1
|
Qiu R, Yang M, Jin X, Liu J, Wang W, Zhang X, Han J, Lei B. AAV2-PDE6B restores retinal structure and function in the retinal degeneration 10 mouse model of retinitis pigmentosa by promoting phototransduction and inhibiting apoptosis. Neural Regen Res 2025; 20:2408-2419. [PMID: 39359097 DOI: 10.4103/nrr.nrr-d-23-01301] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Accepted: 01/30/2024] [Indexed: 10/04/2024] Open
Abstract
JOURNAL/nrgr/04.03/01300535-202508000-00030/figure1/v/2024-09-30T120553Z/r/image-tiff Retinitis pigmentosa is a group of inherited diseases that lead to retinal degeneration and photoreceptor cell death. However, there is no effective treatment for retinitis pigmentosa caused by PDE6B mutation. Adeno-associated virus (AAV)-mediated gene therapy is a promising strategy for treating retinitis pigmentosa. The aim of this study was to explore the molecular mechanisms by which AAV2-PDE6B rescues retinal function. To do this, we injected retinal degeneration 10 (rd10) mice subretinally with AAV2-PDE6B and assessed the therapeutic effects on retinal function and structure using dark- and light-adapted electroretinogram, optical coherence tomography, and immunofluorescence. Data-independent acquisition-mass spectrometry-based proteomic analysis was conducted to investigate protein expression levels and pathway enrichment, and the results from this analysis were verified by real-time polymerase chain reaction and western blotting. AAV2-PDE6B injection significantly upregulated PDE6β expression, preserved electroretinogram responses, and preserved outer nuclear layer thickness in rd10 mice. Differentially expressed proteins between wild-type and rd10 mice were closely related to visual perception, and treating rd10 mice with AAV2-PDE6B restored differentially expressed protein expression to levels similar to those seen in wild-type mice. Kyoto Encyclopedia of Genes and Genome analysis showed that the differentially expressed proteins whose expression was most significantly altered by AAV2-PDE6B injection were enriched in phototransduction pathways. Furthermore, the phototransduction-related proteins Pde6α, Rom1, Rho, Aldh1a1, and Rbp1 exhibited opposite expression patterns in rd10 mice with or without AAV2-PDE6B treatment. Finally, Bax/Bcl-2, p-ERK/ERK, and p-c-Fos/c-Fos expression levels decreased in rd10 mice following AAV2-PDE6B treatment. Our data suggest that AAV2-PDE6B-mediated gene therapy promotes phototransduction and inhibits apoptosis by inhibiting the ERK signaling pathway and upregulating Bcl-2/Bax expression in retinitis pigmentosa.
Collapse
Affiliation(s)
- Ruiqi Qiu
- Henan Eye Hospital, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
- Eye Institute, Henan Academy of Innovations in Medical Science, Zhengzhou, Henan Province, China
| | - Mingzhu Yang
- Henan Eye Hospital, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
- Eye Institute, Henan Academy of Innovations in Medical Science, Zhengzhou, Henan Province, China
| | - Xiuxiu Jin
- Henan Eye Hospital, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
- Eye Institute, Henan Academy of Innovations in Medical Science, Zhengzhou, Henan Province, China
- Branch of National Clinical Research Center for Ocular Disease, Henan Provincial People's Hospital, Zhengzhou, Henan Province, China
| | - Jingyang Liu
- Henan Eye Hospital, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
- Eye Institute, Henan Academy of Innovations in Medical Science, Zhengzhou, Henan Province, China
| | - Weiping Wang
- Henan Eye Hospital, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
- Eye Institute, Henan Academy of Innovations in Medical Science, Zhengzhou, Henan Province, China
| | - Xiaoli Zhang
- Henan Eye Hospital, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
- Academy of Medical Science, Zhengzhou University, Zhengzhou, Henan Province, China
| | - Jinfeng Han
- Henan Eye Hospital, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
- Academy of Medical Science, Zhengzhou University, Zhengzhou, Henan Province, China
| | - Bo Lei
- Henan Eye Hospital, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
- Eye Institute, Henan Academy of Innovations in Medical Science, Zhengzhou, Henan Province, China
- Branch of National Clinical Research Center for Ocular Disease, Henan Provincial People's Hospital, Zhengzhou, Henan Province, China
- Academy of Medical Science, Zhengzhou University, Zhengzhou, Henan Province, China
| |
Collapse
|
2
|
Tong J, Yang L, Liu Y, Yu Y, Zhang L, Zhang Z, Yang Z, Qin Q, Niu J, Gu Y. Empagliflozin attenuates renal tubular ferroptosis in preeclampsia via tazarotene-induced gene 1. Eur J Pharmacol 2025; 986:177140. [PMID: 39551334 DOI: 10.1016/j.ejphar.2024.177140] [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: 11/13/2023] [Revised: 11/07/2024] [Accepted: 11/14/2024] [Indexed: 11/19/2024]
Abstract
Preeclampsia (PE) is a serious pregnancy complication characterized by elevated blood pressure and a major cause of maternal and perinatal morbidities, also known to increase the risk of chronic kidney disease. Mechanisms underlying PE-induced kidney injury remain unclear. Anti-angiotensin II type 1 receptor agonistic autoantibody (AT1-AA) is reported to participate in the pathogenesis of PE-induced kidney injury. Our previous study replicated the major features of PE in pregnant mice by administration of intravenous injection of AT1-AA and found that podocyte senescence plays a role in PE-induced kidney injury. Elevated levels of N-acetyl-β-D glucosaminidase (NAG) and kidney injury molecule-1 (KIM-1) in the urine of patients with PE have been reported, indicating renal tubular injury. In this study, we identified the role of renal proximal tubular epithelial cells (PTECs) in PE-induced kidney injury and the therapeutic value of empagliflozin, an anti-diabetic agent, in a murine model of AT1-AA-induced PE. In our study, higher tubular injury score (Control vs. PE: P < 0.0001) show that PTECs are damaged in AT1-AA-induced PE. We identified ferroptosis as one of the cause of AT1-AA-induced PTEC injury by RNAseq, and confirmed the involvement of ferroptosis by detecting ferrous iron (Control vs. PE: P < 0.0001), reduced glutathione (GSH) (Control vs. PE: P < 0.0001) and lipid peroxidation (Control vs. PE: P < 0.0001). Empagliflozin ameliorates AT1-AA-induced PTEC ferroptosis and injury in PE. Furthermore, we demonstrated that tazarotene-induced gene 1 is involved in AT1-AA-induced PTEC injury. These findings suggest that renal tubules are injured in PE and empagliflozin has therapeutic potential for PE-induced PTEC injury.
Collapse
Affiliation(s)
- Jiahao Tong
- Department of Nephrology, Shanghai Fifth People's Hospital, Fudan University, Shanghai, China
| | - Lin Yang
- Department of Nephrology, Shanghai Fifth People's Hospital, Fudan University, Shanghai, China
| | - Yuan Liu
- Department of Nephrology, Shanghai Fifth People's Hospital, Fudan University, Shanghai, China
| | - Ying Yu
- Department of Nephrology, Shanghai Fifth People's Hospital, Fudan University, Shanghai, China
| | - Lihong Zhang
- Department of Nephrology, Shanghai Fifth People's Hospital, Fudan University, Shanghai, China
| | - Zengzhen Zhang
- Department of Nephrology, Shanghai Fifth People's Hospital, Fudan University, Shanghai, China
| | - Zhenhao Yang
- Department of Nephrology, Shanghai Fifth People's Hospital, Fudan University, Shanghai, China
| | - Qiaojing Qin
- Department of Nephrology, Shanghai Fifth People's Hospital, Fudan University, Shanghai, China
| | - Jianying Niu
- Department of Nephrology, Shanghai Fifth People's Hospital, Fudan University, Shanghai, China.
| | - Yong Gu
- Department of Nephrology, Shanghai Fifth People's Hospital, Fudan University, Shanghai, China; Department of Nephrology, Huashan Hospital, Fudan University, Shanghai, China.
| |
Collapse
|
3
|
Fung L, Dranow DB, Subramanian A, Libby N, Schilling TF. Cellular retinoic acid-binding proteins regulate germ cell proliferation and sex determination in zebrafish. Development 2024; 151:dev202549. [PMID: 39575928 DOI: 10.1242/dev.202549] [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: 11/18/2023] [Accepted: 10/30/2024] [Indexed: 12/14/2024]
Abstract
Cellular retinoic acid (RA)-binding proteins (Crabps) solubilize intracellular RA and transport it to its nuclear receptors or cytoplasmic degradation enzymes. Despite their extreme conservation across chordates, genetic studies of Crabp function have revealed few essential functions. We have generated loss-of-function mutations in all four zebrafish Crabps and find essential roles for Crabp2 proteins in gonad development and sex determination. Transgenic RA reporters show strong RA responses in germ cells at the bipotential stage of gonad development. Double mutants lacking the functions of both Crabp2a and Crabp2b predominantly become male, which correlates with their smaller gonad size and reduced germ cell proliferation during gonad development at late larval and early juvenile stages. In contrast, mutants lacking the functions of both Crabp1a and Crabp1b have normal sex ratios. Exogenous RA treatments at bipotential gonad stages increase germ cell number, consistent with a direct role for RA in promoting germ cell proliferation. Our results suggest essential functions for Crabps in gonad development and sex determination.
Collapse
Affiliation(s)
- Lianna Fung
- Department of Developmental and Cell Biology, University of California, Irvine, CA 92697-2300, USA
| | - Daniel B Dranow
- Department of Developmental and Cell Biology, University of California, Irvine, CA 92697-2300, USA
| | - Arul Subramanian
- Department of Developmental and Cell Biology, University of California, Irvine, CA 92697-2300, USA
| | - Natalia Libby
- Department of Developmental and Cell Biology, University of California, Irvine, CA 92697-2300, USA
| | - Thomas F Schilling
- Department of Developmental and Cell Biology, University of California, Irvine, CA 92697-2300, USA
| |
Collapse
|
4
|
Tao L, He D, Chen Y, Yang K, He B, Cai P, Cai B, Liao C, Liu Z, Li S, Chen J, Wu Y. Transferrin ameliorates retinal degeneration by mediating the dimerization of all-trans-retinal. J Biol Chem 2024; 301:108054. [PMID: 39653244 DOI: 10.1016/j.jbc.2024.108054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Revised: 11/26/2024] [Accepted: 11/29/2024] [Indexed: 12/30/2024] Open
Abstract
High levels of all-trans-retinal (atRAL) in the retina is considered to be responsible for the development of autosomal recessive Stargardt's disease (STGD1) and dry age-related macular degeneration (dAMD). Two bisretinoids, all-trans-retinal dimer (atRAL-dimer) and N-retinyl-N-retinylidene ethanolamine (A2E), form from the dimerization of atRAL in the retina but they possess much lower toxicity and phototoxicity toward retinal pigment epithelium (RPE) cells than atRAL. Here, we introduced a novel function of transferrin (TRF) in mediating the conversion of atRAL into atRAL-dimer and A2E, which effectively protected the retina from damage by atRAL and prevented retinal function decline in mice, and rescued atRAL-loaded RPE cells. Moreover, TRF-mediated conversion of atRAL to atRAL-dimer and A2E required the help of bicarbonate ions (HCO3-). atRAL had the capacity to stimulate the expression of TRF in RPE and photoreceptor cells as well as RPE/choroid and neural retina of mice, reflecting that the elevation of TRF levels by atRAL is most likely to help defy level increase and cytotoxicity of atRAL through facilitating its dimerization and thereby serves as a mechanism of retinal self-protection. Our findings offer a promising avenue for the treatment of retinopathies characterized by disrupted clearance of atRAL.
Collapse
Affiliation(s)
- Lei Tao
- Department of Ophthalmology, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, China; Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Fujian Engineering and Research Center of Eye Regenerative Medicine, Eye Institute of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Danxue He
- Department of Ophthalmology, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, China; Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Fujian Engineering and Research Center of Eye Regenerative Medicine, Eye Institute of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Yuling Chen
- Department of Ophthalmology, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, China; Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Fujian Engineering and Research Center of Eye Regenerative Medicine, Eye Institute of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Kunhuan Yang
- Department of Ophthalmology, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, China; Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Fujian Engineering and Research Center of Eye Regenerative Medicine, Eye Institute of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Beiting He
- Department of Ophthalmology, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, China; Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Fujian Engineering and Research Center of Eye Regenerative Medicine, Eye Institute of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Peixin Cai
- Department of Ophthalmology, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, China; Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Fujian Engineering and Research Center of Eye Regenerative Medicine, Eye Institute of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Binxiang Cai
- Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Fujian Engineering and Research Center of Eye Regenerative Medicine, Eye Institute of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Chunyan Liao
- Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Fujian Engineering and Research Center of Eye Regenerative Medicine, Eye Institute of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Zuguo Liu
- Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Fujian Engineering and Research Center of Eye Regenerative Medicine, Eye Institute of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Shiying Li
- Department of Ophthalmology, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Jingmeng Chen
- School of Medicine, Xiamen University, Xiamen, Fujian, China; Shenzhen Research Institute of Xiamen University, Shenzhen, Guangdong, China
| | - Yalin Wu
- Department of Ophthalmology, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, China; Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Fujian Engineering and Research Center of Eye Regenerative Medicine, Eye Institute of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, China; Shenzhen Research Institute of Xiamen University, Shenzhen, Guangdong, China.
| |
Collapse
|
5
|
Pastok MW, Tomlinson CWE, Turberville S, Butler AM, Baslé A, Noble MEM, Endicott JA, Pohl E, Tatum NJ. Structural requirements for the specific binding of CRABP2 to cyclin D3. Structure 2024; 32:2301-2315.e6. [PMID: 39419021 DOI: 10.1016/j.str.2024.09.020] [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: 12/15/2021] [Revised: 07/31/2024] [Accepted: 09/20/2024] [Indexed: 10/19/2024]
Abstract
Cellular retinoic acid binding protein 2 (CRABP2) transports retinoic acid from the cytoplasm to the nucleus where it then transfers its cargo to retinoic acid receptor-containing complexes leading to activation of gene transcription. We demonstrate using purified proteins that CRABP2 is also a cyclin D3-specific binding protein and that the CRABP2 cyclin D3 binding site and the proposed CRABP2 nuclear localization sequence overlap. Both sequences are within the helix-loop-helix motif that forms a lid to the retinoic acid binding pocket. Mutations within this sequence that block both cyclin D3 and retinoic acid binding promote formation of a CRABP2 structure in which the retinoic acid binding pocket is occupied by an alternative lid conformation. Structural and functional analysis of CRABP2 and cyclin D3 mutants combined with AlphaFold models of the ternary CDK4/6-cyclin D3-CRABP2 complex supports the identification of an α-helical protein binding site on the cyclin D3 C-terminal cyclin box fold.
Collapse
Affiliation(s)
- Martyna W Pastok
- Newcastle University Centre for Cancer, Translational and Clinical Research Institute, Newcastle University, Paul O'Gorman Building, Framlington Place, Newcastle upon Tyne NE2 4HH, UK.
| | - Charles W E Tomlinson
- Department of Chemistry, Durham University, Lower Mountjoy, South Road, Durham DH1 3LE, UK
| | - Shannon Turberville
- Newcastle University Centre for Cancer, Translational and Clinical Research Institute, Newcastle University, Paul O'Gorman Building, Framlington Place, Newcastle upon Tyne NE2 4HH, UK
| | - Abbey M Butler
- Department of Chemistry, Durham University, Lower Mountjoy, South Road, Durham DH1 3LE, UK
| | - Arnaud Baslé
- Biosciences Institute, Newcastle University, Framlington Place, Newcastle upon Tyne NE2 4HH, UK
| | - Martin E M Noble
- Newcastle University Centre for Cancer, Translational and Clinical Research Institute, Newcastle University, Paul O'Gorman Building, Framlington Place, Newcastle upon Tyne NE2 4HH, UK
| | - Jane A Endicott
- Newcastle University Centre for Cancer, Translational and Clinical Research Institute, Newcastle University, Paul O'Gorman Building, Framlington Place, Newcastle upon Tyne NE2 4HH, UK.
| | - Ehmke Pohl
- Department of Chemistry, Durham University, Lower Mountjoy, South Road, Durham DH1 3LE, UK; Department of Biosciences, Durham University, Upper Mountjoy, South Road, Durham DH1 3LE, UK
| | - Natalie J Tatum
- Newcastle University Centre for Cancer, Translational and Clinical Research Institute, Newcastle University, Paul O'Gorman Building, Framlington Place, Newcastle upon Tyne NE2 4HH, UK.
| |
Collapse
|
6
|
Yee C, Bartölke R, Görtemaker K, Schmidt J, Leberecht B, Mouritsen H, Koch KW. Comparison of retinol binding protein 1 with cone specific G-protein as putative effector molecules in cryptochrome signalling. Sci Rep 2024; 14:28326. [PMID: 39550406 PMCID: PMC11569197 DOI: 10.1038/s41598-024-79699-z] [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: 04/15/2024] [Accepted: 11/11/2024] [Indexed: 11/18/2024] Open
Abstract
Vision and magnetoreception in navigating songbirds are strongly connected as recent findings link a light dependent radical-pair mechanism in cryptochrome proteins to signalling pathways in cone photoreceptor cells. A previous yeast-two-hybrid screening approach identified six putative candidate proteins showing binding to cryptochrome type 4a. So far, only the interaction of the cone specific G-protein transducin α-subunit was investigated in more detail. In the present study, we compare the binding features of the G-protein α-subunit with those of another candidate from the yeast-two-hybrid screen, cellular retinol binding protein. Purified recombinant European robin retinol binding protein bound retinol with high affinity, displaying an EC50 of less than 5 nM, thereby demonstrating its functional state. We applied surface plasmon resonance and a Förster resonance transfer analysis to test for interactions between retinol binding protein and cryptochrome 4a. In the absence of retinol, we observed no robust binding events, which contrasts the strong interaction we observed between cryptochrome 4a and the G-protein α-subunit. We conclude that retinol binding protein is unlikely to be involved in the primary magnetosensory signalling cascade.
Collapse
Affiliation(s)
- Chad Yee
- Division of Biochemistry, Department of Neuroscience, Carl von Ossietzky Universität Oldenburg, 26111, Oldenburg, Germany
| | - Rabea Bartölke
- Neurosensorics/Animal Navigation, Institute of Biology and Environmental Sciences, Carl von Ossietzky Universität Oldenburg, 26111, Oldenburg, Germany
| | - Katharina Görtemaker
- Division of Biochemistry, Department of Neuroscience, Carl von Ossietzky Universität Oldenburg, 26111, Oldenburg, Germany
| | - Jessica Schmidt
- Neurosensorics/Animal Navigation, Institute of Biology and Environmental Sciences, Carl von Ossietzky Universität Oldenburg, 26111, Oldenburg, Germany
| | - Bo Leberecht
- Animal Biodiversity and Evolutionary Biology, Institute of Biology and Environmental Sciences, Carl von Ossietzky Universität Oldenburg, 26111, Oldenburg, Germany
| | - Henrik Mouritsen
- Neurosensorics/Animal Navigation, Institute of Biology and Environmental Sciences, Carl von Ossietzky Universität Oldenburg, 26111, Oldenburg, Germany.
- Research Center for Neurosensory Sciences, Carl von Ossietzky Universität Oldenburg, 26111, Oldenburg, Germany.
| | - Karl-Wilhelm Koch
- Division of Biochemistry, Department of Neuroscience, Carl von Ossietzky Universität Oldenburg, 26111, Oldenburg, Germany.
- Research Center for Neurosensory Sciences, Carl von Ossietzky Universität Oldenburg, 26111, Oldenburg, Germany.
| |
Collapse
|
7
|
Walczak-Szeffer A, Piastowska-Ciesielska AW. Endoplasmic reticulum stress as a target for retinoids in cancer treatment. Life Sci 2024; 352:122892. [PMID: 38971363 DOI: 10.1016/j.lfs.2024.122892] [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: 02/29/2024] [Revised: 06/21/2024] [Accepted: 07/03/2024] [Indexed: 07/08/2024]
Abstract
Retinoids, natural and synthetic derivatives of vitamin A, have various regulatory activities including controlling cellular proliferation, differentiation, and death. Furthermore, they have been used to treat specific cancers with satisfying results. Nevertheless, retinoids have yet to be converted into effective systemic therapies for the majority of tumor types. Regulation of unfolded protein response signaling, and persistent activation of endoplasmic reticulum stress (ER-stress) are promising treatment methods for cancer. The present article reviews the current understanding of how vitamin A and its derivatives may aid to cause ER-stress-activated apoptosis, as well as therapeutic options for exploiting ER-stress for achieving beneficial goal. The therapeutic use of some retinoids discussed in this article was related to decreased disease recurrence and improved therapeutic outcomes via ER-stress activation and promotion, indicating that retinoids may play an important role in cancer treatment and prevention. More research is needed to expand the use of vitamin A derivatives in cancer therapy, either alone or in combination with unfolded protein response inducers.
Collapse
Affiliation(s)
- Anna Walczak-Szeffer
- Department of Cell Cultures and Genomic Analysis, Medical University of Lodz, Poland.
| | | |
Collapse
|
8
|
Mambwe B, Mellody KT, Kiss O, O'Connor C, Bell M, Watson REB, Langton AK. Cosmetic retinoid use in photoaged skin: A review of the compounds, their use and mechanisms of action. Int J Cosmet Sci 2024. [PMID: 39128883 DOI: 10.1111/ics.13013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Revised: 07/22/2024] [Accepted: 07/22/2024] [Indexed: 08/13/2024]
Abstract
The inevitable attrition of skin due to ultraviolet radiation, termed photoaging, can be partially restored by treatment with retinoid compounds. Photoaged skin in lightly pigmented individuals, clinically presents with the appearance of wrinkles, increased laxity, and hyper- and hypopigmentation. Underlying these visible signs of ageing are histological features such as epidermal thinning, dermal-epidermal junction flattening, solar elastosis and loss of the dermal fibrillin microfibrillar network, fibrillar collagen and glycosaminoglycans. Retinoid compounds are comprised of three main generations with the first generation (all-trans retinoic acid, retinol, retinaldehyde and retinyl esters) primarily used for the clinical and cosmetic treatment of photoaging, with varying degrees of efficacy, tolerance and stability. All-trans retinoic acid is considered the 'gold standard' for skin rejuvenation; however, it is a prescription-only product largely confined to clinical use. Therefore, retinoid derivatives are readily incorporated into cosmeceutical formulations. The literature reported in this review suggests that retinol, retinyl esters and retinaldehyde that are used in many cosmeceutical products, are efficacious, safe and well-tolerated. Once in the skin, retinoids utilize a complex signalling pathway that promotes remodelling of photoaged epidermis and dermis and leads to the improvement of the cutaneous signs of photoaging.
Collapse
Affiliation(s)
- Bezaleel Mambwe
- Centre for Dermatology Research, Salford Royal NHS Foundation Trust, Manchester Academic Health Science Centre, The University of Manchester, Manchester, UK
| | - Kieran T Mellody
- Centre for Dermatology Research, Salford Royal NHS Foundation Trust, Manchester Academic Health Science Centre, The University of Manchester, Manchester, UK
| | - Orsolya Kiss
- Centre for Dermatology Research, Salford Royal NHS Foundation Trust, Manchester Academic Health Science Centre, The University of Manchester, Manchester, UK
| | - Clare O'Connor
- No7 Beauty Company, Walgreens Boots Alliance, Nottingham, UK
| | - Mike Bell
- No7 Beauty Company, Walgreens Boots Alliance, Nottingham, UK
| | - Rachel E B Watson
- Centre for Dermatology Research, Salford Royal NHS Foundation Trust, Manchester Academic Health Science Centre, The University of Manchester, Manchester, UK
- A*STAR Skin Research Laboratory (A*SRL), Agency for Science, Technology and Research (A*STAR), Singapore City, Singapore
| | - Abigail K Langton
- Centre for Dermatology Research, Salford Royal NHS Foundation Trust, Manchester Academic Health Science Centre, The University of Manchester, Manchester, UK
| |
Collapse
|
9
|
Manoochehri H, Farrokhnia M, Sheykhhasan M, Mahaki H, Tanzadehpanah H. Key target genes related to anti-breast cancer activity of ATRA: A network pharmacology, molecular docking and experimental investigation. Heliyon 2024; 10:e34300. [PMID: 39108872 PMCID: PMC11301165 DOI: 10.1016/j.heliyon.2024.e34300] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Revised: 07/05/2024] [Accepted: 07/08/2024] [Indexed: 01/07/2025] Open
Abstract
All-trans retinoic acid (ATRA) has promising activity against breast cancer. However, the exact mechanisms of ATRA's anticancer effects remain complex and not fully understood. In this study, a network pharmacology and molecular docking approach was applied to identify key target genes related to ATRA's anti-breast cancer activity. Gene/disease enrichment analysis for predicted ATRA targets was performed using the Database for Annotation, Visualization and Integrated Discovery (DAVID), the Comparative Toxicogenomics Database (CTD), and the Gene Set Cancer Analysis (GSCA) database. Protein-Protein Interaction Network (PPIN) generation and analysis was conducted via Search Tool for the Retrieval of Interacting Genes/Proteins (STRING) and cytoscape, respectively. Cancer-associated genes were evaluated using MyGeneVenn from the CTD. Differential expression analysis was conducted using the Tumor, Normal, and Metastatic (TNM) Plot tool and the Human Protein Atlas (HPA). The Glide docking program was used to predict ligand-protein binding. Treatment response predication and clinical profile assessment were performed using Receiver Operating Characteristic (ROC) Plotter and OncoDB databases, respectively. Cytotoxicity and gene expression were measured using MTT/fluorescent assays and Real-Time PCR, respectively. Molecular functions of ATRA targets (n = 209) included eicosanoid receptor activity and transcription factor activity. Some enriched pathways included inclusion body myositis and nuclear receptors pathways. Network analysis revealed 35 hub genes contributing to 3 modules, with 16 of them were associated with breast cancer. These genes were involved in apoptosis, cell cycle, androgen receptor pathway, and ESR-mediated signaling, among others. CCND1, ESR1, MMP9, MDM2, NCOA3, and RARA were significantly overexpressed in tumor samples. ATRA showed a high affinity towards CCND1/CDK4 and MMP9. CCND1, ESR1, and MDM2 were associated with poor treatment response and were downregulated after treatment of the breast cancer cell line with ATRA. CCND1 and ESR1 exhibited differential expression across breast cancer stages. Therefore, some part of ATRA's anti-breast cancer activity may be exerted through the CCND1/CDK4 complex.
Collapse
Affiliation(s)
- Hamed Manoochehri
- The Persian Gulf Marine Biotechnology Research Center, The Persian Gulf Biomedical Sciences Research Institute, Bushehr University of Medical Sciences, Bushehr, Iran
| | - Maryam Farrokhnia
- The Persian Gulf Marine Biotechnology Research Center, The Persian Gulf Biomedical Sciences Research Institute, Bushehr University of Medical Sciences, Bushehr, Iran
| | - Mohsen Sheykhhasan
- Cellular and Molecular Research Center, Qom University of Medical Sciences, Qom, Iran
| | - Hanie Mahaki
- Vascular and Endovascular Surgery Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Hamid Tanzadehpanah
- Antimicrobial Resistance Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
10
|
Chen X, Peng R, Peng D, Liu D, Li R. Helicobacter pylori infection exacerbates metabolic dysfunction-associated steatotic liver disease through lipid metabolic pathways: a transcriptomic study. J Transl Med 2024; 22:701. [PMID: 39075482 PMCID: PMC11288106 DOI: 10.1186/s12967-024-05506-y] [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: 03/31/2024] [Accepted: 07/12/2024] [Indexed: 07/31/2024] Open
Abstract
BACKGROUND The relationship between Helicobacter pylori (H. pylori) infection and metabolic dysfunction-associated steatotic liver disease (MASLD) has attracted increased clinical attention. However, most of those current studies involve cross-sectional studies and meta-analyses, and experimental mechanistic exploration still needs to be improved. This study aimed to investigate the mechanisms by which H. pylori impacts MASLD. METHODS We established two H. pylori-infected (Cag A positive and Cag A negative) mouse models with 16 weeks of chow diet (CD) or high-fat diet (HFD) feeding. Body weight, liver triglyceride, blood glucose, serum biochemical parameters, inflammatory factors, and insulin resistance were measured, and histological analysis of liver tissues was performed. Mouse livers were subjected to transcriptome RNA sequencing analysis. RESULTS Although H. pylori infection could not significantly affect serum inflammatory factor levels and serum biochemical parameters in mice, serum insulin and homeostatic model assessment for insulin resistance levels increased in CD mode. In contrast, H. pylori Cag A + infection significantly aggravated hepatic pathological steatosis induced by HFD and elevated serum inflammatory factors and lipid metabolism parameters. Hepatic transcriptomic analysis in the CD groups revealed 767 differentially expressed genes (DEGs) in the H. pylori Cag A + infected group and 1473 DEGs in the H. pylori Cag A- infected group, and the "nonalcoholic fatty liver disease" pathway was significantly enriched in KEGG analysis. There were 578 DEGs in H. pylori Cag A + infection combined with the HFD feeding group and 820 DEGs in the H. pylori Cag A- infected group. DEGs in the HFD groups were significantly enriched in "fatty acid degradation" and "PPAR pathway." Exploring the effect of different Cag A statuses on mouse liver revealed that fatty acid binding protein 5 was differentially expressed in Cag A- H. pylori. DEG enrichment pathways were concentrated in the "PPAR pathway" and "fatty acid degradation." CONCLUSIONS Clinicians are expected to comprehend the impact of H. pylori on MASLD and better understand and manage MASLD. H. pylori infection may exacerbate the development of MASLD by regulating hepatic lipid metabolism, and the H. pylori virulence factor Cag A plays a vital role in this regulation.
Collapse
Affiliation(s)
- Xingcen Chen
- Department of Gastroenterology, The Second Xiangya Hospital of Central South University, No. 139 Middle Renmin Road, Changsha, 410011, Hunan Province, China
- Research Center of Digestive Diseases, Central South University, No. 139 Middle Renmin Road, Changsha, 410011, Hunan Province, China
- Clinical Research Center for Digestive Diseases in Hunan Province, Changsha, 410011, Hunan Province, China
| | - Ruyi Peng
- Department of Gastroenterology, The Second Xiangya Hospital of Central South University, No. 139 Middle Renmin Road, Changsha, 410011, Hunan Province, China
- Research Center of Digestive Diseases, Central South University, No. 139 Middle Renmin Road, Changsha, 410011, Hunan Province, China
- Clinical Research Center for Digestive Diseases in Hunan Province, Changsha, 410011, Hunan Province, China
| | - Dongzi Peng
- Department of Gastroenterology, The Second Xiangya Hospital of Central South University, No. 139 Middle Renmin Road, Changsha, 410011, Hunan Province, China
- Research Center of Digestive Diseases, Central South University, No. 139 Middle Renmin Road, Changsha, 410011, Hunan Province, China
- Clinical Research Center for Digestive Diseases in Hunan Province, Changsha, 410011, Hunan Province, China
| | - Deliang Liu
- Department of Gastroenterology, The Second Xiangya Hospital of Central South University, No. 139 Middle Renmin Road, Changsha, 410011, Hunan Province, China
- Research Center of Digestive Diseases, Central South University, No. 139 Middle Renmin Road, Changsha, 410011, Hunan Province, China
- Clinical Research Center for Digestive Diseases in Hunan Province, Changsha, 410011, Hunan Province, China
| | - Rong Li
- Department of Gastroenterology, The Second Xiangya Hospital of Central South University, No. 139 Middle Renmin Road, Changsha, 410011, Hunan Province, China.
- Research Center of Digestive Diseases, Central South University, No. 139 Middle Renmin Road, Changsha, 410011, Hunan Province, China.
- Clinical Research Center for Digestive Diseases in Hunan Province, Changsha, 410011, Hunan Province, China.
| |
Collapse
|
11
|
Nhieu J, Wei CW, Ludwig M, Drake JM, Wei LN. CRABP1-complexes in exosome secretion. Cell Commun Signal 2024; 22:381. [PMID: 39075476 PMCID: PMC11285139 DOI: 10.1186/s12964-024-01749-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: 05/10/2024] [Accepted: 07/13/2024] [Indexed: 07/31/2024] Open
Abstract
BACKGROUND Cellular retinoic acid binding protein 1 (CRABP1) mediates rapid, non-canonical activity of retinoic acid (RA) by forming signalosomes via protein-protein interactions. Two signalosomes have been identified previously: CRABP1-MAPK and CRABP1-CaMKII. Crabp1 knockout (CKO) mice exhibited altered exosome profiles, but the mechanism of CRABP1 action was unclear. This study aimed to screen for and identify novel CRABP1 signalosomes that could modulate exosome secretion by using a combinatorial approach involving biochemical, bioinformatic and molecular studies. METHODS Immunoprecipitation coupled with mass spectrometry (IP-MS) identified candidate CRABP1-interacting proteins which were subsequently analyzed using GO Term Enrichment, Functional Annotation Clustering; and Pathway Analysis. Gene expression analysis of CKO samples revealed altered expression of genes related to exosome biogenesis and secretion. The effect of CRABP1 on exosome secretion was then experimentally validated using CKO mice and a Crabp1 knockdown P19 cell line. RESULTS IP-MS identified CRABP1-interacting targets. Bioinformatic analyses revealed significant association with actin cytoskeletal dynamics, kinases, and exosome secretion. The effect of CRABP1 on exosome secretion was experimentally validated by comparing circulating exosome numbers of CKO and wild type (WT) mice, and secreted exosomes from WT and siCRABP1-P19 cells. Pathway analysis identified kinase signaling and Arp2/3 complex as the major pathways where CRABP1-signalosomes modulate exosome secretion, which was validated in the P19 system. CONCLUSION The combinatorial approach allowed efficient screening for and identification of novel CRABP1-signalosomes. The results uncovered a novel function of CRABP1 in modulating exosome secretion, and suggested that CRABP1 could play roles in modulating intercellular communication and signal propagation.
Collapse
Affiliation(s)
- Jennifer Nhieu
- Department of Pharmacology, University of Minnesota, 6-120 Jackson Hall, 321 Church St. SE, Minneapolis, MN, 55455, USA
| | - Chin-Wen Wei
- Department of Pharmacology, University of Minnesota, 6-120 Jackson Hall, 321 Church St. SE, Minneapolis, MN, 55455, USA
| | - Megan Ludwig
- Department of Pharmacology, University of Minnesota, 6-120 Jackson Hall, 321 Church St. SE, Minneapolis, MN, 55455, USA
| | - Justin M Drake
- Department of Pharmacology, University of Minnesota, 6-120 Jackson Hall, 321 Church St. SE, Minneapolis, MN, 55455, USA
| | - Li-Na Wei
- Department of Pharmacology, University of Minnesota, 6-120 Jackson Hall, 321 Church St. SE, Minneapolis, MN, 55455, USA.
| |
Collapse
|
12
|
Abraham E, Volmert B, Roule T, Huang L, Yu J, Williams AE, Cohen HM, Douglas A, Megill E, Morris A, Stronati E, Fueyo R, Zubillaga M, Elrod JW, Akizu N, Aguirre A, Estaras C. A Retinoic Acid:YAP1 signaling axis controls atrial lineage commitment. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.11.602981. [PMID: 39026825 PMCID: PMC11257518 DOI: 10.1101/2024.07.11.602981] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/20/2024]
Abstract
Vitamin A/Retinoic Acid (Vit A/RA) signaling is essential for heart development. In cardiac progenitor cells (CPCs), RA signaling induces the expression of atrial lineage genes while repressing ventricular genes, thereby promoting the acquisition of an atrial cardiomyocyte cell fate. To achieve this, RA coordinates a complex regulatory network of downstream effectors that is not fully identified. To address this gap, we applied a functional genomics approach (i.e scRNAseq and snATACseq) to untreated and RA-treated human embryonic stem cells (hESCs)-derived CPCs. Unbiased analysis revealed that the Hippo effectors YAP1 and TEAD4 are integrated with the atrial transcription factor enhancer network, and that YAP1 is necessary for activation of RA-enhancers in CPCs. Furthermore, in vivo analysis of control and conditionally YAP1 KO mouse embryos (Sox2-cre) revealed that the expression of atrial lineage genes, such as NR2F2, is compromised by YAP1 deletion in the CPCs of the second heart field. Accordingly, we found that YAP1 is required for the formation of an atrial chamber but is dispensable for the formation of a ventricle, in hESC-derived patterned cardiac organoids. Overall, our findings revealed that YAP1 is a non-canonical effector of RA signaling essential for the acquisition of atrial lineages during cardiogenesis.
Collapse
Affiliation(s)
- Elizabeth Abraham
- Department of Cardiovascular Sciences, Aging + Cardiovascular Discovery Center, Temple University, Lewis Katz School of Medicine, Philadelphia, PA, USA
| | - Brett Volmert
- Institute for Quantitative Health Science and Engineering, Division of Developmental and Stem Cell Biology, Michigan State University, East Lansing, MI, USA
| | - Thomas Roule
- Raymond G. Perelman Center for Cellular and Molecular Therapeutics, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Ling Huang
- Integrative Genomics and Bioinformatics Core, Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Jingting Yu
- Integrative Genomics and Bioinformatics Core, Salk Institute for Biological Studies, La Jolla, CA, USA
| | - April E Williams
- Integrative Genomics and Bioinformatics Core, Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Henry M Cohen
- Department of Cardiovascular Sciences, Aging + Cardiovascular Discovery Center, Temple University, Lewis Katz School of Medicine, Philadelphia, PA, USA
| | - Aidan Douglas
- Department of Cardiovascular Sciences, Aging + Cardiovascular Discovery Center, Temple University, Lewis Katz School of Medicine, Philadelphia, PA, USA
| | - Emily Megill
- Department of Cardiovascular Sciences, Aging + Cardiovascular Discovery Center, Temple University, Lewis Katz School of Medicine, Philadelphia, PA, USA
| | - Alex Morris
- Department of Cardiovascular Sciences, Aging + Cardiovascular Discovery Center, Temple University, Lewis Katz School of Medicine, Philadelphia, PA, USA
| | - Eleonora Stronati
- Department of Child and Adolescence Psychiatry, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Raquel Fueyo
- Department of Chemical and Systems Biology, Stanford University School of Medicine, Stanford, CA, USA
| | - Mikel Zubillaga
- Department of Cardiovascular Sciences, Aging + Cardiovascular Discovery Center, Temple University, Lewis Katz School of Medicine, Philadelphia, PA, USA
| | - John W Elrod
- Department of Cardiovascular Sciences, Aging + Cardiovascular Discovery Center, Temple University, Lewis Katz School of Medicine, Philadelphia, PA, USA
| | - Naiara Akizu
- Raymond G. Perelman Center for Cellular and Molecular Therapeutics, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Aitor Aguirre
- Institute for Quantitative Health Science and Engineering, Division of Developmental and Stem Cell Biology, Michigan State University, East Lansing, MI, USA
| | - Conchi Estaras
- Department of Cardiovascular Sciences, Aging + Cardiovascular Discovery Center, Temple University, Lewis Katz School of Medicine, Philadelphia, PA, USA
| |
Collapse
|
13
|
Kibalnyk Y, Afanasiev E, Noble RMN, Watson AES, Poverennaya I, Dittmann NL, Alexiou M, Goodkey K, Greenwell AA, Ussher JR, Adameyko I, Massey J, Graf D, Bourque SL, Stratton JA, Voronova A. The chromatin regulator Ankrd11 controls cardiac neural crest cell-mediated outflow tract remodeling and heart function. Nat Commun 2024; 15:4632. [PMID: 38951500 PMCID: PMC11217281 DOI: 10.1038/s41467-024-48955-1] [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: 02/19/2023] [Accepted: 05/17/2024] [Indexed: 07/03/2024] Open
Abstract
ANKRD11 (Ankyrin Repeat Domain 11) is a chromatin regulator and a causative gene for KBG syndrome, a rare developmental disorder characterized by multiple organ abnormalities, including cardiac defects. However, the role of ANKRD11 in heart development is unknown. The neural crest plays a leading role in embryonic heart development, and its dysfunction is implicated in congenital heart defects. We demonstrate that conditional knockout of Ankrd11 in the murine embryonic neural crest results in persistent truncus arteriosus, ventricular dilation, and impaired ventricular contractility. We further show these defects occur due to aberrant cardiac neural crest cell organization leading to outflow tract septation failure. Lastly, knockout of Ankrd11 in the neural crest leads to impaired expression of various transcription factors, chromatin remodelers and signaling pathways, including mTOR, BMP and TGF-β in the cardiac neural crest cells. In this work, we identify Ankrd11 as a regulator of neural crest-mediated heart development and function.
Collapse
Affiliation(s)
- Yana Kibalnyk
- Department of Medical Genetics, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, AB, T6G 2H7, Canada
- Women and Children's Health Research Institute, 5-083 Edmonton Clinic Health Academy, University of Alberta, 11405 87 Avenue NW, Edmonton, AB, T6G 1C9, Canada
| | - Elia Afanasiev
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, QC, H3A 2B4, Canada
| | - Ronan M N Noble
- Women and Children's Health Research Institute, 5-083 Edmonton Clinic Health Academy, University of Alberta, 11405 87 Avenue NW, Edmonton, AB, T6G 1C9, Canada
- Department of Pediatrics, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, AB, T6G 2G3, Canada
| | - Adrianne E S Watson
- Department of Medical Genetics, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, AB, T6G 2H7, Canada
- Women and Children's Health Research Institute, 5-083 Edmonton Clinic Health Academy, University of Alberta, 11405 87 Avenue NW, Edmonton, AB, T6G 1C9, Canada
| | - Irina Poverennaya
- Department of Neuroimmunology, Center for Brain Research, Medical University of Vienna, 1090, Vienna, Austria
| | - Nicole L Dittmann
- Department of Medical Genetics, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, AB, T6G 2H7, Canada
- Neuroscience and Mental Health Institute, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, AB, T6G 2E1, Canada
| | - Maria Alexiou
- Department of Dentistry, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, AB, T6G 2H7, Canada
| | - Kara Goodkey
- Department of Medical Genetics, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, AB, T6G 2H7, Canada
- Women and Children's Health Research Institute, 5-083 Edmonton Clinic Health Academy, University of Alberta, 11405 87 Avenue NW, Edmonton, AB, T6G 1C9, Canada
| | - Amanda A Greenwell
- Women and Children's Health Research Institute, 5-083 Edmonton Clinic Health Academy, University of Alberta, 11405 87 Avenue NW, Edmonton, AB, T6G 1C9, Canada
- Faculty of Pharmacy & Pharmaceutical Sciences, University of Alberta, Edmonton, Alberta, Edmonton, AB, T6G 2H1, Canada
| | - John R Ussher
- Women and Children's Health Research Institute, 5-083 Edmonton Clinic Health Academy, University of Alberta, 11405 87 Avenue NW, Edmonton, AB, T6G 1C9, Canada
- Faculty of Pharmacy & Pharmaceutical Sciences, University of Alberta, Edmonton, Alberta, Edmonton, AB, T6G 2H1, Canada
| | - Igor Adameyko
- Department of Neuroimmunology, Center for Brain Research, Medical University of Vienna, 1090, Vienna, Austria
- Department of Physiology and Pharmacology, Karolinska Institutet, 17177, Stockholm, Sweden
| | | | - Daniel Graf
- Department of Medical Genetics, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, AB, T6G 2H7, Canada
- Women and Children's Health Research Institute, 5-083 Edmonton Clinic Health Academy, University of Alberta, 11405 87 Avenue NW, Edmonton, AB, T6G 1C9, Canada
- Department of Dentistry, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, AB, T6G 2H7, Canada
| | - Stephane L Bourque
- Women and Children's Health Research Institute, 5-083 Edmonton Clinic Health Academy, University of Alberta, 11405 87 Avenue NW, Edmonton, AB, T6G 1C9, Canada
- Department of Anesthesiology & Pain Medicine, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, AB, T6G 2G3, Canada
| | - Jo Anne Stratton
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, QC, H3A 2B4, Canada
| | - Anastassia Voronova
- Department of Medical Genetics, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, AB, T6G 2H7, Canada.
- Women and Children's Health Research Institute, 5-083 Edmonton Clinic Health Academy, University of Alberta, 11405 87 Avenue NW, Edmonton, AB, T6G 1C9, Canada.
- Neuroscience and Mental Health Institute, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, AB, T6G 2E1, Canada.
- Department of Cell Biology, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, AB, T6G 2H7, Canada.
| |
Collapse
|
14
|
Powała K, Żołek T, Brown G, Kutner A. Molecular Interactions of Selective Agonists and Antagonists with the Retinoic Acid Receptor γ. Int J Mol Sci 2024; 25:6568. [PMID: 38928275 PMCID: PMC11203493 DOI: 10.3390/ijms25126568] [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: 05/21/2024] [Revised: 06/06/2024] [Accepted: 06/11/2024] [Indexed: 06/28/2024] Open
Abstract
All-trans retinoic acid (ATRA), the major active metabolite of all-trans retinol (vitamin A), is a key hormonal signaling molecule. In the adult organism, ATRA has a widespread influence on processes that are crucial to the growth and differentiation of cells and, in turn, the acquisition of mature cell functions. Therefore, there is considerable potential in the use of retinoids to treat diseases. ATRA binds to the retinoic acid receptors (RAR) which, as activated by ATRA, selectively regulate gene expression. There are three main RAR isoforms, RARα, RARβ, and RARγ. They each have a distinct role, for example, RARα and RARγ regulate myeloid progenitor cell differentiation and hematopoietic stem cell maintenance, respectively. Hence, targeting an isoform is crucial to developing retinoid-based therapeutics. In principle, this is exemplified when ATRA is used to treat acute promyelocytic leukemia (PML) and target RARα within PML-RARα oncogenic fusion protein. ATRA with arsenic trioxide has provided a cure for the once highly fatal leukemia. Recent in vitro and in vivo studies of RARγ have revealed the potential use of agonists and antagonists to treat diseases as diverse as cancer, heterotopic ossification, psoriasis, and acne. During the final drug development there may be a need to design newer compounds with added modifications to improve solubility, pharmacokinetics, or potency. At the same time, it is important to retain isotype specificity and activity. Examination of the molecular interactions between RARγ agonists and the ligand binding domain of RARγ has revealed aspects to ligand binding that are crucial to RARγ selectivity and compound activity and key to designing newer compounds.
Collapse
Affiliation(s)
- Katarzyna Powała
- Department of Organic and Physical Chemistry, Faculty of Pharmacy, Medical University of Warsaw, 1 Banacha, 02-097 Warsaw, Poland
| | - Teresa Żołek
- Department of Organic and Physical Chemistry, Faculty of Pharmacy, Medical University of Warsaw, 1 Banacha, 02-097 Warsaw, Poland
| | - Geoffrey Brown
- School of Biomedical Sciences, Institute of Clinical Sciences, College of Medical and Dental Sciences, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK;
| | - Andrzej Kutner
- Department of Drug Chemistry Pharmaceutical and Biomedical Analysis, Faculty of Pharmacy, Medical University of Warsaw, 1 Banacha, 02-097 Warsaw, Poland;
| |
Collapse
|
15
|
Wang D, Pei P, Shea F, Spinney R, Chang A, Lahann J, Mallery SR. Growth modulatory effects of fenretinide encompass keratinocyte terminal differentiation: a favorable outcome for oral squamous cell carcinoma chemoprevention. Carcinogenesis 2024; 45:436-449. [PMID: 38470060 PMCID: PMC11519021 DOI: 10.1093/carcin/bgae022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Revised: 02/19/2024] [Accepted: 03/11/2024] [Indexed: 03/13/2024] Open
Abstract
Oral squamous cell carcinoma (OSCC) is worldwide health problem associated with high morbidity and mortality. From both the patient and socioeconomic perspectives, prevention of progression of premalignant oral intraepithelial neoplasia (OIN) to OSCC is clearly the preferable outcome. Optimal OSCC chemopreventives possess a variety of attributes including high tolerability, bioavailability, efficacy and preservation of an intact surface epithelium. Terminal differentiation, which directs oral keratinocytes leave the proliferative pool to form protective cornified envelopes, preserves the protective epithelial barrier while concurrently eliminating growth-aberrant keratinocytes. This study employed human premalignant oral keratinocytes and an OSCC cell line to evaluate the differentiation-inducing capacity of the synthetic retinoid, fenretinide (4HPR). Full-thickness oral mucosal explants were evaluated for proof of concept differentiation studies. Results of this study characterize the ability of 4HPR to fulfill all requisite components for keratinocyte differentiation, i.e. nuclear import via binding to cellular RA binding protein-II (molecular modeling), binding to and subsequent activation of retinoic acid nuclear receptors (receptor activation assays), increased expression and translation of genes associated with keratinocyte differentiation [Reverse transcription polymerase chain reaction (RT-PCR), immunoblotting] upregulation of a transglutaminase enzyme essential for cornified envelope formation (transglutaminase 3, functional assay) and augmentation of terminal differentiation in human oral epithelial explants (image-analyses quantified corneocyte desquamation). These data build upon the chemoprevention repertoire of 4HPR that includes function as a small molecule kinase inhibitor and inhibition of essential mechanisms necessary for basement membrane invasion. An upcoming clinical trial, which will assess whether a 4HPR-releasing mucoadhesive patch induces histologic, clinical and molecular regression in OIN lesions, will provide essential clinical insights.
Collapse
Affiliation(s)
- Daren Wang
- Division of Oral Maxillofacial Pathology, College of Dentistry, The Ohio State University, Columbus, OH, USA
| | - Ping Pei
- Division of Oral Maxillofacial Pathology, College of Dentistry, The Ohio State University, Columbus, OH, USA
| | - Fortune Shea
- Division of Oral Maxillofacial Pathology, College of Dentistry, The Ohio State University, Columbus, OH, USA
| | - Richard Spinney
- Department of Chemistry and Biochemistry, The Ohio State University, Columbus, OH, USA
| | - Albert Chang
- Department of Chemical Engineering, Biointerfaces Institute, University of Michigan, Ann Arbor, MI, USA
- Department of Material Science and Engineering, Biointerfaces Institute, University of Michigan, Ann Arbor, MI, USA
- Department of Biomedical Engineering, Biointerfaces Institute, University of Michigan, Ann Arbor, MI, USA
- Department of Macromolecular Science and Engineering, Biointerfaces Institute, University of Michigan, Ann Arbor, MI, USA
| | - Joerg Lahann
- Department of Chemical Engineering, Biointerfaces Institute, University of Michigan, Ann Arbor, MI, USA
- Department of Material Science and Engineering, Biointerfaces Institute, University of Michigan, Ann Arbor, MI, USA
- Department of Biomedical Engineering, Biointerfaces Institute, University of Michigan, Ann Arbor, MI, USA
- Department of Macromolecular Science and Engineering, Biointerfaces Institute, University of Michigan, Ann Arbor, MI, USA
| | - Susan R Mallery
- Division of Oral Maxillofacial Pathology, College of Dentistry, The Ohio State University, Columbus, OH, USA
- The Ohio State University Comprehensive Cancer Center, Columbus, OH, USA
| |
Collapse
|
16
|
Yoshioka H, Ueta M, Fukuoka H, Yokoi N, Mizushima K, Naito Y, Kinoshita S, Sotozono C. Alteration of Gene Expression in Pathological Keratinization of the Ocular Surface. Invest Ophthalmol Vis Sci 2024; 65:37. [PMID: 38935029 PMCID: PMC11216254 DOI: 10.1167/iovs.65.6.37] [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: 12/16/2023] [Accepted: 06/03/2024] [Indexed: 06/28/2024] Open
Abstract
Purpose To investigate the molecular mechanism of pathological keratinization in the chronic phase of ocular surface (OS) diseases. Methods In this study, a comprehensive gene expression analysis was performed using oligonucleotide microarrays on OS epithelial cells obtained from three patients with pathological keratinization (Stevens-Johnson syndrome [n = 1 patient], ocular cicatricial pemphigoid [n = 1 patient], and anterior staphyloma [n = 1 patient]). The controls were three patients with conjunctivochalasis. The expression in some transcripts was confirmed using quantitative real-time PCR. Results Compared to the controls, 3118 genes were significantly upregulated by a factor of 2 or more than one-half in the pathological keratinized epithelial cells (analysis of variance P < 0.05). Genes involved in keratinization, lipid metabolism, and oxidoreductase were upregulated, while genes involved in cellular response, as well as known transcription factors (TFs), were downregulated. Those genes were further analyzed with respect to TFs and retinoic acid (RA) through gene ontology analysis and known reports. The expression of TFs MYBL2, FOXM1, and SREBF2, was upregulated, and the TF ELF3 was significantly downregulated. The expression of AKR1B15, RDH12, and CRABP2 (i.e., genes related to RA, which is known to suppress keratinization) was increased more than twentyfold, whereas the expression of genes RARB and RARRES3 was decreased by 1/50. CRABP2, RARB, and RARRES3 expression changes were also confirmed by qRT-PCR. Conclusions In pathological keratinized ocular surfaces, common transcript changes, including abnormalities in vitamin A metabolism, are involved in the mechanism of pathological keratinization.
Collapse
Affiliation(s)
- Hokoru Yoshioka
- Department of Ophthalmology, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Mayumi Ueta
- Department of Ophthalmology, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Hideki Fukuoka
- Department of Ophthalmology, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Norihiko Yokoi
- Department of Ophthalmology, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Katsura Mizushima
- Department of Human Immunology and Nutrition Science, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kamigyo-ku, Kyoto, Japan
| | - Yuji Naito
- Department of Human Immunology and Nutrition Science, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kamigyo-ku, Kyoto, Japan
| | - Shigeru Kinoshita
- Department of Frontier Medical Science and Technology for Ophthalmology, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Chie Sotozono
- Department of Ophthalmology, Kyoto Prefectural University of Medicine, Kyoto, Japan
| |
Collapse
|
17
|
Chen X, Tian J, Zhao C, Wu Y, Li J, Ji Z, Lian D, Jia Z, Chen X, Zhou Z, Zhu B, Hua Z. Resveratrol, a novel inhibitor of fatty acid binding protein 5, inhibits cervical cancer metastasis by suppressing fatty acid transport into nucleus and downstream pathways. Br J Pharmacol 2024; 181:1614-1634. [PMID: 38158217 DOI: 10.1111/bph.16308] [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: 07/27/2023] [Revised: 11/28/2023] [Accepted: 12/14/2023] [Indexed: 01/03/2024] Open
Abstract
BACKGROUND AND PURPOSE Because of cervical cancer (CC) metastasis, the prognosis of diagnosed patients is poor. However, the molecular mechanisms and therapeutic approach for metastatic CC remain elusive. EXPERIMENTAL APPROACH In this study, we first evaluated the effect of resveratrol (RSV) on CC cell migration and metastasis. Via an activity-based protein profiling (ABPP) approach, a photoaffinity probe of RSV (RSV-P) was synthesized, and the protein targets of RSV in HeLa cells were identified. Based on target information and subsequent in vivo and in vitro validation experiments, we finally elucidated the mechanism of RSV corresponding to its antimetastatic activity. KEY RESULTS The results showed that RSV concentration-dependently suppressed CC cell migration and metastasis. A list of proteins was identified as the targets of RSV, through the ABPP approach with RSV-P, among which fatty acid binding protein 5 (FABP5) attracted our attention based on The Cancer Genome Atlas (TCGA) database analysis. Subsequent knockout and overexpression experiments confirmed that RSV directly interacted with FABP5 to inhibit fatty acid transport into the nucleus, thereby suppressing downstream matrix metalloproteinase-2 (MMP2) and matrix metalloproteinase-9 (MMP9) expression, thus inhibiting CC metastasis. CONCLUSIONS AND IMPLICATIONS Our study confirmed the key role of FABP5 in CC metastasis and provided important target information for the design of therapeutic lead compounds for metastatic CC.
Collapse
Affiliation(s)
- Xiao Chen
- School of Biopharmacy, China Pharmaceutical University, Nanjing, China
| | - Jing Tian
- The State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, China
| | - Chunyuan Zhao
- The State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, China
| | - Yanhui Wu
- School of Biopharmacy, China Pharmaceutical University, Nanjing, China
| | - Jiahuang Li
- School of Biopharmacy, China Pharmaceutical University, Nanjing, China
| | - Zehan Ji
- School of Biopharmacy, China Pharmaceutical University, Nanjing, China
| | - Danchen Lian
- School of Biopharmacy, China Pharmaceutical University, Nanjing, China
| | - Zhibo Jia
- School of Biopharmacy, China Pharmaceutical University, Nanjing, China
| | - Xingyu Chen
- School of Biopharmacy, China Pharmaceutical University, Nanjing, China
| | - Zixin Zhou
- School of Biopharmacy, China Pharmaceutical University, Nanjing, China
| | - Bo Zhu
- School of Biopharmacy, China Pharmaceutical University, Nanjing, China
| | - Zichun Hua
- School of Biopharmacy, China Pharmaceutical University, Nanjing, China
- The State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, China
| |
Collapse
|
18
|
Zhang Z, Lu T, Li S, Zhao R, Li H, Zhang X, Li Y, Xia Y, Ni G. Acupuncture Extended the Thrombolysis Window by Suppressing Blood-Brain Barrier Disruption and Regulating Autophagy-Apoptosis Balance after Ischemic Stroke. Brain Sci 2024; 14:399. [PMID: 38672048 PMCID: PMC11048240 DOI: 10.3390/brainsci14040399] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2024] [Revised: 04/14/2024] [Accepted: 04/16/2024] [Indexed: 04/28/2024] Open
Abstract
BACKGROUND Ischemic stroke (IS) is one of the leading causes of death and disability worldwide. The narrow therapeutic window (within 4.5 h) and severe hemorrhagic potential limits therapeutic efficacy of recombinant tissue type plasminogen activator (rt-PA) intravenous thrombolysis for patients. Xingnao Kaiqiao (XNKQ) acupuncture is an integral part of traditional Chinese medicine, specifically designed to address acute ischemic stroke by targeting key acupoints such as Shuigou (GV26) and Neiguan (PC6). In this study, we explored the therapeutic potential of XNKQ acupuncture in extending the time window for thrombolysis and interrogated the molecular mechanisms responsible for this effect. METHODS The effect of extending the thrombolysis window by acupuncture was evaluated via TTC staining, neuronal score evaluation, hemorrhagic transformation assay, and H&E staining. RNA sequencing (RNA-seq) technology was performed to identify the therapeutic targets and intervention mechanisms of acupuncture. Evans blue staining and transmission electron microscopy were used to assess blood-brain barrier (BBB) integrity. Immunofluorescence staining and co-immunoprecipitation were performed to evaluate the level of autophagy and apoptosis and validate their interactions with BBB endothelial cells. RESULTS Acupuncture alleviated infarction and neurological deficits and extended the thrombolysis window to 6 h. The RNA-seq revealed 16 potential therapeutic predictors for acupuncture intervention, which related to suppressing inflammation and restoring the function of BBB and blood vessels. Furthermore, acupuncture suppressed BBB leakage and preserved tight junction protein expression. The protective effect was associated with regulation of the autophagy-apoptosis balance in BBB endothelial cells. Acupuncture intervention dissociated the Beclin1/Bcl-2 complex, thereby promoting autophagy and reducing apoptosis. CONCLUSION XNKQ acupuncture could serve as an adjunctive therapy for rt-PA thrombolysis, aiming to extend the therapeutic time window and mitigate ischemia-reperfusion injury. Acupuncture suppressed BBB disruption by regulating the autophagy-apoptosis balance, which in turn extended the therapeutic window of rt-PA in IS. These findings provide a rationale for further exploration of acupuncture as a complementary candidate co-administered with rt-PA.
Collapse
Affiliation(s)
- Zhihui Zhang
- College of Acupuncture-Moxibustion and Tuina, Nanjing University of Chinese Medicine, No. 138 Xianlin Avenue, Qixia District, Nanjing 210023, China; (Z.Z.)
- Key Laboratory of Acupuncture and Medicine Research of Ministry of Education, Nanjing University of Chinese Medicine, No. 138 Xianlin Avenue, Qixia District, Nanjing 210023, China
| | - Tianliang Lu
- College of Acupuncture-Moxibustion and Tuina, Nanjing University of Chinese Medicine, No. 138 Xianlin Avenue, Qixia District, Nanjing 210023, China; (Z.Z.)
- Key Laboratory of Acupuncture and Medicine Research of Ministry of Education, Nanjing University of Chinese Medicine, No. 138 Xianlin Avenue, Qixia District, Nanjing 210023, China
| | - Shanshan Li
- College of Acupuncture-Moxibustion and Tuina, Nanjing University of Chinese Medicine, No. 138 Xianlin Avenue, Qixia District, Nanjing 210023, China; (Z.Z.)
- Key Laboratory of Acupuncture and Medicine Research of Ministry of Education, Nanjing University of Chinese Medicine, No. 138 Xianlin Avenue, Qixia District, Nanjing 210023, China
| | - Ruyu Zhao
- College of Acupuncture-Moxibustion and Tuina, Nanjing University of Chinese Medicine, No. 138 Xianlin Avenue, Qixia District, Nanjing 210023, China; (Z.Z.)
- Key Laboratory of Acupuncture and Medicine Research of Ministry of Education, Nanjing University of Chinese Medicine, No. 138 Xianlin Avenue, Qixia District, Nanjing 210023, China
| | - Honglei Li
- College of Acupuncture-Moxibustion and Tuina, Nanjing University of Chinese Medicine, No. 138 Xianlin Avenue, Qixia District, Nanjing 210023, China; (Z.Z.)
- Key Laboratory of Acupuncture and Medicine Research of Ministry of Education, Nanjing University of Chinese Medicine, No. 138 Xianlin Avenue, Qixia District, Nanjing 210023, China
| | - Xinchang Zhang
- College of Acupuncture-Moxibustion and Tuina, Nanjing University of Chinese Medicine, No. 138 Xianlin Avenue, Qixia District, Nanjing 210023, China; (Z.Z.)
- Key Laboratory of Acupuncture and Medicine Research of Ministry of Education, Nanjing University of Chinese Medicine, No. 138 Xianlin Avenue, Qixia District, Nanjing 210023, China
| | - Yiyang Li
- College of Acupuncture-Moxibustion and Tuina, Nanjing University of Chinese Medicine, No. 138 Xianlin Avenue, Qixia District, Nanjing 210023, China; (Z.Z.)
- Key Laboratory of Acupuncture and Medicine Research of Ministry of Education, Nanjing University of Chinese Medicine, No. 138 Xianlin Avenue, Qixia District, Nanjing 210023, China
| | - Yawen Xia
- College of Acupuncture-Moxibustion and Tuina, Nanjing University of Chinese Medicine, No. 138 Xianlin Avenue, Qixia District, Nanjing 210023, China; (Z.Z.)
- Key Laboratory of Acupuncture and Medicine Research of Ministry of Education, Nanjing University of Chinese Medicine, No. 138 Xianlin Avenue, Qixia District, Nanjing 210023, China
| | - Guangxia Ni
- College of Acupuncture-Moxibustion and Tuina, Nanjing University of Chinese Medicine, No. 138 Xianlin Avenue, Qixia District, Nanjing 210023, China; (Z.Z.)
- Key Laboratory of Acupuncture and Medicine Research of Ministry of Education, Nanjing University of Chinese Medicine, No. 138 Xianlin Avenue, Qixia District, Nanjing 210023, China
| |
Collapse
|
19
|
He G, Zhang Y, Feng Y, Chen T, Liu M, Zeng Y, Yin X, Qu S, Huang L, Ke Y, Liang L, Yan J, Liu W. SBFI26 induces triple-negative breast cancer cells ferroptosis via lipid peroxidation. J Cell Mol Med 2024; 28:e18212. [PMID: 38516826 PMCID: PMC10958404 DOI: 10.1111/jcmm.18212] [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: 08/18/2023] [Revised: 01/13/2024] [Accepted: 02/09/2024] [Indexed: 03/23/2024] Open
Abstract
SBFI26, an inhibitor of FABP5, has been shown to suppress the proliferation and metastasis of tumour cells. However, the underlying mechanism by which SBFI26 induces ferroptosis in breast cancer cells remains largely unknown. Three breast cancer cell lines were treated with SBFI26 and CCK-8 assessed cytotoxicity. Transcriptome was performed on the Illumina platform and verified by qPCR. Western blot evaluated protein levels. Malondialdehyde (MDA), total superoxide dismutase (T-SOD), Fe, glutathione (GSH) and oxidized glutathione (GSSG) were measured. SBFI26 induced cell death time- and dose-dependent, with a more significant inhibitory effect on MDA-MB-231 cells. Fer-1, GSH and Vitamin C attenuated the effects but not erastin. RNA-Seq analysis revealed that SBFI26 treatment significantly enriched differentially expressed genes related to ferroptosis. Furthermore, SBFI26 increased intracellular MDA, iron ion, and GSSG levels while decreasing T-SOD, total glutathione (T-GSH), and GSH levels.SBFI26 dose-dependently up-regulates the expression of HMOX1 and ALOX12 at both gene and protein levels, promoting ferroptosis. Similarly, it significantly increases the expression of SAT1, ALOX5, ALOX15, ALOXE3 and CHAC1 that, promoting ferroptosis while downregulating the NFE2L2 gene and protein that inhibit ferroptosis. SBFI26 leads to cellular accumulation of fatty acids, which triggers excess ferrous ions and subsequent lipid peroxidation for inducing ferroptosis.
Collapse
Affiliation(s)
- Gang He
- Key Laboratory of Medicinal and Edible Plants Resources Development of Sichuan Education DepartmentSichuan Industrial Institute of Antibiotics, School of Pharmacy, Chengdu UniversityChengduChina
| | - Yiyuan Zhang
- Key Laboratory of Medicinal and Edible Plants Resources Development of Sichuan Education DepartmentSichuan Industrial Institute of Antibiotics, School of Pharmacy, Chengdu UniversityChengduChina
| | - Yanjiao Feng
- Key Laboratory of Medicinal and Edible Plants Resources Development of Sichuan Education DepartmentSichuan Industrial Institute of Antibiotics, School of Pharmacy, Chengdu UniversityChengduChina
| | - Tangcong Chen
- Key Laboratory of Medicinal and Edible Plants Resources Development of Sichuan Education DepartmentSichuan Industrial Institute of Antibiotics, School of Pharmacy, Chengdu UniversityChengduChina
| | - Mei Liu
- Key Laboratory of Medicinal and Edible Plants Resources Development of Sichuan Education DepartmentSichuan Industrial Institute of Antibiotics, School of Pharmacy, Chengdu UniversityChengduChina
| | - Yue Zeng
- Key Laboratory of Medicinal and Edible Plants Resources Development of Sichuan Education DepartmentSichuan Industrial Institute of Antibiotics, School of Pharmacy, Chengdu UniversityChengduChina
| | - Xiaojing Yin
- Key Laboratory of Medicinal and Edible Plants Resources Development of Sichuan Education DepartmentSichuan Industrial Institute of Antibiotics, School of Pharmacy, Chengdu UniversityChengduChina
| | - Shaokui Qu
- Key Laboratory of Medicinal and Edible Plants Resources Development of Sichuan Education DepartmentSichuan Industrial Institute of Antibiotics, School of Pharmacy, Chengdu UniversityChengduChina
| | - Lifen Huang
- Key Laboratory of Medicinal and Edible Plants Resources Development of Sichuan Education DepartmentSichuan Industrial Institute of Antibiotics, School of Pharmacy, Chengdu UniversityChengduChina
| | - Youqiang Ke
- Key Laboratory of Medicinal and Edible Plants Resources Development of Sichuan Education DepartmentSichuan Industrial Institute of Antibiotics, School of Pharmacy, Chengdu UniversityChengduChina
| | - Li Liang
- Key Laboratory of Medicinal and Edible Plants Resources Development of Sichuan Education DepartmentSichuan Industrial Institute of Antibiotics, School of Pharmacy, Chengdu UniversityChengduChina
| | - Jun Yan
- Key Laboratory of Medicinal and Edible Plants Resources Development of Sichuan Education DepartmentSichuan Industrial Institute of Antibiotics, School of Pharmacy, Chengdu UniversityChengduChina
| | - Wei Liu
- Key Laboratory of Medicinal and Edible Plants Resources Development of Sichuan Education DepartmentSichuan Industrial Institute of Antibiotics, School of Pharmacy, Chengdu UniversityChengduChina
| |
Collapse
|
20
|
Yu Y, Xu Z, Zhou H, Xu R, Xu J, Liu W, Wu Y, Qiu Y, Zhang G, Huang X, Chen Y. RBP7 functions as a tumor suppressor in HR + breast cancer by inhibiting the AKT/SREBP1 pathway and reducing fatty acid. Cancer Cell Int 2024; 24:118. [PMID: 38553715 PMCID: PMC10979609 DOI: 10.1186/s12935-024-03299-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Accepted: 03/08/2024] [Indexed: 04/02/2024] Open
Abstract
BACKGROUND Increasing evidence proves that RBP7 plays a significant role in breast cancer (BC). The present study was aimed to investigate the mechanism of RBP7. METHODS Western Blotting and qRT-PCR were performed for evaluating the expression levels. CCK8, colony forming, xenograft mouse model, wound healing and transwell assays were conducted to examine cell ability of proliferation, invasion and migration. Nile red staining and Oil red O staining were used for testing the lipid. RESULTS RBP7 was related to overall survival (OS) in patients with HR + BC. RBP7 protein was significantly decreased in HR + BC tissues and cells. RBP7 suppressed HR + BC cell proliferation in vitro and in vivo, and inhibited migration and invasion. RBP7 reduced fatty acid in HR + BC cells by inhibiting the AKT/SREBP1 pathway. CONCLUSIONS RBP7 may function as a tumor suppressor in HR + BC by inhibiting the AKT/SREBP1 pathway and reducing fatty acid.
Collapse
Affiliation(s)
- Yue Yu
- Department of General Surgery, The First Affiliated Hospital of Soochow University, NO. 899, Pinghai Road, Suzhou, 215006, China
| | - Zhihua Xu
- Department of General Surgery, The First Affiliated Hospital of Soochow University, NO. 899, Pinghai Road, Suzhou, 215006, China
| | - Hao Zhou
- Department of General Surgery, The First Affiliated Hospital of Soochow University, NO. 899, Pinghai Road, Suzhou, 215006, China
| | - Ruyan Xu
- Jiangsu Institute of Clinical Immunology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Jia Xu
- Jiangsu Institute of Clinical Immunology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Wenjun Liu
- Jiangsu Institute of Clinical Immunology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Yuxin Wu
- Department of General Surgery, The First Affiliated Hospital of Soochow University, NO. 899, Pinghai Road, Suzhou, 215006, China
| | - Yue Qiu
- Department of General Surgery, The First Affiliated Hospital of Soochow University, NO. 899, Pinghai Road, Suzhou, 215006, China
| | - Guangbo Zhang
- Jiangsu Institute of Clinical Immunology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Xue Huang
- Jiangsu Institute of Clinical Immunology, The First Affiliated Hospital of Soochow University, Suzhou, China.
| | - Yan Chen
- Department of General Surgery, The First Affiliated Hospital of Soochow University, NO. 899, Pinghai Road, Suzhou, 215006, China.
| |
Collapse
|
21
|
Li G, Yan K, Zhang W, Pan H, Guo P. ARDS and aging: TYMS emerges as a promising biomarker and therapeutic target. Front Immunol 2024; 15:1365206. [PMID: 38558817 PMCID: PMC10978671 DOI: 10.3389/fimmu.2024.1365206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Accepted: 02/29/2024] [Indexed: 04/04/2024] Open
Abstract
Background Acute Respiratory Distress Syndrome (ARDS) is a common condition in the intensive care unit (ICU) with a high mortality rate, yet the diagnosis rate remains low. Recent studies have increasingly highlighted the role of aging in the occurrence and progression of ARDS. This study is committed to investigating the pathogenic mechanisms of cellular and genetic changes in elderly ARDS patients, providing theoretical support for the precise treatment of ARDS. Methods Gene expression profiles for control and ARDS samples were obtained from the Gene Expression Omnibus (GEO) database, while aging-related genes (ARGs) were sourced from the Human Aging Genomic Resources (HAGR) database. Differentially expressed genes (DEGs) were subjected to functional enrichment analysis to understand their roles in ARDS and aging. The Weighted Gene Co-expression Network Analysis (WGCNA) and machine learning pinpointed key modules and marker genes, with ROC curves illustrating their significance. The expression of four ARDS-ARDEGs was validated in lung samples from aged mice with ARDS using qRT-PCR. Gene set enrichment analysis (GSEA) investigated the signaling pathways and immune cell infiltration associated with TYMS expression. Single-nucleus RNA sequencing (snRNA-Seq) explored gene-level differences among cells to investigate intercellular communication during ARDS onset and progression. Results ARDEGs are involved in cellular responses to DNA damage stimuli, inflammatory reactions, and cellular senescence pathways. The MEmagenta module exhibited a significant correlation with elderly ARDS patients. The LASSO, RRF, and XGBoost algorithms were employed to screen for signature genes, including CKAP2, P2RY14, RBP2, and TYMS. Further validation emphasized the potential role of TYMS in the onset and progression of ARDS. Immune cell infiltration indicated differential proportion and correlations with TYMS expression. SnRNA-Seq and cell-cell communication analysis revealed that TYMS is highly expressed in endothelial cells, and the SEMA3 signaling pathway primarily mediates cell communication between endothelial cells and other cells. Conclusion Endothelial cell damage associated with aging could contribute to ARDS progression by triggering inflammation. TYMS emerges as a promising diagnostic biomarker and potential therapeutic target for ARDS.
Collapse
Affiliation(s)
- Gang Li
- Department of Emergency Medicine, The Third Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Ke Yan
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| | - Wanyi Zhang
- Department of Emergency Medicine, The Third Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Haiyan Pan
- Department of Emergency Medicine, The Third Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Pengxiang Guo
- Department of Pharmacology of Chinese Materia Medica, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China
| |
Collapse
|
22
|
Sun L, Zheng M, Gao Y, Brigstock DR, Gao R. Retinoic acid signaling pathway in pancreatic stellate cells: Insight into the anti-fibrotic effect and mechanism. Eur J Pharmacol 2024; 967:176374. [PMID: 38309676 DOI: 10.1016/j.ejphar.2024.176374] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2023] [Revised: 01/15/2024] [Accepted: 01/30/2024] [Indexed: 02/05/2024]
Abstract
Pancreatic stellate cells (PSCs) are activated following loss of cytoplasmic vitamin A (retinol)-containing lipid droplets, which is a key event in the process of fibrogenesis of chronic pancreatitis (CP) and pancreatic ductal adenocarcinoma (PDCA). PSCs are the major source of cancer-associated fibroblasts (CAFs) that produce stroma to induce PDAC cancer cell growth, invasion, and metastasis. As an active metabolite of retinol, retinoic acid (RA) can regulate target gene expression in PSCs through its nuclear receptor complex (RAR/RXR or RXR/RXR) or transcriptional intermediary factor. Additionally, RA also has extranuclear and non-transcriptional effects. In vitro studies have shown that RA induces PSC deactivation which reduces extracellular matrix production through multiple modes of action, such as inhibiting TβRⅡ, PDGFRβ, β-catenin and Wnt production, downregulating ERK1/2 and JNK phosphorylation and suppressing active TGF-β1 release. RA alone or in combination with other reagents have been demonstrated to have an effective anti-fibrotic effect on cerulein-induced mouse CP models in vivo studies. Clinical trial data have shown that repurposing all-trans retinoic acid (ATRA) as a stromal-targeting agent for human pancreatic cancer is safe and tolerable, suggesting the possibility of using RA for the treatment of CP and PDCA in humans. This review focuses on RA signaling pathways in PSCs and the effects and mechanisms of RA in PSC-mediated fibrogenesis as well as the anti-fibrotic and anti-tumor effects of RA targeting PSCs or CAFs in vitro and in vivo, highlighting the potential therapies of RA against CP and PDAC.
Collapse
Affiliation(s)
- Li Sun
- Department of Hepatic Biliary Pancreatic Medicine, First Hospital of Jilin University, Changchun, China; Department of Pathology, First Hospital of Jilin University, Changchun, China
| | - Meifang Zheng
- Department of Hepatic Biliary Pancreatic Medicine, First Hospital of Jilin University, Changchun, China; Zhejiang Cancer Hospital, Hangzhou, Zhejiang, China
| | - Yanhang Gao
- Department of Hepatic Biliary Pancreatic Medicine, First Hospital of Jilin University, Changchun, China; Department of Infectious Diseases, First Hospital of Jilin University, Changchun, China.
| | - David R Brigstock
- The Research Institute at Nationwide Children's Hospital, Columbus, OH, United States
| | - Runping Gao
- Department of Hepatic Biliary Pancreatic Medicine, First Hospital of Jilin University, Changchun, China; Department of Infectious Diseases, First Hospital of Jilin University, Changchun, China.
| |
Collapse
|
23
|
Agellon LB. Importance of fatty acid binding proteins in cellular function and organismal metabolism. J Cell Mol Med 2024; 28:e17703. [PMID: 36876733 PMCID: PMC10902576 DOI: 10.1111/jcmm.17703] [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: 11/01/2022] [Revised: 01/25/2023] [Accepted: 02/14/2023] [Indexed: 03/07/2023] Open
Abstract
Fatty acid binding proteins (Fabps) are small soluble proteins that are abundant in the cytosol. These proteins are known to bind a myriad of small hydrophobic molecules and have been postulated to serve a variety of roles, yet their precise functions have remained an enigma over half a century of study. Here, we consider recent findings, along with the cumulative findings contributed by many laboratories working on Fabps over the last half century, to synthesize a new outlook for what functions Fabps serve in cells and organisms. Collectively, the findings illustrate that Fabps function as versatile multi-purpose devices serving as sensors, conveyors and modulators to enable cells to detect and handle a specific class of metabolites, and to adjust their metabolic capacity and efficiency.
Collapse
Affiliation(s)
- Luis B. Agellon
- School of Human NutritionMcGill UniversitySte. Anne de BellevueQuebecCanada
| |
Collapse
|
24
|
Xiao S, Yan Y, Shao M, Zhou X, Niu Z, Wu Y, Li Y, Cui Y, Long Y, Du Q. Kuijieling decoction regulates the Treg/Th17 cell balance in ulcerative colitis through the RA/RARα signaling pathway. JOURNAL OF ETHNOPHARMACOLOGY 2024; 318:116909. [PMID: 37451490 DOI: 10.1016/j.jep.2023.116909] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Revised: 07/09/2023] [Accepted: 07/11/2023] [Indexed: 07/18/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Ulcerative colitis (UC) is an idiopathic intestinal disease characterized by chronic inflammation with unknown etiology. Kuijieling decoction is a traditional Chinese Medicine with unique therapeutic efficacy for UC. AIM OF THE STUDY To validate the effects of Kuijieling decoction on vitamin A metabolism and retinoic acid (RA) production, along with evaluation of its immunomodulatory activity, and further clarify the upstream mechanisms underlying Treg/Th17 regulation that contribute to therapeutic effects against UC. MATERIALS AND METHODS Network pharmacology and molecular docking analyses were employed to predict the potential anti-UC targets of Kuijieling and associated pathways. A rat model of UC was generated by treatment with trinitrobenzene sulfonic acid (TNBS) to induce inflammation. T lymphocytes were induced to differentiate into Th17 via combined stimulation with the cytokines TGF-β1, IL-6 and IL-23. Expression levels of RA/RARα-related factors (RARα, CRABPII, Smad3, IL-6R and IL-23R) and Treg/Th17 cell-related factors (Foxp3, RORγt) were measured via western blot (WB), quantitative real-time PCR (RT-qPCR), and immunohistochemistry analyses. Components of the vitamin A metabolic pathway (vitamin A, retinol, retinoic acid) and Treg/Th17 cell-related cytokines (IL-10, IL-17, IL-21) were evaluated using ELISA. Flow cytometry was performed to determine the percentages of Treg and Th17 cells. RESULTS The action targets of Kuijieling were significantly associated with T cell activation, Th17 cell differentiation and the immune response. IL-2, IL-6, STAT3 and RARα displayed strong binding affinities with the main components of Kuijieling, suggesting an important role in its therapeutic efficacy. Kuijieling promoted vitamin A metabolism and RA/RARα signaling in UC rats and T lymphocytes. Moreover, Kuijieling effectively regulated the Treg/Th17 cell balance in UC rats and T lymphocytes and relieved inflammation. The protective effect of Kuijieling was weakened after inhibition of the RA/RARα signaling pathway. CONCLUSIONS Kuijieling promotes vitamin A metabolism and RA synthesis, enhances interactions between RA, intracellular binding protein CRABPII and nuclear receptor RARα, and upregulates Smad3 and Foxp3, thus promoting Treg differentiation. Simultaneously, Kuijieling inhibited expression of IL-6R and IL-23R genes and production of RORγt, leading to suppression of Th17 differentiation, and ultimately, reduction of the Th17/Treg cell ratio.
Collapse
Affiliation(s)
- Suting Xiao
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, China.
| | - Yizhen Yan
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, China.
| | - Mingyin Shao
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, China.
| | - Xuan Zhou
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, China.
| | - Zhenyu Niu
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, China.
| | - Yanli Wu
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, China.
| | - Yanwu Li
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, China.
| | - Yong Cui
- School of Medical Device, Shenyang Pharmaceutical University, Shenyang, China.
| | - Yu Long
- School of Traditional Chinese Medicine, Hunan University of Chinese Medicine, Changsha, China.
| | - Qun Du
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, China.
| |
Collapse
|
25
|
Che S, Pham PH, Barbut S, Bienzle D, Susta L. Transcriptomic Profiles of Pectoralis major Muscles Affected by Spaghetti Meat and Woody Breast in Broiler Chickens. Animals (Basel) 2024; 14:176. [PMID: 38254345 PMCID: PMC10812457 DOI: 10.3390/ani14020176] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Revised: 01/03/2024] [Accepted: 01/04/2024] [Indexed: 01/24/2024] Open
Abstract
Spaghetti meat (SM) and woody breast (WB) are breast muscle myopathies of broiler chickens, characterized by separation of myofibers and by fibrosis, respectively. This study sought to investigate the transcriptomic profiles of breast muscles affected by SM and WB. Targeted sampling was conducted on a flock to obtain 10 WB, 10 SM, and 10 Normal Pectoralis major muscle samples from 37-day-old male chickens. Total RNA was extracted, cDNA was used for pair-end sequencing, and differentially expressed genes (DEGs) were determined by a false discovery rate of <0.1 and a >1.5-fold change. Principal component and heatmap cluster analyses showed that the SM and WB samples clustered together. No DEGs were observed between SM and WB fillets, while a total of 4018 and 2323 DEGs were found when comparing SM and WB, respectively, against Normal samples. In both the SM and WB samples, Gene Ontology terms associated with extracellular environment and immune response were enriched. The KEGG analysis showed enrichment of cytokine-cytokine receptor interaction and extracellular matrix-receptor interaction pathways in both myopathies. Although SM and WB are macroscopically different, the similar transcriptomic profiles suggest that these conditions may share a common pathogenesis. This is the first study to compare the transcriptomes of SM and WB, and it showed that, while both myopathies had profiles different from the normal breast muscle, SM and WB were similar, with comparable enriched metabolic pathways and processes despite presenting markedly different macroscopic features.
Collapse
Affiliation(s)
- Sunoh Che
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, ON N1G2W1, Canada; (S.C.); (P.H.P.)
| | - Phuc H. Pham
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, ON N1G2W1, Canada; (S.C.); (P.H.P.)
| | - Shai Barbut
- Department of Food Science, Ontario Agricultural College, University of Guelph, Guelph, ON N1G2W1, Canada;
| | - Dorothee Bienzle
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, ON N1G2W1, Canada; (S.C.); (P.H.P.)
| | - Leonardo Susta
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, ON N1G2W1, Canada; (S.C.); (P.H.P.)
| |
Collapse
|
26
|
Takahashi N. [Prevention and Treatment of Cancer with Vitamin A and Its Derivatives: Cell Differentiation and Proliferation]. YAKUGAKU ZASSHI 2024; 144:203-222. [PMID: 38296498 DOI: 10.1248/yakushi.23-00184] [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: 02/07/2024]
Abstract
Normal differentiation and proliferation of cells are essential for maintaining homeostasis. Following the successful completion of whole genome sequencing, protein modification has been attracted increasing attention in order to understand the roles of protein diversification in protein function and to elucidate molecular targets in mechanisms of signal transduction. Vitamin A is an essential nutrient for health maintenance. It is present as β-carotene in green and yellow vegetables and retinyl ester in animal products and absorbed into the body from the intestines. After ingestion, it is converted to retinol and oxidized in target cells to retinal, which plays critical roles in vision. It is then further oxidized to retinoic acid (RA), which exhibits a number of effects prior to being metabolized by cytochrome P450 and excreted from the body. Since RA exhibits cell differentiation-inducing actions, it is used as a therapeutic agent for patients with acute promyelocytic leukemia. The current paper describes: (1) HL60 cell differentiation and cell differentiation induction therapy by RA; (2) roles played by RA and retinal and their mechanisms of action; (3) retinoylation, post-translational protein-modified by RA, a novel non-genomic RA mechanism of action without RA receptor; (4) new actions of β-carotene and retinol in vivo and (5) potent anticancer effects of p-dodecylaminophenol (p-DDAP), a novel vitamin A derivative created from the RA derivative fenretinide. We propose that nutritional management of vitamin A can be effective at preventing and treating diseases, and that p-DDAP is a promising anticancer drug.
Collapse
Affiliation(s)
- Noriko Takahashi
- Laboratory of Physiological Chemistry, Institute of Medicinal Chemistry, Hoshi University
| |
Collapse
|
27
|
Almaguer J, Hindle A, Lawrence JJ. The Contribution of Hippocampal All-Trans Retinoic Acid (ATRA) Deficiency to Alzheimer's Disease: A Narrative Overview of ATRA-Dependent Gene Expression in Post-Mortem Hippocampal Tissue. Antioxidants (Basel) 2023; 12:1921. [PMID: 38001775 PMCID: PMC10669734 DOI: 10.3390/antiox12111921] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 10/04/2023] [Accepted: 10/05/2023] [Indexed: 11/26/2023] Open
Abstract
There is accumulating evidence that vitamin A (VA) deficiency contributes to the pathogenesis and progression of Alzheimer's disease (AD). All-trans retinoic acid (ATRA), a metabolite of VA in the brain, serves distinct roles in the human hippocampus. Agonists of retinoic acid receptors (RAR), including ATRA, promote activation of the non-amyloidogenic pathway by enhancing expression of α-secretases, providing a mechanistic basis for delaying/preventing amyloid beta (Aβ) toxicity. However, whether ATRA is actually deficient in the hippocampi of patients with AD is not clear. Here, using a publicly available human transcriptomic dataset, we evaluated the extent to which ATRA-sensitive genes are dysregulated in hippocampal tissue from post-mortem AD brains, relative to age-matched controls. Consistent with ATRA deficiency, we found significant dysregulation of many ATRA-sensitive genes and significant upregulation of RAR co-repressors, supporting the idea of transcriptional repression of ATRA-mediated signaling. Consistent with oxidative stress and neuroinflammation, Nrf2 and NfkB transcripts were upregulated, respectively. Interestingly, transcriptional targets of Nrf2 were not upregulated, accompanied by upregulation of several histone deacetylases. Overall, our investigation of ATRA-sensitive genes in the human hippocampus bolsters the scientific premise of ATRA depletion in AD and that epigenetic factors should be considered and addressed as part of VA supplementation.
Collapse
Affiliation(s)
- Joey Almaguer
- Department of Pharmacology and Neuroscience, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA;
| | - Ashly Hindle
- Department of Pharmacology and Neuroscience and Garrison Institute on Aging, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA;
| | - J. Josh Lawrence
- Department of Pharmacology and Neuroscience, Garrison Institute on Aging, Center of Excellence for Translational Neuroscience and Therapeutics, and Center of Excellence for Integrated Health, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
| |
Collapse
|
28
|
Calderon RM, Golczak M, Paik J, Blaner WS. Dietary Vitamin A Affects the Function of Incretin-Producing Enteroendocrine Cells in Male Mice Fed a High-Fat Diet. J Nutr 2023; 153:2901-2914. [PMID: 37648113 PMCID: PMC10613727 DOI: 10.1016/j.tjnut.2023.08.030] [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: 07/01/2023] [Revised: 08/12/2023] [Accepted: 08/24/2023] [Indexed: 09/01/2023] Open
Abstract
BACKGROUND Retinol-binding protein 2 (RBP2) is an intracellular carrier for vitamin A in the absorptive enterocytes. Mice lacking RBP2 (Rbp2-/-) display an unexpected phenotype of obesity, glucose intolerance, and elevated glucose-dependent insulinotropic polypeptide (GIP) levels. GIP and glucagon-like peptide 1 (GLP-1) are incretin hormones secreted by enteroendocrine cells (EECs). We recently demonstrated the presence of RBP2 and other retinoid-related proteins in EECs. OBJECTIVES Given RBP2's role in intracellular retinoid trafficking, we aimed to evaluate whether dietary vitamin A affects incretin-secreting cell function and gene expression. METHODS Male Rbp2-/- mice and sex- and age-matched controls (n = 6-9) were fed a high-fat diet (HFD) for 18 wk containing normal (VAN, 4000 IU/kg of diet) or low (VAL, 25% of normal) vitamin A concentrations. Body weight was recorded biweekly. Plasma GIP and GLP-1 levels were obtained fasting and 30 min after an oral fat gavage at week 16. Glucose tolerance tests were also performed. Mice were killed at week 18, and blood and tissue samples were obtained. RESULTS Rbp2-/- mice displayed greater weight gain on the VAN compared with the VAL diet from week 7 of the intervention (P ≤ 0.01). Stimulated GIP levels were elevated in Rbp2-/- mice compared with their controls fed the VAN diet (P = 0.02), whereas their GIP response was lower when fed the VAL diet (P = 0.03). Although no differences in GLP-1 levels were observed in the VAN diet group, a lower GLP-1 response was seen in Rbp2-/- mice fed the VAL diet (P = 0.02). Changes in incretin gene expression and that of other genes associated with EEC lineage and function were consistent with these observations. Circulating and hepatic retinoid levels revealed no systemic vitamin A deficiency across dietary groups. CONCLUSIONS Our data support a role for RBP2 and dietary vitamin A in incretin secretion and gene expression in mice fed a HFD.
Collapse
Affiliation(s)
- Rossana M Calderon
- Department of Medicine, College of Physicians and Surgeons, Columbia University, New York, NY, United States.
| | - Marcin Golczak
- Department of Pharmacology, Case Western Reserve University, Cleveland, OH, United States; Cleveland Center for Membrane and Structural Biology, School of Medicine, Case Western Reserve University, Cleveland, OH, United States
| | - Jisun Paik
- Department of Comparative Medicine, University of Washington, Seattle, WA, United States
| | - William S Blaner
- Department of Medicine, College of Physicians and Surgeons, Columbia University, New York, NY, United States
| |
Collapse
|
29
|
Nhieu J, Miller MC, Lerdall TA, Mayo KH, Wei LN. Molecular basis for cellular retinoic acid-binding protein 1 in modulating CaMKII activation. Front Mol Biosci 2023; 10:1268843. [PMID: 37822422 PMCID: PMC10562560 DOI: 10.3389/fmolb.2023.1268843] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Accepted: 09/11/2023] [Indexed: 10/13/2023] Open
Abstract
Introduction: Cellular retinoic acid (RA)-binding protein 1 (CRABP1) is a highly conserved protein comprised of an anti-parallel, beta-barrel, and a helix-turn-helix segment outside this barrel. Functionally, CRABP1 is thought to bind and sequester cytosolic RA. Recently, CRABP1 has been established as a major mediator of rapid, non-genomic activity of RA in the cytosol, referred to as "non-canonical" activity. Previously, we have reported that CRABP1 interacts with and dampens the activation of calcium-calmodulin (Ca2+-CaM)-dependent kinase 2 (CaMKII), a major effector of Ca2+ signaling. Through biophysical, molecular, and cellular assays, we, herein, elucidate the molecular and structural mechanisms underlying the action of CRABP1 in dampening CaMKII activation. Results: We identify an interaction surface on CRABP1 for CaMKII binding, located on the beta-sheet surface of the barrel, and an allosteric region within the helix segment outside the barrel, where both are important for interacting with CaMKII. Molecular studies reveal that CRABP1 preferentially associates with the inactive form of CaMKII, thereby dampening CaMKII activation. Alanine mutagenesis of residues implicated in the CaMKII interaction results in either a loss of this preference or a shift of CRABP1 from associating with the inactive CaMKII to associating with the active CaMKII, which corresponds to changes in CRABP1's effect in modulating CaMKII activation. Conclusions: This is the first study to elucidate the molecular and structural basis for CRABP1's function in modulating CaMKII activation. These results further shed insights into CRABP1's functional involvement in multiple signaling pathways, as well as its extremely high sequence conservation across species and over evolution.
Collapse
Affiliation(s)
- Jennifer Nhieu
- Department of Pharmacology, University of Minnesota Medical School, Minneapolis, MN, United States
| | - Michelle C. Miller
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN, United States
| | - Thomas A. Lerdall
- Department of Pharmacology, University of Minnesota Medical School, Minneapolis, MN, United States
| | - Kevin H. Mayo
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN, United States
| | - Li-Na Wei
- Department of Pharmacology, University of Minnesota Medical School, Minneapolis, MN, United States
| |
Collapse
|
30
|
Surmacki JM, Abramczyk H. Confocal Raman imaging reveals the impact of retinoids on human breast cancer via monitoring the redox status of cytochrome c. Sci Rep 2023; 13:15049. [PMID: 37700001 PMCID: PMC10497563 DOI: 10.1038/s41598-023-42301-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Accepted: 09/07/2023] [Indexed: 09/14/2023] Open
Abstract
This paper expands the current state of knowledge on impact of retinoids on redox status of cytochrome c in cancers. Little is known how the expression of cytochromes may influence the development of cancers. We studied the effect of the redox status of the central iron ion in heme of cytochrome c. We determined the redox status of the iron ion in cytochrome c in mitochondria, cytoplasm, lipid droplets, and endoplasmic reticulum of the human breast cancer cells by Raman imaging. We incubated human breast adenocarcinoma cells (SK-BR-3) with retinoic acid, retinol and retinyl ester (palmitate) at concentration of 50 μM for 24 h. We recorded the Raman spectra and images of human breast cancer in vitro SK-BR-3 cells receiving redox stimuli by retinoic acid, retinol and retinyl ester (palmitate). The paper provides evidence that retinoic acid and retinol are pivotally important for mitochondrial energy homeostasis by controlling the redox status of cytochrome c in the electron transport chain controlling oxidative phosphorylation and apoptosis. We discussed the role of retinoids in metabolism and signaling of cancer cells. The paper provides experimental support for theoretical hypothesis how retinoic acid/retinol catalyse resonance energy transfer reactions and controls the activation/inactivation cycle of protein kinase PKCδ.
Collapse
Affiliation(s)
- Jakub Maciej Surmacki
- Laboratory of Laser Molecular Spectroscopy, Institute of Applied Radiation Chemistry, Faculty of Chemistry, Lodz University of Technology, Wroblewskiego 15, 93-590, Lodz, Poland
| | - Halina Abramczyk
- Laboratory of Laser Molecular Spectroscopy, Institute of Applied Radiation Chemistry, Faculty of Chemistry, Lodz University of Technology, Wroblewskiego 15, 93-590, Lodz, Poland.
| |
Collapse
|
31
|
Tian L, Gong L, Hao C, Feng Y, Yao S, Fei B, Wang X, Huang Z. ELOA promotes tumor growth and metastasis by activating RBP1 in gastric cancer. Cancer Med 2023; 12:18946-18959. [PMID: 37694492 PMCID: PMC10557880 DOI: 10.1002/cam4.6516] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Revised: 08/23/2023] [Accepted: 08/30/2023] [Indexed: 09/12/2023] Open
Abstract
BACKGROUND Elongin A (ELOA), our previous work revealed, serves as a novel tumor suppressor in colorectal cancer. However, the function and mechanism of ELOA in other cancer types, including gastric cancer (GC), remain to be elucidated. METHODS The expression of ELOA was measured by quantitative reverse transcription-polymerase chain reaction and western blot. The effects of ELOA on GC growth and metastasis were assessed through a series of in-vitro and in-vivo assays. Furthermore, the potential mechanism of ELOA was revealed by RNA sequencing, dual luciferase reporter assay, chromatin immunoprecipitation, and rescue experiments in GC. RESULTS We uncovered increased expression of ELOA in GC tissues compared with paired normal tissues via bioinformatic analyses and our sample detection. Enhanced ELOA expression in GC tissues was obviously correlated with poor tumor differentiation, lymph node metastasis, advanced tumor stage, and a poor prognosis. A series of functional experiments showed that ELOA promoted the proliferation and metastasis of GC. Mechanistically, we revealed that the decreased levels of miR-490-3p caused the upregulation of ELOA in GC. Both RNA-seq and ChIP assays revealed that ELOA transcriptionally activated retinol-binding protein 1 (RBP1) by binding to its promotor. Furthermore, specific knockdown of RBP1 reduced the tumor-promoting ability of ELOA in GC cells. CONCLUSIONS In summary, our findings demonstrate that ELOA exerts oncogenic properties by activating RBP1 expression, providing the basis for a promising therapeutic target in GC.
Collapse
Affiliation(s)
- Lu Tian
- Laboratory of Cancer Epigenetics, Wuxi School of MedicineJiangnan UniversityWuxiChina
- Wuxi Cancer InstituteAffiliated Hospital of Jiangnan UniversityWuxiChina
| | - Liang Gong
- Laboratory of Cancer Epigenetics, Wuxi School of MedicineJiangnan UniversityWuxiChina
- Wuxi Cancer InstituteAffiliated Hospital of Jiangnan UniversityWuxiChina
- Key Laboratory of Carbohydrate Chemistry & Biotechnology, Ministry of Education, School of BiotechnologyJiangnan UniversityWuxiChina
| | - Chu Hao
- Laboratory of Cancer Epigenetics, Wuxi School of MedicineJiangnan UniversityWuxiChina
- Wuxi Cancer InstituteAffiliated Hospital of Jiangnan UniversityWuxiChina
| | - Yuyang Feng
- Laboratory of Cancer Epigenetics, Wuxi School of MedicineJiangnan UniversityWuxiChina
- Wuxi Cancer InstituteAffiliated Hospital of Jiangnan UniversityWuxiChina
| | - Surui Yao
- Laboratory of Cancer Epigenetics, Wuxi School of MedicineJiangnan UniversityWuxiChina
- Wuxi Cancer InstituteAffiliated Hospital of Jiangnan UniversityWuxiChina
| | - Bojian Fei
- Department of Gastrointestinal SurgeryAffiliated Hospital of Jiangnan UniversityWuxiChina
| | - Xue Wang
- Laboratory of Cancer Epigenetics, Wuxi School of MedicineJiangnan UniversityWuxiChina
- Wuxi Cancer InstituteAffiliated Hospital of Jiangnan UniversityWuxiChina
| | - Zhaohui Huang
- Laboratory of Cancer Epigenetics, Wuxi School of MedicineJiangnan UniversityWuxiChina
- Wuxi Cancer InstituteAffiliated Hospital of Jiangnan UniversityWuxiChina
| |
Collapse
|
32
|
Luo P, Zheng L, Zou J, Chen T, Zou J, Li W, Chen Q, Qian B. Insights into vitamin A in bladder cancer, lack of attention to gut microbiota? Front Immunol 2023; 14:1252616. [PMID: 37711628 PMCID: PMC10497765 DOI: 10.3389/fimmu.2023.1252616] [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: 07/04/2023] [Accepted: 08/14/2023] [Indexed: 09/16/2023] Open
Abstract
Vitamin A has long been associated with bladder cancer, and many exogenous vitamin A supplements, vitamin A derivatives, and synthetic drugs have been investigated over the years. However, the effectiveness of these strategies in clinical practice has not met expectations, and they have not been widely adopted. Recent medical research on intestinal flora has revealed that bladder cancer patients exhibit reduced serum vitamin A levels and an imbalance of gut microbiota. In light of the close relationship between gut microbiota and vitamin A, one can speculate that a complex regulatory mechanism exists between the two in the development and occurrence of bladder cancer. As such, further exploration of their interaction in bladder cancer may help guide the use of vitamin A for preventive purposes. During the course of this review, attention is paid to the influence of intestinal microbiota on the vitamin A metabolism and the RA signaling pathway, as well as the mutual promotion relationships between them in the prevention of bladder cancer, In addition, it emphasizes the importance of intestinal microbiota for bladder cancer prevention and treatment.
Collapse
Affiliation(s)
- Peiyue Luo
- The First Clinical College, Gannan Medical University, Ganzhou, Jiangxi, China
- Department of Urology, The First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, China
- Key Laboratory of Urology and Andrology of Ganzhou, Ganzhou, Jiangxi, China
| | - Liying Zheng
- Department of Graduate, The First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, China
| | - Junrong Zou
- Department of Urology, The First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, China
- Key Laboratory of Urology and Andrology of Ganzhou, Ganzhou, Jiangxi, China
| | - Tao Chen
- The First Clinical College, Gannan Medical University, Ganzhou, Jiangxi, China
- Department of Urology, The First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, China
- Key Laboratory of Urology and Andrology of Ganzhou, Ganzhou, Jiangxi, China
| | - Jun Zou
- The First Clinical College, Gannan Medical University, Ganzhou, Jiangxi, China
- Department of Urology, The First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, China
- Key Laboratory of Urology and Andrology of Ganzhou, Ganzhou, Jiangxi, China
| | - Wei Li
- The First Clinical College, Gannan Medical University, Ganzhou, Jiangxi, China
- Department of Urology, The First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, China
- Key Laboratory of Urology and Andrology of Ganzhou, Ganzhou, Jiangxi, China
| | - Qi Chen
- The First Clinical College, Gannan Medical University, Ganzhou, Jiangxi, China
- Department of Urology, The First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, China
- Key Laboratory of Urology and Andrology of Ganzhou, Ganzhou, Jiangxi, China
| | - Biao Qian
- Department of Urology, The First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, China
- Key Laboratory of Urology and Andrology of Ganzhou, Ganzhou, Jiangxi, China
| |
Collapse
|
33
|
Ghorbanian B, Wong A, Iranpour A. The effect of dietary carbohydrate restriction and aerobic exercise on retinol binding protein 4 (RBP4) and fatty acid binding protein 5 (FABP5) in middle-aged men with metabolic syndrome. Br J Nutr 2023; 130:553-563. [PMID: 36373560 DOI: 10.1017/s0007114522003580] [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: 11/16/2022]
Abstract
Exercise and dietary interventions have been described to positively affect metabolic syndrome (MetS) via molecular-induced changes. The purpose of this study was to investigate the effects of dietary carbohydrate restriction and aerobic exercise (AE) on retinol binding protein 4 (RBP4) and fatty acid binding protein 5 (FABP5) in middle-aged men with MetS. The study had a randomised, double-blinded, parallel-controlled design. Forty middle-aged men with MetS (age: 53·97 ± 2·85 years, BMI = 31·09 ± 1·04 kg/m2) were randomly assigned to four groups, AE (n 10), ketogenic diet (KD; n 10), AE combined with KD (AE + KD; n 10) or control (C; n 10). RBP4, FABP5, body composition (body mass, BMI and body fat), insulin resistance, insulin sensitivity and MetS factors were evaluated prior to and after the 12-week intervention. AE + KD significantly decreased the body fat percentage (P = 0·006), BMI (P = 0·001), Zmets (P = 0·017), RBP4 (P = 0·017) and the homeostasis model of insulin resistance (HOMA-IR) (P = 0·001) as compared with control group and marginally significantly decreased the Zmets as compared with exercise group (P = 0·086). KD significantly decreased RBP4 levels as compared with control group (P = 0·041). Only the AE intervention (P = 0·045) significantly decreased FABP5 levels. Combining intervention of carbohydrate restriction with AE compared with carbohydrate restriction and AE alone improved RBP4, HOMA-IR as well as different body composition and MetS factors in middle-aged men with MetS.
Collapse
Affiliation(s)
- Bahloul Ghorbanian
- Department of Physical Education, Faculty of Educational Sciences and Psychology, Azarbaijan Shahid Madani University, Tabriz, Iran
| | - Alexei Wong
- Department of Health and Human Performance, Marymount University, Arlington, VA, USA
| | - Asgar Iranpour
- Department of Sports Physiology, Faculty of Educational Sciences and Psychology, University of Mohaghegh Ardabili, Ardabil, Iran
| |
Collapse
|
34
|
Viragova S, Aparicio L, Palmerini P, Zhao J, Valencia Salazar LE, Schurer A, Dhuri A, Sahoo D, Moskaluk CA, Rabadan R, Dalerba P. Inverse agonists of retinoic acid receptor/retinoid X receptor signaling as lineage-specific antitumor agents against human adenoid cystic carcinoma. J Natl Cancer Inst 2023; 115:838-852. [PMID: 37040084 PMCID: PMC10323906 DOI: 10.1093/jnci/djad062] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Revised: 03/13/2023] [Accepted: 04/02/2023] [Indexed: 04/12/2023] Open
Abstract
BACKGROUND Adenoid cystic carcinoma (ACC) is a lethal malignancy of exocrine glands, characterized by the coexistence within tumor tissues of 2 distinct populations of cancer cells, phenotypically similar to the myoepithelial and ductal lineages of normal salivary epithelia. The developmental relationship linking these 2 cell types, and their differential vulnerability to antitumor treatments, remains unknown. METHODS Using single-cell RNA sequencing, we identified cell-surface markers (CD49f, KIT) that enabled the differential purification of myoepithelial-like (CD49fhigh/KITneg) and ductal-like (CD49flow/KIT+) cells from patient-derived xenografts (PDXs) of human ACCs. Using prospective xenotransplantation experiments, we compared the tumor-initiating capacity of the 2 cell types and tested whether one could differentiate into the other. Finally, we searched for signaling pathways with differential activation between the 2 cell types and tested their role as lineage-specific therapeutic targets. RESULTS Myoepithelial-like cells displayed higher tumorigenicity than ductal-like cells and acted as their progenitors. Myoepithelial-like and ductal-like cells displayed differential expression of genes encoding for suppressors and activators of retinoic acid signaling, respectively. Agonists of retinoic acid receptor (RAR) or retinoid X receptor (RXR) signaling (all-trans retinoic acid, bexarotene) promoted myoepithelial-to-ductal differentiation, whereas suppression of RAR/RXR signaling with a dominant-negative RAR construct abrogated it. Inverse agonists of RAR/RXR signaling (BMS493, AGN193109) displayed selective toxicity against ductal-like cells and in vivo antitumor activity against PDX models of human ACC. CONCLUSIONS In human ACCs, myoepithelial-like cells act as progenitors of ductal-like cells, and myoepithelial-to-ductal differentiation is promoted by RAR/RXR signaling. Suppression of RAR/RXR signaling is lethal to ductal-like cells and represents a new therapeutic approach against human ACCs.
Collapse
Affiliation(s)
- Sara Viragova
- Department of Pathology and Cell Biology, Columbia University Medical Center, New York, NY, USA
- Herbert Irving Comprehensive Cancer Center, Columbia University Medical Center, New York, NY, USA
- Columbia Stem Cell Initiative, Columbia University Medical Center, New York, NY, USA
- Integrated Program in Cellular, Molecular and Biomedical Studies, Columbia University, New York, NY, USA
| | - Luis Aparicio
- Program for Mathematical Genomics, Department of Systems Biology, Columbia University, New York, NY, USA
- Department of Biomedical Informatics, Columbia University, New York, NY, USA
| | - Pierangela Palmerini
- Department of Pathology and Cell Biology, Columbia University Medical Center, New York, NY, USA
- Herbert Irving Comprehensive Cancer Center, Columbia University Medical Center, New York, NY, USA
- Columbia Stem Cell Initiative, Columbia University Medical Center, New York, NY, USA
| | - Junfei Zhao
- Program for Mathematical Genomics, Department of Systems Biology, Columbia University, New York, NY, USA
- Department of Biomedical Informatics, Columbia University, New York, NY, USA
| | - Luis E Valencia Salazar
- Department of Pathology and Cell Biology, Columbia University Medical Center, New York, NY, USA
- Herbert Irving Comprehensive Cancer Center, Columbia University Medical Center, New York, NY, USA
- Columbia Stem Cell Initiative, Columbia University Medical Center, New York, NY, USA
| | - Alexandra Schurer
- Department of Pathology and Cell Biology, Columbia University Medical Center, New York, NY, USA
- Herbert Irving Comprehensive Cancer Center, Columbia University Medical Center, New York, NY, USA
| | - Anika Dhuri
- Department of Pathology and Cell Biology, Columbia University Medical Center, New York, NY, USA
| | - Debashis Sahoo
- Department of Pediatrics, University of California San Diego, San Diego, CA, USA
- Department of Computer Science and Engineering, University of California San Diego, San Diego, CA, USA
- Rebecca and John Moores Comprehensive Cancer Center, University of California San Diego, San Diego, CA, USA
| | - Christopher A Moskaluk
- Department of Pathology, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Raul Rabadan
- Program for Mathematical Genomics, Department of Systems Biology, Columbia University, New York, NY, USA
- Department of Biomedical Informatics, Columbia University, New York, NY, USA
| | - Piero Dalerba
- Department of Pathology and Cell Biology, Columbia University Medical Center, New York, NY, USA
- Herbert Irving Comprehensive Cancer Center, Columbia University Medical Center, New York, NY, USA
- Columbia Stem Cell Initiative, Columbia University Medical Center, New York, NY, USA
- Department of Medicine, Columbia University Medical Center, New York, NY, USA
- Digestive and Liver Disease Research Center, Columbia University Medical Center, New York, NY, USA
| |
Collapse
|
35
|
Agamia NF, El Mulla KF, Alsayed NM, Ghazala RM, El Maksoud REA, Abdelmeniem IM, Talaat IM, Zaki II, Sabah RM, Melnik BC. Isotretinoin treatment upregulates the expression of p53 in the skin and sebaceous glands of patients with acne vulgaris. Arch Dermatol Res 2023; 315:1355-1365. [PMID: 36585988 PMCID: PMC10205870 DOI: 10.1007/s00403-022-02508-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Revised: 11/29/2022] [Accepted: 12/05/2022] [Indexed: 01/01/2023]
Abstract
The transcriptomic regulation induced by isotretinoin (13-cis retinoic acid) is still a matter of debate as short-term exposures of immortalized sebocytes with isotretinoin produced conflicting results. Based on translational evidence, it has been hypothesized that oral isotretinoin treatment upregulates the expression of the transcription factor p53. Twenty-five patients suffering from acne vulgaris were treated with isotretinoin (0.6 mg/kg body weight) for 6 weeks. Biopsies from back skin were taken before and after isotretinoin treatment for the determination of p53 expression by immunohistochemical staining, quantification of p53 protein concentration by enzyme-linked immunosorbent assay and TP53 gene expression by quantitative reverse transcription real time PCR. Fifteen socio-demographically cross-matched healthy volunteers served as controls. Isotretinoin treatment significantly increased the nuclear expression of p53 in sebaceous glands of treated patients compared to pre-treatment levels and p53 levels of untreated controls. Furthermore, the p53 protein and gene expression significantly increased in the skin after treatment. The magnitude of p53 expression showed an inverse correlation to acne severity score and body mass index. Under clinical conditions, isotretinoin induced the expression of p53, which controls multiple transcription factors involved in the pathogenesis of acne vulgaris including FoxO1, androgen receptor and critical genes involved in the induction of autophagy and apoptosis. Increased p53-FoxO1 signalling enhanced by systemic isotretinoin treatment explains the underlying transcriptomic changes causing sebum suppression but also the adverse effects associated with systemic isotretinoin therapy.
Collapse
Affiliation(s)
- Naglaa Fathi Agamia
- Department of Dermatology, Andrology and Venereology, Faculty of Medicine, University of Alexandria, Alexandria, Egypt.
| | - Khalid Fawzi El Mulla
- Department of Dermatology, Andrology and Venereology, Faculty of Medicine, University of Alexandria, Alexandria, Egypt
| | - Naglaa Mohamed Alsayed
- Department of Dermatology, Andrology and Venereology, Faculty of Medicine, University of Alexandria, Alexandria, Egypt
| | - Rasha Mohamed Ghazala
- Department of Medical Biochemistry, Faculty of Medicine, University of Alexandria, Alexandria, Egypt
| | | | - Iman Mohamed Abdelmeniem
- Department of Dermatology, Andrology and Venereology, Faculty of Medicine, University of Alexandria, Alexandria, Egypt
| | - Iman Mamdouh Talaat
- Department of Pathology, Faculty of Medicine, University of Alexandria, Alexandria, Egypt
- Department of Clinical Sciences, College of Medicine, University of Sharjah, Sharjah, UAE
| | - Inass Ibrahim Zaki
- Department of Pathology, Faculty of Medicine, University of Alexandria, Alexandria, Egypt
| | - Rana Mohamed Sabah
- Department of Dermatology, Andrology and Venereology, Faculty of Medicine, University of Alexandria, Alexandria, Egypt
| | - Bodo Clemens Melnik
- Department of Dermatology, Environmental Medicine and Health Theory, University of Osnabrück, 49076, Osnabrück, Germany
| |
Collapse
|
36
|
Yabut KCB, Isoherranen N. Impact of Intracellular Lipid Binding Proteins on Endogenous and Xenobiotic Ligand Metabolism and Disposition. Drug Metab Dispos 2023; 51:700-717. [PMID: 37012074 PMCID: PMC10197203 DOI: 10.1124/dmd.122.001010] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 03/16/2023] [Accepted: 02/10/2023] [Indexed: 04/05/2023] Open
Abstract
The family of intracellular lipid binding proteins (iLBPs) is comprised of 16 members of structurally related binding proteins that have ubiquitous tissue expression in humans. iLBPs collectively bind diverse essential endogenous lipids and xenobiotics. iLBPs solubilize and traffic lipophilic ligands through the aqueous milieu of the cell. Their expression is correlated with increased rates of ligand uptake into tissues and altered ligand metabolism. The importance of iLBPs in maintaining lipid homeostasis is well established. Fatty acid binding proteins (FABPs) make up the majority of iLBPs and are expressed in major organs relevant to xenobiotic absorption, distribution, and metabolism. FABPs bind a variety of xenobiotics including nonsteroidal anti-inflammatory drugs, psychoactive cannabinoids, benzodiazepines, antinociceptives, and peroxisome proliferators. FABP function is also associated with metabolic disease, making FABPs currently a target for drug development. Yet the potential contribution of FABP binding to distribution of xenobiotics into tissues and the mechanistic impact iLBPs may have on xenobiotic metabolism are largely undefined. This review examines the tissue-specific expression and functions of iLBPs, the ligand binding characteristics of iLBPs, their known endogenous and xenobiotic ligands, methods for measuring ligand binding, and mechanisms of ligand delivery from iLBPs to membranes and enzymes. Current knowledge of the importance of iLBPs in affecting disposition of xenobiotics is collectively described. SIGNIFICANCE STATEMENT: The data reviewed here show that FABPs bind many drugs and suggest that binding of drugs to FABPs in various tissues will affect drug distribution into tissues. The extensive work and findings with endogenous ligands suggest that FABPs may also alter the metabolism and transport of drugs. This review illustrates the potential significance of this understudied area.
Collapse
Affiliation(s)
- King Clyde B Yabut
- Department of Pharmaceutics, School of Pharmacy, University of Washington, Seattle, Washington
| | - Nina Isoherranen
- Department of Pharmaceutics, School of Pharmacy, University of Washington, Seattle, Washington
| |
Collapse
|
37
|
Han X, Sun Y. PROTACs: A novel strategy for cancer drug discovery and development. MedComm (Beijing) 2023; 4:e290. [PMID: 37261210 PMCID: PMC10227178 DOI: 10.1002/mco2.290] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 05/08/2023] [Accepted: 05/09/2023] [Indexed: 06/02/2023] Open
Abstract
Proteolysis targeting chimera (PROTAC) technology has become a powerful strategy in drug discovery, especially for undruggable targets/proteins. A typical PROTAC degrader consists of three components: a small molecule that binds to a target protein, an E3 ligase ligand (consisting of an E3 ligase and its small molecule recruiter), and a chemical linker that hooks first two components together. In the past 20 years, we have witnessed advancement of multiple PROTAC degraders into the clinical trials for anticancer therapies. However, one of the major challenges of PROTAC technology is that only very limited number of E3 ligase recruiters are currently available as E3 ligand for targeted protein degradation (TPD), although human genome encodes more than 600 E3 ligases. Thus, there is an urgent need to identify additional effective E3 ligase recruiters for TPD applications. In this review, we summarized the existing RING-type E3 ubiquitin ligase and their small molecule recruiters that act as effective E3 ligands of PROTAC degraders and their application in anticancer drug discovery. We believe that this review could serve as a reference in future development of efficient E3 ligands of PROTAC technology for cancer drug discovery and development.
Collapse
Affiliation(s)
- Xin Han
- Cancer Institute (Key Laboratory of Cancer Prevention and InterventionChina National Ministry of Education) of the Second Affiliated Hospital and Institute of Translational MedicineZhejiang University School of MedicineHangzhouChina
- Cancer Center of Zhejiang UniversityHangzhouChina
- Zhejiang Provincial Clinical Research Center for CANCERZhejiang ProvinceChina
- Key Laboratory of Molecular Biology in Medical SciencesZhejiang ProvinceChina
| | - Yi Sun
- Cancer Institute (Key Laboratory of Cancer Prevention and InterventionChina National Ministry of Education) of the Second Affiliated Hospital and Institute of Translational MedicineZhejiang University School of MedicineHangzhouChina
- Cancer Center of Zhejiang UniversityHangzhouChina
- Zhejiang Provincial Clinical Research Center for CANCERZhejiang ProvinceChina
- Key Laboratory of Molecular Biology in Medical SciencesZhejiang ProvinceChina
- Research Center for Life Science and Human HealthBinjiang Institute of Zhejiang UniversityHangzhouChina
| |
Collapse
|
38
|
Hawkins MR, Wingert RA. Zebrafish as a Model to Study Retinoic Acid Signaling in Development and Disease. Biomedicines 2023; 11:biomedicines11041180. [PMID: 37189798 DOI: 10.3390/biomedicines11041180] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Revised: 04/06/2023] [Accepted: 04/13/2023] [Indexed: 05/17/2023] Open
Abstract
Retinoic acid (RA) is a metabolite of vitamin A (retinol) that plays various roles in development to influence differentiation, patterning, and organogenesis. RA also serves as a crucial homeostatic regulator in adult tissues. The role of RA and its associated pathways are well conserved from zebrafish to humans in both development and disease. This makes the zebrafish a natural model for further interrogation into the functions of RA and RA-associated maladies for the sake of basic research, as well as human health. In this review, we explore both foundational and recent studies using zebrafish as a translational model for investigating RA from the molecular to the organismal scale.
Collapse
Affiliation(s)
- Matthew R Hawkins
- Department of Biological Sciences, Center for Stem Cells and Regenerative Medicine, Center for Zebrafish Research, Boler-Parseghian Center for Rare and Neglected Diseases, Warren Center for Drug Discovery, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Rebecca A Wingert
- Department of Biological Sciences, Center for Stem Cells and Regenerative Medicine, Center for Zebrafish Research, Boler-Parseghian Center for Rare and Neglected Diseases, Warren Center for Drug Discovery, University of Notre Dame, Notre Dame, IN 46556, USA
| |
Collapse
|
39
|
Liu C, Zeng J, Wu J, Wang J, Wang X, Yao M, Zhang M, Fan J. Identification and validation of key genes associated with atrial fibrillation in the elderly. Front Cardiovasc Med 2023; 10:1118686. [PMID: 37063972 PMCID: PMC10090400 DOI: 10.3389/fcvm.2023.1118686] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Accepted: 03/02/2023] [Indexed: 03/31/2023] Open
Abstract
BackgroundAtrial fibrillation (AF) is the most common cardiac arrhythmia and significantly increases the risk of stroke and heart failure (HF), contributing to a higher mortality rate. Increasing age is a major risk factor for AF; however, the mechanisms of how aging contributes to the occurrence and progression of AF remain unclear. This study conducted weighted gene co-expression network analysis (WGCNA) to identify key modules and hub genes and determine their potential associations with aging-related AF.Materials and methodsWGCNA was performed using the AF dataset GSE2240 obtained from the Gene Expression Omnibus, which contained data from atrial myocardium in cardiac patients with permanent AF or sinus rhythm (SR). Hub genes were identified in clinical samples. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analyses were also performed.ResultsGreen and pink were the most critical modules associated with AF, from which nine hub genes, PTGDS, COLQ, ASTN2, VASH1, RCAN1, AMIGO2, RBP1, MFAP4, and ALDH1A1, were hypothesized to play key roles in the AF pathophysiology in elderly and seven of them have high diagnostic value. Functional enrichment analysis demonstrated that the green module was associated with the calcium, cyclic adenosine monophosphate (cAMP), and peroxisome proliferator-activated receptors (PPAR) signaling pathways, and the pink module may be associated with the transforming growth factor beta (TGF-β) signaling pathway in myocardial fibrosis.ConclusionWe identified nine genes that may play crucial roles in the pathophysiological mechanism of aging-related AF, among which six genes were associated with AF for the first time. This study provided novel insights into the impact of aging on the occurrence and progression of AF, and identified biomarkers and potential therapeutic targets for AF.
Collapse
Affiliation(s)
- Chuanbin Liu
- Western Medical Branch of PLA General Hospital, Beijing, China
| | - Jing Zeng
- Department of Endocrinology, The Second Medical Centre & National Clinical Research Centre for Geriatric Disease, Chinese PLA General Hospital, Beijing, China
| | - Jin Wu
- Tianjin Key Laboratory of Risk Assessment and Control Technology for Environment and Food Safety, Institute of Environmental and Operational Medicine, Tianjin, China
| | - Jing Wang
- Department of General Medicine, The First Medical Center of PLA General Hospital, Beijing, China
| | - Xin Wang
- Department of Ophthalmology, PLA Strategic Support Force Characteristic Medical Center, Beijing, China
| | - Minghui Yao
- Department of Cardiovascular Surgery, the First Medical Center of PLA General Hospital, Beijing, China
| | - Minghua Zhang
- Clinical Pharmacy Laboratory, Chinese PLA General Hospital, Beijing, China
- Correspondence: Minghua Zhang Jiao Fan
| | - Jiao Fan
- Institute of Geriatrics, The Second Medical Centre & National Clinical Research Centre for Geriatric Disease, Chinese PLA General Hospital, Beijing, China
- Correspondence: Minghua Zhang Jiao Fan
| |
Collapse
|
40
|
Kola-Mustapha AT, Raji MA, Adedeji O, Ambrose GO. Network Pharmacology and Molecular Modeling to Elucidate the Potential Mechanism of Neem Oil against Acne vulgaris. Molecules 2023; 28:molecules28062849. [PMID: 36985821 PMCID: PMC10056471 DOI: 10.3390/molecules28062849] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 03/02/2023] [Accepted: 03/03/2023] [Indexed: 03/30/2023] Open
Abstract
Acne vulgaris is a common skin disorder with a complicated etiology. Papules, lesions, comedones, blackheads, and other skin lesions are common physical manifestations of Acne vulgaris, but the individual who has it also regularly has psychological repercussions. Natural oils are being utilized more and more to treat skin conditions since they have fewer negative effects and are expected to provide benefits. Using network pharmacology, this study aims to ascertain if neem oil has any anti-acne benefits and, if so, to speculate on probable mechanisms of action for such effects. The neem leaves (Azadirachta indica) were collected, verified, authenticated, and assigned a voucher number. After steam distillation was used to extract the neem oil, the phytochemical components of the oil were examined using gas chromatography-mass spectrometry (GC-MS). The components of the oil were computationally examined for drug-likeness using Lipinski's criteria. The Pharm Mapper service was used to anticipate the targets. Prior to pathway and protein-protein interaction investigations, molecular docking was performed to predict binding affinity. Neem oil was discovered to be a potential target for STAT1, CSK, CRABP2, and SYK genes in the treatment of Acne vulgaris. In conclusion, it was discovered that the neem oil components with PubChem IDs: ID_610088 (2-(1-adamantyl)-N-methylacetamide), ID_600826 (N-benzyl-2-(2-methyl-5-phenyl-3H-1,3,4-thiadiazol-2-yl)acetamide), and ID_16451547 (N-(3-methoxyphenyl)-2-(1-phenyltetrazol-5-yl)sulfanylpropanamide) have strong affinities for these drug targets and may thus be used as therapeutic agents in the treatment of acne.
Collapse
Affiliation(s)
- Adeola Tawakalitu Kola-Mustapha
- College of Pharmacy, Alfaisal University Riyadh, Riyadh 11461, Saudi Arabia
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmaceutical Sciences, University of Ilorin, Ilorin 240101, Nigeria
| | - Muhabat Adeola Raji
- Department of Microbiology & Immunology, Alfaisal University, Riyadh 11461, Saudi Arabia
| | - Oluwakorede Adedeji
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmaceutical Sciences, University of Ilorin, Ilorin 240101, Nigeria
| | | |
Collapse
|
41
|
Schlotawa L, Tyka K, Kettwig M, Ahrens‐Nicklas RC, Baud M, Berulava T, Brunetti‐Pierri N, Gagne A, Herbst ZM, Maguire JA, Monfregola J, Pena T, Radhakrishnan K, Schröder S, Waxman EA, Ballabio A, Dierks T, Fischer A, French DL, Gelb MH, Gärtner J. Drug screening identifies tazarotene and bexarotene as therapeutic agents in multiple sulfatase deficiency. EMBO Mol Med 2023; 15:e14837. [PMID: 36789546 PMCID: PMC9994482 DOI: 10.15252/emmm.202114837] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Revised: 12/09/2022] [Accepted: 01/09/2023] [Indexed: 02/16/2023] Open
Abstract
Multiple sulfatase deficiency (MSD, MIM #272200) results from pathogenic variants in the SUMF1 gene that impair proper function of the formylglycine-generating enzyme (FGE). FGE is essential for the posttranslational activation of cellular sulfatases. MSD patients display reduced or absent sulfatase activities and, as a result, clinical signs of single sulfatase disorders in a unique combination. Up to date therapeutic options for MSD are limited and mostly palliative. We performed a screen of FDA-approved drugs using immortalized MSD patient fibroblasts. Recovery of arylsulfatase A activity served as the primary readout. Subsequent analysis confirmed that treatment of primary MSD fibroblasts with tazarotene and bexarotene, two retinoids, led to a correction of MSD pathophysiology. Upon treatment, sulfatase activities increased in a dose- and time-dependent manner, reduced glycosaminoglycan content decreased and lysosomal position and size normalized. Treatment of MSD patient derived induced pluripotent stem cells (iPSC) differentiated into neuronal progenitor cells (NPC) resulted in a positive treatment response. Tazarotene and bexarotene act to ultimately increase the stability of FGE variants. The results lay the basis for future research on the development of a first therapeutic option for MSD patients.
Collapse
Affiliation(s)
- Lars Schlotawa
- Department of Paediatrics and Adolescent MedicineUniversity Medical Centre GöttingenGöttingenGermany
| | - Karolina Tyka
- Department of Paediatrics and Adolescent MedicineUniversity Medical Centre GöttingenGöttingenGermany
| | - Matthias Kettwig
- Department of Paediatrics and Adolescent MedicineUniversity Medical Centre GöttingenGöttingenGermany
| | - Rebecca C Ahrens‐Nicklas
- Division of Human Genetics and MetabolismThe Children's Hospital of PhiladelphiaPhiladelphiaPAUSA
| | - Matthias Baud
- School of Chemistry and Institute for Life SciencesUniversity of SouthamptonSouthamptonUK
| | - Tea Berulava
- Department for Epigenetics and Systems Medicine in Neurodegenerative DiseasesGerman Centre for Neurodegenerative DiseasesGöttingenGermany
| | - Nicola Brunetti‐Pierri
- Telethon Institute of Genetics and MedicinePozzuoliItaly
- Department of Translational MedicineUniversity of Naples Federico IINaplesItaly
| | - Alyssa Gagne
- Center for Cellular and Molecular TherapeuticsThe Children's Hospital of PhiladelphiaPhiladelphiaPAUSA
- Department of Pathology and Laboratory MedicineThe Children's Hospital of PhiladelphiaPhiladelphiaPAUSA
| | | | - Jean A Maguire
- Center for Cellular and Molecular TherapeuticsThe Children's Hospital of PhiladelphiaPhiladelphiaPAUSA
- Department of Pathology and Laboratory MedicineThe Children's Hospital of PhiladelphiaPhiladelphiaPAUSA
| | - Jlenia Monfregola
- Telethon Institute of Genetics and MedicinePozzuoliItaly
- Department of Translational MedicineUniversity of Naples Federico IINaplesItaly
| | - Tonatiuh Pena
- Department for Epigenetics and Systems Medicine in Neurodegenerative DiseasesGerman Centre for Neurodegenerative DiseasesGöttingenGermany
- Bioinformatics UnitGerman Centre for Neurodegenerative DiseasesGöttingenGermany
| | | | - Sophie Schröder
- Department for Epigenetics and Systems Medicine in Neurodegenerative DiseasesGerman Centre for Neurodegenerative DiseasesGöttingenGermany
| | - Elisa A Waxman
- Center for Cellular and Molecular TherapeuticsThe Children's Hospital of PhiladelphiaPhiladelphiaPAUSA
- Department of Pathology and Laboratory MedicineThe Children's Hospital of PhiladelphiaPhiladelphiaPAUSA
| | - Andrea Ballabio
- Telethon Institute of Genetics and MedicinePozzuoliItaly
- Department of Translational MedicineUniversity of Naples Federico IINaplesItaly
- Department of Molecular and Human Genetics and Neurological Research InstituteBaylor College of MedicineHoustonTXUSA
| | - Thomas Dierks
- Faculty of Chemistry, Biochemistry IBielefeld UniversityBielefeldGermany
| | - André Fischer
- Department for Epigenetics and Systems Medicine in Neurodegenerative DiseasesGerman Centre for Neurodegenerative DiseasesGöttingenGermany
- Department of Psychiatry and PsychotherapyUniversity Medical Center GöttingenGöttingenGermany
- Multiscale Bioimaging Cluster of Excellence, University Medical Center GöttingenUniversity of GöttingenGöttingenGermany
| | - Deborah L French
- Center for Cellular and Molecular TherapeuticsThe Children's Hospital of PhiladelphiaPhiladelphiaPAUSA
- Department of Pathology and Laboratory MedicineThe Children's Hospital of PhiladelphiaPhiladelphiaPAUSA
| | - Michael H Gelb
- Department of ChemistryUniversity of WashingtonSeattleWAUSA
| | - Jutta Gärtner
- Department of Paediatrics and Adolescent MedicineUniversity Medical Centre GöttingenGöttingenGermany
| |
Collapse
|
42
|
Zeng S, Xu Z, Liang Q, Thakur A, Liu Y, Zhou S, Yan Y. The prognostic gene CRABP2 affects drug sensitivity by regulating docetaxel-induced apoptosis in breast invasive carcinoma: A pan-cancer analysis. Chem Biol Interact 2023; 373:110372. [PMID: 36736488 DOI: 10.1016/j.cbi.2023.110372] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2022] [Revised: 01/21/2023] [Accepted: 01/30/2023] [Indexed: 02/05/2023]
Abstract
Cellular retinoic acid-binding protein 2 (CRABP2), a specific transporter of retinoic acid, has been shown to have an important biological role in human cancers. However, due to the substantial variability among different tumors, the role of CRABP2 remains uncertain and has not yet been subjected to systematic analysis. Utilizing The Cancer Genome Atlas (TCGA), Genotype-Tissue Expression (GTEx), Clinical Proteomic Tumor Analysis Consortium (CPTAC), Human Protein Atlas (HPA), Gene Expression Profiling Interactive Analysis 2 (GEPIA2), Kaplan-Meier Plotter, Biomarker Exploration of Solid Tumors (BEST), Cancer Cell Line Encyclopedia (CCLE), Receiver Operating Characteristic plotter (ROC plotter), and other online public tools, expression levels of CRABP2 in breast invasive carcinoma (BRCA), lung adenocarcinoma (LUAD), and ovarian serous cystadenocarcinoma (OV) were found to be significantly greater than those in adjacent normal tissues, suggesting a correlation to poor prognosis. Among the three, CRABP2 expression in BRCA was most closely associated with clinical prognosis. In a study of docetaxel-treated BRCA patients, CRABP2 expression was significantly higher in the drug-resistant group. Colony formation and flow cytometry analysis were used to further investigate the relationship between CRABP2 and docetaxel sensitivity in BRCA cells MDA-MB-231and BT549. The knockdown of CRABP2 expression significantly reduced cell growth and increased sensitivity to the chemotherapeutic agent docetaxel in BRCA cells. Furthermore, CRABP2 knockdown augmented docetaxel-induced apoptosis. Molecular docking using SwissDock tool revealed that CRABP2 had a greater binding affinity to docetaxel than docetaxel-targeted proteins. This research provides an insight into the expression and prognostic potential of CRABP2 in cancers and suggests that CRABP2 may control docetaxel sensitivity in BRCA cells through apoptosis, warranting further investigation.
Collapse
Affiliation(s)
- Shuangshuang Zeng
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Zhijie Xu
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China; Department of Pathology, Xiangya Changde Hospital, Changde, 415000, Hunan, China
| | - Qiuju Liang
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Abhimanyu Thakur
- Ben May Department for Cancer Research, Pritzker School of Molecular Engineering, University of Chicago, Illinois, USA
| | - Yuanhong Liu
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Shangjun Zhou
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Yuanliang Yan
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China.
| |
Collapse
|
43
|
Guo Z, Zhao Y, Wu Y, Zhang Y, Wang R, Liu W, Zhang C, Yang X. Cellular retinol-binding protein 1: a therapeutic and diagnostic tumor marker. Mol Biol Rep 2023; 50:1885-1894. [PMID: 36515825 DOI: 10.1007/s11033-022-08179-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2022] [Accepted: 12/06/2022] [Indexed: 12/15/2022]
Abstract
Cellular Retinol Binding Protein 1 (CRBP1) gene is a protein coding gene located on human chromosome 3q21, which codifies a protein named CRBP1. CRBP1 is widely expressed in many tissues as a chaperone protein to regulate the uptake, subsequent esterification and bioavailability of retinol. CRBP1 combines retinol and retinaldehyde with high affinity to protect retinoids from non-specific oxidation, and transports retinoids to specific enzymes to promote the biosynthesis of retinoic acid. The vital role of CRBP1 in retinoids metabolism has been gradually discovered, which has been implicated in tumorigenesis. However, the precise functions of CRBP1 in different diseases are still poorly understood. The purpose of this review is to provide an overview of the role of CRBP1 in various diseases, especially in both the promotion and inhibition of cancers, which may also offer a novel biomarker and potential therapeutic target for human diseases.
Collapse
Affiliation(s)
- Zhiyuan Guo
- College of Life Science, Henan Normal University, Xinxiang, 453007, China
- State Key Laboratory Cultivation Base for Cell Differentiation Regulation, Henan Normal University, Xinxiang, 453007, China
| | - Yinshen Zhao
- College of Life Science, Henan Normal University, Xinxiang, 453007, China
- State Key Laboratory Cultivation Base for Cell Differentiation Regulation, Henan Normal University, Xinxiang, 453007, China
| | - Yuqi Wu
- College of Life Science, Henan Normal University, Xinxiang, 453007, China
- State Key Laboratory Cultivation Base for Cell Differentiation Regulation, Henan Normal University, Xinxiang, 453007, China
| | - Yuqi Zhang
- College of Life Science, Henan Normal University, Xinxiang, 453007, China
- State Key Laboratory Cultivation Base for Cell Differentiation Regulation, Henan Normal University, Xinxiang, 453007, China
| | - Ruoyan Wang
- College of Life Science, Henan Normal University, Xinxiang, 453007, China
- State Key Laboratory Cultivation Base for Cell Differentiation Regulation, Henan Normal University, Xinxiang, 453007, China
| | - Wan Liu
- College of Life Science, Henan Normal University, Xinxiang, 453007, China
- State Key Laboratory Cultivation Base for Cell Differentiation Regulation, Henan Normal University, Xinxiang, 453007, China
| | - Chaoyang Zhang
- College of Life Science, Henan Normal University, Xinxiang, 453007, China
- State Key Laboratory Cultivation Base for Cell Differentiation Regulation, Henan Normal University, Xinxiang, 453007, China
| | - Xianguang Yang
- College of Life Science, Henan Normal University, Xinxiang, 453007, China.
- State Key Laboratory Cultivation Base for Cell Differentiation Regulation, Henan Normal University, Xinxiang, 453007, China.
| |
Collapse
|
44
|
Cardeña-Núñez S, Callejas-Marín A, Villa-Carballar S, Rodríguez-Gallardo L, Sánchez-Guardado LÓ, Hidalgo-Sánchez M. CRABP-I Expression Patterns in the Developing Chick Inner Ear. BIOLOGY 2023; 12:biology12010104. [PMID: 36671796 PMCID: PMC9855850 DOI: 10.3390/biology12010104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Revised: 12/30/2022] [Accepted: 01/03/2023] [Indexed: 01/12/2023]
Abstract
The vertebrate inner ear is a complex three-dimensional sensorial structure with auditory and vestibular functions, regarded as an excellent system for analyzing events that occur during development, such as patterning, morphogenesis, and cell specification. Retinoic acid (RA) is involved in all these development processes. Cellular retinoic acid-binding proteins (CRABPs) bind RA with high affinity, buffering cellular free RA concentrations and consequently regulating the activation of precise specification programs mediated by particular regulatory genes. In the otic vesicle, strong CRABP-I expression was detected in the otic wall's dorsomedial aspect, where the endolymphatic apparatus develops, whereas this expression was lower in the ventrolateral aspect, where part of the auditory system forms. Thus, CRABP-I proteins may play a role in the specification of the dorsal-to-ventral and lateral-to-medial axe of the otic anlagen. Regarding the developing sensory patches, a process partly involving the subdivision of a ventromedial pro-sensory domain, the CRABP-I gene displayed different levels of expression in the presumptive territory of each sensory patch, which was maintained throughout development. CRABP-I was also relevant in the acoustic-vestibular ganglion and in the periotic mesenchyme. Therefore, CRABP-I could protect RA-sensitive cells in accordance with its dissimilar concentration in specific areas of the developing chick inner ear.
Collapse
|
45
|
Smith AD, Chen C, Cheung L, Dawson HD. Raw potato starch alters the microbiome, colon and cecal gene expression, and resistance to Citrobacter rodentium infection in mice fed a Western diet. Front Nutr 2023; 9:1057318. [PMID: 36704785 PMCID: PMC9871501 DOI: 10.3389/fnut.2022.1057318] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Accepted: 12/05/2022] [Indexed: 01/11/2023] Open
Abstract
Resistant starches (RS) are fermented in the cecum and colon to produce short-chain fatty acids and other microbial metabolites that can alter host physiology and the composition of the microbiome. We previously showed that mice fed a Total Western Diet (TWD) based on NHANES data that mimics the composition of a typical American diet, containing resistant potato starch (RPS), produced concentration dependent changes to the cecal short-chain fatty acids, the microbiome composition as well as gene expression changes in the cecum and colon that were most prevalent in mice fed the 10% RPS diet. We were then interested in whether feeding TWD/RPS would alter the resistance to bacterial-induced colitis caused by Citrobacter rodentium (Cr), a mouse pathogen that shares 66.7% of encoded genes with Enteropathogenic Escherichia coli. Mice were fed the TWD for 6 weeks followed by a 3-weeks on the RPS diets before infecting with Cr. Fecal Cr excretion was monitored over time and fecal samples were collected for 16S sequencing. Mice were euthanized on day 12 post-infection and cecal contents collected for 16S sequencing. Cecum and colon tissues were obtained for gene expression analysis, histology and to determine the level of mucosa-associated Cr. Feeding RPS increased the percentage of mice productively infected by Cr and fecal Cr excretion on day 4 post-infection. Mice fed the TWD/10% RPS diet also had greater colonization of colonic tissue at day 12 post-infection and colonic pathology. Both diet and infection altered the fecal and cecal microbiome composition with increased levels of RPS resulting in decreased α-diversity that was partially reversed by Cr infection. RNASeq analysis identified several mechanistic pathways that could be associated with the increased colonization of Cr-infected mice fed 10% RPS. In the distal colon we found a decrease in enrichment for genes associated with T cells, B cells, genes associated with the synthesis of DHA-derived SPMs and VA metabolism/retinoic acid signaling. We also found an increase in the expression of the potentially immunosuppressive gene, Ido1. These results suggest that high-level consumption of RPS in the context of a typical American diet, may alter susceptibility to gastrointestinal bacterial infections.
Collapse
|
46
|
Reboul E. Proteins involved in fat-soluble vitamin and carotenoid transport across the intestinal cells: New insights from the past decade. Prog Lipid Res 2023; 89:101208. [PMID: 36493998 DOI: 10.1016/j.plipres.2022.101208] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Revised: 11/28/2022] [Accepted: 11/29/2022] [Indexed: 12/12/2022]
Abstract
It is now well established that vitamins D, E, and K and carotenoids are not absorbed solely through passive diffusion. Broad-specificity membrane transporters such as SR-BI (scavenger receptor class B type I), CD36 (CD36 molecule), NPC1L1 (Niemann Pick C1-like 1) or ABCA1 (ATP-binding cassette A1) are involved in the uptake of these micronutrients from the lumen to the enterocyte cytosol and in their secretion into the bloodstream. Recently, the existence of efflux pathways from the enterocyte back to the lumen or from the bloodstream to the lumen, involving ABCB1 (P-glycoprotein/MDR1) or the ABCG5/ABCG8 complex, has also been evidenced for vitamins D and K. Surprisingly, no membrane proteins have been involved in dietary vitamin A uptake so far. After an overview of the metabolism of fat-soluble vitamins and carotenoids along the gastrointestinal tract (from the mouth to the colon where interactions with microbiota may occur), a focus is placed on the identified and candidate proteins participating in the apical uptake, intracellular transport, basolateral secretion and efflux back to the lumen of fat-soluble vitamins and carotenoids in enterocytes. This review also highlights the mechanisms that remain to be identified to fully unravel the pathways involved in fat-soluble vitamin and carotenoid intestinal absorption.
Collapse
|
47
|
Wei CW, Lerdall T, Najjar F, Wei LN. Depleting Cellular Retinoic Acid Binding Protein 1 Impairs UPR mt. JOURNAL OF CELLULAR SIGNALING 2023; 4:151-162. [PMID: 38706516 PMCID: PMC11068023 DOI: 10.33696/signaling.4.102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 05/07/2024]
Abstract
Mitochondrial dysfunction underlines neurodegenerative diseases which are mostly characterized by progressive degeneration of neurons. We previously reported that Cellular retinoic acid Binding protein 1 (Crabp1) knockout (CKO) mice spontaneously developed age-dependent motor degeneration, with defects accumulated in spinal motor neurons (MNs), the only cell type in spinal cord that expresses CRABP1. Here we uncovered that mitochondrial DNA (mtDNA) content and the expression of genes involved in respiration were significantly reduced in CKO mouse spinal cord, accompanied by significantly elevated reactive oxygen species (ROS) and unfolded protein load, indicating that CRABP1 deficiency caused mitochondrial dysfunction. Further analyses of spinal cord tissues revealed significant reduction in the expression and activity of superoxide dismutase 2 (SOD2), as well as defected mitochondrial unfolded protein response (UPRmt) pathway, specifically an increase in ATF5 mRNA but not its protein level, which suggested failure in the translational response of ATF5 in CKO. Consistently, eukaryotic initiation factor-2α, (eIF2α) phosphorylation was reduced in CKO spinal cord. In a CRABP1 knockdown MN1 model, siCrabp1-MN1, we validated the cell-autonomous function of CRABP1 in modulating the execution of UPRmt. This study reveals a new functional role for CRABP1 in the execution of mitochondrial stress response, that CRABP1 modulates eIF2α phosphorylation thereby contributing to ATF5 translational response that is needed to mitigate mitochondria stress.
Collapse
Affiliation(s)
- Chin-Wen Wei
- Department of Pharmacology, University of Minnesota, Minneapolis, MN 55455, USA
| | - Thomas Lerdall
- Department of Pharmacology, University of Minnesota, Minneapolis, MN 55455, USA
| | - Fatimah Najjar
- Department of Pharmacology, University of Minnesota, Minneapolis, MN 55455, USA
| | - Li-Na Wei
- Department of Pharmacology, University of Minnesota, Minneapolis, MN 55455, USA
| |
Collapse
|
48
|
Pharmacological Inhibition of Lipid Import and Transport Proteins in Ovarian Cancer. Cancers (Basel) 2022; 14:cancers14236004. [PMID: 36497485 PMCID: PMC9737127 DOI: 10.3390/cancers14236004] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Revised: 12/01/2022] [Accepted: 12/02/2022] [Indexed: 12/12/2022] Open
Abstract
Ovarian cancer (OC) is the most lethal gynecological malignancy with a 5-year survival rate of 49%. This is caused by late diagnosis when cells have already metastasized into the peritoneal cavity and to the omentum. OC progression is dependent on the availability of high-energy lipids/fatty acids (FA) provided by endogenous de novo biosynthesis and/or through import from the microenvironment. The blockade of these processes may thus represent powerful strategies against OC. While this has already been shown for inhibition of FA/lipid biosynthesis, evidence of the role of FA/lipid import/transport is still sparse. Therefore, we treated A2780 and SKOV3 OC cells with inhibitors of the lipid uptake proteins fatty acid translocase/cluster of differentiation 36 (FAT/CD36) and low-density lipoprotein (LDL) receptor (LDLR), as well as intracellular lipid transporters of the fatty acid-binding protein (FABP) family, fatty acid transport protein-2 (FATP2/SLC27A2), and ADP-ribosylation factor 6 (ARF6), which are overexpressed in OC. Proliferation was determined by formazan dye labeling/photometry and cell counting. Cell cycle analysis was performed by propidium iodide (PI) staining, and apoptosis was examined by annexin V/PI and active caspase 3 labeling and flow cytometry. RNA-seq data revealed altered stress and metabolism pathways. Overall, the small molecule inhibitors of lipid handling proteins BMS309403, HTS01037, NAV2729, SB-FI-26, and sulfosuccinimidyl oleate (SSO) caused a drug-specific, dose-/time-dependent inhibition of FA/LDL uptake, associated with reduced proliferation, cell cycle arrest, and apoptosis. Our findings indicate that OC cells are very sensitive to lipid deficiency. This dependency should be exploited for development of novel strategies against OC.
Collapse
|
49
|
Yamakawa S, Hayashi Y, Kako K, Sasakura Y, Morino Y, Wada H. Mechanism underlying retinoic acid-dependent metamorphosis in the starfish. Dev Biol 2022; 492:119-125. [DOI: 10.1016/j.ydbio.2022.10.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 09/05/2022] [Accepted: 10/07/2022] [Indexed: 11/03/2022]
|
50
|
Wang Z, Zhang N, Li F, Yue X. Effects of pre-partum dietary crude protein level on colostrum fat globule membrane proteins and the performance of Hu ewes and their offspring. Front Vet Sci 2022; 9:1046214. [DOI: 10.3389/fvets.2022.1046214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Accepted: 11/11/2022] [Indexed: 12/05/2022] Open
Abstract
Dietary proteins play important roles in the growth and reproduction of sheep, and the ewe's demand for proteins increases dramatically during late pregnancy. This research aimed to investigate the effect of dietary crude protein (CP) levels during late pregnancy on colostrum fat globule membrane (MFGM) protein and the growth performance of Hu sheep and their offspring, and provide a reference for the protein intake of ewes during late pregnancy. A total of 108 multiparous Hu sheep (45.6 ± 1.18 kg) were selected for this study, then 60 pregnant ewes confirmed by B-scan ultrasonography were randomly divided into three treatments (20 ewes/treatment) and fed by total mixed ration pellet with CP levels at 9.00% (LP), 12.0% (MP), and 15.0% (HP) during late pregnancy, respectively. The weight and dry matter intake of ewes during late pregnancy were recorded to calculate the average daily gain (ADG) and feed conversion ratio (FCR). Twin lambs were weighed on days 0, 7, 14, 30, 60, and 180 after birth to calculate ADG. Meanwhile, the colostrum of ewes was collected within 12 h after delivery. The colostrum MFGM proteins were identified and quantified by the isobaric tag for relative and absolute quantification (iTRAQ) coupled with liquid chromatography-tandem mass spectrometry methods. In addition, biological functions of differentially expressed proteins (DEPs) were annotated by Gene Ontology annotation and Kyoto Encyclopedia of Genes and Genomes pathway enrichment analysis. The results revealed that a 15.0% CP level had significant effects on the BW of lambs on days 0, 7, and 30 (P < 0.05). Notably, a total of 1,529 MFGM proteins were identified and 286 DEPs were found among three treatments. Functional analysis showed that DEPs were mainly involved in cell growth, differentiation, and tissue repair, and involved in metabolic pathways, such as the porphyrin and chlorophyll metabolism pathways. In this study, lambs in HP treatment had better growth performance; moreover, dietary 15.0% CP level also affected the colostrum MFGM proteins composition of Hu ewes. These observations can facilitate future studies on the feeding regimen of ewes during late pregnancy.
Collapse
|