Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
1.
EMBO J ; 42(16): e113258, 2023 08 15.
Article in English | MEDLINE | ID: mdl-37409632

ABSTRACT

Mitochondrial biogenesis is the process of generating new mitochondria to maintain cellular homeostasis. Here, we report that viruses exploit mitochondrial biogenesis to antagonize innate antiviral immunity. We found that nuclear respiratory factor-1 (NRF1), a vital transcriptional factor involved in nuclear-mitochondrial interactions, is essential for RNA (VSV) or DNA (HSV-1) virus-induced mitochondrial biogenesis. NRF1 deficiency resulted in enhanced innate immunity, a diminished viral load, and morbidity in mice. Mechanistically, the inhibition of NRF1-mediated mitochondrial biogenesis aggravated virus-induced mitochondrial damage, promoted the release of mitochondrial DNA (mtDNA), increased the production of mitochondrial reactive oxygen species (mtROS), and activated the innate immune response. Notably, virus-activated kinase TBK1 phosphorylated NRF1 at Ser318 and thereby triggered the inactivation of the NRF1-TFAM axis during HSV-1 infection. A knock-in (KI) strategy that mimicked TBK1-NRF1 signaling revealed that interrupting the TBK1-NRF1 connection ablated mtDNA release and thereby attenuated the HSV-1-induced innate antiviral response. Our study reveals a previously unidentified antiviral mechanism that utilizes a NRF1-mediated negative feedback loop to modulate mitochondrial biogenesis and antagonize innate immune response.


Subject(s)
Antiviral Agents , Organelle Biogenesis , Animals , Mice , DNA, Mitochondrial/genetics , Immunity, Innate , Nuclear Respiratory Factor 1/genetics
2.
EMBO Rep ; 24(4): e54731, 2023 04 05.
Article in English | MEDLINE | ID: mdl-36847607

ABSTRACT

Ectopic lipid deposition and mitochondrial dysfunction are common etiologies of obesity and metabolic disorders. Excessive dietary uptake of saturated fatty acids (SFAs) causes mitochondrial dysfunction and metabolic disorders, while unsaturated fatty acids (UFAs) counterbalance these detrimental effects. It remains elusive how SFAs and UFAs differentially signal toward mitochondria for mitochondrial performance. We report here that saturated dietary fatty acids such as palmitic acid (PA), but not unsaturated oleic acid (OA), increase lysophosphatidylinositol (LPI) production to impact on the stability of the mitophagy receptor FUNDC1 and on mitochondrial quality. Mechanistically, PA shifts FUNDC1 from dimer to monomer via enhanced production of LPI. Monomeric FUNDC1 shows increased acetylation at K104 due to dissociation of HDAC3 and increased interaction with Tip60. Acetylated FUNDC1 can be further ubiquitinated by MARCH5 for proteasomal degradation. Conversely, OA antagonizes PA-induced accumulation of LPI, and FUNDC1 monomerization and degradation. A fructose-, palmitate-, and cholesterol-enriched (FPC) diet also affects FUNDC1 dimerization and promotes its degradation in a non-alcoholic steatohepatitis (NASH) mouse model. We thus uncover a signaling pathway that orchestrates lipid metabolism with mitochondrial quality.


Subject(s)
Fatty Acids , Mitophagy , Mice , Animals , Fatty Acids/metabolism , Dimerization , Mitochondria/metabolism , Mitochondrial Proteins/metabolism , Membrane Proteins/metabolism
3.
Article in English | MEDLINE | ID: mdl-38925992

ABSTRACT

One-carbon metabolism (1CM), comprising folate metabolism and methionine metabolism, serves as an important mechanism for cellular energy provision and the production of vital signaling molecules, including single-carbon moieties. Its regulation is instrumental in sustaining the proliferation of cancer cells and facilitating metastasis; in addition, recent research has shed light on its impact on the efficacy of T cell-mediated immunotherapy. In this review, we consolidate current insights into how 1CM affects T cell activation, differentiation, and functionality. Furthermore, we delve into the strategies for modulating 1CM in both T cells and tumor cells to enhance the efficacy of adoptively transferred T cells, overcome metabolic challenges in the tumor microenvironment (TME), and maximize the benefits of T cell-mediated immunotherapy.

4.
Cell Rep ; 43(2): 113796, 2024 Feb 27.
Article in English | MEDLINE | ID: mdl-38367240

ABSTRACT

The acidic metabolic byproducts within the tumor microenvironment (TME) hinder T cell effector functions. However, their effects on T cell infiltration remain largely unexplored. Leveraging the comprehensive The Cancer Genome Atlas dataset, we pinpoint 16 genes that correlate with extracellular acidification and establish a metric known as the "tumor acidity (TuAci) score" for individual patients. We consistently observe a negative association between the TuAci score and T lymphocyte score (T score) across various human cancer types. Mechanistically, extracellular acidification significantly impedes T cell motility by suppressing podosome formation. This phenomenon can be attributed to the reduced expression of methyltransferase-like 3 (METTL3) and the modification of RNA N6-methyladenosine (m6A), resulting in a subsequent decrease in the expression of integrin ß1 (ITGB1). Importantly, enforced ITGB1 expression leads to enhanced T cell infiltration and improved antitumor activity. Our study suggests that modulating METTL3 activity or boosting ITGB1 expression could augment T cell infiltration within the acidic TME, thereby improving the efficacy of cell therapy.


Subject(s)
Integrin beta1 , Neoplasms , Humans , Cell- and Tissue-Based Therapy , Hydrogen-Ion Concentration , Integrin beta1/genetics , Methyltransferases/genetics , T-Lymphocytes , Tumor Microenvironment
5.
Nat Metab ; 5(2): 314-330, 2023 02.
Article in English | MEDLINE | ID: mdl-36717749

ABSTRACT

The accumulation of acidic metabolic waste products within the tumor microenvironment inhibits effector functions of tumor-infiltrating lymphocytes (TILs). However, it remains unclear how an acidic environment affects T cell metabolism and differentiation. Here we show that prolonged exposure to acid reprograms T cell intracellular metabolism and mitochondrial fitness and preserves T cell stemness. Mechanistically, elevated extracellular acidosis impairs methionine uptake and metabolism via downregulation of SLC7A5, therefore altering H3K27me3 deposition at the promoters of key T cell stemness genes. These changes promote the maintenance of a 'stem-like memory' state and improve long-term in vivo persistence and anti-tumor efficacy in mice. Our findings not only reveal an unexpected capacity of extracellular acidosis to maintain the stem-like properties of T cells, but also advance our understanding of how methionine metabolism affects T cell stemness.


Subject(s)
Acidosis , Neoplasms , Animals , Mice , Neoplasms/metabolism , Cell Differentiation , Tumor Microenvironment , Acidosis/metabolism , Carbon
6.
Trends Cancer ; 8(3): 159-160, 2022 03.
Article in English | MEDLINE | ID: mdl-35115254

ABSTRACT

Different subsets of tumor-infiltrating dendritic cells (DCs) influence immune response and tolerance in cancer settings. Duong et al. discovered that conventional DC2 subset (cDC2s) can differentiate into stimulatory interferon-stimulated gene (ISG)+ DCs by type I interferon (IFN-I) produced in regressor tumors and acquire and present tumor-derived peptide-MHC (pMHC) class I complexes to increase the protective antitumor CD8+ T cell response.


Subject(s)
CD8-Positive T-Lymphocytes , Dendritic Cells , Bandages , Humans , Immune Tolerance
7.
Cell Rep ; 40(7): 111195, 2022 08 16.
Article in English | MEDLINE | ID: mdl-35977480

ABSTRACT

ATG9A is a highly conserved membrane protein required for autophagy initiation. It is trafficked from the trans-Golgi network (TGN) to the phagophore to act as a membrane source for autophagosome expansion. Here, we show that ATG9A is not just a passenger protein in the TGN but rather works in concert with GRASP55, a stacking factor for Golgi structure, to organize Golgi dynamics and integrity. Upon heat stress, the E3 ubiquitin ligase MARCH9 is promoted to ubiquitinate ATG9A in the form of K63 conjugation, and the nondegradable ubiquitinated ATG9A disperses from the Golgi apparatus to the cytoplasm more intensely, accompanied by inhibiting GRASP55 oligomerization, further resulting in Golgi fragmentation. Knockout of ATG9A or MARCH9 largely prevents Golgi fragmentation and protects Golgi functions under heat and other Golgi stresses. Our results reveal a noncanonical function of ATG9A for Golgi dynamics and suggest the pathway for sensing Golgi stress via the MARCH9/ATG9A axis.


Subject(s)
Autophagosomes , Golgi Apparatus , Autophagosomes/metabolism , Autophagy , Autophagy-Related Proteins/metabolism , Golgi Apparatus/metabolism , Protein Transport , Ubiquitin/metabolism , trans-Golgi Network/metabolism
8.
Elife ; 112022 08 01.
Article in English | MEDLINE | ID: mdl-35913115

ABSTRACT

DBC1 has been characterized as a key regulator of physiological and pathophysiological activities, such as DNA damage, senescence, and tumorigenesis. However, the mechanism by which the functional stability of DBC1 is regulated has yet to be elucidated. Here, we report that the ubiquitination-mediated degradation of DBC1 is regulated by the E3 ubiquitin ligase SIAH2 and deubiquitinase OTUD5 under hypoxic stress. Mechanistically, hypoxia promoted DBC1 to interact with SIAH2 but not OTUD5, resulting in the ubiquitination and subsequent degradation of DBC1 through the ubiquitin-proteasome pathway. SIAH2 knockout inhibited tumor cell proliferation and migration, which could be rescued by double knockout of SIAH2/CCAR2. Human tissue microarray analysis further revealed that the SIAH2/DBC1 axis was responsible for tumor progression under hypoxic stress. These findings define a key role of the hypoxia-mediated SIAH2-DBC1 pathway in the progression of human breast cancer and provide novel insights into the metastatic mechanism of breast cancer.


Subject(s)
Breast Neoplasms , Adaptor Proteins, Signal Transducing/metabolism , Breast/metabolism , Breast Neoplasms/pathology , Female , Humans , Hypoxia/metabolism , Ubiquitin-Protein Ligases/metabolism , Ubiquitination
9.
Cancer Lett ; 506: 55-66, 2021 05 28.
Article in English | MEDLINE | ID: mdl-33662493

ABSTRACT

The persistent antigen stimulation during chronic infections and cancer results in CD8+ T cell exhaustion. The exhausted T (Tex) cells within the tumor microenvironment (TME) are characterized by increased expression of multiple co-inhibitory receptors simultaneously, progressive loss of effector function, poor proliferation and self-renewal capacity, and dysregulated metabolic activity. Emerging insights into molecular mechanisms underlying T cell exhaustion have proposed potential approaches to improve the efficacy of cancer immunotherapy via restoring the effector function of Tex cells. In this review, we summarize the fundamental characteristics (e.g., inhibitory receptors and transcriptional factors) regarding Tex cell differentiation and discuss in particular how those exhaustion features are acquired and shaped by key factors within the TME. Additionally, we discuss the progress and limitations of current cancer immunotherapeutic strategies targeting Tex cells in clinical setting.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Immunotherapy/methods , Neoplasms/therapy , Tumor Microenvironment , Cell Differentiation , Humans , Transcription, Genetic
10.
Cell Metab ; 32(6): 905-907, 2020 12 01.
Article in English | MEDLINE | ID: mdl-33264601

ABSTRACT

Two recent studies published in Nature Immunology map out the link between dysregulated mitochondrial metabolism and terminal exhaustion of tumor-infiltrating T lymphocytes. Yu et al. (2020) and Vardhana et al. (2020) show that defective mitophagy or impaired oxidative phosphorylation triggers mitochondrial reactive oxygen species production, which in turn promotes a T cell exhaustion program, limiting T cell proliferation and self-renewal.


Subject(s)
Oxidative Phosphorylation , T-Lymphocytes , Mitochondria/metabolism , Mitophagy , Reactive Oxygen Species/metabolism
11.
Cell Death Differ ; 27(3): 1036-1051, 2020 03.
Article in English | MEDLINE | ID: mdl-31367011

ABSTRACT

Mitochondria are highly dynamic organelles and respond to stress by changing their fission-fusion cycle, undergoing mitophagy, or releasing apoptotic proteins to initiate cell death. The molecular mechanisms that sense different stresses and coordinate distinct effectors still await full characterization. Here, we show that PGAM5, which exists in an equilibrium between dimeric and multimeric states, dephosphorylates BCL-xL to inhibit apoptosis or FUNDC1 to activate mitofission and mitophagy in response to distinct stresses. In vinblastine-treated cells, PGAM5 dephosphorylates BCL-xL at Ser62 to restore BCL-xL sequestration of BAX and BAK and thereby resistance to apoptosis. Selenite-induced oxidative stress increases the multimerization of PGAM5, resulting in its dissociation from BCL-xL, which causes increased BCL-xL phosphorylation and apoptosis. Once freed, the more multimeric and active PGAM5 dephosphorylates FUNDC1 to initiate mitofission and mitophagy. The reciprocal interaction of PGAM5 with FUNDC1 and BCL-xL, controlled by PGAM5 multimerization, serves as a molecular switch between mitofission/mitophagy and apoptosis.


Subject(s)
Cell Lineage , Membrane Proteins/metabolism , Mitochondria/metabolism , Mitochondrial Proteins/metabolism , Phosphoprotein Phosphatases/metabolism , bcl-X Protein/metabolism , Apoptosis/drug effects , Cell Lineage/drug effects , HeLa Cells , Humans , Mitochondria/drug effects , Mitochondrial Dynamics/drug effects , Mitophagy/drug effects , Models, Biological , Phosphorylation/drug effects , Protein Binding/drug effects , Protein Multimerization/drug effects , Selenious Acid/pharmacology , Serine/metabolism , Vinblastine/pharmacology , bcl-2 Homologous Antagonist-Killer Protein/metabolism , bcl-2-Associated X Protein/metabolism
12.
Nat Commun ; 10(1): 1034, 2019 03 04.
Article in English | MEDLINE | ID: mdl-30833558

ABSTRACT

The interactions between tumor cells with their microenvironments, including hypoxia, acidosis and immune cells, lead to the tumor heterogeneity which promotes tumor progression. Here, we show that SIAH2-NRF1 axis remodels tumor microenvironment through regulating tumor mitochondrial function, tumor-associated macrophages (TAMs) polarization and cell death for tumor maintenance and progression. Mechanistically, low mitochondrial gene expression in breast cancers is associated with a poor clinical outcome. The hypoxia-activated E3 ligase SIAH2 spatially downregulates nuclear-encoded mitochondrial gene expression including pyruvate dehydrogenase beta via degrading NRF1 (Nuclear Respiratory Factor 1) through ubiquitination on lysine 230, resulting in enhanced Warburg effect, metabolic reprogramming and pro-tumor immune response. Dampening NRF1 degradation under hypoxia not only impairs the polarization of TAMs, but also promotes tumor cells to become more susceptible to apoptosis in a FADD-dependent fashion, resulting in secondary necrosis due to the impairment of efferocytosis. These data represent that inhibition of NRF1 degradation is a potential therapeutic strategy against cancer.


Subject(s)
Gene Expression Regulation, Neoplastic , Nuclear Proteins/metabolism , Nuclear Respiratory Factor 1/metabolism , Tumor Microenvironment , Ubiquitin-Protein Ligases/metabolism , Animals , Apoptosis/drug effects , Breast Neoplasms/genetics , Breast Neoplasms/immunology , Breast Neoplasms/pathology , Cell Death/drug effects , Cell Line, Tumor , Cellular Reprogramming , Clustered Regularly Interspaced Short Palindromic Repeats/genetics , Disease Models, Animal , Female , Gene Knockout Techniques , Humans , Hypoxia/metabolism , Macrophages/drug effects , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mitochondria/drug effects , Mitochondria/genetics , Nuclear Proteins/genetics , Nuclear Respiratory Factor 1/genetics , RNA, Small Interfering/genetics , Ubiquitin-Protein Ligases/genetics , Ubiquitination
14.
Nat Commun ; 7: 11123, 2016 Mar 31.
Article in English | MEDLINE | ID: mdl-27030211

ABSTRACT

The evolutionarily conserved Hippo pathway is a regulator that controls organ size, cell growth and tissue homeostasis. Upstream signals of the Hippo pathway have been widely studied, but how microenvironmental factors coordinately regulate this pathway remains unclear. In this study, we identify LIM domain protein Zyxin, as a scaffold protein, that in response to hypoxia and TGF-ß stimuli, forms a ternary complex with Lats2 and Siah2 and stabilizes their interaction. This interaction facilitates Lats2 ubiquitination and degradation, Yap dephosphorylation and subsequently activation. We show that Zyxin is required for TGF-ß and hypoxia-induced Lats2 downregulation and deactivation of Hippo signalling in MDA-MB-231 cells. Depletion of Zyxin impairs the capability of cell migration, proliferation and tumourigenesis in a xenograft model. Zyxin is upregulated in human breast cancer and positively correlates with histological stages and metastasis. Our study demonstrates that Zyxin-Lats2-Siah2 axis may serve as a potential therapeutic target in cancer treatment.


Subject(s)
Nuclear Proteins/physiology , Protein Serine-Threonine Kinases/physiology , Transforming Growth Factor beta/physiology , Tumor Suppressor Proteins/physiology , Ubiquitin-Protein Ligases/physiology , Zyxin/physiology , Adaptor Proteins, Signal Transducing/metabolism , Animals , Carcinogenesis/genetics , Cell Hypoxia , Cell Line, Tumor , Cell Movement , Cell Proliferation , Cellular Microenvironment , Female , HEK293 Cells , Heterografts/metabolism , Heterografts/pathology , Hippo Signaling Pathway , Humans , Mice , Mice, Inbred BALB C , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Phosphoproteins/metabolism , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Proteolysis , Signal Transduction , Transcription Factors , Transforming Growth Factor beta/metabolism , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism , Ubiquitination , YAP-Signaling Proteins , Zyxin/genetics , Zyxin/metabolism
15.
Nat Cell Biol ; 17(1): 95-103, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25438054

ABSTRACT

The Hippo signalling pathway plays important roles in animal development, physiology and tumorigenesis. Understanding how the activity of this pathway is regulated by the cellular microenvironment remains a major challenge. Here we elucidate a molecular mechanism by which hypoxia deactivates Hippo signalling. We demonstrate that the E3 ubiquitin ligase SIAH2 stimulates YAP by destabilizing LATS2, a critical component of the Hippo pathway, in response to hypoxia. Loss of SIAH2 suppresses tumorigenesis in a LATS2-dependent manner in a xenograft mouse model. We further show that YAP complexes with HIF1α and is essential for HIF1α stability and function in tumours in vivo. LATS2 is downregulated in human breast tumours and negatively correlates with SIAH2 expression levels, indicating that the SIAH2-LATS2 pathway may have a role in human cancer. Our data uncover oxygen availability as a microenvironment signal for the Hippo pathway and have implications for understanding the regulation of Hippo signalling in tumorigenesis.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Cell Hypoxia/physiology , Nuclear Proteins/metabolism , Phosphoproteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Tumor Suppressor Proteins/metabolism , Ubiquitin-Protein Ligases/metabolism , Adaptor Proteins, Signal Transducing/biosynthesis , Animals , Breast Neoplasms/genetics , Cell Line, Tumor , Cell Transformation, Neoplastic/genetics , Down-Regulation , Female , HEK293 Cells , HeLa Cells , Hippo Signaling Pathway , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Mice , Mice, Nude , Neoplasm Transplantation , Nuclear Proteins/biosynthesis , Nuclear Proteins/genetics , Phosphoproteins/biosynthesis , Phosphorylation , Protein Serine-Threonine Kinases/biosynthesis , RNA Interference , RNA, Small Interfering , Signal Transduction , Transcription Factors , Transplantation, Heterologous , Tumor Microenvironment , Tumor Suppressor Proteins/biosynthesis , Ubiquitin-Protein Ligases/biosynthesis , Ubiquitin-Protein Ligases/genetics , YAP-Signaling Proteins
SELECTION OF CITATIONS
SEARCH DETAIL