Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 34
Filter
Add more filters










Publication year range
1.
Small ; : e2308836, 2024 Jan 22.
Article in English | MEDLINE | ID: mdl-38258401

ABSTRACT

Mixed-cation perovskite solar cells (PSCs) have attracted much attention because of the advantages of suitable bandgap and stability. It is still a challenge to rationally design and modify the perovskite/tin oxide (SnO2 ) heterogeneous interface for achieving highly efficient and stable PSCs. Herein, a strategy of one-stone-for-three-birds is proposed to achieve multi-functional interface regulation via introducing N-Chlorosuccinimide (NCS) into the solution of SnO2 : i) C═O functional group in NCS can induces strong binding affinity to uncoordinated defects (oxygen vacancies, free lead ions, etc) at the buried interface and passivate them; ii) incomplete in situ hydrolysis reactions can occur spontaneously and adjust the pH value of the SnO2 solution to achieve a more matchable energy level; iii) effectively releasing the residual stress of the underlying perovskite. As a result, a champion power conversion efficiency (PCE) of 24.74% is achieved with a device structure of ITO/SnO2 /Perovskite/Spiro-OMeTAD/Ag, which is one of the highest values for cesium-formamidinium-methylammonium (CsFAMA) triple cation PSCs. Furthermore, the device without encapsulation can sustain 94.6% of its initial PCE after the storage at room temperature and relative humidity (RH) of 20% for 40 days. The research provides a versatile way to manipulate buried interface for achieving efficient and stable PSCs.

2.
Nanomicro Lett ; 15(1): 164, 2023 Jun 29.
Article in English | MEDLINE | ID: mdl-37386337

ABSTRACT

With the rapid rise in perovskite solar cells (PSCs) performance, it is imperative to develop scalable fabrication techniques to accelerate potential commercialization. However, the power conversion efficiencies (PCEs) of PSCs fabricated via scalable two-step sequential deposition lag far behind the state-of-the-art spin-coated ones. Herein, the additive methylammonium chloride (MACl) is introduced to modulate the crystallization and orientation of a two-step sequential doctor-bladed perovskite film in ambient conditions. MACl can significantly improve perovskite film quality and increase grain size and crystallinity, thus decreasing trap density and suppressing nonradiative recombination. Meanwhile, MACl also promotes the preferred face-up orientation of the (100) plane of perovskite film, which is more conducive to the transport and collection of carriers, thereby significantly improving the fill factor. As a result, a champion PCE of 23.14% and excellent long-term stability are achieved for PSCs based on the structure of ITO/SnO2/FA1-xMAxPb(I1-yBry)3/Spiro-OMeTAD/Ag. The superior PCEs of 21.20% and 17.54% are achieved for 1.03 cm2 PSC and 10.93 cm2 mini-module, respectively. These results represent substantial progress in large-scale two-step sequential deposition of high-performance PSCs for practical applications.

3.
Small ; 19(24): e2300374, 2023 Jun.
Article in English | MEDLINE | ID: mdl-36919329

ABSTRACT

Perovskite solar cells (PSCs) have emerged as one of the most promising and competitive photovoltaic technologies, and doctor-blading is a facile and robust deposition technique to efficiently fabricate PSCs in large scale, especially matching with roll-to-roll process. Herein, it demonstrates the encouraging results of one-step, antisolvent-free doctor-bladed methylammonium lead iodide (CH3 NH3 PbI3, MAPbI3 ) PSCs under a wide range of humidity from 45% to 82%. A synergy strategy of ionic-liquid methylammonium acetate (MAAc) and molecular phenylurea additives is developed to modulate the morphology and crystallization process of MAPbI3 perovskite film, leading to high-quality MAPbI3 perovskite film with large-size crystal, low defect density, and ultrasmooth surface. Impressive power conversion efficiency (PCE) of 20.34% is achieved for doctor-bladed PSCs under the humidity over 80% with a device structure of ITO/SnO2 /MAPbI3 /Spiro-OMeTAD/Ag. It is the highest PCEs for one-step solution-processed MAPbI3 PSCs without antisolvent assistance. The research provides a facile and robust large-scale deposition technique to fabricate highly efficient and stable PSCs under a wide range of humidity, even with the humidity over 80%.

4.
IEEE Trans Image Process ; 31: 2809-2823, 2022.
Article in English | MEDLINE | ID: mdl-35312621

ABSTRACT

Existing compression methods typically focus on the removal of signal-level redundancies, while the potential and versatility of decomposing visual data into compact conceptual components still lack further study. To this end, we propose a novel conceptual compression framework that encodes visual data into compact structure and texture representations, then decodes in a deep synthesis fashion, aiming to achieve better visual reconstruction quality, flexible content manipulation, and potential support for various vision tasks. In particular, we propose to compress images by a dual-layered model consisting of two complementary visual features: 1) structure layer represented by structural maps and 2) texture layer characterized by low-dimensional deep representations. At the encoder side, the structural maps and texture representations are individually extracted and compressed, generating the compact, interpretable, inter-operable bitstreams. During the decoding stage, a hierarchical fusion GAN (HF-GAN) is proposed to learn the synthesis paradigm where the textures are rendered into the decoded structural maps, leading to high-quality reconstruction with remarkable visual realism. Extensive experiments on diverse images have demonstrated the superiority of our framework with lower bitrates, higher reconstruction quality, and increased versatility towards visual analysis and content manipulation tasks.

5.
Small ; 17(32): e2102368, 2021 Aug.
Article in English | MEDLINE | ID: mdl-34174144

ABSTRACT

Flexible perovskite solar cells (f-PSCs) have been attracting tremendous attention due to their potentially commercial prospects in flexible energy system and mobile energy system. Reducing the energy barriers and charge extraction losses at the interfaces between perovskite and charge transport layers is essential to improve both efficiency and stability of f-PSCs. Herein, 4-trifluoromethylphenylethylamine iodide (CF3 PEAI) is introduced to form a 2D perovskite at the interface between perovskite and hole transport layer (HTL). It is found that the 2D perovskite plays a dual-functional role in aligning energy band between perovskite and HTL and passivating the traps in the 3D perovskite, thus reducing energy loss and charge carrier recombination at the interface, facilitating the hole transfer from perovskite to the Spiro-OMeTAD. Consequently, the photovoltaic performance of f-PSCs is significantly improved, leading to a power conversion efficiency (PCE) of 21.1% and a certified PCE of 20.5%. Furthermore, the long-term stability of f-PSCs is greatly improved through the protection of 2D perovskite layer to the underlying 3D perovskite. This work provides an excellent strategy to produce efficient and stable f-PSCs, which will accelerate their potential applications.

6.
Sci Rep ; 10(1): 14170, 2020 08 25.
Article in English | MEDLINE | ID: mdl-32843706

ABSTRACT

Chemotherapy-induced peripheral neuropathy is among the most common dose-limiting adverse effects of cancer treatment, leading to dose reduction and discontinuation of life-saving chemotherapy and a permanently impaired quality of life for patients. Currently, no effective treatment or prevention is available. Senescence induced during cancer treatment has been shown to promote the adverse effects. Here, we show that cisplatin induces senescent-like neuronal cells in primary culture and in mouse dorsal root ganglia (DRG), as determined by the characteristic senescence markers including senescence-associated beta-galactosidase, accumulation of cytosolic p16INK4A and HMGB1, as well as increased expression of p16Ink4a, p21, and MMP-9. The accumulation of senescent-like neuronal cells in DRG is associated with cisplatin-induced peripheral neuropathy (CIPN) in mice. To determine if depletion of senescent-like neuronal cells may effectively mitigate CIPN, we used a pharmacological 'senolytic' agent, ABT263, which inhibits the anti-apoptotic proteins BCL-2 and BCL-xL and selectively kills senescent cells. Our results demonstrated that clearance of DRG senescent neuronal cells reverses CIPN, suggesting that senescent-like neurons play a role in CIPN pathogenesis. This finding was further validated using transgenic p16-3MR mice, which permit ganciclovir (GCV) to selectively kill senescent cells expressing herpes simplex virus 1 thymidine kinase (HSV-TK). We showed that CIPN was alleviated upon GCV administration to p16-3MR mice. Together, the results suggest that clearance of senescent DRG neuronal cells following platinum-based cancer treatment might be an effective therapy for the debilitating side effect of CIPN.


Subject(s)
Aniline Compounds/pharmacology , Cellular Senescence/drug effects , Cisplatin/toxicity , Neurons/pathology , Peripheral Nervous System Diseases/prevention & control , Sulfonamides/pharmacology , Animals , Biomarkers , Cells, Cultured , Female , Ganglia, Spinal/cytology , Ganglia, Spinal/drug effects , Genes, Transgenic, Suicide , Hyperalgesia/chemically induced , Hyperalgesia/pathology , Hyperalgesia/prevention & control , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Peripheral Nervous System Diseases/chemically induced , Peripheral Nervous System Diseases/pathology , Primary Cell Culture
7.
Nat Commun ; 11(1): 1996, 2020 04 24.
Article in English | MEDLINE | ID: mdl-32332723

ABSTRACT

Small molecules that selectively kill senescent cells (SCs), termed senolytics, have the potential to prevent and treat various age-related diseases and extend healthspan. The use of Bcl-xl inhibitors as senolytics is largely limited by their on-target and dose-limiting platelet toxicity. Here, we report the use of proteolysis-targeting chimera (PROTAC) technology to reduce the platelet toxicity of navitoclax (also known as ABT263), a Bcl-2 and Bcl-xl dual inhibitor, by converting it into PZ15227 (PZ), a Bcl-xl PROTAC, which targets Bcl-xl to the cereblon (CRBN) E3 ligase for degradation. Compared to ABT263, PZ is less toxic to platelets, but equally or slightly more potent against SCs because CRBN is poorly expressed in platelets. PZ effectively clears SCs and rejuvenates tissue stem and progenitor cells in naturally aged mice without causing severe thrombocytopenia. With further improvement, Bcl-xl PROTACs have the potential to become safer and more potent senolytic agents than Bcl-xl inhibitors.


Subject(s)
Aging/drug effects , Aniline Compounds/pharmacology , Blood Platelets/drug effects , Cellular Senescence/drug effects , Sulfonamides/pharmacology , Adaptor Proteins, Signal Transducing/metabolism , Aniline Compounds/chemistry , Animals , Cell Line , Female , Humans , Male , Mice , Mice, Transgenic , Models, Animal , Primary Cell Culture , Proteolysis/drug effects , Sulfonamides/chemistry , Ubiquitin-Protein Ligases , bcl-X Protein/antagonists & inhibitors , bcl-X Protein/metabolism
8.
Aging Cell ; 18(3): e12927, 2019 06.
Article in English | MEDLINE | ID: mdl-30900385

ABSTRACT

RATIONALE: Age-related changes in the intervertebral discs are the predominant contributors to back pain, a common physical and functional impairment experienced by older persons. Cellular senescence, a process wherein cells undergo growth arrest and chronically secrete numerous inflammatory molecules and proteases, has been reported to cause decline in the health and function of multiple tissues with age. Although senescent cells have been reported to increase in intervertebral degeneration (IDD), it is not known whether they are causative in age-related IDD. OBJECTIVE: The study aimed to elucidate whether a causal relationship exists between cellular senescence and age-related IDD. METHODS AND RESULTS: To examine the impact of senescent cells on age-associated IDD, we used p16-3MR transgenic mice, which enables the selective removal of p16Ink4a -positive senescent cells by the drug ganciclovir. Disc cellularity, aggrecan content and fragmentation alongside expression of inflammatory cytokine (IL-6) and matrix proteases (ADAMTS4 and MMP13) in discs of p16-3MR mice treated with GCV and untreated controls were assessed. In aged mice, reducing the per cent of senescent cells decreased disc aggrecan proteolytic degradation and increased overall proteoglycan matrix content along with improved histological disc features. Additionally, reduction of senescent cells lowered the levels of MMP13, which is purported to promote disc degenerative changes during aging. CONCLUSIONS: The findings of this study suggest that systemic reduction in the number of senescent cells ameliorates multiple age-associated changes within the disc tissue. Cellular senescence could therefore serve as a therapeutic target to restore the health of disc tissue that deteriorates with age.


Subject(s)
Aggrecans/metabolism , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Intervertebral Disc Degeneration/therapy , Intervertebral Disc/cytology , Intervertebral Disc/metabolism , Proteoglycans/metabolism , ADAMTS4 Protein/metabolism , Aging/pathology , Animals , Cell Death/physiology , Cellular Senescence/genetics , Cellular Senescence/physiology , Cyclin-Dependent Kinase Inhibitor p16/genetics , Ganciclovir/pharmacology , Interleukin-6/metabolism , Intervertebral Disc/drug effects , Intervertebral Disc/pathology , Intervertebral Disc Degeneration/metabolism , Matrix Metalloproteinase 13/metabolism , Mice , Mice, Transgenic , Thymidine Kinase/genetics , Thymidine Kinase/metabolism
9.
Aging Cell ; 18(3): e12923, 2019 06.
Article in English | MEDLINE | ID: mdl-30773784

ABSTRACT

Both an increase in osteoclast and a decrease in osteoblast numbers contribute to skeletal aging. Markers of cellular senescence, including expression of the cyclin inhibitor p16, increase with aging in several bone cell populations. The elimination of p16-expressing cells in old mice, using the INK-ATTAC transgene, increases bone mass indicating that senescent cells contribute to skeletal aging. However, the identity of the senescent cells and the extent to which ablation of p16-expressing cells may prevent skeletal aging remain unknown. Using mice expressing the p16-3MR transgene, we examined whether elimination of p16-expressing cells between 12 and 24 months of age could preserve bone mass; and whether elimination of these cells from 20 to 26 months of age could restore bone mass. The activation of the p16-3MR transgene by ganciclovir (GCV) greatly diminished p16 levels in the brain, liver, and osteoclast progenitors from the bone marrow. The age-related increase in osteoclastogenic potential of myeloid cells was also abrogated by GCV. However, GCV did not alter p16 levels in osteocytes-the most abundant cell type in bone-and had no effect on the skeletal aging of p16-3MR mice. These findings indicate that the p16-3MR transgene does not eliminate senescent osteocytes but it does eliminate senescent osteoclast progenitors and senescent cells in other tissues, as described previously. Elimination of senescent osteoclast progenitors, in and of itself, has no effect on the age-related loss of bone mass. Hence, other senescent cell types, such as osteocytes, must be the seminal culprits.


Subject(s)
Aging , Bone Density , Cellular Senescence , Osteoclasts/cytology , Stem Cells/cytology , Animals , Mice , Mice, Inbred C57BL , Mice, Transgenic
10.
Nat Commun ; 9(1): 2377, 2018 06 18.
Article in English | MEDLINE | ID: mdl-29915352

ABSTRACT

The transition of hematopoiesis from the fetal liver (FL) to the bone marrow (BM) is incompletely characterized. We demonstrate that the Wiskott-Aldrich syndrome verprolin-homologous protein (WAVE) complex 2 is required for this transition, as complex degradation via deletion of its scaffold Hem-1 causes the premature exhaustion of neonatal BM hematopoietic stem cells (HSCs). This exhaustion of BM HSC is due to the failure of BM engraftment of Hem-1-/- FL HSCs, causing early death. The Hem-1-/- FL HSC engraftment defect is not due to the lack of the canonical function of the WAVE2 complex, the regulation of actin polymerization, because FL HSCs from Hem-1-/- mice exhibit no defects in chemotaxis, BM homing, or adhesion. Rather, the failure of Hem-1-/- FL HSC engraftment in the marrow is due to the loss of c-Abl survival signaling from degradation of the WAVE2 complex. However, c-Abl activity is dispensable for the engraftment of adult BM HSCs into the BM. These findings reveal a novel function of the WAVE2 complex and define a mechanism for FL HSC fitness in the embryonic BM niche.


Subject(s)
Adaptor Proteins, Signal Transducing/physiology , Bone Marrow/physiology , Hematopoiesis , Liver/embryology , Wiskott-Aldrich Syndrome Protein Family/physiology , Adaptor Proteins, Signal Transducing/metabolism , Animals , Cell Movement , Cytoskeletal Proteins/metabolism , Fetal Development , Hematopoietic Stem Cells/physiology , Liver/physiology , Male , Mice, Inbred C57BL , Mice, Transgenic , Proto-Oncogene Proteins c-abl/metabolism
11.
Aging Cell ; 17(4): e12780, 2018 08.
Article in English | MEDLINE | ID: mdl-29766639

ABSTRACT

The selective depletion of senescent cells (SCs) by small molecules, termed senolytic agents, is a promising therapeutic approach for treating age-related diseases and chemotherapy- and radiotherapy-induced side effects. Piperlongumine (PL) was recently identified as a novel senolytic agent. However, its mechanism of action and molecular targets in SCs was unknown and thus was investigated. Specifically, we used a PL-based chemical probe to pull-down PL-binding proteins from live cells and then mass spectrometry-based proteomic analysis to identify potential molecular targets of PL in SCs. One prominent target was oxidation resistance 1 (OXR1), an important antioxidant protein that regulates the expression of a variety of antioxidant enzymes. We found that OXR1 was upregulated in senescent human WI38 fibroblasts. PL bound to OXR1 directly and induced its degradation through the ubiquitin-proteasome system in an SC-specific manner. The knockdown of OXR1 expression by RNA interference significantly increased the production of reactive oxygen species in SCs in conjunction with the downregulation of antioxidant enzymes such as heme oxygenase 1, glutathione peroxidase 2, and catalase, but these effects were much less significant when OXR1 was knocked down in non-SCs. More importantly, knocking down OXR1 selectively induced apoptosis in SCs and sensitized the cells to oxidative stress caused by hydrogen peroxide. These findings provide new insights into the mechanism by which SCs are highly resistant to oxidative stress and suggest that OXR1 is a novel senolytic target that can be further exploited for the development of new senolytic agents.


Subject(s)
Proteins/metabolism , Apoptosis/genetics , Cell Survival/genetics , Cells, Cultured , Chromatography, Liquid , Humans , Mitochondrial Proteins , Proteins/antagonists & inhibitors , Proteins/genetics , RNA, Small Interfering/genetics , Reactive Oxygen Species/analysis , Reactive Oxygen Species/metabolism , Recombinant Proteins/analysis , Recombinant Proteins/metabolism , Tandem Mass Spectrometry
12.
Blood Adv ; 2(8): 859-870, 2018 04 24.
Article in English | MEDLINE | ID: mdl-29666049

ABSTRACT

Uncovering the cellular and molecular mechanisms by which hematopoietic stem cell (HSC) self-renewal is regulated can lead to the development of new strategies for promoting ex vivo HSC expansion. Here, we report the discovery that alternative (M2)-polarized macrophages (M2-MΦs) promote, but classical (M1)-polarized macrophages (M1-MΦs) inhibit, the self-renewal and expansion of HSCs from mouse bone marrow (BM) in vitro. The opposite effects of M1-MΦs and M2-MΦs on mouse BM HSCs were attributed to their differential expression of nitric oxide synthase 2 (NOS2) and arginase 1 (Arg1), because genetic knockout of Nos2 and Arg1 or inhibition of these enzymes with a specific inhibitor abrogated the differential effects of M1-MΦs and M2-MΦs. The opposite effects of M1-MΦs and M2-MΦs on HSCs from human umbilical cord blood (hUCB) were also observed when hUCB CD34+ cells were cocultured with M1-MΦs and M2-MΦs generated from hUCB CD34- cells. Importantly, coculture of hUCB CD34+ cells with human M2-MΦs for 8 days resulted in 28.7- and 6.6-fold increases in the number of CD34+ cells and long-term SCID mice-repopulating cells, respectively, compared with uncultured hUCB CD34+ cells. Our findings could lead to the development of new strategies to promote ex vivo hUCB HSC expansion to improve the clinical utility and outcome of hUCB HSC transplantation and may provide new insights into the pathogenesis of hematological dysfunctions associated with infection and inflammation that can lead to differential macrophage polarization.


Subject(s)
Cell Proliferation/drug effects , Cell Self Renewal/drug effects , Hematopoietic Stem Cells/cytology , Macrophages/physiology , Animals , Arginase/metabolism , Coculture Techniques , Fetal Blood/cytology , Humans , Male , Mice , Nitric Oxide Synthase Type II/metabolism
13.
PLoS One ; 12(12): e0189466, 2017.
Article in English | MEDLINE | ID: mdl-29232383

ABSTRACT

During deep space missions, astronauts will be exposed to low doses of charged particle irradiation. The long-term health effects of these exposures are largely unknown. We previously showed that low doses of oxygen ion (16O) irradiation induced acute damage to the hematopoietic system, including hematopoietic progenitor and stem cells in a mouse model. However, the chronic effects of low dose 16O irradiation remain undefined. In the current study, we investigated the long-term effects of low dose 16O irradiation on the mouse hematopoietic system. Male C57BL/6J mice were exposed to 0.05 Gy, 0.1 Gy, 0.25 Gy and 1.0 Gy whole body 16O (600 MeV/n) irradiation. The effects of 16O irradiation on bone marrow (BM) hematopoietic progenitor cells (HPCs) and hematopoietic stem cells (HSCs) were examined three months after the exposure. The results showed that the frequencies and numbers of BM HPCs and HSCs were significantly reduced in 0.1 Gy, 0.25 Gy and 1.0 Gy irradiated mice compared to 0.05 Gy irradiated and non-irradiated mice. Exposure of mice to low dose 16O irradiation also significantly reduced the clongenic function of BM HPCs determined by the colony-forming unit assay. The functional defect of irradiated HSCs was detected by cobblestone area-forming cell assay after exposure of mice to 0.1 Gy, 0.25 Gy and 1.0 Gy of 16O irradiation, while it was not seen at three months after 0.5 Gy and 1.0 Gy of γ-ray irradiation. These adverse effects of 16O irradiation on HSCs coincided with an increased intracellular production of reactive oxygen species (ROS). However, there were comparable levels of cellular apoptosis and DNA damage between irradiated and non-irradiated HPCs and HSCs. These data suggest that exposure to low doses of 16O irradiation induces long-term hematopoietic injury, primarily via increased ROS production in HSCs.


Subject(s)
Hematopoietic Stem Cells/radiation effects , Oxygen/administration & dosage , Stem Cells/radiation effects , Animals , Flow Cytometry , Male , Mice , Mice, Inbred C57BL , Oxidative Stress
14.
Int J Radiat Biol ; 93(12): 1312-1320, 2017 12.
Article in English | MEDLINE | ID: mdl-28782442

ABSTRACT

PURPOSE: Exposure to proton irradiation during missions in deep space can lead to bone marrow injury. The acute effects of proton irradiation on hematopoietic stem and progenitor cells remain undefined and thus were investigated. MATERIALS AND METHODS: We exposed male C57BL/6 mice to 0.5 and 1.0 Gy proton total body irradiation (proton-TBI, 150 MeV) and examined changes in peripheral blood cells and bone marrow (BM) progenitors and LSK cells 2 weeks after exposure. RESULTS: 1.0 Gy proton-TBI significantly reduced the numbers of peripheral blood cells compared to 0.5 Gy proton-TBI and unirradiated animals, while the numbers of peripheral blood cell counts were comparable between 0.5 Gy proton-TBI and unirradiated mice. The frequencies and numbers of LSK cells and CMPs in BM of 0.5 and 1.0 Gy irradiated mice were decreased in comparison to those of normal controls. LSK cells and CMPs and their progeny exhibited a radiation-induced impairment in clonogenic function. Exposure to 1.0 Gy increased cellular apoptosis but not the production of reactive oxygen species (ROS) in CMPs two weeks after irradiation. LSK cells from irradiated mice exhibited an increase in ROS production and apoptosis. CONCLUSION: Exposure to proton-TBI can induce acute damage to BM progenitors and LSK cells.


Subject(s)
Bone Marrow Cells/cytology , Hematopoietic Stem Cells/radiation effects , Protons/adverse effects , Whole-Body Irradiation/adverse effects , Animals , Apoptosis/radiation effects , Blood Cell Count , Dose-Response Relationship, Radiation , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Male , Mice , Mice, Inbred C57BL , Oxidative Stress/radiation effects
15.
Int J Mol Sci ; 18(7)2017 Jul 04.
Article in English | MEDLINE | ID: mdl-28677663

ABSTRACT

Long Interspersed Nuclear Element 1 (LINE-1) retrotransposons are the major repetitive elements in mammalian genomes. LINE-1s are well-accepted as driving forces of evolution and critical regulators of the expression of genetic information. Alterations in LINE-1 DNA methylation may lead to its aberrant activity and are reported in virtually all human cancers and in experimental carcinogenesis. In this study, we investigated the endogenous DNA methylation status of the 5' untranslated region (UTR) of LINE-1 elements in the bone marrow hematopoietic stem cells (HSCs), hematopoietic progenitor cells (HPCs), and mononuclear cells (MNCs) in radioresistant C57BL/6J and radiosensitive CBA/J mice and in response to ionizing radiation (IR). We demonstrated that basal levels of DNA methylation within the 5'-UTRs of LINE-1 elements did not differ significantly between the two mouse strains and were negatively correlated with the evolutionary age of LINE-1 elements. Meanwhile, the expression of LINE-1 elements was higher in CBA/J mice. At two months after irradiation to 0.1 or 1 Gy of 137Cs (dose rate 1.21 Gy/min), significant decreases in LINE-1 DNA methylation in HSCs were observed in prone to radiation-induced carcinogenesis CBA/J, but not C57BL/6J mice. At the same time, no residual DNA damage, increased ROS, or changes in the cell cycle were detected in HSCs of CBA/J mice. These results suggest that epigenetic alterations may potentially serve as driving forces of radiation-induced carcinogenesis; however, future studies are needed to demonstrate the direct link between the LINE-1 DNA hypomethylation and radiation carcinogenesis.


Subject(s)
DNA Methylation/radiation effects , Hematopoietic Stem Cells/metabolism , Hematopoietic Stem Cells/radiation effects , Long Interspersed Nucleotide Elements , Radiation, Ionizing , Animals , DNA Damage , Dose-Response Relationship, Radiation , Gene Expression Regulation/radiation effects , Hematopoiesis/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Inbred CBA , Retroelements , Species Specificity
16.
Life Sci Space Res (Amst) ; 13: 39-44, 2017 May.
Article in English | MEDLINE | ID: mdl-28554508

ABSTRACT

Long-term space mission exposes astronauts to a radiation environment with potential health hazards. High-energy charged particles (HZE), including 28Si nuclei in space, have deleterious effects on cells due to their characteristics with high linear energy transfer and dense ionization. The influence of 28Si ions contributes more than 10% to the radiation dose equivalent in the space environment. Understanding the biological effects of 28Si irradiation is important to assess the potential health hazards of long-term space missions. The hematopoietic system is highly sensitive to radiation injury and bone marrow (BM) suppression is the primary life-threatening injuries after exposure to a moderate dose of radiation. Therefore, in the present study we investigated the acute effects of low doses of 28Si irradiation on the hematopoietic system in a mouse model. Specifically, 6-month-old C57BL/6J mice were exposed to 0.3, 0.6 and 0.9Gy 28Si (600MeV) total body irradiation (TBI). The effects of 28Si TBI on BM hematopoietic stem cells (HSCs) and hematopoietic progenitor cells (HPCs) were examined four weeks after the exposure. The results showed that exposure to 28Si TBI dramatically reduced the frequencies and numbers of HSCs in irradiated mice, compared to non-irradiated controls, in a radiation dose-dependent manner. In contrast, no significant changes were observed in BM HPCs regardless of radiation doses. Furthermore, irradiated HSCs exhibited a significant impairment in clonogenic ability. These acute effects of 28Si irradiation on HSCs may be attributable to radiation-induced apoptosis of HSCs, because HSCs, but not HPCs, from irradiated mice exhibited a significant increase in apoptosis in a radiation dose-dependent manner. However, exposure to low doses of 28Si did not result in an increased production of reactive oxygen species and DNA damage in HSCs and HPCs. These findings indicate that exposure to 28Si irradiation leads to acute HSC damage.


Subject(s)
Apoptosis/radiation effects , Bone Marrow/pathology , Hematopoietic Stem Cells/pathology , Whole-Body Irradiation/adverse effects , Animals , Bone Marrow/radiation effects , Cell Cycle/radiation effects , Cells, Cultured , DNA Damage/radiation effects , Hematopoietic Stem Cells/radiation effects , Male , Mice , Mice, Inbred C57BL , Oxidative Stress/radiation effects , Reactive Oxygen Species/metabolism
17.
Aging Cell ; 16(4): 693-703, 2017 08.
Article in English | MEDLINE | ID: mdl-28401730

ABSTRACT

Age-related bone loss in mice results from a decrease in bone formation and an increase in cortical bone resorption. The former is accounted by a decrease in the number of postmitotic osteoblasts which synthesize the bone matrix and is thought to be the consequence of age-dependent changes in mesenchymal osteoblast progenitors. However, there are no specific markers for these progenitors, and conclusions rely on results from in vitro cultures of mixed cell populations. Moreover, the culprits of such changes remain unknown. Here, we have used Osx1-Cre;TdRFP mice in which osteoprogenitors express the TdRFP fluorescent protein. We report that the number of TdRFP-Osx1 cells, freshly isolated from the bone marrow, declines by more than 50% between 6 and 24 months of age in both female and male mice. Moreover, TdRFP-Osx1 cells from old mice exhibited markers of DNA damage and senescence, such as γH2AX foci, G1 cell cycle arrest, phosphorylation of p53, increased p21CIP1 levels, as well as increased levels of GATA4 and activation of NF-κB - two major stimulators of the senescence-associated secretory phenotype (SASP). Bone marrow stromal cells from old mice also exhibited elevated expression of SASP genes, including several pro-osteoclastogenic cytokines, and increased capacity to support osteoclast formation. These changes were greatly attenuated by the senolytic drug ABT263. Together, these findings suggest that the decline in bone mass with age is the result of intrinsic defects in osteoprogenitor cells, leading to decreased osteoblast numbers and increased support of osteoclast formation.


Subject(s)
Aging/genetics , Mesenchymal Stem Cells/metabolism , Osteoblasts/metabolism , Osteoclasts/metabolism , Osteogenesis/genetics , Osteoporosis/genetics , Sp7 Transcription Factor/genetics , Aging/metabolism , Aging/pathology , Aniline Compounds/pharmacology , Animals , Bone and Bones/metabolism , Bone and Bones/pathology , Cell Differentiation , Cyclin-Dependent Kinase Inhibitor p21/genetics , Cyclin-Dependent Kinase Inhibitor p21/metabolism , DNA Damage , Female , G1 Phase Cell Cycle Checkpoints/genetics , GATA4 Transcription Factor/genetics , GATA4 Transcription Factor/metabolism , Gene Expression Regulation , Genes, Reporter , Histones/genetics , Histones/metabolism , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Male , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/pathology , Mice , Mice, Transgenic , NF-kappa B/genetics , NF-kappa B/metabolism , Osteoblasts/drug effects , Osteoblasts/pathology , Osteoclasts/drug effects , Osteoclasts/pathology , Osteoporosis/metabolism , Osteoporosis/pathology , Primary Cell Culture , Signal Transduction , Sp7 Transcription Factor/metabolism , Sulfonamides/pharmacology , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Red Fluorescent Protein
18.
Cancer Discov ; 7(2): 165-176, 2017 02.
Article in English | MEDLINE | ID: mdl-27979832

ABSTRACT

Cellular senescence suppresses cancer by irreversibly arresting cell proliferation. Senescent cells acquire a proinflammatory senescence-associated secretory phenotype. Many genotoxic chemotherapies target proliferating cells nonspecifically, often with adverse reactions. In accord with prior work, we show that several chemotherapeutic drugs induce senescence of primary murine and human cells. Using a transgenic mouse that permits tracking and eliminating senescent cells, we show that therapy-induced senescent (TIS) cells persist and contribute to local and systemic inflammation. Eliminating TIS cells reduced several short- and long-term effects of the drugs, including bone marrow suppression, cardiac dysfunction, cancer recurrence, and physical activity and strength. Consistent with our findings in mice, the risk of chemotherapy-induced fatigue was significantly greater in humans with increased expression of a senescence marker in T cells prior to chemotherapy. These findings suggest that senescent cells can cause certain chemotherapy side effects, providing a new target to reduce the toxicity of anticancer treatments. SIGNIFICANCE: Many genotoxic chemotherapies have debilitating side effects and also induce cellular senescence in normal tissues. The senescent cells remain chronically present where they can promote local and systemic inflammation that causes or exacerbates many side effects of the chemotherapy. Cancer Discov; 7(2); 165-76. ©2016 AACR.This article is highlighted in the In This Issue feature, p. 115.


Subject(s)
Antineoplastic Agents/adverse effects , Breast Neoplasms/drug therapy , Cellular Senescence , Cyclin-Dependent Kinase Inhibitor p16/genetics , Animals , Antineoplastic Agents/pharmacology , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Proliferation/drug effects , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Female , Humans , Mice , Mice, Transgenic , Neoplasm Recurrence, Local
19.
Aging (Albany NY) ; 8(11): 2915-2926, 2016 11 19.
Article in English | MEDLINE | ID: mdl-27913811

ABSTRACT

Accumulating evidence indicates that senescent cells play an important role in many age-associated diseases. The pharmacological depletion of senescent cells (SCs) with a "senolytic agent", a small molecule that selectively kills SCs, is a potential novel therapeutic approach for these diseases. Recently, we discovered ABT-263, a potent and highly selective senolytic agent, by screening a library of rationally-selected compounds. With this screening approach, we also identified a second senolytic agent called piperlongumine (PL). PL is a natural product that is reported to have many pharmacological effects, including anti-tumor activity. We show here that PL preferentially killed senescent human WI-38 fibroblasts when senescence was induced by ionizing radiation, replicative exhaustion, or ectopic expression of the oncogene Ras. PL killed SCs by inducing apoptosis, and this process did not require the induction of reactive oxygen species. In addition, we found that PL synergistically killed SCs in combination with ABT-263, and initial structural modifications to PL identified analogs with improved potency and/or selectivity in inducing SC death. Overall, our studies demonstrate that PL is a novel lead for developing senolytic agents.


Subject(s)
Cellular Senescence/drug effects , Dioxolanes/pharmacology , Aniline Compounds/pharmacology , Antineoplastic Agents/pharmacology , Apoptosis , Fibroblasts/drug effects , Fibroblasts/physiology , Genes, ras , Humans , Reactive Oxygen Species/metabolism , Sulfonamides/pharmacology
20.
PLoS One ; 11(7): e0158097, 2016.
Article in English | MEDLINE | ID: mdl-27367604

ABSTRACT

One of the major health risks to astronauts is radiation on long-duration space missions. Space radiation from sun and galactic cosmic rays consists primarily of 85% protons, 14% helium nuclei and 1% high-energy high-charge (HZE) particles, such as oxygen (16O), carbon, silicon, and iron ions. HZE particles exhibit dense linear tracks of ionization associated with clustered DNA damage and often high relative biological effectiveness (RBE). Therefore, new knowledge of risks from HZE particle exposures must be obtained. In the present study, we investigated the acute effects of low doses of 16O irradiation on the hematopoietic system. Specifically, we exposed C57BL/6J mice to 0.1, 0.25 and 1.0 Gy whole body 16O (600 MeV/n) irradiation and examined the effects on peripheral blood (PB) cells, and bone marrow (BM) hematopoietic stem cells (HSCs) and hematopoietic progenitor cells (HPCs) at two weeks after the exposure. The results showed that the numbers of white blood cells, lymphocytes, monocytes, neutrophils and platelets were significantly decreased in PB after exposure to 1.0 Gy, but not to 0.1 or 0.25 Gy. However, both the frequency and number of HPCs and HSCs were reduced in a radiation dose-dependent manner in comparison to un-irradiated controls. Furthermore, HPCs and HSCs from irradiated mice exhibited a significant reduction in clonogenic function determined by the colony-forming and cobblestone area-forming cell assays. These acute adverse effects of 16O irradiation on HSCs coincided with an increased production of reactive oxygen species (ROS), enhanced cell cycle entry of quiescent HSCs, and increased DNA damage. However, none of the 16O exposures induced apoptosis in HSCs. These data suggest that exposure to low doses of 16O irradiation induces acute BM injury in a dose-dependent manner primarily via increasing ROS production, cell cycling, and DNA damage in HSCs. This finding may aid in developing novel strategies in the protection of the hematopoietic system from space radiation.


Subject(s)
Hematopoietic Stem Cells/radiation effects , Oxygen/adverse effects , Animals , Blood Cell Count , Cell Cycle/radiation effects , DNA Damage , Dose-Response Relationship, Radiation , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Male , Mice , Mice, Inbred C57BL , Oxidative Stress/radiation effects , Reactive Oxygen Species/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...