1
|
Wang X, Huang R, Liu L, Wang X, Zhang X. Evaluation and preservation of fertility in patients with hematologic malignancies. Cancer Lett 2025; 616:217569. [PMID: 39983893 DOI: 10.1016/j.canlet.2025.217569] [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: 10/21/2024] [Revised: 02/14/2025] [Accepted: 02/17/2025] [Indexed: 02/23/2025]
Abstract
For patients with hematologic malignancies, novel therapeutic strategies offer the potential to achieve a complete clinical response and long-term survival. However, declining fertility has become a significant concern, impacting long-term quality of life. Conventional high-dose chemotherapy and radiotherapy are known to reduce fertility or cause sterility. Moreover, limited clinical data are available on the effects of newer therapies, such as targeted treatments and chimeric antigen receptor (CAR)-T cell therapy, on fertility. Additionally, there is no standard method for preserving fertility in these patients. Male patients can opt for sperm cryopreservation, whereas female patients may preserve fertility through embryo, oocyte, or ovarian tissue cryopreservation. However, preserving fertility in prepubescent patients remains particularly challenging. Therefore, hematologists must educate patients about the potential gonadal toxicity of cancer treatments and offer the most appropriate fertility preservation options.
Collapse
Affiliation(s)
- Xiang Wang
- Medical Center of Hematology, Xinqiao Hospital of Army Medical University, Chongqing, 400037, China; Institute of Science Innovation for Blood Ecology and Intelligent Cells, Xinqiao Hospital of Army Medical University, Chongqing, 400037, China; Chongqing Key Laboratory of Hematology and Microenvironment, Chongqing, 400037, China; State Key Laboratory of Trauma and Chemical Poisoning, Army Medical University, Chongqing, 400037, China; Jinfeng Laboratory, Chongqing, 400037, China
| | - Ruihao Huang
- Medical Center of Hematology, Xinqiao Hospital of Army Medical University, Chongqing, 400037, China; Institute of Science Innovation for Blood Ecology and Intelligent Cells, Xinqiao Hospital of Army Medical University, Chongqing, 400037, China; Chongqing Key Laboratory of Hematology and Microenvironment, Chongqing, 400037, China; State Key Laboratory of Trauma and Chemical Poisoning, Army Medical University, Chongqing, 400037, China
| | - Lei Liu
- Medical Center of Hematology, Xinqiao Hospital of Army Medical University, Chongqing, 400037, China; Institute of Science Innovation for Blood Ecology and Intelligent Cells, Xinqiao Hospital of Army Medical University, Chongqing, 400037, China; Chongqing Key Laboratory of Hematology and Microenvironment, Chongqing, 400037, China; State Key Laboratory of Trauma and Chemical Poisoning, Army Medical University, Chongqing, 400037, China; Jinfeng Laboratory, Chongqing, 400037, China
| | - Xiaoqi Wang
- Medical Center of Hematology, Xinqiao Hospital of Army Medical University, Chongqing, 400037, China; Institute of Science Innovation for Blood Ecology and Intelligent Cells, Xinqiao Hospital of Army Medical University, Chongqing, 400037, China; Chongqing Key Laboratory of Hematology and Microenvironment, Chongqing, 400037, China; State Key Laboratory of Trauma and Chemical Poisoning, Army Medical University, Chongqing, 400037, China.
| | - Xi Zhang
- Medical Center of Hematology, Xinqiao Hospital of Army Medical University, Chongqing, 400037, China; Institute of Science Innovation for Blood Ecology and Intelligent Cells, Xinqiao Hospital of Army Medical University, Chongqing, 400037, China; Chongqing Key Laboratory of Hematology and Microenvironment, Chongqing, 400037, China; State Key Laboratory of Trauma and Chemical Poisoning, Army Medical University, Chongqing, 400037, China; Jinfeng Laboratory, Chongqing, 400037, China.
| |
Collapse
|
2
|
Johnson SE, Haney CR, Spann AN, Khurram N, Obeidin F, Lee J, Zhao M. An in vivo imaging approach for simultaneously assessing tumor response and cytotoxicity-induced tissue response in chemotherapy. Apoptosis 2025:10.1007/s10495-025-02118-9. [PMID: 40281308 DOI: 10.1007/s10495-025-02118-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/10/2025] [Indexed: 04/29/2025]
Abstract
In chemotherapeutic treatments, while cancer cells are the primary target, cytotoxic side effects are an important consideration. In the current study, we applied an in vivo imaging tool for characterizing chemotherapeutic response in a preclinical setting. The study focused on simultaneously examining the tumor and tissue response as a result of treatment with bortezomib, a mainstay proteasome inhibitor for treating multiple myeloma, in a preclinical model. OPM-2 tumor-bearing SCID-beige mice were designated as control or treated with bortezomib (1 mg/kg, i.v., every 4 days) (n = 8 per group). 99mTc-duramycin SPECT/CT whole-body scans were acquired 2 days before treatment as baseline and at days 1, 3 and 5 after treatment. Radioactivity uptake in tissues and organs was determined and quantitatively compared between control and bortezomib-treated group at each of the time points. Based on the imaging data, separate groups of tumor-bearing mice (n = 3 each) were included as control and bortezomib treated and the tissues were collected on day 5 for histopathology. In vivo imaging data identified significantly elevated 99mTc-duramycin uptake in the tumor, particularly in tumoral periphery. This was accompanied with signal changes in multiple organs and tissues including the adipose tissue, major bones, abdominal regions, spleen and testes. The imaging findings were consistent with known cytotoxic side effects of bortezomib and were supported by histopathology. The outcome of the study demonstrated potential utilities of the technology by enabling timely determination of the efficacy of anticancer treatments and the effect on collateral tissues as a result of systemic cytotoxic treatment.
Collapse
Affiliation(s)
| | - Chad R Haney
- Chemistry of Life Processes, Northwestern University, Evanston, USA
| | - Alisha N Spann
- Chemistry of Life Processes, Northwestern University, Evanston, USA
| | - Nigar Khurram
- Department of Pathology, Northwestern University, Chicago, USA
| | - Farres Obeidin
- Department of Pathology, Northwestern University, Chicago, USA
| | - Jungwha Lee
- Preventive Medicine, Northwestern University, Chicago, USA
| | - Ming Zhao
- Northwestern University, 320 E. Superior St., Searle 10-519, Chicago, IL, 60611, USA.
| |
Collapse
|
3
|
Abu-Baih RH, Abu-Baih DH, Abdel-Hafez SMN, Fathy M. Activation of SIRT1/Nrf2/HO-1 and Beclin-1/AMPK/mTOR autophagy pathways by eprosartan ameliorates testicular dysfunction induced by testicular torsion in rats. Sci Rep 2024; 14:12566. [PMID: 38822026 PMCID: PMC11143266 DOI: 10.1038/s41598-024-62740-6] [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: 12/22/2023] [Accepted: 05/21/2024] [Indexed: 06/02/2024] Open
Abstract
Testicular torsion carries the ominous prospect of inducing acute scrotal distress and the perilous consequence of testicular atrophy, necessitating immediate surgical intervention to reinstate vital testicular perfusion, notwithstanding the paradoxical detrimental impact of reperfusion. Although no drugs have secured approval for this urgent circumstance, antioxidants emerge as promising candidates. This study aspires to illustrate the influence of eprosartan, an AT1R antagonist, on testicular torsion in rats. Wistar albino rats were meticulously separated into five groups, (n = 6): sham group, eprosartan group, testicular torsion-detorsion (T/D) group, and two groups of T/D treated with two oral doses of eprosartan (30 or 60 mg/kg). Serum testosterone, sperm analysis and histopathological examination were done to evaluate spermatogenesis. Oxidative stress markers were assessed. Bax, BCL-2, SIRT1, Nrf2, HO-1 besides cleaved caspase-3 testicular contents were estimated using ELISA or qRT-PCR. As autophagy markers, SQSTM-1/p62, Beclin-1, mTOR and AMPK were investigated. Our findings highlight that eprosartan effectively improved serum testosterone levels, testicular weight, and sperm count/motility/viability, while mitigating histological irregularities and sperm abnormalities induced by T/D. This recovery in testicular function was underpinned by the activation of the cytoprotective SIRT1/Nrf2/HO-1 axis, which curtailed testicular oxidative stress, indicated by lowering the MDA content and increasing GSH content. In terms of apoptosis, eprosartan effectively countered apoptotic processes by decreasing cleaved caspase-3 content, suppressing Bax and stimulating Bcl-2 gene expression. Simultaneously, it reactivated impaired autophagy by increasing Beclin-1 expression, decreasing the expression of SQSTM-1/p62 and modulate the phosphorylation of AMPK and mTOR proteins. Eprosartan hold promise for managing testicular dysfunction arising from testicular torsion exerting antioxidant, pro-autophagic and anti-apoptotic effect via the activation of SIRT1/Nrf2/HO-1 as well as Beclin-1/AMPK/mTOR pathways.
Collapse
Affiliation(s)
- Rania H Abu-Baih
- Faculty of Pharmacy, Drug Information Center, Minia University, Minia, 61519, Egypt
| | - Dalia H Abu-Baih
- Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, Deraya University, Minia, 61111, Egypt
- Deraya Center for Scientific Research, Deraya University, Minia, 61111, Egypt
| | | | - Moustafa Fathy
- Department of Biochemistry, Faculty of Pharmacy, Minia University, Minia, 61519, Egypt.
| |
Collapse
|
4
|
Shafiey SI, Ahmed KA, Abo-Saif AA, Abo-Youssef AM, Mohamed WR. Galantamine mitigates testicular injury and disturbed spermatogenesis in adjuvant arthritic rats via modulating apoptosis, inflammatory signals, and IL-6/JAK/STAT3/SOCS3 signaling. Inflammopharmacology 2024; 32:405-418. [PMID: 37429998 PMCID: PMC10907493 DOI: 10.1007/s10787-023-01268-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Accepted: 06/17/2023] [Indexed: 07/12/2023]
Abstract
Rheumatoid arthritis (RA) affects the joints and the endocrine system via persistent immune system activation. RA patients have a higher frequency of testicular dysfunction, impotence, and decreased libido. This investigation aimed to evaluate the efficacy of galantamine (GAL) on testicular injury secondary to RA. Rats were allocated into four groups: control, GAL (2 mg/kg/day, p.o), CFA (0.3 mg/kg, s.c), and CFA + GAL. Testicular injury indicators, such as testosterone level, sperm count, and gonadosomatic index, were evaluated. Inflammatory indicators, such as interleukin-6 (IL-6), p-Nuclear factor kappa B (NF-κB p65), and anti-inflammatory cytokine interleukin-10 (IL-10), were assessed. Cleaved caspase-3 expression was immunohistochemically investigated. Protein expressions of Janus kinase (JAK), signal transducers and activators of transcription (STAT3), and Suppressors of Cytokine Signaling 3 (SOCS3) were examined by Western blot analysis. Results show that serum testosterone, sperm count, and gonadosomatic index were increased significantly by GAL. Additionally, GAL significantly diminished testicular IL-6 while improved IL-10 expression relative to CFA group. Furthermore, GAL attenuated testicular histopathological abnormalities by CFA and downregulated cleaved caspase-3 and NF-κB p65 expressions. It also downregulated JAK/STAT3 cascade with SOCS3 upregulation. In conclusion, GAL has potential protective effects on testicular damage secondary to RA via counteracting testicular inflammation, apoptosis, and inhibiting IL-6/JAK/STAT3/SOCS3 signaling.
Collapse
Affiliation(s)
- Sara I Shafiey
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Nahda University, Beni-Suef, 62514, Egypt
| | - Kawkab A Ahmed
- Department of Pathology, Faculty of Veterinary Medicine, Cairo University, Giza, 12211, Egypt
| | - Ali A Abo-Saif
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Nahda University, Beni-Suef, 62514, Egypt
| | - Amira M Abo-Youssef
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Beni-Suef University, Beni-Suef, 62514, Egypt.
| | - Wafaa R Mohamed
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Beni-Suef University, Beni-Suef, 62514, Egypt.
| |
Collapse
|
5
|
Mutluay D, Tenekeci GY, Monsef YA. Bortezomib-Induced Ovarian Toxicity in Mice. Toxicol Pathol 2022; 50:381-389. [DOI: 10.1177/01926233221083527] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Cancer survivors may experience long-term adverse effects of cancer treatments such as premature ovarian failure and infertility. We aimed to investigate the potential effects and toxicity of bortezomib (BTZ) as an effective anticancer drug on ovaries, raise awareness to the negative consequences of the treatment, and help increase the quality of life after treatment. Mice were distributed into bortezomib (BTZ1, BTZ2) and saline-injected control groups (C1, C2) at a dose of 1 mg/kg twice per week for 6 weeks. We sacrificed C1, BTZ1 groups at day 1 and C2, BTZ2 groups at 4 weeks after the last injection. Ovary samples were examined using histopathological and immunohistochemical methods. Ovarian follicle impairment was detected on BTZ-treated mice and was associated with a statistically significant decreased population of primordial and antral follicles compared with control groups. In experimental groups, Caspase-3 and Ki67 expressions were increased, whereas estrogen receptor alpha (ERα) and progesterone receptor (PR) expressions were decreased in various developmental stages of follicles. BTZ specifically targets granulosa cells by inducing granulosa cell apoptosis and may have long-term effects on follicles. Bortezomib treatment may adversely affect ovarian function by accelerating ovarian reserve depletion and changing ERα and PR hormone levels that can cause fertility problems in the long term.
Collapse
Affiliation(s)
- Duygu Mutluay
- Mehmet Akif Ersoy University, Faculty of Veterinary Medicine, Burdur, Turkey
| | | | | |
Collapse
|
6
|
Mendes S, Sá R, Magalhães M, Marques F, Sousa M, Silva E. The Role of ROS as a Double-Edged Sword in (In)Fertility: The Impact of Cancer Treatment. Cancers (Basel) 2022; 14:cancers14061585. [PMID: 35326736 PMCID: PMC8946252 DOI: 10.3390/cancers14061585] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Revised: 03/17/2022] [Accepted: 03/18/2022] [Indexed: 12/22/2022] Open
Abstract
Simple Summary Tumor cells are highly resistant to oxidative stress, but beyond a certain threshold, it may lead to apoptosis/necrosis. Thus, induced loss of redox balance can be a strategy used in anticancer therapies. However, the effectiveness of drugs contrasts with unknown mechanisms involved in the loss of fertility. Considering that cancer patients’ life expectancy is increasing, it raises concerns about the unknown adverse effects. Therefore, new strategies should be pursued alongside explaining to the patients their options regarding the reproduction side effects. Abstract Tumor cells are highly resistant to oxidative stress resulting from the imbalance between high reactive oxygen species (ROS) production and insufficient antioxidant defenses. However, when intracellular levels of ROS rise beyond a certain threshold, largely above cancer cells’ capacity to reduce it, they may ultimately lead to apoptosis or necrosis. This is, in fact, one of the molecular mechanisms of anticancer drugs, as most chemotherapeutic treatments alter redox homeostasis by further elevation of intracellular ROS levels or inhibition of antioxidant pathways. In traditional chemotherapy, it is widely accepted that most therapeutic effects are due to ROS-mediated cell damage, but in targeted therapies, ROS-mediated effects are mostly unknown and data are still emerging. The increasing effectiveness of anticancer treatments has raised new challenges, especially in the field of reproduction. With cancer patients’ life expectancy increasing, many aiming to become parents will be confronted with the adverse effects of treatments. Consequently, concerns about the impact of anticancer therapies on reproductive capacity are of particular interest. In this review, we begin with a short introduction on anticancer therapies, then address ROS physiological/pathophysiological roles in both male and female reproductive systems, and finish with ROS-mediated adverse effects of anticancer treatments in reproduction.
Collapse
Affiliation(s)
- Sara Mendes
- Department of Physical Education and Sports, University Institute of Maia (ISMAI), 4475-690 Maia, Portugal;
- Research Center in Sports Sciences, Health Sciences and Human Development (CIDESD), 5001-801 Vila Real, Portugal
| | - Rosália Sá
- Laboratory of Cell Biology, Department of Microscopy, Institute of Biomedical Sciences Abel Salazar (ICBAS), University of Porto, Rua Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal; (R.S.); (M.S.)
- Unit for Multidisciplinary Research in Biomedicine (UMIB), Laboratory for Integrative and Translational Research in Population Health (ITR), University of Porto, 4099-002 Porto, Portugal;
| | - Manuel Magalhães
- Unit for Multidisciplinary Research in Biomedicine (UMIB), Laboratory for Integrative and Translational Research in Population Health (ITR), University of Porto, 4099-002 Porto, Portugal;
- Department of Oncology, University Hospital Center of Porto (CHUP), Largo do Prof. Abel Salazar, 4099-001 Porto, Portugal;
| | - Franklim Marques
- Department of Oncology, University Hospital Center of Porto (CHUP), Largo do Prof. Abel Salazar, 4099-001 Porto, Portugal;
| | - Mário Sousa
- Laboratory of Cell Biology, Department of Microscopy, Institute of Biomedical Sciences Abel Salazar (ICBAS), University of Porto, Rua Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal; (R.S.); (M.S.)
- Unit for Multidisciplinary Research in Biomedicine (UMIB), Laboratory for Integrative and Translational Research in Population Health (ITR), University of Porto, 4099-002 Porto, Portugal;
| | - Elisabete Silva
- Laboratory of General Physiology, Department of Immuno-Physiology and Pharmacology, Institute of Biomedical Sciences Abel Salazar (ICBAS), University of Porto, Rua Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal
- Institute for Molecular and Cell Biology (IBMC), Institute for Research & Innovation in Health (I3S), University of Porto, Rua Alfredo Allen, 4200-135 Porto, Portugal
- Correspondence:
| |
Collapse
|
7
|
Fraser B, Peters AE, Sutherland JM, Liang M, Rebourcet D, Nixon B, Aitken RJ. Biocompatible Nanomaterials as an Emerging Technology in Reproductive Health; a Focus on the Male. Front Physiol 2021; 12:753686. [PMID: 34858208 PMCID: PMC8632065 DOI: 10.3389/fphys.2021.753686] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Accepted: 10/06/2021] [Indexed: 12/24/2022] Open
Abstract
A growing body of research has confirmed that nanoparticle (NP) systems can enhance delivery of therapeutic and imaging agents as well as prevent potentially damaging systemic exposure to these agents by modifying the kinetics of their release. With a wide choice of NP materials possessing different properties and surface modification options with unique targeting agents, bespoke nanosystems have been developed for applications varying from cancer therapeutics and genetic modification to cell imaging. Although there remain many challenges for the clinical application of nanoparticles, including toxicity within the reproductive system, some of these may be overcome with the recent development of biodegradable nanoparticles that offer increased biocompatibility. In recognition of this potential, this review seeks to present recent NP research with a focus on the exciting possibilities posed by the application of biocompatible nanomaterials within the fields of male reproductive medicine, health, and research.
Collapse
Affiliation(s)
- Barbara Fraser
- Priority Research Centre for Reproductive Science, University of Newcastle, Callaghan, NSW, Australia.,Pregnancy and Reproduction Program, Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
| | - Alexandra E Peters
- Pregnancy and Reproduction Program, Hunter Medical Research Institute, New Lambton Heights, NSW, Australia.,Priority Research Centre for Reproductive Science, School of Biomedical Science and Pharmacy, University of Newcastle, Callaghan, NSW, Australia
| | - Jessie M Sutherland
- Pregnancy and Reproduction Program, Hunter Medical Research Institute, New Lambton Heights, NSW, Australia.,Priority Research Centre for Reproductive Science, School of Biomedical Science and Pharmacy, University of Newcastle, Callaghan, NSW, Australia
| | - Mingtao Liang
- Pregnancy and Reproduction Program, Hunter Medical Research Institute, New Lambton Heights, NSW, Australia.,Priority Research Centre for Reproductive Science, School of Biomedical Science and Pharmacy, University of Newcastle, Callaghan, NSW, Australia
| | - Diane Rebourcet
- Priority Research Centre for Reproductive Science, University of Newcastle, Callaghan, NSW, Australia.,Pregnancy and Reproduction Program, Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
| | - Brett Nixon
- Priority Research Centre for Reproductive Science, University of Newcastle, Callaghan, NSW, Australia.,Pregnancy and Reproduction Program, Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
| | - Robert J Aitken
- Priority Research Centre for Reproductive Science, University of Newcastle, Callaghan, NSW, Australia.,Pregnancy and Reproduction Program, Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
| |
Collapse
|
8
|
Hu L, Zhou Y, Yang J, Zhao X, Mao L, Zheng W, Zhao J, Guo M, Chen C, He Z, Xu L. MicroRNA-7 overexpression positively regulates the CD8 + SP cell development via targeting PIK3R1. Exp Cell Res 2021; 407:112824. [PMID: 34516985 DOI: 10.1016/j.yexcr.2021.112824] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Revised: 08/22/2021] [Accepted: 09/04/2021] [Indexed: 12/21/2022]
Abstract
microRNA-7 (miR-7), a distinct miRNA family member, has been reported to be involved in the biological functions of immune cells. However, the potential role of miR-7 in the CD8+ T cell development remains to be elucidated. In this study, we estimated the potential effects of miR-7 overexpression in the thymic CD8+ SP cell development using miR-7 overexpression mice. Our results showed that compared with those in control wild type (WT) mice, the volume, weight and total cell numbers of thymus in miR-7 overexpression (OE) mice increased significantly. The absolute cell number of CD8+ SP cells in miR-7 OE mice increased and its ability of activation and proliferation enhanced. Futhermore, we clarified that miR-7 overexpression had an intrinsic promote role in CD8+ SP cell development by adoptive cell transfer assay. Mechanistically, the expression level of PIK3R1, a target of miR-7, decreased significantly in CD8+ SP cells of miR-7 OE mice. Moreover, the expression level of phosphorylated (p)-AKT and p-ERK changed inversely and indicating that miR-7 overexpression impaired the balance of AKE and ERK pathways. In summary, our work reveals an essential role of miR-7 in promoting CD8+ SP cell development through the regulation of PIK3R1 and balance of AKT and ERK pathways.
Collapse
Affiliation(s)
- Lin Hu
- Special Key Laboratory of Gene Detection & Therapy of Guizhou Provincial Education Department, Guizhou, 563000, China; Department of Immunology & Talent Base of Biological Therapy of Guizhou Province, Zunyi Medical University, Guizhou, 563000, China
| | - Ya Zhou
- Special Key Laboratory of Gene Detection & Therapy of Guizhou Provincial Education Department, Guizhou, 563000, China; Department of Medical Physics, Zunyi Medical University, Zunyi, Guizhou, 563003, China
| | - Jing Yang
- Special Key Laboratory of Gene Detection & Therapy of Guizhou Provincial Education Department, Guizhou, 563000, China; Department of Immunology & Talent Base of Biological Therapy of Guizhou Province, Zunyi Medical University, Guizhou, 563000, China
| | - Xu Zhao
- Special Key Laboratory of Gene Detection & Therapy of Guizhou Provincial Education Department, Guizhou, 563000, China; Department of Immunology & Talent Base of Biological Therapy of Guizhou Province, Zunyi Medical University, Guizhou, 563000, China
| | - Ling Mao
- Special Key Laboratory of Gene Detection & Therapy of Guizhou Provincial Education Department, Guizhou, 563000, China; Department of Immunology & Talent Base of Biological Therapy of Guizhou Province, Zunyi Medical University, Guizhou, 563000, China
| | - Wen Zheng
- Department of Laboratory Medicine, Qiannan Medical University for Nationalities, Guizhou 558000, China
| | - Juanjuan Zhao
- Special Key Laboratory of Gene Detection & Therapy of Guizhou Provincial Education Department, Guizhou, 563000, China; Department of Immunology & Talent Base of Biological Therapy of Guizhou Province, Zunyi Medical University, Guizhou, 563000, China
| | - Mengmeng Guo
- Special Key Laboratory of Gene Detection & Therapy of Guizhou Provincial Education Department, Guizhou, 563000, China; Department of Immunology & Talent Base of Biological Therapy of Guizhou Province, Zunyi Medical University, Guizhou, 563000, China
| | - Chao Chen
- Special Key Laboratory of Gene Detection & Therapy of Guizhou Provincial Education Department, Guizhou, 563000, China; Department of Immunology & Talent Base of Biological Therapy of Guizhou Province, Zunyi Medical University, Guizhou, 563000, China
| | - Zhixu He
- Department of Paediatrics, Affiliated Hospital of Zunyi Medical University, Guizhou, 563000, China; Key Laboratory of Adult Stem Cell Transformation Research, Chinese Academy of Medical Sciences, Guizhou, 563000, China
| | - Lin Xu
- Special Key Laboratory of Gene Detection & Therapy of Guizhou Provincial Education Department, Guizhou, 563000, China; Department of Immunology & Talent Base of Biological Therapy of Guizhou Province, Zunyi Medical University, Guizhou, 563000, China.
| |
Collapse
|
9
|
The induction strategies administered in the treatment of multiple myeloma exhibit a deleterious effect on the endothelium. Bone Marrow Transplant 2020; 55:2270-2278. [PMID: 32404979 DOI: 10.1038/s41409-020-0947-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Revised: 04/30/2020] [Accepted: 05/04/2020] [Indexed: 02/02/2023]
Abstract
Multiple myeloma induction treatment includes proteasome inhibitors (PI) and immunomodulatory agents at present. The incidence of engraftment syndrome, a transplant complication potentially related to endothelium, has increased in the last years. Our aim was to investigate whether bortezomib (Velcade, V), thalidomide (T), and dexamethasone (D) affect the endothelium, and explore defibrotide (DF) as protective agent. Endothelial cells (ECs) in culture were exposed to the compounds separately or in combination, without (VTD) and with DF (VTD + DF). Changes in markers of: (i) inflammation (ICAM-1 expression and leukocyte adhesion), (ii) VWF production, (iii) cell permeability (VE-cadherin expression and cell monolayer integrity), and (iv) oxidative stress (ROS production and eNOS expression) were measured. ICAM-1 and VWF expression increased significantly in VTD but were similar to controls in VTD + DF. Separately, bortezomib was the main deleterious agent whereas dexamethasone showed no harmful effect. Leukocyte adhesion showed similar trends. VE-cadherin expression was lower in VTD and normalized in VTD + DF. EC permeability increased only with bortezomib. No changes were observed in oxidative stress markers. Our results demonstrate that bortezomib damages the endothelium, and DF prevents this effect. A better knowledge of the induction drugs impact will allow the design of measures to protect the endothelium.
Collapse
|
10
|
Huang W, Cao Z, Yao Q, Ji Q, Zhang J, Li Y. Mitochondrial damage are involved in Aflatoxin B 1-induced testicular damage and spermatogenesis disorder in mice. THE SCIENCE OF THE TOTAL ENVIRONMENT 2020; 701:135077. [PMID: 31733399 DOI: 10.1016/j.scitotenv.2019.135077] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Revised: 10/14/2019] [Accepted: 10/18/2019] [Indexed: 06/10/2023]
Abstract
Aflatoxin B1 (AFB1) is an unavoidable environmental pollutants, which seriously endangers human and animal health. AFB1 has male reproductive toxicity, yet the underlying mechanisms remain inconclusive. Mitochondra are a kind of crucial organelle for maintaining spermatogenesis in testis. Thus, we hypothesized that AFB1 can impair mitochondria to aggravate testicular damage and spermatogenesis disorder. To verify this hypothesis, 48 male mice were intragastrically administered with 0, 0.375, 0.75 or 1.5 mg/kg body weight AFB1 for 30 days, respectively. In this study, we found AFB1 caused testicular histopathological lesions and spermatogenesis abnormalities, with the elevation of oxidative stress (increased H2O2, whereas decreased SOD and GSH). Significant mitochondria structure damage of germ cells and Leydig cells, MMP loss, ATP contents reduction, and inhibited activities of mitochondrial complexes I-IV in mice testis were found in AFB1 treatment groups. Besides, AFB1 inhibited mitochondrial biogenesis and mitochondrial dynamics, presenting as the decreased mRNA and protein expressions of PGC-1α, Nrf1, Tfam, Drp1, Fis1, Mfn1 and Opa1. The results revealed that the mitochondrial damage were involved in AFB1-induced testicular damage and spermatogenesis disorder, providing a considerable direction to clarify potential mechanisms of AFB1 reproductive toxicity.
Collapse
Affiliation(s)
- Wanyue Huang
- Key Laboratory of the Provincial Education, Department of Heilongjiang for Common Animal Disease Prevention and Treatment, College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China
| | - Zheng Cao
- Key Laboratory of the Provincial Education, Department of Heilongjiang for Common Animal Disease Prevention and Treatment, College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China
| | - Qiucheng Yao
- College of Agriculture, Guangdong Ocean University, Zhanjiang 524000, China
| | - Qiang Ji
- Key Laboratory of the Provincial Education, Department of Heilongjiang for Common Animal Disease Prevention and Treatment, College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China
| | - Jian Zhang
- Key Laboratory of the Provincial Education, Department of Heilongjiang for Common Animal Disease Prevention and Treatment, College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China
| | - Yanfei Li
- Key Laboratory of the Provincial Education, Department of Heilongjiang for Common Animal Disease Prevention and Treatment, College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China.
| |
Collapse
|
11
|
Akaras N, Abuc OO, Koc K, Bal T, Geyikoglu F, Atilay H, Erol HS, Yigit S, Gul M. (1 → 3)-β-d-glucan enhances the toxicity induced by Bortezomib in rat testis. J Food Biochem 2020; 44:e13155. [PMID: 31960484 DOI: 10.1111/jfbc.13155] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Revised: 01/01/2020] [Accepted: 01/02/2020] [Indexed: 12/17/2022]
Abstract
We aimed to determine the possible effects of the antioxidant agent (1 → 3)-β-D-glucan on bortezomib-induced rat testis damage. We used five groups of rats; control, (1 → 3)-β-D-glucan (75 mg/kg), bortezomib group, bortezomib + (1 → 3)-β-D-glucan groups (injection of (1 → 3)-β-D-glucan after bortezomib and sacrificed at 48th or 72nd h). The effects of these substances were assessed by measuring the levels of the antioxidant enzymes and LPO, and by performing immunohistochemical analysis with NF-κB. The histology of testis was evaluated using aniline blue staining. (1 → 3)-β-D-glucan leads to significant reductions in the levels of antioxidant enzymes and increased levels of LPO in testes. Moreover, it increased the NF-κB immunopositivity significantly in testis, especially in Bortezomib + (1 → 3)-β-D-glucan group at 48th h. The histological changes were observed in the bortezomib and/or (1 → 3)-β-D-glucan groups. Our results demonstrated that testis damage caused by the treatment with bortezomib was not eliminated by (1 → 3)-β-D-glucan and shockingly it increased the damage. PRACTICAL APPLICATIONS: The testis damage caused by the treatment with bortezomib was not eliminated by (1 → 3)-β-D-glucan and as a result, β-1,3-(D)-glucan enhanced the toxicity by leading a decrease in the levels of GSH, SOD, and CAT, thus caused an elevation in the immunoreactivity of NF-κB and altered the histopathological changes by enhancing the toxic effects of bortezomib. The findings of the previous studies about the antioxidative activity of (1 → 3)-β-D-glucan are controversial. So, it is necessary to consider the cytotoxicity of (1 → 3)-β-D-glucan in testis tissue. Thus, more studies on testis tissue are necessary to confirm that (1 → 3)-β-D-glucan is safe as an antioxidant.
Collapse
Affiliation(s)
- Nurhan Akaras
- Department of Histology and Embryology, Faculty of Medicine, Aksaray University, Aksaray, Turkey
| | - Ozlem Ozgul Abuc
- Department of Histology and Embryology, Faculty of Medicine, Ataturk University, Erzurum, Turkey
| | - Kubra Koc
- Department of Biology, Faculty of Science, Ataturk University, Erzurum, Turkey
| | - Tugba Bal
- Department of Histology and Embryology, Faculty of Medicine, Ataturk University, Erzurum, Turkey
| | - Fatime Geyikoglu
- Department of Biology, Faculty of Science, Ataturk University, Erzurum, Turkey
| | - Hilal Atilay
- Department of Histology and Embryology, Faculty of Medicine, Ataturk University, Erzurum, Turkey
| | - Huseyin Serkan Erol
- Department of Biochemistry, Faculty of Veterinary Medicine, Kastamonu University, Kastamonu, Turkey
| | - Serdar Yigit
- Department of Histology and Embryology, Faculty of Medicine, Kafkas University, Kars, Turkey
| | - Murat Gul
- Department of Urology, Faculty of Medicine, Aksaray University, Aksaray, Turkey
| |
Collapse
|
12
|
Arab HH, Gad AM, Fikry EM, Eid AH. Ellagic acid attenuates testicular disruption in rheumatoid arthritis via targeting inflammatory signals, oxidative perturbations and apoptosis. Life Sci 2019; 239:117012. [PMID: 31678279 DOI: 10.1016/j.lfs.2019.117012] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Revised: 10/22/2019] [Accepted: 10/24/2019] [Indexed: 02/08/2023]
Abstract
BACKGROUND AND PURPOSE Reduced male fertility has been regarded as a serious complication of rheumatoid arthritis. Phytochemicals have been described as protective agents against rheumatoid arthritis-linked testicular impairment. The current study aimed to investigate the potential protective effects of ellagic acid on rheumatoid arthritis-evoked testicular dysfunction vis-à-vis the reference anti-inflammatory celecoxib. EXPERIMENTAL APPROACH Ellagic acid (50 mg/kg/day) and celecoxib (5 mg/kg/day) were administered orally for 20 days in adjuvant-induced arthritic rats. KEY FINDINGS Current data revealed that ellagic acid counteracted rheumatoid arthritis-evoked testicular histopathologic changes, disrupted sperm characteristics and low gonadosomatic index with comparable efficacy to celecoxib. Ellagic acid also enhanced the testicular steroidogenesis via upregulating the gene expression of 3β-hydroxysteroid dehydrogenase, 17β-hydroxysteroid dehydrogenase and steroidogenic acute regulatory protein with consequent boosting of serum testosterone. Notably, ellagic acid attenuated the testicular inflammatory responses through suppression of myeloperoxidase, tumor necrosis factor-α and cyclo-oxygenase-2 protein expression together with enhancing the anti-inflammatory signal interleukin 10. Ellagic acid also curbed the redox alterations via lowering the production of lipid peroxides and nitric oxide and elevation of the anti-oxidant reduced glutathione. In support of cell survival, ellagic acid combated testicular apoptosis through downregulating caspase-3 protein expression. SIGNIFICANCE The present work accentuates the beneficial actions of ellagic acid in rheumatoid arthritis-incurred testicular impairment and disrupted spermatogenesis via combating the inflammatory, oxidative and apoptotic aberrations.
Collapse
Affiliation(s)
- Hany H Arab
- Biochemistry Division and GTMR Unit, Department of Pharmacology and Toxicology, Faculty of Pharmacy, Taif University, Taif, Saudi Arabia; Department of Biochemistry, Faculty of Pharmacy, Cairo University, Cairo, Egypt.
| | - Amany M Gad
- Department of Pharmacology, National Organization for Drug Control and Research, NODCAR, Giza, Egypt
| | - Ebtehal Mohammad Fikry
- Department of Pharmacology, National Organization for Drug Control and Research, NODCAR, Giza, Egypt
| | - Ahmed H Eid
- Department of Pharmacology, National Organization for Drug Control and Research, NODCAR, Giza, Egypt
| |
Collapse
|
13
|
Arab HH, Gad AM, Fikry EM, Eid AH. Ellagic acid attenuates testicular disruption in rheumatoid arthritis via targeting inflammatory signals, oxidative perturbations and apoptosis. Life Sci 2019. [DOI: https://doi.org/10.1016/j.lfs.2019.117012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
|
14
|
Ni FD, Hao SL, Yang WX. Multiple signaling pathways in Sertoli cells: recent findings in spermatogenesis. Cell Death Dis 2019; 10:541. [PMID: 31316051 PMCID: PMC6637205 DOI: 10.1038/s41419-019-1782-z] [Citation(s) in RCA: 166] [Impact Index Per Article: 27.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2019] [Revised: 06/13/2019] [Accepted: 06/17/2019] [Indexed: 12/25/2022]
Abstract
The functions of Sertoli cells in spermatogenesis have attracted much more attention recently. Normal spermatogenesis depends on Sertoli cells, mainly due to their influence on nutrient supply, maintenance of cell junctions, and support for germ cells' mitosis and meiosis. Accumulating evidence in the past decade has highlighted the dominant functions of the MAPK, AMPK, and TGF-β/Smad signaling pathways during spermatogenesis. Among these pathways, the MAPK signaling pathway regulates dynamics of tight junctions and adherens junctions, proliferation and meiosis of germ cells, proliferation and lactate production of Sertoli cells; the AMPK and the TGF-β/Smad signaling pathways both affect dynamics of tight junctions and adherens junctions, as well as the proliferation of Sertoli cells. The AMPK signaling pathway also regulates lactate supply. These signaling pathways combine to form a complex regulatory network for spermatogenesis. In testicular tumors or infertile patients, the activities of these signaling pathways in Sertoli cells are abnormal. Clarifying the mechanisms of signaling pathways in Sertoli cells on spermatogenesis provides new insights into the physiological functions of Sertoli cells in male reproduction, and also serves as a pre-requisite to identify potential therapeutic targets in abnormal spermatogenesis including testicular tumor and male infertility.
Collapse
Affiliation(s)
- Fei-Da Ni
- The Sperm Laboratory, College of Life Sciences, Zhejiang University, 310058, Hangzhou, Zhejiang, China
| | - Shuang-Li Hao
- The Sperm Laboratory, College of Life Sciences, Zhejiang University, 310058, Hangzhou, Zhejiang, China
| | - Wan-Xi Yang
- The Sperm Laboratory, College of Life Sciences, Zhejiang University, 310058, Hangzhou, Zhejiang, China.
| |
Collapse
|
15
|
Homma T, Fujii J. An SOD1 deficiency aggravates proteasome inhibitor bortezomib-induced testicular damage in mice. Biochim Biophys Acta Gen Subj 2019; 1863:1108-1115. [PMID: 30974160 DOI: 10.1016/j.bbagen.2019.04.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Revised: 03/23/2019] [Accepted: 04/05/2019] [Indexed: 10/27/2022]
Abstract
Proteasomes play a key role in maintaining cellular homeostasis by the proteolytic removal of proteins, including ubiquitinated proteins and/or oxidatively-damaged proteins. The proteasome inhibitor bortezomib (BTZ) has been reported to exert testicular toxicity in mice. In the current study, we treated SOD1-knockout (KO) mice with BTZ and investigated the issue of whether oxidative stress is involved in the development of testicular toxicity. The BTZ treatment significantly increased superoxide production and cell death in the testes of SOD1-KO mice compared to wild-type (WT) mice. We also found that high levels of both ubiquitinated proteins and p62 accumulated and underwent aggregation in the seminiferous tubules of BTZ-injected SOD1-KO mice. Furthermore, the proteolytic activities of proteasomes were significantly decreased in the testes of BTZ-injected SOD1-KO mice compared to their WT counterparts. These results suggest that a combination of oxidative stress caused by an SOD1 deficiency and proteasome inhibition by BTZ accelerates the impairment of proteasomes, which results in severe testicular damage in SOD1-KO mice.
Collapse
Affiliation(s)
- Takujiro Homma
- Department of Biochemistry and Molecular Biology, Graduate School of Medical Science, Yamagata University, 2-2-2 Iidanishi, Yamagata 990-9585, Japan.
| | - Junichi Fujii
- Department of Biochemistry and Molecular Biology, Graduate School of Medical Science, Yamagata University, 2-2-2 Iidanishi, Yamagata 990-9585, Japan
| |
Collapse
|
16
|
Eid AH, Gad AM, Fikry EM, Arab HH. Venlafaxine and carvedilol ameliorate testicular impairment and disrupted spermatogenesis in rheumatoid arthritis by targeting AMPK/ERK and PI3K/AKT/mTOR pathways. Toxicol Appl Pharmacol 2019. [DOI: https://doi.org/10.1016/j.taap.2018.12.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
|
17
|
Eid AH, Gad AM, Fikry EM, Arab HH. Venlafaxine and carvedilol ameliorate testicular impairment and disrupted spermatogenesis in rheumatoid arthritis by targeting AMPK/ERK and PI3K/AKT/mTOR pathways. Toxicol Appl Pharmacol 2019; 364:83-96. [PMID: 30578887 DOI: 10.1016/j.taap.2018.12.014] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2018] [Revised: 12/11/2018] [Accepted: 12/18/2018] [Indexed: 02/08/2023]
Abstract
Testicular impairment has been commonly described in long-standing rheumatoid arthritis (RA) patients. Since depression and cardiovascular disorders are the most disturbing co-morbidities of RA, investigating the efficacy of the anti-depressant venlafaxine or the beta-blocker carvedilol in RA-associated testicular dysfunction may add to their clinical utility for RA patients. Previously, both agents have demonstrated significant in vivo anti-oxidant and anti-inflammatory actions. In the current study, venlafaxine (50 mg/kg/day) and carvedilol (10 mg/kg/day) were orally administered to adjuvant arthritic rats for 20 days. Interestingly, venlafaxine and carvedilol effectively suppressed paw edema and mitigated the testicular histopathological aberrations and the disrupted spermatogenesis. Both drugs enhanced testicular steroidogenesis through upregulation of 3β-HSD, 17β-HSD and StAR gene expression with concomitant augmentation of serum testosterone. They also blunted the inflammatory burden via attenuation of myeloperoxidase, TNF-α and the protein expression of NF-κBp65 along with elevation of IL-10. They attenuated testicular oxidative perturbations via lowering lipid peroxides and nitric oxide and boosting glutathione levels. With regard to apoptosis, the two agents lowered the protein expression of caspase-3, cleaved caspase-3, cleaved PARP, Bax and p53, promoting germ cell survival. They also modulated the AMPK/ERK signaling via lowering of p-AMPK and upregulation of p-ERK1/2 along with PI3K/AKT/mTOR transduction by enhancing the PI3Kp110α, p-AKT and p-mTOR protein expression. Together, the present work demonstrates the beneficial effects of venlafaxine and carvedilol in RA testicular dysfunction and impaired spermatogenesis via modulation of AMPK/ERK and PI3K/AKT/mTOR signaling and intervention with the testicular oxidative stress, inflammation and apoptosis.
Collapse
Affiliation(s)
- Ahmed H Eid
- Department of Pharmacology, National Organization for Drug Control and Research, NODCAR, Giza 12553, Egypt
| | - Amany M Gad
- Department of Pharmacology, National Organization for Drug Control and Research, NODCAR, Giza 12553, Egypt
| | - Ebtehal Mohammad Fikry
- Department of Pharmacology, National Organization for Drug Control and Research, NODCAR, Giza 12553, Egypt
| | - Hany H Arab
- Department of Biochemistry, Faculty of Pharmacy, Cairo University, Cairo, Egypt; Biochemistry Division and GTMR Unit, Department of Pharmacology and Toxicology, Faculty of Pharmacy, Taif University, Taif, Saudi Arabia.
| |
Collapse
|
18
|
Soriano-Ursúa MA, Farfán-García ED, Geninatti-Crich S. Turning Fear of Boron Toxicity into Boron-containing Drug Design. Curr Med Chem 2019; 26:5005-5018. [PMID: 30919770 DOI: 10.2174/0929867326666190327154954] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2018] [Revised: 01/24/2019] [Accepted: 02/08/2019] [Indexed: 02/07/2023]
Abstract
BACKGROUND Despite the historical employment of boron-containing compounds (BCCs) with medicinal purposes, the reported cases of BCC toxicity in humans during the twentieth-century drived us towards a "boron-withdrawal" period. Fortunately, the use of boric acid for specific purposes remains, and the discovery of natural BCCs with biological action attractive for therapeutic purposes as well as the introduction of some new BCCs for clinical use has reactivated the interest in studying the properties of these BCCs. METHODS We carried out a structured search of bibliographic databases for scientific peerreviewed research literature regarding boron toxicity and linked that information to that of BCCs in drug design and development. A deductive qualitative content analysis methodology was applied to analyse the interventions and findings of the included studies using a theoretical outline. RESULTS This review recapitulates the following on a timeline: the boron uses in medicine, the data known about the toxicological profiles of some BCCs, the pharmacological properties of some BCCs that are employed in cancer and infectious disease therapies, and the known properties of BCCs recently introduced into clinical assays as well as the identification of their structure-activity relationships for toxicity and therapeutic use. Then, we discuss the use of new approaches taking advantage of some toxicological data to identify potent and efficient BCCs for prevention and therapy while limiting their toxic effects. CONCLUSION Data for boron toxicity can be strategically used for boron-containing drug design.
Collapse
Affiliation(s)
- Marvin A Soriano-Ursúa
- Departamentos de Fisiologia, Bioquimica y Seccion de Estudios de Posgrado e Investigacion, Escuela Superior de Medicina, Plan de San Luis y Diaz Miron s/n, 11340, Mexico City,Mexico
| | - Eunice D Farfán-García
- Departamentos de Fisiologia, Bioquimica y Seccion de Estudios de Posgrado e Investigacion, Escuela Superior de Medicina, Plan de San Luis y Diaz Miron s/n, 11340, Mexico City,Mexico
| | - Simonetta Geninatti-Crich
- Dipartimento di Biotecnologie Molecolari e Scienze per la Salute, Centro, Imaging Molecolare, Universita di Torino, via Nizza 52, Torino, 10126,Italy
| |
Collapse
|
19
|
Martin-Hidalgo D, Hurtado de Llera A, Calle-Guisado V, Gonzalez-Fernandez L, Garcia-Marin L, Bragado MJ. AMPK Function in Mammalian Spermatozoa. Int J Mol Sci 2018; 19:ijms19113293. [PMID: 30360525 PMCID: PMC6275045 DOI: 10.3390/ijms19113293] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2018] [Revised: 10/16/2018] [Accepted: 10/20/2018] [Indexed: 01/03/2023] Open
Abstract
AMP-activated protein kinase AMPK regulates cellular energy by controlling metabolism through the inhibition of anabolic pathways and the simultaneous stimulation of catabolic pathways. Given its central regulator role in cell metabolism, AMPK activity and its regulation have been the focus of relevant investigations, although only a few studies have focused on the AMPK function in the control of spermatozoa's ability to fertilize. This review summarizes the known cellular roles of AMPK that have been identified in mammalian spermatozoa. The involvement of AMPK activity is described in terms of the main physiological functions of mature spermatozoa, particularly in the regulation of suitable sperm motility adapted to the fluctuating extracellular medium, maintenance of the integrity of sperm membranes, and the mitochondrial membrane potential. In addition, the intracellular signaling pathways leading to AMPK activation in mammalian spermatozoa are reviewed. We also discuss the role of AMPK in assisted reproduction techniques, particularly during semen cryopreservation and preservation (at 17 °C). Finally, we reinforce the idea of AMPK as a key signaling kinase in spermatozoa that acts as an essential linker/bridge between metabolism energy and sperm's ability to fertilize.
Collapse
Affiliation(s)
- David Martin-Hidalgo
- Research Group of Intracellular Signaling and Technology of Reproduction (SINTREP), Institute of Biotechnology in Agriculture and Livestock (INBIO G+C), University of Extremadura, 10003 Cáceres, Spain.
- Unit for Multidisciplinary Research in Biomedicine (UMIB), Laboratory of Cell Biology, Department of Microscopy, Institute of Biomedical Sciences Abel Salazar (ICBAS), University of Porto, 40050-313 Porto, Portugal.
| | - Ana Hurtado de Llera
- Research Group of Intracellular Signaling and Technology of Reproduction (SINTREP), Institute of Biotechnology in Agriculture and Livestock (INBIO G+C), University of Extremadura, 10003 Cáceres, Spain.
- Hormones and Metabolism Research Group, Faculty of Health Sciences, University of Beira Interior, 6200-506 Covilhã, Portugal.
| | - Violeta Calle-Guisado
- Research Group of Intracellular Signaling and Technology of Reproduction (SINTREP), Institute of Biotechnology in Agriculture and Livestock (INBIO G+C), University of Extremadura, 10003 Cáceres, Spain.
| | - Lauro Gonzalez-Fernandez
- Research Group of Intracellular Signaling and Technology of Reproduction (SINTREP), Institute of Biotechnology in Agriculture and Livestock (INBIO G+C), University of Extremadura, 10003 Cáceres, Spain.
| | - Luis Garcia-Marin
- Research Group of Intracellular Signaling and Technology of Reproduction (SINTREP), Institute of Biotechnology in Agriculture and Livestock (INBIO G+C), University of Extremadura, 10003 Cáceres, Spain.
| | - M Julia Bragado
- Research Group of Intracellular Signaling and Technology of Reproduction (SINTREP), Institute of Biotechnology in Agriculture and Livestock (INBIO G+C), University of Extremadura, 10003 Cáceres, Spain.
| |
Collapse
|
20
|
Interference with lactate metabolism by mmu-miR-320-3p via negatively regulating GLUT3 signaling in mouse Sertoli cells. Cell Death Dis 2018; 9:964. [PMID: 30237478 PMCID: PMC6148074 DOI: 10.1038/s41419-018-0958-2] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2018] [Revised: 08/13/2018] [Accepted: 08/23/2018] [Indexed: 12/30/2022]
Abstract
Disruption of the nursery function in Sertoli cells (SCs) by reducing lactate production, a preferred energy substrate for developed germ cells (spermatocytes and spermatids), is tightly associated with spermatogenic failure such as SC-only syndrome (SCOS). However, whether this complicated pathogenesis is regulated by certain miRNAs at the post-transcriptional level remain fascinating but largely unknown. Here we show for the first time that mmu-miR-320-3p was exclusively expressed in murine SCs and this expression was significantly induced in busulphan-treated murine testis. The most efficient stimulatory germ cell types for the induction of apoptosis-elicited mmu-miR-320-3p expression were meiotic spermatocytes and haploid spermatids. Functionally, forced expression of the exogenous mmu-miR-320-3p in SCs compromises male fertility by causing oligozoospermia and defection of sperm mobility. Mechanistically, mmu-miR-320-3p negatively regulates lactate production of SCs by directly inhibiting glucose transporter 3 (GLUT3) expression. Thus, dysregulation of mmu-miR-320-3p/GLUT3 cascade and consequently of lactate deficiency may be a key molecular event contributing the germ cell loss by SC dysfunction. Future endeavor in the continuous investigation of this important circulating miRNA may shed novel insights into epigenetic regulation of SCs nursery function and the etiology of azoospermia, and offers novel therapeutic and prognostic targets for SCOS.
Collapse
|
21
|
Bello M, Guadarrama-García C, Velasco-Silveyra LM, Farfán-García ED, Soriano-Ursúa MA. Several effects of boron are induced by uncoupling steroid hormones from their transporters in blood. Med Hypotheses 2018; 118:78-83. [PMID: 30037620 DOI: 10.1016/j.mehy.2018.06.024] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2018] [Accepted: 06/19/2018] [Indexed: 02/07/2023]
Abstract
Boron is increasingly added to food supplements due to multiple effects that have been reported in mammals after boric acid administration. Among these effects are inflammatory process control, bone and muscle strength enhancement, protein expression regulation, and a decreased risk of developing some pathologies in which these processes are key, such as osteoporosis, dermatological inflammatory non-infectious maladies and diseases affecting the central nervous system. Experimental data have suggested that steroid hormone levels in plasma change after boric acid administration, but a clear mechanism behind these variations has not been established. We analyzed possibilities for these changes and hypothesized that boric acid disrupts the interactions between steroid hormones and several carriers in plasma. In particular, we proposed that there is an uncoupling of the interactions between sex hormone binding globulin (SHBG) and estrogens and testosterone and that there are alterations in the binding of hydrophobic ligands by other carrier proteins in plasma. Further experimental and computational studies are required to support the hypothesis that boric acid and probably other boron-containing compounds can displace steroid hormones from their plasma carriers. If such phenomena are confirmed, boron administration with a clear mechanism could be employed as a therapeutic agent in several diseases or physiological events that require modulation of steroid hormone levels in plasma.
Collapse
Affiliation(s)
- Martiniano Bello
- Laboratorio de Modelado Molecular y Sección de Estudios de Posgrado e Investigación, Escuela Superior de Medicina del Instituto Politécnico Nacional, Plan de San Luis y Díaz Mirón s/n, 11340 México City, Mexico
| | - Concepción Guadarrama-García
- Laboratorio de Modelado Molecular y Sección de Estudios de Posgrado e Investigación, Escuela Superior de Medicina del Instituto Politécnico Nacional, Plan de San Luis y Díaz Mirón s/n, 11340 México City, Mexico; Departamento de Fisiología y Sección de Estudios de Posgrado e Investigación, Escuela Superior de Medicina del Instituto Politécnico Nacional, Plan de San Luis y Díaz Mirón s/n, 11340 México City, Mexico
| | - Luz M Velasco-Silveyra
- Departamento de Fisiología y Sección de Estudios de Posgrado e Investigación, Escuela Superior de Medicina del Instituto Politécnico Nacional, Plan de San Luis y Díaz Mirón s/n, 11340 México City, Mexico
| | - Eunice D Farfán-García
- Departamento de Fisiología y Sección de Estudios de Posgrado e Investigación, Escuela Superior de Medicina del Instituto Politécnico Nacional, Plan de San Luis y Díaz Mirón s/n, 11340 México City, Mexico
| | - Marvin A Soriano-Ursúa
- Departamento de Fisiología y Sección de Estudios de Posgrado e Investigación, Escuela Superior de Medicina del Instituto Politécnico Nacional, Plan de San Luis y Díaz Mirón s/n, 11340 México City, Mexico.
| |
Collapse
|
22
|
Akaras N, Bal T, Atilay H, Selli J, Halici MB. Protective effects of agomelatine on testicular damage caused by bortezomib. Biotech Histochem 2017; 92:552-559. [DOI: 10.1080/10520295.2017.1350748] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Affiliation(s)
- N Akaras
- Department of Histology and Embryology, Faculty of Medicine
| | - T Bal
- Department of Histology and Embryology, Faculty of Medicine
| | - H Atilay
- Department of Histology and Embryology, Faculty of Medicine
| | - J Selli
- Department of Histology and Embryology, Faculty of Medicine
| | - MB Halici
- Biochemistry, Faculty of Veterinary Medicine, Ataturk University, Erzurum, Turkey
| |
Collapse
|
23
|
Liu JZ, Yin FY, Yan CY, Wang H, Luo XH. Regulation of Docetaxel Sensitivity in Prostate Cancer Cells by hsa-miR-125a-3p via Modulation of Metastasis-Associated Protein 1 Signaling. Urology 2017; 105:208.e11-208.e17. [PMID: 28088556 DOI: 10.1016/j.urology.2017.01.001] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2016] [Revised: 12/07/2016] [Accepted: 01/02/2017] [Indexed: 01/06/2023]
Abstract
OBJECTIVE To identify the potential downstream targets of hsa-miR-125a-3p, a mature form of miR-125a, during the pathogenesis of chemoresistance in prostate cancer (PCa). MATERIALS AND METHODS The expression levels of hsa-miR-125a-3p were assessed in chemoresistant PCa tissues and experimentally established chemoresistant cells using quantitative reverse transcription-polymerase chain reaction. The effect of hsa-miR-125a-3p knockdown or hsa-miR-125a-3p overexpression on the Dox-induced cell death was evaluated using apoptosis ELISA in chemosensitive PC-3 cells or in chemoresistant PC-3 cells (PC-3R). Finally, using multiple assays, the regulation of metastasis-associated protein 1 (MTA1), an essential component of the Mi-2-nucleosome remodeling deacetylation complex, by hsa-miR-125a-3p was studied at both molecular and functional levels. RESULTS The expression of hsa-miR-125a-3p was significantly downregulated in chemoresistant PCa tissues and cells. Inhibition of hsa-miR-125a-3p significantly increased docetaxel (Dox) resistance in PC-3 cells, whereas upregulation of hsa-miR-125a-3p effectively reduced Dox resistance in PC-3R, suggesting that this microRNA (miRNA) may act as a tumor suppressor along the pathogenesis of drug resistance. Mechanistically, hsa-miR-125a-3p induced apoptosis and Dox sensitivity in PCa cells through regulating MTA1. CONCLUSION Our results collectively indicate that miRNA-MTA1 can form a delicate regulatory loop to maintain a bistable state in the Dox chemosensitivity, and future endeavor in this filed should provide important clues to develop miRNA-based therapies that benefit advanced PCa patients through modulating the functional status of MTA1.
Collapse
Affiliation(s)
- Jian-Zhou Liu
- Department of Urology, Baoji Central Hospital, Baoji, Shaanxi Province, China
| | - Feng-Yan Yin
- Department of Urology, Baoji Central Hospital, Baoji, Shaanxi Province, China
| | - Chang-You Yan
- Xi'an Health Management Service Center, Xi'an, Shaanxi Province, China
| | - Hui Wang
- Department of Medical Psychology, Fourth Military Medical University, Xi'an, Shaanxi Province, China
| | - Xiao-Hui Luo
- Department of Urology, Baoji Central Hospital, Baoji, Shaanxi Province, China.
| |
Collapse
|
24
|
miR-24 suppression of POZ/BTB and AT-hook-containing zinc finger protein 1 (PATZ1) protects endothelial cell from diabetic damage. Biochem Biophys Res Commun 2016; 480:682-689. [DOI: 10.1016/j.bbrc.2016.10.116] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2016] [Accepted: 10/12/2016] [Indexed: 12/16/2022]
|
25
|
Gao ZJ, Min J, Wu XC, Yang T, Yan CY, Dong BH, Zhang T. Repression of neuronal nitric oxide (nNOS) synthesis by MTA1 is involved in oxidative stress-induced neuronal damage. Biochem Biophys Res Commun 2016; 479:40-7. [PMID: 27603575 DOI: 10.1016/j.bbrc.2016.09.016] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2016] [Accepted: 09/03/2016] [Indexed: 12/14/2022]
Abstract
The Metastasis-associated protein 1 (MTA1) coregulator, an essential component of the nucleosome remodeling and deacetylase (NuRD) complex, potentiates neuroprotective effects against ischemia/reperfusion (I/R) injury. But the underlying mechanism(s) remain largely unknown. Here, we discovered that neuronal MTA1 was a target of oxidative stress, and stimulation of neurons with oxygen glucose deprivation (OGD) treatment significantly inhibited MTA1 expression. Additionally, MTA1 depletion augmented ischemic oxidative stress and thus promoted oxidative stress-induced neuronal cell death by OGD. While studying the impact of MTA1 status on global neuronal gene expression, we unexpectedly discovered that MTA1 may modulate OGD-induced neuronal damage via regulation of distinct nitric oxide synthase (NOS) (namely neuronal NOS, nNOS) signaling. We provided in vitro evidence that NOS1 is a chromatin target of MTA1 in OGD-insulted neurons. Mechanistically, neuronal ischemia-mediated repression of NOS1 expression is accompanied by the enhanced recruitment of MTA1 along with histone deacetylases (HDACs) to the NOS1 promoter, which could be effectively blocked by a pharmacological inhibitor of the HDACs. These findings collectively reveal a previously unrecognized, critical homeostatic role of MTA1, both as a target and as a component of the neuronal oxidative stress, in the regulation of acute neuronal responses against brain I/R damage. Our study also provides a molecular mechanistic explanation for the previously reported neurovascular protection by selective nNOS inhibitors.
Collapse
Affiliation(s)
- Zi-Jun Gao
- Department of Anesthesiology, Honghui Hospital, Xi'an Jiaotong University College of Medicine, Xi'an 710054, China
| | - Jie Min
- Department of Ophthalmology, Xi'an No. 4 Hospital, Guangren Hospital of Xi'an Jiaotong University, Xi'an 710004, China
| | - Xu-Cai Wu
- Department of Anesthesiology, Honghui Hospital, Xi'an Jiaotong University College of Medicine, Xi'an 710054, China
| | - Tian Yang
- The 1st Cadet Brigade, Fourth Military Medical University, Xi'an 710032, China
| | - Chang-You Yan
- Xi'an Health Management Service Center, Xi'an 710032, China
| | - Bu-Huai Dong
- Department of Anesthesiology, Honghui Hospital, Xi'an Jiaotong University College of Medicine, Xi'an 710054, China.
| | - Tao Zhang
- Department of Neurosurgery, Tangdu Hospital, Fourth Military Medical University, Xi'an 710038, China.
| |
Collapse
|
26
|
Xu X, Lv YG, Yan CY, Yi J, Ling R. Enforced expression of hsa-miR-125a-3p in breast cancer cells potentiates docetaxel sensitivity via modulation of BRCA1 signaling. Biochem Biophys Res Commun 2016; 479:893-900. [PMID: 27693788 DOI: 10.1016/j.bbrc.2016.09.087] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2016] [Accepted: 09/17/2016] [Indexed: 01/03/2023]
Abstract
Epigenetic gene inactivation by microRNAs (miRNAs) plays a key role in malignant transformation, prevention of apoptosis, drug resistance and metastasis. It has been shown that miR-125a is down-regulated in HER2-amplified and HER2-overexpressing breast cancers (BCa), and this miRNA is believed to serve as an important tumor suppressor. miR-125a has two mature forms: hsa-miR-125a-3p and hsa-miR-125a-5p. However, the functional details of these miRNAs in BCa, particularly during pathogenesis of drug resistance, remain largely unexplored. Herein, we reported that hsa-miR-125a-3p expression was significantly reduced in chemoresistant BCa tissues and in experimentally established chemoresistant BCa cells. hsa-miR-125a-3p knockdown promoted cell proliferation and compromised docetaxel (Dox)-induced cell death, whereas overexpression of hsa-miR-125a-3p attenuated Dox chemoresistance in BCa cells. From a mechanistic standpoint, hsa-miR-125a-3p directly targeted 3'-untranslated regions (3'-UTRs) of breast cancer early onset gene 1 (BRCA1) and inhibits its protein expression via translational repression mechanism. In addition, suppression of BRCA1 expression by siRNA treatment effectively improved hsa-miR-125a-3p deficiency-triggered chemoresistance in BCa cells. Collectively, these findings suggest that hsa-miR-125a-3p may function as a tumor suppressor by regulating the BRCA1 signaling, and reintroduction of hsa-miR-125a-3p analogs could be a potential adjunct therapy for advanced/chemoresistant BCa.
Collapse
Affiliation(s)
- Xin Xu
- Department of Thyroid Gland and Breast Vascular Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, Shaanxi Province, China
| | - Yong-Gang Lv
- Department of Thyroid Gland and Breast Vascular Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, Shaanxi Province, China
| | - Chang-You Yan
- Xi'an Health Management Service Center, Xi'an 710032, Shaanxi Province, China
| | - Jun Yi
- Department of Thyroid Gland and Breast Vascular Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, Shaanxi Province, China.
| | - Rui Ling
- Department of Thyroid Gland and Breast Vascular Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, Shaanxi Province, China.
| |
Collapse
|
27
|
Chen RA, Sun XM, Yan CY, Liu L, Hao MW, Liu Q, Jiao XY, Liang YM. Hyperglycemia-induced PATZ1 negatively modulates endothelial vasculogenesis via repression of FABP4 signaling. Biochem Biophys Res Commun 2016; 477:548-555. [DOI: 10.1016/j.bbrc.2016.06.052] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2016] [Accepted: 06/09/2016] [Indexed: 12/11/2022]
|
28
|
Kwak HJ, Choi HE, Jang J, Park SK, Bae YA, Cheon HG. Bortezomib attenuates palmitic acid-induced ER stress, inflammation and insulin resistance in myotubes via AMPK dependent mechanism. Cell Signal 2016; 28:788-97. [PMID: 27049873 DOI: 10.1016/j.cellsig.2016.03.015] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2015] [Revised: 03/24/2016] [Accepted: 03/28/2016] [Indexed: 11/30/2022]
Abstract
Bortezomib is an anti-cancer agent that induces ER stress by inhibiting proteasomal degradation. However, the effects of bortezomib appear to be dependent on its concentration and cellular context. Since ER stress is closely related to type 2 diabetes, the authors examined the effects of bortezomib on palmitic acid (PA)-induced ER stress in C2C12 murine myotubes. At low concentrations (<20nM), bortezomib protected myotubes from PA (750μM)-induced ER stress and inflammation. Either tunicamycin or thapsigargin-induced ER stress was also reduced by bortezomib. In addition, reduced glucose uptake and Akt phosphorylation induced by PA were prevented by co-treating bortezomib (10nM) both in the presence or absence of insulin. These protective effects of bortezomib were found to be associated with reduced JNK phosphorylation. Furthermore, bortezomib-induced AMPK phosphorylation, and the protective effects of bortezomib were diminished by AMPK knockdown, suggesting that AMPK activation underlies the effects of bortezomib. The in vivo administration of bortezomib at nontoxic levels (at 50 or 200μg/kg, i.p.) twice weekly for 5weeks to ob/ob mice improved insulin resistance, increased AMPK phosphorylation, reduced ER stress marker levels, and JNK inhibition in skeletal muscle. The study shows that bortezomib reduces ER stress, inflammation, and insulin resistance in vitro and in vivo, and suggests that bortezomib has novel applications for the treatment of metabolic disorders.
Collapse
Affiliation(s)
- Hyun Jeong Kwak
- Department of Pharmacology, Gachon University School of Medicine, Incheon 406-799, Republic of Korea
| | - Hye-Eun Choi
- Department of Pharmacology, Gachon University School of Medicine, Incheon 406-799, Republic of Korea
| | - Jinsun Jang
- Department of Pharmacology, Gachon University School of Medicine, Incheon 406-799, Republic of Korea
| | - Soo Kyung Park
- Department of Pharmacology, Gachon University School of Medicine, Incheon 406-799, Republic of Korea
| | - Young-An Bae
- Department of Microbiology, Gachon University, Incheon 406-799, Republic of Korea
| | - Hyae Gyeong Cheon
- Department of Pharmacology, Gachon University School of Medicine, Incheon 406-799, Republic of Korea; Gachon Medical Research Institute, Gil Medical Center, Incheon 405-760, Republic of Korea.
| |
Collapse
|
29
|
Ansari-Pour N, Razaghi-Moghadam Z, Barneh F, Jafari M. Testis-Specific Y-Centric Protein-Protein Interaction Network Provides Clues to the Etiology of Severe Spermatogenic Failure. J Proteome Res 2016; 15:1011-22. [PMID: 26794825 DOI: 10.1021/acs.jproteome.5b01080] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Pinpointing causal genes for spermatogenic failure (SpF) on the Y chromosome has been an ever daunting challenge with setbacks during the past decade. Since complex diseases result from the interaction of multiple genes and also display considerable missing heritability, network analysis is more likely to explicate an etiological molecular basis. We therefore took a network medicine approach by integrating interactome (protein-protein interaction (PPI)) and transcriptome data to reconstruct a Y-centric SpF network. Two sets of seed genes (Y genes and SpF-implicated genes (SIGs)) were used for network reconstruction. Since no PPI was observed among Y genes, we identified their common immediate interactors. Interestingly, 81% (N = 175) of these interactors not only interacted directly with SIGs, but also they were enriched for differentially expressed genes (89.6%; N = 43). The SpF network, formed mainly by the dys-regulated interactors and the two seed gene sets, comprised three modules enriched for ribosomal proteins and nuclear receptors for sex hormones. Ribosomal proteins generally showed significant dys-regulation with RPL39L, thought to be expressed at the onset of spermatogenesis, strongly down-regulated. This network is the first global PPI network pertaining to severe SpF and if experimentally validated on independent data sets can lead to more accurate diagnosis and potential fertility recovery of patients.
Collapse
Affiliation(s)
- Naser Ansari-Pour
- Faculty of New Sciences and Technology, University of Tehran , North Kargar Street, Tehran 143995-7131, Iran.,School of Biological Sciences, Institute for Research in Fundamental Sciences (IPM) , Tehran 19395-5531, Iran
| | - Zahra Razaghi-Moghadam
- Faculty of New Sciences and Technology, University of Tehran , North Kargar Street, Tehran 143995-7131, Iran.,School of Biological Sciences, Institute for Research in Fundamental Sciences (IPM) , Tehran 19395-5531, Iran
| | - Farnaz Barneh
- Faculty of Paramedical Sciences, Shahid Beheshti University of Medical Sciences , Tehran 198396-3113, Iran
| | - Mohieddin Jafari
- Drug Design and Bioinformatics Unit, Medical Biotechnology Department, Biotechnology Research Center, Pasteur Institute of Iran , Tehran 131694-3551, Iran.,School of Biological Sciences, Institute for Research in Fundamental Sciences (IPM) , Tehran 19395-5531, Iran
| |
Collapse
|
30
|
Proteasome inhibitors induce AMPK activation via CaMKKβ in human breast cancer cells. Breast Cancer Res Treat 2015; 153:79-88. [DOI: 10.1007/s10549-015-3512-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2015] [Accepted: 07/18/2015] [Indexed: 01/15/2023]
|