1
|
Luo W, Xu C, Li L, Ji Y, Wang Y, Li Y, Ye Y. Perfluoropentane-based oxygen-loaded nanodroplets reduce microglial activation through metabolic reprogramming. Neural Regen Res 2025; 20:1178-1191. [PMID: 38989955 PMCID: PMC11438333 DOI: 10.4103/nrr.nrr-d-23-01299] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Accepted: 02/05/2024] [Indexed: 07/12/2024] Open
Abstract
JOURNAL/nrgr/04.03/01300535-202504000-00032/figure1/v/2024-07-06T104127Z/r/image-tiff Microglia, the primary immune cells within the brain, have gained recognition as a promising therapeutic target for managing neurodegenerative diseases within the central nervous system, including Parkinson's disease. Nanoscale perfluorocarbon droplets have been reported to not only possess a high oxygen-carrying capacity, but also exhibit remarkable anti-inflammatory properties. However, the role of perfluoropentane in microglia-mediated central inflammatory reactions remains poorly understood. In this study, we developed perfluoropentane-based oxygen-loaded nanodroplets (PFP-OLNDs) and found that pretreatment with these droplets suppressed the lipopolysaccharide-induced activation of M1-type microglia in vitro and in vivo, and suppressed microglial activation in a mouse model of Parkinson's disease. Microglial suppression led to a reduction in the inflammatory response, oxidative stress, and cell migration capacity in vitro. Consequently, the neurotoxic effects were mitigated, which alleviated neuronal degeneration. Additionally, ultrahigh-performance liquid chromatography-tandem mass spectrometry showed that the anti-inflammatory effects of PFP-OLNDs mainly resulted from the modulation of microglial metabolic reprogramming. We further showed that PFP-OLNDs regulated microglial metabolic reprogramming through the AKT-mTOR-HIF-1α pathway. Collectively, our findings suggest that the novel PFP-OLNDs constructed in this study alleviate microglia-mediated central inflammatory reactions through metabolic reprogramming.
Collapse
Affiliation(s)
- Wanxian Luo
- Department of Medicine Ultrasonics, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong Province, China
| | - Chuanhui Xu
- Institute of Neuroscience, Department of Neurosurgery, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong Province, China
| | - Linxi Li
- Institute of Neuroscience, Department of Neurosurgery, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong Province, China
| | - Yunxiang Ji
- Institute of Neuroscience, Department of Neurosurgery, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong Province, China
| | - Yezhong Wang
- Institute of Neuroscience, Department of Neurosurgery, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong Province, China
| | - Yingjia Li
- Department of Medicine Ultrasonics, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong Province, China
| | - Yongyi Ye
- Institute of Neuroscience, Department of Neurosurgery, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong Province, China
| |
Collapse
|
2
|
Bekhbat M. Glycolytic metabolism: Food for immune cells, fuel for depression? Brain Behav Immun Health 2024; 40:100843. [PMID: 39263313 PMCID: PMC11387811 DOI: 10.1016/j.bbih.2024.100843] [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: 12/17/2023] [Revised: 07/16/2024] [Accepted: 08/10/2024] [Indexed: 09/13/2024] Open
Abstract
Inflammation is one biological pathway thought to impact the brain to contribute to major depressive disorder (MDD) and is reliably associated with resistance to standard antidepressant treatments. While peripheral immune cells, particularly monocytes, have been associated with aspects of increased inflammation in MDD and symptom severity, significant gaps in knowledge exist regarding the mechanisms by which these cells are activated to contribute to behavioral symptoms in MDD. One concept that has gained recent appreciation is that metabolic rewiring to glycolysis in activated myeloid cells plays a crucial role in facilitating these cells' pro-inflammatory functions, which may underlie myeloid contribution to systemic inflammation and its effects on the brain. Given emerging evidence from translational studies of depression that peripheral monocytes exhibit signs of glycolytic activation, better understanding the immunometabolic phenotypes of monocytes which are known to be elevated in MDD with high inflammation is a critical step toward comprehending and treating the impact of inflammation on the brain. This narrative review examines the extant literature on glycolytic metabolism of circulating monocytes in depression and discusses the functional implications of immunometabolic shifts at both cellular and systemic levels. Additionally, it proposes potential therapeutic applications of existing immunomodulators that target glycolysis and related metabolic pathways in order to reverse the impact of elevated inflammation on the brain and depressive symptoms.
Collapse
Affiliation(s)
- Mandakh Bekhbat
- Department of Psychiatry and Behavioral Sciences, Emory University, Atlanta, GA, 30322, USA
| |
Collapse
|
3
|
Stoessel MB, Stowell RD, Lowery RL, Le L, Vu AN, Whitelaw BS, Majewska AK. The effects of P2Y12 loss on microglial gene expression, dynamics, and injury response in the cerebellum and cerebral cortex. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.25.614526. [PMID: 39386439 PMCID: PMC11463386 DOI: 10.1101/2024.09.25.614526] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/12/2024]
Abstract
Despite the emerging consensus that microglia are critical to physiological and pathological brain function, it is unclear how microglial roles and their underlying mechanisms differ between brain regions. Microglia throughout the brain express common markers, such as the purinergic receptor P2Y12, that delineate them from peripheral macrophages. P2Y12 is a critical sensor of injury but also contributes to the sensing of neuronal activity and remodeling of synapses, with microglial loss of P2Y12 resulting in behavioral deficits. P2Y12 has largely been studied in cortical microglia, despite the fact that a growing body of evidence suggests that microglia exhibit a high degree of regional specialization. Cerebellar microglia, in particular, exhibit transcriptional, epigenetic, and functional profiles that set them apart from their better studied cortical and hippocampal counterparts. Here, we demonstrate that P2Y12 deficiency does not alter the morphology, distribution, or dynamics of microglia in the cerebellum. In fact, loss of P2Y12 does little to disturb the distinct transcriptomic profiles of cortical and cerebellar microglia. However, unlike in cortex, P2Y12 is not required for a full microglial response to focal injury, suggesting that cerebellar and cortical microglia use different cues to respond to injury. Finally, we show that P2Y12 deficiency impairs cerebellar learning in a delay eyeblink conditioning task, a common test of cerebellar plasticity and circuit function. Our findings suggest not only region-specific roles of microglial P2Y12 signaling in the focal injury response, but also indicate a conserved role for P2Y12 in microglial modulation of plasticity across regions.
Collapse
|
4
|
Ryan AT, Kim M, Lim K. Immune Cell Migration to Cancer. Cells 2024; 13:844. [PMID: 38786066 PMCID: PMC11120175 DOI: 10.3390/cells13100844] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2024] [Revised: 04/27/2024] [Accepted: 05/03/2024] [Indexed: 05/25/2024] Open
Abstract
Immune cell migration is required for the development of an effective and robust immune response. This elegant process is regulated by both cellular and environmental factors, with variables such as immune cell state, anatomical location, and disease state that govern differences in migration patterns. In all cases, a major factor is the expression of cell surface receptors and their cognate ligands. Rapid adaptation to environmental conditions partly depends on intrinsic cellular immune factors that affect a cell's ability to adjust to new environment. In this review, we discuss both myeloid and lymphoid cells and outline key determinants that govern immune cell migration, including molecules required for immune cell adhesion, modes of migration, chemotaxis, and specific chemokine signaling. Furthermore, we summarize tumor-specific elements that contribute to immune cell trafficking to cancer, while also exploring microenvironment factors that can alter these cellular dynamics within the tumor in both a pro and antitumor fashion. Specifically, we highlight the importance of the secretome in these later aspects. This review considers a myriad of factors that impact immune cell trajectory in cancer. We aim to highlight the immunotherapeutic targets that can be harnessed to achieve controlled immune trafficking to and within tumors.
Collapse
Affiliation(s)
- Allison T. Ryan
- Department of Microbiology and Immunology, University of Rochester, Rochester, NY 14642, USA; (A.T.R.); (M.K.)
- David H. Smith Center for Vaccine Biology and Immunology, University of Rochester, Rochester, NY 14642, USA
| | - Minsoo Kim
- Department of Microbiology and Immunology, University of Rochester, Rochester, NY 14642, USA; (A.T.R.); (M.K.)
- David H. Smith Center for Vaccine Biology and Immunology, University of Rochester, Rochester, NY 14642, USA
| | - Kihong Lim
- Department of Microbiology and Immunology, University of Rochester, Rochester, NY 14642, USA; (A.T.R.); (M.K.)
- David H. Smith Center for Vaccine Biology and Immunology, University of Rochester, Rochester, NY 14642, USA
| |
Collapse
|
5
|
Lu J, Chen XZ, Liu Y, Liu YJ, Liu B. Trends in confinement-induced cell migration and multi-omics analysis. Anal Bioanal Chem 2024; 416:2107-2115. [PMID: 38135761 DOI: 10.1007/s00216-023-05109-4] [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: 10/14/2023] [Revised: 11/24/2023] [Accepted: 11/30/2023] [Indexed: 12/24/2023]
Abstract
Cell migration is an essential manner of different cell lines that are involved in embryological development, immune responses, tumorigenesis, and metastasis in vivo. Physical confinement derived from crowded tissue microenvironments has pivotal effects on migratory behaviors. Distinct migration modes under a heterogeneous extracellular matrix (ECM) have been extensively studied, uncovering potential molecular mechanisms involving a series of biological processes. Significantly, multi-omics strategies have been launched to provide multi-angle views of complex biological phenomena, facilitating comprehensive insights into molecular regulatory networks during cell migration. In this review, we describe biomimetic devices developed to explore the migratory behaviors of cells induced by different types of confined microenvironments in vitro. We also discuss the results of multi-omics analysis of intrinsic molecular alterations and critical pathway dysregulations of cell migration under heterogeneous microenvironments, highlighting the significance of physical confinement-triggered intracellular signal transduction in order to regulate cellular behaviors. Finally, we discuss both the challenges and promise of mechanistic analysis in confinement-induced cell migration, promoting the development of early diagnosis and precision therapeutics.
Collapse
Affiliation(s)
- Jiayin Lu
- Department of ChemistryState Key Lab of Molecular Engineering of PolymersShanghai Key Laboratory of Medical Epigenetics, Institutes of Biomedical Sciences, Shanghai Stomatological HospitalShanghai Xuhui Central Hospital, Zhongshan-Xuhui HospitalFudan University, Shanghai, China
| | - Xue-Zhu Chen
- Department of ChemistryState Key Lab of Molecular Engineering of PolymersShanghai Key Laboratory of Medical Epigenetics, Institutes of Biomedical Sciences, Shanghai Stomatological HospitalShanghai Xuhui Central Hospital, Zhongshan-Xuhui HospitalFudan University, Shanghai, China
| | - Yixin Liu
- Department of ChemistryState Key Lab of Molecular Engineering of PolymersShanghai Key Laboratory of Medical Epigenetics, Institutes of Biomedical Sciences, Shanghai Stomatological HospitalShanghai Xuhui Central Hospital, Zhongshan-Xuhui HospitalFudan University, Shanghai, China
| | - Yan-Jun Liu
- Department of ChemistryState Key Lab of Molecular Engineering of PolymersShanghai Key Laboratory of Medical Epigenetics, Institutes of Biomedical Sciences, Shanghai Stomatological HospitalShanghai Xuhui Central Hospital, Zhongshan-Xuhui HospitalFudan University, Shanghai, China.
| | - Baohong Liu
- Department of ChemistryState Key Lab of Molecular Engineering of PolymersShanghai Key Laboratory of Medical Epigenetics, Institutes of Biomedical Sciences, Shanghai Stomatological HospitalShanghai Xuhui Central Hospital, Zhongshan-Xuhui HospitalFudan University, Shanghai, China.
| |
Collapse
|
6
|
Tang JS, Stephens R, Li Y, Cait A, Gell K, Faulkner S, Grooby A, Herst PM, O'Sullivan D, Gasser O. Polyphenol and glucosinolate-derived AhR modulators regulate GPR15 expression on human CD4+ T cells. J Nutr Biochem 2023; 122:109456. [PMID: 37788725 DOI: 10.1016/j.jnutbio.2023.109456] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 08/24/2023] [Accepted: 09/27/2023] [Indexed: 10/05/2023]
Abstract
Diets high in fruit and vegetables are perceived to be beneficial for intestinal homeostasis, in health as well as in the context of inflammatory bowel diseases (IBDs). Recent breakthroughs in the field of immunology have highlighted the importance of the ligand-activated transcription factor aryl hydrocarbon receptor (AhR) as a critical regulator of mucosal immunity, including the intestinal trafficking of CD4+ helper T cells, an immune cell subset implicated in a wide range of homeostatic and pathogenic processes. Specifically, the AhR has been shown to directly regulate the expression of the chemoattractant receptor G Protein-Coupled Receptor 15 (GPR15) on CD4+ T cells. GPR15 is an important gut homing marker whose expression on CD4+ T cells in the peripheral circulation is elevated in patients suffering from ulcerative colitis, raising the possibility that, in this setting, the beneficial effect of a diet rich in fruits and vegetables may be mediated through the modulation of GPR15 expression. To address this, we screened physiologically-relevant polyphenol and glucosinolate metabolites for their ability to affect both AhR activity and GPR15 expression. Our complementary approach and associated findings suggest that polyphenol and glucosinolate metabolites can regulate GPR15 expression on human CD4+ T cells in an AhR-dependent manner.
Collapse
Affiliation(s)
- Jeffry S Tang
- Malaghan Institute of Medical Research, Wellington, New Zealand; High-Value Nutrition National Science Challenge, Auckland, New Zealand.
| | - Ruth Stephens
- Malaghan Institute of Medical Research, Wellington, New Zealand; High-Value Nutrition National Science Challenge, Auckland, New Zealand
| | - Yanyan Li
- Malaghan Institute of Medical Research, Wellington, New Zealand; High-Value Nutrition National Science Challenge, Auckland, New Zealand
| | - Alissa Cait
- Malaghan Institute of Medical Research, Wellington, New Zealand
| | - Katie Gell
- Malaghan Institute of Medical Research, Wellington, New Zealand
| | - Sophie Faulkner
- Malaghan Institute of Medical Research, Wellington, New Zealand; High-Value Nutrition National Science Challenge, Auckland, New Zealand
| | - Alix Grooby
- Malaghan Institute of Medical Research, Wellington, New Zealand; High-Value Nutrition National Science Challenge, Auckland, New Zealand
| | - Patries M Herst
- Malaghan Institute of Medical Research, Wellington, New Zealand; Department of Radiation Therapy, University of Otago, Wellington, New Zealand
| | - David O'Sullivan
- Malaghan Institute of Medical Research, Wellington, New Zealand; High-Value Nutrition National Science Challenge, Auckland, New Zealand
| | - Olivier Gasser
- Malaghan Institute of Medical Research, Wellington, New Zealand; High-Value Nutrition National Science Challenge, Auckland, New Zealand.
| |
Collapse
|
7
|
Espinosa-Ruíz C, Esteban MÁ. Modulation of cell migration and cell tracking of the gilthead seabream (Sparus aurata) SAF-1 cells by probiotics. FISH & SHELLFISH IMMUNOLOGY 2023; 142:109149. [PMID: 37858786 DOI: 10.1016/j.fsi.2023.109149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 02/18/2023] [Accepted: 10/06/2023] [Indexed: 10/21/2023]
Abstract
Cell migration is an essential process in immunity and wound healing. The in vitro scratch assay was optimized for the SAF-1 cell line, obtained from gilthead seabream (Sparus aurata) fin. In addition, selected cells from the cell front were tracked for detailed individual cell movement and morphological analysis. Modulation of migration and cell tracking of the SAF-1 cell line by probiotics was evaluated. Cells were cultured and incubated for 24 h with three species of extremophilic yeasts [Yarrowia lipolytica (D1 and N6) and Debaryomyces hansenii (CBS004)] and the bacterium Shewanella putrefaciens (known as SpPdp11) and then scratch and cell tracking assays were performed. The results indicated that the forward velocity was significantly (p < 0.05) increased in SAF-1 cells incubated with CBS004 or SpPdp11. However, cell velocity, cumulative distance and Euclidean distance were only significantly increased in SAF-1 cells incubated with SpPdp11. Furthermore, to increase our understanding of the genes involved in cell movement, the expression profile of ten structural proteins (α-1β tubulin, vinculin, focal adhesion kinase type, alpha-2 integrin, tetraspanin, integrin-linked kinase 1, tensin 3, tensin 4, paxillin, and light chain 2) was studied by real time-PCR. The expression of these genes was modulated as a function of the probiotic tested and the results indicate that CBS004 and SpPdp11 increase the movement of SAF-1 cells.
Collapse
Affiliation(s)
- Cristóbal Espinosa-Ruíz
- Immunobiology for Aquaculture Group, Department of Cell Biology and Histology, Faculty of Biology, University of Murcia, 30100, Murcia, Spain
| | - Ma Ángeles Esteban
- Immunobiology for Aquaculture Group, Department of Cell Biology and Histology, Faculty of Biology, University of Murcia, 30100, Murcia, Spain.
| |
Collapse
|
8
|
Aderinto N, Abdulbasit MO, Tangmi ADE, Okesanya JO, Mubarak JM. Unveiling the growing significance of metabolism in modulating immune cell function: exploring mechanisms and implications; a review. Ann Med Surg (Lond) 2023; 85:5511-5522. [PMID: 37915697 PMCID: PMC10617839 DOI: 10.1097/ms9.0000000000001308] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2023] [Accepted: 09/06/2023] [Indexed: 11/03/2023] Open
Abstract
Immunometabolism has emerged as a rapidly growing field of research, holding significant promise for personalised medicine and precision immunotherapy. This review explores the intricate relationship between immune function and metabolic processes, emphasising their profound impact on various immune-related disorders. Understanding how metabolic dysregulation contributes to the pathogenesis of these disorders remains a critical research gap. Therefore, this review aims to bridge that gap by examining the key metabolic pathways involved and their specific implications in immune cell function. Key metabolic pathways, including glycolysis, mitochondrial metabolism, fatty acid metabolism, and amino acid metabolism, are discussed in the context of immune cell function. Dysregulation of these pathways can disrupt immune cell activation, differentiation, and overall function, contributing to disease pathogenesis. Understanding these metabolic alterations' molecular mechanisms is essential for developing targeted therapeutic interventions. The review also emphasises the importance of personalised medicine in immune-related disorders. The unique metabolic profiles of individuals can influence treatment outcomes, highlighting the need for tailored approaches. Integrating metabolic profiling into clinical practice can enhance treatment efficacy and improve patient outcomes. Investigating the clinical significance of immunometabolism in diverse disease contexts will facilitate the translation of research findings into clinical practice. Moreover, refining treatment strategies based on individual metabolic profiles will contribute to advancing precision immunotherapy.
Collapse
Affiliation(s)
- Nicholas Aderinto
- Department of Medicine and Surgery, Ladoke Akintola University of Technology, Ogbomoso
| | | | | | | | | |
Collapse
|
9
|
Flati I, Di Vito Nolfi M, Dall’Aglio F, Vecchiotti D, Verzella D, Alesse E, Capece D, Zazzeroni F. Molecular Mechanisms Underpinning Immunometabolic Reprogramming: How the Wind Changes during Cancer Progression. Genes (Basel) 2023; 14:1953. [PMID: 37895302 PMCID: PMC10606647 DOI: 10.3390/genes14101953] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 10/11/2023] [Accepted: 10/12/2023] [Indexed: 10/29/2023] Open
Abstract
Metabolism and the immunological state are intimately intertwined, as defense responses are bioenergetically expensive. Metabolic homeostasis is a key requirement for the proper function of immune cell subsets, and the perturbation of the immune-metabolic balance is a recurrent event in many human diseases, including cancer, due to nutrient fluctuation, hypoxia and additional metabolic changes occurring in the tumor microenvironment (TME). Although much remains to be understood in the field of immunometabolism, here, we report the current knowledge on both physiological and cancer-associated metabolic profiles of immune cells, and the main molecular circuits involved in their regulation, highlighting similarities and differences, and emphasizing immune metabolic liabilities that could be exploited in cancer therapy to overcome immune resistance.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Daria Capece
- Department of Biotechnological and Applied Clinical Sciences (DISCAB), University of L’Aquila, Via Vetoio, Coppito 2, 67100 L’Aquila, Italy; (I.F.); (M.D.V.N.); (F.D.); (D.V.); (D.V.); (E.A.); (F.Z.)
| | | |
Collapse
|
10
|
Rheem HB, Choi H, Yang S, Han S, Rhee SY, Jeong H, Lee KB, Lee Y, Kim IS, Lee H, Choi IS. Fugetaxis of Cell-in-Catalytic-Coat Nanobiohybrids in Glucose Gradients. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2023; 19:e2301431. [PMID: 37282761 DOI: 10.1002/smll.202301431] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Revised: 05/09/2023] [Indexed: 06/08/2023]
Abstract
Manipulation and control of cell chemotaxis remain an underexplored territory despite vast potential in various fields, such as cytotherapeutics, sensors, and even cell robots. Herein is achieved the chemical control over chemotactic movement and direction of Jurkat T cells, as a representative model, by the construction of cell-in-catalytic-coat structures in single-cell nanoencapsulation. Armed with the catalytic power of glucose oxidase (GOx) in the artificial coat, the nanobiohybrid cytostructures, denoted as Jurkat[Lipo_GOx] , exhibit controllable, redirected chemotactic movement in response to d-glucose gradients, in the opposite direction to the positive-chemotaxis direction of naïve, uncoated Jurkat cells in the same gradients. The chemically endowed, reaction-based fugetaxis of Jurkat[Lipo_GOx] operates orthogonally and complementarily to the endogenous, binding/recognition-based chemotaxis that remains intact after the formation of a GOx coat. For instance, the chemotactic velocity of Jurkat[Lipo_GOx] can be adjusted by varying the combination of d-glucose and natural chemokines (CXCL12 and CCL19) in the gradient. This work offers an innovative chemical tool for bioaugmenting living cells at the single-cell level through the use of catalytic cell-in-coat structures.
Collapse
Affiliation(s)
- Hyeong Bin Rheem
- Center for Cell-Encapsulation Research, Department of Chemistry, KAIST, Daejeon, 34141, South Korea
| | - Hyunwoo Choi
- Center for Cell-Encapsulation Research, Department of Chemistry, KAIST, Daejeon, 34141, South Korea
| | - Seoin Yang
- Center for Cell-Encapsulation Research, Department of Chemistry, KAIST, Daejeon, 34141, South Korea
| | - Sol Han
- Center for Cell-Encapsulation Research, Department of Chemistry, KAIST, Daejeon, 34141, South Korea
| | - Su Yeon Rhee
- Center for Cell-Encapsulation Research, Department of Chemistry, KAIST, Daejeon, 34141, South Korea
| | - Hyeongseop Jeong
- Division of Scientific Instrumentation & Management, Korea Basic Science Institute (KBSI), Cheongju, 28119, South Korea
| | - Kyung-Bok Lee
- Division of Scientific Instrumentation & Management, Korea Basic Science Institute (KBSI), Cheongju, 28119, South Korea
| | - Yeji Lee
- Graduate School of Converging Science and Technology, Korea University, Seoul, 02841, South Korea
- Chemical & Biological Integrative Research Center, Biomedical Research Division, Korea Institute of Science and Technology (KIST), Seoul, 02792, South Korea
| | - In-San Kim
- Graduate School of Converging Science and Technology, Korea University, Seoul, 02841, South Korea
- Chemical & Biological Integrative Research Center, Biomedical Research Division, Korea Institute of Science and Technology (KIST), Seoul, 02792, South Korea
| | - Hojae Lee
- Department of Chemistry, Hallym University, Chuncheon, 24252, South Korea
| | - Insung S Choi
- Center for Cell-Encapsulation Research, Department of Chemistry, KAIST, Daejeon, 34141, South Korea
| |
Collapse
|
11
|
Stark M, Levin M, Ulitsky I, Assaraf YG. Folylpolyglutamate synthetase mRNA G-quadruplexes regulate its cell protrusion localization and enhance a cancer cell invasive phenotype upon folate repletion. BMC Biol 2023; 21:13. [PMID: 36721160 PMCID: PMC9889130 DOI: 10.1186/s12915-023-01525-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2022] [Accepted: 01/23/2023] [Indexed: 02/02/2023] Open
Abstract
BACKGROUND Folates are crucial for the biosynthesis of nucleotides and amino acids, essential for cell proliferation and development. Folate deficiency induces DNA damage, developmental defects, and tumorigenicity. The obligatory enzyme folylpolyglutamate synthetase (FPGS) mediates intracellular folate retention via cytosolic and mitochondrial folate polyglutamylation. Our previous paper demonstrated the association of the cytosolic FPGS (cFPGS) with the cytoskeleton and various cell protrusion proteins. Based on these recent findings, the aim of the current study was to investigate the potential role of cFPGS at cell protrusions. RESULTS Here we uncovered a central role for two G-quadruplex (GQ) motifs in the 3'UTR of FPGS mediating the localization of cFPGS mRNA and protein at cell protrusions. Using the MBSV6-loop reporter system and fluorescence microscopy, we demonstrate that following folate deprivation, cFPGS mRNA is retained in the endoplasmic reticulum, whereas upon 15 min of folate repletion, this mRNA is rapidly translocated to cell protrusions in a 3'UTR- and actin-dependent manner. The actin dependency of this folate-induced mRNA translocation is shown by treatment with Latrunculin B and inhibitors of the Ras homolog family member A (RhoA) pathway. Upon folate repletion, the FPGS 3'UTR GQs induce an amoeboid/mesenchymal hybrid cell phenotype during migration and invasion through a collagen gel matrix. Targeted disruption of the 3'UTR GQ motifs by introducing point mutations or masking them by antisense oligonucleotides abrogated cell protrusion targeting of cFPGS mRNA. CONCLUSIONS Collectively, the GQ motifs within the 3'UTR of FPGS regulate its transcript and protein localization at cell protrusions in response to a folate cue, inducing cancer cell invasive phenotype. These novel findings suggest that the 3'UTR GQ motifs of FPGS constitute an attractive druggable target aimed at inhibition of cancer invasion and metastasis.
Collapse
Affiliation(s)
- Michal Stark
- grid.6451.60000000121102151The Fred Wyszkowski Cancer Research Laboratory, Department of Biology, Technion-Israel Institute of Technology, 3200003 Haifa, Israel
| | - May Levin
- grid.6451.60000000121102151The Fred Wyszkowski Cancer Research Laboratory, Department of Biology, Technion-Israel Institute of Technology, 3200003 Haifa, Israel ,grid.507132.2Present address: May Levin, MeMed Diagnostics Ltd, Tirat Carmel, Israel
| | - Igor Ulitsky
- grid.13992.300000 0004 0604 7563Department of Immunology and Regenerative Biology and Department of Molecular Neuroscience, Weizmann Institute of Science, 7610001 Rehovot, Israel
| | - Yehuda G. Assaraf
- grid.6451.60000000121102151The Fred Wyszkowski Cancer Research Laboratory, Department of Biology, Technion-Israel Institute of Technology, 3200003 Haifa, Israel
| |
Collapse
|
12
|
Mao W. Overcoming current challenges to T-cell receptor therapy via metabolic targeting to increase antitumor efficacy, durability, and tolerability. Front Immunol 2022; 13:1056622. [PMID: 36479131 PMCID: PMC9720167 DOI: 10.3389/fimmu.2022.1056622] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Accepted: 10/31/2022] [Indexed: 11/22/2022] Open
Abstract
The antitumor potential of personalized immunotherapy, including adoptive T-cell therapy, has been shown in both preclinical and clinical studies. Combining cell therapy with targeted metabolic interventions can further enhance therapeutic outcomes in terms of magnitude and durability. The ability of a T cell receptor to recognize peptides derived from tumor neoantigens allows for a robust yet specific response against cancer cells while sparing healthy tissue. However, there exist challenges to adoptive T cell therapy such as a suppressive tumor milieu, the fitness and survival of transferred cells, and tumor escape, all of which can be targeted to further enhance the antitumor potential of T cell receptor-engineered T cell (TCR-T) therapy. Here, we explore current strategies involving metabolic reprogramming of both the tumor microenvironment and the cell product, which can lead to increased T cell proliferation, survival, and anti-tumor cytotoxicity. In addition, we highlight potential metabolic pathways and targets which can be leveraged to improve engraftment of transferred cells and obviate the need for lymphodepletion, while minimizing off-target effects. Metabolic signaling is delicately balanced, and we demonstrate the need for thoughtful and precise interventions that are tailored for the unique characteristics of each tumor. Through improved understanding of the interplay between immunometabolism, tumor resistance, and T cell signaling, we can improve current treatment regimens and open the door to potential synergistic combinations.
Collapse
|
13
|
Huang J, Zhou Q. Gene Biomarkers Related to Th17 Cells in Macular Edema of Diabetic Retinopathy: Cutting-Edge Comprehensive Bioinformatics Analysis and In Vivo Validation. Front Immunol 2022; 13:858972. [PMID: 35651615 PMCID: PMC9149582 DOI: 10.3389/fimmu.2022.858972] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Accepted: 04/12/2022] [Indexed: 11/13/2022] Open
Abstract
Background Previous studies have shown that T-helper 17 (Th17) cell-related cytokines are significantly increased in the vitreous of proliferative diabetic retinopathy (PDR), suggesting that Th17 cells play an important role in the inflammatory response of diabetic retinopathy (DR), but its cell infiltration and gene correlation in the retina of DR, especially in diabetic macular edema (DME), have not been studied. Methods The dataset GSE160306 was downloaded from the Gene Expression Omnibus (GEO) database, which contains 9 NPDR samples and 10 DME samples. ImmuCellAI algorithm was used to estimate the abundance of Th17 cells in 24 kinds of infiltrating immune cells. The differentially expressed Th17 related genes (DETh17RGs) between NPDR and DME were documented by difference analysis and correlation analysis. Through aggregate analyses such as gene ontology (GO) and Kyoto Encyclopedia of Gene and Genome (KEGG) pathway enrichment analysis, a protein-protein interaction (PPI) network was constructed to analyze the potential function of DETh17RGs. CytoHubba plug-in algorithm, Lasso regression analysis and support vector machine recursive feature elimination (SVM-RFE) were implemented to comprehensively identify Hub DETh17RGs. The expression archetypes of Hub DETh17RGs were further verified in several other independent datasets related to DR. The Th17RG score was defined as the genetic characterization of six Hub DETh17RGs using the GSVA sample score method, which was used to distinguish early and advanced diabetic nephropathy (DN) as well as normal and diabetic nephropathy. Finally, real-time quantitative PCR (qPCR) was implemented to verify the transcription levels of Hub DETh17RGs in the STZ-induced DR model mice (C57BL/6J). Results 238 DETh17RGs were identified, of which 212 genes were positively correlated while only 26 genes were negatively correlated. Six genes (CD44, CDC42, TIMP1, BMP7, RHOC, FLT1) were identified as Hub DETh17RGs. Because DR and DN have a strong correlation in clinical practice, the verification of multiple independent datasets related to DR and DN proved that Hub DETh17RGs can not only distinguish PDR patients from normal people, but also distinguish DN patients from normal people. It can also identify the initial and advanced stages of the two diseases (NPDR vs DME, Early DN vs Advanced DN). Except for CDC42 and TIMP1, the qPCR transcription levels and trends of other Hub DETh17RGs in STZ-induced DR model mice were consistent with the human transcriptome level in this study. Conclusion This study will improve our understanding of Th17 cell-related molecular mechanisms in the progression of DME. At the same time, it also provides an updated basis for the molecular mechanism of Th17 cell crosstalk in the eye and kidney in diabetes.
Collapse
Affiliation(s)
- Jing Huang
- Department of Ophthalmology, The First Affiliated Hospital of Nanchang University, Jiangxi Center of National Ocular Disease Clinical Research Center, Nanchang, China
| | - Qiong Zhou
- Department of Ophthalmology, The First Affiliated Hospital of Nanchang University, Jiangxi Center of National Ocular Disease Clinical Research Center, Nanchang, China
| |
Collapse
|
14
|
Latorre‐Muro P, Puigserver P. Atossa
: a royal link between OXPHOS metabolism and macrophage migration. EMBO J 2022; 41:e111290. [PMID: 35466422 PMCID: PMC9194787 DOI: 10.15252/embj.2022111290] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2022] [Accepted: 03/29/2022] [Indexed: 11/09/2022] Open
Abstract
The ability of immune cells to penetrate affected tissues is highly dependent on energy provided by mitochondria, yet their involvement in promoting migration remains unclear. Recent work by Emtenani et al (2022) describes a nuclear Atossa-Porthos axis that adjusts transcription and translation of a small subset of OXPHOS genes to increase mitochondrial bioenergetics and allow macrophage tissue invasion in flies.
Collapse
Affiliation(s)
- Pedro Latorre‐Muro
- Department of Cancer Biology Dana‐Farber Cancer Institute Boston MA USA
- Department of Cell Biology Harvard Medical School Boston MA USA
| | - Pere Puigserver
- Department of Cancer Biology Dana‐Farber Cancer Institute Boston MA USA
- Department of Cell Biology Harvard Medical School Boston MA USA
| |
Collapse
|
15
|
Emtenani S, Martin ET, Gyoergy A, Bicher J, Genger JW, Köcher T, Akhmanova M, Guarda M, Roblek M, Bergthaler A, Hurd TR, Rangan P, Siekhaus DE. Macrophage mitochondrial bioenergetics and tissue invasion are boosted by an Atossa-Porthos axis in Drosophila. EMBO J 2022; 41:e109049. [PMID: 35319107 PMCID: PMC9194793 DOI: 10.15252/embj.2021109049] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Revised: 02/14/2022] [Accepted: 02/25/2022] [Indexed: 12/03/2022] Open
Abstract
Cellular metabolism must adapt to changing demands to enable homeostasis. During immune responses or cancer metastasis, cells leading migration into challenging environments require an energy boost, but what controls this capacity is unclear. Here, we study a previously uncharacterized nuclear protein, Atossa (encoded by CG9005), which supports macrophage invasion into the germband of Drosophila by controlling cellular metabolism. First, nuclear Atossa increases mRNA levels of Porthos, a DEAD‐box protein, and of two metabolic enzymes, lysine‐α‐ketoglutarate reductase (LKR/SDH) and NADPH glyoxylate reductase (GR/HPR), thus enhancing mitochondrial bioenergetics. Then Porthos supports ribosome assembly and thereby raises the translational efficiency of a subset of mRNAs, including those affecting mitochondrial functions, the electron transport chain, and metabolism. Mitochondrial respiration measurements, metabolomics, and live imaging indicate that Atossa and Porthos power up OxPhos and energy production to promote the forging of a path into tissues by leading macrophages. Since many crucial physiological responses require increases in mitochondrial energy output, this previously undescribed genetic program may modulate a wide range of cellular behaviors.
Collapse
Affiliation(s)
- Shamsi Emtenani
- Institute of Science and Technology Austria, Klosterneuburg, Austria
| | - Elliot T Martin
- Department of Biological Sciences, RNA Institute, University at Albany, Albany, NY, USA
| | - Attila Gyoergy
- Institute of Science and Technology Austria, Klosterneuburg, Austria
| | - Julia Bicher
- Institute of Science and Technology Austria, Klosterneuburg, Austria
| | - Jakob-Wendelin Genger
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | | | - Maria Akhmanova
- Institute of Science and Technology Austria, Klosterneuburg, Austria
| | - Mariana Guarda
- Institute of Science and Technology Austria, Klosterneuburg, Austria
| | - Marko Roblek
- Institute of Science and Technology Austria, Klosterneuburg, Austria
| | - Andreas Bergthaler
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Thomas R Hurd
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
| | - Prashanth Rangan
- Department of Biological Sciences, RNA Institute, University at Albany, Albany, NY, USA
| | - Daria E Siekhaus
- Institute of Science and Technology Austria, Klosterneuburg, Austria
| |
Collapse
|
16
|
Amiel E, Perona‐Wright G. Sweet talk: Metabolic conversations between host and microbe during infection. Immunology 2021; 162:121-122. [PMID: 33443308 PMCID: PMC7808147 DOI: 10.1111/imm.13301] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
In this issue, we introduce the second part of a series of reviews focusing on how immunometabolism influences host and pathogen interactions during infection. This part of the collection addresses the interface between metabolism and specific types of infection, including immunometabolism in macrophages during helminth infection, the role of metabolism in T-cell exhaustion during chronic viral infections and host immunometabolism in the defence against Mycobacterium tuberculosis infection. These reviews, together with the four articles published in part 1 of the series in November 2020, offer new insights into the complex interactions between mammalian hosts and microbial pathogens through the lens of cellular metabolic regulation.
Collapse
Affiliation(s)
- Eyal Amiel
- Department of Biomedical and Health SciencesUniversity of VermontBurlingtonVermontUSA
| | | |
Collapse
|
17
|
Amiel E, Perona‐Wright G. Metabolic mediators: How immunometabolism directs the immune response to infection. Immunology 2020; 161:163-164. [PMID: 33085098 PMCID: PMC7576874 DOI: 10.1111/imm.13275] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Here we announce the first part of an exciting new series of reviews exploring the impact of immunometabolism in the interaction between host and pathogen, and in the outcome of infection. This collection discusses the links between metabolism and epigenetic control of cell function, post-translation modifications of host proteins that determine protein fate and host cell function, the metabolic determinants of cell migration and immune cell activity, and the tussle for iron as a metabolic mediator of host-pathogen domination. Together these reviews provide engaging new insight into the metabolic signals that guide the dynamic conversation between microbial pathogens and the mammalian hosts they aim to occupy.
Collapse
Affiliation(s)
- Eyal Amiel
- Department of Biomedical and Health SciencesUniversity of VermontBurlingtonVTUSA
| | | |
Collapse
|