Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 81.023
Filter
1.
Pak J Pharm Sci ; 37(2(Special)): 423-428, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38822545

ABSTRACT

This study assessed the inhibitory effect of sodium valproate (VPA) on apoptosis of cardiomyocytes in lethally scalded rats. The model of a 50% total body surface area (TBSA) third-degree full-thickness scald was produced, 48 male SD rats were randomly divided into three groups (n = 16), the sham group and the scald group were given an intraperitoneal injection of 0.25ml of saline, the scald +VPA group was given an intraperitoneal injection of VPA (300 mg/kg) after scalded, Each group was subdivided into two subgroups (n=8) according to the two observation time points of 3h and 6h after scald. Apoptotic cardiomyocytes were observed, and myocardial tissue levels of nitric oxide (NO), cysteine protease-3 (caspase-3) activity, hypoxia-inducible factor-1α (HIF-1α), inducible nitric oxide synthase (iNOS), BCL2/adenovirus E1B interacting protein 3 (BNIP3) and caspase-3 protein were measured. Compared with sham scald group, severe scald elevated CK-MB, cardiomyocyte apoptosis rate, caspase-3 activity and protein levels, NO content, and HIF-1α signalling pathway proteins; whereas VPA decreased CK-MB, cardiomyocyte apoptosis rate and inhibited HIF-1α signalling pathway protein expression. In conclusion, these results suggested that VPA inhibited early cardiomyocyte apoptosis and attenuated myocardial injury in lethally scalded rats, which may be related to the regulation of the HIF-1α signalling pathway.


Subject(s)
Apoptosis , Burns , Caspase 3 , Hypoxia-Inducible Factor 1, alpha Subunit , Myocytes, Cardiac , Nitric Oxide , Rats, Sprague-Dawley , Valproic Acid , Animals , Valproic Acid/pharmacology , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Apoptosis/drug effects , Male , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Burns/drug therapy , Burns/metabolism , Burns/pathology , Caspase 3/metabolism , Nitric Oxide/metabolism , Rats , Nitric Oxide Synthase Type II/metabolism , Membrane Proteins/metabolism , Disease Models, Animal , Mitochondrial Proteins
2.
FASEB J ; 38(11): e23710, 2024 Jun 15.
Article in English | MEDLINE | ID: mdl-38822676

ABSTRACT

Steroidogenic tissues contain cytosolic lipid droplets that are important for steroidogenesis. Perilipin 2 (PLIN2), a structural coat protein located on the surface of lipid droplets in mammalian cells, plays a crucial role in regulating lipid droplet formation and contributing to various cellular processes such as lipid storage and energy homeostasis. Herein, we examine the role that PLIN2 plays in regulating progesterone synthesis in the bovine corpus luteum. Utilizing gene array databases and Western blotting, we have delineated the expression pattern of PLIN2 throughout the follicular to luteal transition. Our findings reveal the presence of PLIN2 in both ovarian follicular and steroidogenic luteal cells, demonstrating an increase in its levels as follicular cells transition into the luteal phase. Moreover, the depletion of PLIN2 via siRNA enhanced progesterone production in small luteal cells, whereas adenovirus-mediated overexpression of both PLIN2 and Perilipin 3 (PLIN3) induced an increase in cytosolic lipid droplet accumulation and decreased hormone-induced progesterone synthesis in these cells. Lastly, in vivo administration of the luteolytic hormone prostaglandin F2α resulted in an upregulation of PLIN2 mRNA and protein expression, accompanied by a decline in serum progesterone. Our findings highlight the pivotal role of PLIN2 in regulating progesterone synthesis in the bovine corpus luteum, as supported by its dynamic expression pattern during the follicular to luteal transition and its responsiveness to luteotropic and luteolytic hormones. We suggest PLIN2 as a potential therapeutic target for modulating luteal function.


Subject(s)
Luteal Cells , Perilipin-2 , Progesterone , Animals , Female , Cattle , Progesterone/metabolism , Perilipin-2/metabolism , Perilipin-2/genetics , Luteal Cells/metabolism , Lipid Droplets/metabolism , Membrane Proteins/metabolism , Membrane Proteins/genetics , Perilipin-3/metabolism , Corpus Luteum/metabolism , Cells, Cultured
3.
Arch Dermatol Res ; 316(6): 323, 2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38822901

ABSTRACT

Refractory diabetic wounds are still a clinical challenge that can cause persistent inflammation and delayed healing. Exosomes of adipose stem cells (ADSC-exos) are the potential strategy for wound repair; however, underlying mechanisms remain mysterious. In this study, we isolated ADSC-exos and identified their characterization. High glucose (HG) stimulated human umbilical vein endothelial cells (HUVECs) to establish in vitro model. The biological behaviors were analyzed by Transwell, wound healing, and tube formation assays. The underlying mechanisms were analyzed using quantitative real-time PCR, co-immunoprecipitation (Co-IP), IP, and western blot. The results showed that ADSC-exos promoted HG-inhibited cell migration and angiogenesis. In addition, ADSC-exos increased the levels of TRIM32 in HG-treated HUVECs, which promoted the ubiquitination of STING and downregulated STING protein levels. Rescue experiments affirmed that ADSC-exos promoted migration and angiogenesis of HG-treated HUVECs by regulating the TRIM32/STING axis. In conclusion, ADSC-exos increased the levels of TRIM32, which interacted with STING and promoted its ubiquitination, downregulating STING levels, thus promoting migration and angiogenesis of HG-treated HUVECs. The findings suggested that ADSC-exos could promote diabetic wound healing and demonstrated a new mechanism of ADSC-exos.


Subject(s)
Cell Movement , Exosomes , Glucose , Human Umbilical Vein Endothelial Cells , Membrane Proteins , Tripartite Motif Proteins , Ubiquitin-Protein Ligases , Wound Healing , Humans , Exosomes/metabolism , Ubiquitin-Protein Ligases/metabolism , Ubiquitin-Protein Ligases/genetics , Tripartite Motif Proteins/metabolism , Tripartite Motif Proteins/genetics , Glucose/metabolism , Membrane Proteins/metabolism , Adipose Tissue/metabolism , Adipose Tissue/cytology , Signal Transduction , Ubiquitination , Neovascularization, Physiologic , Cells, Cultured , Stem Cells/metabolism , Transcription Factors
4.
Proc Natl Acad Sci U S A ; 121(23): e2317790121, 2024 Jun 04.
Article in English | MEDLINE | ID: mdl-38814866

ABSTRACT

The transformation of lung adenocarcinoma to small cell lung cancer (SCLC) is a recognized resistance mechanism and a hindrance to therapies using epidermal growth factor receptor tyrosine kinase inhibitors (TKIs). The paucity of pretranslational/posttranslational clinical samples limits the deeper understanding of resistance mechanisms and the exploration of effective therapeutic strategies. Here, we developed preclinical neuroendocrine (NE) transformation models. Next, we identified a transcriptional reprogramming mechanism that drives resistance to erlotinib in NE transformation cell lines and cell-derived xenograft mice. We observed the enhanced expression of genes involved in the EHMT2 and WNT/ß-catenin pathways. In addition, we demonstrated that EHMT2 increases methylation of the SFRP1 promoter region to reduce SFRP1 expression, followed by activation of the WNT/ß-catenin pathway and TKI-mediated NE transformation. Notably, the similar expression alterations of EHMT2 and SFRP1 were observed in transformed SCLC samples obtained from clinical patients. Importantly, suppression of EHMT2 with selective inhibitors restored the sensitivity of NE transformation cell lines to erlotinib and delayed resistance in cell-derived xenograft mice. We identify a transcriptional reprogramming process in NE transformation and provide a potential therapeutic target for overcoming resistance to erlotinib.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Cell Transformation, Neoplastic , Erlotinib Hydrochloride , Lung Neoplasms , Humans , Animals , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Lung Neoplasms/drug therapy , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/pathology , Carcinoma, Non-Small-Cell Lung/drug therapy , Mice , Erlotinib Hydrochloride/pharmacology , Cell Line, Tumor , Cell Transformation, Neoplastic/genetics , Gene Expression Regulation, Neoplastic , Drug Resistance, Neoplasm/genetics , Wnt Signaling Pathway/genetics , Membrane Proteins/genetics , Membrane Proteins/metabolism , Protein Kinase Inhibitors/pharmacology , Xenograft Model Antitumor Assays , Small Cell Lung Carcinoma/genetics , Small Cell Lung Carcinoma/metabolism , Small Cell Lung Carcinoma/pathology , Transcription, Genetic , Histocompatibility Antigens , Histone-Lysine N-Methyltransferase
5.
J Med Virol ; 96(6): e29712, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38808555

ABSTRACT

Swine acute diarrhea syndrome coronavirus (SADS-CoV) has caused severe intestinal diseases in pigs. It originates from bat coronaviruses HKU2 and has a potential risk of cross-species transmission, raising concerns about its zoonotic potential. Viral entry-related host factors are critical determinants of susceptibility to cells, tissues, or species, and remain to be elucidated for SADS-CoV. Type II transmembrane serine proteases (TTSPs) family is involved in many coronavirus infections and has trypsin-like catalytic activity. Here we examine all 18 members of the TTSPs family through CRISPR-based activation of endogenous protein expression in cells, and find that, in addition to TMPRSS2 and TMPRSS4, TMPRSS13 significantly facilitates SADS-CoV infection. This is confirmed by ectopic expression of TMPRSS13, and specific to trypsin-dependent SADS-CoV. Infection with pseudovirus bearing SADS-CoV spike protein indicates that TMPRSS13 acts at the entry step and is sensitive to serine protease inhibitor Camostat. Moreover, both human and pig TMPRSS13 are able to enhance the cell-cell membrane fusion and cleavage of spike protein. Overall, we demonstrate that TMPRSS13 is another host serine protease promoting the membrane-fusion entry of SADS-CoV, which may expand its host tropism by using diverse TTSPs.


Subject(s)
Membrane Proteins , Serine Endopeptidases , Virus Internalization , Animals , Serine Endopeptidases/metabolism , Serine Endopeptidases/genetics , Swine , Humans , Membrane Proteins/metabolism , Membrane Proteins/genetics , Alphacoronavirus/genetics , Alphacoronavirus/physiology , Coronavirus Infections/virology , Coronavirus Infections/metabolism , Gabexate/analogs & derivatives , Gabexate/pharmacology , Spike Glycoprotein, Coronavirus/metabolism , Spike Glycoprotein, Coronavirus/genetics , HEK293 Cells , Cell Line , Chlorocebus aethiops , Swine Diseases/virology , Esters , Guanidines
6.
Biochim Biophys Acta Mol Cell Res ; 1871(5): 119742, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38702017

ABSTRACT

Peroxisomes are ubiquitous cell organelles involved in various metabolic pathways. In order to properly function, several cofactors, substrates and products of peroxisomal enzymes need to pass the organellar membrane. So far only a few transporter proteins have been identified. We analysed peroxisomal membrane fractions purified from the yeast Hansenula polymorpha by untargeted label-free quantitation mass spectrometry. As expected, several known peroxisome-associated proteins were enriched in the peroxisomal membrane fraction. In addition, several other proteins were enriched, including mitochondrial transport proteins. Localization studies revealed that one of them, the mitochondrial phosphate carrier Mir1, has a dual localization on mitochondria and peroxisomes. To better understand the molecular mechanisms of dual sorting, we localized Mir1 in cells lacking Pex3 or Pex19, two peroxins that play a role in targeting of peroxisomal membrane proteins. In these cells Mir1 only localized to mitochondria, indicating that Pex3 and Pex19 are required to sort Mir1 to peroxisomes. Analysis of the localization of truncated versions of Mir1 in wild-type H. polymorpha cells revealed that most of them localized to mitochondria, but only one, consisting of the transmembrane domains 3-6, was peroxisomal. Peroxisomal localization of this construct was lost in a MIR1 deletion strain, indicating that full-length Mir1 was required for the localization of the truncated protein to peroxisomes. Our data suggest that only full-length Mir1 sorts to peroxisomes, while Mir1 contains multiple regions with mitochondrial sorting information. Data are available via ProteomeXchange with identifier PXD050324.


Subject(s)
Fungal Proteins , Mitochondria , Peroxisomes , Pichia , Peroxisomes/metabolism , Mitochondria/metabolism , Mitochondria/genetics , Fungal Proteins/metabolism , Fungal Proteins/genetics , Pichia/metabolism , Pichia/genetics , Membrane Proteins/metabolism , Membrane Proteins/genetics , Peroxins/metabolism , Peroxins/genetics , Mitochondrial Proteins/metabolism , Mitochondrial Proteins/genetics , Protein Transport
7.
Nan Fang Yi Ke Da Xue Xue Bao ; 44(4): 636-643, 2024 Apr 20.
Article in Chinese | MEDLINE | ID: mdl-38708495

ABSTRACT

OBJECTIVE: To investigate the effect of Jisuikang formula-medicated serum for promoting spinal cord injury (SCI) repair in rats and explore the possible mechanism. METHODS: Thirty adult SD rats were randomized into sham-operated group, SCI (induced using a modified Allen method) model group, and Jisuikang formula-medicated serum treatment group. After the operations, the rats were treated with normal saline or Jisuikang by gavage on a daily basis for 14 days, and the changes in hindlimb motor function of the rats was assessed with Basso-Beattie-Bresnahan (BBB) scores and inclined-plate test. The injured spinal cord tissues were sampled from the SCI rat models for single-cell RNA sequencing, and bioinformatics analysis was performed to identify the target genes of Jisuikang, spinal cord injury and glycolysis. In the cell experiment, cultured astrocytes from neonatal SD rat cortex were treated with SOX2 alone or in combination with Jisuikang-medicated serum for 21 days, and the protein expressions of PKM2, p-PKM2 and YAP and colocalization of PKM2 and YAP in the cells were analyzed with Western blotting and immunofluorescence staining, respectively. RESULTS: The SCI rats with Jisuikang treatment showed significantly improved BBB scores and performance in inclined-plate test. At the injury site, high PKM2 expression was detected in various cell types. Bioinformatic analysis identified the HIPPO-YAP signaling pathway as the target pathway of Jisuikang. In cultured astrocytes, SOX2 combined with the mediated serum, as compared with SOX2 alone, significantly increased PKM2, p-PKM2 and YAP expressions and entry of phosphorylated PKM2 into the nucleus, and promoted PKM2 and YAP co-localization in the cells. CONCLUSION: Jisuikang formula accelerates SCI repair in rats possibly by promoting aerobic glycolysis of the astrocytes via activating the PKM2/YAP axis to induce reprogramming of the astrocytes into neurons.


Subject(s)
Astrocytes , Pyruvate Kinase , Signal Transduction , Spinal Cord Injuries , YAP-Signaling Proteins , Animals , Rats , Astrocytes/metabolism , Astrocytes/drug effects , Carrier Proteins/metabolism , Disease Models, Animal , Drugs, Chinese Herbal/pharmacology , Membrane Proteins/metabolism , Rats, Sprague-Dawley , Signal Transduction/drug effects , Spinal Cord Injuries/metabolism , Spinal Cord Injuries/drug therapy , Thyroid Hormone-Binding Proteins , Thyroid Hormones/metabolism
8.
Nat Commun ; 15(1): 4479, 2024 May 27.
Article in English | MEDLINE | ID: mdl-38802343

ABSTRACT

Deposition of amyloid-ß (Aß) peptides in the brain is a hallmark of Alzheimer's disease. Aßs are generated through sequential proteolysis of the amyloid precursor protein by the γ-secretase complexes (GSECs). Aß peptide length, modulated by the Presenilin (PSEN) and APH-1 subunits of GSEC, is critical for Alzheimer's pathogenesis. Despite high relevance, mechanistic understanding of the proteolysis of Aß, and its modulation by APH-1, remain incomplete. Here, we report cryo-EM structures of human GSEC (PSEN1/APH-1B) reconstituted into lipid nanodiscs in apo form and in complex with the intermediate Aß46 substrate without cross-linking. We find that three non-conserved and structurally divergent APH-1 regions establish contacts with PSEN1, and that substrate-binding induces concerted rearrangements in one of the identified PSEN1/APH-1 interfaces, providing structural basis for APH-1 allosteric-like effects. In addition, the GSEC-Aß46 structure reveals an interaction between Aß46 and loop 1PSEN1, and identifies three other H-bonding interactions that, according to functional validation, are required for substrate recognition and efficient sequential catalysis.


Subject(s)
Amyloid Precursor Protein Secretases , Amyloid beta-Peptides , Cryoelectron Microscopy , Membrane Proteins , Presenilin-1 , Humans , Amyloid Precursor Protein Secretases/metabolism , Amyloid Precursor Protein Secretases/chemistry , Presenilin-1/metabolism , Presenilin-1/chemistry , Presenilin-1/genetics , Amyloid beta-Peptides/metabolism , Amyloid beta-Peptides/chemistry , Membrane Proteins/metabolism , Membrane Proteins/chemistry , Endopeptidases/metabolism , Endopeptidases/chemistry , Amyloid beta-Protein Precursor/metabolism , Amyloid beta-Protein Precursor/chemistry , Protein Binding , Protein Isoforms/metabolism , Protein Isoforms/chemistry , Alzheimer Disease/metabolism , Peptide Fragments/metabolism , Peptide Fragments/chemistry , Peptide Hydrolases/metabolism , Peptide Hydrolases/chemistry , Models, Molecular , Proteolysis
9.
Cell Death Dis ; 15(5): 380, 2024 May 30.
Article in English | MEDLINE | ID: mdl-38816370

ABSTRACT

Senescent cell clearance is emerging as a promising strategy for treating age-related diseases. Senolytics are small molecules that promote the clearance of senescent cells; however, senolytics are uncommon and their underlying mechanisms remain largely unknown. Here, we investigated whether genomic instability is a potential target for senolytic. We screened small-molecule kinase inhibitors involved in the DNA damage response (DDR) in Zmpste24-/- mouse embryonic fibroblasts, a progeroid model characterized with impaired DDR and DNA repair. 4,5,6,7-tetrabromo-2-azabenzamidazole (TBB), which specifically inhibits casein kinase 2 (CK2), was selected and discovered to preferentially trigger apoptosis in Zmpste24-/- cells. Mechanistically, inhibition of CK2 abolished the phosphorylation of heterochromatin protein 1α (HP1α), which retarded the dynamic HP1α dissociation from repressive histone mark H3K9me3 and its relocalization with γH2AX to DNA damage sites, suggesting that disrupting heterochromatin remodeling in the initiation of DDR accelerates apoptosis in senescent cells. Furthermore, feeding Zmpste24-deficient mice with TBB alleviated progeroid features and extended their lifespan. Our study identified TBB as a new class senolytic compound that can reduce age-related symptoms and prolong lifespan in progeroid mice.


Subject(s)
Casein Kinase II , Cellular Senescence , DNA Damage , Longevity , Membrane Proteins , Metalloendopeptidases , Animals , Cellular Senescence/drug effects , Casein Kinase II/metabolism , Casein Kinase II/antagonists & inhibitors , Casein Kinase II/genetics , Mice , Longevity/drug effects , Membrane Proteins/metabolism , Membrane Proteins/genetics , DNA Damage/drug effects , Metalloendopeptidases/metabolism , Metalloendopeptidases/genetics , Metalloendopeptidases/deficiency , Apoptosis/drug effects , Chromobox Protein Homolog 5/metabolism , Histones/metabolism , Mice, Knockout , Fibroblasts/metabolism , Fibroblasts/drug effects , Chromosomal Proteins, Non-Histone/metabolism , Humans , Phosphorylation/drug effects
10.
Cell Death Dis ; 15(5): 379, 2024 May 30.
Article in English | MEDLINE | ID: mdl-38816421

ABSTRACT

CSMD1 (Cub and Sushi Multiple Domains 1) is a well-recognized regulator of the complement cascade, an important component of the innate immune response. CSMD1 is highly expressed in the central nervous system (CNS) where emergent functions of the complement pathway modulate neural development and synaptic activity. While a genetic risk factor for neuropsychiatric disorders, the role of CSMD1 in neurodevelopmental disorders is unclear. Through international variant sharing, we identified inherited biallelic CSMD1 variants in eight individuals from six families of diverse ancestry who present with global developmental delay, intellectual disability, microcephaly, and polymicrogyria. We modeled CSMD1 loss-of-function (LOF) pathogenesis in early-stage forebrain organoids differentiated from CSMD1 knockout human embryonic stem cells (hESCs). We show that CSMD1 is necessary for neuroepithelial cytoarchitecture and synchronous differentiation. In summary, we identified a critical role for CSMD1 in brain development and biallelic CSMD1 variants as the molecular basis of a previously undefined neurodevelopmental disorder.


Subject(s)
Intellectual Disability , Membrane Proteins , Humans , Intellectual Disability/genetics , Intellectual Disability/pathology , Membrane Proteins/genetics , Membrane Proteins/metabolism , Female , Male , Neurodevelopmental Disorders/genetics , Alleles , Malformations of Cortical Development/genetics , Malformations of Cortical Development/pathology , Child , Child, Preschool , Cell Differentiation/genetics , Tumor Suppressor Proteins
11.
Nat Commun ; 15(1): 4609, 2024 May 30.
Article in English | MEDLINE | ID: mdl-38816425

ABSTRACT

The protection of the replication fork structure under stress conditions is essential for genome maintenance and cancer prevention. A key signaling pathway for fork protection involves TRPV2-mediated Ca2+ release from the ER, which is triggered after the generation of cytosolic DNA and the activation of cGAS/STING. This results in CaMKK2/AMPK activation and subsequent Exo1 phosphorylation, which prevent aberrant fork processing, thereby ensuring genome stability. However, it remains poorly understood how the TRPV2 channel is activated by the presence of cytosolic DNA. Here, through a genome-wide CRISPR-based screen, we identify TRPM8 channel-associated factor 1 (TCAF1) as a key factor promoting TRPV2-mediated Ca2+ release under replication stress or other conditions that activate cGAS/STING. Mechanistically, TCAF1 assists Ca2+ release by facilitating the dissociation of STING from TRPV2, thereby relieving TRPV2 repression. Consistent with this function, TCAF1 is required for fork protection, chromosomal stability, and cell survival after replication stress.


Subject(s)
Calcium , Cytosol , DNA Replication , Membrane Proteins , TRPV Cation Channels , Humans , TRPV Cation Channels/metabolism , TRPV Cation Channels/genetics , Calcium/metabolism , Cytosol/metabolism , Membrane Proteins/metabolism , Membrane Proteins/genetics , HEK293 Cells , DNA/metabolism , HeLa Cells , Calcium-Calmodulin-Dependent Protein Kinase Kinase/metabolism , Calcium-Calmodulin-Dependent Protein Kinase Kinase/genetics , Phosphorylation , Genomic Instability , DNA Damage , Animals
12.
Front Immunol ; 15: 1385696, 2024.
Article in English | MEDLINE | ID: mdl-38770013

ABSTRACT

Background: Recent studies have demonstrated a strong association between acute kidney injury (AKI) and chronic kidney disease (CKD), while the unresolved inflammation is believed to be a driving force for this chronic transition process. As a transmembrane pattern recognition receptor, Mincle (macrophage-inducible C-type lectin, Clec4e) was identified to participate in the early immune response after AKI. However, the impact of Mincle on the chronic transition of AKI remains largely unclear. Methods: We performed single-cell RNA sequencing (scRNA-seq) with the unilateral ischemia-reperfusion (UIR) murine model of AKI at days 1, 3, 14 and 28 after injury. Potential effects and mechanism of Mincle on renal inflammation and fibrosis were further validated in vivo utilizing Mincle knockout mice. Results: The dynamic expression of Mincle in macrophages and neutrophils throughout the transition from AKI to CKD was observed. For both cell types, Mincle expression was significantly up-regulated on day 1 following AKI, with a second rise observed on day 14. Notably, we identified distinct subclusters of Minclehigh neutrophils and Minclehigh macrophages that exhibited time-dependent influx with dual peaks characterized with remarkable pro-inflammatory and pro-fibrotic functions. Moreover, we identified that Minclehigh neutrophils represented an "aged" mature neutrophil subset derived from the "fresh" mature neutrophil cluster in kidney. Additionally, we observed a synergistic mechanism whereby Mincle-expressing macrophages and neutrophils sustained renal inflammation by tumor necrosis factor (TNF) production. Mincle-deficient mice exhibited reduced renal injury and fibrosis following AKI. Conclusion: The present findings have unveiled combined persistence of Minclehigh neutrophils and macrophages during AKI-to-CKD transition, contributing to unresolved inflammation followed by fibrosis via TNF-α as a central pro-inflammatory cytokine. Targeting Mincle may offer a novel therapeutic strategy for preventing the transition from AKI to CKD.


Subject(s)
Acute Kidney Injury , Disease Models, Animal , Lectins, C-Type , Macrophages , Membrane Proteins , Mice, Knockout , Neutrophils , Renal Insufficiency, Chronic , Animals , Lectins, C-Type/metabolism , Lectins, C-Type/genetics , Acute Kidney Injury/etiology , Acute Kidney Injury/immunology , Acute Kidney Injury/metabolism , Macrophages/immunology , Macrophages/metabolism , Neutrophils/immunology , Neutrophils/metabolism , Mice , Renal Insufficiency, Chronic/immunology , Renal Insufficiency, Chronic/etiology , Renal Insufficiency, Chronic/metabolism , Renal Insufficiency, Chronic/pathology , Membrane Proteins/genetics , Membrane Proteins/metabolism , Male , Inflammation/immunology , Mice, Inbred C57BL , Reperfusion Injury/immunology , Reperfusion Injury/metabolism , Fibrosis , Disease Progression
13.
Front Immunol ; 15: 1399926, 2024.
Article in English | MEDLINE | ID: mdl-38817608

ABSTRACT

Immune checkpoint inhibitors (ICIs) represent a groundbreaking advance in the treatment of malignancies such as melanoma and non-small cell lung cancer, showcasing substantial therapeutic benefits. Nonetheless, the efficacy of ICIs is limited to a small subset of patients, primarily benefiting those with "hot" tumors characterized by significant immune infiltration. The challenge of converting "cold" tumors, which exhibit minimal immune activity, into "hot" tumors to enhance their responsiveness to ICIs is a critical and complex area of current research. Central to this endeavor is the activation of the cGAS-STING pathway, a pivotal nexus between innate and adaptive immunity. This pathway's activation promotes the production of type I interferon (IFN) and the recruitment of CD8+ T cells, thereby transforming the tumor microenvironment (TME) from "cold" to "hot". This review comprehensively explores the cGAS-STING pathway's role in reconditioning the TME, detailing the underlying mechanisms of innate and adaptive immunity and highlighting the contributions of various immune cells to tumor immunity. Furthermore, we delve into the latest clinical research on STING agonists and their potential in combination therapies, targeting this pathway. The discussion concludes with an examination of the challenges facing the advancement of promising STING agonists in clinical trials and the pressing issues within the cGAS-STING signaling pathway research.


Subject(s)
Immunotherapy , Membrane Proteins , Neoplasms , Nucleotidyltransferases , Signal Transduction , Tumor Microenvironment , Humans , Tumor Microenvironment/immunology , Nucleotidyltransferases/metabolism , Membrane Proteins/metabolism , Immunotherapy/methods , Neoplasms/immunology , Neoplasms/therapy , Neoplasms/metabolism , Animals , Immune Checkpoint Inhibitors/therapeutic use , Immune Checkpoint Inhibitors/pharmacology , Immunity, Innate , Adaptive Immunity
14.
Anticancer Res ; 44(6): 2577-2585, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38821598

ABSTRACT

BACKGROUND/AIM: Nuclear factor erythroid-derived 2-related factor-2 (NRF2) is a transcription factor that regulates stress response genes. It negatively regulates the immune system by acting as a transcriptional repressor of inflammatory genes or suppressing type I interferon (IFN) production pathways. NRF2 is often over-expressed in some tumors, including non-small cell lung cancer, and modulates these tumors via an immune-cold microenvironment. Thus, strategies to convert cold tumors into hot tumors are effective for cancer treatment. MATERIALS AND METHODS: NRF2 was knocked-down or over-expressed in human cancer cells (A549, HeLa, H1299, H1650) and mouse mammary adenocarcinoma TS/A cells. Cells were irradiated or transfected with poly(I:C), and changes in type I IFN levels were examined using quantitative real-time polymerase chain reaction and western blotting. Cytosolic DNA was assayed via PicoGreen staining and immune and cancer cells were co-cultured. RESULTS: Regulation of NRF2 expression altered type I IFN levels in the human lung cancer cell line A549 and several solid tumors. Down-regulation of NRF2 resulted in increased levels of cytosolic DNA and activated the cGAS-STING pathway. We confirmed that type I IFN was induced in NRF2-down-regulated tumor cells using ionizing radiation (IR). Furthermore, when dendritic cells and macrophages were co-cultured with IR-exposed NRF2 knockdown tumor cells, the immune cells produced more IFNB1 and CXCL10. CONCLUSION: The immunosuppressive tumor cell environment is improved by NRF2 down-regulation, and IR treatment may promote immune cell signaling activation.


Subject(s)
Interferon Type I , NF-E2-Related Factor 2 , Radiation, Ionizing , Signal Transduction , NF-E2-Related Factor 2/metabolism , NF-E2-Related Factor 2/genetics , Humans , Interferon Type I/metabolism , Animals , Mice , Cell Line, Tumor , A549 Cells , Lung Neoplasms/radiotherapy , Lung Neoplasms/immunology , Lung Neoplasms/pathology , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Tumor Microenvironment/immunology , Nucleotidyltransferases/genetics , Nucleotidyltransferases/metabolism , Macrophages/immunology , Macrophages/metabolism
15.
Life Sci ; 349: 122719, 2024 Jul 15.
Article in English | MEDLINE | ID: mdl-38759866

ABSTRACT

In the vast landscape of human health, head and neck cancer (HNC) poses a significant health burden globally, necessitating the exploration of novel diagnostics and therapeutics. Metabolic alterations occurring within tumor microenvironment are crucial to understand the foundational cause of HNC. Post-translational modifications (PTMs) have recently emerged as a silent foe exerting a significantly heightened influence on various aspects of the biological processes associated with the onset and advancement of cancer, particularly in the context of HNC. There are numerous targets involved in HNC but recently, the enzyme pyruvate kinase M2 (PKM2) has come out as a hot target due to its involvement in glycolysis resulting in metabolic reprogramming of cancer cells. Various PTMs have been reported to affect the structure and function of PKM2 by modulating its activity. This review aims to investigate the impact of PTMs on the interaction between PKM2 and several signaling pathways and transcription factors in the context of HNC. These interactions possess significant ramification for cellular proliferation, apoptosis, angiogenesis and metastasis. This review primarily explores the role of PTMs influencing PKM2 and its involvement in tumor development. While acknowledging the significance of PKM2 interactions with other tumor regulators, the emphasis lies on dissecting PTM-related mechanisms rather than solely scrutinizing individual regulators. It lays the framework for the development of more sophisticated diagnostic tools and uncovers exciting possibilities for precision medicine essential for effectively addressing the complexity of this malignancy in a precise and focused manner.


Subject(s)
Carrier Proteins , Head and Neck Neoplasms , Membrane Proteins , Protein Processing, Post-Translational , Thyroid Hormone-Binding Proteins , Thyroid Hormones , Humans , Head and Neck Neoplasms/metabolism , Head and Neck Neoplasms/pathology , Thyroid Hormones/metabolism , Membrane Proteins/metabolism , Carrier Proteins/metabolism , Tumor Microenvironment , Animals , Signal Transduction
16.
Synapse ; 78(3): e22293, 2024 May.
Article in English | MEDLINE | ID: mdl-38779935

ABSTRACT

The differentiation of bone marrow stromal cells (BMSCs) into Schwann-like cells (SCLCs) has the potential to promote the structural and functional restoration of injured axons. However, the optimal induction protocol and its underlying mechanisms remain unclear. This study aimed to compare the effectiveness of different induction protocols in promoting the differentiation of rat BMSCs into SCLCs and to explore their potential mechanisms. BMSCs were induced using two distinct methods: a composite factor induction approach (Protocol-1) and a conditioned culture medium induction approach (Protocol-2). The expression of Schwann cells (SCs) marker proteins and neurotrophic factors (NTFs) in the differentiated cells was assessed. Cell proliferation and apoptosis were also measured. During induction, changes in miR-21 and Sprouty RTK signaling antagonist 2 (SPRY2) mRNA were analyzed. Following the transfection of BMSCs with miR-21 agomir or miR-21 antagomir, induction was carried out using both protocols, and the expression of SPRY2, ERK1/2, and SCs marker proteins was examined. The results revealed that NTFs expression was higher in Protocol-1, whereas SCs marker proteins expression did not significantly differ between the two groups. Compared to Protocol-1, Protocol-2 exhibited enhanced cell proliferation and fewer apoptotic and necrotic cells. Both protocols showed a negative correlation between miR-21 and SPRY2 expression throughout the induction stages. After induction, the miR-21 agomir group exhibited reduced SPRY2 expression, increased ERK1/2 expression, and significantly elevated expression of SCs marker proteins. This study demonstrates that Protocol-1 yields higher NTFs expression, whereas Protocol-2 results in stronger SCLCs proliferation. Upregulating miR-21 suppresses SPRY2 expression, activates the ERK1/2 signaling pathway, and promotes BMSC differentiation into SCLCs.


Subject(s)
Cell Differentiation , Cell Proliferation , Membrane Proteins , Mesenchymal Stem Cells , MicroRNAs , Rats, Sprague-Dawley , Schwann Cells , Animals , Schwann Cells/metabolism , Schwann Cells/cytology , MicroRNAs/metabolism , MicroRNAs/genetics , Cell Differentiation/physiology , Rats , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/cytology , Membrane Proteins/metabolism , Membrane Proteins/genetics , Cell Proliferation/physiology , Cells, Cultured , Intracellular Signaling Peptides and Proteins/metabolism , Intracellular Signaling Peptides and Proteins/genetics , Apoptosis/physiology , Nerve Growth Factors/metabolism , Nerve Growth Factors/genetics , Culture Media, Conditioned/pharmacology , Nerve Tissue Proteins
17.
Life Sci Alliance ; 7(7)2024 Jul.
Article in English | MEDLINE | ID: mdl-38719747

ABSTRACT

The differential expression of plasma membrane proteins is integrally analyzed for their diagnosis, prognosis, and therapeutic applications in diverse clinical manifestations. Necessarily, distinct membrane protein enrichment methods and mass spectrometry platforms are employed for their global and relative quantitation. First of its kind to explore, we compiled membrane-associated proteomes in human and mouse systems into a database named, Resource of Experimental Membrane-Enriched Mass spectrometry-derived Proteome (REMEMProt). It currently hosts 14,626 proteins (9,507 proteins in Homo sapiens; 5,119 proteins in Mus musculus) with information on their membrane-protein enrichment methods, experimental/physiological context of detection in cells or tissues, transmembrane domain analysis, and their current attribution as biomarkers. Based on these annotations and the transmembrane domain analysis in proteins or their binary/complex protein-protein interactors, REMEMProt facilitates the assessment of the plasma membrane localization potential of proteins through batch query. A cross-study enrichment analysis platform is enabled in REMEMProt for comparative analysis of proteomes using novel/modified membrane enrichment methods and evaluation of methods for targeted enrichment of membrane proteins. REMEMProt data are made freely accessible to explore and download at https://rememprot.ciods.in/.


Subject(s)
Biomarkers , Databases, Protein , Membrane Proteins , Proteome , Proteomics , Humans , Proteome/metabolism , Membrane Proteins/metabolism , Biomarkers/metabolism , Animals , Mice , Proteomics/methods , Cell Membrane/metabolism , Mass Spectrometry/methods
18.
Proc Natl Acad Sci U S A ; 121(22): e2402764121, 2024 May 28.
Article in English | MEDLINE | ID: mdl-38771879

ABSTRACT

Staphylococcus aureus (S. aureus) can evade antibiotics and host immune defenses by persisting within infected cells. Here, we demonstrate that in infected host cells, S. aureus type VII secretion system (T7SS) extracellular protein B (EsxB) interacts with the stimulator of interferon genes (STING) protein and suppresses the inflammatory defense mechanism of macrophages during early infection. The binding of EsxB with STING disrupts the K48-linked ubiquitination of EsxB at lysine 33, thereby preventing EsxB degradation. Furthermore, EsxB-STING binding appears to interrupt the interaction of 2 vital regulatory proteins with STING: aspartate-histidine-histidine-cysteine domain-containing protein 3 (DHHC3) and TNF receptor-associated factor 6. This persistent dual suppression of STING interactions deregulates intracellular proinflammatory pathways in macrophages, inhibiting STING's palmitoylation at cysteine 91 and its K63-linked ubiquitination at lysine 83. These findings uncover an immune-evasion mechanism by S. aureus T7SS during intracellular macrophage infection, which has implications for developing effective immunomodulators to combat S. aureus infections.


Subject(s)
Bacterial Proteins , Macrophages , Membrane Proteins , Staphylococcal Infections , Staphylococcus aureus , Type VII Secretion Systems , Ubiquitination , Staphylococcus aureus/immunology , Membrane Proteins/metabolism , Membrane Proteins/immunology , Humans , Bacterial Proteins/metabolism , Bacterial Proteins/immunology , Macrophages/immunology , Macrophages/metabolism , Macrophages/microbiology , Animals , Staphylococcal Infections/immunology , Staphylococcal Infections/microbiology , Staphylococcal Infections/metabolism , Type VII Secretion Systems/metabolism , Type VII Secretion Systems/immunology , Type VII Secretion Systems/genetics , Mice , Immune Evasion , Host-Pathogen Interactions/immunology
19.
Nat Commun ; 15(1): 3984, 2024 May 11.
Article in English | MEDLINE | ID: mdl-38734736

ABSTRACT

Greenbeard genetic elements encode rare perceptible signals, signal recognition ability, and altruism towards others that display the same signal. Putative greenbeards have been described in various organisms but direct evidence for all the properties in one system is scarce. The tgrB1-tgrC1 allorecognition system of Dictyostelium discoideum encodes two polymorphic membrane proteins which protect cells from chimerism-associated perils. During development, TgrC1 functions as a ligand-signal and TgrB1 as its receptor, but evidence for altruism has been indirect. Here, we show that mixing wild-type and activated tgrB1 cells increases wild-type spore production and relegates the mutants to the altruistic stalk, whereas mixing wild-type and tgrB1-null cells increases mutant spore production and wild-type stalk production. The tgrB1-null cells cheat only on partners that carry the same tgrC1-allotype. Therefore, TgrB1 activation confers altruism whereas TgrB1 inactivation causes allotype-specific cheating, supporting the greenbeard concept and providing insight into the relationship between allorecognition, altruism, and exploitation.


Subject(s)
Dictyostelium , Protozoan Proteins , Dictyostelium/genetics , Dictyostelium/metabolism , Dictyostelium/physiology , Protozoan Proteins/genetics , Protozoan Proteins/metabolism , Spores, Protozoan/genetics , Spores, Protozoan/metabolism , Signal Transduction , Mutation , Altruism , Membrane Proteins/metabolism , Membrane Proteins/genetics , Chemotaxis/genetics
20.
ACS Nano ; 18(20): 12830-12844, 2024 May 21.
Article in English | MEDLINE | ID: mdl-38709246

ABSTRACT

The immunosuppressive microenvironment of cervical cancer significantly hampers the effectiveness of immunotherapy. Herein, PEGylated manganese-doped calcium sulfide nanoparticles (MCSP) were developed to effectively enhance the antitumor immune response of the cervical cancer through gas-amplified metalloimmunotherapy with dual activation of pyroptosis and STING pathway. The bioactive MCSP exhibited the ability to rapidly release Ca2+, Mn2+, and H2S in response to the tumor microenvironment. H2S disrupted the calcium buffer system of cancer cells by interfering with the oxidative phosphorylation pathway, leading to calcium overload-triggered pyroptosis. On the other hand, H2S-mediated mitochondrial dysfunction further promoted the release of mitochondrial DNA (mtDNA), enhancing the activation effect of Mn2+ on the cGAS-STING signaling axis and thereby activating immunosuppressed dendritic cells. The released H2S acted as an important synergist between Mn2+ and Ca2+ by modulating dual signaling mechanisms to bridge innate and adaptive immune responses. The combination of MCSP NPs and PD-1 immunotherapy achieved synergistic antitumor effects and effectively inhibited tumor growth. This study reveals the potential collaboration between H2S gas therapy and metalloimmunotherapy and provides an idea for the design of nanoimmunomodulators for rational regulation of the immunosuppressive tumor microenvironment.


Subject(s)
Immunotherapy , Membrane Proteins , Pyroptosis , Tumor Microenvironment , Uterine Cervical Neoplasms , Tumor Microenvironment/drug effects , Tumor Microenvironment/immunology , Uterine Cervical Neoplasms/immunology , Uterine Cervical Neoplasms/drug therapy , Uterine Cervical Neoplasms/pathology , Uterine Cervical Neoplasms/metabolism , Uterine Cervical Neoplasms/therapy , Female , Humans , Mice , Animals , Pyroptosis/drug effects , Membrane Proteins/metabolism , Manganese/chemistry , Manganese/pharmacology , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Nanoparticles/chemistry , Signal Transduction/drug effects , Cell Proliferation/drug effects , Calcium/metabolism , Mice, Inbred BALB C , Drug Screening Assays, Antitumor
SELECTION OF CITATIONS
SEARCH DETAIL
...