Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.892
Filter
1.
Sci Adv ; 10(35): eadn9857, 2024 Aug 30.
Article in English | MEDLINE | ID: mdl-39213364

ABSTRACT

Solid tumors, such as triple-negative breast cancer (TNBC), are biologically complex due to cellular heterogeneity, lack of tumor-specific antigens, and an immunosuppressive tumor microenvironment (TME). These challenges restrain chimeric antigen receptor (CAR) T cell efficacy, underlining the importance of armoring. In solid cancers, a localized tumor mass allows alternative administration routes, such as intratumoral delivery with the potential to improve efficacy and safety but may compromise metastatic-site treatment. Using a multi-layered CAR T cell engineering strategy that allowed a synergy between attributes, we show enhanced cytotoxic activity of MUC1 CAR T cells armored with PD1KO, tumor-specific interleukin-12 release, and TGFBR2KO attributes catered towards the TNBC TME. Intratumoral treatment effectively reduced distant tumors, suggesting retention of antigen-recognition benefits at metastatic sites. Overall, we provide preclinical evidence of armored non-alloreactive MUC1 CAR T cells greatly reducing high TNBC tumor burden in a TGFB1- and PD-L1-rich TME both at local and distant sites while preserving safety.


Subject(s)
Immunotherapy, Adoptive , Mucin-1 , Receptors, Chimeric Antigen , Triple Negative Breast Neoplasms , Tumor Microenvironment , Mucin-1/metabolism , Mucin-1/genetics , Mucin-1/immunology , Triple Negative Breast Neoplasms/therapy , Triple Negative Breast Neoplasms/pathology , Triple Negative Breast Neoplasms/immunology , Triple Negative Breast Neoplasms/metabolism , Humans , Female , Animals , Receptors, Chimeric Antigen/immunology , Receptors, Chimeric Antigen/metabolism , Receptors, Chimeric Antigen/genetics , Mice , Immunotherapy, Adoptive/methods , Cell Line, Tumor , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Xenograft Model Antitumor Assays
2.
Cancer Immunol Immunother ; 73(10): 203, 2024 Aug 06.
Article in English | MEDLINE | ID: mdl-39105847

ABSTRACT

BACKGROUND: Chimeric antigen receptor (CAR)-T cells have been used to treat blood cancers by producing a wide variety of cytokines. However, they are not effective in treating solid cancers and can cause severe side-effects, including cytokine release syndrome. TNFα is a tumoricidal cytokine, but it markedly increases the protein levels of cIAP1 and cIAP2, the members of inhibitor of apoptosis protein (IAP) family of E3 ubiquitin ligase that limits caspase-induced apoptosis. Degradation of IAP proteins by an IAP antagonist does not effectively kill cancer cells but enables TNFα to strongly induce cancer cell apoptosis. It would be a promising approach to treat cancers by targeted delivery of TNFα through an inactive adoptive cell in combination with an IAP antagonist. METHODS: Human dendritic cells (DCs) were engineered to express a single tumoricidal factor, TNFα, and a membrane-anchored Mucin1 antibody scFv, named Mucin 1 directed DCs expressing TNFα (M-DCsTNF). The efficacy of M-DCsTNF in recognizing and treating breast cancer was tested in vitro and in vivo. RESULTS: Mucin1 was highly expressed on the surface of a wide range of human breast cancer cell lines. M-DCsTNF directly associated with MDA-MB-231 cells in the bone of NSG mice. M-DCsTNF plus an IAP antagonist, SM-164, but neither alone, markedly induce MDA-MB-231 breast cancer cell apoptosis, which was blocked by TNF antibody. Importantly, M-DCsTNF combined with SM-164, but not SM-164 alone, inhibited the growth of patient-derived breast cancer in NSG mice. CONCLUSION: An adoptive cell targeting delivery of TNFα combined with an IAP antagonist is a novel effective approach to treat breast cancer and could be expanded to treat other solid cancers. Unlike CAR-T cell, this novel adoptive cell is not activated to produce a wide variety of cytokines, except for additional overexpressed TNF, and thus could avoid the severe side effects such as cytokine release syndrome.


Subject(s)
Dendritic Cells , Receptors, Chimeric Antigen , Tumor Necrosis Factor-alpha , Humans , Animals , Mice , Dendritic Cells/immunology , Dendritic Cells/metabolism , Female , Receptors, Chimeric Antigen/immunology , Tumor Necrosis Factor-alpha/metabolism , Mucin-1/immunology , Mucin-1/metabolism , Xenograft Model Antitumor Assays , Cell Line, Tumor , Inhibitor of Apoptosis Proteins/antagonists & inhibitors , Inhibitor of Apoptosis Proteins/metabolism , Immunotherapy, Adoptive/methods , Apoptosis , Breast Neoplasms/therapy , Breast Neoplasms/immunology , Immunotherapy/methods , Neoplasms/therapy , Neoplasms/immunology , Mice, SCID
3.
Cancer Med ; 13(15): e70079, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39118454

ABSTRACT

BACKGROUND: Cancer remains a formidable global health challenge, currently affecting nearly 20 million individuals worldwide. Due to the absence of universally effective treatments, ongoing research explores diverse strategies to combat this disease. Recent efforts have concentrated on developing combined drug regimens and targeted therapeutic approaches. OBJECTIVE: This study aimed to investigate the anticancer efficacy of a conjugated drug system, consisting of doxorubicin and cisplatin (Dox-Cis), encapsulated within niosomes and modified with MUC-1 aptamers to enhance biocompatibility and target specific cancer cells. METHODS: The chemical structure of the Dox-Cis conjugate was characterized using Fourier Transform Infrared Spectroscopy (FTIR) and Liquid Chromatography Quadrupole Time-of-Flight Mass Spectrometry (LC-Q-TOF/MS). The zeta potential and morphological parameters of the niosomal vesicles were determined through Dynamic Light Scattering (DLS) and Transmission Electron Microscopy (TEM). In vitro assessments of cell viability and apoptosis were conducted on MUC-1 positive HeLa cells and MUC-1 negative U87 cells. RESULTS: The findings confirmed the successful conjugation of Dox and Cis within the niosomes. The Nio/Dox-Cis/MUC-1 formulation demonstrated enhanced efficacy compared to the individual drugs and their unencapsulated combination in both cell lines. Notably, the Nio/Dox-Cis/MUC-1 formulation exhibited greater effectiveness on HeLa cells (38.503 ± 1.407) than on U87 cells (46.653 ± 1.297). CONCLUSION: The study underscores the potential of the Dox-Cis conjugate as a promising strategy for cancer treatment, particularly through platforms that facilitate targeted drug delivery to cancer cells. This targeted approach could lead to more effective and personalized cancer therapies.


Subject(s)
Aptamers, Nucleotide , Cell Survival , Cisplatin , Doxorubicin , Liposomes , Mucin-1 , Humans , Doxorubicin/pharmacology , Doxorubicin/chemistry , Mucin-1/metabolism , Mucin-1/chemistry , Liposomes/chemistry , Cisplatin/pharmacology , Cisplatin/chemistry , Aptamers, Nucleotide/chemistry , Aptamers, Nucleotide/pharmacology , Cell Survival/drug effects , Apoptosis/drug effects , Cell Line, Tumor , HeLa Cells , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Drug Compounding/methods
4.
Biochem Biophys Res Commun ; 731: 150402, 2024 Oct 30.
Article in English | MEDLINE | ID: mdl-39024979

ABSTRACT

Given that the corneal epithelium is situated on the outermost part of the eye, its functions can be influenced by external temperatures and chemical substances. This study aimed to elucidate the expression profile of chemosensory receptors in corneal epithelial cells and analyze their role in eye function regulation. A comprehensive analysis of 425 chemosensory receptors in human corneal epithelial cells-transformed (HCE-T) revealed the functional expression of TRPV4. The activation of TRPV4 in HCE-T cells significantly increased the expression of membrane-associated mucins MUC1, MUC4, and MUC16, which are crucial for stabilizing tear films, with efficacy comparable to the active components of dry eye medications. The present study suggests that TRPV4, which is activated by body temperature, regulates mucin expression and proposes it as a novel target for dry eye treatment.


Subject(s)
Epithelium, Corneal , Mucin-1 , Mucin-4 , Mucins , TRPV Cation Channels , Humans , CA-125 Antigen/metabolism , CA-125 Antigen/genetics , Epithelial Cells/metabolism , Epithelial Cells/cytology , Epithelium, Corneal/metabolism , Epithelium, Corneal/cytology , Membrane Proteins/metabolism , Membrane Proteins/genetics , Mucin-1/metabolism , Mucin-1/genetics , Mucin-4/metabolism , Mucin-4/genetics , Mucins/metabolism , Mucins/biosynthesis , TRPV Cation Channels/metabolism , TRPV Cation Channels/genetics
5.
Dermatol Online J ; 30(2)2024 Apr 15.
Article in English | MEDLINE | ID: mdl-38959917

ABSTRACT

Squamoid eccrine ductal carcinoma (SEDC) is a cutaneous adnexal malignancy that is histologically challenging to distinguish from squamous cell carcinoma. We report three cases of this rare entity and review the present literature regarding clinical, histological, and immunohistochemical features. Patients presented with a single nodule or plaque lesion on their back and temple. The shave biopsies for Patient A and C were interpreted as SEDC. Patient B's initial shave biopsy was interpreted as probable surface of squamous cell carcinoma, and subsequent excision revealed SEDC. Ductal differentiation was confirmed by positive expression of epithelial membrane antigen and carcinoembryonic antigen immunostains in all three patients. Review of the 67 previously reported cases emphasizes the importance of diagnosing SEDC accurately and promptly given its potential for distant metastasis and mortality. Perineural or lymphatic invasion is associated with higher rate of recurrence or metastasis. There should be high pathologic suspicion for SEDC in an elderly patient presenting with a palpable lesion, even if located outside of the head and neck area, particularly when there is suggestion of ductal differentiation in a sample of a squamous neoplasm.


Subject(s)
Carcinoma, Squamous Cell , Eccrine Glands , Sweat Gland Neoplasms , Humans , Carcinoembryonic Antigen/analysis , Carcinoembryonic Antigen/metabolism , Carcinoma, Ductal/pathology , Carcinoma, Squamous Cell/pathology , Carcinoma, Squamous Cell/diagnosis , Diagnosis, Differential , Eccrine Glands/pathology , Immunohistochemistry , Mucin-1/analysis , Mucin-1/metabolism , Skin Neoplasms/pathology , Skin Neoplasms/diagnosis , Sweat Gland Neoplasms/pathology , Aged
6.
J Virol ; 98(7): e0049924, 2024 Jul 23.
Article in English | MEDLINE | ID: mdl-38953631

ABSTRACT

Tibroviruses are novel rhabdoviruses detected in humans, cattle, and arthropods. Four tibroviruses are known to infect humans: Bas-Congo virus (BASV), Ekpoma virus 1 (EKV-1), Ekpoma virus 2, and Mundri virus. However, since none of them has been isolated, their biological properties are largely unknown. We aimed to characterize the human tibrovirus glycoprotein (G), which likely plays a pivotal role in viral tropism and pathogenicity. Human tibrovirus Gs were found to share some primary structures and display 14 conserved cysteine residues, although their overall amino acid homology was low (29%-48%). Multiple potential glycosylation sites were found on the G molecules, and endoglycosidase H- and peptide-N-glycosidase F-sensitive glycosylation was confirmed. AlphaFold-predicted three-dimensional (3D) structures of human tibrovirus Gs were overall similar. Membrane fusion mediated by these tibrovirus Gs was induced by acidic pH. The low pH-induced conformational change that triggers fusion was reversible. Virus-like particles (VLPs) were produced by transient expression of Gs in cultured cells and used to produce mouse antisera. Using vesicular stomatitis Indiana virus pseudotyped with Gs, we found that the antisera to the respective tibrovirus VLPs showed limited cross-neutralizing activity. It was also found that human C-type lectins and T-cell immunoglobulin mucin 1 acted as attachment factors for G-mediated entry into cells. Interestingly, BASV-G showed the highest ability to utilize these molecules. The viruses infected a wide range of cell lines with preferential tropism for human-derived cells whereas the preference of EKV-1 was unique compared with the other human tibroviruses. These findings provide fundamental information to understand the biological properties of the human tibroviruses. IMPORTANCE: Human tibroviruses are poorly characterized emerging rhabdoviruses associated with either asymptomatic infection or severe disease with a case fatality rate as high as 60% in humans. However, the extent and burden of human infection as well as factors behind differences in infection outcomes are largely unknown. In this study, we characterized human tibrovirus glycoproteins, which play a key role in virus-host interactions, mainly focusing on their structural and antigenic differences and cellular tropism. Our results provide critical information for understanding the biological properties of these novel viruses and for developing appropriate preparedness interventions such as diagnostic tools, vaccines, and effective therapies.


Subject(s)
Viral Envelope Proteins , Humans , Animals , Viral Envelope Proteins/metabolism , Viral Envelope Proteins/genetics , Mice , Glycosylation , Virus Internalization , Viral Tropism , Cell Line , Mucin-1/metabolism , HEK293 Cells , Antibodies, Viral/immunology , Amino Acid Sequence
7.
Anal Chem ; 96(31): 12767-12775, 2024 08 06.
Article in English | MEDLINE | ID: mdl-39044392

ABSTRACT

Bacteria inherently possess the capability of quorum sensing in response to the environment. In this work, we have proposed a strategy to confer bacteria with the ability to recognize targets with quorum-sensing behavior. Meanwhile, we have successfully achieved artificial control over the target-triggered aggregation of Escherichia coli (E. coli) by modifying the bacteria surface in a new way. Furthermore, by making use of green fluorescent protein (GFP) expressed by E. coli as the output signal, the aggregation of modified E. coli can be observed with the naked eye. Therefore, via the detection of the target, MUC1, an ovarian cancer biomarker, a simple and conveniently operated method to diagnose ovarian cancer is developed in this work. Experimental results show that the developed low-background and enzyme-free amplification method enables the highly sensitive detection of MUC1, achieving a remarkable limit of detection (LOD) of 5.47 fM and a linear detection range spanning from 1 pM to 50 nM and 50 nM to 100 nM, respectively. Clinical samples from healthy donors and patients can give distant assay results, showing great potential for clinical applications of this method.


Subject(s)
Escherichia coli , Mucin-1 , Ovarian Neoplasms , Ovarian Neoplasms/diagnosis , Escherichia coli/isolation & purification , Humans , Female , Mucin-1/analysis , Mucin-1/metabolism , Green Fluorescent Proteins/metabolism , Green Fluorescent Proteins/genetics , Limit of Detection , Biomarkers, Tumor/analysis , Biomarkers, Tumor/metabolism , Quorum Sensing , Biosensing Techniques/methods
8.
Genes (Basel) ; 15(6)2024 Jun 19.
Article in English | MEDLINE | ID: mdl-38927743

ABSTRACT

Ionizing radiation (IR) and chemotherapy with DNA-damaging drugs such as cisplatin are vital cancer treatment options. These treatments induce double-strand breaks (DSBs) as cytotoxic DNA damage; thus, the DSB repair activity in each cancer cell significantly influences the efficacy of the treatments. Pancreatic cancers are known to be resistant to these treatments, and the overexpression of MUC1, a member of the glycoprotein mucins, is associated with IR- and chemo-resistance. Therefore, we investigated the impact of MUC1 on DSB repair. This report examined the effect of the overexpression of MUC1 on homologous recombination (HR) and non-homologous end-joining (NHEJ) using cell-based DSB repair assays. In addition, the therapeutic potential of NHEJ inhibitors including HDAC inhibitors was also studied using pancreatic cancer cell lines. The MUC1-overexpression enhances NHEJ, while partially suppressing HR. Also, MUC1-overexpressed cancer cell lines are preferentially killed by a DNA-PK inhibitor and HDAC1/2 inhibitors. Altogether, MUC1 induces metabolic changes that create an imbalance between NHEJ and HR activities, and this imbalance can be a target for selective killing by HDAC inhibitors. This is a novel mechanism of MUC1-mediated IR-resistance and will form the basis for targeting MUC1-overexpressed pancreatic cancer.


Subject(s)
DNA Breaks, Double-Stranded , DNA End-Joining Repair , Mucin-1 , Pancreatic Neoplasms , Up-Regulation , Humans , Mucin-1/genetics , Mucin-1/metabolism , DNA End-Joining Repair/genetics , Cell Line, Tumor , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Pancreatic Neoplasms/drug therapy , Homologous Recombination , Histone Deacetylase Inhibitors/pharmacology , Gene Expression Regulation, Neoplastic/drug effects
9.
J Control Release ; 372: 417-432, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38908758

ABSTRACT

This study introduces the nanobromhexine lipid particle (NBL) platform designed for effective pulmonary drug delivery. Inspired by respiratory virus transport mechanisms, NBL address challenges associated with mucus permeation and inflammation in pulmonary diseases. Composed of low molecular weight polyethylene glycol-coated lipid nanoparticles with bromhexine hydrochloride, NBL exhibit a size of 118 ± 24 nm, a neutral zeta potential, osmolarity of 358 ± 28 mOsmol/kg, and a pH of 6.5. Nebulizing without leakage and showing no toxicity to epithelial cells, NBL display mucoadhesive properties with a 60% mucin-binding efficiency. They effectively traverse the dense mucus layer of Calu-3 cultures in an air-liquid interface, as supported by a 55% decrease in MUC5AC density and a 29% increase in nanoparticles internalization compared to non-exposed cells. In assessing immunomodulatory effects, NBL treatment in SARS-CoV-2-infected lung cells leads to a 40-fold increase in anti-inflammatory MUC1 gene expression, a proportional reduction in pro-inflammatory IL-6 expression, and elevated anti-inflammatory IL-10 expression. These findings suggest a potential mechanism to regulate the excessive IL-6 expression triggered by virus infection. Therefore, the NBL platform demonstrates promising potential for efficient pulmonary drug delivery and immunomodulation, offering a novel approach to addressing mucus permeation and inflammation in pulmonary diseases.


Subject(s)
Lung , Nanoparticles , SARS-CoV-2 , Nanoparticles/administration & dosage , Humans , Lung/metabolism , SARS-CoV-2/drug effects , Drug Delivery Systems , Immunomodulation , Cell Line , Mucin-1/metabolism , COVID-19 , Lipids/chemistry , Lipids/administration & dosage , Mucus/metabolism , Polyethylene Glycols/chemistry , Epithelial Cells/metabolism , Epithelial Cells/drug effects , COVID-19 Drug Treatment , Mucin 5AC/metabolism , Liposomes
10.
Analyst ; 149(15): 3920-3927, 2024 Jul 22.
Article in English | MEDLINE | ID: mdl-38912896

ABSTRACT

Anti-cancer therapy is crucial in cancer prevention and anti-cancer, and thus, highly sensitive methods for detecting cancer biomarkers are essential for cancer early diagnosis. Herein, an electrochemical aptamer biosensor based on the CRISPR-Cas12a system was constructed for the detection of cancer tumor biomarker MUC1 mucin. The sensitivity was significantly prompted by enzyme-catalyzed signal amplification, and the selectivity was improved by the dual recognition of the aptamer to MUC1 and crRNA-Cas12a system to the aptamer. Glucose oxidase (GOD) was loaded on the surface of magnetic Fe3O4@Au (MGNP) via probe single-stranded DNA (pDNA) with the terminal modification of mercapto (-SH) to form GOD-pDNA/MGNP. The corresponding aptamer of MUC1 (MUC1 Apt) binds to its complementary ssDNA (cDNA) to form the activator Apt/cDNA, which is specifically recognized by crRNA-Cas12a and excites the trans-cleavage function of Cas12a, thus in turn trans-cleaves pDNA and detaches GOD from the magnetic particles. The magnetic beads were separated and transferred into a glucose solution, and the oxidation current of H2O2 produced by the catalytic reaction of GOD was measured on a Pt-modified magnetically-controlled glassy carbon electrode, resulting in an indirect determination of MUC1. The current change was linear with the logarithm of MUC1 concentration in the range from 1.0 × 10-17 g mL-1 to 1.0 × 10-10 g mL-1. The detection limit was as low as 7.01 × 10-18 g mL-1. The method was applied for the detection of MUC1 in medical samples.


Subject(s)
Aptamers, Nucleotide , Biomarkers, Tumor , Biosensing Techniques , Electrochemical Techniques , Glucose Oxidase , Mucin-1 , Humans , Aptamers, Nucleotide/chemistry , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Biomarkers, Tumor/genetics , Biosensing Techniques/methods , CRISPR-Associated Proteins/chemistry , CRISPR-Associated Proteins/metabolism , CRISPR-Cas Systems , DNA, Single-Stranded/chemistry , Electrochemical Techniques/methods , Endodeoxyribonucleases , Enzymes, Immobilized/chemistry , Glucose Oxidase/chemistry , Glucose Oxidase/metabolism , Gold/chemistry , Hydrogen Peroxide/chemistry , Limit of Detection , Magnetite Nanoparticles/chemistry , Mucin-1/metabolism
11.
Anticancer Res ; 44(6): 2689-2698, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38821605

ABSTRACT

BACKGROUND/AIM: There are two main subtypes of mucinous carcinoma (MC) based on the quantification of the mucinous component: the pure variant (pMC) and the mixed variant (mMC). pMC has been subdivided into pure A with a hypocellular variant, and pure B with a hypercellular variant. PATIENTS AND METHODS: We retrospectively analyzed the clinicopathological features of 99 patients with MC who were treated at our institution from January 2002 to December 2014. We evaluated the expression profiles of markers, including mucin (MUC) family members, in the patients groups representing different MC subtypes by performing immunohistochemistry to identify factors involved in the differentiation and progression of MCs. RESULTS: Among the 99 patients, 76 (76.8%) had pure mucinous carcinomas (pMC) and the other 23 (23.2%) had mixed mucinous carcinomas (mMC). Of the pMCs, 54 were pure A and 22 were pure B. The prognosis was worse for pure B than pure A and worse for mMC than pMC. Although there was no significant difference in clinicopathological factors between the pure A and pure B groups, immunohistochemical staining revealed differences in the localization of mucin MUC1 and ß-catenin. A comparison of the pMC and mMC cases revealed more lymphovascular invasion in mMC and differences in the localization of ß-catenin between the two groups. CONCLUSION: The patients' prognoses were significantly poorer depending on the histologic subtype (in the order pure A, pure B, and mixed). MUC1 localization and ß-catenin were revealed as independent predictors contributing to the poorer prognosis.


Subject(s)
Adenocarcinoma, Mucinous , Biomarkers, Tumor , Breast Neoplasms , Mucin-1 , beta Catenin , Humans , Mucin-1/metabolism , Female , beta Catenin/metabolism , Adenocarcinoma, Mucinous/metabolism , Adenocarcinoma, Mucinous/pathology , Middle Aged , Aged , Breast Neoplasms/pathology , Breast Neoplasms/metabolism , Retrospective Studies , Biomarkers, Tumor/metabolism , Prognosis , Adult , Immunohistochemistry , Aged, 80 and over
12.
Int J Mol Med ; 54(1)2024 Jul.
Article in English | MEDLINE | ID: mdl-38785138

ABSTRACT

Ovarian cancer (OC) is one of the most common gynecological malignancies. Currently, chemoradiotherapy is the primary clinical treatment approach for OC; however, it has severe side effects and a high rate of recurrence. Thus, there is an urgent need to develop innovative therapeutic options. Paeoniflorigenone (PFG) is a monoterpene compound isolated from the traditional Chinese medicine Paeoniae Radix Rubra. PFG can inhibit the proliferation of tumor cells; however, its anticancer activity against OC has yet to be elucidated. Mucin 1 (MUC1) is highly expressed in various malignant tumors, and is associated with tumor proliferation, metastasis and epithelial­mesenchymal transition (EMT). In addition, MUC1 affects numerous signaling pathways in tumor cells. In order to develop a possible treatment approach for metastatic OC, the antitumor activity of PFG in OC cells was investigated using Cell Counting Kit­8 assay, Edu assay, flow cytometry, Transwell assay and western blot analysis. In addition, it was assessed how PFG affects MUC1 expression and function. The experiments revealed that PFG significantly inhibited OC cell proliferation, migration, invasion and EMT. PFG also induced S­phase cell cycle arrest in OC cells. Furthermore, PFG inhibited MUC1 promoter activity, which led to a decrease in MUC1 protein expression. By contrast, MUC1 promoted OC progression, including cell proliferation, cell cycle progression and cell migration. Stable knockdown of MUC1 in OC cells improved the ability of PFG to block the Wnt/ß­catenin pathway, and to limit tumor cell invasion and migration, whereas MUC1 overexpression partially counteracted the antitumor effects of PFG. In conclusion, the present study demonstrated that PFG may inhibit the MUC1/Wnt/ß­catenin pathway to induce anti­metastatic, anti­invasive and anti­EMT effects on OC. Notably, MUC1 may be a direct target of PFG. Thus, PFG holds promise as a specific antitumor agent for the treatment of OC.


Subject(s)
Cell Movement , Cell Proliferation , Epithelial-Mesenchymal Transition , Mucin-1 , Ovarian Neoplasms , Wnt Signaling Pathway , Female , Humans , Wnt Signaling Pathway/drug effects , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , Ovarian Neoplasms/drug therapy , Mucin-1/metabolism , Mucin-1/genetics , Cell Line, Tumor , Epithelial-Mesenchymal Transition/drug effects , Cell Proliferation/drug effects , Cell Movement/drug effects , Monoterpenes/pharmacology , Neoplasm Metastasis , Gene Expression Regulation, Neoplastic/drug effects
13.
Cell Death Dis ; 15(5): 330, 2024 May 13.
Article in English | MEDLINE | ID: mdl-38740827

ABSTRACT

The long non-coding RNA X-inactive specific transcript (lncRNA XIST) and MUC1 gene are dysregulated in chronic inflammation and cancer; however, there is no known interaction of their functions. The present studies demonstrate that MUC1-C regulates XIST lncRNA levels by suppressing the RBM15/B, WTAP and METTL3/14 components of the m6A methylation complex that associate with XIST A repeats. MUC1-C also suppresses the YTHDF2-CNOT1 deadenylase complex that recognizes m6A sites and contributes to XIST decay with increases in XIST stability and expression. In support of an auto-regulatory pathway, we show that XIST regulates MUC1-C expression by promoting NF-κB-mediated activation of the MUC1 gene. Of significance, MUC1-C and XIST regulate common genes associated with inflammation and stemness, including (i) miR-21 which is upregulated across pan-cancers, and (ii) TDP-43 which associates with the XIST E repeats. Our results further demonstrate that the MUC1-C/XIST pathway (i) is regulated by TDP-43, (ii) drives stemness-associated genes, and (iii) is necessary for self-renewal capacity. These findings indicate that the MUC1-C/XIST auto-regulatory axis is of importance in cancer progression.


Subject(s)
Gene Expression Regulation, Neoplastic , Mucin-1 , RNA, Long Noncoding , Animals , Humans , Mice , Cell Line, Tumor , Disease Progression , DNA-Binding Proteins/metabolism , DNA-Binding Proteins/genetics , MicroRNAs/metabolism , MicroRNAs/genetics , Mucin-1/metabolism , Mucin-1/genetics , Neoplasms/genetics , Neoplasms/metabolism , Neoplasms/pathology , NF-kappa B/metabolism , RNA, Long Noncoding/metabolism , RNA, Long Noncoding/genetics
14.
Proc Natl Acad Sci U S A ; 121(20): e2322688121, 2024 May 14.
Article in English | MEDLINE | ID: mdl-38709925

ABSTRACT

Brain metastatic breast cancer is particularly lethal largely due to therapeutic resistance. Almost half of the patients with metastatic HER2-positive breast cancer develop brain metastases, representing a major clinical challenge. We previously described that cancer-associated fibroblasts are an important source of resistance in primary tumors. Here, we report that breast cancer brain metastasis stromal cell interactions in 3D cocultures induce therapeutic resistance to HER2-targeting agents, particularly to the small molecule inhibitor of HER2/EGFR neratinib. We investigated the underlying mechanisms using a synthetic Notch reporter system enabling the sorting of cancer cells that directly interact with stromal cells. We identified mucins and bulky glycoprotein synthesis as top-up-regulated genes and pathways by comparing the gene expression and chromatin profiles of stroma-contact and no-contact cancer cells before and after neratinib treatment. Glycoprotein gene signatures were also enriched in human brain metastases compared to primary tumors. We confirmed increased glycocalyx surrounding cocultures by immunofluorescence and showed that mucinase treatment increased sensitivity to neratinib by enabling a more efficient inhibition of EGFR/HER2 signaling in cancer cells. Overexpression of truncated MUC1 lacking the intracellular domain as a model of increased glycocalyx-induced resistance to neratinib both in cell culture and in experimental brain metastases in immunodeficient mice. Our results highlight the importance of glycoproteins as a resistance mechanism to HER2-targeting therapies in breast cancer brain metastases.


Subject(s)
Brain Neoplasms , Breast Neoplasms , Drug Resistance, Neoplasm , Glycocalyx , Quinolines , Receptor, ErbB-2 , Stromal Cells , Humans , Breast Neoplasms/pathology , Breast Neoplasms/metabolism , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Female , Brain Neoplasms/secondary , Brain Neoplasms/metabolism , Brain Neoplasms/drug therapy , Brain Neoplasms/pathology , Brain Neoplasms/genetics , Receptor, ErbB-2/metabolism , Receptor, ErbB-2/genetics , Glycocalyx/metabolism , Animals , Cell Line, Tumor , Stromal Cells/metabolism , Stromal Cells/pathology , Quinolines/pharmacology , Mice , Cell Communication , Coculture Techniques , Mucin-1/metabolism , Mucin-1/genetics , Signal Transduction , ErbB Receptors/metabolism , ErbB Receptors/antagonists & inhibitors
15.
Biomaterials ; 309: 122608, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38744189

ABSTRACT

Necroptotic immunogenic cell death (ICD) can activate the human immune system to treat the metastasis and recurrence of triple-negative breast cancer (TNBC). However, developing the necroptotic inducer and precisely delivering it to the tumor site is the key issue. Herein, we reported that the combination of shikonin (SHK) and chitosan silver nanoparticles (Chi-Ag NPs) effectively induced ICD by triggering necroptosis in 4T1 cells. Moreover, to address the lack of selectivity of drugs for in vivo application, we developed an MUC1 aptamer-targeted nanocomplex (MUC1@Chi-Ag@CPB@SHK, abbreviated as MUC1@ACS) for co-delivering SHK and Chi-Ag NPs. The accumulation of MUC1@ACS NPs at the tumor site showed a 6.02-fold increase compared to the free drug. Subsequently, upon reaching the tumor site, the acid-responsive release of SHK and Chi-Ag NPs from MUC1@ACS NPs cooperatively induced necroptosis in tumor cells by upregulating the expression of RIPK3, p-RIPK3, and tetrameric MLKL, thereby effectively triggering ICD. The sequential maturation of dendritic cells (DCs) subsequently enhanced the infiltration of CD8+ and CD4+ T cells in tumors, while inhibiting regulatory T cells (Treg cells), resulting in the effective treatment of primary and distal tumor growth and the inhibition of TNBC metastasis. This work highlights the importance of nanoparticles in mediating drug interactions during necroptotic ICD.


Subject(s)
Chitosan , Metal Nanoparticles , Naphthoquinones , Necroptosis , Receptor-Interacting Protein Serine-Threonine Kinases , Silver , Triple Negative Breast Neoplasms , Naphthoquinones/pharmacology , Naphthoquinones/chemistry , Triple Negative Breast Neoplasms/pathology , Triple Negative Breast Neoplasms/drug therapy , Chitosan/chemistry , Silver/chemistry , Silver/pharmacology , Animals , Metal Nanoparticles/chemistry , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Cell Line, Tumor , Female , Necroptosis/drug effects , Humans , Mice , Immunogenic Cell Death/drug effects , Mice, Inbred BALB C , Mucin-1/metabolism , Drug Synergism , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Antineoplastic Agents/chemistry
16.
J Mater Chem B ; 12(22): 5551-5560, 2024 Jun 05.
Article in English | MEDLINE | ID: mdl-38747235

ABSTRACT

Timely identification of cancers is pivotal in optimizing treatment efficacy and reducing their widespread impact. This study introduces a novel biosensor for the sensitive electrochemical detection of cancer cells overexpressing mucin 1 (MUC1), a well-established model for breast cancer. The sensor substrate comprises gold columnar nanostructures obtained through glancing angle deposition (GLAD) of copper nanostructures, subsequently replaced by gold via a facile galvanic replacement process. Functionalizing these gold nanostructures with aptamers targeting the MUC1 glycoproteins, a prominent cancer biomarker, enables specific recognition of MCF-7 breast cancer cells. The proposed electrochemical sensing platform offers several advantages, including high selectivity, a wide linear range of detection, a low detection limit of 30 cells per mL, and long-term stability, rendering this sensor highly desirable for definitive breast cancer diagnosis.


Subject(s)
Biosensing Techniques , Breast Neoplasms , Electrochemical Techniques , Gold , Mucin-1 , Humans , Biosensing Techniques/methods , Gold/chemistry , MCF-7 Cells , Breast Neoplasms/diagnosis , Breast Neoplasms/pathology , Mucin-1/analysis , Mucin-1/metabolism , Female , Nanostructures/chemistry , Metal Nanoparticles/chemistry , Aptamers, Nucleotide/chemistry
17.
Anal Chem ; 96(22): 9218-9227, 2024 06 04.
Article in English | MEDLINE | ID: mdl-38781682

ABSTRACT

In situ monitoring of cell secretions and communications plays a fundamental role in screening of disease diagnostic biomarkers and drugs. Quantitative detection of cell secretions and monitoring of intercellular communication have been separately reported, which often rely on target labeling or complex pretreatment steps, inevitably causing damage to the target. Simultaneous in situ noninvasive detection of cell secretions and monitoring of intercellular communication are challenging and have never been reported. Herein, we smartly developed a portable device for in situ label-free monitoring of cell secretions and communications with fluorescence and ion-transport-based nanochannel electrochemistry. Based on the dual signal mode, a series of nonelectroactive secretions were sensitively and accurately quantified. The detection limits for VEGF, MUC1, and ATP were 3.84 pg/mL, 32.7 pg/mL, and 47.4 fM (3σ/S), which were 1/3.9, 1/1.1, and 1/41 of those of commercial ELISA kits, respectively. More interestingly, under the released secretions, the gradual opening of the nanochannel connected the two cells in the left and right chambers of the device; thus, the secretion mediated intercellular communication can be monitored. The proposed platform may provide a promising tool for understanding the mechanism of intercellular communication and discovering new therapeutic targets.


Subject(s)
Electrochemical Techniques , Humans , Electrochemical Techniques/instrumentation , Adenosine Triphosphate/analysis , Adenosine Triphosphate/metabolism , Mucin-1/analysis , Mucin-1/metabolism , Cell Communication , Vascular Endothelial Growth Factor A/analysis , Vascular Endothelial Growth Factor A/metabolism , Fluorescence , Limit of Detection
18.
Anal Chem ; 96(19): 7747-7755, 2024 05 14.
Article in English | MEDLINE | ID: mdl-38691774

ABSTRACT

Accurate classification of tumor cells is of importance for cancer diagnosis and further therapy. In this study, we develop multimolecular marker-activated transmembrane DNA computing systems (MTD). Employing the cell membrane as a native gate, the MTD system enables direct signal output following simple spatial events of "transmembrane" and "in-cell target encounter", bypassing the need of multistep signal conversion. The MTD system comprises two intelligent nanorobots capable of independently sensing three molecular markers (MUC1, EpCAM, and miR-21), resulting in comprehensive analysis. Our AND-AND logic-gated system (MTDAND-AND) demonstrates exceptional specificity, allowing targeted release of drug-DNA specifically in MCF-7 cells. Furthermore, the transformed OR-AND logic-gated system (MTDOR-AND) exhibits broader adaptability, facilitating the release of drug-DNA in three positive cancer cell lines (MCF-7, HeLa, and HepG2). Importantly, MTDAND-AND and MTDOR-AND, while possessing distinct personalized therapeutic potential, share the ability of outputting three imaging signals without any intermediate conversion steps. This feature ensures precise classification cross diverse cells (MCF-7, HeLa, HepG2, and MCF-10A), even in mixed populations. This study provides a straightforward yet effective solution to augment the versatility and precision of DNA computing systems, advancing their potential applications in biomedical diagnostic and therapeutic research.


Subject(s)
DNA , Epithelial Cell Adhesion Molecule , MicroRNAs , Humans , Epithelial Cell Adhesion Molecule/metabolism , DNA/chemistry , MicroRNAs/analysis , MicroRNAs/metabolism , Mucin-1/metabolism , Mucin-1/analysis , Computers, Molecular , MCF-7 Cells , Biomarkers, Tumor/metabolism , Biomarkers, Tumor/analysis , Cell Membrane/metabolism , Cell Membrane/chemistry , Hep G2 Cells
19.
Anal Chem ; 96(21): 8754-8762, 2024 05 28.
Article in English | MEDLINE | ID: mdl-38740024

ABSTRACT

Simultaneous profiling of redox-regulated markers at different cellular sublocations is of great significance for unraveling the upstream and downstream molecular mechanisms of oxidative stress in living cells. Herein, by synchronizing dual target-triggered DNA machineries in one nanoentity, we engineered a DNA walker-driven mass nanotag (MNT) assembly system (w-MNT-AS) that can be sequentially activated by oxidative stress-associated mucin 1 (MUC1) and apurinic/apyrimidinic endonuclease 1 (APE1) from plasma membrane to cytoplasm and induce recycled assembly of MNTs for multiplex detection of the two markers by matrix-assisted laser desorption ionization mass spectrometry (MALDI MS). In the working cascade, the sensing process governs the separate activation of w-MNT-AS by MUC1 and APE1 in diverse locations, while the assembly process contributes to the parallel amplification of the ion signal of the characteristic mass tags. In this manner, the differences between MCF-7, HeLa, HepG2, and L02 cells in membrane MUC1 expression and cytoplasmic APE1 activation were fully characterized. Furthermore, the oxidative stress level and dynamics caused by exogenous H2O2, doxorubicin, and simvastatin were comprehensively demonstrated by tracking the fate of the two markers across different cellular locations. The proposed w-MNT-AS coupled MS method provides an effective route to probe multiple functional molecules that lie at different locations while participating in the same cellular event, facilitating the mechanistic studies on cellular response to oxidative stress and other disease-related cellular processes.


Subject(s)
DNA-(Apurinic or Apyrimidinic Site) Lyase , DNA , Mucin-1 , Oxidative Stress , Humans , Mucin-1/metabolism , DNA/metabolism , DNA/chemistry , DNA-(Apurinic or Apyrimidinic Site) Lyase/metabolism , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Hydrogen Peroxide/metabolism
20.
Oncogene ; 43(28): 2199-2214, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38802648

ABSTRACT

The MUC1 gene evolved in mammals for adaptation of barrier tissues in response to infections and damage. Paraspeckles are nuclear bodies formed on the NEAT1 lncRNA in response to loss of homeostasis. There is no known intersection of MUC1 with NEAT1 or paraspeckles. Here, we demonstrate that the MUC1-C subunit plays an essential role in regulating NEAT1 expression. MUC1-C activates the NEAT1 gene with induction of the NEAT1_1 and NEAT1_2 isoforms by NF-κB- and MYC-mediated mechanisms. MUC1-C/MYC signaling also induces expression of the SFPQ, NONO and FUS RNA binding proteins (RBPs) that associate with NEAT1_2 and are necessary for paraspeckle formation. MUC1-C integrates activation of NEAT1 and RBP-encoding genes by recruiting the PBAF chromatin remodeling complex and increasing chromatin accessibility of their respective regulatory regions. We further demonstrate that MUC1-C and NEAT1 form an auto-inductive pathway that drives common sets of genes conferring responses to inflammation and loss of homeostasis. Of functional significance, we find that the MUC1-C/NEAT1 pathway is of importance for the cancer stem cell (CSC) state and anti-cancer drug resistance. These findings identify a previously unrecognized role for MUC1-C in the regulation of NEAT1, RBPs, and paraspeckles that has been co-opted in promoting cancer progression.


Subject(s)
Disease Progression , Gene Expression Regulation, Neoplastic , Mucin-1 , RNA, Long Noncoding , Humans , RNA, Long Noncoding/genetics , Mucin-1/genetics , Mucin-1/metabolism , Animals , Cell Line, Tumor , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Mice , Neoplasms/genetics , Neoplasms/pathology , Neoplasms/metabolism , NF-kappa B/metabolism , NF-kappa B/genetics , PTB-Associated Splicing Factor/genetics , PTB-Associated Splicing Factor/metabolism , Signal Transduction/genetics , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/metabolism , RNA-Binding Protein FUS/genetics , RNA-Binding Protein FUS/metabolism , DNA-Binding Proteins
SELECTION OF CITATIONS
SEARCH DETAIL