Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 15.421
Filter
1.
Elife ; 132024 Jul 11.
Article in English | MEDLINE | ID: mdl-38990208

ABSTRACT

Rare early-onset lower urinary tract disorders include defects of functional maturation of the bladder. Current treatments do not target the primary pathobiology of these diseases. Some have a monogenic basis, such as urofacial, or Ochoa, syndrome (UFS). Here, the bladder does not empty fully because of incomplete relaxation of its outflow tract, and subsequent urosepsis can cause kidney failure. UFS is associated with biallelic variants of HPSE2, encoding heparanase-2. This protein is detected in pelvic ganglia, autonomic relay stations that innervate the bladder and control voiding. Bladder outflow tracts of Hpse2 mutant mice display impaired neurogenic relaxation. We hypothesized that HPSE2 gene transfer soon after birth would ameliorate this defect and explored an adeno-associated viral (AAV) vector-based approach. AAV9/HPSE2, carrying human HPSE2 driven by CAG, was administered intravenously into neonatal mice. In the third postnatal week, transgene transduction and expression were sought, and ex vivo myography was undertaken to measure bladder function. In mice administered AAV9/HPSE2, the viral genome was detected in pelvic ganglia. Human HPSE2 was expressed and heparanase-2 became detectable in pelvic ganglia of treated mutant mice. On autopsy, wild-type mice had empty bladders, whereas bladders were uniformly distended in mutant mice, a defect ameliorated by AAV9/HPSE2 treatment. Therapeutically, AAV9/HPSE2 significantly ameliorated impaired neurogenic relaxation of Hpse2 mutant bladder outflow tracts. Impaired neurogenic contractility of mutant detrusor smooth muscle was also significantly improved. These results constitute first steps towards curing UFS, a clinically devastating genetic disease featuring a bladder autonomic neuropathy.


Subject(s)
Dependovirus , Disease Models, Animal , Gene Transfer Techniques , Glucuronidase , Urinary Bladder , Animals , Mice , Humans , Urinary Bladder/physiopathology , Glucuronidase/genetics , Glucuronidase/metabolism , Dependovirus/genetics , Genetic Therapy/methods , Genetic Vectors , Intestinal Pseudo-Obstruction/genetics , Intestinal Pseudo-Obstruction/therapy , Intestinal Pseudo-Obstruction/physiopathology , Urologic Diseases , Facies
2.
Front Endocrinol (Lausanne) ; 15: 1405665, 2024.
Article in English | MEDLINE | ID: mdl-38948524

ABSTRACT

Background: Increased levels of serum Klotho have been associated with a reduced risk of several cardiovascular diseases (CVD). However, limited studies exist on the association between serum Klotho and mortality in patients with CVD. Methods: We collected data from CVD patients in the National Health and Nutrition Examination Survey (NHANES) spanning 2007 to 2016. We linked NHANES data with the National Death Index to determine the survival status of participants. Univariate and multivariable Cox regression models were used to investigate the relationship between serum Klotho levels and mortality in CVD patients. The relationship between serum Klotho quartiles and mortality in CVD patients was visualized using Kaplan-Meier (KM) curves and restricted cubic spine. Finally, subgroup analyses were used to examine the association between serum Klotho and all-cause mortality in different populations. Results: 1905 patients with CVD were finally enrolled in our study with a mean follow-up of 7.1 years. The average age of the participants was 63.4 years, with 58.40% being male. KM showed that lower Klotho levels were associated with lower survival rates. After adjusting for potential confounders, patients with higher serum Klotho levels had lower all-cause mortality (Q1: 1.00, Q2: 0.58 (0.42-0.80), Q3: 0.69 (0.47-1.01), and Q4:0.64 (0.45-0.92). However, the relationship between serum Klotho levels and cardiovascular mortality was not statistically significant. Dose-response analysis shows a U-shaped relationship between serum Klotho levels and all-cause mortality in patients with CVD (P nonlinear=0.002). Subgroup analysis indicated that participants with a history of hypertension had a higher risk of all-cause mortality in serum Klotho Q4 compared to Q1 (P trend <0.05). Conclusion: The relationship between serum Klotho levels and all-cause mortality in CVD patients exhibits a U-shaped association. The underlying mechanisms of this association need further investigation.


Subject(s)
Cardiovascular Diseases , Klotho Proteins , Nutrition Surveys , Humans , Male , Female , Cardiovascular Diseases/mortality , Cardiovascular Diseases/blood , Middle Aged , Prospective Studies , Aged , United States/epidemiology , Glucuronidase/blood , Biomarkers/blood , Cause of Death , Follow-Up Studies , Survival Rate
4.
Int J Biol Sci ; 20(9): 3412-3425, 2024.
Article in English | MEDLINE | ID: mdl-38993571

ABSTRACT

Chronic kidney disease (CKD) is linked to greater prevalence and rapid progression of calcific aortic valve disease (CAVD) characterized by valvular leaflet fibrosis and calcification. Fibroblast growth factor 23 (FGF23) level is elevated, and anti-aging protein Klotho is reduced in CKD patients. However, the roles of FGF23 and Klotho in the mechanism of aortic valve fibrosis and calcification remain unclear. We hypothesized that FGF23 mediates CKD-induced CAVD by enhancing aortic valve interstitial cell (AVIC) fibrosis and calcification, while soluble Klotho inhibits FGF23 effect. Methods and Results: In an old mouse model of CKD, kidney damages were accompanied by aortic valve thickening and calcification. FGF23 levels in plasma and aortic valve were increased, while Klotho levels were decreased. Recombinant FGF23 elevated the inflammatory, fibrogenic, and osteogenic activities in AVICs. Neutralizing antibody or shRNA targeting FGF23 suppressed the pathobiological activities in AVICs from valves affected by CAVD. FGF23 exerts its effects on AVICs via FGF receptor (FGFR)/Yes-associated protein (YAP) signaling, and inhibition of FGFR/YAP reduced FGF23's potency in AVICs. Recombinant Klotho downregulated the pathobiological activities in AVICs exposed to FGF23. Incubation of FGF23 with Klotho formed complexes and decreased FGF23's potency. Further, treatment of CKD mice with recombinant Klotho attenuated aortic valve lesions. Conclusion: This study demonstrates that CKD induces FGF23 accumulation, Klotho insufficiency and aortic valve lesions in old mice. FGF23 upregulates the inflammatory, fibrogenic and osteogenic activities in AVICs via the FGFR/YAP signaling pathway. Soluble Klotho suppresses FGF23 effect through molecular interaction and is capable of mitigating CKD-induced CAVD.


Subject(s)
Aortic Valve , Fibroblast Growth Factor-23 , Fibroblast Growth Factors , Glucuronidase , Klotho Proteins , Renal Insufficiency, Chronic , Klotho Proteins/metabolism , Fibroblast Growth Factor-23/metabolism , Animals , Renal Insufficiency, Chronic/metabolism , Glucuronidase/metabolism , Fibroblast Growth Factors/metabolism , Mice , Aortic Valve/metabolism , Aortic Valve/pathology , Calcinosis/metabolism , Male , Signal Transduction , Mice, Inbred C57BL , Humans , Aortic Valve Stenosis/metabolism , Disease Models, Animal
5.
Biol Direct ; 19(1): 45, 2024 Jun 11.
Article in English | MEDLINE | ID: mdl-38863009

ABSTRACT

BACKGROUND: Glioma is a common tumor that occurs in the brain and spinal cord. Hypoxia is a crucial feature of the tumor microenvironment. Tumor-associated macrophages/microglia play a crucial role in the advancement of glioma. This study aims to illuminate the detailed mechanisms by which hypoxia regulates microglia and, consequently, influences the progression of glioma. METHODS: The glioma cell viability and proliferation were analyzed by cell counting kit-8 assay and 5-ethynyl-2'-deoxyuridine assay. Wound healing assay and transwell assay were implemented to detect glioma cell migration and invasion, respectively. Enzyme-linked immunosorbent assay was conducted to detect protein levels in cell culture medium. The protein levels in glioma cells and tumor tissues were evaluated using western blot analysis. The histological morphology of tumor tissue was determined by hematoxylin-eosin staining. The protein expression in tumor tissues was determined using immunohistochemistry. Human glioma xenograft in nude mice was employed to test the influence of hypoxic microglia-derived interleukin-1beta (IL-1ß) and heparanase (HPSE) on glioma growth in vivo. RESULTS: Hypoxic HMC3 cells promoted proliferation, migration, and invasion abilities of U251 and U87 cells by secreting IL-1ß, which was upregulated by hypoxia-induced activation of hypoxia inducible factor-1alpha (HIF-1α). Besides, IL-1ß from HMC3 cells promoted glioma progression and caused activation of nuclear factor-κB (NF-κB) and upregulation of HPSE in vivo. We also confirmed that IL-1ß facilitated HPSE expression in U251 and U87 cells by activating NF-κB. Hypoxic HMC3 cells-secreted IL-1ß facilitated the proliferation, migration, and invasion of U251 and U87 cells via NF-κB-mediated upregulation of HPSE expression. Finally, we revealed that silencing HPSE curbed the proliferation and metastasis of glioma in mice. CONCLUSION: Hypoxia-induced activation of HIF-1α/IL-1ß axis in microglia promoted glioma progression via NF-κB-mediated upregulation of HPSE expression.


Subject(s)
Glioma , Glucuronidase , Hypoxia-Inducible Factor 1, alpha Subunit , Interleukin-1beta , Mice, Nude , Microglia , NF-kappa B , Up-Regulation , Glioma/metabolism , Glioma/genetics , Glioma/pathology , Interleukin-1beta/metabolism , Interleukin-1beta/genetics , Microglia/metabolism , Animals , NF-kappa B/metabolism , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Mice , Glucuronidase/metabolism , Glucuronidase/genetics , Cell Line, Tumor , Disease Progression , Brain Neoplasms/metabolism , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Cell Proliferation , Cell Movement , Hypoxia/metabolism , Hypoxia/physiopathology , Hypoxia/genetics
6.
Funct Plant Biol ; 512024 Jun.
Article in English | MEDLINE | ID: mdl-38870342

ABSTRACT

Genetic transformation is helpful in enhancing crops, utilising promoters that can be constitutive, inducible, or tissue-specific. However, the use of constitutive promoters may hinder plant growth due to energy consumption during cellular processes. To optimise transgene effects, tissue-specific promoters like root-specific ones prove valuable in addressing root-related issues and enhancing productivity. Yet, identified root-specific promoters in crop are limited. To address this gap, the expression pattern of the root-specific SlREO promoter was examined across various crops. Sequencing confirmed its identity and high homology (99%) with the NCBI database, distinct from other plants tested. Using the PLACE database, six motifs associated with root expression were identified, along with several other important elements. The 2.4kb SlREO promoter was linked to a ß-glucuronidase (GUS) reporter gene alongside the CaMV35S promoter in pRI 201-AN-GUS vectors to study its expression. Histochemistry revealed strong root-specific expression in tomato (Solanum lycopersicum ) root tissues and limited expression in stems. However, the SlREO promoter did not consistently maintain its root-specific expression in other plants. Conversely, the CaMV35S promoter exhibited constitutive expression across all tissues in various plants. This study underscores the potential of the SlREO promoter as a root-specific regulatory element, offering avenues for improving crops, particularly against environmental stresses.


Subject(s)
Cloning, Molecular , Gene Expression Regulation, Plant , Plant Roots , Plants, Genetically Modified , Promoter Regions, Genetic , Solanum lycopersicum , Solanum lycopersicum/genetics , Plant Roots/genetics , Plants, Genetically Modified/genetics , Plant Proteins/genetics , Plant Proteins/metabolism , Glucuronidase/genetics , Glucuronidase/metabolism , Base Sequence
7.
Cell Host Microbe ; 32(6): 783-785, 2024 Jun 12.
Article in English | MEDLINE | ID: mdl-38870895

ABSTRACT

Previous studies have explored the role of the gut microbiota in regulating endobiotic homeostasis, but the precise mechanisms remain unclear. In this issue of Cell Host & Microbe, Simpson et al. identified two predominant subtypes of gut microbial ß-Glucuronidase (gmGUS) that can reactivate hormones and neurotransmitters to regulate endobiotic homeostasis.


Subject(s)
Gastrointestinal Microbiome , Glucuronidase , Homeostasis , Glucuronidase/metabolism , Glucuronidase/genetics , Gastrointestinal Microbiome/physiology , Humans , Animals , Gastrointestinal Tract/microbiology , Bacteria/enzymology , Bacteria/metabolism , Bacteria/genetics
8.
J Nanobiotechnology ; 22(1): 325, 2024 Jun 10.
Article in English | MEDLINE | ID: mdl-38858695

ABSTRACT

BACKGROUND: Osteoarthritis (OA) is an aging-related degenerative joint disorder marked by joint discomfort and rigidity. Senescent chondrocytes release pro-inflammatory cytokines and extracellular matrix-degrading proteins, creating an inflammatory microenvironment that hinders chondrogenesis and accelerates matrix degradation. Targeting of senescent chondrocytes may be a promising approach for the treatment of OA. Herein, we describe the engineering of an injectable peptide-hydrogel conjugating a stem cell-homing peptide PFSSTKT for carrying plasmid DNA-laden nanoparticles and Tanshinon IIA (pPNP + TIIA@PFS) that was designed to attenuate OA progression by improving the senescent microenvironment and fostering cartilage regeneration. RESULTS: Specifically, pPNP + TIIA@PFS elevates the concentration of the anti-aging protein Klotho and blocks the transmission of senescence signals to adjacent healthy chondrocytes, significantly mitigating chondrocyte senescence and enhancing cartilage integrity. Additionally, pPNP + TIIA@PFS recruit bone mesenchymal stem cells and directs their subsequent differentiation into chondrocytes, achieving satisfactory chondrogenesis. In surgically induced OA model rats, the application of pPNP + TIIA@PFS results in reduced osteophyte formation and attenuation of articular cartilage degeneration. CONCLUSIONS: Overall, this study introduces a novel approach for the alleviation of OA progression, offering a foundation for potential clinical translation in OA therapy.


Subject(s)
Chondrocytes , Chondrogenesis , Glucuronidase , Hydrogels , Klotho Proteins , Mesenchymal Stem Cells , Osteoarthritis , Plasmids , Rats, Sprague-Dawley , Animals , Osteoarthritis/therapy , Osteoarthritis/drug therapy , Hydrogels/chemistry , Rats , Chondrocytes/metabolism , Chondrocytes/drug effects , Glucuronidase/metabolism , Glucuronidase/pharmacology , Chondrogenesis/drug effects , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/drug effects , Male , Cartilage, Articular/drug effects , Cartilage, Articular/metabolism , Disease Progression , Nanoparticles/chemistry , Humans , DNA , Cellular Senescence/drug effects , Cell Differentiation/drug effects
9.
Commun Biol ; 7(1): 720, 2024 Jun 11.
Article in English | MEDLINE | ID: mdl-38862813

ABSTRACT

Overexpression of the longevity gene Klotho prolongs lifespan, while its knockout shortens lifespan and impairs cognition via perturbation of myelination and synapse formation. However, comprehensive analysis of Klotho knockout effects on mammalian brain transcriptomics is lacking. Here, we report that Klotho knockout alters the levels of aging- and cognition related mRNAs, long non-coding RNAs, microRNAs and tRNA fragments. These include altered neuronal and glial regulators in murine models of aging and Alzheimer's disease and in human Alzheimer's disease post-mortem brains. We further demonstrate interaction of the knockout-elevated tRNA fragments with the spliceosome, possibly affecting RNA processing. Last, we present cell type-specific short RNA-seq datasets from FACS-sorted neurons and microglia of live human brain tissue demonstrating in-depth cell-type association of Klotho knockout-perturbed microRNAs. Together, our findings reveal multiple RNA transcripts in both neurons and glia from murine and human brain that are perturbed in Klotho deficiency and are aging- and neurodegeneration-related.


Subject(s)
Aging , Alzheimer Disease , Brain , Glucuronidase , Klotho Proteins , Longevity , Mice, Knockout , MicroRNAs , RNA, Transfer , Klotho Proteins/metabolism , MicroRNAs/genetics , MicroRNAs/metabolism , Animals , Aging/genetics , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Brain/metabolism , Brain/pathology , Mice , Glucuronidase/genetics , Glucuronidase/metabolism , Humans , Longevity/genetics , RNA, Transfer/genetics , RNA, Transfer/metabolism , Male , Neurons/metabolism , Mice, Inbred C57BL
10.
Lipids Health Dis ; 23(1): 188, 2024 Jun 21.
Article in English | MEDLINE | ID: mdl-38907289

ABSTRACT

BACKGROUND: The anti-aging protein Klotho has diverse functions in antioxidative stress and energy metabolism through several pathways. While it has been reported that α-Klotho is downregulated in patients with insulin resistance (IR), the association between Klotho and IR is complex and controversial. The triglyceride-glucose (TyG) index has provided a practical method for assessing IR. With this in mind, our study aimed to investigate the relationship between the TyG index and soluble α-Klotho protein levels in US populations, both with and without diabetes mellitus. METHODS: This cross-sectional study analyzed data from middle-aged and older participants in the National Health and Nutrition Examination Survey (NHANES) conducted between 2007 and 2016. The participants were divided into two groups based on their diabetes mellitus status: those with diabetes and those without diabetes. To evaluate the relationship between the TyG index and the concentration of the α-Klotho protein in each group, a series of survey-weighted multivariable linear regression models were employed. Furthermore, to examine the association between these two variables, multivariable-adjusted restricted cubic spline curves and subgroup analysis were generated. RESULTS: The study involved 6,439 adults aged 40 years or older, with a mean age of 57.8 ± 10.9 years. Among them, 1577 (24.5%) had diabetes mellitus. A subgroup analysis indicated that the presence of diabetes significantly affected the relationship between the TyG index and the α-Klotho level. After considering all covariables, regression analysis of the participants without diabetes revealed that the α-Klotho concentration decreased by 32.35 pg/ml (95% CI: -50.07, -14.64) with each one unit increase in TyG (p < 0.001). The decline in α-Klotho levels with elevated TyG was more pronounced in the female population. In patients with diabetes mellitus, a non-linear association between the TyG index and α-Klotho was observed. There was no significant correlation observed between the two when TyG index were below 9.7. However, there was an increase in klotho levels of 106.44 pg/ml for each unit increase in TyG index above 9.7 (95% CI: 28.13, 184.74) (p = 0.008). CONCLUSION: Our findings suggested that the presence of diabetes may influence the relationship between the TyG index and soluble α-Klotho. Furthermore, there seem to be sex differences in individuals without diabetes. Further studies are necessary to validate these findings.


Subject(s)
Blood Glucose , Diabetes Mellitus , Glucuronidase , Klotho Proteins , Nutrition Surveys , Triglycerides , Humans , Klotho Proteins/blood , Middle Aged , Male , Female , Aged , Glucuronidase/blood , Cross-Sectional Studies , Blood Glucose/metabolism , Triglycerides/blood , Diabetes Mellitus/blood , Diabetes Mellitus/epidemiology , Insulin Resistance , Adult
11.
Sci Rep ; 14(1): 13189, 2024 06 08.
Article in English | MEDLINE | ID: mdl-38851827

ABSTRACT

The systemic immune-inflammation index (SII), an integrated and ground-breaking inflammatory measure, has been widely used in various fields. We aimed to assess the association between the systemic immune-inflammation index (SII) and α-Klotho (a new anti-aging biomarker). In this cross-sectional investigation, people with complete information on SII and α-Klotho from the National Health and Nutrition Examination Survey (NHANES) between 2007 and 2016 were the study's subject population. SII was calculated by platelet count × neutrophil count/lymphocyte count. The association between SII and α-Klotho was investigated using multivariable linear regression and a generalized additive model. In order to explore the non-linear connection, we employed smoothed curve fitting. Subgroup analysis were also performed. A total of 13,701 participants with an average age of 57.73 ± 10.86 years were enrolled, of whom 51.53% were female. After fully adjustment, SII was negatively associated with serum soluble α-Klotho [ß(95% CI) = - 0.07 (- 0.08, - 0.05)]. Furthermore, we found L-shaped association between SII and klotho protein level, with the inflection point at 255 pg/ml. Subgroup analysis and interaction test revealed that there was no discernible dependence on gender, age, race, smoking, alcohol, diabetes and hypertension (all p for interaction > 0.05). SII level was negatively associated with serum klotho protein concentration in American adults. To verify our findings, more large-scale prospective investigations are still required.


Subject(s)
Biomarkers , Glucuronidase , Inflammation , Klotho Proteins , Nutrition Surveys , Humans , Female , Male , Middle Aged , Inflammation/blood , Cross-Sectional Studies , Prospective Studies , Glucuronidase/blood , Biomarkers/blood , Aged , Adult , Platelet Count
12.
Life Sci ; 351: 122792, 2024 Aug 15.
Article in English | MEDLINE | ID: mdl-38857657

ABSTRACT

AIMS: Drug-induced enteropathy is often associated with the therapeutic use of certain glucuronidated drugs. One such drug is mycophenolic acid (MPA), a well-established immunosuppressant of which gastrointestinal adverse effects are a major concern. The role of bacterial ß-glucuronidase (ß-G) from the gut microbiota in MPA-induced enteropathy has recently been discovered. Bacterial ß-G hydrolyzes MPAG, the glucuronide metabolite of MPA excreted in the bile, leading to the digestive accumulation of MPA that would favor in turn these adverse events. We therefore hypothesized that taming bacterial ß-G activity might reduce MPA digestive exposure and prevent its toxicity. MAIN METHODS: By using a multiscale approach, we evaluated the effect of increasing concentrations of MPA on intestinal epithelial cells (Caco-2 cell line) viability, proliferation, and migration. Then, we investigated the inhibitory properties of amoxapine, a previously described bacterial ß-G inhibitor, by using molecular dynamics simulations, and evaluated its efficiency in blocking MPAG hydrolysis in an Escherichia coli-based ß-G activity assay. The pharmacological effect of amoxapine was evaluated in a mouse model. KEY FINDINGS: We observed that MPA impairs intestinal epithelial cell homeostasis. Amoxapine efficiently blocks the hydrolysis of MPAG to MPA and significantly reduces digestive exposure to MPA in mice. As a result, administration of amoxapine in MPA-treated mice significantly attenuated gastrointestinal lesions. SIGNIFICANCE: Collectively, these results suggest that the digestive accumulation of MPA is involved in the pathophysiology of MPA-gastrointestinal adverse effects. This study provides a proof-of-concept of the therapeutic potential of bacterial ß-G inhibitors in glucuronidated drug-induced enteropathy.


Subject(s)
Biotransformation , Gastrointestinal Microbiome , Glucuronidase , Glucuronides , Mycophenolic Acid , Mycophenolic Acid/metabolism , Mycophenolic Acid/pharmacology , Gastrointestinal Microbiome/drug effects , Glucuronidase/metabolism , Glucuronidase/antagonists & inhibitors , Humans , Animals , Mice , Glucuronides/metabolism , Caco-2 Cells , Male , Immunosuppressive Agents/pharmacology , Immunosuppressive Agents/toxicity , Immunosuppressive Agents/metabolism , Intestinal Diseases/chemically induced , Intestinal Diseases/drug therapy , Intestinal Diseases/metabolism , Intestinal Diseases/microbiology , Cell Proliferation/drug effects , Glycoproteins
13.
Int J Mol Sci ; 25(11)2024 Jun 06.
Article in English | MEDLINE | ID: mdl-38892469

ABSTRACT

Mast cells take up extracellular latent heparanase and store it in secretory granules. The present study examined whether the enzymatic activity of heparanase regulates its uptake efficiency. Recombinant mouse heparanase mimicking both the latent and mature forms (L-Hpse and M-Hpse, respectively) was internalized into mastocytoma MST cells, peritoneal cell-derived mast cells, and bone marrow-derived mast cells. The internalized amount of L-Hpse was significantly higher than that of M-Hpse. In MST cells, L-Hpse was continuously internalized for up to 8 h, while the uptake of M-Hpse was saturated after 2 h of incubation. L-Hpse and M-Hpse are similarly bound to the MST cell surface. The expression level of cell surface heparan sulfate was reduced in MST cells incubated with M-Hpse. The internalized amount of M-Hpse into mast cells was significantly increased in the presence of heparastatin (SF4), a small molecule heparanase inhibitor that does not affect the binding of heparanase to immobilized heparin. Enzymatically quiescent M-Hpse was prepared with a point mutation at Glu335. The internalized amount of mutated M-Hpse was significantly higher than that of wild-type M-Hpse but similar to that of wild-type and mutated L-Hpse. These results suggest that the enzymatic activity of heparanase negatively regulates the mast cell-mediated uptake of heparanase, possibly via the downregulation of cell surface heparan sulfate expression.


Subject(s)
Glucuronidase , Heparitin Sulfate , Mast Cells , Mast Cells/metabolism , Glucuronidase/metabolism , Glucuronidase/genetics , Animals , Heparitin Sulfate/metabolism , Mice , Cell Line, Tumor
14.
Biomacromolecules ; 25(7): 4492-4509, 2024 Jul 08.
Article in English | MEDLINE | ID: mdl-38910355

ABSTRACT

A major shortcoming associated with the application of enzymes in drug synergism originates from the lack of site-specific, multifunctional nanomedicine. This study introduces catalytic nanocompartments (CNCs) made of a mixture of PDMS-b-PMOXA diblock copolymers, decorated with glycooligomer tethers comprising eight mannose-containing repeating units and coencapsulating two enzymes, providing multifunctionality by their in situ parallel reactions. Beta-glucuronidase (GUS) serves for local reactivation of the drug hymecromone, while glucose oxidase (GOx) induces cell starvation through glucose depletion and generation of the cytotoxic H2O2. The insertion of the pore-forming peptide, melittin, facilitates diffusion of substrates and products through the membranes. Increased cell-specific internalization of the CNCs results in a substantial decrease in HepG2 cell viability after 24 h, attributed to simultaneous production of hymecromone and H2O2. Such parallel enzymatic reactions taking place in nanocompartments pave the way to achieve efficient combinatorial cancer therapy by enabling localized drug production along with reactive oxygen species (ROS) elevation.


Subject(s)
Glucose Oxidase , Hydrogen Peroxide , Humans , Glucose Oxidase/chemistry , Glucose Oxidase/metabolism , Hep G2 Cells , Hydrogen Peroxide/chemistry , Hydrogen Peroxide/metabolism , Glucuronidase/metabolism , Cell Survival/drug effects , Catalysis , Reactive Oxygen Species/metabolism , Oligosaccharides/chemistry , Oligosaccharides/metabolism
15.
Cell Host Microbe ; 32(6): 925-944.e10, 2024 Jun 12.
Article in English | MEDLINE | ID: mdl-38754417

ABSTRACT

Hormones and neurotransmitters are essential to homeostasis, and their disruptions are connected to diseases ranging from cancer to anxiety. The differential reactivation of endobiotic glucuronides by gut microbial ß-glucuronidase (GUS) enzymes may influence interindividual differences in the onset and treatment of disease. Using multi-omic, in vitro, and in vivo approaches, we show that germ-free mice have reduced levels of active endobiotics and that distinct gut microbial Loop 1 and FMN GUS enzymes drive hormone and neurotransmitter reactivation. We demonstrate that a range of FDA-approved drugs prevent this reactivation by intercepting the catalytic cycle of the enzymes in a conserved fashion. Finally, we find that inhibiting GUS in conventional mice reduces free serotonin and increases its inactive glucuronide in the serum and intestines. Our results illuminate the indispensability of gut microbial enzymes in sustaining endobiotic homeostasis and indicate that therapeutic disruptions of this metabolism promote interindividual response variabilities.


Subject(s)
Gastrointestinal Microbiome , Glucuronidase , Homeostasis , Animals , Gastrointestinal Microbiome/drug effects , Mice , Glucuronidase/metabolism , Mice, Inbred C57BL , Serotonin/metabolism , Glucuronides/metabolism , Humans , Intestines/microbiology , Male , Germ-Free Life
16.
J Infect Dev Ctries ; 18(4): 618-626, 2024 Apr 30.
Article in English | MEDLINE | ID: mdl-38728647

ABSTRACT

INTRODUCTION: Hepatitis B virus (HBV) infection is a global epidemic that can lead to several liver diseases, seriously affecting people's health. This study aimed to investigate the clinical potential of serum ß-klotho (KLB) as a promising biomarker in HBV-related liver diseases. METHODOLOGY: This study enrolled 30 patients with chronic hepatitis B (CHB), 35 with HBV-related cirrhosis, 66 with HBV-related hepatocellular carcinoma (HCC), and 48 healthy individuals. ELISA measured the levels of serum KLB in the four groups. We then compared the differences in serum KLB levels among the groups and analyzed the relationship between serum KLB and routine clinical parameters. RESULTS: The concentrations of serum KLB levels were increased sequentially among the healthy subjects, the HBV-related CHB group, the HBV-related cirrhosis group, and the HBV-related HCC group (p < 0.05). Expression of KLB was positively correlated with alpha-fetoprotein (AFP), total bilirubin, direct bilirubin, alanine aminotransferase, aspartate aminotransferase, gamma-glutamyl-transferase, alkaline phosphatase, total bile acid, serum markers for liver fibrosis, ascites, cirrhosis, splenomegaly, and model for end-stage liver disease sodium, while negatively correlated with platelet count, albumin, and prothrombin activity (p < 0.05). In addition, serum KLB has better sensitivity in diagnosing HCC than AFP, and serum KLB combined with AFP has higher sensitivity and specificity than AFP alone in diagnosing HCC. CONCLUSIONS: Serum KLB level is associated with the severity of HBV-related liver diseases and has important diagnostic value for HCC. Therefore, it could be a predictive biomarker for monitoring disease progression.


Subject(s)
Biomarkers , Carcinoma, Hepatocellular , Hepatitis B, Chronic , Klotho Proteins , Humans , Male , Female , Biomarkers/blood , Middle Aged , Adult , Hepatitis B, Chronic/blood , Hepatitis B, Chronic/complications , Carcinoma, Hepatocellular/blood , Carcinoma, Hepatocellular/diagnosis , Carcinoma, Hepatocellular/virology , Glucuronidase/blood , Liver Neoplasms/blood , Liver Neoplasms/diagnosis , Liver Neoplasms/virology , Liver Cirrhosis/blood , Liver Cirrhosis/diagnosis , Liver Cirrhosis/virology , Disease Progression , Enzyme-Linked Immunosorbent Assay , Aged
17.
Minerva Med ; 115(3): 320-336, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38727708

ABSTRACT

Phosphate is a key component of mineralized tissues and is also part of many organic compounds. Phosphorus homeostasis depends especially upon intestinal absorption, and renal excretion, which are regulated by various hormones, such as PTH, 1,25-dihydroxyvitamin D, and fibroblast growth factor 23. In this review we provide an update of several genetic disorders that affect phosphate transporters through cell membranes or the phosphate-regulating hormones, and, consequently, result in hypophosphatemia.


Subject(s)
Fibroblast Growth Factor-23 , Fibroblast Growth Factors , Hypophosphatemia , Parathyroid Hormone , Humans , Hypophosphatemia/genetics , Hypophosphatemia/etiology , Fibroblast Growth Factors/metabolism , Fibroblast Growth Factors/genetics , Parathyroid Hormone/metabolism , Phosphates/metabolism , Vitamin D/metabolism , Vitamin D/analogs & derivatives , Klotho Proteins , Phosphate Transport Proteins/genetics , Phosphate Transport Proteins/metabolism , PHEX Phosphate Regulating Neutral Endopeptidase/genetics , Intestinal Absorption/genetics , Glucuronidase/genetics , Glucuronidase/metabolism , Phosphorus/metabolism
18.
Biochem Pharmacol ; 225: 116328, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38815628

ABSTRACT

Early stages of diabetes are characterized by elevations of insulin and glucose concentrations. Both factors stimulate reactive oxygen species (ROS) production, leading to impairments in podocyte function and disruption of the glomerular filtration barrier. Podocytes were recently shown to be an important source of αKlotho (αKL) expression. Low blood Klotho concentrations are also associated with an increase in albuminuria, especially in patients with diabetes. We investigated whether ADAM10, which is known to cleave αKL, is activated in glomeruli and podocytes under diabetic conditions and the potential mechanisms by which ADAM10 mediates ROS production and disturbances of the glomerular filtration barrier. In cultured human podocytes, high glucose increased ADAM10 expression, shedding, and activity, NADPH oxidase activity, ROS production, and albumin permeability. These effects of glucose were inhibited when cells were pretreated with an ADAM10 inhibitor or transfected with short-hairpin ADAM10 (shADAM10) or after the addition soluble Klotho. We also observed increases in ADAM10 activity, NOX4 expression, NADPH oxidase activity, and ROS production in αKL-depleted podocytes. This was accompanied by an increase in albumin permeability in shKL-expressing podocytes. The protein expression and activity of ADAM10 also increased in isolated glomeruli and urine samples from diabetic rats. Altogether, these results reveal a new mechanism by which hyperglycemia in diabetes increases albumin permeability through ADAM10 activation and an increase in oxidative stress via NOX4 enzyme activation. Moreover, αKlotho downregulates ADAM10 activity and supports redox balance, consequently protecting the slit diaphragm of podocyteσ under hyperglycemic conditions.


Subject(s)
ADAM10 Protein , Amyloid Precursor Protein Secretases , Diabetes Mellitus, Experimental , Glucuronidase , Klotho Proteins , Membrane Proteins , Podocytes , Reactive Oxygen Species , Podocytes/metabolism , Podocytes/drug effects , Klotho Proteins/metabolism , ADAM10 Protein/metabolism , ADAM10 Protein/genetics , Reactive Oxygen Species/metabolism , Humans , Animals , Glucuronidase/metabolism , Glucuronidase/genetics , Amyloid Precursor Protein Secretases/metabolism , Membrane Proteins/metabolism , Membrane Proteins/genetics , Rats , Male , Diabetes Mellitus, Experimental/metabolism , NADPH Oxidase 4/metabolism , NADPH Oxidase 4/genetics , NADPH Oxidases/metabolism , Cells, Cultured , Glucose/metabolism , Rats, Sprague-Dawley
19.
Cell Signal ; 120: 111214, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38729322

ABSTRACT

Age-related diseases are intricately linked to the molecular processes underlying aging, with the decline of the antiaging protein Klotho being a key factor. Investigating these processes is crucial for developing therapeutic strategies. The age-associated reduction in Klotho expression, coupled with a decline in the endocrine hormone triiodothyronine (T3), prompted a detailed exploration of their potential interplay. Our research, conducted through both in-vitro and in-vivo studies on BALB/c mice, unveiled a significant capacity of T3 to upregulate various forms of Klotho via ATF-3/p-c-Jun transcription factor. This effect was particularly noteworthy in aged individuals, where Klotho expression had waned compared to their younger counterparts. Importantly, T3 demonstrated a promising therapeutic impact in rejuvenating Klotho expression in this context. Further investigations elucidated the molecular mechanisms underlying T3's impact on aging-related pathways. In-vitro and in-vivo experiments established T3's ability to downregulate the Wnt/ß-Catenin pathway by enhancing Klotho expression. In-silico analyses provided insights into Klotho's intricate role, showing its capacity to inhibit Wnt ligands such as Wnt3 and Wnt8a, consequently disrupting their interaction with the Wnt receptor. Additionally, T3 was found to downregulate kidney-specific GSK-3ß expression through the augmentation of Klotho expression. The study also highlighted T3's role in maintaining calcium and phosphate homeostasis via Klotho. This comprehensive investigation not only sheds light on the intricate mechanisms governing aging processes but also presents promising avenues for therapeutic interventions targeting the Wnt/ß-Catenin pathway implicated in various age-associated diseases.


Subject(s)
Glucuronidase , Kidney , Klotho Proteins , Mice, Inbred BALB C , Triiodothyronine , Wnt Signaling Pathway , Klotho Proteins/metabolism , Animals , Triiodothyronine/metabolism , Triiodothyronine/pharmacology , Glucuronidase/metabolism , Wnt Signaling Pathway/drug effects , Mice , Kidney/metabolism , Humans , Male , beta Catenin/metabolism , Aging/metabolism , Computer Simulation
20.
Endocr Res ; 49(3): 145-153, 2024.
Article in English | MEDLINE | ID: mdl-38725277

ABSTRACT

OBJECTIVE: This study aimed to investigate the relationship between serum α-Klotho levels and insulin resistance (IR), a precursor to type 2 diabetes. METHODS: The study analyzed data from 4,758 adult participants in the National Health and Nutrition Examination Survey (NHANES) spanning from 2007 to 2016. The relationship between α-Klotho concentration and IR was assessed using the Homeostatic Model Assessment of Insulin Resistance (HOMA-IR) and odds ratios (OR) derived from logistic regression models. RESULTS: Results showed that every 1-ln increase in α-Klotho concentration raised the HOMA-IR value by 0.55 (95% confidence interval 0.35-0.74) and the odds of IR by 64% (odds ratio 1.64; 95% confidence interval 1.28-2.1). The odds of IR was 40% greater in highest tertile than in the lowest tertile. CONCLUSION: The findings of this study underscore a significant correlation between increased serum α-Klotho levels and the prevalence of IR.


Subject(s)
Insulin Resistance , Klotho Proteins , Nutrition Surveys , Humans , Insulin Resistance/physiology , Male , Female , Adult , Middle Aged , Glucuronidase/blood , Aged , Cross-Sectional Studies , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL