1
|
Liu Y, Gao H, Chen H, Ji S, Wu L, Zhang H, Wang Y, Fu X, Sun X. Sebaceous gland organoid engineering. BURNS & TRAUMA 2024; 12:tkae003. [PMID: 38699464 PMCID: PMC11063650 DOI: 10.1093/burnst/tkae003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Revised: 12/27/2023] [Indexed: 05/05/2024]
Abstract
Sebaceous glands (SGs), as holocrine-secreting appendages, lubricate the skin and play a central role in the skin barrier. Large full-thickness skin defects cause overall architecture disruption and SG loss. However, an effective strategy for SG regeneration is lacking. Organoids are 3D multicellular structures that replicate key anatomical and functional characteristics of in vivo tissues and exhibit great potential in regenerative medicine. Recently, considerable progress has been made in developing reliable procedures for SG organoids and existing SG organoids recapitulate the main morphological, structural and functional features of their in vivo counterparts. Engineering approaches empower researchers to manipulate cell behaviors, the surrounding environment and cell-environment crosstalk within the culture system as needed. These techniques can be applied to the SG organoid culture system to generate functionally more competent SG organoids. This review aims to provide an overview of recent advancements in SG organoid engineering. It highlights some potential strategies for SG organoid functionalization that are promising to forge a platform for engineering vascularized, innervated, immune-interactive and lipogenic SG organoids. We anticipate that this review will not only contribute to improving our understanding of SG biology and regeneration but also facilitate the transition of the SG organoid from laboratory research to a feasible clinical application.
Collapse
Affiliation(s)
- Yiqiong Liu
- Research Center for Tissue Repair and Regeneration affliated to the Medical Innovation Research Department and 4th Medical Center, PLA General Hospital and PLA Medical College; PLA Key Laboratory of Tissue Repair and Regenerative Medicine and Beijing Key Research Laboratory of Skin Injury, Repair and Regeneration; Research Unit of Trauma Care, Tissue Repair and Regeneration, Chinese Academy of Medical Sciences, 2019RU051, Beijing 100048, P. R. China
| | - Huanhuan Gao
- Research Center for Tissue Repair and Regeneration affliated to the Medical Innovation Research Department and 4th Medical Center, PLA General Hospital and PLA Medical College; PLA Key Laboratory of Tissue Repair and Regenerative Medicine and Beijing Key Research Laboratory of Skin Injury, Repair and Regeneration; Research Unit of Trauma Care, Tissue Repair and Regeneration, Chinese Academy of Medical Sciences, 2019RU051, Beijing 100048, P. R. China
| | - Huating Chen
- Research Center for Tissue Repair and Regeneration affliated to the Medical Innovation Research Department and 4th Medical Center, PLA General Hospital and PLA Medical College; PLA Key Laboratory of Tissue Repair and Regenerative Medicine and Beijing Key Research Laboratory of Skin Injury, Repair and Regeneration; Research Unit of Trauma Care, Tissue Repair and Regeneration, Chinese Academy of Medical Sciences, 2019RU051, Beijing 100048, P. R. China
| | - Shuaifei Ji
- Research Center for Tissue Repair and Regeneration affliated to the Medical Innovation Research Department and 4th Medical Center, PLA General Hospital and PLA Medical College; PLA Key Laboratory of Tissue Repair and Regenerative Medicine and Beijing Key Research Laboratory of Skin Injury, Repair and Regeneration; Research Unit of Trauma Care, Tissue Repair and Regeneration, Chinese Academy of Medical Sciences, 2019RU051, Beijing 100048, P. R. China
| | - Lu Wu
- Research Center for Tissue Repair and Regeneration affliated to the Medical Innovation Research Department and 4th Medical Center, PLA General Hospital and PLA Medical College; PLA Key Laboratory of Tissue Repair and Regenerative Medicine and Beijing Key Research Laboratory of Skin Injury, Repair and Regeneration; Research Unit of Trauma Care, Tissue Repair and Regeneration, Chinese Academy of Medical Sciences, 2019RU051, Beijing 100048, P. R. China
| | - Hongliang Zhang
- Research Center for Tissue Repair and Regeneration affliated to the Medical Innovation Research Department and 4th Medical Center, PLA General Hospital and PLA Medical College; PLA Key Laboratory of Tissue Repair and Regenerative Medicine and Beijing Key Research Laboratory of Skin Injury, Repair and Regeneration; Research Unit of Trauma Care, Tissue Repair and Regeneration, Chinese Academy of Medical Sciences, 2019RU051, Beijing 100048, P. R. China
| | - Yujia Wang
- Queen Mary School of Nanchang University, Nanchang University, Nanchang, Jiangxi 330006, P. R. China
| | - Xiaobing Fu
- Research Center for Tissue Repair and Regeneration affliated to the Medical Innovation Research Department and 4th Medical Center, PLA General Hospital and PLA Medical College; PLA Key Laboratory of Tissue Repair and Regenerative Medicine and Beijing Key Research Laboratory of Skin Injury, Repair and Regeneration; Research Unit of Trauma Care, Tissue Repair and Regeneration, Chinese Academy of Medical Sciences, 2019RU051, Beijing 100048, P. R. China
| | - Xiaoyan Sun
- Research Center for Tissue Repair and Regeneration affliated to the Medical Innovation Research Department and 4th Medical Center, PLA General Hospital and PLA Medical College; PLA Key Laboratory of Tissue Repair and Regenerative Medicine and Beijing Key Research Laboratory of Skin Injury, Repair and Regeneration; Research Unit of Trauma Care, Tissue Repair and Regeneration, Chinese Academy of Medical Sciences, 2019RU051, Beijing 100048, P. R. China
| |
Collapse
|
2
|
Helman SL, Zhou J, Fuqua BK, Lu Y, Collins JF, Chen H, Vulpe CD, Anderson GJ, Frazer DM. The biology of mammalian multi-copper ferroxidases. Biometals 2023; 36:263-281. [PMID: 35167013 PMCID: PMC9376197 DOI: 10.1007/s10534-022-00370-z] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2021] [Accepted: 02/04/2022] [Indexed: 12/24/2022]
Abstract
The mammalian multicopper ferroxidases (MCFs) ceruloplasmin (CP), hephaestin (HEPH) and zyklopen (ZP) comprise a family of conserved enzymes that are essential for body iron homeostasis. Each of these enzymes contains six biosynthetically incorporated copper atoms which act as intermediate electron acceptors, and the oxidation of iron is associated with the four electron reduction of dioxygen to generate two water molecules. CP occurs in both a secreted and GPI-linked (membrane-bound) form, while HEPH and ZP each contain a single C-terminal transmembrane domain. These enzymes function to ensure the efficient oxidation of iron so that it can be effectively released from tissues via the iron export protein ferroportin and subsequently bound to the iron carrier protein transferrin in the blood. CP is particularly important in facilitating iron release from the liver and central nervous system, HEPH is the major MCF in the small intestine and is critical for dietary iron absorption, and ZP is important for normal hair development. CP and HEPH (and possibly ZP) function in multiple tissues. These proteins also play other (non-iron-related) physiological roles, but many of these are ill-defined. In addition to disrupting iron homeostasis, MCF dysfunction perturbs neurological and immune function, alters cancer susceptibility, and causes hair loss, but, despite their importance, how MCFs co-ordinately maintain body iron homeostasis and perform other functions remains incompletely understood.
Collapse
Affiliation(s)
- Sheridan L Helman
- Molecular Nutrition Laboratory, QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | - Jie Zhou
- Department of Physiological Sciences, University of Florida, Gainsville, FL, USA
| | - Brie K Fuqua
- David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Yan Lu
- Iron Metabolism Laboratory, QIMR Berghofer Medical Research Institute, 300 Herston Road, Brisbane, QLD, 4006, Australia
- Mucosal Immunology Group, QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | - James F Collins
- Food Science and Human Nutrition Department, University of Florida, Gainsville, FL, USA
| | - Huijun Chen
- Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University, Nanjing, China
| | - Christopher D Vulpe
- Department of Physiological Sciences, University of Florida, Gainsville, FL, USA
| | - Gregory J Anderson
- Iron Metabolism Laboratory, QIMR Berghofer Medical Research Institute, 300 Herston Road, Brisbane, QLD, 4006, Australia.
- School of Chemistry and Molecular Bioscience, University of Queensland, Brisbane, Australia.
| | - David M Frazer
- Molecular Nutrition Laboratory, QIMR Berghofer Medical Research Institute, Brisbane, Australia
| |
Collapse
|
3
|
The Molecular Mechanism of Natural Products Activating Wnt/β-Catenin Signaling Pathway for Improving Hair Loss. LIFE (BASEL, SWITZERLAND) 2022; 12:life12111856. [PMID: 36430990 PMCID: PMC9693075 DOI: 10.3390/life12111856] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Revised: 10/28/2022] [Accepted: 11/09/2022] [Indexed: 11/16/2022]
Abstract
Hair loss, or alopecia, is a dermatological disorder that causes psychological stress and poor quality of life. Drug-based therapeutics such as finasteride and minoxidil have been clinically used to treat hair loss, but they have limitations due to their several side effects in patients. To solve this problem, there has been meaningful progress in elucidating the molecular mechanisms of hair growth and finding novel targets to develop therapeutics to treat it. Among various signaling pathways, Wnt/β-catenin plays an essential role in hair follicle development, the hair cycle, and regeneration. Thus, much research has demonstrated that various natural products worldwide promote hair growth by stimulating Wnt/β-catenin signaling. This review discusses the functional role of the Wnt/β-catenin pathway and its related signaling molecules. We also review the molecular mechanism of the natural products or compounds that activate Wnt/β-catenin signaling and provide insights into developing therapeutics or cosmeceuticals that treat hair loss.
Collapse
|
4
|
Li K, Zhu J, Li K, Liang W, Zhang J, Zhang Q, Jiao X, Wang X, Wei X, Yang J. High-fat diet blunts T-cell responsiveness in Nile tilapia. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2022; 135:104495. [PMID: 35863514 DOI: 10.1016/j.dci.2022.104495] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Revised: 06/26/2022] [Accepted: 07/14/2022] [Indexed: 06/15/2023]
Abstract
The reduced stress resistance and increased disease risk associated with high-fat diet (HFD) in animals have attracted increasing attention. However, the effects of HFD on adaptive immunity in early vertebrates, especially non-tetrapods, remain unknown. In this study, using Nile tilapia (Oreochromis niloticus) as a model, we investigated the effects of HFD on the primordial T-cell response in fish. Tilapia fed with an HFD for 8 weeks showed impaired lymphocyte homeostasis in the spleen, as indicated by the decreased number of both T and B lymphocytes and increased transcription of proinflammatory cytokines interferon-γ and interleukin-6. Moreover, lymphocytes isolated from HFD-fed fish or cultured in lipid-supplemented medium exhibited diminished T-cell activation in response to CD3ε monoclonal antibody stimulation. Moreover, HFD-fed tilapia infected by Aeromonas hydrophila showed decreased T-cell expansion, increased T-cell apoptosis, reduced granzyme B expression, and impaired infection elimination. Additionally, HFD attenuated adenosine 5'-monophosphate (AMP)-activated protein kinase (AMPK) activity in tilapia lymphocytes, which in turn upregulated fatty acid synthesis but downregulated fatty acid β-oxidation. Altogether, our results suggest that HFD impairs lymphocyte homeostasis and T cell-mediated adaptive immune response in tilapia, which may be associated with the abnormal lipid metabolism in lymphocytes. These findings thus provide a novel perspective for understanding the impact of HFD on the adaptive immune response of early vertebrates.
Collapse
Affiliation(s)
- Kunming Li
- State Key Laboratory of Estuarine and Coastal Research, School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Jiahua Zhu
- State Key Laboratory of Estuarine and Coastal Research, School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Kang Li
- State Key Laboratory of Estuarine and Coastal Research, School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Wei Liang
- State Key Laboratory of Estuarine and Coastal Research, School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Jiansong Zhang
- State Key Laboratory of Estuarine and Coastal Research, School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Qian Zhang
- State Key Laboratory of Estuarine and Coastal Research, School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Xinying Jiao
- State Key Laboratory of Estuarine and Coastal Research, School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Xiaodan Wang
- State Key Laboratory of Estuarine and Coastal Research, School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Xiumei Wei
- State Key Laboratory of Estuarine and Coastal Research, School of Life Sciences, East China Normal University, Shanghai, 200241, China.
| | - Jialong Yang
- State Key Laboratory of Estuarine and Coastal Research, School of Life Sciences, East China Normal University, Shanghai, 200241, China; Laboratory for Marine Biology and Biotechnology, Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266237, China.
| |
Collapse
|
5
|
Shi X, Chen Y, Yang K, Zhu Y, Ma Y, Liu Q, Wang J, Ni C, Zhang Y, Li H, Lin J, Wang J, Wu W. Disrupted citric acid metabolism inhibits hair growth. J Dermatol 2022; 49:1037-1048. [PMID: 35841232 DOI: 10.1111/1346-8138.16509] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Revised: 05/12/2022] [Accepted: 06/19/2022] [Indexed: 11/28/2022]
Abstract
Hair follicles (HFs) play an essential role in sustaining a persistent hair growth cycle. The activities of dermal papilla cells (DPCs) and other cells inside the HFs dominate the process of hair growth. However, the detailed molecular mechanisms remain largely unknown. To investigate the role of citric acid (CA) metabolism in hair growth, we evaluated the effect of citrate synthase (CS)-CA axis on hair growth in vivo and in vitro. Mice hair growth was evaluated by morphology and histopathology analysis. The inflammation and apoptosis levels in mice, HFs, and DPCs were detected by immunohistofluorescence, qPCR, ELISA, western blot, and TUNEL assay. Cell proliferation, cell cycle, and cell apoptosis in DPCs were analyzed by real-time cell analysis and flow cytometer. We found that subcutaneous injection of CA in mice caused significant hair growth suppression, skin lesion, inflammatory response, cell apoptosis, and promotion of catagen entry, compared with the saline control, by activating p-p65 and apoptosis signaling in an NLRP3-dependent manner. In cultured human HFs, CA attenuated the hair shaft production and accelerated HF catagen entry by regulating the above-mentioned pathways. Additionally, CA hampered the proliferation rate of DPCs via inducing cell apoptosis and cell cycle arrest. Considering that citrate synthase (CS) is responsible for CA production and is a rate-limiting enzyme of the tricarboxylic acid cycle, we also investigated the role of CS in CA metabolism and hair growth. As expected, knockdown of CS reduced CA production and reversed CA-induced hair growth inhibition, anagen shrink, inflammation, and apoptosis both in HFs and DPCs. Our experiments demonstrated that CS-CA axis serves as an important mediator and might be a potential therapeutic target in hair growth.
Collapse
Affiliation(s)
- Xiangguang Shi
- Department of Dermatology, Huashan Hospital and Human Phenome Institute, Fudan University, Shanghai, China
| | - Yahui Chen
- Department of Dermatology, Huashan Hospital and Human Phenome Institute, Fudan University, Shanghai, China.,Ministry of Education Key Laboratory of Contemporary Anthropology, Department of Anthropology and Human Genetics, School of Life Sciences, Fudan University, Shanghai, China
| | - Kai Yang
- Department of Dermatology, Jing'an District Central Hospital, Shanghai, China
| | - Yifei Zhu
- Department of Dermatology, Huashan Hospital and Human Phenome Institute, Fudan University, Shanghai, China
| | - Yanyun Ma
- Department of Dermatology, Huashan Hospital and Human Phenome Institute, Fudan University, Shanghai, China
| | - Qingmei Liu
- Department of Dermatology, Huashan Hospital and Human Phenome Institute, Fudan University, Shanghai, China
| | - Ji'an Wang
- Department of Dermatology, Huashan Hospital and Human Phenome Institute, Fudan University, Shanghai, China
| | - Chunya Ni
- Department of Dermatology, Jing'an District Central Hospital, Shanghai, China
| | - Yue Zhang
- Department of Dermatology, Huashan Hospital and Human Phenome Institute, Fudan University, Shanghai, China
| | - Haiyang Li
- Department of Dermatology, Huashan Hospital and Human Phenome Institute, Fudan University, Shanghai, China
| | - Jinran Lin
- Department of Dermatology, Huashan Hospital and Human Phenome Institute, Fudan University, Shanghai, China
| | - Jiucun Wang
- Department of Dermatology, Huashan Hospital and Human Phenome Institute, Fudan University, Shanghai, China.,Research Unit of Dissecting the Population Genetics and Developing New Technologies for Treatment and Prevention of Skin Phenotypes and Dermatological Diseases (2019RU058), Chinese Academy of Medical Sciences, Beijing, China
| | - Wenyu Wu
- Department of Dermatology, Huashan Hospital and Human Phenome Institute, Fudan University, Shanghai, China.,Department of Dermatology, Jing'an District Central Hospital, Shanghai, China
| |
Collapse
|
6
|
Katoh Y, Yaguchi T, Kubo A, Iwata T, Morii K, Kato D, Ohta S, Satomi R, Yamamoto Y, Oyamada Y, Ouchi K, Takahashi S, Ishioka C, Matoba R, Suematsu M, Kawakami Y. Inhibition of stearoyl-CoA desaturase 1 (SCD1) enhances the antitumor T cell response through regulating β-catenin signaling in cancer cells and ER stress in T cells and synergizes with anti-PD-1 antibody. J Immunother Cancer 2022; 10:jitc-2022-004616. [PMID: 35793868 PMCID: PMC9260842 DOI: 10.1136/jitc-2022-004616] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/19/2022] [Indexed: 11/26/2022] Open
Abstract
Background Understanding the mechanisms of non-T cell inflamed tumor microenvironment (TME) and their modulation are important to improve cancer immunotherapies such as immune checkpoint inhibitors. The involvement of various immunometabolisms has recently been indicated in the formation of immunosuppressive TME. In this study, we investigated the immunological roles of stearoyl-CoA desaturase 1 (SCD1), which is essential for fatty acid metabolism, in the cancer immune response. Methods We investigated the roles of SCD1 by inhibition with the chemical inhibitor or genetic manipulation in antitumor T cell responses and the therapeutic effect of anti-programmed cell death protein 1 (anti-PD-1) antibody using various mouse tumor models, and their cellular and molecular mechanisms. The roles of SCD1 in human cancers were also investigated by gene expression analyses of colon cancer tissues and by evaluating the related free fatty acids in sera obtained from patients with non-small cell lung cancer who were treated with anti-PD-1 antibody. Results Systemic administration of a SCD1 inhibitor in mouse tumor models enhanced production of CCL4 by cancer cells through reduction of Wnt/β-catenin signaling and by CD8+ effector T cells through reduction of endoplasmic reticulum stress. It in turn promoted recruitment of dendritic cells (DCs) into the tumors and enhanced the subsequent induction and tumor accumulation of antitumor CD8+ T cells. SCD1 inhibitor was also found to directly stimulate DCs and CD8+ T cells. Administration of SCD1 inhibitor or SCD1 knockout in mice synergized with an anti-PD-1 antibody for its antitumor effects in mouse tumor models. High SCD1 expression was observed in one of the non-T cell-inflamed subtypes in human colon cancer, and serum SCD1 related fatty acids were correlated with response rates and prognosis of patients with non-small lung cancer following anti-PD-1 antibody treatment. Conclusions SCD1 expressed in cancer cells and immune cells causes immunoresistant conditions, and its inhibition augments antitumor T cells and therapeutic effects of anti-PD-1 antibody. Therefore, SCD1 is an attractive target for the development of new diagnostic and therapeutic strategies to improve current cancer immunotherapies including immune checkpoint inhibitors.
Collapse
Affiliation(s)
- Yuki Katoh
- Division of Cellular Signaling, Institute for Advanced Medical Research, Keio University School of Medicine, Tokyo, Japan.,Division of Anatomical Science, Department of Functional Morphology, Nihon University School of Medicine, Tokyo, Japan
| | - Tomonori Yaguchi
- Division of Cellular Signaling, Institute for Advanced Medical Research, Keio University School of Medicine, Tokyo, Japan
| | - Akiko Kubo
- Department of Biochemistry, Keio University School of Medicine, Tokyo, Japan
| | - Takashi Iwata
- Division of Cellular Signaling, Institute for Advanced Medical Research, Keio University School of Medicine, Tokyo, Japan.,Department of Obstetrics and Gynecology, Keio University School of Medicine, Tokyo, Japan
| | - Kenji Morii
- Division of Cellular Signaling, Institute for Advanced Medical Research, Keio University School of Medicine, Tokyo, Japan
| | - Daiki Kato
- Division of Cellular Signaling, Institute for Advanced Medical Research, Keio University School of Medicine, Tokyo, Japan.,Laboratory of Veterinary Surgery, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo, Japan
| | - Shigeki Ohta
- Division of Cellular Signaling, Institute for Advanced Medical Research, Keio University School of Medicine, Tokyo, Japan
| | - Ryosuke Satomi
- National Hospital Organisation Tokyo Medical Center, Tokyo, Japan
| | - Yasuhiro Yamamoto
- Department of Respiratory Medicine, The University of Tokyo, Tokyo, Japan
| | | | - Kota Ouchi
- Department of Medical Oncology, Tohoku University Hospital, Sendai, Japan.,Department of Clinical Oncology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Shin Takahashi
- Department of Medical Oncology, Tohoku University Hospital, Sendai, Japan.,Department of Clinical Oncology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Chikashi Ishioka
- Department of Medical Oncology, Tohoku University Hospital, Sendai, Japan.,Department of Clinical Oncology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | | | - Makoto Suematsu
- Department of Biochemistry, Keio University School of Medicine, Tokyo, Japan
| | - Yutaka Kawakami
- Division of Cellular Signaling, Institute for Advanced Medical Research, Keio University School of Medicine, Tokyo, Japan .,Department of Immunology, International University of Health and Welfare, Chiba, Japan
| |
Collapse
|
7
|
Nuber S, Chung CY, Tardiff DF, Bechade PA, McCaffery TD, Shimanaka K, Choi J, Chang B, Raja W, Neves E, Burke C, Jiang X, Xu P, Khurana V, Dettmer U, Fanning S, Rhodes KJ, Selkoe DJ, Scannevin RH. A Brain-Penetrant Stearoyl-CoA Desaturase Inhibitor Reverses α-Synuclein Toxicity. Neurotherapeutics 2022; 19:1018-1036. [PMID: 35445353 PMCID: PMC9294123 DOI: 10.1007/s13311-022-01199-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/04/2022] [Indexed: 12/01/2022] Open
Abstract
Increasing evidence has shown that Parkinson's disease (PD) impairs midbrain dopaminergic, cortical and other neuronal subtypes in large part due to the build-up of lipid- and vesicle-rich α-synuclein (αSyn) cytotoxic inclusions. We previously identified stearoyl-CoA desaturase (SCD) as a potential therapeutic target for synucleinopathies. A brain-penetrant SCD inhibitor, YTX-7739, was developed and has entered Phase 1 clinical trials. Here, we report the efficacy of YTX-7739 in reversing pathological αSyn phenotypes in various in vitro and in vivo PD models. In cell-based assays, YTX-7739 decreased αSyn-mediated neuronal death, reversed the abnormal membrane interaction of amplified E46K ("3K") αSyn, and prevented pathological phenotypes in A53T and αSyn triplication patient-derived neurospheres, including dysregulated fatty acid profiles and pS129 αSyn accumulation. In 3K PD-like mice, YTX-7739 crossed the blood-brain barrier, decreased unsaturated fatty acids, and prevented progressive motor deficits. Both YTX-7739 treatment and decreasing SCD activity through deletion of one copy of the SCD1 gene (SKO) restored the physiological αSyn tetramer-to-monomer ratio, dopaminergic integrity, and neuronal survival in 3K αSyn mice. YTX-7739 efficiently reduced pS129 + and PK-resistant αSyn in both human wild-type αSyn and 3K mutant mice similar to the level of 3K-SKO. Together, these data provide further validation of SCD as a PD therapeutic target and YTX-7739 as a clinical candidate for treating human α-synucleinopathies.
Collapse
Affiliation(s)
- Silke Nuber
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women Hospital and Harvard Medical School, 60 Fenwood Rd, MA, 02115, Boston, US.
| | - Chee Yeun Chung
- Yumanity Therapeutics, 40 Guest Street, Boston, MA, 02135, US.
| | | | - Pascal A Bechade
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women Hospital and Harvard Medical School, 60 Fenwood Rd, MA, 02115, Boston, US
| | - Thomas D McCaffery
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women Hospital and Harvard Medical School, 60 Fenwood Rd, MA, 02115, Boston, US
| | - Kazuma Shimanaka
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women Hospital and Harvard Medical School, 60 Fenwood Rd, MA, 02115, Boston, US
| | - Jeonghoon Choi
- Yumanity Therapeutics, 40 Guest Street, Boston, MA, 02135, US
| | - Belle Chang
- Yumanity Therapeutics, 40 Guest Street, Boston, MA, 02135, US
- iNeuro Therapeutics, Cambridge, MA, 02116, US
| | - Waseem Raja
- Yumanity Therapeutics, 40 Guest Street, Boston, MA, 02135, US
| | - Esther Neves
- Yumanity Therapeutics, 40 Guest Street, Boston, MA, 02135, US
| | | | - Xin Jiang
- Yumanity Therapeutics, 40 Guest Street, Boston, MA, 02135, US
| | - Ping Xu
- Yumanity Therapeutics, 40 Guest Street, Boston, MA, 02135, US
| | - Vikram Khurana
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women Hospital and Harvard Medical School, 60 Fenwood Rd, MA, 02115, Boston, US
- Yumanity Therapeutics, 40 Guest Street, Boston, MA, 02135, US
| | - Ulf Dettmer
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women Hospital and Harvard Medical School, 60 Fenwood Rd, MA, 02115, Boston, US
| | - Saranna Fanning
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women Hospital and Harvard Medical School, 60 Fenwood Rd, MA, 02115, Boston, US
| | - Kenneth J Rhodes
- Yumanity Therapeutics, 40 Guest Street, Boston, MA, 02135, US
- Wave Life Sciences, 733 Concord Ave, Cambridge, MA, 02138, US
| | - Dennis J Selkoe
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women Hospital and Harvard Medical School, 60 Fenwood Rd, MA, 02115, Boston, US
| | - Robert H Scannevin
- Yumanity Therapeutics, 40 Guest Street, Boston, MA, 02135, US
- Verge Genomics, 2 Tower Pl, South San Francisco, CA, 94080, US
| |
Collapse
|
8
|
Nuber S, Nam AY, Rajsombath MM, Cirka H, Hronowski X, Wang J, Hodgetts K, Kalinichenko LS, Müller CP, Lambrecht V, Winkler J, Weihofen A, Imberdis T, Dettmer U, Fanning S, Selkoe DJ. A Stearoyl-Coenzyme A Desaturase Inhibitor Prevents Multiple Parkinson Disease Phenotypes in α-Synuclein Mice. Ann Neurol 2020; 89:74-90. [PMID: 32996158 PMCID: PMC7756464 DOI: 10.1002/ana.25920] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2020] [Revised: 09/25/2020] [Accepted: 09/25/2020] [Indexed: 12/20/2022]
Abstract
Objective Parkinson disease (PD) has useful symptomatic treatments that do not slow the neurodegenerative process, and no significant disease‐modifying treatments are approved. A key therapeutic target in PD is α‐synuclein (αS), which is both genetically implicated and accumulates in Lewy bodies rich in vesicles and other lipid membranes. Reestablishing αS homeostasis is a central goal in PD. Based on previous lipidomic analyses, we conducted a mouse trial of a stearoyl–coenzyme A desaturase (SCD) inhibitor (“5b”) that prevented αS‐positive vesicular inclusions and cytotoxicity in cultured human neurons. Methods Oral dosing and brain activity of 5b were established in nontransgenic mice. 5b in drinking water was given to mice expressing wild‐type human αS (WT) or an amplified familial PD αS mutation (E35K + E46K + E61K ["3K"]) beginning near the onset of nigral and cortical neurodegeneration and the robust PD‐like motor syndrome in 3K. Motor phenotypes, brain cytopathology, and SCD‐related lipid changes were quantified in 5b‐ versus placebo‐treated mice. Outcomes were compared to effects of crossing 3K to SCD1−/− mice. Results 5b treatment reduced αS hyperphosphorylation in E46K‐expressing human neurons, in 3K neural cultures, and in both WT and 3K αS mice. 5b prevented subtle gait deficits in WT αS mice and the PD‐like resting tremor and progressive motor decline of 3K αS mice. 5b also increased αS tetramers and reduced proteinase K‐resistant lipid‐rich aggregates. Similar benefits accrued from genetically deleting 1 SCD allele, providing target validation. Interpretation Prolonged reduction of brain SCD activity prevented PD‐like neuropathology in multiple PD models. Thus, an orally available SCD inhibitor potently ameliorates PD phenotypes, positioning this approach to treat human α‐synucleinopathies. ANN NEUROL 2021;89:74–90
Collapse
Affiliation(s)
- Silke Nuber
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Alice Y Nam
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Molly M Rajsombath
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Haley Cirka
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | | | - Junmin Wang
- Chemical Biology & Proteomics, Biogen, Cambridge, MA, USA
| | - Kevin Hodgetts
- Laboratory for Drug Discovery in Neurodegeneration, Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Liubov S Kalinichenko
- Department of Psychiatry and Psychotherapy, University Clinic, Friedrich-Alexander University Erlangen-Nuremberg, Erlangen, Germany
| | - Christian P Müller
- Department of Psychiatry and Psychotherapy, University Clinic, Friedrich-Alexander University Erlangen-Nuremberg, Erlangen, Germany
| | - Vera Lambrecht
- Division of Molecular Neurology, Friedrich-Alexander University Erlangen-Nuremberg, Erlangen, Germany
| | - Jürgen Winkler
- Division of Molecular Neurology, Friedrich-Alexander University Erlangen-Nuremberg, Erlangen, Germany
| | - Andreas Weihofen
- Neurodegenerative Diseases Research Unit, Biogen, Cambridge, MA, USA
| | - Thibaut Imberdis
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Ulf Dettmer
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Saranna Fanning
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Dennis J Selkoe
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| |
Collapse
|
9
|
Wang H, Humbatova A, Liu Y, Qin W, Lee M, Cesarato N, Kortüm F, Kumar S, Romano MT, Dai S, Mo R, Sivalingam S, Motameny S, Wu Y, Wang X, Niu X, Geng S, Bornholdt D, Kroisel PM, Tadini G, Walter SD, Hauck F, Girisha KM, Calza AM, Bottani A, Altmüller J, Buness A, Yang S, Sun X, Ma L, Kutsche K, Grzeschik KH, Betz RC, Lin Z. Mutations in SREBF1, Encoding Sterol Regulatory Element Binding Transcription Factor 1, Cause Autosomal-Dominant IFAP Syndrome. Am J Hum Genet 2020; 107:34-45. [PMID: 32497488 DOI: 10.1016/j.ajhg.2020.05.006] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Accepted: 05/04/2020] [Indexed: 12/14/2022] Open
Abstract
IFAP syndrome is a rare genetic disorder characterized by ichthyosis follicularis, atrichia, and photophobia. Previous research found that mutations in MBTPS2, encoding site-2-protease (S2P), underlie X-linked IFAP syndrome. The present report describes the identification via whole-exome sequencing of three heterozygous mutations in SREBF1 in 11 unrelated, ethnically diverse individuals with autosomal-dominant IFAP syndrome. SREBF1 encodes sterol regulatory element-binding protein 1 (SREBP1), which promotes the transcription of lipogenes involved in the biosynthesis of fatty acids and cholesterols. This process requires cleavage of SREBP1 by site-1-protease (S1P) and S2P and subsequent translocation into the nucleus where it binds to sterol regulatory elements (SRE). The three detected SREBF1 mutations caused substitution or deletion of residues 527, 528, and 530, which are crucial for S1P cleavage. In vitro investigation of SREBP1 variants demonstrated impaired S1P cleavage, which prohibited nuclear translocation of the transcriptionally active form of SREBP1. As a result, SREBP1 variants exhibited significantly lower transcriptional activity compared to the wild-type, as demonstrated via luciferase reporter assay. RNA sequencing of the scalp skin from IFAP-affected individuals revealed a dramatic reduction in transcript levels of low-density lipoprotein receptor (LDLR) and of keratin genes known to be expressed in the outer root sheath of hair follicles. An increased rate of in situ keratinocyte apoptosis, which might contribute to skin hyperkeratosis and hypotrichosis, was also detected in scalp samples from affected individuals. Together with previous research, the present findings suggest that SREBP signaling plays an essential role in epidermal differentiation, skin barrier formation, hair growth, and eye function.
Collapse
Affiliation(s)
- Huijun Wang
- 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 100034, China
| | - Aytaj Humbatova
- Institute of Human Genetics, University of Bonn, Medical Faculty & University Hospital Bonn, 53127 Bonn, Germany
| | - Yuanxiang Liu
- Department of Dermatology, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing 100045, China
| | - Wen Qin
- 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 100034, China
| | - Mingyang Lee
- 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 100034, China
| | - Nicole Cesarato
- Institute of Human Genetics, University of Bonn, Medical Faculty & University Hospital Bonn, 53127 Bonn, Germany
| | - Fanny Kortüm
- Institute of Human Genetics, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Sheetal Kumar
- Institute of Human Genetics, University of Bonn, Medical Faculty & University Hospital Bonn, 53127 Bonn, Germany
| | - Maria Teresa Romano
- Institute of Human Genetics, University of Bonn, Medical Faculty & University Hospital Bonn, 53127 Bonn, Germany
| | - Shangzhi Dai
- 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 100034, China
| | - Ran Mo
- 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 100034, China
| | - Sugirthan Sivalingam
- Institute for Medical Biometry, Informatics and Epidemiology, University of Bonn, Medical Faculty, 53127 Bonn, Germany; Institute for Genomic Statistics and Bioinformatics, University of Bonn, Medical Faculty, 53127 Bonn, Germany
| | - Susanne Motameny
- Cologne Center for Genomics, University of Cologne, 50931 Cologne, Germany
| | - Yuan Wu
- Department of Ophthalmology, Peking University First Hospital, Beijing 100034, China
| | - Xiaopeng Wang
- Department of Dermatology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004, China
| | - Xinwu Niu
- Department of Dermatology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004, China
| | - Songmei Geng
- Department of Dermatology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004, China
| | - Dorothea Bornholdt
- Centre for Human Genetics, University of Marburg, 35033 Marburg, Germany
| | - Peter M Kroisel
- Institute of Human Genetics, Medical University of Graz, 8010 Graz, Austria
| | - Gianluca Tadini
- Pediatric Dermatology Unit, Department of Pathophysiology and Transplantation, Fondazione IRCCS Ca' Granda - Ospedale Maggiore Policlinico, University of Milan, Milan, Italy
| | - Scott D Walter
- Retina Consultants, P.C., 43 Woodland Street, Suite 100, Hartford, CT 06105, USA
| | - Fabian Hauck
- Department of Pediatrics, University Hospital Carl Gustav Carus, TU Dresden, 01307 Dresden, Germany; Department of Pediatrics, Dr. von Hauner Children's Hospital, University Hospital, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Katta M Girisha
- Department of Medical Genetics, Kasturba Medical College, Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - Anne-Marie Calza
- Department of Dermatology and Venereology, Geneva University Hospitals, 1205 Geneva, Switzerland
| | - Armand Bottani
- Service of Genetic Medicine, Geneva University Hospitals, 1205 Geneva, Switzerland
| | - Janine Altmüller
- Cologne Center for Genomics, University of Cologne, 50931 Cologne, Germany
| | - Andreas Buness
- Institute for Medical Biometry, Informatics and Epidemiology, University of Bonn, Medical Faculty, 53127 Bonn, Germany; Institute for Genomic Statistics and Bioinformatics, University of Bonn, Medical Faculty, 53127 Bonn, Germany
| | - Shuxia Yang
- 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 100034, China
| | - Xiujuan Sun
- Department of Dermatology, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing 100045, China
| | - Lin Ma
- Department of Dermatology, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing 100045, China
| | - Kerstin Kutsche
- Institute of Human Genetics, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | | | - Regina C Betz
- Institute of Human Genetics, University of Bonn, Medical Faculty & University Hospital Bonn, 53127 Bonn, Germany.
| | - Zhimiao Lin
- 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 100034, China.
| |
Collapse
|
10
|
Li H, Feng Z, He ML. Lipid metabolism alteration contributes to and maintains the properties of cancer stem cells. Theranostics 2020; 10:7053-7069. [PMID: 32641978 PMCID: PMC7330842 DOI: 10.7150/thno.41388] [Citation(s) in RCA: 100] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Accepted: 04/28/2020] [Indexed: 12/11/2022] Open
Abstract
Lipids, the basic components of the cell membrane, execute fundamental roles in almost all the cell activities including cell-cell recognition, signalling transduction and energy supplies. Lipid metabolism is elementary for life sustentation that balances activity between synthesis and degradation. An accumulating amount of data has indicated abnormal lipid metabolism in cancer stem cells (CSCs), and that the alteration of lipid metabolism exerts a great impact on CSCs' properties such as the capability of self-renewal, differentiation, invasion, metastasis, and drug sensitivity and resistance. CSCs' formation and maintenance cannot do without the regulation of fatty acids and cholesterol. In normal cells and embryonic development, fatty acids and cholesterol metabolism are regulated by some important signalling pathways (such as Hedgehog, Notch, Wnt signalling pathways); these signalling pathways also play crucial roles in initiating and/or maintaining CSCs' properties, and such signalling is shown to be commonly modulated by the abnormal lipid metabolism in CSCs; on the other hand, the altered lipid metabolism in turn modifies the cell signalling and generates additional impacts on CSCs. Metabolic rewiring is considered as an ideal hallmark of CSCs, and metabolic alterations would be promising therapeutic targets of CSCs for aggressive tumors. In this review, we summarize the most updated findings of lipid metabolic abnormalities in CSCs and prospect the potential applications of targeting lipid metabolism for anticancer treatment.
Collapse
|
11
|
Clayton R, Göbel K, Niessen C, Paus R, Steensel M, Lim X. Homeostasis of the sebaceous gland and mechanisms of acne pathogenesis. Br J Dermatol 2019; 181:677-690. [DOI: 10.1111/bjd.17981] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/06/2019] [Indexed: 12/13/2022]
Affiliation(s)
- R.W. Clayton
- Skin Research Institute of Singapore Agency for Science, Technology and Research (A*STAR) Singapore
- Centre for Dermatology Research University of Manchester, and NIHR Manchester Biomedical Research Centre Manchester U.K
| | - K. Göbel
- Skin Research Institute of Singapore Agency for Science, Technology and Research (A*STAR) Singapore
- Department of Dermatology Cologne Excellence Cluster on Stress Responses in Aging Associated Diseases (CECAD), and Centre for Molecular Medicine Cologne The University of Cologne Germany
| | - C.M. Niessen
- Department of Dermatology Cologne Excellence Cluster on Stress Responses in Aging Associated Diseases (CECAD), and Centre for Molecular Medicine Cologne The University of Cologne Germany
| | - R. Paus
- Centre for Dermatology Research University of Manchester, and NIHR Manchester Biomedical Research Centre Manchester U.K
- Department of Dermatology and Cutaneous Surgery University of Miami Miller School of Medicine Miami FL U.S.A
| | - M.A.M. Steensel
- Skin Research Institute of Singapore Agency for Science, Technology and Research (A*STAR) Singapore
- Lee Kong Chian School of Medicine Nanyang Technological University Singapore
| | - X. Lim
- Skin Research Institute of Singapore Agency for Science, Technology and Research (A*STAR) Singapore
- Lee Kong Chian School of Medicine Nanyang Technological University Singapore
| |
Collapse
|
12
|
Therapeutic targeting of lipid synthesis metabolism for selective elimination of cancer stem cells. Arch Pharm Res 2018; 42:25-39. [PMID: 30536027 DOI: 10.1007/s12272-018-1098-z] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Accepted: 12/01/2018] [Indexed: 02/06/2023]
Abstract
Cancer stem cells (CSCs) are believed to have an essential role in tumor resistance and metastasis; however, no therapeutic strategy for the selective elimination of CSCs has been established. Recently, several studies have shown that the metabolic regulation for ATP synthesis and biological building block generation in CSCs are different from that in bulk cancer cells and rather similar to that in normal tissue stem cells. To take advantage of this difference for CSC elimination therapy, many studies have tested the effect of blocking these metabolism. Two specific processes for lipid biosynthesis, i.e., fatty acid unsaturation and cholesterol biosynthesis, have been shown to be very effective and selective for CSC targets. In this review, lipid metabolism specific to CSCs are summarized. In addition, how monounsaturated fatty acid and cholesterol synthesis may contribute to CSC maintenance are discussed. Specifically, the molecular mechanism required for lipid synthesis and essential for stem cell biology is highlighted. The limit and preview of the lipid metabolism targeting for CSCs are also discussed.
Collapse
|
13
|
Jain P, Nattakom M, Holowka D, Wang DH, Thomas Brenna J, Ku AT, Nguyen H, Ibrahim SF, Tumbar T. Runx1 Role in Epithelial and Cancer Cell Proliferation Implicates Lipid Metabolism and Scd1 and Soat1 Activity. Stem Cells 2018; 36:1603-1616. [PMID: 29938858 PMCID: PMC6202256 DOI: 10.1002/stem.2868] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2018] [Revised: 06/17/2018] [Accepted: 06/25/2018] [Indexed: 01/12/2023]
Abstract
The role of lipid metabolism in epithelial stem cell (SC) function and carcinogenesis is poorly understood. The transcription factor Runx1 is known to regulate proliferation in mouse epithelial hair follicle (HF) SCs in vivo and in several mouse and human epithelial cancers. We found a novel subset of in vivo Runx1 HFSC target genes related to lipid metabolism and demonstrated changes in distinct classes of lipids driven by Runx1. Inhibition of lipid-enzymes Scd1 and Soat1 activity synergistically reduces proliferation of mouse skin epithelial cells and of human skin and oral squamous cell carcinoma cultured lines. Varying Runx1 levels induces changes in skin monounsaturated fatty acids (e.g., oleate, a product of Scd1) as shown by our lipidome analysis. Furthermore, varying Runx1 levels, the inhibition of Scd1, or the addition of Scd1-product oleate, individually affects the plasma membrane organization (or fluidity) in mouse keratinocytes. These factors also affect the strength of signal transduction through the membranes for Wnt, a pathway that promotes epithelial (cancer) cell proliferation and HFSC activation. Our working model is that HFSC factor Runx1 modulates the fatty acid production, which affects membrane organization, facilitating signal transduction for rapid proliferation of normal and cancer epithelial cells. Stem Cells 2018;36:1603-1616.
Collapse
Affiliation(s)
- Prachi Jain
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, New York, USA
| | - Mary Nattakom
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, New York, USA
| | - David Holowka
- Department of Chemistry & Chemical Biology, Cornell University, Ithaca, New York, USA
| | - Dong Hao Wang
- Division of Nutritional Sciences, Cornell University, Ithaca, New York, USA
- Dell Pediatric Research Institute, University of Texas at Austin, Austin, Texas, USA
| | - J Thomas Brenna
- Division of Nutritional Sciences, Cornell University, Ithaca, New York, USA
- Dell Pediatric Research Institute, University of Texas at Austin, Austin, Texas, USA
| | - Amy Tsu Ku
- Interdepartmental Program in Translational Biology and Molecular Medicine, Baylor College of Medicine, Houston, Texas, USA
| | - Hoang Nguyen
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas, USA
| | - Sherrif F Ibrahim
- Department of Dermatology, School of Medicines & Dentistry, University of Rochester Medical Center, Rochester, New York, USA
| | - Tudorita Tumbar
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, New York, USA
| |
Collapse
|
14
|
Sundberg JP, Hordinsky MK, Bergfeld W, Lenzy YM, McMichael AJ, Christiano AM, McGregor T, Stenn KS, Sivamani RK, Pratt CH, King LE. Cicatricial Alopecia Research Foundation meeting, May 2016: Progress towards the diagnosis, treatment and cure of primary cicatricial alopecias. Exp Dermatol 2018; 27:302-310. [DOI: 10.1111/exd.13495] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/06/2018] [Indexed: 12/11/2022]
Affiliation(s)
- John P. Sundberg
- The Jackson Laboratory; Bar Harbor ME USA
- Department of Dermatology; Vanderbilt University Medical Center; Nashville TN USA
| | | | - Wilma Bergfeld
- Department of Dermatology and Pathology; Cleveland Clinic; Cleveland OH USA
| | | | | | - Angela M. Christiano
- Department of Dermatology; Columbia University College of Physicians & Surgeons; New York NY USA
| | - Tracy McGregor
- Clinical Genetics; Vanderbilt University Medical Center; Nashville TN USA
| | | | - Raja K. Sivamani
- Department of Dermatology; University of California, Davis; Sacramento CA USA
| | | | - Lloyd E. King
- Department of Dermatology; Vanderbilt University Medical Center; Nashville TN USA
| |
Collapse
|