Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
1.
Nature ; 606(7913): 396-405, 2022 06.
Article in English | MEDLINE | ID: mdl-35650435

ABSTRACT

Disseminated cancer cells from primary tumours can seed in distal tissues, but may take several years to form overt metastases, a phenomenon that is termed tumour dormancy. Despite its importance in metastasis and residual disease, few studies have been able to successfully characterize dormancy within melanoma. Here we show that the aged lung microenvironment facilitates a permissive niche for efficient outgrowth of dormant disseminated cancer cells-in contrast to the aged skin, in which age-related changes suppress melanoma growth but drive dissemination. These microenvironmental complexities can be explained by the phenotype switching model, which argues that melanoma cells switch between a proliferative cell state and a slower-cycling, invasive state1-3. It was previously shown that dermal fibroblasts promote phenotype switching in melanoma during ageing4-8. We now identify WNT5A as an activator of dormancy in melanoma disseminated cancer cells within the lung, which initially enables the efficient dissemination and seeding of melanoma cells in metastatic niches. Age-induced reprogramming of lung fibroblasts increases their secretion of the soluble WNT antagonist sFRP1, which inhibits WNT5A in melanoma cells and thereby enables efficient metastatic outgrowth. We also identify the tyrosine kinase receptors AXL and MER as promoting a dormancy-to-reactivation axis within melanoma cells. Overall, we find that age-induced changes in distal metastatic microenvironments promote the efficient reactivation of dormant melanoma cells in the lung.


Subject(s)
Aging , Lung , Melanoma , Neoplasm Metastasis , Stromal Cells , Tumor Microenvironment , Aged , Aging/pathology , Fibroblasts/pathology , Humans , Lung/pathology , Melanoma/pathology , Neoplasm Invasiveness/pathology , Neoplasm Metastasis/pathology , Neoplasm, Residual , Proto-Oncogene Proteins , Receptor Protein-Tyrosine Kinases , Skin/pathology , Stromal Cells/pathology , Wnt-5a Protein , c-Mer Tyrosine Kinase , Axl Receptor Tyrosine Kinase
2.
Mol Cell ; 77(3): 633-644.e5, 2020 02 06.
Article in English | MEDLINE | ID: mdl-31836388

ABSTRACT

Metastatic melanoma is an aggressive disease, despite recent improvements in therapy. Eradicating all melanoma cells even in drug-sensitive tumors is unsuccessful in patients because a subset of cells can transition to a slow-cycling state, rendering them resistant to most targeted therapy. It is still unclear what pathways define these subpopulations and promote this resistant phenotype. In the current study, we show that Wnt5A, a non-canonical Wnt ligand that drives a metastatic, therapy-resistant phenotype, stabilizes the half-life of p53 and uses p53 to initiate a slow-cycling state following stress (DNA damage, targeted therapy, and aging). Inhibiting p53 blocks the slow-cycling phenotype and sensitizes melanoma cells to BRAF/MEK inhibition. In vivo, this can be accomplished with a single dose of p53 inhibitor at the commencement of BRAF/MEK inhibitor therapy. These data suggest that taking the paradoxical approach of inhibiting rather than activating wild-type p53 may sensitize previously resistant metastatic melanoma cells to therapy.


Subject(s)
Melanoma/metabolism , Tumor Suppressor Protein p53/genetics , Wnt-5a Protein/metabolism , Cell Line, Tumor , Drug Resistance, Neoplasm/genetics , Humans , MAP Kinase Kinase Kinases/metabolism , Melanoma/genetics , Melanoma/pathology , Molecular Targeted Therapy , Mutation/drug effects , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins B-raf/metabolism , Signal Transduction/drug effects , Sulfonamides/pharmacology , Tumor Microenvironment/drug effects , Tumor Suppressor Protein p53/physiology
3.
BMC Bioinformatics ; 25(1): 21, 2024 Jan 12.
Article in English | MEDLINE | ID: mdl-38216886

ABSTRACT

BACKGROUND: Metagene plots provide a visualization of biological signal trends over subsections of the genome and are used to perform high-level analysis of experimental data by aggregating genome-level data to create an average profile. The generation of metagene plots is useful for summarizing the results of many sequencing-based applications. Despite their prevalence and utility, the standard metagene plot is blind to conflicting signals within data. If multiple distinct trends occur, they can interact destructively, creating a plot that does not accurately represent any of the underlying trends. RESULTS: We present MetageneCluster, a Python tool to generate a collection of representative metagene plots based on k-means clustering of genomic regions of interest. Clustering the data by similarity allows us to identify patterns within the features of interest. We are then able to summarize each pattern present in the data, rather than averaging across the entire feature space. We show that our method performs well when used to identify conflicting signals in real-world genome-level data. CONCLUSIONS: Overall, MetageneCluster is a user-friendly tool for the creation of metagene plots that capture distinct patterns in underlying sequence data.


Subject(s)
Genome , Genomics , Genomics/methods , Software
5.
JID Innov ; 2(2): 100076, 2022 Mar.
Article in English | MEDLINE | ID: mdl-35146482

ABSTRACT

Murine cancer cell lines are powerful research tools to complement studies in genetically engineered mouse models. We have established 21 melanoma cell lines from embryonic stem cell-genetically engineered mouse models driven by alleles that model the most frequent genetic alterations in human melanoma. In addition, these cell lines harbor regulatory alleles for the genomic integration of transgenes and the regulation of expression of such transgenes. In this study, we report a comprehensive characterization of these cell lines. Specifically, we validated melanocytic origin, driver allele recombination and expression, and activation of the oncogenic MAPK and protein kinase B pathways. We further tested tumor formation in syngeneic immunocompetent recipients as well as the functionality of the integrated Tet-ON system and recombination-mediated cassette exchange homing cassette. Finally, by deleting the transcription factor MAFG with an inducible CRISPR/Cas9 approach, we show the utility of the regulatory alleles for candidate gene modulation. These cell lines will be a valuable resource for studying melanoma biology and therapy.

6.
J Clin Invest ; 131(6)2021 03 15.
Article in English | MEDLINE | ID: mdl-33720046

ABSTRACT

Continued thinning of the atmospheric ozone, which protects the earth from damaging ultraviolet radiation (UVR), will result in elevated levels of UVR reaching the earth's surface, leading to a drastic increase in the incidence of skin cancer. In addition to promoting carcinogenesis in skin cells, UVR is a potent extrinsic driver of age-related changes in the skin known as "photoaging." We are in the preliminary stages of understanding of the role of intrinsic aging in melanoma, and the tumor-permissive effects of photoaging on the skin microenvironment remain largely unexplored. In this Review, we provide an overview of the impact of UVR on the skin microenvironment, addressing changes that converge or diverge with those observed in intrinsic aging. Intrinsic and extrinsic aging promote phenotypic changes to skin cell populations that alter fundamental processes such as melanogenesis, extracellular matrix deposition, inflammation, and immune response. Given the relevance of these processes in cancer, we discuss how photoaging might render the skin microenvironment permissive to melanoma progression.


Subject(s)
Melanoma/etiology , Skin Aging/radiation effects , Skin Neoplasms/etiology , Tumor Microenvironment/radiation effects , Aging/immunology , Aging/metabolism , Aging/pathology , Animals , Disease Progression , Extracellular Matrix/radiation effects , Humans , Immune Tolerance/radiation effects , Melanins/biosynthesis , Melanoma/immunology , Melanoma/metabolism , Mice , Receptors, Aryl Hydrocarbon/metabolism , Skin/immunology , Skin/metabolism , Skin/radiation effects , Skin Neoplasms/immunology , Skin Neoplasms/metabolism , Ultraviolet Rays/adverse effects , Urocanic Acid/metabolism , Vitamin D/metabolism
7.
Cancer Res ; 81(3): 658-670, 2021 02 01.
Article in English | MEDLINE | ID: mdl-33262126

ABSTRACT

Metastatic dissemination remains a significant barrier to successful therapy for melanoma. Wnt5A is a potent driver of invasion in melanoma and is believed to be secreted from the tumor microenvironment (TME). Our data suggest that myeloid-derived suppressor cells (MDSC) in the TME are a major source of Wnt5A and are reliant upon Wnt5A for multiple actions. Knockdown of Wnt5A specifically in the myeloid cells demonstrated a clear decrease in Wnt5A expression within the TME in vivo as well as a decrease in intratumoral MDSC and regulatory T cell (Treg). Wnt5A knockdown also decreased the immunosuppressive nature of MDSC and decreased expression of TGFß1 and arginase 1. In the presence of Wnt5A-depleted MDSC, tumor-infiltrating lymphocytes expressed decreased PD-1 and LAG3, suggesting a less exhausted phenotype. Myeloid-specific Wnt5A knockdown also led to decreased lung metastasis. Tumor-infiltrating MDSC from control animals showed a strong positive correlation with Treg, which was completely ablated in animals with Wnt5A-negative MDSC. Overall, our data suggest that while MDSC contribute to an immunosuppressive and less immunogenic environment, they exhibit an additional function as the major source of Wnt5A in the TME. SIGNIFICANCE: These findings demonstrate that myeloid cells provide a major source of Wnt5A to facilitate metastatic potential in melanoma cells and rely on Wnt5A for their immunosuppressive function.


Subject(s)
Melanoma/metabolism , Myeloid-Derived Suppressor Cells/metabolism , Tumor Microenvironment , Wnt-5a Protein/metabolism , Animals , Antigens, CD/metabolism , Arginase/metabolism , Cell Line, Tumor , Female , Lung Neoplasms/secondary , Lymphocytes, Tumor-Infiltrating/metabolism , Male , Melanoma/secondary , Mice , Mice, Inbred C57BL , Mice, Transgenic , Myeloid-Derived Suppressor Cells/immunology , Neoplasm Invasiveness , Programmed Cell Death 1 Receptor/metabolism , T-Lymphocytes, Regulatory/metabolism , Transforming Growth Factor beta1/metabolism , Lymphocyte Activation Gene 3 Protein
8.
Clin Cancer Res ; 26(21): 5709-5719, 2020 11 01.
Article in English | MEDLINE | ID: mdl-33097493

ABSTRACT

PURPOSE: Angiogenesis is thought to be critical for tumor metastasis. However, inhibiting angiogenesis using antibodies such as bevacizumab (Avastin), has had little impact on melanoma patient survival. We have demonstrated that both angiogenesis and metastasis are increased in older individuals, and therefore sought to investigate whether there was an age-related difference in response to bevacizumab, and if so, what the underlying mechanism could be. EXPERIMENTAL DESIGN: We analyzed data from the AVAST-M trial of 1,343 patients with melanoma treated with bevacizumab to determine whether there is an age-dependent response to bevacizumab. We also examined the age-dependent expression of VEGF and its cognate receptors in patients with melanoma, while using syngeneic melanoma animal models to target VEGF in young versus old mice. We also examined the age-related proangiogenic factor secreted frizzled-related protein 2 (sFRP2) and whether it could modulate response to anti-VEGF therapy. RESULTS: We show that older patients respond poorly to bevacizumab, whereas younger patients show improvement in both disease-free survival and overall survival. We find that targeting VEGF does not ablate angiogenesis in an aged mouse model, while sFRP2 promotes angiogenesis in vitro and in young mice. Targeting sFRP2 in aged mice successfully ablates angiogenesis, while the effects of targeting VEGF in young mice can be overcome by increasing sFRP2. CONCLUSIONS: VEGF is decreased during aging, thereby reducing response to bevacizumab. Despite the decrease in VEGF, angiogenesis is increased because of an increase in sFRP2 in the aged tumor microenvironment. These results stress the importance of considering age as a factor for designing targeted therapies.


Subject(s)
Melanoma/genetics , Membrane Proteins/genetics , Neovascularization, Pathologic/genetics , Vascular Endothelial Growth Factor A/genetics , Age Factors , Aged , Aged, 80 and over , Animals , Bevacizumab/administration & dosage , Cell Line, Tumor , Disease-Free Survival , Gene Expression Regulation, Neoplastic/drug effects , Humans , Melanoma/drug therapy , Melanoma/pathology , Mice , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/pathology , Tumor Microenvironment/drug effects
9.
Cancer Discov ; 10(9): 1282-1295, 2020 09.
Article in English | MEDLINE | ID: mdl-32499221

ABSTRACT

Older patients with melanoma (>50 years old) have poorer prognoses and response rates to targeted therapy compared with young patients (<50 years old), which can be driven, in part, by the aged microenvironment. Here, we show that aged dermal fibroblasts increase the secretion of neutral lipids, especially ceramides. When melanoma cells are exposed to the aged fibroblast lipid secretome, or cocultured with aged fibroblasts, they increase the uptake of lipids via the fatty acid transporter FATP2, which is upregulated in melanoma cells in the aged microenvironment and known to play roles in lipid synthesis and accumulation. We show that blocking FATP2 in melanoma cells in an aged microenvironment inhibits their accumulation of lipids and disrupts their mitochondrial metabolism. Inhibiting FATP2 overcomes age-related resistance to BRAF/MEK inhibition in animal models, ablates tumor relapse, and significantly extends survival time in older animals. SIGNIFICANCE: These data show that melanoma cells take up lipids from aged fibroblasts, via FATP2, and use them to resist targeted therapy. The response to targeted therapy is altered in aged individuals because of the influences of the aged microenvironment, and these data suggest FATP2 as a target to overcome resistance.See related commentary by Montal and White, p. 1255.This article is highlighted in the In This Issue feature, p. 1241.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Coenzyme A Ligases/metabolism , Fibroblasts/metabolism , Melanoma/drug therapy , Skin Neoplasms/drug therapy , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Cell Line, Tumor , Cellular Senescence , Coculture Techniques , Coenzyme A Ligases/antagonists & inhibitors , Dermis/cytology , Dermis/pathology , Drug Resistance, Neoplasm/drug effects , Humans , Keratinocytes/metabolism , Lipid Metabolism , Melanoma/pathology , Molecular Targeted Therapy/methods , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Skin Neoplasms/pathology , Tumor Microenvironment
10.
Pigment Cell Melanoma Res ; 32(2): 237-247, 2019 03.
Article in English | MEDLINE | ID: mdl-30216694

ABSTRACT

This review will focus on the role of the tumor microenvironment (TME) in the development of drug resistance in melanoma. Resistance to mitogen-activated protein kinase inhibitors (MAPKi) in melanoma is observed months after treatment, a phenomenon that is often attributed to the incredible plasticity of melanoma cells but may also depend on the TME. The TME is unique in its cellular composition-it contains fibroblasts, immune cells, endothelial cells, adipocytes, and among others. In addition, the TME provides "non-homeostatic" levels of oxygen, nutrients (hypoxia and metabolic stress), and extracellular matrix proteins, creating a pro-tumorigenic niche that drives resistance to MAPKi treatment. In this review, we will focus on how changes in the tumor microenvironment regulate MAPKi resistance.


Subject(s)
Drug Resistance, Neoplasm , Melanoma/drug therapy , Melanoma/pathology , Molecular Targeted Therapy , Tumor Microenvironment , Animals , Humans , Immunity , Melanoma/immunology , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Mitogen-Activated Protein Kinases/metabolism
11.
Cell Rep ; 27(13): 3760-3769.e4, 2019 06 25.
Article in English | MEDLINE | ID: mdl-31242410

ABSTRACT

In the eukaryotic cell, spliceosomes assemble onto pre-mRNA cotranscriptionally. Spliceosome assembly takes place in the context of the chromatin environment, suggesting that the state of the chromatin may affect splicing. The molecular details and mechanisms through which chromatin affects splicing, however, are still unclear. Here, we show a role for the histone methyltransferase Set2 and its histone modification, H3K36 methylation, in pre-mRNA splicing through high-throughput sequencing. Moreover, the effect of H3K36 methylation on pre-mRNA splicing is mediated through the chromodomain protein Eaf3. We find that Eaf3 is recruited to intron-containing genes and that Eaf3 interacts with the splicing factor Prp45. Eaf3 acts with Prp45 and Prp19 after formation of the precatalytic B complex around the time of splicing activation, thus revealing the step in splicing that is regulated by H3K36 methylation. These studies support a model whereby H3K36 facilitates recruitment of an "adapter protein" to support efficient, constitutive splicing.


Subject(s)
Acetyltransferases/metabolism , Histones/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/metabolism , Spliceosomes/metabolism , Transcription, Genetic , Acetyltransferases/genetics , Histones/genetics , Methylation , RNA Splicing Factors/genetics , RNA Splicing Factors/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/genetics , Spliceosomes/genetics
12.
Cancer Discov ; 9(1): 82-95, 2019 01.
Article in English | MEDLINE | ID: mdl-30279172

ABSTRACT

Older patients with melanoma have lower rates of sentinel lymph node (LN) metastases yet paradoxically have inferior survival. Patient age correlated with an inability to retain Technetium radiotracer during sentinel LN biopsy in more than 1,000 patients, and high Technetium counts correlated to better survival. We hypothesized that loss of integrity in the lymphatic vasculature due to extracellular matrix (ECM) degradation might play a role. We have implicated HAPLN1 in age-dependent ECM degradation in the dermis. Here, we queried whether HAPLN1 could be altered in the lymphatic ECM. Lymphatic HAPLN1 expression was prognostic of long-term patient survival. Adding recombinant HAPLN1 to aged fibroblast ECMs in vitro reduced endothelial permeability via modulation of VE-cadherin junctions, whereas endothelial permeability was increased following HAPLN1 knockdown in young fibroblasts. In vivo, reconstitution of HAPLN1 in aged mice increased the number of LN metastases, but reduced visceral metastases. These data suggest that age-related changes in ECM can contribute to impaired lymphatics. SIGNIFICANCE: Our studies reveal that changes in the stroma during aging may influence the way tumor cells traffic through the lymphatic vasculature. Aging may dictate the route of metastatic dissemination of tumor cells, and understanding these changes may help to reveal targetable moieties in the aging tumor microenvironment.See related commentary by Marie and Merlino, p. 19.This article is highlighted in the In This Issue feature, p. 1.


Subject(s)
Aging , Extracellular Matrix Proteins/metabolism , Melanoma/metabolism , Proteoglycans/metabolism , Skin/metabolism , Adult , Animals , Cells, Cultured , Humans , Immune System , Lymphatic Metastasis , Melanoma/physiopathology , Mice , Mice, Inbred C57BL , Middle Aged , Skin/physiopathology , Tumor Microenvironment
13.
Cancer Discov ; 9(1): 64-81, 2019 01.
Article in English | MEDLINE | ID: mdl-30279173

ABSTRACT

Physical changes in skin are among the most visible signs of aging. We found that young dermal fibroblasts secrete high levels of extracellular matrix (ECM) constituents, including proteoglycans, glycoproteins, and cartilage-linking proteins. The most abundantly secreted was HAPLN1, a hyaluronic and proteoglycan link protein. HAPLN1 was lost in aged fibroblasts, resulting in a more aligned ECM that promoted metastasis of melanoma cells. Reconstituting HAPLN1 inhibited metastasis in an aged microenvironment, in 3-D skin reconstruction models, and in vivo. Intriguingly, aged fibroblast-derived matrices had the opposite effect on the migration of T cells, inhibiting their motility. HAPLN1 treatment of aged fibroblasts restored motility of mononuclear immune cells, while impeding that of polymorphonuclear immune cells, which in turn affected regulatory T-cell recruitment. These data suggest that although age-related physical changes in the ECM can promote tumor cell motility, they may adversely affect the motility of some immune cells, resulting in an overall change in the immune microenvironment. Understanding the physical changes in aging skin may provide avenues for more effective therapy for older patients with melanoma. SIGNIFICANCE: These data shed light on the mechanochemical interactions that occur between aged skin, tumor, and immune cell populations, which may affect tumor metastasis and immune cell infiltration, with implications for the efficacy of current therapies for melanoma.See related commentary by Marie and Merlino, p. 19.This article is highlighted in the In This Issue feature, p. 1.


Subject(s)
Aging , Collagen/metabolism , Melanoma/metabolism , Skin/metabolism , Animals , Cells, Cultured , Extracellular Matrix Proteins/metabolism , Fibroblasts/metabolism , Humans , Immune System , Melanoma/physiopathology , Mice , Mice, Inbred C57BL , Neoplasm Metastasis , Proteoglycans/metabolism , Skin/physiopathology , Tumor Microenvironment
15.
Clin Cancer Res ; 24(21): 5347-5356, 2018 11 01.
Article in English | MEDLINE | ID: mdl-29898988

ABSTRACT

Purpose: We have shown that the aged microenvironment increases melanoma metastasis, and decreases response to targeted therapy, and here we queried response to anti-PD1.Experimental Design: We analyzed the relationship between age, response to anti-PD1, and prior therapy in 538 patients. We used mouse models of melanoma, to analyze the intratumoral immune microenvironment in young versus aged mice and confirmed our findings in human melanoma biopsies.Results: Patients over the age of 60 responded more efficiently to anti-PD-1, and likelihood of response to anti-PD-1 increased with age, even when we controlled for prior MAPKi therapy. Placing genetically identical tumors in aged mice (52 weeks) significantly increased their response to anti-PD1 as compared with the same tumors in young mice (8 weeks). These data suggest that this increased response in aged patients occurs even in the absence of a more complex mutational landscape. Next, we found that young mice had a significantly higher population of regulatory T cells (Tregs), skewing the CD8+:Treg ratio. FOXP3 staining of human melanoma biopsies revealed similar increases in Tregs in young patients. Depletion of Tregs using anti-CD25 increased the response to anti-PD1 in young mice.Conclusions: While there are obvious limitations to our study, including our inability to conduct a meta-analysis due to a lack of available data, and our inability to control for mutational burden, there is a remarkable consistency in these data from over 500 patients across 8 different institutes worldwide. These results stress the importance of considering age as a factor for immunotherapy response. Clin Cancer Res; 24(21); 5347-56. ©2018 AACR See related commentary by Pawelec, p. 5193.


Subject(s)
Antineoplastic Agents, Immunological/pharmacology , Immunomodulation/drug effects , Programmed Cell Death 1 Receptor/antagonists & inhibitors , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/metabolism , Age Factors , Animals , Antineoplastic Agents, Immunological/therapeutic use , Biomarkers, Tumor , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Cell Line, Tumor , Disease Models, Animal , Female , Humans , Melanoma/drug therapy , Melanoma/immunology , Melanoma/metabolism , Melanoma/pathology , Mice , Mice, Transgenic , Neoplasms/drug therapy , Neoplasms/immunology , Neoplasms/metabolism , Neoplasms/pathology , T-Lymphocytes, Regulatory/immunology , Tumor Microenvironment/immunology , Xenograft Model Antitumor Assays
16.
PLoS Negl Trop Dis ; 9(7): e0003841, 2015.
Article in English | MEDLINE | ID: mdl-26204118

ABSTRACT

U-insertion/deletion RNA editing is a post-transcriptional mitochondrial RNA modification phenomenon required for viability of trypanosomatid parasites. Small guide RNAs encoded mainly by the thousands of catenated minicircles contain the information for this editing. We analyzed by NGS technology the mitochondrial genomes and transcriptomes of two strains, the old lab UC strain and the recently isolated LEM125 strain. PacBio sequencing provided complete minicircle sequences which avoided the assembly problem of short reads caused by the conserved regions. Minicircles were identified by a characteristic size, the presence of three short conserved sequences, a region of inherently bent DNA and the presence of single gRNA genes at a fairly defined location. The LEM125 strain contained over 114 minicircles encoding different gRNAs and the UC strain only ~24 minicircles. Some LEM125 minicircles contained no identifiable gRNAs. Approximate copy numbers of the different minicircle classes in the network were determined by the number of PacBio CCS reads that assembled to each class. Mitochondrial RNA libraries from both strains were mapped against the minicircle and maxicircle sequences. Small RNA reads mapped to the putative gRNA genes but also to multiple regions outside the genes on both strands and large RNA reads mapped in many cases over almost the entire minicircle on both strands. These data suggest that minicircle transcription is complete and bidirectional, with 3' processing yielding the mature gRNAs. Steady state RNAs in varying abundances are derived from all maxicircle genes, including portions of the repetitive divergent region. The relative extents of editing in both strains correlated with the presence of a cascade of cognate gRNAs. These data should provide the foundation for a deeper understanding of this dynamic genetic system as well as the evolutionary variation of editing in different strains.


Subject(s)
Genome, Mitochondrial/genetics , Leishmania/genetics , Leishmania/metabolism , RNA, Protozoan/metabolism , Transcriptome/genetics , Base Sequence , Conserved Sequence , DNA, Kinetoplast/genetics , DNA, Protozoan , Leishmania/classification , Molecular Sequence Data , RNA/genetics , RNA Editing , RNA, Mitochondrial , RNA, Protozoan/genetics
SELECTION OF CITATIONS
SEARCH DETAIL