IPI1 is a protein involved in pre-ribosomal RNA (rRNA) processing in Candida glabrata, a fungal pathogen . In Saccharomyces cerevisiae, IPI1 is part of the Rix1 complex, which is essential for processing ITS2 sequences from 35S pre-rRNA and is crucial for cell viability .
IPI1 plays a vital role in cell growth and rRNA processing in C. glabrata . Studies using a C. glabrata strain with IPI1 expression regulated by a tetracycline promoter (tet-IPI1) demonstrated that IPI1 is essential for cell growth . When IPI1 expression was repressed using doxycycline, the cells exhibited a significant growth defect, confirming the protein's importance for cell growth .
ITS2 Processing: The internal transcribed spacer 2 (ITS2) region must be digested for complete rRNA processing . Mutation in IPI1 leads to accumulation of unprocessed ITS2, causing growth defects at elevated temperatures .
IPI1 interacts with several other proteins, including Rix1/Ipi2, Ipi3, and Ssb, which are crucial for its function .
Rix1 Complex: Ipi1 is a component of the Rix1 complex (Ipi1-Rix1/Ipi2-Ipi3), similar to its role in S. cerevisiae .
Ssb Chaperones: Ipi1 interacts physically with ribosome-bound Hsp70 chaperones Ssb and Ssz1, which regulate translation efficiency . The Ipi1-Ssb-Ssz1 complex may function as a negative regulator of Pdr1 in C. glabrata .
A mutation in IPI1, specifically ipi1 R70H, can lead to multidrug resistance (MDR) in C. glabrata .
Pdr1 Activation: The ipi1 R70H mutation disrupts the interaction between Ipi1 and Ssb/Ssz1, which releases the negative regulation of Pdr1, leading to constitutive activation of azole efflux pumps .
Ergosterol Levels: Unlike other mechanisms of Pdr1 activation, the ipi1 R70H strain shows increased ergosterol levels, which may contribute to fluconazole resistance .
The ipi1 R70H mutation does not appear to attenuate the virulence of C. glabrata .
Adhesion: No significant difference in adhesion capacity was observed between the wild-type and ipi1 R70H strains .
Phagocytosis: The ipi1 R70H strain exhibits similar susceptibility to macrophage killing compared to the wild-type strain .
In vivo Models: In mouse and silkworm infection models, the ipi1 R70H mutant showed comparable fungal burdens and survival rates to the wild-type strain .
IPI1 also exists in plants, such as rice, where it regulates plant architecture by controlling the protein levels of IPA1 (IPA1 INTERACTING PROTEIN1) .
IPA1 Regulation: IPI1 affects plant structure by precisely adjusting IPA1 protein levels in various tissues in rice .
Tissue-Specific Ubiquitination: IPI1 promotes the degradation of IPA1 in young panicles but enhances its stability in shoot apexes, regulating downstream genes that determine rice architecture .
Pdr1, a transcriptional activator, controls the expression of the CDR1 gene, which encodes an ABC transporter responsible for azole resistance in C. glabrata .
Transcriptional Control: Pdr1 activity is induced upon loss of the mitochondrial genome and challenge by azole drugs .
Direct Target Genes: Chromatin immunoprecipitation coupled with high-throughput sequencing (ChIP-seq) has identified the promoters and associated genes directly regulated by Pdr1 .
| Feature | Wild-Type Strain | ipi1 R70H Mutant |
|---|---|---|
| Growth | Normal | Defective at elevated temperatures |
| ITS2 Processing | Normal | Impaired |
| Adhesion | Normal | Normal |
| Macrophage Killing | Susceptible | Susceptible |
| Virulence (Mouse Model) | Comparable fungal burdens | Comparable fungal burdens |
| Virulence (Silkworm Model) | Similar survival rates | Similar survival rates |
| Azole Resistance | Susceptible | Resistant |
| Ipi1-Ssb/Ssz1 Interaction | Present | Disrupted |
| Pdr1 Regulation | Negatively regulated by Ipi1-Ssb-Ssz1 complex | Constitutively activated |
| Ergosterol Levels | Normal | Increased |
| Gene | Function |
|---|---|
| IPI1 | Involved in pre-rRNA processing, cell growth, and regulation of Pdr1-mediated drug resistance in Candida glabrata. |
| PDR1 | Transcriptional activator that controls the expression of genes involved in azole resistance, including CDR1, in Candida glabrata. |
| CDR1 | Encodes an ABC transporter responsible for azole resistance in Candida glabrata. |
| SSB/SSZ1 | Encode ribosome-bound Hsp70 chaperones involved in regulating translation efficiency and interacting with Ipi1 to regulate Pdr1 activity. |
| IPA1 | Regulates rice plant architecture by controlling protein levels in different tissues. |
KEGG: cgr:CAGL0M01210g
STRING: 284593.XP_449397.1
IPI1 in Candida glabrata plays a crucial role in pre-rRNA processing, a fundamental step in ribosome biogenesis. The protein is involved in the early stages of ribosomal assembly and maturation, contributing to the proper formation of ribosomes necessary for protein synthesis. IPI1 has been experimentally shown to interact with ribosome-related chaperones Ssb and Ssz1, forming a complex that regulates gene expression related to drug resistance mechanisms . This interaction presents an interesting regulatory pathway where ribosome biosynthesis machinery directly influences stress response and drug resistance. Additionally, IPI1 function affects metabolic activity and cell proliferation rates under stress conditions, suggesting broader roles beyond ribosome biogenesis . Understanding IPI1's function provides insights into fundamental cellular processes in this important fungal pathogen.
IPI1 in C. glabrata shows notable functional divergence from its orthologs in other yeasts, particularly Saccharomyces cerevisiae. In C. glabrata, the Ipi1-Ssb/Ssz1 complex inhibits Pdr1-mediated gene expression and multidrug resistance, while in S. cerevisiae, Ssz1 acts as a positive regulator of Pdr1 . This represents a significant evolutionary adaptation in regulatory mechanisms between these related yeast species. The divergence in function highlights how C. glabrata has evolved unique regulatory pathways despite sharing many core cellular components with S. cerevisiae. These differences must be considered when using S. cerevisiae as a model for C. glabrata studies. Similar proteins involved in pre-rRNA processing, such as RPF1 and ESF1, have been studied in human cells, showing conservation of ribosome biogenesis mechanisms across evolution while maintaining species-specific adaptations .
Several experimental systems are available for studying IPI1 in C. glabrata, including genetic manipulation techniques and heterologous expression systems. For genetic studies, researchers can utilize plasmid-based expression systems such as pGREG576, which can be modified to include promoters suitable for C. glabrata, such as the copper-inducible MTI promoter . This system allows for controlled expression of IPI1 variants and mutants. RNA analysis methods, including northern blotting and ratio analysis of multiple precursors (RAMP), can be employed to analyze the effects of IPI1 mutations or depletion on pre-rRNA processing patterns . For protein interaction studies, co-immunoprecipitation experiments can identify binding partners of IPI1, as demonstrated with the Ipi1-Ssb/Ssz1 complex . Heterologous expression in S. cerevisiae provides another useful system for studying C. glabrata proteins in a well-characterized yeast model . Additionally, in vitro exposure systems to antifungal drugs allow for the study of resistance development, as demonstrated by the identification of the R70H mutation in IPI1 following micafungin exposure .
The R70H mutation in IPI1 represents a sophisticated mechanism of multidrug resistance in C. glabrata that operates through disruption of normal regulatory networks. When C. glabrata cells with this mutation are exposed to micafungin (an echinocandin antifungal), they exhibit resistance to both echinocandins and azole antifungals, demonstrating cross-resistance to different drug classes . Mechanistically, this mutation alters the interaction between Ipi1 and the ribosome-related chaperones Ssb and Ssz1, which normally form a complex that inhibits Pdr1-mediated gene expression . The R70H mutation likely disrupts this inhibitory complex, allowing for increased Pdr1 activity and subsequent upregulation of multidrug transporters. This contributes to azole resistance, while the echinocandin resistance appears to involve additional mechanisms. The mutant also exhibits reduced metabolic activity and cell proliferation when exposed to micafungin, which may contribute to drug tolerance through a persistence-like mechanism . The R70H mutation exemplifies how alterations in ribosome biogenesis factors can have far-reaching effects on cellular drug resistance pathways, revealing unexpected connections between seemingly disparate cellular processes.
Analysis of pre-rRNA processing defects in IPI1 mutants requires a comprehensive approach combining several complementary techniques. The Ratio Analysis of Multiple Precursors (RAMP) method represents one of the most informative approaches, as it quantitatively measures the relative abundance of different pre-rRNA species . In this technique, ratios of precursors are calculated for each RNA sample (e.g., pre-rRNA 41S/45-47S, 12S/32S), and log2 values of these ratios in mutant samples are normalized against control samples . These normalized values can then be plotted on histograms to create RAMP profiles that reveal specific processing defects. Northern blot analysis with probes targeting specific regions of pre-rRNA (such as ITS1 and ITS2) provides visual confirmation of accumulating or depleted precursors. RNA co-immunoprecipitation experiments with IPI1 can identify which pre-rRNA species directly interact with the protein, helping to pinpoint its precise role in the processing pathway . Pulse-chase labeling of nascent RNA followed by gel electrophoresis can track the kinetics of pre-rRNA processing, revealing rate-limiting steps affected by IPI1 mutations. For comprehensive characterization, these methods should be combined with polysome profiling to assess the impact on ribosome assembly and function.
Efficient expression and purification of recombinant C. glabrata IPI1 protein requires careful optimization of expression systems and purification strategies. For heterologous expression, S. cerevisiae offers advantages as a closely related yeast with similar protein folding machinery and post-translational modifications. Using expression vectors like p426GPD with a strong constitutive promoter can yield good protein levels in S. cerevisiae . Alternatively, bacterial expression systems using specialized vectors designed for difficult eukaryotic proteins, such as those with codon optimization and fusion tags to enhance solubility, can be employed. For expression in C. glabrata itself, the copper-inducible MTI promoter system allows for controlled expression of IPI1 variants . This system can be particularly valuable when studying the protein in its native cellular environment. For purification, affinity chromatography using N- or C-terminal tags (His, GST, or FLAG) provides an effective first step, though care must be taken that tags don't interfere with IPI1 function or interactions. Size exclusion chromatography as a second purification step helps separate monomeric IPI1 from aggregates and complexes with other proteins. When studying IPI1 complexes with partners like Ssb and Ssz1, co-expression of these proteins followed by tandem affinity purification can yield intact physiological complexes for structural and functional studies .
Identifying IPI1 interaction partners requires a multi-faceted approach incorporating both in vivo and in vitro techniques. Co-immunoprecipitation (Co-IP) represents a powerful starting point, as demonstrated in studies identifying the interactions between Ipi1 and the ribosome-related chaperones Ssb and Ssz1 . For this approach, epitope-tagged IPI1 (either genomically integrated or plasmid-expressed) is used to pull down interacting proteins from C. glabrata lysates, followed by mass spectrometry to identify binding partners. Yeast two-hybrid screening provides a complementary method to detect binary protein-protein interactions, though it may miss interactions dependent on the nuclear environment or post-translational modifications. Proximity-based labeling methods, such as BioID or APEX2 fused to IPI1, can identify both stable and transient interactions within the cellular context. For validation of specific interactions, bimolecular fluorescence complementation (BiFC) or fluorescence resonance energy transfer (FRET) can confirm proximity of proteins in living cells. RNA-protein interactions can be assessed through RNA co-immunoprecipitation, which has revealed IPI1's association with specific pre-rRNA species . Structural studies using cryo-electron microscopy of purified complexes can provide detailed insights into the molecular basis of these interactions, potentially revealing how mutations like R70H disrupt normal binding patterns.
Establishing a reliable system to study IPI1 mutations and their effects on drug resistance requires careful experimental design addressing several key aspects. First, creating isogenic C. glabrata strains differing only in IPI1 alleles is essential to isolate the specific effects of mutations like R70H . This can be achieved through CRISPR-Cas9 editing or traditional homologous recombination methods to introduce mutations at the native locus. Complementation with wild-type IPI1 in mutant strains confirms the phenotype is specifically due to the mutation. Antifungal susceptibility testing should employ standardized methods (CLSI or EUCAST protocols) to determine minimum inhibitory concentrations (MICs) across multiple drug classes, particularly echinocandins and azoles, to assess cross-resistance patterns . Time-kill assays provide dynamic information about how IPI1 mutations affect the rate of fungal killing by antifungals. Expression analysis of multidrug resistance genes, particularly those regulated by Pdr1, using RT-qPCR or RNA-sequencing helps elucidate the molecular mechanisms connecting IPI1 mutations to resistance . Monitoring the association between mutant IPI1 and its binding partners (Ssb/Ssz1) through co-immunoprecipitation under drug treatment conditions can reveal how mutations affect protein interactions during stress. Finally, in vivo models, such as Galleria mellonella infection systems, can assess how IPI1 mutations impact virulence alongside drug resistance, providing a more complete picture of the clinical significance of these mutations .
Analyzing structural changes caused by the R70H mutation in IPI1 requires a combination of computational, biophysical, and biochemical approaches. Computational methods provide an excellent starting point, with homology modeling based on solved structures of related proteins generating preliminary structural models of wild-type and R70H IPI1. Molecular dynamics simulations can predict how the R70H substitution alters protein flexibility, stability, and electrostatic surface properties over time. For experimental structure determination, X-ray crystallography of purified wild-type and R70H IPI1 proteins would provide high-resolution structures, though crystallization may be challenging. Cryo-electron microscopy offers an alternative for structural analysis, particularly for IPI1 in complex with binding partners like Ssb and Ssz1 . Circular dichroism spectroscopy can detect changes in secondary structure elements (α-helices, β-sheets) caused by the mutation. Differential scanning calorimetry and thermal shift assays measure alterations in protein stability, potentially revealing destabilization by the R70H mutation. Hydrogen-deuterium exchange mass spectrometry can identify regions of altered structural dynamics or solvent accessibility. Limited proteolysis followed by mass spectrometry can reveal changes in exposed cleavage sites, indicating conformational differences. Nuclear magnetic resonance (NMR) spectroscopy, if feasible, would provide atomic-level information about structural perturbations, particularly in the region surrounding position 70. These complementary approaches together can establish how the R70H mutation structurally impacts IPI1 function and interactions.
Interpreting changes in pre-rRNA processing patterns requires systematic analysis and careful consideration of multiple factors. When analyzing northern blot or RNA sequencing data from IPI1 mutants, researchers should first identify which pre-rRNA species accumulate and which are depleted compared to wild-type controls . The pattern of accumulation provides clues about the specific processing steps affected - for example, buildup of longer precursors with depletion of shorter ones indicates a block in processing progression. The Ratio Analysis of Multiple Precursors (RAMP) method provides quantitative measurements by calculating ratios between different pre-rRNA species, with these values compared between mutant and wild-type samples and displayed as log2-transformed values on histograms . This approach helps identify subtle processing defects that might be missed by visual inspection alone. When interpreting these patterns, researchers should consider the possibility of alternate processing pathways that may be activated when the primary pathway is blocked by IPI1 mutation. The appearance of aberrant intermediates, such as the 23S pre-rRNA observed in some ribosome biogenesis factor depletion experiments, suggests the activation of alternative cleavage sites . Time-course experiments are valuable for distinguishing primary from secondary effects, as immediate changes following IPI1 inactivation are likely direct consequences, while later changes may represent cellular adaptations. Finally, researchers should integrate these findings with protein interaction data (such as IPI1's association with Ssb/Ssz1) to build a comprehensive model of how IPI1 mutations affect ribosome biogenesis and potentially connect to phenotypes like drug resistance .
Distinguishing between direct and indirect effects of IPI1 on gene expression profiles presents a significant challenge requiring sophisticated experimental approaches and data analysis. Time-course experiments following IPI1 mutation or depletion provide valuable insights, as immediate transcriptional changes (within minutes to a few hours) are more likely to represent direct effects, while later changes may reflect secondary adaptations. Combining RNA sequencing with chromatin immunoprecipitation sequencing (ChIP-seq) of transcription factors known to be affected by IPI1, such as Pdr1, helps identify which expression changes are mediated through these factors versus other mechanisms . Ribosome profiling alongside RNA-seq can determine whether observed mRNA changes translate to altered protein synthesis, distinguishing transcriptional from post-transcriptional effects. For IPI1's effects on ribosome biogenesis, pulse-chase labeling of pre-rRNA combined with northern blotting provides kinetic information about processing rates, helping establish causality in observed defects . Conditional expression systems, such as the copper-inducible MTI promoter, allow for rapid induction or repression of IPI1, minimizing secondary adaptations that occur with constitutive systems . Genetic epistasis experiments, where double mutants of IPI1 and potential downstream effectors are analyzed, can reveal hierarchical relationships in signaling pathways. Network analysis approaches that integrate transcriptomic, proteomic, and protein interaction data can model the propagation of effects from IPI1 through cellular pathways. Finally, direct binding assays such as RNA-protein or protein-protein interaction studies help establish physical connections that underlie functional relationships, such as the demonstrated interaction between IPI1 and the Ssb/Ssz1 chaperones in mediating drug resistance .
IPI1 presents several promising avenues for antifungal drug development based on its essential role in ribosome biogenesis and unexpected connection to drug resistance mechanisms. As a pre-rRNA processing factor, IPI1 is essential for C. glabrata viability, making it an attractive target for developing fungicidal compounds. The discovery that mutations in IPI1, such as R70H, can promote multidrug resistance suggests that targeting this protein might not only kill fungi but also prevent or reverse resistance to existing antifungals . Structure-based drug design approaches, once crystal structures of IPI1 become available, could yield small molecules that bind to and inhibit IPI1 function. High-throughput screening of chemical libraries against recombinant IPI1 protein or IPI1-dependent cellular assays represents another viable approach for identifying lead compounds. Disrupting the interaction between IPI1 and its binding partners (Ssb/Ssz1) with small molecules or peptide mimetics could potentially restore drug sensitivity in resistant strains . RNA-targeted approaches might also be viable, as IPI1 interacts with pre-rRNA; compounds that block this interaction could inhibit ribosome biogenesis. The fact that human cells utilize distinct mechanisms for aspects of ribosome biogenesis offers potential selectivity for fungal-specific targets within this pathway . Combination therapy approaches, where an IPI1 inhibitor is paired with existing antifungals, might prevent resistance development or resensitize resistant strains. Given the emerging threat of multidrug-resistant C. glabrata invasive infections, developing such targeted therapies addressing resistance mechanisms represents a critical research direction.
The study of IPI1 in C. glabrata provides valuable insights into the evolution of eukaryotic ribosome biogenesis pathways and their diversification across species. Comparative analysis of IPI1 with its orthologs in other fungi reveals both conserved and divergent features of pre-rRNA processing machinery. The finding that the C. glabrata Ipi1-Ssb/Ssz1 complex inhibits Pdr1-mediated gene expression, while in S. cerevisiae Ssz1 acts as a positive regulator of Pdr1, demonstrates how conserved components can evolve different regulatory relationships even between closely related species . This functional divergence likely reflects adaptation to different ecological niches and stress environments. The study of pre-rRNA processing factors like IPI1, RPF1, and ESF1 across different eukaryotes shows that while the core ribosome structure and assembly pathway are conserved, regulatory mechanisms and processing details have evolved distinctly . These differences may provide species-specific control points for ribosome production in response to various environmental conditions. The connection between ribosome biogenesis and drug resistance pathways revealed through IPI1 studies highlights how fundamental cellular processes can become integrated with stress response mechanisms through evolution . The repurposing of cellular machinery is evident in C. glabrata, where components of the mating pathway have been adapted for inter-species communication despite its primarily asexual reproduction strategy . This demonstrates evolutionary flexibility in repurposing existing cellular machinery for new functions. Studying IPI1 and related factors across diverse fungi provides insights into how essential processes diverge during speciation while maintaining core functionality, potentially revealing evolutionary principles applicable to other cellular systems.