1
|
Mannarino M, Cherif H, Ghazizadeh S, Martinez OW, Sheng K, Cousineau E, Lee S, Millecamps M, Gao C, Gilbert A, Peirs C, Naeini RS, Ouellet JA, S Stone L, Haglund L. Senolytic treatment for low back pain. SCIENCE ADVANCES 2025; 11:eadr1719. [PMID: 40085710 PMCID: PMC11908501 DOI: 10.1126/sciadv.adr1719] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Accepted: 02/07/2025] [Indexed: 03/16/2025]
Abstract
Senescent cells (SnCs) accumulate because of aging and external cellular stress throughout the body. They adopt a senescence-associated secretory phenotype (SASP) and release inflammatory and degenerative factors that actively contribute to age-related diseases, such as low back pain (LBP). The senolytics, o-vanillin and RG-7112, remove SnCs in human intervertebral discs (IVDs) and reduce SASP release, but it is unknown whether they can treat LBP. sparc-/- mice, with LBP, were treated orally with o-vanillin and RG-7112 as single or combination treatments. Treatment reduced LBP and SASP factor release and removed SnCs from the IVD and spinal cord. Treatment also lowered degeneration scores in the IVDs, improved vertebral bone quality, and reduced the expression of pain markers in the spinal cord. Together, our data suggest RG-7112 and o-vanillin as potential disease-modifying drugs for LBP and other painful disorders linked to cell senescence.
Collapse
Affiliation(s)
- Matthew Mannarino
- Department of Surgery, Orthopaedic Research Lab, McGill University, Montreal, QC, Canada
- ABC-platform (Animal Behavioral Characterization) at the Alan Edwards Centre for Research on Pain, McGill University, Montreal, QC, Canada
- Alan Edwards Centre for Research on Pain (AECRP), McGill University, Montreal, QC, Canada
| | - Hosni Cherif
- Department of Surgery, Orthopaedic Research Lab, McGill University, Montreal, QC, Canada
- Department of Surgery, McGill Scoliosis and Spine Group, McGill University, Montreal, QC, Canada
| | - Saber Ghazizadeh
- Department of Surgery, Orthopaedic Research Lab, McGill University, Montreal, QC, Canada
| | - Oliver Wu Martinez
- Department of Surgery, Orthopaedic Research Lab, McGill University, Montreal, QC, Canada
| | - Kai Sheng
- Department of Surgery, Orthopaedic Research Lab, McGill University, Montreal, QC, Canada
- Shriner's Hospital for Children, Montreal, QC, Canada
| | - Elsa Cousineau
- Department of Surgery, Orthopaedic Research Lab, McGill University, Montreal, QC, Canada
| | - Seunghwan Lee
- ABC-platform (Animal Behavioral Characterization) at the Alan Edwards Centre for Research on Pain, McGill University, Montreal, QC, Canada
- Alan Edwards Centre for Research on Pain (AECRP), McGill University, Montreal, QC, Canada
- Department of Anesthesiology, University of Minnesota, Minneapolis, MN, USA
| | - Magali Millecamps
- ABC-platform (Animal Behavioral Characterization) at the Alan Edwards Centre for Research on Pain, McGill University, Montreal, QC, Canada
- Alan Edwards Centre for Research on Pain (AECRP), McGill University, Montreal, QC, Canada
| | - Chan Gao
- Division of Physiatry, Department of Medicine, McGill University, Montreal, QC, Canada
| | - Alice Gilbert
- Alan Edwards Centre for Research on Pain (AECRP), McGill University, Montreal, QC, Canada
- Department of Physiology and Cell Information Systems, McGill University, Montreal, QC, Canada
- Université Clermont-Auvergne, CHU Clermont-Ferrand, Inserm, Neuro-Dol, Clermont-Ferrand, France
| | - Cedric Peirs
- Université Clermont-Auvergne, CHU Clermont-Ferrand, Inserm, Neuro-Dol, Clermont-Ferrand, France
| | - Reza Sharif Naeini
- Alan Edwards Centre for Research on Pain (AECRP), McGill University, Montreal, QC, Canada
- Department of Physiology and Cell Information Systems, McGill University, Montreal, QC, Canada
| | - Jean A Ouellet
- Department of Surgery, Orthopaedic Research Lab, McGill University, Montreal, QC, Canada
- Department of Surgery, McGill Scoliosis and Spine Group, McGill University, Montreal, QC, Canada
- Shriner's Hospital for Children, Montreal, QC, Canada
| | - Laura S Stone
- Alan Edwards Centre for Research on Pain (AECRP), McGill University, Montreal, QC, Canada
- Department of Anesthesiology, University of Minnesota, Minneapolis, MN, USA
| | - Lisbet Haglund
- Department of Surgery, Orthopaedic Research Lab, McGill University, Montreal, QC, Canada
- Department of Surgery, McGill Scoliosis and Spine Group, McGill University, Montreal, QC, Canada
- Shriner's Hospital for Children, Montreal, QC, Canada
| |
Collapse
|
2
|
Venkataraman A, Kordic I, Li J, Zhang N, Bharadwaj NS, Fang Z, Das S, Coskun AF. Decoding senescence of aging single cells at the nexus of biomaterials, microfluidics, and spatial omics. NPJ AGING 2024; 10:57. [PMID: 39592596 PMCID: PMC11599402 DOI: 10.1038/s41514-024-00178-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Accepted: 11/05/2024] [Indexed: 11/28/2024]
Abstract
Aging has profound effects on the body, most notably an increase in the prevalence of several diseases. An important aging hallmark is the presence of senescent cells that no longer multiply nor die off properly. Another characteristic is an altered immune system that fails to properly self-surveil. In this multi-player aging process, cellular senescence induces a change in the secretory phenotype, known as senescence-associated secretory phenotype (SASP), of many cells with the intention of recruiting immune cells to accelerate the clearance of these damaged senescent cells. However, the SASP phenotype results in inducing secondary senescence of nearby cells, resulting in those cells becoming senescent, and improper immune activation resulting in a state of chronic inflammation, called inflammaging, in many diseases. Senescence in immune cells, termed immunosenescence, results in further dysregulation of the immune system. An interdisciplinary approach is needed to physiologically assess aging changes of the immune system at the cellular and tissue level. Thus, the intersection of biomaterials, microfluidics, and spatial omics has great potential to collectively model aging and immunosenescence. Each of these approaches mimics unique aspects of the body undergoes as a part of aging. This perspective highlights the key aspects of how biomaterials provide non-cellular cues to cell aging, microfluidics recapitulate flow-induced and multi-cellular dynamics, and spatial omics analyses dissect the coordination of several biomarkers of senescence as a function of cell interactions in distinct tissue environments. An overview of how senescence and immune dysregulation play a role in organ aging, cancer, wound healing, Alzheimer's, and osteoporosis is included. To illuminate the societal impact of aging, an increasing trend in anti-senescence and anti-aging interventions, including pharmacological interventions, medical procedures, and lifestyle changes is discussed, including further context of senescence.
Collapse
Affiliation(s)
- Abhijeet Venkataraman
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
- Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, 315 Ferst Dr NW, Atlanta, GA, 30332, USA
| | - Ivan Kordic
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
- Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| | - JiaXun Li
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Nicholas Zhang
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
- Interdisciplinary Bioengineering Graduate Program, Georgia Institute of Technology, Atlanta, GA, USA
| | - Nivik Sanjay Bharadwaj
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Zhou Fang
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
- Machine Learning Graduate Program, Georgia Institute of Technology, Atlanta, GA, USA
| | - Sandip Das
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Ahmet F Coskun
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA.
- Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, 315 Ferst Dr NW, Atlanta, GA, 30332, USA.
- Interdisciplinary Bioengineering Graduate Program, Georgia Institute of Technology, Atlanta, GA, USA.
| |
Collapse
|
3
|
Park SC, Lee YS, Cho KA, Kim SY, Lee YI, Lee SR, Lim IK. What matters in aging is signaling for responsiveness. Pharmacol Ther 2023; 252:108560. [PMID: 37952903 DOI: 10.1016/j.pharmthera.2023.108560] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2023] [Revised: 10/03/2023] [Accepted: 11/06/2023] [Indexed: 11/14/2023]
Abstract
Biological responsiveness refers to the capacity of living organisms to adapt to changes in both their internal and external environments through physiological and behavioral mechanisms. One of the prominent aspects of aging is the decline in this responsiveness, which can lead to a deterioration in the processes required for maintenance, survival, and growth. The vital link between physiological responsiveness and the essential life processes lies within the signaling systems. To devise effective strategies for controlling the aging process, a comprehensive reevaluation of this connecting loop is imperative. This review aims to explore the impact of aging on signaling systems responsible for responsiveness and introduce a novel perspective on intervening in the aging process by restoring the compromised responsiveness. These innovative mechanistic approaches for modulating altered responsiveness hold the potential to illuminate the development of action plans aimed at controlling the aging process and treating age-related disorders.
Collapse
Affiliation(s)
- Sang Chul Park
- The Future Life & Society Research Center, Advanced Institute of Aging Science, Chonnam National University, Gwangju 61469, Republic of Korea.
| | - Young-Sam Lee
- Department of New Biology, DGIST, Daegu 42988, Republic of Korea; Well Aging Research Center, Division of Biotechnology, DGIST, Daegu 42988, Republic of Korea.
| | - Kyung A Cho
- Department of Biochemistry, Chonnam National University Medical School, Jeollanam-do 58128, Republic of Korea
| | - Sung Young Kim
- Department of Biochemistry, Konkuk University School of Medicine, Seoul 05029, Republic of Korea
| | - Yun-Il Lee
- Well Aging Research Center, Division of Biotechnology, DGIST, Daegu 42988, Republic of Korea; Interdisciplinary Engineering Major, Department of Interdisciplinary Studies, DGIST, Daegu 42988, Republic of Korea
| | - Seung-Rock Lee
- Department of Biochemistry, Chonnam National University Medical School, Jeollanam-do 58128, Republic of Korea; Department of Biomedical Sciences, Research Center for Aging and Geriatrics, Research Institute of Medical Sciences, Chonnam National University Medical School, Gwangju 61469, Republic of Korea
| | - In Kyoung Lim
- Department of Biochemistry and Molecular Biology, Ajou University School of Medicine, Suwon 16499, Republic of Korea
| |
Collapse
|
4
|
Riegger J, Schoppa A, Ruths L, Haffner-Luntzer M, Ignatius A. Oxidative stress as a key modulator of cell fate decision in osteoarthritis and osteoporosis: a narrative review. Cell Mol Biol Lett 2023; 28:76. [PMID: 37777764 PMCID: PMC10541721 DOI: 10.1186/s11658-023-00489-y] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Accepted: 09/11/2023] [Indexed: 10/02/2023] Open
Abstract
During aging and after traumatic injuries, cartilage and bone cells are exposed to various pathophysiologic mediators, including reactive oxygen species (ROS), damage-associated molecular patterns, and proinflammatory cytokines. This detrimental environment triggers cellular stress and subsequent dysfunction, which not only contributes to the development of associated diseases, that is, osteoporosis and osteoarthritis, but also impairs regenerative processes. To counter ROS-mediated stress and reduce the overall tissue damage, cells possess diverse defense mechanisms. However, cellular antioxidative capacities are limited and thus ROS accumulation can lead to aberrant cell fate decisions, which have adverse effects on cartilage and bone homeostasis. In this narrative review, we address oxidative stress as a major driver of pathophysiologic processes in cartilage and bone, including senescence, misdirected differentiation, cell death, mitochondrial dysfunction, and impaired mitophagy by illustrating the consequences on tissue homeostasis and regeneration. Moreover, we elaborate cellular defense mechanisms, with a particular focus on oxidative stress response and mitophagy, and briefly discuss respective therapeutic strategies to improve cell and tissue protection.
Collapse
Affiliation(s)
- Jana Riegger
- Division for Biochemistry of Joint and Connective Tissue Diseases, Department of Orthopedics, Ulm University Medical Center, 89081, Ulm, Germany.
| | - Astrid Schoppa
- Institute of Orthopedic Research and Biomechanics, Ulm University Medical Center, 89081, Ulm, Germany
| | - Leonie Ruths
- Division for Biochemistry of Joint and Connective Tissue Diseases, Department of Orthopedics, Ulm University Medical Center, 89081, Ulm, Germany
| | - Melanie Haffner-Luntzer
- Institute of Orthopedic Research and Biomechanics, Ulm University Medical Center, 89081, Ulm, Germany
| | - Anita Ignatius
- Institute of Orthopedic Research and Biomechanics, Ulm University Medical Center, 89081, Ulm, Germany
| |
Collapse
|
5
|
Yang Y, Mihajlovic M, Masereeuw R. Protein-Bound Uremic Toxins in Senescence and Kidney Fibrosis. Biomedicines 2023; 11:2408. [PMID: 37760849 PMCID: PMC10525416 DOI: 10.3390/biomedicines11092408] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Revised: 08/25/2023] [Accepted: 08/25/2023] [Indexed: 09/29/2023] Open
Abstract
Chronic kidney disease (CKD) is a progressive condition of kidney dysfunction due to diverse causes of injury. In healthy kidneys, protein-bound uremic toxins (PBUTs) are cleared from the systemic circulation by proximal tubule cells through the concerted action of plasma membrane transporters that facilitate their urinary excretion, but the endogenous metabolites are hardly removed with kidney dysfunction and may contribute to CKD progression. Accumulating evidence suggests that senescence of kidney tubule cells influences kidney fibrosis, the common endpoint for CKD with an excessive accumulation of extracellular matrix (ECM). Senescence is a special state of cells characterized by permanent cell cycle arrest and limitation of proliferation, which promotes fibrosis by releasing senescence-associated secretory phenotype (SASP) factors. The accumulation of PBUTs in CKD causes oxidative stress and increases the production of inflammatory (SASP) factors that could trigger fibrosis. Recent studies gave some clues that PBUTs may also promote senescence in kidney tubular cells. This review provides an overview on how senescence contributes to CKD, the involvement of PBUTs in this process, and how kidney senescence can be studied. Finally, some suggestions for future therapeutic options for CKD while targeting senescence are given.
Collapse
Affiliation(s)
- Yi Yang
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, 3584 CG Utrecht, The Netherlands;
| | - Milos Mihajlovic
- Entity of In Vitro Toxicology and Dermato-Cosmetology, Department of Pharmaceutical and Pharmacological Sciences, Vrije Universiteit Brussel, 1090 Brussels, Belgium;
| | - Rosalinde Masereeuw
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, 3584 CG Utrecht, The Netherlands;
| |
Collapse
|
6
|
Rutecki S, Szulc P, Pakuła M, Uruski P, Radziemski A, Naumowicz E, Moszyński R, Tykarski A, Mikuła-Pietrasik J, Książek K. Pro-cancerogenic effects of spontaneous and drug-induced senescence of ovarian cancer cells in vitro and in vivo: a comparative analysis. J Ovarian Res 2022; 15:87. [PMID: 35883110 PMCID: PMC9317468 DOI: 10.1186/s13048-022-01023-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Accepted: 07/19/2022] [Indexed: 11/10/2022] Open
Abstract
Background Clinical outcomes of cancer cell senescence are still elusive. Here, we reveal and compare pro-cancerous activity of spontaneously and drug-inducible senescent ovarian cancer cells. Experiments were performed on tumors and tumor-derived primary epithelial ovarian cancer cells (pEOCs) that were obtained from chemotherapy-naïve patients and from patients who received carboplatin (CPT) and paclitaxel (PCT) before cytoreduction. Results The analysis of tumors showed that senescent cancer cells are present in patients from both groups, albeit most frequently and covering a greater area in tissues from chemotherapy-positive women. This in vivo senescence of pEOCs translated to an expression of senescence markers in early-passage cells in vitro. A conditioned medium from senescent pEOCs fueled the cancer progression, including adhesion of non-senescent pEOCs to normal peritoneal cells, and their increased proliferation, migration, invasion, and EMT. Senescent pEOCs’ secretome promoted angiogenic activity of vascular endothelium, induced senescence of normal peritoneal cells, reprogrammed their secretome towards hypersecretion of cancer-promoting proteins, and stimulated motility of cancer cells subjected to a mesothelium- and fibroblast-derived medium. The most striking finding was, however, that spontaneously senescent pEOCs supported all the above pro-cancerous effects more efficiently than drug-inducible senescent cells, which was plausibly related to augmented release of several cancer spread mediators by these cells. The prevalence of spontaneously senescent pEOCs was most evident in experiments on mice when they were able, unlike the drug-inducible cells, to promote the development of drug-sensitive i.p. xenografts. Conclusions Our study shows that spontaneous senescence of pEOCs should be treated as an independent pathogenetic factor of cancer progression.
Collapse
Affiliation(s)
- Szymon Rutecki
- Department of Pathophysiology of Ageing and Civilization Diseases, Poznań University of Medical Sciences, Długa 1/2 Str, 61-848, Poznań, Poland
| | - Paulina Szulc
- Department of Pathophysiology of Ageing and Civilization Diseases, Poznań University of Medical Sciences, Długa 1/2 Str, 61-848, Poznań, Poland
| | - Martyna Pakuła
- Department of Hypertensiology, Poznań University of Medical Sciences, Długa 1/2 Str., 61-848, Poznań, Poland
| | - Paweł Uruski
- Department of Hypertensiology, Poznań University of Medical Sciences, Długa 1/2 Str., 61-848, Poznań, Poland
| | - Artur Radziemski
- Department of Hypertensiology, Poznań University of Medical Sciences, Długa 1/2 Str., 61-848, Poznań, Poland
| | - Eryk Naumowicz
- General Surgery Ward, Medical Centre HCP, 28 czerwca 1956 r. 223/229 Str., 61-485, Poznań, Poland
| | - Rafał Moszyński
- Division of Gynecological Surgery, Poznań University of Medical Sciences, Polna 33 Str, 60-535, Poznań, Poland
| | - Andrzej Tykarski
- Department of Hypertensiology, Poznań University of Medical Sciences, Długa 1/2 Str., 61-848, Poznań, Poland
| | - Justyna Mikuła-Pietrasik
- Department of Pathophysiology of Ageing and Civilization Diseases, Poznań University of Medical Sciences, Długa 1/2 Str, 61-848, Poznań, Poland
| | - Krzysztof Książek
- Department of Pathophysiology of Ageing and Civilization Diseases, Poznań University of Medical Sciences, Długa 1/2 Str, 61-848, Poznań, Poland.
| |
Collapse
|
7
|
Doxorubicin and doxorubicin-loaded nanoliposome induce senescence by enhancing oxidative stress, hepatotoxicity, and in vivo genotoxicity in male Wistar rats. Naunyn Schmiedebergs Arch Pharmacol 2021; 394:1803-1813. [PMID: 34219194 DOI: 10.1007/s00210-021-02119-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2021] [Accepted: 06/24/2021] [Indexed: 10/20/2022]
Abstract
The senescence phenomenon is historically considered as a tumor-suppressing mechanism that can permanently arrest the proliferation of damaged cells, and prevent tumor eradication by activating cell cycle regulatory pathways. Doxorubicin (DX) as an antineoplastic agent has been used for the treatment of solid and hematological malignancies for a long time, but its clinical use is limited due to irreversible toxicity on off-target tissues. Thereby, the encapsulation of plain drugs in a vehicle may decrease the side effects while increasing their permeability and availability in target cells. Here, we aimed to investigate and compare the effects of DX and DX-loaded nanoliposome (NLDX) on the induction of senescence via assessment of the occurrence of apoptosis/necrosis, genomic damage, oxidative stress, and liver pathologies. The study groups included DX (0.75, 0.5, 0.1 mg/kg/BW), NLDX groups (0.1, 0.05, 0.025 mg/kg/BW), and an untreated control group. The liver tissues were used to investigate the oxidative stress parameters and probable biochemical and histopathological alterations. Annexin V/PI staining was carried out to find the type of cellular death in the liver tissue of healthy rats exposed to different concentrations of DOX and LDOX. Data revealed that the highest dose of NLDX (0.1 mg/kg/BW) could significantly induce cellular senescence throughout significant increasing the level of genotoxic damage (p < 0.0001) and the oxidative stress (p < 0.001) compared with a similar dose of DX, in which the obtained results were further confirmed by flow cytometry and histopathological assessments of the liver tissue. This investigation provides sufficient evidence of improved therapeutic efficacy of NLDX compared with plain DX in male Wistar rats.
Collapse
|
8
|
Park JH, Ryu SJ, Kim BJ, Cho HJ, Park CH, Choi HJC, Jang EJ, Yang EJ, Hwang JA, Woo SH, Lee JH, Park JH, Choi KM, Kwon YY, Lee CK, Park JT, Cho SC, Lee YI, Lee SB, Han JA, Cho KA, Kim MS, Hwang D, Lee YS, Park SC. Disruption of nucleocytoplasmic trafficking as a cellular senescence driver. Exp Mol Med 2021; 53:1092-1108. [PMID: 34188179 PMCID: PMC8257587 DOI: 10.1038/s12276-021-00643-6] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Revised: 04/20/2021] [Accepted: 05/10/2021] [Indexed: 02/06/2023] Open
Abstract
Senescent cells exhibit a reduced response to intrinsic and extrinsic stimuli. This diminished reaction may be explained by the disrupted transmission of nuclear signals. However, this hypothesis requires more evidence before it can be accepted as a mechanism of cellular senescence. A proteomic analysis of the cytoplasmic and nuclear fractions obtained from young and senescent cells revealed disruption of nucleocytoplasmic trafficking (NCT) as an essential feature of replicative senescence (RS) at the global level. Blocking NCT either chemically or genetically induced the acquisition of an RS-like senescence phenotype, named nuclear barrier-induced senescence (NBIS). A transcriptome analysis revealed that, among various types of cellular senescence, NBIS exhibited a gene expression pattern most similar to that of RS. Core proteomic and transcriptomic patterns common to both RS and NBIS included upregulation of the endocytosis-lysosome network and downregulation of NCT in senescent cells, patterns also observed in an aging yeast model. These results imply coordinated aging-dependent reduction in the transmission of extrinsic signals to the nucleus and in the nucleus-to-cytoplasm supply of proteins/RNAs. We further showed that the aging-associated decrease in Sp1 transcription factor expression was critical for the downregulation of NCT. Our results suggest that NBIS is a modality of cellular senescence that may represent the nature of physiological aging in eukaryotes.
Collapse
Affiliation(s)
- Ji-Hwan Park
- grid.249967.70000 0004 0636 3099Korea Bioinformation Center, Korea Research Institute of Bioscience & Biotechnology, Daejeon, 34141 Republic of Korea
| | - Sung Jin Ryu
- grid.419666.a0000 0001 1945 5898Samsung Advanced Institute of Technology, Samsung Electronics Co., Ltd., Suwon, 16677 Republic of Korea ,Present Address: UBLBio Corporation, Suwon, 16679 Republic of Korea
| | - Byung Ju Kim
- grid.417736.00000 0004 0438 6721Present Address: Well Aging Research Center, Division of Biotechnology, DGIST, Daegu, 42988 Republic of Korea ,Present Address: UBLBio Corporation, Suwon, 16679 Republic of Korea
| | - Hyun-Ji Cho
- grid.417736.00000 0004 0438 6721Present Address: Well Aging Research Center, Division of Biotechnology, DGIST, Daegu, 42988 Republic of Korea
| | - Chi Hyun Park
- grid.412010.60000 0001 0707 9039Department of Computer Science and Engineering, Kangwon National University, Chuncheon, 24341 Republic of Korea
| | - Hyo Jei Claudia Choi
- grid.419666.a0000 0001 1945 5898Samsung Advanced Institute of Technology, Samsung Electronics Co., Ltd., Suwon, 16677 Republic of Korea
| | - Eun-Jin Jang
- grid.417736.00000 0004 0438 6721Present Address: Well Aging Research Center, Division of Biotechnology, DGIST, Daegu, 42988 Republic of Korea
| | - Eun Jae Yang
- grid.417736.00000 0004 0438 6721Department of New Biology, DGIST, Daegu, 42988 Republic of Korea
| | - Jeong-A Hwang
- grid.417736.00000 0004 0438 6721Department of New Biology, DGIST, Daegu, 42988 Republic of Korea
| | - Seung-Hwa Woo
- grid.417736.00000 0004 0438 6721Department of New Biology, DGIST, Daegu, 42988 Republic of Korea
| | - Jun Hyung Lee
- grid.417736.00000 0004 0438 6721Department of New Biology, DGIST, Daegu, 42988 Republic of Korea
| | - Ji Hwan Park
- grid.417736.00000 0004 0438 6721Department of New Biology, DGIST, Daegu, 42988 Republic of Korea
| | - Kyung-Mi Choi
- grid.222754.40000 0001 0840 2678Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul, 02841 Republic of Korea
| | - Young-Yon Kwon
- grid.222754.40000 0001 0840 2678Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul, 02841 Republic of Korea
| | - Cheol-Koo Lee
- grid.222754.40000 0001 0840 2678Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul, 02841 Republic of Korea
| | - Joon Tae Park
- grid.412977.e0000 0004 0532 7395Division of Life Sciences, College of Life Sciences and Bioengineering, Incheon National University, Incheon, 22012 Republic of Korea
| | - Sung Chun Cho
- grid.417736.00000 0004 0438 6721Present Address: Well Aging Research Center, Division of Biotechnology, DGIST, Daegu, 42988 Republic of Korea
| | - Yun-Il Lee
- grid.417736.00000 0004 0438 6721Present Address: Well Aging Research Center, Division of Biotechnology, DGIST, Daegu, 42988 Republic of Korea
| | - Sung Bae Lee
- grid.417736.00000 0004 0438 6721Department of Brain & Cognitive Science, DGIST, Daegu, 42988 Republic of Korea
| | - Jeong A. Han
- grid.412010.60000 0001 0707 9039Department of Biochemistry and Molecular Biology, Kangwon National University School of Medicine, Chuncheon, 24341 Republic of Korea
| | - Kyung A Cho
- grid.14005.300000 0001 0356 9399Department of Biochemistry, Chonnam National University, Medical School, Gwangju, 61469 Republic of Korea
| | - Min-Sik Kim
- grid.417736.00000 0004 0438 6721Department of New Biology, DGIST, Daegu, 42988 Republic of Korea
| | - Daehee Hwang
- grid.31501.360000 0004 0470 5905Department of Biological Sciences, Seoul National University, Seoul, 08826 Republic of Korea
| | - Young-Sam Lee
- grid.417736.00000 0004 0438 6721Present Address: Well Aging Research Center, Division of Biotechnology, DGIST, Daegu, 42988 Republic of Korea ,grid.417736.00000 0004 0438 6721Department of New Biology, DGIST, Daegu, 42988 Republic of Korea
| | - Sang Chul Park
- grid.417736.00000 0004 0438 6721Present Address: Well Aging Research Center, Division of Biotechnology, DGIST, Daegu, 42988 Republic of Korea ,grid.14005.300000 0001 0356 9399The Future Life & Society Research Center, Advanced Institute of Aging Science, Chonnam National University, Gwangju, 61469 Republic of Korea
| |
Collapse
|
9
|
Mannarino M, Cherif H, Li L, Sheng K, Rabau O, Jarzem P, Weber MH, Ouellet JA, Haglund L. Toll-like receptor 2 induced senescence in intervertebral disc cells of patients with back pain can be attenuated by o-vanillin. Arthritis Res Ther 2021; 23:117. [PMID: 33863359 PMCID: PMC8051055 DOI: 10.1186/s13075-021-02504-z] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Accepted: 04/03/2021] [Indexed: 01/29/2023] Open
Abstract
BACKGROUND There is an increased level of senescent cells and toll-like teceptor-1, -2, -4, and -6 (TLR) expression in degenerating intervertebral discs (IVDs) from back pain patients. However, it is currently not known if the increase in expression of TLRs is related to the senescent cells or if it is a more general increase on all cells. It is also not known if TLR activation in IVD cells will induce cell senescence. METHODS Cells from non-degenerate human IVD were obtained from spine donors and cells from degenerate IVDs came from patients undergoing surgery for low back pain. Gene expression of TLR-1,2,4,6, senescence and senescence-associated secretory phenotype (SASP) markers was evaluated by RT-qPCR in isolated cells. Matrix synthesis was verified with safranin-O staining and Dimethyl-Methylene Blue Assay (DMMB) confirmed proteoglycan content. Protein expression of p16INK4a, SASP factors, and TLR-2 was evaluated by immunocytochemistry (ICC) and/or by enzyme-linked immunosorbent assay (ELISA). RESULTS An increase in senescent cells was found following 48-h induction with a TLR-2/6 agonist in cells from both non-degenerate and degenerating human IVDs. Higher levels of SASP factors, TLR-2 gene expression, and protein expression were found following 48-h induction with TLR-2/6 agonist. Treatment with o-vanillin reduced the number of senescent cells, and increased matrix synthesis in IVD cells from back pain patients. Treatment with o-vanillin after induction with TLR-2/6 agonist reduced gene and protein expression of SASP factors and TLR-2. Co-localized staining of p16INK4a and TLR-2 demonstrated that senescent cells have a high TLR-2 expression. CONCLUSIONS Taken together our data demonstrate that activation of TLR-2/6 induce senescence and increase TLR-2 and SASP expression in cells from non-degenerate IVDs of organ donors without degeneration and back pain and in cells from degenerating human IVD of patients with disc degeneration and back pain. The senescent cells showed high TLR-2 expression suggesting a link between TLR activation and cell senescence in human IVD cells. The reduction in senescence, SASP, and TLR-2 expression suggest o-vanillin as a potential disease-modifying drug for patients with disc degeneration and back pain.
Collapse
Affiliation(s)
- Matthew Mannarino
- Department of Surgery, Orthopaedic Research Lab, McGill University, Montreal, Canada
- Department of Surgery, McGill Scoliosis and Spine Group, McGill University, Montreal, Canada
- Department of Surgery, The Research Institute of McGill University Health Center, Montreal, Canada
| | - Hosni Cherif
- Department of Surgery, Orthopaedic Research Lab, McGill University, Montreal, Canada
- Department of Surgery, McGill Scoliosis and Spine Group, McGill University, Montreal, Canada
- Department of Surgery, The Research Institute of McGill University Health Center, Montreal, Canada
| | - Li Li
- Department of Surgery, Orthopaedic Research Lab, McGill University, Montreal, Canada
- Department of Surgery, McGill Scoliosis and Spine Group, McGill University, Montreal, Canada
- Department of Surgery, The Research Institute of McGill University Health Center, Montreal, Canada
| | - Kai Sheng
- Department of Surgery, Orthopaedic Research Lab, McGill University, Montreal, Canada
- Department of Surgery, McGill Scoliosis and Spine Group, McGill University, Montreal, Canada
- Shriner's Hospital for Children, Montreal, Canada
| | - Oded Rabau
- Department of Surgery, McGill Scoliosis and Spine Group, McGill University, Montreal, Canada
- Shriner's Hospital for Children, Montreal, Canada
| | - Peter Jarzem
- Department of Surgery, Orthopaedic Research Lab, McGill University, Montreal, Canada
- Department of Surgery, McGill Scoliosis and Spine Group, McGill University, Montreal, Canada
- Department of Surgery, The Research Institute of McGill University Health Center, Montreal, Canada
| | - Michael H Weber
- Department of Surgery, Orthopaedic Research Lab, McGill University, Montreal, Canada
- Department of Surgery, McGill Scoliosis and Spine Group, McGill University, Montreal, Canada
- Department of Surgery, The Research Institute of McGill University Health Center, Montreal, Canada
| | - Jean A Ouellet
- Department of Surgery, Orthopaedic Research Lab, McGill University, Montreal, Canada
- Department of Surgery, McGill Scoliosis and Spine Group, McGill University, Montreal, Canada
- Department of Surgery, The Research Institute of McGill University Health Center, Montreal, Canada
- Shriner's Hospital for Children, Montreal, Canada
| | - Lisbet Haglund
- Department of Surgery, Orthopaedic Research Lab, McGill University, Montreal, Canada.
- Department of Surgery, McGill Scoliosis and Spine Group, McGill University, Montreal, Canada.
- Department of Surgery, The Research Institute of McGill University Health Center, Montreal, Canada.
- Shriner's Hospital for Children, Montreal, Canada.
- Department of Surgery, Montreal General Hospital, McGill University Health Centre, Room C9.173,1650 Cedar Ave, Montreal, QC, H3G 1A4, Canada.
| |
Collapse
|
10
|
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]
|
11
|
The role of adipose tissue senescence in obesity- and ageing-related metabolic disorders. Clin Sci (Lond) 2020; 134:315-330. [PMID: 31998947 DOI: 10.1042/cs20190966] [Citation(s) in RCA: 84] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Revised: 01/07/2020] [Accepted: 01/14/2020] [Indexed: 12/19/2022]
Abstract
Adipose tissue as the largest energy reservoir and endocrine organ is essential for maintenance of systemic glucose, lipid and energy homeostasis, but these metabolic functions decline with ageing and obesity. Adipose tissue senescence is one of the common features in obesity and ageing. Although cellular senescence is a defensive mechanism preventing tumorigenesis, its occurrence in adipose tissue causatively induces defective adipogenesis, inflammation, aberrant adipocytokines production and insulin resistance, leading to adipose tissue dysfunction. In addition to these paracrine effects, adipose tissue senescence also triggers systemic inflammation and senescence as well as insulin resistance in the distal metabolic organs, resulting in Type 2 diabetes and other premature physiological declines. Multiple cell types including mature adipocytes, immune cells, endothelial cells and progenitor cells gradually senesce at different levels in different fat depots with ageing and obesity, highlighting the heterogeneity and complexity of adipose tissue senescence. In this review, we discuss the causes and consequences of adipose tissue senescence, and the major cell types responsible for adipose tissue senescence in ageing and obesity. In addition, we summarize the pharmacological approaches and lifestyle intervention targeting adipose tissue senescence for the treatment of obesity- and ageing-related metabolic diseases.
Collapse
|
12
|
Lee JH, Yoon YM, Song K, Noh H, Lee SH. Melatonin suppresses senescence-derived mitochondrial dysfunction in mesenchymal stem cells via the HSPA1L-mitophagy pathway. Aging Cell 2020; 19:e13111. [PMID: 31965731 PMCID: PMC7059143 DOI: 10.1111/acel.13111] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Revised: 12/18/2019] [Accepted: 01/05/2020] [Indexed: 12/30/2022] Open
Abstract
Mesenchymal stem cells (MSCs) are a popular cell source for stem cell‐based therapy. However, continuous ex vivo expansion to acquire large amounts of MSCs for clinical study induces replicative senescence, causing decreased therapeutic efficacy in MSCs. To address this issue, we investigated the effect of melatonin on replicative senescence in MSCs. In senescent MSCs (late passage), replicative senescence decreased mitophagy by inhibiting mitofission, resulting in the augmentation of mitochondrial dysfunction. Treatment with melatonin rescued replicative senescence by enhancing mitophagy and mitochondrial function through upregulation of heat shock 70 kDa protein 1L (HSPA1L). More specifically, we found that melatonin‐induced HSPA1L binds to cellular prion protein (PrPC), resulting in the recruitment of PrPC into the mitochondria. The HSPA1L‐PrPC complex then binds to COX4IA, which is a mitochondrial complex IV protein, leading to an increase in mitochondrial membrane potential and anti‐oxidant enzyme activity. These protective effects were blocked by knockdown of HSPA1L. In a murine hindlimb ischemia model, melatonin‐treated senescent MSCs enhanced functional recovery by increasing blood flow perfusion, limb salvage, and neovascularization. This study, for the first time, suggests that melatonin protects MSCs against replicative senescence during ex vivo expansion for clinical application via mitochondrial quality control.
Collapse
Affiliation(s)
- Jun Hee Lee
- Medical Science Research Institute Soonchunhyang University Seoul Hospital Seoul Korea
- Departments of Biochemistry Soonchunhyang University College of Medicine Cheonan Korea
| | - Yeo Min Yoon
- Medical Science Research Institute Soonchunhyang University Seoul Hospital Seoul Korea
| | - Keon‐Hyoung Song
- Department of Pharmaceutical Engineering College of Medical Science Soonchunhyang University Asan Korea
| | - Hyunjin Noh
- Department of Internal Medicine Soonchunhyang University Seoul Korea
- Hyonam Kidney Laboratory Soonchunhyang University Seoul Korea
| | - Sang Hun Lee
- Medical Science Research Institute Soonchunhyang University Seoul Hospital Seoul Korea
- Departments of Biochemistry Soonchunhyang University College of Medicine Cheonan Korea
| |
Collapse
|
13
|
Fallah M, Mohammadi H, Shaki F, Hosseini-Khah Z, Moloudizargari M, Dashti A, Ziar A, Mohammadpour A, Mirshafa A, Modanloo M, Shokrzadeh M. Doxorubicin and liposomal doxorubicin induce senescence by enhancing nuclear factor kappa B and mitochondrial membrane potential. Life Sci 2019; 232:116677. [DOI: 10.1016/j.lfs.2019.116677] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Revised: 07/20/2019] [Accepted: 07/20/2019] [Indexed: 01/01/2023]
|
14
|
Vuong NH, Cook DP, Forrest LA, Carter LE, Robineau-Charette P, Kofsky JM, Hodgkinson KM, Vanderhyden BC. Single-cell RNA-sequencing reveals transcriptional dynamics of estrogen-induced dysplasia in the ovarian surface epithelium. PLoS Genet 2018; 14:e1007788. [PMID: 30418965 PMCID: PMC6258431 DOI: 10.1371/journal.pgen.1007788] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2018] [Revised: 11/26/2018] [Accepted: 10/25/2018] [Indexed: 12/30/2022] Open
Abstract
Estrogen therapy increases the risk of ovarian cancer and exogenous estradiol accelerates the onset of ovarian cancer in mouse models. Both in vivo and in vitro, ovarian surface epithelial (OSE) cells exposed to estradiol develop a subpopulation that loses cell polarity, contact inhibition, and forms multi-layered foci of dysplastic cells with increased susceptibility to transformation. Here, we use single-cell RNA-sequencing to characterize this dysplastic subpopulation and identify the transcriptional dynamics involved in its emergence. Estradiol-treated cells were characterized by up-regulation of genes associated with proliferation, metabolism, and survival pathways. Pseudotemporal ordering revealed that OSE cells occupy a largely linear phenotypic spectrum that, in estradiol-treated cells, diverges towards cell state consistent with the dysplastic population. This divergence is characterized by the activation of various cancer-associated pathways including an increase in Greb1 which was validated in fallopian tube epithelium and human ovarian cancers. Taken together, this work reveals possible mechanisms by which estradiol increases epithelial cell susceptibility to tumour initiation.
Collapse
Affiliation(s)
- Nhung H. Vuong
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Canada
- Cancer Therapeutics Program, Ottawa Hospital Research Institute, Ottawa, Canada
| | - David P. Cook
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Canada
- Cancer Therapeutics Program, Ottawa Hospital Research Institute, Ottawa, Canada
| | - Laura A. Forrest
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Canada
- Cancer Therapeutics Program, Ottawa Hospital Research Institute, Ottawa, Canada
| | - Lauren E. Carter
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Canada
- Cancer Therapeutics Program, Ottawa Hospital Research Institute, Ottawa, Canada
| | - Pascale Robineau-Charette
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Canada
- Cancer Therapeutics Program, Ottawa Hospital Research Institute, Ottawa, Canada
| | - Joshua M. Kofsky
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Canada
- Cancer Therapeutics Program, Ottawa Hospital Research Institute, Ottawa, Canada
| | - Kendra M. Hodgkinson
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Canada
- Cancer Therapeutics Program, Ottawa Hospital Research Institute, Ottawa, Canada
| | - Barbara C. Vanderhyden
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Canada
- Cancer Therapeutics Program, Ottawa Hospital Research Institute, Ottawa, Canada
- * E-mail:
| |
Collapse
|
15
|
Meng Q, Gao J, Zhu H, He H, Lu Z, Hong M, Zhou H. The proteomic study of serially passaged human skin fibroblast cells uncovers down-regulation of the chromosome condensin complex proteins involved in replicative senescence. Biochem Biophys Res Commun 2018; 505:1112-1120. [PMID: 30336977 DOI: 10.1016/j.bbrc.2018.10.065] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2018] [Accepted: 10/10/2018] [Indexed: 01/09/2023]
Abstract
Dermal fibroblast is one of the major constitutive cells of skin and plays a central role in skin senescence. The replicative senescence of fibroblasts may cause skin aging, bad wound healing, skin diseases and even cancer. In this study, a label-free quantitative proteomic approach was employed to analyzing the serial passaged human skin fibroblast (CCD-1079Sk) cells, resulting in 3371 proteins identified. Of which, 280 proteins were significantly changed in early passage (6 passages, P6), middle passage (12 passages, P12) and late passage (21 passages, P21), with a time-dependent decrease or increase tendency. Bioinformatic analysis demonstrated that the chromosome condensin complex, including structural maintenance of chromosomes protein 2 (SMC2) and structural maintenance of chromosomes protein 4 (SMC4), were down-regulated in the serially passaged fibroblast cells. The qRT-PCR and Western Blot experiments confirmed that the expression of these two proteins were significantly down-regulated in a time-dependent manner in the subculture of human skin fibroblasts (HSFb cells). In summary, we used serially passaged human skin fibroblast cells coupled with quantitative proteomic approach to profile the protein expression pattern in the temporal progress of replicative senescence in HSFb cells and revealed that the down-regulation of the chromosome condensin complex subunits, such as SMC2 and SMC4, may play an important role in the fibroblast senescence.
Collapse
Affiliation(s)
- Qian Meng
- Department of Analytical Chemistry and CAS Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road, Shanghai, 201203, China; University of Chinese Academy of Sciences, Number 19A Yuquan Road, Beijing, 100049, China
| | - Jing Gao
- Department of Analytical Chemistry and CAS Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road, Shanghai, 201203, China
| | - Hongwen Zhu
- Department of Analytical Chemistry and CAS Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road, Shanghai, 201203, China
| | - Han He
- Department of Analytical Chemistry and CAS Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road, Shanghai, 201203, China
| | - Zhi Lu
- Technology Center, Shanghai Inoherb Co. Ltd, 121 Chengyin Road, Shanghai, 200083, China.
| | - Minhua Hong
- Technology Center, Shanghai Inoherb Co. Ltd, 121 Chengyin Road, Shanghai, 200083, China.
| | - Hu Zhou
- Department of Analytical Chemistry and CAS Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road, Shanghai, 201203, China; University of Chinese Academy of Sciences, Number 19A Yuquan Road, Beijing, 100049, China.
| |
Collapse
|
16
|
Di Rocco G, Baldari S, Gentile A, Capogrossi M, Toietta G. Protein disulfide isomerase as a prosurvival factor in cell therapy for muscular and vascular diseases. Stem Cell Res Ther 2018; 9:250. [PMID: 30257707 PMCID: PMC6158916 DOI: 10.1186/s13287-018-0986-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Revised: 08/14/2018] [Accepted: 08/17/2018] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Cell therapy for degenerative diseases aims at rescuing tissue damage by delivery of precursor cells. Thus far, this strategy has been mostly unsuccessful due to massive loss of donor cells shortly after transplantation. Several strategies have been applied to increase transplanted cell survival but only with limited success. The endoplasmic reticulum (ER) is an organelle involved in protein folding, calcium homeostasis, and lipid biosynthesis. Protein disulfide isomerase (PDI) is a molecular chaperone induced and activated by ER stress. PDI is induced by hypoxia in neuronal, cardiac, and endothelial cells, supporting increased cell survival to hypoxic stress and protection from apoptosis in response to ischemia. METHODS We achieved ex vivo PDI gene transfer into luciferase-expressing myoblasts and endothelial cells. We assessed cell engraftment upon intramuscular transplantation into a mouse model of Duchenne muscular dystrophy (mdx mouse) and into a mouse model of ischemic disease. RESULTS We observed that loss of full-length dystrophin expression in mdx mice muscle leads to an increase of PDI expression, possibly in response to augmented ER protein folding load. Moreover, we determined that overexpression of PDI confers a survival advantage for muscle cells in vitro and in vivo to human myoblasts injected into murine dystrophic muscle and to endothelial cells administered upon hindlimb ischemia damage, improving the therapeutic outcome of the cell therapy treatment. CONCLUSIONS Collectively, these results suggest that overexpression of PDI may protect transplanted cells from hypoxia and other possibly occurring ER stresses, and consequently enhance their regenerative properties.
Collapse
Affiliation(s)
- Giuliana Di Rocco
- Department of Research, Advanced Diagnostic and Technological Innovation, IRCCS Regina Elena National Cancer Institute, via E. Chianesi 53, 00144 Rome, Italy
| | - Silvia Baldari
- Department of Research, Advanced Diagnostic and Technological Innovation, IRCCS Regina Elena National Cancer Institute, via E. Chianesi 53, 00144 Rome, Italy
| | - Antonietta Gentile
- Vascular Pathology, IRCCS Istituto Dermopatico dell’Immacolata, via dei Monti di Creta 104, 00167 Rome, Italy
- Present address: Department of Systems Medicine, Synaptic Immunopathology Laboratory, University of Rome Tor Vergata, Via Montpellier 1, 00133 Rome, Italy
| | - Maurizio Capogrossi
- Vascular Pathology, IRCCS Istituto Dermopatico dell’Immacolata, via dei Monti di Creta 104, 00167 Rome, Italy
| | - Gabriele Toietta
- Department of Research, Advanced Diagnostic and Technological Innovation, IRCCS Regina Elena National Cancer Institute, via E. Chianesi 53, 00144 Rome, Italy
| |
Collapse
|
17
|
Endothelial Cell Aging: How miRNAs Contribute? J Clin Med 2018; 7:jcm7070170. [PMID: 29996516 PMCID: PMC6068727 DOI: 10.3390/jcm7070170] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2018] [Revised: 07/04/2018] [Accepted: 07/09/2018] [Indexed: 12/19/2022] Open
Abstract
Endothelial cells (ECs) form monolayers and line the interior surfaces of blood vessels in the entire body. In most mammalian systems, the capacity of endothelial cells to divide is limited and endothelial cells are prone to be senescent. Aging of ECs and resultant endothelial dysfunction lead to a variety of vascular diseases such as atherosclerosis, diabetes mellites, hypertension, and ischemic injury. However, the mechanism by which ECs get old and become senescent and the impact of endothelial senescence on the vascular function are not fully understood. Recent research has unveiled the crucial roles of miRNAs, which are small non-coding RNAs, in regulating endothelial cellular functions, including nitric oxide production, vascular inflammation, and anti-thromboformation. In this review, how senescent-related miRNAs are involved in controlling the functions of ECs will be discussed.
Collapse
|
18
|
Toutfaire M, Dumortier E, Fattaccioli A, Van Steenbrugge M, Proby CM, Debacq-Chainiaux F. Unraveling the interplay between senescent dermal fibroblasts and cutaneous squamous cell carcinoma cell lines at different stages of tumorigenesis. Int J Biochem Cell Biol 2018; 98:113-126. [DOI: 10.1016/j.biocel.2018.03.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2017] [Revised: 02/08/2018] [Accepted: 03/09/2018] [Indexed: 12/21/2022]
|
19
|
Valentijn FA, Falke LL, Nguyen TQ, Goldschmeding R. Cellular senescence in the aging and diseased kidney. J Cell Commun Signal 2017; 12:69-82. [PMID: 29260442 PMCID: PMC5842195 DOI: 10.1007/s12079-017-0434-2] [Citation(s) in RCA: 102] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2017] [Accepted: 11/03/2017] [Indexed: 12/19/2022] Open
Abstract
The program of cellular senescence is involved in both the G1 and G2 phase of the cell cycle, limiting G1/S and G2/M progression respectively, and resulting in prolonged cell cycle arrest. Cellular senescence is involved in normal wound healing. However, multiple organs display increased senescent cell numbers both during natural aging and after injury, suggesting that senescent cells can have beneficial as well as detrimental effects in organismal aging and disease. Also in the kidney, senescent cells accumulate in various compartments with advancing age and renal disease. In experimental studies, forced apoptosis induction through the clearance of senescent cells leads to better preservation of kidney function during aging. Recent groundbreaking studies demonstrate that senescent cell depletion through INK-ATTAC transgene-mediated or cell-penetrating FOXO4-DRI peptide induced forced apoptosis, reduced age-associated damage and dysfunction in multiple organs, in particular the kidney, and increased performance and lifespan. Senescence is also involved in oncology and therapeutic depletion of senescent cells by senolytic drugs has been studied in experimental and human cancers. Although studies with senolytic drugs in models of kidney injury are lacking, their dose limiting side effects on other organs suggest that targeted delivery might be needed for successful application of senolytic drugs for treatment of kidney disease. In this review, we discuss (i) current understanding of the mechanisms and associated pathways of senescence, (ii) evidence of senescence occurrence and causality with organ injury, and (iii) therapeutic strategies for senescence depletion (senotherapy) including targeting, all in the context of renal aging and disease.
Collapse
Affiliation(s)
- F A Valentijn
- Department of Pathology, University Medical Center Utrecht, H04.312, Heidelberglaan 110, 3584, CX, Utrecht, The Netherlands
| | - L L Falke
- Department of Pathology, University Medical Center Utrecht, H04.312, Heidelberglaan 110, 3584, CX, Utrecht, The Netherlands
- Department of Internal Medicine, Diakonessenhuis, Utrecht, The Netherlands
| | - T Q Nguyen
- Department of Pathology, University Medical Center Utrecht, H04.312, Heidelberglaan 110, 3584, CX, Utrecht, The Netherlands
| | - Roel Goldschmeding
- Department of Pathology, University Medical Center Utrecht, H04.312, Heidelberglaan 110, 3584, CX, Utrecht, The Netherlands.
| |
Collapse
|
20
|
Pantsulaia I, Ciszewski WM, Niewiarowska J. Senescent endothelial cells: Potential modulators of immunosenescence and ageing. Ageing Res Rev 2016; 29:13-25. [PMID: 27235855 DOI: 10.1016/j.arr.2016.05.011] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2016] [Revised: 05/24/2016] [Accepted: 05/24/2016] [Indexed: 02/08/2023]
Abstract
Recent studies have demonstrated that the accumulation of senescent endothelial cells may be the primary cause of cardiovascular diseases. Because of their multifunctional properties, endothelial cells actively take part in stimulating the immune system and inflammation. In addition, ageing is characterized by the progressive deterioration of immune cells and a decline in the activation of the immune response. This results in a loss of the primary function of the immune system, which is eliminating damaged/senescent cells and neutralizing potential sources of harmful inflammatory reactions. In this review, we discuss cellular senescence and the senescence-associated secretory phenotype (SASP) of endothelial cells and summarize the link between endothelial cells and immunosenescence. We describe the possibility that age-related changes in Toll-like receptors (TLRs) and microRNAs can affect the phenotypes of senescent endothelial cells and immune cells via a negative feedback loop aimed at restraining the excessive pro-inflammatory response. This review also addresses the following questions: how do senescent endothelial cells influence ageing or age-related changes in the inflammatory burden; what is the connection between ECs and immunosenescence, and what are the crucial hypothetical pathways linking endothelial cells and the immune system during ageing.
Collapse
|
21
|
Matjusaitis M, Chin G, Sarnoski EA, Stolzing A. Biomarkers to identify and isolate senescent cells. Ageing Res Rev 2016; 29:1-12. [PMID: 27212009 DOI: 10.1016/j.arr.2016.05.003] [Citation(s) in RCA: 104] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2016] [Revised: 05/04/2016] [Accepted: 05/11/2016] [Indexed: 12/25/2022]
Abstract
Aging is the main risk factor for many degenerative diseases and declining health. Senescent cells are part of the underlying mechanism for time-dependent tissue dysfunction. These cells can negatively affect neighbouring cells through an altered secretory phenotype: the senescence-associated secretory phenotype (SASP). The SASP induces senescence in healthy cells, promotes tumour formation and progression, and contributes to other age-related diseases such as atherosclerosis, immune-senescence and neurodegeneration. Removal of senescent cells was recently demonstrated to delay age-related degeneration and extend lifespan. To better understand cell aging and to reap the benefits of senescent cell removal, it is necessary to have a reliable biomarker to identify these cells. Following an introduction to cellular senescence, we discuss several classes of biomarkers in the context of their utility in identifying and/or removing senescent cells from tissues. Although senescence can be induced by a variety of stimuli, senescent cells share some characteristics that enable their identification both in vitro and in vivo. Nevertheless, it may prove difficult to identify a single biomarker capable of distinguishing senescence in all cell types. Therefore, this will not be a comprehensive review of all senescence biomarkers but rather an outlook on technologies and markers that are most suitable to identify and isolate senescent cells.
Collapse
Affiliation(s)
- Mantas Matjusaitis
- Scottish Centre for Regenerative Medicine, The University of Edinburgh, Edinburgh, England, UK
| | - Greg Chin
- Department of Molecular, Cellular, and Developmental Biology, Yale University, New Haven, CT, USA
| | - Ethan Anders Sarnoski
- Department of Molecular, Cellular, and Developmental Biology, Yale University, New Haven, CT, USA
| | - Alexandra Stolzing
- Institute IZBI, University of Leipzig, Leipzig, Germany; Loughborough University, Loughborough, England, UK.
| |
Collapse
|
22
|
Turner JE. Is immunosenescence influenced by our lifetime "dose" of exercise? Biogerontology 2016; 17:581-602. [PMID: 27023222 PMCID: PMC4889625 DOI: 10.1007/s10522-016-9642-z] [Citation(s) in RCA: 72] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2015] [Accepted: 03/03/2016] [Indexed: 02/06/2023]
Abstract
The age-associated decline in immune function, referred to as immunosenescence, is well characterised within the adaptive immune system, and in particular, among T cells. Hallmarks of immunosenescence measured in the T cell pool, include low numbers and proportions of naïve cells, high numbers and proportions of late-stage differentiated effector memory cells, poor proliferative responses to mitogens, and a CD4:CD8 ratio <1.0. These changes are largely driven by infection with Cytomegalovirus, which has been directly linked with increased inflammatory activity, poor responses to vaccination, frailty, accelerated cognitive decline, and early mortality. It has been suggested however, that exercise might exert an anti-immunosenescence effect, perhaps delaying the onset of immunological ageing or even rejuvenating aged immune profiles. This theory has been developed on the basis of evidence that exercise is a powerful stimulus of immune function. For example, in vivo antibody responses to novel antigens can be improved with just minutes of exercise undertaken at the time of vaccination. Further, lymphocyte immune-surveillance, whereby cells search tissues for antigens derived from viruses, bacteria, or malignant transformation, is thought to be facilitated by the transient lymphocytosis and subsequent lymphocytopenia induced by exercise bouts. Moreover, some forms of exercise are anti-inflammatory, and if repeated regularly over the lifespan, there is a lower morbidity and mortality from diseases with an immunological and inflammatory aetiology. The aim of this article is to discuss recent theories for how exercise might influence T cell immunosenescence, exploring themes in the context of hotly debated issues in immunology.
Collapse
Affiliation(s)
- James E Turner
- Department for Health, University of Bath, Claverton Down, Bath, BA2 7AY, UK.
| |
Collapse
|
23
|
Apoptosis or senescence? Which exit route do epithelial cells and fibroblasts preferentially follow? Mech Ageing Dev 2016; 156:17-24. [PMID: 27060261 DOI: 10.1016/j.mad.2016.03.010] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2015] [Revised: 03/24/2016] [Accepted: 03/26/2016] [Indexed: 01/07/2023]
Abstract
Senescence and apoptosis constitute types of cellular responses that normally ensure homeostasis, when endogenous or exogenous signals occur. Their deregulation is often observed in various pathologies, such as age and non-age related diseases including cancer. Although epithelial cells and fibroblasts are capable to exert both functions, under a plethora of insults, the fact that they exhibit notable intrinsic differences in cell/tissue homeostasis properties, might be a crucial determinant of the mode of response to a certain stress signal. Sparse evidence in the literature reveals that in the same tissue/organ context and under the same conditions, the cell type seems to drive the differential counteraction between epithelia and fibroblasts. Based on the above notion we propose that, upon stress insults, human fibroblasts seem to predominantly respond via senescence, while epithelial cells prefer to exert apoptosis. We suggest that considering the tissue as a whole (epithelium and stroma) would benefit research into new therapeutic strategies for chronic diseases and cancer.
Collapse
|
24
|
Succoio M, Comegna M, D'Ambrosio C, Scaloni A, Cimino F, Faraonio R. Proteomic analysis reveals novel common genes modulated in both replicative and stress-induced senescence. J Proteomics 2015. [DOI: 10.1016/j.jprot.2015.07.010] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|