Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 39
Filter
1.
J Med Chem ; 66(2): 1321-1338, 2023 01 26.
Article in English | MEDLINE | ID: mdl-36634271

ABSTRACT

Sulfated glycosaminoglycans (GAGs), or synthetic mimetics thereof, are not favorably viewed as orally bioavailable drugs owing to their high number of anionic sulfate groups. Devising an approach for oral delivery of such highly sulfated molecules would be very useful. This work presents the concept that conjugating cholesterol to synthetic sulfated GAG mimetics enables oral delivery. A focused library of sulfated GAG mimetics was synthesized and found to inhibit the growth of a colorectal cancer cell line under spheroid conditions with a wide range of potencies ( 0.8 to 46 µM). Specific analogues containing cholesterol, either alone or in combination with clinical utilized drugs, exhibited pronounced in vivo anticancer potential with intraperitoneal as well as oral administration, as assessed by ex vivo tertiary and quaternary spheroid growth, cancer stem cell (CSC) markers, and/or self-renewal factors. Overall, cholesterol derivatization of highly sulfated GAG mimetics affords an excellent approach for engineering oral activity.


Subject(s)
Glycosaminoglycans , Sulfates , Glycosaminoglycans/pharmacology , Glycosaminoglycans/metabolism , Neoplastic Stem Cells/metabolism , Biomimetics
2.
Angew Chem Int Ed Engl ; 61(49): e202211320, 2022 12 05.
Article in English | MEDLINE | ID: mdl-36205924

ABSTRACT

The insulin-like growth factor-1 receptor (IGF-1R) is a receptor tyrosine kinase (RTK) that plays critical roles in cancer. Microarray, computational, thermodynamic, and cellular imaging studies reveal that activation of IGF-1R by its cognate ligand IGF1 is inhibited by shorter, soluble heparan sulfate (HS) sequences (e.g., HS06), whereas longer polymeric chains do not inhibit the RTK, a phenomenon directly opposed to the traditional relationship known for GAG-protein systems. The inhibition arises from smaller oligosaccharides binding in a unique pocket in the IGF-1R ectodomain, which competes with the natural cognate ligand IGF1. This work presents a highly interesting observation on preferential and competing inhibition of IGF-1R by smaller sequences, whereas polysaccharides are devoid of this function. These insights will be of major value to glycobiologists and anti-cancer drug discoverers.


Subject(s)
Polysaccharides , Receptors, Somatomedin , Humans , Ligands , Neoplasms/metabolism , Signal Transduction , Receptors, Somatomedin/metabolism
3.
Methods Mol Biol ; 2303: 765-777, 2022.
Article in English | MEDLINE | ID: mdl-34626421

ABSTRACT

Cancer stem-like cells (CSC) have been implicated in resistance to conventional chemotherapy as well as invasion and metastasis resulting in tumor relapse in majority of epithelial cancers including colorectal cancer. Hence, targeting CSC by small molecules is likely to improve therapeutic outcomes. Glycosaminoglycans (GAGs) are long linear polysaccharide molecules with varying degrees of sulfation that allows specific GAG-protein interaction which plays a key role in regulating cancer hallmarks such as cellular growth, angiogenesis, and immune modulation. However, identifying selective CSC-targeting GAG mimetic has been marred by difficulties associated with isolating and enriching CSC in vitro. Herein, we discuss two distinct methods, spheroid growth and EMT-transformed cells, to enrich CSC and set up medium- and high-throughput screen to identify selective CSC-targeting agents.


Subject(s)
Neoplasms , Neoplastic Stem Cells , Cell Line, Tumor , Cell Proliferation , Epithelial-Mesenchymal Transition , Humans
4.
Neoplasia ; 23(3): 348-359, 2021 03.
Article in English | MEDLINE | ID: mdl-33640759

ABSTRACT

High-dose acetaminophen (AAP) with delayed rescue using n-acetylcysteine (NAC), the FDA-approved antidote to AAP overdose, has demonstrated promising antitumor efficacy in early phase clinical trials. However, the mechanism of action (MOA) of AAP's anticancer effects remains elusive. Using clinically relevant AAP concentrations, we evaluated cancer stem cell (CSC) phenotype in vitro and in vivo in lung cancer and melanoma cells with diverse driver mutations. Associated mechanisms were also studied. Our results demonstrated that AAP inhibited 3D spheroid formation, self-renewal, and expression of CSC markers when human cancer cells were grown in serum-free CSC media. Similarly, anti-CSC activity was demonstrated in vivo in xenograft models - tumor formation following in vitro treatment and ex-vivo spheroid formation following in vivo treatment. Intriguingly, NAC, used to mitigate AAP's liver toxicity, did not rescue cells from AAP's anti-CSC effects, and AAP failed to reduce glutathione levels in tumor xenograft in contrast to mice liver tissue suggesting nonglutathione-related MOA. In fact, AAP mediates its anti-CSC effect via inhibition of STAT3. AAP directly binds to STAT3 with an affinity in the low micromolar range and a high degree of specificity for STAT3 relative to STAT1. These findings have high immediate translational significance concerning advancing AAP with NAC rescue to selectively rescue hepatotoxicity while inhibiting CSCs. The novel mechanism of selective STAT3 inhibition has implications for developing rational anticancer combinations and better patient selection (predictive biomarkers) for clinical studies and developing novel selective STAT3 inhibitors using AAP's molecular scaffold.


Subject(s)
Acetaminophen/pharmacology , Antineoplastic Agents/pharmacology , Free Radicals/metabolism , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/metabolism , STAT3 Transcription Factor/antagonists & inhibitors , AC133 Antigen/metabolism , Acetaminophen/administration & dosage , Antineoplastic Agents/administration & dosage , Biomarkers, Tumor , Cell Line, Tumor , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Gene Knockdown Techniques , Humans , Interleukin-6/antagonists & inhibitors , Lung Neoplasms , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/metabolism
5.
Biochim Biophys Acta Rev Cancer ; 1874(2): 188424, 2020 12.
Article in English | MEDLINE | ID: mdl-32956765

ABSTRACT

The development of Castration-Resistant Prostate Cancer (CRPC) remains a major challenge in the treatment of this disease. While Androgen Deprivation Therapy (ADT) can result in tumor shrinkage, a primary response of Prostate Cancer (PCa) cells to ADT is a senescent growth arrest. As a response to cancer therapies, senescence has often been considered as a beneficial outcome due to its association with stable growth abrogation, as well as the potential for immune system activation via the Senescence-Associated Secretory Phenotype (SASP). However, there is increasing evidence that not only can senescent cells regain proliferative capacity, but that senescence contributes to deleterious effects of cancer chemotherapy, including disease recurrence. Notably, the preponderance of work investigating the consequences of therapy-induced senescence on tumor progression has been performed in non-PCa models. Here, we summarize the evidence that ADT promotes a senescent response in PCa and postulate mechanisms by which senescence may contribute to the development of castration-resistance. Primarily, we suggest that ADT-induced senescence may support CRPC development via escape from senescence, by cell autonomous-reprogramming, and by the formation of a pro-tumorigenic SASP. However, due to the scarcity of direct evidence from PCa models, the consequences of ADT-induced senescence outlined here remain speculative until the relationship between senescence and CRPC can be experimentally defined.


Subject(s)
Androgen Antagonists/adverse effects , Prostatic Neoplasms, Castration-Resistant/chemically induced , Cellular Reprogramming , Cellular Senescence , Disease Progression , Humans , Male , Prostatic Neoplasms, Castration-Resistant/pathology , Treatment Outcome
6.
Bioorg Chem ; 101: 103953, 2020 08.
Article in English | MEDLINE | ID: mdl-32474179

ABSTRACT

Curcumin and trans-cinnamaldehyde are acrolein-based Michael acceptor compounds that are commonly found in domestic condiments, and known to cause cancer cell death via redox mechanisms. Based on the structural features of these compounds we designed and synthesized several 2-cinnamamido-N-substituted-cinnamamide (bis-cinnamamide) compounds. One of the derivatives, (Z)-2-[(E)-cinnamamido]-3-phenyl-N-propylacrylamide 8 showed a moderate antiproliferative potency (HCT-116 cell line inhibition of 32.0 µM), no inhibition of normal cell lines C-166, and proven cellular activities leading to apoptosis. SAR studies led to more than 10-fold increase in activity. Our most promising compound, [(Z)-3-(1H-indol-3-yl)-N-propyl-2-[(E)-3-(thien-2-yl)propenamido)propenamide] 45 killed colon cancer cells at IC50 = 0.89 µM (Caco-2), 2.85 µM (HCT-116) and 1.65 µM (HT-29), while exhibiting much weaker potency on C-166 and BHK normal cell lines (IC50 = 71 µM and 77.6 µM, respectively). Cellular studies towards identifying the compounds mechanism of cytotoxic activities revealed that apoptotic induction occurs in part as a result of oxidative stress. Importantly, the compounds showed inhibition of cancer stem cells that are critical for maintaining the potential for self-renewal and stemness. The results presented here show discovery of covalently acting Michael addition compounds that potently kill cancer cells by a defined mechanism, with prominent selectivity profile over non-cancerous cell lines.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Cinnamates/pharmacology , Colonic Neoplasms/pathology , Oxidative Stress/drug effects , Cell Proliferation/drug effects , HCT116 Cells , Humans
7.
Carbohydr Polym ; 205: 385-391, 2019 Feb 01.
Article in English | MEDLINE | ID: mdl-30446119

ABSTRACT

Heparanase, an endo-ß-D-glucuronidase, cleaves cell surface and extracellular matrix heparan sulfate (HS) chains and plays important roles in cellular growth and metastasis. Heparanase assays reported to-date are labor intensive, complex and/or expensive. A simpler assay is critically needed to understand the myriad roles of heparanase. We reasoned that fluorescent heparin could serve as an effective probe of heparanase levels. Following synthesis and screening, a heparin preparation labeled with DABCYL and EDANS was identified, which exhibited a characteristic increase in signal following cleavage by human heparanase. This work describes the synthesis of this heparin substrate, its kinetic and spectrofluorometric properties, optimization of the heparanase assay, use of the assay in inhibitor screening, and elucidation of the state of heparanase in different cell lines. Our FRET-based assay is much simpler and more robust than all assays reported in the literature as well as a commercially available kit.


Subject(s)
Fluorescent Dyes/chemistry , Glucuronidase/chemistry , Heparin/analogs & derivatives , Heparin/chemistry , Naphthalenesulfonates/chemistry , p-Dimethylaminoazobenzene/analogs & derivatives , Animals , Enzyme Assays , Fluorescence Resonance Energy Transfer/methods , HEK293 Cells , Heparin/chemical synthesis , Humans , MCF-7 Cells , Naphthalenesulfonates/chemical synthesis , Sf9 Cells , Spodoptera , p-Dimethylaminoazobenzene/chemical synthesis , p-Dimethylaminoazobenzene/chemistry
8.
Mol Cancer Ther ; 18(1): 51-61, 2019 01.
Article in English | MEDLINE | ID: mdl-30337351

ABSTRACT

Targeting of cancer stem cells (CSC) is expected to be a paradigm-shifting approach for the treatment of cancers. Cell surface proteoglycans bearing sulfated glycosaminoglycan (GAG) chains are known to play a critical role in the regulation of stem cell fate. Here, we show for the first time that G2.2, a sulfated nonsaccharide GAG mimetic (NSGM) of heparin hexasaccharide, selectively inhibits colonic CSCs in vivo G2.2-reduced CSCs (CD133+/CXCR4+, Dual hi) induced HT-29 and HCT 116 colon xenografts' growth in a dose-dependent fashion. G2.2 also significantly delayed the growth of colon xenograft further enriched in CSCs following oxaliplatin and 5-fluorouracil treatment compared with vehicle-treated xenograft controls. In fact, G2.2 robustly inhibited CSCs' abundance (measured by levels of CSC markers, e.g., CD133, DCMLK1, LGR5, and LRIG1) and self-renewal (quaternary spheroids) in colon cancer xenografts. Intriguingly, G2.2 selectively induced apoptosis in the Dual hi CSCs in vivo eluding to its CSC targeting effects. More importantly, G2.2 displayed none to minimal toxicity as observed through morphologic and biochemical studies of vital organ functions, blood coagulation profile, and ex vivo analyses of normal intestinal (and bone marrow) progenitor cell growth. Through extensive in vitro, in vivo, and ex vivo mechanistic studies, we showed that G2.2's inhibition of CSC self-renewal was mediated through activation of p38α, uncovering important signaling that can be targeted to deplete CSCs selectively while minimizing host toxicity. Hence, G2.2 represents a first-in-class (NSGM) anticancer agent to reduce colorectal CSCs.


Subject(s)
Biomimetic Materials/administration & dosage , Colonic Neoplasms/drug therapy , Neoplastic Stem Cells/drug effects , p38 Mitogen-Activated Protein Kinases/metabolism , Animals , Biomimetic Materials/chemistry , Biomimetic Materials/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Colonic Neoplasms/metabolism , Dose-Response Relationship, Drug , HCT116 Cells , HT29 Cells , Heparin/chemistry , Humans , Mice , Neoplastic Stem Cells/metabolism , Signal Transduction/drug effects , Xenograft Model Antitumor Assays
9.
Oncotarget ; 9(65): 32408-32418, 2018 Aug 21.
Article in English | MEDLINE | ID: mdl-30197752

ABSTRACT

C-terminal binding protein 2 (CtBP2) drives intestinal polyposis in the Apcmin mouse model of human Familial Adenomatous Polyposis. As CtBP2 is targetable by an inhibitor of its dehydrogenase domain, understanding CtBP2's role in adenoma formation is necessary to optimize CtBP-targeted therapies in Apc mutated human neoplasia. Tumor initiating cell (TIC) populations were substantially decreased in ApcminCtbp2+/- intestinal epithelia. Moreover, normally nuclear Ctbp2 was mislocalized to the cytoplasm of intestinal crypt stem cells in Ctbp2+/- mice, both Apcmin and wildtype, correlating with low/absent CD133 expression in those cells, and possibly explaining the lower burden of polyps in Apcmin Ctbp2+/- mice. The CtBP inhibitor 4-chloro-hydroxyimino phenylpyruvate (4-Cl-HIPP) also robustly downregulated TIC populations and significantly decreased intestinal polyposis in Apcmin mice. We have therefore demonstrated a critical link between polyposis, intestinal TIC's and Ctbp2 gene dosage or activity, supporting continued efforts targeting CtBP in the treatment or prevention of Apc mutated neoplasia.

10.
PLoS One ; 12(2): e0171619, 2017.
Article in English | MEDLINE | ID: mdl-28182755

ABSTRACT

Glycosaminoglycans (GAGs) are key natural biopolymers that exhibit a range of biological functions including growth and differentiation. Despite this multiplicity of function, natural GAG sequences have not yielded drugs because of problems of heterogeneity and synthesis. Recently, several homogenous non-saccharide glycosaminoglycan mimetics (NSGMs) have been reported as agents displaying major therapeutic promise. Yet, it remains unclear whether sulfated NSGMs structurally mimic sulfated GAGs. To address this, we developed a three-step molecular dynamics (MD)-based algorithm to compare sulfated NSGMs with GAGs. In the first step of this algorithm, parameters related to the range of conformations sampled by the two highly sulfated molecules as free entities in water were compared. The second step compared identity of binding site geometries and the final step evaluated comparable dynamics and interactions in the protein-bound state. Using a test case of interactions with fibroblast growth factor-related proteins, we show that this three-step algorithm effectively predicts the GAG structure mimicking property of NSGMs. Specifically, we show that two unique dimeric NSGMs mimic hexameric GAG sequences in the protein-bound state. In contrast, closely related monomeric and trimeric NSGMs do not mimic GAG in either the free or bound states. These results correspond well with the functional properties of NSGMs. The results show for the first time that appropriately designed sulfated NSGMs can be good structural mimetics of GAGs and the incorporation of a MD-based strategy at the NSGM library screening stage can identify promising mimetics of targeted GAG sequences.


Subject(s)
Glycosaminoglycans/chemistry , Molecular Dynamics Simulation , Small Molecule Libraries/chemistry , Biomimetic Materials/chemistry , Sulfates/chemistry
11.
Oncotarget ; 7(51): 84608-84622, 2016 Dec 20.
Article in English | MEDLINE | ID: mdl-27705927

ABSTRACT

Heparan sulfate (HS) plays a role in the majority of essential hallmarks of cancer, yet its ability to modulate self-renewal, especially of cancer stem cells (CSCs), remains unknown. We have discovered that a non-anticoagulant HS hexasaccharide (HS06) sequence, but not other shorter or longer sequences, selectively inhibited CSC self-renewal and induced apoptosis in colorectal, pancreatic, and breast CSCs suggesting a very general phenomenon. HS06 inhibition of CSCs relied upon early and sustained activation of p38α/ß mitogen activated protein kinase (MAPK) but not other MAPKs family members i.e. ERK and JNK. In contrast, polymeric HS induced exactly opposite changes in MAPK activation and failed to inhibit CSCs. In fact, TCF4 signaling, a critical regulator of CSC self-renewal, was inhibited by HS06 in a p38 activation dependent fashion. In conclusion, HS06 selectively inhibits CSCs self-renewal by causing isoform specific activation of p38MAPK to inhibit TCF4 signaling. These observations on chain length-induced specificity carry major mechanistic implications with regard to HS in cancer biology, while also presenting a novel paradigm for developing novel anti-CSC hexasaccharides that prevent cancer relapse.Heparan sulfate (HS) of specific length, i.e., hexasaccharide (HS06), but not longer or shorter sequences, selectively inhibit cancer stem cells (CSCs) through isoform specific activation of p38 mitogen-activated protein kinase. These findings will have major implication for developing chemical probes to decipher complex signaling events that govern cancer stem cells. Additionally, there are direct implications for designing glycosaminoglycan based cancer therapies to selectively target CSCs that escape killing by traditional chemotherapy threatening cancer relapse.


Subject(s)
Breast Neoplasms/metabolism , Colorectal Neoplasms/metabolism , Heparitin Sulfate/metabolism , Neoplastic Stem Cells/physiology , Pancreatic Neoplasms/metabolism , Cell Self Renewal , Drug Discovery , Enzyme Activation , Female , Gene Expression Regulation, Neoplastic , Heparitin Sulfate/chemistry , Humans , Signal Transduction , Transcription Factor 4/metabolism , Tumor Cells, Cultured , p38 Mitogen-Activated Protein Kinases/metabolism
12.
Oncotarget ; 6(17): 15332-47, 2015 Jun 20.
Article in English | MEDLINE | ID: mdl-25895029

ABSTRACT

We observed a co-upregulation of the insulin-like growth factor receptor (IGF-1R)/AKT/mammalian target of rapamycin (mTOR) [InAT] axis and the mevalonate-isoprenoid biosynthesis (MIB) pathways in colorectal cancer stem cells (CSCs) in an unbiased approach. Hence, we hypothesized that the InAT axis might regulate the MIB pathway to govern colorectal CSCs growth. Stimulation (IGF-1) or inhibition (IGF-1R depletion and pharmacological inhibition of IGF-1R/mTOR) of the InAT axis produced induction or attenuation of CSC growth as well as expression of CSC markers and self-renewal factors respectively. Intriguingly, activation of the InAT axis (IGF-1) caused significant upregulation of the MIB pathway genes (both mRNA and protein); while its inhibition produced the opposite effects in colonospheres. More importantly, supplementation with dimethylallyl- and farnesyl-PP, MIB metabolites downstream of isopentenyl-diphosphate delta isomerase (IDI), but not mevalonate and isopentenyl-pp that are upstream of IDI, resulted in a near-complete reversal of the suppressive effect of the InAT axis inhibitors on CSCs growth. The latter findings suggest a specific regulation of the MIB pathway by the InAT axis distal to the target of statins that inhibit 3-hydroxy-3-methyl-glutaryl-CoA reductase (HMGCR). Effects of IGF-1R inhibition on colonic CSCs proliferation and the MIB pathway were confirmed in an 'in vivo' HCT-116 xenograft model. These observations establish a novel mechanistic link between the InAT axis that is commonly deregulated in colorectal cancer and the MIB pathway in regulation of colonic CSCs growth. Hence, the InAT-MIB corridor is a novel target for developing paradigm shifting optimum anti-CSCs therapies for colorectal cancer.


Subject(s)
Colorectal Neoplasms/pathology , Neoplastic Stem Cells/pathology , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Receptor, IGF Type 1/antagonists & inhibitors , TOR Serine-Threonine Kinases/antagonists & inhibitors , Animals , Apoptosis , Carbon-Carbon Double Bond Isomerases/genetics , Cell Proliferation/physiology , HCT116 Cells , Hemiterpenes/metabolism , Hemiterpenes/pharmacology , Humans , Mevalonic Acid/metabolism , Mice , Mice, Inbred NOD , Mice, SCID , Neoplasm Transplantation , Organophosphorus Compounds/metabolism , Organophosphorus Compounds/pharmacology , Polyisoprenyl Phosphates/metabolism , Polyisoprenyl Phosphates/pharmacology , Proto-Oncogene Proteins c-akt/metabolism , RNA, Messenger/biosynthesis , Receptor, IGF Type 1/biosynthesis , Sesquiterpenes/metabolism , Sesquiterpenes/pharmacology , Spheroids, Cellular , TOR Serine-Threonine Kinases/metabolism , Terpenes/metabolism , Transplantation, Heterologous , Tumor Cells, Cultured
13.
Cardiol Rev ; 23(1): 33-51, 2015.
Article in English | MEDLINE | ID: mdl-25275717

ABSTRACT

Pulmonary arterial hypertension (PAH) is a progressive disease that remains incurable. The past 2 decades have witnessed many advances in PAH-directed therapies. More recently, 3 new oral agents have become available in the United States within the past 2 years. Treprostinil is now available in extended-release oral tablets. Macitentan is the third endothelin receptor antagonist approved for use, demonstrating benefits on morbidity and mortality among patients with PAH in an event-driven study. Riociguat is the first soluble guanylate cyclase stimulator that has been approved for use in the United States. This article reviews the clinical efficacy and safety of these 3 agents and the roles they play in the management of PAH. Additionally, we review the limitations of using surrogate markers such as change in 6-minute walk distance to assess disease progression.


Subject(s)
Antihypertensive Agents/therapeutic use , Disease Progression , Drug Therapy/trends , Hypertension, Pulmonary/drug therapy , Hypertension, Pulmonary/physiopathology , Epoprostenol/analogs & derivatives , Epoprostenol/therapeutic use , Humans , Pyrazoles/therapeutic use , Pyrimidines/therapeutic use , Sulfonamides/therapeutic use , Time Factors , Treatment Outcome , Walking/physiology
14.
Methods Mol Biol ; 1229: 529-41, 2015.
Article in English | MEDLINE | ID: mdl-25325978

ABSTRACT

Cancer stem-like cells (CSC) have been implicated in resistance to conventional chemotherapy as well as invasion and metastasis resulting in tumor relapse in majority of epithelial cancers including colorectal cancer. Hence, targeting CSC by small molecules is likely to improve therapeutic outcomes. Glycosaminoglycans (GAGs) are long linear polysaccharide molecules with varying degrees of sulfation that allows specific GAG-protein interaction which plays a key role in regulating cancer hallmarks such as cellular growth, angiogenesis, and immune modulation. However, identifying selective CSC-targeting GAG mimetic has been marred by difficulties associated with isolating and enriching CSC in vitro. Herein, we discuss two distinct methods, spheroid growth and EMT-transformed cells, to enrich CSC and set up medium- and high-throughput screen to identify selective CSC-targeting agents.


Subject(s)
Antineoplastic Agents/pharmacology , Drug Evaluation, Preclinical/methods , Neoplastic Stem Cells/pathology , Biomarkers, Tumor/metabolism , Cadherins/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Flow Cytometry , Gene Knockdown Techniques , High-Throughput Screening Assays , Humans , Lentivirus/metabolism , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/metabolism , Spheroids, Cellular/drug effects , Spheroids, Cellular/pathology , Transduction, Genetic
15.
Cell Cycle ; 13(22): 3506-18, 2014.
Article in English | MEDLINE | ID: mdl-25483087

ABSTRACT

Selective targeting of cancer stem cells (CSCs), implicated in tumor relapse, holds great promise in the treatment of colorectal cancer. Overexpression of C-terminal binding protein (CtBP), an NADH dependent transcriptional regulator, is often observed in colon cancer. Of note, TCF-4 signaling is also up-regulated in colonic CSCs. We hypothesized that CtBP, whose dehydrogenase activity is amenable to pharmacological inhibition by 4-methylthio-2-oxobutyric acid (MTOB), positively regulates TCF-4 signaling, leading to CSC growth and self-renewal. CSCs demonstrated significant upregulation of CtBP1 and CtBP2 levels (mRNA and protein) and activity partly due to increased NADH/NAD ratio, as well as increased TCF/LEF transcriptional activity, compared to respective controls. Depletion of CtBP2 inhibited, while its overexpression enhanced, CSC growth (1° spheroids) and self-renewal (2°/3° spheroids). Similarly, MTOB caused a robust inhibition of spheroid growth and self-renewal in a dose dependent manner. MTOB displayed significantly greater selectivity for growth inhibition in the spheroids, at least in part through induction of apoptosis, compared to monolayer controls. Moreover, MTOB inhibited basal as well as induced (by GSK-3ß inhibitor) TCF/LEF activity while suppressing mRNA and protein levels of several ß-catenin target genes (CD44, Snail, C-MYC and LGR5). Lastly, CtBP physically interacted with TCF-4, and this interaction was significantly inhibited in the presence of MTOB. The above findings point to a novel role of CtBPs in the promotion of CSC growth and self-renewal through direct regulation of TCF/LEF transcription. Moreover, small molecular inhibition of its function can selectively target CSCs, presenting a novel approach for treatment of colorectal cancer focused on targeting of CSCs.


Subject(s)
Alcohol Oxidoreductases/metabolism , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Colonic Neoplasms/genetics , DNA-Binding Proteins/metabolism , Neoplastic Stem Cells/pathology , Transcription Factors/metabolism , Alcohol Oxidoreductases/genetics , Apoptosis/genetics , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/genetics , Colonic Neoplasms/pathology , DNA-Binding Proteins/genetics , Gene Expression Regulation, Neoplastic , Humans , Protein Binding/genetics , Signal Transduction/genetics , Transcription Factor 4 , Transcription Factors/genetics , beta Catenin/genetics
16.
World J Gastrointest Pathophysiol ; 5(3): 366-72, 2014 Aug 15.
Article in English | MEDLINE | ID: mdl-25133037

ABSTRACT

AIM: To demonstrated the combined effects of aging and carcinogen treatment on cancer stem/stem-like cells (CSCs) of gastric mucosa in an animal model. METHODS: In this study we investigated the effects of aging and Helicobacter pylori (H. pylori) inflammation as a model for inflammation induced carcinogenesis in human and rat gastric mucosa samples. In aging studies, we compared 4-mo old (young) with 22 mo (aged) old Fischer-344 rats. For human studies, gastric biopsies and resection specimens representing normal mucosa or different stages of H. pylori gastritis and gastric adenocarcinomas were used for determining the expression of stem cell markers CD166, ALDH1 and LGR5. In addition we performed immunofluorescent double labeling for B-catenin and Lgr5 in both rat and human gastric tissues to examine the status of Wnt signaling in these cells. RESULTS: CSC markers ALDH1, LGR5, and CD166 were expressed in very low levels in normal human gastric mucosa or young rat gastric mucosa. In contrast, level of expression for all three markers significantly increased in H. pylori gastritis and gastric adenocarcinomas as well as in normal gastric mucosa in aged rats. We also observed cytoplasmic B-catenin staining in both aged rat and human H. pylori inflamed gastric mucosa, which were found to be colocalized with Lgr5 immunoreactive cells. The increased number of ALDH1, CD166 and LGR5 positive cells in H. pylori gastritis indicates that increased number of stem-like cells in gastric mucosa is an early event, and may constitute an important step in the progression to neoplasia. CONCLUSION: Our observation of the age-related increase in cancer stem/stem-like cells in the gastric mucosa may explain the increased incidence of gastric cancer during aging. Combination of aging and H. pylori infection may have additive effects in progression to neoplasia.

17.
ACS Chem Biol ; 9(8): 1826-33, 2014 Aug 15.
Article in English | MEDLINE | ID: mdl-24968014

ABSTRACT

Selective targeting of cancer stem-like cells (CSCs) is a paradigm-shifting approach. We hypothesized that CSCs can be targeted by interfering with functions of sulfated glycosaminoglycans, which play key roles in cancer cell growth, invasion and metastasis. We developed a tandem, dual screen strategy involving (1) assessing inhibition of monolayer versus spheroid growth and (2) assessing inhibition of primary versus secondary spheroid growth to identify G2.2, a unique sulfated nonsaccharide GAG mimetic (NSGM) from a focused library of 53 molecules, as a selective inhibitor of colon CSCs. The NSGM down-regulated several CSC markers through regulation of gene transcription, while closely related, inactive NSGMs G1.4 and G4.1 demonstrated no such changes. G2.2's effects on CSCs were mediated, in part, through induction of apoptosis and inhibition of self-renewal factors. Overall, this work presents the proof-of-principle that CSCs can be selectively targeted through novel NSGMs, which are likely to advance fundamental understanding on CSCs while also aiding development of novel therapeutic agents.


Subject(s)
Colonic Neoplasms/pathology , Glycosaminoglycans/pharmacology , Molecular Mimicry , Neoplastic Stem Cells/drug effects , Cell Division/drug effects , Cell Line, Tumor , Humans , Neoplastic Stem Cells/pathology
18.
Carcinogenesis ; 33(1): 68-76, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22072622

ABSTRACT

Although microRNA-21 (miR-21) is emerging as an oncogene and has been shown to target several tumor suppressor genes, including programmed cell death 4 (PDCD4), its precise mechanism of action on cancer stem cells (CSCs) is unclear. Herein, we report that FOLFOX-resistant HCT-116 and HT-29 cells that are enriched in CSCs show a 3- to 7-fold upregulation of pre- and mature miR-21 and downregulation of PDCD4. Likewise, overexpression of miR-21 in HCT-116 cells, achieved through stable transfection, led to the downregulation of PDCD4 and transforming growth factor beta receptor 2 (TGFßR2). In contrast, the levels of ß-catenin, TCF/LEF activity and the expression of c-Myc, Cyclin-D, which are increased in CSCs, are also augmented in miR-21 overexpressing colon cancer cells, accompanied by an increased sphere forming ability in vitro and tumor formation in SCID mice. Downregulation of TGFßR2 could be attributed to decreased expression of the receptor as evidenced by reduction in the activity of the luciferase gene construct comprising TGFßR2-3' untranslated region (UTR) sequence that binds to miR-21. Moreover, we observed that downregulation of miR-21 enhances luciferase-TGFßR2-3' UTR activity suggesting TGFßR2 as being one of the direct targets of miR-21. Further support is provided by the observation that transfection of TGFßR2 in HCT-116 cells attenuates TCF/LEF luciferase activity, accompanied by decreased expression of ß-catenin, c-Myc and Cyclin-D1. Our current data suggest that miR-21 plays an important role in regulating stemness by modulating TGFßR2 signaling in colon cancer cells.


Subject(s)
Colonic Neoplasms/pathology , MicroRNAs/physiology , Neoplastic Stem Cells/pathology , Protein Serine-Threonine Kinases/physiology , Receptors, Transforming Growth Factor beta/physiology , Apoptosis Regulatory Proteins/physiology , Colonic Neoplasms/drug therapy , Down-Regulation , Drug Resistance, Neoplasm , HCT116 Cells , HT29 Cells , Humans , Protein Serine-Threonine Kinases/antagonists & inhibitors , RNA-Binding Proteins/physiology , Receptor, Transforming Growth Factor-beta Type II , Receptors, Transforming Growth Factor beta/antagonists & inhibitors , Signal Transduction , Wnt Signaling Pathway , beta Catenin/physiology
19.
Am J Physiol Gastrointest Liver Physiol ; 301(2): G347-55, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21596996

ABSTRACT

We have previously demonstrated that expression of the novel gene schlafen-3 (Slfn-3) correlates with intestinal epithelial cell differentiation (Patel VB, Yu Y, Das JK, Patel BB, Majumdar AP. Biochem Biophys Res Commun 388: 752-756, 2009). The present investigation was undertaken to examine whether Slfn-3 plays a role in regulating differentiation of FOLFOX-resistant (5-fluorouracil + oxaliplatin) colon cancer cells that are highly enriched in cancer stem cells (CSCs). Transfection of Slfn-3 in FOLFOX-resistant colon cancer HCT-116 cells resulted in increase of alkaline phosphatase activity, a marker of intestinal differentiation. Additionally, Slfn-3 transfection resulted in reduction of mRNA and protein levels of the CSC markers CD44, CD133, CD166, and aldehyde dehydrogenase 1 in both FOLFOX-resistant HCT-116 and HT-29 cells. This was accompanied by decreased formation of tumorosphere/colonosphere (an in vitro model of tumor growth) in stem cell medium and inhibition of expression of the chemotherapeutic drug transporter protein ABCG2. Additionally, Slfn-3 transfection of FOLFOX-resistant HCT-116 and HT-29 cells reduced Hoechst 33342 dye exclusion. Finally, Slfn-3 transfection inhibited the expression of transforming growth factor-α in both FOLFOX-resistant colon cancer cells, but stimulated apoptosis in response to additional FOLFOX treatment. In summary, our data demonstrate that Slfn-3 expression inhibits multiple characteristics of CSC-enriched, FOLFOX-resistant colon cancer cells, including induction of differentiation and reduction in tumorosphere/colonosphere formation, drug transporter activity, and autocrine stimulation of proliferation. Thus Slfn-3 expression may render colon CSCs more susceptible to cancer chemotherapeutics.


Subject(s)
Autocrine Communication/genetics , Cell Cycle Proteins/genetics , Colonic Neoplasms/drug therapy , Drug Resistance, Neoplasm/genetics , Neoplastic Stem Cells/metabolism , Proteins/genetics , Proteins/physiology , RNA, Messenger/metabolism , AC133 Antigen , ATP Binding Cassette Transporter, Subfamily G, Member 2 , ATP-Binding Cassette Transporters/metabolism , Aldehyde Dehydrogenase 1 Family , Antigens, CD/metabolism , Antineoplastic Combined Chemotherapy Protocols , Apoptosis , Cell Adhesion Molecules, Neuronal/metabolism , Cell Cycle Proteins/physiology , Cell Differentiation/genetics , Colonic Neoplasms/genetics , ErbB Receptors/metabolism , Fetal Proteins/metabolism , Fluorouracil , Glycoproteins/metabolism , HCT116 Cells , HT29 Cells , Humans , Hyaluronan Receptors/metabolism , Isoenzymes/metabolism , Leucovorin , Neoplasm Proteins/metabolism , Organoplatinum Compounds , Peptides/metabolism , Retinal Dehydrogenase/metabolism , Signal Transduction/genetics , Transfection , Transforming Growth Factor alpha/metabolism
20.
J Thorac Oncol ; 6(5): 927-33, 2011 May.
Article in English | MEDLINE | ID: mdl-21415776

ABSTRACT

BACKGROUND: Pemetrexed has emerged as one of the most active agents for the treatment of patients with advanced non-small cell lung cancer (NSCLC). We conducted a phase II study to assess the efficacy and feasibility of integrating pemetrexed in a concurrent therapy plan for patients with stage III NSCLC. METHODS: Patients with stage III NSCLC with performance status 0 to 1, adequate organ function including pulmonary function, and V20 less than 40% were eligible. Patients were treated with cisplatin 75 mg/m² (first five patients 60 mg/m²) and pemetrexed 500 mg/m² every 21 days for three cycles with chest radiotherapy to 66 Gy. Patients then received three cycles of docetaxel 75 mg/m² every 21 days. Tumors were analyzed for Excision Repair Cross Complementation Group 1 and thymidylate synthase. RESULTS: Patient characteristics (N = 28) were median age, 60; males, 68%; stage IIIB, 64%; and squamous cell, 43%. Twenty-four patients (86%) completed all three cycles of cisplatin/pemetrexed. Of the 24 patients eligible for docetaxel, 21 (87%) received it. Grade 3/4 toxicities were neutropenia (39%), febrile neutropenia (14%), esophagitis (14%), and pneumonitis (4%). Median survival was 34 months, and 1-year survival was 66%. Survival was not significantly different in squamous and other histology patients. Tumor analysis in 16 patients showed that moderate/strong expression of thymidylate synthase was significantly associated with progression-free survival and overall survival. CONCLUSION: Integrating pemetrexed in a concurrent therapy regimen for patients with stage III NSCLC is feasible and was associated with a median survival of 34 months.


Subject(s)
Adenocarcinoma/therapy , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Carcinoma, Large Cell/therapy , Carcinoma, Non-Small-Cell Lung/therapy , Carcinoma, Squamous Cell/therapy , Lung Neoplasms/therapy , Adenocarcinoma/drug therapy , Adenocarcinoma/radiotherapy , Adult , Aged , Aged, 80 and over , Carcinoma, Large Cell/drug therapy , Carcinoma, Large Cell/radiotherapy , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/radiotherapy , Carcinoma, Squamous Cell/drug therapy , Carcinoma, Squamous Cell/radiotherapy , Cisplatin/administration & dosage , Combined Modality Therapy , DNA-Binding Proteins/metabolism , Docetaxel , Endonucleases/metabolism , Feasibility Studies , Female , Follow-Up Studies , Glutamates/administration & dosage , Guanine/administration & dosage , Guanine/analogs & derivatives , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/radiotherapy , Male , Middle Aged , Neoplasm Recurrence, Local/drug therapy , Neoplasm Recurrence, Local/radiotherapy , Neoplasm Recurrence, Local/therapy , Neoplasm Staging , Pemetrexed , Survival Rate , Taxoids/administration & dosage , Thymidylate Synthase/metabolism , Treatment Outcome
SELECTION OF CITATIONS
SEARCH DETAIL
...