Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 7.802
Filtrer
1.
Sci Adv ; 10(23): eadj4735, 2024 Jun 07.
Article de Anglais | MEDLINE | ID: mdl-38838150

RÉSUMÉ

Why individuals with Down syndrome (DS) are more susceptible to SARS-CoV-2-induced neuropathology remains elusive. Choroid plexus (ChP) plays critical roles in barrier function and immune response modulation and expresses the ACE2 receptor and the chromosome 21-encoded TMPRSS2 protease, suggesting its substantial role in establishing SARS-CoV-2 infection in the brain. To explore this, we established brain organoids from DS and isogenic euploid iPSC that consist of a core of functional cortical neurons surrounded by a functional ChP-like epithelium (ChPCOs). DS-ChPCOs recapitulated abnormal DS cortical development and revealed defects in ciliogenesis and epithelial cell polarity in ChP-like epithelium. We then demonstrated that the ChP-like epithelium facilitates infection and replication of SARS-CoV-2 in cortical neurons and that this is increased in DS. Inhibiting TMPRSS2 and furin activity reduced viral replication in DS-ChPCOs to euploid levels. This model enables dissection of the role of ChP in neurotropic virus infection and euploid forebrain development and permits screening of therapeutics for SARS-CoV-2-induced neuropathogenesis.


Sujet(s)
Encéphale , COVID-19 , Plexus choroïde , Syndrome de Down , Organoïdes , SARS-CoV-2 , Serine endopeptidases , Plexus choroïde/virologie , Plexus choroïde/métabolisme , Plexus choroïde/anatomopathologie , Organoïdes/virologie , Organoïdes/métabolisme , Organoïdes/anatomopathologie , Humains , SARS-CoV-2/physiologie , COVID-19/virologie , COVID-19/anatomopathologie , COVID-19/métabolisme , Serine endopeptidases/métabolisme , Serine endopeptidases/génétique , Syndrome de Down/métabolisme , Syndrome de Down/anatomopathologie , Syndrome de Down/génétique , Encéphale/virologie , Encéphale/anatomopathologie , Encéphale/métabolisme , Neurones/métabolisme , Neurones/virologie , Neurones/anatomopathologie , Réplication virale , Cellules souches pluripotentes induites/métabolisme , Cellules souches pluripotentes induites/virologie , Furine/métabolisme , Furine/génétique , Angiotensin-converting enzyme 2/métabolisme , Angiotensin-converting enzyme 2/génétique , Tropisme viral
2.
Anal Cell Pathol (Amst) ; 2024: 8810804, 2024.
Article de Anglais | MEDLINE | ID: mdl-38826849

RÉSUMÉ

Head and neck squamous cell carcinoma (HNSCC) poses significant challenges with poor survival rates and limited therapeutic strategies. Our study, using The Cancer Genome Atlas (TCGA) data, assesses cancer-associated fibroblast (CAF) gene signatures' clinical relevance. In our analysis across TCGA tumor types, differential gene expression analysis revealed that fibroblast activation protein (FAP) is upregulated in tumor tissues and associated with poorer survival rates in HNSCC. Furthermore, mechanistic studies employing gene-silencing techniques substantiated that FAP knockout led to a significant decrease in cellular proliferation, invasion, and migration in HNSCC cell lines. Through Gene Ontology and Kyoto Encyclopedia of Genes and Genomes enrichment analyses, we established that high FAP expression correlates with vital biological processes such as extracellular matrix organization, angiogenesis, and cellular motility. Importantly, FAP was found to regulate these processes by promoting the expression of key proteins involved in epithelial-mesenchymal transition-related pathways. Additionally, our analysis revealed a significant correlation between FAP expression and the expression profiles of immune checkpoint molecules, underscoring its potential role in immune modulation. Collectively, our findings illuminate FAP's pivotal role in HNSCC pathogenesis and its potential as a prognostic biomarker and therapeutic target. This research lays the groundwork for understanding the multifaceted roles and regulatory mechanisms of CAFs in HNSCC, thereby offering valuable perspectives for the development of targeted therapeutic strategies aimed at improving patient outcomes.


Sujet(s)
Marqueurs biologiques tumoraux , Endopeptidases , Gelatinases , Régulation de l'expression des gènes tumoraux , Tumeurs de la tête et du cou , Protéines membranaires , Serine endopeptidases , Carcinome épidermoïde de la tête et du cou , Humains , Marqueurs biologiques tumoraux/métabolisme , Marqueurs biologiques tumoraux/génétique , Pronostic , Carcinome épidermoïde de la tête et du cou/génétique , Carcinome épidermoïde de la tête et du cou/métabolisme , Carcinome épidermoïde de la tête et du cou/anatomopathologie , Endopeptidases/métabolisme , Endopeptidases/génétique , Serine endopeptidases/métabolisme , Serine endopeptidases/génétique , Protéines membranaires/métabolisme , Protéines membranaires/génétique , Lignée cellulaire tumorale , Tumeurs de la tête et du cou/génétique , Tumeurs de la tête et du cou/métabolisme , Tumeurs de la tête et du cou/anatomopathologie , Gelatinases/métabolisme , Gelatinases/génétique , Transition épithélio-mésenchymateuse/génétique , Prolifération cellulaire/génétique , Mouvement cellulaire/génétique
3.
Signal Transduct Target Ther ; 9(1): 142, 2024 Jun 03.
Article de Anglais | MEDLINE | ID: mdl-38825657

RÉSUMÉ

Radiotherapy combined with immune checkpoint blockade holds great promise for synergistic antitumor efficacy. Targeted radionuclide therapy delivers radiation directly to tumor sites. LNC1004 is a fibroblast activation protein (FAP)-targeting radiopharmaceutical, conjugated with the albumin binder Evans Blue, which has demonstrated enhanced tumor uptake and retention in previous preclinical and clinical studies. Herein, we demonstrate that 68Ga/177Lu-labeled LNC1004 exhibits increased uptake and prolonged retention in MC38/NIH3T3-FAP and CT26/NIH3T3-FAP tumor xenografts. Radionuclide therapy with 177Lu-LNC1004 induced a transient upregulation of PD-L1 expression in tumor cells. The combination of 177Lu-LNC1004 and anti-PD-L1 immunotherapy led to complete eradication of all tumors in MC38/NIH3T3-FAP tumor-bearing mice, with mice showing 100% tumor rejection upon rechallenge. Immunohistochemistry, single-cell RNA sequencing (scRNA-seq), and TCR sequencing revealed that combination therapy reprogrammed the tumor microenvironment in mice to foster antitumor immunity by suppressing malignant progression and increasing cell-to-cell communication, CD8+ T-cell activation and expansion, M1 macrophage counts, antitumor activity of neutrophils, and T-cell receptor diversity. A preliminary clinical study demonstrated that 177Lu-LNC1004 was well-tolerated and effective in patients with refractory cancers. Further, scRNA-seq of peripheral blood mononuclear cells underscored the importance of addressing immune evasion through immune checkpoint blockade treatment. This was emphasized by the observed increase in antigen processing and presentation juxtaposed with T cell inactivation. In conclusion, our data supported the efficacy of immunotherapy combined with 177Lu-LNC1004 for cancer patients with FAP-positive tumors.


Sujet(s)
Inhibiteurs de points de contrôle immunitaires , Animaux , Souris , Inhibiteurs de points de contrôle immunitaires/pharmacologie , Humains , Protéines membranaires/génétique , Protéines membranaires/immunologie , Microenvironnement tumoral/effets des médicaments et des substances chimiques , Microenvironnement tumoral/immunologie , Endopeptidases/génétique , Cellules NIH 3T3 , Radiopharmaceutiques/usage thérapeutique , Serine endopeptidases/génétique , Serine endopeptidases/immunologie , Tests d'activité antitumorale sur modèle de xénogreffe , Immunothérapie , Gelatinases/génétique , Gelatinases/immunologie , Lutétium/pharmacologie , Lignée cellulaire tumorale
4.
Croat Med J ; 65(3): 220-231, 2024 Jun 13.
Article de Anglais | MEDLINE | ID: mdl-38868968

RÉSUMÉ

AIM: To assess the association of single nucleotide polymorphisms (SNPs) in the ACE2 and TMPRSS2 genes with COVID-19 severity and key biomarkers. METHODS: The study involved 750 COVID-19 patients from Bosnia and Herzegovina, divided into three groups: mild, moderate, and severe cases. Genetic variations within the ACE2 (rs2285666) and TMPRSS2 (rs2070788) genes were examined with real-time polymerase chain reaction. Biochemical markers were determined with standard procedures. RESULTS: There was a significant difference in the rs2070788 genotype distribution between patients with mild and moderate symptoms, but not between other groups. For the rs2285666 polymorphism, no significant difference in genotype distribution was found. In patients with mild symptoms, carriers of the GG genotype of rs2070788 had significantly higher total bilirubin levels than carriers of the AA genotype. Similarly, carriers of the TT genotype of rs2285666 had significantly higher activated partial thromboplastin time and international normalized ratio, and lower lactate dehydrogenase levels compared with the CC genotype. Among patients with severe symptoms, carriers of the GG genotype showed significantly higher potassium levels than carriers of the AA genotype, while carriers of the TT genotype showed significantly higher erythrocyte count as well as hemoglobin and hematocrit levels compared with the CC genotype. CONCLUSION: This study highlights the role of genetic factors, particularly SNPs in the ACE2 and TMPRSS2 genes, in determining COVID-19 severity, aiding patient risk assessment and prognosis.


Sujet(s)
Angiotensin-converting enzyme 2 , Marqueurs biologiques , COVID-19 , Polymorphisme de nucléotide simple , Serine endopeptidases , Indice de gravité de la maladie , Humains , Serine endopeptidases/génétique , COVID-19/génétique , COVID-19/épidémiologie , Angiotensin-converting enzyme 2/génétique , Mâle , Femelle , Bosnie-et-Herzégovine , Adulte d'âge moyen , Marqueurs biologiques/sang , SARS-CoV-2/génétique , Adulte , Sujet âgé , Génotype
5.
PLoS One ; 19(6): e0299445, 2024.
Article de Anglais | MEDLINE | ID: mdl-38870131

RÉSUMÉ

It has been postulated from a combination of evidence that a sudden increase in COVID-19 cases among pediatric patients after onset of the Omicron wave was attributed to a reduced requirement for TMPRSS2-mediated entry in pediatric airways with lower expression levels of TMPRSS2. Epidemic strains were isolated from the indigenous population in an area, and the levels of TMPRSS2 required for Delta and Omicron variants were assessed. As a result, Delta variants proliferated fully in cultures of TMPRSS2-positive Vero cells but not in TMPRSS2-negative Vero cell culture (350-fold, Delta vs 9.6-fold, Omicron). There was no obvious age-dependent selection of Omicron strains affected by the TMPRSS2 (9.6-fold, Adults vs. 12-fold, Children). A phylogenetic tree was generated and Blast searches (up to 100 references) for the spread of strains in the study area showed that each strain had almost identical homology (>99.5%) with foreign isolates, although indigenous strains had obvious differences from each other. This suggested that the differences had been present abroad for a long period. Therefore, the lower requirement for TMPRSS2 by Omicron strains might be applicable to epidemic strains globally. In conclusion, the property of TMPRSS2-independent cleavage makes Omicron proliferate with ease and allows epidemics among children with fewer TMPRSS2 on epithelial surfaces of the respiratory organs.


Sujet(s)
COVID-19 , SARS-CoV-2 , Serine endopeptidases , SARS-CoV-2/génétique , SARS-CoV-2/isolement et purification , Humains , Serine endopeptidases/génétique , Serine endopeptidases/métabolisme , Chlorocebus aethiops , COVID-19/virologie , COVID-19/métabolisme , COVID-19/épidémiologie , Cellules Vero , Enfant , Animaux , Enfant d'âge préscolaire , Phylogenèse , Adulte , Nourrisson , Mâle , Femelle
6.
Am J Pathol ; 194(7): 1162-1170, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38880601

RÉSUMÉ

The placenta plays a crucial role in pregnancy success. ΔNp63α (p63), a transcription factor from the TP53 family, is highly expressed in villous cytotrophoblasts (CTBs), the epithelial stem cells of the human placenta, and is involved in CTB maintenance and differentiation. We examined the mechanisms of action of p63 by identifying its downstream targets. Gene expression changes were evaluated following overexpression and knockdown of p63 in the JEG3 choriocarcinoma cell line, using microarray-based RNA profiling. High-temperature requirement A4 (HTRA4), a placenta-specific serine protease involved in trophoblast differentiation and altered in preeclampsia, was identified as a gene reciprocally regulated by p63, and its expression was characterized in primary human placental tissues by RNA-sequencing and in situ hybridization. Potential p63 DNA-binding motifs were identified in the HTRA4 promoter, and p63 occupancy at some of these sites was confirmed using chromatin immunoprecipitation, followed by quantitative PCR in both JEG3 and trophoblast stem cells. These data begin to identify members of the transcriptional network downstream of p63, thus laying the groundwork for probing mechanisms by which this important transcription factor regulates trophoblast stemness and differentiation.


Sujet(s)
Facteurs de transcription , Trophoblastes , Humains , Trophoblastes/métabolisme , Femelle , Grossesse , Facteurs de transcription/métabolisme , Facteurs de transcription/génétique , Serine endopeptidases/métabolisme , Serine endopeptidases/génétique , Différenciation cellulaire/génétique , Lignée cellulaire tumorale , Protéines suppresseurs de tumeurs/métabolisme , Protéines suppresseurs de tumeurs/génétique , Placenta/métabolisme , Protéases à sérine/métabolisme , Protéases à sérine/génétique , Régions promotrices (génétique)/génétique , Pré-éclampsie/génétique , Pré-éclampsie/métabolisme , Pré-éclampsie/anatomopathologie , Transcription génétique
7.
Respir Res ; 25(1): 238, 2024 Jun 11.
Article de Anglais | MEDLINE | ID: mdl-38862975

RÉSUMÉ

BACKGROUND: TMPRSS2, a key molecule for SARS-CoV-2 invading human host cells, has an association with cancer. However, its association with lung cancer remains insufficiently unexplored. METHODS: In five bulk transcriptomics datasets, one single-cell RNA sequencing (scRNA-seq) dataset and one proteomics dataset for lung adenocarcinoma (LUAD), we explored associations between TMPRSS2 expression and immune signatures, tumor progression phenotypes, genomic features, and clinical prognosis in LUAD by the bioinformatics approach. Furthermore, we performed experimental validation of the bioinformatics findings. RESULTS: TMPRSS2 expression levels correlated negatively with the enrichment levels of both immune-stimulatory and immune-inhibitory signatures, while they correlated positively with the ratios of immune-stimulatory/immune-inhibitory signatures. It indicated that TMPRSS2 levels had a stronger negative correlation with immune-inhibitory than with immune-stimulatory signatures. TMPRSS2 downregulation correlated with increased proliferation, stemness, genomic instability, tumor progression, and worse survival in LUAD. We further validated that TMPRSS2 was downregulated with tumor progression in the LUAD cohort we collected from Jiangsu Cancer Hospital, China. In vitro and in vivo experiments verified the association of TMPRSS2 deficiency with increased tumor cell proliferation and invasion and antitumor immunity in LUAD. Moreover, in vivo experiments demonstrated that TMPRSS2-knockdown tumors were more sensitive to BMS-1, an inhibitor of PD-1/PD-L1. CONCLUSIONS: TMPRSS2 is a tumor suppressor, while its downregulation is a positive biomarker of immunotherapy in LUAD. Our data provide a potential link between lung cancer and pneumonia caused by SARS-CoV-2 infection.


Sujet(s)
Adénocarcinome pulmonaire , Régulation négative , Tumeurs du poumon , Serine endopeptidases , Humains , Adénocarcinome pulmonaire/immunologie , Adénocarcinome pulmonaire/génétique , Adénocarcinome pulmonaire/anatomopathologie , Adénocarcinome pulmonaire/métabolisme , Tumeurs du poumon/immunologie , Tumeurs du poumon/génétique , Tumeurs du poumon/anatomopathologie , Tumeurs du poumon/métabolisme , Serine endopeptidases/génétique , Serine endopeptidases/métabolisme , Animaux , Souris , COVID-19/immunologie , Immunothérapie/méthodes , Régulation de l'expression des gènes tumoraux , Mâle , Gènes suppresseurs de tumeur , Femelle , Souris nude , Lignée cellulaire tumorale , SARS-CoV-2
8.
Front Immunol ; 15: 1348229, 2024.
Article de Anglais | MEDLINE | ID: mdl-38855114

RÉSUMÉ

Introduction: The COVID-19 pandemic represented one of the most significant challenges to researchers and healthcare providers. Several factors determine the disease severity, whereas none alone can explain the tremendous variability. The Single nucleotide variants (SNVs) in angiotensin-converting enzyme-2 (ACE2) and transmembrane serine protease type-2 (TMPRSS2) genes affect the virus entry and are considered possible risk factors for COVID-19. Methods: We compiled a panel of gene variants from both genes and used in-silico analysis to predict their significance. We performed biological validation to assess their capacity to alter the ACE2 interaction with the virus spike protein. Subsequently, we conducted a retrospective comparative genome analysis on those variants in the Emirati patients with different disease severity (total of 96) along with 69 healthy control subjects. Results: Our results showed that the Emirati population lacks the variants that were previously reported as associated with disease severity, whereas a new variant in ACE2 "Chr X:g.15584534" was associated with disease severity specifically among female patients. In-silico analysis revealed that the new variant can determine the ACE2 gene transcription. Several cytokines (GM-CSF and IL-6) and chemokines (MCP-1/CCL2, IL-8/CXCL8, and IP-10/CXCL10) were markedly increased in COVID-19 patients with a significant correlation with disease severity. The newly reported genetic variant of ACE2 showed a positive correlation with CD40L, IL-1ß, IL-2, IL-15, and IL-17A in COVID-19 patients. Conclusion: Whereas COVID-19 represents now a past pandemic, our study underscores the importance of genetic factors specific to a population, which can influence both the susceptibility to viral infections and the level of severity; subsequently expected required preparedness in different areas of the world.


Sujet(s)
Angiotensin-converting enzyme 2 , COVID-19 , Cytokines , Polymorphisme de nucléotide simple , SARS-CoV-2 , Serine endopeptidases , Humains , COVID-19/génétique , Angiotensin-converting enzyme 2/génétique , Femelle , Mâle , SARS-CoV-2/physiologie , Cytokines/sang , Cytokines/génétique , Serine endopeptidases/génétique , Émirats arabes unis/épidémiologie , Adulte d'âge moyen , Adulte , Études rétrospectives , Indice de gravité de la maladie , Sujet âgé
9.
J Med Virol ; 96(6): e29712, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38808555

RÉSUMÉ

Swine acute diarrhea syndrome coronavirus (SADS-CoV) has caused severe intestinal diseases in pigs. It originates from bat coronaviruses HKU2 and has a potential risk of cross-species transmission, raising concerns about its zoonotic potential. Viral entry-related host factors are critical determinants of susceptibility to cells, tissues, or species, and remain to be elucidated for SADS-CoV. Type II transmembrane serine proteases (TTSPs) family is involved in many coronavirus infections and has trypsin-like catalytic activity. Here we examine all 18 members of the TTSPs family through CRISPR-based activation of endogenous protein expression in cells, and find that, in addition to TMPRSS2 and TMPRSS4, TMPRSS13 significantly facilitates SADS-CoV infection. This is confirmed by ectopic expression of TMPRSS13, and specific to trypsin-dependent SADS-CoV. Infection with pseudovirus bearing SADS-CoV spike protein indicates that TMPRSS13 acts at the entry step and is sensitive to serine protease inhibitor Camostat. Moreover, both human and pig TMPRSS13 are able to enhance the cell-cell membrane fusion and cleavage of spike protein. Overall, we demonstrate that TMPRSS13 is another host serine protease promoting the membrane-fusion entry of SADS-CoV, which may expand its host tropism by using diverse TTSPs.


Sujet(s)
Protéines membranaires , Serine endopeptidases , Pénétration virale , Animaux , Serine endopeptidases/métabolisme , Serine endopeptidases/génétique , Suidae , Humains , Protéines membranaires/métabolisme , Protéines membranaires/génétique , Alphacoronavirus/génétique , Alphacoronavirus/physiologie , Infections à coronavirus/virologie , Infections à coronavirus/métabolisme , Gabexate/analogues et dérivés , Gabexate/pharmacologie , Glycoprotéine de spicule des coronavirus/métabolisme , Glycoprotéine de spicule des coronavirus/génétique , Cellules HEK293 , Lignée cellulaire , Chlorocebus aethiops , Maladies des porcs/virologie , Esters , Guanidines
10.
Proc Natl Acad Sci U S A ; 121(23): e2407437121, 2024 Jun 04.
Article de Anglais | MEDLINE | ID: mdl-38814864

RÉSUMÉ

The accessory protease transmembrane protease serine 2 (TMPRSS2) enhances severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) uptake into ACE2-expressing cells, although how increased entry impacts downstream viral and host processes remains unclear. To investigate this in more detail, we performed infection assays in engineered cells promoting ACE2-mediated entry with and without TMPRSS2 coexpression. Electron microscopy and inhibitor experiments indicated TMPRSS2-mediated cell entry was associated with increased virion internalization into endosomes, and partially dependent upon clathrin-mediated endocytosis. TMPRSS2 increased panvariant uptake efficiency and enhanced early rates of virus replication, transcription, and secretion, with variant-specific profiles observed. On the host side, transcriptional profiling confirmed the magnitude of infection-induced antiviral and proinflammatory responses were linked to uptake efficiency, with TMPRSS2-assisted entry boosting early antiviral responses. In addition, TMPRSS2-enhanced infections increased rates of cytopathology, apoptosis, and necrosis and modulated virus secretion kinetics in a variant-specific manner. On the virus side, convergent signatures of cell-uptake-dependent innate immune induction were recorded in viral genomes, manifesting as switches in dominant coupled Nsp3 residues whose frequencies were correlated to the magnitude of the cellular response to infection. Experimentally, we demonstrated that selected Nsp3 mutations conferred enhanced interferon antagonism. More broadly, we show that TMPRSS2 orthologues from evolutionarily diverse mammals facilitate panvariant enhancement of cell uptake. In summary, our study uncovers previously unreported associations, linking cell entry efficiency to innate immune activation kinetics, cell death rates, virus secretion dynamics, and convergent selection of viral mutations. These data expand our understanding of TMPRSS2's role in the SARS-CoV-2 life cycle and confirm its broader significance in zoonotic reservoirs and animal models.


Sujet(s)
COVID-19 , Immunité innée , SARS-CoV-2 , Serine endopeptidases , Pénétration virale , SARS-CoV-2/immunologie , SARS-CoV-2/physiologie , SARS-CoV-2/métabolisme , Humains , Serine endopeptidases/métabolisme , Serine endopeptidases/génétique , COVID-19/virologie , COVID-19/immunologie , COVID-19/métabolisme , Angiotensin-converting enzyme 2/métabolisme , Angiotensin-converting enzyme 2/génétique , Réplication virale , Animaux , Endocytose , Cellules HEK293 , Chlorocebus aethiops , Cytologie
11.
Exp Mol Med ; 56(5): 1221-1229, 2024 May.
Article de Anglais | MEDLINE | ID: mdl-38816566

RÉSUMÉ

Mouse models expressing human ACE2 for coronavirus disease 2019 have been frequently used to understand its pathogenesis and develop therapeutic strategies against SARS-CoV-2. Given that human TMPRSS2 supports viral entry, replication, and pathogenesis, we established a double-transgenic mouse model expressing both human ACE2 and TMPRSS2 for SARS-CoV-2 infection. Co-overexpression of both genes increased viral infectivity in vitro and in vivo. Double-transgenic mice showed significant body weight loss, clinical disease symptoms, acute lung injury, lung inflammation, and lethality in response to viral infection, indicating that they were highly susceptible to SARS-CoV-2. Pretreatment with the TMPRSS2 inhibitor, nafamostat, effectively reduced virus-induced weight loss, viral replication, and mortality in the double-transgenic mice. Moreover, the susceptibility and differential pathogenesis of SARS-CoV-2 variants were demonstrated in this animal model. Together, our results demonstrate that double-transgenic mice could provide a highly susceptible mouse model for viral infection to understand SARS-CoV-2 pathogenesis and evaluate antiviral therapeutics against coronavirus disease 2019.


Sujet(s)
Angiotensin-converting enzyme 2 , COVID-19 , Modèles animaux de maladie humaine , Souris transgéniques , SARS-CoV-2 , Serine endopeptidases , Animaux , Angiotensin-converting enzyme 2/génétique , Angiotensin-converting enzyme 2/métabolisme , Serine endopeptidases/génétique , Serine endopeptidases/métabolisme , COVID-19/virologie , COVID-19/génétique , COVID-19/métabolisme , SARS-CoV-2/physiologie , SARS-CoV-2/génétique , Humains , Souris , Réplication virale , Benzamidines , Guanidines/pharmacologie , Chlorocebus aethiops , Traitements médicamenteux de la COVID-19
12.
Ageing Res Rev ; 98: 102339, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38754634

RÉSUMÉ

Alzheimer's disease (AD) is the most common neurodegenerative disorder that affects the cerebral cortex and hippocampus, and is characterised by progressive cognitive decline and memory loss. A recent report of a patient carrying a novel gain-of-function variant of RELN (H3447R, termed RELN-COLBOS) who developed resilience against presenilin-linked autosomal-dominant AD (ADAD) has generated enormous interest. The RELN-COLBOS variant enhances interactions with the apolipoprotein E receptor 2 (ApoER2) and very-low-density lipoprotein receptor (VLDLR), which are associated with delayed AD onset and progression. These findings were validated in a transgenic mouse model. Reelin is involved in neurodevelopment, neurogenesis, and neuronal plasticity. The evidence accumulated thus far has demonstrated that the Reelin pathway links apolipoprotein E4 (ApoE4), amyloid-ß (Aß), and tubulin-associated unit (Tau), which are key proteins that have been implicated in AD pathogenesis. Reelin and key components of the Reelin pathway have been highlighted as potential therapeutic targets and biomarkers for AD.


Sujet(s)
Maladie d'Alzheimer , Peptides bêta-amyloïdes , Apolipoprotéine E4 , Molécules d'adhérence cellulaire neuronale , Protéines de la matrice extracellulaire , Protéines de tissu nerveux , Protéine reeline , Serine endopeptidases , Protéines tau , Maladie d'Alzheimer/métabolisme , Maladie d'Alzheimer/génétique , Protéines de la matrice extracellulaire/métabolisme , Protéines de la matrice extracellulaire/génétique , Molécules d'adhérence cellulaire neuronale/métabolisme , Molécules d'adhérence cellulaire neuronale/génétique , Humains , Serine endopeptidases/métabolisme , Serine endopeptidases/génétique , Protéines de tissu nerveux/métabolisme , Protéines de tissu nerveux/génétique , Animaux , Peptides bêta-amyloïdes/métabolisme , Protéines tau/métabolisme , Protéines tau/génétique , Apolipoprotéine E4/génétique , Apolipoprotéine E4/métabolisme , Souris
13.
Cancer Res Commun ; 4(6): 1481-1494, 2024 Jun 12.
Article de Anglais | MEDLINE | ID: mdl-38747612

RÉSUMÉ

Cancer-associated fibroblasts (CAF) are a prominent cell type within the tumor microenvironment (TME) where they are known to promote cancer cell growth and survival, angiogenesis, drug resistance, and immunosuppression. The transmembrane prolyl protease fibroblast activation protein (FAP) is expressed on the surface of highly protumorigenic CAFs found in the stroma of nearly every cancer of epithelial origin. The widespread expression of FAP has made it an attractive therapeutic target based on the underlying hypothesis that eliminating protumorigenic CAFs will disrupt the cross-talk between components of TME resulting in cancer cell death and immune infiltration. This hypothesis, however, has never been directly proven. To eliminate FAP-expressing CAFs, we developed an antibody-drug conjugate using our anti-FAP antibody, huB12, coupled to a monomethyl auristatin E (huB12-MMAE) payload. After determining that huB12 was an effective targeting vector, we found that huB12-MMAE potently eliminated FAP-expressing cells as monocultures in vitro and significantly prolonged survival in vivo using a xenograft engineered to overexpress FAP. We investigated the effects of selectively eliminating CAFs using a layered, open microfluidic cell coculture platform, known as the Stacks. Analysis of mRNA and protein expression found that treatment with huB12-MMAE resulted in the increased secretion of the proinflammatory cytokines IL6 and IL8 by CAFs and an associated increase in expression of proinflammatory genes in cancer cells. We also detected increased secretion of CSF1, a cytokine involved in myeloid recruitment and differentiation. Our findings suggest that the mechanism of FAP-targeted therapies is through effects on the immune microenvironment and antitumor immune response. SIGNIFICANCE: The direct elimination of FAP-expressing CAFs disrupts the cross-talk with cancer cells leading to a proinflammatory response and alterations in the immune microenvironment and antitumor immune response.


Sujet(s)
Fibroblastes associés au cancer , Endopeptidases , Immunoconjugués , Microenvironnement tumoral , Humains , Animaux , Immunoconjugués/pharmacologie , Fibroblastes associés au cancer/métabolisme , Fibroblastes associés au cancer/effets des médicaments et des substances chimiques , Fibroblastes associés au cancer/anatomopathologie , Fibroblastes associés au cancer/immunologie , Souris , Microenvironnement tumoral/effets des médicaments et des substances chimiques , Microenvironnement tumoral/immunologie , Endopeptidases/génétique , Endopeptidases/métabolisme , Lignée cellulaire tumorale , Serine endopeptidases/métabolisme , Serine endopeptidases/génétique , Protéines membranaires/génétique , Protéines membranaires/métabolisme , Tests d'activité antitumorale sur modèle de xénogreffe , Gelatinases/métabolisme , Gelatinases/génétique , Oligopeptides/pharmacologie , Femelle
14.
J Affect Disord ; 360: 326-335, 2024 Sep 01.
Article de Anglais | MEDLINE | ID: mdl-38788856

RÉSUMÉ

BACKGROUND: Major depressive disorder (MDD) is notably underdiagnosed and undertreated due to its complex nature and subjective diagnostic methods. Biomarker identification would help provide a clearer understanding of MDD aetiology. Although machine learning (ML) has been implemented in previous studies to study the alteration of microRNA (miRNA) levels in MDD cases, clinical translation has not been feasible due to the lack of interpretability (i.e. too many miRNAs for consideration) and stability. METHODS: This study applied logistic regression (LR) model to the blood miRNA expression profile to differentiate patients with MDD (n = 60) from healthy controls (HCs, n = 60). Embedded (L1-regularised logistic regression) feature selector was utilised to extract clinically relevant miRNAs, and optimized for clinical application. RESULTS: Patients with MDD could be differentiated from HCs with the area under the receiver operating characteristic curve (AUC) of 0.81 on testing data when all available miRNAs were considered (which served as a benchmark). Our LR model selected miRNAs up to 5 (known as LR-5 model) emerged as the best model because it achieved a moderate classification ability (AUC = 0.75), relatively high interpretability (feature number = 5) and stability (ϕ̂Z=0.55) compared to the benchmark. The top-ranking miRNAs identified by our model have demonstrated associations with MDD pathways involving cytokine signalling in the immune system, the reelin signalling pathway, programmed cell death and cellular responses to stress. CONCLUSION: The LR-5 model, which is optimised based on ML design factors, may lead to a robust and clinically usable MDD diagnostic tool.


Sujet(s)
Marqueurs biologiques , Trouble dépressif majeur , Apprentissage machine , microARN , Protéine reeline , Humains , Trouble dépressif majeur/génétique , Trouble dépressif majeur/diagnostic , Trouble dépressif majeur/sang , Trouble dépressif majeur/classification , microARN/sang , microARN/génétique , Mâle , Femelle , Adulte , Adulte d'âge moyen , Marqueurs biologiques/sang , Modèles logistiques , Serine endopeptidases/génétique , Serine endopeptidases/sang , Molécules d'adhérence cellulaire neuronale/génétique , Courbe ROC , Études cas-témoins , Protéines de la matrice extracellulaire/génétique , Protéines de la matrice extracellulaire/sang
15.
Hum Genet ; 143(5): 721-734, 2024 May.
Article de Anglais | MEDLINE | ID: mdl-38691166

RÉSUMÉ

TMPRSS3-related hearing loss presents challenges in correlating genotypic variants with clinical phenotypes due to the small sample sizes of previous studies. We conducted a cross-sectional genomics study coupled with retrospective clinical phenotype analysis on 127 individuals. These individuals were from 16 academic medical centers across 6 countries. Key findings revealed 47 unique TMPRSS3 variants with significant differences in hearing thresholds between those with missense variants versus those with loss-of-function genotypes. The hearing loss progression rate for the DFNB8 subtype was 0.3 dB/year. Post-cochlear implantation, an average word recognition score of 76% was observed. Of the 51 individuals with two missense variants, 10 had DFNB10 with profound hearing loss. These 10 all had at least one of 4 TMPRSS3 variants predicted by computational modeling to be damaging to TMPRSS3 structure and function. To our knowledge, this is the largest study of TMPRSS3 genotype-phenotype correlations. We find significant differences in hearing thresholds, hearing loss progression, and age of presentation, by TMPRSS3 genotype and protein domain affected. Most individuals with TMPRSS3 variants perform well on speech recognition tests after cochlear implant, however increased age at implant is associated with worse outcomes. These findings provide insight for genetic counseling and the on-going design of novel therapeutic approaches.


Sujet(s)
Études d'associations génétiques , Perte d'audition , Protéines membranaires , Serine endopeptidases , Humains , Femelle , Mâle , Serine endopeptidases/génétique , Adulte , Protéines membranaires/génétique , Perte d'audition/génétique , Enfant , Adulte d'âge moyen , Adolescent , Enfant d'âge préscolaire , Génotype , Études de cohortes , Phénotype , Mutation faux-sens , Études transversales , Jeune adulte , Études rétrospectives , Sujet âgé , Protéines tumorales
16.
Placenta ; 152: 53-64, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38805949

RÉSUMÉ

INTRODUCTION: The placenta differs greatly among species, and deep extra-villous trophoblast (EVT) invasion is a unique feature of placentation of higher primates including humans. We reported serine protease HtrA4 being found predominantly in human placentas with aberrant expression linked to preeclampsia. However, it remains unclear where HtrA4 is produced in the placenta, how it is expressed in other species, and whether it is essential for human placentation. METHODS: We first compared HtrA4 protein sequences of over 100 species, then scrutinized the key characteristics of HtrA4 in the human, rhesus macaque and mouse, and determined cellular localization in the placenta. We next investigated functional significance of HtrA4 in EVT differentiation using human trophoblast stem cells (TSCs). RESULTS: Across broader species HtrA4 is well conserved only in higher primates. In humans, only the placenta expressed HtrA4, localising to trophoblasts of villous as well as extra-villous lineages. Rhesus macaques produced HtrA4 but again only in placentas, whereas mice showed no abundant HtrA4 expression anywhere including the placenta, yet it was an active protease if produced. The functional importance of HtrA4 in human EVT was demonstrated using TSCs, which expressed low levels of HtrA4 but significantly up-regulated it during EVT differentiation, and knockdown of HtrA4 severely inhibited the differentiation process. DISCUSSION: HtrA4 is expressed in placentas of humans and macaques but not mice; it is critical for human EVT differentiation. Together with previous reports showing HtrA4 is also indispensable for syncytialization, this study further revealed HtrA4 as a functionally important protease for human placentation.


Sujet(s)
Différenciation cellulaire , Macaca mulatta , Serine endopeptidases , Trophoblastes , Animaux , Trophoblastes/métabolisme , Humains , Femelle , Grossesse , Différenciation cellulaire/physiologie , Souris , Serine endopeptidases/métabolisme , Serine endopeptidases/génétique , Placenta/métabolisme , Placentation/physiologie , Protéases à sérine
18.
Biomolecules ; 14(5)2024 May 17.
Article de Anglais | MEDLINE | ID: mdl-38786001

RÉSUMÉ

During the first and second stages of postnatal development, neocortical neurons exhibit a wide range of spontaneous synchronous activity (SSA). Towards the end of the second postnatal week, the SSA is replaced by a more sparse and desynchronized firing pattern. The developmental desynchronization of neocortical spontaneous neuronal activity is thought to be intrinsically generated, since sensory deprivation from the periphery does not affect the time course of this transition. The extracellular protein reelin controls various aspects of neuronal development through multimodular signaling. However, so far it is unclear whether reelin contributes to the developmental desynchronization transition of neocortical neurons. The present study aims to investigate the role of reelin in postnatal cortical developmental desynchronization using a conditional reelin knockout (RelncKO) mouse model. Conditional reelin deficiency was induced during early postnatal development, and Ca2+ recordings were conducted from organotypic cultures (OTCs) of the somatosensory cortex. Our results show that both wild type (wt) and RelncKO exhibited an SSA pattern during the early postnatal week. However, at the end of the second postnatal week, wt OTCs underwent a transition to a desynchronized network activity pattern, while RelncKO activity remained synchronous. This changing activity pattern suggests that reelin is involved in regulating the developmental desynchronization of cortical neuronal network activity. Moreover, the developmental desynchronization impairment observed in RelncKO was rescued when RelncKO OTCs were co-cultured with wt OTCs. Finally, we show that the developmental transition to a desynchronized state at the end of the second postnatal week is not dependent on glutamatergic signaling. Instead, the transition is dependent on GABAAR and GABABR signaling. The results suggest that reelin controls developmental desynchronization through GABAAR and GABABR signaling.


Sujet(s)
Protéines de la matrice extracellulaire , Souris knockout , Néocortex , Protéines de tissu nerveux , Protéine reeline , Serine endopeptidases , Animaux , Souris , Néocortex/métabolisme , Néocortex/croissance et développement , Protéines de tissu nerveux/métabolisme , Protéines de tissu nerveux/génétique , Serine endopeptidases/métabolisme , Serine endopeptidases/génétique , Protéines de la matrice extracellulaire/métabolisme , Protéines de la matrice extracellulaire/génétique , Molécules d'adhérence cellulaire neuronale/métabolisme , Molécules d'adhérence cellulaire neuronale/génétique , Neurones/métabolisme , Réseau nerveux/métabolisme , Réseau nerveux/croissance et développement , Cortex somatosensoriel/métabolisme , Cortex somatosensoriel/croissance et développement
19.
Nat Commun ; 15(1): 4056, 2024 May 14.
Article de Anglais | MEDLINE | ID: mdl-38744813

RÉSUMÉ

The fusion peptide of SARS-CoV-2 spike protein is functionally important for membrane fusion during virus entry and is part of a broadly neutralizing epitope. However, sequence determinants at the fusion peptide and its adjacent regions for pathogenicity and antigenicity remain elusive. In this study, we perform a series of deep mutational scanning (DMS) experiments on an S2 region spanning the fusion peptide of authentic SARS-CoV-2 in different cell lines and in the presence of broadly neutralizing antibodies. We identify mutations at residue 813 of the spike protein that reduced TMPRSS2-mediated entry with decreased virulence. In addition, we show that an F823Y mutation, present in bat betacoronavirus HKU9 spike protein, confers resistance to broadly neutralizing antibodies. Our findings provide mechanistic insights into SARS-CoV-2 pathogenicity and also highlight a potential challenge in developing broadly protective S2-based coronavirus vaccines.


Sujet(s)
Anticorps neutralisants , COVID-19 , Mutation , SARS-CoV-2 , Glycoprotéine de spicule des coronavirus , Pénétration virale , Glycoprotéine de spicule des coronavirus/immunologie , Glycoprotéine de spicule des coronavirus/génétique , Glycoprotéine de spicule des coronavirus/composition chimique , Glycoprotéine de spicule des coronavirus/métabolisme , Humains , SARS-CoV-2/immunologie , SARS-CoV-2/génétique , Anticorps neutralisants/immunologie , COVID-19/virologie , COVID-19/immunologie , Animaux , Anticorps antiviraux/immunologie , Serine endopeptidases/génétique , Serine endopeptidases/immunologie , Serine endopeptidases/métabolisme , Chlorocebus aethiops , Cellules HEK293 , Cellules Vero , Épitopes/immunologie , Épitopes/génétique , Lignée cellulaire , Souris
20.
Mol Cell Biol ; 44(4): 123-137, 2024.
Article de Anglais | MEDLINE | ID: mdl-38747374

RÉSUMÉ

SREBP transcription factors are central regulators of lipid metabolism. Their proteolytic activation requires ER to the Golgi translocation and subsequent cleavage by site-1-protease (S1P). Produced as a proprotein, S1P undergoes autocatalytic cleavage from its precursor S1PA to mature S1PC form. Here, we report that SPRING (previously C12ORF29) and S1P interact through their ectodomains, and that this facilitates the autocatalytic cleavage of S1PA into its mature S1PC form. Reciprocally, we identified a S1P recognition-motif in SPRING and demonstrate that S1P-mediated cleavage leads to secretion of the SPRING ectodomain in cells, and in liver-specific Spring knockout (LKO) mice transduced with AAV-mSpring. By reconstituting SPRING variants into SPRINGKO cells we show that the SPRING ectodomain supports proteolytic maturation of S1P and SREBP signaling, but that S1P-mediated SPRING cleavage is not essential for these processes. Absence of SPRING modestly diminishes proteolytic maturation of S1PA→C and trafficking of S1PC to the Golgi. However, despite reaching the Golgi in SPRINGKO cells, S1PC fails to rescue SREBP signaling. Remarkably, whereas SREBP signaling was severely attenuated in SPRINGKO cells and LKO mice, that of ATF6, another S1P substrate, was unaffected in these models. Collectively, our study positions SPRING as a dedicated licensing factor for SREBP-specific activation by S1P.


Sujet(s)
Appareil de Golgi , Souris knockout , Proprotein convertases , Animaux , Souris , Appareil de Golgi/métabolisme , Humains , Proprotein convertases/métabolisme , Proprotein convertases/génétique , Serine endopeptidases/métabolisme , Serine endopeptidases/génétique , Transduction du signal , Cellules HEK293 , Foie/métabolisme , Protéolyse , Réticulum endoplasmique/métabolisme , Protéines de liaison à l'élément de régulation des stérols/métabolisme , Protéines de liaison à l'élément de régulation des stérols/génétique
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...