Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 3.638
Filter
Add more filters

Publication year range
1.
Cell ; 185(24): 4654-4673.e28, 2022 11 23.
Article in English | MEDLINE | ID: mdl-36334589

ABSTRACT

Brown adipose tissue (BAT) regulates metabolic physiology. However, nearly all mechanistic studies of BAT protein function occur in a single inbred mouse strain, which has limited the understanding of generalizable mechanisms of BAT regulation over physiology. Here, we perform deep quantitative proteomics of BAT across a cohort of 163 genetically defined diversity outbred mice, a model that parallels the genetic and phenotypic variation found in humans. We leverage this diversity to define the functional architecture of the outbred BAT proteome, comprising 10,479 proteins. We assign co-operative functions to 2,578 proteins, enabling systematic discovery of regulators of BAT. We also identify 638 proteins that correlate with protection from, or sensitivity to, at least one parameter of metabolic disease. We use these findings to uncover SFXN5, LETMD1, and ATP1A2 as modulators of BAT thermogenesis or adiposity, and provide OPABAT as a resource for understanding the conserved mechanisms of BAT regulation over metabolic physiology.


Subject(s)
Adipose Tissue, Brown , Proteome , Humans , Mice , Animals , Adipose Tissue, Brown/metabolism , Proteome/metabolism , Thermogenesis/physiology , Adiposity , Obesity/metabolism , Mice, Inbred C57BL , Proto-Oncogene Proteins/metabolism
2.
Nat Immunol ; 25(1): 102-116, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38012418

ABSTRACT

Chimeric antigen receptor (CAR) T cell therapies have successfully treated hematological malignancies. Macrophages have also gained attention as an immunotherapy owing to their immunomodulatory capacity and ability to infiltrate solid tumors and phagocytize tumor cells. The first-generation CD3ζ-based CAR-macrophages could phagocytose tumor cells in an antigen-dependent manner. Here we engineered induced pluripotent stem cell-derived macrophages (iMACs) with toll-like receptor 4 intracellular toll/IL-1R (TIR) domain-containing CARs resulting in a markedly enhanced antitumor effect over first-generation CAR-macrophages. Moreover, the design of a tandem CD3ζ-TIR dual signaling CAR endows iMACs with both target engulfment capacity and antigen-dependent M1 polarization and M2 resistance in a nuclear factor kappa B (NF-κB)-dependent manner, as well as the capacity to modulate the tumor microenvironment. We also outline a mechanism of tumor cell elimination by CAR-induced efferocytosis against tumor cell apoptotic bodies. Taken together, we provide a second-generation CAR-iMAC with an ability for orthogonal phagocytosis and polarization and superior antitumor functions in treating solid tumors relative to first-generation CAR-macrophages.


Subject(s)
Neoplasms , Receptors, Chimeric Antigen , Humans , Receptors, Antigen, T-Cell , T-Lymphocytes , Cell Line, Tumor , Receptors, Chimeric Antigen/genetics , Immunotherapy, Adoptive/methods , Macrophages/pathology , Tumor Microenvironment
3.
Cell ; 182(3): 578-593.e19, 2020 08 06.
Article in English | MEDLINE | ID: mdl-32679029

ABSTRACT

Piloerection (goosebumps) requires concerted actions of the hair follicle, the arrector pili muscle (APM), and the sympathetic nerve, providing a model to study interactions across epithelium, mesenchyme, and nerves. Here, we show that APMs and sympathetic nerves form a dual-component niche to modulate hair follicle stem cell (HFSC) activity. Sympathetic nerves form synapse-like structures with HFSCs and regulate HFSCs through norepinephrine, whereas APMs maintain sympathetic innervation to HFSCs. Without norepinephrine signaling, HFSCs enter deep quiescence by down-regulating the cell cycle and metabolism while up-regulating quiescence regulators Foxp1 and Fgf18. During development, HFSC progeny secretes Sonic Hedgehog (SHH) to direct the formation of this APM-sympathetic nerve niche, which in turn controls hair follicle regeneration in adults. Our results reveal a reciprocal interdependence between a regenerative tissue and its niche at different stages and demonstrate sympathetic nerves can modulate stem cells through synapse-like connections and neurotransmitters to couple tissue production with demands.


Subject(s)
Accessory Nerve/physiology , Hair Follicle/cytology , Hair/growth & development , Hedgehog Proteins/metabolism , Norepinephrine/metabolism , Signal Transduction/genetics , Stem Cells/metabolism , Stem Cells/physiology , Accessory Nerve/cytology , Animals , Cell Cycle/genetics , Cold Temperature , Female , Fibroblast Growth Factors/metabolism , Forkhead Transcription Factors/metabolism , Gene Expression Profiling , Hair/cytology , Hair/physiology , Hair Follicle/growth & development , Hair Follicle/metabolism , Humans , Male , Mice , Mice, Inbred C57BL , Piloerection , RNA-Seq , Receptors, Adrenergic, beta-2/deficiency , Receptors, Adrenergic, beta-2/genetics , Receptors, Adrenergic, beta-2/metabolism , Repressor Proteins/metabolism , Signal Transduction/drug effects , Smoothened Receptor/genetics , Smoothened Receptor/metabolism , Stem Cell Niche , Stem Cells/cytology , Sympathetic Nervous System/cytology , Sympathetic Nervous System/physiology , Synapses/physiology
5.
Mol Cell ; 83(5): 731-745.e4, 2023 03 02.
Article in English | MEDLINE | ID: mdl-36693379

ABSTRACT

The speckle-type POZ protein (SPOP) functions in the Cullin3-RING ubiquitin ligase (CRL3) as a receptor for the recognition of substrates involved in cell growth, survival, and signaling. SPOP mutations have been attributed to the development of many types of cancers, including prostate and endometrial cancers. Prostate cancer mutations localize in the substrate-binding site of the substrate recognition (MATH) domain and reduce or prevent binding. However, most endometrial cancer mutations are dispersed in seemingly inconspicuous solvent-exposed regions of SPOP, offering no clear basis for their cancer-causing and peculiar gain-of-function properties. Herein, we present the first structure of SPOP in its oligomeric form, uncovering several new interfaces important for SPOP self-assembly and normal function. Given that many previously unaccounted-for cancer mutations are localized in these newly identified interfaces, we uncover molecular mechanisms underlying dysregulation of SPOP function, with effects ranging from gross structural changes to enhanced self-association, and heightened stability and activity.


Subject(s)
Prostatic Neoplasms , Transcription Factors , Male , Humans , Ubiquitination , Transcription Factors/metabolism , Repressor Proteins/genetics , Prostatic Neoplasms/genetics , Mutation
6.
Mol Cell ; 82(13): 2427-2442.e4, 2022 07 07.
Article in English | MEDLINE | ID: mdl-35597238

ABSTRACT

The voltage-gated ion channel activity depends on both activation (transition from the resting state to the open state) and inactivation. Inactivation is a self-restraint mechanism to limit ion conduction and is as crucial to membrane excitability as activation. Inactivation can occur when the channel is open or closed. Although open-state inactivation is well understood, the molecular basis of closed-state inactivation has remained elusive. We report cryo-EM structures of human KV4.2 channel complexes in inactivated, open, and closed states. Closed-state inactivation of KV4 involves an unprecedented symmetry breakdown for pore closure by only two of the four S4-S5 linkers, distinct from known mechanisms of open-state inactivation. We further capture KV4 in a putative resting state, revealing how voltage sensor movements control the pore. Moreover, our structures provide insights regarding channel modulation by KChIP2 and DPP6 auxiliary subunits. Our findings elucidate mechanisms of closed-state inactivation and voltage-dependent activation of the KV4 channel.


Subject(s)
Ion Channel Gating , Shal Potassium Channels , Humans , Ion Channel Gating/physiology , Kinetics , Membrane Potentials/physiology , Shal Potassium Channels/genetics , Shal Potassium Channels/metabolism
7.
Cell ; 156(3): 440-55, 2014 Jan 30.
Article in English | MEDLINE | ID: mdl-24485453

ABSTRACT

Lung stem cells are instructed to produce lineage-specific progeny through unknown factors in their microenvironment. We used clonal 3D cocultures of endothelial cells and distal lung stem cells, bronchioalveolar stem cells (BASCs), to probe the instructive mechanisms. Single BASCs had bronchiolar and alveolar differentiation potential in lung endothelial cell cocultures. Gain- and loss-of-function experiments showed that BMP4-Bmpr1a signaling triggers calcineurin/NFATc1-dependent expression of thrombospondin-1 (Tsp1) in lung endothelial cells to drive alveolar lineage-specific BASC differentiation. Tsp1 null mice exhibited defective alveolar injury repair, confirming a crucial role for the BMP4-NFATc1-TSP1 axis in lung epithelial differentiation and regeneration in vivo. Discovery of this pathway points to methods to direct the derivation of specific lung epithelial lineages from multipotent cells. These findings elucidate a pathway that may be a critical target in lung diseases and provide tools to understand the mechanisms of respiratory diseases at the single-cell level.


Subject(s)
Bronchioles/cytology , Cell Differentiation , Endothelial Cells/metabolism , Pulmonary Alveoli/cytology , Signal Transduction , Stem Cells/metabolism , Animals , Bone Morphogenetic Protein 4/metabolism , Bone Morphogenetic Protein Receptors, Type I/metabolism , Bronchioles/metabolism , Cells, Cultured , Coculture Techniques , Mice , NFATC Transcription Factors/metabolism , Pulmonary Alveoli/metabolism , Stem Cells/cytology , Thrombospondin 1/genetics , Thrombospondin 1/metabolism
8.
Cell ; 158(1): 69-83, 2014 Jul 03.
Article in English | MEDLINE | ID: mdl-24995979

ABSTRACT

Brown fat can reduce obesity through the dissipation of calories as heat. Control of thermogenic gene expression occurs via the induction of various coactivators, most notably PGC-1α. In contrast, the transcription factor partner(s) of these cofactors are poorly described. Here, we identify interferon regulatory factor 4 (IRF4) as a dominant transcriptional effector of thermogenesis. IRF4 is induced by cold and cAMP in adipocytes and is sufficient to promote increased thermogenic gene expression, energy expenditure, and cold tolerance. Conversely, knockout of IRF4 in UCP1(+) cells causes reduced thermogenic gene expression and energy expenditure, obesity, and cold intolerance. IRF4 also induces the expression of PGC-1α and PRDM16 and interacts with PGC-1α, driving Ucp1 expression. Finally, cold, ß-agonists, or forced expression of PGC-1α are unable to cause thermogenic gene expression in the absence of IRF4. These studies establish IRF4 as a transcriptional driver of a program of thermogenic gene expression and energy expenditure.


Subject(s)
Adipose Tissue, Brown/metabolism , Interferon Regulatory Factors/metabolism , Thermogenesis , Transcription Factors/metabolism , Transcriptional Activation , Adipocytes/metabolism , Adipose Tissue, Brown/cytology , Adrenergic beta-3 Receptor Agonists/pharmacology , Animals , Cold Temperature , Cyclic AMP/metabolism , Energy Metabolism , Humans , Ion Channels/genetics , Mice , Mitochondria/metabolism , Mitochondrial Proteins/genetics , Obesity/metabolism , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , Thinness/metabolism , Transcriptional Activation/drug effects , Uncoupling Protein 1
9.
Cell ; 152(3): 479-91, 2013 Jan 31.
Article in English | MEDLINE | ID: mdl-23374344

ABSTRACT

Fast axonal transport (FAT) requires consistent energy over long distances to fuel the molecular motors that transport vesicles. We demonstrate that glycolysis provides ATP for the FAT of vesicles. Although inhibiting ATP production from mitochondria did not affect vesicles motility, pharmacological or genetic inhibition of the glycolytic enzyme GAPDH reduced transport in cultured neurons and in Drosophila larvae. GAPDH localizes on vesicles via a huntingtin-dependent mechanism and is transported on fast-moving vesicles within axons. Purified motile vesicles showed GAPDH enzymatic activity and produced ATP. Finally, we show that vesicular GAPDH is necessary and sufficient to provide on-board energy for fast vesicular transport. Although detaching GAPDH from vesicles reduced transport, targeting GAPDH to vesicles was sufficient to promote FAT in GAPDH deficient neurons. This specifically localized glycolytic machinery may supply constant energy, independent of mitochondria, for the processive movement of vesicles over long distances in axons.


Subject(s)
Axonal Transport , Drosophila melanogaster/metabolism , Glycolysis , Neurons/metabolism , Adenosine Triphosphate/metabolism , Animals , Axons/metabolism , Brain/cytology , Cells, Cultured , Drosophila melanogaster/growth & development , Glyceraldehyde-3-Phosphate Dehydrogenases/metabolism , Mice , Mitochondria/metabolism , Rats
10.
EMBO J ; 42(12): e112675, 2023 06 15.
Article in English | MEDLINE | ID: mdl-37092319

ABSTRACT

Tumor cells surviving hypoxic stress acquire the ability to drive cancer progression. To explore the contribution of dehydrogenases to the low oxygen concentration response, we used siRNAs targeting 163 dehydrogenase-coding genes and discovered that glutamate dehydrogenase 1 (GDH1) plays a critical role in regulating colorectal cancer (CRC) cell survival under hypoxia. We observed that GDH1 deficiency had an inhibitory effect on CRC occurrence and impaired hypoxia-inducible factor 1-alpha (HIF-1α) stability even under hypoxia. Mechanistically, hypoxia triggered p300 recruitment to GDH1, promoting its acetylation at K503 and K527. GDH1 acetylation at K527 induced the formation of a GDH1 complex with EGLN1/HIF-1α; in contrast, GDH1 acetylation at K503 reinforced its affinity for α-ketoglutarate (αKG), and glutamate production. In line with this view, αKG is a product of GDH1 under normoxia, but hypoxia stimulation reversed GDH1 enzyme activity and αKG consumption by the EGLN1/HIF-1α complex, increasing HIF-1α stability and promoting CRC progression. Clinically, hypoxia-modulated GDH1 AcK503/527 can be used as a biomarker of CRC progression and is a potential target for CRC treatment.


Subject(s)
Colorectal Neoplasms , Glutamic Acid , Humans , Glutamic Acid/metabolism , Hypoxia , Cell Hypoxia/genetics , Cell Transformation, Neoplastic , Carcinogenesis , Colorectal Neoplasms/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Cell Line, Tumor
11.
Mol Cell ; 76(1): 148-162.e7, 2019 10 03.
Article in English | MEDLINE | ID: mdl-31447391

ABSTRACT

The rapid proliferation of cancer cells and dysregulated vasculature within the tumor leads to limited nutrient accessibility. Cancer cells often rewire their metabolic pathways for adaption to nutrient stress, and the underlying mechanism remains largely unknown. Glutamate dehydrogenase 1 (GDH1) is a key enzyme in glutaminolysis that converts glutamate to α-ketoglutarate (α-KG). Here, we show that, under low glucose, GDH1 is phosphorylated at serine (S) 384 and interacts with RelA and IKKß. GDH1-produced α-KG directly binds to and activates IKKß and nuclear factor κB (NF-κB) signaling, which promotes glucose uptake and tumor cell survival by upregulating GLUT1, thereby accelerating gliomagenesis. In addition, GDH1 S384 phosphorylation correlates with the malignancy and prognosis of human glioblastoma. Our finding reveals a unique role of α-KG to directly regulate signal pathway, uncovers a distinct mechanism of metabolite-mediated NF-κB activation, and also establishes the critical role of α-KG-activated NF-κB in brain tumor development.


Subject(s)
Brain Neoplasms/metabolism , Cell Proliferation , Energy Metabolism , Glioblastoma/metabolism , Glucose/metabolism , Glutamate Dehydrogenase/metabolism , Ketoglutaric Acids/metabolism , NF-kappa B/metabolism , Adolescent , Adult , Aged , Aged, 80 and over , Animals , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Cell Line, Tumor , Cell Survival , Child , Female , Gene Expression Regulation, Neoplastic , Glioblastoma/genetics , Glioblastoma/pathology , Glucose/deficiency , Glucose Transporter Type 1/genetics , Glucose Transporter Type 1/metabolism , Glutamate Dehydrogenase/genetics , HEK293 Cells , Humans , I-kappa B Kinase/genetics , I-kappa B Kinase/metabolism , Male , Mice, Inbred BALB C , Mice, Nude , Middle Aged , NF-kappa B/genetics , Neoplasm Grading , Phosphorylation , Signal Transduction , Transcription Factor RelA/genetics , Transcription Factor RelA/metabolism , Young Adult
12.
Proc Natl Acad Sci U S A ; 121(14): e2315982121, 2024 Apr 02.
Article in English | MEDLINE | ID: mdl-38536757

ABSTRACT

Throughout evolution, arboviruses have developed various strategies to counteract the host's innate immune defenses to maintain persistent transmission. Recent studies have shown that, in addition to bacteria and fungi, the innate Toll-Dorsal immune system also plays an essential role in preventing viral infections in invertebrates. However, whether the classical Toll immune pathway is involved in maintaining the homeostatic process to ensure the persistent and propagative transmission of arboviruses in insect vectors remain unclear. In this study, we revealed that the transcription factor Dorsal is actively involved in the antiviral defense of an insect vector (Laodelphax striatellus) by regulating the target gene, zinc finger protein 708 (LsZN708), which mediates downstream immune-related effectors against infection with the plant virus (Rice stripe virus, RSV). In contrast, an antidefense strategy involving the use of the nonstructural-protein (NS4) to antagonize host antiviral defense through competitive binding to Dorsal from the MSK2 kinase was employed by RSV; this competitive binding inhibited Dorsal phosphorylation and reduced the antiviral response of the host insect. Our study revealed the molecular mechanism through which Toll-Dorsal-ZN708 mediates the maintenance of an arbovirus homeostasis in insect vectors. Specifically, ZN708 is a newly documented zinc finger protein targeted by Dorsal that mediates the downstream antiviral response. This study will contribute to our understanding of the successful transmission and spread of arboviruses in plant or invertebrate hosts.


Subject(s)
Arboviruses , Hemiptera , Oryza , Tenuivirus , Animals , Arboviruses/genetics , Hemiptera/physiology , Tenuivirus/physiology , Insect Vectors , Antiviral Agents/metabolism , Oryza/genetics , Plant Diseases
13.
N Engl J Med ; 388(8): 694-705, 2023 02 23.
Article in English | MEDLINE | ID: mdl-36812433

ABSTRACT

BACKGROUND: Valoctocogene roxaparvovec delivers a B-domain-deleted factor VIII coding sequence with an adeno-associated virus vector to prevent bleeding in persons with severe hemophilia A. The findings of a phase 3 study of the efficacy and safety of valoctocogene roxaparvovec therapy evaluated after 52 weeks in men with severe hemophilia A have been published previously. METHODS: We conducted an open-label, single-group, multicenter, phase 3 trial in which 134 men with severe hemophilia A who were receiving factor VIII prophylaxis received a single infusion of 6×1013 vector genomes of valoctocogene roxaparvovec per kilogram of body weight. The primary end point was the change from baseline in the annualized rate of treated bleeding events at week 104 after receipt of the infusion. The pharmacokinetics of valoctocogene roxaparvovec were modeled to estimate the bleeding risk relative to the activity of transgene-derived factor VIII. RESULTS: At week 104, a total of 132 participants, including 112 with data that were prospectively collected at baseline, remained in the study. The mean annualized treated bleeding rate decreased by 84.5% from baseline (P<0.001) among the participants. From week 76 onward, the trajectory of the transgene-derived factor VIII activity showed first-order elimination kinetics; the model-estimated typical half-life of the transgene-derived factor VIII production system was 123 weeks (95% confidence interval, 84 to 232). The risk of joint bleeding was estimated among the trial participants; at a transgene-derived factor VIII level of 5 IU per deciliter measured with chromogenic assay, we expected that participants would have 1.0 episode of joint bleeding per year. At 2 years postinfusion, no new safety signals had emerged and no new serious adverse events related to treatment had occurred. CONCLUSIONS: The study data show the durability of factor VIII activity and bleeding reduction and the safety profile of valoctocogene roxaparvovec at least 2 years after the gene transfer. Models of the risk of joint bleeding suggest that the relationship between transgene-derived factor VIII activity and bleeding episodes is similar to that reported with the use of epidemiologic data for persons with mild-to-moderate hemophilia A. (Funded by BioMarin Pharmaceutical; GENEr8-1 ClinicalTrials.gov number, NCT03370913.).


Subject(s)
Factor VIII , Hemophilia A , Humans , Male , Factor VIII/therapeutic use , Gene Transfer Techniques , Half-Life , Hemophilia A/complications , Hemophilia A/drug therapy , Hemorrhage/etiology , Hemorrhage/prevention & control , Recombinant Fusion Proteins/therapeutic use
14.
Circ Res ; 134(7): e17-e33, 2024 Mar 29.
Article in English | MEDLINE | ID: mdl-38420756

ABSTRACT

BACKGROUND: Microvascular complications are the major outcome of type 2 diabetes progression, and the underlying mechanism remains to be determined. METHODS: High-throughput RNA sequencing was performed using human monocyte samples from controls and diabetes. The transgenic mice expressing human CTSD (cathepsin D) in the monocytes was constructed using CD68 promoter. In vivo 2-photon imaging, behavioral tests, immunofluorescence, transmission electron microscopy, Western blot analysis, vascular leakage assay, and single-cell RNA sequencing were performed to clarify the phenotype and elucidate the molecular mechanism. RESULTS: Monocytes expressed high-level CTSD in patients with type 2 diabetes. The transgenic mice expressing human CTSD in the monocytes showed increased brain microvascular permeability resembling the diabetic microvascular phenotype, accompanied by cognitive deficit. Mechanistically, the monocytes release nonenzymatic pro-CTSD to upregulate caveolin expression in brain endothelium triggering caveolae-mediated transcytosis, without affecting the paracellular route of brain microvasculature. The circulating pro-CTSD activated the caveolae-mediated transcytosis in brain endothelial cells via its binding with low-density LRP1 (lipoprotein receptor-related protein 1). Importantly, genetic ablation of CTSD in the monocytes exhibited a protective effect against the diabetes-enhanced brain microvascular transcytosis and the diabetes-induced cognitive impairment. CONCLUSIONS: These findings uncover the novel role of circulatory pro-CTSD from monocytes in the pathogenesis of cerebral microvascular lesions in diabetes. The circulatory pro-CTSD is a potential target for the intervention of microvascular complications in diabetes.


Subject(s)
Cathepsin D , Diabetes Mellitus, Type 2 , Monocytes , Animals , Humans , Mice , Brain/metabolism , Cathepsin D/metabolism , Cathepsin D/pharmacology , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/metabolism , Endothelial Cells/metabolism , Endothelium, Vascular/metabolism , Enzyme Precursors , Mice, Transgenic , Monocytes/metabolism , Transcytosis/physiology
15.
EMBO J ; 40(20): e107480, 2021 10 18.
Article in English | MEDLINE | ID: mdl-34269483

ABSTRACT

The mTORC1 pathway plays key roles in regulating various biological processes, including sensing amino acid deprivation and driving expression of ribosomal protein (RP)-coding genes. In this study, we observed that depletion of glutamate dehydrogenase 1 (GDH1), an enzyme that converts glutamate to α-ketoglutarate (αKG), confers resistance to amino acid deprivation on kidney renal clear cell carcinoma (KIRC) cells. Mechanistically, under conditions of adequate nutrition, GDH1 maintains RP gene expression in a manner dependent on its enzymatic activity. Following amino acid deprivation or mTORC1 inhibition, GDH1 translocates from mitochondria to the cytoplasm, where it becomes ubiquitinated and degraded via the E3 ligase RNF213. GDH1 degradation reduces intracellular αKG levels by more than half and decreases the activity of αKG-dependent lysine demethylases (KDMs). Reduced KDM activity in turn leads to increased histone H3 lysine 9 and 27 methylation, further suppressing RP gene expression and preserving nutrition to support cell survival. In summary, our study exemplifies an economical and efficient strategy of solid tumor cells for coping with amino acid deficiency, which might in the future be targeted to block renal carcinoma progression.


Subject(s)
Carcinoma, Renal Cell/genetics , Glutamate Dehydrogenase/genetics , Glutamic Acid/metabolism , Ketoglutaric Acids/metabolism , Kidney Neoplasms/genetics , Adenosine Triphosphatases/genetics , Adenosine Triphosphatases/metabolism , Animals , Carcinoma, Renal Cell/metabolism , Carcinoma, Renal Cell/mortality , Carcinoma, Renal Cell/pathology , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Line, Tumor , DNA-Binding Proteins/antagonists & inhibitors , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Epithelial Cells/metabolism , Epithelial Cells/pathology , Female , Gene Expression Regulation, Neoplastic , Glutamate Dehydrogenase/antagonists & inhibitors , Glutamate Dehydrogenase/metabolism , Histone Demethylases/genetics , Histone Demethylases/metabolism , Humans , Jumonji Domain-Containing Histone Demethylases/genetics , Jumonji Domain-Containing Histone Demethylases/metabolism , Kidney Neoplasms/metabolism , Kidney Neoplasms/mortality , Kidney Neoplasms/pathology , Mice , Mice, Nude , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Nuclear Proteins/antagonists & inhibitors , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Poly-ADP-Ribose Binding Proteins/genetics , Poly-ADP-Ribose Binding Proteins/metabolism , Protein Isoforms/genetics , Protein Isoforms/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Ribosomal Proteins/genetics , Ribosomal Proteins/metabolism , Signal Transduction , Survival Analysis , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism , Xenograft Model Antitumor Assays
16.
Genome Res ; 32(2): 242-257, 2022 02.
Article in English | MEDLINE | ID: mdl-35042723

ABSTRACT

Single-cell RNA sequencing (scRNA-seq) enables molecular characterization of complex biological tissues at high resolution. The requirement of single-cell extraction, however, makes it challenging for profiling tissues such as adipose tissue, for which collection of intact single adipocytes is complicated by their fragile nature. For such tissues, single-nucleus extraction is often much more efficient and therefore single-nucleus RNA sequencing (snRNA-seq) presents an alternative to scRNA-seq. However, nuclear transcripts represent only a fraction of the transcriptome in a single cell, with snRNA-seq marked with inherent transcript enrichment and detection biases. Therefore, snRNA-seq may be inadequate for mapping important transcriptional signatures in adipose tissue. In this study, we compare the transcriptomic landscape of single nuclei isolated from preadipocytes and mature adipocytes across human white and brown adipocyte lineages, with whole-cell transcriptome. We show that snRNA-seq is capable of identifying the broad cell types present in scRNA-seq at all states of adipogenesis. However, we also explore how and why the nuclear transcriptome is biased and limited, as well as how it can be advantageous. We robustly characterize the enrichment of nuclear-localized transcripts and adipogenic regulatory lncRNAs in snRNA-seq, while also providing a detailed understanding for the preferential detection of long genes upon using this technique. To remove such technical detection biases, we propose a normalization strategy for a more accurate comparison of nuclear and cellular data. Finally, we show successful integration of scRNA-seq and snRNA-seq data sets with existing bioinformatic tools. Overall, our results illustrate the applicability of snRNA-seq for the characterization of cellular diversity in the adipose tissue.


Subject(s)
Adipocytes/cytology , Cell Lineage , Gene Expression Profiling , RNA-Seq , Single-Cell Analysis , Bias , Gene Expression Profiling/methods , Humans , RNA-Seq/methods , Single-Cell Analysis/methods , Transcriptome
17.
Immunity ; 44(4): 913-923, 2016 Apr 19.
Article in English | MEDLINE | ID: mdl-27096320

ABSTRACT

The participation of a specific subset of B cells and how they are regulated in cancer is unclear. Here, we demonstrate that the proportion of CD5(+) relative to interleukin-6 receptor α (IL-6Rα)-expressing B cells was greatly increased in tumors. CD5(+) B cells responded to IL-6 in the absence of IL-6Rα. IL-6 directly bound to CD5, leading to activation of the transcription factor STAT3 via gp130 and its downstream kinase JAK2. STAT3 upregulated CD5 expression, thereby forming a feed-forward loop in the B cells. In mouse tumor models, CD5(+) but not CD5(-) B cells promoted tumor growth. CD5(+) B cells also showed activation of STAT3 in multiple types of human tumor tissues. Thus, our findings demonstrate a critical role of CD5(+) B cells in promoting cancer.


Subject(s)
B-Lymphocytes/immunology , CD5 Antigens/metabolism , Interleukin-6/metabolism , Melanoma, Experimental/pathology , STAT3 Transcription Factor/immunology , Animals , CD5 Antigens/biosynthesis , Cell Line, Tumor , Cytokine Receptor gp130/metabolism , Humans , Interleukin-6/immunology , Janus Kinase 2/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , NIH 3T3 Cells , Protein Binding , Receptors, Interleukin-6/biosynthesis , Receptors, Interleukin-6/genetics , Receptors, Interleukin-6/immunology , Transcriptional Activation/immunology
18.
Circ Res ; 133(4): 353-365, 2023 08 04.
Article in English | MEDLINE | ID: mdl-37462036

ABSTRACT

BACKGROUND: Despite advances in treatment, myocardial infarction (MI) is a leading cause of heart failure and death worldwide, with both ischemia and reperfusion (I/R) causing cardiac injury. A previous study using a mouse model of nonreperfused MI showed activation of brown adipose tissue (BAT). Recent studies showed that molecules secreted by BAT target the heart. We investigated whether BAT attenuates cardiac injury in I/R and sought to identify potential cardioprotective proteins secreted by BAT. METHODS: Myocardial I/R surgery with or without BAT transplantation was performed in wild-type (WT) mice and in mice with impaired BAT function (uncoupling protein 1 [Ucp1]-deficient mice). To identify potential cardioprotective factors produced by BAT, RNA-seq (RNA sequencing) was performed in BAT from WT and Ucp1-/- mice. Subsequently, myocardial I/R surgery with or without BAT transplantation was performed in Bmp3b (bone morphogenetic protein 3b)-deficient mice, and WT mice subjected to myocardial I/R were treated using BMP3b. RESULTS: Dysfunction of BAT in mice was associated with larger MI size after I/R; conversely, augmenting BAT by transplantation decreased MI size. We identified Bmp3b as a protein secreted by BAT after I/R. Compared with WT mice, Bmp3b-deficient mice developed larger MIs. Increasing functional BAT by transplanting BAT from WT mice to Bmp3b-deficient mice reduced I/R injury whereas transplanting BAT from Bmp3b-deficient mice did not. Treatment of WT mice with BMP3b before reperfusion decreased MI size. The cardioprotective effect of BMP3b was mediated through SMAD1/5/8. In humans, the plasma level of BMP3b increased after MI and was positively correlated with the extent of cardiac injury. CONCLUSIONS: The results of this study suggest a cardioprotective role of BAT and BMP3b, a protein secreted by BAT, in a model of I/R injury. Interventions increasing BMP3b levels or targeting Smad 1/5 may represent novel therapeutic approaches to decrease myocardial damage in I/R injury.


Subject(s)
Coronary Artery Disease , Growth Differentiation Factor 10 , Myocardial Infarction , Myocardial Ischemia , Myocardial Reperfusion Injury , Animals , Humans , Mice , Adipose Tissue, Brown/metabolism , Growth Differentiation Factor 10/metabolism , Mice, Inbred C57BL , Myocardial Infarction/metabolism , Myocardial Reperfusion Injury/genetics , Myocardial Reperfusion Injury/prevention & control , Myocardial Reperfusion Injury/drug therapy , Reperfusion
19.
Exp Cell Res ; 440(1): 114125, 2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38880324

ABSTRACT

Bladder cancer(BC) is one of the most prevalent cancers in the urinary tract, with high recurrence and fatality rates. Research indicates that go-ichi-ni-san complex subunit 1 (GINS1) crucially influences cancer progression by regulating DNA replication through cell cycle modulation. Thus, suppressing the active proliferation of cells in tumor tissues may require silencing GINS1. However, the consequences of GINS1 in bladder cancer aren't to be determined. In this paper, we examine the role and mechanism of GINS1 in the development of bladder cancer. GINS1 expression levels and prognostic relevance in bladder cancer were validated using Western blotting, immunohistochemistry, and Kaplan-Meier survival analysis. The influence of GINS1 on bladder cancer was investigated using a variety of approaches, including cell transfection, cell counts, transwell migrations, colony formation, and flow cytometry. Immunohistochemistry studies demonstrate that GINS1 expression is increased in bladder cancer tissues. GINS1 silencing resulted in an arrest of the cell cycle at the phase of G0/G1, which inhibited BC cell growth both in vitro and in vivo. GINS1 knockdown also hindered the AKT/mTOR pathway. Furthermore, increased GINS1 expression affects the cell cycle and stimulates the AKT/mTOR pathway, allowing BC to develop more quickly. Consequently, GINS1 occurs as a latent therapeutic target, particularly for individuals with BC.


Subject(s)
Cell Proliferation , Chromosomal Proteins, Non-Histone , Proto-Oncogene Proteins c-akt , Signal Transduction , TOR Serine-Threonine Kinases , Urinary Bladder Neoplasms , Urinary Bladder Neoplasms/pathology , Urinary Bladder Neoplasms/genetics , Urinary Bladder Neoplasms/metabolism , Humans , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins c-akt/genetics , TOR Serine-Threonine Kinases/metabolism , TOR Serine-Threonine Kinases/genetics , Chromosomal Proteins, Non-Histone/metabolism , Chromosomal Proteins, Non-Histone/genetics , Cell Proliferation/genetics , Animals , Cell Line, Tumor , Proto-Oncogene Proteins c-myc/metabolism , Proto-Oncogene Proteins c-myc/genetics , Gene Expression Regulation, Neoplastic , Mice , Disease Progression , Mice, Nude , Male , Female , Prognosis , Mice, Inbred BALB C , DNA-Binding Proteins
20.
Nature ; 571(7763): 127-131, 2019 07.
Article in English | MEDLINE | ID: mdl-31243371

ABSTRACT

Cancer metastasis is the primary cause of morbidity and mortality, and accounts for up to 95% of cancer-related deaths1. Cancer cells often reprogram their metabolism to efficiently support cell proliferation and survival2,3. However, whether and how those metabolic alterations contribute to the migration of tumour cells remain largely unknown. UDP-glucose 6-dehydrogenase (UGDH) is a key enzyme in the uronic acid pathway, and converts UDP-glucose to UDP-glucuronic acid4. Here we show that, after activation of EGFR, UGDH is phosphorylated at tyrosine 473 in human lung cancer cells. Phosphorylated UGDH interacts with Hu antigen R (HuR) and converts UDP-glucose to UDP-glucuronic acid, which attenuates the UDP-glucose-mediated inhibition of the association of HuR with SNAI1 mRNA and therefore enhances the stability of SNAI1 mRNA. Increased production of SNAIL initiates the epithelial-mesenchymal transition, thus promoting the migration of tumour cells and lung cancer metastasis. In addition, phosphorylation of UGDH at tyrosine 473 correlates with metastatic recurrence and poor prognosis of patients with lung cancer. Our findings reveal a tumour-suppressive role of UDP-glucose in lung cancer metastasis and uncover a mechanism by which UGDH promotes tumour metastasis by increasing the stability of SNAI1 mRNA.


Subject(s)
Lung Neoplasms/genetics , Lung Neoplasms/pathology , Neoplasm Metastasis/genetics , Neoplasm Metastasis/prevention & control , RNA Stability , Snail Family Transcription Factors/genetics , Uridine Diphosphate Glucose/metabolism , Animals , Cell Line, Tumor , Cell Movement , ELAV-Like Protein 1/deficiency , ELAV-Like Protein 1/genetics , ELAV-Like Protein 1/metabolism , Epithelial-Mesenchymal Transition , Female , Humans , Lung Neoplasms/enzymology , Lung Neoplasms/metabolism , Mice , Mice, Nude , Phosphotyrosine/metabolism , Prognosis , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Neoplasm/genetics , RNA, Neoplasm/metabolism , Snail Family Transcription Factors/biosynthesis , Uridine Diphosphate Glucose Dehydrogenase/chemistry , Uridine Diphosphate Glucose Dehydrogenase/genetics , Uridine Diphosphate Glucose Dehydrogenase/metabolism , Uridine Diphosphate Glucuronic Acid/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL