Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 46
Filtrar
Más filtros












Intervalo de año de publicación
1.
Int J Pharm ; 662: 124546, 2024 Sep 05.
Artículo en Inglés | MEDLINE | ID: mdl-39097154

RESUMEN

Biopolymers application in biomedical areas has been limited due to the physicochemical degradation that occurs using conventional processing/sterilization methods (e.g., steam heat, γ-radiation, ethylene oxide). Aiming to avoid/minimize degradation and preserve their properties, supercritical carbon dioxide (scCO2) has been proposed as an alternative sterilization method for such materials. ScCO2 can simultaneously be used as a drying method to produce aerogels (i) and sterilize them (ii). However, a solvent exchange is required to prepare the alcogel from hydrogel, achievable through high-pressure solvent exchange (HPSE) (iii). This study integrated three processes: HPSE, scCO2 drying, and sterilization to prepare alginate-gelatine sterilized aerogels. Two scCO2 sterilization methods were tested. Results showed that sterilization did not compromise the aerogels' chemical, thermal and swelling properties. Conversely, Young's Modulus increased, and BET surface area decreased, due to the structural changes caused by the fast pressurization/depressurization rates applied during sterilization. Regarding the sterilization efficiency, results showed a reduction in contamination throughout the process, achieving a SAL of 10-4. The sterilized aerogels were non-cytotoxic in vitro and showed improved wound-healing properties. The innovative integrated process produced decontaminated/sterile and ready-to-use aerogels reducing process time by 75 %, from 2 days up to 12 h without compromising the aerogel's properties.


Asunto(s)
Alginatos , Dióxido de Carbono , Gelatina , Geles , Esterilización , Alginatos/química , Gelatina/química , Esterilización/métodos , Dióxido de Carbono/química , Geles/química , Animales , Cicatrización de Heridas/efectos de los fármacos , Hidrogeles/química , Ácido Glucurónico/química , Solventes/química , Ratones , Ácidos Hexurónicos/química
2.
J Appl Genet ; 2024 Aug 16.
Artículo en Inglés | MEDLINE | ID: mdl-39150614

RESUMEN

Urofacial syndrome or Ochoa syndrome (UFS or UFOS) is a rare disease characterized by inverted facial expression and bladder dysfunction that was described for the first time in Colombia. It is an autosomal recessive pathology with mutations in the HPSE2 and LRIG2 genes. However, 16% of patients do not have any mutations associated with the syndrome. Despite the importance of neurobiology in its pathophysiology, there are no neurological, neuropsychological, or psychological studies in these patients. A 30-year-old male from Medellín, Colombia, with a significant perinatal history, was diagnosed with grade 4 hydronephrosis on his first ultrasound test. At 4 months of age, symptoms such as hypomimia, lagophthalmos, and recurrent urinary tract infections started to manifest. Imaging studies revealed urinary tract dilatation, vesicoureteral reflux, and a double collector system on his left side, which led to the diagnosis of UFS. Multiple procedures, including vesicostomy, ureterostomy, and enterocystoplasty, were performed. At 20 years of age, he achieved urinary sphincter control. Genetic analysis revealed a founder pathogenic variant, c.1516C > T (p.Arg506Ter), in the HPSE2 gene, which produces a truncated protein that lacks 86 amino acids. This variant is classified as pathogenic according to the ClinVar database for UFS. The mutation age is approximately 260-360 years, and the two alleles share a 7.2-7.4 Mb IBD segment. Moreover, we detected European local ancestry in the IBD segment, which is consistent with a Spanish introduction. Neurological examination, neuropsychological assessment, and psychological testing revealed no abnormalities, except for high stress levels. Clinical analysis of this patient revealed distorted facial expression and detrusor-sphincter dyssynergia, which are typical of patients with UFS. Genetic analysis revealed a pathogenic variant in the HPSE2 gene of European origin and a mutation age of 260-360 years. From a neurological, neuropsychological, and psychological (emotional and personality) perspective, the patient showed no signs or symptoms of clinical interest.

3.
Elife ; 132024 Jul 11.
Artículo en Inglés | MEDLINE | ID: mdl-38990208

RESUMEN

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.


Asunto(s)
Dependovirus , Modelos Animales de Enfermedad , Técnicas de Transferencia de Gen , Glucuronidasa , Vejiga Urinaria , Animales , Ratones , Humanos , Vejiga Urinaria/fisiopatología , Glucuronidasa/genética , Glucuronidasa/metabolismo , Dependovirus/genética , Terapia Genética/métodos , Vectores Genéticos , Seudoobstrucción Intestinal/genética , Seudoobstrucción Intestinal/terapia , Seudoobstrucción Intestinal/fisiopatología , Enfermedades Urológicas , Facies
4.
Technol Cancer Res Treat ; 23: 15330338241261615, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38887096

RESUMEN

This study aimed to investigate the role of miR-558 in tumor angiogenesis by targeting heparinase (HPSE) in tongue squamous cell carcinoma (TSCC)-derived exosomes. In the present study, the role of exosome miR-558 in angiogenesis in vitro and in vivo was investigated by cell proliferation, migration, tube formation, subcutaneous tumor formation in mice, and in vivo Matrigel plug assay. The target genes of miR-558 were detected by means of dual luciferase assay. It was found that TSCC cells secrete miR-558 into the extracellular environment, with exosome as the carrier. Human umbilical vein endothelial cells (HUVEC) ingested exosomes, which not only increased the expression level of miR-558, but also enhanced their proliferation, migration, and tube formation functions. In vivo Matrigel plug assay demonstrated that TSCC cell-derived exosome miR-558 promoted neovascularization in vivo. Compared with negative control cells, TSCC cells overexpressing miR-558 formed subcutaneous tumors in nude mice, with larger volume, heavier mass, and more vascularization. Dual luciferase assay confirmed that HPSE was the direct target gene regulated by miR-558. HPSE promoted the proliferation, migration, and tube formation of HUVECs, and the knockout of HPSE could downregulate the pro-angiogenic effect of miR-558. In summary, miR-558 in TSCC exosomes promotes the proliferation, migration, and tube formation of HUVECs by targeting HPSE, and enhancing tumor angiogenesis.


Asunto(s)
Movimiento Celular , Proliferación Celular , Exosomas , Regulación Neoplásica de la Expresión Génica , Liasa de Heparina , MicroARNs , Neovascularización Patológica , Neoplasias de la Lengua , Humanos , Animales , MicroARNs/genética , Exosomas/metabolismo , Exosomas/genética , Neoplasias de la Lengua/patología , Neoplasias de la Lengua/genética , Neoplasias de la Lengua/metabolismo , Ratones , Neovascularización Patológica/genética , Neovascularización Patológica/metabolismo , Neovascularización Patológica/patología , Movimiento Celular/genética , Línea Celular Tumoral , Liasa de Heparina/metabolismo , Liasa de Heparina/genética , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/patología , Carcinoma de Células Escamosas/metabolismo , Células Endoteliales de la Vena Umbilical Humana , Modelos Animales de Enfermedad , Ensayos Antitumor por Modelo de Xenoinjerto , Carcinoma de Células Escamosas de Cabeza y Cuello/genética , Carcinoma de Células Escamosas de Cabeza y Cuello/patología , Carcinoma de Células Escamosas de Cabeza y Cuello/metabolismo , Angiogénesis
5.
Int J Lab Hematol ; 45(6): 935-944, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37646104

RESUMEN

INTRODUCTION: The heparanase (HPSE) gene is highly polymorphic, but only a minority of its single nucleotide polymorphisms (SNPs) have been studied. Among these, rs4693608 and rs4364254 SNPs are closely associated with mRNA expression and HPSE protein levels in healthy subjects. Given the association between HPSE and inflammatory response, we aimed to evaluate whether HPSE rs4693608 and rs4364254 SNPs could have an impact on graft-versus-host disease after allogeneic stem cell transplants (HSCT). METHODS: A total of 228 consecutive patients who underwent HSCT at our center between 2005 and 2018 were included. The rs4693608 SNP was identified by polymerase chain reaction-restriction fragment length polymorphism (PCR-RFLP) analysis, while the rs4364254 was detected by allele-specific amplification. RESULTS: The recipient-donor discrepancy for rs4364254 HPSE SNP was significantly associated with grade II-IV aGvHD (HR 1.75, p = 0.03). Patients were stratified into risk groups as follows: low-risk group (LDR) including TT-TT, TT-CT, CT-TT, CC-CC; high-risk group (HDR) including CC-CT, CC-TT, CT-CC, CT-CT, TT-CC. Day 100 cumulative incidence of grade II-IV aGvHD was 23.4% in the LDR group and 41.4% in the HDR group (p = 0.01). One-year cumulative incidence of moderate/severe cGvHD was 42.6% in the LDR group and 58.6% in the HDR group (p = 0.04). Independent variables for moderate/severe cGvHD in patients who received myeloablative conditioning included donor rs4693608 SNP (GA/AA vs. GG: HR 6.86, p = 0.008), rs4693608-rs4364254 SNP combination in recipient (HR/MR vs. LR: HR 3.67, p = 0.01), and previous grade II-IV aGvHD (HR 3.28, p = 0.0005). Finally, donors with rs4364254 SNP CC conferred increased transplant-related mortality (TRM) (39.1% vs. 25%, p = 0.03) and decreased graft-relapse free survival (GRFS) (23.5% vs. 34.4%, p = 0.04) compared with CT or TT genotypes. CONCLUSION: The differences in incidence of GvHD according to recipient-donor genotype combinations suggests a possible role for rs4364254 HPSE SNP in predicting GvHD. A high level of HPSE, particularly linked to CC genotype of rs4364254 SNP may promote alloreactive T lymphocytes activation and migration toward target organs.


Asunto(s)
Enfermedad Injerto contra Huésped , Trasplante de Células Madre Hematopoyéticas , Humanos , Polimorfismo de Nucleótido Simple , Trasplante de Células Madre/efectos adversos , Trasplante de Células Madre Hematopoyéticas/efectos adversos , Enfermedad Injerto contra Huésped/genética
6.
Cells ; 12(6)2023 03 16.
Artículo en Inglés | MEDLINE | ID: mdl-36980254

RESUMEN

Multiple myeloma (MM) is a plasma cell malignancy that is accompanied by hypercalcemia, renal failure, anemia, and lytic bone lesions. Heparanase (HPSE) plays an important role in supporting and promoting myeloma progression, maintenance of plasma cell stemness, and resistance to therapy. Previous studies identified functional single nucleotide polymorphisms (SNPs) located in the HPSE gene. In the present study, 5 functional HPSE SNPs and 11 novel HPSE2 SNPs were examined. A very significant association between two enhancer (rs4693608 and rs4693084), and two insulator (rs4364254 and rs4426765) HPSE SNPs and primary paraskeletal disease (PS) was observed. SNP rs657442, located in intron 9 of the HPSE2 gene, revealed a significant protective association with primary paraskeletal disease and lytic bone lesions. The present study demonstrates a promoting (HPSE gene) and protective (HPSE2 gene) role of gene regulatory elements in the development of paraskeletal disease and bone morbidity. The effect of signal discrepancy between myeloma cells and normal cells of the tumor microenvironment is proposed as a mechanism for the involvement of heparanase in primary PS. We suggest that an increase in heparanase-2 expression can lead to effective suppression of heparanase activity in multiple myeloma accompanied by extramedullary and osteolytic bone disease.


Asunto(s)
Glucuronidasa , Mieloma Múltiple , Humanos , Enfermedades Óseas/genética , Glucuronidasa/genética , Intrones , Mieloma Múltiple/genética , Polimorfismo de Nucleótido Simple/genética , Microambiente Tumoral
7.
Anticancer Agents Med Chem ; 23(4): 470-477, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-35980070

RESUMEN

BACKGROUND: Triple-negative breast cancer (TNBC) is considered to be the most malignant subtype of breast cancer (BC). Heparanase (HPSE) has been reported to contribute to tumor development, but its potential function in TNBC is not clear. The intention of this study was to investigate whether HPSE affects TNBC progression and to explore the possible mechanisms. METHODS: Bioinformatics analyses were applied to analyze the expression of HPSE in TNBC samples and normal breast samples. The mRNA and protein levels of HPSE in TNBC cells were detected by RT-qPCR and western blot. Function assays, including CCK-8 assay, colony formation assay, transwell assay and wound healing assay, were conducted to validate the effects of HPSE silencing on TNBC cell proliferation and migration. Mechanism experiments were performed to explore the upstream molecular mechanism of HPSE in TNBC cells. RESULTS: Silencing of HPSE suppressed the proliferation and migration of TNBC cells. Moreover, hypoxia-inducible factor-1 alpha (HIF-1α) interacted with the HPSE promoter and promoted the transcription of HPSE. Isoproterenol (ISO), a pharmacological substitute for chronic stress-induced sympathetic activation, was proven to induce HIF-1α upregulation, so as to transcriptionally activate HPSE in TNBC cells. Furthermore, it manifested that ISO facilitated TNBC cell proliferation and migration in an HPSE-dependent way. CONCLUSION: HPSE activated by ISO-induced HIF-1α promoted TNBC cell proliferation and migration, which might offer a novel insight for TNBC treatment.


Asunto(s)
Neoplasias de la Mama Triple Negativas , Humanos , Neoplasias de la Mama Triple Negativas/patología , Isoproterenol , Regulación hacia Arriba , Proliferación Celular , Movimiento Celular , Línea Celular Tumoral , Regulación Neoplásica de la Expresión Génica
9.
Front Cell Dev Biol ; 10: 742213, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36340029

RESUMEN

Background: Oral cavity cancer is still an important public health problem throughout the world. Oral squamous cell carcinomas (OSCCs) can be quite aggressive and metastatic, with a low survival rate and poor prognosis. However, this is usually related to the clinical stage and histological grade, and molecular prognostic markers for clinical practice are yet to be defined. Heparanase (HPSE1) is an endoglycosidase associated with extracellular matrix remodeling, and although involved in several malignancies, the clinical implications of HPSE1 expression in OSCCs are still unknown. Methods: We sought to investigate HPSE1 expression in a series of primary OSCCs and further explore whether its overexpression plays a relevant role in OSCC tumorigenesis. mRNA and protein expression analyses were performed in OSCC tissue samples and cell lines. A loss-of-function strategy using shRNA and a gain-of-function strategy using an ORF vector targeting HPSE1 were employed to investigate the endogenous modulation of HPSE1 and its effects on proliferation, apoptosis, adhesion, epithelial-mesenchymal transition (EMT), angiogenesis, migration, and invasion of oral cancer in vitro. Results: We demonstrated that HPSE1 is frequently upregulated in OSCC samples and cell lines and is an unfavorable prognostic indicator of disease-specific survival when combined with advanced pT stages. Moreover, abrogation of HPSE1 in OSCC cells significantly promoted apoptosis and inhibited proliferation, migration, invasion, and epithelial-mesenchymal transition by significantly decreasing the expression of N-cadherin and vimentin. Furthermore, a conditioned medium of HPSE1-downregulated cells resulted in reduced vascular endothelial growth. Conclusion: Our results confirm the overexpression of HPSE1 in OSCCs, suggest that HPSE1 expression correlates with disease progression as it is associated with several important biological processes for oral tumorigenesis, and can be managed as a prognostic marker for patients with OSCC.

10.
Front Genet ; 13: 896125, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35812751

RESUMEN

Urofacial (also called Ochoa) syndrome (UFS) is an autosomal recessive congenital disorder of the urinary bladder featuring voiding dysfunction and a grimace upon smiling. Biallelic variants in HPSE2, coding for the secreted protein heparanase-2, are described in around half of families genetically studied. Hpse2 mutant mice have aberrant bladder nerves. We sought to expand the genotypic spectrum of UFS and make insights into its pathobiology. Sanger sequencing, next generation sequencing and microarray analysis were performed in four previously unreported families with urinary tract disease and grimacing. In one, the proband had kidney failure and was homozygous for the previously described pathogenic variant c.429T>A, p.(Tyr143*). Three other families each carried a different novel HPSE2 variant. One had homozygous triplication of exons 8 and 9; another had homozygous deletion of exon 4; and another carried a novel c.419C>G variant encoding the missense p.Pro140Arg in trans with c.1099-1G>A, a previously reported pathogenic splice variant. Expressing the missense heparanase-2 variant in vitro showed that it was secreted as normal, suggesting that 140Arg has aberrant functionality after secretion. Bladder autonomic neurons emanate from pelvic ganglia where resident neural cell bodies derive from migrating neural crest cells. We demonstrated that, in normal human embryos, neuronal precursors near the developing hindgut and lower urinary tract were positive for both heparanase-2 and leucine rich repeats and immunoglobulin like domains 2 (LRIG2). Indeed, biallelic variants of LRIG2 have been implicated in rare UFS families. The study expands the genotypic spectrum in HPSE2 in UFS and supports a developmental neuronal pathobiology.

11.
Gene ; 837: 146654, 2022 Aug 30.
Artículo en Inglés | MEDLINE | ID: mdl-35718240

RESUMEN

Ribosomal protein S23 (RPS23) and Heparanase (HPSE) were located on chromosome 5 and chromosome 6, respectively, which play vital roles in protein synthesis and immunity. The objective of this study was to clone RPS23 and HPSE and to detect the expression levels of RPS23 and HPSE and the polymorphisms of RPS23 and HPSE associated with the hematologic parameters by using qRT-PCR, DNA sequencing and KASPar assay. The quantitative real-time PCR (RT-qPCR) showed that the two genes were expressed widely in the ten tissues of sheep. The expression levels of RPS23 and HPSE were the highest in lung and liver, respectively. The expression levels of RPS23 and HPSE in lung and liver increased from 0 to 3 months, decreased from 3 to 6 months, respectively. Furthermore, two mutations g.720 A > G and g.1077 G > A were detected in the RPS23 and HPSE, respectively, which were confirmed to be significantly associated with hematologic parameters. These results supported RPS23 g.720 A > G and HPSE g.1077 G > A as genetic markers of sheep.


Asunto(s)
Glucuronidasa , Neoplasias Hepáticas , Animales , Glucuronidasa/genética , Neoplasias Hepáticas/genética , Ovinos/genética
12.
Front Immunol ; 13: 916512, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35757776

RESUMEN

Coronavirus disease 2019 (COVID-19) is a systemic disease associated with injury (thinning) of the endothelial glycocalyx (eGC), a protective layer on the vascular endothelium. The aim of this translational study was to investigate the role of the eGC-degrading enzyme heparanase (HPSE), which is known to play a central role in the destruction of the eGC in bacterial sepsis. Excess activity of HPSE in plasma from COVID-19 patients correlated with several markers of eGC damage and perfused boundary region (PBR, an inverse estimate of glycocalyx dimensions of vessels with a diameter 4-25 µm). In a series of translational experiments, we demonstrate that the changes in eGC thickness of cultured cells exposed to COVID-19 serum correlated closely with HPSE activity in concordant plasma samples (R = 0.82, P = 0.003). Inhibition of HPSE by a nonanticoagulant heparin fragment prevented eGC injury in response to COVID-19 serum, as shown by atomic force microscopy and immunofluorescence imaging. Our results suggest that the protective effect of heparin in COVID-19 may be due to an eGC-protective off-target effect.


Asunto(s)
COVID-19 , Glucuronidasa , Glicocálix , COVID-19/metabolismo , COVID-19/patología , Glucuronidasa/metabolismo , Glicocálix/metabolismo , Glicocálix/patología , Heparina/farmacología , Humanos
13.
Int J Mol Sci ; 23(9)2022 Apr 21.
Artículo en Inglés | MEDLINE | ID: mdl-35563015

RESUMEN

It has been accepted for decades that T lymphocytes and metastasising tumour cells traverse basement membranes (BM) by deploying a battery of degradative enzymes, particularly proteases. However, since many redundant proteases can solubilise BM it has been difficult to prove that proteases aid cell migration, particularly in vivo. Recent studies also suggest that other mechanisms allow BM passage of cells. To resolve this issue we exploited heparanase-1 (HPSE-1), the only endoglycosidase in mammals that digests heparan sulfate (HS), a major constituent of BM. Initially we examined the effect of HPSE-1 deficiency on a well-characterised adoptive transfer model of T-cell-mediated inflammation. We found that total elimination of HPSE-1 from this system resulted in a drastic reduction in tissue injury and loss of target HS. Subsequent studies showed that the source of HPSE-1 in the transferred T cells was predominantly activated CD4+ T cells. Based on bone marrow chimeras, two cellular sources of HPSE-1 were identified in T cell recipients, one being haematopoiesis dependent and the other radiation resistant. Collectively our findings unequivocally demonstrate that an acute T-cell-initiated inflammatory response is HPSE-1 dependent and is reliant on HPSE-1 from at least three different cell types.


Asunto(s)
Glicósido Hidrolasas , Linfocitos T , Animales , Glucuronidasa/genética , Glucuronidasa/metabolismo , Heparitina Sulfato/metabolismo , Inflamación , Mamíferos/metabolismo , Péptido Hidrolasas , Linfocitos T/metabolismo
14.
Matrix Biol Plus ; 14: 100106, 2022 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-35392182

RESUMEN

The vascular endothelium is the interface between circulating blood and end organs and thus has a critical role in preserving organ function. The endothelium is lined by a glycan-rich glycocalyx that uniquely contributes to endothelial function through its regulation of leukocyte and platelet interactions with the vessel wall, vascular permeability, coagulation, and vasoreactivity. Degradation of the endothelial glycocalyx can thus promote vascular dysfunction, inflammation propagation, and organ injury. The endothelial glycocalyx and its role in vascular pathophysiology has gained increasing attention over the last decade. While studies characterizing vascular glycocalyx injury and its downstream consequences in a host of adult human diseases and in animal models has burgeoned, studies evaluating glycocalyx damage in pediatric diseases are relatively few. As children have unique physiology that differs from adults, significant knowledge gaps remain in our understanding of the causes and effects of endothelial glycocalyx disintegrity in pediatric critical illness. In this narrative literature overview, we offer a unique perspective on the role of the endothelial glycocalyx in pediatric critical illness, drawing from adult and preclinical data in addition to pediatric clinical experience to elucidate how marked derangement of the endothelial surface layer may contribute to aberrant vascular biology in children. By calling attention to this nascent field, we hope to increase research efforts to address important knowledge gaps in pediatric vascular biology that may inform the development of novel therapeutic strategies.

15.
Matrix Biol Plus ; 12: 100094, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-34917925

RESUMEN

The glycocalyx is a ubiquitous structure found on endothelial cells that extends into the vascular lumen. It is enriched in proteoglycans, which are proteins attached to the glycosaminoglycans heparan sulfate, chondroitin sulfate, dermatan sulfate, keratan sulfate, and hyaluronic acid. In health and disease, the endothelial glycocalyx is a central regulator of vascular permeability, inflammation, coagulation, and circulatory tonicity. During sepsis, a life-threatening syndrome seen commonly in hospitalized patients, the endothelial glycocalyx is degraded, significantly contributing to its many clinical manifestations. In this review we discuss the intrinsically linked mechanisms responsible for septic endothelial glycocalyx destruction: glycosaminoglycan degradation and proteoglycan cleavage. We then examine the consequences of local endothelial glycocalyx loss to several organ systems and the systemic consequences of shed glycocalyx constituents. Last, we explore clinically relevant non-modifiable and modifiable factors that exacerbate or protect against endothelial glycocalyx shedding during sepsis.

16.
Cells ; 10(12)2021 12 10.
Artículo en Inglés | MEDLINE | ID: mdl-34943994

RESUMEN

Heparanase is an endo-ß-glucuronidase that is best known for its pro-cancerous effects but is also implicated in the pathogenesis of various viruses. Activation of heparanase is a common strategy to increase viral spread and trigger the subsequent inflammatory cascade. Using a Single Nucleotide Polymorphisms (SNP)-associated approach we identified enhancer and insulator regions that regulate HPSE expression. Although a role for heparanase in viral infection has been noticed, the impact of HPSE functional SNPs has not been determined. We investigated the effect of cytomegalovirus (CMV) serostatus on the involvement of HPSE enhancer and insulator functional SNPs in the risk of acute graft versus host disease (GVHD) and granulocyte-colony stimulating factor related CD34+ mobilization. A significant correlation between the C alleles of insulator rs4364254 and rs4426765 and CMV seropositivity was found in healthy donors and patients with hematological malignancies. The risk of developing acute GVHD after hematopoietic stem cell transplantation was identified only in CMV-seropositive patients. A significant correlation between the enhancer rs4693608 and insulator rs28649799 and CD34+ cell mobilization was demonstrated in the CMV-seropositive donors. It is thus conceivable that latent CMV infection modulates heparanase regulatory regions and enhances the effect of functional SNPs on heparanase function in normal and pathological processes.


Asunto(s)
Antígenos CD34/metabolismo , Citomegalovirus/fisiología , Glucuronidasa/genética , Enfermedad Injerto contra Huésped/genética , Enfermedad Injerto contra Huésped/virología , Movilización de Célula Madre Hematopoyética , Polimorfismo de Nucleótido Simple/genética , Enfermedad Aguda , Infecciones por Citomegalovirus/genética , Infecciones por Citomegalovirus/virología , Femenino , Frecuencia de los Genes/genética , Factor Estimulante de Colonias de Granulocitos/farmacología , Humanos , Masculino , Factores de Riesgo , Donantes de Tejidos
17.
Bioengineered ; 12(2): 11267-11276, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34753397

RESUMEN

Wound healing is related to proliferation, migration, and angiogenesis of keratinocytes. Insulin-like growth factor 2 mRNA binding protein 2 (IGF2BP2) is an important N6-methyladenosine (m6A) reader, which is involved in multiple processes, including wound healing. However, the function and mechanism of IGF2BP2 in keratinocyte processes are largely uncertain. In the present study, expression levels of IGF2BP2 and heparanase (HPSE) were detected by quantitative reverse transcription polymerase chain reaction and western blotting assays. Cell proliferation was investigated by cell counting kit-8 (CCK-8) analysis. Cell migration was determined through wound healing assay. Angiogenesis was measured by tube formation assay and vascular endothelial growth factor (VEGF) level using enzyme linked immunosorbent assay (ELISA). The interaction between IGF2BP2 and HPSE was analyzed by RNA immunoprecipitation, pull-down and luciferase reporter analyses. The results showed that IGF2BP2 expression was enhanced in wound healing. IGF2BP2 downregulation constrained HaCaT cell proliferation, migration, and angiogenesis. IGF2BP2 knockdown decreased HPSE expression. IGF2BP2 could regulate HPSE stability by binding with 3' untranslated region (UTR) of HPSE. HPSE upregulation attenuated silencing IGF2BP2-mediated suppression of proliferation, migration, and angiogenesis. As a conclusion, IGF2BP2 knockdown repressed proliferation, migration, and angiogenesis of HaCaT cells by decreasing HPSE stability.


Asunto(s)
Movimiento Celular , Glucuronidasa/metabolismo , Queratinocitos/metabolismo , Queratinocitos/patología , Neovascularización Patológica/metabolismo , Proteínas de Unión al ARN/metabolismo , Regiones no Traducidas 3'/genética , Movimiento Celular/genética , Proliferación Celular/genética , Estabilidad de Enzimas , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Técnicas de Silenciamiento del Gen , Células HaCaT , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Neovascularización Patológica/genética , Unión Proteica , Cicatrización de Heridas/genética
18.
Cells ; 10(10)2021 09 23.
Artículo en Inglés | MEDLINE | ID: mdl-34685503

RESUMEN

The HPSE gene encodes heparanase (HPSE), a key player in cancer, inflammation, and autoimmunity. We have previously identified a strong HPSE gene enhancer involved in self-regulation of heparanase by negative feedback exerted in a functional rs4693608 single-nucleotide polymorphism (SNP) dependent manner. In the present study, we analyzed the HPSE gene insulator region, located in intron 9 and containing rs4426765, rs28649799, and rs4364254 SNPs. Our results indicate that this region exhibits HPSE regulatory activity. SNP substitutions lead to modulation of a unique DNA-protein complex that affects insulator activity. Analysis of interactions between enhancer and insulator SNPs revealed that rs4693608 has a major effect on HPSE expression and the risk of post-transplantation acute graft versus host disease (GVHD). The C alleles of insulator SNPs rs4364254 and rs4426765 modify the activity of the HPSE enhancer, resulting in altered HPSE expression and increased risk of acute GVHD. Moreover, rs4426765 correlated with HPSE expression in activated mononuclear cells, as well as with CD3 levels and lymphocyte counts following G-CSF mobilization. rs4363084 and rs28649799 were found to be associated with CD34+ levels. Our study provides new insight into the mechanism of HPSE gene regulation and its impact on normal and pathological processes in the hematopoietic system.


Asunto(s)
Regulación de la Expresión Génica/genética , Glucuronidasa/metabolismo , Enfermedad Injerto contra Huésped/genética , Neoplasias/genética , Células Madre/citología , Alelos , Regulación de la Expresión Génica/fisiología , Frecuencia de los Genes/genética , Genotipo , Movilización de Célula Madre Hematopoyética/métodos , Humanos
19.
Acta Histochem ; 123(7): 151790, 2021 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-34592492

RESUMEN

MicroRNAs (miRNAs) are involved in bone remodeling by regulating the balance of bone formation and resorption. Increasing evidence has confirmed that the communication between osteoclast and osteoblast through secreting exosomes and transferring miRNAs. It has been reported that mineralized osteoblasts release exosomes containing more miR-503-3p. However, the roles and molecular mechanisms of osteoblast exosomes-derived miR-503-3p in osteoclast differentiation remain elusive. Here, we isolated exosomes from the supernatant of osteoblasts and identified the exosome characterization through transmission electron microscopy (TEM), nanoparticle tracking analysis (NTA), and western blot assay. In addition, we found that exosomes and miR-503-3p secreted by osteoblasts inhibited the differentiation of osteoclast progenitor cells. Meanwhile, we found that Hpse (heparanase gene) was a target gene of miR-503-3p and miR-503-3p inhibited the osteoclast differentiation through downregulating the expression of Hpse. In summary, our results demonstrated the roles and the mechanism of osteoblast-derived exosomes inhibited the osteoclast differentiation via miR-503-3p/Hpse axis.


Asunto(s)
Comunicación Celular , Diferenciación Celular , Exosomas/metabolismo , Glucuronidasa/metabolismo , MicroARNs/metabolismo , Osteoblastos/metabolismo , Osteoclastos/metabolismo , Transducción de Señal , Animales , Exosomas/ultraestructura , Glucuronidasa/genética , Ratones , MicroARNs/genética , Osteoblastos/ultraestructura , Osteoclastos/ultraestructura , Células RAW 264.7
20.
Anticancer Res ; 41(7): 3299-3308, 2021 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-34230125

RESUMEN

BACKGROUND/AIM: Heparanase (HPSE) is relevant to therapy resistance in many malignancies yet is largely unstudied in Hodgkin's lymphoma. Here, we investigated links between HPSE, cancer stem cell (CSC) features and radioresistance in KM-H2 and L428 Hodgkin's lymphoma cells. MATERIALS AND METHODS: Firstly, HPSE expression in unsorted and sorted CSCs was assessed. Post-irradiation, HPSE and CSC-related gene expression changes were then quantified. Clonogenic ability was investigated with and without artificial changes in HPSE expression pre and post irradiation. RESULTS: HPSE was highly expressed in L428 but barely present in KM-H2 cells. HPSE was overexpressed in sorted L428 CSCs. Irradiation induced HPSE and expression of CSC markers. High HPSE-expressing L428 cells showed higher clonogenic ability than low HPSE-expressing KM-H2 cells after irradiation. Down-regulation of HPSE in L428 cells reduced their clonogenic capability post-radiation, whilst overexpression of HPSE in KM-H2 cells increased colony formation. CONCLUSION: HPSE expression is associated with CSC features and contributes to radioresistance in Hodgkin's lymphoma cells.


Asunto(s)
Glucuronidasa/metabolismo , Enfermedad de Hodgkin/metabolismo , Células Madre Neoplásicas/metabolismo , Tolerancia a Radiación/fisiología , Adulto , Línea Celular Tumoral , Regulación hacia Abajo/fisiología , Femenino , Regulación Neoplásica de la Expresión Génica/fisiología , Humanos , Masculino
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...