Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Nat Commun ; 12(1): 3898, 2021 06 23.
Article in English | MEDLINE | ID: mdl-34162854

ABSTRACT

One topical area of supramolecular chemistry is the binding of anionic species but despite the importance of anions in diverse cellular processes and for cancer development, anion receptors or 'binders' have received little attention as potential anti-cancer therapeutics. Here we report self-assembling trimetallic cryptands (e.g. [L2(Metal)3]6+ where Metal = Cu2+, Zn2+ or Mn2+) which can encapsulate a range of anions and which show metal-dependent differences in chemical and biological reactivities. In cell studies, both [L2Cu3]6+ and [L2Zn3]6+ complexes are highly toxic to a range of human cancer cell lines and they show significant metal-dependent selective activity towards cancer cells compared to healthy, non-cancerous cells (by up to 2000-fold). The addition of different anions to the complexes (e.g. PO43-, SO42- or PhOPO32-) further alters activity and selectivity allowing the activity to be modulated via a self-assembly process. The activity is attributed to the ability to either bind or hydrolyse phosphate esters and mechanistic studies show differential and selective inhibition of multiple kinases by both [L2Cu3]6+ and [L2Zn3]6+ complexes but via different mechanisms.


Subject(s)
Anions/chemistry , Antineoplastic Agents/chemistry , Coordination Complexes/chemistry , Metals/chemistry , A549 Cells , Antineoplastic Agents/pharmacology , Autophagy/drug effects , Blotting, Western , Cell Line , Cell Line, Tumor , Cell Survival/drug effects , Coordination Complexes/pharmacology , Crystallography, X-Ray , HCT116 Cells , HT29 Cells , Humans , Inhibitory Concentration 50 , Neoplasms/metabolism , Neoplasms/pathology , Phosphotransferases/antagonists & inhibitors , Phosphotransferases/metabolism
2.
Biochem Soc Trans ; 48(3): 733-744, 2020 06 30.
Article in English | MEDLINE | ID: mdl-32573651

ABSTRACT

Nicotinamide adenine dinucleotide (NAD+) and its reduced form NADH are essential coupled redox metabolites that primarily promote cellular oxidative (catabolic) metabolic reactions. This enables energy generation through glycolysis and mitochondrial respiration to support cell growth and survival. In addition, many key enzymes that regulate diverse cell functions ranging from gene expression to proteostasis require NAD+ as a co-substrate for their catalytic activity. This includes the NAD+-dependent sirtuin family of protein deacetylases and the PARP family of DNA repair enzymes. Whilst their vital activity consumes NAD+ which is cleaved to nicotinamide, several pathways exist for re-generating NAD+ and sustaining NAD+ homeostasis. However, there is growing evidence of perturbed NAD+ homeostasis and NAD+-regulated processes contributing to multiple disease states. NAD+ levels decline in the human brain and other organs with age and this is associated with neurodegeneration and other age-related diseases. Dietary supplementation with NAD+ precursors is being investigated to counteract this. Paradoxically, many cancers have increased dependency on NAD+. Clinical efforts to exploit this have so far shown limited success. Emerging new opportunities to exploit dysregulation of NAD+ metabolism in cancers are critically discussed. An update is also provided on other key NAD+ research including perturbation of the NAD+ salvage enzyme NAMPT in the context of the tumour microenvironment (TME), methodology to study subcellular NAD+ dynamics in real-time and the regulation of differentiation by competing NAD+ pools.


Subject(s)
Aging , Cytokines/metabolism , NAD/metabolism , Neoplasms/metabolism , Nicotinamide Phosphoribosyltransferase/metabolism , Oxidation-Reduction , Animals , Catalysis , Gene Expression Profiling , Glycolysis , Humans , Lactate Dehydrogenase 5/metabolism , Mitochondria/metabolism , Neoplasms/drug therapy , Poly (ADP-Ribose) Polymerase-1/metabolism , Signal Transduction , Sirtuins/metabolism , Tumor Microenvironment
3.
J Cell Sci ; 132(19)2019 10 10.
Article in English | MEDLINE | ID: mdl-31515278

ABSTRACT

Tumor stem cells and malignant multicellular networks have been separately implicated in the therapeutic resistance of glioblastoma multiforme (GBM), the most aggressive type of brain cancer in adults. Here, we show that small-molecule inhibition of RHO-associated serine/threonine kinase proteins (ROCKi) significantly promoted the outgrowth of neurite-like cell projections in cultures of heterogeneous patient-derived GBM stem-like cells. These projections formed de novo-induced cellular network (iNet) 'webs', which regressed after withdrawal of ROCKi. Connected cells within the iNet web exhibited long range Ca2+ signal transmission, and significant lysosomal and mitochondrial trafficking. In contrast to their less-connected vehicle control counterparts, iNet cells remained viable and proliferative after high-dose radiation. These findings demonstrate a link between ROCKi-regulated cell projection dynamics and the formation of radiation-resistant multicellular networks. Our study identifies means to reversibly induce iNet webs ex vivo, and may thereby accelerate future studies into the biology of GBM cellular networks.


Subject(s)
Glioblastoma/metabolism , Neoplastic Stem Cells/metabolism , Neurites/metabolism , Calcium Signaling/physiology , Cell Line, Tumor , Cell Movement/physiology , Humans , Immunoblotting , Lysosomes/metabolism , Mitochondria/metabolism , Neuronal Outgrowth/physiology , Phenotype , Protein Serine-Threonine Kinases/metabolism
4.
Medchemcomm ; 10(9): 1620-1634, 2019 Sep 01.
Article in English | MEDLINE | ID: mdl-32952999

ABSTRACT

Traditional cytotoxic agents which act through a DNA-alkylating mechanism are relatively non-specific, resulting in a small therapeutic window and thus limiting their effectiveness. In this study, we evaluate a panel of 24 non-alkylating Strathclyde Minor Groove Binders (S-MGBs), including 14 novel compounds, for in vitro anti-cancer activity against a human colon carcinoma cell line, a cisplatin-sensitive ovarian cancer cell line and a cisplatin-resistant ovarian cancer cell line. A human non-cancerous retinal epithelial cell line was used to measure selectivity of any response. We have identified several S-MGBs with activities comparable to cis-platin and carboplatin, but with better in vitro selectivity indices, particularly S-MGB-4, S-MGB-74 and S-MGB-317. Moreover, a comparison of the cis-platin resistant and cis-platin sensitive ovarian cancer cell lines reveals that our S-MGBs do not show cross resistance with cisplatin or carboplatin and that they likely have a different mechanism of action. Finally, we present an initial investigation into the mechanism of action of one compound from this class, S-MGB-4, demonstrating that neither DNA double strand breaks nor the DNA damage stress sensor protein p53 are induced. This indicates that our S-MGBs are unlikely to act through an alkylating or DNA damage response mechanism.

5.
Sci Transl Med ; 10(454)2018 08 15.
Article in English | MEDLINE | ID: mdl-30111643

ABSTRACT

Pharmacological inhibition of uncontrolled cell growth with small-molecule inhibitors is a potential strategy for treating glioblastoma multiforme (GBM), the most malignant primary brain cancer. We showed that the synthetic small-molecule KHS101 promoted tumor cell death in diverse GBM cell models, independent of their tumor subtype, and without affecting the viability of noncancerous brain cell lines. KHS101 exerted cytotoxic effects by disrupting the mitochondrial chaperone heat shock protein family D member 1 (HSPD1). In GBM cells, KHS101 promoted aggregation of proteins regulating mitochondrial integrity and energy metabolism. Mitochondrial bioenergetic capacity and glycolytic activity were selectively impaired in KHS101-treated GBM cells. In two intracranial patient-derived xenograft tumor models in mice, systemic administration of KHS101 reduced tumor growth and increased survival without discernible side effects. These findings suggest that targeting of HSPD1-dependent metabolic pathways might be an effective strategy for treating GBM.


Subject(s)
Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Energy Metabolism , Glioblastoma/metabolism , Glioblastoma/pathology , Thiazoles/pharmacology , Animals , Apoptosis/drug effects , Autophagy/drug effects , Brain Neoplasms/genetics , Cell Line, Tumor , Cell Proliferation/drug effects , Chaperonin 60/metabolism , Citric Acid Cycle/drug effects , Disease Models, Animal , Energy Metabolism/drug effects , Glioblastoma/genetics , Glycolysis/drug effects , Humans , Metabolic Networks and Pathways/drug effects , Mitochondria/drug effects , Mitochondria/metabolism , Mitochondrial Proteins/metabolism , Neoplasm Invasiveness , Stress, Physiological/drug effects , Survival Analysis , Transcription, Genetic/drug effects , Xenograft Model Antitumor Assays
6.
Angew Chem Int Ed Engl ; 57(31): 9799-9804, 2018 07 26.
Article in English | MEDLINE | ID: mdl-29863754

ABSTRACT

The ligands L1 and L2 both form separable dinuclear double-stranded helicate and mesocate complexes with RuII . In contrast to clinically approved platinates, the helicate isomer of [Ru2 (L1 )2 ]4+ was preferentially cytotoxic to isogenic cells (HCT116 p53-/- ), which lack the critical tumour suppressor gene. The mesocate isomer shows the reverse selectivity, with the achiral isomer being preferentially cytotoxic towards HCT116 p53+/+ . Other structurally similar RuII -containing dinuclear complexes showed very little cytotoxic activity. This study demonstrates that alterations in ligand or isomer can have profound effects on cytotoxicity towards cancer cells of different p53 status and suggests that selectivity can be "tuned" to either genotype. In the search for compounds that can target difficult-to-treat tumours that lack the p53 tumour suppressor gene, [Ru2 (L1 )2 ]4+ is a promising compound for further development.


Subject(s)
Antineoplastic Agents/pharmacology , Colorectal Neoplasms/drug therapy , Organometallic Compounds/pharmacology , Ruthenium/pharmacology , Tumor Suppressor Protein p53/antagonists & inhibitors , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Cell Line, Tumor , Cell Proliferation/drug effects , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Drug Screening Assays, Antitumor , Humans , Molecular Structure , Organometallic Compounds/chemical synthesis , Organometallic Compounds/chemistry , Ruthenium/chemistry , Tumor Suppressor Protein p53/deficiency , Tumor Suppressor Protein p53/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL