Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 62
Filter
Add more filters











Publication year range
1.
Adv Healthc Mater ; : e2303445, 2024 Jan 30.
Article in English | MEDLINE | ID: mdl-38290499

ABSTRACT

The application of oncolytic peptides has become a powerful approach to induce complete and long-lasting remission in multiple types of carcinomas, as affirmed by the appearance of tumor-associated antigens and adenosine triphosphate (ATP) in large quantities, which jumpstarts the cancer-immunity cycle. However, the ATP breakdown product adenosine is a significant contributor to forming the immunosuppressive tumor microenvironment, which substantially weakens peptide-driven oncolytic immunotherapy. In this study, a lipid-coated micelle (CA@TLM) loaded with a stapled oncolytic peptide (PalAno) and an adenosine 2A receptor (A2AR) inhibitor (CPI-444) is devised to enact tumor-targeted oncolytic immunotherapy and to overcome adenosine-mediated immune suppression simultaneously. The CA@TLM micelle accumulates in tumors with high efficiency, and the acidic tumor microenvironment prompts the rapid release of PalAno and CPI-444. Subsequently, PalAno induces swift membrane lysis of tumor cells and the release of antigenic materials. Meanwhile, CPI-444 blocks the activation of the immunosuppressive adenosine-A2AR signaling pathway. This combined approach exhibits pronounced synergy at stalling tumor growth and metastasis in animal models for triple-negative breast cancer and melanoma, providing a novel strategy for enhanced oncolytic immunotherapy.

2.
Small ; 19(18): e2207778, 2023 05.
Article in English | MEDLINE | ID: mdl-36693784

ABSTRACT

Targeted protein degradation (TPD) is an emerging therapeutic strategy with the potential of targeting undruggable pathogenic proteins. After the first proof-of-concept proteolysis-targeting chimeric (PROTAC) molecule was reported, the TPD field has entered a new era. In addition to PROTAC, numerous novel TPD strategies have emerged to expand the degradation landscape. However, their physicochemical properties and uncontrolled off-target side effects have limited their therapeutic efficacy, raising concerns regarding TPD delivery system. The combination of TPD and nanotechnology offers great promise in improving safety and therapeutic efficacy. This review provides an overview of novel TPD technologies, discusses their clinical applications, and highlights the trends and perspectives in TPD nanomedicine.


Subject(s)
Nanomedicine , Neoplasms , Humans , Proteolysis , Proteins/metabolism , Neoplasms/drug therapy , Nanotechnology
3.
Cancer Sci ; 114(5): 1958-1971, 2023 May.
Article in English | MEDLINE | ID: mdl-36692137

ABSTRACT

As one of the common malignant cancer types, gastric cancer (GC) is known for late-stage diagnosis and poor prognosis. Overexpression of the receptor tyrosine kinase MET is associated with poor prognosis among patients with advanced stage GC. However, no MET inhibitor has been used for GC treatment. Like other tyrosine kinase inhibitors that fit the "occupancy-driven" model, current MET inhibitors are prone to acquired resistance. The emerging proteolysis targeting chimera (PROTAC) strategy could overcome such limitations through direct degradation of the target proteins. In this study, we successfully transformed the MET-targeted inhibitor crizotinib into a series of PROTACs, recruiting cereblon/cullin 4A E3 ubiquitin ligase to degrade the MET proteins. The optimized lead PROTAC (PRO-6 E) effectively eliminated MET proteins in vitro and in vivo, inhibiting proliferation and motility of MET-positive GC cells. In the MKN-45 xenograft model, PRO-6 E showed pronounced antitumor efficacy with a well-tolerated dosage regimen. These results validated PRO-6 E as the first oral PROTAC for MET-dependent GC.


Subject(s)
Stomach Neoplasms , Humans , Crizotinib/pharmacology , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Proteolysis , Proteolysis Targeting Chimera , Stomach Neoplasms/drug therapy , Ubiquitin-Protein Ligases/metabolism
4.
JCI Insight ; 7(22)2022 11 22.
Article in English | MEDLINE | ID: mdl-36509291

ABSTRACT

The aberrant activation of STAT3 is associated with the etiology and progression in a variety of malignant epithelial-derived tumors, including head and neck squamous cell carcinoma (HNSCC) and colorectal cancer (CRC). Due to the lack of an enzymatic catalytic site or a ligand-binding pocket, there are no small-molecule inhibitors directly targeting STAT3 that have been approved for clinical translation. Emerging proteolysis targeting chimeric (PROTAC) technology-based approach represents a potential strategy to overcome the limitations of conventional inhibitors and inhibit activation of STAT3 and downstream genes. In this study, the heterobifunctional small-molecule-based PROTACs are successfully prepared from toosendanin (TSN), with 1 portion binding to STAT3 and the other portion binding to an E3 ubiquitin ligase. The optimized lead PROTAC (TSM-1) exhibits superior selectivity, potency, and robust antitumor effects in STAT3-dependent HNSCC and CRC - especially in clinically relevant patient-derived xenografts (PDX) and patient-derived organoids (PDO). The following mechanistic investigation identifies the reduced expression of critical downstream STAT3 effectors, through which TSM-1 promotes cell cycle arrest and apoptosis in tumor cells. These findings provide the first demonstration to our knowledge of a successful PROTAC-targeting strategy in STAT3-dependent epithelial cancer.


Subject(s)
Head and Neck Neoplasms , Ubiquitin-Protein Ligases , Humans , Proteolysis , Squamous Cell Carcinoma of Head and Neck/drug therapy , Ubiquitin-Protein Ligases/metabolism , Head and Neck Neoplasms/drug therapy , STAT3 Transcription Factor/metabolism
5.
Biochim Biophys Acta Rev Cancer ; 1877(5): 188771, 2022 09.
Article in English | MEDLINE | ID: mdl-35931392

ABSTRACT

The transmembrane protein, CD47, is recognized as an important innate immune checkpoint, and CD47-targeted drugs have been in development with the aim of inhibiting the interaction between CD47 and the regulatory glycoprotein SIRPα, for antitumor immunotherapy. Further, CD47 mediates other essential functions such as cell proliferation, caspase-independent cell death (CICD), angiogenesis and other integrin-activation-dependent cell phenotypic responses when bound to thrombospondin-1 (TSP-1) or other ligands. Mounting strategies that target CD47 have been developed in pre-clinical and clinical trials, including antibodies, small molecules, siRNAs, and peptides, and some of them have shown great promise in cancer treatment. Herein, the authors endeavor to provide a retrospective of ligand-mediated CD47 regulatory mechanisms, their roles in controlling antitumor intercellular and intracellular signal transduction, and an overview of CD47-targetd drug design.


Subject(s)
CD47 Antigen , Neoplasms , Caspases/therapeutic use , Humans , Integrins/therapeutic use , Ligands , Neoplasms/pathology , Retrospective Studies , Thrombospondin 1/genetics , Thrombospondin 1/therapeutic use
6.
Theranostics ; 12(7): 3456-3473, 2022.
Article in English | MEDLINE | ID: mdl-35547769

ABSTRACT

Rationale: Scarce tumor mutation burden and neoantigens create tremendous obstacles for an effective immunotherapy of colorectal cancer (CRC). Oncolytic peptides rise as a promising therapeutic approach that boosts tumor-specific immune responses by inducing antigenic substances. However, the clinical application of oncolytic peptides has been hindered because of structural instability, proteolytic degradation, and undesired toxicity when administered systemically. Methods: Based on wasp venom peptide, an optimized stapled oncolytic peptide MP9 was developed with rigid α-helix, protease-resistance, and CRC cell cytotoxicity. By incorporating four functional motifs that include D-peptidomimetic inhibitor of PD-L1, matrix metalloproteinase-2 (MMP-2) cleavable spacer, and MP9 with 4-arm PEG, a novel peptide-polymer conjugate (PEG-MP9-aPDL1) was obtained and identified as the most promising systemic delivery vehicle with PD-L1 targeting specificity and favorable pharmacokinetic properties. Results: We demonstrated that PEG-MP9-aPDL1-driven oncolysis induces a panel of immunogenic cell death (ICD)-relevant damage-associated molecular patterns (DAMPs) both in vitro and in vivo, which are key elements for immunotherapy with PD-L1 inhibitor. Further, PEG-MP9-aPDL1 exhibited prominent immunotherapeutic efficacy in a CRC mouse model characterized by tumor infiltration of CD8+ T cells and induction of cytotoxic lymphocytes (CTLs) in the spleens. Conclusion: Our findings suggest that PEG-MP9-aPDL1 is an all-in-one platform for oncolytic immunotherapy and immune checkpoint blockade (ICB).


Subject(s)
B7-H1 Antigen , Colorectal Neoplasms , Animals , Mice , B7-H1 Antigen/metabolism , Cell Line, Tumor , Colorectal Neoplasms/therapy , Immune Checkpoint Inhibitors , Immunologic Factors , Immunotherapy , Matrix Metalloproteinase 2 , Peptides , Polymers
7.
MedComm (2020) ; 2(3): 297-314, 2021 Sep.
Article in English | MEDLINE | ID: mdl-34766148

ABSTRACT

As key performers in intercellular communication, exosomes released by tumor cells play an important role in cancer development, including angiogenesis, cancer-associated fibroblasts activation, epithelial-mesenchymal transformation (EMT), immune escape, and pre-metastatic niche formation. Meanwhile, other cells in tumor microenvironment (TME) can secrete exosomes and facilitate tumor progression. Elucidating mechanisms regarding these processes may offer perspectives for exosome-based antitumor strategies. In this review, we mainly introduce the versatile roles of tumor or stromal cell derived exosomes in cancer development, with a particular focus on the biological capabilities and functionalities of their diverse contents, such as miRNAs, lncRNAs, and circRNAs. The potential clinical application of exosomes as biomarkers in cancer diagnosis and prognosis is also discussed. Finally, the current antitumor strategies based on exosomes in immunotherapy and targeted delivery for chemotherapeutic or biological agents are summarized.

8.
Theranostics ; 11(17): 8337-8349, 2021.
Article in English | MEDLINE | ID: mdl-34373745

ABSTRACT

Classic small molecule inhibitors that directly target pathogenic proteins typically rely on the accessible binding sites to achieve prolonged occupancy and influence protein functions. The emerging targeted protein degradation (TPD) strategies exemplified by PROteolysis TArgeting Chimeras (PROTACs) are revolutionizing conventional drug discovery modality to target proteins of interest (POIs) that were categorized as "undruggable" before, however, these strategies are limited within intracellular POIs. The novel new degrader technologies such as LYsosome-TArgeting Chimaeras (LYTACs) and Antibody-based PROTACs (AbTACs) have been successfully developed to expand the scope of TPD to extracellular and membrane proteins, fulfilling huge unmet medical needs. Here, we systematically review the currently viable protein degradation strategies, emphasize that LYTACs and AbTACs turn a new avenue for the development of TPD, and highlight the potential challenges and directions in this vibrant field.


Subject(s)
Drug Discovery , Proteins/metabolism , Proteolysis , Animals , Cellular Microenvironment , Drug Delivery Systems/methods , Drug Delivery Systems/trends , Drug Discovery/methods , Drug Discovery/trends , Humans , Lysosomes/metabolism , Membrane Proteins/metabolism , Proteasome Endopeptidase Complex/metabolism
9.
Biomed Pharmacother ; 141: 111833, 2021 Sep.
Article in English | MEDLINE | ID: mdl-34175822

ABSTRACT

Natural products continue to be an unparalleled source of pharmacologically active lead compounds because of their unprecedented structures and unique biological activities. Natural product target discovery is a vital component of natural product-based medicine translation and development and is required to understand and potentially reduce mechanisms that may be associated with off-target side effects and toxicity. Omics-based techniques, including genomics, transcriptomics, proteomics, metabolomics, and bioinformatics, have become recognized as effective tools needed to construct innovative strategies to discover natural product targets. Although considerable progress has been made, the successful discovery of natural product targets remains a challenging time-consuming process that has come to increasingly rely on the effective integration of multi-omics-based technologies to create emerging panomics (a.k.a., integrative omics, pan-omics, multiomics)-based strategies. This review summarizes a series of successful studies regarding the application of integrative omics-based methods in natural product target discovery. The advantages and disadvantages of each technique are discussed, with a particular focus on the systematic integration of multi-omics strategies. Further, emerging micro-scale single-cell-based techniques are introduced, especially to deal with minute natural product samples.


Subject(s)
Biological Products/pharmacology , Drug Discovery/methods , Genomics/methods , Animals , Computational Biology , Humans , Metabolomics , Proteomics , Transcriptome
10.
J Med Chem ; 64(9): 5802-5815, 2021 05 13.
Article in English | MEDLINE | ID: mdl-33844923

ABSTRACT

Peptide stapling chemistry represents an attractive strategy to promote the clinical translation of protein epitope mimetics, but its use has not been applied to natural cytotoxic peptides (NCPs) to produce new oncolytic peptides. Based on a wasp venom peptide, a series of stapled anoplin peptides (StAnos) were prepared. The optimized stapled Ano-3/3s were shown to be protease-resistant and exerted superior tumor cell-selective cytotoxicity by rapid membrane disruption. In addition, Ano-3/3s induced tumor ablation in mice through the direct oncolytic effect and subsequent stimulation of immunogenic cell death. This synergistic oncolytic-immunotherapy effect is more remarkable on melanoma than on triple-negative breast cancer in vivo. The efficacies exerted by Ano-3/3s on melanoma were further characterized by CD8+ T cell infiltration, and the addition of anti-CD8 antibodies diminished the long-term antitumor effects. In summary, these results support stapled peptide chemistry as an advantageous method to enhance the NCP potency for oncolytic therapy.


Subject(s)
Cell Membrane/metabolism , Melanoma, Experimental/therapy , Peptides/therapeutic use , Wasp Venoms/metabolism , Amino Acid Sequence , Animals , Antimicrobial Cationic Peptides/chemistry , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , Cell Line, Tumor , Cell Membrane/drug effects , Drug Design , Female , Hydrophobic and Hydrophilic Interactions , Immunogenic Cell Death/drug effects , Immunotherapy , Melanoma, Experimental/immunology , Mice , Mice, Inbred BALB C , Peptides/chemistry , Peptides/pharmacology , Survival Analysis , Transplantation, Homologous , Triple Negative Breast Neoplasms/immunology , Triple Negative Breast Neoplasms/therapy , Wasp Venoms/chemistry
11.
Nat Prod Rep ; 38(1): 7-17, 2021 01 01.
Article in English | MEDLINE | ID: mdl-32776055

ABSTRACT

Covering: up to 2020Treatment resistance and drug-induced refractory malignancies pose significant challenges for current chemotherapy drugs. There have been increasing research efforts aimed at developing novel chemotherapeutics, especially from natural products and related derivatives. Natural cytotoxic peptides, an emerging source of chemotherapeutics, have exhibited the advantage of overcoming drug resistance and displayed broad-spectrum antitumor activities in the clinic. This highlight examines the increasingly popular cytotoxic peptides from isolated natural products. In-depth review of several peptides provides examples for how this novel strategy can lead to the improved anti-tumor effects. The mechanisms and current application of representative natural cytotoxic peptides (NCPs) have also been discussed, with a particular focus on future directions for interdisciplinary research.


Subject(s)
Antineoplastic Agents/pharmacology , Immunoconjugates/pharmacology , Neoplasms/pathology , Peptides/chemistry , Peptides/pharmacology , Antineoplastic Agents/chemistry , Apoptosis/drug effects , Biological Products/pharmacology , Cell Membrane/drug effects , Cytotoxins/pharmacology , Humans , Immunoconjugates/chemistry , Neoplasms/drug therapy , Neoplasms/therapy , Neovascularization, Pathologic/drug therapy , Oncolytic Virotherapy/methods
12.
Front Pharmacol ; 11: 529962, 2020.
Article in English | MEDLINE | ID: mdl-33101013

ABSTRACT

Coix Seed Oil (CSO) possesses a wide range of pharmacological activities. Kanglaite Injection, a commercial product of CSO, has been used clinically as an anticancer drug in China for decades. However, its molecular mechanisms on triple-negative breast cancer (TNBC) remains to be elucidated. In this study, the effect of CSO was evaluated on murine TNBC 4T1 cells and the orthotopic tumor-bearing mouse model and underlying mechanisms were explored. CSO suppressed cell proliferation, colony formation in vitro, and tumor growth in vivo. miR-205-5p was substantially altered in CSO treated tumor tissues compared to the control group by miRNA-sequencing analysis. Sphingomyelin metabolism (SM) decreased in serum in model group compared to the control group, while it increased by CSO administration by lipid metabolomics analysis. The expression of sphingosine 1 phosphate receptor 1 (S1PR1), the critical effector of SM, was downregulated upon CSO treatment. Mechanically, miRNA-205 directly targeted S1PR1 to regulate SM and cell proliferation. CSO reduced the expression of S1PR1, cyclinD1, and phosphorylation levels of STAT3, MAPK, and AKT while upregulated p27. These results revealed that CSO exerted an anti-TNBC effect via the miR-205/S1PR1 axis to regulate sphingomyelin metabolism, and the downstream STAT3/MAPK/AKT signal pathways were partly involved.

13.
Mar Drugs ; 16(11)2018 Nov 10.
Article in English | MEDLINE | ID: mdl-30423844

ABSTRACT

Breast tumors reprogram their cellular metabolism, nutrient uptake, and utilization-associated biochemical processes. These processes become further transformed as genetically predisposed metastatic breast tumor cells colonize specific organs. Breast tumor cells often metastasize to the brain, bone, lung and liver. Massagué and colleagues isolated organotropic subclones and established organ-specific gene signatures associated with lung-, bone-, and brain-specific metastatic triple-negative breast cancer (TNBC) MDA-MB-231 cells. Using these genetically characterized metastatic subclones specific to lung (LM4175), bone (BoM1833), and brain (BrM-2a), we evaluated marine natural products for the ability to differentially suppress metastatic breast cancer cells in a target organ-dependent manner. Psammaplin-based histone deacetylase (HDAC) inhibitors were found to differentially inhibit HDAC activity, induce activation of hypoxia-inducible factor-1 (HIF-1), and disrupt organotropic metastatic TNBC subclone growth. Further, psammaplins distinctly suppressed the outgrowth of BoM1833 tumor spheroids in 3D-culture systems. Similar results were observed with the prototypical HDAC inhibitor trichostatin A (TSA). These organotropic tumor cell-based studies suggest the potential application of HDAC inhibitors that may yield new directions for anti-metastatic breast tumor research and drug discovery.


Subject(s)
Antineoplastic Agents/pharmacology , Aquatic Organisms , Disulfides/pharmacology , Histone Deacetylase Inhibitors/pharmacology , Porifera , Triple Negative Breast Neoplasms/drug therapy , Tyrosine/analogs & derivatives , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/isolation & purification , Antineoplastic Agents/therapeutic use , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Culture Techniques/methods , Disulfides/chemistry , Disulfides/isolation & purification , Disulfides/therapeutic use , Drug Discovery/methods , Drug Screening Assays, Antitumor/methods , Female , Histone Deacetylase Inhibitors/chemistry , Histone Deacetylase Inhibitors/isolation & purification , Histone Deacetylase Inhibitors/therapeutic use , Histone Deacetylases/metabolism , Humans , Spheroids, Cellular , Tyrosine/chemistry , Tyrosine/isolation & purification , Tyrosine/pharmacology , Tyrosine/therapeutic use
14.
Cell Death Dis ; 9(2): 165, 2018 02 07.
Article in English | MEDLINE | ID: mdl-29416003

ABSTRACT

Tanshinone IIA (Tan IIA), the primary bioactive compound derived from the traditional Chinese medicine (TCM) Salvia miltiorrhiza Bunge, has been reported to possess antitumor activity. However, its antitumor mechanisms are not fully understood. To resolve the potential antitumor mechanism(s) of Tan IIA, its gene expression profiles from our database was analyzed by connectivity map (CMAP) and the CMAP-based mechanistic predictions were confirmed/validated in further studies. Specifically, Tan IIA inhibited total protein kinase C (PKC) activity and selectively suppressed the expression of cytosolic and plasma membrane PKC isoforms ζ and ε. The Ras/MAPK pathway that is closely regulated by the PKC signaling is also inhibited by Tan IIA. While Tan IIA did not inhibit heat shock protein 90 (Hsp90), it synergistically enhanced the antitumor efficacy of the Hsp90 inhibitors 17-AAG and ganetespib in human breast cancer MCF-7 cells. In addition, Tan IIA significantly inhibited PI3K/Akt/mTOR signaling, and induced both cell cycle arrest and autophagy. Collectively, these studies provide new insights into the molecular mechanisms responsible for antitumor activity of Tan IIA.


Subject(s)
Abietanes/pharmacology , Antineoplastic Agents, Phytogenic/pharmacology , Benzoquinones/pharmacology , Biological Products/pharmacology , Lactams, Macrocyclic/pharmacology , Protein Kinase C/antagonists & inhibitors , Abietanes/chemistry , Animals , Antineoplastic Agents, Phytogenic/chemistry , Antineoplastic Agents, Phytogenic/therapeutic use , Apoptosis/drug effects , Autophagy/drug effects , Biological Products/chemistry , Biological Products/therapeutic use , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Cycle Checkpoints/drug effects , Cell Proliferation/drug effects , Drug Synergism , Female , Gene Expression Regulation, Neoplastic/drug effects , HSP90 Heat-Shock Proteins/metabolism , Humans , MAP Kinase Signaling System/drug effects , MCF-7 Cells , Mice, Nude , Phosphatidylinositol 3-Kinases/metabolism , Protein Kinase C/metabolism , Proto-Oncogene Proteins c-akt/metabolism , TOR Serine-Threonine Kinases/metabolism , Triazoles/pharmacology , Xenograft Model Antitumor Assays
15.
Clin Immunol ; 176: 94-99, 2017 03.
Article in English | MEDLINE | ID: mdl-28093362

ABSTRACT

Hypoxia-inducible factor (HIF)-1α is a master regulator of inflammation and is upregulated in alveolar macrophages and lung parenchyma in asthma. HIF-1α regulates select pathways in allergic inflammation, and thus may drive particular asthma phenotypes. This work examines the role of pharmacologic HIF-1α inhibition in allergic inflammatory airway disease (AIAD) pathogenesis in BALB/c mice, which develop an airway hyperresponsiveness (AHR) asthma phenotype. Systemic treatment with HIF-1α antagonist YC-1 suppressed the increase in HIF-1α expression seen in control AIAD mice. Treatment with YC-1 also decreased AHR, blood eosinophilia, and allergic inflammatory gene expression: IL-5, IL-13, myeloperoxidase and iNOS. AIAD mice had elevated BAL levels of NO, and treatment with YC-1 eliminated this response. However, YC-1 did not decrease BAL, lung or bone marrow eosinophilia. We conclude that HIF-1α inhibition in different genetic backgrounds, and thus different AIAD phenotypes, decreases airway resistance and markers of inflammation in a background specific manner. CAPSULE SUMMARY: Asthma is a common disease that can be difficult to control with current therapeutics. We describe how pharmacologic targeting of a specific transcription factor, HIF-1α, suppresses asthmatic airway reactivity and inflammation.


Subject(s)
Asthma/metabolism , Hypersensitivity/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Lung/metabolism , Nitric Oxide/metabolism , Animals , Bronchoalveolar Lavage Fluid/chemistry , Disease Models, Animal , Eosinophils/metabolism , Hypoxia/metabolism , Inflammation/metabolism , Interleukin-13/metabolism , Interleukin-5/metabolism , Mice , Mice, Inbred BALB C , Nitric Oxide Synthase Type II/metabolism
16.
J Nat Prod ; 78(12): 3018-23, 2015 Dec 24.
Article in English | MEDLINE | ID: mdl-26637046

ABSTRACT

The cananga tree alkaloid sampangine (1) has been extensively investigated for its antimicrobial and antitumor potential. Mechanistic studies have linked its biological activities to the reduction of cellular oxygen, the induction of reactive oxygen species (ROS), and alterations in heme biosynthesis. Based on the yeast gene deletion library screening results that indicated mitochondrial gene deletions enhanced the sensitivity to 1, the effects of 1 on cellular respiration were examined. Sampangine increased oxygen consumption rates in both yeast and human tumor cells. Mechanistic investigation indicated that 1 may have a modest uncoupling effect, but predominately acts by increasing oxygen consumption independent of mitochondrial complex IV. Sampangine thus appears to undergo redox cycling that may involve respiratory chain-dependent reduction to a semi-iminoquinone followed by oxidation and consequent superoxide production. Relatively high concentrations of 1 showed significant neurotoxicity in studies conducted with rat cerebellar granule neurons, indicating that sampangine use may be associated with potential neurotoxicity.


Subject(s)
Alkaloids/pharmacology , Anti-Infective Agents/pharmacology , Antineoplastic Agents, Phytogenic/pharmacology , Quinones/pharmacology , Animals , Benzoquinones , Cell Cycle/drug effects , Cell Division , Cell Respiration/drug effects , Electron Transport , Heterocyclic Compounds, 4 or More Rings/pharmacology , Humans , Mitochondria/metabolism , Molecular Structure , Naphthyridines , Oxidation-Reduction , Oxygen , Rats , Reactive Oxygen Species/metabolism , Saccharomyces cerevisiae , Superoxides/metabolism
17.
Mar Drugs ; 13(3): 1552-68, 2015 Mar 20.
Article in English | MEDLINE | ID: mdl-25803180

ABSTRACT

The biologically active lipopeptide kalkitoxin was previously isolated from the marine cyanobacterium Moorea producens (Lyngbya majuscula). Kalkitoxin exhibited N-methyl-D-aspartate (NMDA)-mediated neurotoxicity and acted as an inhibitory ligand for voltage-sensitive sodium channels in cultured rat cerebellar granule neurons. Subsequent studies revealed that kalkitoxin generated a delayed form of colon tumor cell cytotoxicity in 7-day clonogenic cell survival assays. Cell line- and exposure time-dependent cytostatic/cytotoxic effects were previously observed with mitochondria-targeted inhibitors of hypoxia-inducible factor-1 (HIF-1). The transcription factor HIF-1 functions as a key regulator of oxygen homeostasis. Therefore, we investigated the ability of kalkitoxin to inhibit hypoxic signaling in human tumor cell lines. Kalkitoxin potently and selectively inhibited hypoxia-induced activation of HIF-1 in T47D breast tumor cells (IC50 5.6 nM). Mechanistic studies revealed that kalkitoxin inhibits HIF-1 activation by suppressing mitochondrial oxygen consumption at electron transport chain (ETC) complex I (NADH-ubiquinone oxidoreductase). Further studies indicate that kalkitoxin targets tumor angiogenesis by blocking the induction of angiogenic factors (i.e., VEGF) in tumor cells.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Breast Neoplasms/drug therapy , Lipids/pharmacology , Neovascularization, Pathologic/drug therapy , Thiazoles/pharmacology , Angiogenesis Inhibitors/administration & dosage , Breast Neoplasms/pathology , Cell Hypoxia/drug effects , Cell Line, Tumor , Electron Transport Complex I/drug effects , Electron Transport Complex I/metabolism , Female , Humans , Hypoxia-Inducible Factor 1/metabolism , Inhibitory Concentration 50 , Lipids/administration & dosage , Mitochondria/drug effects , Mitochondria/metabolism , Signal Transduction/drug effects , Thiazoles/administration & dosage , Vascular Endothelial Growth Factor A/metabolism
18.
J Nat Prod ; 77(1): 111-7, 2014 Jan 24.
Article in English | MEDLINE | ID: mdl-24328138

ABSTRACT

Certain botanical dietary supplements have been associated with idiosyncratic organ-specific toxicity. Similar toxicological events, caused by drug-induced mitochondrial dysfunction, have forced the withdrawal or U.S. FDA "black box" warnings of major pharmaceuticals. To assess the potential mitochondrial liability of botanical dietary supplements, extracts from 352 authenticated plant samples used in traditional Chinese, Ayurvedic, and Western herbal medicine were evaluated for the ability to disrupt cellular respiration. Blue cohosh (Caulophyllum thalictroides) methanol extract exhibited mitochondriotoxic activity. Used by some U.S. midwives to help induce labor, blue cohosh has been associated with perinatal stroke, acute myocardial infarction, congestive heart failure, multiple organ injury, and neonatal shock. The potential link between mitochondrial disruption and idiosyncratic herbal intoxication prompted further examination. The C. thalictroides methanol extract and three saponins, cauloside A (1), saponin PE (2), and cauloside C (3), exhibited concentration- and time-dependent mitochondriotoxic activities. Upon treatment, cell respiration rate rapidly increased and then dramatically decreased within minutes. Mechanistic studies revealed that C. thalictroides constituents impair mitochondrial function by disrupting membrane integrity. These studies provide a potential etiological link between this mitochondria-sensitive form of cytotoxicity and idiosyncratic organ damage.


Subject(s)
Caulophyllum/chemistry , Cell Respiration/drug effects , Dietary Supplements/toxicity , Membrane Potential, Mitochondrial/drug effects , Oleanolic Acid/analogs & derivatives , Saponins/toxicity , Dose-Response Relationship, Drug , Humans , Molecular Structure , Oleanolic Acid/chemistry , Oleanolic Acid/toxicity , Phytotherapy , Saponins/chemistry , United States
19.
J Nat Prod ; 76(8): 1492-7, 2013 Aug 23.
Article in English | MEDLINE | ID: mdl-23944963

ABSTRACT

The extract of marine sponge Hyrtios communis was found to inhibit activation of the transcription factor hypoxia-inducible factor-1 (HIF-1) in T47D human breast tumor cells. Bioassay-guided isolation led to the identification of six new (1-6) and five previously reported (7-11) sesterterpene analogues and two unrelated sesterterpenes. Two new sesterterpenes, thorectidaeolide A (1) and 4-acetoxythorectidaeolide A (2), and luffariellolide (11) were among the most potent inhibitors of hypoxia (1% O2)-induced HIF-1 activation (IC50 values of 3.2, 3.5, and 3.6 µM, respectively). Luffariellolide (11) exhibited a significant level of cytotoxicity that mirrored its HIF-1 inhibitory activity. Neither compound 1, compound 2, nor any of the other less active sesterterpenes suppressed breast tumor T47D or MDA-MB-231 cell viability.


Subject(s)
Antineoplastic Agents/isolation & purification , Antineoplastic Agents/pharmacology , Porifera/chemistry , Sesterterpenes/isolation & purification , Sesterterpenes/pharmacology , Terpenes/pharmacology , Animals , Antineoplastic Agents/chemistry , Cell Survival/drug effects , Drug Screening Assays, Antitumor , Female , Humans , Hypoxia-Inducible Factor 1/antagonists & inhibitors , Inhibitory Concentration 50 , Marine Biology , Molecular Structure , Oceans and Seas , Palau , Sesterterpenes/chemistry
20.
J Nat Prod ; 76(6): 1175-81, 2013 Jun 28.
Article in English | MEDLINE | ID: mdl-23731014

ABSTRACT

The hypoxia-inducible factor-1 (HIF-1) transcription factor regulates cellular oxygen homeostasis. Agents that activate HIF-1 and downstream HIF targets represent potential drug leads for the prevention and/or treatment of ischemic disorders. In a search for small-molecule HIF-1 activators, 1936 marine invertebrate and algal extract samples (U.S. National Cancer Institute's Open Repository) were evaluated for HIF-1 activation activity in a cell-based reporter assay. Bioassay-guided fractionation of two active extracts of the sponge Dactylospongia elegans afforded four new sesquiterpene quinones (2-5), one new sesquiterpene phenol (6), the known Golgi disruptor ilimaquinone (1), and three previously reported ilimaquinone analogues (7-9). While antiproliferative activity was observed at higher concentrations, the sesquiterpene quinones (1-3) possessing a 2-hydroxy-5-methoxy-1,4-benzoquinone moiety activated HIF-1 and increased the expression of HIF-1 target gene vascular endothelial growth factor (VEGF) in T47D cells.


Subject(s)
Hypoxia-Inducible Factor 1/drug effects , Porifera/chemistry , Quinones/isolation & purification , Quinones/pharmacology , Sesquiterpenes/isolation & purification , Sesquiterpenes/pharmacology , Animals , Dose-Response Relationship, Drug , Female , Humans , Molecular Structure , Nuclear Magnetic Resonance, Biomolecular , Oceans and Seas , Quinones/chemistry , Sesquiterpenes/chemistry , Vascular Endothelial Growth Factor A/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL