-6233-7061
| | - Rongling Wu
- Center for Computational Biology, College of Biological Sciences and Technology, Beijing Forestry University, Beijing 100083, China; (Y.F.); (T.D.); (L.T.); (D.Y.); (M.Z.); (G.Z.); (R.W.)
- Center for Statistical Genetics, The Pennsylvania State University, Hershey, PA 17033, USA
| |
Collapse
|