Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Med Oncol ; 41(6): 138, 2024 May 06.
Article in English | MEDLINE | ID: mdl-38705935

ABSTRACT

Breast cancer (BC) is associated with type 2 diabetes mellitus (T2DM) and obesity. Glucagon-like peptide (GLP)-1 regulates post-prandial insulin secretion, satiety, and gastric emptying. Several GLP-1 analogs have been FDA-approved for the treatment of T2DM and obesity. Moreover, GLP-1 regulates various metabolic activities across different tissues by activating metabolic signaling pathways like adenosine monophosphate (AMP) activated protein kinase (AMPK), and AKT. Rewiring metabolic pathways is a recognized hallmark of cancer, regulated by several cancer-related pathways, including AKT and AMPK. As GLP-1 regulates AKT and AMPK, we hypothesized that it alters BC cells' metabolism, thus inhibiting proliferation. The effect of the GLP-1 analogs exendin-4 (Ex4) and liraglutide on viability, AMPK signaling and metabolism of BC cell lines were assessed. Viability of BC cells was evaluated using colony formation and MTT/XTT assays. Activation of AMPK and related signaling effects were evaluated using western blot. Metabolism effects were measured for glucose, lactate and ATP. Exendin-4 and liraglutide activated AMPK in a cAMP-dependent manner. Blocking Ex4-induced activation of AMPK by inhibition of AMPK restored cell viability. Interestingly, Ex4 and liraglutide reduced the levels of glycolytic metabolites and decreased ATP production, suggesting that GLP-1 analogs impair glycolysis. Notably, inhibiting AMPK reversed the decline in ATP levels, highlighting the role of AMPK in this process. These results establish a novel signaling pathway for GLP-1 in BC cells through cAMP and AMPK modulation affecting proliferation and metabolism. This study suggests that GLP-1 analogs should be considered for diabetic patients with BC.


Subject(s)
Breast Neoplasms , Exenatide , Glucagon-Like Peptide 1 , Liraglutide , Humans , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Exenatide/pharmacology , Female , Liraglutide/pharmacology , Glucagon-Like Peptide 1/metabolism , Glucagon-Like Peptide 1/pharmacology , Glucagon-Like Peptide 1/analogs & derivatives , Cell Line, Tumor , AMP-Activated Protein Kinases/metabolism , Signal Transduction/drug effects , Cell Survival/drug effects , Warburg Effect, Oncologic/drug effects , Cell Proliferation/drug effects , Venoms/pharmacology , Adenylate Kinase/metabolism , Peptides/pharmacology
2.
Harefuah ; 163(2): 102-108, 2024 Feb.
Article in Hebrew | MEDLINE | ID: mdl-38431859

ABSTRACT

INTRODUCTION: Translational research in medicine has undergone significant changes in the last decade, primarily due to the remarkable technological advancements made during this period. Oncology research is at the forefront of translational research in medicine and is heavily influenced by these changes. In this article, we briefly review the technologies that form the basis for the "next generation of translational research" in oncology in the coming decades, as well as the emerging trends in translational research in oncology through the implementation of these technologies.


Subject(s)
Medicine , Translational Research, Biomedical , Humans , Medical Oncology
3.
Oncogene ; 43(5): 354-362, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38040805

ABSTRACT

Klotho, a 1012 amino acid transmembrane protein, is a potent tumor suppressor in different cancer types. Klotho is composed of two internal repeats KL1 and KL2, and the tumor suppressor activity is primarily attributed to the KL1 domain. Despite its significant role in regulating various cancer-related pathways, the precise mechanism underlying its tumor suppressor activity remains unresolved. In this study, we aimed to identify the sequence responsible for the tumor suppressor function of Klotho and gain insights into its mechanism of action. To accomplish this, we generated expression vectors of truncated KL1 at the C and N-terminal regions and evaluated their ability to inhibit the colony formation of several cancer cell lines. Our findings demonstrated that truncated KL1 1-340 (KL340) effectively inhibited colony formation similar to KL1, while truncated KL1 1-320 (KL320) lost this activity. Furthermore, this correlated with the inhibitory effect of KL1 and KL340 on the Wnt/ß-catenin pathway, whereas KL320 had no effect. Transcriptomic analysis of MCF-7 cells expressing the constructs revealed enriched pathways associated with tumor suppressor activity in KL1 and KL340. Interestingly, the α-fold predictor tool highlighted distinct differences in the α and ß sheets of the TIM barrel fold of the truncated Klotho constructs, adding to our understanding of their structural variations. In summary, this study identified the 340 N-terminal amino acids as the sequence that possesses Klotho's tumor suppressor activity and reveals a critical role in the 320-340 sequence for this function. It also provides a foundation for the development of Klotho-based therapeutic approaches for cancer treatment.


Subject(s)
Gene Expression Profiling , Glucuronidase , Humans , Glucuronidase/genetics , Glucuronidase/metabolism , MCF-7 Cells , Hormones
4.
Oncogene ; 41(40): 4487-4497, 2022 09.
Article in English | MEDLINE | ID: mdl-36038662

ABSTRACT

Klotho is a well-established longevity hormone. Its most prominent function is the regulation of phosphate homeostasis. However, klotho possesses multiple pleiotropic activities, including inhibition of major signaling pathways, reducing oxidative stress and suppressing inflammation. These activities are tightly associated with cancer, and klotho was discovered as a universal tumor suppressor. We review here novel molecular aspects of klotho activity in cancer, focusing on its structure-function relationships and clinical aspects regarding its expression, blood levels, clinical risk, and prognostic value in the clinical setting. In addition, the potential benefit of klotho treatment combined with chemotherapy, biological therapy, or immunotherapy, are discussed. Finally, as klotho was shown in preclinical models to inhibit cancer development and growth, we discuss various approaches to developing klotho-based therapies.


Subject(s)
Glucuronidase , Neoplasms , Glucuronidase/genetics , Glucuronidase/metabolism , Hormones , Humans , Klotho Proteins , Neoplasms/genetics , Neoplasms/therapy , Phosphates/metabolism
5.
J Natl Cancer Inst ; 114(2): 203-209, 2022 Feb 07.
Article in English | MEDLINE | ID: mdl-34453830

ABSTRACT

BACKGROUND: Activity and safety of the SARS-CoV-2 BNT162b2 vaccine in actively treated patients with solid tumors is currently unknown. METHODS: We conducted a retrospective study of 326 patients with solid tumors treated with anticancer medications to determine the proportion of cancer patients with immunogenicity against SARS-CoV-2 following 2 doses of the BNT162b2 vaccine. The control group comprised 164 vaccinated healthy adults. Anti-SARS-CoV-2 S immunoglobulin G antibodies were measured using a level greater than 50 AU/mL as a cutoff for seropositivity. Information on adverse effects was collected using a questionnaire. All statistical tests were 2-sided. RESULTS: Most patients (205, 62.9%) were treated with chemotherapy either alone or with additional therapy; 55 (16.9%) were treated with immune checkpoint inhibitors and 38 (11.7%) with targeted therapy alone; 28 (8.6%) received other combinations. The vaccine was well tolerated, and no severe side effects were reported. Among patients with cancer, 39 (11.9%) were seronegative compared with 5 (3.0%) of the control group (P = .001). Median immunoglobulin G titers were statistically significantly lower among patients with cancer compared with control (931 AU/mL vs 2817 AU/mL, P = .003). Seronegativity proportions were higher in the chemotherapy-treated group (n = 19; 18.8%) compared with the immune checkpoint inhibitor-treated patients (n = 5; 9.1%) and with those treated with targeted therapy (n = 1; 2.6%) (P = .02). Titers were also statistically significantly different among treatment types (P = .002). CONCLUSIONS: The BNT162b2 vaccine is safe and effective in actively treated patients with cancer. The relatively lower antibody titers and lower proportion of seropositive patients, especially among chemotherapy-treated patients, call for continuing the use of personal protective measures in these patients, even following vaccination.


Subject(s)
COVID-19 , Neoplasms , Adult , Antibodies, Viral , BNT162 Vaccine , COVID-19 Vaccines , Humans , Immunogenicity, Vaccine , Neoplasms/drug therapy , Prospective Studies , RNA, Messenger , Retrospective Studies , SARS-CoV-2
7.
Mol Cancer Res ; 13(10): 1398-407, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26113466

ABSTRACT

UNLABELLED: Klotho is a transmembrane protein containing two internal repeats, KL1 and KL2, both displaying significant homology to members of the ß-glycosidase family. Klotho is expressed in the kidney, brain, and various endocrine tissues, but can also be cleaved and act as a circulating hormone. Klotho is an essential cofactor for binding of fibroblast growth factor 23 (FGF23) to the FGF receptor and can also inhibit the insulin-like growth factor-1 (IGF-1) pathway. Data from a wide array of malignancies indicate klotho as a tumor suppressor; however, the structure-function relationships governing its tumor suppressor activities have not been deciphered. Here, the tumor suppressor activities of the KL1 and KL2 domains were examined. Overexpression of either klotho or KL1, but not of KL2, inhibited colony formation by MCF-7 and MDA-MB-231 cells. Moreover, in vivo administration of KL1 was not only well tolerated but significantly slowed tumor formation in nude mice. Further studies indicated that KL1, but not KL2, interacted with the IGF-1R and inhibited the IGF-1 pathway. Based on computerized structural modeling, klotho constructs were generated in which critical amino acids have been mutated. Interestingly, the mutated proteins retained their tumor suppressor activity but showed reduced ability to modulate FGF23 signaling. These data indicate differential activity of the klotho domains, KL1 and KL2, in breast cancer and reveal that the tumor suppressor activities of klotho can be dissected from its physiologic activities. IMPLICATIONS: These findings pave the way for a rational design of safe klotho-based molecules for the treatment of breast cancer.


Subject(s)
Breast Neoplasms/metabolism , Glucuronidase/metabolism , Animals , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Female , Fibroblast Growth Factor-23 , Glucuronidase/chemistry , Glucuronidase/genetics , HEK293 Cells , Heterografts , Humans , Klotho Proteins , MCF-7 Cells , Male , Mice , Mice, Nude , Models, Molecular , Signal Transduction , Structure-Activity Relationship , Transfection
8.
Sci Transl Med ; 5(185): 185ra62, 2013 May 15.
Article in English | MEDLINE | ID: mdl-23677591

ABSTRACT

Premature ovarian failure and infertility are major side effects of chemotherapy treatments in young cancer patients. A more thorough understanding of the mechanism behind chemotherapy-induced follicle loss is necessary to develop new methods to preserve fertility in these patients. We show that the alkylating agent cyclophosphamide (Cy) activates the growth of the quiescent primordial follicle population in mice, resulting in loss of ovarian reserve. Despite the initial massive apoptosis observed in growing, though not in resting, follicles of Cy-treated mice, differential follicle counts demonstrated both a decrease in primordial follicles and an increase in early growing follicles. Immunohistochemistry showed that granulosa cells were undergoing proliferation. Analysis of the phosphatidylinositol 3-kinase signaling pathway demonstrated that Cy increased phosphorylation of proteins that stimulate follicle activation in the oocytes and granulosa cells. Coadministration of an immunomodulator, AS101, reduced follicle activation, thereby increasing follicle reserve and rescuing fertility after Cy, and also increased the efficacy of Cy against breast cancer cell lines. These findings suggest that the mechanism in Cy-induced loss of ovarian reserve is accelerated primordial follicle activation, which results in a "burnout" effect and follicle depletion. By preventing this activation, AS101 shows potential as an ovarian-protective agent, which may be able to preserve fertility in female cancer patients.


Subject(s)
Cyclophosphamide/adverse effects , Ethylenes/pharmacology , Fertility/drug effects , Ovarian Follicle/pathology , Animals , Anti-Mullerian Hormone/blood , Apoptosis/drug effects , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Cell Line, Tumor , Enzyme Activation/drug effects , Ethylenes/therapeutic use , Female , Mice , Mice, Inbred BALB C , Models, Biological , Ovarian Follicle/drug effects , Ovarian Follicle/enzymology , Ovarian Follicle/growth & development , PTEN Phosphohydrolase/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/drug effects
9.
Breast Cancer Res Treat ; 132(2): 449-61, 2012 Apr.
Article in English | MEDLINE | ID: mdl-21638053

ABSTRACT

The incretin hormone glucagon-like peptide (GLP)-1 is secreted from intestinal L cells in response to food intake, and promotes insulin secretion and pancreatic ß-cell proliferation. Reduced GLP-1 levels are observed in obesity and type 2 diabetes mellitus (T2DM) and are associated with reduced insulin secretion and increased insulin resistance. GLP-1 mediates its activities through activation of a G-protein coupled receptor, which is expressed in the pancreas, as well as other tissues. Long-acting GLP-1 receptor (GLP-1R) agonists, such as exendin-4, are currently approved for the treatment of T2DM. As obesity and T2DM are associated with increased risk of breast cancer, we aimed to explore the effects of GLP-1 and exendin-4, on breast cancer cells. Treatment with GLP-1 or exendin-4 reduced viability and enhanced apoptosis of breast cancer cells but did not affect viability of nontumorigenic cells. Moreover, exendin-4 attenuated tumor formation by breast cancer cells in athymic mice. Treatment with either GLP-1 or exendin-4 elevated cAMP levels, activated the down-stream target CREB, and enhanced CRE promoter transcription, in breast cancer cells. Moreover, inhibition of exendin-4-induced adenylate cyclase activation restored cell viability, thus suggesting cAMP as a principle mediator of exendin-4 anti-tumorigenic activity. While the pancreatic form of the GLP-1R could not be detected in breast cancer cells, several lines of evidence indicated the existence of an alternative GLP-1R in mammary cells. Thus, internalization of GLP-1 into MCF-7 cells was evidenced, infection of MCF-7 cells with the pancreatic receptor enhanced proliferation, and treatment with exendin-(9-39), a GLP-1R antagonist, further increased cAMP levels. Our studies indicate the incretin hormone GLP-1 as a potent inducer of cAMP and an inhibitor of breast cancer cell proliferation. Reduced GLP-1 levels may, therefore, serve as a novel link between obesity, diabetes mellitus, and breast cancer.


Subject(s)
Antineoplastic Agents/pharmacology , Breast Neoplasms/drug therapy , Cell Proliferation/drug effects , Cyclic AMP/metabolism , Glucagon-Like Peptide 1/pharmacology , Peptides/pharmacology , Signal Transduction/drug effects , Venoms/pharmacology , Adenylyl Cyclase Inhibitors , Adenylyl Cyclases/metabolism , Animals , Apoptosis/drug effects , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Survival/drug effects , Cyclic AMP Response Element-Binding Protein/metabolism , Dose-Response Relationship, Drug , Enzyme Activation , Enzyme Activators/pharmacology , Enzyme Inhibitors/pharmacology , Exenatide , Female , Gene Expression Regulation, Neoplastic/drug effects , Glucagon-Like Peptide-1 Receptor , Humans , Mice , Mice, Nude , Receptors, Glucagon/agonists , Receptors, Glucagon/genetics , Receptors, Glucagon/metabolism , Response Elements/drug effects , Time Factors , Transcriptional Activation/drug effects , Transfection , Up-Regulation , Xenograft Model Antitumor Assays , p38 Mitogen-Activated Protein Kinases/metabolism
10.
Clin Cancer Res ; 17(13): 4254-66, 2011 Jul 01.
Article in English | MEDLINE | ID: mdl-21571866

ABSTRACT

PURPOSE: Klotho is a transmembrane protein which can be shed, act as a circulating hormone and modulate the insulin-like growth factor (IGF)-I and the fibroblast growth factor (FGF) pathways. We have recently identified klotho as a tumor suppressor in breast cancer. Klotho is expressed in the normal pancreas and both the IGF-I and FGF pathways are involved in pancreatic cancer development. We, therefore, undertook to study the expression and activity of klotho in pancreatic cancer. EXPERIMENTAL DESIGN: Klotho expression was studied using immunohistochemistry and quantitative RT-PCR. Effects of klotho on cell growth were assessed in the pancreatic cancer cells Panc1, MiaPaCa2, and Colo357, using colony and MTT assays and xenograft models. Signaling pathway activity was measured by Western blotting. RESULTS: Klotho expression is downregulated in pancreatic adenocarcinoma. Overexpression of klotho, or treatment with soluble klotho, reduced growth of pancreatic cancer cells in vitro and in vivo, and inhibited activation of the IGF-I and the bFGF pathways. KL1 is a klotho subdomain formed by cleavage or alternative splicing. Compared with the full-length protein, KL1 showed similar growth inhibitory activity but did not promote FGF23 signaling. Thus, its administration to mice showed favorable safety profile. CONCLUSIONS: These studies indicate klotho as a potential tumor suppressor in pancreatic cancer, and suggest, for the first time, that klotho tumor suppressive activities are mediated through its KL1 domain. These results suggest the use of klotho or KL1 as potential strategy for the development of novel therapeutic interventions for pancreatic cancer.


Subject(s)
Adenocarcinoma , Fibroblast Growth Factor 2/antagonists & inhibitors , Glucuronidase/metabolism , Insulin-Like Growth Factor I/antagonists & inhibitors , Pancreatic Neoplasms , Signal Transduction/genetics , Tumor Suppressor Proteins/metabolism , Adenocarcinoma/genetics , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Animals , Cell Line, Tumor , Cell Survival/genetics , Down-Regulation/genetics , Female , Fibroblast Growth Factor 2/genetics , Fibroblast Growth Factor-23 , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Glucuronidase/genetics , HEK293 Cells , Humans , Insulin-Like Growth Factor I/genetics , Klotho Proteins , Mice , Mice, Nude , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Tumor Suppressor Proteins/genetics
11.
Proc Natl Acad Sci U S A ; 103(23): 8628-33, 2006 Jun 06.
Article in English | MEDLINE | ID: mdl-16731619

ABSTRACT

In Alzheimer's disease, both acetylcholinesterase (AChE) and butyrylcholinesterase (BChE) colocalize with brain fibrils of amyloid-beta (Abeta) peptides, and synaptic AChE-S facilitates fibril formation by association with insoluble Abeta fibrils. Here, we report that human BChE and BSP41, a synthetic peptide derived from the BChE C terminus, inversely associate with the soluble Abeta conformers and delay the onset and decrease the rate of Abeta fibril formation in vitro, at a 1:100 BChE/Abeta molar ratio and in a dose-dependent manner. The corresponding AChE synthetic peptide (ASP)40 peptide, derived from the homologous C terminus of synaptic human (h)AChE-S, failed to significantly affect Abeta fibril formation, attributing the role of enhancing this process to an AChE domain other than the C terminus. Circular dichroism and molecular modeling confirmed that both ASP40 and BChE synthetic peptide (BSP)41 are amphipathic alpha-helices. However, ASP40 shows symmetric amphipathicity, whereas BSP41 presented an aromatic tryptophan residue in the polar side of the C terminus. That this aromatic residue is causally involved in the attenuating effect of BChE was further supported by mutagenesis experiments in which (W8R) BSP41 showed suppressed capacity to attenuate fibril formation. In Alzheimer's disease, BChE may have thus acquired an inverse role to that of AChE by adopting imperfect amphipathic characteristics of its C terminus.


Subject(s)
Amyloid/chemistry , Amyloid/metabolism , Butyrylcholinesterase/metabolism , Acetylcholinesterase/chemistry , Amino Acid Sequence , Amyloid/biosynthesis , Butyrylcholinesterase/chemistry , Conserved Sequence , Humans , Kinetics , Models, Molecular , Molecular Sequence Data , Peptides/chemistry , Solubility , Structure-Activity Relationship
SELECTION OF CITATIONS
SEARCH DETAIL
...