Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 5.051
Filter
1.
J Vet Sci ; 25(3): e35, 2024 May.
Article in English | MEDLINE | ID: mdl-38834505

ABSTRACT

IMPORTANCE: Experimental autoimmune encephalomyelitis (EAE) is an animal model of multiple sclerosis characterized by inflammation within the central nervous system. However, inflammation in non-neuronal tissues, including the lungs, has not been fully evaluated. OBJECTIVE: This study evaluated the inflammatory response in lungs of EAE mice by immunohistochemistry and histochemistry. METHODS: Eight adult C57BL/6 mice were injected with myelin oligodendrocyte glycoprotein35-55 to induce the EAE. Lungs and spinal cords were sampled from the experimental mice at the time of sacrifice and used for the western blotting, histochemistry, and immunohistochemistry. RESULTS: Histopathological examination revealed inflammatory lesions in the lungs of EAE mice, characterized by infiltration of myeloperoxidase (MPO)- and galectin-3-positive cells, as determined by immunohistochemistry. Increased numbers of collagen fibers in the lungs of EAE mice were confirmed by histopathological analysis. Western blotting revealed significantly elevated level of osteopontin (OPN), cluster of differentiation 44 (CD44), MPO and galectin-3 in the lungs of EAE mice compared with normal controls (p < 0.05). Immunohistochemical analysis revealed both OPN and CD44 in ionized calcium-binding adapter molecule 1-positive macrophages within the lungs of EAE mice. CONCLUSIONS AND RELEVANCE: Taken together, these findings suggest that the increased OPN level in lungs of EAE mice led to inflammation; concurrent increases in proinflammatory factors (OPN and galectin-3) caused pulmonary impairment.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental , Lung , Mice, Inbred C57BL , Animals , Encephalomyelitis, Autoimmune, Experimental/pathology , Mice , Lung/pathology , Female , Immunohistochemistry , Osteopontin/metabolism , Galectin 3/metabolism , Peroxidase/metabolism , Hyaluronan Receptors/metabolism , Spinal Cord/pathology , Inflammation/pathology , Blotting, Western
2.
Sci Adv ; 10(23): eadl6083, 2024 Jun 07.
Article in English | MEDLINE | ID: mdl-38838151

ABSTRACT

Hepatocellular carcinoma (HCC) acquires an immunosuppressive microenvironment, leading to unbeneficial therapeutic outcomes. Hyaluronan-mediated motility receptor (HMMR) plays a crucial role in tumor progression. Here, we found that aberrant expression of HMMR could be a predictive biomarker for the immune suppressive microenvironment of HCC, but the mechanism remains unclear. We established an HMMR-/- liver cancer mouse model to elucidate the HMMR-mediated mechanism of the dysregulated "don't eat me" signal. HMMR knockout inhibited liver cancer growth and induced phagocytosis. HMMRhigh liver cancer cells escaped from phagocytosis via sustaining CD47 signaling. Patients with HMMRhighCD47high expression showed a worse prognosis than those with HMMRlowCD47low expression. HMMR formed a complex with FAK/SRC in the cytoplasm to activate NF-κB signaling, which could be independent of membrane interaction with CD44. Notably, targeting HMMR could enhance anti-PD-1 treatment efficiency by recruiting CD8+ T cells. Overall, our data revealed a regulatory mechanism of the "don't eat me" signal and knockdown of HMMR for enhancing anti-PD-1 treatment.


Subject(s)
CD47 Antigen , Carcinoma, Hepatocellular , Hyaluronan Receptors , Liver Neoplasms , Phagocytes , Phagocytosis , Carcinoma, Hepatocellular/pathology , Carcinoma, Hepatocellular/immunology , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/genetics , Liver Neoplasms/pathology , Liver Neoplasms/immunology , Liver Neoplasms/metabolism , Liver Neoplasms/genetics , Animals , Humans , Mice , Hyaluronan Receptors/metabolism , Hyaluronan Receptors/genetics , Phagocytes/metabolism , Phagocytes/immunology , CD47 Antigen/metabolism , CD47 Antigen/genetics , Cell Line, Tumor , Signal Transduction , Tumor Microenvironment/immunology , Immune Evasion , NF-kappa B/metabolism , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Mice, Knockout , Focal Adhesion Kinase 1/metabolism , Focal Adhesion Kinase 1/genetics , Tumor Escape
3.
Int J Nanomedicine ; 19: 4893-4906, 2024.
Article in English | MEDLINE | ID: mdl-38828202

ABSTRACT

Introduction: The tumor microenvironment (TME) has attracted considerable attention as a potential therapeutic target for cancer. High levels of reactive oxygen species (ROS) in the TME may act as a stimulus for drug release. In this study, we have developed ROS-responsive hyaluronic acid-bilirubin nanoparticles (HABN) loaded with doxorubicin (DOX@HABN) for the specific delivery and release of DOX in tumor tissue. The hyaluronic acid shell of the nanoparticles acts as an active targeting ligand that can specifically bind to CD44-overexpressing tumors. The bilirubin core has intrinsic anti-cancer activity and ROS-responsive solubility change properties. Methods & Results: DOX@HABN showed the HA shell-mediated targeting ability, ROS-responsive disruption leading to ROS-mediated drug release, and synergistic anti-cancer activity against ROS-overproducing CD44-overexpressing HeLa cells. Additionally, intravenously administered HABN-Cy5.5 showed remarkable tumor-targeting ability in HeLa tumor-bearing mice with limited distribution in major organs. Finally, intravenous injection of DOX@HABN into HeLa tumor-bearing mice showed synergistic anti-tumor efficacy without noticeable side effects. Conclusion: These findings suggest that DOX@HABN has significant potential as a cancer-targeting and TME ROS-responsive nanomedicine for targeted cancer treatment.


Subject(s)
Bilirubin , Doxorubicin , Hyaluronan Receptors , Hyaluronic Acid , Nanomedicine , Nanoparticles , Reactive Oxygen Species , Tumor Microenvironment , Hyaluronic Acid/chemistry , Tumor Microenvironment/drug effects , Animals , Reactive Oxygen Species/metabolism , Humans , Doxorubicin/pharmacology , Doxorubicin/chemistry , Doxorubicin/pharmacokinetics , Doxorubicin/administration & dosage , Nanoparticles/chemistry , Mice , HeLa Cells , Hyaluronan Receptors/metabolism , Bilirubin/chemistry , Bilirubin/pharmacology , Bilirubin/pharmacokinetics , Drug Liberation , Mice, Inbred BALB C , Mice, Nude , Xenograft Model Antitumor Assays , Drug Carriers/chemistry , Drug Carriers/pharmacokinetics , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/administration & dosage , Neoplasms/drug therapy , Neoplasms/metabolism
4.
Mol Biol Rep ; 51(1): 641, 2024 May 10.
Article in English | MEDLINE | ID: mdl-38727798

ABSTRACT

BACKGROUND: The interrelationship between cellular metabolism and the epithelial-to-mesenchymal transition (EMT) process has made it an interesting topic to investigate the adjuvant effect of therapeutic diets in the treatment of cancers. However, the findings are controversial. In this study, the effects of glucose limitation along and with the addition of beta-hydroxybutyrate (bHB) were examined on the expression of specific genes and proteins of EMT, Wnt, Hedgehog, and Hippo signaling pathways, and also on cellular behavior of gastric cancer stem-like (MKN-45) and non-stem-like (KATO III) cells. METHODS AND RESULTS: The expression levels of chosen genes and proteins studied in cancer cells gradually adopted a low-glucose condition of one-fourth, along and with the addition of bHB, and compared to the unconditioned control cells. The long-term switching of the metabolic fuels successfully altered the expression profiles and behaviors of both gastric cancer cells. However, the results for some changes were the opposite. Glucose limitation along and with the addition of bHB reduced the CD44+ population in MKN-45 cells. In KATO III cells, glucose restriction increased the CD44+ population. Glucose deprivation alleviated EMT-related signaling pathways in MKN-45 cells but stimulated EMT in KATO III cells. Interestingly, bHB enrichment reduced the beneficial effect of glucose starvation in MKN-45 cells, but also alleviated the adverse effects of glucose restriction in KATO III cells. CONCLUSIONS: The findings of this research clearly showed that some controversial results in clinical trials for ketogenic diet in cancer patients stemmed from the different signaling responses of various cells to the metabolic changes in a heterogeneous cancer mass.


Subject(s)
3-Hydroxybutyric Acid , Epithelial-Mesenchymal Transition , Glucose , Signal Transduction , Stomach Neoplasms , Epithelial-Mesenchymal Transition/genetics , Stomach Neoplasms/metabolism , Stomach Neoplasms/genetics , Stomach Neoplasms/pathology , Humans , Cell Line, Tumor , 3-Hydroxybutyric Acid/pharmacology , 3-Hydroxybutyric Acid/metabolism , Glucose/metabolism , Ketosis/metabolism , Gene Expression Regulation, Neoplastic , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Hyaluronan Receptors/metabolism , Hyaluronan Receptors/genetics
5.
Nat Commun ; 15(1): 3904, 2024 May 09.
Article in English | MEDLINE | ID: mdl-38724502

ABSTRACT

Chronic wounds are a major complication in patients with diabetes. Here, we identify a therapeutic circRNA and load it into small extracellular vesicles (sEVs) to treat diabetic wounds in preclinical models. We show that circCDK13 can stimulate the proliferation and migration of human dermal fibroblasts and human epidermal keratinocytes by interacting with insulin-like growth factor 2 mRNA binding protein 3 in an N6-Methyladenosine-dependent manner to enhance CD44 and c-MYC expression. We engineered sEVs that overexpress circCDK13 and show that local subcutaneous injection into male db/db diabetic mouse wounds and wounds of streptozotocin-induced type I male diabetic rats could accelerate wound healing and skin appendage regeneration. Our study demonstrates that the delivery of circCDK13 in sEVs may present an option for diabetic wound treatment.


Subject(s)
Cell Proliferation , Diabetes Mellitus, Experimental , Extracellular Vesicles , Fibroblasts , Keratinocytes , RNA, Circular , Wound Healing , Animals , Extracellular Vesicles/metabolism , Extracellular Vesicles/transplantation , Wound Healing/drug effects , Humans , Male , Mice , Rats , Fibroblasts/metabolism , RNA, Circular/genetics , RNA, Circular/metabolism , Keratinocytes/metabolism , Cell Movement , Skin/metabolism , Hyaluronan Receptors/metabolism , Hyaluronan Receptors/genetics , Proto-Oncogene Proteins c-myc/metabolism , Proto-Oncogene Proteins c-myc/genetics , Mice, Inbred C57BL , Disease Models, Animal , Rats, Sprague-Dawley , RNA-Binding Proteins/metabolism , RNA-Binding Proteins/genetics
6.
Cell Commun Signal ; 22(1): 291, 2024 May 27.
Article in English | MEDLINE | ID: mdl-38802835

ABSTRACT

A promising new therapy option for acute kidney injury (AKI) is mesenchymal stem cells (MSCs). However, there are several limitations to the use of MSCs, such as low rates of survival, limited homing capacity, and unclear differentiation. In search of better therapeutic strategies, we explored all-trans retinoic acid (ATRA) pretreatment of MSCs to observe whether it could improve the therapeutic efficacy of AKI. We established a renal ischemia/reperfusion injury model and treated mice with ATRA-pretreated MSCs via tail vein injection. We found that AKI mice treated with ATRA-MSCs significantly improved renal function compared with DMSO-MSCs treatment. RNA sequencing screened that hyaluronic acid (HA) production from MSCs promoted by ATRA. Further validation by chromatin immunoprecipitation experiments verified that retinoic acid receptor RARα/RXRγ was a potential transcription factor for hyaluronic acid synthase 2. Additionally, an in vitro hypoxia/reoxygenation model was established using human proximal tubular epithelial cells (HK-2). After co-culturing HK-2 cells with ATRA-pretreated MSCs, we observed that HA binds to cluster determinant 44 (CD44) and activates the PI3K/AKT pathway, which enhances the anti-inflammatory, anti-apoptotic, and proliferative repair effects of MSCs in AKI. Inhibition of the HA/CD44 axis effectively reverses the renal repair effect of ATRA-pretreated MSCs. Taken together, our study suggests that ATRA pretreatment promotes HA production by MSCs and activates the PI3K/AKT pathway in renal tubular epithelial cells, thereby enhancing the efficacy of MSCs against AKI.


Subject(s)
Acute Kidney Injury , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells , Tretinoin , Acute Kidney Injury/therapy , Acute Kidney Injury/pathology , Acute Kidney Injury/metabolism , Acute Kidney Injury/drug therapy , Animals , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/cytology , Tretinoin/pharmacology , Tretinoin/therapeutic use , Humans , Mice , Male , Mice, Inbred C57BL , Hyaluronic Acid/pharmacology , Hyaluronan Receptors/metabolism , Hyaluronan Receptors/genetics , Proto-Oncogene Proteins c-akt/metabolism , Cell Line , Phosphatidylinositol 3-Kinases/metabolism , Signal Transduction/drug effects , Reperfusion Injury/therapy , Reperfusion Injury/drug therapy , Reperfusion Injury/pathology , Reperfusion Injury/metabolism , Disease Models, Animal , Apoptosis/drug effects
7.
Biomolecules ; 14(5)2024 May 20.
Article in English | MEDLINE | ID: mdl-38786011

ABSTRACT

The development of hydrogels that allow vascular endothelial cells to form capillary-like networks is critical for advancing tissue engineering and drug discovery. In this study, we developed hydrogels composed of phenolated hyaluronic acid (HA-Ph) with an average molecular weight of 490-159 kDa via sonication in an aqueous solution. These hydrogels were synthesized by the horseradish peroxidase-catalyzed crosslinking of phenol moieties in the presence of hydrogen peroxide and phenolated gelatin. The sonication-degraded HA-Ph (198 kDa) significantly enhanced the migration ability of human umbilical vein endothelial cells (HUVECs) on cell culture plates when added to the medium compared to the original HA-Ph (490 kDa) and less-degraded HA-Ph (312-399 kDa). In addition, HUVECs cultured on these hydrogels formed networks that did not occur on hydrogels made from the original HA-Ph. CD44 expression and PI3K gene expression, both markers related to angiogenesis, were 3.5- and 1.8-fold higher, respectively, in cells cultured on sonication-degraded HA-Ph hydrogels than in those cultured on hydrogels comprising the original HA-Ph. These results highlight the potential of hydrogels containing sonication-degraded HA-Ph for tissue engineering and drug-screening applications involving human vascular endothelial cells.


Subject(s)
Human Umbilical Vein Endothelial Cells , Hyaluronan Receptors , Hyaluronic Acid , Hydrogels , Neovascularization, Physiologic , Humans , Hyaluronic Acid/chemistry , Hyaluronic Acid/pharmacology , Hydrogels/chemistry , Hydrogels/pharmacology , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , Hyaluronan Receptors/metabolism , Neovascularization, Physiologic/drug effects , Cell Movement/drug effects , Tissue Engineering/methods , Phosphatidylinositol 3-Kinases/metabolism , Ultrasonic Waves
8.
Front Immunol ; 15: 1354992, 2024.
Article in English | MEDLINE | ID: mdl-38736891

ABSTRACT

CD44 is a ubiquitous leukocyte adhesion molecule involved in cell-cell interaction, cell adhesion, migration, homing and differentiation. CD44 can mediate the interaction between leukemic stem cells and the surrounding extracellular matrix, thereby inducing a cascade of signaling pathways to regulate their various behaviors. In this review, we focus on the impact of CD44s/CD44v as biomarkers in leukemia development and discuss the current research and prospects for CD44-related interventions in clinical application.


Subject(s)
Biomarkers, Tumor , Hyaluronan Receptors , Leukemia , Neoplastic Stem Cells , Humans , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/immunology , Hyaluronan Receptors/metabolism , Leukemia/metabolism , Leukemia/therapy , Leukemia/immunology , Biomarkers, Tumor/metabolism , Animals , Signal Transduction , Molecular Targeted Therapy
9.
Int J Mol Sci ; 25(9)2024 Apr 26.
Article in English | MEDLINE | ID: mdl-38731968

ABSTRACT

Cluster of differentiation 44 (CD44), a multi-functional cell surface receptor, has several variants and is ubiquitously expressed in various cells and tissues. CD44 is well known for its function in cell adhesion and is also involved in diverse cellular responses, such as proliferation, migration, differentiation, and activation. To date, CD44 has been extensively studied in the field of cancer biology and has been proposed as a marker for cancer stem cells. Recently, growing evidence suggests that CD44 is also relevant in non-cancer diseases. In liver disease, it has been shown that CD44 expression is significantly elevated and associated with pathogenesis by impacting cellular responses, such as metabolism, proliferation, differentiation, and activation, in different cells. However, the mechanisms underlying CD44's function in liver diseases other than liver cancer are still poorly understood. Hence, to help to expand our knowledge of the role of CD44 in liver disease and highlight the need for further research, this review provides evidence of CD44's effects on liver physiology and its involvement in the pathogenesis of liver disease, excluding cancer. In addition, we discuss the potential role of CD44 as a key regulator of cell physiology.


Subject(s)
Hyaluronan Receptors , Liver Diseases , Liver , Humans , Hyaluronan Receptors/metabolism , Liver/metabolism , Liver/pathology , Liver Diseases/metabolism , Liver Diseases/pathology , Animals , Cell Differentiation
10.
Mol Biol Rep ; 51(1): 693, 2024 May 25.
Article in English | MEDLINE | ID: mdl-38796656

ABSTRACT

BACKGROUND: Testicular germ cell tumors (TGCTs) exhibit diverse biological and pathological features and are divided in two main types, seminomas and nonseminomatous germ cell tumors (NSGCTs). CD44 is a cell surface receptor, which is highly expressed in malignancies and is implicated in tumorigenesis affecting cell-matrix interactions and cell signaling. METHODS AND RESULTS: Here, we examined the expression of CD44 in tumor cell lines and in patients' material. We found that CD44 is over-expressed in TGCTs compared to normal tissues. Immunohistochemical staining in 71 tissue specimens demonstrated increased expression of CD44 in some patients, whereas CD44 was absent in normal tissue. In seminomas, a high percentage of tumor and stromal cells showed cytoplasmic and/or cell surface staining for CD44 as well as increased staining for CD44 in the tumor stroma was found in some cases. The increased expression of CD44 either in tumor cells or in stromal components was associated with tumor size, nodal metastasis, vascular/lymphatic invasion, and disease stage only in seminomas. The increased stromal expression of CD44 in TGCTs was positively associated with angiogenesis. CONCLUSIONS: CD44 may exhibit diverse biological functions in seminomas and NSGCTs. The expression of CD44 in tumor cells as well as in tumor stroma fosters an aggressive phenotype in seminomas and should be considered in disease treatment.


Subject(s)
Hyaluronan Receptors , Seminoma , Testicular Neoplasms , Humans , Hyaluronan Receptors/metabolism , Seminoma/metabolism , Seminoma/pathology , Seminoma/genetics , Male , Testicular Neoplasms/metabolism , Testicular Neoplasms/pathology , Adult , Cell Line, Tumor , Middle Aged , Neoplasms, Germ Cell and Embryonal/metabolism , Neoplasms, Germ Cell and Embryonal/pathology , Neoplasms, Germ Cell and Embryonal/genetics , Biomarkers, Tumor/metabolism , Gene Expression Regulation, Neoplastic , Immunohistochemistry/methods
11.
Sci Rep ; 14(1): 11431, 2024 05 19.
Article in English | MEDLINE | ID: mdl-38763930

ABSTRACT

Our current study reports the successful synthesis of thiolated chitosan-based nanoparticles for targeted drug delivery of 5-Fluorouracil. This process was achieved through the ionic gelation technique, aiming to improve the efficacy of the chemotherapeutic moiety by modifying the surface of the nanoparticles (NPs) with a ligand. We coated these NPs with hyaluronic acid (HA) to actively target the CD44 receptor, which is frequently overexpressed in various solid malignancies, including breast cancer. XRD, FTIR, SEM, and TEM were used for the physicochemical analysis of the NPs. These 5-Fluorouracil (5-FU) loaded NPs were evaluated on MDA-MB-231 (a triple-negative breast cell line) and MCF-10A (normal epithelial breast cells) to determine their in vitro efficacy. The developed 5-FU-loaded NPs exhibited a particle size within a favorable range (< 300 nm). The positive zeta potential of these nanoparticles facilitated their uptake by negatively charged cancer cells. Moreover, they demonstrated robust stability and achieved high encapsulation efficiency. These nanoparticles exhibited significant cytotoxicity compared to the crude drug (p < 0.05) and displayed a promising release pattern consistent with the basic diffusion model. These traits improve the pharmacokinetic profile, efficacy, and ability to precisely target these nanoparticles, offering a potentially successful anticancer treatment for breast cancer. However, additional in vivo assessments of these formulations are obligatory to confirm these findings.


Subject(s)
Chitosan , Fluorouracil , Hyaluronan Receptors , Nanoparticles , Triple Negative Breast Neoplasms , Fluorouracil/administration & dosage , Fluorouracil/pharmacology , Fluorouracil/chemistry , Chitosan/chemistry , Humans , Hyaluronan Receptors/metabolism , Triple Negative Breast Neoplasms/drug therapy , Triple Negative Breast Neoplasms/metabolism , Triple Negative Breast Neoplasms/pathology , Nanoparticles/chemistry , Cell Line, Tumor , Female , Drug Carriers/chemistry , Hyaluronic Acid/chemistry , Drug Delivery Systems , Antimetabolites, Antineoplastic/pharmacology , Antimetabolites, Antineoplastic/administration & dosage , Antimetabolites, Antineoplastic/pharmacokinetics , Antimetabolites, Antineoplastic/chemistry , Cell Survival/drug effects , Particle Size
12.
J Endocrinol ; 262(1)2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38692289

ABSTRACT

CD44, a cell surface adhesion receptor and stem cell biomarker, is recently implicated in chronic metabolic diseases. Ablation of CD44 ameliorates adipose tissue inflammation and insulin resistance in obesity. Here, we investigated cell type-specific CD44 expression in human and mouse adipose tissue and further studied how CD44 in preadipocytes regulates adipocyte function. Using Crispr Cas9-mdediated gene deletion and lentivirus-mediated gene re-expression, we discovered that deletion of CD44 promotes adipocyte differentiation and adipogenesis, whereas re-expression of CD44 abolishes this effect and decreases insulin responsiveness and adiponectin secretion in 3T3-L1 cells. Mechanistically, CD44 does so via suppressing Pparg expression. Using quantitative proteomics analysis, we further discovered that cell cycle-regulated pathways were mostly decreased by deletion of CD44. Indeed, re-expression of CD44 moderately restored expression of proteins involved in all phases of the cell cycle. These data were further supported by increased preadipocyte proliferation rates in CD44-deficient cells and re-expression of CD44 diminished this effect. Our data suggest that CD44 plays a crucial role in regulating adipogenesis and adipocyte function possibly through regulating PPARγ and cell cycle-related pathways. This study provides evidence for the first time that CD44 expressed in preadipocytes plays key roles in regulating adipocyte function outside immune cells where CD44 is primarily expressed. Therefore, targeting CD44 in (pre)adipocytes may provide therapeutic potential to treat obesity-associated metabolic complications.


Subject(s)
3T3-L1 Cells , Adipocytes , Adipogenesis , Cell Cycle , Hyaluronan Receptors , PPAR gamma , Adipogenesis/genetics , Adipogenesis/physiology , Hyaluronan Receptors/metabolism , Hyaluronan Receptors/genetics , Animals , PPAR gamma/metabolism , PPAR gamma/genetics , Mice , Cell Cycle/genetics , Cell Cycle/physiology , Humans , Adipocytes/metabolism , Gene Deletion , Cell Differentiation/genetics , Male , Adipose Tissue/metabolism , Adipose Tissue/cytology , Signal Transduction/physiology
13.
Int J Biol Macromol ; 269(Pt 1): 132074, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38705320

ABSTRACT

Treatment for triple negative breast cancer (TNBC) remains a huge challenge due to the lack of targeted therapeutics and tumor heterogenicity. Cisplatin (Cis) have demonstrated favorable therapeutic response in TNBC and thus is used together with various kinase inhibitors to fight the heterogenicity of TNBC. The combination of Cis with SRC inhibitor dasatinib (DAS) has shown encouraging anti-TNBC efficacy although the additive toxicity was commonly observed. To overcome the severe side effects of this Cis involved therapy, here we co-encapsulated Cis and DAS into a self-assembled hyaluronan (HA) nanogel (designated as HA/Cis/DAS (HCD) nanogel) to afford the TNBC targeted delivery by using the 4T1 mouse model. The acquired HCD nanogel was around 181 nm in aqueous solution, demonstrating the pharmacological activities of both Cis and DAS. Taking advantages of HA's targeting capability towards CD44 that is overexpressed on many TNBC cells, the HCD could well maintain the anticancer efficacy of the Cis and DAS combination, significantly increase the maximum tolerated dose and relieve the renal toxicity in vivo. The current HCD nanogel provides a potent strategy to improve the therapeutic outcome of Cis and DAS combination and thus representing a new targeted treatment option for TNBC.


Subject(s)
Cisplatin , Dasatinib , Hyaluronic Acid , Nanogels , Triple Negative Breast Neoplasms , Triple Negative Breast Neoplasms/drug therapy , Triple Negative Breast Neoplasms/pathology , Hyaluronic Acid/chemistry , Animals , Dasatinib/pharmacology , Dasatinib/chemistry , Mice , Cisplatin/pharmacology , Cisplatin/chemistry , Female , Nanogels/chemistry , Cell Line, Tumor , Humans , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Polyethyleneimine/chemistry , Mice, Inbred BALB C , Hyaluronan Receptors/metabolism
14.
J Biomed Sci ; 31(1): 54, 2024 May 24.
Article in English | MEDLINE | ID: mdl-38790021

ABSTRACT

BACKGROUND: Alcohol-related liver disease (ALD) is a major health concern worldwide, but effective therapeutics for ALD are still lacking. Tumor necrosis factor-inducible gene 6 protein (TSG-6), a cytokine released from mesenchymal stem cells, was shown to reduce liver fibrosis and promote successful liver repair in mice with chronically damaged livers. However, the effect of TSG-6 and the mechanism underlying its activity in ALD remain poorly understood. METHODS: To investigate its function in ALD mice with fibrosis, male mice chronically fed an ethanol (EtOH)-containing diet for 9 weeks were treated with TSG-6 (EtOH + TSG-6) or PBS (EtOH + Veh) for an additional 3 weeks. RESULTS: Severe hepatic injury in EtOH-treated mice was markedly decreased in TSG-6-treated mice fed EtOH. The EtOH + TSG-6 group had less fibrosis than the EtOH + Veh group. Activation of cluster of differentiation 44 (CD44) was reported to promote HSC activation. CD44 and nuclear CD44 intracellular domain (ICD), a CD44 activator which were upregulated in activated HSCs and ALD mice were significantly downregulated in TSG-6-exposed mice fed EtOH. TSG-6 interacted directly with the catalytic site of MMP14, a proteolytic enzyme that cleaves CD44, inhibited CD44 cleavage to CD44ICD, and reduced HSC activation and liver fibrosis in ALD mice. In addition, a novel peptide designed to include a region that binds to the catalytic site of MMP14 suppressed CD44 activation and attenuated alcohol-induced liver injury, including fibrosis, in mice. CONCLUSIONS: These results demonstrate that TSG-6 attenuates alcohol-induced liver damage and fibrosis by blocking CD44 cleavage to CD44ICD and suggest that TSG-6 and TSG-6-mimicking peptide could be used as therapeutics for ALD with fibrosis.


Subject(s)
Cell Adhesion Molecules , Hyaluronan Receptors , Liver Cirrhosis , Liver Diseases, Alcoholic , Animals , Hyaluronan Receptors/metabolism , Hyaluronan Receptors/genetics , Mice , Cell Adhesion Molecules/metabolism , Cell Adhesion Molecules/genetics , Male , Liver Diseases, Alcoholic/metabolism , Liver Diseases, Alcoholic/drug therapy , Liver Cirrhosis/metabolism , Liver Cirrhosis/drug therapy , Liver Cirrhosis/chemically induced , Mice, Inbred C57BL , Peptides/pharmacology , Peptides/metabolism , Ethanol
15.
Genes (Basel) ; 15(5)2024 Apr 24.
Article in English | MEDLINE | ID: mdl-38790166

ABSTRACT

Clear cell RCC (ccRCC) represents the most common type of kidney cancer, with surgery being the only potential curative treatment. Almost one-third of ccRCC patients relapse either locally or as cases of distant metastases. Several biomarkers have been employed in order to separate ccRCC patients with better prognosis or to predict treatment outcomes, with limited results. CD44 is a membrane glycoprotein with multiple roles in normal development but also cancer. Recently, the CD44 standard isoform has been implicated in tumor progression and the metastasis cascade through microenvironment interactions. Here, through CD44 immunohistochemical staining of ccRCC patient samples and TCGA data analysis, we sought to elucidate the expression patterns (mRNA and protein) of CD44 in clear cell RCC and correlate its expression with clinicopathological parameters. We were able to show that CD44 expression presents a positive association with tumor grade and overall survival, predicting a worse patient outcome in ccRCC. In addition, our data indicate that the CD44 mRNA upregulation can be attributed to reduced gene methylation, implicating epigenetic gene regulation in ccRCC development and progression.


Subject(s)
Carcinoma, Renal Cell , DNA Methylation , Gene Expression Regulation, Neoplastic , Hyaluronan Receptors , Kidney Neoplasms , Humans , Carcinoma, Renal Cell/genetics , Carcinoma, Renal Cell/pathology , Carcinoma, Renal Cell/mortality , Carcinoma, Renal Cell/metabolism , Hyaluronan Receptors/genetics , Hyaluronan Receptors/metabolism , Kidney Neoplasms/genetics , Kidney Neoplasms/pathology , Kidney Neoplasms/mortality , Kidney Neoplasms/metabolism , Female , Male , Middle Aged , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Neoplasm Grading , Aged , Prognosis , Adult
16.
ACS Appl Mater Interfaces ; 16(21): 27055-27064, 2024 May 29.
Article in English | MEDLINE | ID: mdl-38757711

ABSTRACT

A major contributing cause to breast cancer related death is metastasis. Moreover, breast cancer metastasis often shows little symptoms until a large area of the organs is occupied by metastatic cancer cells. Breast cancer multimodal imaging is attractive since it integrates advantages from several modalities, enabling more accurate cancer detection. Glycoprotein CD44 is overexpressed on most breast cancer cells and is the primary cell surface receptor for hyaluronan (HA). To facilitate breast cancer diagnosis, we report an indocyanine green (ICG) and HA conjugated iron oxide nanoparticle (NP-ICG-HA), which enabled active targeting to breast cancer by HA-CD44 interaction and detected metastasis with magnetic particle imaging (MPI) and near-infrared fluorescence imaging (NIR-FI). When evaluated in a transgenic breast cancer mouse model, NP-ICG-HA enabled the detection of multiple breast tumors in MPI and NIR-FI, providing more comprehensive images and a diagnosis of breast cancer. Furthermore, NP-ICG-HAs were evaluated in a lung metastasis model. Upon NP-ICG-HA administration, MPI showed clear signals in the lungs, indicating the tumor sites. This is the first time that HA-based NPs have enabled MPI of cancer. NP-ICG-HAs are an attractive platform for noninvasive detection of primary breast cancer and lung metastasis.


Subject(s)
Breast Neoplasms , Hyaluronic Acid , Indocyanine Green , Lung Neoplasms , Optical Imaging , Hyaluronic Acid/chemistry , Animals , Lung Neoplasms/diagnostic imaging , Lung Neoplasms/secondary , Lung Neoplasms/pathology , Female , Mice , Breast Neoplasms/diagnostic imaging , Breast Neoplasms/pathology , Humans , Indocyanine Green/chemistry , Hyaluronan Receptors/metabolism , Cell Line, Tumor , Magnetite Nanoparticles/chemistry , Magnetic Iron Oxide Nanoparticles/chemistry
17.
Sci Rep ; 14(1): 10583, 2024 05 08.
Article in English | MEDLINE | ID: mdl-38719848

ABSTRACT

Identifying marker combinations for robust prognostic validation in primary tumour compartments remains challenging. We aimed to assess the prognostic significance of CSC markers (ALDH1, CD44, p75NTR, BMI-1) and E-cadherin biomarkers in OSCC. We analysed 94 primary OSCC and 67 metastatic lymph node samples, including central and invasive tumour fronts (ITF), along with clinicopathological data. We observed an increase in ALDH1+/CD44+/BMI-1- tumour cells in metastatic lesions compared to primary tumours. Multivariate analysis highlighted that elevated p75NTR levels (at ITF) and reduced E-cadherin expression (at the tumour centre) independently predicted metastasis, whilst ALDH1high exhibited independent predictive lower survival at the ITF, surpassing the efficacy of traditional tumour staging. Then, specifically at the ITF, profiles characterized by CSChighE-cadherinlow (ALDH1highp75NTRhighE-cadherinlow) and CSCintermediateE-cadherinlow (ALDH1 or p75NTRhighE-cadherinlow) were significantly associated with worsened overall survival and increased likelihood of metastasis in OSCC patients. In summary, our study revealed diverse tumour cell profiles in OSCC tissues, with varying CSC and E-cadherin marker patterns across primary tumours and metastatic sites. Given the pivotal role of reduced survival rates as an indicator of unfavourable prognosis, the immunohistochemistry profile identified as CSChighE-cadherinlow at the ITF of primary tumours, emerges as a preferred prognostic marker closely linked to adverse outcomes in OSCC.


Subject(s)
Aldehyde Dehydrogenase 1 Family , Biomarkers, Tumor , Cadherins , Carcinoma, Squamous Cell , Mouth Neoplasms , Adult , Aged , Female , Humans , Male , Middle Aged , Aldehyde Dehydrogenase 1 Family/metabolism , Biomarkers, Tumor/metabolism , Cadherins/metabolism , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/pathology , Carcinoma, Squamous Cell/mortality , Hyaluronan Receptors/metabolism , Immunohistochemistry , Lymphatic Metastasis , Mouth Neoplasms/pathology , Mouth Neoplasms/metabolism , Mouth Neoplasms/mortality , Mouth Neoplasms/diagnosis , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Nerve Tissue Proteins/metabolism , Polycomb Repressive Complex 1/metabolism , Polycomb Repressive Complex 1/genetics , Prognosis , Receptors, Nerve Growth Factor/metabolism , Retinal Dehydrogenase/metabolism
18.
Mol Med ; 30(1): 61, 2024 May 17.
Article in English | MEDLINE | ID: mdl-38760717

ABSTRACT

BACKGROUND: Triple negative breast cancer (TNBC) is a heterogeneous and aggressive disease characterized by a high risk of mortality and poor prognosis. It has been reported that Laminin γ2 (LAMC2) is highly expressed in a variety of tumors, and its high expression is correlated with cancer development and progression. However, the function and mechanism by which LAMC2 influences TNBC remain unclear. METHODS: Kaplan-Meier survival analysis and Immunohistochemical (IHC) staining were used to examine the expression level of LAMC2 in TNBC. Subsequently, cell viability assay, wound healing and transwell assay were performed to detect the function of LAMC2 in cell proliferation and migration. A xenograft mouse model was used to assess tumorigenic function of LAMC2 in vivo. Luciferase reporter assay and western blot were performed to unravel the underlying mechanism. RESULTS: In this study, we found that higher expression of LAMC2 significantly correlated with poor survival in the TNBC cohort. Functional characterization showed that LAMC2 promoted cell proliferation and migration capacity of TNBC cell lines via up-regulating CD44. Moreover, LAMC2 exerted oncogenic roles in TNBC through modulating the expression of epithelial-mesenchymal transition (EMT) markers. Luciferase reporter assay verified that LAMC2 targeted ZEB1 to promote its transcription. Interestingly, LAMC2 regulated cell migration in TNBC via STAT3 signaling pathway. CONCLUSION: LAMC2 targeted ZEB1 via activating CD44/STAT3 signaling pathway to promote TNBC proliferation and migration, suggesting that LAMC2 could be a potential therapeutic target in TNBC patients.


Subject(s)
Cell Proliferation , Gene Expression Regulation, Neoplastic , Hyaluronan Receptors , Laminin , STAT3 Transcription Factor , Signal Transduction , Triple Negative Breast Neoplasms , Zinc Finger E-box-Binding Homeobox 1 , Humans , STAT3 Transcription Factor/metabolism , STAT3 Transcription Factor/genetics , Animals , Triple Negative Breast Neoplasms/genetics , Triple Negative Breast Neoplasms/metabolism , Triple Negative Breast Neoplasms/pathology , Triple Negative Breast Neoplasms/mortality , Cell Line, Tumor , Female , Hyaluronan Receptors/metabolism , Hyaluronan Receptors/genetics , Zinc Finger E-box-Binding Homeobox 1/metabolism , Zinc Finger E-box-Binding Homeobox 1/genetics , Laminin/metabolism , Laminin/genetics , Mice , Epithelial-Mesenchymal Transition/genetics , Cell Movement/genetics , Middle Aged , Biomarkers, Tumor/metabolism , Biomarkers, Tumor/genetics
19.
Carbohydr Polym ; 336: 122129, 2024 Jul 15.
Article in English | MEDLINE | ID: mdl-38670770

ABSTRACT

Hyaluronan, a linear glycosaminoglycan comprising D-N-acetylglucosamine and D-glucuronic acid, is the main component of the extracellular matrix. Its influence on cell proliferation, migration, inflammation, signalling, and other functions, depends heavily on its molecular weight and chemical modification. Unsaturated HA oligosaccharides are available in defined length and purity. Their potential therapeutic utility can be further improved by chemical modification, e. g., reduction. No synthesis of such modified oligosaccharides, either stepwise or by hyaluronan cleavage, has been reported yet. Here we show a three-step synthesis (esterification, depolymerization and reduction) of unsaturated even numbered hyaluronan oligosaccharides with carboxylates and the reducing terminus reduced to an alcohol. Particular oligosaccharides were synthesised. The modified oligosaccharides are not cleaved by mammalian or bacterial hyaluronidase and do not affect the growth of mouse and human fibroblasts. Further, MTT and NRU viability tests showed that they inhibit the growth of human colon carcinoma cells HT-29 by 20-50 % in concentrations 500-1000 µg/mL. Interestingly, this effect takes place regardless of CD44 receptor expression and was not observed with unmodified HA oligosaccharides. These compounds could serve as enzymatically stable building blocks for biologically active substances.


Subject(s)
Cell Proliferation , Cytostatic Agents , Hyaluronic Acid , Hyaluronoglucosaminidase , Oligosaccharides , Hyaluronic Acid/chemistry , Hyaluronic Acid/pharmacology , Humans , Oligosaccharides/chemistry , Oligosaccharides/pharmacology , Animals , Mice , Cell Proliferation/drug effects , Hyaluronoglucosaminidase/metabolism , Hyaluronoglucosaminidase/antagonists & inhibitors , Cytostatic Agents/pharmacology , Cytostatic Agents/chemistry , Cytostatic Agents/chemical synthesis , HT29 Cells , Hyaluronan Receptors/metabolism , Fibroblasts/drug effects
20.
Biomolecules ; 14(4)2024 Apr 10.
Article in English | MEDLINE | ID: mdl-38672482

ABSTRACT

Hyaluronic acid (HA), a major glycosaminoglycan of the brain extracellular matrix, modulates cell behaviors through binding its receptor, Cd44. In this study, we assessed the influence of HA on high-grade brain tumors in vitro. The model comprised cell cultures derived from six rodent carcinogen-induced brain tumors, forming 3D spheroids prone to spontaneous fusion. Supplementation of the standard culture medium with 0.25% HA significantly inhibited the fusion rates, preserving the shape and size uniformity of spheroids. The 3D cultures were assigned to two groups; a Cd44lo group had a tenfold decreased relative expression of Cd44 than another (Cd44hi) group. In addition, these two groups differed by expression levels of Sox2 transcription factor; the correlation analysis revealed a tight negative association for Cd44 and Sox2. Transcriptomic responses of spheroids to HA exposure also depended on Cd44 expression levels, from subtle in Cd44lo to more pronounced and specific in Cd44hi, involving cell cycle progression, PI3K/AKT/mTOR pathway activation, and multidrug resistance genes. The potential HA-induced increase in brain tumor 3D models' resistance to anticancer drug therapy should be taken into account when designing preclinical studies using HA scaffold-based models. The property of HA to prevent the fusion of brain-derived spheroids can be employed in CNS regenerative medicine and experimental oncology to ensure the production of uniform, controllably fusing neurospheres when creating more accurate in vitro brain models.


Subject(s)
Brain Neoplasms , Hyaluronan Receptors , Hyaluronic Acid , SOXB1 Transcription Factors , Spheroids, Cellular , Hyaluronic Acid/pharmacology , Spheroids, Cellular/drug effects , Spheroids, Cellular/metabolism , Brain Neoplasms/metabolism , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Animals , Hyaluronan Receptors/metabolism , Hyaluronan Receptors/genetics , SOXB1 Transcription Factors/metabolism , SOXB1 Transcription Factors/genetics , Rats , Transcriptome/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Tumor Cells, Cultured , Cell Fusion
SELECTION OF CITATIONS
SEARCH DETAIL
...