Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Daohong Zhou is active.

Publication


Featured researches published by Daohong Zhou.


Nature Medicine | 2016

Clearance of senescent cells by ABT263 rejuvenates aged hematopoietic stem cells in mice

Jianhui Chang; Yingying Wang; Lijian Shao; Remi Martin Laberge; Marco Demaria; Judith Campisi; Krishnamurthy Janakiraman; Norman E. Sharpless; Sheng Ding; Wei Feng; Yi Luo; Xiaoyan Wang; Nukhet Aykin-Burns; Kimberly J. Krager; Usha Ponnappan; Martin Hauer-Jensen; Aimin Meng; Daohong Zhou

Senescent cells (SCs) accumulate with age and after genotoxic stress, such as total-body irradiation (TBI). Clearance of SCs in a progeroid mouse model using a transgenic approach delays several age-associated disorders, suggesting that SCs play a causative role in certain age-related pathologies. Thus, a senolytic pharmacological agent that can selectively kill SCs holds promise for rejuvenating tissue stem cells and extending health span. To test this idea, we screened a collection of compounds and identified ABT263 (a specific inhibitor of the anti-apoptotic proteins BCL-2 and BCL-xL) as a potent senolytic drug. We show that ABT263 selectively kills SCs in culture in a cell type– and species-independent manner by inducing apoptosis. Oral administration of ABT263 to either sublethally irradiated or normally aged mice effectively depleted SCs, including senescent bone marrow hematopoietic stem cells (HSCs) and senescent muscle stem cells (MuSCs). Notably, this depletion mitigated TBI-induced premature aging of the hematopoietic system and rejuvenated the aged HSCs and MuSCs in normally aged mice. Our results demonstrate that selective clearance of SCs by a pharmacological agent is beneficial in part through its rejuvenation of aged tissue stem cells. Thus, senolytic drugs may represent a new class of radiation mitigators and anti-aging agents.


Cancer Discovery | 2017

Cellular Senescence Promotes Adverse Effects of Chemotherapy and Cancer Relapse

Marco Demaria; Monique N. O'Leary; Jianhui Chang; Lijian Shao; Su Liu; Fatouma Alimirah; Kristin Koenig; Catherine Le; Natalia Mitin; Allison M. Deal; Shani Alston; Emmeline C. Academia; Sumner Kilmarx; Alexis Valdovinos; Boshi Wang; Alain de Bruin; Brian K. Kennedy; Simon Melov; Daohong Zhou; Norman E. Sharpless; Hyman B. Muss; Judith Campisi

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.nnnSIGNIFICANCEnMany 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.


Aging (Albany NY) | 2016

Discovery of piperlongumine as a potential novel lead for the development of senolytic agents

Yingying Wang; Jianhui Chang; Xingui Liu; Xuan Zhang; Suping Zhang; Xin Zhang; Daohong Zhou; Guangrong Zheng

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.


International Journal of Radiation Oncology Biology Physics | 2017

Inhibition of Bcl-2/xl With ABT-263 Selectively Kills Senescent Type II Pneumocytes and Reverses Persistent Pulmonary Fibrosis Induced by Ionizing Radiation in Mice

Jin Pan; Deguan Li; Yanfeng Xu; Junling Zhang; Yueying Wang; Mengyi Chen; Shuai Lin; Lan Huang; Eun Joo Chung; Deborah Citrin; Yingying Wang; Martin Hauer-Jensen; Daohong Zhou; Aimin Meng

PURPOSEnIonizing radiation (IR)-induced pulmonary fibrosis (PF) is an irreversible and severe late effect of thoracic radiation therapy. The goal of this study was to determine whether clearance of senescent cells with ABT-263, a senolytic drug that can selectively kill senescent cells, can reverse PF.nnnMETHODS AND MATERIALSnC57BL/6J mice were exposed to a single dose of 17xa0Gy on the right side of the thorax. Sixteen weeks after IR, they were treated with 2 cycles of vehicle or ABT-263 (50xa0mg/kg per day for 5xa0days per cycle) by gavage. The effects of ABT-263 on IR-induced increases in senescent cells; elevation in the expression of selective inflammatory cytokines, matrix metalloproteinases, and tissue inhibitors of matrix metalloproteinases; and the severity of the tissue injury and fibrosis in the irradiated lungs were evaluated 3xa0weeks after the last treatment, in comparison with the changes observed in the irradiated lungs before treatment or after vehicle treatment.nnnRESULTSnAt 16xa0weeks after exposure of C57BL/6 mice to a single dose of 17xa0Gy, thoracic irradiation resulted in persistent PF associated with a significant increase in senescent cells. Treatment of the irradiated mice with ABT-263 after persistent PF had developed reduced senescent cells and reversed the disease.nnnCONCLUSIONSnTo our knowledge, this is the first study to demonstrate that PF can be reversed by a senolytic drug such as ABT-263 after it becomes a progressive disease.xa0Therefore, ABT-263 has the potential to be developed as a new treatment for PF.


PLOS Genetics | 2015

EEPD1 Rescues Stressed Replication Forks and Maintains Genome Stability by Promoting End Resection and Homologous Recombination Repair.

Yuehan Wu; Suk Hee Lee; Elizabeth A. Williamson; Brian L. Reinert; Ju Hwan Cho; Fen Xia; Aruna S. Jaiswal; Gayathri Srinivasan; Bhavita Patel; Alexis Brantley; Daohong Zhou; Lijian Shao; Rupak Pathak; Martin Hauer-Jensen; Sudha B. Singh; Kimi Y. Kong; Xaiohua Wu; Hyun Suk Kim; Tim Beissbarth; Jochen Gaedcke; Sandeep Burma; Jac A. Nickoloff; Robert Hromas

Replication fork stalling and collapse is a major source of genome instability leading to neoplastic transformation or cell death. Such stressed replication forks can be conservatively repaired and restarted using homologous recombination (HR) or non-conservatively repaired using micro-homology mediated end joining (MMEJ). HR repair of stressed forks is initiated by 5’ end resection near the fork junction, which permits 3’ single strand invasion of a homologous template for fork restart. This 5’ end resection also prevents classical non-homologous end-joining (cNHEJ), a competing pathway for DNA double-strand break (DSB) repair. Unopposed NHEJ can cause genome instability during replication stress by abnormally fusing free double strand ends that occur as unstable replication fork repair intermediates. We show here that the previously uncharacterized Exonuclease/Endonuclease/Phosphatase Domain-1 (EEPD1) protein is required for initiating repair and restart of stalled forks. EEPD1 is recruited to stalled forks, enhances 5’ DNA end resection, and promotes restart of stalled forks. Interestingly, EEPD1 directs DSB repair away from cNHEJ, and also away from MMEJ, which requires limited end resection for initiation. EEPD1 is also required for proper ATR and CHK1 phosphorylation, and formation of gamma-H2AX, RAD51 and phospho-RPA32 foci. Consistent with a direct role in stalled replication fork cleavage, EEPD1 is a 5’ overhang nuclease in an obligate complex with the end resection nuclease Exo1 and BLM. EEPD1 depletion causes nuclear and cytogenetic defects, which are made worse by replication stress. Depleting 53BP1, which slows cNHEJ, fully rescues the nuclear and cytogenetic abnormalities seen with EEPD1 depletion. These data demonstrate that genome stability during replication stress is maintained by EEPD1, which initiates HR and inhibits cNHEJ and MMEJ.


PLOS ONE | 2012

A Sensitive and Quantitative Polymerase Chain Reaction-Based Cell Free In Vitro Non-Homologous End Joining Assay for Hematopoietic Stem Cells

Lijian Shao; Wei Feng; Kyung Jong Lee; Benjamin P C Chen; Daohong Zhou

Hematopoietic stem cells (HSCs) are responsible for sustaining hematopoietic homeostasis and regeneration after injury for the entire lifespan of an organism. Maintenance of genomic stability is crucial for the preservation of HSCs, which depends on their efficient repair of DNA damage, particularly DNA double strand breaks (DSBs). Because of the paucity of HSCs and lack of sensitive assays, directly measuring the ability of HSCs to repair DSBs has been difficult. Therefore, we developed a sensitive and quantitative cell free in vitro non-homologous end joining (NHEJ) assay using linearized plasmids as the substrates and quantitative polymerase chain reaction (qPCR) technique. This assay can sensitively detect DSB repair via NHEJ in less than 1 µg 293T cell nuclear proteins or nuclear extracts from about 5,000 to 10,000 human BM CD34(+) hematopoietic cells. Using this assay, we confirmed that human bone marrow HSCs (CD34(+)CD38(-) cells) are less proficient in the repair of DSBs by NHEJ than HPCs (CD34(+)CD38(+) cells). In contrast, mouse quiescent HSCs (Pyronin-Y(low) LKS(+) cells) and cycling HSCs (Pyronin-Y(hi) LKS(+) cells) repaired the damage more efficiently than HPCs (LKS(-) cells). The difference in the abilities of human and mouse HSCs and HPCs to repair DSBs through NHEJ is likely attributed to their differential expression of key NHEJ DNA damage repair genes such as LIG4. These findings suggest that the qPCR-based cell free in vitro NHEJ assay can be used to sensitively measure the ability of human and mouse HSCs to repair DSBs.


Aging Cell | 2017

DNA damage and senescence in osteoprogenitors expressing Osx1 may cause their decrease with age

Ha Neui Kim; Jianhui Chang; Lijian Shao; Li Han; Srividhya Iyer; Stavros C. Manolagas; Charles A. O'Brien; Robert L. Jilka; Daohong Zhou; Maria Almeida

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.


Blood | 2016

Timing of the loss of Pten protein determines disease severity in a mouse model of myeloid malignancy

Y. Lucy Liu; Yan Yan; Cody Webster; Lijian Shao; Shelly Lensing; Hongyu Ni; Wei Feng; Natalia Colorado; Rupak Pathak; Zhifu Xiang; Martin Hauer-Jensen; Shaoguang Li; Daohong Zhou; Peter D. Emanuel

Juvenile myelomonocytic leukemia (JMML) is an aggressive pediatric mixed myelodysplastic/myeloproliferative neoplasm (MDS/MPN). JMML leukemogenesis is linked to a hyperactivated RAS pathway, with driver mutations in the KRAS, NRAS, NF1, PTPN11, or CBL genes. Previous murine models demonstrated how those genes contributed to the selective hypersensitivity of JMML cells to granulocyte macrophage-colony-stimulating factor (GM-CSF), a unifying characteristic in the disease. However, it is unclear what causes the early death in children with JMML, because transformation to acute leukemia is rare. Here, we demonstrate that loss of Pten (phosphatase and tensin homolog) protein at postnatal day 8 in mice harboring Nf1 haploinsufficiency results in an aggressive MPN with death at a murine prepubertal age of 20 to 35 days (equivalent to an early juvenile age in JMML patients). The death in the mice was due to organ infiltration with monocytes/macrophages. There were elevated activities of protein kinase B (Akt) and mitogen-activated protein kinase (MAPK) in cells at physiological concentrations of GM-CSF. These were more pronounced in mice with Nf1 haploinsufficiency than in littermates with wild-type Nf1,but this model is insufficient to cause cells to be GM-CSF hypersensitive. This new model represents a murine MPN model with features of a pediatric unclassifiable mixed MDS/MPN and mimics many clinical manifestations of JMML in terms of age of onset, aggressiveness, and organ infiltration with monocytes/macrophages. Our data suggest that the timing of the loss of PTEN protein plays a critical role in determining the disease severity in myeloid malignancies. This model may be useful for studying the pathogenesis of pediatric diseases with alterations in the Ras pathway.


Radiotherapy and Oncology | 2016

Marrow damage and hematopoietic recovery following allogeneic bone marrow transplantation for acute leukemias: Effect of radiation dose and conditioning regimen

Christopher Wilke; Shernan G. Holtan; Leslie C. Sharkey; Todd E. DeFor; Mukta Arora; Priya Premakanthan; Sophia Yohe; S. Vagge; Daohong Zhou; Jennifer Holter Chakrabarty; Marc Mahe; R. Corvò; Kathryn E. Dusenbery; Guy Storme; Daniel J. Weisdorf; Michael R. Verneris; Susanta K. Hui

BACKGROUND AND PURPOSEnTotal body irradiation (TBI) is a common component of hematopoietic cell transplantation (HCT) conditioning regimens. Preclinical studies suggest prolonged bone marrow (BM) injury after TBI could contribute to impaired engraftment and poor hematopoietic function.nnnMATERIALS AND METHODSnWe studied the longitudinal changes in the marrow environment in patients receiving allogeneic HCT with myeloablative (MA, n=42) and reduced intensity (RIC, n=56) doses of TBI from 2003-2013, including BM cellularity, histologic features of injury and repair, hematologic and immunologic recovery.nnnRESULTSnFollowing MA conditioning, a 30% decrease in the marrow cellularity persisted at 1 year post-transplant (p=0.03). RIC HCT marrow cellularity transiently decreased but returned to baseline by 6 months even though the RIC group received mostly umbilical cord blood (UCB) grafts (82%, vs. 17% in the MA cohort, p<0.01). There was no evidence of persistent marrow vascular damage or inflammation. Recipients of more intensive conditioning did not show more persistent cytopenias with the exception of a tendency for minimal thrombocytopenia. Immune recovery was similar between MA and RIC.nnnCONCLUSIONSnThese findings suggest that TBI associated with MA conditioning leads to prolonged reductions in marrow cellularity, but does not show additional histological evidence of long-term injury, which is further supported by similar peripheral counts and immunologic recovery.


Radiotherapy and Oncology | 2017

Early assessment of dosimetric and biological differences of total marrow irradiation versus total body irradiation in rodents

Susanta K. Hui; Yutaka Takahashi; Shernan G. Holtan; Rezvan Azimi; Davis M. Seelig; Masashi Yagi; Jessie Ingvalson; Parham Alaei; Leslie C. Sharkey; Behiye Kodal; Nicholas D. Peterson; Carolyn Meyer; Lindsey Godin; Michael J. Ehrhardt; Guy Storme; Daohong Zhou; Angela Panoskaltsis-Mortari

PURPOSEnTo develop a murine total marrow irradiation (TMI) model in comparison with the total body irradiation (TBI) model.nnnMATERIALS AND METHODSnMyeloablative TMI and TBI were administered in mice using a custom jig, and the dosimetric differences between TBI and TMI were evaluated. The early effects of TBI/TMI on bone marrow (BM) and organs were evaluated using histology, FDG-PET, and cytokine production. TMI and TBI with and without cyclophosphamide (Cy) were evaluated for donor cell engraftment and tissue damage early after allogeneic hematopoietic cell transplantation (HCT). Stromal derived factor-1 (SDF-1) expression was evaluated.nnnRESULTSnTMI resulted in similar dose exposure to bone and 50% reduction in dose to bystander organs. BM histology was similar between the groups. In the non-HCT model, TMI mice had significantly less acute intestinal and lung injury compared to TBI. In the HCT model, recipients of TMI had significantly less acute intestinal injury and spleen GVHD, but increased early donor cell engraftment and BM:organ SDF-1 ratio compared to TBI recipients.nnnCONCLUSIONSnThe expected BM damage was similar in both models, but the damage to other normal tissues was reduced by TMI. However, BM engraftment was improved in the TMI group compared to TBI, which may be due to enhanced production of SDF-1 in BM relative to other organs after TMI.

Collaboration


Dive into the Daohong Zhou's collaboration.

Top Co-Authors

Avatar

Lijian Shao

University of Arkansas for Medical Sciences

View shared research outputs
Top Co-Authors

Avatar

Martin Hauer-Jensen

University of Arkansas for Medical Sciences

View shared research outputs
Top Co-Authors

Avatar

Jianhui Chang

University of Arkansas for Medical Sciences

View shared research outputs
Top Co-Authors

Avatar

Rupak Pathak

University of Arkansas for Medical Sciences

View shared research outputs
Top Co-Authors

Avatar

Wei Feng

University of Arkansas for Medical Sciences

View shared research outputs
Top Co-Authors

Avatar

Yingying Wang

University of Arkansas for Medical Sciences

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge