12 research outputs found

    Benzyl isothiocyanate induces apoptosis and inhibits tumor growth in canine mammary carcinoma via down-regulation of the cyclin B1/Cdk1 pathway

    Get PDF
    Background: Canine mammary carcinoma is common in female dogs, and its poor prognosis remains a serious clinical challenge, especially in developing countries. Benzyl isothiocyanate (BITC) has attracted great interest because of its inhibitory effect against tumor activity. However, its effect and the underlying mechanisms of action in canine mammary cancer are not well-understood. Here, we show that BITC suppresses mammary tumor growth, both in vivo and in vitro, and reveal some of the potential mechanisms involved. Methods: The effect of BITC on canine mammary cancer was evaluated on CIPp and CMT-7364, canine mammary carcinoma lines. The cell lines were treated with BITC and then subjected to wound healing and invasion assays. Cell cycles and apoptosis were measured using flow cytometry; TUNEL assay; immunohistochemistry (IHC) for caspase 3, caspase 9, and cyclin D1; hematoxylin and eosin (H&E) staining; and/or quantitative polymerase chain reaction (qPCR). Results: BITC showed a strong suppressive effect in both CIPp and CMT-7364 cells by inhibiting cell growth in vitro; these effects were both dose- and time-dependent. BITC also inhibited migration and invasion of CIPp and CMT-7364 cells. BITC induced G2 arrest and apoptosis, decreasing tumor growth in nude mice by downregulation of cyclin B1 and Cdk1 expression. Conclusion: BITC suppressed both invasion and migration of CIPp and CMT-7364 cells and induced apoptosis. BITC inhibited canine mammary tumor growth by suppressing cyclinB1 and Cdk1 expression in nude mice

    Long Non-Coding RNA as a Potential Biomarker for Canine Tumors

    No full text
    Cancer is the leading cause of death in both humans and companion animals. Long non-coding RNA (lncRNA) plays a crucial role in the progression of various types of cancers in humans, involving tumor proliferation, metastasis, angiogenesis, and signaling pathways, and acts as a potential biomarker for diagnosis and targeted treatment. However, research on lncRNAs related to canine tumors is in an early stage. Dogs have long been considered a promising natural model for human disease. This article summarizes the molecular function of lncRNAs as novel biomarkers in various types of canine tumors, providing new insights into canine tumor diagnosis and treatment. Further research on the function and mechanism of lncRNAs is needed, which will benefit both human and veterinary medicine

    Lamellar Keratoplasty Using Acellular Bioengineering Cornea (BioCorneaVet<sup>TM</sup>) for the Treatment of Feline Corneal Sequestrum: A Retrospective Study of 62 Eyes (2018–2021)

    No full text
    To retrospectively evaluate the effectiveness and outcome of lamellar keratoplasty using acellular bioengineering cornea (BioCorneaVetTM) for the treatment of feline corneal sequestrum (FCS). The medical records of cats diagnosed with FCS that underwent lamellar keratoplasty with BioCorneaVetTM between 2018 and 2021 with a minimum of 3 months of follow-up were reviewed. Follow-up examinations were performed weekly for 3 months, and then optical coherence tomography (OCT) examination was performed on select patients at 0, 3, 6, and 12 months post-operatively. A total of 61 cats (30 left eyes and 32 right eyes) were included. The Persian breed was overrepresented, 48/61 (78.69%). Four different thicknesses of acellular bioengineering cornea were used (200, 300, 400, or 450 microns), and the mean graft size was 8.23 mm (range, 5.00–12.00 mm). Minor complications were composed of partial dehiscence, and protrusion of the graft occurred in 7/62 eyes (11.29%). The median postoperative follow-up was 12.00 months (range, 3–41 months). A good visual outcome was achieved in 60/62 eyes (96.77%), and a mild to moderate corneal opacification occurred in 2/62 (3.23%). No recurrence of corneal sequestrum was observed. From the results, lamellar keratoplasty using acellular bioengineering cornea (BioCorneaVetTM) is an effective treatment for FCS, providing a good tectonic support and natural collagen framework, and resulting in satisfactory visual and cosmetic effects

    <i>P</i>. <i>mirabilis</i> treatment alters markers of hypoxia in the tumor microenvironment.

    No full text
    <p>(A) Western blotting demonstrated reduced expressions of CA IX and HIF-1a in the bacteria treatment group after 21 days of treatment; (B) The result was confirmed by immunohistochemistry for CA IX and HIF-1a in tumor sections of treated and control mice. CA IX positivity was localized in the cytoplasm and HIF-1a in the nucleus. (C and D) Quantitative analysis of immunohistochemistry staining indicated that CA IX and HIF-1a expression were lower in the bacteria treatment group than those in the control group (p < 0.05).</p

    The adherence and growth inhibition by <i>P</i>. <i>mirabilis in vitro</i>.

    No full text
    <p>(A) <i>P</i>. <i>mirabilis</i> adhered to 4T1, MDA-MB-231, MCF7, and CHO cells <i>in vitro</i>. (B) The cells were counted by stained with Trypan Blue. Bacteria treatment showed dose- and time-dependent antiproliferative effects in 4T1 cell lines (C) In the colony formation assay, <i>P</i>. <i>mirabilis</i> inhibited colony formation of breast tumor cells and CHO cells by crystal violet staining. Data were expressed as the mean ± SD.</p

    <i>P</i>. <i>mirabilis</i> suppressed spontaneous pulmonary metastases <i>in vivo</i>.

    No full text
    <p>Bacterial treatment inhibited tumor spontaneous pulmonary metastases by observing fresh lung tissues (A) at the end of the experiment. (B) Quantitative statistics of metastatic foci showed significant differences between <i>P</i>. <i>mirabilis</i> treatment group and PBS control group (n = 6) (p < 0.05). (C) Quantitative statistics showed lung weight of mice in the control group was significantly higher than those in the <i>P</i>. <i>mirabilis</i> treatment group (p < 0.05). (D) Those results were further confirmed by analyzing H&E stained sections. Data were expressed as the mean ± SD.</p

    <i>P</i>. <i>mirabilis</i> treatment regulated the immune system <i>in vivo</i>.

    No full text
    <p>Results of IHC indicated that the expression of NKp46 (A) and CD11c (B) in spleen sections was significantly increased after 24 hours bacteria treatment (p < 0.05). There were no differences of the expression of CD11b (C) and Ly-6G (D) between the control groups and the groups after 24 hours bacteria administration. (E) Quantitative analysis of immunohistochemistry staining for the expression of NKp46, CD11c, CD11b, and Ly-6G in the mice spleen tissues, respectively.</p

    The therapeutic effect of <i>P</i>. <i>mirabilis</i> in 4T1 solid tumor model.

    No full text
    <p>(A) Bacterial treatment suppressed tumor growth at the end of the experiment. Quantitative analysis demonstrated that the volume (B) and weight (C) of tumors in the <i>P</i>. <i>mirabilis</i> and CTX treatment groups were significantly less than those in the control group (p < 0.05). Corresponding to the images in (D) and (E) quantitative analysis of Ki-67 staining further confirmed that tumor proliferation was significantly inhibited in the treatment group (p < 0.05). (F) The body weights of mice significantly decreased at the beginning of treatment, but at the end of the experiment, they were not significantly different among these groups. (J) Morphological analysis of mice kidney tissues: there were no obvious differences in morphology between the <i>P</i>. <i>mirabilis</i> treatment group and the PBS control group. Data were expressed as mean ± SD.</p

    <i>Proteus mirabilis</i> inhibits cancer growth and pulmonary metastasis in a mouse breast cancer model

    No full text
    <div><p>A variety of bacteria have been used as agents and vectors for antineoplastic therapy. A series of mechanisms, including native bacterial toxicity, sensitization of the immune system and competition for nutrients, may contribute to antitumor effects. However, the antitumor effects of <i>Proteus</i> species have been minimally studied, and it is not clear if bacteria can alter tumor hypoxia as a component of their antineoplastic effect. In the present study, <i>Proteus mirabilis</i> bacteria were evaluated for the ability to proliferate and accumulate in murine tumors after intravenous injection. To further investigate the efficacy and safety of bacterial injection, mice bearing 4T1 tumors were treated with an intravenous dose of 5×10<sup>7</sup> CFU <i>Proteus mirabilis</i> bacteria via the tail vein weekly for three treatments. Histopathology, immunohistochemistry (IHC) and western analysis were then performed on excised tumors. The results suggested <i>Proteus mirabilis</i> localized preferentially to tumor tissues and remarkably suppressed the growth of primary breast cancer and pulmonary metastasis in murine 4T1 models. Results showed that the expression of NKp46 and CD11c was significantly increased after bacteria treatment. Furthermore, tumor expression of carbonic anhydrase IX (CA IX) and hypoxia inducible factor-1a (HIF-1a), surrogates for hypoxia, was significantly lower in the treated group than the control group mice as assessed by IHC and western analysis. These findings demonstrated that <i>Proteus mirabilis</i> may a promising bacterial strain for used against primary tumor growth and pulmonary metastasis, and the immune system and reduction of tumor hypoxia may contribute to the antineoplastic and antimetastatic effects observed.</p></div
    corecore