LIVIVO - The Search Portal for Life Sciences

zur deutschen Oberfläche wechseln
Advanced search

Search results

Result 1 - 10 of total 43

Search options

  1. Article ; Online: How the Unfolded Protein Response Is a Boon for Tumors and a Bane for the Immune System.

    Raines, Lydia N / Huang, Stanley Ching-Cheng

    ImmunoHorizons

    2023  Volume 7, Issue 4, Page(s) 256–264

    Abstract: The correct folding of proteins is essential for appropriate cell function and is tightly regulated within the endoplasmic reticulum (ER). Environmental challenges and cellular conditions disrupt ER homeostasis and induce ER stress, which adversely ... ...

    Abstract The correct folding of proteins is essential for appropriate cell function and is tightly regulated within the endoplasmic reticulum (ER). Environmental challenges and cellular conditions disrupt ER homeostasis and induce ER stress, which adversely affect protein folding and activate the unfolded protein response (UPR). It is now becoming recognized that cancer cells can overcome survival challenges posed within the tumor microenvironment by activating the UPR. Furthermore, the UPR has also been found to impose detrimental effects on immune cells by inducing immunoinhibitory activity in both tumor-infiltrating innate and adaptive immune cells. This suggests that these signaling axes may be important therapeutic targets, resulting in multifaceted approaches to eradicating tumor cells. In this mini-review, we discuss the role of the UPR in driving tumor progression and modulating the immune system's ability to target cancer cells. Additionally, we highlight some of the key unanswered questions that may steer future UPR research.
    MeSH term(s) Humans ; Unfolded Protein Response ; Endoplasmic Reticulum Stress ; Neoplasms/pathology ; Endoplasmic Reticulum/metabolism ; Endoplasmic Reticulum/pathology ; Immune System ; Tumor Microenvironment
    Language English
    Publishing date 2023-04-17
    Publishing country United States
    Document type Review ; Journal Article ; Research Support, N.I.H., Extramural ; Research Support, Non-U.S. Gov't
    ISSN 2573-7732
    ISSN (online) 2573-7732
    DOI 10.4049/immunohorizons.2200064
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  2. Article ; Online: Control of immune cell function by the unfolded protein response.

    Di Conza, Giusy / Ho, Ping-Chih / Cubillos-Ruiz, Juan R / Huang, Stanley Ching-Cheng

    Nature reviews. Immunology

    2023  Volume 23, Issue 9, Page(s) 546–562

    Abstract: Initiating and maintaining optimal immune responses requires high levels of protein synthesis, folding, modification and trafficking in leukocytes, which are processes orchestrated by the endoplasmic reticulum. Importantly, diverse extracellular and ... ...

    Abstract Initiating and maintaining optimal immune responses requires high levels of protein synthesis, folding, modification and trafficking in leukocytes, which are processes orchestrated by the endoplasmic reticulum. Importantly, diverse extracellular and intracellular conditions can compromise the protein-handling capacity of this organelle, inducing a state of 'endoplasmic reticulum stress' that activates the unfolded protein response (UPR). Emerging evidence shows that physiological or pathological activation of the UPR can have effects on immune cell survival, metabolism, function and fate. In this Review, we discuss the canonical role of the adaptive UPR in immune cells and how dysregulation of this pathway in leukocytes contributes to diverse pathologies such as cancer, autoimmunity and metabolic disorders. Furthermore, we provide an overview as to how pharmacological approaches that modulate the UPR could be harnessed to control or activate immune cell function in disease.
    MeSH term(s) Humans ; Unfolded Protein Response ; Endoplasmic Reticulum Stress ; Neoplasms/pathology ; Immunity ; Endoplasmic Reticulum/metabolism
    Language English
    Publishing date 2023-02-08
    Publishing country England
    Document type Journal Article ; Review ; Research Support, N.I.H., Extramural ; Research Support, Non-U.S. Gov't ; Research Support, U.S. Gov't, Non-P.H.S.
    ZDB-ID 2062776-2
    ISSN 1474-1741 ; 1474-1733
    ISSN (online) 1474-1741
    ISSN 1474-1733
    DOI 10.1038/s41577-023-00838-0
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  3. Article ; Online: Breathe In, Breathe Out: Metabolic Regulation of Lung Macrophages in Host Defense Against Bacterial Infection.

    Andrews, J Tucker / Voth, Daniel E / Huang, Stanley Ching-Cheng / Huang, Lu

    Frontiers in cellular and infection microbiology

    2022  Volume 12, Page(s) 934460

    Abstract: Lung macrophages are substantially distinct from other tissue-resident macrophages. They act as frontier sentinels of the alveolar-blood interface and are constantly exposed to various pathogens. Additionally, they precisely regulate immune responses ... ...

    Abstract Lung macrophages are substantially distinct from other tissue-resident macrophages. They act as frontier sentinels of the alveolar-blood interface and are constantly exposed to various pathogens. Additionally, they precisely regulate immune responses under homeostatic and pathological conditions to curtail tissue damage while containing respiratory infections. As a highly heterogeneous population, the phenotypes and functions of lung macrophages with differing developmental ontogenies are linked to both intrinsic and extrinsic metabolic processes. Importantly, targeting these metabolic pathways greatly impacts macrophage functions, which in turn leads to different disease outcomes in the lung. In this review, we will discuss underlying metabolic regulation of lung macrophage subsets and how metabolic circuits, together with epigenetic modifications, dictate lung macrophage function during bacterial infection.
    MeSH term(s) Bacterial Infections/pathology ; Humans ; Immunity ; Lung/microbiology ; Macrophages ; Macrophages, Alveolar
    Language English
    Publishing date 2022-07-08
    Publishing country Switzerland
    Document type Journal Article ; Review ; Research Support, N.I.H., Extramural ; Research Support, Non-U.S. Gov't
    ZDB-ID 2619676-1
    ISSN 2235-2988 ; 2235-2988
    ISSN (online) 2235-2988
    ISSN 2235-2988
    DOI 10.3389/fcimb.2022.934460
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  4. Article ; Online: Is glucose the scapegoat for tumor evasion?

    Raines, Lydia N / Huang, Stanley Ching-Cheng

    Cancer cell

    2021  Volume 39, Issue 7, Page(s) 907–909

    Abstract: Tumor cells undergo rapid aerobic glycolysis to fuel growth and proliferation. A recent finding in Nature reveals that tumor-infiltrating myeloid cells demonstrate greater capacity for glucose uptake than tumor cells and consume significant amounts ... ...

    Abstract Tumor cells undergo rapid aerobic glycolysis to fuel growth and proliferation. A recent finding in Nature reveals that tumor-infiltrating myeloid cells demonstrate greater capacity for glucose uptake than tumor cells and consume significant amounts within the tumor milieu. Unexpectedly, tumor cells account for the highest glutamine utilization in the tumor microenvironment.
    MeSH term(s) Glucose ; Glutamine/metabolism ; Glycolysis ; Humans ; Neoplasms ; Tumor Microenvironment
    Chemical Substances Glutamine (0RH81L854J) ; Glucose (IY9XDZ35W2)
    Language English
    Publishing date 2021-07-13
    Publishing country United States
    Document type Journal Article ; Comment
    ZDB-ID 2078448-X
    ISSN 1878-3686 ; 1535-6108
    ISSN (online) 1878-3686
    ISSN 1535-6108
    DOI 10.1016/j.ccell.2021.06.006
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  5. Article: Molecular Chaperones: Molecular Assembly Line Brings Metabolism and Immunity in Shape.

    Zhao, Haoxin / Raines, Lydia N / Huang, Stanley Ching-Cheng

    Metabolites

    2020  Volume 10, Issue 10

    Abstract: Molecular chaperones are a set of conserved proteins that have evolved to assist the folding of many newly synthesized proteins by preventing their misfolding under conditions such as elevated temperatures, hypoxia, acidosis and nutrient deprivation. ... ...

    Abstract Molecular chaperones are a set of conserved proteins that have evolved to assist the folding of many newly synthesized proteins by preventing their misfolding under conditions such as elevated temperatures, hypoxia, acidosis and nutrient deprivation. Molecular chaperones belong to the heat shock protein (HSP) family. They have been identified as important participants in immune functions including antigen presentation, immunostimulation and immunomodulation, and play crucial roles in metabolic rewiring and epigenetic circuits. Growing evidence has accumulated to indicate that metabolic pathways and their metabolites influence the function of immune cells and can alter transcriptional activity through epigenetic modification of (de)methylation and (de)acetylation. However, whether molecular chaperones can regulate metabolic programs to influence immune activity is still largely unclear. In this review, we discuss the available data on the biological function of molecular chaperones to immune responses during inflammation, with a specific focus on the interplay between molecular chaperones and metabolic pathways that drive immune cell fate and function.
    Language English
    Publishing date 2020-10-03
    Publishing country Switzerland
    Document type Journal Article ; Review
    ZDB-ID 2662251-8
    ISSN 2218-1989
    ISSN 2218-1989
    DOI 10.3390/metabo10100394
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  6. Article ; Online: Carbohydrate and Amino Acid Metabolism as Hallmarks for Innate Immune Cell Activation and Function.

    Zhao, Haoxin / Raines, Lydia N / Huang, Stanley Ching-Cheng

    Cells

    2020  Volume 9, Issue 3

    Abstract: Immune activation is now understood to be fundamentally linked to intrinsic and/or extrinsic metabolic processes which are essential for immune cells to survive, proliferate, and perform their effector functions. Moreover, disruption or dysregulation of ... ...

    Abstract Immune activation is now understood to be fundamentally linked to intrinsic and/or extrinsic metabolic processes which are essential for immune cells to survive, proliferate, and perform their effector functions. Moreover, disruption or dysregulation of these pathways can result in detrimental outcomes and underly a number of pathologies in both communicable and non-communicable diseases. In this review, we discuss how the metabolism of carbohydrates and amino acids in particular can modulate innate immunity and how perturbations in these pathways can result in failure of these immune cells to properly function or induce unfavorable phenotypes.
    MeSH term(s) Amino Acids/metabolism ; Animals ; Carbohydrate Metabolism ; Humans ; Immunity, Innate ; Models, Biological ; Myeloid-Derived Suppressor Cells/metabolism
    Chemical Substances Amino Acids
    Language English
    Publishing date 2020-02-27
    Publishing country Switzerland
    Document type Journal Article ; Research Support, N.I.H., Extramural ; Research Support, Non-U.S. Gov't ; Review
    ZDB-ID 2661518-6
    ISSN 2073-4409 ; 2073-4409
    ISSN (online) 2073-4409
    ISSN 2073-4409
    DOI 10.3390/cells9030562
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  7. Article ; Online: Circles of Life: linking metabolic and epigenetic cycles to immunity.

    Lio, Chan-Wang Jerry / Huang, Stanley Ching-Cheng

    Immunology

    2020  Volume 161, Issue 3, Page(s) 165–174

    Abstract: Metabolites are the essential substrates for epigenetic modification enzymes to write or erase the epigenetic blueprint in cells. Hence, the availability of nutrients and activity of metabolic pathways strongly influence the enzymatic function. Recent ... ...

    Abstract Metabolites are the essential substrates for epigenetic modification enzymes to write or erase the epigenetic blueprint in cells. Hence, the availability of nutrients and activity of metabolic pathways strongly influence the enzymatic function. Recent studies have shed light on the choreography between metabolome and epigenome in the control of immune cell differentiation and function, with a major focus on histone modifications. Yet, despite its importance in gene regulation, DNA methylation and its relationship with metabolism is relatively unclear. In this review, we will describe how the metabolic flux can influence epigenetic networks in innate and adaptive immune cells, with a focus on the DNA methylation cycle and the metabolites S-adenosylmethionine and α-ketoglutarate. Future directions will be discussed for this rapidly emerging field.
    MeSH term(s) Adaptive Immunity ; Animals ; Citric Acid Cycle/immunology ; DNA Methylation ; Epigenesis, Genetic ; Epigenome/immunology ; Humans ; Immunity, Innate ; Ketoglutaric Acids/metabolism ; Metabolome/immunology ; S-Adenosylmethionine/metabolism
    Chemical Substances Ketoglutaric Acids ; S-Adenosylmethionine (7LP2MPO46S)
    Keywords covid19
    Language English
    Publishing date 2020-06-03
    Publishing country England
    Document type Journal Article ; Research Support, N.I.H., Extramural ; Research Support, Non-U.S. Gov't ; Review
    ZDB-ID 80124-0
    ISSN 1365-2567 ; 0019-2805 ; 0953-4954
    ISSN (online) 1365-2567
    ISSN 0019-2805 ; 0953-4954
    DOI 10.1111/imm.13207
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  8. Article ; Online: Fatty acids secreted from head and neck cancer induce M2-like Macrophages.

    Albakri, Marwah M / Huang, Stanley Ching-Cheng / Tashkandi, Hammad N / Sieg, Scott F

    Journal of leukocyte biology

    2022  Volume 112, Issue 4, Page(s) 617–628

    Abstract: Tumor-infiltrating monocytes can mature into Macrophages that support tumor survival or that display antitumor properties. To explore mechanisms steering Macrophage maturation, we assessed the effects of supernatants from squamous cell carcinoma cell ... ...

    Abstract Tumor-infiltrating monocytes can mature into Macrophages that support tumor survival or that display antitumor properties. To explore mechanisms steering Macrophage maturation, we assessed the effects of supernatants from squamous cell carcinoma cell lines (FaDu and SCC) on monocyte-derived Macrophage maturation. Purified monocytes were incubated in medium or medium supplemented with supernatants from FaDu and SCC9 or the leukemia monocytic cell line, THP-1. Macrophages were examined for markers of maturation (CD14, CD68), activation (HLA-DR, CD86, IL15R), scavenger receptor (CD36), toll-like receptor (TLR4), M2 marker (CD206), immune checkpoint (PD-L1), and intracellular chemokine expression (IP-10). Compared to other conditions, cells incubated with FaDu or SCC9 supernatants displayed enhanced survival, down-regulation of cell surface HLA-DR, CD86, IL-15R, CD36, and intracellular IP-10 expression, and increased cell surface PD-L1, CD14, and CD206 expression. Despite expressing TLR4 and CD14, Macrophages matured in tumor supernatants failed to respond to stimulation with the canonical TLR4 agonist, LPS. These changes were accompanied by a decrease in intracellular phospho-p38 expression in tumor supernatant conditioned Macrophages. Depletion of fatty acids from tumor supernatants or treatment of cell cultures with an inhibitor of fatty acid oxidation, Etomoxir, reversed a number of these phenotypic changes induced by tumor supernatants. Additionally, Macrophages incubated with either palmitic acid or oleic acid developed similar phenotypes as cells incubated in tumor supernatants. Together, these data suggest that fatty acids derived from tumor cells can mediate the maturation of Macrophages into a cell type with limited pro-inflammatory characteristics.
    MeSH term(s) B7-H1 Antigen/metabolism ; Chemokine CXCL10/metabolism ; Fatty Acids/metabolism ; HLA-DR Antigens/metabolism ; Head and Neck Neoplasms ; Humans ; Lipopolysaccharides/pharmacology ; Macrophages/metabolism ; Oleic Acids/metabolism ; Oleic Acids/pharmacology ; Palmitic Acids/metabolism ; Palmitic Acids/pharmacology ; Toll-Like Receptor 4/metabolism
    Chemical Substances B7-H1 Antigen ; Chemokine CXCL10 ; Fatty Acids ; HLA-DR Antigens ; Lipopolysaccharides ; Oleic Acids ; Palmitic Acids ; Toll-Like Receptor 4
    Language English
    Publishing date 2022-02-25
    Publishing country England
    Document type Journal Article ; Research Support, Non-U.S. Gov't
    ZDB-ID 605722-6
    ISSN 1938-3673 ; 0741-5400
    ISSN (online) 1938-3673
    ISSN 0741-5400
    DOI 10.1002/JLB.1A0521-251R
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  9. Article ; Online: Navigating metabolic pathways to enhance antitumour immunity and immunotherapy.

    Li, Xiaoyun / Wenes, Mathias / Romero, Pedro / Huang, Stanley Ching-Cheng / Fendt, Sarah-Maria / Ho, Ping-Chih

    Nature reviews. Clinical oncology

    2019  Volume 16, Issue 7, Page(s) 425–441

    Abstract: The development of immunotherapies over the past decade has resulted in a paradigm shift in the treatment of cancer. However, the majority of patients do not benefit from immunotherapy, presumably owing to insufficient reprogramming of the ... ...

    Abstract The development of immunotherapies over the past decade has resulted in a paradigm shift in the treatment of cancer. However, the majority of patients do not benefit from immunotherapy, presumably owing to insufficient reprogramming of the immunosuppressive tumour microenvironment (TME) and thus limited reinvigoration of antitumour immunity. Various metabolic machineries and nutrient-sensing mechanisms orchestrate the behaviour of immune cells in response to nutrient availability in the TME. Notably, tumour-infiltrating immune cells typically experience metabolic stress as a result of the dysregulated metabolic activity of tumour cells, leading to impaired antitumour immune responses. Moreover, the immune checkpoints that are often exploited by tumour cells to evade immunosurveillance have emerging roles in modulating the metabolic and functional activity of T cells. Thus, repurposing of drugs targeting cancer metabolism might synergistically enhance immunotherapy via metabolic reprogramming of the TME. In addition, interventions targeting the metabolic circuits that impede antitumour immunity have been developed, with several clinical trials underway. Herein, we discuss how these metabolic circuits regulate antitumour immunity and the possible approaches to targeting these pathways in the context of anticancer immunotherapy. We also describe hypothetical combination treatments that could be used to better unleash the potential of adoptive cell therapies by enhancing T cell metabolism.
    MeSH term(s) Antineoplastic Agents/pharmacology ; Antineoplastic Agents/therapeutic use ; Combined Modality Therapy ; Drug Repositioning ; Drug Synergism ; Humans ; Immunotherapy/methods ; Metabolic Networks and Pathways/drug effects ; Neoplasms/drug therapy ; Neoplasms/immunology ; Tumor Microenvironment/drug effects
    Chemical Substances Antineoplastic Agents
    Language English
    Publishing date 2019-03-26
    Publishing country England
    Document type Journal Article ; Research Support, Non-U.S. Gov't ; Review
    ZDB-ID 2491410-1
    ISSN 1759-4782 ; 1759-4774
    ISSN (online) 1759-4782
    ISSN 1759-4774
    DOI 10.1038/s41571-019-0203-7
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  10. Article ; Online: ILC3s integrate glycolysis and mitochondrial production of reactive oxygen species to fulfill activation demands.

    Di Luccia, Blanda / Gilfillan, Susan / Cella, Marina / Colonna, Marco / Huang, Stanley Ching-Cheng

    The Journal of experimental medicine

    2019  Volume 216, Issue 10, Page(s) 2231–2241

    Abstract: Group 3 innate lymphoid cells (ILC3s) are the innate counterparts of Th17 that require the transcription factor RORγt for development and contribute to the defense against pathogens through IL-22 and IL-17 secretion. Proliferation and effector functions ... ...

    Abstract Group 3 innate lymphoid cells (ILC3s) are the innate counterparts of Th17 that require the transcription factor RORγt for development and contribute to the defense against pathogens through IL-22 and IL-17 secretion. Proliferation and effector functions of Th17 require a specific mTOR-dependent metabolic program that utilizes high-rate glycolysis, while mitochondrial lipid oxidation and production of reactive oxygen species (mROS) support alternative T reg cell differentiation. Whether ILC3s employ a specific metabolic program is not known. Here, we find that ILC3s rely on mTOR complex 1 (mTORC1) for proliferation and production of IL-22 and IL-17A after in vitro activation and
    MeSH term(s) Animals ; Citrobacter rodentium/immunology ; Enterobacteriaceae Infections/genetics ; Enterobacteriaceae Infections/immunology ; Enterobacteriaceae Infections/pathology ; Glycolysis/genetics ; Glycolysis/immunology ; Hypoxia-Inducible Factor 1, alpha Subunit/genetics ; Hypoxia-Inducible Factor 1, alpha Subunit/immunology ; Interleukin-17/genetics ; Interleukin-17/immunology ; Interleukins/genetics ; Interleukins/immunology ; Lymphocyte Activation ; Mechanistic Target of Rapamycin Complex 1/genetics ; Mechanistic Target of Rapamycin Complex 1/immunology ; Mice ; Mice, Knockout ; Mitochondria/genetics ; Mitochondria/immunology ; Mitochondria/pathology ; Nuclear Receptor Subfamily 1, Group F, Member 3/genetics ; Nuclear Receptor Subfamily 1, Group F, Member 3/immunology ; Reactive Oxygen Species/immunology ; Th17 Cells/immunology ; Th17 Cells/pathology ; Interleukin-22
    Chemical Substances Hif1a protein, mouse ; Hypoxia-Inducible Factor 1, alpha Subunit ; Il17a protein, mouse ; Interleukin-17 ; Interleukins ; Nuclear Receptor Subfamily 1, Group F, Member 3 ; Reactive Oxygen Species ; Rorc protein, mouse ; Mechanistic Target of Rapamycin Complex 1 (EC 2.7.11.1)
    Language English
    Publishing date 2019-07-11
    Publishing country United States
    Document type Journal Article ; Research Support, N.I.H., Extramural ; Research Support, Non-U.S. Gov't
    ZDB-ID 218343-2
    ISSN 1540-9538 ; 0022-1007
    ISSN (online) 1540-9538
    ISSN 0022-1007
    DOI 10.1084/jem.20180549
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

To top