Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 10.325
Filter
1.
Arq Bras Oftalmol ; 88(1): e20230163, 2024.
Article in English | MEDLINE | ID: mdl-39109744

ABSTRACT

PURPOSE: The epithelial-mesenchymal transition of human lens epithelial cells plays a role in posterior capsule opacification, a fibrotic process that leads to a common type of cataract. Hyaluronic acid has been implicated in this fibrosis. Studies have investigated the role of transforming growth factor (TGF)-ß2 in epithelial-mesenchymal transition. However, the role of TGF-ß2 in hyaluronic acid-mediated fibrosis of lens epithelial cell remains unknown. We here examined the role of TGF-ß2 in the hyaluronic acid-mediated epithelial-mesenchymal transition of lens epithelial cells. METHODS: Cultured human lens epithelial cells (HLEB3) were infected with CD44-siRNA by using the Lipofectamine 3000 transfection reagent. The CCK-8 kit was used to measure cell viability, and the scratch assay was used to determine cell migration. Cell oxidative stress was analyzed in a dichloro-dihydro-fluorescein diacetate assay and by using a flow cytometer. The TGF-ß2 level in HLEB3 cells was examined through immunohistochemical staining. The TGF-ß2 protein level was determined through western blotting. mRNA expression levels were determined through quantitative real-time polymerase chain reaction. RESULTS: Treatment with hyaluronic acid (1.0 µM, 24 h) increased the epithelial-mesenchymal transition of HLEB3 cells. The increase in TGF-ß2 levels corresponded to an increase in CD44 levels in the culture medium. However, blocking the CD44 function significantly reduced the TGF-ß2-mediated epithelial-mesenchymal transition response of HLEB3 cells. CONCLUSIONS: Our study showed that both CD44 and TGF-ß2 are critical contributors to the hyaluronic acid-mediated epithelial-mesenchymal transition of lens epithelial cells, and that TGF-ß2 in epithelial-mesenchymal transition is regulated by CD44. These results suggest that CD44 could be used as a target for preventing hyaluronic acid-induced posterior capsule opacification. Our findings suggest that CD44/TGF-ß2 is crucial for the hyaluronic acid-induced epithelial-mesenchymal transition of lens epithelial cells.


Subject(s)
Cell Movement , Epithelial Cells , Epithelial-Mesenchymal Transition , Hyaluronan Receptors , Hyaluronic Acid , Lens, Crystalline , Transforming Growth Factor beta2 , Humans , Epithelial-Mesenchymal Transition/drug effects , Epithelial-Mesenchymal Transition/physiology , Hyaluronic Acid/pharmacology , Hyaluronan Receptors/metabolism , Transforming Growth Factor beta2/pharmacology , Transforming Growth Factor beta2/metabolism , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Lens, Crystalline/cytology , Lens, Crystalline/drug effects , Lens, Crystalline/metabolism , Cell Movement/drug effects , Cell Survival/drug effects , Oxidative Stress/drug effects , Oxidative Stress/physiology , Blotting, Western , Capsule Opacification/metabolism , Capsule Opacification/pathology , Real-Time Polymerase Chain Reaction , Flow Cytometry , Immunohistochemistry , Cells, Cultured
2.
J Neuroinflammation ; 21(1): 190, 2024 Aug 02.
Article in English | MEDLINE | ID: mdl-39095775

ABSTRACT

Retinitis pigmentosa (RP), an inherited retinal disease, affects 1,5 million people worldwide. The initial mutation-driven photoreceptor degeneration leads to chronic inflammation, characterized by Müller cell activation and upregulation of CD44. CD44 is a cell surface transmembrane glycoprotein and the primary receptor for hyaluronic acid. It is involved in many pathological processes, but little is known about CD44's retinal functions. CD44 expression is also increased in Müller cells from our Pde6bSTOP/STOP RP mouse model. To gain a more detailed understanding of CD44's role in healthy and diseased retinas, we analyzed Cd44-/- and Cd44-/-Pde6bSTOP/STOP mice, respectively. The loss of CD44 led to enhanced photoreceptor degeneration, reduced retinal function, and increased inflammatory response. To understand the underlying mechanism, we performed proteomic analysis on isolated Müller cells from Cd44-/- and Cd44-/-Pde6bSTOP/STOP retinas and identified a significant downregulation of glutamate transporter 1 (SLC1A2). This downregulation was accompanied by higher glutamate levels, suggesting impaired glutamate homeostasis. These novel findings indicate that CD44 stimulates glutamate uptake via SLC1A2 in Müller cells, which in turn, supports photoreceptor survival and function.


Subject(s)
Ependymoglial Cells , Hyaluronan Receptors , Retinitis Pigmentosa , Signal Transduction , Animals , Hyaluronan Receptors/metabolism , Hyaluronan Receptors/genetics , Mice , Ependymoglial Cells/metabolism , Signal Transduction/physiology , Retinitis Pigmentosa/metabolism , Retinitis Pigmentosa/pathology , Retinitis Pigmentosa/genetics , Mice, Knockout , Mice, Inbred C57BL , Photoreceptor Cells, Vertebrate/metabolism , Cell Survival/physiology , Mice, Transgenic , Retina/metabolism , Retina/pathology
3.
Adv Gerontol ; 37(3): 259-265, 2024.
Article in Russian | MEDLINE | ID: mdl-39139118

ABSTRACT

The review describes the involvement of various hyaluronic acid receptors, including CD44, RHAMM, HARE, TLR, LYVE-1, in maintaining normal homeostasis and aging, as well as in the development of age-associated inflammatory processes (inflamaging) and malignant tumors. The association of CD44 receptor activation with immune cells and the development of coronary heart disease has been shown. In addition, a link between the CD44 receptor and osteoarthritis has been shown, via TLR2 and TLR4. The oncogenic potential of RHAMM in relation to breast, prostate, leukemia, pancreas, lung and glioblastoma cancers has been described, with the strongest expression observed in metastatic tumors. In vivo and in vitro experiments, it was found that fragments of hyaluronic acid with a length of 4 to 25 disaccharides can contribute to the proliferation of lymphatic endothelial cells and lymphangiogenesis. Thus, hyaluronic acid receptors play an important role in the aging process through the regulation of inflamaging and in the development of malignant neoplasms.


Subject(s)
Aging , Hyaluronan Receptors , Neoplasms , Humans , Aging/metabolism , Aging/physiology , Hyaluronan Receptors/metabolism , Neoplasms/metabolism , Neoplasms/pathology , Hyaluronic Acid/metabolism
4.
Sci Rep ; 14(1): 19560, 2024 08 22.
Article in English | MEDLINE | ID: mdl-39174579

ABSTRACT

Three-dimensional (3D) bioprinting culture models capable of reproducing the pathological architecture of diseases are increasingly advancing. In this study, 3D scaffolds were created using extrusion-based bioprinting method with alginate, gelatin, and hyaluronic acid to investigate the effects of hyaluronic acid on the physical properties of the bioscaffold as well as on the formation of liver cancer spheroids. Conformational analysis, rheological characterization, and swelling-degradation tests were performed to characterize the scaffolds. After generating spheroids from hepatocellular carcinoma cells on the 3D scaffolds, cell viability and proliferation assays were performed. Flow cytometry and immunofluorescence microscopy were used into examine the expression of albumin, CD44, and E-cadherin to demonstrate functional capability and maturation levels of the spheroid-forming cells. The results show that hyaluronic acid in the scaffolds correlates with spheroid formation and provides high survival rates. It is also associated with an increase in CD44 expression and a decrease in E-cadherin, while there is no significant change in the albumin expression in the cells. Overall, the findings demonstrate that hyaluronic acid in a 3D hydrogel scaffold supports spheroid formation and may induce stemness. We present a promising 3D scaffold model for enhancing liver cancer spheroid formation and mimicking solid tumors. This model also has the potential for further studies to examine stem cell properties in 3D models.


Subject(s)
Hyaluronan Receptors , Hyaluronic Acid , Neoplastic Stem Cells , Spheroids, Cellular , Tissue Scaffolds , Hyaluronic Acid/pharmacology , Hyaluronic Acid/chemistry , Humans , Spheroids, Cellular/drug effects , Spheroids, Cellular/metabolism , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/pathology , Tissue Scaffolds/chemistry , Hyaluronan Receptors/metabolism , Liver Neoplasms/pathology , Liver Neoplasms/metabolism , Cell Survival/drug effects , Cadherins/metabolism , Cell Proliferation/drug effects , Bioprinting/methods , Carcinoma, Hepatocellular/pathology , Carcinoma, Hepatocellular/metabolism , Cell Line, Tumor , Gelatin/chemistry , Hydrogels/chemistry , Hydrogels/pharmacology
5.
Oncoimmunology ; 13(1): 2393442, 2024.
Article in English | MEDLINE | ID: mdl-39175947

ABSTRACT

The inflammatory tumor microenvironment (TME) is a key driver for tumor-promoting processes. Tumor-associated macrophages are one of the main immune cell types in the TME and their increased density is related to poor prognosis in prostate cancer. Here, we investigated the influence of pro-inflammatory (M1) and immunosuppressive (M2) macrophages on prostate cancer lineage plasticity. Our findings reveal that M1 macrophage secreted factors upregulate genes related to stemness while downregulating genes associated with androgen response in prostate cancer cells. The expression of cancer stem cell (CSC) plasticity markers NANOG, KLF4, SOX2, OCT4, and CD44 was stimulated by the secreted factors from M1 macrophages. Moreover, AR and its target gene PSA were observed to be suppressed in LNCaP cells treated with secreted factors from M1 macrophages. Inhibition of NFκB signaling using the IKK16 inhibitor resulted in downregulation of NANOG, SOX2, and CD44 and CSC plasticity. Our study highlights that the secreted factors from M1 macrophages drive prostate cancer cell plasticity by upregulating the expression of CSC plasticity markers through NFκB signaling pathway.


Subject(s)
Hyaluronan Receptors , Kruppel-Like Factor 4 , Macrophages , NF-kappa B , Nanog Homeobox Protein , Neoplastic Stem Cells , Prostatic Neoplasms , SOXB1 Transcription Factors , Signal Transduction , Male , Humans , Prostatic Neoplasms/pathology , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/genetics , Nanog Homeobox Protein/metabolism , Nanog Homeobox Protein/genetics , SOXB1 Transcription Factors/metabolism , SOXB1 Transcription Factors/genetics , Hyaluronan Receptors/metabolism , Hyaluronan Receptors/genetics , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Kruppel-Like Factor 4/metabolism , NF-kappa B/metabolism , Cell Line, Tumor , Macrophages/metabolism , Up-Regulation , Tumor Microenvironment/immunology , Cell Plasticity/genetics , Gene Expression Regulation, Neoplastic , Tumor-Associated Macrophages/metabolism , Tumor-Associated Macrophages/immunology , Animals , Mice
6.
J Clin Invest ; 134(16)2024 Aug 15.
Article in English | MEDLINE | ID: mdl-39145451

ABSTRACT

CD44 is associated with a high risk of metastasis, recurrence, and drug resistance in various cancers. Here we report that platelet endothelial aggregation receptor 1 (PEAR1) is a CD44 chaperone protein that protected CD44 from endocytosis-mediated degradation and enhances cleavage of the CD44 intracellular domain (CD44-ICD). Furthermore, we found that lysyl oxidase-like protein 2 (LOXL2), an endogenous ligand of PEAR1, bound to the PEAR1-EMI domain and facilitated the interaction between PEAR1 and CD44 by inducing PEAR1 Ser891 phosphorylation in a manner that was independent of its enzyme activity. Levels of PEAR1 protein and PEAR1 phosphorylation at Ser891 were increased in patients with triple-negative breast cancer (TNBC), were positively correlated with expression of LOXL2 and CD44, and were negatively correlated with overall survival. The level of PEAR1 Ser891 phosphorylation was identified as the best independent prognostic factor in TNBC patients. The prognostic efficacy of the combination of PEAR1 phosphorylation at Ser891 and CD44 expression was superior to that of PEAR1 phosphorylation at Ser891 alone. Blocking the interaction between LOXL2 and PEAR1 with monoclonal antibodies significantly inhibited TNBC metastasis, representing a promising therapeutic strategy for TNBC.


Subject(s)
Amino Acid Oxidoreductases , Hyaluronan Receptors , Neoplasm Metastasis , Receptors, Cell Surface , Triple Negative Breast Neoplasms , Humans , Triple Negative Breast Neoplasms/pathology , Triple Negative Breast Neoplasms/metabolism , Triple Negative Breast Neoplasms/genetics , Hyaluronan Receptors/metabolism , Hyaluronan Receptors/genetics , Female , Phosphorylation , Receptors, Cell Surface/metabolism , Receptors, Cell Surface/genetics , Amino Acid Oxidoreductases/metabolism , Amino Acid Oxidoreductases/genetics , Animals , Cell Line, Tumor , Mice , Proteolysis , Neoplasm Proteins/metabolism , Neoplasm Proteins/genetics
7.
Neoplasia ; 56: 101034, 2024 10.
Article in English | MEDLINE | ID: mdl-39128424

ABSTRACT

BACKGROUND: Radiotherapy is the primary treatment for patients with nasopharyngeal carcinoma (NPC); however, almost 20% of patients experience treatment failure due to radioresistance. Therefore, understanding the mechanisms of radioresistance is imperative. HOTAIRM1 is deregulated in various human cancers, yet its role in NPC radioresistance are largely unclear. METHODS: This study investigated the association between HOTAIRM1 and radioresistance using CCK8, flow cytometry, and comet assays. Additionally, xenograft mice and patient-derived xenografts (PDX) models were employed to elucidate the biological functions of HOTAIRM1, and transcriptomic RNA sequencing was utilized to identify its target genes. RESULTS: Our study revealed an upregulation of HOTAIRM1 levels in radioresistant NPC cell lines and tissues. Furthermore, a positive correlation was noted between high HOTAIRM1 expression and increased NPC cell proliferation, reduced apoptosis, G2/M cell cycle arrest, and diminished cellular DNA damage following radiotherapy. HOTAIRM1 modulates the acetylation and stability of the FTO protein, and inhibiting FTO elevates the m6A methylation level of CD44 precursor transcripts in NPC cells. Additionally, silencing the m6A reading protein YTHDC1 was found to increase the expression of CD44V. HOTAIRM1 enhances NPC cell resistance to ferroptosis and irradiation through the HOTAIRM1-FTO-YTHDC1-CD44 axis. Mechanistically, HOTAIRM1 interacts with the FTO protein and induces m6A demethylation of the CD44 transcript. The absence of m6A modification in the CD44 transcript prevents its recognition by YTHDC1, resulting in the transition from CD44S to CD44V. An abundance of CD44V suppresses ferroptosis induced by irradiation and contributes to NPC radioresistance. CONCLUSIONS: In conclusion, the results in this study support the idea that HOTAIRM1 stimulates CD44 alternative splicing via FTO-mediated demethylation, thereby attenuating ferroptosis induced by irradiation and promoting NPC radioresistance.


Subject(s)
Alpha-Ketoglutarate-Dependent Dioxygenase FTO , Alternative Splicing , Gene Expression Regulation, Neoplastic , Hyaluronan Receptors , Nasopharyngeal Carcinoma , Nasopharyngeal Neoplasms , Radiation Tolerance , Humans , Nasopharyngeal Carcinoma/genetics , Nasopharyngeal Carcinoma/pathology , Nasopharyngeal Carcinoma/metabolism , Nasopharyngeal Carcinoma/radiotherapy , Alpha-Ketoglutarate-Dependent Dioxygenase FTO/genetics , Alpha-Ketoglutarate-Dependent Dioxygenase FTO/metabolism , Mice , Radiation Tolerance/genetics , Hyaluronan Receptors/genetics , Hyaluronan Receptors/metabolism , Animals , Cell Line, Tumor , Acetylation , Nasopharyngeal Neoplasms/genetics , Nasopharyngeal Neoplasms/radiotherapy , Nasopharyngeal Neoplasms/pathology , Nasopharyngeal Neoplasms/metabolism , Cell Proliferation , Apoptosis/genetics , Xenograft Model Antitumor Assays , MicroRNAs
8.
Med Gas Res ; 14(3): 121-126, 2024 Sep 01.
Article in English | MEDLINE | ID: mdl-39073340

ABSTRACT

Tumor hypoxia may compromise the results of chemotherapy for treating colorectal cancer because it stimulates angiogenesis and the release of tumor growth factors. Hyperbaric oxygen (HBO) supplementation may potentiate the effects of chemotherapy in such cases. This study aimed to assess the effect of HBO therapy combined with chemotherapy on the treatment of colorectal cancer in mice. C57BL6 mice were submitted to the intrarectal instillation of N-methyl-N-nitrosoguanidine (MNNG) and treated with 5-fluorouracil (5FU) and/or HBO therapy. The MNNG group presented the highest dysplastic crypt rate. The 5FU + HBO group presented the highest rate of apoptotic cells per dysplastic crypt. The 5FU group presented the highest expression of hypoxia-inducible factor-1 alpha and CD44. HBO therapy increased the effect of 5FU on the treatment of the experimental colorectal neoplasia in mice.


Subject(s)
Colorectal Neoplasms , Fluorouracil , Hyperbaric Oxygenation , Mice, Inbred C57BL , Animals , Fluorouracil/pharmacology , Mice , Colorectal Neoplasms/therapy , Colorectal Neoplasms/pathology , Colorectal Neoplasms/drug therapy , Male , Antimetabolites, Antineoplastic/pharmacology , Apoptosis/drug effects , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Hyaluronan Receptors/metabolism , Combined Modality Therapy , Methylnitronitrosoguanidine/pharmacology
9.
Int Immunopharmacol ; 138: 112613, 2024 Sep 10.
Article in English | MEDLINE | ID: mdl-38959542

ABSTRACT

Cancer stem cells (CSCs) significantly interfere with immunotherapy, leading to challenges such as low response rates and acquired resistance. PD-L1 expression is associated with the CSC population's overexpression of CD44. Mounting evidence suggests that the breast cancer stem cell (BCSC) marker CD44 and the immune checkpoint PD-L1 contribute to treatment failure through their networks. Natural compounds can overcome therapy resistance in breast cancer by targeting mechanisms underlying resistance in BCSCs. This review provides an updated insight into the CD44 and PD-L1 networks of BCSCs in mediating metastasis and immune evasion. The review critically examines existing literature, providing a comprehensive understanding of the topic and emphasizing the impact of natural flavones on the signaling pathways of BCSCs. Additionally, the review discusses the potential of natural compounds in targeting CD44 and PD-L1 in breast cancer (BC). Natural compounds consistently show potential in targeting regulatory mechanisms of BCSCs, inducing loss of stemness, and promoting differentiation. They offer a promising approach for developing alternative therapeutic strategies to manage breast cancer.


Subject(s)
B7-H1 Antigen , Breast Neoplasms , Drug Resistance, Neoplasm , Hyaluronan Receptors , Immune Evasion , Neoplastic Stem Cells , Humans , Hyaluronan Receptors/metabolism , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/immunology , Breast Neoplasms/drug therapy , Breast Neoplasms/immunology , Breast Neoplasms/metabolism , B7-H1 Antigen/metabolism , Drug Resistance, Neoplasm/drug effects , Female , Animals , Biological Products/pharmacology , Biological Products/therapeutic use , Signal Transduction/drug effects
10.
Nat Commun ; 15(1): 5689, 2024 Jul 07.
Article in English | MEDLINE | ID: mdl-38971796

ABSTRACT

Leukemia is a kind of hematological malignancy originating from bone marrow, which provides essential signals for initiation, progression, and recurrence of leukemia. However, how to specifically deliver drugs to the bone marrow remains elusive. Here, we develop biomimetic vesicles by infusing hematopoietic stem and progenitor cell (HSPC) membrane with liposomes (HSPC liposomes), which migrate to the bone marrow of leukemic mice via hyaluronic acid-CD44 axis. Moreover, the biomimetic vesicles exhibit superior binding affinity to leukemia cells through intercellular cell adhesion molecule-1 (ICAM-1)/integrin ß2 (ITGB2) interaction. Further experiments validate that the vesicles carrying chemotherapy drug cytarabine (Ara-C@HSPC-Lipo) markedly inhibit proliferation, induce apoptosis and differentiation of leukemia cells, and decrease number of leukemia stem cells. Mechanically, RNA-seq reveals that Ara-C@HSPC-Lipo treatment induces apoptosis and differentiation and inhibits the oncogenic pathways. Finally, we verify that HSPC liposomes are safe in mice. This study provides a method for targeting bone marrow and treating leukemia.


Subject(s)
Apoptosis , Bone Marrow , Cytarabine , Drug Delivery Systems , Hematopoietic Stem Cells , Leukemia , Liposomes , Animals , Hematopoietic Stem Cells/drug effects , Hematopoietic Stem Cells/metabolism , Mice , Cytarabine/pharmacology , Bone Marrow/drug effects , Bone Marrow/pathology , Bone Marrow/metabolism , Apoptosis/drug effects , Leukemia/drug therapy , Leukemia/pathology , Humans , Cell Differentiation/drug effects , Cell Membrane/metabolism , Cell Membrane/drug effects , Cell Line, Tumor , CD18 Antigens/metabolism , Cell Proliferation/drug effects , Hyaluronan Receptors/metabolism , Hyaluronic Acid/chemistry , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/pathology , Neoplastic Stem Cells/metabolism
11.
J Mol Neurosci ; 74(3): 64, 2024 Jul 09.
Article in English | MEDLINE | ID: mdl-38981928

ABSTRACT

Circular RNAs (circRNAs) are a subclass of non-coding RNAs which have demonstrated potential as biomarkers for Alzheimer's disease (AD). In this study, we conducted a comprehensive exploration of the circRNA transcriptome within AD brain tissues. Specifically, we assessed circRNA expression patterns in the dorsolateral prefrontal cortex collected from nine AD-afflicted individuals and eight healthy controls. Utilising two circRNA detection tools, CIRI2 and CIRCexplorer2, we detected thousands of circRNAs and performed a differential expression analysis. CircRNAs which exhibited statistically significantly differential expression were identified as AD-specific differentially expressed circRNAs. Notably, our investigation revealed 120 circRNAs with significant upregulation and 1325 circRNAs displaying significant downregulation in AD brains when compared to healthy brain tissue. Additionally, we explored the expression profiles of the linear RNA counterparts corresponding to differentially expressed circRNAs in AD-afflicted brains and discovered that the linear RNA counterparts exhibited no significant changes in the levels of expression. We used CRAFT tool to predict that circUBE4B had potential to target miRNA named as hsa-miR-325-5p, ultimately regulated CD44 gene. This study provides a comprehensive overview of differentially expressed circRNAs in the context of AD brains, underscoring their potential as molecular biomarkers for AD. These findings significantly enhance our comprehension of AD's underlying pathophysiological mechanisms, offering promising avenues for future diagnostic and therapeutic developments.


Subject(s)
Alzheimer Disease , MicroRNAs , RNA, Circular , Transcriptome , Humans , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , RNA, Circular/genetics , RNA, Circular/metabolism , MicroRNAs/genetics , MicroRNAs/metabolism , Male , Female , Aged , Hyaluronan Receptors/genetics , Hyaluronan Receptors/metabolism , Brain/metabolism , Biomarkers/metabolism
12.
Zhongguo Zhong Yao Za Zhi ; 49(11): 2906-2919, 2024 Jun.
Article in Chinese | MEDLINE | ID: mdl-39041150

ABSTRACT

Rheumatoid arthritis(RA) is a condition in which the joints are in a weakly acidic environment. In RA, RA fibroblastlike synoviocytes( RAFLS) in the joints become abnormally activated and secrete a large amount of matrix metalloproteinases(MMPs), and the receptor protein CD44 on the cell membrane is specifically upregulated. Xuetongsu(XTS), an active ingredient in the Tujia ethnomedicine Xuetong, is known to inhibit the proliferation of RAFLS. However, its development and utilization have been limited due to poor targeting ability. A biomimetic XTS-Prussian blue nanoparticles(PB NPs) drug delivery system called THMPX NPs which can target CD44 was constructed in this study. The surface of THMPX NPs was modified with hyaluronic acid(HA) and a long chain of triglycerol monostearate(TGMS) and 3-aminobenzeneboronic acid(PBA)(PBA-TGMS). The overexpressed MMPs and H+ in inflammatory RAFLS can synergistically cleave the PBA-TGMS on the surface of the nanoparticles, exposing HA to interact with CD44. This allows THMPX NPs to accumulate highly in RAFLS, and upon near-infrared light irradiation, generate heat and release XTS, thereby inhibiting the proliferation and migration of RAFLS. Characterization revealed that THMPX NPs were uniform cubes with a diameter of(190. 3±4. 7) nm and an average potential of(-15. 3± 2. 3) m V. Upon near-infrared light irradiation for 5 min, the temperature of THMPX NPs reached 41. 5 ℃, indicating MMPs and H+-triggered drug release. Safety assessments showed that THMPX NPs had a hemolysis rate of less than 4% and exhibited no cytotoxicity against normal RAW264. 7 and human fibroblast-like synoviocytes(HFLS). In vitro uptake experiments demonstrated the significant targeting ability of THMPX NPs to RAFLS. Free radical scavenging experiments revealed excellent free radical clearance capacity of THMPX NPs, capable of removing reactive oxygen species in RAFLS. Cell counting kit-8 and scratch assays demonstrated that THMPX NPs significantly suppressed the viability and migratory ability of RAFLS. This study provides insights into the development of innovative nanoscale targeted drugs from traditional ethnic medicines for RA treatment.


Subject(s)
Cell Movement , Cell Proliferation , Matrix Metalloproteinases , Nanoparticles , Cell Proliferation/drug effects , Cell Proliferation/radiation effects , Nanoparticles/chemistry , Humans , Cell Movement/drug effects , Cell Movement/radiation effects , Matrix Metalloproteinases/metabolism , Matrix Metalloproteinases/genetics , Ferrocyanides/chemistry , Hydrogen-Ion Concentration , Synoviocytes/drug effects , Synoviocytes/radiation effects , Synoviocytes/metabolism , Lasers , Hyaluronan Receptors/metabolism , Hyaluronan Receptors/genetics , Drugs, Chinese Herbal/chemistry , Drugs, Chinese Herbal/pharmacology , Arthritis, Rheumatoid/drug therapy , Arthritis, Rheumatoid/metabolism
13.
Int J Mol Sci ; 25(14)2024 Jul 11.
Article in English | MEDLINE | ID: mdl-39062846

ABSTRACT

Hyaluronan (HA) has gained significant attention in cancer research for its role in modulating chemoresistance. This review aims to elucidate the mechanisms by which HA contributes to chemoresistance, focusing on its interactions within the tumor microenvironment. HA is abundantly present in the extracellular matrix (ECM) and binds to cell-surface receptors such as CD44 and RHAMM. These interactions activate various signaling pathways, including PI3K/Akt, MAPK, and NF-κB, which are implicated in cell survival, proliferation, and drug resistance. HA also influences the physical properties of the tumor stroma, enhancing its density and reducing drug penetration. Additionally, HA-mediated signaling contributes to the epithelial-mesenchymal transition (EMT), a process associated with increased metastatic potential and resistance to apoptosis. Emerging therapeutic strategies aim to counteract HA-induced chemoresistance by targeting HA synthesis, degradation, metabolism, or its binding to CD44. This review underscores the complexity of HA's role in chemoresistance and highlights the potential for HA-targeted therapies to improve the efficacy of conventional chemotherapeutics.


Subject(s)
Drug Resistance, Neoplasm , Epithelial-Mesenchymal Transition , Extracellular Matrix , Hyaluronic Acid , Neoplasms , Signal Transduction , Tumor Microenvironment , Humans , Hyaluronic Acid/metabolism , Neoplasms/drug therapy , Neoplasms/metabolism , Neoplasms/pathology , Signal Transduction/drug effects , Extracellular Matrix/metabolism , Hyaluronan Receptors/metabolism , Antineoplastic Agents/therapeutic use , Antineoplastic Agents/pharmacology , Animals
14.
Mol Pharm ; 21(8): 4098-4115, 2024 Aug 05.
Article in English | MEDLINE | ID: mdl-39047292

ABSTRACT

Triple-negative breast cancer (TNBC) is the deadliest form of breast cancer with limited treatment options. The persistence of highly tumorigenic CD44-expressing subpopulation referred to as cancer stem cells (CSCs), endowed with the self-renewal capacity, has been associated with therapeutic resistance, hence clinical relapses. To mitigate these undesired events, targeted immunotherapies using antibody-photoconjugate (APC) or antibody-drug conjugate (ADC), were developed to specifically release cytotoxic payloads within targeted cells overexpressing cognate antigen receptors. Therefore, an αCD44(scFv)-SNAP-tag antibody fusion protein was engineered through genetic fusion of a single-chain antibody fragment (scFv) to a SNAPf-tag fusion protein, capable of self-conjugating with benzylguanine-modified light-sensitive near-infrared (NIR) phthalocyanine dye IRDye700DX (BG-IR700) or the small molecule toxin auristatin-F (BG-AURIF). Binding of the αCD44(scFv)-SNAPf-IR700 photoimmunoconjugate to antigen-positive cells was demonstrated by confocal microscopy and flow cytometry. By switching to NIR irradiation, CD44-expressing TNBC was selectively killed through induced phototoxic activities. Likewise, the αCD44(scFv)-SNAPf-AURIF immunoconjugate was able to selectively accumulate within targeted cells and significantly reduced cell viability through antimitotic activities at nano- to micromolar drug concentrations. This study provides an in vitro proof-of-concept for a future strategy to selectively destroy light-accessible superficial CD44-expressing TNBC tumors and their metastatic lesions which are inaccessible to therapeutic light.


Subject(s)
Aminobenzoates , Hyaluronan Receptors , Immunoconjugates , Oligopeptides , Triple Negative Breast Neoplasms , Humans , Triple Negative Breast Neoplasms/drug therapy , Triple Negative Breast Neoplasms/therapy , Triple Negative Breast Neoplasms/pathology , Hyaluronan Receptors/metabolism , Immunoconjugates/pharmacology , Cell Line, Tumor , Aminobenzoates/pharmacology , Aminobenzoates/chemistry , Female , Oligopeptides/pharmacology , Oligopeptides/chemistry , Single-Chain Antibodies/pharmacology , Immunotherapy/methods , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/metabolism
15.
J Nanobiotechnology ; 22(1): 452, 2024 Jul 30.
Article in English | MEDLINE | ID: mdl-39080641

ABSTRACT

Drug resistance to chemotherapy in cancers remains significant clinical challenges. CD44 modulates cellular adhesion, migration and growth, which plays a pivotal role in driving cancer resistance and even recurrence. Despite ongoing efforts, accurate, safe, and real-time dynamic monitoring techniques for CD44 expression remain inadequate in guiding the management of drug-resistant cancer treatment. In this study, we developed a nano-quenching and recovery detector of CD44 (Cy3-AptCD44@BPNSs) for visualizing cancer drug resistance. The fluorescence recovery of the detector is directly related to the CD44 expression level on cancer cells, which can be used to indicate the degree of drug resistance. It's confirmed that downregulating CD44 expression on cancer cells results in a corresponding decrease in the fluorescence intensity of the detector, which enables precise and dynamic monitoring of CD44. In addition, the Cy3-AptCD44@BPNSs also exhibited specificity in detecting CD44. This visualizing strategy may open up a wide range of possibilities for rapid recognition to cancer drug resistance, which is more efficient and flexible.


Subject(s)
Drug Resistance, Neoplasm , Hyaluronan Receptors , Hyaluronan Receptors/metabolism , Humans , Cell Line, Tumor , Neoplasms/drug therapy , Fluorescence , Antineoplastic Agents/pharmacology
16.
J Cell Mol Med ; 28(13): e18525, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38982317

ABSTRACT

Triple-negative breast cancer (TNBC) is often considered one of the most aggressive subtypes of breast cancer, characterized by a high recurrence rate and low overall survival (OS). It is notorious for posing challenges related to drug resistance. While there has been progress in TNBC research, the mechanisms underlying chemotherapy resistance in TNBC remain largely elusive. We collect single-cell RNA sequencing (scRNA-seq) data from five TNBC patients susceptible to chemotherapy and five resistant cases. Comprehensive analyses involving copy number variation (CNV), pseudotime trajectory, cell-cell interactions, pseudospace analysis, as well as transcription factor and functional enrichment are conducted specifically on macrophages and malignant cells. Furthermore, we performed validation experiments on clinical samples using multiplex immunofluorescence. We identified a subset of SPP1+ macrophages that secrete SPP1 signals interacting with CD44 on malignant cell surfaces, potentially activating the PDE3B pathway within malignant cells via the integrin pathway, leading to chemotherapy resistance. The abnormally enhanced SPP1 signal between macrophages and malignant cells may serve as a factor promoting chemotherapy resistance in TNBC patients. Therefore, SPP1+ macrophages could potentially serve as a therapeutic target to reduce chemotherapy resistance.


Subject(s)
Cell Communication , Drug Resistance, Neoplasm , Hyaluronan Receptors , Macrophages , Osteopontin , Single-Cell Analysis , Transcriptome , Triple Negative Breast Neoplasms , Humans , Hyaluronan Receptors/metabolism , Hyaluronan Receptors/genetics , Drug Resistance, Neoplasm/genetics , Osteopontin/metabolism , Osteopontin/genetics , Single-Cell Analysis/methods , Macrophages/metabolism , Triple Negative Breast Neoplasms/genetics , Triple Negative Breast Neoplasms/metabolism , Triple Negative Breast Neoplasms/pathology , Triple Negative Breast Neoplasms/drug therapy , Female , Gene Expression Regulation, Neoplastic , Cell Line, Tumor , Gene Expression Profiling
17.
Ren Fail ; 46(2): 2384586, 2024 Dec.
Article in English | MEDLINE | ID: mdl-39082695

ABSTRACT

Peritoneal dialysis (PD) is a widely used sustainable kidney replacement therapy. Prolonged use of PD fluids is associated with mesothelial-mesenchymal transition, peritoneal fibrosis, and eventual ultrafiltration (UF) failure. However, the impact of pressure on the peritoneum remains unclear. In the present study, we hypothesized increased pressure is a potential contributing factor to peritoneal fibrosis and investigated the possible mechanisms. In vitro experiments found that pressurization led to a mesenchymal phenotype, the expression of fibrotic markers and inflammatory factors in human mesothelial MeT-5A cells. Pressure also increased cell proliferation and augmented cell migration potential in MeT-5A cells. The mouse PD model and human peritoneum equilibrium test (PET) data both showed a positive association between higher pressure and increased small solute transport, along with decreased net UF. Mechanistically, we found that significant upregulation of CD44 in mesothelial cells upon pressurization. Notably, the treatment of CD44 neutralizing antibodies prevented pressure-induced phenotypic changes in mesothelial cells, while a CD44 inhibitor oligo-fucoidan ameliorated pressure-induced peritoneal thickening, fibrosis, and inflammation in PD mice. To conclude, intraperitoneal pressure results in peritoneal fibrosis in PD via CD44-mediated mesothelial changes and inflammation. CD44 blockage can be utilized as a novel preventive approach for PD-related peritoneal fibrosis and UF failure.


Subject(s)
Hyaluronan Receptors , Peritoneal Dialysis , Peritoneal Fibrosis , Peritoneum , Signal Transduction , Peritoneal Fibrosis/metabolism , Peritoneal Fibrosis/etiology , Peritoneal Fibrosis/pathology , Animals , Mice , Hyaluronan Receptors/metabolism , Humans , Peritoneum/pathology , Peritoneum/metabolism , Peritoneal Dialysis/adverse effects , Disease Models, Animal , Inflammation/metabolism , Pressure/adverse effects , Male , Cell Proliferation , Epithelial-Mesenchymal Transition , Mice, Inbred C57BL , Cell Line , Cell Movement
18.
Sheng Wu Gong Cheng Xue Bao ; 40(7): 2211-2222, 2024 Jul 25.
Article in Chinese | MEDLINE | ID: mdl-39044585

ABSTRACT

Renal pelvic carcinoma is a common upper urothelial cancer. The lack of an ideal in vitro model seriously hinders the research progress in the treatment for this disease. This study established a pipeline for the culture of renal pelvic carcinoma organoids based on the tumor tissue samples derived from the patients and tested the organoids to chemotherapeutic drugs. The results of immunohistochemistry and fluorescence experiments confirmed that the renal pelvic carcinoma organoids obtained from culture presented obvious nuclear heteromorphism, which was consistent with the tissue samples from renal pelvic carcinoma patients. The tumor marker molecule CD44 and the cell proliferation marker molecule Ki67 were positive in the organoids, indicating that the organoids were enriched with tumor stem cells and had strong proliferative ability. The renal pelvic carcinoma organoids were highly sensitive to pirarubicin, which had obvious killing effects. In brief, this study successfully established an in vitro model of renal pelvic cancer organoids and tested the sensitivity of the model to chemotherapeutic drugs. The results provide a new laboratory model for the individualized diagnosis and treatment of epithelial carcinomas represented by renal pelvic carcinoma.


Subject(s)
Antineoplastic Agents , Doxorubicin , Kidney Neoplasms , Organoids , Humans , Organoids/drug effects , Kidney Neoplasms/pathology , Kidney Neoplasms/drug therapy , Antineoplastic Agents/pharmacology , Doxorubicin/pharmacology , Doxorubicin/analogs & derivatives , Kidney Pelvis/pathology , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/pathology , Hyaluronan Receptors/metabolism , Cell Proliferation/drug effects , Carcinoma, Renal Cell/pathology , Carcinoma, Renal Cell/drug therapy
19.
BMC Oral Health ; 24(1): 820, 2024 Jul 19.
Article in English | MEDLINE | ID: mdl-39030509

ABSTRACT

BACKGROUND: There are 54,000 new cases of oral cavity and oropharyngeal cancer in the United States and more than 476,000 worldwide each year. Oral cavity and oropharyngeal squamous cell carcinoma make up most tumors with five-year survival rates of 50% due to prevalence of late-stage diagnoses. Improved methods of early detection in high-risk individuals are urgently needed. We aimed to assess the tumorigenic biomarkers soluble CD44 (solCD44) and total protein (TP) measured using oral rinses as affordable convenient screening tools for cancer detection. METHODS: In this prospective cohort study, we recruited 150 healthy current or former smokers through a community screening program. Baseline and four annual visits were conducted from March 2011-January 2016 with records followed until August 2020. Participants provided oral rinses, received head and neck exams, and completed questionnaires. SolCD44 and TP levels were measured and compared across groups and time. Participants were placed in the cancer group if malignancy developed in the study period, the suspicious group if physical exams were concerning for premalignant disease or cancer in the head and neck, and the healthy group if there were no suspicious findings. This analysis used two-sample t-test for comparison of means and two-sample Wilcoxon Test for comparison of medians. For subjects with follow-ups, estimated means of biomarkers were obtained from a fitted Repeated Measures Analysis of Variance (RANOVA) model including group, visit, and their interaction. Pairwise comparisons of mean solCD44 were made, including intergroup and intragroup comparison of values at different years. RESULTS: Most participants were males (58.7%), < 60 years of age. (90.7%), and Black (100%). Baseline mean solCD44 was elevated (2.781 ng/ml) in the cancer group compared to the suspicious group (1.849 ng/ml) and healthy group (1.779 ng/ml). CONCLUSION: This study supports the feasibility of a CD44-based oral rinse test as an affordable and convenient adjunctive tool for early detection of aerodigestive tract and other cancers in high-risk populations.


Subject(s)
Biomarkers, Tumor , Early Detection of Cancer , Hyaluronan Receptors , Mouth Neoplasms , Mouthwashes , Humans , Hyaluronan Receptors/analysis , Prospective Studies , Male , Female , Middle Aged , Mouth Neoplasms/diagnosis , Mouthwashes/therapeutic use , Biomarkers, Tumor/analysis , Biomarkers, Tumor/blood , Adult , Oropharyngeal Neoplasms , Aged
20.
Aging (Albany NY) ; 16(13): 10765-10783, 2024 Jul 09.
Article in English | MEDLINE | ID: mdl-38985127

ABSTRACT

The calcitonin receptor (CALCR) is an essential protein for maintaining calcium homeostasis and has been reported to be upregulated in numerous cancers. However, the molecular role of CALCR in renal cell carcinoma (RCC) is not well understood. In this study, we identified the overexpression of CALCR in RCC using human tissue chip by immunohistochemical (IHC) staining, which was associated with a poor prognosis. Functionally, CALCR depletion inhibited RCC cell proliferation and migration, and induced cell apoptosis and cycle arrest. CALCR is also essential for in vivo tumor formation. Mechanistically, we demonstrated that CALCR could directly bind to CD44, preventing CD44 protein degradation and thereby upregulating CD44 expression. Moreover, a deficiency in CD44 significantly attenuated the promoting role of CALCR on RCC cell proliferation, migration and anti-apoptosis capacities. Collectively, CALCR exacerbates RCC progression via stabilizing CD44, offering a fundamental basis for considering CALCR as a potential therapeutic target for RCC patients.


Subject(s)
Apoptosis , Carcinoma, Renal Cell , Cell Proliferation , Disease Progression , Hyaluronan Receptors , Kidney Neoplasms , Humans , Carcinoma, Renal Cell/pathology , Carcinoma, Renal Cell/metabolism , Carcinoma, Renal Cell/genetics , Hyaluronan Receptors/metabolism , Hyaluronan Receptors/genetics , Kidney Neoplasms/pathology , Kidney Neoplasms/metabolism , Kidney Neoplasms/genetics , Cell Line, Tumor , Animals , Cell Movement , Mice , Male , Gene Expression Regulation, Neoplastic , Female
SELECTION OF CITATIONS
SEARCH DETAIL