1
|
Banerjee A, Thekkekkara D, Manjula SN, Nair SP, Lalitha MS. Correlation of autophagy and Alzheimer's disease with special emphasis on the role of phosphodiesterase-4. 3 Biotech 2025; 15:139. [PMID: 40292249 PMCID: PMC12018668 DOI: 10.1007/s13205-025-04306-5] [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: 02/14/2025] [Accepted: 04/02/2025] [Indexed: 04/30/2025] Open
Abstract
Autophagy disruption is important in Alzheimer's disease (AD) as it prevents misfolded proteins from being removed, which leads to the accumulation of amyloid plaques and neurofibrillary tangles (NFTs). Restoring autophagy improves neuronal survival and cognitive function, according to experimental models. In AD models, mTOR inhibition and AMPK activation enhance synaptic plasticity and lessen learning deficits. Inhibitors of phosphodiesterase-4 (PDE4) improve cognition and reduce neuroinflammation via altering cyclic adenosine monophosphate (cAMP) transmission. Furthermore, autophagic-lysosomal clearance is encouraged by upregulating transcription factor EB (TFEB), which lessens the pathogenic damage linked to AD. These results point to autophagy modification as a promising therapeutic approach, with the mTOR, AMPK, cAMP, and TFEB pathways being possible targets for drugs. Though much evidence is based on animal studies, these findings provide valuable insights into autophagy's role in AD pathology, offering promising directions for future research and drug development.
Collapse
Affiliation(s)
- Aniruddha Banerjee
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Mysore, Karnataka 570 015 India
| | - Dithu Thekkekkara
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Mysore, Karnataka 570 015 India
| | - S. N. Manjula
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Mysore, Karnataka 570 015 India
| | - Salini P. Nair
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Mysore, Karnataka 570 015 India
| | - Mankala Sree Lalitha
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Mysore, Karnataka 570 015 India
| |
Collapse
|
2
|
Yi H, Liang W, Yang S, Liu H, Deng J, Han S, Feng X, Cheng W, Chen Y, Hang J, Lu H, Ran R. Melanin deposition and key molecular features in Xenopus tropicalis oocytes. BMC Biol 2025; 23:62. [PMID: 40016733 PMCID: PMC11866844 DOI: 10.1186/s12915-025-02168-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Accepted: 02/18/2025] [Indexed: 03/01/2025] Open
Abstract
BACKGROUND Melanin pigmentation in oocytes is a critical feature for both the esthetic and developmental aspects of oocytes, influencing their polarity and overall development. Despite substantial knowledge of melanogenesis in melanocytes and retinal pigment epithelium cells, the molecular mechanisms underlying oocyte melanogenesis remain largely unknown. RESULTS Here, we compare the oocytes of wild-type, tyr-/- and mitf-/- Xenopus tropicalis and found that mitf-/- oocytes exhibit normal melanin deposition at the animal pole, whereas tyr-/- oocytes show no melanin deposition at this site. Transmission electron microscopy confirmed that melanogenesis in mitf-/- oocytes proceeds normally, similar to wild-type oocytes. Transcriptomic analysis revealed that mitf-/- oocytes still express melanogenesis-related genes, enabling them to complete melanogenesis. Additionally, in Xenopus tropicalis oocytes, the expression of the MiT subfamily factor tfe3 is relatively high, while tfeb, mitf, and tfec levels are extremely low. The expression pattern of tfe3 is similar to that of tyr and other melanogenesis-related genes. Thus, melanogenesis in Xenopus tropicalis oocytes is independent of Mitf and may be regulated by other MiT subfamily factors such as Tfe3, which control the expression of genes like tyr, dct, and tyrp1. Furthermore, transcriptomic data revealed that changes in the expression of genes related to mitochondrial cloud formation represent the most significant molecular changes during oocyte development. CONCLUSIONS Overall, these findings suggest that further elucidation of Tyr-dependent and Mitf-independent mechanisms of melanin deposition at the animal pole will enhance our understanding of melanogenesis and Oogenesis.
Collapse
Affiliation(s)
- Hongyang Yi
- National Clinical Research Centre for Infectious Diseases, the Third People'S Hospital of Shenzhenand, the Second Affiliated Hospital of Southern University of Science and Technologyaq , Shenzhen, 518112, China
- Department of Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Weizheng Liang
- Hebei Provincial Key Laboratory of Systems Biology and Gene Regulation, Central Laboratory, The First Affiliated Hospital of Hebei North University, Zhangjiakou, Hebei, 075000, China
| | - Sumei Yang
- National Clinical Research Centre for Infectious Diseases, the Third People'S Hospital of Shenzhenand, the Second Affiliated Hospital of Southern University of Science and Technologyaq , Shenzhen, 518112, China
| | - Han Liu
- National Clinical Research Centre for Infectious Diseases, the Third People'S Hospital of Shenzhenand, the Second Affiliated Hospital of Southern University of Science and Technologyaq , Shenzhen, 518112, China
| | - Jiayu Deng
- National Clinical Research Centre for Infectious Diseases, the Third People'S Hospital of Shenzhenand, the Second Affiliated Hospital of Southern University of Science and Technologyaq , Shenzhen, 518112, China
| | - Shuhong Han
- National Clinical Research Centre for Infectious Diseases, the Third People'S Hospital of Shenzhenand, the Second Affiliated Hospital of Southern University of Science and Technologyaq , Shenzhen, 518112, China
| | - Xiaohui Feng
- Department of Obstetrics and Gynecology, The Fifth Affiliated Hospital of Sun Yat-Sen University, Zhuhai, 519000, China
| | - Wenjie Cheng
- Department of Urology, The Fifth Affiliated Hospital of Sun Yat-Sen University, Zhuhai, 519000, China
| | - Yonglong Chen
- Department of Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, 518055, China.
| | - Jing Hang
- State Key Laboratory of Female Fertility Promotion, Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China.
| | - Hongzhou Lu
- National Clinical Research Centre for Infectious Diseases, the Third People'S Hospital of Shenzhenand, the Second Affiliated Hospital of Southern University of Science and Technologyaq , Shenzhen, 518112, China.
| | - Rensen Ran
- Department of Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, 518055, China.
- State Key Laboratory of Female Fertility Promotion, Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China.
| |
Collapse
|
3
|
Huang T, Sun C, Du F, Chen ZJ. STING-induced noncanonical autophagy regulates endolysosomal homeostasis. Proc Natl Acad Sci U S A 2025; 122:e2415422122. [PMID: 39982740 PMCID: PMC11874320 DOI: 10.1073/pnas.2415422122] [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: 08/01/2024] [Accepted: 01/21/2025] [Indexed: 02/22/2025] Open
Abstract
The cGAS-STING pathway mediates innate immune responses to cytosolic DNA. In addition to its well-established role in inducing inflammatory cytokines, activation of the cGAS-STING pathway also induces noncanonical autophagy, a process involving the conjugation of the ATG8 family of ubiquitin-like proteins to membranes of the endolysosomal system. The mechanisms and functions of STING-induced autophagy remain poorly understood. In this study, we demonstrated that STING activation induced formation of pH-elevated Golgi-derived vesicles that led to ATG16L1 and V-ATPase-dependent noncanonical autophagy. We showed that STING-induced noncanonical autophagy resulted in activation of the MiT/TFE family of transcription factors (TFEB, TFE3, and MITF), which regulate lysosome biogenesis. We found that lipidation of the ATG8 proteins, particularly GABARAPs, inhibited phosphorylation of MiT/TFE transcription factors by mTORC1. The lipidated GABARAPs bound to the Folliculin-interacting proteins (FNIPs), thereby sequestering the FNIP-folliculin protein complexes from activating mTORC1, resulting in dephosphorylation and nuclear translocation of MiT/TFE transcription factors. Furthermore, we found that STING-induced autophagy activated Leucine-rich repeat kinase 2 (LRRK2), a protein implicated in Parkinson's disease, through GABARAPs lipidation. We further showed that STING-induced autophagy induced ALIX-mediated ESCRT machinery recruitment to mitigate endolysosomal perturbation. These results reveal the multifaceted functions of STING-induced noncanonical autophagy in regulating endolysosomal homeostasis.
Collapse
Affiliation(s)
- Tuozhi Huang
- Department of Molecular Biology, University of Texas, Southwestern Medical Center, Dallas, TX75390-9148
- Center for Inflammation Research, University of Texas, Southwestern Medical Center, Dallas, TX75390-9148
| | - Chenglong Sun
- Department of Molecular Biology, University of Texas, Southwestern Medical Center, Dallas, TX75390-9148
- Center for Inflammation Research, University of Texas, Southwestern Medical Center, Dallas, TX75390-9148
| | - Fenghe Du
- Department of Molecular Biology, University of Texas, Southwestern Medical Center, Dallas, TX75390-9148
- Center for Inflammation Research, University of Texas, Southwestern Medical Center, Dallas, TX75390-9148
- HHMI, University of Texas, Southwestern Medical Center, Dallas, TX75390-9148
| | - Zhijian J. Chen
- Department of Molecular Biology, University of Texas, Southwestern Medical Center, Dallas, TX75390-9148
- Center for Inflammation Research, University of Texas, Southwestern Medical Center, Dallas, TX75390-9148
- HHMI, University of Texas, Southwestern Medical Center, Dallas, TX75390-9148
| |
Collapse
|
4
|
Zhang Y, Wang S, Yang A. Hydrolyzed conchiolin protein inhibits melanogenesis through PKA/CREB and MEK/ERK signalling pathways. Int J Cosmet Sci 2025; 47:31-44. [PMID: 39128885 PMCID: PMC11787999 DOI: 10.1111/ics.13012] [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: 04/20/2024] [Revised: 07/02/2024] [Accepted: 07/17/2024] [Indexed: 08/13/2024]
Abstract
OBJECTIVE Hydrolyzed conchiolin protein (HCP) derived from pearl and nacre extracts exerts skin-lightening effects; however, the underlying molecular mechanisms are not fully understood. Herein, we investigated the effect of HCP on melanogenesis and the signalling pathways involved. METHODS B16F10 cells and PIG cells were treated with HCP to verify its ability to inhibit melanin. Western Blot, immunofluorescence, and flow cytometry methods were performed to investigate the effect of HCP on melanogenesis signalling pathway proteins. The inhibitors were used to further validate the effect of HCP on PKA/CREB and MEK/ERK signalling pathways. To further evaluate the whitening ability of HCP, changes in melanin were detected using 3D melanin skin model and zebrafish model. RESULTS HCP was found to significantly inhibit melanin synthesis and decrease the expression of melanogenesis-related proteins, such as microphthalmia-associated transcription factor (MITF), tyrosinase, and tyrosinase-related protein-2, in a dose-dependent manner. Additionally, we revealed that HCP suppresses melanogenesis via the regulation of the PKA/cAMP response element-binding (CREB) and MEK/extracellular signalling-regulated kinase (ERK) signalling pathways. Using 3D melanin skin models, we demonstrated that HCP can achieve skin-lightening effects by improving apparent chroma, increasing apparent brightness, and inhibiting melanin synthesis. Furthermore, HCP exhibits skin-whitening effects in a zebrafish model. CONCLUSION These results suggest that HCP suppresses the melanogenesis signalling cascade by inhibiting the PKA/CREB, MEK/ERK signalling pathway and downregulating MITF and its downstream signalling pathways, resulting in decreased melanin synthesis. In summary, HCP is a potential anti-pigmentation agent with promising applications in cosmetics and pharmaceutical products.
Collapse
Affiliation(s)
| | - Sisi Wang
- Osmum Biological Co., LtdHuzhouChina
| | | |
Collapse
|
5
|
Niu B, An X, Chen Y, He T, Zhan X, Zhu X, Ping F, Zhang W, Zhou J. Nigella sativa L. seed extract alleviates oxidative stress-induced cellular senescence and dysfunction in melanocytes. Chin J Nat Med 2025; 23:203-213. [PMID: 39986696 DOI: 10.1016/s1875-5364(25)60824-7] [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: 04/26/2024] [Revised: 06/29/2024] [Accepted: 07/19/2024] [Indexed: 02/24/2025]
Abstract
Nigella sativa L. seeds have been traditionally utilized in Chinese folk medicine for centuries to treat vitiligo. This study revealed that the ethanolic extract of Nigella sativa L. (HZC) enhances melanogenesis and mitigates oxidative stress-induced cellular senescence and dysfunction in melanocytes. In accordance with established protocols, the ethanol fraction from Nigella sativa L. seeds was extracted, concentrated, and lyophilized to evaluate its herbal effects via 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assays, tyrosinase activity evaluation, measurement of cellular melanin contents, scratch assays, senescence-associated β-galactosidase (SA-β-gal) staining, enzyme-linked immunosorbent assay (ELISA), and Western blot analysis for expression profiling of experimentally relevant proteins. The results indicated that HZC significantly enhanced tyrosinase activity and melanin content while notably increasing the protein expression levels of Tyr, Mitf, and gp100 in B16F10 cells. Furthermore, HZC effectively mitigated oxidative stress-induced cellular senescence, improved melanocyte condition, and rectified various functional impairments associated with melanocyte dysfunction. These findings suggest that HZC increases melanin synthesis in melanocytes through the activation of the MAPK, PKA, and Wnt signaling pathways. In addition, HZC attenuates oxidative damage induced by H2O2 therapy by activating the nuclear factor E2-related factor 2-antioxidant response element (Nrf2-ARE) pathway and enhancing the activity of downstream antioxidant enzymes, thus preventing premature senescence and dysfunction in melanocytes.
Collapse
Affiliation(s)
- Ben Niu
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, China; Wuxi People's Hospital, Nanjing Medical University, Wuxi 214023, China
| | - Xiaohong An
- Yunnan Characteristic Plant Extraction Laboratory, Yunnan Yunke Characteristic Plant Extraction Laboratory Co., Ltd., Kunming 650106, China
| | - Yongmei Chen
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Ting He
- Drug Discovery and Development Laboratories, Ningxia Hui Medicine Research Institute, Yinchuan, 750021, China
| | - Xiao Zhan
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Xiuqi Zhu
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Fengfeng Ping
- Wuxi People's Hospital, Nanjing Medical University, Wuxi 214023, China
| | - Wei Zhang
- Hospital for Skin Diseases Institute of Dermatology, Chinese Academy of Medical Sciences & Peking Union Medical College, Nanjing 210042, China.
| | - Jia Zhou
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, China.
| |
Collapse
|
6
|
Hong Z, Wang D, Qiao X, Xie Y, Yang S, Hao K, Han C, Liu H, Liu Z. Wnt5a negatively regulates melanogenesis in primary Arctic fox epidermal melanocytes. Gene 2025; 934:149045. [PMID: 39461575 DOI: 10.1016/j.gene.2024.149045] [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: 07/12/2024] [Revised: 09/16/2024] [Accepted: 10/22/2024] [Indexed: 10/29/2024]
Abstract
Melanocytes, which are mainly found in the epidermis, are responsible for the melanin of skin and hair, and thereby contribute to the appearance of skin and provide protection from damage by ultraviolet radiation. Our previous study revealed that the Wnt5a, one of the many genes that affect melanin production, might be involved in the coat color seasonal change of the Arctic fox by influencing skin melanogenesis. Although the role of Wnt5a in melanocyte lines and melanoma cells has been extensively studied, its role in primary epidermal melanocytes has not been explored. This study aimed to investigate the role and mechanism of the Wnt5a in influencing melanogenesis in Arctic fox primary epidermal melanocytes. We constructed the Clustered Regularly Interspaced Short Palindromic Repeats (CRISPR) knockout plasmid targeting exons of the Wnt5a and transfected it into primary epidermal melanocytes. The results of the amplification knockout region assay, RT-qPCR assay, and western blot assay showed the success of Wnt5a knockout. RT-qPCR assay and melanin content assay showed that melanin production in melanocytes was significantly increased after Wnt5a knockout, and melanin-related key genes, such as microphthalmia-associated transcription factor, tyrosinase and tyrosinase-related protein 1, were significantly elevated. In addition, we also found that the expression of the β-catenin gene of the Wnt canonical pathway was significantly elevated after Wnt5a knockout. In conclusion, our results indicate that the Wnt5a plays a negative regulatory role in melanogenesis in primary epidermal melanocytes, and is presumably involved in antagonizing or inhibiting canonical Wnt signaling.
Collapse
Affiliation(s)
- Zhilin Hong
- College of Animal Science and Technology, Hebei Normal University of Science & Technology, Hebei Key Laboratory of Special Animal Germplasm Resources Mining and Innovation, Qinhuangdao, Hebei 066004, China
| | - Dongxian Wang
- College of Animal Science and Technology, Hebei Normal University of Science & Technology, Hebei Key Laboratory of Special Animal Germplasm Resources Mining and Innovation, Qinhuangdao, Hebei 066004, China
| | - Xian Qiao
- College of Animal Science and Technology, Hebei Normal University of Science & Technology, Hebei Key Laboratory of Special Animal Germplasm Resources Mining and Innovation, Qinhuangdao, Hebei 066004, China
| | - Yuchun Xie
- College of Animal Science and Technology, Hebei Normal University of Science & Technology, Hebei Key Laboratory of Special Animal Germplasm Resources Mining and Innovation, Qinhuangdao, Hebei 066004, China
| | - Shanshan Yang
- College of Animal Science and Technology, Hebei Normal University of Science & Technology, Hebei Key Laboratory of Special Animal Germplasm Resources Mining and Innovation, Qinhuangdao, Hebei 066004, China
| | - Kexing Hao
- College of Animal Science and Technology, Hebei Normal University of Science & Technology, Hebei Key Laboratory of Special Animal Germplasm Resources Mining and Innovation, Qinhuangdao, Hebei 066004, China
| | - Cong Han
- College of Animal Science and Technology, Hebei Normal University of Science & Technology, Hebei Key Laboratory of Special Animal Germplasm Resources Mining and Innovation, Qinhuangdao, Hebei 066004, China
| | - Huayun Liu
- College of Animal Science and Technology, Hebei Normal University of Science & Technology, Hebei Key Laboratory of Special Animal Germplasm Resources Mining and Innovation, Qinhuangdao, Hebei 066004, China
| | - Zhengzhu Liu
- College of Animal Science and Technology, Hebei Normal University of Science & Technology, Hebei Key Laboratory of Special Animal Germplasm Resources Mining and Innovation, Qinhuangdao, Hebei 066004, China.
| |
Collapse
|
7
|
Tedeschi V, Sapienza S, Ciancio R, Canzoniero LMT, Pannaccione A, Secondo A. Lysosomal Channels as New Molecular Targets in the Pharmacological Therapy of Neurodegenerative Diseases via Autophagy Regulation. Curr Neuropharmacol 2025; 23:375-383. [PMID: 38766825 DOI: 10.2174/1570159x22666240517101846] [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: 11/20/2023] [Revised: 12/20/2023] [Accepted: 01/16/2024] [Indexed: 05/22/2024] Open
Abstract
Besides controlling several organellar functions, lysosomal channels also guide the catabolic "self-eating" process named autophagy, which is mainly involved in protein and organelle quality control. Neuronal cells are particularly sensitive to the rate of autophagic flux either under physiological conditions or during the degenerative process. Accordingly, neurodegeneration occurring in Parkinson's (PD), Alzheimer's (AD), and Huntington's Diseases (HD), and Amyotrophic Lateral Sclerosis (ALS) as well as Lysosomal Storage Diseases (LSD) is partially due to defective autophagy and accumulation of toxic aggregates. In this regard, dysfunction of lysosomal ionic homeostasis has been identified as a putative cause of aberrant autophagy. From a therapeutic perspective, Transient Receptor Potential Channel Mucolipin 1 (TRPML1) and Two-Pore Channel isoform 2 (TPC2), regulating lysosomal homeostasis, are now considered promising druggable targets in neurodegenerative diseases. Compelling evidence suggests that pharmacological modulation of TRPML1 and TPC2 may rescue the pathological phenotype associated with autophagy dysfunction in AD, PD, HD, ALS, and LSD. Although pharmacological repurposing has identified several already used drugs with the ability to modulate TPC2, and several tools are already available for the modulation of TRPML1, many efforts are necessary to design and test new entities with much higher specificity in order to reduce dysfunctional autophagy during neurodegeneration.
Collapse
Affiliation(s)
- Valentina Tedeschi
- Department of Neuroscience, Reproductive and Odontostomatological Sciences, Federico II University of Naples, Via S. Pansini 5, 80131 Naples, Italy
| | - Silvia Sapienza
- Department of Neuroscience, Reproductive and Odontostomatological Sciences, Federico II University of Naples, Via S. Pansini 5, 80131 Naples, Italy
| | - Raffaella Ciancio
- Department of Neuroscience, Reproductive and Odontostomatological Sciences, Federico II University of Naples, Via S. Pansini 5, 80131 Naples, Italy
| | | | - Anna Pannaccione
- Department of Neuroscience, Reproductive and Odontostomatological Sciences, Federico II University of Naples, Via S. Pansini 5, 80131 Naples, Italy
| | - Agnese Secondo
- Department of Neuroscience, Reproductive and Odontostomatological Sciences, Federico II University of Naples, Via S. Pansini 5, 80131 Naples, Italy
| |
Collapse
|
8
|
El Mir J, Nasrallah A, Thézé N, Cario M, Fayyad‐Kazan H, Thiébaud P, Rezvani H. Xenopus as a model system for studying pigmentation and pigmentary disorders. Pigment Cell Melanoma Res 2025; 38:e13178. [PMID: 38849973 PMCID: PMC11681847 DOI: 10.1111/pcmr.13178] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Revised: 04/17/2024] [Accepted: 05/24/2024] [Indexed: 06/09/2024]
Abstract
Human pigmentary disorders encompass a broad spectrum of phenotypic changes arising from disruptions in various stages of melanocyte formation, the melanogenesis process, or the transfer of pigment from melanocytes to keratinocytes. A large number of pigmentation genes associated with pigmentary disorders have been identified, many of them awaiting in vivo confirmation. A more comprehensive understanding of the molecular basis of pigmentary disorders requires a vertebrate animal model where changes in pigmentation are easily observable in vivo and can be combined to genomic modifications and gain/loss-of-function tools. Here we present the amphibian Xenopus with its unique features that fulfill these requirements. Changes in pigmentation are particularly easy to score in Xenopus embryos, allowing whole-organism based phenotypic screening. The development and behavior of Xenopus melanocytes closely mimic those observed in mammals. Interestingly, both Xenopus and mammalian skins exhibit comparable reactions to ultraviolet radiation. This review highlights how Xenopus constitutes an alternative and complementary model to the more commonly used mouse and zebrafish, contributing to the advancement of knowledge in melanocyte cell biology and related diseases.
Collapse
Affiliation(s)
- Joudi El Mir
- University of Bordeaux, Inserm, BRIC, UMR 1312BordeauxFrance
| | - Ali Nasrallah
- University of Bordeaux, Inserm, BRIC, UMR 1312BordeauxFrance
| | - Nadine Thézé
- University of Bordeaux, Inserm, BRIC, UMR 1312BordeauxFrance
| | - Muriel Cario
- University of Bordeaux, Inserm, BRIC, UMR 1312BordeauxFrance
- Aquiderm, University of BordeauxBordeauxFrance
| | - Hussein Fayyad‐Kazan
- Laboratory of Cancer Biology and Molecular ImmunologyLebanese UniversityHadathLebanon
| | - Pierre Thiébaud
- University of Bordeaux, Inserm, BRIC, UMR 1312BordeauxFrance
| | - Hamid‐Reza Rezvani
- University of Bordeaux, Inserm, BRIC, UMR 1312BordeauxFrance
- Aquiderm, University of BordeauxBordeauxFrance
| |
Collapse
|
9
|
Leng D, Yang M, Miao X, Huang Z, Li M, Liu J, Wang T, Li D, Feng C. Dynamic changes in the skin transcriptome for the melanin pigmentation in embryonic chickens. Poult Sci 2025; 104:104210. [PMID: 39693959 PMCID: PMC11720608 DOI: 10.1016/j.psj.2024.104210] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Revised: 07/12/2024] [Accepted: 08/08/2024] [Indexed: 12/20/2024] Open
Abstract
Dermal hyperpigmentation stands out among the various skin pigmentation phenotypes in chickens, where most other pigmentation variants affect feather color and patterning predominantly. Despite numerous black chicken breeds worldwide, only a select few exhibit comprehensive black pigmentation, which encompasses the skin, meat, flesh, and bones. The process of skin melanin pigmentation is intricate and develops successively. Historically, research has concentrated primarily on specific developmental points or stages, but fewer studies have examined the entire transcriptome across the timeline of the development of the embryo integument. In our investigation, we undertook the sequencing of chicken embryo skin samples from d 4 to d 13 of incubation. Our results showed that melanoblasts continued to migrate from E4 to the epidermis until E12. Beginning with E6, melanin was synthesized and transferred to epidermal cells and feather follicles in large quantities, and genes such as DCT, TYR, TYRP1, and MITF played a key role in this process, which is significantly different from that of white-skinned chickens. There were 854 differentially expressed genes between E7 and E8. At this stage, melanocytes formed dendritic forms and transferred melanin to keratinocytes, while the dorsal skin became visibly dark. In addition, CDH3, which is a core factor involved in a variety of biological processes, may have an important impact on skin melanin pigmentation. Collectively, our findings unveiled a phased relationship between the canonical pathway and the noncanonical pathway from E4 to E13. These analyses illuminated the gene regulatory mechanism and provided foundational data that pertained to pigmentation in chickens.
Collapse
Affiliation(s)
- Dong Leng
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China; School of Pharmacy, Chengdu University, Chengdu 610106, China
| | - Maosen Yang
- School of Pharmacy, Chengdu University, Chengdu 610106, China; College of Animal Science and Technology, Guangxi University, Nanning 530004, China
| | - Xiaomeng Miao
- Institute of Animal Husbandry and Veterinary Medicine, Guizhou Academy of Agricultural Sciences, Guiyang 550005, China
| | - Zhiying Huang
- School of Pharmacy, Chengdu University, Chengdu 610106, China; College of Animal Science, Shanxi Agricultural University, Taiyuan 030031, China
| | - Mengmeng Li
- School of Pharmacy, Chengdu University, Chengdu 610106, China
| | - Jia Liu
- Guizhou Province Livestock and Poultry Genetic Resources Management Station, Guizhou Provincial Department of Agriculture and Rural Affairs, Guiyang 550001, China
| | - Tao Wang
- School of Pharmacy, Chengdu University, Chengdu 610106, China
| | - Diyan Li
- School of Pharmacy, Chengdu University, Chengdu 610106, China
| | - Chungang Feng
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China.
| |
Collapse
|
10
|
Mechmet F, Steingrímsson E, Petersen PH. Reduction in the olfactory ability in aging Mitf mutant mice without evidence of neurodegeneration. FRONTIERS IN AGING 2024; 5:1462900. [PMID: 39524487 PMCID: PMC11543575 DOI: 10.3389/fragi.2024.1462900] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Accepted: 09/30/2024] [Indexed: 11/16/2024]
Abstract
Age-related decline occurs in most brain structures and sensory systems. An illustrative case is olfaction. The olfactory bulb (OB) undergoes deterioration with age, resulting in reduced olfactory ability. A decline in olfaction is also associated with early symptoms of neurodegenerative diseases, including Alzheimer's disease (AD) and Parkinson's disease (PD). However, the underlying reasons are unclear. The microphthalmia-associated transcription factor (MITF) is expressed in the projection neurons (PNs) of the OB-the mitral and tufted (M/T) cells. Primary M/T cells from Mitf mutant mice show hyperactivity, potentially attributed to the reduced expression of a key potassium channel subunit, Kcnd3/Kv4.3. This influences intrinsic plasticity, an essential mechanism involving the non-synaptic regulation of neuronal activity. As neuronal hyperactivity often precedes neurodegenerative conditions, the current study aimed to determine whether the absence of Mitf causes degenerative effects during aging. Aged Mitf mutant mice showed reduced olfactory ability without inflammation. However, an increase in the expression of potassium channel subunit genes in the OBs of aged Mitf mi-vga9/mi-vga9 mice suggests that during aging, compensatory mechanisms lead to stabilization.
Collapse
Affiliation(s)
- Fatich Mechmet
- Department of Anatomy, Biomedical Center, Faculty of Medicine, University of Iceland, Reykjavik, Iceland
| | - Eiríkur Steingrímsson
- Department of Biochemistry and Molecular Biology, Biomedical Center, Faculty of Medicine, University of Iceland, Reykjavik, Iceland
| | - Petur Henry Petersen
- Department of Anatomy, Biomedical Center, Faculty of Medicine, University of Iceland, Reykjavik, Iceland
| |
Collapse
|
11
|
Markos A, Kubovciak J, Mikula Mrstakova S, Zitova A, Paces J, Machacova S, Kozmik-Jr Z, Kozmik Z, Kozmikova I. Cell type and regulatory analysis in amphioxus illuminates evolutionary origin of the vertebrate head. Nat Commun 2024; 15:8859. [PMID: 39402029 PMCID: PMC11473876 DOI: 10.1038/s41467-024-52938-7] [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: 01/26/2024] [Accepted: 09/25/2024] [Indexed: 10/17/2024] Open
Abstract
To shed light on the enigmatic origin of the vertebrate head, our study employs an integrated approach that combines single-cell transcriptomics, perturbations in signaling pathways, and cis-regulatory analysis in amphioxus. As a representative of a basal lineage within the chordate phylum, amphioxus retains many characteristics thought to have been present in the common chordate ancestor. Through cell type characterization, we identify the presence of prechordal plate-like, pre-migratory, and migratory neural crest-like cell populations in the developing amphioxus embryo. Functional analysis establishes conserved roles of the Nodal and Hedgehog signaling pathways in prechordal plate-like populations, and of the Wnt signaling pathway in neural crest-like populations' development. Furthermore, our trans-species transgenic experiments highlight similarities in the regulatory environments that drive neural crest-like and prechordal plate-like developmental programs in both vertebrates and amphioxus. Our findings provide evidence that the key features of vertebrate head development can be traced back to the common ancestor of all chordates.
Collapse
Affiliation(s)
- Anna Markos
- Laboratory of Transcriptional Regulation, Institute of Molecular Genetics of the Czech Academy of Sciences, Videnska 1083, Prague, Czech Republic
| | - Jan Kubovciak
- Laboratory of Genomics and Bioinformatics, Institute of Molecular Genetics of the Czech Academy of Sciences, Videnska 1083, Prague, Czech Republic
| | - Simona Mikula Mrstakova
- Laboratory of Transcriptional Regulation, Institute of Molecular Genetics of the Czech Academy of Sciences, Videnska 1083, Prague, Czech Republic
| | - Anna Zitova
- Laboratory of Transcriptional Regulation, Institute of Molecular Genetics of the Czech Academy of Sciences, Videnska 1083, Prague, Czech Republic
| | - Jan Paces
- Laboratory of Genomics and Bioinformatics, Institute of Molecular Genetics of the Czech Academy of Sciences, Videnska 1083, Prague, Czech Republic
| | - Simona Machacova
- Laboratory of Transcriptional Regulation, Institute of Molecular Genetics of the Czech Academy of Sciences, Videnska 1083, Prague, Czech Republic
| | - Zbynek Kozmik-Jr
- Laboratory of Genomics and Bioinformatics, Institute of Molecular Genetics of the Czech Academy of Sciences, Videnska 1083, Prague, Czech Republic
| | - Zbynek Kozmik
- Laboratory of Transcriptional Regulation, Institute of Molecular Genetics of the Czech Academy of Sciences, Videnska 1083, Prague, Czech Republic
| | - Iryna Kozmikova
- Laboratory of Transcriptional Regulation, Institute of Molecular Genetics of the Czech Academy of Sciences, Videnska 1083, Prague, Czech Republic.
| |
Collapse
|
12
|
Yan X, Yang L, Fu X, Luo X, Wang C, Xie QP, OuYang F. Transcription factor EB, a promising therapeutic target in cardiovascular disease. PeerJ 2024; 12:e18209. [PMID: 39403192 PMCID: PMC11472789 DOI: 10.7717/peerj.18209] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Accepted: 09/10/2024] [Indexed: 10/19/2024] Open
Abstract
Cardiovascular disease (CVD) remains the major cause of morbidity and mortality around the world. Transcription factor EB (TFEB) is a master regulator of lysosome biogenesis and autophagy. Emerging studies revealed that TFEB also mediates cellular adaptation responses to various stimuli, such as mitochondrial dysfunction, pathogen infection and metabolic toxin. Based on its significant capability to modulate the autophagy-lysosome process (ALP), TFEB plays a critical role in the development of CVD. In this review, we briefly summarize that TFEB regulates cardiac dysfunction mainly through ameliorating lysosomal and mitochondrial dysfunction and reducing inflammation.
Collapse
Affiliation(s)
- Xin Yan
- Department of Cardiovascular Medicine, Zhuzhou Hospital Affiliated to Xiangya School of Medicine, Central South University, Zhuzhou, China
| | - Li Yang
- Department of Cardiovascular Medicine, Zhuzhou Hospital Affiliated to Xiangya School of Medicine, Central South University, Zhuzhou, China
| | - Xiaolei Fu
- Department of Cardiovascular Medicine, Zhuzhou Hospital Affiliated to Xiangya School of Medicine, Central South University, Zhuzhou, China
| | - Xin Luo
- Department of Cardiovascular Medicine, Zhuzhou Hospital Affiliated to Xiangya School of Medicine, Central South University, Zhuzhou, China
| | - Chengming Wang
- Department of Cardiovascular Medicine, Zhuzhou Hospital Affiliated to Xiangya School of Medicine, Central South University, Zhuzhou, China
| | - Qiu Ping Xie
- Department of Cardiovascular Medicine, Zhuzhou Hospital Affiliated to Xiangya School of Medicine, Central South University, Zhuzhou, China
| | - Fan OuYang
- Department of Cardiovascular Medicine, Zhuzhou Hospital Affiliated to Xiangya School of Medicine, Central South University, Zhuzhou, China
| |
Collapse
|
13
|
Moritz RKC, Ebelt N, Rattay T, Ehrenreich J, Sunderkötter C, Gerloff D. The Expression of miR-211-5p in Sentinel Lymph Node Metastases of Malignant Melanoma Is a Potential Marker for Poor Prognosis. Int J Mol Sci 2024; 25:10859. [PMID: 39409187 PMCID: PMC11477290 DOI: 10.3390/ijms251910859] [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: 08/20/2024] [Revised: 09/30/2024] [Accepted: 10/07/2024] [Indexed: 10/20/2024] Open
Abstract
Metastatic primary cutaneous melanoma is a frequently fatal disease despite recent therapeutic advances. Biomarkers to stratify patients' prognosis are lacking. MicroRNAs (miRNAs) are small, non-coding RNAs. We aimed to determine the expression of miR-211-5p in primary tumors and metastases of malignant melanoma and its potential use as a prognostic biomarker. We performed in situ hybridization for miRNA-211-5p on 109 FFPE melanoma samples from 76 patients, including 31 paired primary tumor/metastasis samples. For validation, we performed in silico analyses of TCGA skin cutaneous melanoma (SKCM) cohort. High miR-211-5p expression was more frequent in primary tumors (70.8%) compared to metastases (39.3%). In metastases, it was associated with a significantly worse overall survival. Data from TCGA SKCM cohort confirmed that high miR-211-5p expression in melanoma metastases, but not primary tumors, is associated with worse overall survival. MiR-211-5p expression in metastases is associated with a shorter survival, emphasizing the potential of miR-211-5p as a risk predictor for a less favorable clinical outcome in metastatic disease. In situ hybridization could be implemented in a routine laboratory workflow and can be performed on diagnostic tissue.
Collapse
Affiliation(s)
- Rose Kathrin Caroline Moritz
- Department of Dermatology and Venereology, University Hospital Halle (Saale), Martin-Luther-University Halle-Wittenberg, 06120 Halle (Saale), Germany; (R.K.C.M.); (N.E.); (T.R.); (J.E.); (C.S.)
- Department of Dermatology, Venereology and Allergology, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt—Universität zu Berlin, 10117 Berlin, Germany
| | - Nicole Ebelt
- Department of Dermatology and Venereology, University Hospital Halle (Saale), Martin-Luther-University Halle-Wittenberg, 06120 Halle (Saale), Germany; (R.K.C.M.); (N.E.); (T.R.); (J.E.); (C.S.)
| | - Tina Rattay
- Department of Dermatology and Venereology, University Hospital Halle (Saale), Martin-Luther-University Halle-Wittenberg, 06120 Halle (Saale), Germany; (R.K.C.M.); (N.E.); (T.R.); (J.E.); (C.S.)
| | - Jovine Ehrenreich
- Department of Dermatology and Venereology, University Hospital Halle (Saale), Martin-Luther-University Halle-Wittenberg, 06120 Halle (Saale), Germany; (R.K.C.M.); (N.E.); (T.R.); (J.E.); (C.S.)
| | - Cord Sunderkötter
- Department of Dermatology and Venereology, University Hospital Halle (Saale), Martin-Luther-University Halle-Wittenberg, 06120 Halle (Saale), Germany; (R.K.C.M.); (N.E.); (T.R.); (J.E.); (C.S.)
| | - Dennis Gerloff
- Department of Dermatology and Venereology, University Hospital Halle (Saale), Martin-Luther-University Halle-Wittenberg, 06120 Halle (Saale), Germany; (R.K.C.M.); (N.E.); (T.R.); (J.E.); (C.S.)
| |
Collapse
|
14
|
Vu HN, Valdimarsson MM, Sigurbjörnsdóttir S, Bergsteinsdóttir K, Debbache J, Bismuth K, Swing DA, Hallsson JH, Larue L, Arnheiter H, Copeland NG, Jenkins NA, Heidarsson PO, Steingrímsson E. Novel mechanisms of MITF regulation identified in a mouse suppressor screen. EMBO Rep 2024; 25:4252-4280. [PMID: 39169200 PMCID: PMC11467436 DOI: 10.1038/s44319-024-00225-3] [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: 02/22/2024] [Revised: 07/08/2024] [Accepted: 07/17/2024] [Indexed: 08/23/2024] Open
Abstract
MITF, a basic Helix-Loop-Helix Zipper (bHLHZip) transcription factor, plays vital roles in melanocyte development and functions as an oncogene. We perform a genetic screen for suppressors of the Mitf-associated pigmentation phenotype in mice and identify an intragenic Mitf mutation that terminates MITF at the K316 SUMOylation site, leading to loss of the C-end intrinsically disordered region (IDR). The resulting protein is more nuclear but less stable than wild-type MITF and retains DNA-binding ability. As a dimer, it can translocate wild-type and mutant MITF partners into the nucleus, improving its own stability thus ensuring nuclear MITF supply. smFRET analysis shows interactions between K316 SUMOylation and S409 phosphorylation sites across monomers; these interactions largely explain the observed effects. The recurrent melanoma-associated E318K mutation in MITF, which affects K316 SUMOylation, also alters protein regulation in concert with S409. This suggests that residues K316 and S409 of MITF are impacted by SUMOylation and phosphorylation, respectively, mediating effects on nuclear localization and stability through conformational changes. Our work provides a novel mechanism of genetic suppression, and an example of how apparently deleterious mutations lead to normal phenotypes.
Collapse
Affiliation(s)
- Hong Nhung Vu
- Department of Biochemistry and Molecular Biology, BioMedical Center, Faculty of Medicine, University of Iceland, Sturlugata 8, 102, Reykjavík, Iceland
| | - Matti Már Valdimarsson
- Department of Biochemistry, Science Institute, School of Engineering and Natural Sciences, University of Iceland, Sturlugata 7, 102, Reykjavík, Iceland
| | - Sara Sigurbjörnsdóttir
- Department of Biochemistry and Molecular Biology, BioMedical Center, Faculty of Medicine, University of Iceland, Sturlugata 8, 102, Reykjavík, Iceland
| | - Kristín Bergsteinsdóttir
- Department of Biochemistry and Molecular Biology, BioMedical Center, Faculty of Medicine, University of Iceland, Sturlugata 8, 102, Reykjavík, Iceland
| | - Julien Debbache
- Mammalian Development Section, NINDS, NIH, Bethesda, MD, 20892-3706, USA
| | - Keren Bismuth
- Mammalian Development Section, NINDS, NIH, Bethesda, MD, 20892-3706, USA
| | - Deborah A Swing
- Mouse Cancer Genetics Program, NCI, Frederick, MD, 21702-1201, USA
| | - Jón H Hallsson
- Department of Biochemistry and Molecular Biology, BioMedical Center, Faculty of Medicine, University of Iceland, Sturlugata 8, 102, Reykjavík, Iceland
| | - Lionel Larue
- Institut Curie, PSL Research University, INSERM U1021, Normal and Pathological Development of Melanocytes, 91405, Orsay, France
| | - Heinz Arnheiter
- Mammalian Development Section, NINDS, NIH, Bethesda, MD, 20892-3706, USA
| | - Neal G Copeland
- Mouse Cancer Genetics Program, NCI, Frederick, MD, 21702-1201, USA
- Genetics Department, MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Nancy A Jenkins
- Mouse Cancer Genetics Program, NCI, Frederick, MD, 21702-1201, USA
- Genetics Department, MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Petur O Heidarsson
- Department of Biochemistry, Science Institute, School of Engineering and Natural Sciences, University of Iceland, Sturlugata 7, 102, Reykjavík, Iceland
| | - Eiríkur Steingrímsson
- Department of Biochemistry and Molecular Biology, BioMedical Center, Faculty of Medicine, University of Iceland, Sturlugata 8, 102, Reykjavík, Iceland.
| |
Collapse
|
15
|
Fischer KD, Tiwari S, Thier B, Qiu LC, Lin TC, Paschen A, Imig J. Long non-coding RNA GRASLND links melanoma differentiation and interferon-gamma response. Front Mol Biosci 2024; 11:1471100. [PMID: 39398277 PMCID: PMC11466874 DOI: 10.3389/fmolb.2024.1471100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Accepted: 09/13/2024] [Indexed: 10/15/2024] Open
Abstract
Melanoma is a highly malignant tumor, that stands as the most lethal form of skin cancer and is characterized by notable phenotypic plasticity and intratumoral heterogeneity. Melanoma plasticity is involved in tumor growth, metastasis and therapy resistance. Long non-coding RNAs (lncRNAs) could influence plasticity due to their regulatory function. However, their role and mode of action are poorly studied. Here, we show a relevance of lncRNA GRASLND in melanoma differentiation and IFNγ signaling. GRASLND knockdown revealed switching of differentiated, melanocytic melanoma cells towards a dedifferentiated, slow-proliferating and highly-invasive cell state. Interestingly, GRASLND is overexpressed in differentiated melanomas and associated with poor prognosis. Accordingly, we found GRASLND expressed in immunological "cold" tumors and it negatively correlates with gene signatures of immune response activation. In line, silencing of GRASLND under IFNγ enhanced the expression of IFNγ-stimulated genes, including HLA-I antigen presentation, demonstrating suppressive activity of GRASLND on IFNγ signaling. Our findings demonstrate that in differentiated melanomas elevated expression of GRASLND interferes with anti-tumor effects of IFNγ, suggesting a role of GRASLND in tumor immune evasion.
Collapse
Affiliation(s)
- Kim Denise Fischer
- Chemical Genomics Centre, Max Planck Institute of Molecular Physiology, Dortmund, Germany
- Faculty of Chemistry and Chemical Biology, Technical University of Dortmund, Dortmund, Germany
| | - Shashank Tiwari
- Chemical Genomics Centre, Max Planck Institute of Molecular Physiology, Dortmund, Germany
| | - Beatrice Thier
- Department of Dermatology, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Lin Christina Qiu
- Chemical Genomics Centre, Max Planck Institute of Molecular Physiology, Dortmund, Germany
- Faculty of Chemistry and Chemical Biology, Technical University of Dortmund, Dortmund, Germany
| | - Tzu-Chen Lin
- Faculty of Chemistry and Chemical Biology, Technical University of Dortmund, Dortmund, Germany
| | - Annette Paschen
- Department of Dermatology, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Jochen Imig
- Chemical Genomics Centre, Max Planck Institute of Molecular Physiology, Dortmund, Germany
| |
Collapse
|
16
|
García-Llorca A, Eysteinsson T. The Microphthalmia-Associated Transcription Factor (MITF) and Its Role in the Structure and Function of the Eye. Genes (Basel) 2024; 15:1258. [PMID: 39457382 PMCID: PMC11508060 DOI: 10.3390/genes15101258] [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: 09/06/2024] [Revised: 09/21/2024] [Accepted: 09/24/2024] [Indexed: 10/28/2024] Open
Abstract
BACKGROUND/OBJECTIVES The microphthalmia-associated transcription factor (Mitf) has been found to play an important role in eye development, structure, and function. The Mitf gene is responsible for controlling cellular processes in a range of cell types, contributing to multiple eye development processes. In this review, we survey what is now known about the impact of Mitf on eye structure and function in retinal disorders. Several mutations in the human and mouse Mitf gene are now known, and the effects of these on eye phenotype are addressed. We discuss the importance of Mitf in regulating ion transport across the retinal pigment epithelium (RPE) and the vasculature of the eye. METHODS The literature was searched using the PubMed, Scopus, and Google Scholar databases. Fundus and Optical Coherence Tomography (OCT) images from mice were obtained with a Micron IV rodent imaging system. RESULTS Defects in neural-crest-derived melanocytes resulting from any Mitf mutations lead to hypopigmentation in the eye, coat, and inner functioning of the animals. While many Mitf mutations target RPE cells in the eye, fewer impact osteoclasts at the same time. Some of the mutations in mice lead to microphthalmia, and ultimately vision loss, while other mice show a normal eye size; however, the latter, in some cases, show hypopigmentation in the fundus and the choroid is depigmented and thickened, and in rare cases Mitf mutations lead to progressive retinal degeneration. CONCLUSIONS The Mitf gene has an impact on the structure and function of the retina and its vasculature, the RPE, and the choroid in the adult eye.
Collapse
Affiliation(s)
- Andrea García-Llorca
- Department of Physiology, Biomedical Center, Faculty of Medicine, University of Iceland, 101 Reykjavík, Iceland;
| | - Thor Eysteinsson
- Department of Physiology, Biomedical Center, Faculty of Medicine, University of Iceland, 101 Reykjavík, Iceland;
- Department of Ophthalmology, Biomedical Center, Faculty of Medicine, University of Iceland, 101 Reykjavík, Iceland
| |
Collapse
|
17
|
Rubin CJ, Hodge M, Naboulsi R, Beckman M, Bellone RR, Kallenberg A, J'Usrey S, Ohmura H, Seki K, Furukawa R, Ohnuma A, Davis BW, Tozaki T, Lindgren G, Andersson L. An intronic copy number variation in Syntaxin 17 determines speed of greying and melanoma incidence in Grey horses. Nat Commun 2024; 15:7510. [PMID: 39209879 PMCID: PMC11362437 DOI: 10.1038/s41467-024-51898-2] [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: 11/09/2023] [Accepted: 08/20/2024] [Indexed: 09/04/2024] Open
Abstract
The Greying with age phenotype in horses involves loss of hair pigmentation whereas skin pigmentation is not reduced, and a predisposition to melanoma. The causal mutation was initially reported as a duplication of a 4.6 kb intronic sequence in Syntaxin 17. The speed of greying varies considerably among Grey horses. Here we demonstrate the presence of two different Grey alleles, G2 carrying two tandem copies of the duplicated sequence and G3 carrying three. The latter is by far the most common allele, probably due to strong selection for the striking white phenotype. Our results reveal a remarkable dosage effect where the G3 allele is associated with fast greying and high incidence of melanoma whereas G2 is associated with slow greying and low incidence of melanoma. The copy number expansion transforms a weak enhancer to a strong melanocyte-specific enhancer that underlies hair greying (G2 and G3) and a drastically elevated risk of melanoma (G3 only). Our direct pedigree-based observation of the origin of a G2 allele from a G3 allele by copy number contraction demonstrates the dynamic evolution of this locus and provides the ultimate evidence for causality of the copy number variation of the 4.6 kb intronic sequence.
Collapse
Affiliation(s)
- Carl-Johan Rubin
- Science for Life Laboratory, Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden
- Institute of Marine Research, Bergen, Norway
| | - McKaela Hodge
- Department of Veterinary Integrative Biosciences, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, Texas, USA
| | - Rakan Naboulsi
- Department of Animal Sciences, Swedish University of Agricultural Sciences, Uppsala, Sweden
- Childhood Cancer Research Unit, Department of Women's and Children's Health, Karolinska Institute, Tomtebodavägen 18A, 17177, Stockholm, Sweden
| | | | - Rebecca R Bellone
- Veterinary Genetics Laboratory, School of Veterinary Medicine, University of California-Davis, Davis, California, USA
- Department of Population Health and Reproduction, School of Veterinary Medicine, University of California, Davis, CA, USA
| | - Angelica Kallenberg
- Veterinary Genetics Laboratory, School of Veterinary Medicine, University of California-Davis, Davis, California, USA
| | - Stephanie J'Usrey
- Veterinary Genetics Laboratory, School of Veterinary Medicine, University of California-Davis, Davis, California, USA
| | - Hajime Ohmura
- Racehorse hospital, Miho Training Center, Japan Racing Association, Ibaraki, Japan
| | - Kazuhiro Seki
- Hidaka Training and Research Center, Japan Racing Association, Hokkaido, Japan
| | - Risako Furukawa
- Genetic Analysis Department, Laboratory of Racing Chemistry, Tochigi, Japan
| | - Aoi Ohnuma
- Genetic Analysis Department, Laboratory of Racing Chemistry, Tochigi, Japan
| | - Brian W Davis
- Department of Veterinary Integrative Biosciences, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, Texas, USA
| | - Teruaki Tozaki
- Genetic Analysis Department, Laboratory of Racing Chemistry, Tochigi, Japan
| | - Gabriella Lindgren
- Department of Animal Sciences, Swedish University of Agricultural Sciences, Uppsala, Sweden
| | - Leif Andersson
- Science for Life Laboratory, Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden.
- Department of Veterinary Integrative Biosciences, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, Texas, USA.
| |
Collapse
|
18
|
Zhao JF, Shpiro N, Sathe G, Brewer A, Macartney TJ, Wood NT, Negoita F, Sakamoto K, Sapkota GP. Targeted dephosphorylation of TFEB promotes its nuclear translocation. iScience 2024; 27:110432. [PMID: 39081292 PMCID: PMC11284556 DOI: 10.1016/j.isci.2024.110432] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Revised: 03/07/2024] [Accepted: 06/28/2024] [Indexed: 08/02/2024] Open
Abstract
Reversible phosphorylation of the transcription factor EB (TFEB) coordinates cellular responses to metabolic and other stresses. During nutrient replete and stressor-free conditions, phosphorylated TFEB is primarily localized to the cytoplasm. Stressor-mediated reduction of TFEB phosphorylation promotes its nuclear translocation and context-dependent transcriptional activity. In this study, we explored targeted dephosphorylation of TFEB as an approach to activate TFEB in the absence of nutrient deprivation or other cellular stress. Through an induction of proximity between TFEB and several phosphatases using the AdPhosphatase system, we demonstrate targeted dephosphorylation of TFEB in cells. Furthermore, by developing a heterobifunctional molecule BDPIC (bromoTAG-dTAG proximity-inducing chimera), we demonstrate targeted dephosphorylation of TFEB-dTAG through induced proximity to bromoTAG-PPP2CA. Targeted dephosphorylation of TFEB-dTAG by bromoTAG-PPP2CA with BDPIC at the endogenous levels is sufficient to induce nuclear translocation and some transcriptional activity of TFEB.
Collapse
Affiliation(s)
- Jin-Feng Zhao
- Medical Research Council (MRC) Protein Phosphorylation & Ubiquitylation Unit, School of Life Sciences, University of Dundee, Dundee DD1 5EH, UK
| | - Natalia Shpiro
- Medical Research Council (MRC) Protein Phosphorylation & Ubiquitylation Unit, School of Life Sciences, University of Dundee, Dundee DD1 5EH, UK
| | - Gajanan Sathe
- Medical Research Council (MRC) Protein Phosphorylation & Ubiquitylation Unit, School of Life Sciences, University of Dundee, Dundee DD1 5EH, UK
| | - Abigail Brewer
- Medical Research Council (MRC) Protein Phosphorylation & Ubiquitylation Unit, School of Life Sciences, University of Dundee, Dundee DD1 5EH, UK
| | - Thomas J. Macartney
- Medical Research Council (MRC) Protein Phosphorylation & Ubiquitylation Unit, School of Life Sciences, University of Dundee, Dundee DD1 5EH, UK
| | - Nicola T. Wood
- Medical Research Council (MRC) Protein Phosphorylation & Ubiquitylation Unit, School of Life Sciences, University of Dundee, Dundee DD1 5EH, UK
| | - Florentina Negoita
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Kei Sakamoto
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Gopal P. Sapkota
- Medical Research Council (MRC) Protein Phosphorylation & Ubiquitylation Unit, School of Life Sciences, University of Dundee, Dundee DD1 5EH, UK
| |
Collapse
|
19
|
Münchhalfen M, Görg R, Haberl M, Löber J, Willenbrink J, Schwarzt L, Höltermann C, Ickes C, Hammermann L, Kus J, Chapuy B, Ballabio A, Reichardt SD, Flügel A, Engels N, Wienands J. TFEB activation hallmarks antigenic experience of B lymphocytes and directs germinal center fate decisions. Nat Commun 2024; 15:6971. [PMID: 39138218 PMCID: PMC11322606 DOI: 10.1038/s41467-024-51166-3] [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: 03/28/2024] [Accepted: 07/31/2024] [Indexed: 08/15/2024] Open
Abstract
Ligation of the B cell antigen receptor (BCR) initiates humoral immunity. However, BCR signaling without appropriate co-stimulation commits B cells to death rather than to differentiation into immune effector cells. How BCR activation depletes potentially autoreactive B cells while simultaneously primes for receiving rescue and differentiation signals from cognate T lymphocytes remains unknown. Here, we use a mass spectrometry-based proteomic approach to identify cytosolic/nuclear shuttling elements and uncover transcription factor EB (TFEB) as a central BCR-controlled rheostat that drives activation-induced apoptosis, and concurrently promotes the reception of co-stimulatory rescue signals by supporting B cell migration and antigen presentation. CD40 co-stimulation prevents TFEB-driven cell death, while enhancing and prolonging TFEB's nuclear residency, which hallmarks antigenic experience also of memory B cells. In mice, TFEB shapes the transcriptional landscape of germinal center B cells. Within the germinal center, TFEB facilitates the dark zone entry of light-zone-residing centrocytes through regulation of chemokine receptors and, by balancing the expression of Bcl-2/BH3-only family members, integrates antigen-induced apoptosis with T cell-provided CD40 survival signals. Thus, TFEB reprograms antigen-primed germinal center B cells for cell fate decisions.
Collapse
Affiliation(s)
- Matthias Münchhalfen
- Institute of Cellular & Molecular Immunology, University Medical Center Göttingen, Göttingen, Germany
| | - Richard Görg
- Institute of Cellular & Molecular Immunology, University Medical Center Göttingen, Göttingen, Germany
| | - Michael Haberl
- Institute for Neuroimmunology and Multiple Sclerosis Research, University Medical Center Göttingen, Göttingen, Germany
| | - Jens Löber
- Department of Medical Hematology and Oncology, University Medical Center Göttingen, Göttingen, Germany
- Department of Hematology, Oncology, and Tumor Immunology, Charité, Campus Benjamin Franklin, University Medical Center Berlin, Berlin, Germany
| | - Jakob Willenbrink
- Institute of Cellular & Molecular Immunology, University Medical Center Göttingen, Göttingen, Germany
| | - Laura Schwarzt
- Institute of Cellular & Molecular Immunology, University Medical Center Göttingen, Göttingen, Germany
| | - Charlotte Höltermann
- Institute of Cellular & Molecular Immunology, University Medical Center Göttingen, Göttingen, Germany
| | - Christian Ickes
- Institute of Cardiovascular Physiology, University Medical Center Göttingen, Georg August University, Göttingen, Germany
| | - Leonard Hammermann
- Institute for Neuroimmunology and Multiple Sclerosis Research, University Medical Center Göttingen, Göttingen, Germany
| | - Jan Kus
- Institute of Cellular & Molecular Immunology, University Medical Center Göttingen, Göttingen, Germany
| | - Björn Chapuy
- Department of Medical Hematology and Oncology, University Medical Center Göttingen, Göttingen, Germany
- Department of Hematology, Oncology, and Tumor Immunology, Charité, Campus Benjamin Franklin, University Medical Center Berlin, Berlin, Germany
| | - Andrea Ballabio
- Telethon Institute of Genetics and Medicine (TIGEM), Naples, Italy
- Department of Translational Medical Sciences, Federico II University, Naples, Italy
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, USA
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, USA
| | - Sybille D Reichardt
- Institute of Cellular & Molecular Immunology, University Medical Center Göttingen, Göttingen, Germany
| | - Alexander Flügel
- Institute for Neuroimmunology and Multiple Sclerosis Research, University Medical Center Göttingen, Göttingen, Germany
| | - Niklas Engels
- Institute of Cellular & Molecular Immunology, University Medical Center Göttingen, Göttingen, Germany
| | - Jürgen Wienands
- Institute of Cellular & Molecular Immunology, University Medical Center Göttingen, Göttingen, Germany.
| |
Collapse
|
20
|
Brunialti E, Rizzi N, Pinto-Costa R, Villa A, Panzeri A, Meda C, Rebecchi M, Di Monte DA, Ciana P. Design and validation of a reporter mouse to study the dynamic regulation of TFEB and TFE3 activity through in vivo imaging techniques. Autophagy 2024; 20:1879-1894. [PMID: 38522425 PMCID: PMC11262230 DOI: 10.1080/15548627.2024.2334111] [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: 07/14/2023] [Accepted: 03/18/2024] [Indexed: 03/26/2024] Open
Abstract
TFEB and TFE3 belong to the MiT/TFE family of transcription factors that bind identical DNA responsive elements in the regulatory regions of target genes. They are involved in regulating lysosomal biogenesis, function, exocytosis, autophagy, and lipid catabolism. Precise control of TFEB and TFE3 activity is crucial for processes such as senescence, stress response, energy metabolism, and cellular catabolism. Dysregulation of these factors is implicated in various diseases, thus researchers have explored pharmacological approaches to modulate MiT/TFE activity, considering these transcription factors as potential therapeutic targets. However, the physiological complexity of their functions and the lack of suitable in vivo tools have limited the development of selective MiT/TFE modulating agents. Here, we have created a reporter-based biosensor, named CLEARoptimized, facilitating the pharmacological profiling of TFEB- and TFE3-mediated transcription. This innovative tool enables the measurement of TFEB and TFE3 activity in living cells and mice through imaging and biochemical techniques. CLEARoptimized consists of a promoter with six coordinated lysosomal expression and regulation motifs identified through an in-depth bioinformatic analysis of the promoters of 128 TFEB-target genes. The biosensor drives the expression of luciferase and tdTomato reporter genes, allowing the quantification of TFEB and TFE3 activity in cells and in animals through optical imaging and biochemical assays. The biosensor's validity was confirmed by modulating MiT/TFE activity in both cell culture and reporter mice using physiological and pharmacological stimuli. Overall, this study introduces an innovative tool for studying autophagy and lysosomal pathway modulation at various biological levels, from individual cells to the entire organism.Abbreviations: CLEAR: coordinated lysosomal expression and regulation; MAR: matrix attachment regions; MiT: microphthalmia-associated transcription factor; ROI: region of interest; TBS: tris-buffered saline; TF: transcription factor; TFE3: transcription factor binding to IGHM enhancer 3; TFEB: transcription factor EB; TH: tyrosine hydroxylase; TK: thymidine kinase; TSS: transcription start site.
Collapse
Affiliation(s)
| | | | - Rita Pinto-Costa
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Alessandro Villa
- Department of Health Sciences, University of Milan, Milan, Italy
| | - Alessia Panzeri
- Department of Health Sciences, University of Milan, Milan, Italy
| | - Clara Meda
- Department of Health Sciences, University of Milan, Milan, Italy
| | - Monica Rebecchi
- Department of Health Sciences, University of Milan, Milan, Italy
| | | | - Paolo Ciana
- Department of Health Sciences, University of Milan, Milan, Italy
| |
Collapse
|
21
|
Min Y, Li Q, Yu H, Du S. Examination of wnt signaling mediated melanin transport and shell color formation in Pacific oyster ( Crassostrea gigas). MARINE LIFE SCIENCE & TECHNOLOGY 2024; 6:488-501. [PMID: 39219677 PMCID: PMC11358575 DOI: 10.1007/s42995-024-00221-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Accepted: 03/07/2024] [Indexed: 09/04/2024]
Abstract
Mollusca exhibit remarkable diversity in shell coloration, attributed to the presence of melanin, a widely distributed pigment with various essential roles, such as mechanical strengthening, antioxidation and thermoregulation. However, the regulatory network governing melanogenesis and melanin transport in molluscs remains poorly understood. In this study, we conducted a systematic analysis of melanin distribution and transport in the Pacific oyster, utilizing light microscopy and high-resolution transmission electron microscopy. In addition, we characterized CgWnt1 and CgWnt2b-a in Crassostrea gigas, and analyzed Wnt signaling in melanocyte formation. Expression analysis revealed that these genes were predominantly expressed in the mantle of black-shelled individuals, particularly in the outer fold of the mantle. Furthermore, we employed RNA interference and inhibitors to specifically inhibit Wnt signaling in both in vivo and in vitro. The results revealed impaired melanogenesis and diminished tyrosinase activity upon Wnt signaling inhibition. These findings suggest the crucial role of Wnt ligands and downstream factors in melanogenesis. In summary, our study provides valuable insights into the regulatory mechanism of shell pigmentation in C. gigas. By demonstrating the promotion of melanogenesis through Wnt signaling modulation, we contribute to a better understanding of the complex processes underlying molluscan melanin production and shell coloration. Supplementary Information The online version contains supplementary material available at 10.1007/s42995-024-00221-5.
Collapse
Affiliation(s)
- Yue Min
- Key Laboratory of Mariculture, Ministry of Education, and College of Fisheries, Ocean University of China, Qingdao, 266003 China
| | - Qi Li
- Key Laboratory of Mariculture, Ministry of Education, and College of Fisheries, Ocean University of China, Qingdao, 266003 China
- Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266237 China
| | - Hong Yu
- Key Laboratory of Mariculture, Ministry of Education, and College of Fisheries, Ocean University of China, Qingdao, 266003 China
| | - Shaojun Du
- Institute of Marine and Environmental Technology, Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, 21240 USA
| |
Collapse
|
22
|
Lasota J, Thompson LDR, Chłopek M, Kowalik A, Miettinen M. Unusual PEComa With PRCC :: TFE3 Fusion Mimicking Sinonasal Tract Melanoma. Appl Immunohistochem Mol Morphol 2024; 32:322-325. [PMID: 38975712 DOI: 10.1097/pai.0000000000001211] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Accepted: 06/05/2024] [Indexed: 07/09/2024]
Abstract
BACKGROUND We report a nasal cavity unusual perivascular epithelioid cell tumor (PEComa) mimicking mucosal melanoma. METHODS Immunohistochemistry was performed using BenchMark Ultra and panel of antibodies. The Ion Torrent platform and Ion AmpliSeq cancer hotspot panel were utilized for DNA genotyping. Target-specific RNA libraries for the detection of fusion transcripts were constructed using Archer Universal RNA Reagent Kit v2 and Archer FusionPlex Solid Tumor panel and sequenced on the MiSeqDx instrument. RESULTS The tumor, diagnosed in 46-year-old female, was composed of spindle cells, and lacked pigmentation. Immunohistochemically, it showed a patchy HMB-45 positivity. Other melanocytic markers (S100 protein, Melan-A, SOX10) were negative. The tumor cells were weakly positive for KIT (CD117) while negative for smooth muscle actin, pancytokeratin cocktail (AE1/AE3), and synaptophysin. Diagnosis of primary sinonasal tract mucosal melanoma was favored. Additional molecular studies detected PRCC :: TFE3 fusion as the sole genetic change, and suggested the diagnosis of unusual PEComa. Previously, TFE3 fusions were reported in a subset of PEComas but not in melanomas, while PRCC involvement has only been documented once in an ocular PEComa. Immunohistochemistry revealed strong nuclear TFE3 expression concordant with the molecular findings. CONCLUSIONS This report emphasis the importance of molecular testing in the differential diagnosis between PEComa and melanoma, especially when the tumor arises in a site typical of melanoma but showing an unusual morphology and immunophenotype. The detection of TFE3 fusion transcripts suggested the diagnosis of SNT PEComa, although it cannot be excluded that this and similar tumors represent a distinct diagnostic category.
Collapse
Affiliation(s)
- Jerzy Lasota
- Laboratory of Pathology, National Cancer Institute, Bethesda, MD
| | | | - Małgorzata Chłopek
- Laboratory of Pathology, National Cancer Institute, Bethesda, MD
- Department of Molecular Diagnostics, Holycross Cancer Center
| | - Artur Kowalik
- Department of Molecular Diagnostics, Holycross Cancer Center
- Division of Medical Biology, Institute of Biology Jan Kochanowski University, Kielce, Poland
| | - Markku Miettinen
- Laboratory of Pathology, National Cancer Institute, Bethesda, MD
| |
Collapse
|
23
|
Chatzi D, Kyriakoudi SA, Dermitzakis I, Manthou ME, Meditskou S, Theotokis P. Clinical and Genetic Correlation in Neurocristopathies: Bridging a Precision Medicine Gap. J Clin Med 2024; 13:2223. [PMID: 38673496 PMCID: PMC11050951 DOI: 10.3390/jcm13082223] [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: 02/27/2024] [Revised: 04/09/2024] [Accepted: 04/10/2024] [Indexed: 04/28/2024] Open
Abstract
Neurocristopathies (NCPs) encompass a spectrum of disorders arising from issues during the formation and migration of neural crest cells (NCCs). NCCs undergo epithelial-mesenchymal transition (EMT) and upon key developmental gene deregulation, fetuses and neonates are prone to exhibit diverse manifestations depending on the affected area. These conditions are generally rare and often have a genetic basis, with many following Mendelian inheritance patterns, thus making them perfect candidates for precision medicine. Examples include cranial NCPs, like Goldenhar syndrome and Axenfeld-Rieger syndrome; cardiac-vagal NCPs, such as DiGeorge syndrome; truncal NCPs, like congenital central hypoventilation syndrome and Waardenburg syndrome; and enteric NCPs, such as Hirschsprung disease. Additionally, NCCs' migratory and differentiating nature makes their derivatives prone to tumors, with various cancer types categorized based on their NCC origin. Representative examples include schwannomas and pheochromocytomas. This review summarizes current knowledge of diseases arising from defects in NCCs' specification and highlights the potential of precision medicine to remedy a clinical phenotype by targeting the genotype, particularly important given that those affected are primarily infants and young children.
Collapse
Affiliation(s)
| | | | | | | | | | - Paschalis Theotokis
- Department of Histology-Embryology, School of Medicine, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece; (D.C.); (S.A.K.); (I.D.); (M.E.M.); (S.M.)
| |
Collapse
|
24
|
Ojalvo-Pacheco J, Yakhine-Diop SMS, Fuentes JM, Paredes-Barquero M, Niso-Santano M. Role of TFEB in Huntington's Disease. BIOLOGY 2024; 13:238. [PMID: 38666850 PMCID: PMC11048341 DOI: 10.3390/biology13040238] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Revised: 03/26/2024] [Accepted: 04/02/2024] [Indexed: 04/28/2024]
Abstract
Huntington's disease (HD) is an autosomal dominant neurodegenerative disease caused by an expansion of the CAG trinucleotide repeat in exon 1 of the huntingtin (HTT) gene. This expansion leads to a polyglutamine (polyQ) tract at the N-terminal end of HTT, which reduces the solubility of the protein and promotes its accumulation. Inefficient clearance of mutant HTT (mHTT) by the proteasome or autophagy-lysosomal system leads to accumulation of oligomers and toxic protein aggregates in neurons, resulting in impaired proteolytic systems, transcriptional dysregulation, impaired axonal transport, mitochondrial dysfunction and cellular energy imbalance. Growing evidence suggests that the accumulation of mHTT aggregates and autophagic and/or lysosomal dysfunction are the major pathogenic mechanisms underlying HD. In this context, enhancing autophagy may be an effective therapeutic strategy to remove protein aggregates and improve cell function. Transcription factor EB (TFEB), a master transcriptional regulator of autophagy, controls the expression of genes critical for autophagosome formation, lysosomal biogenesis, lysosomal function and autophagic flux. Consequently, the induction of TFEB activity to promote intracellular clearance may be a therapeutic strategy for HD. However, while some studies have shown that overexpression of TFEB facilitates the clearance of mHTT aggregates and ameliorates the disease phenotype, others indicate such overexpression may lead to mHTT co-aggregation and worsen disease progression. Further studies are necessary to confirm whether TFEB modulation could be an effective therapeutic strategy against mHTT-mediated toxicity in different disease models.
Collapse
Affiliation(s)
- Javier Ojalvo-Pacheco
- Departamento de Bioquímica y Biología Molecular y Genética, Facultad de Enfermería y Terapia Ocupacional, Universidad de Extremadura, 10003 Caceres, Spain; (J.O.-P.); (S.M.S.Y.-D.); (J.M.F.)
| | - Sokhna M. S. Yakhine-Diop
- Departamento de Bioquímica y Biología Molecular y Genética, Facultad de Enfermería y Terapia Ocupacional, Universidad de Extremadura, 10003 Caceres, Spain; (J.O.-P.); (S.M.S.Y.-D.); (J.M.F.)
- Centro de Investigación Biomédica en Red en Enfermedades Neurodegenerativa, Instituto de Salud Carlos III (CIBER-CIBERNED-ISCIII), 28029 Madrid, Spain
- Instituto Universitario de Investigación Biosanitaria de Extremadura (INUBE), 10003 Caceres, Spain
| | - José M. Fuentes
- Departamento de Bioquímica y Biología Molecular y Genética, Facultad de Enfermería y Terapia Ocupacional, Universidad de Extremadura, 10003 Caceres, Spain; (J.O.-P.); (S.M.S.Y.-D.); (J.M.F.)
- Centro de Investigación Biomédica en Red en Enfermedades Neurodegenerativa, Instituto de Salud Carlos III (CIBER-CIBERNED-ISCIII), 28029 Madrid, Spain
- Instituto Universitario de Investigación Biosanitaria de Extremadura (INUBE), 10003 Caceres, Spain
| | - Marta Paredes-Barquero
- Centro de Investigación Biomédica en Red en Enfermedades Neurodegenerativa, Instituto de Salud Carlos III (CIBER-CIBERNED-ISCIII), 28029 Madrid, Spain
- Instituto Universitario de Investigación Biosanitaria de Extremadura (INUBE), 10003 Caceres, Spain
| | - Mireia Niso-Santano
- Departamento de Bioquímica y Biología Molecular y Genética, Facultad de Enfermería y Terapia Ocupacional, Universidad de Extremadura, 10003 Caceres, Spain; (J.O.-P.); (S.M.S.Y.-D.); (J.M.F.)
- Centro de Investigación Biomédica en Red en Enfermedades Neurodegenerativa, Instituto de Salud Carlos III (CIBER-CIBERNED-ISCIII), 28029 Madrid, Spain
- Instituto Universitario de Investigación Biosanitaria de Extremadura (INUBE), 10003 Caceres, Spain
| |
Collapse
|
25
|
Ran R, Li L, Xu T, Huang J, He H, Chen Y. Revealing mitf functions and visualizing allografted tumor metastasis in colorless and immunodeficient Xenopus tropicalis. Commun Biol 2024; 7:275. [PMID: 38443437 PMCID: PMC10915148 DOI: 10.1038/s42003-024-05967-3] [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: 07/17/2023] [Accepted: 02/23/2024] [Indexed: 03/07/2024] Open
Abstract
Transparent immunodeficient animal models not only enhance in vivo imaging investigations of visceral organ development but also facilitate in vivo tracking of transplanted tumor cells. However, at present, transparent and immunodeficient animal models are confined to zebrafish, presenting substantial challenges for real-time, in vivo imaging studies addressing specific biological inquiries. Here, we employed a mitf-/-/prkdc-/-/il2rg-/- triple-knockout strategy to establish a colorless and immunodeficient amphibian model of Xenopus tropicalis. By disrupting the mitf gene, we observed the loss of melanophores, xanthophores, and granular glands in Xenopus tropicalis. Through the endogenous mitf promoter to drive BRAFV600E expression, we confirmed mitf expression in melanophores, xanthophores and granular glands. Moreover, the reconstruction of the disrupted site effectively reinstated melanophores, xanthophores, and granular glands, further highlighting the crucial role of mitf as a regulator in their development. By crossing mitf-/- frogs with prkdc-/-/il2rg-/- frogs, we generated a mitf-/-/prkdc-/-/il2rg-/- Xenopus tropicalis line, providing a colorless and immunodeficient amphibian model. Utilizing this model, we successfully observed intravital metastases of allotransplanted xanthophoromas and migrations of allotransplanted melanomas. Overall, colorless and immunodeficient Xenopus tropicalis holds great promise as a valuable platform for tumorous and developmental biology research.
Collapse
Affiliation(s)
- Rensen Ran
- Department of Chemical Biology, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Shenzhen Key Laboratory of Cell Microenvironment, School of Life Sciences, Southern University of Science and Technology, 518055, Shenzhen, China.
- Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital of Sun Yat-sen University, 519000, Zhuhai, China.
| | - Lanxin Li
- Department of Chemical Biology, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Shenzhen Key Laboratory of Cell Microenvironment, School of Life Sciences, Southern University of Science and Technology, 518055, Shenzhen, China
| | - Tingting Xu
- Fujian Medical University Union Hospital, 350001, Fuzhou, China
| | - Jixuan Huang
- Department of Chemical Biology, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Shenzhen Key Laboratory of Cell Microenvironment, School of Life Sciences, Southern University of Science and Technology, 518055, Shenzhen, China
| | - Huanhuan He
- Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital of Sun Yat-sen University, 519000, Zhuhai, China
| | - Yonglong Chen
- Department of Chemical Biology, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Shenzhen Key Laboratory of Cell Microenvironment, School of Life Sciences, Southern University of Science and Technology, 518055, Shenzhen, China.
| |
Collapse
|
26
|
Zhu Y, Li Q. Mitf involved in shell pigmentation by activating tyrosinase-mediated melanin synthesis in Pacific oyster (Crassostrea gigas). Gene 2024; 897:148086. [PMID: 38104952 DOI: 10.1016/j.gene.2023.148086] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Revised: 12/05/2023] [Accepted: 12/13/2023] [Indexed: 12/19/2023]
Abstract
Pigmentation is frequently observed in the molluscan shells, whereas the molecular regulation about these shell pigments formation is not clear. The microphthalmia-associated transcription factor (Mitf) is an important transactivator in melanin synthesis in vertebrates. Here, the Mitf containing a highly conserved basic helix-loop-helixleucine zipper (bHLH-LZ) domain was identified in an economically important marine bivalve Pacific oyster Crassostrea gigas. The Mitf was found to widespread tissue distribution and the expression was higher in the marginal mantle than in the central mantle. Particularly, the expression level of Mitf was high in black shell color oysters compared with white shell oysters. After injecting siRNA, the expression of Mitf decreased significantly, and the efficiency of RNA interference reached 53%. Besides, knockdown Mitf obviously decreased expression of tyrosinase family genes and tyrosinase activity of mantles, indicating a potential regulatory relationship between Mitf and Tyr or Typs. Simultaneously, there was a sharply reduce in the number of the melanosomes in the outer fold of mantle by silencing of Mitf. Luciferase assays in cell culture further verified that Mitf was involved in transcriptional regulation of Typ-2 and Typ-3 genes through binding to their specific promoter regions. These data argue that Mitf is involved in shell pigmentation through activating tyrosinase-mediated melanin synthesis in C. gigas.
Collapse
Affiliation(s)
- Yijing Zhu
- Key Laboratory of Mariculture, Ministry of Education, Ocean University of China, Qingdao 266003, China; Marine Science Research Institute of Shandong Province (National Oceanographic Center, Qingdao), Qingdao 266104, China
| | - Qi Li
- Key Laboratory of Mariculture, Ministry of Education, Ocean University of China, Qingdao 266003, China; Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, Qingdao 266237, China.
| |
Collapse
|
27
|
Tanaka Y, Sato-Matsubara M, Tsuruta D, Tanaka H, Kadono C, Sugawara K, Kawada N, Wakamatsu K, Ito S, Yoshizato K. Cytoglobin functions as a redox regulator of melanogenesis in normal epidermal melanocytes. Pigment Cell Melanoma Res 2024; 37:276-285. [PMID: 37920136 DOI: 10.1111/pcmr.13146] [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: 05/15/2023] [Revised: 09/21/2023] [Accepted: 10/15/2023] [Indexed: 11/04/2023]
Abstract
Epidermal melanocytes are continuously exposed to sunlight-induced reactive oxygen species (ROS) and oxidative stress generated during the synthesis of melanin. Therefore, they have developed mechanisms that maintain normal redox homeostasis. Cytoglobin (CYGB), a ubiquitously expressed intracellular iron hexacoordinated globin, exhibits antioxidant activity and regulates the redox state of mammalian cells through its activities as peroxidase and nitric oxide (NO) dioxygenase. We postulated that CYGB functions in the melanogenic process as a regulator that maintains oxidative stress within a physiological level. This was examined by characterizing normal human melanocytes with the knockdown (KD) of CYGB using morphological and molecular biological criteria. CYGB-KD cells were larger, had more dendrites, and generated more melanin granules in the advanced stages of melanogenesis than control cells. The expression levels of major melanogenesis-associated genes and proteins were higher in CYGB-KD melanocytes than in wild type (WT) cells. As expected, CYGB-KD melanocytes generated more ROS and NO than WT cells. In conclusion, CYGB physiologically contributes to maintaining redox homeostasis in the melanogenic activity of normal melanocytes by controlling the intracellular levels of ROS and NO.
Collapse
Affiliation(s)
- Yo Tanaka
- Research Laboratories, Nippon Menard Cosmetic Co., Ltd., Nagoya, Japan
| | - Misako Sato-Matsubara
- Department of Hepatology, Graduate School of Medicine, Osaka Metropolitan University, Osaka, Japan
- Donated Synthetic Biology Laboratory, Osaka Metropolitan University Graduate School of Medicine, Osaka, Japan
| | - Daisuke Tsuruta
- Department of Dermatology, Osaka Metropolitan University Graduate School of Medicine, Osaka, Japan
| | - Hiroshi Tanaka
- Research Laboratories, Nippon Menard Cosmetic Co., Ltd., Nagoya, Japan
| | - Chiho Kadono
- Donated Synthetic Biology Laboratory, Osaka Metropolitan University Graduate School of Medicine, Osaka, Japan
| | - Koji Sugawara
- Department of Dermatology, Osaka Metropolitan University Graduate School of Medicine, Osaka, Japan
| | - Norifumi Kawada
- Department of Hepatology, Graduate School of Medicine, Osaka Metropolitan University, Osaka, Japan
| | - Kazumasa Wakamatsu
- Institute for Melanin Chemistry, Fujita Health University, Toyoake, Japan
| | - Shosuke Ito
- Institute for Melanin Chemistry, Fujita Health University, Toyoake, Japan
| | - Katsutoshi Yoshizato
- Donated Synthetic Biology Laboratory, Osaka Metropolitan University Graduate School of Medicine, Osaka, Japan
| |
Collapse
|
28
|
Chen H, Gong S, Zhang H, Chen Y, Liu Y, Hao J, Liu H, Li X. From the regulatory mechanism of TFEB to its therapeutic implications. Cell Death Discov 2024; 10:84. [PMID: 38365838 PMCID: PMC10873368 DOI: 10.1038/s41420-024-01850-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Revised: 02/01/2024] [Accepted: 02/05/2024] [Indexed: 02/18/2024] Open
Abstract
Transcription factor EB (TFEB), known as a major transcriptional regulator of the autophagy-lysosomal pathway, regulates target gene expression by binding to coordinated lysosomal expression and regulation (CLEAR) elements. TFEB are regulated by multiple links, such as transcriptional regulation, post-transcriptional regulation, translational-level regulation, post-translational modification (PTM), and nuclear competitive regulation. Targeted regulation of TFEB has been victoriously used as a treatment strategy in several disease models such as ischemic injury, lysosomal storage disorders (LSDs), cancer, metabolic disorders, neurodegenerative diseases, and inflammation. In this review, we aimed to elucidate the regulatory mechanism of TFEB and its applications in several disease models by targeting the regulation of TFEB as a treatment strategy.
Collapse
Affiliation(s)
- Huixia Chen
- Institute of Nephrology, and Guangdong Provincial Key Laboratory of Autophagy and Major Chronic Non-Communicable Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, China
| | - Siqiao Gong
- Institute of Nephrology, and Guangdong Provincial Key Laboratory of Autophagy and Major Chronic Non-Communicable Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, China
| | - Hongyong Zhang
- Zhanjiang Institute of Clinical Medicine, Central People's Hospital of Zhanjiang, Guangdong Medical University Zhan-jiang Central Hospital, Zhanjiang, 524001, China
| | - Yongming Chen
- Institute of Nephrology, and Guangdong Provincial Key Laboratory of Autophagy and Major Chronic Non-Communicable Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, China
| | - Yonghan Liu
- Institute of Nephrology, and Guangdong Provincial Key Laboratory of Autophagy and Major Chronic Non-Communicable Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, China
| | - Junfeng Hao
- Institute of Nephrology, and Guangdong Provincial Key Laboratory of Autophagy and Major Chronic Non-Communicable Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, China.
| | - Huafeng Liu
- Institute of Nephrology, and Guangdong Provincial Key Laboratory of Autophagy and Major Chronic Non-Communicable Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, China.
| | - Xiaoyu Li
- Institute of Nephrology, and Guangdong Provincial Key Laboratory of Autophagy and Major Chronic Non-Communicable Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, China.
| |
Collapse
|
29
|
Sabaté San José A, Petersen PH. Absence of meningeal mast cells in the Mitf mutant mouse. Front Cell Neurosci 2024; 18:1337621. [PMID: 38405598 PMCID: PMC10884230 DOI: 10.3389/fncel.2024.1337621] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Accepted: 01/17/2024] [Indexed: 02/27/2024] Open
Abstract
Mast cells (MCs) are located in the meninges of the central nervous system (CNS), where they play key roles in the immune response. MC-deficient mice are advantageous in delineating the role of MCs in the immune response in vivo. In this study, we illustrate that a mutation in microphthalmia-associated transcription factor (Mitf) affects meningeal MC number in a dosage-dependent manner. C57BL/6J Mitf null mice lack meningeal MCs completely, whereas heterozygous mice have on average 25% fewer MCs. Mitf heterozygous mice might be a valuable tool to study the role of MCs in the meninges.
Collapse
Affiliation(s)
- Alba Sabaté San José
- Department of Anatomy, Biomedical Center, Faculty of Medicine, University of Iceland, Reykjavik, Iceland
- ULB Neuroscience Institute (UNI), Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Petur Henry Petersen
- Department of Anatomy, Biomedical Center, Faculty of Medicine, University of Iceland, Reykjavik, Iceland
| |
Collapse
|
30
|
Durbin HJ, Yampara-Iquise H, Rowan TN, Schnabel RD, Koltes JE, Powell JG, Decker JE. Genomic loci involved in sensing environmental cues and metabolism affect seasonal coat shedding in Bos taurus and Bos indicus cattle. G3 (BETHESDA, MD.) 2024; 14:jkad279. [PMID: 38092373 PMCID: PMC10849337 DOI: 10.1093/g3journal/jkad279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Accepted: 11/17/2023] [Indexed: 02/09/2024]
Abstract
Seasonal shedding of winter hair at the start of summer is well studied in wild and domesticated populations. However, the genetic influences on this trait and their interactions are poorly understood. We use data from 13,364 cattle with 36,899 repeated phenotypes to investigate the relationship between hair shedding and environmental variables, single nucleotide polymorphisms, and their interactions to understand quantitative differences in seasonal shedding. Using deregressed estimated breeding values from a repeated records model in a genome-wide association analysis (GWAA) and meta-analysis of year-specific GWAA gave remarkably similar results. These GWAA identified hundreds of variants associated with seasonal hair shedding. There were especially strong associations between chromosomes 5 and 23. Genotype-by-environment interaction GWAA identified 1,040 day length-by-genotype interaction associations and 17 apparent temperature-by-genotype interaction associations with hair shedding, highlighting the importance of day length on hair shedding. Accurate genomic predictions of hair shedding were created for the entire dataset, Angus, Hereford, Brangus, and multibreed datasets. Loci related to metabolism and light-sensing have a large influence on seasonal hair shedding. This is one of the largest genetic analyses of a phenological trait and provides insight into both agriculture production and basic science.
Collapse
Affiliation(s)
- Harly J Durbin
- Genetics Area Program, University of Missouri, Columbia, MO 65211, USA
- Syngenta, Research Triangle Park, NC 27709, USA
| | | | - Troy N Rowan
- Genetics Area Program, University of Missouri, Columbia, MO 65211, USA
- University of Tennessee Institute of Agriculture, Knoxville, TN 37996, USA
| | - Robert D Schnabel
- Genetics Area Program, University of Missouri, Columbia, MO 65211, USA
- Division of Animal Sciences, University of Missouri, Columbia, MO 65211, USA
- Institute for Data Science and Informatics, University of Missouri, Columbia, MO 65211, USA
| | - James E Koltes
- Department of Animal Science, Iowa State University, Ames, IA 50010, USA
- Department of Animal Science, University of Arkansas, Fayetteville, AR 72701, USA
| | - Jeremy G Powell
- Department of Animal Science, University of Arkansas, Fayetteville, AR 72701, USA
| | - Jared E Decker
- Genetics Area Program, University of Missouri, Columbia, MO 65211, USA
- Division of Animal Sciences, University of Missouri, Columbia, MO 65211, USA
- Institute for Data Science and Informatics, University of Missouri, Columbia, MO 65211, USA
| |
Collapse
|
31
|
Tzika AC. On the role of TFEC in reptilian coloration. Front Cell Dev Biol 2024; 12:1358828. [PMID: 38385026 PMCID: PMC10879265 DOI: 10.3389/fcell.2024.1358828] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Accepted: 01/22/2024] [Indexed: 02/23/2024] Open
Abstract
Reptilian species, particularly snakes and lizards, are emerging models of animal coloration. Here, I focus on the role of the TFEC transcription factor in snake and lizard coloration based on a study on wild-type and piebald ball pythons. Genomic mapping previously identified a TFEC mutation linked to the piebald ball python phenotype. The association of TFEC with skin coloration was further supported by gene-editing experiments in the brown anole lizard. However, novel histological analyses presented here reveal discrepancies between the ball python and the anole TFEC mutants phenotype, cautioning against broad generalizations. Indeed, both wild-type and piebald ball pythons completely lack iridophores, whereas the TFEC anole lizard mutants lose their iridophores compared to the wild-type anole. Based on these findings, I discuss the potential role of the MiT/TFE family in skin pigmentation across vertebrate lineages and advocate the need for developmental analyses and additional gene-editing experiments to explore the reptilian coloration diversity.
Collapse
Affiliation(s)
- Athanasia C. Tzika
- Laboratory of Artificial and Natural Evolution (LANE), Department of Genetics and Evolution, University of Geneva, Geneva, Switzerland
| |
Collapse
|
32
|
Zhao M, Huang Y, Yin X, Xu J, Sun Y, Wang J. PEComa with ASPSCR1::TFE3 fusion: expanding the molecular genetic spectrum of TFE3-rearranged PEComa with an emphasis on overlap with alveolar soft part sarcoma. Histopathology 2024; 84:482-491. [PMID: 37936565 DOI: 10.1111/his.15087] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Revised: 10/11/2023] [Accepted: 10/16/2023] [Indexed: 11/09/2023]
Abstract
AIMS Mesenchymal neoplasms involving TFE3 gene fusions are diverse, mainly include alveolar soft part sarcoma (ASPS) that is characterised by ASPSCR1::TFE3 fusion, and a small subset of perivascular epithelioid cell tumours (PEComas) referred to as TFE3-rearranged PEComa, that most frequently harbours SFPQ::TFE3 fusion. Historically, ASPS and TFE3-rearranged PEComa are considered two distinctive entities despite their known morphological overlap. However, recent studies have suggested a potential histogenetic relationship between them, and several neoplasms that showed morphological features more closely fit PEComa rather than ASPS but harboured ASPSCR1::TFE3 fusion have been documented. In this study, we report three cases of PEComa with ASPSCR1::TFE3 fusion. METHODS AND RESULTS Clinicopathological features were assessed and partner agnostic targeted next-generation sequencing on clinically validated platforms were performed. The patients are two females and one male with age at presentation ranging from 21 to 51 years. All three tumours were located in the viscera (rectum, kidney and cervix). On a relatively limited follow-up period (range = 9-15 months), all patients are alive without evidence of recurrent or metastatic disease. The neoplasms were composed of tight nested architecture of epithelioid clear cells separated by a delicate vascular network, two of which were associated with sheets of plump spindle cells, and none showed significant discohesive tumour morphology. Immunohistochemically, in addition to TFE3 protein, all three neoplasms demonstrated co-expression of melan-A and smooth muscle actin. RNA-sequencing identified ASPSCR1::TFE3 fusion in all three cases that were confirmed by subsequent fluorescence in-situ hybridisation analyses. CONCLUSIONS Our study expands the molecular genetic spectrum of TFE3-rearranged PEComa and further indicates its close relationship to ASPS.
Collapse
Affiliation(s)
- Ming Zhao
- Ningbo Clinical Pathology Diagnosis Center, Ningbo, China
| | - Yan Huang
- Department of Pathology, The First People's Hospital of Linping District, Hangzhou, China
| | - Xiaona Yin
- Ningbo Clinical Pathology Diagnosis Center, Ningbo, China
| | - Jiayun Xu
- Ningbo Clinical Pathology Diagnosis Center, Ningbo, China
| | - Yuefang Sun
- Department of Pathology, Shanghai Tenth People's Hospital, Tenth People's Hospital of Tongji University, Shanghai, China
| | - Jian Wang
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Institute of Pathology, Shanghai Medical College, Fudan University, Shanghai, China
| |
Collapse
|
33
|
Wen J, Song J, Chen J, Feng Z, Jing Q, Gong W, Kang X, Mei L, He C, Ma L, Feng Y. Modeling of pigmentation disorders associated with MITF mutation in Waardenburg syndrome revealed an impaired melanogenesis pathway in iPS-derived melanocytes. Pigment Cell Melanoma Res 2024; 37:21-35. [PMID: 37559350 DOI: 10.1111/pcmr.13118] [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: 09/20/2022] [Revised: 06/18/2023] [Accepted: 07/26/2023] [Indexed: 08/11/2023]
Abstract
Waardenburg Syndrome (WS) is a rare genetic disorder that leads to congenital hearing loss and pigmentation defects. Microphthalmia-associated transcription factor (MITF) is one of its significant pathogenic genes. Despite the comprehensive investigation in animal models, the pathogenic mechanism is still poorly described in humans due to difficulties accessing embryonic tissues. In this work, we used induced pluripotent stem cells derived from a WS patient carrying a heterozygous mutation in the MITF gene c.626A>T (p.His209Leu), and differentiated toward melanocyte lineage, which is the most affected cell type involved in WS. Compared with the wild-type cell line, the MITFmut cell line showed a reduced expression of the characteristic melanocyte-related genes and a lesser proportion of mature, fully pigmented melanosomes. The transcriptome analysis also revealed widespread gene expression changes at the melanocyte stage in the MITFmut cell line. The differentially expressed genes were enriched in melanogenesis and cell proliferation-related pathways. Interestingly, ion transport-related genes also showed a significant difference in MITFmut -induced melanocytes, indicating that the MITF mutant may lead to the dysfunction of potassium channels and transporters produced by intermediate cells in the cochlea, further causing the associated phenotype of deafness. Altogether, our study provides valuable insights into how MITF mutation affects WS patients, which might result in defective melanocyte development and the related phenotype based on the patient-derived iPSC model.
Collapse
Affiliation(s)
- Jie Wen
- Department of Otorhinolaryngology, The Affiliated Changsha Central Hospital, Hengyang Medical School, University of South China, Changsha, China
- Institute of Otorhinolaryngology, Head and Neck Surgery, University of South China, Changsha, China
| | - Jian Song
- Department of Otorhinolaryngology, Xiangya Hospital Central South University, Changsha, China
- Province Key Laboratory of Otolaryngology Critical Diseases, Changsha, China
| | - Jiale Chen
- Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha, China
| | - Zhili Feng
- Department of Otorhinolaryngology, The Affiliated Changsha Central Hospital, Hengyang Medical School, University of South China, Changsha, China
- Institute of Otorhinolaryngology, Head and Neck Surgery, University of South China, Changsha, China
| | - Qiancheng Jing
- Department of Otorhinolaryngology, The Affiliated Changsha Central Hospital, Hengyang Medical School, University of South China, Changsha, China
- Institute of Otorhinolaryngology, Head and Neck Surgery, University of South China, Changsha, China
| | - Wei Gong
- Department of Otorhinolaryngology, The Affiliated Changsha Central Hospital, Hengyang Medical School, University of South China, Changsha, China
- Institute of Otorhinolaryngology, Head and Neck Surgery, University of South China, Changsha, China
| | - Xiaoming Kang
- Department of Otorhinolaryngology, The Affiliated Changsha Central Hospital, Hengyang Medical School, University of South China, Changsha, China
- Institute of Otorhinolaryngology, Head and Neck Surgery, University of South China, Changsha, China
| | - Lingyun Mei
- Department of Otorhinolaryngology, Xiangya Hospital Central South University, Changsha, China
- Province Key Laboratory of Otolaryngology Critical Diseases, Changsha, China
| | - Chufeng He
- Department of Otorhinolaryngology, Xiangya Hospital Central South University, Changsha, China
- Province Key Laboratory of Otolaryngology Critical Diseases, Changsha, China
| | - Lu Ma
- Institute of Otorhinolaryngology, Head and Neck Surgery, University of South China, Changsha, China
- The Hengyang Key Laboratory of Cellular Stress Biology, Institute of Cytology and Genetics, Hengyang Medical School, University of South China, Hengyang, China
| | - Yong Feng
- Department of Otorhinolaryngology, The Affiliated Changsha Central Hospital, Hengyang Medical School, University of South China, Changsha, China
- Institute of Otorhinolaryngology, Head and Neck Surgery, University of South China, Changsha, China
- Province Key Laboratory of Otolaryngology Critical Diseases, Changsha, China
- Hengyang Medical School, University of South China, Hengyang, China
| |
Collapse
|
34
|
Shao J, Lang Y, Ding M, Yin X, Cui L. Transcription Factor EB: A Promising Therapeutic Target for Ischemic Stroke. Curr Neuropharmacol 2024; 22:170-190. [PMID: 37491856 PMCID: PMC10788889 DOI: 10.2174/1570159x21666230724095558] [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: 05/08/2022] [Revised: 12/14/2022] [Accepted: 12/15/2022] [Indexed: 07/27/2023] Open
Abstract
Transcription factor EB (TFEB) is an important endogenous defensive protein that responds to ischemic stimuli. Acute ischemic stroke is a growing concern due to its high morbidity and mortality. Most survivors suffer from disabilities such as numbness or weakness in an arm or leg, facial droop, difficulty speaking or understanding speech, confusion, impaired balance or coordination, or loss of vision. Although TFEB plays a neuroprotective role, its potential effect on ischemic stroke remains unclear. This article describes the basic structure, regulation of transcriptional activity, and biological roles of TFEB relevant to ischemic stroke. Additionally, we explore the effects of TFEB on the various pathological processes underlying ischemic stroke and current therapeutic approaches. The information compiled here may inform clinical and basic studies on TFEB, which may be an effective therapeutic drug target for ischemic stroke.
Collapse
Affiliation(s)
- Jie Shao
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Jilin University, Changchun, China
| | - Yue Lang
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Jilin University, Changchun, China
| | - Manqiu Ding
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Jilin University, Changchun, China
| | - Xiang Yin
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Jilin University, Changchun, China
| | - Li Cui
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Jilin University, Changchun, China
| |
Collapse
|
35
|
Santasusagna S, Zhu S, Jawalagatti V, Carceles-Cordon M, Ertel A, Garcia-Longarte S, Song WM, Fujiwara N, Li P, Mendizabal I, Petrylak DP, Kelly WK, Reddy EP, Wang L, Schiewer MJ, Lujambio A, Karnes J, Knudsen KE, Cordon-Cardo C, Dong H, Huang H, Carracedo A, Hoshida Y, Rodriguez-Bravo V, Domingo-Domenech J. Master Transcription Factor Reprogramming Unleashes Selective Translation Promoting Castration Resistance and Immune Evasion in Lethal Prostate Cancer. Cancer Discov 2023; 13:2584-2609. [PMID: 37676710 PMCID: PMC10714140 DOI: 10.1158/2159-8290.cd-23-0306] [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: 03/15/2023] [Revised: 07/28/2023] [Accepted: 09/05/2023] [Indexed: 09/08/2023]
Abstract
Signaling rewiring allows tumors to survive therapy. Here we show that the decrease of the master regulator microphthalmia transcription factor (MITF) in lethal prostate cancer unleashes eukaryotic initiation factor 3B (eIF3B)-dependent translation reprogramming of key mRNAs conferring resistance to androgen deprivation therapy (ADT) and promoting immune evasion. Mechanistically, MITF represses through direct promoter binding eIF3B, which in turn regulates the translation of specific mRNAs. Genome-wide eIF3B enhanced cross-linking immunoprecipitation sequencing (eCLIP-seq) showed specialized binding to a UC-rich motif present in subsets of 5' untranslated regions. Indeed, translation of the androgen receptor and major histocompatibility complex I (MHC-I) through this motif is sensitive to eIF3B amount. Notably, pharmacologic targeting of eIF3B-dependent translation in preclinical models sensitizes prostate cancer to ADT and anti-PD-1 therapy. These findings uncover a hidden connection between transcriptional and translational rewiring promoting therapy-refractory lethal prostate cancer and provide a druggable mechanism that may transcend into effective combined therapeutic strategies. SIGNIFICANCE Our study shows that specialized eIF3B-dependent translation of specific mRNAs released upon downregulation of the master transcription factor MITF confers castration resistance and immune evasion in lethal prostate cancer. Pharmacologic targeting of this mechanism delays castration resistance and increases immune-checkpoint efficacy. This article is featured in Selected Articles from This Issue, p. 2489.
Collapse
Affiliation(s)
- Sandra Santasusagna
- Department of Urology, Mayo Comprehensive Cancer Center, Rochester, Minnesota
- Department of Biochemistry and Molecular Biology, Mayo Comprehensive Cancer Center, Rochester, Minnesota
| | - Shijia Zhu
- Department of Medicine, Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, Texas
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, Minnesota
| | - Vijayakumar Jawalagatti
- Department of Urology, Mayo Comprehensive Cancer Center, Rochester, Minnesota
- Department of Biochemistry and Molecular Biology, Mayo Comprehensive Cancer Center, Rochester, Minnesota
| | | | - Adam Ertel
- Department of Oncology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Saioa Garcia-Longarte
- Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Derio, Spain
| | - Won-Min Song
- Department of Genetics and Genome Sciences, Tisch Cancer Center, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Naoto Fujiwara
- Department of Medicine, Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Peiyao Li
- Department of Oncology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Isabel Mendizabal
- Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Derio, Spain
| | - Daniel P. Petrylak
- Department of Oncology, Yale Comprehensive Cancer Center, Yale School of Medicine, New Haven, Connecticut
| | - William Kevin Kelly
- Department of Oncology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - E. Premkumar Reddy
- Department of Oncological Sciences, Tisch Cancer Center, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Liguo Wang
- Department of Biochemistry and Molecular Biology, Mayo Comprehensive Cancer Center, Rochester, Minnesota
| | - Matthew J. Schiewer
- Department of Pharmacology, Physiology, and Cancer Biology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Amaia Lujambio
- Department of Oncological Sciences, Tisch Cancer Center, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Jeffrey Karnes
- Department of Urology, Mayo Comprehensive Cancer Center, Rochester, Minnesota
| | - Karen E. Knudsen
- Department of Pharmacology, Physiology, and Cancer Biology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Carlos Cordon-Cardo
- Department of Pathology. Tisch Cancer Center, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Haidong Dong
- Department of Urology, Mayo Comprehensive Cancer Center, Rochester, Minnesota
- Department of Immunology, Mayo Comprehensive Cancer Center, Rochester, Minnesota
| | - Haojie Huang
- Department of Urology, Mayo Comprehensive Cancer Center, Rochester, Minnesota
- Department of Biochemistry and Molecular Biology, Mayo Comprehensive Cancer Center, Rochester, Minnesota
| | - Arkaitz Carracedo
- Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Derio, Spain
- Ikerbasque, Basque Foundation for Science, Bilbao, Spain
- Traslational prostate cancer Research Lab, CIC bioGUNE-Basurto, Biocruces Bizkaia Health Research Institute CIC bioGUNE, Bizkaia Technology Park, Derio, Spain
- CIBERONC, Madrid, Spain
- Biochemistry and Molecular Biology Department, University of the Basque Country (UPV/EHU), Bilbao, Spain
| | - Yujin Hoshida
- Department of Medicine, Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Veronica Rodriguez-Bravo
- Department of Urology, Mayo Comprehensive Cancer Center, Rochester, Minnesota
- Department of Biochemistry and Molecular Biology, Mayo Comprehensive Cancer Center, Rochester, Minnesota
| | - Josep Domingo-Domenech
- Department of Urology, Mayo Comprehensive Cancer Center, Rochester, Minnesota
- Department of Biochemistry and Molecular Biology, Mayo Comprehensive Cancer Center, Rochester, Minnesota
| |
Collapse
|
36
|
Johnson MR, Li S, Guerrero-Juarez CF, Miller P, Brack BJ, Mereby SA, Moreno JA, Feigin CY, Gaska J, Rivera-Perez JA, Nie Q, Ploss A, Shvartsman SY, Mallarino R. A multifunctional Wnt regulator underlies the evolution of rodent stripe patterns. Nat Ecol Evol 2023; 7:2143-2159. [PMID: 37813945 PMCID: PMC10839778 DOI: 10.1038/s41559-023-02213-7] [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: 03/29/2023] [Accepted: 08/27/2023] [Indexed: 10/11/2023]
Abstract
Animal pigment patterns are excellent models to elucidate mechanisms of biological organization. Although theoretical simulations, such as Turing reaction-diffusion systems, recapitulate many animal patterns, they are insufficient to account for those showing a high degree of spatial organization and reproducibility. Here, we study the coat of the African striped mouse (Rhabdomys pumilio) to uncover how periodic stripes form. Combining transcriptomics, mathematical modelling and mouse transgenics, we show that the Wnt modulator Sfrp2 regulates the distribution of hair follicles and establishes an embryonic prepattern that foreshadows pigment stripes. Moreover, by developing in vivo gene editing in striped mice, we find that Sfrp2 knockout is sufficient to alter the stripe pattern. Strikingly, mutants exhibited changes in pigmentation, revealing that Sfrp2 also regulates hair colour. Lastly, through evolutionary analyses, we find that striped mice have evolved lineage-specific changes in regulatory elements surrounding Sfrp2, many of which may be implicated in modulating the expression of this gene. Altogether, our results show that a single factor controls coat pattern formation by acting both as an orienting signalling mechanism and a modulator of pigmentation. More broadly, our work provides insights into how spatial patterns are established in developing embryos and the mechanisms by which phenotypic novelty originates.
Collapse
Affiliation(s)
- Matthew R Johnson
- Department of Molecular Biology, Princeton University, Princeton, NJ, USA
| | - Sha Li
- Department of Molecular Biology, Princeton University, Princeton, NJ, USA
| | - Christian F Guerrero-Juarez
- Carle Illinois College of Medicine, University of Illinois at Urbana-Champaign, Urbana, IL, USA
- Department of Developmental and Cell Biology, University of California, Irvine, CA, USA
- Department of Mathematics, University of California, Irvine, CA, USA
- Center for Complex Biological Systems, University of California, Irvine, CA, USA
- NSF-Simons Center for Multiscale Cell Fate Research, University of California, Irvine, CA, USA
| | - Pearson Miller
- Center for Computational Biology, Flatiron Institute, New York, NY, USA
| | - Benjamin J Brack
- Department of Molecular Biology, Princeton University, Princeton, NJ, USA
| | - Sarah A Mereby
- Department of Molecular Biology, Princeton University, Princeton, NJ, USA
| | - Jorge A Moreno
- Department of Molecular Biology, Princeton University, Princeton, NJ, USA
| | - Charles Y Feigin
- Department of Molecular Biology, Princeton University, Princeton, NJ, USA
| | - Jenna Gaska
- Department of Molecular Biology, Princeton University, Princeton, NJ, USA
| | | | - Qing Nie
- Department of Developmental and Cell Biology, University of California, Irvine, CA, USA
- Department of Mathematics, University of California, Irvine, CA, USA
- Center for Complex Biological Systems, University of California, Irvine, CA, USA
- NSF-Simons Center for Multiscale Cell Fate Research, University of California, Irvine, CA, USA
| | - Alexander Ploss
- Department of Molecular Biology, Princeton University, Princeton, NJ, USA
| | - Stanislav Y Shvartsman
- Department of Molecular Biology, Princeton University, Princeton, NJ, USA
- Center for Computational Biology, Flatiron Institute, New York, NY, USA
- The Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ, USA
| | - Ricardo Mallarino
- Department of Molecular Biology, Princeton University, Princeton, NJ, USA.
| |
Collapse
|
37
|
Zhang Y, Chen Q, Fu X, Zhu S, Huang Q, Li C. Current Advances in the Regulatory Effects of Bioactive Compounds from Dietary Resources on Nonalcoholic Fatty Liver Disease: Role of Autophagy. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2023; 71:17554-17569. [PMID: 37955247 DOI: 10.1021/acs.jafc.3c04692] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/14/2023]
Abstract
Nonalcoholic fatty liver disease (NAFLD) is the most prevalent chronic liver disease characterized by lipid metabolic disorder primarily due to sedentary lifestyles and excessive food consumption. However, there are currently no approved and effective drugs available to treat NAFLD. In recent years, research has shown that dietary bioactive compounds, such as polysaccharides, polyphenols, flavones, and alkaloids, have the potential to improve NAFLD by regulating autophagy. However, there is no up-to-date review of research progress in this field. This review aims to systematically summarize and discuss the regulatory effects and molecular mechanisms of dietary bioactive compounds on NAFLD through the modulation of autophagy. The existing research has demonstrated that some dietary bioactive compounds can effectively improve various aspects of NAFLD progression, such as lipid metabolism, insulin resistance (IR), endoplasmic reticulum (ER) stress, oxidative stress, mitochondrial homeostasis, and inflammation. Molecular mechanism studies have revealed that they exert their beneficial effects on NAFLD through autophagy-mediated signaling pathways, predominantly involving transcription factor EB (TFEB), mammalian target of rapamycin (mTOR), adenosine monophosphate-activated protein kinase (AMPK), peroxisome proliferator-activated receptors (PPARs), SIRT, and PTEN-induced kinase 1 (PINK1)/parkin. Furthermore, the challenges and prospects of current research in this field are highlighted. Overall, this review provides valuable insights into the potential treatment of NAFLD using dietary bioactive compounds that can modulate autophagy.
Collapse
Affiliation(s)
- Yue Zhang
- School of Food Science and Engineering, Guangdong Province Key Laboratory for Green Processing of Natural Products and Product Safety, South China University of Technology, Guangzhou 510640, China
| | - Qing Chen
- School of Food Science and Engineering, Guangdong Province Key Laboratory for Green Processing of Natural Products and Product Safety, South China University of Technology, Guangzhou 510640, China
- School of Food Science and Dietetics, Guangzhou City Polytechnic, Guangzhou 510405, China
| | - Xiong Fu
- School of Food Science and Engineering, Guangdong Province Key Laboratory for Green Processing of Natural Products and Product Safety, South China University of Technology, Guangzhou 510640, China
| | - Siming Zhu
- School of Food Science and Engineering, Guangdong Province Key Laboratory for Green Processing of Natural Products and Product Safety, South China University of Technology, Guangzhou 510640, China
| | - Qiang Huang
- School of Food Science and Engineering, Guangdong Province Key Laboratory for Green Processing of Natural Products and Product Safety, South China University of Technology, Guangzhou 510640, China
| | - Chao Li
- School of Food Science and Engineering, Guangdong Province Key Laboratory for Green Processing of Natural Products and Product Safety, South China University of Technology, Guangzhou 510640, China
| |
Collapse
|
38
|
Takla M, Keshri S, Rubinsztein DC. The post-translational regulation of transcription factor EB (TFEB) in health and disease. EMBO Rep 2023; 24:e57574. [PMID: 37728021 PMCID: PMC10626434 DOI: 10.15252/embr.202357574] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Revised: 08/10/2023] [Accepted: 08/25/2023] [Indexed: 09/21/2023] Open
Abstract
Transcription factor EB (TFEB) is a basic helix-loop-helix leucine zipper transcription factor that acts as a master regulator of lysosomal biogenesis, lysosomal exocytosis, and macro-autophagy. TFEB contributes to a wide range of physiological functions, including mitochondrial biogenesis and innate and adaptive immunity. As such, TFEB is an essential component of cellular adaptation to stressors, ranging from nutrient deprivation to pathogenic invasion. The activity of TFEB depends on its subcellular localisation, turnover, and DNA-binding capacity, all of which are regulated at the post-translational level. Pathological states are characterised by a specific set of stressors, which elicit post-translational modifications that promote gain or loss of TFEB function in the affected tissue. In turn, the resulting increase or decrease in survival of the tissue in which TFEB is more or less active, respectively, may either benefit or harm the organism as a whole. In this way, the post-translational modifications of TFEB account for its otherwise paradoxical protective and deleterious effects on organismal fitness in diseases ranging from neurodegeneration to cancer. In this review, we describe how the intracellular environment characteristic of different diseases alters the post-translational modification profile of TFEB, enabling cellular adaptation to a particular pathological state.
Collapse
Affiliation(s)
- Michael Takla
- Department of Medical Genetics, Cambridge Institute for Medical Research (CIMR)University of CambridgeCambridgeUK
- UK Dementia Research Institute, Cambridge Institute for Medical Research (CIMR)University of CambridgeCambridgeUK
| | - Swati Keshri
- Department of Medical Genetics, Cambridge Institute for Medical Research (CIMR)University of CambridgeCambridgeUK
- UK Dementia Research Institute, Cambridge Institute for Medical Research (CIMR)University of CambridgeCambridgeUK
| | - David C Rubinsztein
- Department of Medical Genetics, Cambridge Institute for Medical Research (CIMR)University of CambridgeCambridgeUK
- UK Dementia Research Institute, Cambridge Institute for Medical Research (CIMR)University of CambridgeCambridgeUK
| |
Collapse
|
39
|
Medda A, Compagnoni M, Spini G, Citro S, Croci O, Campaner S, Tagliabue M, Ansarin M, Chiocca S. c-MYC-dependent transcriptional inhibition of autophagy is implicated in cisplatin sensitivity in HPV-positive head and neck cancer. Cell Death Dis 2023; 14:719. [PMID: 37925449 PMCID: PMC10625625 DOI: 10.1038/s41419-023-06248-3] [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: 01/25/2023] [Revised: 10/13/2023] [Accepted: 10/25/2023] [Indexed: 11/06/2023]
Abstract
Autophagy is important for the removal, degradation and recycling of damaged organelles, proteins, and lipids through the degradative action of lysosomes. In addition to its catabolic function, autophagy is important in cancer and viral-mediated tumorigenesis, including Human Papillomavirus (HPV) positive cancers. HPV infection is a major risk factor in a subset of head and neck cancer (HNC), for which no targeted therapies are currently available. Herein, we assessed autophagy function in HPV-positive HNC. We showed that HPV-positive HNC cells presented a transcriptional and functional impairment of the autophagic process compared to HPV-negative cells, which were reactivated by knocking down HPV E6/E7 oncoproteins, the drivers of cellular transformation. We found that the oncoprotein c-MYC was stabilized and triggered in HPV-positive cell lines. This resulted in the reduced binding of the MiT/TFE transcription factors to their autophagy targets due to c-MYC competition. Thus, the knock-down of c-MYC induced the upregulation of autophagic and lysosomal genes in HPV-positive HNC cells, as well as the increase of autophagic markers at the protein level. Moreover, HPV oncoprotein E7 upregulated the expression of the phosphatase inhibitor CIP2A, accounting for c-MYC upregulation and stability in HPV+ HNC cells. CIP2A mRNA expression negatively correlated with autophagy gene expression in tumor tissues from HNC patients, showing, for the first time, its implication in a transcriptional autophagic context. Both CIP2A and c-MYC knock-down, as well as pharmacological downregulation of c-MYC, resulted in increased resistance to cisplatin treatment. Our results not only show a novel way by which HPV oncoproteins manipulate the host machinery but also provide more insights into the role of autophagy in chemoresistance, with possible implications for targeted HPV-positive HNC therapy.
Collapse
Affiliation(s)
- Alessandro Medda
- Department of Experimental Oncology, IEO, European Institute of Oncology IRCCS, IEO Campus, Via Adamello 16, 20139, Milan, Italy
| | - Micaela Compagnoni
- Department of Experimental Oncology, IEO, European Institute of Oncology IRCCS, IEO Campus, Via Adamello 16, 20139, Milan, Italy
| | - Giorgio Spini
- Department of Experimental Oncology, IEO, European Institute of Oncology IRCCS, IEO Campus, Via Adamello 16, 20139, Milan, Italy
| | - Simona Citro
- Department of Experimental Oncology, IEO, European Institute of Oncology IRCCS, IEO Campus, Via Adamello 16, 20139, Milan, Italy
| | - Ottavio Croci
- Center for Genomic Science of IIT, CGS@SEMM (Istituto Italiano di Tecnologia at European School of Molecular Medicine), Fondazione Istituto Italiano di Tecnologia (IIT), Milan, Italy
| | - Stefano Campaner
- Center for Genomic Science of IIT, CGS@SEMM (Istituto Italiano di Tecnologia at European School of Molecular Medicine), Fondazione Istituto Italiano di Tecnologia (IIT), Milan, Italy
| | - Marta Tagliabue
- Division of Otolaryngology Head & Neck Surgery, IEO, European Institute of Oncology IRCCS, 20141, Milan, Italy
- Department of Biomedical Sciences, University of Sassari, Sassari, Italy
| | - Mohssen Ansarin
- Division of Otolaryngology Head & Neck Surgery, IEO, European Institute of Oncology IRCCS, 20141, Milan, Italy
| | - Susanna Chiocca
- Department of Experimental Oncology, IEO, European Institute of Oncology IRCCS, IEO Campus, Via Adamello 16, 20139, Milan, Italy.
| |
Collapse
|
40
|
Cao M, Xu T, Song Y, Wang H, Wei S, Yin D. 2,2',4,4'-tetrabromodiphenyl ether causes depigmentation in zebrafish larvae via a light-mediated pathway. THE SCIENCE OF THE TOTAL ENVIRONMENT 2023; 897:165382. [PMID: 37422226 DOI: 10.1016/j.scitotenv.2023.165382] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Revised: 07/04/2023] [Accepted: 07/05/2023] [Indexed: 07/10/2023]
Abstract
Polybrominated diphenyl ethers (PBDEs) are organic pollutants widely detected in various environmental media due to their high persistence and bioaccumulation. PBDE-induced visual impairment and neurotoxicity were previously demonstrated using zebrafish (Danio rerio) models, and recent research reported the phenotypic depigmentation effect of PBDEs at high concentrations on zebrafish, but whether those effects are still present at environment-relevant levels is still unclear. Herein, we performed both phenotypic examination and mechanism investigation in zebrafish embryos (48 hpf) and larvae (5 dpf) about their pigmentation status when exposing to PBDE congener BDE-47 (2,2',4,4'-tetrabrominated diphenyl ether) at levels from 0.25 to 25 μg/L. Results showed that low-level BDE-47 can restrain the relative melanin abundance of zebrafish larvae to 70.47% (p < 0.05) and 61.54% (p < 0.01) respectively under 2.5 and 25 μg/L BDE-47 compared with control, and the thickness of retinal pigment epithelium (RPE) remarkably reduced from 571.4 nm to 350.3 nm (p < 0.001) under 25 μg/L BDE-47 exposure. We also observed disrupted expressions of melanin synthesis genes and disorganized mitfa differentiation patterns based on Tg(mifta:EGFP), as well as visual impairment resulting from thinner RPE. Considering both processes of visual development and melanin synthesis are highly sensitive to ambient light conditions, we prolonged the light regime of maintaining zebrafish larvae from 14 hours light versus 10 hours dark (14L:10D) to 18 hours light versus 6 hours dark (18L:6D). Lengthening photoperiod successfully rescued the fluorescent level of mitfa in zebrafish epidermis and most gene expressions associated with melanin synthesis under 25 μg/L BDE-47 exposure to the normal level. In conclusion, our work reported the effects of low-level PBDEs on melanin production using zebrafish embryos and larvae, and identified the potential role of a light-mediated pathway in the neurotoxic mechanism of PBDEs.
Collapse
Affiliation(s)
- Miao Cao
- Key Laboratory of Yangtze River Water Environment, Ministry of Education, College of Environmental Science and Engineering, Tongji University, Shanghai 200092, China
| | - Ting Xu
- Key Laboratory of Yangtze River Water Environment, Ministry of Education, College of Environmental Science and Engineering, Tongji University, Shanghai 200092, China; Shanghai Institute of Pollution Control and Ecological Security, Shanghai 200092, China
| | - Yiqun Song
- Key Laboratory of Yangtze River Water Environment, Ministry of Education, College of Environmental Science and Engineering, Tongji University, Shanghai 200092, China
| | - Huan Wang
- Key Laboratory of Yangtze River Water Environment, Ministry of Education, College of Environmental Science and Engineering, Tongji University, Shanghai 200092, China
| | - Sheng Wei
- Key Laboratory of Yangtze River Water Environment, Ministry of Education, College of Environmental Science and Engineering, Tongji University, Shanghai 200092, China
| | - Daqiang Yin
- Key Laboratory of Yangtze River Water Environment, Ministry of Education, College of Environmental Science and Engineering, Tongji University, Shanghai 200092, China; Shanghai Institute of Pollution Control and Ecological Security, Shanghai 200092, China.
| |
Collapse
|
41
|
Bouchard KV, Costin GE. Promoting New Approach Methodologies (NAMs) for research on skin color changes in response to environmental stress factors: tobacco and air pollution. FRONTIERS IN TOXICOLOGY 2023; 5:1256399. [PMID: 37886123 PMCID: PMC10598764 DOI: 10.3389/ftox.2023.1256399] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Accepted: 08/25/2023] [Indexed: 10/28/2023] Open
Abstract
Aging is one of the most dynamic biological processes in the human body and is known to carry significant impacts on individuals' self-esteem. Skin pigmentation is a highly heritable trait made possible by complex, strictly controlled cellular and molecular mechanisms. Genetic, environmental and endocrine factors contribute to the modulation of melanin's amount, type and distribution in the skin layers. One of the hallmarks of extrinsic skin aging induced by environmental stress factors is the alteration of the constitutive pigmentation pattern clinically defined as senile lentigines and/or melasma or other pigmentary dyschromias. The complexity of pollutants and tobacco smoke as environmental stress factors warrants a thorough understanding of the mechanisms by which they impact skin pigmentation through repeated and long-term exposure. Pre-clinical and clinical studies demonstrated that pollutants are known to induce reactive oxygen species (ROS) or inflammatory events that lead directly or indirectly to skin hyperpigmentation. Another mechanistic direction is provided by Aryl hydrocarbon Receptors (AhR) which were shown to mediate processes leading to skin hyperpigmentation in response to pollutants by regulation of melanogenic enzymes and transcription factors involved in melanin biosynthesis pathway. In this context, we will discuss a diverse range of New Approach Methodologies (NAMs) capable to provide mechanistic insights of the cellular and molecular pathways involved in the action of environmental stress factors on skin pigmentation and to support the design of raw ingredients and formulations intended to counter their impact and of any subsequently needed clinical studies.
Collapse
|
42
|
Li C, Yang Y, Li L, Chen Y, Shi Q, Zhang H, Zhang L, Chen Y, Li R, Li Z, Liu S, Ye Z, Zhao X, Liang X. Role of TFEB in regulation of the podocyte actin cytoskeleton. Arch Biochem Biophys 2023; 747:109752. [PMID: 37714254 DOI: 10.1016/j.abb.2023.109752] [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: 04/28/2023] [Revised: 08/16/2023] [Accepted: 09/11/2023] [Indexed: 09/17/2023]
Abstract
Podocyte injury is linked to the pathogenesis and progression of renal disease. The Transcription Factor EB (TFEB), a master regulator of the autophagy and lysosomal pathways, has been found to exert cell- and tissue-specific biological function. To explore TFEB function and underlying mechanisms in podocytes, a total of 4645 differentially expressed genes (DEGs) were detected in TFEB-knockdown mouse podocytes by transcriptome sequencing. Gene Ontology, Kyoto Encyclopedia of Genes and Genomes, and Ingenuity Pathway Analysis showed that, apart from the enrichment in autophagy and lysosomal pathways, DEGs were enriched in cytoskeleton structure (Actin Cytoskeleton, Focal Adhesion, and Adherens Junction), as well as cytoskeleton regulatory molecular signaling (Hippo and Rho GTPase Signaling). In vitro, TFEB knockdown resulted in podocyte cytoskeletal rearrangement, which was disorganized with cortical distribution of actin filaments. Further, TFEB knockdown decreased mRNA and protein levels of Synaptopodin and led to the rearrangement of Synaptopodin. Inhibition of TFEB decreased mRNA levels for proteins involved in actin cytoskeleton dynamics. Moreover, apoptosis was increased by TFEB knockdown in podocyte. In summary, this study initiated a comprehensive analysis of the role of TFEB in podocyte function and the potential underlying mechanisms, and identified a novel role for TFEB in regulation of the podocyte actin cytoskeleton.
Collapse
Affiliation(s)
- Cuili Li
- School of Medicine, South China University of Technology, Guangzhou, 510006, China; Department of Nephrology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China
| | - Yan Yang
- Department of Nephrology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China; The Second School of Clinical Medicine, Southern Medical University, Guangzhou, 510515, China
| | - Luan Li
- School of Medicine, South China University of Technology, Guangzhou, 510006, China; Department of Nephrology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China
| | - Yingwen Chen
- School of Medicine, South China University of Technology, Guangzhou, 510006, China; Department of Nephrology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China
| | - Qingying Shi
- Department of Nephrology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China; The Second School of Clinical Medicine, Southern Medical University, Guangzhou, 510515, China
| | - Hong Zhang
- School of Medicine, South China University of Technology, Guangzhou, 510006, China; Department of Nephrology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China
| | - Li Zhang
- Department of Nephrology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China
| | - Yuanhan Chen
- Department of Nephrology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China
| | - Ruizhao Li
- Department of Nephrology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China
| | - Zhilian Li
- Department of Nephrology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China
| | - Shuangxin Liu
- Department of Nephrology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China
| | - Zhiming Ye
- Department of Nephrology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China
| | - Xingchen Zhao
- Department of Nephrology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China.
| | - Xinling Liang
- School of Medicine, South China University of Technology, Guangzhou, 510006, China; Department of Nephrology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China.
| |
Collapse
|
43
|
Brown AD, Vergunst KL, Branch M, Blair CM, Dupré DJ, Baillie GS, Langelaan DN. Structural basis of CBP/p300 recruitment by the microphthalmia-associated transcription factor. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2023; 1870:119520. [PMID: 37353163 DOI: 10.1016/j.bbamcr.2023.119520] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 05/19/2023] [Accepted: 06/08/2023] [Indexed: 06/25/2023]
Abstract
The microphthalmia-associated transcription factor (MITF) is a master regulator of the melanocyte cell lineage. Aberrant MITF activity can lead to multiple malignancies including skin cancer, where it modulates the progression and invasiveness of melanoma. MITF-regulated gene expression requires recruitment of the transcriptional co-regulator CBP/p300, but details of this process are not fully defined. In this study, we investigate the structural and functional interaction between the MITF N-terminal transactivation domain (MITFTAD) and CBP/p300. Using pulldown assays and nuclear magnetic resonance spectroscopy we determined that MITFTAD is intrinsically disordered and binds to the TAZ1 and TAZ2 domains of CBP/p300 with moderate affinity. The solution-state structure of the MITFTAD:TAZ2 complex reveals that MITF interacts with a hydrophobic surface of TAZ2, while remaining somewhat dynamic. Peptide array and mutagenesis experiments determined that an acidic motif is integral to the MITFTAD:TAZ2 interaction and is necessary for transcriptional activity of MITF. Peptides that bind to the same surface of TAZ2 as MITFTAD, such as the adenoviral protein E1A, are capable of displacing MITF from TAZ2 and inhibiting transactivation. These findings provide insight into co-activator recruitment by MITF that are fundamental to our understanding of MITF targeted gene regulation and melanoma biology.
Collapse
Affiliation(s)
- Alexandra D Brown
- Department of Biochemistry & Molecular Biology, Dalhousie University, Halifax, NS B3H 4R2, Canada
| | - Kathleen L Vergunst
- Department of Biochemistry & Molecular Biology, Dalhousie University, Halifax, NS B3H 4R2, Canada
| | - Makenzie Branch
- Department of Biochemistry & Molecular Biology, Dalhousie University, Halifax, NS B3H 4R2, Canada
| | - Connor M Blair
- School of Cardiovascular & Metabolic Health, University of Glasgow, Glasgow G12 8QQ, Scotland, United Kingdom of Great Britain and Northern Ireland
| | - Denis J Dupré
- Department of Pharmacology, Dalhousie University, Halifax, NS B3H 4R2, Canada
| | - George S Baillie
- School of Cardiovascular & Metabolic Health, University of Glasgow, Glasgow G12 8QQ, Scotland, United Kingdom of Great Britain and Northern Ireland
| | - David N Langelaan
- Department of Biochemistry & Molecular Biology, Dalhousie University, Halifax, NS B3H 4R2, Canada.
| |
Collapse
|
44
|
Zheng PH, Lu YP, Zhang XX, Luan KE, Zhang ZL, Li JJ, Xu T, Li JT, Xian JA, Guo H, Wang AL. New insights into the regulation mechanism of Pacific white shrimp (Litopenaeus vannamei) hepatopancreas under 4-nonylphenol exposure using transcriptome analysis. FISH & SHELLFISH IMMUNOLOGY 2023; 141:109050. [PMID: 37666313 DOI: 10.1016/j.fsi.2023.109050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Revised: 08/28/2023] [Accepted: 09/01/2023] [Indexed: 09/06/2023]
Abstract
4-Nonylphenol (4-NP) is one of the common endocrine-disrupting chemicals (EDCs) in estuaries and coastal zones, which can exert detrimental effects on the physiological function of aquatic organisms. However, the molecular response triggered by 4-NP remains largely unknown in Pacific white shrimp (Litopenaeus vannamei). In this study, transcriptomic analysis was performed to investigate the underlying mechanisms of 4-NP toxicity in the hepatopancreas of L. vannamei. Nine RNA-Seq libraries were generated from L. vannamei at 0 h, 24 h, and 48 h following exposure to 4-NP. Compared with 0 h vs 24 h, 962 up- and 463 down-regulated differentially expressed genes (DEGs) were identified, indicating that many genes in L. vannamei were induced to resist adverse circumstances by 4-NP exposure. In contrast, 902 up- and 1027 down-regulated DEGs were revealed in the comparison of 0 h vs 48 h, demonstrating that prolonged exposure to the stress from 4-NP resulted in more inhibited genes. To validate the accuracy of the transcriptome data, eight DEGs were selected for quantitative real-time polymerase chain reaction (qRT-PCR), which were consistent with the RNA-Seq results. Through KEGG pathway enrichment analysis, three specific pathways related to hormonal effects and endocrine function of L. vannamei were enriched significantly, including tyrosine metabolism, insect hormone biosynthesis, and melanogenesis. After 4-NP stress, genes involved in tyrosine metabolism (Tyr) and melanogenesis pathway (AC, CBP, Wnt, Frizzled, Tcf, and Ras) were induced to promote melanin pigment to help shrimp resist adverse environments. In the insect hormone biosynthesis, ALDH, CYP15A1, CYP15A1/C1, and JHE genes were activated to synthesize juvenile hormone (JH), while Spook, Phm, Sad, and CYP18A1 were induced to generate molting hormone. There is an enhanced interaction between the molting hormone and JH, with JH playing a dominant role and maintaining its "classic status quo action". Our study demonstrated that 4-NP exposure led to impairments of biological functions in L. vannamei hepatopancreas. The genes and pathways identified provide novel insights into the molecular mechanisms underlying 4-NP toxicity effects in prawns and enrich the information on the toxicity mechanism of crustaceans in response to EDCs exposure.
Collapse
Affiliation(s)
- Pei-Hua Zheng
- Hainan Provincial Key Laboratory for Functional Components Research and Utilization of Marine Bio-resources, Institute of Tropical Biosciences and Biotechnology, Chinese Academy of Tropical Agricultural Sciences & Key Laboratory for Biology and Genetic Resources of Tropical Crops of Hainan Province, Hainan Institute of Tropical Agricultural Resources, Haikou, 571101, PR China
| | - Yao-Peng Lu
- Hainan Provincial Key Laboratory for Functional Components Research and Utilization of Marine Bio-resources, Institute of Tropical Biosciences and Biotechnology, Chinese Academy of Tropical Agricultural Sciences & Key Laboratory for Biology and Genetic Resources of Tropical Crops of Hainan Province, Hainan Institute of Tropical Agricultural Resources, Haikou, 571101, PR China
| | - Xiu-Xia Zhang
- Hainan Provincial Key Laboratory for Functional Components Research and Utilization of Marine Bio-resources, Institute of Tropical Biosciences and Biotechnology, Chinese Academy of Tropical Agricultural Sciences & Key Laboratory for Biology and Genetic Resources of Tropical Crops of Hainan Province, Hainan Institute of Tropical Agricultural Resources, Haikou, 571101, PR China
| | - Ke-Er Luan
- Hainan Provincial Key Laboratory for Functional Components Research and Utilization of Marine Bio-resources, Institute of Tropical Biosciences and Biotechnology, Chinese Academy of Tropical Agricultural Sciences & Key Laboratory for Biology and Genetic Resources of Tropical Crops of Hainan Province, Hainan Institute of Tropical Agricultural Resources, Haikou, 571101, PR China
| | - Ze-Long Zhang
- Hainan Provincial Key Laboratory for Functional Components Research and Utilization of Marine Bio-resources, Institute of Tropical Biosciences and Biotechnology, Chinese Academy of Tropical Agricultural Sciences & Key Laboratory for Biology and Genetic Resources of Tropical Crops of Hainan Province, Hainan Institute of Tropical Agricultural Resources, Haikou, 571101, PR China
| | - Jia-Jun Li
- Hainan Provincial Key Laboratory for Functional Components Research and Utilization of Marine Bio-resources, Institute of Tropical Biosciences and Biotechnology, Chinese Academy of Tropical Agricultural Sciences & Key Laboratory for Biology and Genetic Resources of Tropical Crops of Hainan Province, Hainan Institute of Tropical Agricultural Resources, Haikou, 571101, PR China
| | - Tong Xu
- Hainan Provincial Key Laboratory for Functional Components Research and Utilization of Marine Bio-resources, Institute of Tropical Biosciences and Biotechnology, Chinese Academy of Tropical Agricultural Sciences & Key Laboratory for Biology and Genetic Resources of Tropical Crops of Hainan Province, Hainan Institute of Tropical Agricultural Resources, Haikou, 571101, PR China
| | - Jun-Tao Li
- Hainan Provincial Key Laboratory for Functional Components Research and Utilization of Marine Bio-resources, Institute of Tropical Biosciences and Biotechnology, Chinese Academy of Tropical Agricultural Sciences & Key Laboratory for Biology and Genetic Resources of Tropical Crops of Hainan Province, Hainan Institute of Tropical Agricultural Resources, Haikou, 571101, PR China
| | - Jian-An Xian
- Hainan Provincial Key Laboratory for Functional Components Research and Utilization of Marine Bio-resources, Institute of Tropical Biosciences and Biotechnology, Chinese Academy of Tropical Agricultural Sciences & Key Laboratory for Biology and Genetic Resources of Tropical Crops of Hainan Province, Hainan Institute of Tropical Agricultural Resources, Haikou, 571101, PR China.
| | - Hui Guo
- Guangdong South China Sea Key Laboratory of Aquaculture for Aquatic Economic Animals, College of Fisheries, Guangdong Ocean University, Zhanjiang, 524025, PR China.
| | - An-Li Wang
- School of Life Sciences, South China Normal University, Guangzhou, 510631, PR China.
| |
Collapse
|
45
|
Wen W, Zheng H, Li W, Huang G, Chen P, Zhu X, Cao Y, Li J, Huang X, Huang Y. Transcription factor EB: A potential integrated network regulator in metabolic-associated cardiac injury. Metabolism 2023; 147:155662. [PMID: 37517793 DOI: 10.1016/j.metabol.2023.155662] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Revised: 07/19/2023] [Accepted: 07/21/2023] [Indexed: 08/01/2023]
Abstract
With the worldwide pandemic of metabolic diseases, such as obesity, diabetes, and non-alcoholic fatty liver disease (NAFLD), cardiometabolic disease (CMD) has become a significant cause of death in humans. However, the pathophysiology of metabolic-associated cardiac injury is complex and not completely clear, and it is important to explore new strategies and targets for the treatment of CMD. A series of pathophysiological disturbances caused by metabolic disorders, such as insulin resistance (IR), hyperglycemia, hyperlipidemia, mitochondrial dysfunction, oxidative stress, inflammation, endoplasmic reticulum stress (ERS), autophagy dysfunction, calcium homeostasis imbalance, and endothelial dysfunction, may be related to the incidence and development of CMD. Transcription Factor EB (TFEB), as a transcription factor, has been extensively studied for its role in regulating lysosomal biogenesis and autophagy. Recently, the regulatory role of TFEB in other biological processes, including the regulation of glucose homeostasis, lipid metabolism, etc. has been gradually revealed. In this review, we will focus on the relationship between TFEB and IR, lipid metabolism, endothelial dysfunction, oxidative stress, inflammation, ERS, calcium homeostasis, autophagy, and mitochondrial quality control (MQC) and the potential regulatory mechanisms among them, to provide a comprehensive summary for TFEB as a potential new therapeutic target for CMD.
Collapse
Affiliation(s)
- Weixing Wen
- Department of Cardiology, Shunde Hospital, Southern Medical University (The First People's Hospital of Shunde), NO. 1 Jiazi Road, Lunjiao, Shunde District, Foshan City, Guangdong 528308, China; Medical Research Center, Shunde Hospital, Southern Medical University (The First People's Hospital of Shunde), NO. 1 Jiazi Road, Lunjiao, Shunde District, Foshan City, Guangdong 528308, China
| | - Haoxiao Zheng
- Department of Cardiology, Shunde Hospital, Southern Medical University (The First People's Hospital of Shunde), NO. 1 Jiazi Road, Lunjiao, Shunde District, Foshan City, Guangdong 528308, China; Medical Research Center, Shunde Hospital, Southern Medical University (The First People's Hospital of Shunde), NO. 1 Jiazi Road, Lunjiao, Shunde District, Foshan City, Guangdong 528308, China.
| | - Weiwen Li
- Department of Cardiology, Shunde Hospital, Southern Medical University (The First People's Hospital of Shunde), NO. 1 Jiazi Road, Lunjiao, Shunde District, Foshan City, Guangdong 528308, China; Medical Research Center, Shunde Hospital, Southern Medical University (The First People's Hospital of Shunde), NO. 1 Jiazi Road, Lunjiao, Shunde District, Foshan City, Guangdong 528308, China
| | - Guolin Huang
- Department of Cardiology, Shunde Hospital, Southern Medical University (The First People's Hospital of Shunde), NO. 1 Jiazi Road, Lunjiao, Shunde District, Foshan City, Guangdong 528308, China; Medical Research Center, Shunde Hospital, Southern Medical University (The First People's Hospital of Shunde), NO. 1 Jiazi Road, Lunjiao, Shunde District, Foshan City, Guangdong 528308, China
| | - Peng Chen
- Department of Cardiology, Shunde Hospital, Southern Medical University (The First People's Hospital of Shunde), NO. 1 Jiazi Road, Lunjiao, Shunde District, Foshan City, Guangdong 528308, China; Medical Research Center, Shunde Hospital, Southern Medical University (The First People's Hospital of Shunde), NO. 1 Jiazi Road, Lunjiao, Shunde District, Foshan City, Guangdong 528308, China
| | - Xiaolin Zhu
- Department of Cardiology, Shunde Hospital, Southern Medical University (The First People's Hospital of Shunde), NO. 1 Jiazi Road, Lunjiao, Shunde District, Foshan City, Guangdong 528308, China; Medical Research Center, Shunde Hospital, Southern Medical University (The First People's Hospital of Shunde), NO. 1 Jiazi Road, Lunjiao, Shunde District, Foshan City, Guangdong 528308, China.
| | - Yue Cao
- Department of Cardiology, Shunde Hospital, Southern Medical University (The First People's Hospital of Shunde), NO. 1 Jiazi Road, Lunjiao, Shunde District, Foshan City, Guangdong 528308, China; Medical Research Center, Shunde Hospital, Southern Medical University (The First People's Hospital of Shunde), NO. 1 Jiazi Road, Lunjiao, Shunde District, Foshan City, Guangdong 528308, China
| | - Jiahuan Li
- Department of Cardiology, Shunde Hospital, Southern Medical University (The First People's Hospital of Shunde), NO. 1 Jiazi Road, Lunjiao, Shunde District, Foshan City, Guangdong 528308, China; Medical Research Center, Shunde Hospital, Southern Medical University (The First People's Hospital of Shunde), NO. 1 Jiazi Road, Lunjiao, Shunde District, Foshan City, Guangdong 528308, China
| | - Xiaohui Huang
- Department of Cardiology, Shunde Hospital, Southern Medical University (The First People's Hospital of Shunde), NO. 1 Jiazi Road, Lunjiao, Shunde District, Foshan City, Guangdong 528308, China; Medical Research Center, Shunde Hospital, Southern Medical University (The First People's Hospital of Shunde), NO. 1 Jiazi Road, Lunjiao, Shunde District, Foshan City, Guangdong 528308, China
| | - Yuli Huang
- Department of Cardiology, Shunde Hospital, Southern Medical University (The First People's Hospital of Shunde), NO. 1 Jiazi Road, Lunjiao, Shunde District, Foshan City, Guangdong 528308, China; The George Institute for Global Health, Faculty of Medicine, University of New South Wales, Sydney, Australia; Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation Research, Guangzhou, China; Medical Research Center, Shunde Hospital, Southern Medical University (The First People's Hospital of Shunde), NO. 1 Jiazi Road, Lunjiao, Shunde District, Foshan City, Guangdong 528308, China.
| |
Collapse
|
46
|
Brown AD, Lynch K, Langelaan DN. The C-terminal transactivation domain of MITF interacts promiscuously with co-activator CBP/p300. Sci Rep 2023; 13:16094. [PMID: 37752231 PMCID: PMC10522771 DOI: 10.1038/s41598-023-43207-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Accepted: 09/21/2023] [Indexed: 09/28/2023] Open
Abstract
The microphthalmia-associated transcription factor (MITF) is one of four closely related members of the MiT/TFE family (TFEB, TFE3, TFEC) that regulate a wide range of cellular processes. MITF is a key regulator of melanocyte-associated genes, and essential to proper development of the melanocyte cell lineage. Abnormal MITF activity can contribute to the onset of several diseases including melanoma, where MITF is an amplified oncogene. To enhance transcription, MITF recruits the co-activator CREB-binding protein (CBP) and its homolog p300 to gene promoters, however the molecular determinants of their interaction are not yet fully understood. Here, we characterize the interactions between the C-terminal MITF transactivation domain and CBP/p300. Using NMR spectroscopy, protein pulldown assays, and isothermal titration calorimetry we determine the C-terminal region of MITF is intrinsically disordered and binds with high-affinity to both TAZ1 and TAZ2 of CBP/p300. Mutagenesis studies revealed two conserved motifs within MITF that are necessary for TAZ2 binding and critical for MITF-dependent transcription of a reporter gene. Finally, we observe the transactivation potential of the MITF C-terminal region is reliant on the N-terminal transactivation domain for function. Taken together, our study helps elucidate the molecular details of how MITF interacts with CBP/p300 through multiple redundant interactions that lend insight into MITF function in melanocytes and melanoma.
Collapse
Affiliation(s)
- Alexandra D Brown
- Department of Biochemistry and Molecular Biology, Dalhousie University, Halifax, NS, B3H 4R2, Canada
| | - Kyle Lynch
- Department of Biochemistry and Molecular Biology, Dalhousie University, Halifax, NS, B3H 4R2, Canada
| | - David N Langelaan
- Department of Biochemistry and Molecular Biology, Dalhousie University, Halifax, NS, B3H 4R2, Canada.
| |
Collapse
|
47
|
Zheng HY, Wang YX, Zhou K, Xie HL, Ren Z, Liu HT, Ou YS, Zhou ZX, Jiang ZS. Biological functions of CRTC2 and its role in metabolism-related diseases. J Cell Commun Signal 2023; 17:495-506. [PMID: 36856929 PMCID: PMC10409973 DOI: 10.1007/s12079-023-00730-5] [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: 08/04/2022] [Accepted: 02/01/2023] [Indexed: 03/02/2023] Open
Abstract
CREB-regulated transcription coactivator2 (CRTC2 or TORC2) is a transcriptional coactivator of CREB(cAMP response element binding protein), which affects human energy metabolism through cyclic adenosine phosphate pathway, Mammalian target of rapamycin (mTOR) pathway, Sterol regulatory element binding protein 1(SREBP1), Sterol regulatory element binding protein 2 (SREBP2) and other substances Current studies on CRTC2 mainly focus on glucose and lipid metabolism, relevant studies show that CRTC2 can participate in the occurrence and development of related diseases by affecting metabolic homeostasis. It has been found that Crtc2 acts as a signaling regulator for cAMP and Ca2 + signaling pathways in many cell types, and phosphorylation at ser171 and ser275 can regulate downstream biological functions by controlling CRTC2 shuttling between cytoplasm and nucleus.
Collapse
Affiliation(s)
- Hong-Yu Zheng
- Institute of Cardiovascular Disease, Key Lab for Arteriosclerology of Hunan Province, International Joint Laboratory for Arteriosclerotic Disease Research of Hunan Province, University of South China, Hengyang, 421001, China
| | - Yan-Xia Wang
- Institute of Cardiovascular Disease, Key Lab for Arteriosclerology of Hunan Province, International Joint Laboratory for Arteriosclerotic Disease Research of Hunan Province, University of South China, Hengyang, 421001, China
| | - Kun Zhou
- Institute of Cardiovascular Disease, Key Lab for Arteriosclerology of Hunan Province, International Joint Laboratory for Arteriosclerotic Disease Research of Hunan Province, University of South China, Hengyang, 421001, China
| | - Hai-Lin Xie
- Institute of Cardiovascular Disease, Key Lab for Arteriosclerology of Hunan Province, International Joint Laboratory for Arteriosclerotic Disease Research of Hunan Province, University of South China, Hengyang, 421001, China
| | - Zhong Ren
- Institute of Cardiovascular Disease, Key Lab for Arteriosclerology of Hunan Province, International Joint Laboratory for Arteriosclerotic Disease Research of Hunan Province, University of South China, Hengyang, 421001, China
| | - Hui-Ting Liu
- Institute of Cardiovascular Disease, Key Lab for Arteriosclerology of Hunan Province, International Joint Laboratory for Arteriosclerotic Disease Research of Hunan Province, University of South China, Hengyang, 421001, China
| | - Yang-Shao Ou
- Institute of Cardiovascular Disease, Key Lab for Arteriosclerology of Hunan Province, International Joint Laboratory for Arteriosclerotic Disease Research of Hunan Province, University of South China, Hengyang, 421001, China
| | - Zhi-Xiang Zhou
- Institute of Cardiovascular Disease, Key Lab for Arteriosclerology of Hunan Province, International Joint Laboratory for Arteriosclerotic Disease Research of Hunan Province, University of South China, Hengyang, 421001, China
| | - Zhi-Sheng Jiang
- Institute of Cardiovascular Disease, Key Lab for Arteriosclerology of Hunan Province, International Joint Laboratory for Arteriosclerotic Disease Research of Hunan Province, University of South China, Hengyang, 421001, China.
| |
Collapse
|
48
|
Wang S, Long H, Hou L, Feng B, Ma Z, Wu Y, Zeng Y, Cai J, Zhang DW, Zhao G. The mitophagy pathway and its implications in human diseases. Signal Transduct Target Ther 2023; 8:304. [PMID: 37582956 PMCID: PMC10427715 DOI: 10.1038/s41392-023-01503-7] [Citation(s) in RCA: 218] [Impact Index Per Article: 109.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 05/03/2023] [Accepted: 05/16/2023] [Indexed: 08/17/2023] Open
Abstract
Mitochondria are dynamic organelles with multiple functions. They participate in necrotic cell death and programmed apoptotic, and are crucial for cell metabolism and survival. Mitophagy serves as a cytoprotective mechanism to remove superfluous or dysfunctional mitochondria and maintain mitochondrial fine-tuning numbers to balance intracellular homeostasis. Growing evidences show that mitophagy, as an acute tissue stress response, plays an important role in maintaining the health of the mitochondrial network. Since the timely removal of abnormal mitochondria is essential for cell survival, cells have evolved a variety of mitophagy pathways to ensure that mitophagy can be activated in time under various environments. A better understanding of the mechanism of mitophagy in various diseases is crucial for the treatment of diseases and therapeutic target design. In this review, we summarize the molecular mechanisms of mitophagy-mediated mitochondrial elimination, how mitophagy maintains mitochondrial homeostasis at the system levels and organ, and what alterations in mitophagy are related to the development of diseases, including neurological, cardiovascular, pulmonary, hepatic, renal disease, etc., in recent advances. Finally, we summarize the potential clinical applications and outline the conditions for mitophagy regulators to enter clinical trials. Research advances in signaling transduction of mitophagy will have an important role in developing new therapeutic strategies for precision medicine.
Collapse
Affiliation(s)
- Shouliang Wang
- The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan City People's Hospital, Qingyuan, Guangdong, China
| | - Haijiao Long
- The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan City People's Hospital, Qingyuan, Guangdong, China
- Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Lianjie Hou
- The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan City People's Hospital, Qingyuan, Guangdong, China
| | - Baorong Feng
- The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan City People's Hospital, Qingyuan, Guangdong, China
| | - Zihong Ma
- The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan City People's Hospital, Qingyuan, Guangdong, China
| | - Ying Wu
- The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan City People's Hospital, Qingyuan, Guangdong, China
| | - Yu Zeng
- The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan City People's Hospital, Qingyuan, Guangdong, China
| | - Jiahao Cai
- The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan City People's Hospital, Qingyuan, Guangdong, China
| | - Da-Wei Zhang
- Group on the Molecular and Cell Biology of Lipids and Department of Pediatrics, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada.
| | - Guojun Zhao
- The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan City People's Hospital, Qingyuan, Guangdong, China.
| |
Collapse
|
49
|
Lovero D, Porcelli D, Giordano L, Lo Giudice C, Picardi E, Pesole G, Pignataro E, Palazzo A, Marsano RM. Structural and Comparative Analyses of Insects Suggest the Presence of an Ultra-Conserved Regulatory Element of the Genes Encoding Vacuolar-Type ATPase Subunits and Assembly Factors. BIOLOGY 2023; 12:1127. [PMID: 37627011 PMCID: PMC10452791 DOI: 10.3390/biology12081127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Revised: 07/28/2023] [Accepted: 08/09/2023] [Indexed: 08/27/2023]
Abstract
Gene and genome comparison represent an invaluable tool to identify evolutionarily conserved sequences with possible functional significance. In this work, we have analyzed orthologous genes encoding subunits and assembly factors of the V-ATPase complex, an important enzymatic complex of the vacuolar and lysosomal compartments of the eukaryotic cell with storage and recycling functions, respectively, as well as the main pump in the plasma membrane that energizes the epithelial transport in insects. This study involves 70 insect species belonging to eight insect orders. We highlighted the conservation of a short sequence in the genes encoding subunits of the V-ATPase complex and their assembly factors analyzed with respect to their exon-intron organization of those genes. This study offers the possibility to study ultra-conserved regulatory elements under an evolutionary perspective, with the aim of expanding our knowledge on the regulation of complex gene networks at the basis of organellar biogenesis and cellular organization.
Collapse
Affiliation(s)
- Domenica Lovero
- Dipartimento di Bioscienze Biotecnologie e Ambiente, Università Degli Studi di Bari Aldo Moro, Via Orabona 4, 70125 Bari, Italy; (D.L.); (D.P.); (E.P.); (G.P.); (E.P.); (A.P.)
- MASMEC Biomed S.p.A., Via Delle Violette 14, 70026 Modugno, Italy
| | - Damiano Porcelli
- Dipartimento di Bioscienze Biotecnologie e Ambiente, Università Degli Studi di Bari Aldo Moro, Via Orabona 4, 70125 Bari, Italy; (D.L.); (D.P.); (E.P.); (G.P.); (E.P.); (A.P.)
- METALABS S.R.L., Corso A. De Gasperi 381/1, 70125 Bari, Italy
| | - Luca Giordano
- Cardio-Pulmonary Institute (CPI), Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Justus-Liebig-University, Aulweg 130, 35392 Giessen, Germany;
| | - Claudio Lo Giudice
- Istituto di Tecnologie Biomediche (ITB), Consiglio Nazionale Delle Ricerche, Via Giovanni Amendola, 122, 70126 Bari, Italy;
| | - Ernesto Picardi
- Dipartimento di Bioscienze Biotecnologie e Ambiente, Università Degli Studi di Bari Aldo Moro, Via Orabona 4, 70125 Bari, Italy; (D.L.); (D.P.); (E.P.); (G.P.); (E.P.); (A.P.)
| | - Graziano Pesole
- Dipartimento di Bioscienze Biotecnologie e Ambiente, Università Degli Studi di Bari Aldo Moro, Via Orabona 4, 70125 Bari, Italy; (D.L.); (D.P.); (E.P.); (G.P.); (E.P.); (A.P.)
| | - Eugenia Pignataro
- Dipartimento di Bioscienze Biotecnologie e Ambiente, Università Degli Studi di Bari Aldo Moro, Via Orabona 4, 70125 Bari, Italy; (D.L.); (D.P.); (E.P.); (G.P.); (E.P.); (A.P.)
| | - Antonio Palazzo
- Dipartimento di Bioscienze Biotecnologie e Ambiente, Università Degli Studi di Bari Aldo Moro, Via Orabona 4, 70125 Bari, Italy; (D.L.); (D.P.); (E.P.); (G.P.); (E.P.); (A.P.)
| | - René Massimiliano Marsano
- Dipartimento di Bioscienze Biotecnologie e Ambiente, Università Degli Studi di Bari Aldo Moro, Via Orabona 4, 70125 Bari, Italy; (D.L.); (D.P.); (E.P.); (G.P.); (E.P.); (A.P.)
| |
Collapse
|
50
|
Vu HN, Valdimarsson MM, Sigurbjörnsdóttir S, Bergsteinsdóttir K, Debbache J, Bismuth K, Swing DA, Hallsson JH, Larue L, Arnheiter H, Copeland NG, Jenkins NA, Heidarsson PO, Steingrímsson E. Novel mechanisms of MITF regulation and melanoma predisposition identified in a mouse suppressor screen. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.04.551952. [PMID: 37786677 PMCID: PMC10541597 DOI: 10.1101/2023.08.04.551952] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/04/2023]
Abstract
MITF, a basic-Helix-Loop-Helix Zipper (bHLHZip) transcription factor, plays vital roles in melanocyte development and functions as an oncogene. To explore MITF regulation and its role in melanoma, we conducted a genetic screen for suppressors of the Mitf-associated pigmentation phenotype. An intragenic Mitf mutation was identified, leading to termination of MITF at the K316 SUMOylation site and loss of the C-end intrinsically disordered region (IDR). The resulting protein is more nuclear but less stable than wild-type MITF and retains DNA-binding ability. Interestingly, as a dimer, it can translocate wild-type and mutant MITF partners into the nucleus, improving its own stability and ensuring an active nuclear MITF supply. Interactions between K316 SUMOylation and S409 phosphorylation sites across monomers largely explain the observed effects. Notably, the recurrent melanoma-associated E318K mutation in MITF, which affects K316 SUMOylation, also alters protein regulation in concert with S409, unraveling a novel regulatory mechanism with unexpected disease insights.
Collapse
Affiliation(s)
- Hong Nhung Vu
- Department of Biochemistry and Molecular Biology, BioMedical Center, Faculty of Medicine, University of Iceland, Sturlugata 8, 102 Reykjavík, Iceland
| | - Matti Már Valdimarsson
- Department of Biochemistry, Science Institute, School of Engineering and Natural Sciences, University of Iceland, Sturlugata 7, 102 Reykjavík, Iceland
| | - Sara Sigurbjörnsdóttir
- Department of Biochemistry and Molecular Biology, BioMedical Center, Faculty of Medicine, University of Iceland, Sturlugata 8, 102 Reykjavík, Iceland
| | - Kristín Bergsteinsdóttir
- Department of Biochemistry and Molecular Biology, BioMedical Center, Faculty of Medicine, University of Iceland, Sturlugata 8, 102 Reykjavík, Iceland
| | - Julien Debbache
- Mammalian Development Section, NINDS, NIH, Bethesda, MD 20892-3706
| | - Keren Bismuth
- Mammalian Development Section, NINDS, NIH, Bethesda, MD 20892-3706
| | | | - Jón H. Hallsson
- Department of Biochemistry and Molecular Biology, BioMedical Center, Faculty of Medicine, University of Iceland, Sturlugata 8, 102 Reykjavík, Iceland
| | - Lionel Larue
- Institut Curie, PSL Research University, INSERM U1021, Normal and Pathological Development of Melanocytes, 91405, Orsay, France
| | - Heinz Arnheiter
- Mammalian Development Section, NINDS, NIH, Bethesda, MD 20892-3706
| | - Neal G. Copeland
- Mouse Cancer Genetics Program, NCI, Frederick, MD 21702-1201
- Current address: Genetics Department, MD Anderson Cancer Center, Houston, TX 77030
| | - Nancy A. Jenkins
- Mouse Cancer Genetics Program, NCI, Frederick, MD 21702-1201
- Current address: Genetics Department, MD Anderson Cancer Center, Houston, TX 77030
| | - Petur O. Heidarsson
- Department of Biochemistry, Science Institute, School of Engineering and Natural Sciences, University of Iceland, Sturlugata 7, 102 Reykjavík, Iceland
| | - Eiríkur Steingrímsson
- Department of Biochemistry and Molecular Biology, BioMedical Center, Faculty of Medicine, University of Iceland, Sturlugata 8, 102 Reykjavík, Iceland
| |
Collapse
|