Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 10.220
Filter
1.
Dent Med Probl ; 61(2): 225-231, 2024.
Article in English | MEDLINE | ID: mdl-38567731

ABSTRACT

BACKGROUND: Type 2 diabetes mellitus (DM) is a known systemic risk factor for periodontitis. An increased expression of CD44 has been suggested in type 2 diabetics and periodontitis patients. OBJECTIVES: The present study aimed to assess the expression of CD44 antigen in patients with chronic periodontitis (CP) and type 2 DM in a South Indian urban population. Additionally, the relationships between the expression of CD44 antigen in gingival tissues, periodontal clinical parameters, and the random blood sugar (RBS) and glycated hemoglobin (HbA1c) levels were assessed. MATERIAL AND METHODS: A total of 63 subjects were divided into 3 groups: systemically and periodontally healthy controls (group H); CP patients, otherwise healthy (group CP); and CP patients with type 2 DM (group CP+DM). Periodontal parameters were recorded for all groups, and additionally the RBS and HbA1c levels for group CP+DM. Gingival tissue samples were obtained and subjected to immunohistochemical analysis for CD44. RESULTS: The expression of CD44 was significantly higher in the diseased groups. Epithelial CD44 expression was significantly stronger in group CP+DM as compared to groups CP and H (p < 0.001), whereas connective tissue CD44 expression was similar in groups CP and CP+DM (p = 0.657). Furthermore, an inverse relationship was observed between blood glucose parameters and CD44 expression in the epithelium and connective tissue. CONCLUSIONS: The expression of CD44 increased with the severity of periodontal disease. Additionally, glycemic control in patients with CP and type 2 DM had an impact on CD44 expression. Our findings indicate a possible destructive role of CD44 in the pathogenesis of periodontal diseases in individuals with type 2 DM.


Subject(s)
Chronic Periodontitis , Diabetes Mellitus, Type 2 , Gingiva , Glycated Hemoglobin , Hyaluronan Receptors , Humans , Hyaluronan Receptors/metabolism , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/complications , Male , Female , Chronic Periodontitis/metabolism , Adult , Glycated Hemoglobin/metabolism , Middle Aged , Gingiva/metabolism , Immunohistochemistry , Blood Glucose/metabolism , Periodontal Index , Case-Control Studies , India
2.
Sci Rep ; 14(1): 9156, 2024 04 21.
Article in English | MEDLINE | ID: mdl-38644369

ABSTRACT

Intervertebral Disc (IVD) degeneration has been associated with a chronic inflammatory response, but knowledge on the contribution of distinct IVD cells, namely CD44, to the progression of IVD degeneration remains elusive. Here, bovine nucleus pulposus (NP) CD44 cells were sorted and compared by gene expression and proteomics with the negative counterpart. NP cells were then stimulated with IL-1b (10 ng/ml) and dynamics of CD44 gene and protein expression was analyzed upon pro-inflammatory treatment. The results emphasize that CD44 has a multidimensional functional role in IVD metabolism, ECM synthesis and production of neuropermissive factors. CD44 widespread expression in NP was partially associated with CD14 and CD45, resulting in the identification of distinct cell subsets. In conclusion, this study points out CD44 and CD44-based cell subsets as relevant targets in the modulation of the IVD pro-inflammatory/degenerative cascade.


Subject(s)
Hyaluronan Receptors , Inflammation , Intervertebral Disc Degeneration , Nucleus Pulposus , Animals , Cattle , Nucleus Pulposus/metabolism , Nucleus Pulposus/pathology , Hyaluronan Receptors/metabolism , Hyaluronan Receptors/genetics , Inflammation/metabolism , Inflammation/pathology , Intervertebral Disc Degeneration/metabolism , Intervertebral Disc Degeneration/pathology , Cells, Cultured , Interleukin-1beta/metabolism , Proteomics/methods
3.
J Nanobiotechnology ; 22(1): 198, 2024 Apr 22.
Article in English | MEDLINE | ID: mdl-38649957

ABSTRACT

Heat shock protein 90 (HSP90) is overexpressed in numerous cancers, promotes the maturation of numerous oncoproteins and facilitates cancer cell growth. Certain HSP90 inhibitors have entered clinical trials. Although less than satisfactory clinical effects or insurmountable toxicity have compelled these trials to be terminated or postponed, these results of preclinical and clinical studies demonstrated that the prospects of targeting therapeutic strategies involving HSP90 inhibitors deserve enough attention. Nanoparticulate-based drug delivery systems have been generally supposed as one of the most promising formulations especially for targeting strategies. However, so far, no active targeting nano-formulations have succeeded in clinical translation, mainly due to complicated preparation, complex formulations leading to difficult industrialization, incomplete biocompatibility or nontoxicity. In this study, HSP90 and CD44-targeted A6 peptide functionalized biomimetic nanoparticles (A6-NP) was designed and various degrees of A6-modification on nanoparticles were fabricated to evaluate targeting ability and anticancer efficiency. With no excipients, the hydrophobic HSP90 inhibitor G2111 and A6-conjugated human serum albumin could self-assemble into nanoparticles with a uniform particle size of approximately 200 nm, easy fabrication, well biocompatibility and avoidance of hepatotoxicity. Besides, G2111 encapsulated in A6-NP was only released less than 5% in 12 h, which may avoid off-target cell toxicity before entering into cancer cells. A6 peptide modification could significantly enhance uptake within a short time. Moreover, A6-NP continues to exert the broad anticancer spectrum of Hsp90 inhibitors and displays remarkable targeting ability and anticancer efficacy both in hematological malignancies and solid tumors (with colon tumors as the model cancer) both in vitro and in vivo. Overall, A6-NP, as a simple, biomimetic and active dual-targeting (CD44 and HSP90) nanomedicine, displays high potential for clinical translation.


Subject(s)
Antineoplastic Agents , Colonic Neoplasms , HSP90 Heat-Shock Proteins , Hyaluronan Receptors , Leukemia, Myeloid, Acute , HSP90 Heat-Shock Proteins/antagonists & inhibitors , HSP90 Heat-Shock Proteins/metabolism , Humans , Hyaluronan Receptors/metabolism , Animals , Cell Line, Tumor , Mice , Colonic Neoplasms/drug therapy , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/metabolism , Nanoparticles/chemistry , Drug Delivery Systems/methods , Mice, Nude , Mice, Inbred BALB C , Peptides/chemistry , Peptides/pharmacology
4.
Int J Mol Sci ; 25(7)2024 Mar 31.
Article in English | MEDLINE | ID: mdl-38612715

ABSTRACT

Breast cancer (BC) represents one of the most prevalent malignant threats to women globally. Tumor relapse or metastasis is facilitated by BC stemness progression, contributing to tumorigenicity. Therefore, comprehending the characteristics of stemness progression and the underlying molecular mechanisms is pivotal for BC advancement. Hinokitiol (ß-thujaplicin), a tropolone-related compound abundant in the heartwood of cupressaceous plants, exhibits antimicrobial activity. In our study, we employed three BC cell lines (MDA-MB-231, MCF-7, and T47D) to assess the expression of stemness-, apoptosis-, and autophagy-related proteins. Hinokitiol significantly reduced the viability of cancer cells in a dose-dependent manner. Furthermore, we observed that hinokitiol enhances apoptosis by increasing the levels of cleaved poly-ADP-ribose polymerase (PARP) and phospho-p53. It also induces dysfunction in autophagy through the upregulation of LC3B and p62 protein expression. Additionally, hinokitiol significantly suppressed the number and diameter of cancer cell line spheres by reducing the expression of cluster of differentiation44 (CD44) and key transcription factors. These findings underscore hinokitiol's potential as a therapeutic agent for breast cancer, particularly as a stemness-progression inhibitor. Further research and clinical studies are warranted to explore the full therapeutic potential of hinokitiol in the treatment of breast cancer.


Subject(s)
Breast Neoplasms , Monoterpenes , Tropolone , Tropolone/analogs & derivatives , Humans , Female , Tropolone/pharmacology , Breast Neoplasms/drug therapy , Neoplasm Recurrence, Local , Apoptosis , Autophagy , MCF-7 Cells , Hyaluronan Receptors , SOXB1 Transcription Factors
5.
Front Immunol ; 15: 1363834, 2024.
Article in English | MEDLINE | ID: mdl-38633247

ABSTRACT

Background: Hyaluronan-mediated motility receptor (HMMR) is overexpressed in multiple carcinomas and influences the development and treatment of several cancers. However, its role in hepatocellular carcinoma (HCC) remains unclear. Methods: The "limma" and "GSVA" packages in R were used to perform differential expression analysis and to assess the activity of signalling pathways, respectively. InferCNV was used to infer copy number variation (CNV) for each hepatocyte and "CellChat" was used to analyse intercellular communication networks. Recursive partitioning analysis (RPA) was used to re-stage HCC patients. The IC50 values of various drugs were evaluated using the "pRRophetic" package. In addition, quantitative reverse transcription polymerase chain reaction (qRT-PCR) was performed to confirm HMMR expression in an HCC tissue microarray. Flow cytometry (FCM) and cloning, Edu and wound healing assays were used to explore the capacity of HMMR to regulate HCC tumour. Results: Multiple cohort studies and qRT-PCR demonstrated that HMMR was overexpressed in HCC tissue compared with normal tissue. In addition, HMMR had excellent diagnostic performance. HMMR knockdown inhibited the proliferation and migration of HCC cells in vitro. Moreover, high HMMR expression was associated with "G2M checkpoint" and "E2F targets" in bulk RNA and scRNA-seq, and FCM confirmed that HMMR could regulate the cell cycle. In addition, HMMR was involved in the regulation of the tumour immune microenvironment via immune cell infiltration and intercellular interactions. Furthermore, HMMR was positively associated with genomic heterogeneity with patients with high HMMR expression potentially benefitting more from immunotherapy. Moreover, HMMR was associated with poor prognosis in patients with HCC and the re-staging by recursive partitioning analysis (RPA) gave a good prognosis prediction value and could guide chemotherapy and targeted therapy. Conclusion: The results of the present study show that HMMR could play a role in the diagnosis, prognosis, and treatments of patients with HCC based on bulk RNA-seq and scRAN-seq analyses and is a promising molecular marker for HCC.


Subject(s)
Carcinoma, Hepatocellular , Extracellular Matrix Proteins , Hyaluronan Receptors , Liver Neoplasms , Humans , DNA Copy Number Variations , Single-Cell Gene Expression Analysis , Tumor Microenvironment
6.
Anal Chim Acta ; 1303: 342520, 2024 May 15.
Article in English | MEDLINE | ID: mdl-38609255

ABSTRACT

BACKGROUND: Cluster of Differentiation 44 (CD44) is considered an important biomarker for various cancers, and achieving highly sensitive detection of CD44 is crucial, which plays a significant role in tumor invasion and metastasis, providing essential information for clinical tumor diagnosis. Commonly used methods for analysis include fluorescence spectroscopy (FL), photoelectrochemical analysis (PEC), electrochemical analysis (EC), and commercial ELISA kits. Although these methods offer high sensitivity, they can be relatively complex to perform experimentally. Electrochemiluminescence (ECL) has gained widespread research attention due to its high sensitivity, ease of operation, effective spatiotemporal control, and close to zero background signal. RESULTS: In this work, a sandwich-type ECL immunosensor for detecting CD44 was constructed using luminol as a luminophore. In this sensing platform, bimetallic MOFs (Pd@FeNi-MIL-88B) loaded with palladium nanoparticles (Pd NPs) were used as a novel enzyme mimic, exhibiting excellent catalytic performance towards the electroreduction of H2O2. The hybrids provided a strong support platform for luminol and antibodies, significantly enhancing the initial ECL signal of luminol. Subsequently, core-shell Au@MnO2 nanocomposites were synthesised by gold nanoparticles (Au NPs) encapsulated in manganese dioxide (MnO2) thin layers, as labels. In the luminol/H2O2 system, Au@MnO2 exhibited strong light absorption in the broad UV-vis spectrum, similar to the black body effect, and the scavenging effect of Mn2+ on O2•-, which achieved the dual-quenching of ECL signal. Under the optimal experimental conditions, the immunosensor demonstrated a detection range of 0.1 pg mL-1 - 100 ng mL-1, with a detection limit of 0.069 pg mL-1. SIGNIFICANCE: Based on Pd@FeNi-MIL-88B nanoenzymes and Au@MnO2 nanocomposites, a dual-quenching sandwich-type ECL immunosensor for the detection of CD44 was constructed. The proposed immunosensor exhibited excellent reproducibility, stability, selectivity, and sensitivity, and provided a valuable analytical strategy and technical platform for the accurate detection of disease biomarkers, and opened up potential application prospects for early clinical treatment.


Subject(s)
Biosensing Techniques , Metal Nanoparticles , Neoplasms , Humans , Manganese Compounds , Gold , Hydrogen Peroxide , Luminol , Reproducibility of Results , Immunoassay , Oxides , Palladium , Hyaluronan Receptors
7.
Methods Mol Biol ; 2777: 71-81, 2024.
Article in English | MEDLINE | ID: mdl-38478336

ABSTRACT

CD44 has been described in many malignancies as a marker of cancer stem cells (CSCs). Several techniques can be used to detect these cells. Here we detail CD44 detection by flow cytometry, a precise technique allowing to determine the percentage of positive cells and the mean fluorescent intensity reflecting the CD44 expression by cells in the samples. The protocol explained here can be used to detect CD44 from cell suspensions prepared from tissues or in vitro cell cultures.


Subject(s)
Hyaluronan Receptors , Neoplastic Stem Cells , Flow Cytometry , Hyaluronan Receptors/metabolism , Neoplastic Stem Cells/metabolism
8.
Prostate ; 84(8): 738-746, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38528654

ABSTRACT

BACKGROUND: The occurrence of castration-resistant prostate cancer (CRPC) varies in patients with advanced prostate cancer (PCa) undergoing androgen deprivation therapy (ADT). The rate of occurrence of CRPC may be related to the presence of prostate cancer stem cells (CSC). Thus, this study aims to evaluate the presence of CSC markers (CD44 and CD133) in histopathology tissue at the time of diagnosis and their correlation with the occurrence of CRPC in patients with advanced PCa within 2 years of ADT. METHOD: A retrospective case-control study was conducted to evaluate the incidence of CRPC within 2 years. The inclusion criteria were patients with PCa who had received treatment with ADT and a first-generation anti-androgen (AA) for 2 years. We classified patients based on whether they developed CRPC within 2 years (CRPC) of the therapy or did not experience CRPC within 2 years (non-CRPC) of the therapy. We performed immunohistochemical (IHC) staining for CD44 and CD133 on the prostate biopsy tissue samples. RESULTS: Data were collected from records spanning 2011-2019. We analyzed a total of 65 samples, including 22 patients with CRPC and 43 patients with non-CRPC who had received treatment with LHRH agonists and AA for up to 2 years. Our findings showed a significant H-score difference in CD44 protein expression between CRPC prostate adenocarcinoma samples 869 (200-1329) and non-CRPC 524 (154-1166) (p = 0.033). There was no significant difference in CD133 protein expression between the two groups (p = 0.554). However, there was a significant difference in the nonoccurrence of CRPC between the high expressions of both CD44 and CD133 groups with other expressions of CD44/CD133 groups (25% vs. 75%; p = 0.011; odds ratio = 4.29; 95% confidence interval [1.34, 13.76]). CONCLUSION: This study found a low expression of at least one CD44/CD133 protein in the patients without early occurrence of CRPC. This result might suggest that CD44/CD133 may function as a potential prognostic marker for PCa, especially in a low expression, to identify patients who have a better prognosis regarding the occurrence of early CRPC.


Subject(s)
AC133 Antigen , Androgen Antagonists , Biomarkers, Tumor , Hyaluronan Receptors , Prostatic Neoplasms, Castration-Resistant , Humans , Male , Hyaluronan Receptors/metabolism , Hyaluronan Receptors/analysis , Hyaluronan Receptors/biosynthesis , Prostatic Neoplasms, Castration-Resistant/pathology , Prostatic Neoplasms, Castration-Resistant/metabolism , AC133 Antigen/metabolism , Retrospective Studies , Aged , Prognosis , Case-Control Studies , Androgen Antagonists/therapeutic use , Biomarkers, Tumor/metabolism , Middle Aged , Aged, 80 and over , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology
9.
Biomolecules ; 14(3)2024 Mar 19.
Article in English | MEDLINE | ID: mdl-38540786

ABSTRACT

In pulpitis, dentinal restorative processes are considerably associated with undifferentiated mesenchymal cells in the pulp. This study aimed to investigate strategies to improve the odonto/osteogenic differentiation of dental pulp stem cells (DPSCs) in an inflammatory environment. After pretreatment of DPSCs with 20 ng/mL tumor necrosis factor-induced protein-6 (TSG-6), DPSCs were cultured in an inflammation-inducing solution. Real-time polymerase chain reaction and Western blotting were performed to measure the expression levels of nuclear factor kappa B (NF-κB) and odonto/osteogenic differentiation markers, respectively. Cell Counting Kit-8 and 5-ethynyl-2'-deoxyuridine assays were used to assess cell proliferation and activity. Subcutaneous ectopic osteogenesis and mandibular bone cultures were performed to assess the effects of TSG-6 in vivo. The expression levels of odonto/osteogenic markers were higher in TSG-6-pre-treated DPSCs than nontreated DPSCs, whereas NF-κB-related proteins were lower after the induction of inflammation. An anti-CD44 antibody counteracted the rescue effect of TSG-6 on DPSC activity and mineralization in an inflammatory environment. Exogenous administration of TSG-6 enhanced the anti-inflammatory properties of DPSCs and partially restored their mineralization function by inhibiting NF-κB signaling. The mechanism of action of TSG-6 was attributed to its interaction with CD44. These findings reveal novel mechanisms by which DPSCs counter inflammation and provide a basis for the treatment of pulpitis.


Subject(s)
NF-kappa B , Pulpitis , Humans , NF-kappa B/metabolism , Osteogenesis , Pulpitis/metabolism , Dental Pulp/metabolism , Signal Transduction , Cell Differentiation , Inflammation/metabolism , Stem Cells , Cells, Cultured , Cell Proliferation , Hyaluronan Receptors/metabolism
10.
Sensors (Basel) ; 24(6)2024 Mar 21.
Article in English | MEDLINE | ID: mdl-38544254

ABSTRACT

The accuracy and efficacy of medical treatment would be greatly improved by the continuous and real-time monitoring of protein biomarkers. Identification of cancer biomarkers in patients with solid malignant tumors is receiving increasing attention. Existing techniques for detecting cancer proteins, such as the enzyme-linked immunosorbent assay, require a lot of work, are not multiplexed, and only allow for single-time point observations. In order to get one step closer to clinical usage, a dynamic platform for biosensing the cancer biomarker CD44 using a single-mode optical fiber-based ball resonator biosensor was designed, constructed and evaluated in this work. The main novelty of the work is an in-depth study of the capability of an in-house fabricated optical fiber biosensor for in situ detection of a cancer biomarker (CD44 protein) by conducting several types of experiments. The main results of the work are as follows: (1) Calibration of the fabricated fiber-optic ball resonator sensors in both static and dynamic conditions showed similar sensitivity to the refractive index change demonstrating its usefulness as a biosensing platform for dynamic measurements; (2) The fabricated sensors were shown to be insensitive to pressure changes further confirming their utility as an in situ sensor; (3) The sensor's packaging and placement were optimized to create a better environment for the fabricated ball resonator's performance in blood-mimicking environment; (4) Incubating increasing protein concentrations with antibody-functionalized sensor resulted in nearly instantaneous signal change indicating a femtomolar detection limit in a dynamic range from 7.1 aM to 16.7 nM; (5) The consistency of the obtained signal change was confirmed by repeatability studies; (6) Specificity experiments conducted under dynamic conditions demonstrated that the biosensors are highly selective to the targeted protein; (7) Surface morphology studies by AFM measurements further confirm the biosensor's exceptional sensitivity by revealing a considerable shift in height but no change in surface roughness after detection. The biosensor's ability to analyze clinically relevant proteins in real time with high sensitivity offers an advancement in the detection and monitoring of malignant tumors, hence improving patient diagnosis and health status surveillance.


Subject(s)
Biosensing Techniques , Neoplasms , Humans , Biomarkers, Tumor , Biosensing Techniques/methods , Fiber Optic Technology/methods , Optical Fibers , Proteins , Neoplasms/diagnosis , Hyaluronan Receptors
11.
Am J Alzheimers Dis Other Demen ; 39: 15333175241238577, 2024.
Article in English | MEDLINE | ID: mdl-38491918

ABSTRACT

Dysregulation of the peripheral immune system is be involved in the neuroinflammation in Alzheimer disease (AD) and accelerate the disease progression. The contribution of immune cells, particularly B cells, to AD pathogenesis has gained attention in recent research. In this study, we investigated the role of Peripheral Blood Memory B cells (PBMBs) and their secreted Migration Inhibition Factor (MIF) in driving macrophage behavior in AD based on the scRNA-seq technique, immunofluorescence and flow cytometry. We discovered that MIF binds to the CD74-CD44 receptor complex on macrophages, influencing their behavior. The dysregulated macrophage response hampers the clearance of amyloid-beta (Aß) plaques, exacerbating AD pathology. Targeting the MIF-CD74-CD44 signal pathway may hold therapeutic potential in modulating macrophage activity and mitigating neuroinflammation in AD. This study provides a further understanding of peripheral immune cells dysregulated in AD.


Subject(s)
Alzheimer Disease , Macrophage Migration-Inhibitory Factors , Humans , Memory B Cells , Neuroinflammatory Diseases , Macrophage Migration-Inhibitory Factors/metabolism , Hyaluronan Receptors/metabolism
12.
Int J Mol Sci ; 25(6)2024 Mar 08.
Article in English | MEDLINE | ID: mdl-38542115

ABSTRACT

Cluster of differentiation 44 (CD44), a cell surface adhesion molecule overexpressed in cancer stem cells, has been implicated in chemoresistance. This scoping review, following PRISMA-ScR guidelines, systematically identified and evaluated clinical studies on the impact of CD44 expression on chemotherapy treatment outcomes across various cancer types. The search encompassed PubMed (1985-2023) and SCOPUS (1936-2023) databases, yielding a total of 12,659 articles, of which 40 met the inclusion criteria and were included in the qualitative synthesis using a predefined data extraction table. Data collected included the cancer type, sample size, interventions, control, treatment outcome, study type, expression of CD44 variants and isoforms, and effect of CD44 on chemotherapy outcome. Most of the studies demonstrated an association between increased CD44 expression and negative chemotherapeutic outcomes such as shorter overall survival, increased tumor recurrence, and resistance to chemotherapy, indicating a potential role of CD44 upregulation in chemoresistance in cancer patients. However, a subset of studies also reported non-significant relationships or conflicting results. In summary, this scoping review highlighted the breadth of the available literature investigating the clinical association between CD44 and chemotherapeutic outcomes. Further research is required to elucidate this relationship to aid clinicians in managing CD44-positive cancer patients.


Subject(s)
Drug Resistance, Neoplasm , Hyaluronan Receptors , Humans , Hyaluronan Receptors/genetics , Hyaluronan Receptors/metabolism , Treatment Outcome
13.
Int J Biol Sci ; 20(4): 1314-1331, 2024.
Article in English | MEDLINE | ID: mdl-38385088

ABSTRACT

Peritoneal metastasis (PM) continues to limit the clinical efficacy of gastric cancer (GC). Early growth response 1 (EGR1) plays an important role in tumor cell proliferation, angiogenesis and invasion. However, the role of EGR1 derived from the tumor microenvironment in reshaping the phenotypes of GC cells and its specific molecular mechanisms in increasing the potential for PM are still unclear. In this study, we reported that EGR1 was significantly up-regulated in mesothelial cells from GC peritoneal metastases, leading to enhanced epithelial-mesenchymal transformation (EMT) and stemness phenotypes of GC cells under co-culture conditions. These phenotypes were achieved through the transcription and secretion of TGF-ß1 by EGR1 in mesothelial cells, which could regulate the expression and internalization of CD44s. After being internalized into the cytoplasm, CD44s interacted with STAT3 to promote STAT3 phosphorylation and activation, and induced EMT and stemness gene transcription, thus positively regulating the metastasis of GC cells. Moreover, TGF-ß1 secretion in the PM microenvironment was significantly increased compared with the matched primary tumor. The blocking effect of SHR-1701 on TGF-ß1 was verified by inhibiting peritoneal metastases in xenografts. Collectively, the interplay of EGR1/TGF-ß1/CD44s/STAT3 signaling between mesothelial cells and GC cells induces EMT and stemness phenotypes, offering potential as a therapeutic target for PM of GC.


Subject(s)
Early Growth Response Protein 1 , Peritoneal Neoplasms , Stomach Neoplasms , Humans , Cell Line, Tumor , Cell Movement , Early Growth Response Protein 1/genetics , Early Growth Response Protein 1/metabolism , Epithelial-Mesenchymal Transition/physiology , Hyaluronan Receptors/genetics , Hyaluronan Receptors/metabolism , Peritoneum/pathology , Signal Transduction/genetics , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/metabolism , Stomach Neoplasms/metabolism , Transforming Growth Factor beta1/metabolism , Tumor Microenvironment/genetics , Animals
14.
Nanoscale ; 16(10): 5280-5293, 2024 Mar 07.
Article in English | MEDLINE | ID: mdl-38369899

ABSTRACT

Macrophage-driven inflammation is the central player in a range of pathological conditions, comprising autoimmune disorders, various cancers, as well as chronic inflammatory states like rheumatoid arthritis. Therapeutic strategies tailored to specifically target macrophage behavior have acquired substantial interest for their potential to alleviate chronic inflammation effectively. In this study, we introduce a pioneering therapeutic approach utilizing specialized CD44-targeted immunoliposomes carrying bortezomib to address inflammation at the cellular level and the significance of this strategy lies in its precision nature. Bortezomib's inhibition of the proteasome interferes with the finely-tuned mechanism that controls NFκB activation, ultimately leading to a downregulation of the inflammatory response. After performing computational docking demonstrating its strong binding affinity to the proteasome molecule, the resulting nano-construct displayed a hydrodynamic size of 144.26 ± 74.4 nm and a quasi-spherical morphology. Moreover, the nano-construct ensured a minimum shelf-life of 30 days, aiming for targeted delivery with practical longevity. Upon internalization of immunoliposomes, the interaction with CD44 receptors exhibited downstream signaling events. This included the activation of Jun amino-terminal kinases 1/2 (JNK1/2) and the extracellular-signal-regulated kinases (ERK) pathway. JNK1/2 activation may lead to the release of mitochondrial pro-apoptotic factors, triggering the intrinsic apoptotic pathway and activation of caspases, which was confirmed from the level of apoptotic gene and protein expression. The precise targeting and anti-inflammatory action of this therapy against macrophages hold promise for therapeutic interventions in a wide range of inflammatory conditions, offering a novel avenue for precision medicine in the battle against excessive inflammation.


Subject(s)
Inflammation , Proteasome Endopeptidase Complex , Humans , Bortezomib/pharmacology , Proteasome Endopeptidase Complex/metabolism , Proteasome Endopeptidase Complex/pharmacology , Inflammation/drug therapy , Inflammation/metabolism , Liposomes/metabolism , Macrophages/metabolism , Hyaluronan Receptors/metabolism
15.
Molecules ; 29(3)2024 Feb 05.
Article in English | MEDLINE | ID: mdl-38338483

ABSTRACT

Hyaluronan (HA) is a non-sulfated glycosaminoglycan that is present in a variety of body tissues and organs. Hyaluronan has a wide range of biological activities that are frequently influenced by molar mass; however, they also depend greatly on the source, purity, and kind of impurities in hyaluronan. High-molar-mass HA has anti-inflammatory, immunosuppressive, and antiangiogenic properties, while low-molar-mass HA has opposite properties. A number of chemical modifications have been performed to enhance the stability of HA and its applications in medical practice. Hyaluronan is widely applied in medicine, such as viscosupplementation, ophthalmology, otolaryngology, wound healing, cosmetics, and drug delivery. In this review, we summarized several medical applications of polymers based on the hyaluronan backbone.


Subject(s)
Cosmetics , Hyaluronic Acid , Hyaluronic Acid/chemistry , Wound Healing , Pharmaceutical Preparations , Drug Delivery Systems , Hyaluronan Receptors
16.
Phytother Res ; 38(4): 1830-1837, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38353369

ABSTRACT

CD44+ cancer stem cells (CSCs) are believed to account for drug resistance and tumour recurrence due to their potential to self-renew and differentiate into heterogeneous lineages. Therefore, efficient treatment strategies targeting and eliminating these CSCs are required. The flavonolignan, Silibinin, has gained immense attention in targeting CD44+ CSCs as it alters functional properties like cell cycle arrest, apoptosis, inhibition of invasion and metastasis and also inhibits a range of molecular pathways. However, its limited bioavailability is a major hurdle in asserting Silibinin as a translational therapeutic agent. Combinatorial therapy of Silibinin with conventional chemotherapeutic drugs is an alternative approach in targeting CD44+ CSCs as it increases the efficacy and reduces the cytotoxicity of chemotherapeutic drugs, thus preventing drug resistance. Certain Silibinin-conjugated nano-formulations have also been successfully developed, through which there is improved absorptivity/bioavailability of Silibinin and a decrease in the concentration of therapeutic drugs leading to reduced cytotoxicity. In this review, we summarise the effectiveness of the synergistic therapeutic approach for Silibinin in targeting the molecular mechanisms of CD44+ CSCs and emphasise the potential role of Silibinin as a novel therapeutic agent.


Subject(s)
Neoplasms , Humans , Hyaluronan Receptors/metabolism , Hyaluronan Receptors/therapeutic use , Neoplasms/drug therapy , Neoplastic Stem Cells , Silybin/pharmacology
17.
J Control Release ; 367: 540-556, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38301927

ABSTRACT

Cancer presents a high mortality rate due to ineffective treatments and tumour relapse with progression. Cancer vaccines hold tremendous potential due to their capability to eradicate tumour and prevent relapse. In this study, we present a novel glycovaccine for precise targeting and immunotherapy of aggressive solid tumours that overexpress CD44 standard isoform (CD44s) carrying immature Tn and sialyl-Tn (sTn) O-glycans. We describe an enzymatic method and an enrichment strategy to generate libraries of well-characterized cancer-specific CD44s-Tn and/or sTn glycoproteoforms, which mimic the heterogeneity found in tumours. We conjugated CD44-Tn-derived glycopeptides with carrier proteins making them more immunogenic, with further demonstration of the importance of this conjugation to overcome the glycopeptides' intrinsic toxicity. We have optimized the glycopeptide-protein maleimide-thiol conjugation chemistry to avoid undesirable cross-linking between carrier proteins and CD44s glycopeptides. The resulting glycovaccines candidates were well-tolerated in vivo, inducing both humoral and cellular immunity, including immunological memory. The generated antibodies exhibited specific reactivity against synthetic CD44s-Tn glycopeptides, CD44s-Tn glycoengineered cells, and human tumours. In summary, we present a promising prototype of a cancer glycovaccine for future therapeutical pre-clinical efficacy validation.


Subject(s)
Cancer Vaccines , Neoplasms , Humans , Vaccines, Combined , Antigens, Tumor-Associated, Carbohydrate/chemistry , Glycoconjugates , Neoplasms/therapy , Immunotherapy , Glycopeptides/chemistry , Carrier Proteins , Recurrence , Hyaluronan Receptors
18.
Environ Int ; 185: 108494, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38364571

ABSTRACT

Hexavalent chromium [Cr(VI)] is a common environmental pollutant and chronic exposure to Cr(VI) causes lung cancer in humans, however, the mechanism of Cr(VI) carcinogenesis has not been well understood. Lung cancer is the leading cause of cancer-related death, although the mechanisms of how lung cancer develops and progresses have been poorly understood. While long non-coding RNAs (lncRNAs) are found abnormally expressed in cancer, how dysregulated lncRNAs contribute to carcinogenesis remains largely unknown. The goal of this study is to investigate the mechanism of Cr(VI)-induced lung carcinogenesis focusing on the role of the lncRNA ABHD11 antisense RNA 1 (tail to tail) (ABHD11-AS1). It was found that the lncRNA ABHD11-AS1 expression levels are up-regulated in chronic Cr(VI) exposure-transformed human bronchial epithelial cells, chronically Cr(VI)-exposed mouse lung tissues, and human lung cancer cells as well. Bioinformatics analysis revealed that ABHD11-AS1 levels are up-regulated in lung adenocarcinomas (LUADs) tissues and associated with worse overall survival of LUAD patients but not in lung squamous cell carcinomas. It was further determined that up-regulation of ABHD11-AS1 expression plays an important role in chronic Cr(VI) exposure-induced cell malignant transformation and tumorigenesis, and the stemness of human lung cancer cells. Mechanistically, it was found that ABHD11-AS1 directly binds SART3 (spliceosome associated factor 3, U4/U6 recycling protein). The interaction of ABHD11-AS1 with SART3 promotes USP15 (ubiquitin specific peptidase 15) nuclear localization. Nuclear localized USP15 interacts with pre-mRNA processing factor 19 (PRPF19) to increase CD44 RNA alternative splicing activating ß-catenin and enhancing cancer stemness. Together, these findings indicate that lncRNA ABHD11-AS1 interacts with SART3 and regulates CD44 RNA alternative splicing to promote cell malignant transformation and lung carcinogenesis.


Subject(s)
Chromium , DNA Repair Enzymes , Hyaluronan Receptors , Lung Neoplasms , Nuclear Proteins , RNA, Long Noncoding , Serine Proteases , Ubiquitin-Specific Proteases , Humans , Animals , Mice , RNA, Antisense/metabolism , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Alternative Splicing , Carcinogenesis/genetics , Cell Transformation, Neoplastic , Lung , Lung Neoplasms/genetics , Cell Proliferation/genetics , Cell Line, Tumor , Antigens, Neoplasm/metabolism , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , RNA Splicing Factors/genetics , RNA Splicing Factors/metabolism
19.
Biomacromolecules ; 25(3): 1959-1971, 2024 Mar 11.
Article in English | MEDLINE | ID: mdl-38379131

ABSTRACT

Triple-negative breast cancer (TNBC) presents treatment challenges due to a lack of detectable surface receptors. Natural killer (NK) cell-based adaptive immunotherapy is a promising treatment because of the characteristic anticancer effects of killing malignant cells directly by secreting cytokines and lytic granules. To maximize the cancer recognition ability of NK cells, biomaterial-mediated ex vivo cell surface engineering has been developed for sufficient cell membrane immobilization of tumor-targeting ligands via hydrophobic anchoring. In this study, we optimized amphiphilic balances of NK cell coating materials composed of CD44-targeting hyaluronic acid (HA)-poly(ethylene glycol) (PEG)-lipid to improve TNBC recognition and the anticancer effect. Changes in the modular design of our material by differentiating hydrophilic PEG length and incorporating lipid amount into HA backbones precisely regulated the amphiphilic nature of HA-PEG-lipid conjugates. The optimized biomaterial demonstrated improved anchoring into NK cell membranes and facilitating the surface presentation level of HA onto NK cell surfaces. This led to enhanced cancer targeting via increasing the formation of immune synapse, thereby augmenting the anticancer capability of NK cells specifically toward CD44-positive TNBC cells. Our approach addresses targeting ability of NK cell to solid tumors with a deficiency of surface tumor-specific antigens while offering a valuable material design strategy using amphiphilic balance in immune cell surface engineering techniques.


Subject(s)
Hyaluronic Acid , Triple Negative Breast Neoplasms , Humans , Hyaluronic Acid/chemistry , Cell Line, Tumor , Biocompatible Materials/pharmacology , Killer Cells, Natural , Lipids , Hyaluronan Receptors/metabolism
20.
Cell Signal ; 118: 111120, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38417636

ABSTRACT

Vestigial-like family member 1 (VGLL1) is one of the X-linked genes whose expression is elevated in basal-like breast cancer (BLBC) because of X-chromosome isodisomy. As an approach towards understanding its function, we performed correlation study using transcript data of breast cancer patients from cBioPortal for Cancer Genomics. Our analysis identified EGFR as the most correlated transcript with VGLL1. We demonstrate that VGLL1 promotes EGFR expression and increases the frequency of breast tumor initiating cells (CD44high/+CD24low/-). These findings are crucial because an elevated EGFR expression and high frequency of CD44high/+CD24low/- cells are defining features of BLBC, and we provide a new mechanistic insight into how their expressions are controlled. Importantly, VGLL1 regulation of EGFR and CD44high/+CD24low/- population is mediated by the hippo-transducer TAZ which exerts its oncogenic roles by binding and activating TEAD transcription factors. A crucial finding is that TEAD-binding domain of TAZ is dispensable for its regulation of EGFR and CD44high/+CD24low/- cells. Instead, VGLL1 stabilization of cytoplasmic TAZ is essential for these functions. Also, we show that VGLL1/TAZ restricts the surface expression of CD24 which contributes to the increased number of CD44high/+CD24low/- cells. In addition, we observed that VGLL1 represses AXL expression and suppresses claudin-low phenotype, and that is caused by the VGLL1 mediated nuclear expulsion of TAZ. Therefore, EGFR and AXL seem to represent two different breast tumor subtypes, and their differential expressions is controlled by VGLL1.


Subject(s)
Breast Neoplasms , Female , Humans , Breast Neoplasms/pathology , CD24 Antigen/metabolism , Cytoplasm/metabolism , DNA-Binding Proteins/metabolism , ErbB Receptors/genetics , ErbB Receptors/metabolism , Hyaluronan Receptors/metabolism , Neoplastic Stem Cells/metabolism , Transcription Factors/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...