Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 58
Filter
1.
Proc Natl Acad Sci U S A ; 121(20): e2316271121, 2024 May 14.
Article in English | MEDLINE | ID: mdl-38709929

ABSTRACT

Random mutagenesis, including when it leads to loss of gene function, is a key mechanism enabling microorganisms' long-term adaptation to new environments. However, loss-of-function mutations are often deleterious, triggering, in turn, cellular stress and complex homeostatic stress responses, called "allostasis," to promote cell survival. Here, we characterize the differential impacts of 65 nonlethal, deleterious single-gene deletions on Escherichia coli growth in three different growth environments. Further assessments of select mutants, namely, those bearing single adenosine triphosphate (ATP) synthase subunit deletions, reveal that mutants display reorganized transcriptome profiles that reflect both the environment and the specific gene deletion. We also find that ATP synthase α-subunit deleted (ΔatpA) cells exhibit elevated metabolic rates while having slower growth compared to wild-type (wt) E. coli cells. At the single-cell level, compared to wt cells, individual ΔatpA cells display near normal proliferation profiles but enter a postreplicative state earlier and exhibit a distinct senescence phenotype. These results highlight the complex interplay between genomic diversity, adaptation, and stress response and uncover an "aging cost" to individual bacterial cells for maintaining population-level resilience to environmental and genetic stress; they also suggest potential bacteriostatic antibiotic targets and -as select human genetic diseases display highly similar phenotypes, - a bacterial origin of some human diseases.


Subject(s)
Escherichia coli , Stress, Physiological , Escherichia coli/genetics , Escherichia coli/metabolism , Stress, Physiological/genetics , Mutation , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , Gene Deletion , Transcriptome , Gene Expression Regulation, Bacterial , Adaptation, Physiological/genetics , Loss of Function Mutation
2.
Genes (Basel) ; 15(5)2024 Apr 23.
Article in English | MEDLINE | ID: mdl-38790156

ABSTRACT

BRAF mutation identification is important for the diagnosis and treatment of several tumor types, both solid and hematologic. Rapid identification of BRAF mutations is required to determine eligibility for targeted BRAF inhibitor therapy. The Idylla BRAF mutation assay is a rapid, multiplex allele-specific PCR test designed to detect the most common oncogenic BRAF V600 mutations in formalin-fixed paraffin-embedded (FFPE) tissue samples. Here, we describe the validation of the Idylla BRAF mutation assay in our laboratory. During routine clinical practice, we noticed cases in which BRAF V600 mutations were identified with unusual amplification curves, with three cases displaying a delayed amplification within a double amplification pattern and two false-positive calls. We therefore initiated a quality improvement effort to systematically and retrospectively evaluate next-generation sequencing (NGS)-tested cases with BRAF mutations identified within five amino acids of BRAF codon V600 and did not identify additional false-positive cases. We hypothesize that late amplification in a double amplification pattern may represent non-specific amplification, whereas cases displaying single delayed amplification curves may stem from the presence of either non-V600 variants, very low-level V600 variants, cytosine deamination artifacts, and/or non-specific amplification by an allele-specific PCR primer. Regardless, we recommend that Idylla BRAF cases with non-classical amplification curves undergo reflex NGS testing. These findings are likely relevant for other Idylla assays interrogating hotspot mutations in genes such as EGFR, IDH1/2, KRAS, and NRAS.


Subject(s)
High-Throughput Nucleotide Sequencing , Mutation , Proto-Oncogene Proteins B-raf , Humans , Proto-Oncogene Proteins B-raf/genetics , High-Throughput Nucleotide Sequencing/methods , DNA Mutational Analysis/methods , Retrospective Studies , Multiplex Polymerase Chain Reaction/methods , Neoplasms/genetics
3.
Biomedicines ; 12(3)2024 Mar 21.
Article in English | MEDLINE | ID: mdl-38540310

ABSTRACT

Metastatic melanoma has a very poor prognosis. Statins, 3-hydroxy-3-methyl-glutaryl-coenzyme A reductase (HMGCR) inhibitors, are cholesterol-lowering agents with a potential for cancer treatment. The inhibition of HMGCR by statins, however, induces feedback, which paradoxically upregulates HMGCR expression via sterol regulatory element-binding protein-2 (SREBP2). Dipyridamole, an antiplatelet agent, is known to inhibit SREBP2 upregulation. We aimed to demonstrate the efficacy of statin-dipyridamole combination treatment in both human and spontaneously occurring canine melanoma cell lines. The half maximal inhibitory concentration (IC50) of atorvastatin showed a 68-92% reduction when combined with dipyridamole, compared with that of atorvastatin alone. In some melanoma cell lines, cell proliferation was suppressed to almost zero by the combination treatment (≥3 µM atorvastatin). Finally, the BRAF inhibitor, vemurafenib, further potentiated the effects of the combined statin-dipyridamole treatment in BRAF V600E mutation-bearing human melanoma cell lines. In conclusion, the inexpensive and frequently prescribed statin-dipyridamole combination therapy may lead to new developments in the treatment of melanoma and may potentiate the effects of vemurafenib for the targeted therapy of BRAF V600E-mutation bearing melanoma patients. The concordance between the data from canine and human melanoma cell lines reinforces this possibility.

4.
Biochem Biophys Res Commun ; 677: 13-19, 2023 10 15.
Article in English | MEDLINE | ID: mdl-37541087

ABSTRACT

Statins are cholesterol-lowering drugs that have exhibited potential as cancer therapeutic agents. However, as some cancer cells are resistant to statins, broadening an anticancer spectrum of statins is desirable. The upregulated expression of the statin target enzyme, 3-hydroxy-3-methyl-glutaryl-coenzyme A (HMG-CoA) reductase (HMGCR), in statin-treated cancer cells is a well-known mechanism of statin resistance, which can be counteracted by the downregulation of HMGCR gene expression, or degradation of the HMGCR protein. However, the mechanism by which HMGCR degradation influences the anticancer effects of statins remain unreported. We tested the effect of the HMGCR degrader compound SR-12813 at a concentration that did not affect the growth of eight diverse tumor cell lines. Combined treatment with atorvastatin and a low concentration of SR-12813 led to lowering of increased HMGCR expression, and augmented the cytostatic effect of atorvastatin in both statin-resistant and -sensitive cancer cells compared with that of atorvastatin treatment alone. Dual-targeting of HMGCR using statins and SR-12813 (or similar compounds) could provide an improved anticancer therapeutic approach.


Subject(s)
Hydroxymethylglutaryl-CoA Reductase Inhibitors , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Atorvastatin/pharmacology , Up-Regulation , Hydroxymethylglutaryl CoA Reductases/genetics , Hydroxymethylglutaryl CoA Reductases/metabolism
5.
Genes (Basel) ; 14(8)2023 07 28.
Article in English | MEDLINE | ID: mdl-37628603

ABSTRACT

Molecular diagnostics for lung cancer is a well-established standard of care, but how to use the available diagnostic tools for optimal and cost-effective patient care remains unresolved. Here, we show that DNA-only, small gene next-generation sequencing (sNGS) panels (<50 genes) combined with ultra-rapid reflex testing for common fusion transcripts using the Idylla Genefusion assay provide a cost-effective and sufficiently comprehensive testing modality for the majority of lung cancer cases. We also demonstrate the need for additional reflex testing capability on larger DNA and fusion panels for a small subset of lung cancers bearing rare single-nucleotide variants, indels and fusion transcripts and secondary, post-treatment resistance mutations. A similar testing workflow could be adopted for other solid tumor types for which extensive gene/fusion variant profiles are available both in the treatment-naïve and post-therapy settings.


Subject(s)
High-Throughput Nucleotide Sequencing , Lung Neoplasms , Humans , Pathology, Molecular , Lung Neoplasms/diagnosis , Lung Neoplasms/genetics , Biological Assay , Reflex
6.
Heliyon ; 9(7): e18017, 2023 Jul.
Article in English | MEDLINE | ID: mdl-37501994

ABSTRACT

Statins have anticancer effects and may be used as anticancer agents via drug repositioning. In reverse transcription-quantitative polymerase chain reaction (RT-qPCR) assays, the internal reference gene must not be affected by any experimental conditions. As statins exert a wide range of effects on cells by inhibiting the mevalonate pathway, it is possible that statin treatment might alter the expression of housekeeping genes used as internal reference genes, thereby misleading the assessment of obtained gene expression data. Here, we evaluated the expression stability of internal reference genes in atorvastatin-treated cancer cell lines. We treated both statin-sensitive and statin-resistant cancer cell lines with atorvastatin at seven different concentrations and performed RT-qPCR on 15 housekeeping genes whose expression stability was then assessed using five different algorithms. In both statin-sensitive and statin-resistant cancer cell lines, atorvastatin affected the expression of certain internal reference genes in a dose-dependent and cancer cell line-dependent manner; therefore, caution should be exercised when comparing target gene expression between cells. Our findings emphasize the importance of the validation of internal reference genes in gene expression analyses in drug treatment-based cancer research.

7.
Mod Pathol ; 36(11): 100295, 2023 11.
Article in English | MEDLINE | ID: mdl-37517480

ABSTRACT

Poorly differentiated neuroendocrine carcinomas (NECs) are rare malignant neoplasms with aggressive behavior. The diagnosis remains challenging due to ever-changing terminologies and morphologic overlaps with other disease entities. Herein, we seek to better define anorectal NECs by high-risk human papillomavirus (HPV) status and molecular profiling. Fourteen cases, including 3 men and 11 women with a median age of 63 years, were included. High-risk HPV RNA in situ hybridization was diffusely positive (+) in 7 cases, focal rarely positive (+/-) in 2 cases, and completely negative (-) in 5 cases. By morphology, all HPV(-) NECs were large-cell type, 3 mixed with a tubular adenoma/dysplasia or invasive adenocarcinoma. HPV-related (+ or +/-) NECs were mostly small-cell type, 3 mixed with squamous dysplasia and/or squamous cell carcinoma. Immunohistochemically, all NECs were positive for at least 2 neuroendocrine markers. The HPV(-) NECs were also positive for CDX2, whereas all HPV-related NECs were negative or only focally positive for CDX2, p40, and p63. Overexpression of p53 was found in 3 HPV(-) and 2 HPV(+/-) NECs but not in any HPV(+) NECs. Molecular analysis revealed MYC gene amplification in 4 cases: 2 HPV(-), 1 HPV(+/-), and 1 HPV(+). This was confirmed by fluorescence in situ hybridization in all but 1 HPV(-) NEC, which showed polysomy 8 but no true MYC amplification. Interestingly, only 2 of the 4 MYC amplification-bearing cases, both p53 normal/wild-type, expressed c-Myc protein by immunohistochemistry. The other 2 cases, both p53 overexpressed, did not show c-Myc expression despite true MYC amplification. Our study demonstrates that anorectal NECs arise in HPV-dependent or -independent pathways, with heterogeneous expression of other lineage markers and different molecular signatures. Expressions of p53 and c-Myc proteins appear to be mutually exclusive regardless of HPV status, likely mediating alternative mechanisms of NEC carcinogenesis.


Subject(s)
Carcinoma, Neuroendocrine , Papillomavirus Infections , Male , Humans , Female , Middle Aged , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Human Papillomavirus Viruses , Papillomavirus Infections/complications , Papillomavirus Infections/pathology , In Situ Hybridization, Fluorescence , Carcinoma, Neuroendocrine/pathology , Carcinogenesis
8.
Article in English | MEDLINE | ID: mdl-37160316

ABSTRACT

Autologous and allogeneic hematopoietic stem cell transplantation (HSCT) has revolutionized the therapy of hematolymphoid malignancies. Yet, how to best detect or predict the emergence of HSCT-related complications remain unresolved. Here, we describe a case of donor-derived, transient Alpha Beta (αß) T-cell large granular clonal lymphocytosis and cytopenia that emerged post-HSCT in a patient with a history of gamma delta (γδ) T-cell large granular lymphocytic leukemia (T-LGLL). Clonal unrelatedness of post-transplant T-LGL lymphocytosis to the patient's pretransplant T-LGLL was first identified by T-cell receptor (TCR) PCR showing different sized fragments of rearranged gamma chains, in addition to shift from γδ to αß TCR expression by flow cytometry analyses. Donor-derivation of the patient's post-transplant clonal lymphocytosis was confirmed by serial chimerism analyses of recipient's blood specimens demonstrating 100% donor DNA. Moreover, oncogenic DNMT3A and RUNX1 mutations were detected by next-generation sequencing (NGS) only in post-transplant specimens. Intriguingly, despite continued increase in DNMT3A and RUNX1 mutation load, the patient's clonal lymphocytosis and anemia eventually largely resolved; yet, the observed mutation profile with persistent thrombocytopenia indicated secondary clonal cytopenia of undetermined significance (CCUS) in the absence of overt morphologic evidence of myeloid neoplasm in the marrow. This case illustrates the utility of longitudinal chimerism analysis and NGS testing combined with flow cytometric immunophenotyping to evaluate emerging donor-derived hematolymphoid processes and to properly interpret partial functional engraftment. It may also support the notion that driver mutation-induced microenvironmental changes may paradoxically contribute to reestablishing tissue homeostasis.


Subject(s)
Leukemia, Large Granular Lymphocytic , Lymphocytosis , Humans , Leukemia, Large Granular Lymphocytic/genetics , Lymphocytosis/genetics , Core Binding Factor Alpha 2 Subunit , Clonal Hematopoiesis , DNA Modification Methylases , T-Lymphocytes
9.
Life Sci ; 312: 121249, 2023 Jan 01.
Article in English | MEDLINE | ID: mdl-36455649

ABSTRACT

AIMS: Statins, cholesterol-lowering drugs, are potential therapeutic agents for inhibiting cancer proliferation. However, the mechanisms that mediate the effects of statins, the homeostatic responses of tumor cells to statin therapy, and the modes underlying the antitumor effects of statins remain unclear. MAIN METHODS: To uncover the effects of statins on cancer cells in vitro, we performed transcriptome and metabolome analyses on atorvastatin-treated statin-resistant and statin-sensitive lung cancer cells. KEY FINDINGS: The results of Gene Ontology terms and pathway enrichment analyses showed that after 24 h of atorvastatin treatment, the expression of cell cycle- and DNA replication-related genes was significantly decreased in the statin-sensitive cancer cells. The results of metabolome analysis showed that the components of polyamine metabolism and purine metabolism, glycolysis, and pentose phosphate pathway were decreased in the statin-sensitive cancer cells. SIGNIFICANCE: Differences in cellular properties between statin-sensitive and statin-resistant cancer cells revealed additional candidates for therapeutic targets in statin-treated cancer cells and suggested that inhibiting these metabolic pathways could improve efficacy. In conclusion, combining statins with inhibitors of polyamine metabolism (cell proliferation and protein translation), purine metabolism (DNA synthesis), glycolytic system (energy production), and pentose phosphate pathway (antioxidant stress) might enhance the anticancer effects of statins.


Subject(s)
Hydroxymethylglutaryl-CoA Reductase Inhibitors , Neoplasms , Mevalonic Acid , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Hydroxymethylglutaryl-CoA Reductase Inhibitors/therapeutic use , Atorvastatin/pharmacology , Polyamines , Purines , Neoplasms/drug therapy , Neoplasms/genetics
10.
Article in English | MEDLINE | ID: mdl-35732498

ABSTRACT

Liquid biopsy is considered an alternative to standard next-generation sequencing (NGS) of solid tumor samples when biopsy tissue is inadequate for testing or when testing of a peripheral blood sample is preferred. A common assumption of liquid biopsies is that the NGS data obtained on circulating cell-free DNA is a high-fidelity reflection of what would be found by solid tumor testing. Here, we describe a case that challenges this widely held assumption. A patient diagnosed with lung carcinoma showed pathogenic IDH1 and TP53 mutations by liquid biopsy NGS at an outside laboratory. Subsequent in-house NGS of a metastatic lymph node fine-needle aspiration (FNA) sample revealed two pathogenic EGFR mutations. Morphologic and immunophenotypic assessment of the patient's blood sample identified acute myeloid leukemia, with in-house NGS confirming and identifying pathogenic IDH1, TP53, and BCOR mutations, respectively. This case, together with a few similar reports, demonstrates that caution is needed when interpreting liquid biopsy NGS results, especially if they are inconsistent with the presumptive diagnosis. Our case suggests that routine parallel sequencing of peripheral white blood cells would substantially increase the fidelity of the obtained liquid biopsy results.


Subject(s)
Leukemia, Myeloid, Acute , Lung Neoplasms , Biopsy, Fine-Needle/methods , DNA Mutational Analysis/methods , High-Throughput Nucleotide Sequencing/methods , Humans , Incidental Findings , Leukemia, Myeloid, Acute/diagnosis , Leukemia, Myeloid, Acute/genetics , Liquid Biopsy/methods , Lung/pathology , Lung Neoplasms/diagnosis , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Mutation
11.
Genes (Basel) ; 13(5)2022 04 29.
Article in English | MEDLINE | ID: mdl-35627184

ABSTRACT

Testing of tumors by next generation sequencing (NGS) is impacted by relatively long turnaround times and a need for highly trained personnel. Recently, Idylla oncology assays were introduced to test for BRAF, EGFR, KRAS, and NRAS common hotspot mutations that do not require specialized trained personnel. Moreover, the interpretation of results is fully automated, with rapid turnaround time. Though Idylla testing and NGS have been shown to have high concordance in identifying EGFR, BRAF, KRAS, and NRAS hotspot mutations, there is limited experience on optimal ways the Idylla system can be used in routine practice. We retrospectively evaluated all cases with EGFR, BRAF, KRAS, or NRAS mutations identified in clinical specimens sequenced on two different NGS panels at the University of Rochester Medical Center (URMC) molecular diagnostics laboratory between July 2020 and July 2021 and assessed if these mutations would be detected by the Idylla cartridges if used. We found that the Idylla system could accurately identify Tier 1 or 2 actionable genomic alterations in select associated disease pathologies if used. Yet, in a minority of cases, we would have been unable to detect NGS-identified pathogenic mutations due to their absence on the Idylla panels. We derived algorithmic practice guidelines for the use of the Idylla cartridges. Overall, Idylla molecular testing could be implemented either as a first-line standalone diagnostic tool in select indications or for orthogonal confirmation of uncertain results.


Subject(s)
Neoplasms , Proto-Oncogene Proteins B-raf , DNA Mutational Analysis/methods , ErbB Receptors/genetics , High-Throughput Nucleotide Sequencing/methods , Humans , Multiplex Polymerase Chain Reaction , Mutation , Neoplasms/diagnosis , Neoplasms/genetics , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins p21(ras)/genetics , Retrospective Studies
12.
Sci Rep ; 11(1): 12763, 2021 06 17.
Article in English | MEDLINE | ID: mdl-34140545

ABSTRACT

Epithelial-mesenchymal transition (EMT) in primary tumor cells is a key prerequisite for metastasis initiation. Statins, cholesterol-lowering drugs, can delay metastasis formation in vivo and attenuate the growth and proliferation of tumor cells in vitro. The latter effect is stronger in tumor cells with a mesenchymal-like phenotype than in those with an epithelial one. However, the effect of statins on epithelial cancer cells treated with EMT-inducing growth factors such as transforming growth factor-ß (TGF-ß) remains unclear. Here, we examined the effect of atorvastatin on two epithelial cancer cell lines following TGF-ß treatment. Atorvastatin-induced growth inhibition was stronger in TGF-ß-treated cells than in cells not thusly treated. Moreover, treatment of cells with atorvastatin prior to TGF-ß treatment enhanced this effect, which was further potentiated by the simultaneous reduction in the expression of the statin target enzyme, 3-hydroxy-3-methylglutaryl coenzyme A reductase (HMGCR). Dual pharmacological targeting of HMGCR can thus strongly inhibit the growth and proliferation of epithelial cancer cells treated with TGF-ß and may also improve statin therapy-mediated attenuation of metastasis formation in vivo.


Subject(s)
Atorvastatin/pharmacology , Hydroxymethylglutaryl CoA Reductases/metabolism , Neoplasms/pathology , Transforming Growth Factor beta/pharmacology , Biomarkers, Tumor/metabolism , Cell Count , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Size/drug effects , Epithelial-Mesenchymal Transition/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Humans , Models, Biological , RNA, Messenger/genetics , RNA, Messenger/metabolism
13.
Article in English | MEDLINE | ID: mdl-33832922

ABSTRACT

Somatic mutations in hotspot regions of the cytosolic or mitochondrial isoforms of the isocitrate dehydrogenase gene (IDH1 and IDH2, respectively) contribute to the pathogenesis of acute myeloid leukemia (AML) by producing the oncometabolite 2-hydroxyglutarate (2-HG). The allosteric IDH1 inhibitor, ivosidenib, suppresses 2-HG production and induces clinical responses in relapsed/refractory IDH1-mutant AML. Herein, we describe a clinical case of AML in which we detected the neomorphic IDH1 p.R132C mutation in consecutive patient samples with a mutational hotspot targeted next-generation sequencing (NGS) assay. The patient had a clinical response to ivosidenib, followed by relapse and disease progression. Subsequent sequencing of the relapsed sample using a newly developed all-exon, hybrid-capture-based NGS panel identified an additional IDH1 p.S280F mutation known to cause renewed 2-HG production and drug resistance. Structural modeling confirmed that serine-to-phenylalanine substitution at this codon sterically hinders ivosidenib from binding to the mutant IDH1 dimer interface and predicted a similar effect on the pan-IDH inhibitor AG-881. Joint full-exon NGS and structural modeling enables monitoring IDH1 inhibitor-treated AML patients for acquired drug resistance and choosing follow-up therapy.


Subject(s)
Drug Resistance, Neoplasm/genetics , Enzyme Inhibitors/pharmacology , Exons , Isocitrate Dehydrogenase/drug effects , Isocitrate Dehydrogenase/genetics , Leukemia, Myeloid, Acute/genetics , Aged , Binding Sites , Enzyme Inhibitors/chemistry , Female , Genetic Predisposition to Disease/genetics , Glycine/analogs & derivatives , Glycine/therapeutic use , High-Throughput Nucleotide Sequencing , Humans , Isocitrate Dehydrogenase/chemistry , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/pathology , Mutation , Pyridines , Recurrence
14.
Article in English | MEDLINE | ID: mdl-32532878

ABSTRACT

The ATP-binding cassette transporter member A3 (ABCA3) is a lipid transporter with a critical function in pulmonary surfactant biogenesis. Biallelic loss-of-function mutations in ABCA3 result in severe surfactant deficiency leading to neonatal respiratory failure with death in the first year of life. Herein, we describe a newborn with severe respiratory distress at birth progressing to respiratory failure requiring transplant. This patient was found to have a maternally inherited frameshift loss-of-function ABCA3 mutation and a paternally inherited synonymous variant in ABCA3 predicted to create a cryptic splice site. Additional studies showed reduced ABCA3 expression in hyperplastic alveolar epithelial type II cells and lamellar body alterations characteristic of ABCA3 deficiency, leading to a diagnosis of autosomal recessive ABCA3-related pulmonary surfactant dysfunction. This case highlights the need for an integrated, comprehensive approach for the diagnosis of inherited diseases when in silico modeling is utilized in the interpretation of key novel genetic mutations.


Subject(s)
ATP-Binding Cassette Transporters/genetics , Genetic Association Studies , Heterozygote , Mutation , Phenotype , Respiratory Insufficiency/diagnosis , Respiratory Insufficiency/genetics , ATP-Binding Cassette Transporters/metabolism , Biopsy , DNA Mutational Analysis , Disease Progression , Genetic Association Studies/methods , High-Throughput Nucleotide Sequencing , Humans , Immunohistochemistry , Infant, Newborn , Lung Transplantation , Male , Respiratory Function Tests , Respiratory Insufficiency/surgery , Treatment Outcome
15.
Bioinformatics ; 36(10): 3084-3092, 2020 05 01.
Article in English | MEDLINE | ID: mdl-32101277

ABSTRACT

MOTIVATION: The biological effects of human missense variants have been studied experimentally for decades but predicting their effects in clinical molecular diagnostics remains challenging. Available computational tools are usually based on the analysis of sequence conservation and structural properties of the mutant protein. We recently introduced a new machine learning method that demonstrated for the first time the significance of protein dynamics in determining the pathogenicity of missense variants. RESULTS: Here, we present a new interface (Rhapsody) that enables fully automated assessment of pathogenicity, incorporating both sequence coevolution data and structure- and dynamics-based features. Benchmarked against a dataset of about 20 000 annotated variants, the methodology is shown to outperform well-established and/or advanced prediction tools. We illustrate the utility of Rhapsody by in silico saturation mutagenesis studies of human H-Ras, phosphatase and tensin homolog and thiopurine S-methyltransferase. AVAILABILITY AND IMPLEMENTATION: The new tool is available both as an online webserver at http://rhapsody.csb.pitt.edu and as an open-source Python package (GitHub repository: https://github.com/prody/rhapsody; PyPI package installation: pip install prody-rhapsody). Links to additional resources, tutorials and package documentation are provided in the 'Python package' section of the website. SUPPLEMENTARY INFORMATION: Supplementary data are available at Bioinformatics online.


Subject(s)
Documentation , Software , Computational Biology , Computer Simulation , Humans , Virulence
16.
Proc Natl Acad Sci U S A ; 116(24): 11770-11775, 2019 06 11.
Article in English | MEDLINE | ID: mdl-31127043

ABSTRACT

The mechanisms of bacterial chemotaxis have been extensively studied for several decades, but how the physical environment influences the collective migration of bacterial cells remains less understood. Previous models of bacterial chemotaxis have suggested that the movement of migrating bacteria across obstacle-laden terrains may be slower compared with terrains without them. Here, we show experimentally that the size or density of evenly spaced obstacles do not alter the average exit rate of Escherichia coli cells from microchambers in response to external attractants, a function that is dependent on intact cell-cell communication. We also show, both by analyzing a revised theoretical model and by experimentally following single cells, that the reduced exit time in the presence of obstacles is a consequence of reduced tumbling frequency that is adjusted by the E. coli cells in response to the topology of their environment. These findings imply operational short-term memory of bacteria while moving through complex environments in response to chemotactic stimuli and motivate improved algorithms for self-autonomous robotic swarms.


Subject(s)
Chemotaxis/physiology , Escherichia coli/physiology , Cell Communication/physiology , Movement/physiology
17.
Br J Cancer ; 119(9): 1094-1105, 2018 10.
Article in English | MEDLINE | ID: mdl-30401978

ABSTRACT

BACKGROUND: Metastasis in breast cancer foreshadows mortality, as clinically evident disease is aggressive and generally chemoresistant. Disseminated breast cancer cells often enter a period of dormancy for years to decades before they emerge as detectable cancers. Harboring of these dormant cells is not individually predictable, and available information suggests that these micrometastatic foci cannot be effectively targeted by existing therapies. As such, long-term, relatively non-toxic interventions that prevent metastatic outgrowth would be an advance in treatment. Epidemiological studies have found that statins reduce breast cancer specific mortality but not the incidence of primary cancer. However, the means by which statins reduce mortality without affecting primary tumor development remains unclear. METHODS: We examine statin efficacy against two breast cancer cell lines in models of breast cancer metastasis: a 2D in vitro co-culture model of breast cancer cell interaction with the liver, a 3D ex vivo microphysiological system model of breast cancer metastasis, and two independent mouse models of spontaneous breast cancer metastasis to the lung and liver, respectively. RESULTS: We demonstrate that statins can directly affect the proliferation of breast cancer cells, specifically at the metastatic site. In a 2D co-culture model of breast cancer cell interaction with the liver, we demonstrate that atorvastatin can directly suppress proliferation of mesenchymal but not epithelial breast cancer cells. Further, in an ex vivo 3D liver microphysiological system of breast cancer metastasis, we found that atorvastatin can block stimulated emergence of dormant breast cancer cells. In two independent models of spontaneous breast cancer metastasis to the liver and to the lung, we find that statins significantly reduce proliferation of the metastatic but not primary tumor cells. CONCLUSIONS: As statins can block metastatic tumor outgrowth, they should be considered for use as long-term adjuvant drugs to delay clinical emergence and decrease mortality in breast cancer patients.


Subject(s)
Atorvastatin/administration & dosage , Breast Neoplasms/drug therapy , Hydroxymethylglutaryl-CoA Reductase Inhibitors/administration & dosage , Liver Neoplasms/drug therapy , Lung Neoplasms/drug therapy , Animals , Atorvastatin/pharmacology , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Coculture Techniques , Epithelial-Mesenchymal Transition , Female , Hepatocytes/cytology , Hepatocytes/drug effects , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Liver Neoplasms/secondary , Lung Neoplasms/secondary , MCF-7 Cells , Mice , Xenograft Model Antitumor Assays
18.
Breast Cancer Res ; 20(1): 144, 2018 11 20.
Article in English | MEDLINE | ID: mdl-30458856

ABSTRACT

Epidemiologic studies have, variably, shown the concomitant use of statin drugs to be beneficial to cancer outcomes. Statin drugs have been FDA approved for three decades for the treatment of high cholesterol and atherosclerotic coronary artery disease and are widely used. This has engendered studies as to their influence on concomitant diseases, including cancers. In this context, statin use has been correlated, variably, with a decrease in deaths from breast cancer. However, there is no extant model for this effect, and the extent of efficacy is open to question.The overarching goal of this article is to communicate to the reader of the potential of statins to reduce breast cancer progression and mortality. This is the use as a secondary prevention measure, and not as a therapy to directly counter active cancer. First, salient aspects of statin pharmacology, as relates to cardiovascular disease, will be discussed. Second, the basic and clinical research studies that investigate statin usage in breast cancer will be presented. Additionally, statin effects in other cancer types will be included for context. Finally, proposals for future basic and clinical research studies to determine the role of statins in breast cancer management will be presented.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Breast Neoplasms/drug therapy , Cardiovascular Diseases/drug therapy , Hydroxymethylglutaryl-CoA Reductase Inhibitors/therapeutic use , Neoplasm Recurrence, Local/prevention & control , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Breast Neoplasms/mortality , Breast Neoplasms/prevention & control , Cardiovascular Diseases/epidemiology , Clinical Trials as Topic , Comorbidity , Disease Progression , Female , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Incidence , Neoplasm Recurrence, Local/epidemiology , Treatment Outcome
19.
Oncotarget ; 9(50): 29304-29315, 2018 Jun 29.
Article in English | MEDLINE | ID: mdl-30034619

ABSTRACT

HMG-CoA reductase (HMGCR) inhibitors, statins, are potent cholesterol reducing drugs that exhibit anti-tumor effects in vitro and in animal models, including attenuation of metastasis formation, and their use correlates with reduced cancer-specific mortality in retrospective human cohort studies. However, E-cadherin expressing epithelial- and mixed epithelial-mesenchymal cancer cell lines (reflective of primary and outgrowing metastatic tumor cells, respectively) require higher statin concentrations than mesenchymal-like tumor cells (reflective of in-circulation metastatic tumor cells) to achieve the same degree of growth inhibition. Here, we show that attenuation of HMGCR expression in the presence of atorvastatin leads to stronger growth inhibition than dual target blockade of the mevalonate pathway in relatively statin resistant cell lines, mainly through inhibition of protein prenylation pathways. Thus, combined inhibition of the mevalonate pathway's rate-limiting enzyme, HMGCR, can improve atorvastatin's growth inhibitory effect on epithelial- and mixed mesenchymal-epithelial cancer cells, a finding that may have implications for the design of future anti-metastatic cancer therapies.

20.
Sci Rep ; 8(1): 9388, 2018 06 20.
Article in English | MEDLINE | ID: mdl-29925909

ABSTRACT

Cell-to-cell differences in protein expression in normal tissues and tumors are a common phenomenon, but the underlying principles that govern this heterogeneity are largely unknown. Here, we show that in monolayer cancer cell-line cultures, the expression of the five metabolic enzymes of serine-glycine synthesis (SGS), including its rate-limiting enzyme, phosphoglycerate dehydrogenase (PHGDH), displays stochastic cell-to-cell variation. By contrast, in cancer cell line-derived three-dimensional (3D) microtumors PHGDH expression is restricted to the outermost part of the microtumors' outer proliferative cell layer, while the four other SGS enzymes display near uniform expression throughout the microtumor. A mathematical model suggests that metabolic stress in the microtumor core activates factors that restrict PHGDH expression. Thus, intracellular enzyme expression in growing cell ecosystems can shift to spatially ordered patterns in 3D structured environments due to emergent cell-cell communication, with potential implications for the design of effective anti-metabolic cancer therapies.


Subject(s)
Phosphoglycerate Dehydrogenase/metabolism , Cell Communication/physiology , Cell Line, Tumor , Cell Proliferation/physiology , Ecosystem , Glycine/metabolism , Humans , Ki-67 Antigen/metabolism , Models, Theoretical , Serine/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...