به جمع مشترکان مگیران بپیوندید!

تنها با پرداخت 70 هزارتومان حق اشتراک سالانه به متن مقالات دسترسی داشته باشید و 100 مقاله را بدون هزینه دیگری دریافت کنید.

برای پرداخت حق اشتراک اگر عضو هستید وارد شوید در غیر این صورت حساب کاربری جدید ایجاد کنید

عضویت
جستجوی مطالب مجلات
ردیف ۱۰-۱ از ۸۱۱۷۴ عنوان مطلب
|
  • Wenfei Gu, Guangtao Qi, Li Chen *
    Background
    Spi-B transcription factor (SPIB) is an E-twenty-six (ETS) transcription factor associated with tumor immunity.
    Objective
    To investigate the functions and mechanisms of SPIB in ovarian cancer (OC) cells.
    Methods
    Cell proliferation, apoptosis, migration, and invasion were determined using colony formation, EdU, flow cytometry, and transwell assays, respectively. The binding sites of programmed death-ligand 1 (PD-L1) and SPIB were predicted using the JASPAR database and verified using the ChIP and luciferase reporter assays.
    Results
    SPIB knockdown inhibited OC cell proliferation, migration, and invasion, and significantly boosted apoptosis (p<0.05). SPIB directly enhanced PD-L1 transcription in OVCAR-3 and SKOV3 cells (p<0.05). Importantly, the JAK/STAT pathway was markedly inactivated in OC cells upon SPIB knockdown. SPIB knockdown markedly decreased JAK2 and STAT1 phosphorylation in OVCAR-3 and SKOV3 cells (p<0.05).
    Conclusion
    These data indicate that SPIB knockdown inhibits OC cell progression by downregulating PD-L1 and inactivating the JAK/STAT pathway.
    Keywords: JAK, STAT Pathway, Ovarian Cancer, PD-L1 (CD274), SPIB
  • Akbar Oghalaie, Alireza Shoari, Fatemeh Kazemi-Lomedasht, Fatemeh Rahimi-Jamnani, Fereidoun Mahboudi, Hajarossadat Ghaderi, Mohammad Hosseininejad-Chafi, Reza Moaazami, Arghavan Ardalan, Somayeh Piri-Gavgani, Delavar Shabazzadeh, Mahdi Behdani *
    Programmed death ligand-1 (PD-L1, CD274 and B7-H1) has been described as a ligand for immune inhibitory receptor programmed death protein 1 (PD-1). With binding to PD-1 on activated T cells, PD-L1 can prevent T cell responses via motivating apoptosis. Consequently, it causes cancers immune evasion and helps the tumor growth; hence, PD-L1 is regarded as a therapeutic target for malignant cancers. The anti-PD-L1 monoclonal antibody targeting PD-1/PD-L1 immune checkpoint has attained remarkable outcomes in clinical application and has turned to one of the most prevalent anti-cancer drugs. The present study aimed to develop polyclonal heavy chain antibodies targeting PD-L1via Camelus dromedarius immunization. The extra-cellular domain of human PD-L1 (hPD-L1) protein was cloned, expressed, and purified. Afterwards, this recombinant protein was utilized as an antigen for camel immunization to acquire polyclonal camelid sera versus this protein. Our outcomes showed that hPD-L1 protein was effectively expressed in the prokaryotic system. The antibody-based techniques, such as enzyme-linked immunosorbent assay, western blotting, and flow cytometry displayed that the hPD-L1 protein was detected by generated polyclonal antibody. Due to the advantages of multi-epitope-binding ability, our study exhibited that camelid antibody is effective to be applied significantly for detection of PD-L1 protein in essential antibody-based studies.
    Keywords: Camelid heavy-chain antibody, Immunization, Polyclonal antibody, Programmed death ligand-1
  • Niloofar Namvar, Mehdi Montazer, Simin Ahmadvand, Bahare Sadeghian, Abbas Ghaderi

    There is an urgent need to discover novel prognostic biomarkers and treatment strategies for gastric cancer (GC) patients. Several immune-related markers have been proposed as prognostic tools and immunotherapeutic targets to manage diseases. In this regard, we evaluated the expression pattern and prognostic significance of programmed death-1 (PD-1), programmed death-ligand 1 (PD-L1), CD45RO+ tumor-infiltrating lymphocytes (TILs), and DNA mismatch repair (MMR) proteins (MLH1, MSH2, PMS2, and MSH6) in non-metastatic intestinal-type gastric adenocarcinoma. Samples and data from 70 GC patients were collected. Immunohistochemistry staining was used to detect the markers. We then evaluated the prognosis significance of each marker and their intercorrelation. Cytoplasmic PD-1 expressed by tumor cells was significantly associated with poorer survival. However, multivariate analysis indicated stronger prognostic values for TNM stage, tumor location, and extracellular mucin. A significant positive association was found between CD45ROhigh TILs and PD-1 expression on tumor-infiltrating cells (TICs). All GC patients with deficient MMR (d‑MMR) had a higher number of CD45RO+ TILs and were associated with PD-1+ TICs and PD‑L1+ tumor cells (TCs). However, the difference was not statistically significant. Despite the association of PD‑1 overexpression on TCs with shorter overall survival, histopathological factors, including tumor location, TNM stage, and extracellular mucin, remain the most decisive prognostic factors in non-metastatic intestinal-type gastric adenocarcinoma. Additionally, our data support a prognostic role for d-MMR and CD45RO, but not PD-1 and PD-L1 expression on TICs.

    Keywords: Immunohistochemistry, CD45RO antigens, DNA mismatch repair, PD-L1, PD-1, Stomach neoplasms
  • Mohsen Soltanshahi, Saeid Taghiloo, Hossein Asgarian-Omran*

    Tumor-targeted therapy with small-molecule inhibitors (SMIs) has been demonstrated to be a highly effective therapeutic strategy for various cancers. However, their possible associations with immune evasion mechanisms remain unknown. This study examined the association of inhibitors of the protein kinase B (AKT), mammalian target of rapamycin (mTOR), and Bruton’s tyrosine kinase (BTK) signaling pathways with the expression of immune checkpoint ligands programmed death-ligand 1 (PD-L1), CD155, and galectin-9 (Gal-9) in a breast cancer cell line. MCF-7 cells were treated with everolimus, MK-2206, and ibrutinib. AnMTT assay was used to determine the optimal dose for all drugs. A real-time polymerase chain reaction was utilized to measure the mRNA expression of PD-L1, CD155, and Gal-9. The western blot technique was also employed to evaluate the protein expression of the phosphorylated signal transducer and activator of transcription 3 (STAT3). The optimal doses of everolimus, MK-2206, and ibrutinib were observed to be 200, 320, and 2000 nM, respectively. The PD-L1 and CD155 mRNA expression was significantly decreased following the treatment with everolimus and ibrutinib, but not with MK-2206. There were no differences in Gal-9 expression between the single-treated and control groups; however, combined treatment with everolimus and ibrutinib increased its mRNA expression. Everolimus and ibrutinib both inhibited constitutive STAT3 phosphorylation in MCF-7, which was more pronounced in combination treatment. The findings regarding the modulation of PD-L1, CD155, and Gal-9 molecules by SMIs emphasize the crosstalk between the expression of these immune checkpoint molecules and AKT/mTOR/BTK signaling pathways through STAT3 as a critical transcription factor.

    Keywords: Breast Cancer, Small-Molecule Inhibitors, PD-L1, CD155, Galectin-9, STAT3
  • Mohsen Soltanshahi, Saeid Taghiloo, Hossein Asgarian-Omran*

    Tumor-targeted therapy with small-molecule inhibitors (SMIs) has been demonstrated to be a highly effective therapeutic strategy for various cancers. However, their possible associations with immune evasion mechanisms remain unknown. This study examined the association of inhibitors of the protein kinase B (AKT), mammalian target of rapamycin (mTOR), and Bruton’s tyrosine kinase (BTK) signaling pathways with the expression of immune checkpoint ligands programmed death-ligand 1 (PD-L1), CD155, and galectin-9 (Gal-9) in a breast cancer cell line. MCF-7 cells were treated with everolimus, MK-2206, and ibrutinib. AnMTT assay was used to determine the optimal dose for all drugs. A real-time polymerase chain reaction was utilized to measure the mRNA expression of PD-L1, CD155, and Gal-9. The western blot technique was also employed to evaluate the protein expression of the phosphorylated signal transducer and activator of transcription 3 (STAT3). The optimal doses of everolimus, MK-2206, and ibrutinib were observed to be 200, 320, and 2000 nM, respectively. The PD-L1 and CD155 mRNA expression was significantly decreased following the treatment with everolimus and ibrutinib, but not with MK-2206. There were no differences in Gal-9 expression between the single-treated and control groups; however, combined treatment with everolimus and ibrutinib increased its mRNA expression. Everolimus and ibrutinib both inhibited constitutive STAT3 phosphorylation in MCF-7, which was more pronounced in combination treatment. The findings regarding the modulation of PD-L1, CD155, and Gal-9 molecules by SMIs emphasize the crosstalk between the expression of these immune checkpoint molecules and AKT/mTOR/BTK signaling pathways through STAT3 as a critical transcription factor.

    Keywords: Breast Cancer, Small-Molecule Inhibitors, PD-L1, CD155, Galectin-9, STAT3
  • یعقوب فتح الهی، بهاره دبیر منش*، خسرو خواجه، عاطفه خداکرمی

    نقاط بازرسی ایمنی، مولکول هایی هستند که سیستم ایمنی را تنظیم می کنند. برخی از سلول های تومور می توانند لیگاندهای متصل شونده به این نقاط بازرسی ایمنی را برای فرار از پاسخ های ایمنی ضد توموری بیان کنند. برخی از عوامل، مانند آنتی بادی ها، می توانند این نقاط بازرسی را مهار کنند. هدف از این مطالعه بیان یک دیا بادی دو منظوره جدید در فضای پری پلاسمیک باکتری E. coli BL21(DE3)  برای مهار همزمان دو نقطه بازرسی ایمنی، پروتئین مرتبط با 4 (CTLA 4) و لیگاند متصل شونده (PD L1)  است. دیا بادی دو منظوره بر اساس ژن نواحی متغیر آنتی بادی های anti PD-L1 و anti CTLA 4 طراحی و جهت بیان درباکتری  E. coli BL21 (DE3) بهینه سازی و در پلاسمید بیانی pET21 کلون شد. پس از ترنسفرم کردن به داخل سویه بیانی E. coli BL21(DE3) اثر شرایط بیانی مختلف کشت مورد بررسی قرار گرفت، بیانDia-21 با وزن مولکولی 55کیلو دالتون با الکتروفورز ژل SDS-PAGE و وسترن بلات تایید شد. بهترین شرایط برای بیان پری پلاسمیک به فرم محلول بیان با 0/5 میلی مولار IPTG در دمای 23 درجه سانتی گراد محیط کشتLB  و زمان 18 ساعت به دست آمد. پروتئین بیان شده سپس با استفاده از کروماتوگرافی تمایلی نیکل آگارز با بازده نهایی 0/4 میلی گرم در لیتر تخلیص و نهایت میل ترکیبی برهمکنش پروتئین خالص شده توسط  Cell-ELISA  بررسی شد. پروتئین دیا بادی نوترکیب در یک سیستم باکتریایی بیان و خالص سازی و برهم کنش پروتئین دیا بادی نوترکیب  باگیرنده سطح سلولی PDL-1 از طریق Cell-Elisa بررسی شد.

    کلید واژگان: نقاط بازرسی, دیابادی, CTLA-4, PDL-1
    Yaghoub Fathollahi, Bahareh Dabirmanesh*, Khosrow Khajeh, Atefeh Khodakarami

    Immune checkpoints are molecules that regulators  the immune system. However, some tumor cells can express the ligands of immune checkpoints to escape from antitumor immune responses. Some agents, such as antibodies, can inhibit these checkpoints that prevent the immune system from targeting and killing cancer cells. The aim of this study was to express a novel bispecific diabody in periplasmic space of E.coli for simultaneous targeting of two immune checkpoints, cytotoxic T‑lymphocyte‑associated protein 4 (CTLA‑4) and programmed death- ligand 1 (PD‑L1). The bispecific diabody was constructed based on the variable regions gene of anti PD-L1 and anti CTLA‑4 antibodies. The optimum codon for expression in E. coli was chemically synthesized and subcloned in pET21 expression plasmid. After transformation, the effect of cultivation conditions on periplasmic expression of the protein in E. coli BL21(DE3) was evaluated. Then, the bispecific diabody was purified . Expression of diabody with a molecular weight of 55 kDa was verified by Sodium dodecyl sulfate‑polyacrylamide gel electrophoresis and western blotting analysis. The best condition for soluble periplasmic expression was obtained to be incubation with 0.5 mM isopropyl β‑D‑1‑thiogalactopyranoside at 23°C. The protein was successfully purified using affinity chromatography with a final yield of 0.4 mg/L. The affinity of the purified protein interaction were checked by ELISA. Recombinant Diabody protein was cloned, expressed, and purified in a bacterial system and Diabody Interaction with PDL-1 receptor conformed by Cell-Elisa.

    Keywords: Immune Checkpoints, Recombinant Diabody, CTLA 4, PD-L1
  • Mohammad Reza Dehghani, Reza Ahangari Cohan, Kobra Omidfar, Mohammad Hossein Ghahremani *
    Tri-functional antibodies, as an effective novel tumor targeting agents, are composed of anti-CD3 rat IgG2b and an anti-tumor antigen antibody. We have intended to develop a novel drug system to induce the apoptosis of HER2-expressing tumor cells, activate and engage cytotoxic T lymphocytes, natural killer cells, and macrophages. In addition, the drug can inhibit programmed death ligand 1 (PDL-1)-expressing tumor cells. We have designed a mammalian vector system suitable to express the trifunctional antibody composed of antiHER2×human-CD80: human-IgG1Fc antibody. This antibody contains four chains of anti-HER2/CL, anti-HER2/CH1-3, B7.1/CL, and B7.1/CH1-3 within the human IgG1 framework and so the vector system will simultaneously express the four chains. The amino acid/nucleotide sequences datasets were retrieved from the GenBank, UniProtKB and PDB databases. The heavy and light chains variable domain framework regions and complementarity determining regions of scA21 antibody were determined by IMGT/V-QUEST and Paratome software. The amino acid sequences of tri-functional antibody chains manually were assembled and converted to DNA sequences by sequence manipulation suite software. The adapting codon usage of these DNA sequences was performed by JCAT software. Finally, the secondary structures of obtained RNAs from the DNAs were individually analyzed by RNAfold program. The Prodigy equilibrium dissociation constant, a ratio of koff/kon, between the antibody and its antigen for hantiHER2VHCH-HER2, hantiHER2VLCL-HER2, CD80CH-CD28, and CD80CL-CD28 were equal to 3.60E-10, 3.10E-9, 1.10E-8 and 2.70E-10; respectively. These findings were confirmed the composition and nano-molar affinity of the respective constructs. A single specific no-cloning expression vector, pHuchiTriomAb, was designed in silico with a desirable length of 8.292 Kbps and bidirectional expression potential for the four chimeric antibody chains. This construct was designed, and gene codon usage adapted to be expressed within CHO cells and secreted a trifunctional antibody into the cell culture medium by interleukin 21 signal peptide.This robust expression system for rapid production of tri-functional antibody has been designed to substitute hybrid hybridoma technology (quadroma and trioma) with the facilitated purification of the trifunctional antibody from the antibody variants.
    Keywords: Insilico, Tri-functional antibody, Cancer, Immunotherapy, Biotechnology
  • Aarya Unnikrishnan, Vijaya Basavaraj *
    Background & Objective

    Head and Neck Squamous Cell Carcinoma (HNSCC) is a highly frequent malignancy worldwide and is also the leading cause of death. The prognosis for individuals with HNSCC remains dismal, with a five-year survival rate of less than 50%. The novel anti-PD-L1 immunotherapy is found to be promising, and immunohistochemistry (IHC) has been established as a reliable method for patient stratification. We intend to evaluate the prognostic significance of the expression of programmed death ligand-1 (PD-L1) in HNSCC and determine its association with clinicopathological variables.

    Methods

    A total of 50 cases of biopsy-confirmed HNSCC were studied in a tertiary hospital between Dec 2020 and June 2022. The specimens were tested for PD-L1 IHC expression with antibody clone CAL-10 (Biocare) and scored by Combined Positive Score (CPS). The association between PD-L1 expression and clinicopathological variables was evaluated.

    Results

    PD-L1 was positive in 92% of the cases, and a significant association (P= 0.024) was seen between PD-L1 expression and tumor-infiltrating lymphocytes (TILs). PD-L1 did not show any significant association with patient demographics, tumor site, grade, or stage.

    Conclusion

    In the present study, evaluation of the immunohistochemical expression of PD-L1 on the tumor cells and TILs in HNSCC revealed a high prevalence of PD-L1 expression. PD-L1 IHC studies for patient selection for immunotherapy would be a promising technique. Frequent PD-L1 expression in tumors with significant TILs may be useful in identifying patients who may benefit from anti-PD-1/PD-L1 therapy.

    Keywords: Head, Neck, Programmed death ligand-1, Squamous cell carcinoma, Tumor-infiltrating Lymphocytes
  • Shruti Tadachina, Sheela Shivalingaiah *, Mahesh Shetty
    Background & Objective

    Colorectal carcinoma (CRC) is one of the most common cancers worldwide. The interaction of programmed cell death receptor 1 (PD-1) and programmed death ligand 1 (PD-L1) plays an important role by inhibiting the immune mechanism by which cancer cells escape antitumor immunity. Immunotherapy using checkpoint inhibitors is a growing treatment modality in many cancers; one such is anti-PD1/PD-L1. The present study aimed to study the immunohistochemical (IHC) expression of PD-L1 in CRC and evaluate the association of PD-L1 expression in CRC with various known clinicopathological parameters.

    Methods

    It was a 2-year prospective study and included 34 colectomy specimens diagnosed as colorectal adenocarcinoma. The expression of PD-L1 was evaluated on tumor cells & tumor-infiltrating immune cells (TIICs) and correlated with various clinicopathological parameters.

    Results

    Immunohistochemical expression of PD-L1 on tumor cells and tumor microenvironment in CRC revealed positivity in 17.65% of cases each. The PD-L1 expression on tumor cells was associated with lymphovascular invasion (LVI) and perineural invasion (PNI) with P- values of 0.012 and 0.005, respectively, while PD-L1 expression on TIICs was associated with tumor budding with P-value 0.022.

    Conclusion

    IHC expression of PD-L1 on tumor cells and immune cells was associated with some known poor prognostic factors. Since anti-PD1/PD-L1 is used for targeted therapy, it may be beneficial and economically feasible to evaluate PD-L1 in CRC and establish its role as a prognostic factor.

    Keywords: Colorectal carcinoma, PD-L1 Expression, IHC, tumor cells, Tumour infiltrating immune cells
نکته:
  • از آنجا که گزینه «جستجوی دقیق» غیرفعال است همه کلمات به تنهایی جستجو و سپس با الگوهای استاندارد، رتبه‌ای بر حسب کلمات مورد نظر شما به هر نتیجه اختصاص داده شده‌است‌.
  • نتایج بر اساس میزان ارتباط مرتب شده‌اند و انتظار می‌رود نتایج اولیه به موضوع مورد نظر شما بیشتر نزدیک باشند. تغییر ترتیب نمایش به تاریخ در جستجوی چندکلمه چندان کاربردی نیست!
  • جستجوی عادی ابزار ساده‌ای است تا با درج هر کلمه یا عبارت، مرتبط ترین مطلب به شما نمایش داده‌شود. اگر هر شرطی برای جستجوی خود در نظر دارید لازم است از جستجوی پیشرفته استفاده کنید. برای نمونه اگر به دنبال نوشته‌های نویسنده خاصی هستید، یا می‌خواهید کلمات فقط در عنوان مطلب جستجو شود یا دوره زمانی خاصی مدنظر شماست حتما از جستجوی پیشرفته استفاده کنید تا نتایج مطلوب را ببینید.
در صورت تمایل نتایج را فیلتر کنید:
* با توجه به بالا بودن تعداد نتایج یافت‌شده، آمار تفکیکی نمایش داده نمی‌شود. بهتراست برای بهینه‌کردن نتایج، شرایط جستجو را تغییر دهید یا از فیلترهای زیر استفاده کنید.
* ممکن است برخی از فیلترهای زیر دربردارنده هیچ نتیجه‌ای نباشند.
نوع نشریه
اعتبار نشریه
زبان مطلب
درخواست پشتیبانی - گزارش اشکال