1
|
Shi J, Zhao F, Qiu T, Ren D, Li Z, Ma J, Zhao J. High-altitude hypoxia exacerbates chemotherapy-induced myelosuppression by lowering serum G-CSF/GM-CSF and regulating apoptosis and proliferation. Discov Oncol 2025; 16:938. [PMID: 40434597 PMCID: PMC12119416 DOI: 10.1007/s12672-025-02611-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Accepted: 05/07/2025] [Indexed: 05/29/2025] Open
Abstract
The unique hypoxic environment in high-altitude regions is increasingly drawing attention for its impact on the health of residents, particularly in patients post-chemotherapy. This study aimed to investigate the effects and potential mechanisms of high-altitude hypoxia on myelosuppression following chemotherapy, with the goal of providing a theoretical basis for clinical treatment. A retrospective clinical study of 80 patients with breast cancer revealed that patients in the plateau exhibited a significantly higher incidence of grade 3 or higher neutropenia and any level of neutropenia post-chemotherapy than those in the plain, with propensity score matching (PSM) confirming these associations. Animal experiments revealed that high-altitude hypoxia reduced the white blood cell (WBC) count, granulocyte count, lymphocyte count, and number of bone marrow nucleated cells (BMNCs) in cyclophosphamide (CTX)-treated mice. Additionally, high-altitude hypoxia induced a significant reduction in the proliferation index and an elevation in apoptosis rates in BMNCs. High-altitude hypoxia also significantly reduced serum levels of granulocyte colony-stimulating factor (G-CSF) and granulocyte-macrophage colony-stimulating factor (GM-CSF). Transcriptomic analysis of BMNCs demonstrated that high-altitude hypoxia might modulate the hematopoietic function in CTX-induced myelosuppression mice through pathways related to hematopoiesis, such as porphyrin metabolism, hematopoietic cell lineage, ECM-receptor interaction, and PI3K-Akt signaling pathway. Our results suggest that high-altitude hypoxia exacerbates chemotherapy-induced myelosuppression, possibly through reducing the serum level of G-CSF/GM-CSF and regulating apoptosis and proliferation by PI3K-Akt signaling pathway, highlighting that cancer patients undergoing chemotherapy in hypoxic environments may require enhanced supportive care to mitigate these adverse effects.
Collapse
Affiliation(s)
- Jing Shi
- School of Clinical Medicine of Qinghai University, Affiliated Hospital of Qinghai University, Xining, 810000, China
- Breast Disease Diagnosis and Treatment Center of Affiliated Hospital of Qinghai University, Affiliated Cancer Hospital of Qinghai University, Xining, 810000, China
- Affiliated Hospital of Jining Medical University, Jining, 272029, Shandong, China
| | - Fuxing Zhao
- Breast Disease Diagnosis and Treatment Center of Affiliated Hospital of Qinghai University, Affiliated Cancer Hospital of Qinghai University, Xining, 810000, China
| | - Tianlei Qiu
- Breast Disease Diagnosis and Treatment Center of Affiliated Hospital of Qinghai University, Affiliated Cancer Hospital of Qinghai University, Xining, 810000, China
| | - Dengfeng Ren
- Breast Disease Diagnosis and Treatment Center of Affiliated Hospital of Qinghai University, Affiliated Cancer Hospital of Qinghai University, Xining, 810000, China
| | - Zitao Li
- Breast Disease Diagnosis and Treatment Center of Affiliated Hospital of Qinghai University, Affiliated Cancer Hospital of Qinghai University, Xining, 810000, China
| | - Junli Ma
- Affiliated Hospital of Jining Medical University, Jining, 272029, Shandong, China
| | - Jiuda Zhao
- Breast Disease Diagnosis and Treatment Center of Affiliated Hospital of Qinghai University, Affiliated Cancer Hospital of Qinghai University, Xining, 810000, China.
| |
Collapse
|
2
|
Zhou X, Bao Q, Cui Y, Li X, Yang C, Yang Y, Gao Y, Chen D, Huang J. Life destiny of erythrocyte in high altitude erythrocytosis: mechanisms underlying the progression from physiological (moderate) to pathological (excessive) high-altitude erythrocytosis. Front Genet 2025; 16:1528935. [PMID: 40242475 PMCID: PMC12000012 DOI: 10.3389/fgene.2025.1528935] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Accepted: 03/17/2025] [Indexed: 04/18/2025] Open
Abstract
High-altitude polycythemia (HAPC) represents a pathological escalation of the physiological erythrocytosis induced by chronic hypoxia exposure. While moderate erythroid expansion enhances oxygen delivery, HAPC manifests as hematologic disorder characterized by hemoglobin thresholds (≥21 g/dL males; ≥19 g/dL females) and multi-organ complications including microcirculatory thrombosis, right ventricular hypertrophy, and uric acid dysmetabolism. This review critically evaluates the continuum between adaptive and maladaptive polycythemia through multiscale analysis of erythrocyte biology. We integrate genomic predisposition patterns, bone marrow erythroid kinetic studies, and peripheral erythrocyte pathophenotypes revealed by multi-omics profiling (iron-redox proteome, hypoxia-metabolome crosstalk). Current diagnostic limitations are highlighted, particularly the oversimplification of hemoglobin cutoffs that neglect transitional dynamics in erythrocyte turnover. By reconstructing the erythroid life cycle-from hypoxia-sensitive progenitor commitment to senescent cell clearance-we propose a phase transition model where cumulative epigenetic-metabolic derangements overcome homeostatic buffers, triggering pathological erythroid amplification. These insights reframe HAPC as a systems biology failure of erythroid adaptation, informing predictive biomarkers and targeted interventions to preserve hematological homeostasis in hypoxic environments.
Collapse
Affiliation(s)
- Xiaoying Zhou
- Department of High Altitude Physiology and Pathology, College of High Altitude Military Medicine, Army Medical University, Chongqing, China
- Key Laboratory of Extreme Environmental Medicine, Ministry of Education, Chongqing, China
| | - Quanwei Bao
- Department of Emergency Medicine, Daping Hospital, Army Medical University, Chongqing, China
| | - Yu Cui
- Department of High Altitude Physiology and Pathology, College of High Altitude Military Medicine, Army Medical University, Chongqing, China
- Key Laboratory of Extreme Environmental Medicine, Ministry of Education, Chongqing, China
| | - Xiaoxu Li
- Department of High Altitude Physiology and Pathology, College of High Altitude Military Medicine, Army Medical University, Chongqing, China
- Key Laboratory of Extreme Environmental Medicine, Ministry of Education, Chongqing, China
| | - Chengzhong Yang
- Department of High Altitude Physiology and Pathology, College of High Altitude Military Medicine, Army Medical University, Chongqing, China
- Key Laboratory of Extreme Environmental Medicine, Ministry of Education, Chongqing, China
| | - Yidong Yang
- Department of High Altitude Physiology and Pathology, College of High Altitude Military Medicine, Army Medical University, Chongqing, China
- Key Laboratory of Extreme Environmental Medicine, Ministry of Education, Chongqing, China
| | - Yuqi Gao
- Key Laboratory of Extreme Environmental Medicine, Ministry of Education, Chongqing, China
- College of High Altitude Military Medicine, Army Medical University, Chongqing, China
| | - Dewei Chen
- Department of High Altitude Physiology and Pathology, College of High Altitude Military Medicine, Army Medical University, Chongqing, China
- Key Laboratory of Extreme Environmental Medicine, Ministry of Education, Chongqing, China
| | - Jian Huang
- Department of High Altitude Physiology and Pathology, College of High Altitude Military Medicine, Army Medical University, Chongqing, China
- Key Laboratory of Extreme Environmental Medicine, Ministry of Education, Chongqing, China
| |
Collapse
|
3
|
Abdolmaleky HM, Nohesara S, Zhou JR, Thiagalingam S. Epigenetics in evolution and adaptation to environmental challenges: pathways for disease prevention and treatment. Epigenomics 2025; 17:317-333. [PMID: 39948759 PMCID: PMC11970782 DOI: 10.1080/17501911.2025.2464529] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Accepted: 02/04/2025] [Indexed: 04/02/2025] Open
Abstract
Adaptation to challenging environmental conditions is crucial for the survival/fitness of all organisms. Alongside genetic mutations that provide adaptive potential during environmental challenges, epigenetic modifications offer dynamic, reversible, and rapid mechanisms for regulating gene expression in response to environmental changes in both evolution and daily life, without altering DNA sequences or relying on accidental favorable mutations. The widespread conservation of diverse epigenetic mechanisms - like DNA methylation, histone modifications, and RNA interference across diverse species, including plants - underscores their significance in evolutionary biology. Remarkably, environmentally induced epigenetic alterations are passed to daughter cells and inherited transgenerationally through germline cells, shaping offspring phenotypes while preserving adaptive epigenetic memory. Throughout anthropoid evolution, epigenetic modifications have played crucial roles in: i) suppressing transposable elements and viral genomes intruding into the host genome; ii) inactivating one of the X chromosomes in female cells to balance gene dosage; iii) genetic imprinting to ensure expression from one parental allele; iv) regulating functional alleles to compensate for dysfunctional ones; and v) modulating the epigenome and transcriptome in response to influence from the gut microbiome among other functions. Understanding the interplay between environmental factors and epigenetic processes may provide valuable insights into developmental plasticity, evolutionary dynamics, and disease susceptibility.
Collapse
Affiliation(s)
- Hamid Mostafavi Abdolmaleky
- Department of Medicine (Biomedical Genetics), Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- Department of Medicine, Division of Gastroenterology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Shabnam Nohesara
- Department of Medicine (Biomedical Genetics), Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Jin-Rong Zhou
- Nutrition/Metabolism Laboratory, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Sam Thiagalingam
- Department of Medicine (Biomedical Genetics), Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- Department of Pathology & Laboratory Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| |
Collapse
|
4
|
Wang Z, Gomchok D, Ye Y, Wen Y, Wuren T. Platelet Reduction in Rats Exposed to Chronic Hypoxia Is Associated with Interaction of Glycoprotein Ib Alpha von Willebrand Factor. Hamostaseologie 2025. [PMID: 40154511 DOI: 10.1055/a-2462-6667] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/01/2025] Open
Abstract
Chronic high-altitude hypoxia is associated with reduced platelet count, but it is unclear whether the decrease in platelet count is due to impaired production or increased clearance. This study examines how hypoxia affects platelet production and apoptosis and elucidates the impact of glycoprotein Ibα-von Willebrand factor interaction on platelets in rats using a hypobaric hypoxia chamber. The results showed that the number of megakaryocytes increased under hypoxia; however, the levels of differentiation and polyploidy decreased, while those of apoptosis increased. Platelet production did not reduce according to the reticulated platelet percentage, while platelet apoptosis enhanced; these results suggest that increased platelet clearance was the main reason behind platelet reduction. Our previous microarray results indicated that glycoprotein Ibα (GPIbα) expression increased under hypoxia, which was a protein involved in platelet clearance; therefore, we examined the interaction of platelet GPIbα with the von Willebrand factor (vWF) both in vivo and in vitro to explore the effect of this process on platelets and whether it is related to platelet apoptosis. Under hypoxia, the stronger interaction between GPIbα and vWF promoted platelet apoptosis; inhibiting this interaction reduced platelet apoptosis and increased platelet counts. Platelet reduction is associated with apoptosis induced by the interaction between GPIbα and vWF.
Collapse
Affiliation(s)
- Zhuoya Wang
- School of Medicine, Research Center for High Altitude Medicine, Qinghai University, Xining, China
- Geriatric department, Qinghai University Affiliated Hospital, Xining, China
- Key Laboratory of Application and Foundation for High Altitude Medicine Research, Qinghai Province, Xining, China
| | - Drolma Gomchok
- School of Medicine, Research Center for High Altitude Medicine, Qinghai University, Xining, China
- Key Laboratory of Application and Foundation for High Altitude Medicine Research, Qinghai Province, Xining, China
| | - Yi Ye
- School of Medicine, Research Center for High Altitude Medicine, Qinghai University, Xining, China
- Key Laboratory of Application and Foundation for High Altitude Medicine Research, Qinghai Province, Xining, China
| | - Yi Wen
- Department of General Surgery & Pancreatic Injury and Repair Key Laboratory of Sichuan Province, The General Hospital of Western Theater Command (Chengdu Military General Hospital), Chengdu, China
| | - Tana Wuren
- School of Medicine, Research Center for High Altitude Medicine, Qinghai University, Xining, China
- Key Laboratory of Application and Foundation for High Altitude Medicine Research, Qinghai Province, Xining, China
| |
Collapse
|
5
|
Tang S, Zhou W, Chen L, Yan H, Chen L, Luo F. High altitude polycythemia and its maladaptive mechanisms: an updated review. Front Med (Lausanne) 2024; 11:1448654. [PMID: 39257892 PMCID: PMC11383785 DOI: 10.3389/fmed.2024.1448654] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Accepted: 08/12/2024] [Indexed: 09/12/2024] Open
Abstract
High altitude polycythemia is a maladaptation of highlanders exposed to hypoxic environment, leading to high blood viscosity and severe cardiorespiratory dysfunction. Prolonged hypoxia causes respiratory depression and severe hypoxemia, and further mediates changes in genetic and molecular mechanisms that regulate erythropoiesis and apoptosis, ultimately resulting in excessive erythrocytosis (EE). This updated review investigated the maladaptive mechanisms of EE, including respiratory chemoreceptor passivation, sleep-related breathing disorders, sex hormones, iron metabolism, and hypoxia-related factors and pathways.
Collapse
Affiliation(s)
- Shijie Tang
- Department of High Altitude Medicine, Center for High Altitude Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- High Altitude Medicine Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Department of Pulmonary and Critical Care Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Wenwen Zhou
- Department of High Altitude Medicine, Center for High Altitude Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- High Altitude Medicine Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Ling Chen
- Department of High Altitude Medicine, Center for High Altitude Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- High Altitude Medicine Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Hui Yan
- Department of High Altitude Medicine, Center for High Altitude Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- High Altitude Medicine Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Lei Chen
- Department of High Altitude Medicine, Center for High Altitude Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- High Altitude Medicine Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Department of Pulmonary and Critical Care Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Fengming Luo
- Department of High Altitude Medicine, Center for High Altitude Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- High Altitude Medicine Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Department of Pulmonary and Critical Care Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
6
|
Hajiaqaei M, Ranjbaran M, Kadkhodaee M, Shafie A, Abdi A, Lorian K, Kianian F, Seifi B. Hydrogen sulfide upregulates hypoxia inducible factors and erythropoietin production in chronic kidney disease induced by 5/6 nephrectomized rats. Mol Biol Rep 2024; 51:916. [PMID: 39158746 DOI: 10.1007/s11033-024-09824-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Accepted: 07/23/2024] [Indexed: 08/20/2024]
Abstract
INTRODUCTION In end stage renal disease )ESRD(, reduced EPO production resulted in decreased oxygen diffusion that cause Hypoxia-inducible factors (HIFs) stabilization. The mechanism of beneficial effects of H2S in chronic kidney disease (CKD) is the aim of the present study to examine the effects of the H2S donor sodium hydrosulfide (NaHS) on renal function parameters, oxidative stress indices and expression levels of HIF-2α gene and erythropoietin protein in 5/6 nephrectomy-induced chronic renal failure in rats. METHODS AND MATERIALS Male rats were assigned into 3 groups (n = 8): Sham, CKD and NaHS groups. In the CKD group, 5/6 nephrectomy was performed. In the sham group, rats were anesthetized but 5/6 nephrectomy was not induced. In the NaHS group, 30 µmol/L of NaHS in drinking water for 8 weeks was adminstrated 4 weeks after 5/6 nephrectomy induction. At the end of the 12 week, blood and renal tissues were taken to evaluate renal function parameters, oxidative stress indices and expression levels of HIF-2α gene and erythropoietin protein. RESULTS The induction of 5/6 nephrectomy significantly caused renal dysfunction, oxidative stress, increased HIF-2α gene expression and decreased erythropoietin levels in renal tissue samples. NaHS administration resulted in a marked improvement in renal function and oxidative stress indicators, a marked reduction in HIF-2α gene expression as well as an increase in erythropoietin protein levels in comparison with the CKD group. CONCLUSION In this study, regional hypoxia and oxidative stress in CKD, may cause the stabilization of the HIFs complexes, although erythropoietin synthesis was not increased due to destructive effects of CKD on the kidney tissues. Administration of NaHS caused up-regulating HIF-erythropoietin signaling pathway.
Collapse
Affiliation(s)
- Mahdi Hajiaqaei
- Department of Physiology, School of Medicine, Tehran University of Medical Sciences, Poorsina Ave, Tehran, Iran
| | - Mina Ranjbaran
- Department of Physiology, School of Medicine, Tehran University of Medical Sciences, Poorsina Ave, Tehran, Iran
| | - Mehri Kadkhodaee
- Department of Physiology, School of Medicine, Tehran University of Medical Sciences, Poorsina Ave, Tehran, Iran
| | - Anahid Shafie
- Department of Physiology, School of Medicine, Tehran University of Medical Sciences, Poorsina Ave, Tehran, Iran
| | - Arash Abdi
- Department of Physiology, School of Medicine, Tehran University of Medical Sciences, Poorsina Ave, Tehran, Iran
| | - Keivan Lorian
- Research and Clinical Center for Infertility, Yazd Rreproductive Sciences Institute, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Farzaneh Kianian
- Department of Physiology, School of Medicine, Tehran University of Medical Sciences, Poorsina Ave, Tehran, Iran
| | - Behjat Seifi
- Department of Physiology, School of Medicine, Tehran University of Medical Sciences, Poorsina Ave, Tehran, Iran.
| |
Collapse
|
7
|
Zheng GP, Nian W, Shi XF, Xie YB. [Progress in multiomics research on high altitude polycythemia]. ZHONGHUA XUE YE XUE ZA ZHI = ZHONGHUA XUEYEXUE ZAZHI 2024; 45:795-800. [PMID: 39307731 PMCID: PMC11535562 DOI: 10.3760/cma.j.cn121090-20240318-00100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Indexed: 12/06/2024]
Abstract
Chronic mountain sickness (CMS) or Monge syndrome is a disease that is prevalent at altitude above 2 500 meters. High altitude polycythemia (HAPC) is one subtype of CMS. EPAS1 and EGNL1 are the most critical high-altitude adaptation genes in the genome of the Tibetan population. The HIF-PHD-VHL system plays an important role in the pathogenesis of HAPC. The protease encoded by the SENP1 gene regulates hypoxia related transcription factors such as HIF and GATA to affect the expression of EPO or EPOR involved in red blood cell generation. With the development of genetic testing and omics technology, new progress in the fields of metabolomics, proteomics and metabolomics has been made in the pathogenesis of HAPC. The above new research results provide a preliminary basis for bone marrow hematoecology and hematopoietic regulation of HAPC. The diagnostic criteria for CMS have certain limitations, especially in patients with excessive erythrocytosis who should undergo genetic testing recommended for congenital and polycythemia vera. This article provides a review of the latest research on HAPC in various omics techniques, hematopoietic regulation and diagnostic processes which is more conducive to understand the pathogenesis. The clinical diagnosis of excessive erythrocytosis emphasizes the importance of genetic testing.
Collapse
Affiliation(s)
- G P Zheng
- Department of Hematology, Qinghai Provincial People's Hospital, Xining 810007, China
| | - W Nian
- Department of Geriatrics, Qinghai Provincial People's Hospital, Xining 810007, China
| | - X F Shi
- Department of Respiratory and Critical Care Medicine, Qinghai Provincial People's Hospital, Xining 810007, China
| | - Y B Xie
- Department of Hematology, Qinghai Provincial People's Hospital, Xining 810007, China
| |
Collapse
|
8
|
Kuang H, Yang L, Li Z, Wang J, Zheng K, Mei J, Sun H, Huang Y, Yang C, Luo W. DNA methyltransferase 3A induces the occurrence of oral submucous fibrosis by promoting the methylation of the von Hippel-Lindau. Oral Dis 2024; 30:2325-2336. [PMID: 37743610 DOI: 10.1111/odi.14725] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 07/25/2023] [Accepted: 08/15/2023] [Indexed: 09/26/2023]
Abstract
BACKGROUND Oral submucous fibrosis (OSF) is associated with malignant disorders. DNA methyltransferase 3A (DNMT3A) is a DNA methylesterase reported to be upregulated in multiple organs and shown to inhibit fibrosis. However, the detailed effect of DNMT3A on OSF remains unclear. METHODS To mimic OSF in vitro, oral fibroblasts were exposed to arecoline and molecular biological experiments were performed to detect the function of DNMT3A in OSF. RESULTS We found that von Hippel-Lindau (VHL) was downregulated and highly methylated in OSF. Arecoline remarkably increased the viability, invasiveness, and migration of oral fibroblasts, but upregulation of VHL partially reversed these effects. DNMT3A induces DNA hypermethylation in the VHL promoter, and VHL markedly inhibits the level of tenascin-C (TNC) by inducing the ubiquitination of TNC. TNC reversed the inhibitory effect of VHL upregulation on the differentiation of oral fibroblasts into myofibroblasts. CONCLUSION DNMT3A induces OSF by promoting methylation of the VHL promoter. Hence, our study provides novel insights into the discovery of novel strategies that can be employed against OSF.
Collapse
Affiliation(s)
- Huifang Kuang
- Department of Stomatology, The First Affiliated Hospital of Hainan Medical University, Haikou, China
- School of Stomatology, Hainan Medical University, Haikou, China
| | - Liyan Yang
- Department of Stomatology, The First Affiliated Hospital of Hainan Medical University, Haikou, China
- School of Stomatology, Hainan Medical University, Haikou, China
| | - Zhixin Li
- Department of Stomatology, The First Affiliated Hospital of Hainan Medical University, Haikou, China
- School of Stomatology, Hainan Medical University, Haikou, China
| | - Jinrong Wang
- Department of Stomatology, The First Affiliated Hospital of Hainan Medical University, Haikou, China
- School of Stomatology, Hainan Medical University, Haikou, China
| | - Kaiyue Zheng
- Department of Stomatology, The First Affiliated Hospital of Hainan Medical University, Haikou, China
- School of Stomatology, Hainan Medical University, Haikou, China
| | - Jie Mei
- Department of Stomatology, The First Affiliated Hospital of Hainan Medical University, Haikou, China
- School of Stomatology, Hainan Medical University, Haikou, China
| | - Honglan Sun
- Department of Stomatology, The First Affiliated Hospital of Hainan Medical University, Haikou, China
- School of Stomatology, Hainan Medical University, Haikou, China
| | - Yuqi Huang
- Department of Stomatology, The First Affiliated Hospital of Hainan Medical University, Haikou, China
- School of Stomatology, Hainan Medical University, Haikou, China
| | - Chao Yang
- Department of Stomatology, The People's Hospital of Longhua, Shenzhen, China
- Research and Development Department, Shenzhen Uni-Medica Technology Co., Ltd, Shenzhen, China
| | - Wen Luo
- Department of Stomatology, The First Affiliated Hospital of Hainan Medical University, Haikou, China
- School of Stomatology, Hainan Medical University, Haikou, China
| |
Collapse
|
9
|
Wang S, Ma J, Qiu H, Liu S, Zhang S, Liu H, Zhang P, Ge RL, Li G, Cui S. Plasma exosomal microRNA expression profiles in patients with high-altitude polycythemia. Blood Cells Mol Dis 2023; 98:102707. [DOI: 10.1016/j.bcmd.2022.102707] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2022] [Revised: 10/19/2022] [Accepted: 10/26/2022] [Indexed: 11/06/2022]
|
10
|
Liu X, Zhang H, Yan J, Li X, Li J, Hu J, Shang X, Yang H. Deciphering the Efficacy and Mechanism of Astragalus membranaceus on High Altitude Polycythemia by Integrating Network Pharmacology and In Vivo Experiments. Nutrients 2022; 14:4968. [PMID: 36500998 PMCID: PMC9740273 DOI: 10.3390/nu14234968] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Revised: 11/11/2022] [Accepted: 11/17/2022] [Indexed: 11/25/2022] Open
Abstract
Hypoxic exposure makes plateau migrators susceptible to high altitude polycythemia (HAPC). Astragalus membranaceus (AM) is an edible and medicinal plant with remarkable immunomodulatory activities. The purpose of this study was to discover if AM could be a candidate for the prevention of HAPC and its mechanism. Here, network pharmacology was applied to screen active compounds, key targets, and enriched pathways of AM in the treatment of HAPC. Molecular docking evaluated the affinity between compounds and core targets. Subsequently, the mechanisms of AM were further verified using the hypoxia exposure-induced mice model of HAPC. The network pharmacology analysis and molecular docking results identified 14 core targets of AM on HAPC, which were predominantly mainly enriched in the HIF-1 pathway. In the HAPC animal models, we found that AM inhibited the differentiation of hematopoietic stem cells into the erythroid lineage. It also suppressed the production of erythrocytes and hemoglobin in peripheral blood by reducing the expression of HIF-1α, EPO, VEGFA, and Gata-1 mRNA. Furthermore, AM downregulated the expression of IL-6, TNF-α, and IFN-γ mRNA, thereby alleviating organ inflammation. In conclusion, AM supplementation alleviates hypoxia-induced HAPC in mice, and TNF-α, AKT1, HIF-1α, VEGFA, IL-6, and IL-1B may be the key targets.
Collapse
Affiliation(s)
- Xiru Liu
- School of Life Sciences, Northwestern Polytechnical University, Xi’an 710072, China
- Research Center of Special Environmental Biomechanics & Medical Engineering, Northwestern Polytechnical University, Xi’an 710072, China
| | - Hao Zhang
- School of Life Sciences, Northwestern Polytechnical University, Xi’an 710072, China
- Research Center of Special Environmental Biomechanics & Medical Engineering, Northwestern Polytechnical University, Xi’an 710072, China
| | - Jinxiao Yan
- School of Life Sciences, Northwestern Polytechnical University, Xi’an 710072, China
- Research Center of Special Environmental Biomechanics & Medical Engineering, Northwestern Polytechnical University, Xi’an 710072, China
| | - Xiang Li
- School of Life Sciences, Northwestern Polytechnical University, Xi’an 710072, China
- Research Center of Special Environmental Biomechanics & Medical Engineering, Northwestern Polytechnical University, Xi’an 710072, China
| | - Jie Li
- General Station for Drug & Instrument Supervision and Control, Joint Logistics Support Force, PLA, Dalian 116041, China
| | - Jialu Hu
- School of Computer Science, Northwestern Polytechnical University, Xi’an 710072, China
| | - Xuequn Shang
- School of Computer Science, Northwestern Polytechnical University, Xi’an 710072, China
| | - Hui Yang
- School of Life Sciences, Northwestern Polytechnical University, Xi’an 710072, China
- Research Center of Special Environmental Biomechanics & Medical Engineering, Northwestern Polytechnical University, Xi’an 710072, China
| |
Collapse
|
11
|
Chen P, Liu Y, Liu W, Wang Y, Liu Z, Rong M. Impact of High-Altitude Hypoxia on Bone Defect Repair: A Review of Molecular Mechanisms and Therapeutic Implications. Front Med (Lausanne) 2022; 9:842800. [PMID: 35620712 PMCID: PMC9127390 DOI: 10.3389/fmed.2022.842800] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Accepted: 04/15/2022] [Indexed: 11/23/2022] Open
Abstract
Reaching areas at altitudes over 2,500–3,000 m above sea level has become increasingly common due to commerce, military deployment, tourism, and entertainment. The high-altitude environment exerts systemic effects on humans that represent a series of compensatory reactions and affects the activity of bone cells. Cellular structures closely related to oxygen-sensing produce corresponding functional changes, resulting in decreased tissue vascularization, declined repair ability of bone defects, and longer healing time. This review focuses on the impact of high-altitude hypoxia on bone defect repair and discusses the possible mechanisms related to ion channels, reactive oxygen species production, mitochondrial function, autophagy, and epigenetics. Based on the key pathogenic mechanisms, potential therapeutic strategies have also been suggested. This review contributes novel insights into the mechanisms of abnormal bone defect repair in hypoxic environments, along with therapeutic applications. We aim to provide a foundation for future targeted, personalized, and precise bone regeneration therapies according to the adaptation of patients to high altitudes.
Collapse
Affiliation(s)
- Pei Chen
- Department of Periodontology and Implantology, Stomatological Hospital, Southern Medical University, Guangzhou, China
| | - Yushan Liu
- Department of Periodontology and Implantology, Stomatological Hospital, Southern Medical University, Guangzhou, China
| | - Wenjing Liu
- Department of Prosthodontics, Stomatological Hospital, Southern Medical University, Guangzhou, China
| | - Yarong Wang
- Department of Periodontology and Implantology, Stomatological Hospital, Southern Medical University, Guangzhou, China
| | - Ziyi Liu
- Department of Periodontology and Implantology, Stomatological Hospital, Southern Medical University, Guangzhou, China
| | - Mingdeng Rong
- Department of Periodontology and Implantology, Stomatological Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
12
|
Padmasekar M, Savai R, Seeger W, Pullamsetti SS. Exposomes to Exosomes: Exosomes as Tools to Study Epigenetic Adaptive Mechanisms in High-Altitude Humans. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2021; 18:8280. [PMID: 34444030 PMCID: PMC8392481 DOI: 10.3390/ijerph18168280] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 07/30/2021] [Accepted: 07/31/2021] [Indexed: 12/29/2022]
Abstract
Humans on earth inhabit a wide range of environmental conditions and some environments are more challenging for human survival than others. However, many living beings, including humans, have developed adaptive mechanisms to live in such inhospitable, harsh environments. Among different difficult environments, high-altitude living is especially demanding because of diminished partial pressure of oxygen and resulting chronic hypobaric hypoxia. This results in poor blood oxygenation and reduces aerobic oxidative respiration in the mitochondria, leading to increased reactive oxygen species generation and activation of hypoxia-inducible gene expression. Genetic mechanisms in the adaptation to high altitude is well-studied, but there are only limited studies regarding the role of epigenetic mechanisms. The purpose of this review is to understand the epigenetic mechanisms behind high-altitude adaptive and maladaptive phenotypes. Hypobaric hypoxia is a form of cellular hypoxia, which is similar to the one suffered by critically-ill hypoxemia patients. Thus, understanding the adaptive epigenetic signals operating in in high-altitude adjusted indigenous populations may help in therapeutically modulating signaling pathways in hypoxemia patients by copying the most successful epigenotype. In addition, we have summarized the current information about exosomes in hypoxia research and prospects to use them as diagnostic tools to study the epigenome of high-altitude adapted healthy or maladapted individuals.
Collapse
Affiliation(s)
- Manju Padmasekar
- Max-Planck Institute for Heart and Lung Research, Member of the German Center for Lung Research (DZL), Member of the Cardio-Pulmonary Institute (CPI), 61231 Bad Nauheim, Germany; (M.P.); (R.S.); (W.S.)
| | - Rajkumar Savai
- Max-Planck Institute for Heart and Lung Research, Member of the German Center for Lung Research (DZL), Member of the Cardio-Pulmonary Institute (CPI), 61231 Bad Nauheim, Germany; (M.P.); (R.S.); (W.S.)
- Institute for Lung Health (ILH), Justus Liebig University, 35392 Giessen, Germany
- Department of Internal Medicine, Justus-Liebig University Giessen, Member of the DZL, Member of CPI, 35392 Giessen, Germany
- Frankfurt Cancer Institute (FCI), Goethe University, 60438 Frankfurt am Main, Germany
| | - Werner Seeger
- Max-Planck Institute for Heart and Lung Research, Member of the German Center for Lung Research (DZL), Member of the Cardio-Pulmonary Institute (CPI), 61231 Bad Nauheim, Germany; (M.P.); (R.S.); (W.S.)
- Institute for Lung Health (ILH), Justus Liebig University, 35392 Giessen, Germany
- Department of Internal Medicine, Justus-Liebig University Giessen, Member of the DZL, Member of CPI, 35392 Giessen, Germany
| | - Soni Savai Pullamsetti
- Max-Planck Institute for Heart and Lung Research, Member of the German Center for Lung Research (DZL), Member of the Cardio-Pulmonary Institute (CPI), 61231 Bad Nauheim, Germany; (M.P.); (R.S.); (W.S.)
- Institute for Lung Health (ILH), Justus Liebig University, 35392 Giessen, Germany
- Department of Internal Medicine, Justus-Liebig University Giessen, Member of the DZL, Member of CPI, 35392 Giessen, Germany
| |
Collapse
|
13
|
Tomc J, Debeljak N. Molecular Pathways Involved in the Development of Congenital Erythrocytosis. Genes (Basel) 2021; 12:1150. [PMID: 34440324 PMCID: PMC8391844 DOI: 10.3390/genes12081150] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Revised: 07/24/2021] [Accepted: 07/26/2021] [Indexed: 01/08/2023] Open
Abstract
Patients with idiopathic erythrocytosis are directed to targeted genetic testing including nine genes involved in oxygen sensing pathway in kidneys, erythropoietin signal transduction in pre-erythrocytes and hemoglobin-oxygen affinity regulation in mature erythrocytes. However, in more than 60% of cases the genetic cause remains undiagnosed, suggesting that other genes and mechanisms must be involved in the disease development. This review aims to explore additional molecular mechanisms in recognized erythrocytosis pathways and propose new pathways associated with this rare hematological disorder. For this purpose, a comprehensive review of the literature was performed and different in silico tools were used. We identified genes involved in several mechanisms and molecular pathways, including mRNA transcriptional regulation, post-translational modifications, membrane transport, regulation of signal transduction, glucose metabolism and iron homeostasis, which have the potential to influence the main erythrocytosis-associated pathways. We provide valuable theoretical information for deeper insight into possible mechanisms of disease development. This information can be also helpful to improve the current diagnostic solutions for patients with idiopathic erythrocytosis.
Collapse
Affiliation(s)
| | - Nataša Debeljak
- Medical Centre for Molecular Biology, Institute of Biochemistry and Molecular Genetics, Faculty of Medicine, University of Ljubljana, SI-1000 Ljubljana, Slovenia;
| |
Collapse
|
14
|
Tomc J, Debeljak N. Molecular Insights into the Oxygen-Sensing Pathway and Erythropoietin Expression Regulation in Erythropoiesis. Int J Mol Sci 2021; 22:ijms22137074. [PMID: 34209205 PMCID: PMC8269393 DOI: 10.3390/ijms22137074] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 06/25/2021] [Accepted: 06/26/2021] [Indexed: 02/07/2023] Open
Abstract
Erythropoiesis is regulated by several factors, including the oxygen-sensing pathway as the main regulator of erythropoietin (EPO) synthesis in the kidney. The release of EPO from the kidney and its binding to the EPO receptor (EPOR) on erythrocyte progenitor cells in the bone marrow results in increased erythropoiesis. Any imbalance in these homeostatic mechanisms can lead to dysregulated erythropoiesis and hematological disorders. For example, mutations in genes encoding key players of oxygen-sensing pathway and regulation of EPO production (HIF-EPO pathway), namely VHL, EGLN, EPAS1 and EPO, are well known causative factors that contribute to the development of erythrocytosis. We aimed to investigate additional molecular mechanisms involved in the HIF-EPO pathway that correlate with erythropoiesis. To this end, we conducted an extensive literature search and used several in silico tools. We identified genes encoding transcription factors and proteins that control transcriptional activation or repression; genes encoding kinases, deacetylases, methyltransferases, conjugating enzymes, protein ligases, and proteases involved in post-translational modifications; and genes encoding nuclear transport receptors that regulate nuclear transport. All these genes may modulate the stability or activity of HIF2α and its partners in the HIF-EPO pathway, thus affecting EPO synthesis. The theoretical information we provide in this work can be a valuable tool for a better understanding of one of the most important regulatory pathways in the process of erythropoiesis. This knowledge is necessary to discover the causative factors that may contribute to the development of hematological diseases and improve current diagnostic and treatment solutions in this regard.
Collapse
Affiliation(s)
- Jana Tomc
- Medical Centre for Molecular Biology, Institute of Biochemistry and Molecular Genetics, Faculty of Medicine, University of Ljubljana, 1000 Ljubljana, Slovenia;
- Centre for Functional Genomics and Bio-Chips, Institute of Biochemistry and Molecular Genetics, Faculty of Medicine, University of Ljubljana, 1000 Ljubljana, Slovenia
| | - Nataša Debeljak
- Medical Centre for Molecular Biology, Institute of Biochemistry and Molecular Genetics, Faculty of Medicine, University of Ljubljana, 1000 Ljubljana, Slovenia;
- Correspondence: ; Tel.: +386-1-543-7645
| |
Collapse
|
15
|
Chen K, Li N, Fan F, Geng Z, Zhao K, Wang J, Zhang Y, Tang C, Wang X, Meng X. Tibetan Medicine Duoxuekang Capsule Ameliorates High-Altitude Polycythemia Accompanied by Brain Injury. Front Pharmacol 2021; 12:680636. [PMID: 34045970 PMCID: PMC8144525 DOI: 10.3389/fphar.2021.680636] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Accepted: 04/22/2021] [Indexed: 12/15/2022] Open
Abstract
Objective: Duoxuekang (DXK) capsule is an empirical prescription for Tibetan medicine in the treatment of hypobaric hypoxia (HH)-induced brain injury in the plateau. This study aimed to investigate the protective effects and underlying molecular mechanisms of DXK on HH-induced brain injury. Methods: UPLC–Q-TOF/MS was performed for chemical composition analysis of DXK. The anti-hypoxia and anti-fatigue effects of DXK were evaluated by the normobaric hypoxia test, sodium nitrite toxicosis test, and weight-loaded swimming test in mice. Simultaneously, SD rats were used for the chronic hypobaric hypoxia (CHH) test. RBC, HGB, HCT, and the whole blood viscosity were evaluated. The activities of SOD and MDA in the brain, and EPO and LDH levels in the kidney were detected using ELISA. H&E staining was employed to observe the pathological morphology in the hippocampus and cortex of rats. Furthermore, immunofluorescence and Western blot were carried out to detect the protein expressions of Mapk10, RASGRF1, RASA3, Ras, and IGF-IR in the brain of rats. Besides, BALB/c mice were used for acute hypobaric hypoxia (AHH) test, and Western blot was employed to detect the protein expression of p-ERK/ERK, p-JNK/JNK, and p-p38/p38 in the cerebral cortex of mice. Results: 23 different chemical compositions of DXK were identified by UPLC–Q-TOF/MS. The anti-hypoxia test verified that DXK can prolong the survival time of mice. The anti-fatigue test confirmed that DXK can prolong the swimming time of mice, decrease the level of LDH, and increase the hepatic glycogen level. Synchronously, DXK can decrease the levels of RBC, HGB, HCT, and the whole blood viscosity under the CHH condition. Besides, DXK can ameliorate CHH-induced brain injury, decrease the levels of EPO and LDH in the kidney, reduce MDA, and increase SOD in the hippocampus. Furthermore, DXK can converse HH-induced marked increase of Mapk10, RASGRF1, and RASA3, and decrease of Ras and IGF-IR. In addition, DXK can suppress the ratio of p-ERK/ERK, p-JNK/JNK, and p-p38/p38 under the HH condition. Conclusion: Together, the cerebral protection elicited by DXK was due to the decrease of hematological index, suppressing EPO, by affecting the MAPK signaling pathway in oxidative damage, and regulating the RAS signaling pathway.
Collapse
Affiliation(s)
- Ke Chen
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Ning Li
- School of Ethnic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Fangfang Fan
- School of Ethnic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - ZangJia Geng
- School of Pharmacy, Southwest Minzu University, Chengdu, China
| | - Kehui Zhao
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jing Wang
- School of Management, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yi Zhang
- School of Ethnic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China.,Ethnic Medicine Academic Heritage Innovation Research Center, Chengdu University of Traditional Chinese Medicine, Chengdu, China.,NMPA Key Laboratory for Quality Evaluation of Traditional Chinese Medicine (Traditional Chinese Patent Medicine), Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Ce Tang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Innovative Institute of Chinese Medicine and Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xiaobo Wang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Innovative Institute of Chinese Medicine and Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xianli Meng
- State Key Laboratory of Southwestern Chinese Medicine Resources, Innovative Institute of Chinese Medicine and Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
16
|
Wu H, Chu Y, Sun S, Li G, Xu S, Zhang X, Jiang Y, Gao S, Wang Q, Zhang J, Pang D. Hypoxia-Mediated Complement 1q Binding Protein Regulates Metastasis and Chemoresistance in Triple-Negative Breast Cancer and Modulates the PKC-NF-κB-VCAM-1 Signaling Pathway. Front Cell Dev Biol 2021; 9:607142. [PMID: 33708767 PMCID: PMC7940382 DOI: 10.3389/fcell.2021.607142] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Accepted: 01/29/2021] [Indexed: 12/24/2022] Open
Abstract
Objectives Complement 1q binding protein (C1QBP/HABP1/p32/gC1qR) has been found to be overexpressed in triple-negative breast cancer (TNBC). However, the underlying mechanisms of high C1QBP expression and its role in TNBC remain largely unclear. Hypoxia is a tumor-associated microenvironment that promotes metastasis and paclitaxel (PTX) chemoresistance in tumor cells. In this study, we aimed to assess C1QBP expression and explore its role in hypoxia-related metastasis and chemoresistance in TNBC. Materials and Methods RNA-sequencing of TNBC cells under hypoxia was performed to identify C1QBP. The effect of hypoxia inducible factor 1 subunit alpha (HIF-1α) on C1QBP expression was investigated using chromatin immunoprecipitation (ChIP) assay. The role of C1QBP in mediating metastasis, chemoresistance to PTX, and regulation of metastasis-linked vascular cell adhesion molecule 1 (VCAM-1) expression were studied using in vitro and in vivo experiments. Clinical tissue microarrays were used to verify the correlation of C1QBP with the expression of HIF-1α, VCAM-1, and RELA proto-oncogene nuclear factor-kappa B subunit (P65). Results We found that hypoxia-induced HIF-1α upregulated C1QBP. The inhibition of C1QBP notably blocked metastasis of TNBC cells and increased their sensitivity to PTX under hypoxic conditions. Depletion of C1QBP decreased VCAM-1 expression by reducing the amount of P65 in the nucleus and suppressed the activation of hypoxia-induced protein kinase C-nuclear factor-kappa B (PKC-NF-κB) signaling.immunohistochemistry (IHC) staining of the tissue microarray showed positive correlations between the C1QBP level and those of HIF-1α, P65, and VCAM-1. Conclusion Targeting C1QBP along with PTX treatment might be a potential treatment for TNBC patients.
Collapse
Affiliation(s)
- Hao Wu
- Sino-Russian Medical Research Center, Harbin Medical University Cancer Hospital, Harbin, China.,Translational Medicine Research and Cooperation Center of Northern China, Harbin Medical University, Harbin, China.,Heilongjiang Academy of Medical Sciences, Harbin, China
| | - Yijun Chu
- Translational Medicine Research and Cooperation Center of Northern China, Harbin Medical University, Harbin, China.,Heilongjiang Academy of Medical Sciences, Harbin, China.,Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin, China
| | - Shanshan Sun
- Translational Medicine Research and Cooperation Center of Northern China, Harbin Medical University, Harbin, China.,Heilongjiang Academy of Medical Sciences, Harbin, China.,Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin, China
| | - Guozheng Li
- Translational Medicine Research and Cooperation Center of Northern China, Harbin Medical University, Harbin, China.,Heilongjiang Academy of Medical Sciences, Harbin, China.,Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin, China
| | - Shouping Xu
- Translational Medicine Research and Cooperation Center of Northern China, Harbin Medical University, Harbin, China.,Heilongjiang Academy of Medical Sciences, Harbin, China.,Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin, China
| | - Xianyu Zhang
- Translational Medicine Research and Cooperation Center of Northern China, Harbin Medical University, Harbin, China.,Heilongjiang Academy of Medical Sciences, Harbin, China.,Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin, China
| | - Yongdong Jiang
- Translational Medicine Research and Cooperation Center of Northern China, Harbin Medical University, Harbin, China.,Heilongjiang Academy of Medical Sciences, Harbin, China.,Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin, China
| | - Song Gao
- Translational Medicine Research and Cooperation Center of Northern China, Harbin Medical University, Harbin, China.,Heilongjiang Academy of Medical Sciences, Harbin, China.,Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin, China
| | - Qin Wang
- Sino-Russian Medical Research Center, Harbin Medical University Cancer Hospital, Harbin, China.,Translational Medicine Research and Cooperation Center of Northern China, Harbin Medical University, Harbin, China.,Heilongjiang Academy of Medical Sciences, Harbin, China
| | - Jian Zhang
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin, China
| | - Da Pang
- Sino-Russian Medical Research Center, Harbin Medical University Cancer Hospital, Harbin, China.,Translational Medicine Research and Cooperation Center of Northern China, Harbin Medical University, Harbin, China.,Heilongjiang Academy of Medical Sciences, Harbin, China.,Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin, China
| |
Collapse
|