Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 5.572
Filter
1.
FASEB J ; 38(16): e23862, 2024 Aug 31.
Article in English | MEDLINE | ID: mdl-39162681

ABSTRACT

Anterior cruciate ligament (ACL) injuries pose a significant challenge due to their limited healing potential, often resulting in premature arthritis. The factors and mechanisms contributing to this inadequate healing process remain elusive. During the acute phase of injury, ACL tissues express elevated periostin levels that decline over time. The functional significance of periostin in ligament biology remains understudied. In this study, we investigated the functional and mechanistic implications of periostin deficiency in ACL biology, utilizing ligament fibroblasts derived from patients and a murine model of ACL rupture. Our investigations unveiled that periostin knockdown compromised fibroblast growth characteristics, hindered the egress of progenitor cells from explants, and arrested cell-cycle progression, resulting in the accumulation of cells in the G0/G1 phase and moderate apoptosis. Concurrently, a significant reduction in the expression of cell-cycle and matrix-related genes was observed. Moreover, periostin deficiency triggered apoptosis through STAT3Y705/p38MAPK signaling and induced cellular senescence through increased production of reactive oxygen species (ROS). Mechanistically, inhibition of ROS production mitigated cell senescence in these cells. Notably, in vivo data revealed that ACL in Postn-/- mice exhibited a higher tearing frequency than wild-type mice under equivalent loading conditions. Furthermore, injured ACL with silenced periostin expression, achieved through nanoparticle-siRNA complex delivery, displayed an elevated propensity for apoptosis and senescence compared to intact ACL in C57BL/6 mice. Together, our findings underscore the pivotal role of periostin in ACL health, injury, and potential for healing.


Subject(s)
Anterior Cruciate Ligament Injuries , Anterior Cruciate Ligament , Cellular Senescence , Fibroblasts , Periostin , Reactive Oxygen Species , Animals , Female , Humans , Male , Mice , Anterior Cruciate Ligament/metabolism , Anterior Cruciate Ligament Injuries/metabolism , Anterior Cruciate Ligament Injuries/pathology , Apoptosis , Cells, Cultured , Cellular Senescence/physiology , Fibroblasts/metabolism , Mice, Inbred C57BL , Periostin/genetics , Periostin/metabolism , Reactive Oxygen Species/metabolism , STAT3 Transcription Factor/metabolism
2.
Cell Syst ; 15(8): 738-752.e5, 2024 Aug 21.
Article in English | MEDLINE | ID: mdl-39173586

ABSTRACT

Cellular longevity is regulated by both genetic and environmental factors. However, the interactions of these factors in the context of aging remain largely unclear. Here, we formulate a mathematical model for dynamic glucose modulation of a core gene circuit in yeast aging, which not only guided the design of pro-longevity interventions but also revealed the theoretical principles underlying these interventions. We introduce the dynamical systems theory to capture two general means for promoting longevity-the creation of a stable fixed point in the "healthy" state of the cell and the "dynamic stabilization" of the system around this healthy state through environmental oscillations. Guided by the model, we investigate how both of these can be experimentally realized by dynamically modulating environmental glucose levels. The results establish a paradigm for theoretically analyzing the trajectories and perturbations of aging that can be generalized to aging processes in diverse cell types and organisms.


Subject(s)
Glucose , Saccharomyces cerevisiae , Saccharomyces cerevisiae/genetics , Glucose/metabolism , Models, Biological , Gene Regulatory Networks , Cellular Senescence/physiology , Cellular Senescence/genetics , Longevity/physiology , Longevity/genetics , Environment
3.
Respir Res ; 25(1): 299, 2024 Aug 07.
Article in English | MEDLINE | ID: mdl-39113018

ABSTRACT

BACKGROUND: Although recent studies provide mechanistic understanding to the pathogenesis of radiation induced lung injury (RILI), rare therapeutics show definitive promise for treating this disease. Type II alveolar epithelial cells (AECII) injury in various manner results in an inflammation response to initiate RILI. RESULTS: Here, we reported that radiation (IR) up-regulated the TNKS1BP1, causing progressive accumulation of the cellular senescence by up-regulating EEF2 in AECII and lung tissue of RILI mice. Senescent AECII induced Senescence-Associated Secretory Phenotype (SASP), consequently activating fibroblasts and macrophages to promote RILI development. In response to IR, elevated TNKS1BP1 interacted with and decreased CNOT4 to suppress EEF2 degradation. Ectopic expression of EEF2 accelerated AECII senescence. Using a model system of TNKS1BP1 knockout (KO) mice, we demonstrated that TNKS1BP1 KO prevents IR-induced lung tissue senescence and RILI. CONCLUSIONS: Notably, this study suggested that a regulatory mechanism of the TNKS1BP1/CNOT4/EEF2 axis in AECII senescence may be a potential strategy for RILI.


Subject(s)
Alveolar Epithelial Cells , Cellular Senescence , Mice, Inbred C57BL , Mice, Knockout , Animals , Mice , Cellular Senescence/radiation effects , Cellular Senescence/physiology , Alveolar Epithelial Cells/metabolism , Alveolar Epithelial Cells/radiation effects , Alveolar Epithelial Cells/pathology , Lung Injury/metabolism , Lung Injury/genetics , Lung Injury/pathology , Elongation Factor 2 Kinase/metabolism , Elongation Factor 2 Kinase/genetics , Humans , Radiation Injuries, Experimental/metabolism , Radiation Injuries, Experimental/pathology , Radiation Injuries, Experimental/genetics , Cells, Cultured , Male
4.
Int J Biol Sci ; 20(10): 4055-4073, 2024.
Article in English | MEDLINE | ID: mdl-39113713

ABSTRACT

In the context of diabetes, endothelial cells frequently exhibit compromised intercellular junctions and accelerated cellular senescence simultaneously. The precise mechanisms underlying these issues and the identification of effective treatments remain largely undefined. Our findings reveal that human umbilical vein endothelial cells (HUVECs) can counteract senescence and uphold the integrity of intercellular junctions under mildly to moderately elevated glucose levels (10 mM and 15 mM) via two primary mechanisms: i) The acetylation of NRF2 at lysine residues K56, K68, and K52 prevents its ubiquitination, enhancing the transcription of antioxidant genes GST, SOD1, and GPX1. This activity diminishes cytoplasmic oxidative stress, thereby mitigating endothelial cell senescence. ii) The interaction between the Neh2 domain of NRF2 and the PAS-B domain of HIF-2α within the nucleus curtails the attachment of HIF-2α to the NOX4/p22phox promoter. This action lessens oxidative stress near the cell membrane, maintaining intercellular junctions by safeguarding the disulfide bonds in occludin and E-cadherin from disruption. However, these protective strategies prove insufficient under severe hyperglycemic conditions (25 mM). Further investigation has identified Oltipraz, an activator of NRF2, as also promoting the degradation of HIF-2α. Through its simultaneous modulation of NRF2 and HIF-2α, Oltipraz significantly reduces cellular senescence and prevents the deterioration of intercellular junctions in HUVECs subjected to high glucose concentrations (25 mM). Our research positions Oltipraz as a promising therapeutic candidate for mitigating diabetes-induced vascular endothelial damage, potentially offering benefits against diabetes-related atherosclerosis and valvular calcification.


Subject(s)
Cellular Senescence , Human Umbilical Vein Endothelial Cells , Intercellular Junctions , NF-E2-Related Factor 2 , Signal Transduction , Humans , NF-E2-Related Factor 2/metabolism , Cellular Senescence/physiology , Human Umbilical Vein Endothelial Cells/metabolism , Intercellular Junctions/metabolism , Basic Helix-Loop-Helix Transcription Factors/metabolism , Oxidative Stress , Diabetes Mellitus/metabolism , Glucose/metabolism
5.
Invest Ophthalmol Vis Sci ; 65(10): 29, 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-39167399

ABSTRACT

Purpose: Dysregulated cholesterol metabolism is critical in the pathogenesis of AMD. Cellular senescence contributes to the development of numerous age-associated diseases. In this study, we investigated the link between cholesterol burden and the cellular senescence of photoreceptors. Methods: Retinas from rod-specific ATP binding cassette subfamily A member 1 (Abca1) and G member 1 (Abcg1) (Abca1/g1-rod/-rod) knockout mice fed with a high-fat diet were analyzed for the signs of cellular senescence. Real-time quantitative PCR and immunofluorescence were used to characterize the senescence profile of the retina and cholesterol-treated photoreceptor cell line (661W). Inducible elimination of p16(Ink4a)-positive senescent cells (INK-ATTAC) mice or the administration of senolytic drugs (dasatinib and quercetin: D&Q) were used to examine the impact of senolytics on AMD-like phenotypes in Abca1/g1-rod/-rod retina. Results: Increased accumulation of senescent cells as measured by markers of cellular senescence was found in Abca1/g1-rod/-rod retina. Exogenous cholesterol also induced cellular senescence in 661W cells. Selective elimination of senescent cells in Abca1/g1-rod/-rod;INK-ATTAC mice or by administration of D&Q improved visual function, lipid accumulation in retinal pigment epithelium, and Bruch's membrane thickening. Conclusions: Cholesterol accumulation promotes cellular senescence in photoreceptors. Eliminating senescent photoreceptors improves visual function in a model of retinal neurodegeneration, and senotherapy offers a novel therapeutic avenue for further investigation.


Subject(s)
ATP Binding Cassette Transporter 1 , Cellular Senescence , Cholesterol , Disease Models, Animal , Mice, Knockout , Retinal Degeneration , Animals , Mice , Cellular Senescence/physiology , Cholesterol/metabolism , ATP Binding Cassette Transporter 1/metabolism , Retinal Degeneration/metabolism , Retinal Degeneration/pathology , Real-Time Polymerase Chain Reaction , Mice, Inbred C57BL , ATP Binding Cassette Transporter, Subfamily G, Member 1/metabolism , ATP Binding Cassette Transporter, Subfamily G, Member 1/genetics , Retinal Rod Photoreceptor Cells/metabolism , Retinal Rod Photoreceptor Cells/pathology , Retinal Rod Photoreceptor Cells/physiology
6.
Cell Syst ; 15(8): 676-678, 2024 Aug 21.
Article in English | MEDLINE | ID: mdl-39173583

ABSTRACT

How do variations in nutrient levels influence cellular lifespan? A dynamical systems model of a core circuit involved in yeast aging suggests principles underlying lifespan extension observed at static and alternating glucose levels that are reminiscent of intermittent fasting regimens.


Subject(s)
Cellular Senescence , Saccharomyces cerevisiae , Saccharomyces cerevisiae/physiology , Saccharomyces cerevisiae/genetics , Cellular Senescence/physiology , Glucose/metabolism , Models, Biological , Single-Cell Analysis/methods
7.
Biol Res ; 57(1): 51, 2024 Aug 08.
Article in English | MEDLINE | ID: mdl-39118171

ABSTRACT

Obesity, a global health crisis, disrupts multiple systemic processes, contributing to a cascade of metabolic dysfunctions by promoting the pathological expansion of visceral adipose tissue (VAT). This expansion is characterized by impaired differentiation of pre-adipocytes and an increase in senescent cells, leading to a pro-inflammatory state and exacerbated oxidative stress. Particularly, the senescence-associated secretory phenotype (SASP) and adipose tissue hypoxia further impair cellular function, promoting chronic disease development. This review delves into the potential of autophagy modulation and the therapeutic application of senolytics and senomorphics as novel strategies to mitigate adipose tissue senescence. By exploring the intricate mechanisms underlying adipocyte dysfunction and the emerging role of natural compounds in senescence modulation, we underscore the promising horizon of senotherapeutics in restoring adipose health. This approach not only offers a pathway to combat the metabolic complications of obesity, but also opens new avenues for enhancing life quality and managing the global burden of obesity-related conditions. Our analysis aims to bridge the gap between current scientific progress and clinical application, offering new perspectives on preventing and treating obesity-induced adipose dysfunction.


Subject(s)
Adipose Tissue , Autophagy , Cellular Senescence , Obesity , Senotherapeutics , Humans , Obesity/drug therapy , Cellular Senescence/physiology , Cellular Senescence/drug effects , Autophagy/physiology , Autophagy/drug effects , Senotherapeutics/pharmacology , Animals , Adipocytes
8.
Mech Ageing Dev ; 220: 111959, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38950628

ABSTRACT

Oligodendrocyte precursor cells (OPCs) comprise 5-8 % of the adult glial cell population and stand out as the most proliferative cell type in the central nervous system (CNS). OPCs are responsible for generating oligodendrocytes (OLs), the myelinating cells of the CNS. However, OPC functions decline as we age, resulting in impaired differentiation and inadequate remyelination. This review explores the cellular and molecular changes associated with OPC aging, and their impact on OPC differentiation and functionality. Furthermore, it examines the impact of OPC aging within the context of multiple sclerosis and Alzheimer's disease, both neurodegenerative conditions wherein aged OPCs exacerbate disease progression by impeding remyelination. Moreover, various pharmacological interventions targeting pathways related to senescence and differentiation are discussed as potential strategies to rejuvenate aged OPCs. Enhancing our understanding of OPC aging mechanisms holds promise for developing new therapies to improve remyelination and repair in age-related neurodegenerative disorders.


Subject(s)
Brain , Cell Differentiation , Cellular Senescence , Oligodendrocyte Precursor Cells , Humans , Oligodendrocyte Precursor Cells/metabolism , Oligodendrocyte Precursor Cells/physiology , Cellular Senescence/physiology , Animals , Brain/metabolism , Brain/pathology , Cell Differentiation/physiology , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Multiple Sclerosis/pathology , Multiple Sclerosis/metabolism , Aging/physiology , Aging/metabolism , Aging/pathology , Oligodendroglia/metabolism , Remyelination/physiology
9.
CNS Neurosci Ther ; 30(7): e14819, 2024 Jul.
Article in English | MEDLINE | ID: mdl-39056208

ABSTRACT

AIMS: Astrocytic senescence is inextricably linked to aging and neurodegenerative disorders, including Parkinson's disease (PD). P7C3 is a small, neuroprotective aminopropyl carbazole compound that exhibits anti-inflammatory properties. However, the effects of P7C3 on astrocytic senescence in PD remain to be elucidated. METHODS: An in vitro, long culture-induced, replicative senescence cell model and a 1-methyl-4-phenylpyridinium (MPP+)/rotenone-induced premature senescence cell model were used to investigate the effects of P7C3 on astrocytic senescence. An in vivo, 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced mouse PD model was used to study the role of P7C3 in astrocytic senescence. Immunoblotting, real-time quantitative RT-PCR (qPCR), immunofluorescence, subcellular fractionation assays, and immunohistochemistry were utilized to confirm the effects of P7C3 on astrocytic senescence and elucidate its underlying mechanisms. RESULTS: This study determined that P7C3 suppressed the senescence-associated secretory phenotype (SASP) in both cell models, as demonstrated by the reduction in the critical senescence marker p16 and proinflammatory factors (IL-6, IL-1ß, CXCL10, and MMP9) and increased laminB1 levels, implying that P7C3 inhibited replicative astrocytic senescence and MPP+/rotenone-induced premature astrocytic senescence, Most importantly, we demonstrated that P7C3 prevented the death of dopamine (DA) neurons and reduced the behavioral deficits in the MPTP-induced mouse model of PD, which is accompanied by a decrease in senescent astrocytes in the substantia nigra compacta (SNc). Mechanistically, P7C3 promoted Nrf2/Sirt3-mediated mitophagy and reduced mitochondrial reactive oxygen species (mitoROS) generation, which contributed to the suppression of astrocytic senescence. Furthermore, Sirt3 deficiency obviously abolished the inhibitory effects of P7C3 on astrocytic senescence. CONCLUSION: This study revealed that P7C3 inhibited astrocytic senescence via increased Nrf2/Sirt3-mediated mitophagy and suppression of mitoROS, which further protected against DA neuronal loss. These observations provide a prospective theoretical basis for P7C3 in the treatment of age-associated neurodegenerative diseases, such as PD.


Subject(s)
Astrocytes , Cellular Senescence , Dopaminergic Neurons , Mice, Inbred C57BL , Animals , Mice , Dopaminergic Neurons/drug effects , Dopaminergic Neurons/pathology , Dopaminergic Neurons/metabolism , Astrocytes/drug effects , Astrocytes/metabolism , Astrocytes/pathology , Cellular Senescence/drug effects , Cellular Senescence/physiology , Male , Neuroprotective Agents/pharmacology , Carbazoles/pharmacology , Disease Models, Animal
10.
Medicina (Kaunas) ; 60(7)2024 Jul 08.
Article in English | MEDLINE | ID: mdl-39064537

ABSTRACT

Background and Objectives: The presence and contribution of senescent cells in premalignant lesions is well documented, but not in germ cell neoplasia in situ. The purpose of this study is to identify the presence of senescent cells in pre-malignant testicular conditions and in different histological types of testicular cancer. Materials and Methods: Thirty patients who underwent orchiectomy due to testicular tumors were included. Formalin-fixed paraffin-embedded (FFPE) testicular tissue for each patient was available. Sections from these specimens were examined by immunohistochemical analysis with the following markers: GL13 for cellular senescence, p21WAF1/Cip1 for cell cycle arrest, and Ki67 for cell proliferation. Results: Thirteen (43.3%) suffered from seminoma with a mean total proportion of GCNIS senescence of 20.81 ± 6.81%. In the group of embryonal testicular tumors, nine (30%) patients were included, with an average rate of 6.64 ± 5.42% of senescent cells in GCNIS. One (3.3%) patient suffered from chondrosarcoma in which 7.9% of GL13+ cells were detected in GCNIS. Four (13.4%) patients suffered from teratoma and three (10%) from yolk sac tumors, while GCNIS senescence was detected in a range of 4.43 ± 1.78% and 3.76 ± 1.37%, respectively. Conclusions: Cellular senescence was detected in both germ cell neoplasia in situ and testicular cancer, but was more prevalent within the premalignant lesions.


Subject(s)
Cellular Senescence , Neoplasms, Germ Cell and Embryonal , Testicular Neoplasms , Humans , Male , Testicular Neoplasms/pathology , Testicular Neoplasms/surgery , Cellular Senescence/physiology , Adult , Neoplasms, Germ Cell and Embryonal/pathology , Neoplasms, Germ Cell and Embryonal/surgery , Middle Aged , Orchiectomy , Immunohistochemistry
11.
Exp Gerontol ; 194: 112508, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38986855

ABSTRACT

hTERT gene therapies hold significant promise for treating age-related diseases. However, further research is required to address the challenges of delivery and ethical considerations. We hypothesized that exosomes derived from hTERT-immortalized cells could function similarly to hTERT gene therapies by maintaining telomere length and attenuating cellular senescence biomarkers. In this study, we overexpressed the hTERT gene in Human Foreskin Fibroblast-1 cells (HFF cells) to produce hTERT-immortalized HFF cells (hT-HFF cells). We then used exosomes derived from these hT-HFF cells to treat human fibroblasts, HFF cells. Our results demonstrated that these exosomes effectively attenuated biomarkers of cellular senescence in HFF cells. Furthermore, analysis revealed that hTERT mRNA was indeed packaged into the exosomes from hT-HFF cells. This mRNA was capable of elongating telomeres and delaying cellular senescence in HFF cells. Therefore, exosomes from hT-HFF cells show potential as a treatment for age-related diseases.


Subject(s)
Cellular Senescence , Exosomes , Fibroblasts , Telomerase , Humans , Telomerase/metabolism , Telomerase/genetics , Cellular Senescence/physiology , Exosomes/metabolism , Fibroblasts/metabolism , RNA, Messenger/metabolism , Telomere/metabolism , Telomere Homeostasis/physiology , Cell Line
12.
J Am Heart Assoc ; 13(15): e033341, 2024 Aug 06.
Article in English | MEDLINE | ID: mdl-39023057

ABSTRACT

Cellular senescence, a permanent halt in cell division due to stress, spurs functional and structural changes, contributing to vascular aging characterized by endothelial dysfunction and vascular remodeling. This process raises the risk of ischemic stroke (IS) in older individuals, with its mechanisms still not completely understood despite ongoing research efforts. In this review, we have analyzed the impact of vascular aging on increasing susceptibility and exacerbating the pathology of IS. We have emphasized the detrimental effects of endothelial dysfunction and vascular remodeling influenced by oxidative stress and inflammatory response on vascular aging and IS. Our goal is to aid the understanding of vascular aging and IS pathogenesis, particularly benefiting older adults with high risk of IS.


Subject(s)
Aging , Ischemic Stroke , Oxidative Stress , Humans , Ischemic Stroke/physiopathology , Ischemic Stroke/etiology , Aging/physiology , Aging/pathology , Vascular Remodeling/physiology , Endothelium, Vascular/physiopathology , Endothelium, Vascular/pathology , Risk Factors , Cellular Senescence/physiology , Animals
13.
J Orthop Surg Res ; 19(1): 421, 2024 Jul 22.
Article in English | MEDLINE | ID: mdl-39034400

ABSTRACT

BACKGROUND: Cellular senescence features irreversible growth arrest and secretion of multiple proinflammatory cytokines. Cyclic GMP-AMP synthase (cGAS) detects DNA damage and activates the DNA-sensing pathway, resulting in the upregulation of inflammatory genes and induction of cellular senescence. This study aimed to investigate the effect of cGAS in regulating senescence of nucleus pulposus (NP) cells under inflammatory microenvironment. METHODS: The expression of cGAS was evaluated by immunohistochemical staining in rat intervertebral disc (IVD) degeneration model induced by annulus stabbing. NP cells were harvested from rat lumbar IVD and cultured with 10ng/ml IL-1ß for 48 h to induce premature senescence. cGAS was silenced by cGAS specific siRNA in NP cells and cultured with IL-1ß. Cellular senescence was evaluated by senescence-associated beta-galactosidase (SA-ß-gal) staining and flow cytometry. The expression of senescence-associated secretory phenotype including IL-6, IL-8, and TNF-a was evaluated by ELISA and western blotting. RESULTS: cGAS was detected in rat NP cells in cytoplasm and the expression was significantly increased in degenerated IVD. Culturing in 10ng/ml IL-1ß for 48 h induced cellular senescence in NP cells with attenuation of G1-S phase transition. In senescent NP cells the expression of cGAS, p53, p16, NF-kB, IL-6, IL-8, TNF-α was significantly increased while aggrecan and collagen type II was reduced than in normal NP cells. In NP cells with silenced cGAS, the expression of p53, p16, NF-kB, IL-6, IL-8, and TNF-α was reduced in inflammatory culturing with IL-1ß. CONCLUSION: cGAS was increased by NP cells in degenerated IVD promoting cellular senescence and senescent inflammatory phenotypes. Targeting cGAS may alleviate IVD degeneration by reducing NP cell senescence.


Subject(s)
Cellular Senescence , Intervertebral Disc Degeneration , Nucleotidyltransferases , Nucleus Pulposus , Rats, Sprague-Dawley , Cellular Senescence/physiology , Animals , Nucleus Pulposus/metabolism , Nucleus Pulposus/pathology , Intervertebral Disc Degeneration/pathology , Intervertebral Disc Degeneration/metabolism , Nucleotidyltransferases/metabolism , Nucleotidyltransferases/genetics , Cells, Cultured , Rats , Male , Inflammation/metabolism , Inflammation/pathology , Interleukin-1beta/metabolism
14.
Biomater Adv ; 163: 213938, 2024 Oct.
Article in English | MEDLINE | ID: mdl-38959650

ABSTRACT

Endothelial cells are constantly exposed to mechanical stimuli, of which mechanical stretch has shown various beneficial or deleterious effects depending on whether loads are within physiological or pathological levels, respectively. Vascular properties change with age, and on a cell-scale, senescence elicits changes in endothelial cell mechanical properties that together can impair its response to stretch. Here, high-rate uniaxial stretch experiments were performed to quantify and compare the stretch-induced damage of monolayers consisting of young, senescent, and aged endothelial populations. The aged and senescent phenotypes were more fragile to stretch-induced damage. Prominent damage was detected by immunofluorescence and scanning electron microscopy as intercellular and intracellular void formation. Damage increased proportionally to the applied level of deformation and, for the aged and senescent phenotype, induced significant detachment of cells at lower levels of stretch compared to the young counterpart. Based on the phenotypic difference in cell-substrate adhesion of senescent cells indicating more mature focal adhesions, a discrete network model of endothelial cells being stretched was developed. The model showed that the more affine deformation of senescent cells increased their intracellular energy, thus enhancing the tendency for cellular damage and impending detachment. Next to quantifying for the first-time critical levels of endothelial stretch, the present results indicate that young cells are more resilient to deformation and that the fragility of senescent cells may be associated with their stronger adhesion to the substrate.


Subject(s)
Cellular Senescence , Endothelial Cells , Stress, Mechanical , Humans , Endothelial Cells/pathology , Endothelial Cells/physiology , Cellular Senescence/physiology , Cell Adhesion , Human Umbilical Vein Endothelial Cells , Cells, Cultured , Microscopy, Electron, Scanning
15.
Bone Res ; 12(1): 38, 2024 Jul 04.
Article in English | MEDLINE | ID: mdl-38961077

ABSTRACT

Bone marrow adipocytes (BMAds) affect bone homeostasis, but the mechanism remains unclear. Here, we showed that exercise inhibited PCNA clamp-associated factor (PCLAF) secretion from the bone marrow macrophages to inhibit BMAds senescence and thus alleviated skeletal aging. The genetic deletion of PCLAF in macrophages inhibited BMAds senescence and delayed skeletal aging. In contrast, the transplantation of PCLAF-mediated senescent BMAds into the bone marrow of healthy mice suppressed bone turnover. Mechanistically, PCLAF bound to the ADGRL2 receptor to inhibit AKT/mTOR signaling that triggered BMAds senescence and subsequently spread senescence among osteogenic and osteoclastic cells. Of note, we developed a PCLAF-neutralizing antibody and showed its therapeutic effects on skeletal health in old mice. Together, these findings identify PCLAF as an inducer of BMAds senescence and provide a promising way to treat age-related osteoporosis.


Subject(s)
Adipocytes , Aging , Cellular Senescence , Animals , Adipocytes/metabolism , Cellular Senescence/physiology , Mice , Aging/physiology , Mice, Inbred C57BL , Bone Marrow Cells/metabolism , Bone and Bones/metabolism , Bone and Bones/physiology , Male , Osteogenesis/physiology , Signal Transduction , Macrophages/metabolism
16.
FASEB J ; 38(14): e23824, 2024 Jul 31.
Article in English | MEDLINE | ID: mdl-39012304

ABSTRACT

The regenerative ability of limb bones after injury decreases during aging, but whether a similar phenomenon occurs in jawbones and whether autophagy plays a role in this process remain unclear. Through retrospective analysis of clinical data and studies on a mouse model of jawbone defects, we confirmed the presence of delayed or impaired bone regeneration in the jawbones of old individuals and mice. Subsequently, osteoblasts (OBs) derived from mouse jawbones were isolated, showing reduced osteogenesis in senescent osteoblasts (S-OBs). We observed a reduction in autophagy within both aged jawbones and S-OBs. Additionally, pharmacological inhibition of autophagy in normal OBs (N-OBs) led to cell aging and decreased osteogenesis, while autophagic activation reversed the aging phenotype of S-OBs. The activator rapamycin (RAPA) increased the autophagy level and bone regeneration in aged jawbones. Finally, we found that fatty acid-binding protein 3 (FABP3) was degraded by autolysosomes through its interaction with sequestosome 1 (P62/SQSTM1). Autophagy inhibition within senescent jawbones and S-OBs led to the excessive accumulation of FABP3, and FABP3 knockdown partially rescued the decreased osteogenesis in S-OBs and alleviated age-related compromised jawbone regeneration. In summary, we confirmed that autophagy inhibition plays an important role in delaying bone regeneration in aging jawbones. Autophagic activation or FABP3 knockdown can partially rescue the osteogenesis of S-OBs and the regeneration of aging jawbones, providing insight into jawbone aging.


Subject(s)
Aging , Autophagy , Bone Regeneration , Fatty Acid-Binding Proteins , Osteoblasts , Osteogenesis , Animals , Female , Humans , Male , Mice , Aging/physiology , Aging/metabolism , Autophagy/physiology , Cellular Senescence/physiology , Fatty Acid-Binding Proteins/metabolism , Fatty Acid-Binding Proteins/genetics , Jaw , Mice, Inbred C57BL , Osteoblasts/metabolism , Osteogenesis/physiology
17.
Cytometry A ; 105(8): 580-594, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38995093

ABSTRACT

Senescence is an irreversible arrest of the cell cycle that can be characterized by markers of senescence such as p16, p21, and KI-67. The characterization of different senescence-associated phenotypes requires selection of the most relevant senescence markers to define reliable cytometric methodologies. Mass cytometry (a.k.a. Cytometry by time of flight, CyTOF) can monitor up to 40 different cell markers at the single-cell level and has the potential to integrate multiple senescence and other phenotypic markers to identify senescent cells within a complex tissue such as skeletal muscle, with greater accuracy and scalability than traditional bulk measurements and flow cytometry-based measurements. This article introduces an analysis framework for detecting putative senescent cells based on clustering, outlier detection, and Boolean logic for outliers. Results show that the pipeline can identify putative senescent cells in skeletal muscle with well-established markers such as p21 and potential markers such as GAPDH. It was also found that heterogeneity of putative senescent cells in skeletal muscle can partly be explained by their cell type. Additionally, autophagy-related proteins ATG4A, LRRK2, and GLB1 were identified as important proteins in predicting the putative senescent population, providing insights into the association between autophagy and senescence. It was observed that sex did not affect the proportion of putative senescent cells among total cells. However, age did have an effect, with a higher proportion observed in fibro/adipogenic progenitors (FAPs), satellite cells, M1 and M2 macrophages from old mice. Moreover, putative senescent cells from muscle of old and young mice show different expression levels of senescence-related proteins, with putative senescent cells of old mice having higher levels of p21 and GAPDH, whereas putative senescent cells of young mice had higher levels of IL-6. Overall, the analysis framework prioritizes multiple senescence-associated proteins to characterize putative senescent cells sourced from tissue made of different cell types.


Subject(s)
Biomarkers , Cellular Senescence , Flow Cytometry , Muscle, Skeletal , Animals , Cellular Senescence/physiology , Mice , Muscle, Skeletal/cytology , Muscle, Skeletal/metabolism , Flow Cytometry/methods , Biomarkers/metabolism , Female , Male , Mice, Inbred C57BL , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Single-Cell Analysis/methods
18.
Bone ; 187: 117198, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39002837

ABSTRACT

Early B cell factor 1 (EBF1) is a transcription factor expressed by multiple lineages of stromal cells within the bone marrow. While cultures of Ebf1-deficient cells have been demonstrated to have impaired differentiation into either the osteoblast or adipogenic lineage in vitro by several groups, in vivo there has been a nominal consequence of the loss of EBF1 on skeletal development. In this study we used Prx-cre driven deletion of Ebf1 to eliminate EBF1 from the entire mesenchymal lineage of the skeleton and resolve this discrepancy. We report here that EBF1 is expressed primarily in the Mesenchymal Stem and Progenitor Cell (MSPC)-Adipo, MSPC-Osteo, and the Early Mesenchymal Progenitors, and that loss of EBF1 has a plethora of consequences to maintenance of the skeleton throughout adulthood. Stroma from the Prx-cre;Ebf1fl/fl bones had impaired osteogenic differentiation, an age-dependent loss of CFU-F, and elevated senescence accompanying Ebf1-deletion. New bone formation was reduced after 3 months, and resulted in a quiescent bone environment with fewer osteoblasts and an accompanied reduction in osteoclast-mediated remodeling. Consequently, bones were less ductile at a younger age, and deletion of EBF1 dramatically impaired fracture repair. Disruption of EBF1 in perivascular populations also rearranged the vascular network within these bones and disrupted cytokine signaling from key hematopoietic niches resulting in anemia, reductions in B cells, and myeloid skewing of marrow hematopoietic lineages. Mechanistically we observed disrupted BMP signaling within Ebf1-deficient progenitors with reduced SMAD1-phosphorylation, and elevated secretion of the soluble BMP-inhibitor Gremlin from the MSPC-Adipo cells. Ebf1-deficient progenitors also exhibited posttranslational suppression of glucocorticoid receptor expression. Together, these results suggest that EBF1 signaling is required for mesenchymal progenitor mobilization to maintain the adult skeleton, and that the primary action of EBF1 in the early mesenchymal lineage is to promote proliferation, and differentiation of these perivascular cells to sustain a healthy tissue.


Subject(s)
Cell Differentiation , Homeostasis , Osteogenesis , Trans-Activators , Animals , Trans-Activators/metabolism , Trans-Activators/genetics , Osteogenesis/genetics , Bone and Bones/metabolism , Bone and Bones/pathology , Mice , Mesenchymal Stem Cells/metabolism , Gene Deletion , Stromal Cells/metabolism , Bone Marrow/metabolism , Osteoblasts/metabolism , Mice, Inbred C57BL , Cellular Senescence/physiology , Aging/metabolism , Cellular Microenvironment
19.
Aging (Albany NY) ; 16(14): 11128-11133, 2024 Jul 19.
Article in English | MEDLINE | ID: mdl-39033779

ABSTRACT

Parkinson's disease (PD) is an age-related movement disorder caused by the loss of dopaminergic (DA) neurons of the substantia nigra pars compacta (SNpc) of the midbrain, however, the underlying cause(s) of this DA neuron loss in PD is unknown and there are currently no effective treatment options to prevent or slow neuronal loss or the progression of related symptoms. It has been shown that both environmental factors as well as genetic predispositions underpin PD development and recent research has revealed that lysosomal dysfunction and lipid accumulation are contributors to disease progression, where an age-related aggregation of alpha-synuclein as well as lipids have been found in PD patients. Interestingly, the most common genetic risk factor for PD is Glucosylceramidase Beta 1 (GBA), which encodes a lysosomal glucocerebrosidase (GCase) that cleaves the beta-glucosidic linkage of lipids known as glucocerebrosides (GluCer). We have recently discovered that artificial induction of GluCer accumulation leads to cellular senescence of DA neurons, suggesting that lipid aggregation plays a crucial role in the pathology of PD by driving senescence in these vulnerable DA neurons. Here, we discuss the relevance of the age-related aggregation of lipids as well as the direct functional link between general lipid aggregation, cellular senescence, and inflammaging of DA neurons. We propose that the expression of a cellular senescence phenotype in the most vulnerable neurons in PD can be triggered by lysosomal impairment and lipid aggregation. Importantly, we highlight additional data that perilipin (PLIN2) is significantly upregulated in senescent DA neurons, suggesting an overall enrichment of lipid droplets (LDs) in these cells. These findings align with our previous results in dopaminergic neurons in highlighting a central role for lipid accumulation in the senescence of DA neurons. Importantly, general lipid droplet aggregation and global lysosomal impairment have been implicated in many neurodegenerative diseases including PD. Taken together, our data suggest a connection between age-related lysosomal impairment, lipid accumulation, and cellular senescence in DA neurons that in turn drives inflammaging in the midbrain and ultimately leads to neurodegeneration and PD.


Subject(s)
Cellular Senescence , Dopaminergic Neurons , Parkinson Disease , Dopaminergic Neurons/metabolism , Humans , Cellular Senescence/physiology , Parkinson Disease/metabolism , Parkinson Disease/pathology , Parkinson Disease/genetics , Animals , Lipid Metabolism , Glucosylceramidase/metabolism , Glucosylceramidase/genetics , Aging/metabolism , Lysosomes/metabolism , alpha-Synuclein/metabolism , alpha-Synuclein/genetics
20.
Neurosci Lett ; 837: 137916, 2024 Aug 10.
Article in English | MEDLINE | ID: mdl-39059459

ABSTRACT

After peripheral nerve injury (PNI), the long-term healing process at the injury site involves a progressive accumulation of collagen fibers and the development of localized scar tissue. Excessive formation of scar tissue within nerves hinders the process of nerve repair. In this study, we demonstrate that scar formation following nerve injury induces alterations in the local physical microenvironment, specifically an increase in nerve stiffness. Recent research has indicated heightened expression of Piezo1 in Schwann cells (SCs). Our findings also indicate Piezo1 expression in SCs and its association with suppressed proliferation and migration. Transcriptomic data suggests that activation of Piezo1 results in elevated expression of senescence-associated genes. GO enrichment analysis reveals upregulation of the TGF-ß pathway. Overall, our study highlights the potential for Piezo1-induced signaling to regulate SC senescence and its potential significance in the pathophysiology of fibrotic scar formation surrounding peripheral nerves.


Subject(s)
Cellular Senescence , Cicatrix , Fibrosis , Ion Channels , Peripheral Nerve Injuries , Schwann Cells , Schwann Cells/metabolism , Schwann Cells/pathology , Animals , Cicatrix/metabolism , Cicatrix/pathology , Cellular Senescence/physiology , Ion Channels/metabolism , Ion Channels/genetics , Peripheral Nerve Injuries/metabolism , Peripheral Nerve Injuries/pathology , Cell Proliferation , Mice , Transforming Growth Factor beta/metabolism , Male
SELECTION OF CITATIONS
SEARCH DETAIL