Displaying all 7 publications

Abstract:
Sort:
  1. Al-Hatamleh MAI, Ahmad S, Boer JC, Lim J, Chen X, Plebanski M, et al.
    J Oncol, 2019;2019:6313242.
    PMID: 31239840 DOI: 10.1155/2019/6313242
    In the past decade, nanomedicine research has provided us with highly useful agents (nanoparticles) delivering therapeutic drugs to target cancer cells. The present review highlights nanomedicine applications for breast cancer immunotherapy. Recent studies have suggested that tumour necrosis factor (TNF) and its receptor 2 (TNFR2) expressed on breast cancer cells have important functional consequences. This cytokine/receptor interaction is also critical for promoting highly immune-suppressive phenotypes by regulatory T cells (Tregs). This review generally provides a background for nanoparticles as potential drug delivery agents for immunomodulators and further discusses in depth the potential of TNF antagonists delivery to modulate TNF-TNFR2 interactions and inhibit breast cancer progression.
  2. Roslan Z, Muhamad M, Selvaratnam L, Ab-Rahim S
    J Oncol, 2019;2019:4536302.
    PMID: 31031810 DOI: 10.1155/2019/4536302
    Low-density lipoprotein receptor (LDLR) has been an object of research since the 1970s because of its role in various cell functions. The LDLR family members include LRP5, LRP6, and LRP8. Even though LRP5, 6, and 8 are in the same family, intriguingly, these three proteins have various roles in physiological events, as well as in regulating different mechanisms in various kinds of cancers. LRP5, LRP6, and LRP8 have been shown to play important roles in a broad panel of cancers. LRP5 is highly expressed in many tissues and is involved in the modulation of glucose-induced insulin secretion, bone development, and cholesterol metabolism, as well as cancer progression. Recently, LRP5 has also been shown to play a role in chondroblastic subtype of osteosarcoma (OS) and prostate cancer and also in noncancer case such as osteoporosis. LRP6, which has been previously discovered to share the same structures as LRP5, has also been associated with many cancer progressions such as human triple negative breast cancer (TNBC), primary chronic lymphocytic leukemia (CLL), nonsmall cell lung cancer (NSCL), lung squamous cell carcinoma (LSCC), and hepatocellular carcinoma (HCC). In addition to its role in cancer progression, LRP8 (apolipoprotein E receptor 2 [APOER2]) has also been demonstrated to regulate canonical Wnt/β-catenin signaling pathway whereby this pathway plays a role in cell migration and development. Therefore, this review aimed to elucidate the role of LRP 5, 6, and 8 in regulating the cancer progression.
  3. Abd-Aziz N, Kamaruzman NI, Poh CL
    J Oncol, 2020;2020:8029721.
    PMID: 32733559 DOI: 10.1155/2020/8029721
    MicroRNAs (miRNAs) are small noncoding RNAs that function at the posttranscriptional level in the cellular regulation process. miRNA expression exerts vital effects on cell growth such as cell proliferation and survival. In cancers, miRNAs have been shown to initiate carcinogenesis, where overexpression of oncogenic miRNAs (oncomiRs) or reduced expression of tumor suppressor miRNAs has been reported. In this review, we discuss the involvement of miRNAs in tumorigenesis, the role of synthetic miRNAs as either mimics or antagomirs to overcome cancer growth, miRNA delivery, and approaches to enhance their therapeutic potentials.
  4. Al-Koshab M, Alabsi AM, Mohd Bakri M, Ali-Saeed R, Selvi Naicker M
    J Oncol, 2020;2020:5490468.
    PMID: 32104177 DOI: 10.1155/2020/5490468
    Background: The aim of this study is to evaluate the chemopreventive and chemotherapeutic activities of Ficus deltoidea (FD) in an animal model induced for oral cancer using 4-nitroquinoline-1-oxide (4NQO).

    Methods: Male Sprague-Dawley (SD) rats were randomized into six groups (n = 7 per group): Group 1 (untreated group); Group 2 (control cancer group) received 4NQO only for 8 weeks in their drinking water; Groups 3 and 4 (chemopreventive) received 4NQO for 8 weeks and were simultaneously treated with FD extract at 250 and 500 mg/kg, respectively, by oral gavage; Groups 5 and 6 (chemotherapeutic) received 4NQO for 8 weeks followed by the administration of FD extract at 250 and 500 mg/kg, respectively, for another 10 weeks. The incidence of oral cancer was microscopically evaluated. Moreover, immunohistochemical expression was analysed in tongue specimens using an image analyser computer system, while the RT2 profiler PCR array method was employed for gene expression analysis.

    Results: The results of the present study showed a beneficial regression effect of the FD extract on tumor progression. The FD extract significantly reduced the incidence of oral squamous cell carcinoma (OSCC) from 100% to 14.3% in the high-dose groups. The immunohistochemical analysis showed that the FD extract had significantly decreased the expression of the key tumor marker cyclin D1 and had significantly increased the expression of the β-catenin and e-cadherin antibodies that are associated with enhanced cellular adhesion. Based on the gene expression analysis, FD extract had reduced the expression of the TWIST1 and RAC1 genes associated with epithelial-mesenchymal transition (EMT) and had significantly downregulated the COX-2 and EGFR genes associated with cancer angiogenesis, metastasis, and chemoresistance. Our data suggest that the FD extract exerts chemopreventive and chemotherapeutic activities in an animal model induced for oral cancer using 4NQO, thus having the potential to be developed as chemopreventive and chemotherapeutic agents.β-catenin and e-cadherin antibodies that are associated with enhanced cellular adhesion. Based on the gene expression analysis, FD extract had reduced the expression of the TWIST1 and RAC1 genes associated with epithelial-mesenchymal transition (EMT) and had significantly downregulated the COX-2 and EGFR genes associated with cancer angiogenesis, metastasis, and chemoresistance. Our data suggest that the FD extract exerts chemopreventive and chemotherapeutic activities in an animal model induced for oral cancer using 4NQO, thus having the potential to be developed as chemopreventive and chemotherapeutic agents.TWIST1 and RAC1 genes associated with epithelial-mesenchymal transition (EMT) and had significantly downregulated the COX-2 and EGFR genes associated with cancer angiogenesis, metastasis, and chemoresistance. Our data suggest that the FD extract exerts chemopreventive and chemotherapeutic activities in an animal model induced for oral cancer using 4NQO, thus having the potential to be developed as chemopreventive and chemotherapeutic agents.RAC1 genes associated with epithelial-mesenchymal transition (EMT) and had significantly downregulated the COX-2 and EGFR genes associated with cancer angiogenesis, metastasis, and chemoresistance. Our data suggest that the FD extract exerts chemopreventive and chemotherapeutic activities in an animal model induced for oral cancer using 4NQO, thus having the potential to be developed as chemopreventive and chemotherapeutic agents.COX-2 and EGFR genes associated with cancer angiogenesis, metastasis, and chemoresistance. Our data suggest that the FD extract exerts chemopreventive and chemotherapeutic activities in an animal model induced for oral cancer using 4NQO, thus having the potential to be developed as chemopreventive and chemotherapeutic agents.EGFR genes associated with cancer angiogenesis, metastasis, and chemoresistance. Our data suggest that the FD extract exerts chemopreventive and chemotherapeutic activities in an animal model induced for oral cancer using 4NQO, thus having the potential to be developed as chemopreventive and chemotherapeutic agents.

  5. Tang W, Liu H, Li X, Ooi TC, Rajab NF, Cao H, et al.
    J Oncol, 2023;2023:2611105.
    PMID: 36908705 DOI: 10.1155/2023/2611105
    BACKGROUND: Approximately 10% of cancer patients worldwide have colorectal cancer (CRC), a prevalent gastrointestinal malignancy with substantial mortality and morbidity. The purpose of this work was to investigate the APOC1 gene's expression patterns in the CRC tumor microenvironment and, using the findings from bioinformatics, to assess the biological function of APOC1 in the development of CRC.

    METHODS: The TCGA portal was employed in this investigation to find APOC1 expression in CRC. Its correlation with other genes and clinicopathological data was examined using the UALCAN database. To validate APOC1's cellular location, the Human Protein was employed. In order to forecast the relationship between APOC1 expression and prognosis in CRC patients, the Kaplan-Meier plotter database was used. TISIDB was also employed to evaluate the connection between immune responses and APOC1 expression in CRC. The interactions of APOC1 with other proteins were predicted using STRING. In order to understand the factors that contribute to liver metastasis from CRC, single-cell RNA sequencing (scRNA-seq) was done on one patient who had the disease. This procedure included sampling preoperative blood and the main colorectal cancer tissues, surrounding colorectal cancer normal tissues, liver metastatic cancer tissues, and normal liver tissues. Finally, an in vitro knockdown method was used to assess how APOC1 expression in tumor-associated macrophages (TAMs) affected CRC cancer cell growth and migration.

    RESULTS: When compared to paracancerous tissues, APOC1 expression was considerably higher in CRC tissues. The clinicopathological stage and the prognosis of CRC patients had a positive correlation with APOC1 upregulation and a negative correlation, respectively. APOC1 proteins are mostly found in cell cytosols where they may interact with APOE, RAB42, and TREM2. APOC1 was also discovered to have a substantial relationship with immunoinhibitors (CD274, IDO1, and IL10) and immunostimulators (PVR, CD86, and ICOS). According to the results of scRNA-seq, we found that TAMs of CRC tissues had considerably more APOC1 than other cell groups. The proliferation and migration of CRC cells were impeded in vitro by APOC1 knockdown in TAMs.

    CONCLUSION: Based on scRNA-seq research, the current study shows that APOC1 was overexpressed in TAMs from CRC tissues. By inhibiting APOC1 in TAMs, CRC progression was reduced in vitro, offering a new tactic and giving CRC patients fresh hope.

  6. Ramli S, Sim MS, Guad RM, Gopinath SCB, Subramaniyan V, Fuloria S, et al.
    J Oncol, 2021;2021:5519720.
    PMID: 33936199 DOI: 10.1155/2021/5519720
    The rising trend of gastrointestinal (GI) cancer has become a global burden due to its aggressive nature and poor prognosis. Long noncoding RNAs (lncRNAs) have recently been reported to be overexpressed in different GI cancers and may contribute to cancer progression and chemoresistance. They are featured with more than 200 nucleotides, commonly polyadenylated, and lacking an open reading frame. LncRNAs, particularly urothelial carcinoma-associated 1 (UCA1), are oncogenes involved in regulating cancer progression, such as cell proliferation, invasion, migration, and chemoresistance, particularly in GI cancer. This review was aimed to present an updated focus on the molecular regulatory roles and patterns of lncRNA UCA1 in progression and chemoresistance of different GI cancers, as well as deciphering the underlying mechanisms and its interactions with key molecules involved, together with a brief presentation on its diagnostic and prognostic values. The regulatory roles of lncRNA UCA1 are implicated in esophageal cancer, gastric cancer, pancreatic cancer, hepatobiliary cancer, and colorectal cancer, where they shared similar molecular mechanisms in regulating cancer phenotypes and chemoresistance. Comparatively, gastric cancer is the most intensively studied type in GI cancer. LncRNA UCA1 is implicated in biological roles of different GI cancers via interactions with various molecules, particularly microRNAs, and signaling pathways. In conclusion, lncRNA UCA1 is a potential molecular target for GI cancer, which may lead to the development of a novel chemotherapeutic agent. Hence, it also acts as a potential diagnostic and prognostic marker for GI cancer patients.
  7. Tune BXJ, Sim MS, Poh CL, Guad RM, Woon CK, Hazarika I, et al.
    J Oncol, 2022;2022:3249766.
    PMID: 35586209 DOI: 10.1155/2022/3249766
    Cancer is one of the major causes of death worldwide. Its treatments usually fail when the tumor has become malignant and metastasized. Metastasis is a key source of cancer recurrence, which often leads to resistance towards chemotherapeutic agents. Hence, most cancer-related deaths are linked to the occurrence of chemoresistance. Although chemoresistance can emerge through a multitude of mechanisms, chemoresistance and metastasis share a similar pathway, which is an epithelial-to-mesenchymal transition (EMT). Matrix metalloproteinases (MMPs), a class of zinc and calcium-chelated enzymes, are found to be key players in driving cancer migration and metastasis through EMT induction. The aim of this review is to discuss the regulatory roles and associated molecular mechanisms of specific MMPs in regulating chemoresistance, particularly EMT initiation and resistance to apoptosis. A brief presentation on their potential diagnostic and prognostic values was also deciphered. It also aimed to describe existing MMP inhibitors and the potential of utilizing other strategies to inhibit MMPs to reduce chemoresistance, such as upstream inhibition of MMP expressions and MMP-responsive nanomaterials to deliver drugs as well as epigenetic regulations. Hence, manipulation of MMP expression can be a powerful tool to aid in treating patients with chemo-resistant cancers. However, much still needs to be done to bring the solution from bench to bedside.
Related Terms
Filters
Contact Us

Please provide feedback to Administrator (afdal@afpm.org.my)

External Links