首页 | 本学科首页   官方微博 | 高级检索  
相似文献
 共查询到20条相似文献,搜索用时 890 毫秒
1.
2.
3.
An optimal treatment regimen for localized prostate cancer (PCa) is yet to be determined. Increasing evidence reveals a lower α/β ratio for PCa with hypofractionated radiation therapy (HFRT) regimens introduced to exploit this change in therapeutic ratio. HFRT also results in shortened overall treatment times of 4 to 5 weeks, thus reducing staffing and machine burden, and, more importantly, patient stress. This review evaluates pretreatment characteristics, outcomes, and toxicity for 15 HFRT studies on localized PCa. HFRT results in comparable or better biochemical relapse-free survival and toxicity and is a viable option for localized PCa.Key words: Localized prostate cancer, Hypofractionation, Short-course radiotherapy, Dose escalation, Biologic equivalenceMultiple randomized dose-escalation trials for localized prostate cancer (PCa) have shown improved biochemical relapse-free survival (bRFS) rates for higher total doses using conventionally fractionated radiotherapy (CFRT), though at a cost of longer treatment duration.14 The increased treatment time requires increased access to radiation treatment facilities, with additional burden on both patients and staff. To address the issue of prolonged treatment duration while maintaining equivalent bRFS, an increasing number of studies have pursued the role of hypofractionated radiotherapy (HFRT) with higher daily doses delivered in a shorter total amount of time. This treatment paradigm assumes a low α/β ratio for PCa, as demonstrated in several recent studies, with higher α/β ratios for normal surrounding tissues.57 By employing HFRT, the increased daily radiation doses exploit the aforementioned α/β ratios by allowing equivalent tumor kill as with CFRT, while also allowing for normal tissue repair.With longer-term and randomized HFRT data now reported in the literature, it seems appropriate to address whether the time has come to make HFRT the new standard. This article seeks to review the current literature and the role of HFRT in the modern era of radiotherapy for localized PCa.  相似文献   

4.
5.
6.
Although cancer cell secretome profiling is a promising strategy used to identify potential body fluid-accessible cancer biomarkers, questions remain regarding the depth to which the cancer cell secretome can be mined and the efficiency with which researchers can select useful candidates from the growing list of identified proteins. Therefore, we analyzed the secretomes of 23 human cancer cell lines derived from 11 cancer types using one-dimensional SDS-PAGE and nano-LC-MS/MS performed on an LTQ-Orbitrap mass spectrometer to generate a more comprehensive cancer cell secretome. A total of 31,180 proteins was detected, accounting for 4,584 non-redundant proteins, with an average of 1,300 proteins identified per cell line. Using protein secretion-predictive algorithms, 55.8% of the proteins appeared to be released or shed from cells. The identified proteins were selected as potential marker candidates according to three strategies: (i) proteins apparently secreted by one cancer type but not by others (cancer type-specific marker candidates), (ii) proteins released by most cancer cell lines (pan-cancer marker candidates), and (iii) proteins putatively linked to cancer-relevant pathways. We then examined protein expression profiles in the Human Protein Atlas to identify biomarker candidates that were simultaneously detected in the secretomes and highly expressed in cancer tissues. This analysis yielded 6–137 marker candidates selective for each tumor type and 94 potential pan-cancer markers. Among these, we selectively validated monocyte differentiation antigen CD14 (for liver cancer), stromal cell-derived factor 1 (for lung cancer), and cathepsin L1 and interferon-induced 17-kDa protein (for nasopharyngeal carcinoma) as potential serological cancer markers. In summary, the proteins identified from the secretomes of 23 cancer cell lines and the Human Protein Atlas represent a focused reservoir of potential cancer biomarkers.Cancer is a major cause of mortality worldwide, accounting for 10 million new cases and more than 6 million deaths per year. In developing countries, cancer is the second most common cause of death, accounting for 23–25% of the overall mortality rate (1). Notwithstanding improvements in diagnostic imaging technologies and medical treatments, the long term survival of most cancer patients is poor. Cancer therapy is often challenging because the majority of cancers are initially diagnosed in their advanced stages. For example, the 5-year survival rate for patients with HNC1 is less than 50%. More than 50% of all HNC patients have advanced disease at the time of diagnosis (2, 3). Enormous effort has been devoted to screening and characterizing cancer markers for the early detection of cancer. Thus far, these markers include carcinoembryonic antigen, prostate-specific antigen, α-fetoprotein, CA 125, CA 15-3, and CA 19-9. Unfortunately, most biomarkers have limited specificity, sensitivity, or both (4). Thus, there is a growing consensus that marker panels, which are more sensitive and specific than individual markers, would increase the efficacy and accuracy of early stage cancer detection (48). The development of novel and useful biomarker panels is therefore an urgent need in the field of cancer management.Proteomics technology platforms are promising tools for the discovery of new cancer biomarkers (9). Over the past decade, serum and plasma have been the major targets of proteomics studies aimed at identifying potential cancer biomarkers (1013). However, the progress of these studies has been hampered by the complex nature of serum/plasma samples and the large dynamic range between the concentrations of different proteins (14). As cancer biomarkers are likely to be present in low amounts in blood samples, the direct isolation of these markers from plasma and serum samples requires a labor-intensive process involving the depletion of abundant proteins and extensive protein fractionation prior to mass spectrometric analysis (1518). Alternatively, the secretome, or group of proteins secreted by cancer cells (19), can be analyzed to identify circulating molecules present at elevated levels in serum or plasma samples from cancer patients. These proteins have the potential to act as cancer-derived marker candidates, which are distinct from host-responsive marker candidates. We, along with other groups, have demonstrated the efficacy of secretome-based strategies in a variety of cancer types, including NPC (20), breast cancer (21, 22), lung cancer (23, 24), CRC (25, 26), oral cancer (27), prostate cancer (28, 29), ovarian cancer (30), and Hodgkin lymphoma (31). In these studies, proteins secreted from cancer cells into serum-free media were resolved by one- or two-dimensional gels followed by in-gel tryptic digestion and analysis via MALDI-TOF MS or LC-MS/MS. Alternatively, the proteins were trypsin-digested in solution and analyzed by LC-MS/MS. In general, more proteins were detected in the secretome using the LC-MS/MS method than the MALDI-TOF MS method. Advanced protein separation and identification technologies have made it possible to detect more proteins in the secretomes of cancer cells, thereby facilitating the discovery of cancer biomarkers.Although the cancer cell secretomes of various tumor types have been individually analyzed by different groups using distinct protocols, few studies have used the same protocol to compare cancer cell secretomes derived from different tumor types. We previously assessed the secretomes of 21 cancer cell lines derived from 12 cancer types (i.e. consisting of 795 protein identities and 325 non-redundant proteins) by one-dimensional gel and MALDI-TOF MS (25). Our preliminary findings revealed that different cell lines have distinct secreted protein profiles and that several putative biomarkers, such as Mac-2BP (20, 26, 27, 29) and cathepsin D (21, 23, 32), present in the secretome of a given cancer cell type are commonly shared among different cancers. These observations suggest that an in-depth comparison of secretomes derived from different tumor types may identify marker candidates common to most cancers as well as markers for specific cancer types. As an increasing number of proteins are identified in the secretomes of various cancer cell lines, scientists are faced with the challenge of quickly and efficiently narrowing down the list to candidates with higher chances of success during validation testing with precious clinical specimens.In the present study, we applied one-dimensional SDS-PAGE in conjunction with nano-LC-MS/MS (GeLC-MS/MS) (33, 34) to analyze the conditioned media of 23 cancer cell lines derived from 11 cancer types, including NPC, breast cancer, bladder cancer, cervical cancer, CRC, epidermoid carcinoma, liver cancer, lung cancer, T cell lymphoma, oral cancer, and pancreatic cancer. Within this data set, 4,584 non-redundant proteins were identified from a total of 23 cell lines, yielding an average of ∼1,300 proteins per cell line. Potential marker candidates were identified via the comparative analysis of different cell line secretomes and by putative linkages to cancer-relevant pathways. The selected proteins were further compared with the HPA (35) to generate a focused data set of proteins that are secreted or released, cancer type-specific, and highly expressed in human cancer tissues. Finally, we selectively validated four proteins as potential serological cancer markers using blood samples from cancer patients.  相似文献   

7.
Approximately 0.2% of Americans aged 20 to 39 years are childhood cancer survivors. Advances in cancer detection and therapy have greatly improved survival rates for young cancer patients; however, treatment of childhood cancers can adversely impact reproductive function. Many cancer patients report a strong desire to be informed of existing options for fertility preservation and future reproduction prior to initiation of gonadotoxic cancer therapies, including surgery, chemotherapy, and radiotherapy. This article discusses, in detail, the effects of cancer treatment on fertility in men and women, and outlines both current and experimental methods of fertility preservation among cancer patients.Key words: Fertility preservation, Childhood cancer, Sperm cryopreservation, Testicular tissue cryopreservation, Spermatogonial stem cell cryopreservation, Embryo cryopreservation, Oocyte cryopreservation, Ovarian tissue cryopreservationIn 2014, an estimated 15,780 new cancer cases were diagnosed among children and adolescents younger than age 20 years, resulting in 1960 deaths. In addition, 1 in 285 children will be diagnosed with cancer before age 20, and approximately 0.2% of Americans aged 20 to 39 years are childhood cancer survivors.1 Advances in cancer detection and therapy have greatly improved survival rates for young cancer patients; however, treatment of childhood cancers can adversely impact reproductive function (eg, men who survive childhood cancer are half as likely as their siblings to father a child).2 Many cancer patients report a strong desire to be informed of existing options for fertility preservation and future reproduction.3 Therefore, the American Society of Clinical Oncology and the American Society for Reproductive Medicine recommend that consideration of fertility preservation be included prior to initiation of gonadotoxic cancer therapies, including surgery, chemotherapy, and radiotherapy.46Infertility as a result of cancer treatment can be psycho logically upsetting for many patients,3,7,8 and data suggest that those who pursued fertility preservation usually cope better with their cancer treatment.9 Infertile cancer survivors have an option to become parents through adoption or gamete donation, but most declare a preference for having a biological child.3,10 Schover and colleagues3 found that 51% of newly diagnosed young male cancer patients reported a desire to have children in the future, and this rate increased to 77% for those who did not have children at the time of diagnosis. The desire to become a biological parent persists in male cancer survivors, as 70% reported wanting to father a child after chemotherapy treatment.9 A history of cancer treatment may be perceived by some to pose an increased risk to the health of future offspring; however, several studies have shown that male cancer survivors have not demonstrated an increased risk for having a child with birth defects or cancer.11,12 Recently, a retrospective cohort study conducted in the United States showed no increased risk of malformations or premature birth in the offspring of male cancer survivors.13The optimal time for consideration of fertility preservation is before the initiation of any oncologic therapy that can affect gametogenesis; thus, it is critical that fertility preservation is discussed with all patients at the time of diagnosis and before treatment starts. Practitioners who provide care for cancer patients should be aware of the relationship between cancer treatment and infertility. Moreover, they need to be able to appropriately refer patients to a reproductive medicine specialist in a timely fashion for further counseling and fertility preservation. Although fertility concerns are paramount to young adults with cancer, many oncologists still do not routinely address these concerns.3,14 In a survey of 200 young male cancer survivors who were primarily treated at a comprehensive cancer center, only 51% recalled being offered sperm cryopreservation prior to their cancer treatment.3 Further, it is important to recognize the psychologic stressors associated with a new cancer diagnosis and associated late effects of cancer treatment, such as infertility or early menopause. Findings from several studies support the importance of counseling patients regarding their risk for fertility issues and educating providers regarding the potential fertility preservation options that are available. For example, Babb and colleagues15 found that, at many institutions, this counseling is already taking place and there is a high rate of discussion with newly diagnosed patients regarding infertility.  相似文献   

8.
There is a mounting evidence of the existence of autoantibodies associated to cancer progression. Antibodies are the target of choice for serum screening because of their stability and suitability for sensitive immunoassays. By using commercial protein microarrays containing 8000 human proteins, we examined 20 sera from colorectal cancer (CRC) patients and healthy subjects to identify autoantibody patterns and associated antigens. Forty-three proteins were differentially recognized by tumoral and reference sera (p value <0.04) in the protein microarrays. Five immunoreactive antigens, PIM1, MAPKAPK3, STK4, SRC, and FGFR4, showed the highest prevalence in cancer samples, whereas ACVR2B was more abundant in normal sera. Three of them, PIM1, MAPKAPK3, and ACVR2B, were used for further validation. A significant increase in the expression level of these antigens on CRC cell lines and colonic mucosa was confirmed by immunoblotting and immunohistochemistry on tissue microarrays. A diagnostic ELISA based on the combination of MAPKAPK3 and ACVR2B proteins yielded specificity and sensitivity values of 73.9 and 83.3% (area under the curve, 0.85), respectively, for CRC discrimination after using an independent sample set containing 94 sera representative of different stages of progression and control subjects. In summary, these studies confirmed the presence of specific autoantibodies for CRC and revealed new individual markers of disease (PIM1, MAPKAPK3, and ACVR2B) with the potential to diagnose CRC with higher specificity and sensitivity than previously reported serum biomarkers.Colorectal cancer (CRC)1 is the second most prevalent cancer in the western world. The development of this disease takes decades and involves multiple genetic events. CRC remains a major cause of mortality in developed countries because most of the patients are diagnosed at advanced stages because of the reluctance to use highly invasive diagnostic tools like colonoscopy. Actually only a few proteins have been described as biomarkers in CRC (carcinoembryonic antigen (CEA), CA19.9, and CA125 (13)), although none of them is recommended for clinical screening (4). Proteomics analysis is actively used for the identification of new biomarkers. In previous studies, the use of two-dimensional DIGE and antibody microarrays allowed the identification of differentially expressed proteins in CRC tissue, including isoforms and post-translational modifications responsible for modifications in signaling pathways (58). Both approaches resulted in the identification of a collection of potential tumoral tissue biomarkers that is currently being investigated.However, the implementation of simpler, non-invasive methods for the early detection of CRC should be based on the identification of proteins or antibodies in serum or plasma (913). There is ample evidence of the existence of an immune response to cancer in humans as demonstrated by the presence of autoantibodies in cancer sera. Self-proteins (autoantigens) altered before or during tumor formation can elicit an immune response (1319). These tumor-specific autoantibodies can be detected at early cancer stages and prior to cancer diagnosis revealing a great potential as biomarkers (14, 15, 20). Tumor proteins can be affected by specific point mutations, misfolding, overexpression, aberrant glycosylation, truncation, or aberrant degradation (e.g. p53, HER2, NY-ESO1, or MUC1 (16, 2125)). In fact, a number of tumor-associated autoantigens (TAAs) were identified previously in different studies involving autoantibody screening in CRC (2628).Several approaches have been used to identify TAAs in cancer, including natural protein arrays prepared with fractions obtained from two-dimensional LC separations of tumoral samples (29, 30) or protein extracts from cancer cells or tissue (9, 31) probed by Western blot with patient sera, cancer tissue peptide libraries expressed as cDNA expression libraries for serological screening (serological analysis of recombinant cDNA expression libraries (SEREX)) (22, 32), or peptides expressed on the surface of phages in combination with microarrays (17, 18, 33, 34). However, these approaches suffer from several drawbacks. In some cases TAAs have to be isolated and identified from the reactive protein lysate by LC-MS techniques, or in the phage display approach, the reactive TAA could be a mimotope without a corresponding linear amino acid sequence. Moreover, cDNA libraries might not be representative of the protein expression levels in tumors as there is a poor correspondence between mRNA and protein levels.Protein arrays provide a novel platform for the identification of both autoantibodies and their respective TAAs for diagnostic purposes in cancer serum patients. They present some advantages. Proteins printed on the microarray are known “a priori,” avoiding the need for later identifications and the discovery of mimotopes. There is no bias in protein selection as the proteins are printed at a similar concentration. This should result in a higher sensitivity for biomarker identification (13, 35, 36).In this study, we used commercially available high density protein microarrays for the identification of autoantibody signatures and tumor-associated antigens in colorectal cancer. We screened 20 CRC patient and control sera with protein microarrays containing 8000 human proteins to identify the CRC-associated autoantibody repertoire and the corresponding TAAs. Autoantibody profiles that discriminated the different types of CRC metastasis were identified. Moreover a set of TAAs showing increased or decreased expression in cancer cell lines and paired tumoral tissues was found. Finally an ELISA was set up to test the ability of the most immunoreactive proteins to detect colorectal adenocarcinoma. On the basis of the antibody response, combinations of three antigens, PIM1, MAPKAPK3, and ACVR2B, showed a great potential for diagnosis.  相似文献   

9.
10.
11.
12.
The discovery of novel early detection biomarkers of disease could offer one of the best approaches to decrease the morbidity and mortality of ovarian and other cancers. We report on the use of a single-chain variable fragment antibody library for screening ovarian serum to find novel biomarkers for the detection of cancer. We alternately panned the library with ovarian cancer and disease-free control sera to make a sublibrary of antibodies that bind proteins differentially expressed in cancer. This sublibrary was printed on antibody microarrays that were incubated with labeled serum from multiple sets of cancer patients and controls. The antibodies that performed best at discriminating disease status were selected, and their cognate antigens were identified using a functional protein microarray. Overexpression of some of these antigens was observed in cancer serum, tumor proximal fluid, and cancer tissue via dot blot and immunohistochemical staining. Thus, our use of recombinant antibody microarrays for unbiased discovery found targets for ovarian cancer detection in multiple sample sets, supporting their further study for disease diagnosis.Despite many advances in the treatment of cancer, early detection and tumor removal remains the best prospect for overcoming disease. Ovarian cancer is an excellent example of the potential prognostic value of early detection because diagnosis at a localized stage has a 5-year survival rate of 93%. However, only 19% of cases are diagnosed at this stage, and by the time the disease has evolved to an advanced stage, the 5-year survival rate drops to 31% (1).Much effort has been expended to find early detection markers of ovarian cancer, and some success has been achieved. Most notable is CA125, the only approved marker for the detection of recurrence of ovarian cancer (2). Other leading targets are mesothelin and HE4, which have been examined by several groups for their efficacy as early detection markers (38). Nevertheless, several conditions necessitate the discovery of more specific and sensitive ovarian cancer markers: the heterogeneity of this disease, the ambiguity of its symptoms, its low incidence in the general population, and the low sensitivity and specificity of currently available markers.One of the difficulties in finding markers in blood is the complexity of the plasma/serum proteome, estimated in the tens to hundreds of thousands of proteins, as well as its large range in constituent protein concentrations, which can span 12 orders of magnitude (9). However, along with its easy accessibility, the fact that blood is in contact with virtually every tissue and contains tissue- and tumor-derived proteins makes it a preferred source for disease biomarker discovery.Our previous results (10, 11) and those of others (1214) using high density, full-length IgG antibody microarrays to validate and discover cancer serum biomarkers demonstrated that this platform is valuable for simultaneously comparing the levels of hundreds of proteins on dozens of serum samples from cancer patients and healthy controls. We confirmed overexpression of CA125, mesothelin, and HE4 in ovarian cancer samples using this high density microarray platform, validating our array methodology for measurement of cancer serum biomarkers and yielding new putative biomarkers for this disease (10, 11).Previously reported approaches are typically limited to a few hundred antibodies. The methodology reported here allows us to exploit the specific advantages of antibodies as high affinity capture reagents to detect differential expression of thousands of tumor biomarkers using a diverse (2 × 108 binding agents) single-chain variable fragment antibody (scFv)1 library for detection of ovarian cancer markers in serum, tumor cyst fluid, and ascites fluid. Our results build on previous reports of phage display library microarrays to discover autoantibody (1518) and other protein (12, 19, 20) cancer biomarkers. Our scFv are high affinity capture reagents consisting of the variable regions of human antibody heavy and light chains joined by a flexible linker peptide. These recombinant antibodies are able to recognize a wide variety of antigens, including many previously thought difficult, such as self-antigens and proteins that are not normally immunogenic in animals (2124). Using a highly diverse recombinant antibody library, one has the ability to overcome the complexity of the serum proteome. It has been calculated that for an immune repertoire to be complete (at least one antibody in the repertoire has reasonable affinity for every epitope possible in nature) it requires a diversity of at least 106 antibodies (25). The reported diversity of our scFv library exceeds this value by 100-fold (21).To enrich for antibodies that differentiate disease status, we performed a selection or panning of the naïve library for proteins that are differentially expressed in cyst fluid, ascites fluid, or serum of cancer patients with respect to healthy serum. We printed this sublibrary on activated hydrogel slides that were queried with three different sets of labeled case and control sera to further select those that discriminate cancer status in a statistically significant manner. Next, we identified some of the targets that bind to the individual scFv using high density nucleic acid programmable protein arrays (NAPPAs) expressing a total of over 7000 proteins. Finally, we validated the effectiveness of the selection process by confirming overexpression of these targets in cancer serum, cyst fluid, and ascites fluid as well as in tumor sections.  相似文献   

13.
Drug resistance poses a major challenge to ovarian cancer treatment. Understanding mechanisms of drug resistance is important for finding new therapeutic targets. In the present work, a cisplatin-resistant ovarian cancer cell line A2780-DR was established with a resistance index of 6.64. The cellular accumulation of cisplatin was significantly reduced in A2780-DR cells as compared with A2780 cells consistent with the general character of drug resistance. Quantitative proteomic analysis identified 340 differentially expressed proteins between A2780 and A2780-DR cells, which involve in diverse cellular processes, including metabolic process, cellular component biogenesis, cellular processes, and stress responses. Expression levels of Ras-related proteins Rab 5C and Rab 11B in A2780-DR cells were lower than those in A2780 cells as confirmed by real-time quantitative PCR and Western blotting. The short hairpin (sh)RNA-mediated knockdown of Rab 5C in A2780 cells resulted in markedly increased resistance to cisplatin whereas overexpression of Rab 5C in A2780-DR cells increases sensitivity to cisplatin, demonstrating that Rab 5C-dependent endocytosis plays an important role in cisplatin resistance. Our results also showed that expressions of glycolytic enzymes pyruvate kinase, glucose-6-phosphate isomerase, fructose-bisphosphate aldolase, lactate dehydrogenase, and phosphoglycerate kinase 1 were down-regulated in drug resistant cells, indicating drug resistance in ovarian cancer is directly associated with a decrease in glycolysis. Furthermore, it was found that glutathione reductase were up-regulated in A2780-DR, whereas vimentin, HSP90, and Annexin A1 and A2 were down-regulated. Taken together, our results suggest that drug resistance in ovarian cancer cell line A2780 is caused by multifactorial traits, including the down-regulation of Rab 5C-dependent endocytosis of cisplatin, glycolytic enzymes, and vimentin, and up-regulation of antioxidant proteins, suggesting Rab 5C is a potential target for treatment of drug-resistant ovarian cancer. This constitutes a further step toward a comprehensive understanding of drug resistance in ovarian cancer.Ovarian cancer is the major cause of death in women with gynecological cancer. Early diagnosis of ovarian cancer is difficult, while its progression is fast. The standard treatment is surgical removal followed by platinum-taxane chemotherapy. However, the efficacy of the traditional surgery and chemotherapy is rather compromised and platinum resistant cancer recurs in ∼25% of patients within six months, and the overall five-year survival rate is about 31% (13). Virtually no efficient second line treatment is available. In order to increase survival rates from ovarian cancer and enhance patients'' quality of life, new therapeutic targets are urgently required, necessitating a deeper understanding of molecular mechanisms of drug resistance.Mechanisms of drug-resistance in ovarian cancer have been extensively studied over the last 30 years. Earlier studies have found that multiple factors are linked to drug resistance in human ovarian cancer including reduced intracellular drug accumulation, intracellular cisplatin inactivation, and increased DNA repair (4). Reduced cellular drug accumulation is mediated by the copper transporter-1 responsible for the influx of cisplatin (59) and MDR1, which encodes an integral membrane protein named P-glycoprotein for the active efflux of platinum drugs. Up-regulation of MDR1 has been observed in cisplatin-treated ovarian cancer cells although cisplatin is not a substrate of P-glycoprotein (1013). A fraction of intracellular cisplatin can be converted into cisplatin-thiol conjugates by glutathione-S-transferase (GST) π, leading to inactivation of cisplatin. Up-regulation of both GSTπ and γ-glutamylcysteine synthetase has been associated with cisplatin resistance in ovarian, cervical and lung cancer cell lines (1418). Binding of cisplatin to DNA leads to intrastrand or interstrand cross-links that alter the structure of the DNA molecule causing DNA damage. It has been amply documented that pathways for recognition and repair of damaged DNA are up-regulated in drug-resistant cancer cells (1926). Furthermore, the secondary mutations have been identified, which restore the wild-type BRCA2 reading frame enhancing the acquired resistance to platinum-based chemotherapy (24). Alternations in other signaling pathways have also been found in drug resistant ovarian cancer (2729). For example, DNA-PK phosphorylates RAC-alpha serine/threonine-protein kinase (AKT) and inhibits cisplatin-mediated apoptosis (28); and silencing of HDAC4 increases acetyl-STAT1 levels to prevent platinum-induced STAT1 activation and restore cisplatin sensitivity (29).Proteomics is playing an increasingly important role in identifying differentially expressed proteins between drug-resistant and drug sensitive ovarian cancer cells (3035). An earlier study has identified 57 differentially expressed proteins in human ovarian cancer cells and their platinum-resistant sublines, including annexin A3, destrin, cofilin 1, Glutathione-S-transferase omega 1, and cytosolic NADP+-dependent isocitrate dehydrogenase using 2D gel electrophoresis (30). Employing a similar 2D gel electrophoresis approach, changes in protein expressions of capsid glycoprotein, fructose-bisphosphate aldolase C, heterogeneous nuclear ribonucleoproteins A2/B1, putative RNA-binding protein 3, Ran-specific GTPase-activating protein, ubiquitin carboxyl-terminal hydrolase isozyme L1, stathmin, ATPSH protein, chromobox protein homolog3, and phosphoglycerate kinase 1 (PGK)1 were found in A2780 and drug-resistant A2780 cells (32). It is worth mentioning that ALDO and PGK are glycolytic enzymes, indicating that glycolysis plays a role in drug resistance. Studies have demonstrated that resistance to platinum drugs in ovarian cancer cells is linked to mitochondrial dysfunctions in oxidative phosphorylation and energy production (3640). Mitochondrial proteomic analysis of drug-resistant cells has shown that five mitochondrial proteins (ATP-a, PRDX3, PHB, ETF, and ALDH) that participate in the electron transport respiratory chain are down-regulated in drug-resistant cell lines (41). PRDX3 is involved in redox regulation of the cell to protect radical-sensitive enzymes from oxidative damage. However, it is not clear how down-regulation of PRDX3 is associated with drug-resistance. A more recent study showed that activated leukocyte cell adhesion molecule (ALCA) and A kinase anchoring protein 12 (AKAP12) are elevated in drug-resistant A2780-CP20 cells by quantifying the mitochondrial proteins (42). Despite these efforts, the drug-resistance mechanisms are not yet well understood.In this work, we established and characterized a drug-resistant cell line A2780-DR from A2780 cells. We employed a quantitative proteomic method to identify the differentially expressed proteins between A2780 and A2780-DR cells. Expression changes of selected proteins were confirmed by qPCR and Western blotting. We also used shRNA silencing to explore functions of Rab 5C and Rab 11B proteins in drug resistance. Our data indicate that the differentially expressed proteins participate in a variety of cellular processes and enhance our understanding of the mechanisms of drug resistance in ovarian cancer cells.  相似文献   

14.
15.
16.
17.
Gastric cardia cancer (GCC), which occurs at the gastric-esophageal boundary, is one of the most malignant tumors. Despite its high mortality and morbidity, the molecular mechanism of initiation and progression of this disease is largely unknown. In this study, using proteomics and metabolomics approaches, we found that the level of several enzymes and their related metabolic intermediates involved in glucose metabolism were deregulated in GCC. Among these enzymes, two subunits controlling pyruvic acid efflux, lactate dehydrogenase A (LDHA) and pyruvate dehydrogenase B (PDHB), were further analyzed in vitro. Either down-regulation of LDH subunit LDHA or overexpression of PDH subunit PDHB could force pyruvic acid into the Krebs cycle rather than the glycolysis process in AGS gastric cancer cells, which inhibited cell growth and cell migration. Our results reflect an important glucose metabolic signature, especially the dysregulation of pyruvic acid efflux in the development of GCC. Forced transition from glycolysis to the Krebs cycle had an inhibitory effect on GCC progression, providing potential therapeutic targets for this disease.Gastric cardia cancer (GCC),1 which occurs at the gastric-esophageal boundary, is one of the most malignant tumors. Despite the steadily falling incidence of gastric non-cardia cancer in the past two decades (1), the rate of GCC has risen rapidly, establishing gastric cancer as the second major cause of cancer-related deaths throughout the world (2). GCC has become a significant cause of mortality and morbidity both in the west (35) and in Asia (6, 7), especially in China (8). Although this cancer has become an important health problem worldwide, the its pathogenesis has not been well characterized (1). Most patients are diagnosed at an advanced stage, contributing to the high mortality rate of the disease.Systematic proteomics analysis has proved to be a powerful approach in a variety of human cancer research, including lung (9), esophagus (10), gastric (11), liver (12), breast (13), and brain cancer (14). Metabolomics, another new bio-omics technology recently introduced into cancer research (15), is the global analysis of the small metabolites produced by normal or pathologic cellular processes. Some metabolic intermediates have been identified as new cancer biomarkers (16).Using proteomics and metabolomics methods in this study, we found that a series of proteins and metabolic intermediates, mainly involved in glucose metabolism, were altered during the development of GCC. The high activity of anaerobic glycolysis and the impairment of aerobic respiration occurring in these cells recapitulated the Warburg effect (17). Further studies using a gastric cancer cell line demonstrated that the predominant anaerobic glycolysis was essential for tumor cells to sustain rapid proliferation, whereas forced transition from anaerobic glycolysis to aerobic respiration inhibited the growth of tumor cells. In conclusion, our study revealed the major metabolic alterations essential for the development of GCC and discovered a biomarker signature of GCC. Such a finding has the potential to improve early diagnosis and prognosis and helps to identify new therapeutic targets.  相似文献   

18.
Insulin plays a central role in the regulation of vertebrate metabolism. The hormone, the post-translational product of a single-chain precursor, is a globular protein containing two chains, A (21 residues) and B (30 residues). Recent advances in human genetics have identified dominant mutations in the insulin gene causing permanent neonatal-onset DM2 (14). The mutations are predicted to block folding of the precursor in the ER of pancreatic β-cells. Although expression of the wild-type allele would in other circumstances be sufficient to maintain homeostasis, studies of a corresponding mouse model (57) suggest that the misfolded variant perturbs wild-type biosynthesis (8, 9). Impaired β-cell secretion is associated with ER stress, distorted organelle architecture, and cell death (10). These findings have renewed interest in insulin biosynthesis (1113) and the structural basis of disulfide pairing (1419). Protein evolution is constrained not only by structure and function but also by susceptibility to toxic misfolding.Insulin plays a central role in the regulation of vertebrate metabolism. The hormone, the post-translational product of a single-chain precursor, is a globular protein containing two chains, A (21 residues) and B (30 residues). Recent advances in human genetics have identified dominant mutations in the insulin gene causing permanent neonatal-onset DM2 (14). The mutations are predicted to block folding of the precursor in the ER of pancreatic β-cells. Although expression of the wild-type allele would in other circumstances be sufficient to maintain homeostasis, studies of a corresponding mouse model (57) suggest that the misfolded variant perturbs wild-type biosynthesis (8, 9). Impaired β-cell secretion is associated with ER stress, distorted organelle architecture, and cell death (10). These findings have renewed interest in insulin biosynthesis (1113) and the structural basis of disulfide pairing (1419). Protein evolution is constrained not only by structure and function but also by susceptibility to toxic misfolding.  相似文献   

19.
20.
设为首页 | 免责声明 | 关于勤云 | 加入收藏

Copyright©北京勤云科技发展有限公司  京ICP备09084417号