Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 754
Filter
1.
BMC Nephrol ; 25(1): 161, 2024 May 10.
Article in English | MEDLINE | ID: mdl-38730296

ABSTRACT

BACKGROUND: Previous observational studies have reported that systemic inflammatory regulators are related to the development of chronic kidney disease (CKD); however, whether these associations are causal remains unclear. The current study aimed to investigate the potential causal relationships between systemic inflammatory regulators and CKD and kidney function. METHOD: We performed bidirectional two-sample Mendelian randomization (MR) analyses to infer the underlying causal associations between 41 systemic inflammatory regulators and CKD and kidney function. The inverse-variance weighting (IVW) test was used as the primary analysis method. In addition, sensitivity analyses were executed via the Mendelian randomization pleiotropy residual sum and outlier (MR-PRESSO) test and the weighted median test. RESULTS: The findings revealed 12 suggestive associations between 11 genetically predicted systemic inflammatory regulators and CKD or kidney function in the forward analyses, including 4 for CKD, 3 for blood urea nitrogen (BUN), 4 for eGFRcrea and 1 for eGFRcys. In the other direction, we identified 6 significant causal associations, including CKD with granulocyte-colony stimulating factor (GCSF) (IVW ß = 0.145; 95% CI, 0.042 to 0.248; P = 0.006), CKD with stem cell factor (SCF) (IVW ß = 0.228; 95% CI, 0.133 to 0.323; P = 2.40 × 10- 6), eGFRcrea with SCF (IVW ß =-2.90; 95% CI, -3.934 to -1.867; P = 3.76 × 10- 8), eGFRcys with GCSF (IVW ß =-1.382; 95% CI, -2.404 to -0.361; P = 0.008), eGFRcys with interferon gamma (IFNg) (IVW ß =-1.339; 95% CI, -2.313 to -0.366; P = 0.007) and eGFRcys with vascular endothelial growth factor (VEGF) (IVW ß =-1.709; 95% CI, -2.720 to -0.699; P = 9.13 × 10- 4). CONCLUSIONS: Our findings support causal links between systemic inflammatory regulators and CKD or kidney function both in the forward and reverse MR analyses.


Subject(s)
Mendelian Randomization Analysis , Renal Insufficiency, Chronic , Humans , Renal Insufficiency, Chronic/genetics , Renal Insufficiency, Chronic/blood , Glomerular Filtration Rate , Inflammation/genetics , Granulocyte Colony-Stimulating Factor/blood , Stem Cell Factor/genetics , Stem Cell Factor/blood , Kidney/metabolism , Kidney/physiopathology , Blood Urea Nitrogen
2.
Cancer Lett ; 589: 216795, 2024 May 01.
Article in English | MEDLINE | ID: mdl-38556106

ABSTRACT

The immune microenvironment constructed by tumor-infiltrating immune cells and the molecular phenotype defined by hormone receptors (HRs) have been implicated as decisive factors in the regulation of breast cancer (BC) progression. Here, we found that the infiltration of mast cells (MCs) informed impaired prognoses in HR(+) BC but predicted improved prognoses in HR(-) BC. However, molecular features of MCs in different BC remain unclear. We next discovered that HR(-) BC cells were prone to apoptosis under the stimulation of MCs, whereas HR(+) BC cells exerted anti-apoptotic effects. Mechanistically, in HR(+) BC, the KIT ligand (KITLG), a major mast cell growth factor in recruiting and activating MCs, could be transcriptionally upregulated by the progesterone receptor (PGR), and elevate the production of MC-derived granulin (GRN). GRN attenuates TNFα-induced apoptosis in BC cells by competitively binding to TNFR1. Furthermore, disruption of PGR-KITLG signaling by knocking down PGR or using the specific KITLG-cKIT inhibitor iSCK03 potently enhanced the sensitivity of HR(+) BC cells to MC-induced apoptosis and exerted anti-tumor activity. Collectively, these results demonstrate that PGR-KITLG signaling in BC cells preferentially induces GRN expression in MCs to exert anti-apoptotic effects, with potential value in developing precision medicine approaches for diagnosis and treatment.


Subject(s)
Breast Neoplasms , Stem Cell Factor , Humans , Female , Stem Cell Factor/genetics , Stem Cell Factor/metabolism , Mast Cells/pathology , Breast Neoplasms/pathology , Feedback , Apoptosis , Tumor Microenvironment
3.
Cardiovasc Res ; 120(7): 745-755, 2024 May 29.
Article in English | MEDLINE | ID: mdl-38507654

ABSTRACT

AIMS: In hypoxia, endothelial cells (ECs) proliferate, migrate, and form new vasculature in a process called angiogenesis. Recent studies have suggested that ECs rely on glycolysis to meet metabolic needs for angiogenesis in ischaemic tissues, and several studies have investigated the molecular mechanisms integrating angiogenesis and endothelial metabolism. Here, we investigated the role of stem cell factor (SCF) and its receptor, cKIT, in regulating endothelial glycolysis during hypoxia-driven angiogenesis. METHODS AND RESULTS: SCF and cKIT signalling increased the glucose uptake, lactate production, and glycolysis in human ECs under hypoxia. Mechanistically, SCF and cKIT signalling enhanced the expression of genes encoding glucose transporter 1 (GLUT1) and glycolytic enzymes via Akt- and ERK1/2-dependent increased translation of hypoxia inducible factor 1A (HIF1A). In hypoxic conditions, reduction of glycolysis and HIF-1α expression using chemical inhibitors significantly reduced the SCF-induced in vitro angiogenesis in human ECs. Compared with normal mice, mice with oxygen-induced retinopathy (OIR), characterized by ischaemia-driven pathological retinal neovascularization, displayed increased levels of SCF, cKIT, HIF-1α, GLUT1, and glycolytic enzymes in the retina. Moreover, cKIT-positive neovessels in the retina of mice with OIR showed elevated expression of GLUT1 and glycolytic enzymes. Further, blocking SCF and cKIT signalling using anti-SCF neutralizing IgG and cKIT mutant mice significantly reduced the expression of HIF-1α, GLUT1, and glycolytic enzymes and decreased the pathological neovascularization in the retina of mice with OIR. CONCLUSION: We demonstrated that SCF and cKIT signalling regulate angiogenesis by controlling endothelial glycolysis in hypoxia and elucidated the SCF/cKIT/HIF-1α axis as a novel metabolic regulation pathway during hypoxia-driven pathological angiogenesis.


Subject(s)
Cell Hypoxia , Glucose Transporter Type 1 , Glycolysis , Hypoxia-Inducible Factor 1, alpha Subunit , Proto-Oncogene Proteins c-kit , Signal Transduction , Stem Cell Factor , Animals , Humans , Stem Cell Factor/metabolism , Stem Cell Factor/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Glucose Transporter Type 1/metabolism , Glucose Transporter Type 1/genetics , Proto-Oncogene Proteins c-kit/metabolism , Proto-Oncogene Proteins c-kit/genetics , Human Umbilical Vein Endothelial Cells/metabolism , Mice, Inbred C57BL , Endothelial Cells/metabolism , Endothelial Cells/pathology , Retinal Neovascularization/metabolism , Retinal Neovascularization/pathology , Retinal Neovascularization/genetics , Mice , Neovascularization, Physiologic , Cells, Cultured , Disease Models, Animal , Glucose/metabolism
4.
Exp Hematol ; 128: 38-47, 2023 12.
Article in English | MEDLINE | ID: mdl-37722652

ABSTRACT

Sterile α-motif domain-14 (Samd14) protein expression increases the regenerative capacity of the erythroid system. Samd14 is transcriptionally upregulated and promotes cell signaling via the receptor tyrosine kinase Kit in a critical window of acute erythroid regeneration. We generated a hematopoietic-specific conditional Samd14 knockout mouse model (Samd14-CKO) to study the role of Samd14 in hematopoiesis. The Samd14-CKO mouse was viable and exhibited no steady-state hematopoietic phenotype. Samd14-CKO mice were hypersensitive to 5-fluorouracil, resulting in more severe anemia during recovery and impaired erythroid progenitor colony formation. Ex vivo, Samd14-CKO hematopoietic progenitors were defective in their ability to form mast cells. Samd14-CKO mast cells exhibited altered Kit/stem cell factor (SCF), IL-3/IL-3R signaling, and less granularity than Samd14-FL/FL cells. Our findings indicate that Samd14 promotes both erythroid and mast cell functions. The Samd14-CKO mouse phenotype exhibits striking similarities to the KitW/W-v mice, which carry Kit mutations resulting in reduced tyrosine kinase-dependent signaling, causing mast cell and erythroid abnormalities. The Samd14-CKO mouse model is a new tool for studying hematologic pathologies involving Kit signaling.


Subject(s)
Anemia , Cytoskeletal Proteins , Hematopoiesis , Animals , Mice , Anemia/metabolism , Hematopoiesis/genetics , Proteins , Proto-Oncogene Proteins c-kit/genetics , Proto-Oncogene Proteins c-kit/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Signal Transduction , Stem Cell Factor/genetics , Cytoskeletal Proteins/genetics , Cytoskeletal Proteins/metabolism
5.
J Cell Mol Med ; 27(19): 2983-2994, 2023 10.
Article in English | MEDLINE | ID: mdl-37603611

ABSTRACT

Short-chain fatty acid butyrate is produced from the bacterial fermentation of indigestible fiber in the intestinal lumen, and it has been shown to attenuate lung inflammation in murine asthma models. Mast cells (MCs) are initiators of inflammatory response to allergens, and they play an important role in asthma. MC survival and proliferation is regulated by its growth factor stem cell factor (SCF), which acts through the receptor, KIT. It has previously been shown that butyrate attenuates the activation of MCs by allergen stimulation. However, how butyrate mechanistically influences SCF signalling to impact MC function remains unknown. Here, we report that butyrate treatment triggered the modification of MC histones via butyrylation and acetylation, and inhibition of histone deacetylase (HDAC) activity. Further, butyrate treatment caused downregulation of SCF receptor KIT and associated phosphorylation, leading to significant attenuation of SCF-mediated MC proliferation, and pro-inflammatory cytokine secretion. Mechanistically, butyrate inhibited MC function by suppressing KIT and downstream p38 and Erk phosphorylation, and it mediated these effects via modification of histones, acting as an HDAC inhibitor and not via its traditional GPR41 (FFAR3) or GPR43 (FFAR2) butyrate receptors. In agreement, the pharmacological inhibition of Class I HDAC (HDAC1/3) mirrored butyrate's effects, suggesting that butyrate impacts MC function by HDAC1/3 inhibition. Taken together, butyrate epigenetically modifies histones and downregulates the SCF/KIT/p38/Erk signalling axis, leading to the attenuation of MC function, validating its ability to suppress MC-mediated inflammation. Therefore, butyrate supplementations could offer a potential treatment strategy for allergy and asthma via epigenetic alterations in MCs.


Subject(s)
Asthma , Histones , Humans , Mice , Animals , Histones/metabolism , Mast Cells/metabolism , Butyrates/pharmacology , Histone Code , Stem Cell Factor/genetics , Stem Cell Factor/metabolism , Epigenesis, Genetic , Asthma/metabolism
6.
Endocr Relat Cancer ; 30(10)2023 10 01.
Article in English | MEDLINE | ID: mdl-37578265

ABSTRACT

Gastrointestinal stromal tumors (GISTs) are mesenchymal neoplasms, believed to originate from the interstitial cells of Cajal (ICC), often caused by overexpression of tyrosine kinase receptors (TKR) KIT or PDGFRA. Here, we present evidence that the embryonic stem cell factor FOXD3, first identified as 'Genesis' and involved in both gastrointestinal and neural crest cell development, is implicated in GIST pathogenesis; its involvement is investigated both in vitro and in zebrafish and a mouse model of FOXD3 deficiency. Samples from a total of 58 patients with wild-type GISTs were used for molecular analyses, including Sanger sequencing, comparative genomic hybridization, and methylation analysis. Immunohistochemistry and western blot evaluation were used to assess FOXD3 expression. Additionally, we conducted in vitro functional studies in tissue samples and in transfected cells to confirm the pathogenicity of the identified genetic variants. Germline partially inactivating FOXD3 sequence variants (p.R54H and p.Ala88_Gly91del) were found in patients with isolated GISTs. Chromosome 1p loss was the most frequent chromosomal abnormality identified in tumors. In vitro experiments demonstrate the impairment of FOXD3 in the presence of those variants. Animal studies showed disruption of the GI neural network and changes in the number and distribution in the ICC. FOXD3 suppresses KIT expression in human cells; its inactivation led to an increase in ICC in zebrafish, as well as mice, providing evidence for a functional link between FOXD3 defects and KIT overexpression leading to GIST formation.


Subject(s)
Gastrointestinal Neoplasms , Gastrointestinal Stromal Tumors , Humans , Animals , Mice , Gastrointestinal Stromal Tumors/genetics , Zebrafish/genetics , Zebrafish/metabolism , Stem Cell Factor/genetics , Comparative Genomic Hybridization , Proto-Oncogene Proteins c-kit/genetics , Receptor, Platelet-Derived Growth Factor alpha/genetics , Transcription Factors/genetics , Embryonic Stem Cells/chemistry , Embryonic Stem Cells/metabolism , Embryonic Stem Cells/pathology , Mutation , Gastrointestinal Neoplasms/genetics , Forkhead Transcription Factors/genetics
7.
Oncogene ; 42(34): 2578-2588, 2023 08.
Article in English | MEDLINE | ID: mdl-37468679

ABSTRACT

Gastrointestinal stromal tumor (GIST) is the most common human sarcoma and is typically driven by a single mutation in the Kit or PDGFRA receptor. While highly effective, tyrosine kinase inhibitors (TKIs) are not curative. The natural ligand for the Kit receptor is Kit ligand (KitL), which exists in both soluble and membrane-bound forms. While KitL is known to stimulate human GIST cell lines in vitro, we used a genetically engineered mouse model of GIST containing a common human KIT mutation to investigate the intratumoral sources of KitL, importance of KitL during GIST oncogenesis, and contribution of soluble KitL to tumor growth in vivo. We discovered that in addition to tumor cells, endothelia and smooth muscle cells produced KitL in KitV558Δ/+ tumors, even after imatinib therapy. Genetic reduction of total KitL in tumor cells of KitV558Δ/+ mice impaired tumor growth in vivo. Similarly, genetic reduction of tumor cell soluble KitL in KitV558Δ/+ mice decreased tumor size. By RNA sequencing, quantitative PCR, and immunohistochemistry, KitL expression was heterogeneous in human GIST specimens. In particular, PDGFRA-mutant tumors had much higher KitL expression than Kit-mutant tumors, suggesting the benefit of Kit activation in the absence of mutant KIT. Serum KitL was higher in GIST patients with tumors resistant to imatinib and in those with tumors expressing more KitL RNA. Overall, KitL supports the growth of GIST at baseline and after imatinib therapy and remains a potential biomarker and therapeutic target.


Subject(s)
Antineoplastic Agents , Gastrointestinal Stromal Tumors , Humans , Mice , Animals , Imatinib Mesylate/pharmacology , Imatinib Mesylate/therapeutic use , Gastrointestinal Stromal Tumors/drug therapy , Gastrointestinal Stromal Tumors/genetics , Gastrointestinal Stromal Tumors/pathology , Stem Cell Factor/genetics , Stem Cell Factor/pharmacology , Stem Cell Factor/therapeutic use , Pyrimidines/pharmacology , Piperazines/pharmacology , Piperazines/therapeutic use , Benzamides/pharmacology , Drug Resistance, Neoplasm/genetics , Proto-Oncogene Proteins c-kit , Mutation , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use
8.
Genes (Basel) ; 14(5)2023 05 02.
Article in English | MEDLINE | ID: mdl-37239395

ABSTRACT

Breeding ornamental white sika deer is a new notion that can be used to broaden the sika deer industry However, it is very rare for other coat phenotypes to occur, especially white (apart from albinism), due to the genetic stability and homogeneity of its coat color phenotype, making it difficult to breed white sika deer between species. We found a white sika deer and sequenced its whole genome. Then, the clean data obtained were analyzed on the basis of gene frequency, and a cluster of coat color candidate genes containing 92 coat color genes, one SV (structure variation), and five nonsynonymous SNPs (single nucleotide polymorphisms) was located. We also discovered a lack of melanocytes in the skin tissue of the white sika deer through histological examination, initially proving that the white phenotype of sika deer is caused by a 10.099 kb fragment deletion of the SCF gene(stem cell factor). By designing SCF-specific primers to detect genotypes of family members of the white sika deer, and then combining them with their phenotypes, we found that the genotype of the white sika deer is SCF789/SCF789, whereas that of individuals with white patches on their faces is SCF789/SCF1-9. All these results showed that the SCF gene plays an important role in the development of melanocytes in sika deer and is responsible for the appearance of the white coat color. This study reveals the genetic mechanism of the white coat color in sika deer and supplies data as a reference for breeding white ornamental sika deer.


Subject(s)
Deer , Stem Cell Factor , Animals , Stem Cell Factor/genetics , Deer/genetics , Phenotype , Genotype , Gene Frequency
9.
Nat Commun ; 14(1): 2754, 2023 05 13.
Article in English | MEDLINE | ID: mdl-37179330

ABSTRACT

Active thermogenesis in the brown adipose tissue (BAT) facilitating the utilization of lipids and glucose is critical for maintaining body temperature and reducing metabolic diseases, whereas inactive BAT accumulates lipids in brown adipocytes (BAs), leading to BAT whitening. Although cellular crosstalk between endothelial cells (ECs) and adipocytes is essential for the transport and utilization of fatty acid in BAs, the angiocrine roles of ECs mediating this crosstalk remain poorly understood. Using single-nucleus RNA sequencing and knock-out male mice, we demonstrate that stem cell factor (SCF) derived from ECs upregulates gene expressions and protein levels of the enzymes for de novo lipogenesis, and promotes lipid accumulation by activating c-Kit in BAs. In the early phase of lipid accumulation induced by denervation or thermoneutrality, transiently expressed c-Kit on BAs increases the protein levels of the lipogenic enzymes via PI3K and AKT signaling. EC-specific SCF deletion and BA-specific c-Kit deletion attenuate the induction of the lipogenic enzymes and suppress the enlargement of lipid droplets in BAs after denervation or thermoneutrality in male mice. These data provide insight into SCF/c-Kit signaling as a regulator that promotes lipid accumulation through the increase of lipogenic enzymes in BAT when thermogenesis is inhibited.


Subject(s)
Adipocytes, Brown , Hypercholesterolemia , Animals , Male , Mice , Adipocytes, Brown/metabolism , Adipose Tissue, Brown/metabolism , Endothelial Cells/metabolism , Fatty Acids/metabolism , Hypercholesterolemia/metabolism , Lipogenesis/genetics , Mice, Knockout , Receptor Protein-Tyrosine Kinases/metabolism , Stem Cell Factor/genetics , Stem Cell Factor/metabolism , Thermogenesis/genetics , Proto-Oncogene Proteins c-kit
10.
Int J Biol Sci ; 19(6): 1910-1924, 2023.
Article in English | MEDLINE | ID: mdl-37063428

ABSTRACT

The stem cell factor (SCF) binds to c-Kit in endothelial cells, thus activating downstream signaling and angiogenesis. Herein, we examined the role of G protein subunit alpha inhibitory (Gαi) proteins in this process. In MEFs and HUVECs, Gαi1/3 was associated with SCF-activated c-Kit, promoting c-Kit endocytosis, and binding of key adaptor proteins, subsequently transducing downstream signaling. SCF-induced Akt-mTOR and Erk activation was robustly attenuated by Gαi1/3 silencing or knockout (KO), or due to dominant negative mutations but was strengthened substantially following ectopic overexpression of Gαi1/3. SCF-induced HUVEC proliferation, migration, and capillary tube formation were suppressed after Gαi1/3 silencing or KO, or due to dominant negative mutations. In vivo, endothelial knockdown of Gαi1/3 by intravitreous injection of endothelial-specific shRNA adeno-associated virus (AAV) potently reduced SCF-induced signaling and retinal angiogenesis in mice. Moreover, mRNA and protein expressions of SCF increased significantly in the retinal tissues of streptozotocin-induced diabetic retinopathy (DR) mice. SCF silencing, through intravitreous injection of SCF shRNA AAV, inhibited pathological retinal angiogenesis and degeneration of retinal ganglion cells in DR mice. Finally, the expression of SCF and c-Kit increased in proliferative retinal tissues of human patients with proliferative DR. Taken together, Gαi1/3 mediate SCF/c-Kit-activated signaling and angiogenesis.


Subject(s)
Endothelial Cells , Signal Transduction , Animals , Humans , Mice , Adaptor Proteins, Signal Transducing/metabolism , Endothelial Cells/metabolism , Proto-Oncogene Proteins c-kit/genetics , Proto-Oncogene Proteins c-kit/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , RNA, Small Interfering/metabolism , Signal Transduction/genetics , Stem Cell Factor/genetics , Stem Cell Factor/metabolism , GTP-Binding Protein alpha Subunits, Gi-Go/metabolism
11.
Development ; 150(6)2023 03 15.
Article in English | MEDLINE | ID: mdl-36861441

ABSTRACT

Several cell types have been proposed to create the required microenvironment for spermatogenesis. However, expression patterns of the key growth factors produced by these somatic cells have not been systematically studied and no such factor has been conditionally deleted from its primary source(s), raising the question of which cell type(s) are the physiological sources of these growth factors. Here, using single-cell RNA sequencing and a series of fluorescent reporter mice, we found that stem cell factor (Scf), one of the essential growth factors for spermatogenesis, was broadly expressed in testicular stromal cells, including Sertoli, endothelial, Leydig, smooth muscle and Tcf21-CreER+ stromal cells. Both undifferentiated and differentiating spermatogonia were associated with Scf-expressing Sertoli cells in the seminiferous tubule. Conditional deletion of Scf from Sertoli cells, but not any other Scf-expressing cells, blocked the differentiation of spermatogonia, leading to complete male infertility. Conditional overexpression of Scf in Sertoli cells, but not endothelial cells, significantly increased spermatogenesis. Our data reveal the importance of anatomical localization for Sertoli cells in regulating spermatogenesis and that SCF produced specifically by Sertoli cells is essential for spermatogenesis.


Subject(s)
Sertoli Cells , Stem Cell Factor , Male , Animals , Mice , Sertoli Cells/metabolism , Stem Cell Factor/genetics , Stem Cell Factor/metabolism , Spermatogenesis/genetics , Testis/metabolism , Spermatogonia/metabolism
12.
Proc Natl Acad Sci U S A ; 120(13): e2300054120, 2023 03 28.
Article in English | MEDLINE | ID: mdl-36943885

ABSTRACT

The receptor tyrosine kinase KIT and its ligand stem cell factor (SCF) are required for the development of hematopoietic stem cells, germ cells, and other cells. A variety of human cancers, such as acute myeloid leukemia, gastrointestinal stromal tumor, and mast cell leukemia, are driven by somatic gain-of-function KIT mutations. Here, we report cryo electron microscopy (cryo-EM) structural analyses of full-length wild-type and two oncogenic KIT mutants, which show that the overall symmetric arrangement of the extracellular domain of ligand-occupied KIT dimers contains asymmetric D5 homotypic contacts juxtaposing the plasma membrane. Mutational analysis of KIT reveals in D5 region an "Achilles heel" for therapeutic intervention. A ligand-sensitized oncogenic KIT mutant exhibits a more comprehensive and stable D5 asymmetric conformation. A constitutively active ligand-independent oncogenic KIT mutant adopts a V-shaped conformation solely held by D5-mediated contacts. Binding of SCF to this mutant fully restores the conformation of wild-type KIT dimers, including the formation of salt bridges responsible for D4 homotypic contacts and other hallmarks of SCF-induced KIT dimerization. These experiments reveal an unexpected structural plasticity of oncogenic KIT mutants and a therapeutic target in D5.


Subject(s)
Neoplasms , Proto-Oncogene Proteins c-kit , Humans , Proto-Oncogene Proteins c-kit/genetics , Proto-Oncogene Proteins c-kit/metabolism , Ligands , Cryoelectron Microscopy , Receptor Protein-Tyrosine Kinases/metabolism , Stem Cell Factor/genetics , Stem Cell Factor/metabolism , Phosphorylation
13.
Gerontology ; 69(5): 628-640, 2023.
Article in English | MEDLINE | ID: mdl-36720215

ABSTRACT

INTRODUCTION: Youthful blood environment was shown to decelerate the aging process of the kidney and to attenuate senile renal fibrosis in a young-old parabiotic animal model; in addition, we identified a stem cell factor (SCF) that is closely linked with the process. This research was to investigate the effect of youthful blood environment on senile renal interstitial fibrosis and the role of SCF. METHODS: We bred SCF receptor c-Kit gene loss-of-function Wps/Wps mice and established a combination mice model that was subjected to unilateral ureteral obstructive (UUO) and parabiotic surgeries. Parabiotic mice were divided into isochronic parabiotic (young-young [Y-IP] and old-old [O-IP]) and heterochronic parabiotic (young-old [HP]) groups. UUO surgery was performed in one of the parabiotic pairs in the IP group (Y-IPuuo and O-IPuuo) and in the elderly mice in the HP group (O-HPuuo). In order to study the role of SCF/c-kit on renal interstitial fibrosis, UUO surgery was performed in wildtype (WT) and Wps/Wps mice. RESULTS: Fourteen days after UUO surgery, the kidney interstitial fibrosis area, kidney function, and the expressions of SCF/c-Kit, pNF-κB, and fibrosis-related proteins in the O-HPuuo group were significantly lower than those in the Ouuo and O-IPuuo groups. Compared with WT UUO mice, the expressions of pNF-κB and fibrosis-related proteins and the kidney function were all significantly decreased in Wps/Wps UUO mice. CONCLUSION: Youthful blood environment downregulated the expressions of SCF/c-Kit in elderly UUO mice and ameliorated UUO-induced kidney fibrosis and function loss.


Subject(s)
Kidney Diseases , Ureteral Obstruction , Mice , Animals , Stem Cell Factor/genetics , Stem Cell Factor/metabolism , Stem Cell Factor/pharmacology , Ureteral Obstruction/complications , Ureteral Obstruction/metabolism , Ureteral Obstruction/pathology , Kidney Diseases/genetics , Kidney Diseases/metabolism , Kidney/pathology , Fibrosis , Disease Models, Animal
14.
Arterioscler Thromb Vasc Biol ; 43(2): 367-378, 2023 02.
Article in English | MEDLINE | ID: mdl-36579647

ABSTRACT

BACKGROUND: Genome-wide association studies have reported 23 gene loci related to abdominal aortic aneurysm (AAA)-a potentially lethal condition characterized by a weakened dilated vessel wall. This study aimed to identify proteomic signatures and pathways related to these risk loci to better characterize AAA genetic susceptibility. METHODS: Plasma concentrations of 4870 proteins were determined using a DNA aptamer-based array. Linear regression analysis estimated the associations between the 23 risk alleles and plasma protein levels with adjustments for potential confounders in a race-stratified analysis of 1671 Black and 7241 White participants. Significant proteins were then evaluated for their prediction of clinical AAA (454 AAA events in 11 064 individuals), and those significantly associated with AAA were further interrogated using Mendelian randomization analysis. RESULTS: Risk variants proximal to PSRC1-CELSR2-SORT1, PCIF1-ZNF335-MMP9, RP11-136O12.2/TRIB1, ZNF259/APOA5, IL6R, PCSK9, LPA, and APOE were associated with 118 plasma proteins in Whites and 59 were replicated in Black participants. Novel associations with clinical AAA incidence were observed for kit ligand (HR, 0.59 [95% CI, 0.42-0.82] for top versus first quintiles) and neogenin (HR, 0.64 [95% CI, 0.46-0.88]) over a median 21.2-year follow-up; neogenin was also associated with ultrasound-detected asymptomatic AAA (N=4295; 57 asymptomatic AAA cases). Mendelian randomization inverse variance weighted estimates suggested that AAA risk is promoted by lower levels of kit ligand (OR per SD=0.67; P=1.4×10-5) and neogenin (OR per SD=0.50; P=0.03). CONCLUSIONS: Low levels of neogenin and kit ligand may be novel risk factors for AAA development in potentially causal pathways. These findings provide insights and potential targets to reduce AAA susceptibility.


Subject(s)
Aortic Aneurysm, Abdominal , Proprotein Convertase 9 , Humans , Proprotein Convertase 9/metabolism , Stem Cell Factor/genetics , Genome-Wide Association Study , Proteomics , Aortic Aneurysm, Abdominal/epidemiology , Aortic Aneurysm, Abdominal/genetics , Aortic Aneurysm, Abdominal/metabolism , Transcription Factors/metabolism , Risk Factors , Nuclear Proteins/genetics , Adaptor Proteins, Signal Transducing/genetics , Protein Serine-Threonine Kinases/metabolism , Intracellular Signaling Peptides and Proteins/genetics
15.
Anim Genet ; 53(5): 549-556, 2022 Oct.
Article in English | MEDLINE | ID: mdl-35811453

ABSTRACT

Since domestication, a wide variety of phenotypes including coat color variation has developed in livestock. This variation is mostly based on selective breeding. During the beginning of selective breeding, potential negative consequences did not become immediately evident due to low frequencies of homozygous animals and have been occasionally neglected. However, numerous studies of coat color genetics have been carried out over more than a century and, meanwhile, pleiotropic effects for several coat color genes, including disorders of even lethal impact, were described. Similar coat color phenotypes can often be found across species, caused either by conserved genes or by different genes. Even in the same species, more than one gene could cause the same or similar coat color phenotype. The roan coat color in livestock species is characterized by a mixture of white and colored hair in cattle, pig, sheep, goat, alpaca, and horse. So far, the genetic background of this phenotype is not fully understood, but KIT and its ligand KITLG (MGF) are major candidate genes in livestock species. For some of these species, pleiotropic effects such as subfertility in homozygous roan cattle or homozygous embryonic lethality in certain horse breeds have been described. This review aims to point out the similarities and differences of the roan phenotype across the following livestock species: cattle, pig, sheep, goat, alpaca, and horse; and provides the current state of knowledge on genetic background and pleiotropic effects.


Subject(s)
Camelids, New World , Livestock , Animals , Cattle/genetics , Color , Goats/genetics , Hair Color/genetics , Horses/genetics , Livestock/genetics , Phenotype , Sheep , Stem Cell Factor/genetics , Swine
16.
J Allergy Clin Immunol ; 149(6): 1845-1854, 2022 06.
Article in English | MEDLINE | ID: mdl-35469840

ABSTRACT

The discovery in 1987/1988 and 1990 of the cell surface receptor KIT and its ligand, stem cell factor (SCF), was a critical achievement in efforts to understand the development and function of multiple distinct cell lineages. These include hematopoietic progenitors, melanocytes, germ cells, and mast cells, which all are significantly affected by loss-of-function mutations of KIT or SCF. Such mutations also influence the development and/or function of additional cells, including those in parts of the central nervous system and the interstitial cells of Cajal (which control gut motility). Many other cells can express KIT constitutively or during immune responses, including dendritic cells, eosinophils, type 2 innate lymphoid cells, and taste cells. Yet the biological importance of KIT in many of these cell types largely remains to be determined. We here review the history of work investigating mice with mutations affecting the white spotting locus (which encodes KIT) or the steel locus (which encodes SCF), focusing especially on the influence of such mutations on mast cells. We also briefly review efforts to target the KIT/SCF pathway with anti-SCF or anti-Kit antibodies in mouse models of allergic disorders, parasite immunity, or fibrosis in which mast cells are thought to play significant roles.


Subject(s)
Mast Cells , Proto-Oncogene Proteins c-kit , Animals , Cell Lineage , Humans , Immunity, Innate , Lymphocytes/cytology , Lymphocytes/immunology , Lymphocytes/metabolism , Mast Cells/metabolism , Mice , Proto-Oncogene Proteins c-kit/genetics , Proto-Oncogene Proteins c-kit/metabolism , Stem Cell Factor/genetics , Stem Cell Factor/metabolism
17.
J Biomed Nanotechnol ; 18(1): 218-224, 2022 Jan 01.
Article in English | MEDLINE | ID: mdl-35180915

ABSTRACT

MicroRNA (miRNA)-124 inhibitor may enhance cerebral blood vessel formation in lacunar cerebral infarction (LCI) and mesoporous silica nanoparticles are highlighted as a drug carrier which improves patients' outcome. This study explored the impact of miR-124 and its promising targeted gene P2Y12 encapsulated by mesoporous silica nanoparticles (MSNs) on progression of LCI, and its interaction between SCF/c-kit signaling pathway. After establishment of animal models, the animals were divided into 6 groups, namely: model group, blank group, empty carrier group, carrier + miR-124 inhibitor group, SCF/c-kit inhibitor group, and P2Y12 agonist group. Western blot analysis and microscope determined the expression level of miR-124 in the rat brain tissue slices. MVD, SCF and c-kit P2Y12 protein expression levels were detected and their targeting relationship was verified. miR-124 was poorly expressed in the cells of rats with LCI upon injection of MSNs carrying miR-124-inhibitor. The LCI model group had the highest number of VEGF-positive. Compared with the model group, the number in the carrier + miR-124 inhibitor group was lowest. Moreover, treatment with SCF/c-kit inhibitor and P2Y12 agonist also obtained reduction in the number of VEGF-positive cells with less prominent effect (P < 0.05). With elevation of MVD in the LCI rats, injection of P2Y12 agonist or SCF/c-kit inhibitor significantly decreased the amount of MVD, while miR-124 inhibitor-loaded MSNs better reduced the MVD level. Besides, the LCI rats exhibited up-regulated level of P2Y12 protein. Injection of P2Y12 agonist or SCF/c-kit inhibitor dramatically decreased the level of P2Y12, where the level was still higher than that of carrier + miR-124 inhibitor group. Moreover, administration of miR-124 inhibitor-loaded MSNs resulted in increased SCF and c-kit protein level, and SCF/c-kit inhibitor group and P2Y12 agonist group also had increased SCF and c-kit protein level, compared to the model group. Mechanistically, the miR-124 was indicated to target P2Y12 with stronger fluorescence intensity in mutant plasmid (P < 0.05). MSN-encapsulated miR-124 inhibitor increased the expression of SCF/c-kit protein by targeting P2Y12, thereby enhancing regeneration of cerebral blood vessels in LCI.


Subject(s)
MicroRNAs , Nanoparticles , Animals , Cerebral Infarction , Humans , MicroRNAs/genetics , MicroRNAs/metabolism , Rats , Signal Transduction , Silicon Dioxide , Stem Cell Factor/genetics , Stem Cell Factor/metabolism
18.
Nat Commun ; 13(1): 648, 2022 02 03.
Article in English | MEDLINE | ID: mdl-35115536

ABSTRACT

In the bone marrow, classical and plasmacytoid dendritic cells (DC) develop from the macrophage-DC precursor (MDP) through a common DC precursor (CDP) step. This developmental process receives essential input from the niche in which it takes place, containing endothelial cells (EC) among other cell types. Here we show that targeted deletion of serine/threonine kinase 11 (Stk11) encoding tumor suppressor liver kinase b1 (Lkb1) in mouse ECs but not DCs, results in disrupted differentiation of MDPs to CDPs, severe reduction in mature DC numbers and spontaneous tumorigenesis. In wild type ECs, Lkb1 phosphorylates polypyrimidine tract binding protein 1 (Ptbp1) at threonine 138, which regulates stem cell factor (Scf) pre-mRNA splicing. In the absence of Lkb1, exon 6 of Scf is spliced out, leading to the loss of Scf secretion. Adeno-associated-virus-mediated delivery of genes encoding either soluble Scf or the phosphomimetic mutant Ptbp1T138E proteins rescued the defects of MDP to CDP differentiation and DC shortage in the endothelium specific Stk11 knockout mice. In summary, endothelial Stk11 expression regulates DC differentiation via modulation of Scf splicing, marking the Stk11-soluble-Scf axis as a potential cause of DC deficiency syndromes.


Subject(s)
AMP-Activated Protein Kinases/genetics , Cell Differentiation/genetics , Cell Transformation, Neoplastic/genetics , Dendritic Cells/metabolism , Endothelial Cells/metabolism , AMP-Activated Protein Kinase Kinases/genetics , AMP-Activated Protein Kinase Kinases/metabolism , AMP-Activated Protein Kinases/metabolism , Animals , Blotting, Western , Bone Marrow Cells/metabolism , Cell Survival/genetics , Cells, Cultured , Fluorescent Antibody Technique , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Mice, Knockout , Mice, Transgenic , RNA Interference , Reverse Transcriptase Polymerase Chain Reaction , Stem Cell Factor/genetics , Stem Cell Factor/metabolism
19.
Elife ; 102021 12 01.
Article in English | MEDLINE | ID: mdl-34851822

ABSTRACT

Cell cycle gene expression programs fuel proliferation and are universally dysregulated in cancer. The retinoblastoma (RB)-family of proteins, RB1, RBL1/p107, and RBL2/p130, coordinately represses cell cycle gene expression, inhibiting proliferation, and suppressing tumorigenesis. Phosphorylation of RB-family proteins by cyclin-dependent kinases is firmly established. Like phosphorylation, ubiquitination is essential to cell cycle control, and numerous proliferative regulators, tumor suppressors, and oncoproteins are ubiquitinated. However, little is known about the role of ubiquitin signaling in controlling RB-family proteins. A systems genetics analysis of CRISPR/Cas9 screens suggested the potential regulation of the RB-network by cyclin F, a substrate recognition receptor for the SCF family of E3 ligases. We demonstrate that RBL2/p130 is a direct substrate of SCFcyclin F. We map a cyclin F regulatory site to a flexible linker in the p130 pocket domain, and show that this site mediates binding, stability, and ubiquitination. Expression of a mutant version of p130, which cannot be ubiquitinated, severely impaired proliferative capacity and cell cycle progression. Consistently, we observed reduced expression of cell cycle gene transcripts, as well a reduced abundance of cell cycle proteins, analyzed by quantitative, iterative immunofluorescent imaging. These data suggest a key role for SCFcyclin F in the CDK-RB network and raise the possibility that aberrant p130 degradation could dysregulate the cell cycle in human cancers.


Subject(s)
Cyclins/genetics , Retinoblastoma-Like Protein p130/genetics , Retinoblastoma/genetics , Stem Cell Factor/genetics , Cyclins/metabolism , HEK293 Cells , HeLa Cells , Humans , MCF-7 Cells , Retinoblastoma-Like Protein p130/metabolism , Stem Cell Factor/metabolism
20.
Mol Genet Genomic Med ; 9(12): e1841, 2021 12.
Article in English | MEDLINE | ID: mdl-34716665

ABSTRACT

Familial Progressive Hyper- and Hypopigmentation is a pigmentary disorder characterized by a mix of hypo- and hyperpigmented lesions, café-au-lait spots and hypopigmented ash-leaf macules. The disorder was previously linked to KITLG and various mutations have been reported to segregate in different families. Furthermore, association between KITLG mutations and malignancies was also suggested. Exome and SANGER sequencing were performed for identification of KITLG mutations. Functional in silico analyses were additionally performed to assess the findings. We identified a de novo mutation in exon 4 of KITLG gene causing NM_000899.4:c.[329A>T] (chr12:88912508A>T) leading to NP_000890.1:p.(Asp110Val) substitution in the 3rd alpha helix. It was predicted as pathogenic, located in a conserved region and causing an increase in hydrophobicity in the KITLG protein. Our findings clearly confirm an additional hot spot of KITLG mutations in the 3rd alpha helix, which very likely increases the risk of malignancies. To our knowledge the present study provides the strongest evidence of association of the KITLG mutation with both Familial Progressive Hyper- and Hypopigmentation and malignancy due to its' location on somatic cancer mutation locus. Additionally we also address difficulties with classification of the unique phenotype and propose a subtype within broader diagnosis.


Subject(s)
Genetic Association Studies , Genetic Predisposition to Disease , Hyperpigmentation/diagnosis , Hyperpigmentation/genetics , Hypopigmentation/diagnosis , Hypopigmentation/genetics , Mutation , Stem Cell Factor/genetics , Amino Acid Sequence , Genetic Association Studies/methods , Humans , Immunohistochemistry , Pedigree , Phenotype , Sequence Analysis, DNA , Skin/pathology , Stem Cell Factor/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...