1
|
Pibuel MA, Poodts D, Molinari Y, Díaz M, Amoia S, Byrne A, Hajos S, Lompardía S, Franco P. The importance of RHAMM in the normal brain and gliomas: physiological and pathological roles. Br J Cancer 2023; 128:12-20. [PMID: 36207608 PMCID: PMC9814267 DOI: 10.1038/s41416-022-01999-w] [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: 05/16/2022] [Revised: 09/19/2022] [Accepted: 09/22/2022] [Indexed: 01/27/2023] Open
Abstract
Although the literature about the functions of hyaluronan and the CD44 receptor in the brain and brain tumours is extensive, the role of the receptor for hyaluronan-mediated motility (RHAMM) in neural stem cells and gliomas remain poorly explored. RHAMM is considered a multifunctional receptor which performs various biological functions in several normal tissues and plays a significant role in cancer development and progression. RHAMM was first identified for its ability to bind to hyaluronate, the extracellular matrix component associated with cell motility control. Nevertheless, additional functions of this protein imply the interaction with different partners or cell structures to regulate other biological processes, such as mitotic-spindle assembly, gene expression regulation, cell-cycle control and proliferation. In this review, we summarise the role of RHAMM in normal brain development and the adult brain, focusing on the neural stem and progenitor cells, and discuss the current knowledge on RHAMM involvement in glioblastoma progression, the most aggressive glioma of the central nervous system. Understanding the implications of RHAMM in the brain could be useful to design new therapeutic approaches to improve the prognosis and quality of life of glioblastoma patients.
Collapse
Affiliation(s)
- Matías A Pibuel
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica; Departamento de Microbiología, Inmunología y Biotecnología; Instituto de Estudios de la Inmunidad Humoral (IDEHU)-CONICET, Capital Federal (1113), Buenos Aires, Argentina.
| | - Daniela Poodts
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica; Departamento de Microbiología, Inmunología y Biotecnología; Instituto de Estudios de la Inmunidad Humoral (IDEHU)-CONICET, Capital Federal (1113), Buenos Aires, Argentina
| | - Yamila Molinari
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica; Departamento de Química Biológica. Instituto de Química y Fisicoquímica Biológicas (IQUIFIB)-CONICET, Capital Federal (1113), Buenos Aires, Argentina
| | - Mariángeles Díaz
- Instituto de Estudios de la Inmunidad Humoral (IDEHU)- CONICET, Universidad de Buenos Aires, Capital Federal (1113), Buenos Aires, Argentina
| | - Sofía Amoia
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica; Departamento de Microbiología, Inmunología y Biotecnología; Instituto de Estudios de la Inmunidad Humoral (IDEHU)-CONICET, Capital Federal (1113), Buenos Aires, Argentina
| | - Agustín Byrne
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica; Departamento de Química Biológica. Instituto de Química y Fisicoquímica Biológicas (IQUIFIB)-CONICET, Capital Federal (1113), Buenos Aires, Argentina
| | - Silvia Hajos
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica; Departamento de Microbiología, Inmunología y Biotecnología; Instituto de Estudios de la Inmunidad Humoral (IDEHU)-CONICET, Capital Federal (1113), Buenos Aires, Argentina
| | - Silvina Lompardía
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica; Departamento de Microbiología, Inmunología y Biotecnología; Instituto de Estudios de la Inmunidad Humoral (IDEHU)-CONICET, Capital Federal (1113), Buenos Aires, Argentina
| | - Paula Franco
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica; Departamento de Química Biológica. Instituto de Química y Fisicoquímica Biológicas (IQUIFIB)-CONICET, Capital Federal (1113), Buenos Aires, Argentina
| |
Collapse
|
2
|
Al-Maghrabi J, Khabaz MN. Cyclooxygenase-2 immunohistochemical expression is associated with worse prognosis in breast cancer: Retrospective study and literature review. Saudi Med J 2022; 43:687-693. [PMID: 35830999 PMCID: PMC9749694 DOI: 10.15537/smj.2022.43.7.20220052] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2022] [Accepted: 05/28/2022] [Indexed: 11/16/2022] Open
Abstract
OBJECTIVES To assess the immunohistochemistry phenotype of cyclooxygenase-2 (COX-2) in breast cancer (BC) and to correlate it with histological and clinical prognostic factors. METHODS This retrospective study utilized COX-2 monoclonal antibody in an immunohistochemistry staining of tissue microarrays slides of 570 cases of previously diagnosed BC and with 52 of normal breast tissues from breast specimens resected for benign lesions or reconstruction (fibroadenoma and normal breast epithelium). This project was carried out in the Laboratory of pathology, King Abdulaziz University, Jeddah, Saudi Arabia, between September 2019 and September 2021. RESULTS The present data showed an important connection between the COX-2 expression phenotype and BC compared to benign breast tissues (p=0.034). The expression pattern of COX-2 was allied significantly with some factors which distinguished aggressive subtypes of BC, such as stage, distant metastases, lymphovascular invasion, and poor survival. CONCLUSION Cyclooxygenase-2 is a valuable marker that could facilitate BC diagnosis and prognosis.
Collapse
Affiliation(s)
- Jaudah Al-Maghrabi
- From the Department of Pathology (Al-Maghrabi), Faculty of Medicine; from the Department of Pathology (Khabaz), Rabigh Faculty of Medicine, King Abdulaziz University, and from the Department of Pathology (Al-Maghrabi), King Faisal Specialist Hospital and Research Centre, Jeddah, Kingdom of Saudi Arabia.
- Address correspondence and reprint request to: Dr. Mohamad N. Khabaz, Department of Pathology, Rabigh Faculty of Medicine, King Abdulaziz University, Jeddah, Kingdom of Saudi Arabia. E-mail: ORCID ID: https://orcid.org/0000-0002-5298-7690
| | - Mohamad N. Khabaz
- From the Department of Pathology (Al-Maghrabi), Faculty of Medicine; from the Department of Pathology (Khabaz), Rabigh Faculty of Medicine, King Abdulaziz University, and from the Department of Pathology (Al-Maghrabi), King Faisal Specialist Hospital and Research Centre, Jeddah, Kingdom of Saudi Arabia.
| |
Collapse
|
3
|
Jankiewicz WK, Barnett SD, Stavniichuk A, Hwang SH, Hammock BD, Belayet JB, Khan AH, Imig JD. Dual sEH/COX-2 Inhibition Using PTUPB-A Promising Approach to Antiangiogenesis-Induced Nephrotoxicity. Front Pharmacol 2021; 12:744776. [PMID: 34955823 PMCID: PMC8695932 DOI: 10.3389/fphar.2021.744776] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Accepted: 10/21/2021] [Indexed: 01/11/2023] Open
Abstract
Kidney injury from antiangiogenic chemotherapy is a significant clinical challenge, and we currently lack the ability to effectively treat it with pharmacological agents. Thus, we set out to investigate whether simultaneous soluble epoxide hydrolase (sEH) and cyclooxygenase-2 (COX-2) inhibition using a dual sEH/COX-2 inhibitor PTUPB could be an effective strategy for treating antiangiogenic therapy-induced kidney damage. We used a multikinase inhibitor, sorafenib, which is known to cause serious renal side effects. The drug was administered to male Sprague-Dawley rats that were on a high-salt diet. Sorafenib was administered over the course of 56 days. The study included three experimental groups; 1) control group (naïve rats), 2) sorafenib group [rats treated with sorafenib only (20 mg/kg/day p.o.)], and 3) sorafenib + PTUPB group (rats treated with sorafenib only for the initial 28 days and subsequently coadministered PTUPB (10 mg/kg/day i.p.) from days 28 through 56). Blood pressure was measured every 2 weeks. After 28 days, sorafenib-treated rats developed hypertension (161 ± 4 mmHg). Over the remainder of the study, sorafenib treatment resulted in a further elevation in blood pressure through day 56 (200 ± 7 mmHg). PTUPB treatment attenuated the sorafenib-induced blood pressure elevation and by day 56, blood pressure was 159 ± 4 mmHg. Urine was collected every 2 weeks for biochemical analysis. After 28 days, sorafenib rats developed pronounced proteinuria (9.7 ± 0.2 P/C), which intensified significantly (35.8 ± 3.5 P/C) by the end of day 56 compared with control (2.6 ± 0.4 P/C). PTUPB mitigated sorafenib-induced proteinuria, and by day 56, it reduced proteinuria by 73%. Plasma and kidney tissues were collected on day 56. Kidney histopathology revealed intratubular cast formation, interstitial fibrosis, glomerular injury, and glomerular nephrin loss at day 56 in sorafenib-treated rats. PTUPB treatment reduced histological features by 30%-70% compared with the sorafenib-treated group and restored glomerular nephrin levels. Furthermore, PTUPB also acted on the glomerular permeability barrier by decreasing angiotensin-II-induced glomerular permeability to albumin. Finally, PTUPB improved in vitro the viability of human mesangial cells. Collectively, our data demonstrate the potential of using PTUPB or dual sEH/COX-2 inhibition as a therapeutic strategy against sorafenib-induced glomerular nephrotoxicity.
Collapse
Affiliation(s)
- Wojciech K. Jankiewicz
- Drug Discovery Center and Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Scott D. Barnett
- Drug Discovery Center and Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Anna Stavniichuk
- Drug Discovery Center and Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Sung Hee Hwang
- Department of Entomology and Nematology and Comprehensive Cancer Center, University of California, Davis, Davis, CA, United States
| | - Bruce D. Hammock
- Department of Entomology and Nematology and Comprehensive Cancer Center, University of California, Davis, Davis, CA, United States
| | - Jawad B. Belayet
- Department of Chemistry and Biochemistry, University of Wisconsin Milwaukee, Milwaukee, WI, United States
| | - A. H. Khan
- Drug Discovery Center and Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI, United States
| | - John D. Imig
- Drug Discovery Center and Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI, United States
| |
Collapse
|
4
|
Khan AH, Hwang SH, Barnett SD, Burkhan A, Jankiewicz WK, Hammock BD, Imig JD. Multitarget molecule, PTUPB, to treat diabetic nephropathy in rats. Br J Pharmacol 2021; 178:4468-4484. [PMID: 34255857 PMCID: PMC8863090 DOI: 10.1111/bph.15623] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Revised: 06/28/2021] [Accepted: 07/02/2021] [Indexed: 01/02/2023] Open
Abstract
BACKGROUND AND PURPOSE Diabetic nephropathy is a common complications related to high morbidity and mortality in type 2 diabetes. We investigated the action of the dual modulator, PTUPB, a soluble epoxide hydrolase and cyclooxygenase-2 inhibitor against diabetic nephropathy. EXPERIMENTAL APPROACH Sixteen-week-old type 2 diabetic and proteinuric obese ZSF1 rats were treated with vehicle, PTUPB or enalapril for 8 weeks. Measurements were made of epoxyeicosatrienoic acids, thromboxane B2 (TBX2 ) and prostaglandin E2 (PGE2 ) in the kidney of these and lean ZSF1 rats along with their blood pressure. KEY RESULT Obese ZSF1 rats were diabetic with fivefold higher fasting blood glucose levels and markedly higher HbA1c levels compared with lean ZSF1 rats. PTUPB nor enalapril reduced fasting blood glucose or HbA1c but alleviated the development of diabetic nephropathy. In PTUPB-treated obese ZSF1 rats, glomerular nephrin expression was preserved. Enalapril also alleviated diabetic nephropathy. Diabetic renal injury in obese ZSF1 rats was accompanied by renal inflammation with six to sevenfold higher urinary MCP-1 (CCR2) level and renal infiltration of CD-68 positive cells. PTUPB and enalapril significantly reduced urinary MCP-1 levels and renal mRNA expression of cytokines. Both PTUPB and enalapril lowered blood pressure. PTUPB but not enalapril decreased hyperlipidaemia and liver injury in obese ZSF1 rats. CONCLUSION AND IMPLICATIONS Overall, the dual modulator PTUPB does not treat hyperglycaemia but can effectively alleviate hypertension, diabetic nephropathy, hyperlipidaemia and liver injury in type 2 diabetic rats. Our data further demonstrate that the renal actions of PTUPB are comparable with a current standard diabetic nephropathy treatment.
Collapse
Affiliation(s)
- Abdul Hye Khan
- Drug Discovery Center and Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Sung Hee Hwang
- Department of Entomology and Nematology and Comprehensive Cancer Center, University of California, Davis, CA 95616, USA
| | - Scott D. Barnett
- Drug Discovery Center and Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Anna Burkhan
- Drug Discovery Center and Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Wojciech K. Jankiewicz
- Drug Discovery Center and Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Bruce D. Hammock
- Department of Entomology and Nematology and Comprehensive Cancer Center, University of California, Davis, CA 95616, USA
| | - John D. Imig
- Drug Discovery Center and Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| |
Collapse
|
5
|
Pibuel MA, Poodts D, Díaz M, Molinari YA, Franco PG, Hajos SE, Lompardía SL. Antitumor effect of 4MU on glioblastoma cells is mediated by senescence induction and CD44, RHAMM and p-ERK modulation. Cell Death Discov 2021; 7:280. [PMID: 34628469 PMCID: PMC8502173 DOI: 10.1038/s41420-021-00672-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Revised: 09/08/2021] [Accepted: 09/23/2021] [Indexed: 01/10/2023] Open
Abstract
The extracellular matrix plays a key role in cancer progression. Hyaluronan, the main glycosaminoglycan of the extracellular matrix, has been related to several tumor processes. Hyaluronan acts through the interaction with cell membrane receptors as CD44 and RHAMM and triggers signaling pathways as MEK/ERK. 4-methylumbelliferone (4MU), a well-known hyaluronan synthesis inhibitor, is a promising alternative for cancer therapy. 4MU is a coumarin derivative without adverse effects that has been studied in several tumors. However, little is known about its use in glioblastoma (GBM), the most malignant primary brain tumor in adults. Glioblastoma is characterized by fast growth, migration and tissue invasiveness, and a poor median survival of the patients after treatment. Several reports linked glioblastoma progression with HA levels and even with CD44 and RHAMM expression, as well as MEK/ERK activation. Previously, we showed on a murine GBM cell line that HA enhances GBM migration, while 4MU markedly inhibits it. In this work we showed for the first time, that 4MU decreases cell migration and induces senescence in U251 and LN229 human GBM cell lines. Furthermore, we observed that HA promotes GBM cell migration on both cell lines and that such effects depend on CD44 and RHAMM, as well as MEK/ERK signaling pathway. Interestingly, we observed that the exogenous HA failed to counteract the effects of 4MU, indicating that 4MU effects are independent of HA synthesis inhibition. We found that 4MU decreases total CD44 and RHAMM membrane expression, which could explain the effect of 4MU on cell migration. Furthermore, we observed that 4MU increases the levels of RHAMM inside the cell while decreases the nucleus/cytoplasm relation of p-ERK, associated with 4MU effects on cell proliferation and senescence induction. Overall, 4MU should be considered as a promising therapeutic alternative to improve the outcome of patients with GBM.
Collapse
Grants
- PIP N°0289 Consejo Nacional de Investigaciones Científicas y Técnicas (National Scientific and Technical Research Council)
- PIP N°053 Consejo Nacional de Investigaciones Científicas y Técnicas (National Scientific and Technical Research Council)
- PIP N°053 Consejo Nacional de Investigaciones Científicas y Técnicas (National Scientific and Technical Research Council)
- PIP N°0289 Consejo Nacional de Investigaciones Científicas y Técnicas (National Scientific and Technical Research Council)
- PIP N°053 Consejo Nacional de Investigaciones Científicas y Técnicas (National Scientific and Technical Research Council)
- PIP N°0289 Consejo Nacional de Investigaciones Científicas y Técnicas (National Scientific and Technical Research Council)
- PIP N°053 Consejo Nacional de Investigaciones Científicas y Técnicas (National Scientific and Technical Research Council)
- PIP N°0289 Consejo Nacional de Investigaciones Científicas y Técnicas (National Scientific and Technical Research Council)
- PIP N°053 Consejo Nacional de Investigaciones Científicas y Técnicas (National Scientific and Technical Research Council)
- PIP N°0289 Consejo Nacional de Investigaciones Científicas y Técnicas (National Scientific and Technical Research Council)
- PIP N°0289 Consejo Nacional de Investigaciones Científicas y Técnicas (National Scientific and Technical Research Council)
- PIP N°053 Consejo Nacional de Investigaciones Científicas y Técnicas (National Scientific and Technical Research Council)
- PIP N°0289 Consejo Nacional de Investigaciones Científicas y Técnicas (National Scientific and Technical Research Council)
- PIP N°053 Consejo Nacional de Investigaciones Científicas y Técnicas (National Scientific and Technical Research Council)
- UBACYT 20020170100454BA Universidad de Buenos Aires (University of Buenos Aires)
- UBACYT 20020170100454BA Universidad de Buenos Aires (University of Buenos Aires)
- UBACYT 20020170100454BA Universidad de Buenos Aires (University of Buenos Aires)
- UBACYT 20020170100454BA Universidad de Buenos Aires (University of Buenos Aires)
- UBACYT 20020170100454BA Universidad de Buenos Aires (University of Buenos Aires)
- UBACYT 20020170100454BA Universidad de Buenos Aires (University of Buenos Aires)
- PICT-2017- 2971 Ministry of Science, Technology and Productive Innovation, Argentina | Agencia Nacional de Promoción Científica y Tecnológica (National Agency for Science and Technology, Argentina)
- PICT-2017- 2971 Ministry of Science, Technology and Productive Innovation, Argentina | Agencia Nacional de Promoción Científica y Tecnológica (National Agency for Science and Technology, Argentina)
- PICT-2017- 2971 Ministry of Science, Technology and Productive Innovation, Argentina | Agencia Nacional de Promoción Científica y Tecnológica (National Agency for Science and Technology, Argentina)
- PICT-2017- 2971 Ministry of Science, Technology and Productive Innovation, Argentina | Agencia Nacional de Promoción Científica y Tecnológica (National Agency for Science and Technology, Argentina)
- PICT-2017- 2971 Ministry of Science, Technology and Productive Innovation, Argentina | Agencia Nacional de Promoción Científica y Tecnológica (National Agency for Science and Technology, Argentina)
- PICT-2017- 2971 Ministry of Science, Technology and Productive Innovation, Argentina | Agencia Nacional de Promoción Científica y Tecnológica (National Agency for Science and Technology, Argentina)
- PICT-2017- 2971 Ministry of Science, Technology and Productive Innovation, Argentina | Agencia Nacional de Promoción Científica y Tecnológica (National Agency for Science and Technology, Argentina)
Collapse
Affiliation(s)
- Matías Arturo Pibuel
- Instituto de Estudios de la Inmunidad Humoral (IDEHU)- CONICET; Departamento de Microbiología, Inmunología y Biotecnología, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Capital Federal, Argentina.
| | - Daniela Poodts
- Instituto de Estudios de la Inmunidad Humoral (IDEHU)- CONICET; Departamento de Microbiología, Inmunología y Biotecnología, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Capital Federal, Argentina
| | - Mariángeles Díaz
- Instituto de Estudios de la Inmunidad Humoral (IDEHU)- CONICET, Universidad de Buenos Aires, Capital Federal, Argentina
| | - Yamila Azul Molinari
- Instituto de Química y Fisicoquímica Biológicas (IQUIFIB)-CONICET; Departamento de Química Biológica, Cátedra de Química Biológica Patológica, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Capital Federal, Argentina
| | - Paula Gabriela Franco
- Instituto de Química y Fisicoquímica Biológicas (IQUIFIB)-CONICET; Departamento de Química Biológica, Cátedra de Química Biológica Patológica, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Capital Federal, Argentina
| | - Silvia Elvira Hajos
- Instituto de Estudios de la Inmunidad Humoral (IDEHU)- CONICET; Departamento de Microbiología, Inmunología y Biotecnología, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Capital Federal, Argentina
| | - Silvina Laura Lompardía
- Instituto de Estudios de la Inmunidad Humoral (IDEHU)- CONICET; Departamento de Microbiología, Inmunología y Biotecnología, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Capital Federal, Argentina
| |
Collapse
|
6
|
Fishbein A, Hammock BD, Serhan CN, Panigrahy D. Carcinogenesis: Failure of resolution of inflammation? Pharmacol Ther 2021; 218:107670. [PMID: 32891711 PMCID: PMC7470770 DOI: 10.1016/j.pharmthera.2020.107670] [Citation(s) in RCA: 101] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/19/2020] [Indexed: 02/06/2023]
Abstract
Inflammation in the tumor microenvironment is a hallmark of cancer and is recognized as a key characteristic of carcinogens. However, the failure of resolution of inflammation in cancer is only recently being understood. Products of arachidonic acid and related fatty acid metabolism called eicosanoids, including prostaglandins, leukotrienes, lipoxins, and epoxyeicosanoids, critically regulate inflammation, as well as its resolution. The resolution of inflammation is now appreciated to be an active biochemical process regulated by endogenous specialized pro-resolving lipid autacoid mediators which combat infections and stimulate tissue repair/regeneration. Environmental and chemical human carcinogens, including aflatoxins, asbestos, nitrosamines, alcohol, and tobacco, induce tumor-promoting inflammation and can disrupt the resolution of inflammation contributing to a devastating global cancer burden. While mechanisms of carcinogenesis have focused on genotoxic activity to induce mutations, nongenotoxic mechanisms such as inflammation and oxidative stress promote genotoxicity, proliferation, and mutations. Moreover, carcinogens initiate oxidative stress to synergize with inflammation and DNA damage to fuel a vicious feedback loop of cell death, tissue damage, and carcinogenesis. In contrast, stimulation of resolution of inflammation may prevent carcinogenesis by clearance of cellular debris via macrophage phagocytosis and inhibition of an eicosanoid/cytokine storm of pro-inflammatory mediators. Controlling the host inflammatory response and its resolution in carcinogen-induced cancers will be critical to reducing carcinogen-induced morbidity and mortality. Here we review the recent evidence that stimulation of resolution of inflammation, including pro-resolution lipid mediators and soluble epoxide hydrolase inhibitors, may be a new chemopreventive approach to prevent carcinogen-induced cancer that should be evaluated in humans.
Collapse
Affiliation(s)
- Anna Fishbein
- Center for Vascular Biology Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA; Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA.
| | - Bruce D. Hammock
- Department of Entomology and Nematology, and UCD Comprehensive Cancer Center, University of California, Davis, CA 95616, USA
| | - Charles N. Serhan
- Center for Experimental Therapeutics and Reperfusion Injury, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Dipak Panigrahy
- Center for Vascular Biology Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA,Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| |
Collapse
|
7
|
Pibuel MA, Poodts D, Díaz M, Hajos SE, Lompardía SL. The scrambled story between hyaluronan and glioblastoma. J Biol Chem 2021; 296:100549. [PMID: 33744285 PMCID: PMC8050860 DOI: 10.1016/j.jbc.2021.100549] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Revised: 03/12/2021] [Accepted: 03/16/2021] [Indexed: 02/06/2023] Open
Abstract
Advances in cancer biology are revealing the importance of the cancer cell microenvironment on tumorigenesis and cancer progression. Hyaluronan (HA), the main glycosaminoglycan in the extracellular matrix, has been associated with the progression of glioblastoma (GBM), the most frequent and lethal primary tumor in the central nervous system, for several decades. However, the mechanisms by which HA impacts GBM properties and processes have been difficult to elucidate. In this review, we provide a comprehensive assessment of the current knowledge on HA's effects on GBM biology, introducing its primary receptors CD44 and RHAMM and the plethora of relevant downstream signaling pathways that can scramble efforts to directly link HA activity to biological outcomes. We consider the complexities of studying an extracellular polymer and the different strategies used to try to capture its function, including 2D and 3D in vitro studies, patient samples, and in vivo models. Given that HA affects not only migration and invasion, but also cell proliferation, adherence, and chemoresistance, we highlight the potential role of HA as a therapeutic target. Finally, we review the different existing approaches to diminish its protumor effects, such as the use of 4-methylumbelliferone, HA oligomers, and hyaluronidases and encourage further research along these lines in order to improve the survival and quality of life of GBM patients.
Collapse
Affiliation(s)
- Matías Arturo Pibuel
- Departamento de Microbiología, Inmunología, Biotecnología y Genética, Facultad de Farmacia y Bioquímica, Instituto de Estudios de la Inmunidad Humoral (IDEHU)-CONICET, Universidad de Buenos Aires, Capital Federal, Argentina.
| | - Daniela Poodts
- Departamento de Microbiología, Inmunología, Biotecnología y Genética, Facultad de Farmacia y Bioquímica, Instituto de Estudios de la Inmunidad Humoral (IDEHU)-CONICET, Universidad de Buenos Aires, Capital Federal, Argentina
| | - Mariángeles Díaz
- Instituto de Estudios de la Inmunidad Humoral (IDEHU)-CONICET, Universidad de Buenos Aires, Capital Federal, Argentina
| | - Silvia Elvira Hajos
- Departamento de Microbiología, Inmunología, Biotecnología y Genética, Facultad de Farmacia y Bioquímica, Instituto de Estudios de la Inmunidad Humoral (IDEHU)-CONICET, Universidad de Buenos Aires, Capital Federal, Argentina
| | - Silvina Laura Lompardía
- Departamento de Microbiología, Inmunología, Biotecnología y Genética, Facultad de Farmacia y Bioquímica, Instituto de Estudios de la Inmunidad Humoral (IDEHU)-CONICET, Universidad de Buenos Aires, Capital Federal, Argentina.
| |
Collapse
|
8
|
Sun CP, Zhang XY, Morisseau C, Hwang SH, Zhang ZJ, Hammock BD, Ma XC. Discovery of Soluble Epoxide Hydrolase Inhibitors from Chemical Synthesis and Natural Products. J Med Chem 2020; 64:184-215. [PMID: 33369424 DOI: 10.1021/acs.jmedchem.0c01507] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Soluble epoxide hydrolase (sEH) is an α/β hydrolase fold protein and widely distributed in numerous organs including the liver, kidney, and brain. The inhibition of sEH can effectively maintain endogenous epoxyeicosatrienoic acids (EETs) levels and reduce dihydroxyeicosatrienoic acids (DHETs) levels, resulting in therapeutic potentials for cardiovascular, central nervous system, and metabolic diseases. Therefore, since the beginning of this century, the development of sEH inhibitors is a hot research topic. A variety of potent sEH inhibitors have been developed by chemical synthesis or isolated from natural sources. In this review, we mainly summarized the interconnected aspects of sEH with cardiovascular, central nervous system, and metabolic diseases and then focus on representative inhibitors, which would provide some useful guidance for the future development of potential sEH inhibitors.
Collapse
Affiliation(s)
- Cheng-Peng Sun
- Dalian Key Laboratory of Metabolic Target Characterization and Traditional Chinese Medicine Intervention, College (Institute) of Integrative Medicine, College of Pharmacy, Dalian Medical University, Dalian 116044, People's Republic of China
| | - Xin-Yue Zhang
- Dalian Key Laboratory of Metabolic Target Characterization and Traditional Chinese Medicine Intervention, College (Institute) of Integrative Medicine, College of Pharmacy, Dalian Medical University, Dalian 116044, People's Republic of China
| | - Christophe Morisseau
- Department of Entomology and Nematology, UC Davis Comprehensive Cancer Center, University of California, Davis, California 95616, United States
| | - Sung Hee Hwang
- Department of Entomology and Nematology, UC Davis Comprehensive Cancer Center, University of California, Davis, California 95616, United States
| | - Zhan-Jun Zhang
- State Key Laboratory of Cognitive Neuroscience and Learning, Beijing Normal University, Beijing 100875, People's Republic of China
| | - Bruce D Hammock
- Department of Entomology and Nematology, UC Davis Comprehensive Cancer Center, University of California, Davis, California 95616, United States
| | - Xiao-Chi Ma
- Dalian Key Laboratory of Metabolic Target Characterization and Traditional Chinese Medicine Intervention, College (Institute) of Integrative Medicine, College of Pharmacy, Dalian Medical University, Dalian 116044, People's Republic of China.,College of Pharmacy, School of Medicine, Hangzhou Normal University, Hangzhou 311121, People's Republic of China
| |
Collapse
|
9
|
Imig JD, Jankiewicz WK, Khan AH. Epoxy Fatty Acids: From Salt Regulation to Kidney and Cardiovascular Therapeutics: 2019 Lewis K. Dahl Memorial Lecture. Hypertension 2020; 76:3-15. [PMID: 32475311 PMCID: PMC7448548 DOI: 10.1161/hypertensionaha.120.13898] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Epoxyeicosatrienoic acids (EETs) are epoxy fatty acids that have biological actions that are essential for maintaining water and electrolyte homeostasis. An inability to increase EETs in response to a high-salt diet results in salt-sensitive hypertension. Vasodilation, inhibition of epithelial sodium channel, and inhibition of inflammation are the major EET actions that are beneficial to the heart, resistance arteries, and kidneys. Genetic and pharmacological means to elevate EETs demonstrated antihypertensive, anti-inflammatory, and organ protective actions. Therapeutic approaches to increase EETs were then developed for cardiovascular diseases. sEH (soluble epoxide hydrolase) inhibitors were developed and progressed to clinical trials for hypertension, diabetes mellitus, and other diseases. EET analogs were another therapeutic approach taken and these drugs are entering the early phases of clinical development. Even with the promise for these therapeutic approaches, there are still several challenges, unexplored areas, and opportunities for epoxy fatty acids.
Collapse
Affiliation(s)
- John D Imig
- From the Department of Pharmacology and Toxicology, Cardiovascular Center, Medical College of Wisconsin, Milwaukee
| | - Wojciech K Jankiewicz
- From the Department of Pharmacology and Toxicology, Cardiovascular Center, Medical College of Wisconsin, Milwaukee
| | - Abdul H Khan
- From the Department of Pharmacology and Toxicology, Cardiovascular Center, Medical College of Wisconsin, Milwaukee
| |
Collapse
|
10
|
Yang HH, Duan JX, Liu SK, Xiong JB, Guan XX, Zhong WJ, Sun CC, Zhang CY, Luo XQ, Zhang YF, Chen P, Hammock BD, Hwang SH, Jiang JX, Zhou Y, Guan CX. A COX-2/sEH dual inhibitor PTUPB alleviates lipopolysaccharide-induced acute lung injury in mice by inhibiting NLRP3 inflammasome activation. Theranostics 2020; 10:4749-4761. [PMID: 32308747 PMCID: PMC7163435 DOI: 10.7150/thno.43108] [Citation(s) in RCA: 141] [Impact Index Per Article: 28.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Accepted: 03/08/2020] [Indexed: 01/11/2023] Open
Abstract
Rationale: Dysregulation of arachidonic acid (ARA) metabolism results in inflammation; however, its role in acute lung injury (ALI) remains elusive. In this study, we addressed the role of dysregulated ARA metabolism in cytochromes P450 (CYPs) /cyclooxygenase-2 (COX-2) pathways in the pathogenesis of lipopolysaccharide (LPS)-induced ALI in mice. Methods: The metabolism of CYPs/COX-2-derived ARA in the lungs of LPS-induced ALI was investigated in C57BL/6 mice. The COX-2/sEH dual inhibitor PTUPB was used to establish the function of CYPs/COX-2 dysregulation in ALI. Primary murine macrophages were used to evaluate the underlying mechanism of PTUPB involved in the activation of NLRP3 inflammasome in vitro. Results: Dysregulation of CYPs/COX-2 metabolism of ARA occurred in the lungs and in primary macrophages under the LPS challenge. Decrease mRNA expression of Cyp2j9, Cyp2j6, and Cyp2j5 was observed, which metabolize ARA into epoxyeicosatrienoic acids (EETs). The expressions of COX-2 and soluble epoxide hydrolase (sEH), on the other hand, was significantly upregulated. Pre-treatment with the dual COX-2 and sEH inhibitor, PTUPB, attenuated the pathological injury of lung tissues and reduced the infiltration of inflammatory cells. Furthermore, PTUPB decreased the pro-inflammatory factors, oxidative stress, and activation of NACHT, LRR, and PYD domains-containing protein 3 (NLRP3) inflammasome in LPS-induced ALI mice. PTUPB pre-treatment remarkably reduced the activation of macrophages and NLRP3 inflammasome in vitro. Significantly, both preventive and therapeutic treatment with PTUPB improved the survival rate of mice receiving a lethal dose of LPS. Conclusion: The dysregulation of CYPs/COX-2 metabolized ARA contributes to the uncontrolled inflammatory response in ALI. The dual COX-2 and sEH inhibitor PTUPB exerts anti-inflammatory effects in treating ALI by inhibiting the NLRP3 inflammasome activation.
Collapse
Affiliation(s)
- Hui-Hui Yang
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, Hunan 410078, China
| | - Jia-Xi Duan
- Department of Pulmonary and Critical Care Medicine, the Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
- Research Unit of Respiratory Disease, Central South University, Changsha, Hunan 410011, China
- Hunan Diagnosis and Treatment Center of Respiratory Disease, Central South University, Changsha, Hunan 410011, China
| | - Shao-Kun Liu
- Department of Pulmonary and Critical Care Medicine, the Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
- Research Unit of Respiratory Disease, Central South University, Changsha, Hunan 410011, China
- Hunan Diagnosis and Treatment Center of Respiratory Disease, Central South University, Changsha, Hunan 410011, China
| | - Jian-Bing Xiong
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, Hunan 410078, China
| | - Xin-Xin Guan
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, Hunan 410078, China
| | - Wen-Jing Zhong
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, Hunan 410078, China
| | - Chen-Chen Sun
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, Hunan 410078, China
| | - Chen-Yu Zhang
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, Hunan 410078, China
| | - Xiao-Qin Luo
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, Hunan 410078, China
| | - Yan-Feng Zhang
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, Hunan 410078, China
| | - Ping Chen
- Department of Pulmonary and Critical Care Medicine, the Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
- Research Unit of Respiratory Disease, Central South University, Changsha, Hunan 410011, China
- Hunan Diagnosis and Treatment Center of Respiratory Disease, Central South University, Changsha, Hunan 410011, China
| | - Bruce D. Hammock
- Department of Entomology and Nematology and UC Davis Comprehensive Cancer Center, University of California, Davis, One Shields Avenue, Davis, CA 95616, USA
| | - Sung Hee Hwang
- Department of Entomology and Nematology and UC Davis Comprehensive Cancer Center, University of California, Davis, One Shields Avenue, Davis, CA 95616, USA
| | - Jian-Xin Jiang
- State Key Laboratory of Trauma, Burns, and Combined Injury, Army Medical University, Chongqing, 400038, China
| | - Yong Zhou
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, Hunan 410078, China
| | - Cha-Xiang Guan
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, Hunan 410078, China
| |
Collapse
|
11
|
Zhang YF, Sun CC, Duan JX, Yang HH, Zhang CY, Xiong JB, Zhong WJ, Zu C, Guan XX, Jiang HL, Hammock BD, Hwang SH, Zhou Y, Guan CX. A COX-2/sEH dual inhibitor PTUPB ameliorates cecal ligation and puncture-induced sepsis in mice via anti-inflammation and anti-oxidative stress. Biomed Pharmacother 2020; 126:109907. [PMID: 32114358 DOI: 10.1016/j.biopha.2020.109907] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Revised: 12/31/2019] [Accepted: 12/31/2019] [Indexed: 12/14/2022] Open
Abstract
Arachidonic acid can be metabolized to prostaglandins and epoxyeicosatrienoic acids (EETs) by cyclooxygenase-2 (COX-2) and cytochrome P450 (CYP), respectively. While protective EETs are degraded by soluble epoxide hydrolase (sEH) very fast. We have reported that dual inhibition of COX-2 and sEH with specific inhibitor PTUPB shows anti-pulmonary fibrosis and renal protection. However, the effect of PTUPB on cecal ligation and puncture (CLP)-induced sepsis remains unclear. The current study aimed to investigate the protective effects of PTUPB against CLP-induced sepsis in mice and the underlying mechanisms. We found that COX-2 expressions were increased, while CYPs expressions were decreased in the liver, lung, and kidney of mice undergone CLP. PTUPB treatment significantly improved the survival rate, reduced the clinical scores and systemic inflammatory response, alleviated liver and kidney dysfunction, and ameliorated the multiple-organ injury of the mice with sepsis. Besides, PTUPB treatment reduced the expression of hypoxia-inducible factor-1α in the liver, lung, and kidney of septic mice. Importantly, we found that PTUPB treatment suppressed the activation of NLRP3 inflammasome in the liver and lung of septic mice. Meanwhile, we found that PTUPB attenuated the oxidative stress, which contributed to the activation of NLRP3 inflammasome. Altogether, our data, for the first time, demonstrate that dual inhibition of COX-2 and sEH with PTUPB ameliorates the multiple organ dysfunction in septic mice.
Collapse
Affiliation(s)
- Yan-Feng Zhang
- Department of Cardiovascular Surgery, Xiangya Hospital, Central South University, Changsha, Hunan 410078, China
| | - Chen-Chen Sun
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, Hunan 410078, China
| | - Jia-Xi Duan
- Department of Pulmonary and Critical Care Medicine, the Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China; Research Unit of Respiratory Disease, Central South University, Changsha, Hunan 410011, China; Hunan Diagnosis and Treatment Center of Respiratory Disease, Central South University, Changsha, Hunan 410011, China
| | - Hui-Hui Yang
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, Hunan 410078, China
| | - Chen-Yu Zhang
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, Hunan 410078, China
| | - Jian-Bing Xiong
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, Hunan 410078, China
| | - Wen-Jing Zhong
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, Hunan 410078, China
| | - Cheng Zu
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, Hunan 410078, China
| | - Xin-Xin Guan
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, Hunan 410078, China
| | - Hui-Ling Jiang
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, Hunan 410078, China
| | - Bruce D Hammock
- Department of Entomology and Nematology and UC Davis Comprehensive Cancer Center, University of California, Davis, CA 95616, USA
| | - Sung Hee Hwang
- Department of Entomology and Nematology and UC Davis Comprehensive Cancer Center, University of California, Davis, CA 95616, USA
| | - Yong Zhou
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, Hunan 410078, China.
| | - Cha-Xiang Guan
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, Hunan 410078, China.
| |
Collapse
|
12
|
Zhang CY, Duan JX, Yang HH, Sun CC, Zhong WJ, Tao JH, Guan XX, Jiang HL, Hammock BD, Hwang SH, Zhou Y, Guan CX. COX-2/sEH dual inhibitor PTUPB alleviates bleomycin-induced pulmonary fibrosis in mice via inhibiting senescence. FEBS J 2019; 287:1666-1680. [PMID: 31646730 DOI: 10.1111/febs.15105] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2019] [Revised: 08/30/2019] [Accepted: 10/23/2019] [Indexed: 02/02/2023]
Abstract
Pulmonary fibrosis (PF) is a senescence-associated disease with poor prognosis. Currently, there is no effective therapeutic strategy for preventing and treating the disease process. Mounting evidence suggests that arachidonic acid (ARA) metabolites are involved in the pathogenesis of various fibrosis. However, the relationship between the metabolism of ARA and PF is still elusive. In this study, we observed a disorder in the cyclooxygenase-2/cytochrome P450 (COX-2/CYP) metabolism of ARA in the lungs of PF mice induced by bleomycin (BLM). Therefore, we aimed to explore the role of COX-2/CYP-derived ARA metabolic disorders in PF. PTUPB, a dual COX-2 and soluble epoxide hydrolase (sEH) inhibitor, was used to restore the balance of COX-2/CYP metabolism. sEH is an enzyme hydrolyzing epoxyeicosatrienoic acids derived from ARA by CYP. We found that PTUPB alleviated the pathological changes in lung tissue and collagen deposition, as well as reduced senescence marker molecules (p16Ink4a and p53-p21Waf1/Cip1 ) in the lungs of mice treated by BLM. In vitro, we found that PTUPB pretreatment remarkably reduced the expression of senescence-related molecules in the alveolar epithelial cells (AECs) induced by BLM. In conclusion, our study supports the notion that the COX-2/CYP-derived ARA metabolic disorders may be a potential therapeutic target for PF via inhibiting the cellular senescence in AECs.
Collapse
Affiliation(s)
- Chen-Yu Zhang
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, China
| | - Jia-Xi Duan
- Department of Pulmonary and Critical Care Medicine, The Second Xiangya Hospital, Central South University, Changsha, China.,Research Unit of Respiratory Disease, Central South University, Changsha, China.,Hunan Diagnosis and Treatment Center of Respiratory Disease, Central South University, Changsha, Hunan, China
| | - Hui-Hui Yang
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, China
| | - Chen-Chen Sun
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, China
| | - Wen-Jing Zhong
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, China
| | - Jia-Hao Tao
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, China
| | - Xin-Xin Guan
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, China
| | - Hui-Ling Jiang
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, China
| | - Bruce D Hammock
- Department of Entomology and Nematology, UC Davis Comprehensive Cancer Center, University of California, Davis, CA, USA
| | - Sung Hee Hwang
- Department of Entomology and Nematology, UC Davis Comprehensive Cancer Center, University of California, Davis, CA, USA
| | - Yong Zhou
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, China
| | - Cha-Xiang Guan
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, China
| |
Collapse
|
13
|
Rand AA, Rajamani A, Kodani SD, Harris TR, Schlatt L, Barnych B, Passerini AG, Hammock BD. Epoxyeicosatrienoic acid (EET)-stimulated angiogenesis is mediated by epoxy hydroxyeicosatrienoic acids (EHETs) formed from COX-2. J Lipid Res 2019; 60:1996-2005. [PMID: 31641036 DOI: 10.1194/jlr.m094219] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Revised: 09/27/2019] [Indexed: 12/19/2022] Open
Abstract
Epoxyeicosatrienoic acids (EETs) are formed from the metabolism of arachidonic acid by cytochrome P450s. EETs promote angiogenesis linked to tumor growth in various cancer models that is attenuated in vivo by cyclooxygenase 2 (COX-2) inhibitors. This study further defines a role for COX-2 in mediating endothelial EET metabolism promoting angiogenesis. Using human aortic endothelial cells (HAECs), we quantified 8,9-EET-induced tube formation and cell migration as indicators of angiogenic potential in the presence and absence of a COX-2 inducer [phorbol 12,13-dibutyrate (PDBu)]. The angiogenic response to 8,9-EET in the presence of PDBu was 3-fold that elicited by 8,9-EET stabilized with a soluble epoxide hydrolase inhibitor (t-TUCB). Contributing to this response was the COX-2 metabolite of 8,9-EET, the 11-hydroxy-8,9-EET (8,9,11-EHET), which exogenously enhanced angiogenic responses in HAECs at levels comparable to those elicited by vascular endothelial growth factor (VEGF). In contrast, the 15-hydroxy-8,9-EET isomer was also formed but inactive. The 8,9,11-EHET also promoted expression of the VEGF family of tyrosine kinase receptors. These results indicate that 8,9-EET-stimulated angiogenesis is enhanced by COX-2 metabolism in the endothelium through the formation of 8,9,11-EHET. This alternative pathway for the metabolism of 8,9-EET may be particularly important in regulating angiogenesis under circumstances in which COX-2 is induced, such as in cancer tumor growth and inflammation.
Collapse
Affiliation(s)
- Amy A Rand
- Department of Chemistry, Carleton University, Ottawa, ON, Canada.,Department of Entomology and Nematology, University of California, Davis, Davis, CA.,UC Davis Comprehensive Cancer Center, University of California, Davis, Davis, CA
| | - Anita Rajamani
- Department of Biomedical Engineering, University of California, Davis, Davis, CA
| | - Sean D Kodani
- Department of Entomology and Nematology, University of California, Davis, Davis, CA.,UC Davis Comprehensive Cancer Center, University of California, Davis, Davis, CA
| | - Todd R Harris
- Department of Chemistry, Carleton University, Ottawa, ON, Canada.,Department of Entomology and Nematology, University of California, Davis, Davis, CA.,UC Davis Comprehensive Cancer Center, University of California, Davis, Davis, CA
| | - Lukas Schlatt
- Department of Entomology and Nematology, University of California, Davis, Davis, CA.,UC Davis Comprehensive Cancer Center, University of California, Davis, Davis, CA
| | - Bodgan Barnych
- Department of Entomology and Nematology, University of California, Davis, Davis, CA.,UC Davis Comprehensive Cancer Center, University of California, Davis, Davis, CA
| | - Anthony G Passerini
- Department of Biomedical Engineering, University of California, Davis, Davis, CA
| | - Bruce D Hammock
- Department of Entomology and Nematology, University of California, Davis, Davis, CA .,UC Davis Comprehensive Cancer Center, University of California, Davis, Davis, CA
| |
Collapse
|
14
|
Dileepan M, Rastle-Simpson S, Greenberg Y, Wijesinghe DS, Kumar NG, Yang J, Hwang SH, Hammock BD, Sriramarao P, Rao SP. Effect Of Dual sEH/COX-2 Inhibition on Allergen-Induced Airway Inflammation. Front Pharmacol 2019; 10:1118. [PMID: 31611798 PMCID: PMC6777353 DOI: 10.3389/fphar.2019.01118] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Accepted: 08/30/2019] [Indexed: 12/20/2022] Open
Abstract
Arachidonic acid metabolites resulting from the cyclooxygenase (COX), lipoxygenase, and cytochrome P450 oxidase enzymatic pathways play pro- and anti-inflammatory roles in allergic airway inflammation (AAI) and asthma. Expression of COX-2 and soluble epoxide hydrolase (sEH) are elevated in allergic airways and their enzymatic products (e.g., prostaglandins and diols of epoxyeicosatrienoic acids, respectively) have been shown to participate in the pathogenesis of AAI. Here, we evaluated the outcome of inhibiting the COX-2 and sEH enzymatic pathways with a novel dual inhibitor, PTUPB, in A. alternata-induced AAI. Allergen-challenged mice were administered with 10 or 30 mg/kg of PTUPB, celecoxib (selective COX-2 inhibitor), t-TUCB (selective sEH inhibitor) or vehicle daily by gavage and evaluated for various features of AAI. PTUPB and t-TUCB at 30 mg/kg, but not celecoxib, inhibited eosinophilic infiltration and significantly increased levels of anti-inflammatory EETs in the lung tissue of allergen-challenged mice. t-TUCB significantly inhibited allergen-induced IL-4 and IL-13, while a less pronounced reduction was noted with PTUPB and celecoxib. Additionally, t-TUCB markedly inhibited eotaxin-2, an eosinophil-specific chemokine, which was only marginally reduced by PTUPB and remained elevated in celecoxib-treated mice. PTUPB or t-TUCB administration reversed allergen-induced reduction in levels of various lipid mediators in the lungs, with only a minimal effect noted with celecoxib. Despite the anti-inflammatory effects, PTUPB or t-TUCB did not reduce allergen-induced airway hyperresponsiveness (AHR). However, development of structural changes in the allergic airways, such as mucus hypersecretion and smooth muscle hypertrophy, was significantly inhibited by both inhibitors. Celecoxib, on the other hand, inhibited only airway smooth muscle hypertrophy, but not mucus hypersecretion. In conclusion, dual inhibition of COX-2 and sEH offers no additional advantage relative to sEH inhibition alone in attenuating various features associated with A. alternata-induced AAI, while COX-2 inhibition exerts only moderate or no effect on several of these features. Dual sEH/COX-2 inhibition may be useful in treating conditions where eosinophilic inflammation co-exists with pain-associated inflammation.
Collapse
Affiliation(s)
- Mythili Dileepan
- Department of Veterinary & Biomedical Sciences, University of Minnesota, St. Paul, MN, United States
| | - Stephanie Rastle-Simpson
- Department of Veterinary & Biomedical Sciences, University of Minnesota, St. Paul, MN, United States
| | - Yana Greenberg
- Department of Veterinary & Biomedical Sciences, University of Minnesota, St. Paul, MN, United States
| | - Dayanjan S Wijesinghe
- Department of Pharmacotherapy and Outcomes Sciences, School of Pharmacy, Virginia Commonwealth University, Richmond, VA, United States
| | - Naren Gajenthra Kumar
- Department of Pharmacotherapy and Outcomes Sciences, School of Pharmacy, Virginia Commonwealth University, Richmond, VA, United States
| | - Jun Yang
- Department of Entomology, Nematology and Comprehensive Cancer Center, University of California, Davis, CA, United States
| | - Sung Hee Hwang
- Department of Entomology, Nematology and Comprehensive Cancer Center, University of California, Davis, CA, United States
| | - Bruce D Hammock
- Department of Entomology, Nematology and Comprehensive Cancer Center, University of California, Davis, CA, United States
| | - P Sriramarao
- Department of Veterinary & Biomedical Sciences, University of Minnesota, St. Paul, MN, United States
| | - Savita P Rao
- Department of Veterinary & Biomedical Sciences, University of Minnesota, St. Paul, MN, United States
| |
Collapse
|
15
|
Jones RD, Liao J, Tong X, Xu D, Sun L, Li H, Yang GY. Epoxy-Oxylipins and Soluble Epoxide Hydrolase Metabolic Pathway as Targets for NSAID-Induced Gastroenteropathy and Inflammation-Associated Carcinogenesis. Front Pharmacol 2019; 10:731. [PMID: 31293429 PMCID: PMC6603234 DOI: 10.3389/fphar.2019.00731] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Accepted: 06/05/2019] [Indexed: 12/14/2022] Open
Abstract
Polyunsaturated fatty acids (PUFAs) including epoxide-modified ω-3 and ω-6 fatty acids are made via oxidation to create highly polarized carbon-oxygen bonds crucial to their function as signaling molecules. A critical PUFA, arachidonic acid (ARA), is metabolized to a diverse set of lipids signaling molecules through cyclooxygenase (COX), lipoxygenase (LOX), cytochrome P450 epoxygenase, or cytochrome P450 hydroxylase; however, the majority of ARA is metabolized into anti-inflammatory epoxides via cytochrome P450 enzymes. These short-lived epoxide lipids are rapidly metabolized or inactivated by the soluble epoxide hydrolase (sEH) into diol-containing products. sEH inhibition or knockout has been a practical approach to study the biology of the epoxide lipids, and has been shown to effectively treat inflammatory conditions in the preclinical models including gastrointestinal ulcers and colitis by shifting oxylipins to epoxide profiles, inhibiting inflammatory cell infiltration and activation, and enhancing epithelial cell defense via increased mucin production, thus providing further evidence for the role of sEH as a pro-inflammatory protein. Non-steroidal anti-inflammatory drugs (NSAIDs) with COX-inhibitor activity are among the most commonly used analgesics and have demonstrated applications in the management of cardiovascular disease and intriguingly cancer. Major side effects of NSAIDs however are gastrointestinal ulcers which frequently precludes their long-term application. In this review, we hope to bridge the gap between NSAID toxicity and sEH-mediated metabolic pathways to focus on the role of epoxy fatty acid metabolic pathway of PUFAs in NSAIDS-ulcer formation and healing as well as inflammation-related carcinogenesis. Specifically we address the potential application of sEH inhibition to enhance ulcer healing at the site of inflammation via their activity on altered lipid signaling, mitochondrial function, and diminished reactive oxygen species, and further discuss the significance of dual COX and sEH inhibitor in anti-inflammation and carcinogenesis.
Collapse
Affiliation(s)
- Ryan D Jones
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Jie Liao
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Xin Tong
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Dandan Xu
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Leyu Sun
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Haonan Li
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Guang-Yu Yang
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| |
Collapse
|
16
|
Sausville LN, Williams SM, Pozzi A. Cytochrome P450 epoxygenases and cancer: A genetic and a molecular perspective. Pharmacol Ther 2019; 196:183-194. [DOI: 10.1016/j.pharmthera.2018.11.009] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
|
17
|
Hiesinger K, Wagner KM, Hammock BD, Proschak E, Hwang SH. Development of multitarget agents possessing soluble epoxide hydrolase inhibitory activity. Prostaglandins Other Lipid Mediat 2019; 140:31-39. [PMID: 30593866 PMCID: PMC6345559 DOI: 10.1016/j.prostaglandins.2018.12.003] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2018] [Revised: 11/27/2018] [Accepted: 12/24/2018] [Indexed: 02/08/2023]
Abstract
Over the last two decades polypharmacology has emerged as a new paradigm in drug discovery, even though developing drugs with high potency and selectivity toward a single biological target is still a major strategy. Often, targeting only a single enzyme or receptor shows lack of efficacy. High levels of inhibitor of a single target also can lead to adverse side effects. A second target may offer additive or synergistic effects to affecting the first target thereby reducing on- and off-target side effects. Therefore, drugs that inhibit multiple targets may offer a great potential for increased efficacy and reduced the adverse effects. In this review we summarize recent findings of rationally designed multitarget compounds that are aimed to improve efficacy and safety profiles compared to those that target a single enzyme or receptor. We focus on dual inhibitors/modulators that target the soluble epoxide hydrolase (sEH) as a common part of their design to take advantage of the beneficial effects of sEH inhibition.
Collapse
Affiliation(s)
- Kerstin Hiesinger
- Institute of Pharmaceutical Chemistry, Goethe-University of Frankfurt, Max-von-Laue Str. 9, D-60439, Frankfurt am Main, Germany
| | - Karen M Wagner
- Department of Entomology and Nematology and UC Davis Comprehensive Cancer Center, University of California, Davis, One Shields Avenue, Davis, CA 95616, USA
| | - Bruce D Hammock
- Department of Entomology and Nematology and UC Davis Comprehensive Cancer Center, University of California, Davis, One Shields Avenue, Davis, CA 95616, USA
| | - Ewgenij Proschak
- Institute of Pharmaceutical Chemistry, Goethe-University of Frankfurt, Max-von-Laue Str. 9, D-60439, Frankfurt am Main, Germany
| | - Sung Hee Hwang
- Department of Entomology and Nematology and UC Davis Comprehensive Cancer Center, University of California, Davis, One Shields Avenue, Davis, CA 95616, USA.
| |
Collapse
|
18
|
Kodani SD, Morisseau C. Role of epoxy-fatty acids and epoxide hydrolases in the pathology of neuro-inflammation. Biochimie 2019; 159:59-65. [PMID: 30716359 DOI: 10.1016/j.biochi.2019.01.020] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2018] [Accepted: 01/29/2019] [Indexed: 12/31/2022]
Abstract
Neuroinflammation is a physiologic response aimed at protecting the central nervous system during injury. However, unresolved and chronic neuroinflammation can lead to long term damage and eventually neurologic disease including Parkinson's disease, Alzheimer's disease and dementia. Recently, enhancing the concentration of epoxyeicosatrienoic acids (EETs) through blocking their hydrolytic degradation by soluble epoxide hydrolase (sEH) has been applied towards reducing the long-term damage associated with central neurologic insults. Evidence suggests this protective effect is mediated, at least in part, through polarization of microglia to an anti-inflammatory phenotype that blocks the inflammatory actions of prostaglandins and promotes wound repair. This mini-review overviews the epidemiologic basis for using sEH inhibition towards neuroinflammatory disease and pharmacologic studies testing sEH inhibition in several neurologic diseases. Additionally, the combination of sEH inhibition with other eicosanoid signaling pathways is considered as an enhanced approach for developing potent neuroprotectants.
Collapse
Affiliation(s)
- Sean D Kodani
- Department of Entomology and Nematology, UCD Comprehensive Cancer Center, University of California Davis, One Shields Avenue, Davis, CA, 95616, USA
| | - Christophe Morisseau
- Department of Entomology and Nematology, UCD Comprehensive Cancer Center, University of California Davis, One Shields Avenue, Davis, CA, 95616, USA.
| |
Collapse
|
19
|
Suppression of chemotherapy-induced cytokine/lipid mediator surge and ovarian cancer by a dual COX-2/sEH inhibitor. Proc Natl Acad Sci U S A 2019; 116:1698-1703. [PMID: 30647111 DOI: 10.1073/pnas.1803999116] [Citation(s) in RCA: 84] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Although chemotherapy is a conventional cancer treatment, it may induce a protumorigenic microenvironment by triggering the release of proinflammatory mediators. In this study, we demonstrate that ovarian tumor cell debris generated by first-line platinum- and taxane-based chemotherapy accelerates tumor progression by stimulating a macrophage-derived "surge" of proinflammatory cytokines and bioactive lipids. Thus, targeting a single inflammatory mediator or pathway is unlikely to prevent therapy-induced tumor progression. Here, we show that combined pharmacological abrogation of the cyclooxygenase-2 (COX-2) and soluble epoxide hydrolase (sEH) pathways prevented the debris-induced surge of both cytokines and lipid mediators by macrophages. In animal models, the dual COX-2/sEH inhibitor PTUPB delayed the onset of debris-stimulated ovarian tumor growth and ascites leading to sustained survival over 120 days postinjection. Therefore, dual inhibition of COX-2/sEH may be an approach to suppress debris-stimulated ovarian tumor growth by preventing the therapy-induced surge of cytokines and lipid mediators.
Collapse
|
20
|
Hashemi Goradel N, Najafi M, Salehi E, Farhood B, Mortezaee K. Cyclooxygenase-2 in cancer: A review. J Cell Physiol 2018; 234:5683-5699. [PMID: 30341914 DOI: 10.1002/jcp.27411] [Citation(s) in RCA: 476] [Impact Index Per Article: 68.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2018] [Accepted: 08/22/2018] [Indexed: 12/17/2022]
Abstract
Cyclooxygenase-2 (COX-2) is frequently expressed in many types of cancers exerting a pleiotropic and multifaceted role in genesis or promotion of carcinogenesis and cancer cell resistance to chemo- and radiotherapy. COX-2 is released by cancer-associated fibroblasts (CAFs), macrophage type 2 (M2) cells, and cancer cells to the tumor microenvironment (TME). COX-2 induces cancer stem cell (CSC)-like activity, and promotes apoptotic resistance, proliferation, angiogenesis, inflammation, invasion, and metastasis of cancer cells. COX-2 mediated hypoxia within the TME along with its positive interactions with YAP1 and antiapoptotic mediators are all in favor of cancer cell resistance to chemotherapeutic drugs. COX-2 exerts most of the functions through its metabolite prostaglandin E2. In some and limited situations, COX-2 may act as an antitumor enzyme. Multiple signals are contributed to the functions of COX-2 on cancer cells or its regulation. Members of mitogen-activated protein kinase (MAPK) family, epidermal growth factor receptor (EGFR), and nuclear factor-κβ are main upstream modulators for COX-2 in cancer cells. COX-2 also has interactions with a number of hormones within the body. Inhibition of COX-2 provides a high possibility to exert therapeutic outcomes in cancer. Administration of COX-2 inhibitors in a preoperative setting could reduce the risk of metastasis in cancer patients. COX-2 inhibition also sensitizes cancer cells to treatments like radio- and chemotherapy. Chemotherapeutic agents adversely induce COX-2 activity. Therefore, choosing an appropriate chemotherapy drugs along with adjustment of the type and does for COX-2 inhibitors based on the type of cancer would be an effective adjuvant strategy for targeting cancer.
Collapse
Affiliation(s)
- Nasser Hashemi Goradel
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Masoud Najafi
- Radiology and Nuclear Medicine Department, School of Paramedical Sciences, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Eniseh Salehi
- Department of Anatomy, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Bagher Farhood
- Departments of Medical Physics and Radiology, Faculty of Paramedical Sciences, Kashan University of Medical Sciences, Kashan, Iran
| | - Keywan Mortezaee
- Department of Anatomy, School of Medicine, Kurdistan University of Medical Sciences, Sanandaj, Iran
| |
Collapse
|
21
|
Mendes M, Sousa JJ, Pais A, Vitorino C. Targeted Theranostic Nanoparticles for Brain Tumor Treatment. Pharmaceutics 2018; 10:E181. [PMID: 30304861 PMCID: PMC6321593 DOI: 10.3390/pharmaceutics10040181] [Citation(s) in RCA: 66] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2018] [Revised: 09/21/2018] [Accepted: 09/27/2018] [Indexed: 12/13/2022] Open
Abstract
The poor prognosis and rapid recurrence of glioblastoma (GB) are associated to its fast-growing process and invasive nature, which make difficult the complete removal of the cancer infiltrated tissues. Additionally, GB heterogeneity within and between patients demands a patient-focused method of treatment. Thus, the implementation of nanotechnology is an attractive approach considering all anatomic issues of GB, since it will potentially improve brain drug distribution, due to the interaction between the blood⁻brain barrier and nanoparticles (NPs). In recent years, theranostic techniques have also been proposed and regarded as promising. NPs are advantageous for this application, due to their respective size, easy surface modification and versatility to integrate multiple functional components in one system. The design of nanoparticles focused on therapeutic and diagnostic applications has increased exponentially for the treatment of cancer. This dual approach helps to understand the location of the tumor tissue, the biodistribution of nanoparticles, the progress and efficacy of the treatment, and is highly useful for personalized medicine-based therapeutic interventions. To improve theranostic approaches, different active strategies can be used to modulate the surface of the nanotheranostic particle, including surface markers, proteins, drugs or genes, and take advantage of the characteristics of the microenvironment using stimuli responsive triggers. This review focuses on the different strategies to improve the GB treatment, describing some cell surface markers and their ligands, and reports some strategies, and their efficacy, used in the current research.
Collapse
Affiliation(s)
- Maria Mendes
- Faculty of Pharmacy, University of Coimbra, 3000-548 Coimbra, Portugal.
- Center for Neurosciences and Cell Biology (CNC), University of Coimbra, 3004-504 Coimbra, Portugal.
| | - João José Sousa
- Faculty of Pharmacy, University of Coimbra, 3000-548 Coimbra, Portugal.
- LAQV, REQUIMTE, Group of Pharmaceutical Technology, 3000-548 Coimbra, Portugal.
| | - Alberto Pais
- Coimbra Chemistry Centre, Department of Chemistry, University of Coimbra, 3004-535 Coimbra, Portugal.
| | - Carla Vitorino
- Faculty of Pharmacy, University of Coimbra, 3000-548 Coimbra, Portugal.
- Center for Neurosciences and Cell Biology (CNC), University of Coimbra, 3004-504 Coimbra, Portugal.
- LAQV, REQUIMTE, Group of Pharmaceutical Technology, 3000-548 Coimbra, Portugal.
| |
Collapse
|
22
|
Li J, Ji X, Wang H. Targeting Long Noncoding RNA HMMR-AS1 Suppresses and Radiosensitizes Glioblastoma. Neoplasia 2018; 20:456-466. [PMID: 29574252 PMCID: PMC5915996 DOI: 10.1016/j.neo.2018.02.010] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2018] [Revised: 02/11/2018] [Accepted: 02/14/2018] [Indexed: 01/07/2023] Open
Abstract
Emergent evidences revealed that long noncoding RNAs (lncRNAs) participate in neoplastic progression. HMMR is an oncogene that is highly expressed in glioblastoma (GBM) and supports GBM growth. Whether lncRNAs regulate HMMR in GBM remains unknown. Herein, we identify that an HMMR antisense lncRNA, HMMR-AS1, is hyperexpressed in GBM cell lines and stabilizes HMMR mRNA. Knockdown of HMMR-AS1 reduces HMMR expression; inhibits cell migration, invasion, and mesenchymal phenotypes; and suppresses GBM cell growth both in vitro and in vivo. Moreover, knockdown of HMMR-AS1 radiosensitizes GBM by reducing DNA repair proteins ATM, RAD51, and BMI1. Our data demonstrate a mechanism of sense-antisense interference between HMMR and HMMR-AS1 in GBM and suggest that targeting HMMR-AS1 is a potential strategy for GBM treatment.
Collapse
Affiliation(s)
- Junyang Li
- Department of Neurosurgery, Jinling Hospital, Medical School of Nanjing University, Nanjing, 210002, China
| | - Xiangjun Ji
- Department of Neurosurgery, Jinling Hospital, Medical School of Nanjing University, Nanjing, 210002, China
| | - Handong Wang
- Department of Neurosurgery, Jinling Hospital, Medical School of Nanjing University, Nanjing, 210002, China.
| |
Collapse
|