Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 64.771
Filter
1.
Adipocyte ; 13(1): 2339418, 2024 Dec.
Article in English | MEDLINE | ID: mdl-38706095

ABSTRACT

A Disintegrin And Metalloproteinase domain-containing protein 10 (ADAM10), is involved in several metabolic and inflammatory pathways. We speculated that ADAM10 plays a modulatory role in adipose tissue inflammation and metabolism. To this end, we studied adipose tissue-specific ADAM10 knock-out mice (aKO). While young, regular chow diet-fed aKO mice showed increased insulin sensitivity, following prolonged (33 weeks) high-fat diet (HFD) exposure, aKO mice developed obesity and insulin resistance. Compared to controls, aKO mice showed less inflammatory adipokine profile despite the significant increase in adiposity. In brown adipose tissue, aKO mice on HFD had changes in CD8+ T cell populations indicating a lesser inflammatory pattern. Following HFD, both aKO and control littermates demonstrated decreased adipose tissue pro-inflammatory macrophages, and increased anti-inflammatory accumulation, without differences between the genotypes. Collectively, our observations indicate that selective deletion of ADAM10 in adipocytes results in a mitigated inflammatory response, leading to increased insulin sensitivity in young mice fed with regular diet. This state of insulin sensitivity, following prolonged HFD, facilitates energy storage resulting in increased fat accumulation which ultimately leads to the development of a phenotype of obesity and insulin resistance. In conclusion, the data indicate that ADAM10 has a modulatory effect of inflammation and whole-body energy metabolism.


Subject(s)
ADAM10 Protein , Adipose Tissue , Diet, High-Fat , Mice, Knockout , Animals , Male , Mice , ADAM10 Protein/metabolism , ADAM10 Protein/genetics , Adipocytes/metabolism , Adipose Tissue/metabolism , Amyloid Precursor Protein Secretases/metabolism , Diet, High-Fat/adverse effects , Inflammation/metabolism , Insulin Resistance , Membrane Proteins/metabolism , Membrane Proteins/genetics , Obesity/metabolism , Obesity/etiology , Phenotype
3.
Int J Biol Sci ; 20(7): 2370-2387, 2024.
Article in English | MEDLINE | ID: mdl-38725841

ABSTRACT

The pathogenesis of Intervertebral Disc Degeneration (IDD) is complex and multifactorial, with cellular senescence of nucleus pulposus (NP) cells and inflammation playing major roles in the progression of IDD. The stimulator of interferon genes (STING) axis is a key mediator of inflammation during infection, cellular stress, and tissue damage. Here, we present a progressive increase in STING in senescent NP cells with the degradation disorder. The STING degradation function in normal NP cells can prevent IDD. However, the dysfunction of STING degradation through autophagy causes the accumulation and high expression of STING in senescent NP cells as well as inflammation continuous activation together significantly promotes IDD. In senescent NP cells and intervertebral discs (IVDs), we found that STING autophagy degradation was significantly lower than that of normal NP cells and IVDs when STING was activated by 2'3'-cGAMP. Also, the above phenomenon was found in STINGgt/gt, cGAS-/- mice with models of age-induced, lumbar instability-induced IDD as well as found in the rat caudal IVD puncture models. Taken together, we suggested that the promotion of STING autophagy degradation in senescent NP Cells demonstrated a potential therapeutic modality for the treatment of IDD.


Subject(s)
Autophagy , Cellular Senescence , Intervertebral Disc Degeneration , Membrane Proteins , Nucleus Pulposus , Intervertebral Disc Degeneration/metabolism , Intervertebral Disc Degeneration/pathology , Nucleus Pulposus/metabolism , Animals , Autophagy/physiology , Membrane Proteins/metabolism , Membrane Proteins/genetics , Mice , Cellular Senescence/physiology , Rats , Male , Rats, Sprague-Dawley , Humans , Mice, Inbred C57BL
4.
PLoS One ; 19(5): e0301082, 2024.
Article in English | MEDLINE | ID: mdl-38722977

ABSTRACT

Branching morphogenesis is a complex process shared by many organs including the lungs, kidney, prostate, as well as several exocrine organs including the salivary, mammary and lacrimal glands. This critical developmental program ensures the expansion of an organ's surface area thereby maximizing processes of cellular secretion or absorption. It is guided by reciprocal signaling from the epithelial and mesenchymal cells. While signaling pathways driving salivary gland branching morphogenesis have been relatively well-studied, our understanding of the underlying transcriptional regulatory mechanisms directing this program, is limited. Here, we performed in vivo and ex vivo studies of the embryonic mouse submandibular gland to determine the function of the transcription factor ΔNp63, in directing branching morphogenesis. Our studies show that loss of ΔNp63 results in alterations in the differentiation program of the ductal cells which is accompanied by a dramatic reduction in branching morphogenesis that is mediated by dysregulation of WNT signaling. We show that ΔNp63 modulates WNT signaling to promote branching morphogenesis by directly regulating Sfrp1 expression. Collectively, our findings have revealed a novel role for ΔNp63 in the regulation of this critical process and offers a better understanding of the transcriptional networks involved in branching morphogenesis.


Subject(s)
Gene Expression Regulation, Developmental , Membrane Proteins , Morphogenesis , Animals , Mice , Morphogenesis/genetics , Membrane Proteins/metabolism , Membrane Proteins/genetics , Intercellular Signaling Peptides and Proteins/metabolism , Intercellular Signaling Peptides and Proteins/genetics , Salivary Glands/metabolism , Salivary Glands/embryology , Wnt Signaling Pathway , Submandibular Gland/metabolism , Submandibular Gland/embryology , Trans-Activators/metabolism , Trans-Activators/genetics , Cell Differentiation
5.
Viral Immunol ; 37(4): 186-193, 2024 05.
Article in English | MEDLINE | ID: mdl-38717821

ABSTRACT

Coronavirus disease 2019 (COVID-19) represented an international health risk. Variants of the interferon-induced transmembrane protein-3 (IFITM3) gene can increase the risk of developing severe viral infections. This cross-sectional study investigated the association between IFITM3 rs12252A>G single nucleotide polymorphism (SNP) and COVID-19 severity and mortality in 100 Egyptian patients. All participants were subjected to serum interleukin-6 (IL-6) determination by ELISA and IFITM3 rs12252 genotyping by real-time polymerase chain reaction. Of all participants, 85.0% had the IFITM3 rs12252 homozygous AA genotype, whereas 15.0% had the heterozygous AG genotype. None of our participants had the homozygous GG genotype. The IFITM3 rs12252A allele was found in 92.5% and the G allele in only 7.5%. There was no significant association (p > 0.05) between the IFITM3 rs12252 SNP and COVID-19 severity, intensive care unit (ICU) admission, or IL-6 serum levels. The heterozygous AG genotype frequency showed a significant increase among participants who died (32.0%) compared with those who had been cured (9.3%). The mutant G allele was associated with patients' death. Its frequency among cured participants was 8.5%, whereas in those who died was 24.2% (p = 0.024) with 3.429 odds ratio [95% confidence interval: 1.1-10.4]. In conclusion, this study revealed a significant association between the G allele variant of IFITM3 rs12252 and COVID-19 mortality. However, results were unable to establish a significant link between rs12252 polymorphism, disease severity, ICU admission, or serum IL-6 levels.


Subject(s)
COVID-19 , Genotype , Interleukin-6 , Membrane Proteins , Polymorphism, Single Nucleotide , RNA-Binding Proteins , SARS-CoV-2 , Humans , COVID-19/mortality , COVID-19/genetics , Female , Male , Egypt , Middle Aged , Membrane Proteins/genetics , Adult , Interleukin-6/blood , Interleukin-6/genetics , Cross-Sectional Studies , SARS-CoV-2/genetics , RNA-Binding Proteins/genetics , Genetic Predisposition to Disease , Alleles , Severity of Illness Index , Gene Frequency , Aged
6.
Mol Cancer ; 23(1): 94, 2024 May 08.
Article in English | MEDLINE | ID: mdl-38720298

ABSTRACT

BACKGROUND: The hypoxic tumor microenvironment is a key factor that promotes metabolic reprogramming and vascular mimicry (VM) in ovarian cancer (OC) patients. ESM1, a secreted protein, plays an important role in promoting proliferation and angiogenesis in OC. However, the role of ESM1 in metabolic reprogramming and VM in the hypoxic microenvironment in OC patients has not been determined. METHODS: Liquid chromatography coupled with tandem MS was used to analyze CAOV3 and OV90 cells. Interactions between ESM1, PKM2, UBA2, and SUMO1 were detected by GST pull-down, Co-IP, and molecular docking. The effects of the ESM1-PKM2 axis on cell glucose metabolism were analyzed based on an ECAR experiment. The biological effects of the signaling axis on OC cells were detected by tubule formation, transwell assay, RT‒PCR, Western blot, immunofluorescence, and in vivo xenograft tumor experiments. RESULTS: Our findings demonstrated that hypoxia induces the upregulation of ESM1 expression through the transcription of HIF-1α. ESM1 serves as a crucial mediator of the interaction between PKM2 and UBA2, facilitating the SUMOylation of PKM2 and the subsequent formation of PKM2 dimers. This process promotes the Warburg effect and facilitates the nuclear translocation of PKM2, ultimately leading to the phosphorylation of STAT3. These molecular events contribute to the promotion of ovarian cancer glycolysis and vasculogenic mimicry. Furthermore, our study revealed that Shikonin effectively inhibits the molecular interaction between ESM1 and PKM2, consequently preventing the formation of PKM2 dimers and thereby inhibiting ovarian cancer glycolysis, fatty acid synthesis and vasculogenic mimicry. CONCLUSION: Our findings demonstrated that hypoxia increases ESM1 expression through the transcriptional regulation of HIF-1α to induce dimerization via PKM2 SUMOylation, which promotes the OC Warburg effect and VM.


Subject(s)
Carrier Proteins , Fatty Acids , Membrane Proteins , Neoplasm Proteins , Ovarian Neoplasms , Thyroid Hormone-Binding Proteins , Thyroid Hormones , Tumor Microenvironment , Female , Humans , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , Ovarian Neoplasms/genetics , Animals , Thyroid Hormones/metabolism , Mice , Membrane Proteins/metabolism , Membrane Proteins/genetics , Cell Line, Tumor , Fatty Acids/metabolism , Neoplasm Proteins/metabolism , Neoplasm Proteins/genetics , Carrier Proteins/metabolism , Carrier Proteins/genetics , Warburg Effect, Oncologic , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Gene Expression Regulation, Neoplastic , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/pathology , Xenograft Model Antitumor Assays , Cell Proliferation , Proteoglycans
7.
Nat Commun ; 15(1): 3933, 2024 May 10.
Article in English | MEDLINE | ID: mdl-38730243

ABSTRACT

As a strategy to improve the therapeutic success of chimeric antigen receptor T cells (CART) directed against solid tumors, we here test the combinatorial use of CART and IMSA101, a newly developed stimulator of interferon genes (STING) agonist. In two syngeneic tumor models, improved overall survival is observed when mice are treated with intratumorally administered IMSA101 in addition to intravenous CART infusion. Transcriptomic analyses of CART isolated from tumors show elevated T cell activation, as well as upregulated cytokine pathway signatures, in particular IL-18, in the combination treatment group. Also, higher levels of IL-18 in serum and tumor are detected with IMSA101 treatment. Consistent with this, the use of IL-18 receptor negative CART impair anti-tumor responses in mice receiving combination treatment. In summary, we find that IMSA101 enhances CART function which is facilitated through STING agonist-induced IL-18 secretion.


Subject(s)
Interleukin-18 , Membrane Proteins , Receptors, Chimeric Antigen , Animals , Interleukin-18/metabolism , Membrane Proteins/agonists , Membrane Proteins/metabolism , Membrane Proteins/genetics , Mice , Receptors, Chimeric Antigen/metabolism , Receptors, Chimeric Antigen/immunology , Humans , Cell Line, Tumor , Mice, Inbred C57BL , T-Lymphocytes/immunology , T-Lymphocytes/drug effects , T-Lymphocytes/metabolism , Lymphocyte Activation/drug effects , Immunotherapy, Adoptive/methods , Female , Neoplasms/immunology , Neoplasms/therapy , Neoplasms/drug therapy
8.
Cell Mol Life Sci ; 81(1): 209, 2024 May 06.
Article in English | MEDLINE | ID: mdl-38710967

ABSTRACT

As an integral lysosomal transmembrane protein, transmembrane protein 106B (TMEM106B) regulates several aspects of lysosomal function and is associated with neurodegenerative diseases. The TMEM106B gene mutations lead to lysosomal dysfunction and accelerate the pathological progression of Neurodegenerative diseases. Yet, the precise mechanism of TMEM106B in Neurodegenerative diseases remains unclear. Recently, different research teams discovered that TMEM106B is an amyloid protein and the C-terminal domain of TMEM106B forms amyloid fibrils in various Neurodegenerative diseases and normally elderly individuals. In this review, we discussed the physiological functions of TMEM106B. We also included TMEM106B gene mutations that cause neurodegenerative diseases. Finally, we summarized the identification and cryo-electronic microscopic structure of TMEM106B fibrils, and discussed the promising therapeutic strategies aimed at TMEM106B fibrils and the future directions for TMEM106B research in neurodegenerative diseases.


Subject(s)
Membrane Proteins , Mutation , Nerve Tissue Proteins , Neurodegenerative Diseases , Humans , Membrane Proteins/metabolism , Membrane Proteins/genetics , Membrane Proteins/chemistry , Neurodegenerative Diseases/metabolism , Neurodegenerative Diseases/genetics , Neurodegenerative Diseases/pathology , Nerve Tissue Proteins/metabolism , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/chemistry , Animals , Lysosomes/metabolism , Lysosomes/genetics , Amyloid/metabolism , Amyloid/genetics , Amyloid/chemistry
9.
Pediatr Allergy Immunol ; 35(5): e14136, 2024 May.
Article in English | MEDLINE | ID: mdl-38747707

ABSTRACT

BACKGROUND: Familial hemophagocytic lymphohistiocytosis type 3 (FHL3) is caused by UNC13D variants. The clinical manifestations of FHL3 are highly diverse and complex. Some patients exhibit atypical or incomplete phenotypes, making accurate diagnosis difficult. Our study aimed to broaden the understanding of the atypical FHL3 clinical spectrum. METHODS: In our study, we analyzed in detail the clinical features of four Chinese patients with UNC13D variants. Additionally, we conducted a comprehensive review of the existing literature on previously reported atypical manifestations and summarized the findings. RESULTS: Two of our patients presented with muscle involvement, while the other two had hematological involvement; none of them met the diagnostic criteria for hemophagocytic lymphohistiocytosis (HLH). However, protein expression and functional analysis ultimately confirmed diagnostic criteria for FHL3 in all patients. From the literature we reviewed, many atypical FHL3 patients had neurological involvement, especially isolated neurological manifestations. At the same time, arthritis and hypogammaglobulinemia were also prone to occur. CONCLUSION: Our study highlights that the expression of the Munc13-4 protein may not fully indicate the pathogenicity of UNC13D variants, whereas CD107a analysis could be more sensitive for disease diagnosis. These findings contribute to a broader understanding of the FHL3 clinical spectrum and may offer new insights into the underlying pathogenesis of UNC13D variants. It is crucial to prioritize the timely and accurate diagnosis of atypical patients, as they may often be overlooked among individuals with rheumatic or hematological diseases.


Subject(s)
Lymphohistiocytosis, Hemophagocytic , Membrane Proteins , Humans , Lymphohistiocytosis, Hemophagocytic/diagnosis , Lymphohistiocytosis, Hemophagocytic/genetics , Male , Membrane Proteins/genetics , Female , Child, Preschool , Infant , Child , Mutation , Phenotype , China/epidemiology
10.
Clin Lab ; 70(5)2024 May 01.
Article in English | MEDLINE | ID: mdl-38747923

ABSTRACT

BACKGROUND: Familial hemophagocytic lymphohistiocytosis (FHL) onset in the fetal and neonatal periods is sporadic, and infants are susceptible to intrauterine death. Early and accurate diagnosis and treatment are the keys to preventing complications and death in FHL patients due to the complex and diverse clinical manifestations of the disease. METHODS: We report a rare case of a preterm infant with a low birth weight of 2,010 g and a gestational age of 32 + 4 weeks who presented with a leaky syndrome similar to sepsis after birth. Anti-infective, other support, and symptomatic treatments were not effective. Bone marrow examination results on day 13 suggested hemophago-cytosis. RESULTS: Various compound heterozygous UNC13D genes were found by exome sequencing, which confirmed the diagnosis of FHL type 3. Genetic variants of this locus have never been reported in the literature. CONCLUSIONS: Neonatal onset FHL is challenging to diagnose, especially in premature infants. It is necessary to complete exome sequencing if the patient has no apparent pathogen infection or effective treatment.


Subject(s)
Infant, Low Birth Weight , Infant, Premature , Lymphohistiocytosis, Hemophagocytic , Humans , Lymphohistiocytosis, Hemophagocytic/genetics , Lymphohistiocytosis, Hemophagocytic/diagnosis , Infant, Newborn , Exome Sequencing , Membrane Proteins/genetics , Male , Female , Gestational Age
11.
PLoS One ; 19(5): e0302817, 2024.
Article in English | MEDLINE | ID: mdl-38743659

ABSTRACT

BACKGROUND: IgG subclass levels in hemochromatosis are incompletely characterized. METHODS: We characterized IgG subclass levels of referred hemochromatosis probands with HFE p.C282Y/p.C282Y (rs1800562) and human leukocyte antigen (HLA)-A and -B typing/haplotyping and compared them with IgG subclass levels of eight published cohorts of adults unselected for hemochromatosis. RESULTS: There were 157 probands (82 men, 75 women; mean age 49±13 y). Median serum ferritin, mean body mass index (BMI), median IgG4, and median phlebotomy units to achieve iron depletion were significantly higher in men. Diabetes, cirrhosis, and HLA-A*03,-B*44, -A*03,B*07, and -A*01,B*08 prevalences and median absolute lymphocyte counts in men and women did not differ significantly. Mean IgG subclass levels [95% confidence interval] were: IgG1 5.31 g/L [3.04, 9.89]; IgG2 3.56 g/L [1.29, 5.75]; IgG3 0.61 g/L [0.17, 1.40]; and IgG4 0.26 g/L [<0.01, 1.25]. Relative IgG subclasses were 54.5%, 36.6%, 6.3%, and 2.7%, respectively. Median IgG4 was higher in men than women (0.34 g/L [0.01, 1.33] vs. 0.19 g/L [<0.01, 0.75], respectively; p = 0.0006). A correlation matrix with Bonferroni correction revealed the following positive correlations: IgG1 vs. IgG3 (p<0.01); IgG2 vs. IgG3 (p<0.05); and IgG2 vs. IgG4 (p<0.05). There was also a positive correlation of IgG4 vs. male sex (p<0.01). Mean IgG1 was lower and mean IgG2 was higher in probands than seven of eight published adult cohorts unselected for hemochromatosis diagnoses. CONCLUSIONS: Mean IgG subclass levels of hemochromatosis probands were 5.31, 3.56, 0.61, and 0.26 g/L, respectively. Median IgG4 was higher in men than women. There were positive associations of IgG subclass levels. Mean IgG1 may be lower and mean IgG2 may be higher in hemochromatosis probands than adults unselected for hemochromatosis.


Subject(s)
Hemochromatosis Protein , Hemochromatosis , Immunoglobulin G , Humans , Male , Hemochromatosis/blood , Hemochromatosis/genetics , Hemochromatosis/immunology , Female , Immunoglobulin G/blood , Middle Aged , Hemochromatosis Protein/genetics , Adult , Aged , Membrane Proteins/immunology , Membrane Proteins/genetics , Histocompatibility Antigens Class I/genetics , Histocompatibility Antigens Class I/immunology
12.
Chirurgia (Bucur) ; 119(2): 136-155, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38743828

ABSTRACT

Background: Colorectal cancer (CRC) exhibits molecular and morphological diversity, involving genetic, epigenetic alterations, and disruptions in signaling pathways. This necessitates a comprehensive review synthesizing recent advancements in molecular mechanisms, established biomarkers, as well as emerging ones like CDX2 for enhanced CRC assessment. Material and Methods: This review analyzes the last decade's literature and current guidelines to study CRC's molecular intricacies. It extends the analysis beyond traditional biomarkers to include emerging ones like CDX2, examining their interaction with carcinogenic mechanisms and molecular pathways, alongside reviewing current testing methodologies. Results: A multi-biomarker strategy, incorporating both traditional and emerging biomarkers like CDX2, is crucial for optimizing CRC management. This strategy elucidates the complex interaction between biomarkers and the tumor's molecular pathways, significantly influencing prognostic evaluations, therapeutic decision-making, and paving the way for personalized medicine in CRC. Conclusions: This review proposes CDX2 as an emerging prognostic biomarker and emphasizes the necessity of thorough molecular profiling for individualized treatment strategies. By enhancing CRC treatment approaches and prognostic evaluation, this effort marks a step forward in precision oncology, leveraging an enriched understanding of tumor behavior.


Subject(s)
Biomarkers, Tumor , CDX2 Transcription Factor , Colorectal Neoplasms , Membrane Proteins , Microsatellite Instability , Proto-Oncogene Proteins B-raf , Proto-Oncogene Proteins p21(ras) , Humans , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/classification , CDX2 Transcription Factor/metabolism , CDX2 Transcription Factor/genetics , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Proto-Oncogene Proteins p21(ras)/genetics , Proto-Oncogene Proteins B-raf/genetics , Prognosis , Membrane Proteins/genetics , Membrane Proteins/metabolism , GTP Phosphohydrolases/genetics , GTP Phosphohydrolases/metabolism , DNA Mismatch Repair , Predictive Value of Tests , Precision Medicine
13.
Cell Death Dis ; 15(5): 334, 2024 May 14.
Article in English | MEDLINE | ID: mdl-38744890

ABSTRACT

The prevalence of diabetes steadily increases worldwide mirroring the prevalence of obesity. Endoplasmic reticulum (ER) stress is activated in diabetes and contributes to ß-cell dysfunction and apoptosis through the activation of a terminal unfolded protein response (UPR). Our results uncover a new role for Bax Inhibitor-One (BI-1), a negative regulator of inositol-requiring enzyme 1 (IRE1α) in preserving ß-cell health against terminal UPR-induced apoptosis and pyroptosis in the context of supraphysiological loads of insulin production. BI-1-deficient mice experience a decline in endocrine pancreatic function in physiological and pathophysiological conditions, namely obesity induced by high-fat diet (HFD). We observed early-onset diabetes characterized by hyperglycemia, reduced serum insulin levels, ß-cell loss, increased pancreatic lipases and pro-inflammatory cytokines, and the progression of metabolic dysfunction. Pancreatic section analysis revealed that BI-1 deletion overburdens unfolded proinsulin in the ER of ß-cells, confirmed by ultrastructural signs of ER stress with overwhelmed IRE1α endoribonuclease (RNase) activity in freshly isolated islets. ER stress led to ß-cell dysfunction and islet loss, due to an increase in immature proinsulin granules and defects in insulin crystallization with the presence of Rod-like granules. These results correlated with the induction of autophagy, ER phagy, and crinophagy quality control mechanisms, likely to alleviate the atypical accumulation of misfolded proinsulin in the ER. In fine, BI-1 in ß-cells limited IRE1α RNase activity from triggering programmed ß-cell death through apoptosis and pyroptosis (caspase-1, IL-1ß) via NLRP3 inflammasome activation and metabolic dysfunction. Pharmaceutical IRE1α inhibition with STF-083010 reversed ß-cell failure and normalized the metabolic phenotype. These results uncover a new protective role for BI-1 in pancreatic ß-cell physiology as a stress integrator to modulate the UPR triggered by accumulating unfolded proinsulin in the ER, as well as autophagy and programmed cell death, with consequences on ß-cell function and insulin secretion. In pancreatic ß-cells, BI-1-/- deficiency perturbs proteostasis with proinsulin misfolding, ER stress, terminal UPR with overwhelmed IRE1α/XBP1s/CHOP activation, inflammation, ß-cell programmed cell death, and diabetes.


Subject(s)
Apoptosis , Endoplasmic Reticulum Stress , Insulin-Secreting Cells , Membrane Proteins , Proinsulin , Proteostasis , Unfolded Protein Response , Animals , Insulin-Secreting Cells/metabolism , Insulin-Secreting Cells/pathology , Proinsulin/metabolism , Mice , Membrane Proteins/metabolism , Membrane Proteins/genetics , Protein Folding , Endoribonucleases/metabolism , Mice, Inbred C57BL , Diet, High-Fat , Mice, Knockout , Male
14.
Sci Rep ; 14(1): 11020, 2024 05 14.
Article in English | MEDLINE | ID: mdl-38745067

ABSTRACT

The absence of stimulator of interferon genes (STING) in 129.B6.Fcgr2b-deficient mice rescue lupus phenotypes. The administration of a STING inhibitor (ISD017) into the young 129.B6.Fcgr2b-deficient mice prevents lupus nephritis development. This study mainly aimed to evaluate the effects of STING inhibition (ISD107) on established SLE in mice to prove that ISD017 could be a good therapeutic drug to reverse the already set-up autoimmunity and kidney impairment. Twenty-four-week-old Fcgr2b-deficient mice were treated with cyclophosphamide (25 mg/kg, intraperitoneal, once per week), ISD017 (10 mg/kg, intraperitoneal, three times per week), or control vehicle for 8 weeks, and were analyzed for phenotypes. Both ISD017 and cyclophosphamide treatment increased long-term survival and reduced the severity of glomerulonephritis in Fcgr2b-deficient mice. While cyclophosphamide reduced activated B cells (B220+GL-7+), ISD017 decreased activated T cells (CD4+CD69+) and neutrophils (Ly6c+Ly6g+) in Fcgr2b-deficient mice. In addition, ISD017 reduced IL-1ß and interferon-inducible genes. In summary, ISD017 treatment in symptomatic 129.B6.Fcgr2b-deficient mice reduced the severity of glomerulonephritis and increased long-term survival. ISD017 worked comparably to cyclophosphamide for treating lupus nephritis in 129.B6.Fcgr2b-deficient mice. ISD017 reduced activated T cells and neutrophils, while cyclophosphamide targeted activated B cells. These results suggested that STING inhibitors can potentially be a new therapeutic drug for treating lupus.


Subject(s)
Cyclophosphamide , Membrane Proteins , Receptors, IgG , Animals , Mice , Membrane Proteins/genetics , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/deficiency , Membrane Proteins/metabolism , Cyclophosphamide/pharmacology , Receptors, IgG/genetics , Receptors, IgG/metabolism , Lupus Nephritis/drug therapy , Lupus Nephritis/pathology , Glomerulonephritis/drug therapy , Mice, Knockout , Female , Disease Models, Animal , B-Lymphocytes/drug effects , B-Lymphocytes/metabolism , B-Lymphocytes/immunology , Lupus Erythematosus, Systemic/drug therapy , Lupus Erythematosus, Systemic/genetics , Mice, Inbred C57BL
15.
Front Immunol ; 15: 1376629, 2024.
Article in English | MEDLINE | ID: mdl-38715613

ABSTRACT

ORMDL3 is a prominent member of a family of highly conserved endoplasmic reticulum resident proteins, ORMs (ORM1 and ORM2) in yeast, dORMDL in Drosophila and ORMDLs (ORMDL1, ORMDL2, and ORMDL3) in mammals. ORMDL3 mediates feedback inhibition of de novo sphingolipid synthesis. Expression levels of ORMDL3 are associated with the development of inflammatory and autoimmune diseases including asthma, systemic lupus erythematosus, type 1 diabetes mellitus and others. It has been shown that simultaneous deletions of other ORMDL family members could potentiate ORMDL3-induced phenotypes. To understand the complex function of ORMDL proteins in immunity in vivo, we analyzed mice with single or double deletions of Ormdl genes. In contrast to other single and double knockouts, simultaneous deletion of ORMDL1 and ORMDL3 proteins disrupted blood homeostasis and reduced immune cell content in peripheral blood and spleens of mice. The reduced number of splenocytes was not caused by aberrant immune cell homing. A competitive bone marrow transplantation assay showed that the development of Ormdl1-/-/Ormdl3-/- B cells was dependent on lymphocyte intrinsic factors. Highly increased sphingolipid production was observed in the spleens and bone marrow of Ormdl1-/-/Ormdl3-/- mice. Slight, yet significant, increase in some sphingolipid species was also observed in the spleens of Ormdl3-/- mice and in the bone marrow of both, Ormdl1-/- and Ormdl3-/- single knockout mice. Taken together, our results demonstrate that the physiological expression of ORMDL proteins is critical for the proper development and circulation of lymphocytes. We also show cell-type specific roles of individual ORMDL family members in the production of different sphingolipid species.


Subject(s)
Homeostasis , Membrane Proteins , Mice, Knockout , Animals , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Sphingolipids/metabolism , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Gene Deletion , Mice, Inbred C57BL , Spleen/immunology , Spleen/metabolism
16.
Clin Epigenetics ; 16(1): 62, 2024 May 07.
Article in English | MEDLINE | ID: mdl-38715103

ABSTRACT

BACKGROUND: Temple syndrome (TS14) is a rare imprinting disorder caused by maternal UPD14, imprinting defects or paternal microdeletions which lead to an increase in the maternal expressed genes and a silencing the paternally expressed genes in the 14q32 imprinted domain. Classical TS14 phenotypic features include pre- and postnatal short stature, small hands and feet, muscular hypotonia, motor delay, feeding difficulties, weight gain, premature puberty along and precocious puberty. METHODS: An exon array comparative genomic hybridization was performed on a patient affected by psychomotor and language delay, muscular hypotonia, relative macrocephaly, and small hand and feet at two years old. At 6 years of age, the proband presented with precocious thelarche. Genes dosage and methylation within the 14q32 region were analyzed by MS-MLPA. Bisulfite PCR and pyrosequencing were employed to quantification methylation at the four known imprinted differentially methylated regions (DMR) within the 14q32 domain: DLK1 DMR, IG-DMR, MEG3 DMR and MEG8 DMR. RESULTS: The patient had inherited a 69 Kb deletion, encompassing the entire DLK1 gene, on the paternal allele. Relative hypermethylation of the two maternally methylated intervals, DLK1 and MEG8 DMRs, was observed along with normal methylation level at IG-DMR and MEG3 DMR, resulting in a phenotype consistent with TS14. Additional family members with the deletion showed modest methylation changes at both the DLK1 and MEG8 DMRs consistent with parental transmission. CONCLUSION: We describe a girl with clinical presentation suggestive of Temple syndrome resulting from a small paternal 14q32 deletion that led to DLK1 whole-gene deletion, as well as hypermethylation of the maternally methylated DLK1-DMR.


Subject(s)
Calcium-Binding Proteins , Chromosomes, Human, Pair 14 , DNA Methylation , Genomic Imprinting , Intercellular Signaling Peptides and Proteins , Humans , Calcium-Binding Proteins/genetics , DNA Methylation/genetics , Chromosomes, Human, Pair 14/genetics , Intercellular Signaling Peptides and Proteins/genetics , Genomic Imprinting/genetics , Membrane Proteins/genetics , Child , Male , Comparative Genomic Hybridization/methods , Female , Chromosome Deletion , Child, Preschool , Phenotype , Abnormalities, Multiple/genetics , Imprinting Disorders , Muscle Hypotonia , Facies
17.
Elife ; 122024 May 02.
Article in English | MEDLINE | ID: mdl-38695862

ABSTRACT

Here, we investigated the mechanisms by which aging-related reductions of the levels of Numb in skeletal muscle fibers contribute to loss of muscle strength and power, two critical features of sarcopenia. Numb is an adaptor protein best known for its critical roles in development, including asymmetric cell division, cell-type specification, and termination of intracellular signaling. Numb expression is reduced in old humans and mice. We previously showed that, in mouse skeletal muscle fibers, Numb is localized to sarcomeres where it is concentrated near triads; conditional inactivation of Numb and a closely related protein Numb-like (Numbl) in mouse myofibers caused weakness, disorganization of sarcomeres, and smaller mitochondria with impaired function. Here, we found that a single knockout of Numb in myofibers causes reduction in tetanic force comparable to a double Numb, Numbl knockout. We found by proteomics analysis of protein complexes isolated from C2C12 myotubes by immunoprecipitation using antibodies against Numb that Septin 7 is a potential Numb-binding partner. Septin 7 is a member of the family of GTP-binding proteins that organize into filaments, sheets, and rings, and is considered part of the cytoskeleton. Immunofluorescence evaluation revealed a partial overlap of staining for Numb and Septin 7 in myofibers. Conditional, inducible knockouts of Numb led to disorganization of Septin 7 staining in myofibers. These findings indicate that Septin 7 is a Numb-binding partner and suggest that interactions between Numb and Septin 7 are critical for structural organization of the sarcomere and muscle contractile function.


Subject(s)
Intracellular Signaling Peptides and Proteins , Membrane Proteins , Mice, Knockout , Muscle Contraction , Nerve Tissue Proteins , Sarcomeres , Septins , Animals , Septins/metabolism , Septins/genetics , Sarcomeres/metabolism , Mice , Muscle Contraction/physiology , Membrane Proteins/metabolism , Membrane Proteins/genetics , Nerve Tissue Proteins/metabolism , Nerve Tissue Proteins/genetics , Protein Binding , Muscle Fibers, Skeletal/metabolism , Muscle Fibers, Skeletal/physiology
18.
Nat Commun ; 15(1): 2869, 2024 May 01.
Article in English | MEDLINE | ID: mdl-38693144

ABSTRACT

Only ~20% of heavy drinkers develop alcohol cirrhosis (AC). While differences in metabolism, inflammation, signaling, microbiome signatures and genetic variations have been tied to the pathogenesis of AC, the key underlying mechanisms for this interindividual variability, remain to be fully elucidated. Induced pluripotent stem cell-derived hepatocytes (iHLCs) from patients with AC and healthy controls differ transcriptomically, bioenergetically and histologically. They include a greater number of lipid droplets (LDs) and LD-associated mitochondria compared to control cells. These pre-pathologic indicators are effectively reversed by Aramchol, an inhibitor of stearoyl-CoA desaturase. Bioenergetically, AC iHLCs have lower spare capacity, slower ATP production and their mitochondrial fuel flexibility towards fatty acids and glutamate is weakened. MARC1 and PNPLA3, genes implicated by GWAS in alcohol cirrhosis, show to correlate with lipid droplet-associated and mitochondria-mediated oxidative damage in AC iHLCs. Knockdown of PNPLA3 expression exacerbates mitochondrial deficits and leads to lipid droplets alterations. These findings suggest that differences in mitochondrial bioenergetics and lipid droplet formation are intrinsic to AC hepatocytes and can play a role in its pathogenesis.


Subject(s)
Acyltransferases , Energy Metabolism , Hepatocytes , Induced Pluripotent Stem Cells , Lipase , Lipid Droplets , Liver Cirrhosis, Alcoholic , Mitochondria , Phospholipases A2, Calcium-Independent , Humans , Hepatocytes/metabolism , Hepatocytes/pathology , Induced Pluripotent Stem Cells/metabolism , Lipid Droplets/metabolism , Liver Cirrhosis, Alcoholic/metabolism , Liver Cirrhosis, Alcoholic/pathology , Liver Cirrhosis, Alcoholic/genetics , Lipase/metabolism , Lipase/genetics , Mitochondria/metabolism , Male , Membrane Proteins/metabolism , Membrane Proteins/genetics , Female , Middle Aged , Adult , Oxidative Stress
19.
Cancer Discov ; 14(5): 698-700, 2024 May 01.
Article in English | MEDLINE | ID: mdl-38692265

ABSTRACT

SUMMARY: In this issue, Rubinson, Tanaka, and colleagues demonstrate that differences among G12C inhibitors rely on their ability to covalently bind not only G12C mutant KRAS but also NRAS and HRAS, proposing sotorasib as a potent NRAS G12C inhibitor. See related article by Rubinson et al., p. 727 (6).


Subject(s)
Piperazines , Proto-Oncogene Proteins p21(ras) , Pyridines , Pyrimidines , Humans , Proto-Oncogene Proteins p21(ras)/genetics , Proto-Oncogene Proteins p21(ras)/antagonists & inhibitors , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/genetics , Mutation , GTP Phosphohydrolases/genetics , Antineoplastic Agents/therapeutic use , Antineoplastic Agents/pharmacology
20.
BMC Cancer ; 24(1): 551, 2024 May 01.
Article in English | MEDLINE | ID: mdl-38693472

ABSTRACT

OBJECTIVE: We aimed to screen novel gene signatures for ovarian cancer (OC) and explore the role of biomarkers in OC via regulating pyroptosis using bioinformatics analysis. METHODS: Differentially expressed genes (DEGs) of OC were screened from GSE12470 and GSE16709 datasets. Hub genes were determined from protein-protein interaction networks after bioinformatics analysis. The role of Centromeric protein M (CENPM) in OC was assessed by subcutaneous tumor experiment using hematoxylin-eosin and immunohistochemical staining. Tumor metastasis was evaluated by detecting epithelial-mesenchymal transition-related proteins. The proliferation, migration, and invasion were determined using cell counting kit and transwell assay. Enzyme-linked immunosorbent assay was applied to measure inflammatory factors. The mRNA and protein expression were detected using real-time quantitative PCR and western blot. RESULTS: We determined 9 hub genes (KIFC1, PCLAF, CDCA5, KNTC1, MCM3, OIP5, CENPM, KIF15, and ASF1B) with high prediction value for OC. In SKOV3 and A2780 cells, the expression levels of hub genes were significantly up-regulated, compared with normal ovarian cells. CENPM was selected as a key gene. Knockdown of CENPM suppressed proliferation, migration, and invasion of OC cells. Subcutaneous tumor experiment revealed that CENPM knockdown significantly suppressed tumor growth and metastasis. Additionally, pyroptosis was promoted in OC cells and xenograft tumors after CENPM knockdown. Furthermore, CENPM knockdown activated cGAS-STING pathway and the pathway inhibitor reversed the inhibitory effect of CENPM knockdown on viability, migration, and invasion of OC cells. CONCLUSION: CENPM was a novel biomarker of OC, and knockdown of CENPM inhibited OC progression by promoting pyroptosis and activating cGAS-STING pathway.


Subject(s)
Membrane Proteins , Nucleotidyltransferases , Ovarian Neoplasms , Pyroptosis , Signal Transduction , Humans , Female , Pyroptosis/genetics , Membrane Proteins/genetics , Membrane Proteins/metabolism , Ovarian Neoplasms/genetics , Ovarian Neoplasms/pathology , Ovarian Neoplasms/metabolism , Animals , Mice , Nucleotidyltransferases/metabolism , Nucleotidyltransferases/genetics , Cell Line, Tumor , Gene Knockdown Techniques , Cell Proliferation/genetics , Gene Expression Regulation, Neoplastic , Chromosomal Proteins, Non-Histone/metabolism , Chromosomal Proteins, Non-Histone/genetics , Cell Movement/genetics , Xenograft Model Antitumor Assays , Mice, Nude
SELECTION OF CITATIONS
SEARCH DETAIL
...