1
|
Li Z, Hu F, Xiong L, Zhou X, Dong C, Zheng Y. Underlying mechanisms of traditional Chinese medicine in the prevention and treatment of diabetic retinopathy: Evidences from molecular and clinical studies. JOURNAL OF ETHNOPHARMACOLOGY 2024; 335:118641. [PMID: 39084273 DOI: 10.1016/j.jep.2024.118641] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Revised: 07/26/2024] [Accepted: 07/28/2024] [Indexed: 08/02/2024]
Abstract
As one of the most serious microvascular complications of diabetes mellitus (DM), diabetic retinopathy (DR) can cause visual impairment and even blindness. With the rapid increase in the prevalence of DM, the incidence of DR is also rising year by year. Preventing and effectively treating DR has become a major focus in the medical field. Traditional Chinese medicine (TCM) has a wealth of experience in treating DR and has achieved significant results with various herbs and TCM prescriptions. Traditional Chinese Medicine (TCM) provides a comprehensive therapeutic strategy for diabetic retinopathy (DR), encompassing anti-inflammatory and antioxidant actions, anti-neovascularization, neuroprotection, regulation of glucose metabolism, and inhibition of apoptosis. This review provides an overview of the current status of TCM treatment for DR in recent years, including experimental studies and clinical researches, to explore the clinical efficacy and the underlying modern mechanisms of herbs and TCM prescriptions. Besides, we also discussed the challenges TCM faces in treating DR, such as drug-drug interactions among TCM components and the lack of high-quality evidence-based medicine practice, which pose significant obstacles to TCM's application in DR.
Collapse
Affiliation(s)
- Zhengpin Li
- Anhui University of Traditional Chinese Medicine, College of Traditional Chinese Medicine, Hefei, China
| | - Faquan Hu
- Anhui University of Traditional Chinese Medicine, College of Traditional Chinese Medicine, Hefei, China
| | - Liyuan Xiong
- Anhui University of Traditional Chinese Medicine, College of Traditional Chinese Medicine, Hefei, China
| | - Xuemei Zhou
- Anhui University of Traditional Chinese Medicine, College of Traditional Chinese Medicine, Hefei, China
| | - Changwu Dong
- The Second Clinical Medical School, Anhui University of Traditional Chinese Medicine, Hefei, China
| | - Yujiao Zheng
- Anhui University of Traditional Chinese Medicine, College of Traditional Chinese Medicine, Hefei, China.
| |
Collapse
|
2
|
Misra R, Barman P, Bhabak KP. Esterase-Responsive Fluorogenic Prodrugs of Aldose Reductase Inhibitor Epalrestat: An Innovative Strategy toward Enhanced Anticancer Activity. ACS APPLIED BIO MATERIALS 2024; 7:6542-6553. [PMID: 39146213 DOI: 10.1021/acsabm.4c00719] [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] [Indexed: 08/17/2024]
Abstract
In addition to the conventional chemotherapeutic drugs, potent inhibitors of key enzymes that are differentially overexpressed in cancer cells and associated with its progression are often considered as the drugs of choice for treating cancer. Aldose reductase (AR), which is primarily associated with complications of diabetes, is known to be closely related to the development of cancer and drug resistance. Epalrestat (EPA), an FDA-approved drug, is a potent inhibitor of AR and exhibits anticancer activity. However, its poor pharmacokinetic properties limit its bioavailability and therapeutic benefits. We report herein the first examples of esterase-responsive turn-on fluorogenic prodrugs for the sustained release of EPA to cancer cells with a turn-on fluorescence readout. Carboxylesterases are known to be overexpressed in several organ-specific cancer cells and help in selective uncaging of drug from the prodrugs. The prodrugs were synthesized using a multistep organic synthesis and successfully characterized. Absorption and emission spectroscopic studies indicated successful activation of the prodrugs in the presence of porcine liver esterase (PLE) under physiological condition. HPLC studies revealed a simultaneous release of both the drug and the fluorophore from the prodrugs over time with mechanistic insights. While the inhibitory potential of EPA released from the prodrugs toward the enzyme AR was validated in the aqueous medium, the anticancer activity of the prodrugs was studied in a representative cervical cancer cell line. Interestingly, our results revealed that the development of the prodrugs can significantly enhance the anticancer potential of EPA. Finally, the drug uncaging process from the prodrugs by the intracellular esterases was studied in the cellular medium by measuring the turn-on fluorescence using fluorescence microscopy. Therefore, the present study highlights the rational development of the fluorogenic prodrugs of EPA, which will help enhance its anticancer potential with better therapeutic potential.
Collapse
Affiliation(s)
- Roopjyoti Misra
- Department of Chemistry, Indian Institute of Technology Guwahati, Guwahati 781039, Assam, India
| | - Pallavi Barman
- Department of Chemistry, Indian Institute of Technology Guwahati, Guwahati 781039, Assam, India
| | - Krishna P Bhabak
- Department of Chemistry, Indian Institute of Technology Guwahati, Guwahati 781039, Assam, India
| |
Collapse
|
3
|
NAGINI SIDDAVARAM, KALLAMADI PRATHAPREDDY, TANAGALA KRANTHIKIRANKISHORE, REDDY GEEREDDYBHANUPRAKASH. Aldo-keto reductases: Role in cancer development and theranostics. Oncol Res 2024; 32:1287-1308. [PMID: 39055885 PMCID: PMC11267078 DOI: 10.32604/or.2024.049918] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Accepted: 05/08/2024] [Indexed: 07/28/2024] Open
Abstract
Aldo-keto reductases (AKRs) are a superfamily of enzymes that play crucial roles in various cellular processes, including the metabolism of xenobiotics, steroids, and carbohydrates. A growing body of evidence has unveiled the involvement of AKRs in the development and progression of various cancers. AKRs are aberrantly expressed in a wide range of malignant tumors. Dysregulated expression of AKRs enables the acquisition of hallmark traits of cancer by activating oncogenic signaling pathways and contributing to chemoresistance. AKRs have emerged as promising oncotherapeutic targets given their pivotal role in cancer development and progression. Inhibition of aldose reductase (AR), either alone or in combination with chemotherapeutic drugs, has evolved as a pragmatic therapeutic option for cancer. Several classes of synthetic aldo-keto reductase (AKR) inhibitors have been developed as potential anticancer agents, some of which have shown promise in clinical trials. Many AKR inhibitors from natural sources also exhibit anticancer effects. Small molecule inhibitors targeting specific AKR isoforms have shown promise in preclinical studies. These inhibitors disrupt the activation of oncogenic signaling by modulating transcription factors and kinases and sensitizing cancer cells to chemotherapy. In this review, we discuss the physiological functions of human AKRs, the aberrant expression of AKRs in malignancies, the involvement of AKRs in the acquisition of cancer hallmarks, and the role of AKRs in oncogenic signaling, and drug resistance. Finally, the potential of aldose reductase inhibitors (ARIs) as anticancer drugs is summarized.
Collapse
|
4
|
Shanbhag AP, Bhowmik P. Cancer to Cataracts: The Mechanistic Impact of Aldo-Keto Reductases in Chronic Diseases. THE YALE JOURNAL OF BIOLOGY AND MEDICINE 2024; 97:179-204. [PMID: 38947111 PMCID: PMC11202113 DOI: 10.59249/vtbv6559] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 07/02/2024]
Abstract
Aldo-keto reductases (AKRs) are a superfamily of promiscuous enzymes that have been chiseled by evolution to act as catalysts for numerous regulatory pathways in humans. However, they have not lost their promiscuity in the process, essentially making them a double-edged sword. The superfamily is involved in multiple metabolic pathways and are linked to chronic diseases such as cataracts, diabetes, and various cancers. Unlike other detoxifying enzymes such as cytochrome P450s (CYP450s), short-chain dehydrogenases (SDRs), and medium-chain dehydrogenases (MDRs), that participate in essential pathways, AKRs are more widely distributed and have members with interchangeable functions. Moreover, their promiscuity is ubiquitous across all species and participates in the resistance of pathogenic microbes. Moreover, the introduction of synthetic substrates, such as synthetic molecules and processed foods, results in unwanted "toxification" due to enzyme promiscuity, leading to chronic diseases.
Collapse
Affiliation(s)
- Anirudh P. Shanbhag
- Bugworks Research India Pvt. Ltd., Bengaluru,
Karnataka, India
- Novartis Healthcare Pvt. Ltd., Hyderabad, Telangana,
India
| | - Purnendu Bhowmik
- Bugworks Research India Pvt. Ltd., Bengaluru,
Karnataka, India
- Centre for Cellular and Molecular Platforms (C-CAMP),
National Centre for Biological Sciences (NCBS), Bengaluru, Karnataka,
India
| |
Collapse
|
5
|
Alahmari H, Liu CC, Rubin E, Lin VY, Rodriguez P, Chang KC. Vitamin C alleviates hyperglycemic stress in retinal pigment epithelial cells. Mol Biol Rep 2024; 51:637. [PMID: 38727927 DOI: 10.1007/s11033-024-09595-2] [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: 02/02/2024] [Accepted: 04/29/2024] [Indexed: 05/13/2024]
Abstract
BACKGROUND Retinal pigment epithelial cells (RPECs) are a type of retinal cells that structurally and physiologically support photoreceptors. However, hyperglycemia has been shown to play a critical role in the progression of diabetic retinopathy (DR), which is one of the leading causes of vision impairment. In the diabetic eye, the high glucose environment damages RPECs via the induction of oxidative stress, leading to the release of excess reactive oxygen species (ROS) and triggering apoptosis. In this study, we aim to investigate the antioxidant mechanism of Vitamin C in reducing hyperglycemia-induced stress and whether this mechanism can preserve the function of RPECs. METHODS AND RESULTS ARPE-19 cells were treated with high glucose in the presence or absence of Vitamin C. Cell viability was measured by MTT assay. Cleaved poly ADP-ribose polymerase (PARP) was used to identify apoptosis in the cells. ROS were detected by the DCFH-DA reaction. The accumulation of sorbitol in the aldose reductase (AR) polyol pathway was determined using the sorbitol detection assay. Primary mouse RPECs were isolated from adult mice and identified by Rpe65 expression. The mitochondrial damage was measured by mitochondrial membrane depolarization. Our results showed that high glucose conditions reduce cell viability in RPECs while Vitamin C can restore cell viability, compared to the vehicle treatment. We also demonstrated that Vitamin C reduces hyperglycemia-induced ROS production and prevents cell apoptosis in RPECs in an AR-independent pathway. CONCLUSIONS These results suggest that Vitamin C is not only a nutritional necessity but also an adjuvant that can be combined with AR inhibitors for alleviating hyperglycemic stress in RPECs.
Collapse
Affiliation(s)
- Hamid Alahmari
- Department of Ophthalmology, Louis J. Fox Center for Vision Restoration, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15219, USA
| | - Chia-Chun Liu
- Department of Ophthalmology, Louis J. Fox Center for Vision Restoration, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15219, USA
| | - Elizabeth Rubin
- Department of Ophthalmology, Louis J. Fox Center for Vision Restoration, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15219, USA
| | - Venice Y Lin
- Department of Ophthalmology, Louis J. Fox Center for Vision Restoration, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15219, USA
- North Allegheny Senior High School, Wexford, PA, 15090, USA
| | - Paul Rodriguez
- Department of Ophthalmology, Louis J. Fox Center for Vision Restoration, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15219, USA
| | - Kun-Che Chang
- Department of Ophthalmology, Louis J. Fox Center for Vision Restoration, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15219, USA.
- Department of Neurobiology, Center of Neuroscience, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15213, USA.
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan.
| |
Collapse
|
6
|
Rao M, Huang YK, Liu CC, Meadows C, Cheng HC, Zhou M, Chen YC, Xia X, Goldberg JL, Williams AM, Kuwajima T, Chang KC. Aldose reductase inhibition decelerates optic nerve degeneration by alleviating retinal microglia activation. Sci Rep 2023; 13:5592. [PMID: 37019993 PMCID: PMC10076364 DOI: 10.1038/s41598-023-32702-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Accepted: 03/31/2023] [Indexed: 04/07/2023] Open
Abstract
As part of the central nervous system (CNS), retinal ganglion cells (RGCs) and their axons are the only neurons in the retina that transmit visual signals from the eye to the brain via the optic nerve (ON). Unfortunately, they do not regenerate upon injury in mammals. In ON trauma, retinal microglia (RMG) become activated, inducing inflammatory responses and resulting in axon degeneration and RGC loss. Since aldose reductase (AR) is an inflammatory response mediator highly expressed in RMG, we investigated if pharmacological inhibition of AR can attenuate ocular inflammation and thereby promote RGC survival and axon regeneration after ON crush (ONC). In vitro, we discovered that Sorbinil, an AR inhibitor, attenuates BV2 microglia activation and migration in the lipopolysaccharide (LPS) and monocyte chemoattractant protein-1 (MCP-1) treatments. In vivo, Sorbinil suppressed ONC-induced Iba1 + microglia/macrophage infiltration in the retina and ON and promoted RGC survival. Moreover, Sorbinil restored RGC function and delayed axon degeneration one week after ONC. RNA sequencing data revealed that Sorbinil protects the retina from ONC-induced degeneration by suppressing inflammatory signaling. In summary, we report the first study demonstrating that AR inhibition transiently protects RGC and axon from degeneration, providing a potential therapeutic strategy for optic neuropathies.
Collapse
Affiliation(s)
- Mishal Rao
- Department of Ophthalmology, Louis J. Fox Center for Vision Restoration, University of Pittsburgh School of Medicine, 203 Lothrop, Pittsburgh, PA, 15213, USA
| | - Yu-Kai Huang
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan
- Division of Neurosurgery, Department of Surgery, Kaohsiung Medical University Hospital, Kaohsiung, 80708, Taiwan
- Department of Surgery, Kaohsiung Municipal Ta-Tung Hospital, Kaohsiung, 80145, Taiwan
| | - Chia-Chun Liu
- Department of Ophthalmology, Louis J. Fox Center for Vision Restoration, University of Pittsburgh School of Medicine, 203 Lothrop, Pittsburgh, PA, 15213, USA
| | - Chandler Meadows
- Department of Ophthalmology, Louis J. Fox Center for Vision Restoration, University of Pittsburgh School of Medicine, 203 Lothrop, Pittsburgh, PA, 15213, USA
| | - Hui-Chun Cheng
- Department of Ophthalmology, Louis J. Fox Center for Vision Restoration, University of Pittsburgh School of Medicine, 203 Lothrop, Pittsburgh, PA, 15213, USA
| | - Mengli Zhou
- Department of Computational and Systems Biology, Hillman Cancer Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15260, USA
| | - Yu-Chih Chen
- Department of Computational and Systems Biology, Hillman Cancer Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15260, USA
| | - Xin Xia
- Spencer Center for Vision Research, Byers Eye Institute, School of Medicine, Stanford University, Palo Alto, CA, 94304, USA
| | - Jeffrey L Goldberg
- Spencer Center for Vision Research, Byers Eye Institute, School of Medicine, Stanford University, Palo Alto, CA, 94304, USA
| | - Andrew M Williams
- Department of Ophthalmology, Louis J. Fox Center for Vision Restoration, University of Pittsburgh School of Medicine, 203 Lothrop, Pittsburgh, PA, 15213, USA
| | - Takaaki Kuwajima
- Department of Ophthalmology, Louis J. Fox Center for Vision Restoration, University of Pittsburgh School of Medicine, 203 Lothrop, Pittsburgh, PA, 15213, USA
| | - Kun-Che Chang
- Department of Ophthalmology, Louis J. Fox Center for Vision Restoration, University of Pittsburgh School of Medicine, 203 Lothrop, Pittsburgh, PA, 15213, USA.
- Department of Neurobiology, Center of Neuroscience, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15213, USA.
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan.
| |
Collapse
|
7
|
Sardelli G, Scali V, Signore G, Balestri F, Cappiello M, Mura U, Del Corso A, Moschini R. Response of a Human Lens Epithelial Cell Line to Hyperglycemic and Oxidative Stress: The Role of Aldose Reductase. Antioxidants (Basel) 2023; 12:antiox12040829. [PMID: 37107204 PMCID: PMC10135174 DOI: 10.3390/antiox12040829] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 03/23/2023] [Accepted: 03/25/2023] [Indexed: 03/31/2023] Open
Abstract
A common feature of different types of diabetes is the high blood glucose levels, which are known to induce a series of metabolic alterations, leading to damaging events in different tissues. Among these alterations, both increased polyol pathway flux and oxidative stress are considered to play relevant roles in the response of different cells. In this work, the effect on a human lens epithelial cell line of stress conditions, consisting of exposure to either high glucose levels or to the lipid peroxidation product 4-hydroxy-2-nonenal, is reported. The occurrence of osmotic imbalance, alterations of glutathione levels, and expression of inflammatory markers was monitored. A common feature of the two stress conditions was the expression of COX-2, which, only in the case of hyperglycemic stress, occurred through NF-κB activation. In our cell model, aldose reductase activity, which is confirmed as the only activity responsible for the osmotic imbalance occurring in hyperglycemic conditions, seemed to have no role in controlling the onset of the inflammatory phenomena. However, it played a relevant role in cellular detoxification against lipid peroxidation products. These results, in confirming the multifactorial nature of the inflammatory phenomena, highlight the dual role of aldose reductase as having both damaging but also protecting activity, depending on stress conditions.
Collapse
Affiliation(s)
- Gemma Sardelli
- Biochemistry Unit, Department of Biology, University of Pisa, 56123 Pisa, Italy
| | - Viola Scali
- Biochemistry Unit, Department of Biology, University of Pisa, 56123 Pisa, Italy
| | - Giovanni Signore
- Biochemistry Unit, Department of Biology, University of Pisa, 56123 Pisa, Italy
- Interdepartmental Research Center Nutrafood “Nutraceuticals and Food for Health”, University of Pisa, 56124 Pisa, Italy
| | - Francesco Balestri
- Biochemistry Unit, Department of Biology, University of Pisa, 56123 Pisa, Italy
- Interdepartmental Research Center Nutrafood “Nutraceuticals and Food for Health”, University of Pisa, 56124 Pisa, Italy
| | - Mario Cappiello
- Biochemistry Unit, Department of Biology, University of Pisa, 56123 Pisa, Italy
- Interdepartmental Research Center Nutrafood “Nutraceuticals and Food for Health”, University of Pisa, 56124 Pisa, Italy
| | - Umberto Mura
- Biochemistry Unit, Department of Biology, University of Pisa, 56123 Pisa, Italy
| | - Antonella Del Corso
- Biochemistry Unit, Department of Biology, University of Pisa, 56123 Pisa, Italy
- Interdepartmental Research Center Nutrafood “Nutraceuticals and Food for Health”, University of Pisa, 56124 Pisa, Italy
- Correspondence: ; Tel.: +39-050-2211450
| | - Roberta Moschini
- Biochemistry Unit, Department of Biology, University of Pisa, 56123 Pisa, Italy
- Interdepartmental Research Center Nutrafood “Nutraceuticals and Food for Health”, University of Pisa, 56124 Pisa, Italy
| |
Collapse
|
8
|
The Role of Aldose Reductase in Beta-Amyloid-Induced Microglia Activation. Int J Mol Sci 2022; 23:ijms232315088. [PMID: 36499422 PMCID: PMC9739496 DOI: 10.3390/ijms232315088] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Revised: 11/28/2022] [Accepted: 11/29/2022] [Indexed: 12/03/2022] Open
Abstract
The occurrence of Alzheimer's disease has been associated with the accumulation of beta-amyloid (β-amyloid) plaques. These plaques activate microglia to secrete inflammatory molecules, which damage neurons in the brain. Thus, understanding the underlying mechanism of microglia activation can provide a therapeutic strategy for alleviating microglia-induced neuroinflammation. The aldose reductase (AR) enzyme catalyzes the reduction of glucose to sorbitol in the polyol pathway. In addition to mediating diabetic complications in hyperglycemic environments, AR also helps regulate inflammation in microglia. However, little is known about the role of AR in β-amyloid-induced inflammation in microglia and subsequent neuronal death. In this study, we confirmed that AR inhibition attenuates increased β-amyloid-induced reactive oxygen species and tumor necrosis factor α secretion by suppressing ERK signaling in BV2 cells. In addition, we are the first to report that AR inhibition reduced the phagocytotic capability and cell migration of BV2 cells in response to β-amyloid. To further investigate the protective role of the AR inhibitor sorbinil in neurons, we co-cultured β-amyloid-induced microglia with stem cell-induced neurons. sorbinil ameliorated neuronal damage in both cells in the co-culture system. In summary, our findings reveal AR regulation of microglia activation as a novel therapeutic target for Alzheimer's disease.
Collapse
|
9
|
Bailly C. Moving toward a new horizon for the aldose reductase inhibitor epalrestat to treat drug-resistant cancer. Eur J Pharmacol 2022; 931:175191. [PMID: 35964660 DOI: 10.1016/j.ejphar.2022.175191] [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: 06/15/2022] [Revised: 07/18/2022] [Accepted: 08/05/2022] [Indexed: 11/19/2022]
Abstract
Epalrestat (EPA) is a potent inhibitor of aldose reductases AKR1B1 and AKR1B10, used for decades in Japan for the treatment of diabetic peripheral neuropathy. This orally-active, brain-permeable small molecule, with a relatively rare and essential 2-thioxo-4-thiazolidinone motif, functions as a regulator intracellular carbonyl species. The repurposing of EPA for the treatment of pediatric rare diseases, brain disorders and cancer has been proposed. A detailed analysis of the mechanism of action, and the benefit of EPA to combat advanced malignancies is offered here. EPA has revealed marked anticancer activities, alone and in combination with cytotoxic chemotherapy and targeted therapeutics, in experimental models of liver, colon, and breast cancers. Through inhibition of AKR1B1 and/or AKR1B10 and blockade of the epithelial-mesenchymal transition, EPA largely enhances the sensitivity of cancer cells to drugs like doxorubicin and sorafenib. EPA has revealed a major anticancer effect in an experimental model of basal-like breast cancer and clinical trials have been developed in patients with triple-negative breast cancer. The repurposing of the drug to treat chemo-resistant solid tumors seems promising, but more studies are needed to define the best trajectory for the positioning of EPA in oncology.
Collapse
Affiliation(s)
- Christian Bailly
- OncoWitan, Scientific Consulting Office, Lille, Wasquehal, 59290, France.
| |
Collapse
|
10
|
Chang KC, Liu PF, Chang CH, Lin YC, Chen YJ, Shu CW. The interplay of autophagy and oxidative stress in the pathogenesis and therapy of retinal degenerative diseases. Cell Biosci 2022; 12:1. [PMID: 34980273 PMCID: PMC8725349 DOI: 10.1186/s13578-021-00736-9] [Citation(s) in RCA: 63] [Impact Index Per Article: 31.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Accepted: 12/19/2021] [Indexed: 12/27/2022] Open
Abstract
Oxidative stress is mainly caused by intracellular reactive oxygen species (ROS) production, which is highly associated with normal physiological homeostasis and the pathogenesis of diseases, particularly ocular diseases. Autophagy is a self-clearance pathway that removes oxidized cellular components and regulates cellular ROS levels. ROS can modulate autophagy activity through transcriptional and posttranslational mechanisms. Autophagy further triggers transcription factor activation and degrades impaired organelles and proteins to eliminate excessive ROS in cells. Thus, autophagy may play an antioxidant role in protecting ocular cells from oxidative stress. Nevertheless, excessive autophagy may cause autophagic cell death. In this review, we summarize the mechanisms of interaction between ROS and autophagy and their roles in the pathogenesis of several ocular diseases, including glaucoma, age-related macular degeneration (AMD), diabetic retinopathy (DR), and optic nerve atrophy, which are major causes of blindness. The autophagy modulators used to treat ocular diseases are further discussed. The findings of the studies reviewed here might shed light on the development and use of autophagy modulators for the future treatment of ocular diseases.
Collapse
Affiliation(s)
- Kun-Che Chang
- Department of Ophthalmology and Neurobiology, Louis J. Fox Center for Vision Restoration, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.,Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Pei-Feng Liu
- Department of Biomedical Science and Environmental Biology, PhD Program in Life Science, College of Life Science, Kaohsiung Medical University, Kaohsiung, Taiwan.,Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan.,Center for Cancer Research, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Chia-Hsuan Chang
- Institute of BioPharmaceutical Sciences, National Sun Yat-Sen University, No. 70, Lianhai Rd., Gushan Dist., Kaohsiung, 80424, Taiwan
| | - Ying-Cheng Lin
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Taichung Veterans General Hospital, Taichung, Taiwan
| | - Yen-Ju Chen
- Institute of Clinical Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan.,Department of Medical Research, Taichung Veterans General Hospital, Taichung, Taiwan.,Division of Allergy, Immunology and Rheumatology, Department of Internal Medicine, Taichung Veterans General Hospital, Taichung, Taiwan
| | - Chih-Wen Shu
- Institute of BioPharmaceutical Sciences, National Sun Yat-Sen University, No. 70, Lianhai Rd., Gushan Dist., Kaohsiung, 80424, Taiwan.
| |
Collapse
|
11
|
Sabatier P, Beusch CM, Gencheva R, Cheng Q, Zubarev R, Arnér ESJ. Comprehensive chemical proteomics analyses reveal that the new TRi-1 and TRi-2 compounds are more specific thioredoxin reductase 1 inhibitors than auranofin. Redox Biol 2021; 48:102184. [PMID: 34788728 PMCID: PMC8591550 DOI: 10.1016/j.redox.2021.102184] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Revised: 11/06/2021] [Accepted: 11/09/2021] [Indexed: 12/16/2022] Open
Abstract
Anticancer drugs that target cellular antioxidant systems have recently attracted much attention. Auranofin (AF) is currently evaluated in several clinical trials as an anticancer agent that targets the cytosolic and mitochondrial forms of the selenoprotein thioredoxin reductase, TXNRD1 and TXNRD2. Recently, two novel TXNRD1 inhibitors (TRi-1 and TRi-2) have been developed that showed anticancer efficacy comparable to AF, but with lower mitochondrial toxicity. However, the cellular action mechanisms of these drugs have not yet been thoroughly studied. Here we used several proteomics approaches to determine the effects of AF, TRi-1 and TRi-2 when used at IC50 concentrations with the mouse B16 melanoma and LLC lung adenocarcinoma cells, as these are often used for preclinical mouse models in evaluation of anticancer drugs. The results demonstrate that TRi-1 and TRi-2 are more specific TXNRD1 inhibitors than AF and reveal additional AF-specific effects on the cellular proteome. Interestingly, AF triggered stronger Nrf2-driven antioxidant responses than the other two compounds. Furthermore, AF affected several additional proteins, including GSK3A, GSK3B, MCMBP and EEFSEC, implicating additional effects on glycogen metabolism, cellular differentiation, inflammatory pathways, DNA replication and selenoprotein synthesis processes. Our proteomics data provide a resource for researchers interested in the multidimensional analysis of proteome changes associated with oxidative stress in general, and the effects of TXNRD1 inhibitors and AF protein targets in particular.
Collapse
Affiliation(s)
- Pierre Sabatier
- Chemistry I, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, 17177, Sweden
| | - Christian M Beusch
- Chemistry I, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, 17177, Sweden
| | - Radosveta Gencheva
- Division of Biochemistry, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, 17177, Sweden
| | - Qing Cheng
- Division of Biochemistry, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, 17177, Sweden
| | - Roman Zubarev
- Chemistry I, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, 17177, Sweden; Department of Pharmacological & Technological Chemistry, I.M. Sechenov First Moscow State Medical University, Moscow, 119146, Russia; The National Medical Research Center for Endocrinology, 115478, Moscow, Russia.
| | - Elias S J Arnér
- Division of Biochemistry, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, 17177, Sweden; Department of Selenoprotein Research, National Institute of Oncology, 1122, Budapest, Hungary.
| |
Collapse
|
12
|
Maksimovic I, David Y. Non-enzymatic Covalent Modifications as a New Chapter in the Histone Code. Trends Biochem Sci 2021; 46:718-730. [PMID: 33965314 PMCID: PMC8364488 DOI: 10.1016/j.tibs.2021.04.004] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Revised: 03/26/2021] [Accepted: 04/02/2021] [Indexed: 12/29/2022]
Abstract
The interior of the cell abounds with reactive species that can accumulate as non-enzymatic covalent modifications (NECMs) on biological macromolecules. These adducts interfere with many cellular processes, for example, by altering proteins' surface topology, enzymatic activity, or interactomes. Here, we discuss dynamic NECMs on chromatin, which serves as the cellular blueprint. We first outline the chemistry of NECM formation and then focus on the recently identified effects of their accumulation on chromatin structure and transcriptional output. We next describe the known cellular regulatory mechanisms that prevent or reverse NECM formation. Finally, we discuss recently developed chemical biology platforms for probing and manipulating these NECMs in vitro and in vivo.
Collapse
Affiliation(s)
- Igor Maksimovic
- Tri-Institutional PhD Program in Chemical Biology, New York, NY, USA; Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Yael David
- Tri-Institutional PhD Program in Chemical Biology, New York, NY, USA; Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Department of Pharmacology, Weill Cornell Medicine, New York, NY, USA; Department of Physiology, Biophysics, and Systems Biology, Weill Cornell Medicine, New York, NY, USA.
| |
Collapse
|
13
|
Nanogel-Facilitated In-Situ Delivery of a Cataract Inhibitor. Biomolecules 2021; 11:biom11081150. [PMID: 34439816 PMCID: PMC8391309 DOI: 10.3390/biom11081150] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Revised: 07/28/2021] [Accepted: 07/30/2021] [Indexed: 01/05/2023] Open
Abstract
Cataracts are a leading cause of blindness worldwide. Surgical removal of cataracts is a safe and effective procedure to restore vision. However, a large number of patients later develop vision loss due to regrowth of lens cells and subsequent degradation of the visual axis leading to visual disability. This postsurgical complication, known as posterior capsular opacification (PCO), occurs in up to 30% of cataract patients and has no clinically proven pharmacological means of prevention. Despite the availability of many compounds capable of preventing early steps in PCO development, there is currently no effective means to deliver such therapies into the eye for a suitable duration. To model a solution to this unmet medical need, we fabricated acrylic substrates as intraocular lens (IOL) mimics scaled to place into the capsular bag of the mouse lens following a mock-cataract surgery. Substrates were coated with a hydrophilic crosslinked acrylate nanogel designed to elute Sorbinil, an aldose reductase inhibitor previously shown to suppress PCO. Insertion of the Sorbinil-eluting device into the lens capsule at the time of cataract surgery resulted in substantial prevention of cellular changes associated with PCO development. This model demonstrates that a cataract inhibitor can be delivered into the postsurgical lens capsule at therapeutic levels.
Collapse
|
14
|
Petrash JM, Shieh B, Ammar DA, Pedler MG, Orlicky DJ. Diabetes-Independent Retinal Phenotypes in an Aldose Reductase Transgenic Mouse Model. Metabolites 2021; 11:metabo11070450. [PMID: 34357344 PMCID: PMC8305400 DOI: 10.3390/metabo11070450] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Revised: 06/21/2021] [Accepted: 06/26/2021] [Indexed: 11/16/2022] Open
Abstract
Aldose reductase (AR), the first and rate-limiting enzyme of the polyol pathway, has been implicated in the onset and development of the ocular complications of diabetes, including cataracts and retinopathy. Despite decades of research conducted to address possible mechanisms, questions still persist in understanding if or how AR contributes to imbalances leading to diabetic eye disease. To address these questions, we created a strain of transgenic mice engineered for the overexpression of human AR (AR-Tg). In the course of monitoring these animals for age-related retinal phenotypes, we observed signs of Müller cell gliosis characterized by strong immunostaining for glial fibrillary acidic protein. In addition, we observed increased staining for Iba1, consistent with an increase in the number of retinal microglia, a marker of retinal inflammation. Compared to age-matched nontransgenic controls, AR-Tg mice showed an age-dependent loss of Brn3a-positive retinal ganglion cells and an associated decrease in PERG amplitude. Both RGC-related phenotypes were rescued in animals treated with Sorbinil in drinking water. These results support the hypothesis that increased levels of AR may be a risk factor for structural and functional changes known to accompany retinopathy in humans.
Collapse
Affiliation(s)
- Jonathan Mark Petrash
- Department of Ophthalmology, University of Colorado School of Medicine, 12800 E. 19th Ave., Aurora, CO 80045, USA; (B.S.); (M.G.P.)
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, 1635 Aurora Ct, Aurora, CO 80045, USA
- Correspondence:
| | - Biehuoy Shieh
- Department of Ophthalmology, University of Colorado School of Medicine, 12800 E. 19th Ave., Aurora, CO 80045, USA; (B.S.); (M.G.P.)
| | - David A. Ammar
- Lions Eye Institute for Transplant and Research, 1410 N 21st St, Tampa, FL 33605, USA;
| | - Michelle G. Pedler
- Department of Ophthalmology, University of Colorado School of Medicine, 12800 E. 19th Ave., Aurora, CO 80045, USA; (B.S.); (M.G.P.)
| | - David J. Orlicky
- Department of Pathology, University of Colorado School of Medicine, 12800 E. 19th Ave., Aurora, CO 80045, USA;
| |
Collapse
|
15
|
Protective Effect of Aldo-keto Reductase 1B1 Against Neuronal Cell Damage Elicited by 4'-Fluoro-α-pyrrolidinononanophenone. Neurotox Res 2021; 39:1360-1371. [PMID: 34043181 DOI: 10.1007/s12640-021-00380-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Revised: 05/17/2021] [Accepted: 05/21/2021] [Indexed: 10/21/2022]
Abstract
Chronic exposure to cathinone derivatives increases the risk of severe health hazards, whereas little is known about the detailed pathogenic mechanisms triggered by the derivatives. We have recently shown that treatment with α-pyrrolidinononanophenone (α-PNP, a highly lipophilic cathinone derivative possessing a long hydrocarbon main chain) provokes neuronal cell apoptosis and its 4'-fluorinated analog (F-α-PNP) potently augments the apoptotic effect. In this study, we found that neuronal SK-N-SH cell damage elicited by F-α-PNP treatment is augmented most potently by pre-incubation with an AKR1B1 inhibitor tolrestat, among specific inhibitors of four aldo-keto reductase (AKR) family members (1B1, 1C1, 1C2, and 1C3) expressed in the neuronal cells. In addition, forced overexpression of AKR1B1 remarkably lowered the cell sensitivity to F-α-PNP toxicity, clearly indicating that AKR1B1 protects from neurotoxicity of the derivative. Treatment of SK-N-SH cells with F-α-PNP resulted in a dose-dependent up-regulation of AKR1B1 expression and activation of its transcription factor NF-E2-related factor 2. Metabolic analyses using liquid chromatography/mass spectrometry/mass spectrometry revealed that AKR1B1 is hardly involved in the F-α-PNP metabolism. The F-α-PNP treatment resulted in production of reactive oxygen species and lipid peroxidation byproduct 4-hydroxy-2-nonenal (HNE) in the cells. The enhanced HNE level was reduced by overexpression of AKR1B1, which also lessened the cell damage elicited by HNE. These results suggest that the AKR1B1-mediated neuronal cell protection is due to detoxification of HNE formed by F-α-PNP treatment, but not to metabolism of the derivative.
Collapse
|
16
|
Endo S, Matsunaga T, Nishinaka T. The Role of AKR1B10 in Physiology and Pathophysiology. Metabolites 2021; 11:332. [PMID: 34063865 PMCID: PMC8224097 DOI: 10.3390/metabo11060332] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Revised: 05/17/2021] [Accepted: 05/19/2021] [Indexed: 12/16/2022] Open
Abstract
AKR1B10 is a human nicotinamide adenine dinucleotide phosphate (NADPH)-dependent reductase belonging to the aldo-keto reductase (AKR) 1B subfamily. It catalyzes the reduction of aldehydes, some ketones and quinones, and interacts with acetyl-CoA carboxylase and heat shock protein 90α. The enzyme is highly expressed in epithelial cells of the stomach and intestine, but down-regulated in gastrointestinal cancers and inflammatory bowel diseases. In contrast, AKR1B10 expression is low in other tissues, where the enzyme is upregulated in cancers, as well as in non-alcoholic fatty liver disease and several skin diseases. In addition, the enzyme's expression is elevated in cancer cells resistant to clinical anti-cancer drugs. Thus, growing evidence supports AKR1B10 as a potential target for diagnosing and treating these diseases. Herein, we reviewed the literature on the roles of AKR1B10 in a healthy gastrointestinal tract, the development and progression of cancers and acquired chemoresistance, in addition to its gene regulation, functions, and inhibitors.
Collapse
Affiliation(s)
- Satoshi Endo
- Laboratory of Biochemistry, Gifu Pharmaceutical University, Gifu 501-1196, Japan
| | - Toshiyuki Matsunaga
- Education Center of Green Pharmaceutical Sciences, Gifu Pharmaceutical University, Gifu 502-8585, Japan;
| | - Toru Nishinaka
- Laboratory of Biochemistry, Faculty of Pharmacy, Osaka Ohtani University, Tondabayashi 584-8540, Osaka, Japan;
| |
Collapse
|
17
|
Hupy ML, Pedler MG, Shieh B, Wang D, Wang XJ, Petrash JM. Suppression of epithelial to mesenchymal transition markers in mouse lens by a Smad7-based recombinant protein. Chem Biol Interact 2021; 344:109495. [PMID: 33961834 DOI: 10.1016/j.cbi.2021.109495] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Revised: 04/14/2021] [Accepted: 04/22/2021] [Indexed: 12/17/2022]
Abstract
Cataracts, a clouding of the eye lens, are a leading cause of visual impairment and are responsible for one of the most commonly performed surgical procedures worldwide. Although generally safe and effective, cataract surgery can lead to a secondary lens abnormality due to transition of lens epithelial cells to a mesenchymal phenotype (EMT) and opacification of the posterior lens capsular bag. Occurring in up to 40% of cataract cases over time, posterior capsule opacification (PCO) introduces additional treatment costs and reduced quality of life for patients. Studies have shown that PCO pathogenesis is driven in part by TGF-β, signaling through the action of the family of Smad coactivators to effect changes in gene transcription. In the present study, we evaluated the ability of Smad-7, a well characterized inhibitor of TGF-β -mediated Smad signaling, to suppress the EMT response in lens epithelial cells associated with PCO pathogenesis. Treatment of lens epithelial cells with a cell-permeable form of Smad7 variant resulted in suppressed expression of EMT markers such as alpha smooth muscle actin and fibronectin. A single application of cell-permeable Smad7 variant in the capsular bag of a mouse cataract surgery model resulted in suppression of gene transcripts encoding alpha smooth muscle actin and fibronectin. These results point to Smad7 as a promising biotherapeutic agent for prevention or substantial reduction in the incidence of PCO following cataract surgery.
Collapse
Affiliation(s)
- Matthew L Hupy
- Department of Ophthalmology, University of Colorado, Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Michelle G Pedler
- Department of Ophthalmology, University of Colorado, Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Biehuoy Shieh
- Department of Ophthalmology, University of Colorado, Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Dongyan Wang
- Department of Pathology, University of Colorado, Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Xiao-Jing Wang
- Department of Pathology, University of Colorado, Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - J Mark Petrash
- Department of Ophthalmology, University of Colorado, Anschutz Medical Campus, Aurora, CO, 80045, USA; Department of Pharmaceutical Sciences, University of Colorado, Anschutz Medical Campus, Aurora, CO, 80045, USA.
| |
Collapse
|
18
|
Cappiello M, Balestri F, Moschini R, Mura U, Del-Corso A. Intra-site differential inhibition of multi-specific enzymes. J Enzyme Inhib Med Chem 2020; 35:840-846. [PMID: 32208768 PMCID: PMC7144184 DOI: 10.1080/14756366.2020.1743988] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Revised: 03/12/2020] [Accepted: 03/12/2020] [Indexed: 12/17/2022] Open
Abstract
The ability to catalyse a reaction acting on different substrates, known as "broad-specificity" or "multi-specificity", and to catalyse different reactions at the same active site ("promiscuity") are common features among the enzymes. These properties appear to go against the concept of extreme specificity of the catalytic action of enzymes and have been re-evaluated in terms of evolution and metabolic adaptation. This paper examines the potential usefulness of a differential inhibitory action in the study of the susceptibility to inhibition of multi-specific or promiscuous enzymes acting on different substrates. Aldose reductase is a multi-specific enzyme that catalyses the reduction of both aldoses and hydrophobic cytotoxic aldehydes and is used here as a concrete case to deal with the differential inhibition approach.
Collapse
Affiliation(s)
- Mario Cappiello
- Department of Biology, Biochemistry Unit, University of Pisa, Pisa, Italy
- Interdepartmental Research Center Nutrafood “Nutraceuticals and Food for Health”, University of Pisa, Pisa, Italy
| | - Francesco Balestri
- Department of Biology, Biochemistry Unit, University of Pisa, Pisa, Italy
- Interdepartmental Research Center Nutrafood “Nutraceuticals and Food for Health”, University of Pisa, Pisa, Italy
| | - Roberta Moschini
- Department of Biology, Biochemistry Unit, University of Pisa, Pisa, Italy
- Interdepartmental Research Center Nutrafood “Nutraceuticals and Food for Health”, University of Pisa, Pisa, Italy
| | - Umberto Mura
- Department of Biology, Biochemistry Unit, University of Pisa, Pisa, Italy
| | - Antonella Del-Corso
- Department of Biology, Biochemistry Unit, University of Pisa, Pisa, Italy
- Interdepartmental Research Center Nutrafood “Nutraceuticals and Food for Health”, University of Pisa, Pisa, Italy
| |
Collapse
|
19
|
Franko A, Berti L, Hennenlotter J, Rausch S, Scharpf MO, de Angelis MH, Stenzl A, Birkenfeld AL, Peter A, Lutz SZ, Häring HU, Heni M. Transcript Levels of Aldo-Keto Reductase Family 1 Subfamily C (AKR1C) Are Increased in Prostate Tissue of Patients with Type 2 Diabetes. J Pers Med 2020; 10:jpm10030124. [PMID: 32932589 PMCID: PMC7564141 DOI: 10.3390/jpm10030124] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 09/03/2020] [Accepted: 09/09/2020] [Indexed: 02/07/2023] Open
Abstract
Aldo-keto reductase family 1 (AKR1) enzymes play a crucial role in diabetic complications. Since type 2 diabetes (T2D) is associated with cancer progression, we investigated the impact of diabetes on AKR1 gene expression in the context of prostate cancer (PCa) development. In this study, we analyzed benign (BEN) prostate and PCa tissue of patients with and without T2D. Furthermore, to replicate hyperglycemia in vitro, we treated the prostate adenocarcinoma cell line PC3 with increasing glucose concentrations. Gene expression was quantified using real-time qPCR. In the prostate tissue of patients with T2D, AKR1C1 and AKR1C2 transcripts were higher compared to samples of patients without diabetes. In PC3 cells, high glucose treatment induced the gene expression levels of AKR1C1, C2, and C3. Furthermore, both in human tissue and in PC3 cells, the transcript levels of AKR1C1, C2, and C3 showed positive associations with oncogenes, which are involved in proliferation processes and HIF1α and NFκB pathways. These results indicate that in the prostate glands of patients with T2D, hyperglycemia could play a pivotal role by inducing the expression of AKR1C1, C2, and C3. The higher transcript level of AKR1C was furthermore associated with upregulated HIF1α and NFκB pathways, which are major drivers of PCa carcinogenesis.
Collapse
Affiliation(s)
- Andras Franko
- Department of Internal Medicine IV, Division of Endocrinology, Diabetology and Nephrology, University Hospital Tübingen, 72076 Tübingen, Germany; (A.F.); (A.L.B.); (S.Z.L.); (H.-U.H.)
- Institute for Diabetes Research and Metabolic Diseases, Helmholtz Centre Munich, University of Tübingen, 72076 Tübingen, Germany;
- German Center for Diabetes Research (DZD), 85764 Neuherberg, Germany;
| | - Lucia Berti
- Institute for Diabetes Research and Metabolic Diseases, Helmholtz Centre Munich, University of Tübingen, 72076 Tübingen, Germany;
- German Center for Diabetes Research (DZD), 85764 Neuherberg, Germany;
| | - Jörg Hennenlotter
- Department of Urology, University Hospital Tübingen, 72076 Tübingen, Germany; (J.H.); (S.R.); (A.S.)
| | - Steffen Rausch
- Department of Urology, University Hospital Tübingen, 72076 Tübingen, Germany; (J.H.); (S.R.); (A.S.)
| | - Marcus O. Scharpf
- Institute of Pathology, University Hospital Tübingen, 72076 Tübingen, Germany;
| | - Martin Hrabĕ de Angelis
- German Center for Diabetes Research (DZD), 85764 Neuherberg, Germany;
- Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, 85764 Neuherberg, Germany
| | - Arnulf Stenzl
- Department of Urology, University Hospital Tübingen, 72076 Tübingen, Germany; (J.H.); (S.R.); (A.S.)
| | - Andreas L. Birkenfeld
- Department of Internal Medicine IV, Division of Endocrinology, Diabetology and Nephrology, University Hospital Tübingen, 72076 Tübingen, Germany; (A.F.); (A.L.B.); (S.Z.L.); (H.-U.H.)
- Institute for Diabetes Research and Metabolic Diseases, Helmholtz Centre Munich, University of Tübingen, 72076 Tübingen, Germany;
- German Center for Diabetes Research (DZD), 85764 Neuherberg, Germany;
| | - Andreas Peter
- Institute for Diabetes Research and Metabolic Diseases, Helmholtz Centre Munich, University of Tübingen, 72076 Tübingen, Germany;
- German Center for Diabetes Research (DZD), 85764 Neuherberg, Germany;
- Institute for Clinical Chemistry and Pathobiochemistry, Department for Diagnostic Laboratory Medicine, University Hospital Tübingen, 72076 Tübingen, Germany; (A.P.)
| | - Stefan Z. Lutz
- Department of Internal Medicine IV, Division of Endocrinology, Diabetology and Nephrology, University Hospital Tübingen, 72076 Tübingen, Germany; (A.F.); (A.L.B.); (S.Z.L.); (H.-U.H.)
- Clinic for Geriatric and Orthopedic Rehabilitation Bad Sebastiansweiler, 72116 Mössingen, Germany
| | - Hans-Ulrich Häring
- Department of Internal Medicine IV, Division of Endocrinology, Diabetology and Nephrology, University Hospital Tübingen, 72076 Tübingen, Germany; (A.F.); (A.L.B.); (S.Z.L.); (H.-U.H.)
- Institute for Diabetes Research and Metabolic Diseases, Helmholtz Centre Munich, University of Tübingen, 72076 Tübingen, Germany;
- German Center for Diabetes Research (DZD), 85764 Neuherberg, Germany;
| | - Martin Heni
- Department of Internal Medicine IV, Division of Endocrinology, Diabetology and Nephrology, University Hospital Tübingen, 72076 Tübingen, Germany; (A.F.); (A.L.B.); (S.Z.L.); (H.-U.H.)
- Institute for Diabetes Research and Metabolic Diseases, Helmholtz Centre Munich, University of Tübingen, 72076 Tübingen, Germany;
- German Center for Diabetes Research (DZD), 85764 Neuherberg, Germany;
- Institute for Clinical Chemistry and Pathobiochemistry, Department for Diagnostic Laboratory Medicine, University Hospital Tübingen, 72076 Tübingen, Germany; (A.P.)
- Correspondence: ; Tel.: +49-7071-29-82714
| |
Collapse
|
20
|
Frankfater C, Bozeman SL, Hsu FF, Andley UP. Alpha-crystallin mutations alter lens metabolites in mouse models of human cataracts. PLoS One 2020; 15:e0238081. [PMID: 32833997 PMCID: PMC7446835 DOI: 10.1371/journal.pone.0238081] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2020] [Accepted: 08/10/2020] [Indexed: 12/24/2022] Open
Abstract
Cataracts are a major cause of blindness worldwide and commonly occur in individuals over 70 years old. Cataracts can also appear earlier in life due to genetic mutations. The lens proteins, αA- and αB-crystallins, are chaperone proteins that have important roles maintaining protein solubility to prevent cataract formation. Mutations in the CRYAA and CRYAB crystallin genes are associated with autosomal dominant early onset human cataracts. Although studies about the proteomic and genomic changes that occur in cataracts have been reported, metabolomics studies are very limited. Here, we directly investigated cataract metabolism using gas-chromatography-mass spectrometry (GC-MS) to analyze the metabolites in adult Cryaa-R49C and Cryab-R120G knock-in mouse lenses. The most abundant metabolites were myo-inositol, L-(+)-lactic acid, cholesterol, phosphate, glycerol phosphate, palmitic and 9-octadecenoic acids, α-D-mannopyranose, and β-D-glucopyranose. Cryaa-R49C knock-in mouse lenses had a significant decrease in the number of sugars and minor sterols, which occurred in concert with an increase in lactic acid. Cholesterol composition was unchanged. In contrast, Cryab-R120G knock-in lenses exhibited increased total amino acid content including valine, alanine, serine, leucine, isoleucine, glycine, and aspartic acid. Minor sterols, including cholest-7-en-3-ol and glycerol phosphate were decreased. These studies indicate that lenses from Cryaa-R49C and Cryab-R120G knock-in mice, which are models for human cataracts, have unique amino acid and metabolite profiles.
Collapse
Affiliation(s)
- Cheryl Frankfater
- Mass Spectrometry Resource, Division of Endocrinology, Diabetes, Metabolism, and Lipid Research, Department of Medicine, Washington University School of Medicine, St. Louis, MO, United States of America
| | - Stephanie L. Bozeman
- Departments of Ophthalmology and Visual Sciences, Washington University School of Medicine, St. Louis, MO, United States of America
| | - Fong-Fu Hsu
- Mass Spectrometry Resource, Division of Endocrinology, Diabetes, Metabolism, and Lipid Research, Department of Medicine, Washington University School of Medicine, St. Louis, MO, United States of America
| | - Usha P. Andley
- Departments of Ophthalmology and Visual Sciences, Washington University School of Medicine, St. Louis, MO, United States of America
| |
Collapse
|
21
|
Balestri F, Barracco V, Renzone G, Tuccinardi T, Pomelli CS, Cappiello M, Lessi M, Rotondo R, Bellina F, Scaloni A, Mura U, Del Corso A, Moschini R. Stereoselectivity of Aldose Reductase in the Reduction of Glutathionyl-Hydroxynonanal Adduct. Antioxidants (Basel) 2019; 8:antiox8100502. [PMID: 31652566 PMCID: PMC6827081 DOI: 10.3390/antiox8100502] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2019] [Revised: 10/18/2019] [Accepted: 10/21/2019] [Indexed: 12/11/2022] Open
Abstract
The formation of the adduct between the lipid peroxidation product 4-hydroxy-2-nonenal (HNE) and glutathione, which leads to the generation of 3-glutathionyl-4-hydroxynonane (GSHNE), is one of the main routes of HNE detoxification. The aldo-keto reductase AKR1B1 is involved in the reduction of the aldehydic group of both HNE and GSHNE. In the present study, the effect of chirality on the recognition by aldose reductase of HNE and GSHNE was evaluated. AKR1B1 discriminates very modestly between the two possible enantiomers of HNE as substrates. Conversely, a combined kinetic analysis of the glutathionyl adducts obtained starting from either 4R- or 4S-HNE and mass spectrometry analysis of GSHNE products obtained from racemic HNE revealed that AKR1B1 possesses a marked preference toward the 3S,4R-GSHNE diastereoisomer. Density functional theory and molecular modeling studies revealed that this diastereoisomer, besides having a higher tendency to be in an open aldehydic form (the one recognized by AKR1B1) in solution than other GSHNE diastereoisomers, is further stabilized in its open form by a specific interaction with the enzyme active site. The relevance of this stereospecificity to the final metabolic fate of GSHNE is discussed.
Collapse
Affiliation(s)
- Francesco Balestri
- Biochemistry Unit, Department of Biology, University of Pisa, via S. Zeno 51, 56127 Pisa, Italy.
- Interdepartmental Research Center Nutrafood "Nutraceuticals and Food for Health", University of Pisa, 56124 Pisa, Italy.
| | - Vito Barracco
- Biochemistry Unit, Department of Biology, University of Pisa, via S. Zeno 51, 56127 Pisa, Italy.
| | - Giovanni Renzone
- Proteomics & Mass Spectrometry Laboratory, ISPAAM-CNR, Via Argine 1085, 80147 Napoli, Italy.
| | - Tiziano Tuccinardi
- Department of Pharmacy, University of Pisa, via Bonanno 6, 56126 Pisa, Italy.
| | | | - Mario Cappiello
- Biochemistry Unit, Department of Biology, University of Pisa, via S. Zeno 51, 56127 Pisa, Italy.
- Interdepartmental Research Center Nutrafood "Nutraceuticals and Food for Health", University of Pisa, 56124 Pisa, Italy.
| | - Marco Lessi
- Department of Chemistry and Industrial Chemistry, University of Pisa, via G. Moruzzi, 13, 56124 Pisa, Italy.
| | - Rossella Rotondo
- Biochemistry Unit, Department of Biology, University of Pisa, via S. Zeno 51, 56127 Pisa, Italy.
| | - Fabio Bellina
- Department of Chemistry and Industrial Chemistry, University of Pisa, via G. Moruzzi, 13, 56124 Pisa, Italy.
| | - Andrea Scaloni
- Proteomics & Mass Spectrometry Laboratory, ISPAAM-CNR, Via Argine 1085, 80147 Napoli, Italy.
| | - Umberto Mura
- Biochemistry Unit, Department of Biology, University of Pisa, via S. Zeno 51, 56127 Pisa, Italy.
| | - Antonella Del Corso
- Biochemistry Unit, Department of Biology, University of Pisa, via S. Zeno 51, 56127 Pisa, Italy.
- Interdepartmental Research Center Nutrafood "Nutraceuticals and Food for Health", University of Pisa, 56124 Pisa, Italy.
| | - Roberta Moschini
- Biochemistry Unit, Department of Biology, University of Pisa, via S. Zeno 51, 56127 Pisa, Italy.
- Interdepartmental Research Center Nutrafood "Nutraceuticals and Food for Health", University of Pisa, 56124 Pisa, Italy.
| |
Collapse
|
22
|
Sugar Alcohols of Polyol Pathway Serve as Alarmins to Mediate Local-Systemic Innate Immune Communication in Drosophila. Cell Host Microbe 2019; 26:240-251.e8. [PMID: 31350199 DOI: 10.1016/j.chom.2019.07.001] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Revised: 05/30/2019] [Accepted: 07/02/2019] [Indexed: 12/18/2022]
Abstract
Interorgan immunological communication is critical to connect the local-systemic innate immune response and orchestrate a homeostatic host defense. However, the factors and their roles in this process remain unclear. We find Drosophila IMD response in guts can sequentially trigger a systemic IMD reaction in the fat body. Sugar alcohols of the polyol pathway are essential for the spatiotemporal regulation of gut-fat body immunological communication (GFIC). IMD activation in guts causes elevated levels of sorbitol and galactitol in hemolymph. Aldose reductase (AR) in hemocytes, the rate-limiting enzyme of the polyol pathway, is necessary and sufficient for the increase of plasma sugar alcohols. Sorbitol relays GFIC by subsequent activation of Metalloprotease 2, which cleaves PGRP-LC to activate IMD response in fat bodies. Thus, this work unveils how GFIC relies on the intermediate activation of the polyol pathway in hemolymph and demonstrates that AR provides a critical metabolic checkpoint in the global inflammatory response.
Collapse
|