Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
Recent Pat Anticancer Drug Discov ; 14(3): 258-267, 2019.
Article in English | MEDLINE | ID: mdl-31538905

ABSTRACT

BACKGROUND: The high mobility group A proteins modulate the transcription of numerous genes by interacting with transcription factors and/or altering the structure of chromatin. These proteins are involved in both benign and malignant neoplasias as a result of several pathways. A large amount of benign human mesenchymal tumors has rearrangements of HMGA genes. On the contrary, malignant tumors show unarranged HMGA overexpression that is frequently and causally related to neoplastic cell transformation. Here, we review the function of the HMGA proteins in human neoplastic disorders, the pathways by which they contribute to carcinogenesis and the new patents focused on targeting HMGA proteins. OBJECTIVE: Current review was conducted to check the involvement of HMGA as a druggable target in cancer treatment. METHODS: We reviewed the most recent patents focused on targeting HMGA in cancer treatment analyzing patent literature published during the last years, including the World Intellectual Property Organization (WIPO®), United States Patent Trademark Office (USPTO®), Espacenet®, and Google Patents. RESULTS: HMGA proteins are intriguing targets for cancer therapy and are objects of different patents based on the use of DNA aptamers, inhibitors, oncolytic viruses, antisense molecules able to block their oncogenic functions. CONCLUSION: Powerful strategies able to selectively interfere with HMGA expression and function could represent a helpful approach in the development of new anti-cancer therapies.


Subject(s)
Aptamers, Nucleotide/pharmacology , HMGA Proteins/antagonists & inhibitors , Neoplasms/therapy , RNA, Small Interfering/pharmacology , Databases, Factual , HMGA Proteins/genetics , HMGA Proteins/metabolism , Humans , Molecular Targeted Therapy , Neoplasms/genetics , Neoplasms/metabolism , Neoplasms/pathology , Oncolytic Virotherapy , Patents as Topic , Transcription Factors/metabolism
2.
Reprod Biol Endocrinol ; 16(1): 25, 2018 Mar 20.
Article in English | MEDLINE | ID: mdl-29558965

ABSTRACT

BACKGROUND: Ovarian retinoid homeostasis plays an important role in the physiological function of the ovary. Retinol-binding protein 4 (RBP4) acts as the mediator for the systemic and intercellular transport of retinol and is heavily involved in cellular retinol influx, efflux, and exchange. However, the expression patterns and regulatory mechanisms of Rbp4 in the ovary remain unclear. METHODS: The expression pattern of ovarian Rbp4 was examined in immature mice during different developmental stages and in adult mice during different stages of the estrous cycle. The potential regulation and mechanisms of ovarian Rbp4 expression by estrogen and related gonadotropins in mouse ovaries were also investigated. RESULTS: The present study demonstrated that the ovarian expression of Rbp4 remained constant before puberty and increased significantly in the peripubertal period. In adult female mice, the expression of Rbp4 increased at proestrus and peaked at estrus at both the mRNA and protein levels. The protein distribution of RBP4 was mainly localized in the granulosa cell and theca cell layer in follicles. In addition, the expression of Rbp4 was significantly induced by follicle-stimulating hormone (FSH) or FSH + luteinizing hormone (LH) in combination in immature mouse (3 weeks old) ovaries in vivo and in granulosa cells cultured in vitro, both at the mRNA and protein levels. In contrast, treatment with LH or 17ß-estradiol did not exhibit any observable effects on ovarian Rbp4 expression. Transcription factors high-mobility group AT-hook 1 (HMGA1), steroidogenic factor 1 (SF-1), and liver receptor homolog 1 (LRH-1) (which have been previously shown to be involved in activation of Rbp4 transcription), also responded to FSH stimulation. In addition, H-89, an inhibitor of protein kinase A (PKA), and the depletion of HMGA1, SF-1, and LRH-1 by small interfering RNAs (siRNAs), resulted in a dramatic loss of the induction of Rbp4 expression by FSH at both the mRNA and protein levels. CONCLUSIONS: These data indicate that the dynamic expression of Rbp4 is mainly regulated by FSH through the cAMP-PKA pathway, involving transcriptional factors HMGA1, SF-1, and LRH-1, in the mouse ovary during different stages of development and the estrous cycle.


Subject(s)
Follicle Stimulating Hormone/pharmacology , Gene Expression/drug effects , Ovary/metabolism , Retinol-Binding Proteins, Plasma/genetics , Animals , Cells, Cultured , Cyclic AMP-Dependent Protein Kinases/antagonists & inhibitors , Estrous Cycle , Female , Granulosa Cells/chemistry , HMGA Proteins/antagonists & inhibitors , HMGA Proteins/physiology , Mice , Mice, Inbred BALB C , Ovary/growth & development , RNA, Messenger/analysis , RNA, Small Interfering/pharmacology , Receptors, Cytoplasmic and Nuclear/antagonists & inhibitors , Receptors, Cytoplasmic and Nuclear/physiology , Retinol-Binding Proteins, Plasma/analysis , Sexual Maturation , Steroidogenic Factor 1/antagonists & inhibitors , Steroidogenic Factor 1/physiology , Theca Cells/chemistry
3.
PLoS One ; 13(2): e0192882, 2018.
Article in English | MEDLINE | ID: mdl-29462157

ABSTRACT

The HGMA1 architectural transcription factor is highly overexpressed in many human cancers. Because HMGA1 is a hub for regulation of many oncogenes, its overexpression in cancer plays a central role in cancer progression and therefore HMGA1 is gaining increasing attention as a target for development of therapeutic approaches to suppress either its expression or action in cancer cells. We have developed the strategy of introducing decoy hyper binding sites for HMGA1 into the nucleus of cancer cells with the goal of competetively sequestering overexpressed HMGA1 and thus suppressing its oncogenic action. Towards achieving this goal, we have introduced an HMGA1 decoy hyper binding site composed of six copies of a high affinity HMGA1 binding site into the genome of the replication defective adenovirus serotype 5 genome and shown that the engineered virus effectively reduces the viability of human pancreatic and cancer cells. Here we report the first pre-clinical measures of toxicity and biodistribution of the engineered virus in C57BL/6J Black 6 mice. The immune response to exposure of the engineered virus was determined by assaying the serum levels of key cytokines, IL-6 and TNF-α. Toxicity due to exposure to the virus was determined by measuring the serum levels of the liver enzymes aspartate aminotransferase and alanine aminotransferase. Biodistribution was measured following direct injection into the pancreas or liver by quantifying viral loads in the pancreas, liver, spleen and brain.


Subject(s)
Adenoviridae/genetics , Genetic Engineering , Genetic Therapy/methods , HMGA Proteins/antagonists & inhibitors , Neoplasms/therapy , Animals , Binding Sites , Female , HEK293 Cells , HMGA Proteins/genetics , HMGA Proteins/metabolism , Humans , Interleukin-6/blood , Liver/virology , Mice, Inbred C57BL , Models, Animal , Pancreas/virology , Transaminases/blood , Tumor Necrosis Factor-alpha/blood , Viral Load
4.
J Biomol Struct Dyn ; 34(4): 877-91, 2016.
Article in English | MEDLINE | ID: mdl-26084422

ABSTRACT

High mobility group A1 (HMGA1), a non-histone chromosomal protein, is highly expressed in a wide range of human cancers including cervical, breast, and prostate cancers. Therefore, hmga1 gene is considered as an attractive potential target for anticancer drugs. We have chosen 27 bp DNA sequence from a regulatory region of hmga1 promoter and studied its interaction with adriamycin (ADM) and in vitro expression of HMGA1 in the presence of ADM in HeLa cell line. A variety of biophysical techniques were employed to understand the characteristics of [DNA-ADM] complex. Spectrophotometric titration data, DNA denaturation profiles, and quenching of fluorescence of ADM in the presence of DNA demonstrated a strong complexation between DNA and ADM with a high binding affinity (Ka) of 1.3 × 10(6) M(-1) and a stoichiometry of 1:3 (drug:nucleotide). The energetics of binding obtained from isothermal titration calorimetry and differential scanning calorimetry suggest the binding to be exothermic and enthalpy (∆H, -6.7 ± 2.4 kcal M(-1)) and entropy (TΔS, 18.5 ± 6.4 kcal M(-1)) driven (20°C), which is typical of intercalative mode of binding. Further, results on decreased expression (by ~70%) of HMGA1 both at mRNA and protein levels in association with the observed cell death (by ~75%) in HeLa cell line, clearly confirm that ADM does target hmga1; however, the effect of ADM on genes other than hmga1 either directly or via hmga1-mediated pathways cannot be ruled out in the observed cytotoxicity. Therefore, hmga1 in general and particularly the regulatory region is a promising target for therapeutic strategy in combating cancer.


Subject(s)
Antibiotics, Antineoplastic/chemistry , Doxorubicin/chemistry , HMGA Proteins/genetics , Regulatory Sequences, Nucleic Acid , Antibiotics, Antineoplastic/pharmacology , Calorimetry, Differential Scanning , Cell Death/genetics , Cell Line, Tumor , DNA/chemistry , DNA/metabolism , Doxorubicin/pharmacology , Female , Gene Expression Regulation, Neoplastic/drug effects , HMGA Proteins/antagonists & inhibitors , Humans , Molecular Structure , Nucleic Acid Conformation , Protein Binding , Structure-Activity Relationship , Thermodynamics , Transition Temperature , Uterine Cervical Neoplasms/drug therapy , Uterine Cervical Neoplasms/genetics , Uterine Cervical Neoplasms/metabolism
5.
J Biol Chem ; 285(51): 40012-8, 2010 Dec 17.
Article in English | MEDLINE | ID: mdl-20961861

ABSTRACT

High mobility group A1 (HMGA1) proteins belong to a group of architectural transcription factors that are overexpressed in a range of human malignancies, including pancreatic adenocarcinoma. They promote anchorage-independent growth and epithelial-mesenchymal transition and are therefore suggested as potential therapeutic targets. Employing in vitro selection techniques against a chosen fragment of HMGA1, we have generated biostable l-RNA oligonucleotides, so-called Spiegelmers, that specifically bind HMGA1b with low nanomolar affinity. We demonstrate that the best binding Spiegelmers, NOX-A50 and NOX-f33, compete HMGA1b from binding to its natural binding partner, AT-rich double-stranded DNA. We describe a formulation method based on polyplex formation with branched polyethylenimine for efficient delivery of polyethylene glycol-modified Spiegelmers and show improved tissue distribution and persistence in mice. In a xenograft mouse study using the pancreatic cancer cell line PSN-1, subcutaneous administration of 2 mg/kg per day NOX-A50 formulated in polyplexes showed an enhanced delivery of NOX-A50 to the tumor and a significant reduction of tumor volume. Our results demonstrate that intracellular targets can be successfully addressed with a Spiegelmer using polyethylenimine-based delivery and underline the importance of HMGA1 as a therapeutic target in pancreatic cancer.


Subject(s)
Adenocarcinoma/drug therapy , Aptamers, Nucleotide/pharmacology , Drug Delivery Systems , HMGA Proteins/antagonists & inhibitors , Pancreatic Neoplasms/drug therapy , Xenograft Model Antitumor Assays , Animals , Cell Line, Tumor , DNA, Neoplasm/metabolism , Dose-Response Relationship, Drug , HMGA Proteins/metabolism , Humans , Mice , Mice, Mutant Strains , Protein Binding
SELECTION OF CITATIONS
SEARCH DETAIL
...