ijms-logo

Journal Browser

Journal Browser

Cellular Senescence in Physiological and Pathological Processes 2.0

A special issue of International Journal of Molecular Sciences (ISSN 1422-0067). This special issue belongs to the section "Molecular Pathology, Diagnostics, and Therapeutics".

Deadline for manuscript submissions: closed (31 July 2023) | Viewed by 16056

Special Issue Editors


E-Mail Website
Guest Editor
Department of Molecular Medicine and Medical Biotechnology, Medical School, University of Naples Federico II, Via Pansini, 80131 Naples, Italy
Interests: stem cells; cell culture; apoptosis; authophagy
Special Issues, Collections and Topics in MDPI journals

E-Mail Website
Guest Editor
Research Institute on Terrestrial Ecosystems (IRET), National Research Council of Italy (CNR), Via Pietro Castellino 111, 80131 Naples, Italy
Interests: stem cell; bioactive molecules; pulmonary disease; aging related disease; senescence; miRNA
Special Issues, Collections and Topics in MDPI journals

Special Issue Information

Dear Colleagues,

This Special Issue is the continuation of our previous Special Issue “Cellular Senescence in Physiological and Pathological Processes”.

Senescence is the permanent cell cycle arrest that occurs in response to extracellular or intracellular stress. It determines the loss of cellular functions over time. A senescent cell loses its original function and acquires new activities. Senescent cells secrete senescence-associated secretory phenotype (SASP) proteins to carry out several functions, such as sensitizing normal neighboring cells to senesce, reinforcing the senescence process through autocrine signaling, inducing tissue remodeling and repair, and promoting wound healing and immune cell recruitment. Cellular senescence is also believed to promote protective anticancer mechanisms resulting in tumor growth arrest, or, paradoxically, persistence of senescent cells in tissues may promote cancer onset. In addition, numerous studies correlate cellular senescence to aging, as it limits the proliferation of damaged cells, largely contributing to the reduction in tissue functions and renewal. Senescence has also been indicated as a major cause of age-related diseases. Recent evidence has shown that pharmacological ablation of senescent cells improves longevity and promotes health span.

This Special issue welcomes original research and critical reviews on the role of cellular senescence in physiological and pathological processes. Topics of this Special Issue include but are not limited to regulatory pathways of senescence; senescence and stem cells; links between cellular senescence and aging; novel therapeutic treatment to remove senescent cells; and senescence and cancer.

Dr. Tiziana Squillaro
Dr. Mauro Finicelli
Guest Editors

Manuscript Submission Information

Manuscripts should be submitted online at www.mdpi.com by registering and logging in to this website. Once you are registered, click here to go to the submission form. Manuscripts can be submitted until the deadline. All submissions that pass pre-check are peer-reviewed. Accepted papers will be published continuously in the journal (as soon as accepted) and will be listed together on the special issue website. Research articles, review articles as well as short communications are invited. For planned papers, a title and short abstract (about 100 words) can be sent to the Editorial Office for announcement on this website.

Submitted manuscripts should not have been published previously, nor be under consideration for publication elsewhere (except conference proceedings papers). All manuscripts are thoroughly refereed through a single-blind peer-review process. A guide for authors and other relevant information for submission of manuscripts is available on the Instructions for Authors page. International Journal of Molecular Sciences is an international peer-reviewed open access semimonthly journal published by MDPI.

Please visit the Instructions for Authors page before submitting a manuscript. There is an Article Processing Charge (APC) for publication in this open access journal. For details about the APC please see here. Submitted papers should be well formatted and use good English. Authors may use MDPI's English editing service prior to publication or during author revisions.

Published Papers (8 papers)

Order results
Result details
Select all
Export citation of selected articles as:

Editorial

Jump to: Research, Review, Other

3 pages, 183 KiB  
Editorial
Cellular Senescence in Physiological and Pathological Processes
by Mauro Finicelli, Gianfranco Peluso and Tiziana Squillaro
Int. J. Mol. Sci. 2022, 23(21), 13342; https://doi.org/10.3390/ijms232113342 - 01 Nov 2022
Viewed by 1254
Abstract
This Special Issue aims to address the impact of cellular senescence on human biology, looking at both physiological and pathological processes [...] Full article
(This article belongs to the Special Issue Cellular Senescence in Physiological and Pathological Processes 2.0)

Research

Jump to: Editorial, Review, Other

17 pages, 3560 KiB  
Article
Impact of Senolytic Treatment on Gene Expression in Aged Lung
by Soo Jung Cho, Alexander Pronko, Jianjun Yang and Heather Stout-Delgado
Int. J. Mol. Sci. 2023, 24(8), 7628; https://doi.org/10.3390/ijms24087628 - 21 Apr 2023
Viewed by 1484
Abstract
Cellular senescence plays a key role in mediating tissue remodeling and modulation of host responses to pathogenic stimuli. Our current study was designed to gain a better understanding of the impact of short-term senolytic treatment or inflammatory stimulation on lung senescence. The results [...] Read more.
Cellular senescence plays a key role in mediating tissue remodeling and modulation of host responses to pathogenic stimuli. Our current study was designed to gain a better understanding of the impact of short-term senolytic treatment or inflammatory stimulation on lung senescence. The results of our study demonstrate that short term treatment of aged adult mice (20 months of age) with senolytics, quercetin, and dasatinib decreases p16 and p21 expression in lung tissue. Short-term treatment with senolytics also significantly improved the expression of genes associated with genomic instability, telomere attrition, mitochondrial dysfunction, DNA binding, and the inflammatory response. In contrast, in response to low-dose LPS administration, there was increased expression of genes associated with genomic instability, mitochondrial dysfunction, and heightened inflammatory responses in young adult murine lung (3 months of age). Taken together, the results of our current study illustrate the efficacy of senolytic treatment on modulating responses in aged lung and the potential role of chronic low dose inflammation on senescence induction in the lung. Full article
(This article belongs to the Special Issue Cellular Senescence in Physiological and Pathological Processes 2.0)
Show Figures

Figure 1

24 pages, 7305 KiB  
Article
Modulation of Cellular Senescence in HEK293 and HepG2 Cells by Ultrafiltrates UPla and ULu Is Partly Mediated by Modulation of Mitochondrial Homeostasis under Oxidative Stress
by Junxian Zhou, Kang Liu, Chris Bauer, Gerald Bendner, Heike Dietrich, Jakub Peter Slivka, Michael Wink, Michelle B. F. Wong, Mike K. S. Chan and Thomas Skutella
Int. J. Mol. Sci. 2023, 24(7), 6748; https://doi.org/10.3390/ijms24076748 - 04 Apr 2023
Cited by 1 | Viewed by 2197
Abstract
Protein probes, including ultrafiltrates from the placenta (UPla) and lung (ULu) of postnatal rabbits, were investigated in premature senescent HEK293 and HepG2 cells to explore whether they could modulate cellular senescence. Tris-Tricine–PAGE, gene ontology (GO), and LC–MS/MS analysis were applied to describe the [...] Read more.
Protein probes, including ultrafiltrates from the placenta (UPla) and lung (ULu) of postnatal rabbits, were investigated in premature senescent HEK293 and HepG2 cells to explore whether they could modulate cellular senescence. Tris-Tricine–PAGE, gene ontology (GO), and LC–MS/MS analysis were applied to describe the characteristics of the ultrafiltrates. HEK293 and HepG2 cells (both under 25 passages) exposed to a sub-toxic concentration of hydrogen peroxide (H2O2, 300 μM) became senescent; UPla (10 μg/mL), ULu (10 μg/mL), as well as positive controls lipoic acid (10 μg/mL) and transferrin (10 μg/mL) were added along with H2O2 to the cells. Cell morphology; cellular proliferation; senescence-associated beta-galactosidase (SA-β-X-gal) activity; expression of senescence biomarkers including p16 INK4A (p16), p21 Waf1/Cip1 (p21), HMGB1, MMP-3, TNF-α, IL-6, lamin B1, and phospho-histone H2A.X (γ-H2AX); senescence-related gene expression; reactive oxygen species (ROS) levels; and mitochondrial fission were examined. Tris-Tricine–PAGE revealed prominent detectable bands between 10 and 100 kDa. LC–MS/MS identified 150–180 proteins and peptides in the protein probes, and GO analysis demonstrated a distinct enrichment of proteins associated with “extracellular space” and “proteasome core complex”. UPla and ULu modulated senescent cell morphology, improved cell proliferation, and decreased beta-galactosidase activity, intracellular and mitochondrial ROS production, and mitochondrial fission caused by H2O2. The results from this study demonstrated that UPla and Ulu, as well as lipoic acid and transferrin, could protect HEK293 and HepG2 cells from H2O2-induced oxidative damage via protecting mitochondrial homeostasis and thus have the potential to be explored in anti-aging therapies. Full article
(This article belongs to the Special Issue Cellular Senescence in Physiological and Pathological Processes 2.0)
Show Figures

Figure 1

12 pages, 11346 KiB  
Article
Generation of a p21 Reporter Mouse and Its Use to Identify and Eliminate p21high Cells In Vivo
by Zimei Yi, Le Ren, Yu Wei, Siyi Chen, Jiechen Zhao, Jiayu Zhu and Junhua Wu
Int. J. Mol. Sci. 2023, 24(6), 5565; https://doi.org/10.3390/ijms24065565 - 14 Mar 2023
Cited by 1 | Viewed by 1805
Abstract
P21 and p16 have been identified as inducers of senescence. Many transgenic mouse models have been developed to target cells expressing high levels of p16Ink4a (p16high) and investigate their potential contribution to tissue dysfunction in aging, obesity, and other pathological [...] Read more.
P21 and p16 have been identified as inducers of senescence. Many transgenic mouse models have been developed to target cells expressing high levels of p16Ink4a (p16high) and investigate their potential contribution to tissue dysfunction in aging, obesity, and other pathological conditions. However, the specific roles of p21 in various senescence-driven processes remain unclear. To gain a deeper understanding of p21, we built a p21-3MR mouse model containing a p21 promoter-driven module that allowed us to target cells with high p21Chip expression (p21high). Using this transgenic mouse, we monitored, imaged, and eliminated p21high cells in vivo. We also applied this system to chemically induced weakness and found that the clearance of p21high cells improved doxorubicin (DOXO)-induced multi-organ toxicity in mice. By recognizing p21 transcriptional activation spatially and temporally, the p21-3MR mouse model can be a valuable and powerful tool for studying p21high cells to further understand senescence biology. Full article
(This article belongs to the Special Issue Cellular Senescence in Physiological and Pathological Processes 2.0)
Show Figures

Figure 1

Review

Jump to: Editorial, Research, Other

12 pages, 273 KiB  
Review
The Cytoprotective Role of Autophagy in Response to BRAF-Targeted Therapies
by Ahmed M. Elshazly and David A. Gewirtz
Int. J. Mol. Sci. 2023, 24(19), 14774; https://doi.org/10.3390/ijms241914774 - 30 Sep 2023
Cited by 2 | Viewed by 1188
Abstract
BRAF-targeted therapies are widely used for the treatment of melanoma patients with BRAF V600 mutations. Vemurafenib, dabrafenib as well as encorafenib have demonstrated substantial therapeutic activity; however, as is the case with other chemotherapeutic agents, the frequent development of resistance limits their efficacy. [...] Read more.
BRAF-targeted therapies are widely used for the treatment of melanoma patients with BRAF V600 mutations. Vemurafenib, dabrafenib as well as encorafenib have demonstrated substantial therapeutic activity; however, as is the case with other chemotherapeutic agents, the frequent development of resistance limits their efficacy. Autophagy is one tumor survival mechanism that could contribute to BRAF inhibitor resistance, and multiple studies support an association between vemurafenib-induced and dabrafenib-induced autophagy and tumor cell survival. Clinical trials have also demonstrated a potential benefit from the inclusion of autophagy inhibition as an adjuvant therapy. This review of the scientific literature relating to the role of autophagy that is induced in response to BRAF-inhibitors supports the premise that autophagy targeting or modulation could be an effective adjuvant therapy. Full article
(This article belongs to the Special Issue Cellular Senescence in Physiological and Pathological Processes 2.0)
13 pages, 1098 KiB  
Review
Role of Senescent Astrocytes in Health and Disease
by Jacopo Meldolesi
Int. J. Mol. Sci. 2023, 24(10), 8498; https://doi.org/10.3390/ijms24108498 - 09 May 2023
Cited by 2 | Viewed by 2600
Abstract
For many decades after their discovery, astrocytes, the abundant glial cells of the brain, were believed to work as a glue, supporting the structure and metabolic functions of neurons. A revolution that started over 30 years ago revealed many additional functions of these [...] Read more.
For many decades after their discovery, astrocytes, the abundant glial cells of the brain, were believed to work as a glue, supporting the structure and metabolic functions of neurons. A revolution that started over 30 years ago revealed many additional functions of these cells, including neurogenesis, gliosecretion, glutamate homeostasis, assembly and function of synapses, neuronal metabolism with energy production, and others. These properties have been confirmed, limited however, to proliferating astrocytes. During their aging or following severe brain stress lesions, proliferating astrocytes are converted into their no-longer-proliferating, senescent forms, similar in their morphology but profoundly modified in their functions. The changed specificity of senescent astrocytes is largely due to their altered gene expression. The ensuing effects include downregulation of many properties typical of proliferating astrocytes, and upregulation of many others, concerned with neuroinflammation, release of pro-inflammatory cytokines, dysfunction of synapses, etc., specific to their senescence program. The ensuing decrease in neuronal support and protection by astrocytes induces the development, in vulnerable brain regions, of neuronal toxicity together with cognitive decline. Similar changes, ultimately reinforced by astrocyte aging, are also induced by traumatic events and molecules involved in dynamic processes. Senescent astrocytes play critical roles in the development of many severe brain diseases. The first demonstration, obtained for Alzheimer’s disease less than 10 years ago, contributed to the elimination of the previously predominant neuro-centric amyloid hypothesis. The initial astrocyte effects, operating a considerable time before the appearance of known Alzheimer’s symptoms evolve with the severity of the disease up to their proliferation during the final outcome. Involvement of astrocytes in other neurodegenerative diseases and cancer is now intensely investigated. Full article
(This article belongs to the Special Issue Cellular Senescence in Physiological and Pathological Processes 2.0)
Show Figures

Figure 1

26 pages, 2014 KiB  
Review
Senescence of Tumor Cells in Anticancer Therapy—Beneficial and Detrimental Effects
by Wiktoria Monika Piskorz and Marzanna Cechowska-Pasko
Int. J. Mol. Sci. 2022, 23(19), 11082; https://doi.org/10.3390/ijms231911082 - 21 Sep 2022
Cited by 8 | Viewed by 3141
Abstract
Cellular senescence process results in stable cell cycle arrest, which prevents cell proliferation. It can be induced by a variety of stimuli including metabolic stress, DNA damage, telomeres shortening, and oncogenes activation. Senescence is generally considered as a process of tumor suppression, both [...] Read more.
Cellular senescence process results in stable cell cycle arrest, which prevents cell proliferation. It can be induced by a variety of stimuli including metabolic stress, DNA damage, telomeres shortening, and oncogenes activation. Senescence is generally considered as a process of tumor suppression, both by preventing cancer cells proliferation and inhibiting cancer progression. It can also be a key effector mechanism for many types of anticancer therapies such as chemotherapy and radiotherapy, both directly and through bioactive molecules released by senescent cells that can stimulate an immune response. Senescence is characterized by a senescence-associated secretory phenotype (SASP) that can have both beneficial and detrimental impact on cancer progression. Despite the negatives, attempts are still being made to use senescence to fight cancer, especially when it comes to senolytics. There is a possibility that a combination of prosenescence therapy—which targets tumor cells and causes their senescence—with senotherapy—which targets senescent cells, can be promising in cancer treatment. This review provides information on cellular senescence, its connection with carcinogenesis and therapeutic possibilities linked to this process. Full article
(This article belongs to the Special Issue Cellular Senescence in Physiological and Pathological Processes 2.0)
Show Figures

Figure 1

Other

13 pages, 2000 KiB  
Brief Report
Doxorubicin–Mediated miR–433 Expression on Exosomes Promotes Bystander Senescence in Multiple Myeloma Cells in a DDR–Independent Manner
by Elisabetta Vulpis, Lorenzo Cuollo, Cristiana Borrelli, Fabrizio Antonangeli, Laura Masuelli, Marco Cippitelli, Cinzia Fionda, Giulio Caracciolo, Maria Teresa Petrucci, Angela Santoni, Alessandra Zingoni and Alessandra Soriani
Int. J. Mol. Sci. 2023, 24(7), 6862; https://doi.org/10.3390/ijms24076862 - 06 Apr 2023
Cited by 3 | Viewed by 1758
Abstract
The success of senescence-based anticancer therapies relies on their anti-proliferative power and on their ability to trigger anti-tumor immune responses. Indeed, genotoxic drug-induced senescence increases the expression of NK cell-activating ligands on multiple myeloma (MM) cells, boosting NK cell recognition and effector functions. [...] Read more.
The success of senescence-based anticancer therapies relies on their anti-proliferative power and on their ability to trigger anti-tumor immune responses. Indeed, genotoxic drug-induced senescence increases the expression of NK cell-activating ligands on multiple myeloma (MM) cells, boosting NK cell recognition and effector functions. Senescent cells undergo morphological change and context-dependent functional diversification, acquiring the ability to secrete a vast pool of molecules termed the senescence-associated secretory phenotype (SASP), which affects neighboring cells. Recently, exosomes have been recognized as SASP factors, contributing to modulating a variety of cell functions. In particular, evidence suggests a key role for exosomal microRNAs in influencing many hallmarks of cancer. Herein, we demonstrate that doxorubicin treatment of MM cells leads to the enrichment of miR-433 into exosomes, which in turn induces bystander senescence. Our analysis reveals that the establishment of the senescent phenotype on neighboring MM cells is p53- and p21-independent and is related to CDK-6 down-regulation. Notably, miR-433-dependent senescence does not induce the up-regulation of activating ligands on MM cells. Altogether, our findings highlight the possibility of miR-433-enriched exosomes to reinforce doxorubicin-mediated cellular senescence. Full article
(This article belongs to the Special Issue Cellular Senescence in Physiological and Pathological Processes 2.0)
Show Figures

Figure 1

Back to TopTop