1
|
Yang Y, Wu Y, Xiang L, Picardo M, Zhang C. Deciphering the role of skin aging in pigmentary disorders. Free Radic Biol Med 2025; 227:638-655. [PMID: 39674424 DOI: 10.1016/j.freeradbiomed.2024.12.032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Revised: 12/08/2024] [Accepted: 12/11/2024] [Indexed: 12/16/2024]
Abstract
Skin aging is a complex biological process involving intrinsic and extrinsic factors. Skin aging contains alterations at the tissue, cellular, and molecular levels. Currently, there is increasing evidence that skin aging occurs not only in time-dependent chronological aging but also plays a role in skin pigmentary disorders. This review provides an in-depth analysis of the impact of skin aging on different types of pigmentary disorders, including both hyperpigmentation disorders such as melasma and senile lentigo and hypopigmentation disorders such as vitiligo, idiopathic guttate hypomelanosis and graying of hair. In addition, we explore the mechanisms of skin aging on pigmentation regulation and suggest several potential therapeutic approaches for skin aging and aging-related pigmentary disorders.
Collapse
Affiliation(s)
- Yiwen Yang
- Department of Dermatology, Huashan Hospital, Fudan University, No.12 Wulumuqi Zhong Road, Shanghai 200040, PR China
| | - Yue Wu
- Department of Dermatology, Huashan Hospital, Fudan University, No.12 Wulumuqi Zhong Road, Shanghai 200040, PR China
| | - Leihong Xiang
- Department of Dermatology, Huashan Hospital, Fudan University, No.12 Wulumuqi Zhong Road, Shanghai 200040, PR China
| | - Mauro Picardo
- Istituto Dermopatico Immacolata, IDI-RCCS, Rome, Italy.
| | - Chengfeng Zhang
- Department of Dermatology, Huashan Hospital, Fudan University, No.12 Wulumuqi Zhong Road, Shanghai 200040, PR China.
| |
Collapse
|
2
|
Zhao N, Xiong Q, Li P, Chen G, Xiao H, Wu C. TSC complex decrease the expression of mTOR by regulated miR-199b-3p. Sci Rep 2025; 15:1892. [PMID: 39806027 PMCID: PMC11730325 DOI: 10.1038/s41598-025-85706-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Accepted: 01/06/2025] [Indexed: 01/16/2025] Open
Abstract
The TSC complex formed by TSC1 and TSC2 is the most important upstream negative regulator of mTORC1. Genetic variations in either TSC1 or TSC2 cause tuberous sclerosis complex (TSC) disease which is a rare autosomal dominant disorder resulting in impairment of multiple organ systems. In this study, besides a reported variation, c.2509_2512del (p.Asn837Valfs*11, p.N837fs) in TSC1, we found a de novo TSC2 variation c.1113delG (p.Gln371Hisfs*18, p.Q371fs), which these two mutation influence the formation of TSC complex. We found that the decrease of TSC complex with the appearance of the decreased miR-199b-3p expression. At the same time, the reduction of miR-199b-3p increased the expression of mTOR and the activation of mTORC1 and mTORC2, the additional miR-199b-3p caused the decrease the expression of mTOR and the activation of mTORC1 and mTORC2. In brief, our results may illustrate a novel mechanism of TSC caused by variations in either TSC1 or TSC2, and a new mTOR expression regulator, miR-199b-3p.
Collapse
Affiliation(s)
- Na Zhao
- Key Laboratory of Chemical Biology and Molecular Engineering of National Ministry of Education, Institutes of Biomedical Sciences, Shanxi University, Taiyuan, 030006, China
- Department of Pathology, The Second Hospital of ShanXi Medical University, No.382 WuYi Road, Tai Yuan, ShanXi, Taiyuan, 030000, China
| | - Qiuhong Xiong
- Key Laboratory of Chemical Biology and Molecular Engineering of National Ministry of Education, Institutes of Biomedical Sciences, Shanxi University, Taiyuan, 030006, China
| | - Ping Li
- Key Laboratory of Chemical Biology and Molecular Engineering of National Ministry of Education, Institutes of Biomedical Sciences, Shanxi University, Taiyuan, 030006, China
| | - Guangxin Chen
- Key Laboratory of Chemical Biology and Molecular Engineering of National Ministry of Education, Institutes of Biomedical Sciences, Shanxi University, Taiyuan, 030006, China
| | - Han Xiao
- Key Laboratory of Chemical Biology and Molecular Engineering of National Ministry of Education, Institutes of Biomedical Sciences, Shanxi University, Taiyuan, 030006, China.
| | - Changxin Wu
- Key Laboratory of Chemical Biology and Molecular Engineering of National Ministry of Education, Institutes of Biomedical Sciences, Shanxi University, Taiyuan, 030006, China.
| |
Collapse
|
3
|
Yang H, Zhang X, Wang W, Ge Y, Yang Y, Lin T. miR-25-5p in exosomes derived from UVB-induced fibroblasts regulates melanogenesis via TSC2-dominated cellular organelle dysfunction. J Dermatol Sci 2024; 115:75-84. [PMID: 38969533 DOI: 10.1016/j.jdermsci.2024.06.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2023] [Revised: 05/16/2024] [Accepted: 06/04/2024] [Indexed: 07/07/2024]
Abstract
BACKGROUND Few reports have confirmed whether exosomes derived from fibroblasts can regulate the process of melanogenesis. We wondered whether exosomes derived from fibroblasts could have a potent regulatory effect on melanogenesis and explored the underlying mechanisms. OBJECTIVE This study aimed to find the role of fibroblasts in melanocytes and revealed the related mechanisms. METHODS RT-qPCR, Western blot analysis were conducted to measure the RNA and protein expression level of various related genes. miRNA sequencing, mass spectrum analysis and subsequent bioinformatics analysis were employed to find the underlying targets. Zebrafish were employed to measure the melanin synthesis related process in vivo. Furthermore, electron microscopy, ROS measurement and dual-luciferase reporter assay were adopted to investigate the relationship between these processes. RESULTS We found that exosomes derived from human primary dermal fibroblasts were internalized by human primary melanocytes and MNT1 cells and that the melanin content and the expression of melanin synthesis-related proteins TYR and MITF was inhibited by exosomes derived from UVB-induced human primary dermal fibroblasts. The miRNA expression profile in secreted exosomes changed significantly, with miR-25-5p identified as capable of regulating TSC2 expression via the CDS region. The miR-25-5p-TSC2 axis could affect the melanin content through subsequent cellular organelle dysfunction, such as mitochondrial dysfunction, endoplasmic reticulum stress and dysregulation of lysosomal cysteine proteases. CONCLUSION We unveiled a novel regulatory role of fibroblasts in melanocytes, facilitated by the secretion of exosomes. miR-25-5p within exosomes plays a pivotal role in regulating melanogenesis via TSC2-induced cellular organelle dysfunction.
Collapse
Affiliation(s)
- Hedan Yang
- Department of Cosmetic Laser Surgery, Hospital of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, China
| | - Xiaoli Zhang
- Department of Cosmetic Laser Surgery, Hospital of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, China
| | - Wenzhu Wang
- Department of Cosmetic Laser Surgery, Hospital of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, China
| | - Yiping Ge
- Department of Cosmetic Laser Surgery, Hospital of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, China
| | - Yin Yang
- Department of Cosmetic Laser Surgery, Hospital of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, China.
| | - Tong Lin
- Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Hospital of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, China.
| |
Collapse
|
4
|
Jiang X, Yang C, Wang Z, Liang L, Gong Z, Huang S, Xu Z, Zhang B, Pei X, Cai L, Wang H, Lin Z. Loss-of-function variants in GLMN are associated with generalized skin hyperpigmentation with or without glomuvenous malformation. Br J Dermatol 2024; 191:107-116. [PMID: 38489583 DOI: 10.1093/bjd/ljae108] [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: 12/08/2023] [Revised: 01/30/2024] [Accepted: 03/08/2024] [Indexed: 03/17/2024]
Abstract
BACKGROUND Inherited hyperpigmented skin disorders comprise a group of entities with considerable clinical and genetic heterogenicity. The genetic basis of a majority of these disorders remains to be elucidated. OBJECTIVES This study aimed to identify the underlying gene for an unclarified disorder of autosomal-dominant generalized skin hyperpigmentation with or without glomuvenous malformation. METHODS Whole-exome sequencing was performed in five unrelated families with autosomal-dominant generalized skin hyperpigmentation. Variants were confirmed using Sanger sequencing and a minigene assay was employed to evaluate the splicing alteration. Immunofluorescence and transmission electron microscopy (TEM) were used to determine the quantity of melanocytes and melanosomes in hyperpigmented skin lesions. GLMN knockdown by small interfering RNA assays was performed in human MNT-1 cells to examine melanin concentration and the underlying molecular mechanism. RESULTS We identified five variants in GLMN in five unrelated families, including c.995_996insAACA(p.Ser333Thrfs*11), c.632 + 4delA, c.1470_1473dup(p.Thr492fs*12), c.1319G > A(p.Trp440*) and c.1613_1614insTA(Thr540*). The minigene assay confirmed that the c.632 + 4delA mutant resulted in abolishment of the canonical donor splice site. Although the number of melanocytes remained unchanged in skin lesions, as demonstrated by immunofluorescent staining of tyrosinase and premelanosome protein, TEM revealed an increased number of melanosomes in the skin lesion of a patient. The GLMN knockdown MNT-1 cells demonstrated a higher melanin concentration, a higher proportion of stage III and IV melanosomes, upregulation of microphthalmia-associated transcription factor and tyrosinase, and downregulation of phosphorylated p70S6 K vs. mock-transfected cells. CONCLUSIONS We found that loss-of-function variants in GLMN are associated with generalized skin hyperpigmentation with or without glomuvenous malformation. Our study implicates a potential role of glomulin in human skin melanogenesis, in addition to vascular morphogenesis.
Collapse
Affiliation(s)
- Xingyuan Jiang
- Dermatology Hospital, Southern Medical University, Guangzhou, China
- Department of Dermatology, Peking University First Hospital, Beijing Key Laboratory of Molecular Diagnosis on Dermatoses, National Clinical Research Center for Skin and Immune Diseases, Beijing, China
| | - Chao Yang
- Dermatology Hospital, Southern Medical University, Guangzhou, China
| | - Zhaoyang Wang
- Department of Dermatology, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
| | - Lina Liang
- Dermatology Hospital, Southern Medical University, Guangzhou, China
| | - Zhuoqing Gong
- Department of Dermatology, Peking University First Hospital, Beijing Key Laboratory of Molecular Diagnosis on Dermatoses, National Clinical Research Center for Skin and Immune Diseases, Beijing, China
| | - Shimiao Huang
- Dermatology Hospital, Southern Medical University, Guangzhou, China
| | - Zigang Xu
- Department of Dermatology, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
| | - Bin Zhang
- Department of Dermatology, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
| | - Xiaoping Pei
- Dermatology Hospital, Southern Medical University, Guangzhou, China
| | - Liangqi Cai
- Department of Dermatology, The First Affiliated Hospital of Xiamen University, Xiamen, China
| | - Huijun Wang
- Dermatology Hospital, Southern Medical University, Guangzhou, China
| | - Zhimiao Lin
- Dermatology Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
5
|
Maruta H, He H. Rapamycin vs TORin-1 or Gleevec vs Nilotinib: Simple chemical evolution that converts PAK1-blockers to TOR-blockers or vice versa? Drug Discov Ther 2024; 18:134-139. [PMID: 38569833 DOI: 10.5582/ddt.2023.01097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/05/2024]
Abstract
Both PAK1 (RAC/CDC42-activating kinase 1) and TOR (Target of Rapamycin) are among the major oncogenic/ageing kinases. However, they play the opposite role in our immune system, namely immune system is suppressed by PAK1, while it requires TOR. Thus, PAK1-blockers, would be more effective for therapy of cancers, than TOR-blockers. Since 2015 when we discovered genetically that PDGF-induced melanogenesis depends on "PAK1", we are able to screening a series of PAK1-blockers as melanogenesis-inhibitors which could eventually promote longevity. Interestingly, rapamycin, the first TOR-inhibitor, promotes melanogenesis, clearly indicating that TOR suppresses melanogenesis. However, a new TOR-inhibitor called TORin-1 no longer suppresses immune system, and blocks melanogenesis in cell culture. These observations strongly indicate that TORin-1 acts as PAK1-blockers, instead of TOR-blockers, in vivo. Thus, it is most likely that melanogenesis in cell culture could enable us to discriminate PAK1-blockers from TORblockers.
Collapse
Affiliation(s)
| | - Hong He
- Melbourne University Hospital (Austin Health), Melbourne, Australia
| |
Collapse
|
6
|
Zhang H, Zhu C, Zhao J, Zheng R, Xing J, Li Z, Zhang Y, Xu Q. The enhanced hepatotoxicity of isobavachalcone in depigmented zebrafish due to calcium signaling dysregulation and lipid metabolism disorder. J Appl Toxicol 2024; 44:919-932. [PMID: 38400677 DOI: 10.1002/jat.4593] [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: 01/02/2024] [Revised: 01/31/2024] [Accepted: 02/07/2024] [Indexed: 02/25/2024]
Abstract
Isobavachalcone (IBC) is a flavonoid component derived from Psoraleae Fructus that can increase skin pigmentation and treat vitiligo. However, IBC has been reported to be hepatotoxic. Current studies on IBC hepatotoxicity are mostly on normal organisms but lack studies on hepatotoxicity in patients. This study established the depigmented zebrafish model by using phenylthiourea (PTU) and investigated the difference in hepatotoxicity between normal and depigmented zebrafish caused by IBC and the underlying mechanism. Morphological, histological, and ultrastructural examination and RT-qPCR verification were used to evaluate the effects of IBC on the livers of zebrafish larvae. IBC significantly decreased liver volume, altered lipid metabolism, and induced pathological and ultrastructural changes in the livers of zebrafish with depigmentation compared with normal zebrafish. The RNA-sequencing and RT-qPCR results showed that the difference in hepatotoxicity between normal and depigmented zebrafish caused by IBC was closely related to the calcium signaling pathway, lipid decomposition and metabolism, and oxidative stress. This work delved into the mechanism of the enhanced IBC-induced hepatotoxicity in depigmented zebrafish and provided a new insight into the hepatotoxicity of IBC.
Collapse
Affiliation(s)
- Huiwen Zhang
- Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), Jinan, China
| | - Chengyue Zhu
- Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), Jinan, China
| | - Jingcheng Zhao
- Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), Jinan, China
- College of Medicine, Xin Jiang Medical University, Urumqi, China
| | - Ruifang Zheng
- Institute of Medicine of Xinjiang Uygur Autonomous Region, Urumqi, China
| | - Jianguo Xing
- Institute of Medicine of Xinjiang Uygur Autonomous Region, Urumqi, China
| | - Zhijian Li
- College of Medicine, Xin Jiang Medical University, Urumqi, China
- Hospital of Xin Jiang Traditional UYGMJR Medicine, Urumqi, China
| | - Yun Zhang
- Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), Jinan, China
| | - Qian Xu
- Jinan Municipal Hospital of Traditional Chinese Medicine, Jinan, China
| |
Collapse
|
7
|
Lin C, Traets JJH, Vredevoogd DW, Visser NL, Peeper DS. TSC2 regulates tumor susceptibility to TRAIL-mediated T-cell killing by orchestrating mTOR signaling. EMBO J 2023; 42:e111614. [PMID: 36715448 PMCID: PMC9975943 DOI: 10.15252/embj.2022111614] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2022] [Revised: 12/08/2022] [Accepted: 12/14/2022] [Indexed: 01/31/2023] Open
Abstract
Resistance to cancer immunotherapy continues to impair common clinical benefit. Here, we use whole-genome CRISPR-Cas9 knockout data to uncover an important role for Tuberous Sclerosis Complex 2 (TSC2) in determining tumor susceptibility to cytotoxic T lymphocyte (CTL) killing in human melanoma cells. TSC2-depleted tumor cells had disrupted mTOR regulation following CTL attack, which was associated with enhanced cell death. Wild-type tumor cells adapted to CTL attack by shifting their mTOR signaling balance toward increased mTORC2 activity, circumventing apoptosis, and necroptosis. TSC2 ablation strongly augmented tumor cell sensitivity to CTL attack in vitro and in vivo, suggesting one of its functions is to critically protect tumor cells. Mechanistically, TSC2 inactivation caused elevation of TRAIL receptor expression, cooperating with mTORC1-S6 signaling to induce tumor cell death. Clinically, we found a negative correlation between TSC2 expression and TRAIL signaling in TCGA patient cohorts. Moreover, a lower TSC2 immune response signature was observed in melanomas from patients responding to immune checkpoint blockade. Our study uncovers a pivotal role for TSC2 in the cancer immune response by governing crosstalk between TSC2-mTOR and TRAIL signaling, aiding future therapeutic exploration of this pathway in immuno-oncology.
Collapse
Affiliation(s)
- Chun‐Pu Lin
- Division of Molecular Oncology and ImmunologyOncode Institute, The Netherlands Cancer InstituteAmsterdamThe Netherlands
| | - Joleen J H Traets
- Division of Molecular Oncology and ImmunologyOncode Institute, The Netherlands Cancer InstituteAmsterdamThe Netherlands
- Division of Tumor Biology and ImmunologyThe Netherlands Cancer InstituteAmsterdamThe Netherlands
| | - David W Vredevoogd
- Division of Molecular Oncology and ImmunologyOncode Institute, The Netherlands Cancer InstituteAmsterdamThe Netherlands
| | - Nils L Visser
- Division of Molecular Oncology and ImmunologyOncode Institute, The Netherlands Cancer InstituteAmsterdamThe Netherlands
| | - Daniel S Peeper
- Division of Molecular Oncology and ImmunologyOncode Institute, The Netherlands Cancer InstituteAmsterdamThe Netherlands
| |
Collapse
|
8
|
Treichel AM, Boeszoermenyi B, Lee CCR, Moss J, Kwiatkowski DJ, Darling TN. Diagnosis of Mosaic Tuberous Sclerosis Complex Using Next-Generation Sequencing of Subtle or Unusual Cutaneous Findings. JID INNOVATIONS 2023; 3:100180. [PMID: 36960317 PMCID: PMC10030254 DOI: 10.1016/j.xjidi.2023.100180] [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: 10/05/2021] [Revised: 11/08/2021] [Accepted: 11/10/2021] [Indexed: 01/11/2023] Open
Abstract
Skin findings can be critical to determining whether a patient with lymphangioleiomyomatosis (LAM), a progressive pulmonary disease that predominantly affects adult women, has sporadic LAM or LAM in association with tuberous sclerosis complex (TSC). Three individuals with LAM underwent evaluation for TSC-associated mucocutaneous and internal findings. We used our previously published algorithm to confirm the clinical suspicion for mosaicism and guide the selection of tissue specimens and genetic workup. Next-generation sequencing of cutaneous findings was used to confirm clinical suspicion for mosaic TSC in individuals with LAM. Two individuals previously thought to have sporadic LAM were diagnosed with mosaic TSC-associated LAM upon next-generation sequencing of unilateral angiofibromas in one and an unusual cutaneous hamartoma in the other. A third individual, diagnosed with TSC in childhood, was found to have a mosaic pathogenic variant in TSC2 in cutaneous tissue from a digit with macrodactyly. Accurate diagnosis of mosaic TSC-associated LAM may require enhanced genetic testing and is important because of the implications regarding surveillance, prognosis, and risk of transmission to offspring.
Collapse
Affiliation(s)
- Alison M. Treichel
- Department of Dermatology, University Hospitals Cleveland Medical Center, Case Western Reserve University, Cleveland, Ohio, USA
| | - Barbara Boeszoermenyi
- Cancer Genetics Laboratory, Division of Pulmonary and Critical Care Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Chyi-Chia Richard Lee
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Joel Moss
- Pulmonary Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - David J. Kwiatkowski
- Cancer Genetics Laboratory, Division of Pulmonary and Critical Care Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Thomas N. Darling
- Department of Dermatology, F. Edward Hebert School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA
| |
Collapse
|
9
|
Fernandes B, Cavaco-Paulo A, Matamá T. A Comprehensive Review of Mammalian Pigmentation: Paving the Way for Innovative Hair Colour-Changing Cosmetics. BIOLOGY 2023; 12:biology12020290. [PMID: 36829566 PMCID: PMC9953601 DOI: 10.3390/biology12020290] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Revised: 01/26/2023] [Accepted: 02/09/2023] [Indexed: 02/15/2023]
Abstract
The natural colour of hair shafts is formed at the bulb of hair follicles, and it is coupled to the hair growth cycle. Three critical processes must happen for efficient pigmentation: (1) melanosome biogenesis in neural crest-derived melanocytes, (2) the biochemical synthesis of melanins (melanogenesis) inside melanosomes, and (3) the transfer of melanin granules to surrounding pre-cortical keratinocytes for their incorporation into nascent hair fibres. All these steps are under complex genetic control. The array of natural hair colour shades are ascribed to polymorphisms in several pigmentary genes. A myriad of factors acting via autocrine, paracrine, and endocrine mechanisms also contributes for hair colour diversity. Given the enormous social and cosmetic importance attributed to hair colour, hair dyeing is today a common practice. Nonetheless, the adverse effects of the long-term usage of such cosmetic procedures demand the development of new methods for colour change. In this context, case reports of hair lightening, darkening and repigmentation as a side-effect of the therapeutic usage of many drugs substantiate the possibility to tune hair colour by interfering with the biology of follicular pigmentary units. By scrutinizing mammalian pigmentation, this review pinpoints key targetable processes for the development of innovative cosmetics that can safely change the hair colour from the inside out.
Collapse
Affiliation(s)
- Bruno Fernandes
- CEB—Centre of Biological Engineering, University of Minho, Campus of Gualtar, 4710-057 Braga, Portugal
| | - Artur Cavaco-Paulo
- CEB—Centre of Biological Engineering, University of Minho, Campus of Gualtar, 4710-057 Braga, Portugal
- LABBELS—Associate Laboratory, 4710-057 Braga, Portugal
- Correspondence: (A.C.-P.); (T.M.); Tel.: +351-253-604-409 (A.C.-P.); +351-253-601-599 (T.M.)
| | - Teresa Matamá
- CEB—Centre of Biological Engineering, University of Minho, Campus of Gualtar, 4710-057 Braga, Portugal
- LABBELS—Associate Laboratory, 4710-057 Braga, Portugal
- Correspondence: (A.C.-P.); (T.M.); Tel.: +351-253-604-409 (A.C.-P.); +351-253-601-599 (T.M.)
| |
Collapse
|
10
|
Shining Light on Autophagy in Skin Pigmentation and Pigmentary Disorders. Cells 2022; 11:cells11192999. [PMID: 36230960 PMCID: PMC9563738 DOI: 10.3390/cells11192999] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 09/13/2022] [Accepted: 09/23/2022] [Indexed: 01/18/2023] Open
Abstract
Autophagy is a vital process for cell survival and it preserves homeostasis by recycling or disassembling unnecessary or dysfunctional cellular constituents. Autophagy ameliorates skin integrity, regulating epidermal differentiation and constitutive pigmentation. It induces melanogenesis and contributes to skin color through melanosome turnover. Autophagy activity is involved in skin phenotypic plasticity and cell function maintenance and, if altered, it concurs to the onset and/or progression of hypopigmentary and hyperpigmentary disorders. Overexpression of autophagy exerts a protective role against the intrinsic metabolic stress occurring in vitiligo skin, while its dysfunction has been linked to the tuberous sclerosis complex hypopigmentation. Again, autophagy impairment reduces melanosome degradation by concurring to pigment accumulation characterizing senile lentigo and melasma. Here we provide an updated review that describes recent findings on the crucial role of autophagy in skin pigmentation, thus revealing the complex interplay among melanocyte biology, skin environment and autophagy. Hence, targeting this process may also represent a promising strategy for treating pigmentary disorders.
Collapse
|
11
|
Wong H, Bowie S, Handisides S, Escardó-Paton J. Case report: tuberous sclerosis and persistent hyperplastic primary vitreous. BMC Ophthalmol 2022; 22:308. [PMID: 35842607 PMCID: PMC9288079 DOI: 10.1186/s12886-022-02526-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2022] [Accepted: 07/07/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Persistent hyperplastic primary vitreous (PHPV) in a patient with tuberous sclerosis (TS) has been described in one previous case report in 1999. Otherwise, there is no literature around this potential association. We describe a case of an infant with TS and PHPV. CASE PRESENTATION An 11-month old male was under investigation for leukocoria, microphthalmia and suspected PHPV after being seen in ophthalmology clinic. He presented to hospital with seizures and was diagnosed with TS on imaging. Imaging also showed the known microphthalmia and a mass associated with the lens. Subsequent paediatric ophthalmology review and examination under anaesthesia confirmed microphthalmia, PHPV and a retrolental mass which was thought to represent total retinal detachment or a retinal hamartoma within a retinal detachment. CONCLUSIONS This is the second case report of PHPV in a patient with TS. The previous case report postulated that the atypical location of the retinal hamartoma was secondary to the abnormal globe development in PHPV.
Collapse
Affiliation(s)
- Hayley Wong
- Radiology Department, Middlemore Hospital, Auckland, New Zealand.
| | - Sarah Bowie
- Radiology Department, Middlemore Hospital, Auckland, New Zealand
| | - Shona Handisides
- Radiology Department, Middlemore Hospital, Auckland, New Zealand
| | - Julia Escardó-Paton
- Ophthalmology Department, Auckland City Hospital and Middlemore Hospital, Auckland, New Zealand
| |
Collapse
|
12
|
Renal organoid modeling of tuberous sclerosis complex reveals lesion features arise from diverse developmental processes. Cell Rep 2022; 40:111048. [PMID: 35793620 DOI: 10.1016/j.celrep.2022.111048] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Revised: 04/15/2022] [Accepted: 06/13/2022] [Indexed: 02/06/2023] Open
Abstract
Tuberous sclerosis complex (TSC) is a multisystem tumor-forming disorder caused by loss of TSC1 or TSC2. Renal manifestations predominately include cysts and angiomyolipomas. Despite a well-described monogenic etiology, the cellular pathogenesis remains elusive. We report a genetically engineered human renal organoid model that recapitulates pleiotropic features of TSC kidney disease in vitro and upon orthotopic xenotransplantation. Time course single-cell RNA sequencing demonstrates that loss of TSC1 or TSC2 affects multiple developmental processes in the renal epithelial, stromal, and glial compartments. First, TSC1 or TSC2 ablation induces transitional upregulation of stromal-associated genes. Second, epithelial cells in the TSC1-/- and TSC2-/- organoids exhibit a rapamycin-insensitive epithelial-to-mesenchymal transition. Third, a melanocytic population forms exclusively in TSC1-/- and TSC2-/- organoids, branching from MITF+ Schwann cell precursors. Together, these results illustrate the pleiotropic developmental consequences of biallelic inactivation of TSC1 or TSC2 and offer insight into TSC kidney lesion pathogenesis.
Collapse
|
13
|
Secco LP, Coubes C, Meyer P, Chenine L, Roubertie A, Malinge MC, Bessis D. Dermatological and genetic data in tuberous sclerosis: A prospective single-center study of 38 patients. Ann Dermatol Venereol 2022; 149:241-244. [PMID: 35527063 DOI: 10.1016/j.annder.2022.02.007] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 11/20/2021] [Accepted: 02/09/2022] [Indexed: 11/15/2022]
Abstract
BACKGROUND Tuberous sclerosis complex (TSC) is a genetic disorder involving the TSC1 or TSC2 gene. Skin signs are prominent, but dermatological data are scarce. This study aims to describe the cutaneous signs of TSC with the genotype. METHODS We studied the dermatological characteristics of 38 patients with TSC at the University Hospital of Montpellier. We collected details of genotypic features. RESULTS All the patients presented at least one cutaneous sign. The dermatological examination alone was sufficient to establish a definite diagnosis of TSC based on the diagnostic criteria for 34/38 patients. No association was found between cutaneous signs and the presence of a TSC1 or TSC2 mutation. We noted skin signs that were poorly described in the disease, namely epidermal nevus in 3 patients, vascular malformation in 2 patients, and keratosis pilaris in 9 patients. DISCUSSION While several studies demonstrate a more severe neurological phenotype in TSC2 mutated patients, skin expression does not appear to differ according to the mutated gene. Further case reports and molecular genetic studies are needed to determine the link between epidermal nevus, vascular malformations, keratosis pilaris and TSC.
Collapse
Affiliation(s)
- L-P Secco
- Department of Dermatology, Montpellier University Hospital Center, 34295 Montpellier, France.
| | - C Coubes
- Department of Genetics, Montpellier University Hospital Center, 34295 Montpellier, France
| | - P Meyer
- Department of Pediatric Neurology, Montpellier University Hospital Center, 34295 Montpellier, France
| | - L Chenine
- Department of Nephrology, Montpellier University Hospital Center, 34295 Montpellier, France
| | - A Roubertie
- Department of Pediatric Neurology, Montpellier University Hospital Center, 34295 Montpellier, France
| | - M-C Malinge
- Department of Genetics, Angers University Hospital Center, 49100 Angers, France
| | - D Bessis
- Department of Dermatology, Montpellier University Hospital Center, 34295 Montpellier, France
| |
Collapse
|
14
|
She Q, Dong Y, Li D, An R, Zhou T, Nie X, Pan R, Deng Y. ABCB6 knockdown suppresses melanogenesis through the GSK3-β/β-catenin signaling axis in human melanoma and melanocyte cell lines. J Dermatol Sci 2022; 106:101-110. [DOI: 10.1016/j.jdermsci.2022.04.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Revised: 04/04/2022] [Accepted: 04/08/2022] [Indexed: 10/18/2022]
|
15
|
[Epithelioid angiomyolipoma of the kidney after successfully treated malignant melanoma]. Urologe A 2021; 61:183-186. [PMID: 34605930 PMCID: PMC8831349 DOI: 10.1007/s00120-021-01662-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/09/2021] [Indexed: 11/25/2022]
Abstract
Hintergrund Die Therapie der epitheloiden Angiomyolipome (eAML) kann eine Herausforderung darstellen, da bei dieser sehr seltenen Unterform der gutartigen mesenchymalen Angiomyolipome anders als bei den klassischen Angiomyolipomen bei bis zu 30 % der Fälle Lymphknotenmetastasen, lokale Rezidive und Fernmetastasen auftreten. Ziel der Arbeit Wir berichten hier nach unserer Recherche erstmals in Deutschland von einem Fall von eAML nach stattgehabtem malignem Melanom. Material und Methoden Neben der Klinik und Histologie wird die genetische Untersuchung des Tumorgewebes dargestellt. Ergebnisse Es fand sich eine somatische, trunkierende Mutation des TSC2-Gens („tuberous sclerosis complex“) im Angiomyolipom. Schlussfolgerung Die Beziehung zu verwandten Tumorentitäten in der histologischen Diagnostik wird dargestellt und eine mögliche Rolle der genetischen Diagnostik für die Therapieplanung diskutiert.
Collapse
|
16
|
Carmignac V, Mignot C, Blanchard E, Kuentz P, Aubriot-Lorton MH, Parker VER, Sorlin A, Fraitag S, Courcet JB, Duffourd Y, Rodriguez D, Knox RG, Polubothu S, Boland A, Olaso R, Delepine M, Darmency V, Riachi M, Quelin C, Rollier P, Goujon L, Grotto S, Capri Y, Jacquemont ML, Odent S, Amram D, Chevarin M, Vincent-Delorme C, Catteau B, Guibaud L, Arzimanoglou A, Keddar M, Sarret C, Callier P, Bessis D, Geneviève D, Deleuze JF, Thauvin C, Semple RK, Philippe C, Rivière JB, Kinsler VA, Faivre L, Vabres P. Clinical spectrum of MTOR-related hypomelanosis of Ito with neurodevelopmental abnormalities. Genet Med 2021; 23:1484-1491. [PMID: 33833411 PMCID: PMC8354853 DOI: 10.1038/s41436-021-01161-6] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Revised: 03/16/2021] [Accepted: 03/16/2021] [Indexed: 11/13/2022] Open
Abstract
PURPOSE Hypomelanosis of Ito (HI) is a skin marker of somatic mosaicism. Mosaic MTOR pathogenic variants have been reported in HI with brain overgrowth. We sought to delineate further the pigmentary skin phenotype and clinical spectrum of neurodevelopmental manifestations of MTOR-related HI. METHODS From two cohorts totaling 71 patients with pigmentary mosaicism, we identified 14 patients with Blaschko-linear and one with flag-like pigmentation abnormalities, psychomotor impairment or seizures, and a postzygotic MTOR variant in skin. Patient records, including brain magnetic resonance image (MRI) were reviewed. Immunostaining (n = 3) for melanocyte markers and ultrastructural studies (n = 2) were performed on skin biopsies. RESULTS MTOR variants were present in skin, but absent from blood in half of cases. In a patient (p.[Glu2419Lys] variant), phosphorylation of p70S6K was constitutively increased. In hypopigmented skin of two patients, we found a decrease in stage 4 melanosomes in melanocytes and keratinocytes. Most patients (80%) had macrocephaly or (hemi)megalencephaly on MRI. CONCLUSION MTOR-related HI is a recognizable neurocutaneous phenotype of patterned dyspigmentation, epilepsy, intellectual deficiency, and brain overgrowth, and a distinct subtype of hypomelanosis related to somatic mosaicism. Hypopigmentation may be due to a defect in melanogenesis, through mTORC1 activation, similar to hypochromic patches in tuberous sclerosis complex.
Collapse
Affiliation(s)
- Virginie Carmignac
- INSERM UMR1231, Bourgogne Franche-Comté University, Dijon, France.
- MAGEC-Mosaïque Reference Center, Dijon University Hospital, Dijon, France.
| | - Cyril Mignot
- Neuropaediatrics and Development Pathology Department, Trousseau Hospital, AP-HP, Paris, France
- Genetics Department and Reference Center for rare causes of Intellectual Disability, Pitié-Salpêtrière hospital, AP-HP, Paris, France
| | - Emmanuelle Blanchard
- Plateforme IBiSA de Microscopie Electronique, Anatomie et cytologie pathologique, Université et CHRU de Tours, Tours, France
- INSERM U1259 MAVIVH, Université et CHRU de Tours, Tours, France
| | - Paul Kuentz
- INSERM UMR1231, Bourgogne Franche-Comté University, Dijon, France
- Fédération Hospitalo-Universitaire Médecine Translationnelle et Anomalies du Développement (TRANSLAD), Dijon-Burgundy University Hospital, Dijon, France
| | | | - Victoria E R Parker
- The University of Cambridge Metabolic Research Laboratories, Institute of Metabolic Science, Cambridge, UK
| | - Arthur Sorlin
- INSERM UMR1231, Bourgogne Franche-Comté University, Dijon, France
- Fédération Hospitalo-Universitaire Médecine Translationnelle et Anomalies du Développement (TRANSLAD), Dijon-Burgundy University Hospital, Dijon, France
- Pediatrics and Medical Genetics Department, Dijon-Bourgogne University Hospital, Dijon, France
| | - Sylvie Fraitag
- Service d'Anatomie et Cytologie Pathologique, Necker-Enfants Malades Hospital, Paris, France
| | - Jean-Benoît Courcet
- INSERM UMR1231, Bourgogne Franche-Comté University, Dijon, France
- Fédération Hospitalo-Universitaire Médecine Translationnelle et Anomalies du Développement (TRANSLAD), Dijon-Burgundy University Hospital, Dijon, France
- Pediatrics and Medical Genetics Department, Dijon-Bourgogne University Hospital, Dijon, France
| | - Yannis Duffourd
- INSERM UMR1231, Bourgogne Franche-Comté University, Dijon, France
- Fédération Hospitalo-Universitaire Médecine Translationnelle et Anomalies du Développement (TRANSLAD), Dijon-Burgundy University Hospital, Dijon, France
| | - Diana Rodriguez
- Genetics Department and Reference Center for rare causes of Intellectual Disability, Pitié-Salpêtrière hospital, AP-HP, Paris, France
| | - Rachel G Knox
- The University of Cambridge Metabolic Research Laboratories, Institute of Metabolic Science, Cambridge, UK
| | - Satyamaanasa Polubothu
- Paediatric Dermatology, Great Ormond St Hospital for Children NHS Foundation Trust, London, UK
- UCL GOS Institute of Child Health, London, UK
- Mosaicism and Precision Medicine laboratory, Francis Crick Institute, London, UK
| | - Anne Boland
- National Genotyping Center, Genomic Institute, CEA, Evry, France
| | - Robert Olaso
- National Genotyping Center, Genomic Institute, CEA, Evry, France
| | - Marc Delepine
- National Genotyping Center, Genomic Institute, CEA, Evry, France
| | - Véronique Darmency
- Pediatrics and Medical Genetics Department, Dijon-Bourgogne University Hospital, Dijon, France
| | - Melissa Riachi
- UCL GOS Institute of Child Health, London, UK
- Mosaicism and Precision Medicine laboratory, Francis Crick Institute, London, UK
| | - Chloé Quelin
- Clinical Genetics department, Rennes University Hospital, Rennes, France
| | - Paul Rollier
- Clinical Genetics department, Rennes University Hospital, Rennes, France
| | - Louise Goujon
- Clinical Genetics department, Rennes University Hospital, Rennes, France
| | - Sarah Grotto
- Genetics Department, AP-HP, Robert-Debré University Hospital, Paris, France
| | - Yline Capri
- Genetics Department, AP-HP, Robert-Debré University Hospital, Paris, France
| | | | - Sylvie Odent
- Clinical Genetics department, Rennes University Hospital, Rennes, France
| | - Daniel Amram
- Clinical Genetics Department, Créteil Hospital, Créteil, France
| | - Martin Chevarin
- INSERM UMR1231, Bourgogne Franche-Comté University, Dijon, France
- Unité Fonctionnelle Innovation en Diagnostic Génomique des Maladies Rares, FHU-TRANSLAD, CHU Dijon Bourgogne University Hospital, Dijon, France
| | | | - Benoît Catteau
- Dermatology department, Lille University Hospital, Lille, France
| | - Laurent Guibaud
- Pediatric and Fetal Imaging Department, Hospices Civils de Lyon, Bron, France
| | - Alexis Arzimanoglou
- Department of Paediatric Clinical Epileptology, Sleep Disorders and Functional Neurology, University Hospitals of Lyon (HCL), Lyon, France
- Brain Dynamics and Cognition (DYCOG) Team, Lyon Neuroscience Research Centre, Lyon, France
| | - Malika Keddar
- Cytogenetics Department, Dijon University Hospital, Dijon, France
| | - Catherine Sarret
- Medical genetics department, Pôle Femme et Enfant, Clermont-Ferrand University Hospital-Hôpital d'Estaing, Clermont-Ferrand, France
| | - Patrick Callier
- INSERM UMR1231, Bourgogne Franche-Comté University, Dijon, France
- Fédération Hospitalo-Universitaire Médecine Translationnelle et Anomalies du Développement (TRANSLAD), Dijon-Burgundy University Hospital, Dijon, France
- Cytogenetics Department, Dijon University Hospital, Dijon, France
| | - Didier Bessis
- Dermatology Department, Montpellier University Hospital, Montpellier, France
| | - David Geneviève
- Medical Genetics Department, Montpellier University Hospital, Montpellier, France
| | | | - Christel Thauvin
- INSERM UMR1231, Bourgogne Franche-Comté University, Dijon, France
- Fédération Hospitalo-Universitaire Médecine Translationnelle et Anomalies du Développement (TRANSLAD), Dijon-Burgundy University Hospital, Dijon, France
- Centre de Référence Déficiences Intellectuelles de Causes Rares, Hôpital d'Enfants, Dijon, France
| | - Robert K Semple
- The University of Cambridge Metabolic Research Laboratories, Institute of Metabolic Science, Cambridge, UK
- Center for Cardiovascular Science, University of Edinburgh, Edinburgh, UK
| | | | - Jean-Baptiste Rivière
- INSERM UMR1231, Bourgogne Franche-Comté University, Dijon, France
- Fédération Hospitalo-Universitaire Médecine Translationnelle et Anomalies du Développement (TRANSLAD), Dijon-Burgundy University Hospital, Dijon, France
| | - Veronica A Kinsler
- Paediatric Dermatology, Great Ormond St Hospital for Children NHS Foundation Trust, London, UK
- UCL GOS Institute of Child Health, London, UK
- Mosaicism and Precision Medicine laboratory, Francis Crick Institute, London, UK
| | - Laurence Faivre
- INSERM UMR1231, Bourgogne Franche-Comté University, Dijon, France
- Fédération Hospitalo-Universitaire Médecine Translationnelle et Anomalies du Développement (TRANSLAD), Dijon-Burgundy University Hospital, Dijon, France
- Centre de Référence Anomalies du Développement et Syndromes Malformatifs, Hôpital d'Enfants, Dijon, France
| | - Pierre Vabres
- INSERM UMR1231, Bourgogne Franche-Comté University, Dijon, France
- MAGEC-Mosaïque Reference Center, Dijon University Hospital, Dijon, France
- Fédération Hospitalo-Universitaire Médecine Translationnelle et Anomalies du Développement (TRANSLAD), Dijon-Burgundy University Hospital, Dijon, France
| |
Collapse
|
17
|
Evaluation of a resorufin-based fluorescent probe for tyrosinase detection in skin pigmentation disorders. Biodes Manuf 2021. [DOI: 10.1007/s42242-021-00138-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
18
|
de la Calle Arregui C, Plata-Gómez AB, Deleyto-Seldas N, García F, Ortega-Molina A, Abril-Garrido J, Rodriguez E, Nemazanyy I, Tribouillard L, de Martino A, Caleiras E, Campos-Olivas R, Mulero F, Laplante M, Muñoz J, Pende M, Sabio G, Sabatini DM, Efeyan A. Limited survival and impaired hepatic fasting metabolism in mice with constitutive Rag GTPase signaling. Nat Commun 2021; 12:3660. [PMID: 34135321 PMCID: PMC8209044 DOI: 10.1038/s41467-021-23857-8] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Accepted: 05/17/2021] [Indexed: 12/16/2022] Open
Abstract
The mechanistic target of rapamycin complex 1 (mTORC1) integrates cellular nutrient signaling and hormonal cues to control metabolism. We have previously shown that constitutive nutrient signaling to mTORC1 by means of genetic activation of RagA (expression of GTP-locked RagA, or RagAGTP) in mice resulted in a fatal energetic crisis at birth. Herein, we rescue neonatal lethality in RagAGTP mice and find morphometric and metabolic alterations that span glucose, lipid, ketone, bile acid and amino acid homeostasis in adults, and a median lifespan of nine months. Proteomic and metabolomic analyses of livers from RagAGTP mice reveal a failed metabolic adaptation to fasting due to a global impairment in PPARα transcriptional program. These metabolic defects are partially recapitulated by restricting activation of RagA to hepatocytes, and revert by pharmacological inhibition of mTORC1. Constitutive hepatic nutrient signaling does not cause hepatocellular damage and carcinomas, unlike genetic activation of growth factor signaling upstream of mTORC1. In summary, RagA signaling dictates dynamic responses to feeding-fasting cycles to tune metabolism so as to match the nutritional state.
Collapse
Affiliation(s)
- Celia de la Calle Arregui
- Metabolism and Cell Signaling Laboratory, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Ana Belén Plata-Gómez
- Metabolism and Cell Signaling Laboratory, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Nerea Deleyto-Seldas
- Metabolism and Cell Signaling Laboratory, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Fernando García
- Proteomics Unit, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Ana Ortega-Molina
- Metabolism and Cell Signaling Laboratory, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Julio Abril-Garrido
- Metabolism and Cell Signaling Laboratory, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Elena Rodriguez
- Myocardial Pathophysiology, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Ivan Nemazanyy
- Platform for Metabolic Analyses, Structure Fédérative de Recherche Necker, INSERM US24/CNRS UMS 3633, Paris, France
| | - Laura Tribouillard
- Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec (CRIUCPQ), Faculté de Médecine, Université Laval, Québec, QC, Canada
- Centre de recherche sur le cancer de l'Université Laval, Université Laval, Québec, QC, Canada
| | - Alba de Martino
- Histopathology Unit. Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Eduardo Caleiras
- Histopathology Unit. Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Ramón Campos-Olivas
- Spectroscopy and NMR Unit, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Francisca Mulero
- Molecular Imaging Unit, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Mathieu Laplante
- Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec (CRIUCPQ), Faculté de Médecine, Université Laval, Québec, QC, Canada
- Centre de recherche sur le cancer de l'Université Laval, Université Laval, Québec, QC, Canada
| | - Javier Muñoz
- Proteomics Unit, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Mario Pende
- Institut Necker Enfants Malades, INSERM U1151, Université de Paris, Paris, France
| | - Guadalupe Sabio
- Myocardial Pathophysiology, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - David M Sabatini
- Whitehead Institute for Biomedical Research, Nine Cambridge Center, Cambridge, MA, USA
- Department of Biology, Massachusetts Institute of Technology (MIT), Cambridge, MA, USA
- David H. Koch Institute for Integrative Cancer Research at MIT, Cambridge, MA, USA
- Broad Institute, Seven Cambridge Center, Cambridge, MA, USA
- Howard Hughes Medical Institute, MIT, Cambridge, MA, USA
| | - Alejo Efeyan
- Metabolism and Cell Signaling Laboratory, Spanish National Cancer Research Centre (CNIO), Madrid, Spain.
- Whitehead Institute for Biomedical Research, Nine Cambridge Center, Cambridge, MA, USA.
- Department of Biology, Massachusetts Institute of Technology (MIT), Cambridge, MA, USA.
| |
Collapse
|
19
|
Dong Z, Dai H, Gao Y, Feng Z, Liu W, Liu F, Zhang Z, Ma F, Xie X, Zhu Z, Liu W, Liu B. Inhibition of the Wnt/β-catenin signaling pathway reduces autophagy levels in complement treated podocytes. Exp Ther Med 2021; 22:737. [PMID: 34055054 PMCID: PMC8138266 DOI: 10.3892/etm.2021.10169] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2020] [Accepted: 01/25/2021] [Indexed: 01/12/2023] Open
Abstract
In idiopathic membranous nephropathy, the complement membrane attack complex, more commonly referred to as complement 5b-9 (C5b-9), induces glomerular epithelial cell injury and proteinuria. C5b-9 can also activate numerous mechanisms that restrict or facilitate injury. Recent studies suggest that autophagy and the canonical Wnt signaling pathway serve an important role in repairing podocyte injury. However, the effect of C5b-9 on these pathways and the relationship between them remains unclear. The aim of the present study was to show the effect of C5b-9 on the Wnt/β-catenin signaling pathway and autophagy in podocytes in vitro. Levels of relevant indicators were detected by immunofluorescence staining and capillary western immunoassay. C5b-9 serum significantly activated the Wnt/β-catenin signaling pathway and promoted autophagy. Treatment with Dickkopf-related protein 1 (DKK1), a Wnt/β-catenin pathway blocker, protected podocytes from injury and significantly inhibited autophagy. The results indicated that inhibition of the Wnt/β-catenin pathway physiologically activated autophagy. The results indicated that C5b-9 resulted in a decrease in Akt in podocytes. However, the podocytes preincubated with DKK1 and then attacked by C5b-9 showed an increase in Akt levels. This may explain the observation that blocking the Wnt/β-catenin signaling pathway attenuated C5b-9 podocyte damage, while inhibiting autophagy. The results of the present study also suggest that regulation of these two pathways may serve as a novel method for the treatment of idiopathic membranous nephropathy.
Collapse
Affiliation(s)
- Zhaocheng Dong
- Beijing University of Chinese Medicine, Chaoyang, Beijing 100029, P.R. China.,Beijing Hospital of Traditional Chinese Medicine Affiliated to Capital Medical University, Dongcheng, Beijing 100010, P.R. China
| | - Haoran Dai
- Shunyi Branch, Beijing Traditional Chinese Medicine Hospital, Shunyi, Beijing 101300, P.R. China
| | - Yu Gao
- Beijing Hospital of Traditional Chinese Medicine Affiliated to Capital Medical University, Dongcheng, Beijing 100010, P.R. China.,Capital Medical University, Fengtai, Beijing 100069, P.R. China
| | - Zhendong Feng
- Beijing Chinese Medicine Hospital, Pinggu Hospital, Pinggu, Beijing 101200, P.R. China
| | - Wenbin Liu
- Beijing Hospital of Traditional Chinese Medicine Affiliated to Capital Medical University, Dongcheng, Beijing 100010, P.R. China.,Capital Medical University, Fengtai, Beijing 100069, P.R. China
| | - Fei Liu
- Beijing University of Chinese Medicine, Chaoyang, Beijing 100029, P.R. China.,Beijing Hospital of Traditional Chinese Medicine Affiliated to Capital Medical University, Dongcheng, Beijing 100010, P.R. China
| | - Zihan Zhang
- Beijing University of Chinese Medicine, Chaoyang, Beijing 100029, P.R. China.,Beijing Hospital of Traditional Chinese Medicine Affiliated to Capital Medical University, Dongcheng, Beijing 100010, P.R. China
| | - Fang Ma
- China Academy of Traditional Chinese Medicine, Guanganmen Hospital, Xicheng, Beijing 100053, P.R. China
| | - Xinran Xie
- Beijing Institute of Traditional Chinese Medicine, Beijing Hospital of Traditional Chinese Medicine Affiliated to Capital Medical University, Dongcheng, Beijing 100010, P.R. China
| | - Zebing Zhu
- Beijing University of Chinese Medicine, Chaoyang, Beijing 100029, P.R. China.,Key Laboratory of Chinese Internal Medicine of the Ministry of Education, Beijing University of Chinese Medicine Affiliated to Dongzhimen Hospital, Dongchen, Beijing 100700, P.R. China
| | - Weijing Liu
- Key Laboratory of Chinese Internal Medicine of the Ministry of Education, Beijing University of Chinese Medicine Affiliated to Dongzhimen Hospital, Dongchen, Beijing 100700, P.R. China
| | - Baoli Liu
- Beijing Hospital of Traditional Chinese Medicine Affiliated to Capital Medical University, Dongcheng, Beijing 100010, P.R. China
| |
Collapse
|
20
|
Treichel AM, Pithadia DJ, Lee CCR, Oyerinde O, Moss J, Darling TN. Histopathological features of fibrous cephalic plaques in tuberous sclerosis complex. Histopathology 2021; 79:619-628. [PMID: 33882161 DOI: 10.1111/his.14392] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2021] [Revised: 04/15/2021] [Accepted: 04/17/2021] [Indexed: 02/03/2023]
Abstract
AIMS Fibrous cephalic plaques (FCPs) in individuals with tuberous sclerosis complex (TSC) may be excised for cosmetic reasons or biopsied to confirm lesion identification and TSC diagnosis. The aim of this study was to determine the range of histopathological features of FCPs. METHODS AND RESULTS A retrospective analysis was conducted on 119 adults with TSC. Twenty-one lesions from 16 individuals were evaluated by a dermatopathologist. Additionally, we assessed whether lesion colour or histology varied by anatomical location. Seventy-six lesions were observed in 36 of 119 individuals. Erythematous lesions were more commonly found on the forehead, face or neck than on the scalp (odds ratio = 12.6, P = 0.0001). Thickened and disorganised collagen fibre bundles were present in 95% (20/21) of lesions. Perifollicular fibrosis was observed in 95% (20/21) of lesions, enhanced vascularity was observed in 52% (11/21) of lesions, and features of fibrofolliculoma were observed in 43% (9/21) of lesions. Other abnormalities included features similar to trichofolliculoma, follicular-derived, infundibular-type cysts, and abnormally arranged primitive hair follicles. CONCLUSIONS FCPs in TSC show thickened bundles of collagen, and hamartomatous changes involving hair follicles. Recognition of these histopathological features may raise the possibility of unsuspected TSC or confirm FCP identification.
Collapse
Affiliation(s)
- Alison M Treichel
- Pulmonary Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA.,Department of Dermatology, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Deeti J Pithadia
- Pulmonary Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA.,Department of Dermatology, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Chyi-Chia R Lee
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Oyetewa Oyerinde
- Pulmonary Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA.,Department of Dermatology, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Joel Moss
- Pulmonary Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Thomas N Darling
- Department of Dermatology, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| |
Collapse
|
21
|
Lee AY. Skin Pigmentation Abnormalities and Their Possible Relationship with Skin Aging. Int J Mol Sci 2021; 22:ijms22073727. [PMID: 33918445 PMCID: PMC8038212 DOI: 10.3390/ijms22073727] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Revised: 03/24/2021] [Accepted: 04/01/2021] [Indexed: 12/13/2022] Open
Abstract
Skin disorders showing abnormal pigmentation are often difficult to manage because of their uncertain etiology or pathogenesis. Abnormal pigmentation is a common symptom accompanying aging skin. The association between skin aging and skin pigmentation abnormalities can be attributed to certain inherited disorders characterized by premature aging and abnormal pigmentation in the skin and some therapeutic modalities effective for both. Several molecular mechanisms, including oxidative stress, mitochondrial DNA mutations, DNA damage, telomere shortening, hormonal changes, and autophagy impairment, have been identified as involved in skin aging. Although each of these skin aging-related mechanisms are interconnected, this review examined the role of each mechanism in skin hyperpigmentation or hypopigmentation to propose the possible association between skin aging and pigmentation abnormalities.
Collapse
Affiliation(s)
- Ai-Young Lee
- Department of Dermatology, College of Medicine, Dongguk University Ilsan Hospital, 814 Siksa-dong, Ilsandong-gu, Goyang-si 410-773, Gyeonggi-do, Korea
| |
Collapse
|
22
|
Evans JF, Obraztsova K, Lin SM, Krymskaya VP. CrossTORC and WNTegration in Disease: Focus on Lymphangioleiomyomatosis. Int J Mol Sci 2021; 22:ijms22052233. [PMID: 33668092 PMCID: PMC7956553 DOI: 10.3390/ijms22052233] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Revised: 02/18/2021] [Accepted: 02/21/2021] [Indexed: 02/07/2023] Open
Abstract
The mechanistic target of rapamycin (mTOR) and wingless-related integration site (Wnt) signal transduction networks are evolutionarily conserved mammalian growth and cellular development networks. Most cells express many of the proteins in both pathways, and this review will briefly describe only the key proteins and their intra- and extracellular crosstalk. These complex interactions will be discussed in relation to cancer development, drug resistance, and stem cell exhaustion. This review will also highlight the tumor-suppressive tuberous sclerosis complex (TSC) mutated, mTOR-hyperactive lung disease of women, lymphangioleiomyomatosis (LAM). We will summarize recent advances in the targeting of these pathways by monotherapy or combination therapy, as well as future potential treatments.
Collapse
|
23
|
Ferreira LDS, Calderipe CB, Maass JB, Carrard VC, Martins MD, Abreu LG, Schuch LF, Uchoa Vasconcelos AC. Oral pigmented lesions in syndromic individuals: A systematic review. Oral Dis 2021; 28:531-540. [PMID: 33394507 DOI: 10.1111/odi.13769] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Revised: 12/07/2020] [Accepted: 12/29/2020] [Indexed: 01/15/2023]
Abstract
OBJECTIVE To systematically integrate the available data published in the literature on oral pigmented lesions (OPL) associated with syndromes, summarizing the clinical and demographic features of the individuals. MATERIALS AND METHODS An electronic search was undertaken in six databases. Eligibility criteria were articles in English, Spanish, and Portuguese describing case reports or case series of OPL associated with syndromes. Data were aggregated and statistically evaluated. RESULTS About 108 articles reporting 149 cases of individuals with syndromes were identified. Among the affected individuals, nine syndromes were reported. The mean age at diagnosis was 35.93 years (0.41 to 83 years), with a predilection for white (n = 85/85.86%) female (n = 102/68.46%) individuals. As regards the number of lesions, 109 (73.15%) were multiple and 40 (26.85%) were single. Lip represented the anatomical location more affected (122 cases/38.01%), followed by the buccal mucosa (100 cases/31.15%). Brownish lesions accounted for 82 (69.49%) cases. The mean time of evolution was 10.52 years (0.16 to 56 years). OPL preceding diagnosis of the syndrome was observed in 111 (74.50%) cases. CONCLUSIONS Although these syndromes are uncommon, dentists should be able to recognize their manifestations, since oral manifestations can represent an important aspect in early diagnosis.
Collapse
Affiliation(s)
- Luíse Dos Santos Ferreira
- Diagnostic Center for Oral Diseases, School of Dentistry, Universidade Federal de Pelotas, Pelotas, Brazil
| | - Camila Barcellos Calderipe
- Diagnostic Center for Oral Diseases, School of Dentistry, Universidade Federal de Pelotas, Pelotas, Brazil
| | - Julianne Bartz Maass
- Diagnostic Center for Oral Diseases, School of Dentistry, Universidade Federal de Pelotas, Pelotas, Brazil
| | - Vinicius Coelho Carrard
- Department of Oral Pathology, School of Dentistry, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Manoela Domingues Martins
- Department of Oral Pathology, School of Dentistry, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil.,Department of Oral Diagnosis, Piracicaba Dental School, Universidade de Campinas, Piracicaba, Brazil
| | - Lucas Guimarães Abreu
- Department of Child's and Adolescent's Oral Health, School of Dentistry, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Lauren Frenzel Schuch
- Department of Oral Diagnosis, Piracicaba Dental School, Universidade de Campinas, Piracicaba, Brazil
| | | |
Collapse
|
24
|
Koguchi-Yoshioka H, Tanemura A, Katayama I, Fujimoto M, Wataya-Kaneda M. Hypohidrosis in the macules in tuberous sclerosis complex and neurofibromatosis. J Dermatol 2020; 48:418-419. [PMID: 33368630 DOI: 10.1111/1346-8138.15708] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Revised: 10/26/2020] [Accepted: 10/31/2020] [Indexed: 11/30/2022]
Affiliation(s)
- Hanako Koguchi-Yoshioka
- Department of Dermatology, Course of Integrated Medicine, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Atsushi Tanemura
- Department of Dermatology, Course of Integrated Medicine, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Ichiro Katayama
- Department of Dermatology, Course of Integrated Medicine, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Manabu Fujimoto
- Department of Dermatology, Course of Integrated Medicine, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Mari Wataya-Kaneda
- Department of Dermatology, Course of Integrated Medicine, Graduate School of Medicine, Osaka University, Osaka, Japan
| |
Collapse
|
25
|
Li X, Hu T, Liu J, Fang B, Geng X, Xiong Q, Zhang L, Jin Y, Liu X, Li L, Wang Y, Li R, Bai X, Yang H, Dai Y. A Bama miniature pig model of monoallelic TSC1 mutation for human tuberous sclerosis complex. J Genet Genomics 2020; 47:735-742. [PMID: 33612456 DOI: 10.1016/j.jgg.2020.11.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Revised: 11/09/2020] [Accepted: 11/20/2020] [Indexed: 01/18/2023]
Abstract
Tuberous sclerosis complex (TSC) is a dominant genetic neurocutaneous syndrome characterized by multiple organ hamartomas. Although rodent models bearing a germline mutation in either TSC1 or TSC2 gene have been generated, they do not develop pathogenic lesions matching those seen in patients with TSC because of the significant differences between mice and humans, highlighting the need for an improved large animal model of TSC. Here, we successfully generate monoallelic TSC1-modified Bama miniature pigs using the CRISPR/Cas9 system along with somatic cell nuclear transfer (SCNT) technology. The expression of phosphorylated target ribosomal protein S6 is significantly enhanced in the piglets, indicating that disruption of a TSC1 allele activate the mechanistic target of rapamycin (mTOR) signaling pathway. Notably, differing from the mouse TSC models reported previously, the TSC1+/- Bama miniature pig developed cardiac rhabdomyoma and subependymal nodules, resembling the major clinical features that occur in patients with TSC. These TSC1+/- Bama miniature pigs could serve as valuable large animal models for further elucidation of the pathogenesis of TSC and the development of therapeutic strategies for TSC disease.
Collapse
Affiliation(s)
- Xiaoxue Li
- Jiangsu Key Laboratory of Xenotransplantation, Nanjing Medical University, Nanjing 211166, China
| | - Tingdong Hu
- Jiangsu Key Laboratory of Xenotransplantation, Nanjing Medical University, Nanjing 211166, China
| | - Jiying Liu
- Jiangsu Key Laboratory of Xenotransplantation, Nanjing Medical University, Nanjing 211166, China
| | - Bin Fang
- Jiangsu Key Laboratory of Xenotransplantation, Nanjing Medical University, Nanjing 211166, China
| | - Xue Geng
- Jiangsu Key Laboratory of Xenotransplantation, Nanjing Medical University, Nanjing 211166, China
| | - Qiang Xiong
- Jiangsu Key Laboratory of Xenotransplantation, Nanjing Medical University, Nanjing 211166, China
| | - Lining Zhang
- Jiangsu Key Laboratory of Xenotransplantation, Nanjing Medical University, Nanjing 211166, China
| | - Yong Jin
- Jiangsu Key Laboratory of Xenotransplantation, Nanjing Medical University, Nanjing 211166, China
| | - Xiaorui Liu
- Jiangsu Key Laboratory of Xenotransplantation, Nanjing Medical University, Nanjing 211166, China
| | - Lin Li
- Jiangsu Key Laboratory of Xenotransplantation, Nanjing Medical University, Nanjing 211166, China; Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing 211166, China
| | - Ying Wang
- Jiangsu Key Laboratory of Xenotransplantation, Nanjing Medical University, Nanjing 211166, China; Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing 211166, China
| | - Rongfeng Li
- Jiangsu Key Laboratory of Xenotransplantation, Nanjing Medical University, Nanjing 211166, China; Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing 211166, China
| | - Xiaochun Bai
- Department of Cell Biology, Guangdong Provincial Key Laboratory of Bone and Joint Degenerative Diseases, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Haiyuan Yang
- Jiangsu Key Laboratory of Xenotransplantation, Nanjing Medical University, Nanjing 211166, China; Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing 211166, China.
| | - Yifan Dai
- Jiangsu Key Laboratory of Xenotransplantation, Nanjing Medical University, Nanjing 211166, China; Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing 211166, China; State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing 211166, China; Shenzhen Xenotransplantation Medical Engineering Research and Development Center, Institute of Translational Medicine, Shenzhen Second People's Hospital, First Affiliated Hospital of Shenzhen University, Shenzhen 518035, China.
| |
Collapse
|
26
|
Matozaki T, Kotani T, Murata Y, Saito Y. Roles of Src family kinase, Ras, and mTOR signaling in intestinal epithelial homeostasis and tumorigenesis. Cancer Sci 2020; 112:16-21. [PMID: 33073467 PMCID: PMC7780047 DOI: 10.1111/cas.14702] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Revised: 10/12/2020] [Accepted: 10/14/2020] [Indexed: 12/15/2022] Open
Abstract
The turnover of intestinal epithelial cells (IECs) is relatively rapid (3-5 days in mouse and human), and this short existence and other aspects of the homeostasis of IECs are tightly regulated by various signaling pathways including Wnt-β-catenin signaling. Dysregulation of IEC homeostasis likely contributes to the development of intestinal inflammation and intestinal cancer. The roles of receptor protein tyrosine kinases and their downstream signaling molecules such as Src family kinases, Ras, and mTOR in homeostatic regulation of IEC turnover have recently been evaluated. These signaling pathways have been found to promote not only the proliferation of IECs but also the differentiation of progenitor cells into secretory cell types such as goblet cells. Of note, signaling by Src family kinases, Ras, and mTOR has been shown to oppose the Wnt-β-catenin signaling pathway and thereby to limit the number of Lgr5+ intestinal stem cells or of Paneth cells. Such cross-talk of signaling pathways is important not only for proper regulation of IEC homeostasis but for the development of intestinal tumors and potentially for anticancer therapy.
Collapse
Affiliation(s)
- Takashi Matozaki
- Division of Molecular and Cellular Signaling, Department of Biochemistry and Molecular Biology, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Takenori Kotani
- Division of Molecular and Cellular Signaling, Department of Biochemistry and Molecular Biology, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Yoji Murata
- Division of Molecular and Cellular Signaling, Department of Biochemistry and Molecular Biology, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Yasuyuki Saito
- Division of Molecular and Cellular Signaling, Department of Biochemistry and Molecular Biology, Kobe University Graduate School of Medicine, Kobe, Japan
| |
Collapse
|
27
|
Li G, Wang M, Caulk AW, Cilfone NA, Gujja S, Qin L, Chen PY, Chen Z, Yousef S, Jiao Y, He C, Jiang B, Korneva A, Bersi MR, Wang G, Liu X, Mehta S, Geirsson A, Gulcher JR, Chittenden TW, Simons M, Humphrey JD, Tellides G. Chronic mTOR activation induces a degradative smooth muscle cell phenotype. J Clin Invest 2020; 130:1233-1251. [PMID: 32039915 DOI: 10.1172/jci131048] [Citation(s) in RCA: 67] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2019] [Accepted: 12/03/2019] [Indexed: 01/01/2023] Open
Abstract
Smooth muscle cell (SMC) proliferation has been thought to limit the progression of thoracic aortic aneurysm and dissection (TAAD) because loss of medial cells associates with advanced disease. We investigated effects of SMC proliferation in the aortic media by conditional disruption of Tsc1, which hyperactivates mTOR complex 1. Consequent SMC hyperplasia led to progressive medial degeneration and TAAD. In addition to diminished contractile and synthetic functions, fate-mapped SMCs displayed increased proteolysis, endocytosis, phagocytosis, and lysosomal clearance of extracellular matrix and apoptotic cells. SMCs acquired a limited repertoire of macrophage markers and functions via biogenesis of degradative organelles through an mTOR/β-catenin/MITF-dependent pathway, but were distinguishable from conventional macrophages by an absence of hematopoietic lineage markers and certain immune effectors even in the context of hyperlipidemia. Similar mTOR activation and induction of a degradative SMC phenotype in a model of mild TAAD due to Fbn1 mutation greatly worsened disease with near-uniform lethality. The finding of increased lysosomal markers in medial SMCs from clinical TAAD specimens with hyperplasia and matrix degradation further supports the concept that proliferation of degradative SMCs within the media causes aortic disease, thus identifying mTOR-dependent phenotypic modulation as a therapeutic target for combating TAAD.
Collapse
Affiliation(s)
- Guangxin Li
- Department of Surgery, Yale School of Medicine, New Haven, Connecticut, USA.,Department of Breast and Thyroid Surgery, Peking University Shenzhen Hospital, Shenzhen, Guangdong Province, China
| | - Mo Wang
- Department of Surgery, Yale School of Medicine, New Haven, Connecticut, USA
| | - Alexander W Caulk
- Department of Biomedical Engineering, Yale School of Engineering and Applied Science, New Haven, Connecticut, USA
| | - Nicholas A Cilfone
- Computational Statistics and Bioinformatics Group, Advanced Artificial Intelligence Research Laboratory, WuXi NextCODE, Cambridge, Massachusetts, USA
| | - Sharvari Gujja
- Computational Statistics and Bioinformatics Group, Advanced Artificial Intelligence Research Laboratory, WuXi NextCODE, Cambridge, Massachusetts, USA
| | - Lingfeng Qin
- Department of Surgery, Yale School of Medicine, New Haven, Connecticut, USA
| | | | - Zehua Chen
- Computational Statistics and Bioinformatics Group, Advanced Artificial Intelligence Research Laboratory, WuXi NextCODE, Cambridge, Massachusetts, USA
| | - Sameh Yousef
- Department of Surgery, Yale School of Medicine, New Haven, Connecticut, USA
| | - Yang Jiao
- Department of Surgery, Yale School of Medicine, New Haven, Connecticut, USA
| | - Changshun He
- Department of Surgery, Yale School of Medicine, New Haven, Connecticut, USA
| | - Bo Jiang
- Department of Surgery, Yale School of Medicine, New Haven, Connecticut, USA
| | - Arina Korneva
- Department of Biomedical Engineering, Yale School of Engineering and Applied Science, New Haven, Connecticut, USA
| | - Matthew R Bersi
- Department of Biomedical Engineering, Yale School of Engineering and Applied Science, New Haven, Connecticut, USA
| | | | - Xinran Liu
- Cell Biology, Yale School of Medicine, New Haven, Connecticut, USA.,Center for Cellular and Molecular Imaging, EM Core Facility, Yale School of Medicine, New Haven, Connecticut, USA
| | | | - Arnar Geirsson
- Department of Surgery, Yale School of Medicine, New Haven, Connecticut, USA.,Program in Vascular Biology and Therapeutics, Yale School of Medicine, New Haven, Connecticut, USA
| | - Jeffrey R Gulcher
- Computational Statistics and Bioinformatics Group, Advanced Artificial Intelligence Research Laboratory, WuXi NextCODE, Cambridge, Massachusetts, USA
| | - Thomas W Chittenden
- Computational Statistics and Bioinformatics Group, Advanced Artificial Intelligence Research Laboratory, WuXi NextCODE, Cambridge, Massachusetts, USA
| | - Michael Simons
- Internal Medicine.,Program in Vascular Biology and Therapeutics, Yale School of Medicine, New Haven, Connecticut, USA
| | - Jay D Humphrey
- Department of Biomedical Engineering, Yale School of Engineering and Applied Science, New Haven, Connecticut, USA.,Program in Vascular Biology and Therapeutics, Yale School of Medicine, New Haven, Connecticut, USA
| | - George Tellides
- Department of Surgery, Yale School of Medicine, New Haven, Connecticut, USA.,Program in Vascular Biology and Therapeutics, Yale School of Medicine, New Haven, Connecticut, USA.,Veterans Affairs Connecticut Healthcare System, West Haven, Connecticut, USA
| |
Collapse
|
28
|
Kotani T, Setiawan J, Konno T, Ihara N, Okamoto S, Saito Y, Murata Y, Noda T, Matozaki T. Regulation of colonic epithelial cell homeostasis by mTORC1. Sci Rep 2020; 10:13810. [PMID: 32796887 PMCID: PMC7427982 DOI: 10.1038/s41598-020-70655-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Accepted: 07/30/2020] [Indexed: 12/29/2022] Open
Abstract
Cell signaling important for homeostatic regulation of colonic epithelial cells (CECs) remains poorly understood. Mammalian target of rapamycin complex 1 (mTORC1), a protein complex that contains the serine-threonine kinase mTOR, mediates signaling that underlies the control of cellular functions such as proliferation and autophagy by various external stimuli. We here show that ablation of tuberous sclerosis complex 2 (Tsc2), a negative regulator of mTORC1, specifically in intestinal epithelial cells of mice resulted in increased activity of mTORC1 of, as well as increased proliferative activity of, CECs. Such Tsc2 ablation also reduced the population of Lgr5-positive colonic stem cells and the expression of Wnt target genes in CECs. The stimulatory phosphorylation of the kinase Akt and inhibitory phosphorylation of glycogen synthase kinase 3β were both markedly decreased in the colon of the Tsc2 conditional knockout (CKO) mice. Development of colonic organoids with cryptlike structures was enhanced for Tsc2 CKO mice compared with control mice. Finally, Tsc2 CKO mice manifested increased susceptibility to dextran sulfate sodium-induced colitis. Our results thus suggest that mTORC1 activity promotes the proliferation of, as well as the expression of Wnt target genes in, CECs and thereby contributes to colonic organogenesis and homeostasis.
Collapse
Affiliation(s)
- Takenori Kotani
- Division of Molecular and Cellular Signaling, Department of Biochemistry and Molecular Biology, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Jajar Setiawan
- Division of Molecular and Cellular Signaling, Department of Biochemistry and Molecular Biology, Kobe University Graduate School of Medicine, Kobe, Japan.,Department of Physiology, Faculty of Medicine, Public Health, and Nursing, Universitas Gadjah Mada, Yogyakarta, Indonesia
| | - Tasuku Konno
- Division of Molecular and Cellular Signaling, Department of Biochemistry and Molecular Biology, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Noriko Ihara
- Division of Molecular and Cellular Signaling, Department of Biochemistry and Molecular Biology, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Saki Okamoto
- Division of Molecular and Cellular Signaling, Department of Biochemistry and Molecular Biology, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Yasuyuki Saito
- Division of Molecular and Cellular Signaling, Department of Biochemistry and Molecular Biology, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Yoji Murata
- Division of Molecular and Cellular Signaling, Department of Biochemistry and Molecular Biology, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Tetsuo Noda
- Department of Cell Biology, Cancer Institute, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Takashi Matozaki
- Division of Molecular and Cellular Signaling, Department of Biochemistry and Molecular Biology, Kobe University Graduate School of Medicine, Kobe, Japan.
| |
Collapse
|
29
|
Happle R, Torrelo A. Superimposed mosaicism in tuberous sclerosis complex: a key to understanding all of the manifold manifestations? J Eur Acad Dermatol Venereol 2020; 34:2511-2517. [DOI: 10.1111/jdv.16603] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Accepted: 04/10/2020] [Indexed: 12/27/2022]
Affiliation(s)
- R. Happle
- Department of Dermatology Medical Center University of Freiburg Freiburg Germany
| | - A. Torrelo
- Department. of Dermatology Hospital Infantil Universitario Niño Jesús Madrid Spain
| |
Collapse
|
30
|
Crawford M, Liu N, Mahdipour E, Barr K, Heit B, Dagnino L. Integrin-linked kinase regulates melanosome trafficking and melanin transfer in melanocytes. Mol Biol Cell 2020; 31:768-781. [PMID: 32049584 PMCID: PMC7185957 DOI: 10.1091/mbc.e19-09-0510] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Melanosomes are melanin-containing organelles that provide pigmentation and protection from solar UV radiation to the skin. In melanocytes, melanosomes mature and traffic to dendritic tips, where they are transferred to adjacent epidermal keratinocytes through pathways that involve microtubule networks and the actin cytoskeleton. However, the role of scaffold proteins in these processes is poorly understood. Integrin-linked kinase (ILK) is a scaffold protein that regulates microtubule stability and F-actin dynamics. Here we show that ILK is necessary for normal trafficking of melanosomes along microtubule tracks. In the absence of ILK, immature melanosomes are not retained in perinuclear regions, and mature melanosome trafficking along microtubule tracks is impaired. These deficits can be attenuated by microtubule stabilization. Microtubules are also necessary for the formation of dendrites in melanocytes, and Ilk inactivation reduces melanocyte dendricity. Activation of glycogen synthase kinase-3 (GSK-3) interferes with microtubule assembly. Significantly, inhibition of GSK-3 activity or exogenous expression of the GSK-3 substrate collapsin response mediator protein 2 (CRMP2) in ILK-deficient melanocytes restored dendricity. ILK is also required for normal melanin transfer, and GSK-3 inhibition in melanocytes partially restored melanin transfer to neighboring keratinocytes. Thus, our work shows that ILK is a central modulator of melanosome movements in primary epidermal melanocytes and identifies ILK and GSK-3 as important modulators of melanin transfer to keratinocytes, a key process for epidermal UV photoprotection.
Collapse
Affiliation(s)
- Melissa Crawford
- Department of Physiology and Pharmacology, Children's Health Research Institute and Lawson Health Research Institute, University of Western Ontario, London, ON N6G 2C4, Canada
| | - Nancy Liu
- Department of Physiology and Pharmacology, Children's Health Research Institute and Lawson Health Research Institute, University of Western Ontario, London, ON N6G 2C4, Canada
| | - Elahe Mahdipour
- Department of Physiology and Pharmacology, Children's Health Research Institute and Lawson Health Research Institute, University of Western Ontario, London, ON N6G 2C4, Canada.,Department of Medical Biotechnology and Nanotechnology, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Kevin Barr
- Department of Physiology and Pharmacology, Children's Health Research Institute and Lawson Health Research Institute, University of Western Ontario, London, ON N6G 2C4, Canada
| | - Bryan Heit
- Department of Microbiology and Immunology and Robarts Research Institute, University of Western Ontario, London, ON N6G 2C4, Canada
| | - Lina Dagnino
- Department of Physiology and Pharmacology, Children's Health Research Institute and Lawson Health Research Institute, University of Western Ontario, London, ON N6G 2C4, Canada.,Department of Oncology, University of Western Ontario, London, ON N6G 2C4, Canada
| |
Collapse
|
31
|
Zhang J, Zhu S, Tan Q, Cheng D, Dai Q, Yang Z, Zhang L, Li F, Zuo Y, Dai W, Chen L, Gu E, Xu G, Wei Z, Cao Y, Liu X. Combination therapy with ropivacaine-loaded liposomes and nutrient deprivation for simultaneous cancer therapy and cancer pain relief. Am J Cancer Res 2020; 10:4885-4899. [PMID: 32308756 PMCID: PMC7163441 DOI: 10.7150/thno.43932] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Accepted: 03/13/2020] [Indexed: 01/02/2023] Open
Abstract
Autophagy allows cancer cells to respond changes in nutrient status by degrading and recycling non-essential intracellular contents. Inhibition of autophagy combined with nutrient deprivation is an effective strategy to treat cancer. Pain is a primary determinant of poor quality of life in advanced cancer patients, but there is currently no satisfactory treatment. In addition, effective treatment of cancer does not efficiently relieve cancer pain, but may increase pain in many cases. Hence, few studies focus on simultaneous cancer therapy and pain relief, and made this situation even worse. Method: Ropivacaine was loaded into tumor-active targeted liposomes. The cytotoxicity of ropivacaine-based combination therapy in B16 and HeLa cells were tested. Moreover, a mice model of cancer pain which was induced by inoculation of melanoma near the sciatic nerve was constructed to assess the cancer suppression and pain relief effects of ropivacaine-based combination therapy. Results: Ropivacaine and ropivacaine-loaded liposomes (Rop-DPRL) were novelly found to damage autophagic degradation. Replicated administration of Rop-DPRL and calorie restriction (CR) could efficiently repress the development of tumor. In addition, administration of Rop-DPRL could relieve cancer pain with its own analgestic ability in a short duration, while repeated administration of Rop-DPRL and CR resulted in continuous alleviation of cancer pain through reduction of VEGF-A levels in advanced cancer mice. Further, dual inhibition of phosphorylation of STAT3 at Tyr705 and Ser727 by Rop-DPRL and CR contribute to the reduction of VEGF-A. Conclusion: Combination therapy with Rop-DPRL and nutrient deprivation simultaneously suppresses cancer growth and relieves cancer pain.
Collapse
|
32
|
Grabacka M, Plonka PM, Reiss K. Melanoma-Time to fast or time to feast? An interplay between PPARs, metabolism and immunity. Exp Dermatol 2020; 29:436-445. [PMID: 31957066 DOI: 10.1111/exd.14072] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2019] [Revised: 12/30/2019] [Accepted: 01/10/2020] [Indexed: 12/15/2022]
Abstract
Development and progression of melanoma can be accelerated by intensification of particular metabolic pathways, such as aerobic glycolysis and avid amino acid catabolism, and is accompanied by aberrant immune responses within the tumor microenvironment. Contrary to other cancer types, melanoma reveals some unique tissue-specific features, such as melanogenesis, which is intertwined with metabolism. Nuclear peroxisome proliferator-activated receptors (PPARs) take part in regulation of systemic and cellular metabolism, inflammation and melanogenesis. They appear as a focal regulatory point for these three distinct processes by occupying the intersection among AMP-dependent protein kinase (AMPK), mammalian target of rapamycin (mTOR) and PPAR gamma coactivator 1-alpha (PGC-1α) signalling pathways. When deregulated, they may accelerate melanoma malignant growth. Presenting the contribution of PPARα and PPARγ in melanoma biology, we attempt to ask how two contrasting metabolic states: obesity and fasting, can change progression of the disease and possible outcome of the treatment. This short essay is aimed to provoke a discussion about some practical implications for melanoma prevention and treatment, especially: how metabolic manipulation may be exploited to overcome immunosuppression and support immune checkpoint blockade efficacy.
Collapse
Affiliation(s)
- Maja Grabacka
- Department of Biotechnology and General Technology of Foods, Faculty of Food Technology, University of Agriculture, Kraków, Poland
| | - Przemyslaw M Plonka
- Department of Biophysics, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Kraków, Poland
| | - Krzysztof Reiss
- Stanley S. Scott Cancer Center, Louisiana State University Health Sciences Center, New Orleans, USA
| |
Collapse
|
33
|
Zhang L, Huang J, Lin Q, Ma Y, Xia R, Zhu Y, Abudubari S. Serum Proteomic Profiling Analysis of Rats Chronically Exposed to Arsenic. Med Sci Monit 2019; 25:9923-9932. [PMID: 31874112 PMCID: PMC6941779 DOI: 10.12659/msm.918696] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
Background Arsenic (As) is an environmental contaminant, and As pollution in water and soil is a public health issue worldwide. As exposure is associated with the incidence of many disorders, such as arteriosclerosis, diabetes, neurodegenerative diseases, and renal dysfunction. However, the mechanism of As toxicity remains unclear. Material/Methods We investigated the changes in serum protein profiles of rats chronically exposed to As. Twenty healthy rats were randomly divided into 4 groups, and sodium arsenite of varying final concentrations (0, 2, 10, and 50 mg/L, respectively) was add into the drinking water for each group. The administration lasted for 3 months. Two proteomic strategies, isobaric tags for relative and absolute quantitation (iTRAQ), and 2-dimensional gel electrophoresis (2-DE), were employed to screen the differential serum proteins between control and arsenite exposure groups. Results We identified a total of 27 differentially-expressed proteins, among which 9 proteins were significantly upregulated and 18 were downregulated by As exposure. Many of the differentially-expressed proteins were related to fat digestion and absorption, including 5 apolipoproteins, which indicated lipid metabolism may be the most affected by As exposure. Conclusions This study revealed the influence of As on lipid metabolism, suggesting an increased potential risk of relevant diseases in subjects chronically exposed to As.
Collapse
Affiliation(s)
- Ling Zhang
- Division of Endemic Disease Prevention, Xinjiang Uighur Autonomous Region Center for Disease Control and Prevention, Urumqi, Xinjiang, China (mainland)
| | - Jia Huang
- Division of Endemic Disease Prevention, Xinjiang Uighur Autonomous Region Center for Disease Control and Prevention, Urumqi, Xinjiang, China (mainland).,School of Public Health, Xinjiang Medical University, Urumqi, Xinjiang, China (mainland)
| | - Qin Lin
- Division of Endemic Disease Prevention, Xinjiang Uighur Autonomous Region Center for Disease Control and Prevention, Urumqi, Xinjiang, China (mainland)
| | - Yan Ma
- School of Public Health, Xinjiang Medical University, Urumqi, Xinjiang, China (mainland)
| | - Rongxiang Xia
- Division of Endemic Disease Prevention, Xinjiang Uighur Autonomous Region Center for Disease Control and Prevention, Urumqi, Xinjiang, China (mainland)
| | - Yuming Zhu
- Division of Endemic Disease Prevention, Xinjiang Uighur Autonomous Region Center for Disease Control and Prevention, Urumqi, Xinjiang, China (mainland)
| | - Saimaitikari Abudubari
- School of Public Health, Xinjiang Medical University, Urumqi, Xinjiang, China (mainland)
| |
Collapse
|
34
|
Improvement in Impaired Social Cognition but Not Seizures by Everolimus in a Child with Tuberous Sclerosis-Associated Autism through Increased Serum Antioxidant Proteins and Oxidant/Antioxidant Status. Case Rep Pediatr 2019; 2019:2070619. [PMID: 31871809 PMCID: PMC6907049 DOI: 10.1155/2019/2070619] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Revised: 07/11/2019] [Accepted: 08/01/2019] [Indexed: 12/23/2022] Open
Abstract
We investigated the effect of the mammalian target of rapamycin (mTOR) inhibitor everolimus on tuberous sclerosis complex- (TSC-) associated autistic symptoms and focal seizures with impaired awareness in a female child with TSC. We further evaluated the relationship between improved autistic symptoms and seizures and increased the serum levels of the antioxidant proteins, ceruloplasmin (Cp) and transferrin (Tf), and oxidant-antioxidant status indicated by the oxidant marker oxidized low-density lipoprotein (ox-LDL) and the antioxidant marker total antioxidant power (TAP). Everolimus treatment improved impaired social cognition and autistic behaviors; however, seizure and epileptic activity persisted. Serum Cp and Tf levels gradually increased in response to improved autistic symptoms. Serum TAP levels gradually decreased from baseline to the lowest value at 16 weeks and then increased at 24 weeks, showing a trend toward decreased total score of the Aberrant Behavior Checklist. This study revealed that everolimus treatment improved impaired social cognition with increased serum levels of the copper mediator (Cp) and iron mediator (Tf) via homeostatic control of mTOR activity accompanied by overlap of the oxidant-antioxidant system. Everolimus had no effect on TSC-related epileptiform discharges, and thus, the autistic symptoms and epileptic activity may be two independent end results of a common central nervous system disorder including mTOR hyperactivity. This trial is registered with JMAS-IIA00258.
Collapse
|
35
|
Kinslechner K, Schütz B, Pistek M, Rapolter P, Weitzenböck HP, Hundsberger H, Mikulits W, Grillari J, Röhrl C, Hengstschläger M, Stangl H, Mikula M. Loss of SR-BI Down-Regulates MITF and Suppresses Extracellular Vesicle Release in Human Melanoma. Int J Mol Sci 2019; 20:E1063. [PMID: 30823658 PMCID: PMC6429474 DOI: 10.3390/ijms20051063] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Revised: 02/22/2019] [Accepted: 02/26/2019] [Indexed: 02/06/2023] Open
Abstract
Melanoma is a skin tumor with a high tendency for metastasis and thus is one of the deadliest cancers worldwide. Here, we investigated the expression of the scavenger receptor class B type 1 (SR-BI), a high-density lipoprotein (HDL) receptor, and tested for its role in melanoma pigmentation as well as extracellular vesicle release. We first analyzed the expression of SR-BI in patient samples and found a strong correlation with MITF expression as well as with the melanin synthesis pathway. Hence, we asked whether SR-BI could also play a role for the secretory pathway in metastatic melanoma cells. Interestingly, gain- and loss-of-function of SR-BI revealed regulation of the proto-oncogene MET. In line, SR-BI knockdown reduced expression of the small GTPase RABB22A, the ESCRT-II protein VPS25, and SNAP25, a member of the SNARE complex. Accordingly, reduced overall extracellular vesicle generation was detected upon loss of SR-BI. In summary, SR-BI expression in human melanoma enhances the formation and transport of extracellular vesicles, thereby contributing to the metastatic phenotype. Therapeutic targeting of SR-BI would not only interfere with cholesterol uptake, but also with the secretory pathway, therefore suppressing a key hallmark of the metastatic program.
Collapse
Affiliation(s)
- Katharina Kinslechner
- Center for Pathobiochemistry and Genetics, Medical University of Vienna, 1090 Vienna, Austria.
| | - Birgit Schütz
- Center for Pathobiochemistry and Genetics, Medical University of Vienna, 1090 Vienna, Austria.
| | - Martina Pistek
- Center for Pathobiochemistry and Genetics, Medical University of Vienna, 1090 Vienna, Austria.
| | - Philipp Rapolter
- Center for Pathobiochemistry and Genetics, Medical University of Vienna, 1090 Vienna, Austria.
| | - Hans P Weitzenböck
- Medical and Pharmaceutical Biotechnology, IMC University of Applied Sciences, 3500 Krems, Austria.
| | - Harald Hundsberger
- Medical and Pharmaceutical Biotechnology, IMC University of Applied Sciences, 3500 Krems, Austria.
| | - Wolfgang Mikulits
- Department of Medicine I, Division: Institute of Cancer Research, Comprehensive Cancer Center, Medical University of Vienna, 1090 Vienna, Austria.
| | - Johannes Grillari
- Department of Biotechnology, BOKU -University of Natural Resources and Life Sciences, 1190 Vienna, Austria.
| | - Clemens Röhrl
- Center for Pathobiochemistry and Genetics, Medical University of Vienna, 1090 Vienna, Austria.
| | - Markus Hengstschläger
- Center for Pathobiochemistry and Genetics, Medical University of Vienna, 1090 Vienna, Austria.
| | - Herbert Stangl
- Center for Pathobiochemistry and Genetics, Medical University of Vienna, 1090 Vienna, Austria.
| | - Mario Mikula
- Center for Pathobiochemistry and Genetics, Medical University of Vienna, 1090 Vienna, Austria.
| |
Collapse
|
36
|
mTOR-dependent upregulation of xCT blocks melanin synthesis and promotes tumorigenesis. Cell Death Differ 2019; 26:2015-2028. [PMID: 30760873 DOI: 10.1038/s41418-019-0274-0] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2016] [Revised: 12/14/2018] [Accepted: 12/20/2018] [Indexed: 12/26/2022] Open
Abstract
Loss of either TSC1 or TSC2 causes tuberous sclerosis complex (TSC) via activation of mTOR signaling pathway. The two prominent features of TSC are skin lesions including hypomelanic macules and benign tumors in multiple organs, whose molecular alterations are largely unknown. We report here that Xc- cystine/glutamate antiporter (xCT) was elevated in Tsc2-/- or Pten-/- cells, Tsc1 knockout mouse tissues and TSC2-deficient human kidney tumor. xCT was transcriptionally boosted by mTOR-mediated Oct1 signaling cascade. Augmented xCT led to reduction of eumelanin and elevation of pheomelanin in Tsc1 skin knockout mice through mTOR signaling pathway. Disruption of xCT suppressed the proliferation and tumorigenesis of Pten-null cells and Tsc2-null cells. mTOR hyperactive cells were more sensitive to inhibitors of mTOR or xCT. Combined inhibition of mTOR and xCT synergistically blocked the propagation and oncogenesis of mTOR hyperactive cells. Therefore, oncogenic mTOR activation of xCT is a key connection between aberrant melanin synthesis and tumorigenesis. We suggest that xCT is a novel therapeutic target for TSC and other aberrant mTOR-related diseases.
Collapse
|
37
|
Ogórek B, Lam HC, Khabibullin D, Liu HJ, Nijmeh J, Triboulet R, Kwiatkowski DJ, Gregory RI, Henske EP. TSC2 regulates microRNA biogenesis via mTORC1 and GSK3β. Hum Mol Genet 2019; 27:1654-1663. [PMID: 29509898 DOI: 10.1093/hmg/ddy073] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2017] [Accepted: 02/26/2018] [Indexed: 12/21/2022] Open
Abstract
Tuberous sclerosis complex (TSC) is an autosomal dominant disease caused by germline inactivating mutations of TSC1 or TSC2. In TSC-associated tumors of the brain, heart, skin, kidney and lung, inactivation of both alleles of TSC1 or TSC2 leads to hyperactivation of the mTORC1 pathway. The TSC/mTORC1 pathway is a key regulator of cellular processes related to growth, proliferation and autophagy. We and others have previously found that mTORC1 regulates microRNA biogenesis, but the mechanisms are not fully understood. Microprocessor, a multi-protein complex including the nuclease Drosha, processes the primary miR transcript. Using a dual-luciferase reporter, we found that inhibition of mTORC1 or downregulation of Raptor decreased Microprocessor activity, while loss of TSC2 led to a striking increase (∼5-fold) in Microprocessor activity. To determine the global impact of TSC2 on microRNAs we quantitatively analyzed 752 microRNAs in Tsc2-expressing and Tsc2-deficient cells. Out of 259 microRNAs expressed in both cell lines, 137 were significantly upregulated and 24 were significantly downregulated in Tsc2-deficient cells, consistent with the increased Microprocessor activity. Microprocessor activity is known to be regulated in part by GSK3β. We found that total GSK3β levels were higher in Tsc2-deficient cells, and the increase in Microprocessor activity associated with Tsc2 loss was reversed by three different GSK3β inhibitors. Furthermore, mTOR inhibition increased the levels of phospho-GSK3β (S9), which negatively affects Microprocessor activity. Taken together these data reveal that TSC2 regulates microRNA biogenesis and Microprocessor activity via GSK3β.
Collapse
Affiliation(s)
- Barbara Ogórek
- Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Hilaire C Lam
- Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Damir Khabibullin
- Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Heng-Jia Liu
- Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Julie Nijmeh
- Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Robinson Triboulet
- Stem Cell Program, Division of Hematology/Oncology, Boston Children's Hospital, Boston, MA 02115, USA.,Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02115, USA
| | - David J Kwiatkowski
- Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Richard I Gregory
- Stem Cell Program, Division of Hematology/Oncology, Boston Children's Hospital, Boston, MA 02115, USA.,Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02115, USA
| | - Elizabeth P Henske
- Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
38
|
Abnormal glycogen storage in tuberous sclerosis complex caused by impairment of mTORC1-dependent and -independent signaling pathways. Proc Natl Acad Sci U S A 2019; 116:2977-2986. [PMID: 30728291 DOI: 10.1073/pnas.1812943116] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Tuberous sclerosis complex (TSC) is an autosomal dominant syndrome that causes tumor formation in multiple organs. TSC is caused by inactivating mutations in the genes encoding TSC1/2, negative regulators of the mammalian target of rapamycin complex 1 (mTORC1). Diminished TSC function is associated with excess glycogen storage, but the causative mechanism is unknown. By studying human and mouse cells with defective or absent TSC2, we show that complete loss of TSC2 causes an increase in glycogen synthesis through mTORC1 hyperactivation and subsequent inactivation of glycogen synthase kinase 3β (GSK3β), a negative regulator of glycogen synthesis. Specific TSC2 pathogenic mutations, however, result in elevated glycogen levels with no changes in mTORC1 or GSK3β activities. We identify mTORC1-independent lysosomal depletion and impairment of autophagy as the driving causes underlying abnormal glycogen storage in TSC irrespective of the underlying mutation. The defective autophagic degradation of glycogen is associated with abnormal ubiquitination and degradation of essential proteins of the autophagy-lysosome pathway, such as LC3 and lysosomal associated membrane protein 1 and 2 (LAMP1/2) and is restored by the combined use of mTORC1 and Akt pharmacological inhibitors. In complementation to current models that place mTORC1 as the central therapeutic target for TSC pathogenesis, our findings identify mTORC1-independent pathways that are dysregulated in TSC and that should therefore be taken into account in the development of a therapeutic treatment.
Collapse
|
39
|
Huang J, Gu S, Chen M, Zhang SJ, Jiang Z, Chen X, Jiang C, Liu G, Radu RA, Sun X, Vollrath D, Du J, Yan B, Zhao C. Abnormal mTORC1 signaling leads to retinal pigment epithelium degeneration. Am J Cancer Res 2019; 9:1170-1180. [PMID: 30867823 PMCID: PMC6401408 DOI: 10.7150/thno.26281] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2018] [Accepted: 12/24/2018] [Indexed: 12/13/2022] Open
Abstract
Retinal pigment epithelial (RPE) degeneration is potentially involved in the pathogenesis of several retinal degenerative diseases. mTORC1 signaling is shown as a crucial regulator of many biological processes and disease progression. In this study, we aimed at investigating the role of mTORC1 signaling in RPE degeneration. Methods: Western blots were conducted to detect mTORC1 expression pattern during RPE degeneration. Cre-loxP system was used to generate RPE-specific mTORC1 activation mice. Fundus, immunofluorescence staining, transmission electron microscopy, and targeted metabolomic analysis were conducted to determine the effects of mTORC1 activation on RPE degeneration in vivo. Electroretinography, spectral-domain optical coherence tomography, and histological experiments were conducted to determine the effects of mTORC1 activation on choroidal and retinal function in vivo. Results: RPE-specific activation of mTORC1 led to RPE degeneration as shown by the loss of RPE-specific marker, compromised cell junction integrity, and intracellular accumulation of lipid droplets. RPE degeneration further led to abnormal choroidal and retinal function. The inhibition of mTORC1 signaling with rapamycin could partially reverse RPE degeneration. Targeted metabolomics analysis further revealed that mTORC1 activation affected the metabolism of purine, carboxylic acid, and niacin in RPE. Conclusion: This study revealed that abnormal activation of mTORC1 signaling leads to RPE degeneration, which could provide a promising target for the treatment of RPE dysfunction-related diseases.
Collapse
|
40
|
mTORC1 signaling suppresses Wnt/β-catenin signaling through DVL-dependent regulation of Wnt receptor FZD level. Proc Natl Acad Sci U S A 2018; 115:E10362-E10369. [PMID: 30297426 DOI: 10.1073/pnas.1808575115] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Wnt/β-catenin signaling plays pivotal roles in cell proliferation and tissue homeostasis by maintaining somatic stem cell functions. The mammalian target of rapamycin (mTOR) signaling functions as an integrative rheostat that orchestrates various cellular and metabolic activities that shape tissue homeostasis. Whether these two fundamental signaling pathways couple to exert physiological functions still remains mysterious. Using a genome-wide CRISPR-Cas9 screening, we discover that mTOR complex 1 (mTORC1) signaling suppresses canonical Wnt/β-catenin signaling. Deficiency in tuberous sclerosis complex 1/2 (TSC1/2), core negative regulators of mTORC1 activity, represses Wnt/β-catenin target gene expression, which can be rescued by RAD001. Mechanistically, mTORC1 signaling regulates the cell surface level of Wnt receptor Frizzled (FZD) in a Dishevelled (DVL)-dependent manner by influencing the association of DVL and clathrin AP-2 adaptor. Sustained mTORC1 activation impairs Wnt/β-catenin signaling and causes loss of stemness in intestinal organoids ex vivo and primitive intestinal progenitors in vivo. Wnt/β-catenin-dependent liver metabolic zonation gene expression program is also down-regulated by mTORC1 activation. Our study provides a paradigm that mTORC1 signaling cell autonomously regulates Wnt/β-catenin pathway to influence stem cell maintenance.
Collapse
|
41
|
耿 忆, 王 亚, 邓 蓉, 傅 楷, 邓 燕. [Shufeng Huoxue Formula suppresses proliferation and regulates melanin metabolism in murine B16 melanoma cells in vitro through autophagy pathway]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2018; 38:630-634. [PMID: 29891464 PMCID: PMC6743889 DOI: 10.3969/j.issn.1673-4254.2018.05.21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 11/06/2017] [Indexed: 06/08/2023]
Abstract
OBJECTIVE To investigate the role of autophagy in the regulatory effect of Shufeng Huoxue Fumula (SFHXF) on the proliferation and melanin metabolism in cultured murine B16 melanoma cells. METHODS B16 cells were treated with solutions containing 0.12, 0.25, 0.49, 0.98, or 1.96 mg/mL SFHXF preparations, rapamycin (an autophagy inducer), or rapamycin+SFHXF. The changes in the proliferation of B16 cells were assessed using MTT assay, and tyrosinase activity and melanin content in the cells were determined. The expressions of autophagy-related proteins P62, p-mTOR, LC3B, and beclin 1 in the cells were detected using Western blotting. RESULT Compared with the blank control cells, treatments with SFHXF both in the presence and in the absence of rapamycin concentration-dependently inhibited the cell proliferation (P<0.05) and obviously increased tyrosinase activity and melanogenesis in B16 cells (P<0.05); 0.98 mg/mL SFHLF, rapamycin+0.98 mg/mL SFHXF, and 50 nmol/L rapamycin all significantly up-regulated the expressions of LC3B-II and beclin 1 and down-regulated the expressions of P62 and p-mTOR in the cells. CONCLUSION SFHXF can regulate melanin metabolism and enhance tyrosinase activity and melanogenesis through the autophagy pathway to inhibit the proliferation of B16 cells in vitro.
Collapse
Affiliation(s)
- 忆薇 耿
- />南方医科大学中医药学院,广东 广州 510515School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, China
| | - 亚兰 王
- />南方医科大学中医药学院,广东 广州 510515School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, China
| | - 蓉 邓
- />南方医科大学中医药学院,广东 广州 510515School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, China
| | - 楷历 傅
- />南方医科大学中医药学院,广东 广州 510515School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, China
| | - 燕 邓
- />南方医科大学中医药学院,广东 广州 510515School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, China
| |
Collapse
|
42
|
|
43
|
Yang F, Yang L, Wataya-Kaneda M, Yoshimura T, Tanemura A, Katayama I. Uncoupling of ER/Mitochondrial Oxidative Stress in mTORC1 Hyperactivation-Associated Skin Hypopigmentation. J Invest Dermatol 2017; 138:669-678. [PMID: 29080681 DOI: 10.1016/j.jid.2017.10.007] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2017] [Revised: 09/28/2017] [Accepted: 10/08/2017] [Indexed: 11/30/2022]
Abstract
Accumulating evidence has described the involvement of mTORC1 signaling in pigmentation regulation; however, the precise mechanism is not fully understood. Here, we generated mice with conditional deletion of the mTORC1 suppressor Tsc2 in melanocytes. It resulted in constitutive hyperactivation of mTORC1 and reduced skin pigmentation. Mechanistically, neither the number of melanocytes nor the expression of melanogenesis-related enzymes was decreased; however, endoplasmic reticulum and mitochondrial oxidative stress and lower melanization in melanosomes were observed. By contrast, abrogation of mTORC1 by rapamycin completely reversed the reduced pigmentation, and alleviation of endoplasmic reticulum stress by SMER28 or 4-phenylbutyrate (PBA) or alleviation of mitochondrial oxidative stress by administration of adenosine triphosphate partially reversed the reduced pigmentation in these mice. In addition, we showed that these mechanisms were involved in reduced pigmentation of TSC2 small interfering RNA-transfected cultured human primary melanocytes and skin lesions of patients with the TSC gene mutation.
Collapse
Affiliation(s)
- Fei Yang
- Department of Dermatology, Course of Integrated Medicine, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Lingli Yang
- Department of Dermatology, Course of Integrated Medicine, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Mari Wataya-Kaneda
- Department of Dermatology, Course of Integrated Medicine, Graduate School of Medicine, Osaka University, Osaka, Japan.
| | - Takuji Yoshimura
- Laboratory of Reproductive Engineering, The Institute of Experimental Animal Sciences, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Atsushi Tanemura
- Department of Dermatology, Course of Integrated Medicine, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Ichiro Katayama
- Department of Dermatology, Course of Integrated Medicine, Graduate School of Medicine, Osaka University, Osaka, Japan
| |
Collapse
|
44
|
Armstrong LC, Westlake G, Snow JP, Cawthon B, Armour E, Bowman AB, Ess KC. Heterozygous loss of TSC2 alters p53 signaling and human stem cell reprogramming. Hum Mol Genet 2017; 26:4629-4641. [PMID: 28973543 PMCID: PMC5886307 DOI: 10.1093/hmg/ddx345] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2017] [Revised: 08/28/2017] [Accepted: 09/01/2017] [Indexed: 12/21/2022] Open
Abstract
Tuberous sclerosis complex (TSC) is a pediatric disorder of dysregulated growth and differentiation caused by loss of function mutations in either the TSC1 or TSC2 genes, which regulate mTOR kinase activity. To study aberrations of early development in TSC, we generated induced pluripotent stem cells using dermal fibroblasts obtained from patients with TSC. During validation, we found that stem cells generated from TSC patients had a very high rate of integration of the reprogramming plasmid containing a shRNA against TP53. We also found that loss of one allele of TSC2 in human fibroblasts is sufficient to increase p53 levels and impair stem cell reprogramming. Increased p53 was also observed in TSC2 heterozygous and homozygous mutant human stem cells, suggesting that the interactions between TSC2 and p53 are consistent across cell types and gene dosage. These results support important contributions of TSC2 heterozygous and homozygous mutant cells to the pathogenesis of TSC and the important role of p53 during reprogramming.
Collapse
Affiliation(s)
- Laura C Armstrong
- Division of Pediatric Neurology, Department of Pediatrics, Vanderbilt University Medical Center, D4105 Medical Center North, Nashville, TN 37232, USA
| | - Grant Westlake
- Division of Pediatric Neurology, Department of Pediatrics, Vanderbilt University Medical Center, D4105 Medical Center North, Nashville, TN 37232, USA
| | - John P Snow
- Division of Pediatric Neurology, Department of Pediatrics, Vanderbilt University Medical Center, D4105 Medical Center North, Nashville, TN 37232, USA
| | - Bryan Cawthon
- Division of Pediatric Neurology, Department of Pediatrics, Vanderbilt University Medical Center, D4105 Medical Center North, Nashville, TN 37232, USA
| | - Eric Armour
- Division of Pediatric Neurology, Department of Pediatrics, Vanderbilt University Medical Center, D4105 Medical Center North, Nashville, TN 37232, USA
| | - Aaron B Bowman
- Division of Pediatric Neurology, Department of Pediatrics, Vanderbilt University Medical Center, D4105 Medical Center North, Nashville, TN 37232, USA
| | - Kevin C Ess
- Division of Pediatric Neurology, Department of Pediatrics, Vanderbilt University Medical Center, D4105 Medical Center North, Nashville, TN 37232, USA
| |
Collapse
|
45
|
Yang F, Yang L, Wataya-Kaneda M, Hasegawa J, Yoshimori T, Tanemura A, Tsuruta D, Katayama I. Dysregulation of autophagy in melanocytes contributes to hypopigmented macules in tuberous sclerosis complex. J Dermatol Sci 2017; 89:155-164. [PMID: 29146131 DOI: 10.1016/j.jdermsci.2017.11.002] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2017] [Revised: 10/05/2017] [Accepted: 11/01/2017] [Indexed: 12/15/2022]
Abstract
BACKGROUND Tuberous sclerosis complex (TSC) gene mutations lead to constitutive activation of the mammalian target of rapamycin (mTOR) pathway, resulting in a broad range of symptoms. Hypopigmented macules are the earliest sign. Although we have already confirmed that topical rapamycin treatment (an mTOR inhibitor) protects patients with TSC against macular hypopigmentation, the pathogenesis of such lesions remains poorly understood. OBJECTIVE Recently emerging evidence supports a role for autophagy in skin pigmentation. Herein, we investigated the impact of autophagic dysregulation on TSC-associated hypopigmentation. METHODS Skin samples from 10 patients with TSC, each bearing characteristic hypopigmented macules, and 6 healthy donors were subjected to immunohistochemical and electron microscopic analyses. In addition, TSC2-knockdown (KD) was investigated in human epidermal melanocytes by melanin content examination, real-time PCR, western blotting analyses, and intracellular immunofluorescence staining. RESULTS Activation of the mTOR signaling pathway decreased melanocytic pigmentation in hypopigmented macules of patients with TSC and in TSC2-KD melanocytes. In addition, LC3 expression (a marker of autophagy) and autophagosome counts increased, whereas, intracellular accumulation of autophagic degradative substrates (p62 and ubiquitinated proteins) was evident in TSC2-KD melanocytes. Furthermore, depigmentation in TSC2-KD melanocytes was accelerated by inhibiting autophagy (ATG7-KD or bafilomycin A1-pretreatment) and was completely reversed by induction of autophagy via mTOR-dependent (rapamycin) or mTOR-independent (SMER28) exposure. Finally, dysregulation of autophagy, marked by increased LC3 expression and accumulation of ubiquitinated proteins, was also observed in melanocytes of TSC-related hypopigmented macules. CONCLUSION Our data demonstrate that melanocytes of patients with TSC display autophagic dysregulation, which thereby reduced pigmentation, serving as the basis for the hypomelanotic macules characteristic of TSC.
Collapse
Affiliation(s)
- Fei Yang
- Department of Dermatology, Course of Integrated Medicine, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Lingli Yang
- Department of Dermatology, Course of Integrated Medicine, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Mari Wataya-Kaneda
- Department of Dermatology, Course of Integrated Medicine, Graduate School of Medicine, Osaka University, Osaka, Japan.
| | - Junya Hasegawa
- Laboratory of Intracellular Membrane Dynamics, Graduate School of Frontier Biosciences, Osaka University, Osaka, Japan
| | - Tamotsu Yoshimori
- Laboratory of Intracellular Membrane Dynamics, Graduate School of Frontier Biosciences, Osaka University, Osaka, Japan
| | - Atsushi Tanemura
- Department of Dermatology, Course of Integrated Medicine, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Daisuke Tsuruta
- Department of Dermatology, Graduate School of Medicine, Osaka City University, Osaka, Japan
| | - Ichiro Katayama
- Department of Dermatology, Course of Integrated Medicine, Graduate School of Medicine, Osaka University, Osaka, Japan
| |
Collapse
|
46
|
Figlia G, Norrmén C, Pereira JA, Gerber D, Suter U. Dual function of the PI3K-Akt-mTORC1 axis in myelination of the peripheral nervous system. eLife 2017; 6:e29241. [PMID: 28880149 PMCID: PMC5589416 DOI: 10.7554/elife.29241] [Citation(s) in RCA: 68] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2017] [Accepted: 07/31/2017] [Indexed: 01/24/2023] Open
Abstract
Myelination is a biosynthetically demanding process in which mTORC1, the gatekeeper of anabolism, occupies a privileged regulatory position. We have shown previously that loss of mTORC1 function in Schwann cells (SCs) hampers myelination. Here, we genetically disrupted key inhibitory components upstream of mTORC1, TSC1 or PTEN, in mouse SC development, adult homeostasis, and nerve injury. Surprisingly, the resulting mTORC1 hyperactivity led to markedly delayed onset of both developmental myelination and remyelination after injury. However, if mTORC1 was hyperactivated after myelination onset, radial hypermyelination was observed. At early developmental stages, physiologically high PI3K-Akt-mTORC1 signaling suppresses expression of Krox20 (Egr2), the master regulator of PNS myelination. This effect is mediated by S6K and contributes to control mechanisms that keep SCs in a not-fully differentiated state to ensure proper timing of myelination initiation. An ensuing decline in mTORC1 activity is crucial to allow myelination to start, while remaining mTORC1 activity drives myelin growth.
Collapse
Affiliation(s)
- Gianluca Figlia
- Department of BiologyInstitute of Molecular Health Sciences, Swiss Federal Institute of TechnologyZürichSwitzerland
| | - Camilla Norrmén
- Department of BiologyInstitute of Molecular Health Sciences, Swiss Federal Institute of TechnologyZürichSwitzerland
| | - Jorge A Pereira
- Department of BiologyInstitute of Molecular Health Sciences, Swiss Federal Institute of TechnologyZürichSwitzerland
| | - Daniel Gerber
- Department of BiologyInstitute of Molecular Health Sciences, Swiss Federal Institute of TechnologyZürichSwitzerland
| | - Ueli Suter
- Department of BiologyInstitute of Molecular Health Sciences, Swiss Federal Institute of TechnologyZürichSwitzerland
| |
Collapse
|
47
|
Julian LM, Delaney SP, Wang Y, Goldberg AA, Doré C, Yockell-Lelièvre J, Tam RY, Giannikou K, McMurray F, Shoichet MS, Harper ME, Henske EP, Kwiatkowski DJ, Darling TN, Moss J, Kristof AS, Stanford WL. Human Pluripotent Stem Cell-Derived TSC2-Haploinsufficient Smooth Muscle Cells Recapitulate Features of Lymphangioleiomyomatosis. Cancer Res 2017; 77:5491-5502. [PMID: 28830860 DOI: 10.1158/0008-5472.can-17-0925] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2017] [Revised: 06/22/2017] [Accepted: 08/16/2017] [Indexed: 01/06/2023]
Abstract
Lymphangioleiomyomatosis (LAM) is a progressive destructive neoplasm of the lung associated with inactivating mutations in the TSC1 or TSC2 tumor suppressor genes. Cell or animal models that accurately reflect the pathology of LAM have been challenging to develop. Here, we generated a robust human cell model of LAM by reprogramming TSC2 mutation-bearing fibroblasts from a patient with both tuberous sclerosis complex (TSC) and LAM (TSC-LAM) into induced pluripotent stem cells (iPSC), followed by selection of cells that resemble those found in LAM tumors by unbiased in vivo differentiation. We established expandable cell lines under smooth muscle cell (SMC) growth conditions that retained a patient-specific genomic TSC2+/- mutation and recapitulated the molecular and functional characteristics of pulmonary LAM cells. These include multiple indicators of hyperactive mTORC1 signaling, presence of specific neural crest and SMC markers, expression of VEGF-D and female sex hormone receptors, reduced autophagy, and metabolic reprogramming. Intriguingly, the LAM-like features of these cells suggest that haploinsufficiency at the TSC2 locus contributes to LAM pathology, and demonstrated that iPSC reprogramming and SMC lineage differentiation of somatic patient cells with germline mutations was a viable approach to generate LAM-like cells. The patient-derived SMC lines we have developed thus represent a novel cellular model of LAM that can advance our understanding of disease pathogenesis and develop therapeutic strategies against LAM. Cancer Res; 77(20); 5491-502. ©2017 AACR.
Collapse
Affiliation(s)
- Lisa M Julian
- Ottawa Hospital Research Institute, Regenerative Medicine Program, Ottawa, Ontario, Canada.,University of Ottawa, Ottawa, Ontario, Canada
| | - Sean P Delaney
- Ottawa Hospital Research Institute, Regenerative Medicine Program, Ottawa, Ontario, Canada.,University of Ottawa, Ottawa, Ontario, Canada.,Ottawa Institute of Systems Biology, Ottawa, Ontario, Canada
| | - Ying Wang
- Ottawa Hospital Research Institute, Regenerative Medicine Program, Ottawa, Ontario, Canada
| | | | - Carole Doré
- Ottawa Hospital Research Institute, Regenerative Medicine Program, Ottawa, Ontario, Canada
| | | | - Roger Y Tam
- Ottawa Hospital Research Institute, Regenerative Medicine Program, Ottawa, Ontario, Canada.,University of Ottawa, Ottawa, Ontario, Canada.,University of Toronto, Donnelly Centre for Cellular & Biomolecular Research, Boston, Massachusetts
| | - Krinio Giannikou
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Fiona McMurray
- University of Ottawa, Ottawa, Ontario, Canada.,Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Health Sciences, Bethesda, Maryland
| | - Molly S Shoichet
- University of Toronto, Donnelly Centre for Cellular & Biomolecular Research, Boston, Massachusetts
| | - Mary-Ellen Harper
- University of Ottawa, Ottawa, Ontario, Canada.,Ottawa Institute of Systems Biology, Ottawa, Ontario, Canada.,Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Health Sciences, Bethesda, Maryland
| | - Elizabeth P Henske
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - David J Kwiatkowski
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Thomas N Darling
- Uniformed Services University of Health Sciences, Bethesda, Maryland
| | - Joel Moss
- National Heart, Lung, and Blood Institute, NIH, Bethesda, Maryland
| | - Arnold S Kristof
- Research Institute of McGill University Health Centre, Montreal, Quebec, Canada
| | - William L Stanford
- Ottawa Hospital Research Institute, Regenerative Medicine Program, Ottawa, Ontario, Canada. .,University of Ottawa, Ottawa, Ontario, Canada.,Ottawa Institute of Systems Biology, Ottawa, Ontario, Canada
| |
Collapse
|
48
|
[Effect of TSC2 gene expression downregulation by lentivirus induced RNA interference on U937 cell line and its mechanism]. ZHONGHUA XUE YE XUE ZA ZHI = ZHONGHUA XUEYEXUE ZAZHI 2017; 38:612-617. [PMID: 28810331 PMCID: PMC7342282 DOI: 10.3760/cma.j.issn.0253-2727.2017.07.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
目的 研究下调TSC2基因表达对白血病U937细胞系的生物学作用及其对mTOR通路活性的影响。 方法 选择TSC2高表达的U937细胞系,通过慢病毒介导的RNA干扰技术下调TSC2基因表达;采用CCK-8比色法、细胞集落形成实验和流式细胞术检测其对细胞增殖、分化和凋亡的影响;采用Western blot法和实时荧光定量PCR(RQ-PCR)法检测TSC2表达下调对mTOR通路蛋白表达及活性的影响。 结果 TSC2基因表达降低能够促进U937细胞的增殖和集落形成(P<0.05);能够使U937细胞G1期[(52.53±3.75)%对(75.10±4.33)%,t=6.829,P=0.002]比例明显降低,G2/M期[(22.43±1.00)%对(15.47±1.20)%,t=−5.581,P=0.019]、S期[(25.03±4.34)%对(14.33±0.91)%,t=−5.413,P=0.013]比例升高;对细胞分化和细胞凋亡没有明显影响(P>0.05)。TSC2基因表达下调后,mTOR活性升高,磷酸化的4EBP1和S6K1蛋白活性升高,而AKT蛋白活性没有明显变化;与细胞增殖相关的基因cyclin D1、c-myc表达升高,PTEN基因表达升高,P27KIP基因和凋亡相关基因BCL-XL的表达没有明显的改变。 结论 TSC2基因表达下调可以通过调节mTOR通路活性促进白血病细胞的增殖。
Collapse
|