Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
1.
Nat Cell Biol ; 25(12): 1848-1859, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37957324

ABSTRACT

Breast cancer brain metastasis (BCBM) is a lethal disease with no effective treatments. Prior work has shown that brain cancers and metastases are densely infiltrated with anti-inflammatory, protumourigenic tumour-associated macrophages, but the role of brain-resident microglia remains controversial because they are challenging to discriminate from other tumour-associated macrophages. Using single-cell RNA sequencing, genetic and humanized mouse models, we specifically identify microglia and find that they play a distinct pro-inflammatory and tumour-suppressive role in BCBM. Animals lacking microglia show increased metastasis, decreased survival and reduced natural killer and T cell responses, showing that microglia are critical to promote anti-tumour immunity to suppress BCBM. We find that the pro-inflammatory response is conserved in human microglia, and markers of their response are associated with better prognosis in patients with BCBM. These findings establish an important role for microglia in anti-tumour immunity and highlight them as a potential immunotherapy target for brain metastasis.


Subject(s)
Brain Neoplasms , Breast Neoplasms , Mice , Animals , Humans , Female , Microglia , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Brain Neoplasms/pathology , Brain/pathology , Treatment Outcome
2.
Nat Genet ; 55(4): 595-606, 2023 04.
Article in English | MEDLINE | ID: mdl-36914836

ABSTRACT

Women with germline BRCA1 mutations (BRCA1+/mut) have increased risk for hereditary breast cancer. Cancer initiation in BRCA1+/mut is associated with premalignant changes in breast epithelium; however, the role of the epithelium-associated stromal niche during BRCA1-driven tumor initiation remains unclear. Here we show that the premalignant stromal niche promotes epithelial proliferation and mutant BRCA1-driven tumorigenesis in trans. Using single-cell RNA sequencing analysis of human preneoplastic BRCA1+/mut and noncarrier breast tissues, we show distinct changes in epithelial homeostasis including increased proliferation and expansion of basal-luminal intermediate progenitor cells. Additionally, BRCA1+/mut stromal cells show increased expression of pro-proliferative paracrine signals. In particular, we identify pre-cancer-associated fibroblasts (pre-CAFs) that produce protumorigenic factors including matrix metalloproteinase 3 (MMP3), which promotes BRCA1-driven tumorigenesis in vivo. Together, our findings demonstrate that precancerous stroma in BRCA1+/mut may elevate breast cancer risk through the promotion of epithelial proliferation and an accumulation of luminal progenitor cells with altered differentiation.


Subject(s)
Breast Neoplasms , Mammary Glands, Human , Female , Humans , Mutation , BRCA1 Protein/genetics , Breast Neoplasms/pathology , Cell Transformation, Neoplastic/metabolism , Mammary Glands, Human/metabolism , Carcinogenesis/pathology , Stromal Cells/pathology
4.
Commun Biol ; 4(1): 1268, 2021 11 05.
Article in English | MEDLINE | ID: mdl-34741115

ABSTRACT

Metastasis is a fatal disease where research progress has been hindered by a lack of authentic experimental models. Here, we develop a 3D tumor sphere culture-transplant system that facilitates the growth and engineering of patient-derived xenograft (PDX) tumor cells for functional metastasis assays in vivo. Orthotopic transplantation and RNA sequencing (RNA-seq) analyses show that PDX tumor spheres maintain tumorigenic potential, and the molecular marker and global transcriptome signatures of native tumor cells. Tumor spheres display robust capacity for lentiviral engineering and dissemination in spontaneous and experimental metastasis assays in vivo. Inhibition of pathways previously reported to attenuate metastasis also inhibit metastasis after sphere culture, validating our approach for authentic investigations of metastasis. Finally, we demonstrate a new role for the metabolic enzyme NME1 in promoting breast cancer metastasis, providing proof-of-principle that our culture-transplant system can be used for authentic propagation and engineering of patient tumor cells for functional studies of metastasis.


Subject(s)
Breast Neoplasms/pathology , Heterografts , Neoplasm Metastasis , Xenograft Model Antitumor Assays , Animals , Disease Models, Animal , Female , Mice , Neoplasms, Experimental , Tumor Microenvironment
5.
Nat Methods ; 18(9): 1091-1102, 2021 09.
Article in English | MEDLINE | ID: mdl-34413523

ABSTRACT

Mitochondria display complex morphology and movements, which complicates their segmentation and tracking in time-lapse images. Here, we introduce Mitometer, an algorithm for fast, unbiased, and automated segmentation and tracking of mitochondria in live-cell two-dimensional and three-dimensional time-lapse images. Mitometer requires only the pixel size and the time between frames to identify mitochondrial motion and morphology, including fusion and fission events. The segmentation algorithm isolates individual mitochondria via a shape- and size-preserving background removal process. The tracking algorithm links mitochondria via differences in morphological features and displacement, followed by a gap-closing scheme. Using Mitometer, we show that mitochondria of triple-negative breast cancer cells are faster, more directional, and more elongated than those in their receptor-positive counterparts. Furthermore, we show that mitochondrial motility and morphology in breast cancer, but not in normal breast epithelia, correlate with metabolic activity. Mitometer is an unbiased and user-friendly tool that will help resolve fundamental questions regarding mitochondrial form and function.


Subject(s)
Breast Neoplasms/pathology , Imaging, Three-Dimensional/methods , Mitochondria , Software , Time-Lapse Imaging/methods , Algorithms , Breast Neoplasms/metabolism , Cells, Cultured , Female , Humans , Mammary Glands, Human/cytology , Mitochondria/metabolism , NAD/metabolism , Reproducibility of Results , Triple Negative Breast Neoplasms/pathology
6.
Nat Cell Biol ; 22(3): 310-320, 2020 03.
Article in English | MEDLINE | ID: mdl-32144411

ABSTRACT

Although metastasis remains the cause of most cancer-related mortality, mechanisms governing seeding in distal tissues are poorly understood. Here, we establish a robust method for the identification of global transcriptomic changes in rare metastatic cells during seeding using single-cell RNA sequencing and patient-derived-xenograft models of breast cancer. We find that both primary tumours and micrometastases display transcriptional heterogeneity but micrometastases harbour a distinct transcriptome program conserved across patient-derived-xenograft models that is highly predictive of poor survival of patients. Pathway analysis revealed mitochondrial oxidative phosphorylation as the top pathway upregulated in micrometastases, in contrast to higher levels of glycolytic enzymes in primary tumour cells, which we corroborated by flow cytometric and metabolomic analyses. Pharmacological inhibition of oxidative phosphorylation dramatically attenuated metastatic seeding in the lungs, which demonstrates the functional importance of oxidative phosphorylation in metastasis and highlights its potential as a therapeutic target to prevent metastatic spread in patients with breast cancer.


Subject(s)
Breast Neoplasms/genetics , Breast Neoplasms/pathology , Transcriptome , Animals , Breast Neoplasms/metabolism , Energy Metabolism , Female , Humans , Mice, Inbred NOD , Mice, SCID , Mitochondria/metabolism , Neoplasm Metastasis , Oxidative Phosphorylation , Sequence Analysis, RNA , Single-Cell Analysis , Transcription, Genetic
7.
Sci Immunol ; 5(44)2020 02 21.
Article in English | MEDLINE | ID: mdl-32086381

ABSTRACT

Myeloid-derived suppressor cells (MDSCs) are innate immune cells that acquire the capacity to suppress adaptive immune responses during cancer. It remains elusive how MDSCs differ from their normal myeloid counterparts, which limits our ability to specifically detect and therapeutically target MDSCs during cancer. Here, we sought to determine the molecular features of breast cancer-associated MDSCs using the widely studied mouse model based on the mouse mammary tumor virus (MMTV) promoter-driven expression of the polyomavirus middle T oncoprotein (MMTV-PyMT). To identify MDSCs in an unbiased manner, we used single-cell RNA sequencing to compare MDSC-containing splenic myeloid cells from breast tumor-bearing mice with wild-type controls. Our computational analysis of 14,646 single-cell transcriptomes revealed that MDSCs emerge through an aberrant neutrophil maturation trajectory in the spleen that confers them an immunosuppressive cell state. We establish the MDSC-specific gene signature and identify CD84 as a surface marker for improved detection and enrichment of MDSCs in breast cancers.


Subject(s)
Breast Neoplasms/pathology , Myeloid-Derived Suppressor Cells/pathology , Single-Cell Analysis , Transcriptome , Animals , Biomarkers, Tumor/genetics , Biomarkers, Tumor/immunology , Breast Neoplasms/immunology , Cell Differentiation/genetics , Female , Humans , Mice , Mice, Inbred Strains , Mice, Transgenic , Myeloid-Derived Suppressor Cells/immunology , RNA, Neoplasm/genetics , RNA, Neoplasm/immunology , Signaling Lymphocytic Activation Molecule Family/genetics , Signaling Lymphocytic Activation Molecule Family/immunology
8.
J Alzheimers Dis ; 69(3): 631-649, 2019.
Article in English | MEDLINE | ID: mdl-31127770

ABSTRACT

Neurons consume the highest amount of oxygen, depend on oxidative metabolism for energy, and survive for the lifetime of an individual. Therefore, neurons are vulnerable to death caused by oxidative-stress, accumulation of damaged and dysfunctional proteins and organelles. There is an exponential increase in the number of patients diagnosed with neurodegenerative diseases such as Alzheimer's (AD) as the number of elderly increases exponentially. Development of AD pathology is a complex phenomenon characterized by neuronal death, accumulation of extracellular amyloid-ß plaques and neurofibrillary tangles, and most importantly loss of memory and cognition. These pathologies are most likely caused by mechanisms including oxidative stress, mitochondrial dysfunction/stress, accumulation of misfolded proteins, and defective organelles due to impaired proteasome and autophagy mechanisms. Currently, there are no effective treatments to halt the progression of this disease. In order to treat this complex disease with multiple biochemical pathways involved, a complex treatment regimen targeting different mechanisms should be investigated. Furthermore, as AD is a progressive disease-causing morbidity over many years, any chemo-modulator for treatment must be used over long period of time. Therefore, treatments must be safe and non-interfering with other processes. Ideally, a treatment like medicinal food or a supplement that can be taken regularly without any side effect capable of reducing oxidative stress, stabilizing mitochondria, activating autophagy or proteasome, and increasing energy levels of neurons would be the best solution. This review summarizes progress in research on different mechanisms of AD development and some of the potential therapeutic development strategies targeting the aforementioned pathologies.


Subject(s)
Alzheimer Disease/pathology , Signal Transduction/drug effects , Alzheimer Disease/drug therapy , Animals , Autophagy , Disease Progression , Humans , Oxidative Stress
9.
Nat Commun ; 10(1): 650, 2019 02 08.
Article in English | MEDLINE | ID: mdl-30737373

ABSTRACT

During wound healing in adult mouse skin, hair follicles and then adipocytes regenerate. Adipocytes regenerate from myofibroblasts, a specialized contractile wound fibroblast. Here we study wound fibroblast diversity using single-cell RNA-sequencing. On analysis, wound fibroblasts group into twelve clusters. Pseudotime and RNA velocity analyses reveal that some clusters likely represent consecutive differentiation states toward a contractile phenotype, while others appear to represent distinct fibroblast lineages. One subset of fibroblasts expresses hematopoietic markers, suggesting their myeloid origin. We validate this finding using single-cell western blot and single-cell RNA-sequencing on genetically labeled myofibroblasts. Using bone marrow transplantation and Cre recombinase-based lineage tracing experiments, we rule out cell fusion events and confirm that hematopoietic lineage cells give rise to a subset of myofibroblasts and rare regenerated adipocytes. In conclusion, our study reveals that wounding induces a high degree of heterogeneity among fibroblasts and recruits highly plastic myeloid cells that contribute to adipocyte regeneration.


Subject(s)
Adipocytes/cytology , Adipocytes/metabolism , Fibroblasts/cytology , Fibroblasts/metabolism , Single-Cell Analysis/methods , Skin/cytology , Stem Cells/cytology , Animals , Blotting, Western , Cells, Cultured , Female , Male , Mice , Sequence Analysis, RNA , Stem Cells/metabolism , Wound Healing/physiology
10.
Oxid Med Cell Longev ; 2019: 7404815, 2019.
Article in English | MEDLINE | ID: mdl-31934268

ABSTRACT

Alzheimer's disease (AD) is the most prevalent form of dementia and is associated with loss of memory, amyloid-beta plaque buildup, and neurofibrillary tangles. These features might be a result of neuronal cell death in the cerebral cortex and hippocampal regions of the brain. AD pathologies can be attributed to a variety of biochemical consequences including mitochondrial dysfunction, increased oxidative stress, and autophagy inhibition. Unfortunately, current therapeutics are limited only to symptomatic relief and do not halt the progression of neurodegeneration. Previous in vitro experiments have shown that a water-soluble formulation of coenzyme-Q10, Ubisol-Q10, can stabilize the mitochondria, prevent oxidative stress, and inhibit premature senescence in fibroblasts of AD patients. Since autophagy plays a critical role in maintenance and survival of neurons, we hypothesized that Ubisol-Q10 treatment could result in resumption of autophagy. Indeed, we observed induction of autophagy by Ubisol-Q10 treatment in AD fibroblasts as well as in the brains of transgenic AD mice. We found increased expression of autophagy-related genes beclin-1 and JNK1 following Ubisol-Q10 treatment of AD fibroblasts. These results were confirmed at the protein level by immunofluorescence and Western blotting. Interestingly, despite reduction of oxidative stress in cells due to Ubisol-Q10 treatment, autophagy inhibition leads to resumption of premature senescence in these PS-1 mutated fibroblasts indicating that autophagy is critical to prevent the senescence phenotype. Withdrawal of Ubisol-Q10 treatment also leads to the return of the senescence phenotype in AD fibroblasts indicating that constant supplementation of Ubisol-Q10 is required. Additionally, Ubisol-Q10 supplementation in the drinking water of double transgenic AD mice leads to increased expression of beclin-1 and JNK1 in the cortical region. Thus, the activation of autophagy by Ubisol-Q10 could be the mechanism for its ability to halt the progression of AD pathology in transgenic AD mice shown previously.


Subject(s)
Alzheimer Disease/drug therapy , Cerebral Cortex/metabolism , Fibroblasts/physiology , Mutation/genetics , Presenilin-1/genetics , Ubiquinone/analogs & derivatives , Animals , Autophagy , Beclin-1/genetics , Beclin-1/metabolism , Cell Death , Cellular Senescence/drug effects , Cerebral Cortex/drug effects , Disease Models, Animal , Fibroblasts/drug effects , Humans , Mice , Mice, Transgenic , Mitogen-Activated Protein Kinase 8/genetics , Mitogen-Activated Protein Kinase 8/metabolism , Neuroprotection , Ubiquinone/chemistry , Ubiquinone/therapeutic use , Up-Regulation
11.
BMC Health Serv Res ; 18(1): 970, 2018 Dec 17.
Article in English | MEDLINE | ID: mdl-30558605

ABSTRACT

BACKGROUND: Chronic back disorders (CBD) are prevalent, costly, and among the most common reasons for seeking primary care; however, little is known regarding the comparative use of family physician, chiropractic, and physiotherapy services among people with CBD in Canada. Elucidating these differences may identify potential gaps in access to care and inform the development of strategies to improve access. The research objectives were to investigate patterns of health care use and to profile factors associated with self-reported use of family physicians, chiropractors, and physiotherapists among adult Canadians with CBD. METHODS: The combined 2009 and 2010 Canadian Community Health Surveys conducted by Statistics Canada were used to investigate self-reported health care use among adults with CBD. This complex survey employs population weights and bootstrapping to be representative of the Canadian population. Following descriptive analyses, we used multiple logistic regression to profile self-reported health care use while statistically controlling for possible confounding effects. RESULTS: The majority of adult respondents with CBD sought care only with a family physician (53.8%), with 20.9% and 16.2% seeking care with combined family physician/chiropractor or family physician/physiotherapist, respectively. Few respondents sought care only with a chiropractor (2.5%) or physiotherapist (1.0%). After adjustment, differential patterns of utilization (p < 0.05) were evident between provider groups with respect to age, gender, socioeconomic status, rural/urban residence, functional limitations, and presence of co-morbidities. CONCLUSIONS: This research highlights potential inequities in access to physiotherapists and chiropractors in relation to family physicians among adult Canadians with CBD, particularly among lower socioeconomic status and rural/remote populations.


Subject(s)
Back Pain/therapy , Chiropractic/statistics & numerical data , Family Practice/statistics & numerical data , Patient Acceptance of Health Care/statistics & numerical data , Physical Therapy Modalities/statistics & numerical data , Adolescent , Adult , Age Distribution , Aged , Back Pain/epidemiology , Canada/epidemiology , Chronic Pain/epidemiology , Community Health Services/statistics & numerical data , Comorbidity , Facilities and Services Utilization , Female , Health Surveys , Humans , Logistic Models , Male , Middle Aged , Physical Therapists/statistics & numerical data , Physicians, Family/statistics & numerical data , Prevalence , Rural Health/statistics & numerical data , Self Report , Surveys and Questionnaires , Young Adult
13.
Nat Commun ; 9(1): 2028, 2018 05 23.
Article in English | MEDLINE | ID: mdl-29795293

ABSTRACT

Breast cancer arises from breast epithelial cells that acquire genetic alterations leading to subsequent loss of tissue homeostasis. Several distinct epithelial subpopulations have been proposed, but complete understanding of the spectrum of heterogeneity and differentiation hierarchy in the human breast remains elusive. Here, we use single-cell mRNA sequencing (scRNAseq) to profile the transcriptomes of 25,790 primary human breast epithelial cells isolated from reduction mammoplasties of seven individuals. Unbiased clustering analysis reveals the existence of three distinct epithelial cell populations, one basal and two luminal cell types, which we identify as secretory L1- and hormone-responsive L2-type cells. Pseudotemporal reconstruction of differentiation trajectories produces one continuous lineage hierarchy that closely connects the basal lineage to the two differentiated luminal branches. Our comprehensive cell atlas provides insights into the cellular blueprint of the human breast epithelium and will form the foundation to understand how the system goes awry during breast cancer.


Subject(s)
Breast Neoplasms/genetics , Breast/cytology , Epithelial Cells/physiology , Gene Expression Profiling/methods , Transcriptome/genetics , Adult , Biomarkers, Tumor/genetics , Breast/pathology , Breast Neoplasms/pathology , Cell Differentiation/genetics , Cell Lineage/genetics , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/pathology , Cluster Analysis , Female , Humans , Sequence Analysis, RNA/methods , Single-Cell Analysis/methods
14.
FASEB J ; 32(1): 417-430, 2018 01.
Article in English | MEDLINE | ID: mdl-28928246

ABSTRACT

Harsh adverse effects as a result of nonspecific targeting of chemotherapeutics currently pose obstacles in cancer therapy; thus, it would be invaluable to devise novel approaches to specifically target cancer cells. The natural compound pancratistatin (PST) has been shown to preferentially induce apoptosis in a variety of cancer cell types. Recently, several analogs of PST were shown to be efficacious in inducing apoptosis in a variety of aggressive cancer cell types via cancer cell mitochondrial targeting; it caused dissipation of mitochondrial membrane potential and decreased oxygen consumption, and with isolated mitochondria, it induced the release of apoptogenic factors. The natural compound piperlongumine has been shown to target the stress response to reactive oxygen species in cancer cells. We explored the combinatorial potential of two small molecules (SVTH-6 and piperlongumine) that target these vulnerabilities in cancer cells. Interestingly, when combined with the PST analog, SVTH-6, an increase in mitochondrial dysfunction was observed, leading to an enhanced cytotoxic effect against several human cancer cell types. Additionally, this combination treatment was effective in reducing cancer cell growth in physiologically more relevant 3-dimensional spheroid cell cultures. This enhanced effect was found to be dependent on reactive oxygen species generation because an antioxidant could rescue cancer cells from this combination treatment. Importantly, noncancerous cells were markedly less sensitive to this combination treatment. Thus, targeting mitochondrial and oxidative stress vulnerabilities of cancer cells could be an effective strategy for cancer therapy.-Ma, D., Gilbert, T., Pignanelli, C., Tarade, D., Noel, M., Mansour, F., Gupta, M., Ma, S., Ropat, J., Curran, C., Vshyvenko, S., Hudlicky, T., Pandey. S. Exploiting mitochondrial and oxidative vulnerabilities with a synthetic analog of pancratistatin in combination with piperlongumine for cancer therapy.


Subject(s)
Amaryllidaceae Alkaloids/administration & dosage , Dioxolanes/administration & dosage , Isoquinolines/administration & dosage , Neoplasms/drug therapy , Amaryllidaceae Alkaloids/chemistry , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/chemistry , Apoptosis/drug effects , Cell Line, Tumor , Drug Synergism , HCT116 Cells , HT29 Cells , Humans , Isoquinolines/chemistry , Membrane Potential, Mitochondrial/drug effects , Mitochondria/drug effects , Neoplasms/metabolism , Neoplasms/pathology , Oxidative Stress/drug effects , U937 Cells
15.
Sci Rep ; 7(1): 1105, 2017 04 24.
Article in English | MEDLINE | ID: mdl-28439094

ABSTRACT

Recently, research has focused on targeting the oxidative and metabolic vulnerabilities in cancer cells. Natural compounds like curcumin that target such susceptibilities have failed further clinical advancements due to the poor stability and bioavailability as well as the need of high effective doses. We have synthesized and evaluated the anti-cancer activity of several monocarbonyl analogs of curcumin. Interestingly, two novel analogs (Compound A and I) in comparison to curcumin, have increased chemical stability and have greater anti-cancer activity in a variety of human cancer cells, including triple-negative, inflammatory breast cancer cells. In particular, the generation of reactive oxygen species was selective to cancer cells and occurred upstream of mitochondrial collapse and execution of apoptosis. Furthermore, Compound A in combination with another cancer-selective/pro-oxidant, piperlongumine, caused an enhanced anti-cancer effect. Most importantly, Compound A was well tolerated by mice and was effective in inhibiting the growth of human triple-negative breast cancer and leukemia xenografts in vivo when administered intraperitoneally. Thus, exploiting oxidative vulnerabilities in cancer cells could be a selective and efficacious means to eradicate malignant cells as demonstrated by the curcumin analogs presented in this report with high therapeutic potential.


Subject(s)
Antineoplastic Agents/pharmacology , Curcumin/analogs & derivatives , Curcumin/pharmacology , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/chemical synthesis , Breast Neoplasms/drug therapy , Cell Line, Tumor , Cell Survival/drug effects , Curcumin/administration & dosage , Curcumin/chemical synthesis , Disease Models, Animal , Humans , Leukemia/drug therapy , Mice , Neoplasm Transplantation , Reactive Oxygen Species/analysis , Reactive Oxygen Species/toxicity , Treatment Outcome
16.
PLoS One ; 12(3): e0171806, 2017.
Article in English | MEDLINE | ID: mdl-28253265

ABSTRACT

The cis-stilbene, combretastatin A4 (CA4), is a potent microtubule targeting and vascular damaging agent. Despite promising results at the pre-clinical level and extensive clinical evaluation, CA4 has yet to be approved for therapeutic use. One impediment to the development of CA4 is an inherent conformational instability about the ethylene linker, which joins two aromatic rings. We have previously published preliminary data regarding structurally simplified biphenyl derivatives of CA4, lacking an ethylene linker, which retain anti-proliferative and pro-apoptotic activity, albeit at higher doses. Our current study provides a more comprehensive evaluation regarding the anti-proliferative and pro-apoptotic properties of biphenyl CA4 derivatives in both 2D and 3D cancerous and non-cancerous cell models. Computational analysis has revealed that cytotoxicity of CA4 and biphenyl analogues correlates with predicted tubulin affinity. Additional mechanistic evaluation of the biphenyl derivatives found that their anti-cancer activity is dependent on prolonged mitotic arrest, in a similar manner to CA4. Lastly, we have shown that cancer cells deficient in the extrinsic pathway of apoptosis experience delayed cell death following treatment with CA4 or analogues. Biphenyl derivatives of CA4 represent structurally simplified analogues of CA4, which retain a similar mechanism of action. The biphenyl analogues warrant in vivo examination to evaluate their potential as vascular damaging agents.


Subject(s)
Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Biphenyl Compounds/chemistry , Cell Cycle Checkpoints/drug effects , Mitosis/drug effects , Stilbenes/chemistry , Stilbenes/pharmacology , Antineoplastic Agents/metabolism , Apoptosis/drug effects , Cell Proliferation/drug effects , DNA Damage , Humans , Jurkat Cells , Membrane Potential, Mitochondrial/drug effects , Models, Molecular , Protein Conformation , Stilbenes/metabolism , Tubulin/chemistry , Tubulin/metabolism
17.
Sci Rep ; 7: 42957, 2017 02 21.
Article in English | MEDLINE | ID: mdl-28220885

ABSTRACT

Enhanced mitochondrial stability and decreased dependence on oxidative phosphorylation confer an acquired resistance to apoptosis in cancer cells, but may present opportunities for therapeutic intervention. The compound pancratistatin (PST) has been shown to selectively induce apoptosis in cancer cells. However, its low availability in nature has hindered its clinical advancement. We synthesized PST analogs and a medium-throughput screen was completed. Analogs SVTH-7, -6, and -5 demonstrated potent anti-cancer activity greater than PST and several standard chemotherapeutics. They disrupted mitochondrial function, activated the intrinsic apoptotic pathway, and reduced growth of tumor xenografts in vivo. Interestingly, the pro-apoptotic effects of SVTH-7 on cancer cells and mitochondria were abrogated with the inhibition of mitochondrial complex II and III, suggesting mitochondrial or metabolic vulnerabilities may be exploited by this analog. This work provides a scaffold for characterizing distinct mitochondrial and metabolic features of cancer cells and reveals several lead compounds with high therapeutic potential.


Subject(s)
Amaryllidaceae Alkaloids/pharmacology , Antineoplastic Agents/pharmacology , Electron Transport Complex III/metabolism , Electron Transport Complex II/metabolism , Isoquinolines/pharmacology , Mitochondria/drug effects , Amaryllidaceae Alkaloids/chemistry , Amaryllidaceae Alkaloids/therapeutic use , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/therapeutic use , Apoptosis/drug effects , Body Weight/drug effects , Caspases/metabolism , Cell Culture Techniques , Cell Line, Tumor , Electron Transport Complex II/antagonists & inhibitors , Electron Transport Complex III/antagonists & inhibitors , Humans , Isoquinolines/chemistry , Isoquinolines/therapeutic use , Mice , Mice, Nude , Mitochondria/metabolism , Neoplasms/drug therapy , Neoplasms/pathology , Reactive Oxygen Species/metabolism , Spheroids, Cellular/drug effects , Spheroids, Cellular/pathology , Transplantation, Heterologous
19.
Oncotarget ; 8(67): 110756-110773, 2017 Dec 19.
Article in English | MEDLINE | ID: mdl-29340014

ABSTRACT

Cancer cells are reported to have elevated levels of reactive oxygen species (ROS) and are highly dependent on cellular defense mechanisms against oxidative stress. Numerous nutraceuticals and natural polyphenolic compounds have a wide range of abilities to alter cellular redox states with potential implications in various diseases. Furthermore, therapeutic options for cancers are mostly nonselective treatments including genotoxic or tubulin-targeting compounds. Some of the natural extracts, containing multiple bioactive compounds, could target multiple pathways in cancer cells to selectively induce cell death. Cymbopogon citratus (lemongrass) and Camellia sinensis (white tea) extracts have been shown to have medicinal properties, however, their activity against lymphoma and leukemia, as well as mechanistic details, have not been fully characterized. Herein, we report potent anti-cancer properties in dose and time-dependent manners of ethanolic lemongrass and hot water white tea extracts in lymphoma and leukemia models. Both extracts were able to effectively induce apoptosis selectively in these human cancer cell types. Interestingly, ethanolic lemongrass extract induces apoptosis primarily by the extrinsic pathway and was found to be dependent on the generation of ROS. Conversely, apoptotic induction by hot water white tea extract was independent of ROS. Furthermore, both of these extracts caused mitochondrial depolarization and decreased rates of oxygen consumption in lymphoma and leukemia cells, leading to cell death. Most importantly, both these extracts were effective in reducing tumor growth in human lymphoma xenograft models when administered orally. Thus, these natural extracts could have potential for being nontoxic alternatives for the treatment of cancer.

20.
BMC Oral Health ; 16: 3, 2016 Jan 13.
Article in English | MEDLINE | ID: mdl-26762247

ABSTRACT

BACKGROUND: Numerous strategies have been proposed to decrease orthodontic treatment time. Photobiomodulation (PBM) has previously been demonstrated to assist in this objective. The aim of this study was to test if intraoral PBM increases the rate of tooth alignment and reduces the time required to resolve anterior dental crowding. METHODS: Nineteen orthodontic subjects with Class I or Class II malocclusion and Little's Irregularity Index (LII) ≥ 3 mm were selected from a pool of applicants, providing 28 total arches. No cases required extraction. The test group (N = 11, 18 arches, 10 upper, 8 lower) received daily PBM treatment with an intraoral LED device (OrthoPulse™, Biolux Research Ltd.) during orthodontic treatment, while the control group (N = 8, 10 arches, 3 upper, 7 lower) received only orthodontic treatment. The PBM device exposed the buccal side of the gums to near-infrared light with a continuous 850-nm wavelength, generating an average daily energy density of 9.5 J/cm(2). LII was measured at the start (T0) of orthodontic treatment until alignment was reached (T1, where LII ≤ 1 mm). The control group was mostly bonded with 0.018-in slot self-ligating SPEED brackets (Hespeler Orthodontics, Cambridge, ON. Canada), while conventionally-ligating Ormco Mini-Diamond twins were used on the PBM group (Ormco, Glendora, Calif. USA). Both groups progressed through alignment with NiTi arch-wires from 0.014-in through to 0.018-in (Ormco), with identical arch-wire changes. The rate of anterior alignment, in LII mm/week, and total treatment time was collected for both groups. Cox proportional hazards models were used to compare groups and while considering age, sex, ethnicity, arch and degree of crowding. RESULTS: The mean alignment rate for the PBM group was significantly higher than that of the control group, with an LII change rate of 1.27 mm/week (SD 0.53, 95 % CI ± 0.26) versus 0.44 mm/week (SD 0.20, 95 % CI ± 0.12), respectively (p = 0.0002). The treatment time to alignment was significantly smaller for the PBM group, which achieved alignment in 48 days (SD 39, 95 % CI ± 39), while the control group took 104 days (SD 55, 95 % CI ±19, p = 0.0053) on average. These results demonstrated that intraoral PBM increased the average rate of tooth movement by 2.9-fold, resulting in a 54 % average decrease in alignment duration versus control. The average PBM compliance to daily treatments was 93 % during alignment. CONCLUSIONS: Under the limitations of this study, the findings suggest that intraoral PBM could be used to decrease anterior alignment treatment time, which could consequently decrease full orthodontic treatment time. However, due to its limitations, further research in the form of a large, randomized trial is needed. TRIAL REGISTRATION: ClinicalTrials.gov NCT02267837 . Registered 10 October 2014.


Subject(s)
Malocclusion/therapy , Phototherapy/methods , Tooth Movement Techniques/methods , Humans , Orthodontic Brackets , Orthodontic Wires , Radiation Dosage , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...