Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Cells ; 12(15)2023 08 05.
Article in English | MEDLINE | ID: mdl-37566086

ABSTRACT

Cellular senescence contributes importantly to aging and aging-related diseases, including idiopathic pulmonary fibrosis (IPF). Alveolar epithelial type II (ATII) cells are progenitors of alveolar epithelium, and ATII cell senescence is evident in IPF. Previous studies from this lab have shown that increased expression of plasminogen activator inhibitor 1 (PAI-1), a serine protease inhibitor, promotes ATII cell senescence through inducing p53, a master cell cycle repressor, and activating p53-p21-pRb cell cycle repression pathway. In this study, we further show that PAI-1 binds to proteasome components and inhibits proteasome activity and p53 degradation in human lung epithelial A549 cells and primary mouse ATII cells. This is associated with a senescence phenotype of these cells, manifested as increased p53 and p21 expression, decreased phosphorylated retinoblastoma protein (pRb), and increased senescence-associated beta-galactose (SA-ß-gal) activity. Moreover, we find that, although overexpression of wild-type PAI-1 (wtPAI-1) or a secretion-deficient, mature form of PAI-1 (sdPAI-1) alone induces ATII cell senescence (increases SA-ß-gal activity), only wtPAI-1 induces p53, suggesting that the premature form of PAI-1 is required for the interaction with the proteasome. In summary, our data indicate that PAI-1 can bind to proteasome components and thus inhibit proteasome activity and p53 degradation in ATII cells. As p53 is a master cell cycle repressor and PAI-1 expression is increased in many senescent cells, the results from this study will have a significant impact not only on ATII cell senescence/lung fibrosis but also on the senescence of other types of cells in different diseases.


Subject(s)
Alveolar Epithelial Cells , Idiopathic Pulmonary Fibrosis , Plasminogen Activator Inhibitor 1 , Tumor Suppressor Protein p53 , Animals , Humans , Mice , Alveolar Epithelial Cells/metabolism , Idiopathic Pulmonary Fibrosis/metabolism , Plasminogen Activator Inhibitor 1/metabolism , Proteasome Endopeptidase Complex/metabolism , Tumor Suppressor Protein p53/metabolism
2.
Aging Dis ; 14(2): 515-528, 2023 Apr 01.
Article in English | MEDLINE | ID: mdl-37008063

ABSTRACT

The etiology for late-onset Alzheimer's disease (LOAD), which accounts for >95% of Alzheimer's disease (AD) cases, is unknown. Emerging evidence suggests that cellular senescence contributes importantly to AD pathophysiology, although the mechanisms underlying brain cell senescence and by which senescent cells promote neuro-pathophysiology remain unclear. In this study we show for the first time that the expression of plasminogen activator inhibitor 1 (PAI-1), a serine protease inhibitor, is increased, in correlation with the increased expression of cell cycle repressors p53 and p21, in the hippocampus/cortex of senescence accelerated mouse prone 8 (SAMP8) mice and LOAD patients. Double immunostaining results show that astrocytes in the brain of LOAD patients and SAMP8 mice express higher levels of senescent markers and PAI-1, compared to astrocytes in the corresponding controls. In vitro studies further show that overexpression of PAI-1 alone, intracellularly or extracellularly, induced senescence, whereas inhibition or silencing PAI-1 attenuated H2O2-induced senescence, in primary mouse and human astrocytes. Treatment with the conditional medium (CM) from senescent astrocytes induced neuron apoptosis. Importantly, the PAI-1 deficient CM from senescent astrocytes that overexpress a secretion deficient PAI-1 (sdPAI-1) has significantly reduced effect on neurons, compared to the PAI-1 containing CM from senescent astrocytes overexpressing wild type PAI-1 (wtPAI-1), although sdPAI-1 and wtPAI-1 induce similar degree of astrocyte senescence. Together, our results suggest that increased PAI-1, intracellularly or extracellularly, may contribute to brain cell senescence in LOAD and that senescent astrocytes can induce neuron apoptosis through secreting pathologically active molecules, including PAI-1.

3.
Ann N Y Acad Sci ; 1479(1): 148-158, 2020 11.
Article in English | MEDLINE | ID: mdl-32602122

ABSTRACT

Exposure of rats to 2-chloroethyl ethyl sulfide (CEES), an analog of sulfur mustard, can cause acute lung injury (ALI), resulting in increased inflammation and coagulation and altered levels of plasma microRNAs (miRNAs). Rats were exposed to aerosolized CEES and euthanized 12 h later for collection of tissue and plasma. Profiling of miRNAs in plasma, using a TaqMan-based RT-PCR array, revealed 14 differentially expressed miRNAs. Target gene prediction and pathway analysis revealed miRNA-mediated regulation of organismal injury, inflammation, and respiratory diseases. miR-140-5p, a marker of ALI, was downregulated in the plasma, lung, liver, and kidney of CEES-exposed rats, with a concomitant increase in the expression of the inflammation markers IL-6 and IL-1α and the coagulation marker tissue factor (F3). Exposure of rat airway epithelial cells (RL-65) to CEES (0.5 mM) caused cell death and a decrease in miR-140-5p both in cells and media supernatant. This was accompanied by an increase in cellular mRNA levels of IL-6, IL-1α, and F3, as well as FGF9 and EGR2, putative targets of miR-140. Knockdown of miR-140 by specific oligos in RL-65 cells mimicked the in vivo CEES-mediated effects, leading to significantly increased mRNA levels of IL-6, IL-1α, F3, FGF9, and EGR2. Our study identifies miR-140-5p as a mediator of CEES-induced ALI, which could potentially be targeted for therapy.


Subject(s)
Acute Lung Injury , Blood Coagulation/drug effects , Chemical Warfare Agents/toxicity , MicroRNAs/metabolism , Mustard Gas/analogs & derivatives , Acute Lung Injury/chemically induced , Acute Lung Injury/genetics , Acute Lung Injury/metabolism , Animals , Early Growth Response Protein 2/genetics , Early Growth Response Protein 2/metabolism , Fibroblast Growth Factor 9/genetics , Fibroblast Growth Factor 9/metabolism , Inflammation/chemically induced , Inflammation/genetics , Inflammation/metabolism , Interleukin-1alpha/genetics , Interleukin-1alpha/metabolism , Interleukin-6/genetics , Interleukin-6/metabolism , Male , MicroRNAs/genetics , Mustard Gas/toxicity , Rats , Rats, Sprague-Dawley
4.
Am J Respir Cell Mol Biol ; 62(3): 319-330, 2020 03.
Article in English | MEDLINE | ID: mdl-31513752

ABSTRACT

Senescence of alveolar type II (ATII) cells, progenitors of the alveolar epithelium, is a pathological feature and contributes importantly to the pathogenesis of idiopathic pulmonary fibrosis. Despite recognition of the importance of ATII cell senescence in idiopathic pulmonary fibrosis pathogenesis, how ATII cell senescence is regulated and how senescent ATII cells contribute to lung fibrogenesis remain unclear. In this study, we show that TGF-ß1 (transforming growth factor-ß1), a most ubiquitous and potent profibrotic cytokine, induces plasminogen activator inhibitor-1 (PAI-1), a cell senescence and fibrosis mediator, and p16 as well as senescence, but not apoptosis, in primary mouse ATII cells. We also found that senescent ATII cells secrete various cytokines and chemokines, including IL-4 and IL-13, which stimulate the expression of genes associated with a profibrotic phenotype in alveolar macrophages. Similar responses were also observed in TGF-ß1-treated rat ATII (L2) and rat macrophage NR8383 cells. Deletion of PAI-1 or inhibition of PAI-1 activity with a small molecule PAI-1 inhibitor, however, blocks TGF-ß1-induced senescence as well as a senescence-associated secretory phenotype in ATII and L2 cells and, consequently, the stimulatory effects of the conditioned medium from senescent ATII/L2 cells on macrophages. Moreover, we show that silencing p16 ameliorates PAI-1 protein-induced ATII cell senescence and secretion of profibrotic mediators. Our data suggest that PAI-1 mediates TGF-ß1-induced ATII cell senescence and secretion of profibrotic mediators through inducing p16, and they also suggest that senescent ATII cells contribute to lung fibrogenesis in part by activating alveolar macrophages through secreting profibrotic and proinflammatory mediators.


Subject(s)
Alveolar Epithelial Cells/cytology , Cellular Senescence/physiology , Macrophage Activation/physiology , Macrophages, Alveolar/physiology , Serpin E2/physiology , Transforming Growth Factor beta1/physiology , Alveolar Epithelial Cells/metabolism , Animals , Cells, Cultured , Chemokines/metabolism , Culture Media, Conditioned/pharmacology , Cyclin-Dependent Kinase Inhibitor p16/physiology , Cytokines/metabolism , Genes, p16 , Mice , Mice, Knockout , Pulmonary Fibrosis/pathology , RNA Interference , RNA, Small Interfering/genetics , Rats , Serpin E2/deficiency , Serpin E2/genetics
5.
Biochim Biophys Acta Mol Basis Dis ; 1863(1): 92-102, 2017 01.
Article in English | MEDLINE | ID: mdl-27750040

ABSTRACT

Expression of T17M rhodopsin (T17M) in rods activates the Unfolded Protein Response (UPR) and leads to the development of autosomal dominant retinitis pigmentosa (adRP). The rod death occurs in adRP retinas prior to cone photoreceptor death, so the mechanism by which cone photoreceptors die remains unclear. Therefore, the goal of the study was to verify whether UPR in rods induces TNFa-mediated signaling to the cones and to determine whether the TNFa deficit could prevent adRP cone cell death. Primary rod photoreceptors and cone-derived 661W cells transfected with siRNA against TNFa were treated with tunicamycin to mimic activation of UPR in T17M retinas expressing normal and reduced TNFa levels. The 661W cells were then exposed to recombinant TNFa to evaluate cell viability. In vivo, the role of TNFa was assessed in T17M TNFa+/- mice by electroretinography, optical coherence tomography, histology, immunohistochemistry, and a cytokine enzyme-linked immunosorbent assay. Rods overexpressed and secreted TNFa in response to UPR activation. The recombinant TNFa treatment lowered the number of viable cones, inducing cell death through elevation of pro-inflammatory cytokines and caspase-3/7 activity. The TNFa deficiency significantly protected adRP retinas. The photopic ERG amplitudes and the number of surviving cones dramatically increased in T17M TNFa+/- mice. This neuroprotection was associated with a reduced level of pro-inflammatory cytokines. Our results indicate that rod photoreceptors, following UPR activation during adRP progression, secrete TNFa and signal a self-destructive program to the cones, resulting in their cell death. TNFa therefore holds promise as a therapeutic target for treatment of adRP.


Subject(s)
Gene Knockdown Techniques , Retinal Cone Photoreceptor Cells/pathology , Retinitis Pigmentosa/genetics , Retinitis Pigmentosa/pathology , Tumor Necrosis Factor-alpha/genetics , Animals , Cell Survival , Cells, Cultured , Female , Inflammation/genetics , Inflammation/immunology , Inflammation/pathology , Male , Mice , Mice, Inbred C57BL , Retinal Cone Photoreceptor Cells/cytology , Retinal Cone Photoreceptor Cells/immunology , Retinal Rod Photoreceptor Cells , Retinitis Pigmentosa/immunology , Tumor Necrosis Factor-alpha/immunology , Unfolded Protein Response
6.
PLoS One ; 8(10): e78043, 2013.
Article in English | MEDLINE | ID: mdl-24205081

ABSTRACT

Breast cancer patients, who are already at increased risk of developing bone metastases and osteolytic bone damage, are often treated with doxorubicin. Unfortunately, doxorubicin has been reported to induce damage to bone. Moreover, we have previously reported that doxorubicin treatment increases circulating levels of TGFß in murine pre-clinical models. TGFß has been implicated in promoting osteolytic bone damage, a consequence of increased osteoclast-mediated resorption and suppression of osteoblast differentiation. Therefore, we hypothesized that in a preclinical breast cancer bone metastasis model, administration of doxorubicin would accelerate bone loss in a TGFß-mediated manner. Administration of doxorubicin to 4T1 tumor-bearing mice produced an eightfold increase in osteolytic lesion areas compared untreated tumor-bearing mice (P = 0.002) and an almost 50% decrease in trabecular bone volume expressed in BV/TV (P = 0.0005), both of which were rescued by anti-TGFß antibody (1D11). Doxorubicin, which is a known inducer of oxidative stress, decreased osteoblast survival and differentiation, which was rescued by N-acetyl cysteine (NAC). Furthermore, doxorubicin treatment decreased Cu-ZnSOD (SOD1) expression and enzyme activity in vitro, and treatment with anti-TGFß antibody was able to rescue both. In conclusion, a combination therapy using doxorubicin and anti-TGFß antibody might be beneficial for preventing therapy-related bone loss in cancer patients.


Subject(s)
Bone Neoplasms/metabolism , Breast Neoplasms/complications , Breast Neoplasms/metabolism , Doxorubicin/adverse effects , Doxorubicin/therapeutic use , Oxidative Stress/drug effects , Transforming Growth Factor beta/metabolism , Animals , Antibodies/therapeutic use , Bone Neoplasms/secondary , Cell Differentiation/drug effects , Cell Survival/drug effects , Disease Models, Animal , Female , Mice , Osteoblasts/cytology , Osteoblasts/drug effects , Transforming Growth Factor beta/antagonists & inhibitors
7.
Free Radic Biol Med ; 53(12): 2298-307, 2012 Dec 15.
Article in English | MEDLINE | ID: mdl-23085426

ABSTRACT

Radiation therapy is an integral part of treatment for cancer patients; however, major side effects of this modality include aberrant bone remodeling and bone loss. Ionizing radiation (IR) is a major external factor that contributes to a significant increase in oxidative stress such as reactive oxygen species (ROS), has been implicated in osteoporotic phenotypes, and has been implicated in osteoporotic phenotypes, bone loss, and fracture risk. One of the major cellular defenses against heightened oxidative stress is mediated by nuclear factor (erythroid-derived 2)-like 2 (Nrf2), a master transcription factor that regulates induction of antioxidant gene expression and phase II antioxidant enzymes. Our objective was to test the hypothesis that loss of functional Nrf2 increases radiation-induced bone loss. We irradiated (single dose, 20Gy) the hindlegs of age- and sex-matched Nrf2(+/+) and Nrf2(-/-) mice. After 1 month, microCT analysis and histology revealed a drastic overall decrease in the bone volume after irradiation of mice lacking Nrf2. Although radiation exposure led to bone loss in mice with intact Nrf2, it was dramatically enhanced by loss of Nrf2. Furthermore, in the absence of Nrf2, a decrease in osteoblast mineralization was noted in calvarial osteoblasts compared with wild-type controls, and treatment with a common antioxidant, N-acetyl-l-cysteine (NAC), was able to rescue the mineralization. As expected, we observed a higher number of osteoclasts in Nrf2(-/-) mice compared to Nrf2(+/+) mice, and after irradiation, the trend remained the same. RT-PCR analysis of calvarial osteoblasts revealed that in the absence of Nrf2, the expression of RANKL was increased after irradiation. Interestingly, RANKL expression was suppressed when the calvarial osteoblasts were treated with NAC before IR exposure. Taken together, our data suggest that loss of Nrf2 leads to heightened oxidative stress and increased susceptibility to radiation-induced bone loss.


Subject(s)
NF-E2-Related Factor 2/genetics , Osteoporosis/genetics , RANK Ligand/metabolism , Radiation Injuries, Experimental/genetics , Animals , Bone Marrow Cells/physiology , Bone Marrow Cells/radiation effects , Cell Survival/radiation effects , Mice , Mice, Knockout , NF-E2-Related Factor 2/deficiency , Osteoblasts/physiology , Osteoblasts/radiation effects , Osteoclasts/physiology , Osteoclasts/radiation effects , Osteoporosis/metabolism , Oxidative Stress , RANK Ligand/genetics , Radiation Injuries, Experimental/metabolism , Radiation Tolerance , Skull/metabolism , Skull/pathology , Skull/radiation effects , Tibia/metabolism , Tibia/pathology , Tibia/radiation effects , Up-Regulation/radiation effects
8.
PLoS One ; 6(11): e27090, 2011.
Article in English | MEDLINE | ID: mdl-22096521

ABSTRACT

Breast cancer often metastasizes to bone causing osteolytic bone resorption which releases active TGFß. Because TGFß favors progression of breast cancer metastasis to bone, we hypothesized that treatment using anti-TGFß antibody may reduce tumor burden and rescue tumor-associated bone loss in metastatic breast cancer. In this study we have tested the efficacy of an anti-TGFß antibody 1D11 preventing breast cancer bone metastasis. We have used two preclinical breast cancer bone metastasis models, in which either human breast cancer cells or murine mammary tumor cells were injected in host mice via left cardiac ventricle. Using several in vivo, in vitro and ex vivo assays, we have demonstrated that anti-TGFß antibody treatment have significantly reduced tumor burden in the bone along with a statistically significant threefold reduction in osteolytic lesion number and tenfold reduction in osteolytic lesion area. A decrease in osteoclast numbers (p = 0.027) in vivo and osteoclastogenesis ex vivo were also observed. Most importantly, in tumor-bearing mice, anti-TGFß treatment resulted in a twofold increase in bone volume (p<0.01). In addition, treatment with anti-TGFß antibody increased the mineral-to-collagen ratio in vivo, a reflection of improved tissue level properties. Moreover, anti-TGFß antibody directly increased mineralized matrix formation in calverial osteoblast (p = 0.005), suggesting a direct beneficial role of anti-TGFß antibody treatment on osteoblasts. Data presented here demonstrate that anti-TGFß treatment may offer a novel therapeutic option for tumor-induced bone disease and has the dual potential for simultaneously decreasing tumor burden and rescue bone loss in breast cancer to bone metastases. This approach of intervention has the potential to reduce skeletal related events (SREs) in breast cancer survivors.


Subject(s)
Antibodies/therapeutic use , Bone Neoplasms/prevention & control , Bone and Bones/drug effects , Bone and Bones/metabolism , Breast Neoplasms/complications , Transforming Growth Factor beta/antagonists & inhibitors , Animals , Antibodies/pharmacology , Bone Neoplasms/metabolism , Bone Neoplasms/secondary , Bone and Bones/cytology , Cell Differentiation/drug effects , Cell Line, Tumor , Collagen/metabolism , Female , Humans , Mice , Mice, Nude , Osteoblasts/cytology , Osteoblasts/drug effects , Osteoclasts/cytology , Osteoclasts/drug effects , Osteogenesis/drug effects , Real-Time Polymerase Chain Reaction
9.
PLoS One ; 6(9): e25215, 2011.
Article in English | MEDLINE | ID: mdl-21949884

ABSTRACT

Cutaneous wound healing is a normal physiological process and comprises different phases. Among these phases, angiogenesis or new blood vessel formation in wound tissue plays an important role. Skin is richly supplied by sympathetic nerves and evidences indicate the significant role of the sympathetic nervous system in cutaneous wound healing. Dopamine (DA) is an important catecholamine neurotransmitter released by the sympathetic nerve endings and recent studies have demonstrated the potent anti-angiogenic action of DA, which is mediated through its D(2) DA receptors. We therefore postulate that this endogenous catecholamine neurotransmitter may have a role in the neovascularization of dermal wound tissues and subsequently in the process of wound healing. In the present study, the therapeutic efficacy of D(2) DA receptor antagonist has been investigated for faster wound healing in a murine model of full thickness dermal wound. Our results indicate that treatment with specific D(2) DA receptor antagonist significantly expedites the process of full thickness normal dermal wound healing in mice by inducing angiogenesis in wound tissues. The underlined mechanisms have been attributed to the up-regulation of homeobox transcription factor HoxD3 and its target α5ß1 integrin, which play a pivotal role in wound angiogenesis. Since D(2) DA receptor antagonists are already in clinical use for other disorders, these results have significant translational value from the bench to the bedside for efficient wound management along with other conventional treatment modalities.


Subject(s)
Dermis/blood supply , Dermis/injuries , Dopamine/pharmacology , Neovascularization, Physiologic/drug effects , Neovascularization, Physiologic/physiology , Wound Healing/physiology , Wounds and Injuries/prevention & control , Animals , Blotting, Western , Cells, Cultured , DNA-Binding Proteins/metabolism , Dermis/drug effects , Dopamine Agents/pharmacology , Dopamine Antagonists/pharmacology , Dopamine D2 Receptor Antagonists , Endothelium, Vascular/cytology , Endothelium, Vascular/drug effects , Endothelium, Vascular/metabolism , Flow Cytometry , Integrin alpha5beta1/metabolism , Mice , Receptors, Dopamine D2/metabolism , Salicylamides/pharmacology , Umbilical Veins/cytology , Umbilical Veins/drug effects , Umbilical Veins/metabolism , Wounds and Injuries/pathology
10.
Asian Pac J Cancer Prev ; 8(4): 578-82, 2007.
Article in English | MEDLINE | ID: mdl-18260732

ABSTRACT

Globally, colorectal cancer is the third commonest cancer in men since 1975.The present study focuses on the preventive strategies aimed at reducing the incidences and mortality of large bowel cancer. Chemoprevention of colon cancer appears to be a very realistic possibility because various intermediate stages have been identified preceding the development of malignant colonic tumors. Several studies have demonstrated that generous consumption of vegetables reduces the risk of colon cancer. This idea has prompted the present investigation to search for some novel plant products, which may have possible anticarcinogenic activity. It has already been proved from various experiments that chemopreventive agents, by virtue of their anti-oxidant, anti-inflammatory, anti-proliferative, apoptosis-inducing activity, act at various levels including molecular, cellular, tissue and organ levels to interfere with carcinogens. Previous studies from our laboratory have already reported the inhibitory effect of cinnamon and cardamom on azoxymethane induced colon carcinogenesis by virtue of their anti-inflammatory, anti-proliferative and pro-apoptotic activity. This particular experiment was carried out to assess the anti-oxidative potential of these spices. Aqueous suspensions of cinnamon and cardamom have been shown to enhance the level of detoxifying enzyme (GST activity) with simultaneous decrease in lipid peroxidation levels in the treatment groups when compared to that of the carcinogen control group.


Subject(s)
Cinnamomum zeylanicum , Colonic Neoplasms/prevention & control , Elettaria , Glutathione Transferase/metabolism , Lipid Peroxidation/drug effects , Phytotherapy , Precancerous Conditions/prevention & control , Animals , Azoxymethane/toxicity , Carcinogens/toxicity , Colon/enzymology , Colonic Neoplasms/chemically induced , Colonic Neoplasms/enzymology , Liver/enzymology , Mice , Precancerous Conditions/chemically induced , Precancerous Conditions/enzymology
SELECTION OF CITATIONS
SEARCH DETAIL
...