Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 345
Filter
1.
Clin Chim Acta ; 564: 119948, 2025 Jan 01.
Article in English | MEDLINE | ID: mdl-39214396

ABSTRACT

Chronic renal failure (CRF) is an incurable disease with unique challenges. Anemia is a frequent complication affecting dialysis patients. Erythropoietin (EPO) is used to treat anemia, but a poor response may result. We investigated genetic polymorphisms of store-operated calcium channel (SOC) signaling, an important erythropoietin-activated pathway that may induce EPO resistance in patients with renal failure. A total of 108 end stage renal disease (ESRD) patients were selected for this study. Patients were divided into two groups according to their erythropoietin resistance index (ERI): 39 patients with an ERI>10 and 69 patients with an ERI<10. We selected four tagging single nucleotide polymorphisms (tSNPs) in STIM1 and five in ORAI1 in our study. A polymerase chain reaction was performed, and genotyping against EPO resistance was correlated. Patients with the AG genotype of rs1561876 in STIM1, the TC genotype of rs6486795 in ORAI1, and the TG or GG genotypes of rs12320939 in ORAI1 were associated with an increased risk of erythropoietin resistance. Overall, we reported a moderately significant relationship between genetic polymorphisms of STIM1 and EPO resistance. We also reported a highly significant relationship between genetic polymorphisms of ORAI1 and EPO resistance. The (A-A-G) haplotype of STIM1 and the (G-T-G-T-A, G-C-G-C-G, or G-T-T-C-G) haplotypes of ORAI1 were significantly associated with EPO resistance.


Subject(s)
Erythropoietin , Kidney Failure, Chronic , Neoplasm Proteins , ORAI1 Protein , Polymorphism, Single Nucleotide , Stromal Interaction Molecule 1 , Humans , Stromal Interaction Molecule 1/genetics , Egypt , Kidney Failure, Chronic/genetics , Male , Erythropoietin/genetics , Female , ORAI1 Protein/genetics , Middle Aged , Neoplasm Proteins/genetics , Adult , Drug Resistance/genetics
2.
Proc Natl Acad Sci U S A ; 121(39): e2400531121, 2024 Sep 24.
Article in English | MEDLINE | ID: mdl-39292746

ABSTRACT

It is well established that DNA Damage Regulated Autophagy Modulator 1 (DRAM1), a lysosomal protein and a target of p53, participates in autophagy. The cellular functions of DRAM1 beyond autophagy remain elusive. Here, we show p53-dependent upregulation of DRAM1 in mitochondrial damage-induced Parkinson's disease (PD) models and exacerbation of disease phenotypes by DRAM1. We find that the lysosomal location of DRAM1 relies on its intact structure including the cytosol-facing C-terminal domain. Excess DRAM1 disrupts endoplasmic reticulum (ER) structure, triggers ER stress, and induces protective ER-phagy. Mechanistically, DRAM1 interacts with stromal interacting molecule 1 (STIM1) to tether lysosomes to the ER and perturb STIM1 function in maintaining intracellular calcium homeostasis. STIM1 overexpression promotes cellular health by restoring calcium homeostasis, ER stress response, ER-phagy, and AMP-activated protein kinase (AMPK)-Unc-51 like autophagy activating kinase 1 (ULK1) signaling in cells with excess DRAM1. Thus, by promoting organelle contact between lysosomes and the ER, DRAM1 modulates ER structure and function and cell survival under stress. Our results suggest that DRAM1 as a lysosomal protein performs diverse roles in cellular homeostasis and stress response. These findings may have significant implications for our understanding of the role of the p53/DRAM1 axis in human diseases, from cancer to neurodegenerative diseases.


Subject(s)
Calcium , Endoplasmic Reticulum Stress , Endoplasmic Reticulum , Homeostasis , Lysosomes , Membrane Proteins , Stromal Interaction Molecule 1 , Tumor Suppressor Protein p53 , Lysosomes/metabolism , Stromal Interaction Molecule 1/metabolism , Stromal Interaction Molecule 1/genetics , Humans , Calcium/metabolism , Endoplasmic Reticulum/metabolism , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Protein p53/genetics , Endoplasmic Reticulum Stress/physiology , Membrane Proteins/metabolism , Membrane Proteins/genetics , Animals , Mice , Autophagy/physiology , Autophagy-Related Protein-1 Homolog/metabolism , Autophagy-Related Protein-1 Homolog/genetics , Neoplasm Proteins/metabolism , Neoplasm Proteins/genetics , Parkinson Disease/metabolism , Parkinson Disease/genetics , Parkinson Disease/pathology
3.
Discov Med ; 36(187): 1678-1691, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39190383

ABSTRACT

BACKGROUND: Remodeling of vascular smooth muscle cells (VSMCs), as a pathological hallmark of cardiovascular diseases, is related to the molecular rewiring of Calcium signaling, which induces upregulation of stromal interaction molecule (STIM) proteins. This study analyzed the influence of STIM1 proteins on the remodeling of VSMCs in atherosclerosis (AS). METHODS: After oxidized low-density lipoprotein (ox-LDL) treatment and transfection, VSMC viability, migration, and invasion were separately measured using Cell Counting Kit-8, Scratch assay, and Transwell assay. An animal AS model was constructed, and histological analysis via hematoxylin-eosin staining was conducted on the aorta. RESULTS: Ox-LDL promoted expression of STIM1 and Orai calcium release-activated calcium modulator 1 (Orai1). STIM1 or Orai1 downregulation suppressed viability, migration, invasion, and phenotypic switching of ox-LDL-treated VSMCs, whereas STIM1 or Orai1 upregulation had opposite effects. Orai1 level was upregulated by STIM1 overexpression. Orai1 silencing reversed the effects of STIM1 overexpression in VSMCs. STIM1 deficiency alleviated AS and regulated expression of Orai1 and phenotypic switch-related factors in vivo. CONCLUSION: STIM1 deficiency suppresses viability, migration, invasion, and phenotypic switching of ox-LDL-induced VSMCs and alleviates AS by inhibiting Orai1.


Subject(s)
Atherosclerosis , Cell Movement , Lipoproteins, LDL , Muscle, Smooth, Vascular , Myocytes, Smooth Muscle , ORAI1 Protein , Stromal Interaction Molecule 1 , Animals , Humans , Male , Mice , Atherosclerosis/pathology , Atherosclerosis/metabolism , Cell Movement/drug effects , Cell Survival/drug effects , Disease Models, Animal , Lipoproteins, LDL/metabolism , Muscle, Smooth, Vascular/pathology , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/pathology , Neoplasm Proteins/metabolism , Neoplasm Proteins/genetics , ORAI1 Protein/metabolism , ORAI1 Protein/genetics , ORAI1 Protein/antagonists & inhibitors , Stromal Interaction Molecule 1/metabolism , Stromal Interaction Molecule 1/genetics , Vascular Remodeling/drug effects
4.
Proc Natl Acad Sci U S A ; 121(35): e2407324121, 2024 Aug 27.
Article in English | MEDLINE | ID: mdl-39178228

ABSTRACT

Learning and memory require coordinated structural and functional plasticity at neuronal glutamatergic synapses located on dendritic spines. Here, we investigated how the endoplasmic reticulum (ER) controls postsynaptic Ca2+ signaling and long-term potentiation of dendritic spine size, i.e., sLTP that accompanies functional strengthening of glutamatergic synaptic transmission. In most ER-containing (ER+) spines, high-frequency optical glutamate uncaging (HFGU) induced long-lasting sLTP that was accompanied by a persistent increase in spine ER content downstream of a signaling cascade engaged by N-methyl-D-aspartate receptors (NMDARs), L-type Ca2+ channels (LTCCs), and Orai1 channels, the latter being activated by stromal interaction molecule 1 (STIM1) in response to ER Ca2+ release. In contrast, HFGU stimulation of ER-lacking (ER-) spines expressed only transient sLTP and exhibited weaker Ca2+ signals noticeably lacking Orai1 and ER contributions. Consistent with spine ER regulating structural metaplasticity, delivery of a second stimulus to ER- spines induced ER recruitment along with persistent sLTP, whereas ER+ spines showed no additional increases in size or ER content in response to sequential stimulation. Surprisingly, the physical interaction between STIM1 and Orai1 induced by ER Ca2+ release, but not the resulting Ca2+ entry through Orai1 channels, proved necessary for the persistent increases in both spine size and ER content required for expression of long-lasting late sLTP.


Subject(s)
Calcium Channels, L-Type , Dendritic Spines , Endoplasmic Reticulum , Neuronal Plasticity , ORAI1 Protein , Stromal Interaction Molecule 1 , Stromal Interaction Molecule 1/metabolism , Stromal Interaction Molecule 1/genetics , Endoplasmic Reticulum/metabolism , Dendritic Spines/metabolism , Animals , ORAI1 Protein/metabolism , ORAI1 Protein/genetics , Neuronal Plasticity/physiology , Calcium Channels, L-Type/metabolism , Long-Term Potentiation/physiology , Calcium Signaling/physiology , Receptors, N-Methyl-D-Aspartate/metabolism , Calcium/metabolism , Mice , Signal Transduction/physiology , Rats
5.
Cell Calcium ; 123: 102926, 2024 Nov.
Article in English | MEDLINE | ID: mdl-38959763

ABSTRACT

Two recent papers have highlighted that STIM1, a key component of Store-operated Ca2+-entry, is able to translocate to the nucleus and participate in nuclear Ca2+-handling and in DNA repair. These finding opens new avenues on the role that this Ca2+-sensing protein may have in health and disease.


Subject(s)
Calcium , Cell Nucleus , Neoplasm Proteins , Stromal Interaction Molecule 1 , Animals , Humans , Calcium/metabolism , Calcium Signaling , Cell Nucleus/metabolism , DNA Repair , Membrane Proteins/metabolism , Myopathies, Structural, Congenital/metabolism , Myopathies, Structural, Congenital/pathology , Myopathies, Structural, Congenital/genetics , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Stromal Interaction Molecule 1/genetics , Stromal Interaction Molecule 1/metabolism , Mice
6.
Hypertension ; 81(9): 1895-1909, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38989583

ABSTRACT

BACKGROUND: STIM1 (stromal interaction molecule 1) regulates store-operated calcium entry and is involved in pulmonary artery vasoconstriction and pulmonary artery smooth muscle cell proliferation, leading to pulmonary arterial hypertension (PAH). METHODS: Bioinformatics analysis and a 2-stage matched case-control study were conducted to screen for noncoding variants that may potentially affect STIM1 transcriptional regulation in 242 patients with idiopathic PAH and 414 healthy controls. Luciferase reporter assay, real-time quantitative polymerase chain reaction, western blot, 5-ethynyl-2'-deoxyuridine (EdU) assay, and intracellular Ca2+ measurement were performed to study the mechanistic roles of those STIM1 noncoding variants in PAH. RESULTS: Five noncoding variants (rs3794050, rs7934581, rs3750996, rs1561876, and rs3750994) were identified and genotyped using Sanger sequencing. Rs3794050, rs7934581, and rs1561876 were associated with idiopathic PAH (recessive model, all P<0.05). Bioinformatics analysis showed that these 3 noncoding variants possibly affect the enhancer function of STIM1 or the microRNA (miRNA) binding to STIM1. Functional validation performed in HEK293 and pulmonary artery smooth muscle cells demonstrated that the noncoding variant rs1561876-G (STIM1 mutant) had significantly stronger transcriptional activity than the wild-type counterpart, rs1561876-A, by affecting the transcriptional regulatory function of both hsa-miRNA-3140-5p and hsa-miRNA-4766-5p. rs1561876-G enhanced intracellular Ca2+ signaling in human pulmonary artery smooth muscle cells secondary to calcium-sensing receptor activation and promoted proliferation of pulmonary artery smooth muscle cells under both normoxia and hypoxia conditions, suggesting a possible contribution to PAH development. CONCLUSIONS: The potential clinical implications of the 3 noncoding variants of STIM1, rs3794050, rs7934581, and rs1561876, are 2-fold, as they may help predict the risk and prognosis of idiopathic PAH and guide investigations on novel therapeutic pathway(s).


Subject(s)
Pulmonary Artery , Stromal Interaction Molecule 1 , Humans , Stromal Interaction Molecule 1/genetics , Stromal Interaction Molecule 1/metabolism , Male , Female , Case-Control Studies , Middle Aged , Adult , Neoplasm Proteins/genetics , Myocytes, Smooth Muscle/metabolism , Familial Primary Pulmonary Hypertension/genetics , Familial Primary Pulmonary Hypertension/physiopathology , Genetic Predisposition to Disease , Muscle, Smooth, Vascular/metabolism , Gene Expression Regulation , Cell Proliferation/genetics , Polymorphism, Single Nucleotide
7.
Mol Cell ; 84(14): 2596-2597, 2024 07 25.
Article in English | MEDLINE | ID: mdl-39059368

ABSTRACT

In a recent publication in Cell, Woo et al.1 report that stimulator of interferon genes (STING) links inflammation with glutamate-driven excitotoxicity to induce ferroptosis, identifying a mechanism of inflammation-induced neurodegeneration and also a novel candidate therapeutic target for multiple sclerosis.


Subject(s)
Ferroptosis , Membrane Proteins , Multiple Sclerosis , Neuroprotection , Humans , Membrane Proteins/metabolism , Membrane Proteins/genetics , Animals , Multiple Sclerosis/genetics , Multiple Sclerosis/immunology , Multiple Sclerosis/drug therapy , Multiple Sclerosis/metabolism , Ferroptosis/drug effects , Ferroptosis/genetics , Stromal Interaction Molecule 1/metabolism , Stromal Interaction Molecule 1/genetics , Glutamic Acid/metabolism , Inflammation , Signal Transduction
8.
Int J Mol Sci ; 25(14)2024 Jul 10.
Article in English | MEDLINE | ID: mdl-39062821

ABSTRACT

Sensing the lowering of endoplasmic reticulum (ER) calcium (Ca2+), STIM1 mediates a ubiquitous Ca2+ influx process called the store-operated Ca2+ entry (SOCE). Dysregulated STIM1 function or abnormal SOCE is strongly associated with autoimmune disorders, atherosclerosis, and various forms of cancers. Therefore, uncovering the molecular intricacies of post-translational modifications, such as oxidation, on STIM1 function is of paramount importance. In a recent proteomic screening, we identified three protein disulfide isomerases (PDIs)-Prolyl 4-hydroxylase subunit beta (P4HB), protein disulfide-isomerase A3 (PDIA3), and thioredoxin domain-containing protein 5 (TXNDC5)-as the ER-luminal interactors of STIM1. Here, we demonstrated that these PDIs dynamically associate with STIM1 and STIM2. The mutation of the two conserved cysteine residues of STIM1 (STIM1-2CA) decreased its Ca2+ affinity both in cellulo and in situ. Knockdown of PDIA3 or P4HB increased the Ca2+ affinity of wild-type STIM1 while showing no impact on the STIM1-2CA mutant, indicating that PDIA3 and P4HB regulate STIM1's Ca2+ affinity by acting on ER-luminal cysteine residues. This modulation of STIM1's Ca2+ sensitivity was further confirmed by Ca2+ imaging experiments, which showed that knockdown of these two PDIs does not affect STIM1-mediated SOCE upon full store depletion but leads to enhanced SOCE amplitudes upon partial store depletion. Thus, P4HB and PDIA3 dynamically modulate STIM1 activation by fine-tuning its Ca2+ binding affinity, adjusting the level of activated STIM1 in response to physiological cues. The coordination between STIM1-mediated Ca2+ signaling and redox responses reported herein may have implications for cell physiology and pathology.


Subject(s)
Calcium , Neoplasm Proteins , Oxidation-Reduction , Procollagen-Proline Dioxygenase , Protein Disulfide-Isomerases , Stromal Interaction Molecule 1 , Stromal Interaction Molecule 1/metabolism , Stromal Interaction Molecule 1/genetics , Humans , Protein Disulfide-Isomerases/metabolism , Protein Disulfide-Isomerases/genetics , Calcium/metabolism , Procollagen-Proline Dioxygenase/metabolism , Procollagen-Proline Dioxygenase/genetics , Neoplasm Proteins/metabolism , Neoplasm Proteins/genetics , Endoplasmic Reticulum/metabolism , HEK293 Cells , Protein Binding , Calcium Signaling , Stromal Interaction Molecule 2/metabolism , Stromal Interaction Molecule 2/genetics
9.
Clin Immunol ; 265: 110306, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38977117

ABSTRACT

Store-operated calcium entry (SOCE) plays a crucial role in maintaining cellular calcium homeostasis. This mechanism involves proteins, such as stromal interaction molecule 1 (STIM1) and ORAI1. Mutations in the genes encoding these proteins, especially STIM1, can lead to various diseases, including CRAC channelopathies associated with severe combined immunodeficiency. Herein, we describe a novel homozygous mutation, NM_003156 c.792-3C > G, in STIM1 in a patient with a clinical profile of CRAC channelopathy, including immune system deficiencies and muscle weakness. Functional analyses revealed three distinct spliced forms in the patient cells: wild-type, exon 7 skipping, and intronic retention. Calcium influx analysis revealed impaired SOCE in the patient cells, indicating a loss of STIM1 function. We developed an antisense oligonucleotide treatment that improves STIM1 splicing and highlighted its potential as a therapeutic approach. Our findings provide insights into the complex effects of STIM1 mutations and shed light on the multifaceted clinical presentation of the patient.


Subject(s)
Calcium , Mutation , Neoplasm Proteins , Stromal Interaction Molecule 1 , Humans , Stromal Interaction Molecule 1/genetics , Stromal Interaction Molecule 1/metabolism , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Calcium/metabolism , Channelopathies/genetics , Male , Calcium Release Activated Calcium Channels/genetics , Calcium Release Activated Calcium Channels/metabolism , Female , Severe Combined Immunodeficiency/genetics , ORAI1 Protein/genetics , ORAI1 Protein/metabolism
10.
Eur J Pharmacol ; 979: 176832, 2024 Sep 15.
Article in English | MEDLINE | ID: mdl-39038639

ABSTRACT

The contractile function of vascular smooth muscle cells (VSMCs) typically undergoes significant changes with advancing age, leading to severe vascular aging-related diseases. The precise role and mechanism of stromal interaction molecule-1 (STIM1) in age-mediated Ca2+ signaling and vasocontraction remain unclear. The connection between STIM1 and age-related vascular dysfunction was investigated using a multi-myograph system, immunohistochemical analysis, protein blotting, and SA-ß-gal staining. Results showed that vasoconstrictor responses in the thoracic aorta, intrarenal artery, and coronary artery decreased with age. STIM1 knockdown in the intrarenal and coronary arteries reduced vascular tone in young mice, while no change was observed in the thoracic aorta. A significant reduction in vascular tone occurred in the STIM1 knockout group with nifedipine. In the thoracic aorta, vasoconstriction significantly decreased with age following the use of nifedipine and thapsigargin and almost disappeared after STIM1 knockdown. The proportion of senescent VSMCs increased significantly in aged mice and further increased in sm-STIM1 KO aged mice. Moreover, the expression of senescence markers p21, p16, and IL-6 significantly increased with age, with p21 expression further increased in the STIM1 knockdown aged group, but not p16 or IL-6. These findings indicate that different arteries exhibit distinct organ-specific features and that STIM1 downregulation may contribute to age-related vasoconstrictive dysfunction through activation of the p21 pathway.


Subject(s)
Aging , Coronary Vessels , Down-Regulation , Stromal Interaction Molecule 1 , Vasoconstriction , Animals , Stromal Interaction Molecule 1/metabolism , Stromal Interaction Molecule 1/genetics , Vasoconstriction/drug effects , Mice , Coronary Vessels/metabolism , Coronary Vessels/physiopathology , Aging/metabolism , Male , Mice, Knockout , Mice, Inbred C57BL , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/drug effects , Renal Artery/metabolism , Cellular Senescence/drug effects , Interleukin-6/metabolism , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/drug effects , Aorta/metabolism , Aorta/drug effects
11.
Neurotoxicology ; 103: 134-145, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38901802

ABSTRACT

Methamphetamine (METH) is a widely abused amphetamine-type psychoactive drug that causes serious health problems. Previous studies have demonstrated that METH can induce neuron autophagy and apoptosis in vivo and in vitro. However, the molecular mechanisms underlying METH-induced neuron autophagy and apoptosis remain poorly understood. Stromal interacting molecule 1 (STIM1) was hypothesized to be involved in METH-induced neuron autophagy and apoptosis. Therefore, the expression of STIM1 protein was measured and the effect of blocking STIM1 expression with siRNA was investigated in cultured neuronal cells, and the hippocampus and striatum of mice exposed to METH. Furthermore, intracellular calcium concentration and endoplasmic reticulum (ER) stress-related proteins were determined in vitro and in vivo in cells treated with METH. The results suggested that STIM1 mediates METH-induced neuron autophagy by activating the p-Akt/p-mTOR pathway. METH exposure also resulted in increased expression of Orai1, which was reversed after STIM1 silencing. Moreover, the disruption of intracellular calcium homeostasis induced ER stress and up-regulated the expression of pro-apoptotic protein CCAAT/enhancer-binding protein homologous protein (CHOP), resulting in classic mitochondria apoptosis. METH exposure can cause neuronal autophagy and apoptosis by increasing the expression of STIM1 protein; thus, STIM1 may be a potential gene target for therapeutics in METH-caused neurotoxicity.


Subject(s)
Apoptosis , Autophagy , Endoplasmic Reticulum Stress , Methamphetamine , Neurons , Stromal Interaction Molecule 1 , Methamphetamine/toxicity , Animals , Stromal Interaction Molecule 1/metabolism , Stromal Interaction Molecule 1/genetics , Autophagy/drug effects , Apoptosis/drug effects , Neurons/drug effects , Neurons/pathology , Neurons/metabolism , Endoplasmic Reticulum Stress/drug effects , Endoplasmic Reticulum Stress/physiology , Mice , Mice, Inbred C57BL , Central Nervous System Stimulants/toxicity , Calcium/metabolism , Cells, Cultured , Male , Hippocampus/drug effects , Hippocampus/metabolism , Hippocampus/pathology , ORAI1 Protein/metabolism , ORAI1 Protein/genetics , TOR Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Transcription Factor CHOP/metabolism , Transcription Factor CHOP/genetics , Corpus Striatum/drug effects , Corpus Striatum/metabolism , Corpus Striatum/pathology
12.
Article in English | MEDLINE | ID: mdl-38918943

ABSTRACT

INTRODUCTION: This study explores the immunogenetic associations of human leukocyte antigens (HLA) and the calcium release-activated calcium modulator 1 (ORAI1) and stromal interaction molecule 1 (STIM1) genes in HIV-1‒positive patients with HIV-related skin disorders. METHODS: This study assessed the distribution of variants of HLA class II alleles and expression levels of ORAI1 and STIM1 genes in the blood between HIV-1‒positive patients with HIV-related skin disorders and the control group with no HIV within the Latvian population. RESULTS: The research group comprised 115 HIV-1‒positive patients with HIV-related skin disorders, and the control group included 80 healthy individuals. Risk alleles (HLA- DQB1*02:01-0301 and HLA-DQA1*01:01-0501) and protective alleles (HLA-DRB1*07-13, DRB1*01-13, DRB1*04-11, and HLA-DQA1*05:01-0501) showed statistical significance in the groups. In 38 out of 115 patients, higher expression levels of ORAI1 and STIM1 genes were detected in the blood at the beginning of treatment. A significantly higher level of the microribonucleic acid (mRNA) ORAI1 gene was also found in the control group. CONCLUSIONS: The results demonstrate that HLA class II alleles are associated with a trend toward risk/protection concerning HIV-related skin disorders in HIV-1‒positive patients. It was also shown that a low level of ORAI1 mRNA and the risk allele HLA-DQB1*0201-0301 were simultaneously present in the research group.


Subject(s)
HIV Infections , ORAI1 Protein , Polymorphism, Genetic , Stromal Interaction Molecule 1 , Humans , ORAI1 Protein/genetics , Male , Latvia , Female , Stromal Interaction Molecule 1/genetics , Adult , HIV Infections/genetics , Middle Aged , Skin Diseases/genetics , Case-Control Studies , Neoplasm Proteins/genetics , Genetic Predisposition to Disease
13.
Sci Rep ; 14(1): 11243, 2024 05 16.
Article in English | MEDLINE | ID: mdl-38755179

ABSTRACT

Immune thrombocytopenia (ITP) is an autoimmune disease caused by T-cell dysfunction. Recently, several studies have shown that a disturbed Th17/Treg balance contributes to the development of ITP. MicroRNAs (miRNAs) are small noncoding RNA moleculesthat posttranscriptionally regulate gene expression. Emerging evidences have demonstrated that miRNAs play an important role in regulating the Th17/Treg balance. In the present study, we found that miR-641 was upregulated in ITP patients. In primary T cells, overexpression of miR-641 could cause downregulation of its target genes STIM1 and SATB1, thus inducing a Th17 (upregulated)/Treg (downregulated) imbalance. Inhibition of miR-641 by a miR-641 sponge in primary T cells of ITP patients or by antagomiR-641 in an ITP murine model could cause upregulation of STIM1 and SATB1, thus restoring Th17/Treg homeostasis. These results suggested that the miR-641-STIM/SATB1 axis plays an important role in regulating the Th17/Treg balance in ITP.


Subject(s)
Matrix Attachment Region Binding Proteins , MicroRNAs , Purpura, Thrombocytopenic, Idiopathic , Stromal Interaction Molecule 1 , T-Lymphocytes, Regulatory , Th17 Cells , Adult , Animals , Female , Humans , Male , Mice , Middle Aged , Disease Models, Animal , Gene Expression Regulation , Matrix Attachment Region Binding Proteins/genetics , Matrix Attachment Region Binding Proteins/metabolism , MicroRNAs/genetics , MicroRNAs/metabolism , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Purpura, Thrombocytopenic, Idiopathic/immunology , Purpura, Thrombocytopenic, Idiopathic/genetics , Purpura, Thrombocytopenic, Idiopathic/metabolism , Stromal Interaction Molecule 1/genetics , Stromal Interaction Molecule 1/metabolism , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Th17 Cells/immunology , Th17 Cells/metabolism
14.
Am J Physiol Lung Cell Mol Physiol ; 327(2): L150-L159, 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-38771147

ABSTRACT

Alteration in the normal mechanical forces of breathing can contribute to changes in contractility and remodeling characteristic of airway diseases, but the mechanisms that mediate these effects in airway cells are still under investigation. Airway smooth muscle (ASM) cells contribute to both contractility and extracellular matrix (ECM) remodeling. In this study, we explored ASM mechanisms activated by mechanical stretch, focusing on mechanosensitive piezo channels and the key Ca2+ regulatory protein stromal interaction molecule 1 (STIM1). Expression of Ca2+ regulatory proteins, including STIM1, Orai1, and caveolin-1, mechanosensitive ion channels Piezo-1 and Piezo-2, and NLRP3 inflammasomes were upregulated by 10% static stretch superimposed on 5% cyclic stretch. These effects were blunted by STIM1 siRNA. Histamine-induced [Ca2+]i responses and inflammasome activation were similarly blunted by STIM1 knockdown. These data show that the effects of mechanical stretch in human ASM cells are mediated through STIM1, which activates multiple pathways, including Piezo channels and the inflammasome, leading to potential downstream changes in contractility and ECM remodeling.NEW & NOTEWORTHY Mechanical forces on the airway can contribute to altered contractility and remodeling in airway diseases, but the mechanisms are not clearly understood. Using human airway smooth muscle cells exposed to cyclic forces with static stretch to mimic breathing and static pressure, we found that the effects of stretch are mediated through STIM1, resulting in the activation of multiple pathways, including Piezo channels and the inflammasome, with potential downstream influences on contractility and remodeling.


Subject(s)
Myocytes, Smooth Muscle , Stromal Interaction Molecule 1 , Humans , Stromal Interaction Molecule 1/metabolism , Stromal Interaction Molecule 1/genetics , Myocytes, Smooth Muscle/metabolism , Neoplasm Proteins/metabolism , Neoplasm Proteins/genetics , Inflammasomes/metabolism , Stress, Mechanical , Mechanotransduction, Cellular , Muscle, Smooth/metabolism , Ion Channels/metabolism , Caveolin 1/metabolism , Caveolin 1/genetics , Signal Transduction , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , Calcium/metabolism , Cells, Cultured , Muscle Contraction/physiology , Airway Remodeling/physiology , ORAI1 Protein/metabolism , ORAI1 Protein/genetics
15.
J Biol Chem ; 300(7): 107422, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38815866

ABSTRACT

Infiltration of monocyte-derived cells to sites of infection and injury is greater in males than in females, due in part, to increased chemotaxis, the process of directed cell movement toward a chemical signal. The mechanisms governing sexual dimorphism in chemotaxis are not known. We hypothesized a role for the store-operated calcium entry (SOCE) pathway in regulating chemotaxis by modulating leading and trailing edge membrane dynamics. We measured the chemotactic response of bone marrow-derived macrophages migrating toward complement component 5a (C5a). Chemotactic ability was dependent on sex and inflammatory phenotype (M0, M1, and M2), and correlated with SOCE. Notably, females exhibited a significantly lower magnitude of SOCE than males. When we knocked out the SOCE gene, stromal interaction molecule 1 (STIM1), it eliminated SOCE and equalized chemotaxis across both sexes. Analysis of membrane dynamics at the leading and trailing edges showed that STIM1 influences chemotaxis by facilitating retraction of the trailing edge. Using BTP2 to pharmacologically inhibit SOCE mirrored the effects of STIM1 knockout, demonstrating a central role of STIM/Orai-mediated calcium signaling. Importantly, by monitoring the recruitment of adoptively transferred monocytes in an in vivo model of peritonitis, we show that increased infiltration of male monocytes during infection is dependent on STIM1. These data support a model in which STIM1-dependent SOCE is necessary and sufficient for mediating the sex difference in monocyte recruitment and macrophage chemotactic ability by regulating trailing edge dynamics.


Subject(s)
Calcium , Chemotaxis , Macrophages , Monocytes , Stromal Interaction Molecule 1 , Animals , Female , Male , Mice , Calcium/metabolism , Calcium Signaling , Macrophages/metabolism , Mice, Inbred C57BL , Mice, Knockout , Monocytes/metabolism , Sex Characteristics , Stromal Interaction Molecule 1/metabolism , Stromal Interaction Molecule 1/genetics
16.
Proc Natl Acad Sci U S A ; 121(21): e2318874121, 2024 May 21.
Article in English | MEDLINE | ID: mdl-38753510

ABSTRACT

The single-pass transmembrane protein Stromal Interaction Molecule 1 (STIM1), located in the endoplasmic reticulum (ER) membrane, possesses two main functions: It senses the ER-Ca2+ concentration and directly binds to the store-operated Ca2+ channel Orai1 for its activation when Ca2+ recedes. At high resting ER-Ca2+ concentration, the ER-luminal STIM1 domain is kept monomeric but undergoes di/multimerization once stores are depleted. Luminal STIM1 multimerization is essential to unleash the STIM C-terminal binding site for Orai1 channels. However, structural basis of the luminal association sites has so far been elusive. Here, we employed molecular dynamics (MD) simulations and identified two essential di/multimerization segments, the α7 and the adjacent region near the α9-helix in the sterile alpha motif (SAM) domain. Based on MD results, we targeted the two STIM1 SAM domains by engineering point mutations. These mutations interfered with higher-order multimerization of ER-luminal fragments in biochemical assays and puncta formation in live-cell experiments upon Ca2+ store depletion. The STIM1 multimerization impeded mutants significantly reduced Ca2+ entry via Orai1, decreasing the Ca2+ oscillation frequency as well as store-operated Ca2+ entry. Combination of the ER-luminal STIM1 multimerization mutations with gain of function mutations and coexpression of Orai1 partially ameliorated functional defects. Our data point to a hydrophobicity-driven binding within the ER-luminal STIM1 multimer that needs to switch between resting monomeric and activated multimeric state. Altogether, these data reveal that interactions between SAM domains of STIM1 monomers are critical for multimerization and activation of the protein.


Subject(s)
Neoplasm Proteins , Protein Multimerization , Stromal Interaction Molecule 1 , Humans , Binding Sites , Calcium/metabolism , Endoplasmic Reticulum/metabolism , HEK293 Cells , Molecular Dynamics Simulation , Neoplasm Proteins/metabolism , Neoplasm Proteins/genetics , Neoplasm Proteins/chemistry , ORAI1 Protein/metabolism , ORAI1 Protein/genetics , ORAI1 Protein/chemistry , Protein Binding , Protein Domains , Stromal Interaction Molecule 1/metabolism , Stromal Interaction Molecule 1/genetics , Stromal Interaction Molecule 1/chemistry
17.
J Clin Immunol ; 44(4): 94, 2024 Apr 05.
Article in English | MEDLINE | ID: mdl-38578569

ABSTRACT

PURPOSE: Deficiency of stromal interaction molecule 1 (STIM1) results in combined immunodeficiency accompanied by extra-immunological findings like enamel defects and myopathy. We here studied a patient with a STIM1 loss-of-function mutation who presented with severe lymphoproliferation. We sought to explore the efficacy of the mTOR inhibitor rapamycin in controlling disease manifestations and reversing aberrant T-cell subsets and functions, which has never been used previously in this disorder. METHODS: Clinical findings of the patient were collected over time. We performed immunological evaluations before and after initiation of rapamycin treatment, including detailed lymphocyte subset analyses, alterations in frequencies of circulating T follicular helper (cTFH) and regulatory T (Treg) cells and their subtypes as well as T cell activation and proliferation capacities. RESULTS: A novel homozygous exon 2 deletion in STIM1 was detected in a 3-year-old girl with severe lymphoproliferation, recurrent infections, myopathy, iris hypoplasia, and enamel hypoplasia. Lymphoproliferation was associated with severe T-cell infiltrates. The deletion resulted in a complete loss of protein expression, associated with a lack of store-operated calcium entry response, defective T-cell activation, proliferation, and cytokine production. Interestingly, patient blood contained fewer cTFH and increased circulating follicular regulatory (cTFR) cells. Abnormal skewing towards TH2-like responses in certain T-cell subpopulations like cTFH, non-cTFH memory T-helper, and Treg cells was associated with increased eosinophil numbers and serum IgE levels. Treatment with rapamycin controlled lymphoproliferation, improved T-cell activation and proliferation capacities, reversed T-cell responses, and repressed high IgE levels and eosinophilia. CONCLUSIONS: This study enhances our understanding of STIM1 deficiency by uncovering additional abnormal T-cell responses, and reveals for the first time the potential therapeutic utility of rapamycin for this disorder.


Subject(s)
Muscular Diseases , Sirolimus , Female , Humans , Child, Preschool , Stromal Interaction Molecule 1/genetics , T-Lymphocyte Subsets , Immunoglobulin E , Neoplasm Proteins
18.
Ann Clin Lab Sci ; 54(1): 17-25, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38514055

ABSTRACT

OBJECTIVE: Diabetic cardiomyopathy (DCM) is the most common cardiovascular complication of type 2 diabetes mellitus (T2DM). Patients affected with DCM face a notably higher risk of progressing to congestive heart failure compared to other populations. Myocardial hypertrophy, a clearly confirmed pathological change in DCM, plays an important role in the development of DCM, with abnormal Ca2+ homeostasis serving as the key signal to induce myocardial hypertrophy. Therefore, investigating the mechanism of Ca2+ transport is of great significance for the prevention and treatment of myocardial hypertrophy in T2DM. METHODS: The rats included in the experiment were divided into wild type (WT) group and T2DM group. The T2DM rat model was established by feeding the rats with high-fat and high-sugar diets for three months combined with low dose of streptozotocin (100mg/kg). Afterwards, primary rat cardiomyocytes were isolated and cultured, and cardiomyocyte hypertrophy was induced through high-glucose treatment. Subsequently, mechanistic investigations were carried out through transfection with si-STIM1 and oe-STIM1. Western blot (WB) was used to detect the expression of the STIM1, Orai1 and p-CaMKII. qRT-PCR was used to detect mRNA levels of myocardial hypertrophy marker proteins. Cell surface area was detected using TRITC-Phalloidin staining, and intracellular Ca2+ concentration in cardiomyocytes was measured using Fluo-4 fluorescence staining. RESULTS: Through animal experiments, an upregulation of Orai1 and STIM1 was revealed in the rat model of myocardial hypertrophy induced by T2DM. Meanwhile, through cell experiments, it was found that in high glucose (HG)-induced hypertrophic cardiomyocytes, the expression of STIM1, Orai1, and p-CaMKII was upregulated, along with increased levels of store-operated Ca2+ entry (SOCE) and abnormal Ca2+ homeostasis. However, when STIM1 was downregulated in HG-induced cardiomyocytes, SOCE levels decreased and p-CaMKII was downregulated, resulting in an improvement in myocardial hypertrophy. To further elucidate the mechanism of action involving SOCE and CaMKII in T2DM-induced myocardial hypertrophy, high-glucose cardiomyocytes were respectively treated with BTP2 (SOCE blocker) and KN-93 (CaMKII inhibitor), and the results showed that STIM1 can mediate SOCE, thereby affecting the phosphorylation level of CaMKII and improving cardiomyocyte hypertrophy. CONCLUSION: STIM1/Orai1-mediated SOCE regulates p-CaMKII levels, thereby inducing myocardial hypertrophy in T2DM.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinase Type 2 , Calcium , Cardiomegaly , Diabetes Mellitus, Type 2 , Diabetic Cardiomyopathies , Glucose , ORAI1 Protein , Stromal Interaction Molecule 1 , Animals , Rats , Calcium/metabolism , Calcium-Calmodulin-Dependent Protein Kinase Type 2/genetics , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Cardiomegaly/etiology , Cardiomegaly/metabolism , Diabetes Mellitus, Type 2/complications , Glucose/metabolism , Glucose/pharmacology , ORAI1 Protein/genetics , ORAI1 Protein/metabolism , Stromal Interaction Molecule 1/genetics , Stromal Interaction Molecule 1/metabolism , Up-Regulation , Diabetic Cardiomyopathies/complications , Rats, Sprague-Dawley , Male
19.
Cell Calcium ; 119: 102871, 2024 May.
Article in English | MEDLINE | ID: mdl-38537434

ABSTRACT

The stromal interaction molecules (STIMs) are the sarcoplasmic reticulum (SR) Ca2+ sensors that trigger store-operated Ca2+ entry (SOCE) in a variety of cell types. While STIM1 isoform has been the focus of the research in cardiac pathophysiology, the function of the homolog STIM2 remains unknown. Using Ca2+ imaging and patch-clamp techniques, we showed that knockdown (KD) of STIM2 by siRNAs increased SOCE and the ISOC current in neonatal rat ventricular cardiomyocytes (NRVMs). Within this cardiomyocyte model, we identified the transcript expression of Stim2.1 and Stim2.2 splice variants, with predominance for Stim2.2. Using conventional and super-resolution confocal microscopy (STED), we found that exogenous STIM2.1 and STIM2.2 formed pre-clusters with a reticular organization at rest. Following SR Ca2+ store depletion, some STIM2.1 and STIM2.2 clusters were translocated to SR-plasma membrane (PM) junctions and co-localized with Orai1. The overexpression strategy revealed that STIM2.1 suppressed Orai1-mediated SOCE and the ISOC current while STIM2.2 enhanced SOCE. STIM2.2-enhanced SOCE was also dependent on TRPC1 and TRPC4. Even if STIM2 KD or splice variants overexpression did not affect cytosolic Ca2+ cycling, we observed, using Rhod-2/AM Ca2+ imaging, that Orai1 inhibition or STIM2.1 overexpression abolished the mitochondrial Ca2+ (mCa2+) uptake, as opposed to STIM2 KD. We also found that STIM2 was present in the mitochondria-associated endoplasmic reticulum (ER) membranes (MAMs) by interacting with the inositol trisphosphate receptors (IP3Rs), voltage-dependent anion channel (VDAC), mitochondrial Ca2+ uniporter (MCU), and mitofusin-2 (MNF2). Our results suggested that, in NRVMs, STIM2.1 constitutes the predominant functional variant that negatively regulates Orai1-generated SOCE. It participates in the control of mCa2+ uptake capacity possibly via the STIM2-IP3Rs-VDAC-MCU and MNF2 complex.


Subject(s)
Calcium , Myocytes, Cardiac , Stromal Interaction Molecule 1 , Animals , Rats , Biological Transport , Calcium/metabolism , Calcium Channels/metabolism , Calcium Signaling , Homeostasis , Mitochondria/metabolism , Myocytes, Cardiac/metabolism , ORAI1 Protein/metabolism , Stromal Interaction Molecule 1/genetics , Stromal Interaction Molecule 1/metabolism
20.
J Clin Invest ; 134(7)2024 Feb 01.
Article in English | MEDLINE | ID: mdl-38300705

ABSTRACT

Stromal interaction molecule 1 (STIM1) is a Ca2+ sensor located in the sarcoplasmic reticulum (SR) of skeletal muscle, where it is best known for its role in store-operated Ca2+ entry (SOCE). Genetic syndromes resulting from STIM1 mutations are recognized as a cause of muscle weakness and atrophy. Here, we focused on a gain-of-function mutation that occurs in humans and mice (STIM1+/D84G mice), in which muscles exhibited constitutive SOCE. Unexpectedly, this constitutive SOCE did not affect global Ca2+ transients, SR Ca2+ content, or excitation-contraction coupling (ECC) and was therefore unlikely to underlie the reduced muscle mass and weakness observed in these mice. Instead, we demonstrate that the presence of D84G STIM1 in the nuclear envelope of STIM1+/D84G muscle disrupted nuclear-cytosolic coupling, causing severe derangement in nuclear architecture, DNA damage, and altered lamina A-associated gene expression. Functionally, we found that D84G STIM1 reduced the transfer of Ca2+ from the cytosol to the nucleus in myoblasts, resulting in a reduction of [Ca2+]N. Taken together, we propose a novel role for STIM1 in the nuclear envelope that links Ca2+ signaling to nuclear stability in skeletal muscle.


Subject(s)
Muscle Weakness , Nuclear Envelope , Stromal Interaction Molecule 1 , Animals , Humans , Mice , Calcium/metabolism , Calcium Signaling , Muscle Weakness/genetics , Muscle Weakness/metabolism , Muscle, Skeletal/metabolism , Mutation , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Nuclear Envelope/genetics , Nuclear Envelope/metabolism , ORAI1 Protein/genetics , Stromal Interaction Molecule 1/genetics , Stromal Interaction Molecule 1/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL