Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
1.
EMBO J ; 32(12): 1681-701, 2013 Jun 12.
Article in English | MEDLINE | ID: mdl-23685356

ABSTRACT

Telomeres are repetitive DNA structures that, together with the shelterin and the CST complex, protect the ends of chromosomes. Telomere shortening is mitigated in stem and cancer cells through the de novo addition of telomeric repeats by telomerase. Telomere elongation requires the delivery of the telomerase complex to telomeres through a not yet fully understood mechanism. Factors promoting telomerase-telomere interaction are expected to directly bind telomeres and physically interact with the telomerase complex. In search for such a factor we carried out a SILAC-based DNA-protein interaction screen and identified HMBOX1, hereafter referred to as homeobox telomere-binding protein 1 (HOT1). HOT1 directly and specifically binds double-stranded telomere repeats, with the in vivo association correlating with binding to actively processed telomeres. Depletion and overexpression experiments classify HOT1 as a positive regulator of telomere length. Furthermore, immunoprecipitation and cell fractionation analyses show that HOT1 associates with the active telomerase complex and promotes chromatin association of telomerase. Collectively, these findings suggest that HOT1 supports telomerase-dependent telomere elongation.


Subject(s)
Homeodomain Proteins/metabolism , Multiprotein Complexes/metabolism , Telomerase/metabolism , Telomere-Binding Proteins/metabolism , Telomere/metabolism , Chromatin/genetics , Chromatin/metabolism , HeLa Cells , Homeodomain Proteins/genetics , Humans , Multiprotein Complexes/genetics , Repetitive Sequences, Nucleic Acid/physiology , Telomerase/genetics , Telomere/genetics , Telomere-Binding Proteins/genetics
2.
Trends Biochem Sci ; 37(9): 391-9, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22727244

ABSTRACT

The stability of the ends of linear eukaryotic chromosomes is ensured by functional telomeres, which are composed of short, species-specific direct repeat sequences. The maintenance of telomeres depends on a specialized ribonucleoprotein (RNP) called telomerase. Both telomeres and telomerase are dynamic entities with different physical behaviors and, given their substrate-enzyme relation, they must establish a productive interaction. Regulatory mechanisms controlling this interaction are key missing elements in our understanding of telomere functions. Here, we review the dynamic properties of telomeres and the maturing telomerase RNPs, and summarize how tracking the timing of their dance during the cell cycle will yield insights into chromosome stability mechanisms. Cancer cells often display loss of genome integrity; therefore, these issues are of particular interest for our understanding of cancer initiation or progression.


Subject(s)
Cell Cycle , Telomerase/metabolism , Telomere/metabolism , Animals , Cell Nucleus/enzymology , Humans , Ribonucleoproteins/metabolism
3.
Semin Cancer Biol ; 23(2): 116-24, 2013 Apr.
Article in English | MEDLINE | ID: mdl-22330096

ABSTRACT

Telomeres are crucial for the maintenance of genome stability through "capping" of chromosome ends to prevent their recognition as double-strand breaks, thus avoiding end-to-end fusions or illegitimate recombination [1-3]. Similar to other genomic regions, telomeres participate to the nuclear architecture while being highly mobile. The interaction of telomeres with nuclear domains or compartments greatly differs not only between organisms but also between cells within the same organism. It is also expected that biological processes like replication, repair or telomere elongation impact the distribution of chromosome extremities within the nucleus, as they probably do with other regions of the genome. Pathological processes such as cancer induce profound changes in the nuclear architecture, which also affects telomere dynamics and spatial organization. Here we will expose our present knowledge on the relationship between telomeres and nuclear architecture and on how this relationship is affected by normal or abnormal telomere metabolisms.


Subject(s)
Cell Nucleus/genetics , Telomere/physiology , Animals , Cell Nucleus/metabolism , Cell Nucleus/physiology , Cell Nucleus/ultrastructure , Chromatin Assembly and Disassembly/physiology , DNA Replication/genetics , DNA Replication/physiology , Genomic Instability/physiology , Humans , Models, Biological , Neoplasms/genetics , Neoplasms/pathology , Telomere/chemistry , Telomere/genetics , Telomere/metabolism , Telomere Homeostasis/genetics
4.
FASEB J ; 22(2): 622-32, 2008 Feb.
Article in English | MEDLINE | ID: mdl-17898088

ABSTRACT

The skin human papillomavirus (HPV) types belonging to the genus beta of the HPV phylogenetic tree appear to be associated with nonmelanoma skin cancer. We previously showed that the beta HPV type 38 E6 and E7 oncoproteins are able to inactivate the tumor suppressors p53 and retinoblastoma. Here, both viral proteins were expressed in primary human skin keratinocytes in order to study their effects on the telomere/telomerase system. We show that immortalization of skin keratinocytes induced by HPV38 E6/E7 is associated with hTERT gene overexpression. This event is, in part, explained by the accumulation of the p53-related protein, DeltaNp73. Despite elevated levels of hTERT mRNA, the telomerase activity detected in HPV38 E6/E7 keratinocytes was lower than that observed in HPV16 E6/E7 keratinocytes. The low telomerase activation in highly proliferative HPV38 E6/E7 keratinocytes resulted in the presence of extremely short and unstable telomeres. In addition, we observed anaphase bridges, mitotic multipolarity, and dramatic genomic aberrations. Interestingly, the ectopic expression of hTERT prevents both telomere erosion and genomic instability. Thus, we showed that in HPV38 E6/E7 keratinocytes characterized by unscheduled proliferation, suboptimal activation of telomerase and subsequent extensive telomere shortening result in genomic instability facilitating cellular immortalization.


Subject(s)
Genomic Instability/genetics , Keratinocytes/metabolism , Papillomaviridae/physiology , Skin Diseases/metabolism , Telomerase/metabolism , Telomere/genetics , Telomere/metabolism , Anaphase , Cell Survival , Cells, Cultured , Chromosomes, Human/genetics , Female , Humans , Keratinocytes/cytology , Meiosis , Oncogene Proteins, Viral/metabolism , Polyploidy , RNA, Messenger/genetics , Skin Diseases/genetics , Skin Diseases/pathology , Up-Regulation
5.
Mech Ageing Dev ; 129(1-2): 17-26, 2008.
Article in English | MEDLINE | ID: mdl-18054991

ABSTRACT

Telomere shortening accompanies human aging, and premature aging syndromes are often associated with short telomeres. These two observations are central to the hypothesis that telomere length directly influences longevity. If true, genetically determined mechanisms of telomere length homeostasis should significantly contribute to variations of longevity in the human population. On the other hand, telomere shortening is also observed in the course of many aging-associated disorders but determining whether it is a cause or a consequence is not an easy task. Here, we review the most relevant experimental and descriptive data relating telomere length, as a quantitative trait, to aging and longevity.


Subject(s)
Aging/genetics , Telomere/metabolism , Animals , Caenorhabditis elegans/genetics , Dyskeratosis Congenita/genetics , Humans , Longevity/genetics , Mice , Models, Animal , Quantitative Trait, Heritable , Saccharomyces cerevisiae/genetics , Telomere/chemistry , Werner Syndrome/genetics
6.
Biochimie ; 90(1): 73-82, 2008 Jan.
Article in English | MEDLINE | ID: mdl-17728038

ABSTRACT

Telomeres are required to preserve genome integrity, chromosome stability, nuclear architecture and chromosome pairing during meiosis. Given that telomerase activity is limiting or absent in most somatic tissues, shortening of telomeres during development and aging is the rule. In vitro, telomere length operates as a mechanism to prevent uncontrolled cell growth and therefore defines the proliferation potential of a cell. In vitro, in somatic cells that have lost proliferation control, shortening of telomeres becomes the main source of genome instability leading to genetic or epigenetic changes that may allow cells to become immortal and to acquire tumor phenotypes. In vivo, mice models have indisputably shown both the protective and the promoting role of very short telomeres in cancer development. In humans, although telomere shortening and other types of telomere dysfunction probably contribute to the genome instability often detected in tumors, the specific contributions of such instability to the development of cancer remain largely undetermined.


Subject(s)
Chromosomal Instability , Genomic Instability , Neoplasms/etiology , Telomerase/metabolism , Telomere-Binding Proteins/metabolism , Telomere/physiology , Aging , Animals , Cell Proliferation , Cell Transformation, Neoplastic , Epigenesis, Genetic , Humans , Mutation , Neoplasms/genetics , Telomere/enzymology , Telomere/genetics
7.
Nucleic Acids Res ; 33(17): 5446-57, 2005.
Article in English | MEDLINE | ID: mdl-16192571

ABSTRACT

The phylogenetically-derived secondary structures of telomerase RNAs (TR) from ciliates, yeasts and vertebrates are surprisingly conserved and contain a pseudoknot domain at a similar location downstream of the template. As the pseudoknot domains of Tetrahymena TR (tTR) and human TR (hTR) mediate certain similar functions, we hypothesized that they might be functionally interchangeable. We constructed a chimeric TR (htTR) by exchanging the hTR pseudoknot sequences for the tTR pseudoknot region. The chimeric RNA reconstituted human telomerase activity when coexpressed with hTERT in vitro, but exhibited defects in repeat addition processivity and levels of DNA synthesis compared to hTR. Activity was dependent on tTR sequences within the chimeric RNA. htTR interacted with hTERT in vitro and dimerized predominantly via a region of its hTR backbone, the J7b/8a loop. Introduction of htTR in telomerase-negative cells stably expressing hTERT did not reconstitute an active enzyme able to elongate telomeres. Thus, our results indicate that the chimeric RNA reconstituted a weakly active nonprocessive human telomerase enzyme in vitro that was defective in telomere elongation in vivo. This suggests that there may be species-specific requirements for pseudoknot functions.


Subject(s)
RNA/chemistry , RNA/metabolism , Telomerase/chemistry , Telomerase/metabolism , Telomere/metabolism , Tetrahymena/genetics , Animals , Cell Line , DNA-Binding Proteins/metabolism , Dimerization , Humans , Mutation , Nucleic Acid Conformation , Polymerase Chain Reaction , RNA/genetics , Telomerase/genetics , Tetrahymena/enzymology
8.
Mol Cancer Ther ; 5(7): 1669-75, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16891452

ABSTRACT

Breast cancer is the most common malignancy among women. Current therapies for breast tumors are based on the use of chemotherapeutic drugs that are quite toxic for the patients and often result in resistance. Telomerase is up-regulated in 95% of breast carcinomas but not in adjacent normal tissues. Therefore, it represents a very promising target for anticancer therapies. Unfortunately, the antiproliferative effects of telomerase inhibition require extensive telomere shortening before they are fully present. Combining telomerase inhibition with common chemotherapeutic drugs can be used to reduce this lag phase and induce tumor cell death more effectively. Few studies have analyzed the effects of telomerase inhibition in combination with anticancer drugs in breast cancer cells. In this study, we inhibited telomerase activity in two breast cancer cell lines using a dominant-negative human telomerase reverse transcriptase and analyzed cell viability after treatment with different anticancer compounds. We found that dominant-negative human telomerase reverse transcriptase efficiently inhibits telomerase activity and causes telomere shortening over time. Moreover, cells in which telomerase was suppressed were more sensitive to anticancer agents independently of their mechanism of action and this sensitization was dependent on the presence of shorter telomeres. Altogether, our data show that blocking telomere length maintenance in combination with anticancer drugs can be used as an effective way to induce death of breast cancer cells.


Subject(s)
Antineoplastic Agents/therapeutic use , Breast Neoplasms/drug therapy , Breast Neoplasms/enzymology , DNA-Binding Proteins/antagonists & inhibitors , DNA-Binding Proteins/genetics , Telomerase/antagonists & inhibitors , Telomerase/genetics , Breast Neoplasms/genetics , Cell Line, Tumor , Drug Resistance, Neoplasm , Female , Humans , Telomere/drug effects , Telomere/metabolism , Transfection
9.
Oncogene ; 24(53): 7893-901, 2005 Nov 24.
Article in English | MEDLINE | ID: mdl-16116482

ABSTRACT

In human somatic cells proliferation results in telomere shortening due to the end replication problem and the absence of adequate levels of telomerase activity. The progressive loss of telomeric DNA has been associated with replicative senescence. Maintenance of telomere structure and function is, therefore, an essential requisite for cells that proliferate indefinitely. Human cells that have acquired the immortal phenotype mostly rely on telomerase to compensate for telomere shortening with cell division. However, a certain percentage of immortalized cell lines and human tumors maintain their telomeres by Alternative Lengthening of Telomeres (ALT), a mechanism not fully understood but apparently based on homologous recombination. Here, we report the isolation of an immortal human cell line that is derived from an ALT cell line but maintains telomeres in the absence of key features of ALT and of telomerase. The properties of these cells suggest that the identification of ALT cells may not be reliably based on known ALT markers. This finding is of relevance for discriminating between the mortal and immortal phenotype among telomerase-negative cells in vitro and in vivo, particularly in regard to the development of pharmacological approaches for cancer treatment based on telomerase inhibition.


Subject(s)
Telomerase/metabolism , Telomere/ultrastructure , Tumor Cells, Cultured , Biomarkers/analysis , Cell Proliferation , Humans , Phenotype , Transfection
10.
Cancer Res ; 64(7): 2324-7, 2004 Apr 01.
Article in English | MEDLINE | ID: mdl-15059879

ABSTRACT

Telomere maintenance activity is a hallmark of cancer. In some telomerase-negative tumors, telomeres become lengthened by alternative lengthening of telomeres (ALT), a recombination-mediated DNA replication process in which telomeres use other telomeric DNA as a copy template. Using chromosome orientation fluorescence in situ hybridization, we found that postreplicative exchange events involving a telomere and another TTAGGG-repeat tract occur at remarkably high frequencies in ALT cells (range 28-280/100 metaphases) and rarely or never in non-ALT cells, including cell lines with very long telomeres. Like the ALT phenotype itself, the telomeric exchanges were not suppressed when telomerase was activated in ALT cells. These exchanges are telomere specific because there was no correlation with sister chromatid exchange rates at interstitial locations, and they were not observed in non-ALT Bloom syndrome cells with very high sister chromatid exchange rates.


Subject(s)
Neoplasms/genetics , Telomere/genetics , Animals , Cell Line, Tumor , Humans , In Situ Hybridization, Fluorescence , Mice , NIH 3T3 Cells , Sister Chromatid Exchange
11.
Oncogene ; 21(5): 769-77, 2002 Jan 24.
Article in English | MEDLINE | ID: mdl-11850805

ABSTRACT

In this work, the full-length hTERT gene was isolated and the sequence of the previously unknown region in intron 6 as well as that of upstream and downstream hTERT regions was determined. We have shown that intron 6 includes a variable number of tandem repeats (VNTR) of a 38 bp sequence, (hTERT-VNTR 6-1). Eight alleles of hTERT-VNTR 6-1 were identified among 103 unrelated individuals, ranging from 27 to 47 repeats. hTERT-VNTR 2-2 is another new 61 bp minisatellite repeat found in intron 2 of hTERT. At least four alleles of hTERT-VNTR 2-2 can be distinguished. Previous studies have described polymorphisms for minisatellites hTERT-VNTR 2-1, a 42 bp repeat in intron 2, and hTERT-VNTR 6-2, a 36 bp repeat in intron 6. These, together with another minisatellite found in intron 12, add up to five such structures within the hTERT gene. The segregation of hTERT minisatellites was analysed in families, revealing that the VNTRs are transmitted through meiosis following a Mendelian inheritance. Minisatellites in hTERT were also analysed in matching normal and cancer tissues from patients with tumors; in one patient with a kidney tumor, the two VNTRs in intron 6 had undergone concomitant rearrangements. This observation suggests that chromosomal rearrangements implicating these VNTRs may be associated with the activation of telomerase expression in cancer cells.


Subject(s)
Minisatellite Repeats , Polymorphism, Genetic , Telomerase/genetics , Base Sequence , DNA-Binding Proteins , Genes , Genomic Library , Humans , Introns , Molecular Sequence Data , Neoplasms/genetics , Sequence Analysis, DNA
12.
Cancer Lett ; 212(2): 135-44, 2004 Aug 30.
Article in English | MEDLINE | ID: mdl-15341022

ABSTRACT

Chromosome aberrations are the hallmark of cancer cells. Although a few specific chromosome aberrations are frequently detected in some types of cancer, the majority of karyotypic abnormalities tend to differ between different histological types and between individuals with the same type of cancer. Recent work indicates that telomeres may be directly involved in shaping the karyotypes of tumor cells. In particular, the heterogeneity of telomere lengths within cells may have direct influence on the frequency with which chromosomes engage in telomeric fusions and in subsequent breakage-fusion-bridge cycles. Since telomere length distribution among chromosome arms is a polymorphic trait, difference in distributions between individuals may account, at least in part, for the karyotypic differences found among tumors of the same type. Conversely, if single telomere lengths happen to be inherited, the segregation of particularly short telomeres in families may increase the incidence of specific chromosome aberrations during tumor evolution, and perhaps contribute, along with other factors, to cancer pre-disposition.


Subject(s)
Chromosome Aberrations , Chromosomes/ultrastructure , Telomere/ultrastructure , Alleles , Blotting, Southern , Humans , Karyotyping , Polymorphism, Genetic
14.
Autoimmun Rev ; 9(10): 646-51, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20435169

ABSTRACT

Telomeres are specialized nucleoproteic structures that cap and protect the ends of chromosomes. They can be elongated by the telomerase enzyme, but in telomerase negative cells, telomeres shorten after each cellular division because of the end replicating problem. This phenomenon leads ultimately to cellular senescence, conferring to the telomeres a role of biological clock. Oxidative stress, inflammation and increased cell renewal are supplementary environmental factors that accelerate age-related telomere shortening. Similar to other types of DNA damage, very short/dysfunctional telomeres activate a DNA response pathway leading to different outcomes: DNA repair, cell senescence or apoptosis. During the last 10 years, studies on the telomere/telomerase system in autoimmune and/or systemic immune-mediated diseases have revealed its involvement in relevant physiopathological processes. Here, we present a literature review of telomere and telomerase homeostasis in systemic inflammatory diseases including systemic lupus erythematosus, rheumatoid arthritis and granulomatous diseases. The available data indicate that both telomerase activity and telomere length are modified in various systemic immune-mediated diseases and appear to be connected with premature immunosenescence. Studies on the telomere/telomerase system open new research avenues for the basic understanding and for therapeutic approaches of these pathologies.


Subject(s)
Autoimmune Diseases/genetics , Cellular Senescence/immunology , Telomerase/metabolism , Telomere/immunology , Aging, Premature/genetics , Animals , Apoptosis , Cellular Senescence/genetics , DNA Damage , Enzyme Activation , Humans , Telomerase/genetics , Telomere/genetics
15.
Cancer Lett ; 275(2): 266-76, 2009 Mar 18.
Article in English | MEDLINE | ID: mdl-19056167

ABSTRACT

Cellular viability requires telomere maintenance, which, in mammals, is mainly mediated by the reverse transcriptase telomerase. Telomerase core components are a catalytic subunit TERT and an RNA subunit TR (hTR in humans, mTR in mouse) that carries the template to generate telomeres de novo. Telomere dysfunction can lead to senescence or apoptosis and impairs the continued growth of immortal cancerous cell lines. The introduction of a template-mutated hTR in telomerase-positive and telomerase-negative human cell lines results in dramatic growth defects. No study has addressed the consequences of expressing a template-mutated mTR in mouse immortal cell lines. Therefore, we analyzed the effects of long-term expression of a template-mutated mTR in the telomerase-positive and telomerase-negative murine cell lines CB17 and DKO301, respectively. Whereas the CB17 clones expressing the template-mutated mTR did not demonstrate any growth impairment, many of the DKO301 clones expressing the template-mutated mTR underwent growth and cell cycle defects and eventual cell death. These results suggest that in the absence of wild-type telomerase, the expression of the template-mutated mTR likely perturbs telomere function, leading to decreased cellular viability. Furthermore, whereas the expression of template-mutated hTR in telomerase-negative human cell lines leads to immediate cellular toxicity, the expression of the template-mutated mTR in the telomerase-negative mouse cell line did not.


Subject(s)
Mutation , RNA/genetics , Telomerase/genetics , Animals , Apoptosis , Base Sequence , Cell Division , Cell Line, Transformed , DNA Primers , In Situ Hybridization, Fluorescence , Mice , Reverse Transcriptase Polymerase Chain Reaction
16.
Ai Zheng ; 27(7): 775-83, 2008 Jul.
Article in Zh | MEDLINE | ID: mdl-18606076

ABSTRACT

Telomeres play fundamental roles in genome stability, nuclear architecture and chromosome pairing during meiosis. They shorten at every cell division and may be re-elongated or not depending on the presence of the dedicated enzyme, telomerase. Since in most human somatic cells telomerase is not expressed, shortening of telomeres during development and aging is the rule. Short telomeres being, under physiological conditions, incompatible with extended cell proliferation, telomere length defines the proliferation potential of a cell and operates as a mechanism to prevent uncontrolled cell growth. Conversely, in cells in which proliferation checkpoints have been abolished, shortening of telomeres causes chromosomes to fuse and to initiate cycles of breakage-fusion-bridge thus becoming a strong driving force for genome instability. In vitro, transformed cells with highly unstable genomes because of severe telomere shortening accumulate deleterious genetic changes and die (crisis). At the same time, random genetic or epigenetic changes may allow cells to acquire a telomere maintenance mechanism (as well as other tumor phenotypes) and to become immortal. Although telomere shortening and other types of telomere dysfunction probably contribute to the genome instability detected in early tumors in vivo, the direct contributions of dysfunctional telomeres to the acquisition of tumor phenotypes in humans remain largely unspecified.


Subject(s)
Neoplasms/etiology , Telomere , Aging , Cell Proliferation , Evolution, Molecular , Genomic Instability , Humans , Mutation , Neoplasms/genetics , Phenotype
17.
Cell Cycle ; 4(4): 585-9, 2005 Apr.
Article in English | MEDLINE | ID: mdl-15753647

ABSTRACT

Dyskeratosis congenita (DC) is a rare multi-system syndrome characterized by nail dystrophy, abnormal skin pigmentation and mucosal leukoplakia. The gene mutated in the X-linked form of human DC encodes for dyskerin, a nucleolar pseudourydilase that is involved in rRNA maturation. Dyskerin is also involved in telomerase function through its interaction with the telomerase RNA (hTR). Mutations in dyskerin result in low levels of hTR, decreased telomerase activity and telomere shortening. Autosomal dominant DC is characterized by mutations in hTR, supporting the hypothesis that the DC phenotype may be caused by impaired telomere maintenance. Several mutations have been identified in different regions of hTR in patients affected by autosomal dominant DC. Recent reports have shown that coexpression of wild-type hTR with hTR harboring mutations found in the pseudoknot domain does not affect telomerase activity in vitro. However, these studies did not assess the consequences of mutant hTR expression at the telomeres. Here we provide the first direct in vivo evidence that a mutant hTR carrying the GC to AG double substitution in the pseudoknot at nucleotides 107-108 found in patients affected by autosomal dominant DC does not behave as a dominant-negative for telomere maintenance. Rather it reconstitutes a weakly active telomerase enzyme, which is defective in telomere elongation.


Subject(s)
Dyskeratosis Congenita/genetics , Mutation , RNA/genetics , Telomerase/genetics , Telomere/ultrastructure , Cell Cycle Proteins/chemistry , Cell Line, Tumor , Chromosome Mapping , Chromosomes/ultrastructure , Genes, Dominant , Humans , In Situ Hybridization, Fluorescence , Nuclear Proteins/chemistry , RNA/chemistry , RNA, Ribosomal/chemistry , Reverse Transcriptase Polymerase Chain Reaction , Sequence Analysis, DNA , Telomerase/metabolism , Transfection
18.
Microbiology (Reading) ; 143 ( Pt 8): 2753-2761, 1997 Aug.
Article in English | MEDLINE | ID: mdl-9274028

ABSTRACT

Intercellular communication is a crucial phenomenon during spore development in Bacillus subtilis. It couples the establishment of a compartment-specific genetic program to the transcriptional activity of a sigma factor in the other compartment. It also keeps sigma factor activation in register with the morphological process. This study used directed mutagenesis to analyse the pathway that couples sigma E activation in the mother-cell to activation of sigma F in the forespore following asymmetric septation. Targets for mutagenesis in SpoIIGA (the receptor) were chosen based on the predicted topology of the protein when associated with the cell membrane. The results showed that a residue near the N terminus (D6), predicted to be exposed outside the cell, is required for receptor activity, whereas the major extracellular loop (between membrane domains IV and V) is dispensable for function. In contrast, mutations in SpoIIR (the signal) that partially blocked protein release (but not membrane translocation) had no effect on signal transduction. These results do not rule out the possibility that uncharacterized molecules intervene in the signalling pathway that establishes the mother-cell-specific developmental program during the early stage of sporulation.


Subject(s)
Bacillus subtilis/physiology , Bacterial Proteins/genetics , Cell Communication/genetics , Membrane Proteins/genetics , Peptide Hydrolases , Sigma Factor/antagonists & inhibitors , Alkaline Phosphatase/genetics , Amino Acid Sequence , DNA Mutational Analysis , Genes, Bacterial , Genes, Reporter , Molecular Sequence Data , Mutagenesis, Site-Directed , Sigma Factor/metabolism , Spores, Bacterial/physiology , Transcription Factors/metabolism , beta-Galactosidase/genetics
SELECTION OF CITATIONS
SEARCH DETAIL