Difference between revisions of "Part:BBa K5108009"

 
(11 intermediate revisions by the same user not shown)
Line 12: Line 12:
 
     <li style="color: blue;">Usage and Biology</li>
 
     <li style="color: blue;">Usage and Biology</li>
 
<li style="color: blue;">Sequence and Features</li>
 
<li style="color: blue;">Sequence and Features</li>
<li style="color: blue;">Modeling</li>
 
 
     <li style="color: blue;">Characterization and Measurements
 
     <li style="color: blue;">Characterization and Measurements
 
         <ol>
 
         <ol>
 
             <li style="color: blue;">SDS-PAGE</li>
 
             <li style="color: blue;">SDS-PAGE</li>
 
             <li style="color: blue;">Growth analysis</li>
 
             <li style="color: blue;">Growth analysis</li>
             <li style="color: blue;">Consumption analysis of sarcosine, creatine and creatinine by NMR spectroscopy</li>
+
             <li style="color: blue;">Consumption analysis of creatinine by NMR spectroscopy</li>
 
         </ol>
 
         </ol>
 
     </li>
 
     </li>
Line 33: Line 32:
 
<h2 style="color: blue;"> <b>Usage and Biology</b></h2>
 
<h2 style="color: blue;"> <b>Usage and Biology</b></h2>
  
<p>Creatinine is carbon compound rich in nitrogen excreted in human urine [1]. During recent years countless research has been done on the topic of waste-upcycling and revalorization. Creatinine is one of the few human waste products still to be valorized during space missions. In our project, we wanted to use it as carbon and nitrogen source to support the growth of <i>Pseudomonas fluorescens</i>, which serves as biostimulant for plant. Certain species of <i>Pseudomonas</i>, such as <i>Pseudomonas putida</i> can degrade creatinine and use it as carbon and nitrogen sources to ensure its growth. There is no bibliography on this pathway being present in <i>P. fluorescens</i>. The two enzymes permitting creatinine metabolization are creatinine amidohydrolase (CrnA, <a href="https://www.uniprot.org/uniprotkb/P83772/entry" target="blank">EC 3.5.2.10</a>) and creatinase (CreA, <a href="https://www.uniprot.org/uniprotkb/P38488/entry" target="blank">EC 3.5.3.3</a>), both expressed in the same operon. The first catalyzes the hydrolysis of creatinine into creatine. Then, the creatinase catalyzes the hydrolysis of creatine into sarcosine and urea. Finally, in <i>P. putida</i>, sarcosine is degraded by a sarcosine oxidase to join the glycine metabolism [2] (<b>Figure 1</b>).</p>
+
<p>Creatinine is a urinary human waste, rich in carbon and nitrogen [1]. During recent years countless research has been done on the topic of waste-upcycling and revalorization. Creatinine is one of the few human waste products still to be valorized during space missions. In our project, we wanted to use it as a carbon and nitrogen source to support the growth of <i>Pseudomonas fluorescens</i>, which serves as a biostimulant for plants. Certain species of <i>Pseudomonas</i> like <i>Pseudomonas putida</i> can degrade creatinine and use it as carbon and nitrogen sources to sustain growth. There is no bibliography on the presence of this pathway in <i>P. fluorescens</i>. The two enzymes permitting creatinine metabolization are creatinine amidohydrolase (CrnA, <a href="https://www.uniprot.org/uniprotkb/P83772/entry" target="blank">EC 3.5.2.10</a>) and creatinase (CreA, <a href="https://www.uniprot.org/uniprotkb/P38488/entry" target="blank">EC 3.5.3.3</a>), both expressed from an operon. CrnA catalyzes the hydrolysis of creatinine into creatine. Then, CreA catalyzes the hydrolysis of creatine into sarcosine and urea. Finally, in <i>P. putida</i> - but this was not reported in <i>P. fluorescens</i> - sarcosine is degraded by a sarcosine oxidase to enter the glycine metabolism (figure 1).</p>
 +
 
  
 
<div align="center">
 
<div align="center">
Line 47: Line 47:
 
<h2 style="color: blue;"><b>Sequence and Features</b></h2>
 
<h2 style="color: blue;"><b>Sequence and Features</b></h2>
  
<p>The part BBa_K5108009 permits the utilization of creatinine as sole carbon and nitrogen sources to ensure the growth of <i>P. fluorescens</i>. This part is composed of the creatinase and creatinine amidohydrolase ORFs (<i>creA</i> <a href="https://parts.igem.org/Part:BBa_K5108003">BBa_K5108003</a>, <i>crnA</i> <a href="https://parts.igem.org/Part:BBa_K5108004">BBa_K5108004</a>) keeping the natural order from <i>P. putida</i>, and two RBS (<a href="https://parts.igem.org/Part:BBa_K5108006" target="blank">BBa_K5108006</a>, <a href="https://parts.igem.org/Part:BBa_K5108007">BBa_K5108007</a>) allowing their expression in <i>P. fluorescens</i>. This part was expressed under the <i>Pm</i> promoter control into the pSEVA438-Ptet vector (<b>Figure 2</b>). The constitutive repression by the XylS is lifted by the <i>m</i>-toluic acid. We demonstrated efficient enzyme activity to support <i>P. fluorescens</i> growth.</p>
+
<p>The part BBa_K5108009 permits the utilization of creatinine as sole carbon and nitrogen sources to sustain growth of <i>P. fluorescens</i>. This part is composed of the creatinase and creatinine amidohydrolase ORFs (<i>creA</i> <a href="https://parts.igem.org/Part:BBa_K5108003">BBa_K5108003</a>, <i>crnA</i> <a href="https://parts.igem.org/Part:BBa_K5108004">BBa_K5108004</a>) keeping the natural order found in <i>P. putida</i> in the form of an operon, and two RBS (<a href="https://parts.igem.org/Part:BBa_K5108006" target="blank">BBa_K5108006</a>, <a href="https://parts.igem.org/Part:BBa_K5108007">BBa_K5108007</a>) enabling their expression in <i>P. fluorescens</i>. This part was expressed under control of the <i>Pm</i> promoter into the pSEVA438-Ptet vector (<b>Figure 2</b>). Repression by the XylS transcription factor can be lifted by addition of <i>m</i>-toluic acid.</p>
  
 
<div align="center">
 
<div align="center">
Line 54: Line 54:
 
             style="width:60%;"
 
             style="width:60%;"
 
             src="https://static.igem.wiki/teams/5108/lea/cloning-strategy-crea-crna-part.png"><br><br>  
 
             src="https://static.igem.wiki/teams/5108/lea/cloning-strategy-crea-crna-part.png"><br><br>  
             <figcaption class="normal"><span class="titre-image"><b>Figure 2: Representation of the cloning strategy for pSEVA438-Ptet-creA-crnA plasmid.</b></span></figcaption>
+
             <figcaption class="normal"><span class="titre-image"><b>Figure 2: Schematic of the cloning strategy for pSEVA438-Ptet-creA-crnA plasmid.</b></span></figcaption>
 
         </figure>
 
         </figure>
 
</div>
 
</div>
 
<br>
 
<br>
  
<p>To create the functional vector containing the <i>creA-crnA</i> operon, the cloning of the <i>creA-crnA</i> synthetized gBlocks into the pSEVA438-Ptet linearized vector was performed following In-Fusion Assembly (Takara). <b>Figure 3</b> demonstrates the successful cloning by restriction digest with EcoRI and HindIII enzymes (New England Biolabs R3101S, R3104S) (Figure 3). The construct was confirmed by Sanger sequencing (Genewyz, <b>Figure 4</b>).</p>
+
<p>To create the expression plasmid containing the <i>creA-crnA</i> operon, we cloned of the PCR-amplified <i>creA-crnA</i> gBlocks into the linearized pSEVA438-Ptet vector by In-Fusion Assembly (Takara, France). <b>Figure 3</b> shows the expected digestion pattern using EcoRI and HindIII enzymes (New England Biolabs, France, R3101S, R3104S). The construct was confirmed by Sanger sequencing (Genewyz, Germany).</p>
 +
  
 
<div align="center">
 
<div align="center">
Line 67: Line 68:
 
             src="https://static.igem.wiki/teams/5108/lea/crea-crna-plamsid-digestion.png"><br><br>  
 
             src="https://static.igem.wiki/teams/5108/lea/crea-crna-plamsid-digestion.png"><br><br>  
 
             <figcaption class="normal"><span class="titre-image"><b>Figure 3: Restriction digest of pSEVA438-Ptet-creA-crnA plasmid.</b> The plasmid was digested with EcoRI and HindIII separately or in combination. The expected (left) and experimental (right) digestion patterns are shown.</span></figcaption>
 
             <figcaption class="normal"><span class="titre-image"><b>Figure 3: Restriction digest of pSEVA438-Ptet-creA-crnA plasmid.</b> The plasmid was digested with EcoRI and HindIII separately or in combination. The expected (left) and experimental (right) digestion patterns are shown.</span></figcaption>
        </figure>
 
</div>
 
<br>
 
 
<div align="center">
 
        <figure class="normal mx-auto">   
 
            <img class="d-block"
 
            style="width:100%;"
 
            src="https://static.igem.wiki/teams/5108/lea/sanger-sequencing-ccrea.png"><br><br>
 
            <figcaption class="normal"><span class="titre-image"><b>Figure 4: <i>creA-crnA</i> locus’ sequencing of pSEVA438-Ptet-creA-crnA plasmid.</b> The <i>creA-crnA</i> operon was sequenced by two Sanger sequencing using two flanking primers.</span></figcaption>
 
        </figure>
 
</div>
 
<br>
 
 
 
<h2 style="color: blue;"><b>Modeling</b></h2>
 
 
 
<div align="center">
 
        <figure class="normal mx-auto">   
 
            <img class="d-block"
 
            style="width:100%;"
 
            src="https://static.igem.wiki/teams/5108/lea/modelo-growth-rate.png"><br><br>
 
            <figcaption class="normal"><span class="titre-image"><b>Figure 5: A COMPLETER </b>
 
</span></figcaption>
 
 
         </figure>
 
         </figure>
 
</div>
 
</div>
Line 99: Line 75:
 
<h3 style="color: blue;">SDS-PAGE</h3>
 
<h3 style="color: blue;">SDS-PAGE</h3>
  
<p>The pSEVA438-MBPeGFP plasmid, originally used in <i>P. putida</i> KT2440, was employed as positive control of <i>Pm</i> promoter's inducibility in <i>P. fluorescens</i> SBW25. This construct encodes the fusion protein MBPeGFP (Maltose-Binding Protein enhanced Green Fluorescent Protein) under the control of the <i>Pm</i> promoter. Based on the results of <i>Vogeleer P. et al. (2024)</i> [3], the pSEVA438-MBPeGFP- and pSEVA438-Ptet-creA-crnA-transformed <i>P. fluorescens</i> SBW25 strains were cultured in M9 minimal medium supplemented with glucose (28 mM), with or without 0.5 mM of <i>m</i>-toluic acid inducer. After incubation, a whole-protein extraction was performed for each strain to assess the level of expression, as well as the solubility of our proteins.</p>  
+
<p>To assess protein production, the strain was grown on M9 minimal medium with glucose (28 mM), with or without 0.5 mM of <i>m</i>-toluic acid (as inducer of the <i>Pm</i> promoter). <i>P. fluorescens</i> transformed with a pSEVA438-MBPeGFP plasmid was used as positive control of <i>Pm</i> promoter's inducibility in <i>P. fluorescens</i>. This construct encodes a maltose-binding protein (MBP) fused to enhanced Green Fluorescent Protein (eGFP) under the control of the <i>Pm</i> promoter (gift from P. Vogeleer) [3]. </p>
  
<p>The obtained SDS-PAGE is presented in <b>Figure 5</b>. Both soluble and insoluble fractions contain MBPeGFP, with the majority of protein being in the soluble fraction independently of the presence of the inducer. Although transcriptional leakage was clearly observed without the inducer, MBPeGFP was overproduced when the <i>Pm</i> promoter was activated with 0.5 mM of <i>m</i>-toluic acid, confirming the functionality of the <i>Pm</i> promoter in <i>P. fluorescens</i>. The presence of insoluble MBPeGFP can be caused by its overexpression leading to protein aggregation.
+
<p>The obtained SDS-PAGE is presented in <b>Figure 4</b>. Both soluble and insoluble fractions contain MBPeGFP, with the majority of protein being in the soluble fraction independently of the presence of the inducer. Although transcriptional leakage was clearly observed without the inducer, MBPeGFP was produced at higher yield when the <i>Pm</i> promoter was activated with 0.5 mM of <I>m</i>-toluic acid, confirming the functionality of the <i>Pm</i> promoter in <i>P. fluorescens</i>. The presence of insoluble MBPeGFP can be caused by its overexpression leading to protein aggregation.<br>
SDS-PAGE analysis of the cell lysate derived from <i>P. fluorescens</i> transformed with pSEVA438-Ptet-creA-crnA revealed a clearly visible band at the expected size of CreA in both soluble and insoluble fractions when its expression is induced. As expected based on the leaky expression of MBPeGFP, CreA is also produced without the inducer in the soluble protein fraction. In contrast, there is no visible band at the expected size of CrnA, suggesting that the <i>crnA</i> gene is not or poorly expressed.</p>  
+
<br>
 +
SDS-PAGE analysis of the cell lysate derived from <i>P. fluorescens</i> transformed with pSEVA438-Ptet-creA-crnA revealed a clearly visible band at the expected size of CreA in both soluble and insoluble fractions when its expression was induced. As expected based on the leaky expression of MBPeGFP, CreA is also produced without the inducer in the soluble protein fraction. In contrast, there is no visible band at the expected size of CrnA, suggesting that the <i>crnA</i> gene is not or poorly expressed.</p>
  
 
<div align="center">
 
<div align="center">
Line 109: Line 86:
 
             style="width:60%;"
 
             style="width:60%;"
 
             src="https://static.igem.wiki/teams/5108/lea/sds-page-crea-crna.png"><br><br>  
 
             src="https://static.igem.wiki/teams/5108/lea/sds-page-crea-crna.png"><br><br>  
             <figcaption class="normal"><span class="titre-image"><b>Figure 5: SDS-PAGE of soluble and insoluble protein fractions from cultures of <i>Pseudomonas fluorescens</i> transformed with pSEVA438-MBPeGFP or pSEVA438-Ptet-creA-crnA.</b> <i>P. fluorescens</i> was cultured with and without inducer, m-toluic acid.  Arrows show expected size of MBPeGFP, creatinine amidohydrolase (CrnA) and creatinase (CreA).</span></figcaption>
+
             <figcaption class="normal"><span class="titre-image"><b>Figure 4: SDS-PAGE of soluble and insoluble protein fractions from cultures of <i>Pseudomonas fluorescens</i> transformed with pSEVA438-MBPeGFP or pSEVA438-Ptet-creA-crnA.</b> <i>P. fluorescens</i> was cultured with and without inducer.  Arrows show expected size of MBPeGFP, creatinine amidohydrolase (CrnA) and creatinase (CreA).</span></figcaption>
 
         </figure>
 
         </figure>
 
</div>
 
</div>
Line 116: Line 93:
 
<h3 style="color: blue;">Growth analysis</h3>
 
<h3 style="color: blue;">Growth analysis</h3>
  
<p>To challenge the efficiency of our construction, <i>P. fluorescens</i> SBW25 WT was transformed with the empty plasmid pSEVA438-Ptet as a control or plasmid pSEVA438-Ptet-creA-crnA, or no plasmid as a second control. The three strains were all cultured in M9 minimal medium without NH4Cl and supplemented either with creatinine (44 mM), creatine (38 mM), or sarcosine (67 mM). The <i>Pm</i> inducer <i>m</i>-toluic acid was added at 0.5 mM into all the culture media. The growth curves of each condition are presented in <b>Figure 6</b>.</p>
+
<p>WT <i>P. fluorescens</i> SBW25 was transformed with either the empty plasmid pSEVA438-Ptet as a control or the plasmid pSEVA438-Ptet-creA-crnA, or no plasmid as a second control. The three strains were cultured in M9 minimal medium without NH<sub>4</sub>Cl (as it would provide a nitrogen source) and supplemented either with creatinine (44 mM), creatine (38 mM), or sarcosine (67 mM). The inducer <i>m</i>-toluic acid was added at 0.5 mM. The growth curves for each condition are presented in <b>Figure 5</b>.</p>
  
 
<div align="center">
 
<div align="center">
Line 123: Line 100:
 
             style="width:80%;"
 
             style="width:80%;"
 
             src="https://static.igem.wiki/teams/5108/lea/growth-curves-crea.jpg"><br><br>  
 
             src="https://static.igem.wiki/teams/5108/lea/growth-curves-crea.jpg"><br><br>  
             <figcaption class="normal"><span class="titre-image"><b>Figure 6: Growth of the engineered <i>Pseudomonas fluorescens</i> SBW25 strain with <i>creA-crnA</i> operon on M9 minimal medium complemented with creatinine, creatine or sarcosine.</b> Measurement of the OD at 600 nm every 30 minutes. The mean value of the biological and technical replicates for each condition is shown (n ≥ 3). The term plasmid refers to pSEVA438-Ptet.</span></figcaption>
+
             <figcaption class="normal"><span class="titre-image"><b>Figure 5: Growth of the engineered <i>Pseudomonas fluorescens</i> SBW25 strain with <i>creA-crnA</i> operon on M9 minimal medium complemented with creatinine, creatine or sarcosine.</b> Optical density (OD) at 600 nm was measured every 30 minutes. The mean value of the biological and technical replicates for each condition is shown (n ≥ 3).</span></figcaption>
 
         </figure>
 
         </figure>
 
</div>
 
</div>
  
<p>No growth was observed in either the WT or the negative control strains when cultured with creatinine, confirming that the WT <i>P. fluorescens</i> SBW25 strain does not naturally metabolize this compound. In contrast, the engineered strain harboring the <i>creA</i> and <i>crnA</i> genes exhibited clear growth on creatinine, demonstrating that the introduced metabolic pathway is functional with a growth rate of 0.06 h⁻¹.
+
<p>No growth was observed in the WT and the negative control strains when cultured with <b>creatinine</b>, confirming that our strain <i>P. fluorescens</i> SBW25 does not naturally metabolize this compound. In contrast, the engineered strain harboring the <i>creA</i> and <i>crnA</i> genes exhibited clear growth on creatinine, demonstrating that <b>the introduced metabolic pathway is functional</b> with a growth rate of 0.06 h⁻¹ (<b>Figure 5</b>). These physiology results indicate that the <i>crnA</i> gene is well expressed, although the yield of protein production was too low to be detected by SDS-PAGE (<b>Figure 4</b>).</p>
<br>
+
<br>
+
 
+
In analogy with the results obtained with creatinine, neither the WT nor the negative control strains grew on creatine. However, the engineered strain with <i>creA-crnA</i> operon showed moderate growth, confirming that the transformed bacteria could also metabolize creatine. The same growth has been observed for growth on creatine as a sole source of carbon and nitrogen.<br>
+
<br>
+
 
+
However, the growth rate on sarcosine (0.10 h⁻¹) was higher than on creatinine, indicating that the pathway for creatinine degradation may not be as efficient as for sarcosine. This suggests a need for optimization of the metabolic pathway, as differences in growth rates point to potential bottlenecks or suboptimal expression of the enzymes involved in creatinine degradation.<br>
+
<br>
+
  
Using sarcosine as a substrate, all three strains could grow. Growth rate of the strains transformed with the empty plasmid (0.11 h⁻¹) or with the pSEVA438-Ptet-creA-crnA plasmid (0.10 h⁻¹) was lower than that of the WT strain (0.15 h⁻¹), indicating that plasmid introduction had an adverse effect on the growth rate. This was not surprising, as the production of CreA and CrnA may impose a metabolic burden on the bacteria, along with the cost of maintaining antibiotic resistance.
 
</p>
 
 
<br>
 
<br>
  
<h3 style="color: blue;">Consumption analysis of sarcosine, creatine and creatinine by NMR spectroscopy</h3>
+
<h3 style="color: blue;">Consumption analysis of creatinine by NMR spectroscopy</h3>
  
<p>The engineered strain with <i>creA-crnA</i> operon was cultivated of in M9 minimal medium supplemented with creatinine (44 mM), creatine (38 mM), or sarcosine (67 mM). Supernatants during the exponential growth phase were collected and analyzed using 500 MHz NMR spectroscopy to monitor the concentrations of sarcosine, creatine, and creatinine over time. Growth rates and uptake rates were calculated using Physiofit.</p>
+
<p>Supernatants from bacterial cultures were collected during the exponential growth phase and analyzed using 500 MHz NMR spectroscopy to monitor the concentrations of sarcosine, creatine, and creatinine over time. Growth rates and uptake rates were calculated using Physiofit.</p>
  
<h5 style="color: darkblue;">Sarcosine degradation:</h5>
 
  
<p>As shown in <b>Figure 7</b>, the experimental data reveals a clear decrease in sarcosine concentration over time. The initial concentration of sarcosine started at approximately 40 mM, although the expected concentration based on initial conditions was 56 mM. NMR analysis was conducted on the supernatants collected during the exponential growth phase of the bacteria. A sarcosine uptake rate of 3.6 mmol.h⁻¹.gDW⁻¹ was obtained, while the measured growth rate under this condition was 0.105 h⁻¹. The lower growth rate observed in the transformed <i>P. fluorescens</i> strain on sarcosine (0.105 h⁻¹) compared to the WT strain (0.186 h⁻¹) in the same condition (growth in Erlenmeyer flasks) can likely be attributed to the metabolic burden imposed by the plasmid carrying the <i>creA-crnA</i> operon. This effect was similarly observed in microplate experiments, where the growth rate of the transformed strain (0.10 h⁻¹) was also lower than that of the WT strain (0.15 h⁻¹) as presented in <b>Figure 6</b>.<br>
+
<p>The consumption of creatinine was analyzed (<b>Figure 6</b>). Starting at an initial concentration of 26 mM, the creatinine uptake rate was established at 3.58 mmol.h⁻¹.gDW⁻¹. These results demonstrate that <i>P. fluorescens</i> carrying the <i>creA-crnA</i> plasmid can degrade creatinine and utilize it as a sole source of carbon and nitrogen.</p>  
To validate the predictability of our metabolic model, we used the experimentally measured sarcosine uptake rate as an input parameter and the model predicted a growth rate of 0.113 h⁻¹, which is in perfect agreement with the experimentally observed rate of 0.105 h⁻¹.</p>  
+
  
 
<div align="center">
 
<div align="center">
 
         <figure class="normal mx-auto">     
 
         <figure class="normal mx-auto">     
 
             <img class="d-block"
 
             <img class="d-block"
             style="width:60%;"
+
             style="width:50%;"
             src="https://static.igem.wiki/teams/5108/lea/rmn-sarcosine.png"><br><br>  
+
             src="https://static.igem.wiki/teams/5108/results/fig12-resu-12.png"><br><br>  
             <figcaption class="normal"><span class="titre-image"><b>Figure 7: Uptake of sarcosine by the transformed pSEVA438-Ptet-creA-crnA <i>Pseudomonas fluorescens</i> SBW25 strain on M9 minimal medium complemented with sarcosine.</b> Samples of supernatants were taken every hour during the exponential phase and sarcosine concentration was analyzed by 500 MHz NMR spectrometry. Simulation of sarcosine uptake (dashed line) was done with Physiofit with the following characteristics. The shaded area indicates the 95% confidence interval. The growth rate on sarcosine is 0.105 h-1 (n=1).</span></figcaption>
+
             <figcaption class="normal"><span class="titre-image"><b>Figure 6: Uptake of creatinine by the transformed pSEVA438-Ptet-creA-crnA <i>Pseudomonas fluorescens</i> SBW25 strain on M9 minimal medium complemented with creatinine.</b> Samples of supernatants were taken every hour during the exponential phase and creatine concentration was analyzed by 500 MHz NMR spectroscopy. Simulation of creatine uptake (dashed line) was done with Physiofit. The shaded area indicates the 95% confidence interval.</span></figcaption>
 
         </figure>
 
         </figure>
 
</div>
 
</div>
  
<h5 style="color: darkblue;">Creatine degradation:</h5>
+
<p>Similar results are obtained for the creatine and sarcosine consumption (<a href="https://2024.igem.wiki/toulouse-insa-ups/results">iGEM ToulouseINSA-UPS 2024</a>).</p>
 
+
<p>The degradation of creatine was also monitored through NMR (<b>Figure 8</b>). The initial creatine concentration was lower than expected (18 mM, with an expected value of 38 mM), but similar trends were observed in terms of degradation. The experimental data fit well with the simulated predictions, further validating our metabolic model for creatine degradation. A creatine uptake rate of 2.05 mmol.h⁻¹.gDW⁻¹ was obtained, while the measured growth rate under this condition was 0.054 h⁻¹. To validate the predictability of our metabolic model, we also used the experimentally measured sarcosine uptake rate as an input parameter and the model predicted a growth rate of 0.078 h⁻¹, which remains in agreement with the experimentally observed rate of 0.054 h⁻¹.</p>
+
 
+
<div align="center">
+
        <figure class="normal mx-auto">   
+
            <img class="d-block"
+
            style="width:60%;"
+
            src="https://static.igem.wiki/teams/5108/lea/rmn-creatine.png"><br><br>
+
            <figcaption class="normal"><span class="titre-image"><b>Figure 8: Uptake of creatine by the transformed pSEVA438-Ptet-creA-crnA <i>Pseudomonas fluorescens</i> SBW25 strain on M9 minimal medium complemented with creatine.</b> Samples of supernatants were taken every hour during the exponential phase and creatine concentration was analyzed by 500 MHz NMR spectrometry. Simulation of creatine uptake (dashed line) was done with Physiofit with the following characteristics. The shaded area indicates the 95% confidence interval. The growth rate on creatine is 0.054 h⁻¹ (n=1).</span></figcaption>
+
        </figure>
+
</div>
+
 
+
 
+
<h5 style="color: darkblue;">Creatinine degradation:</h5>
+
 
+
<p>Lastly, the degradation of creatinine was analyzed (<b>Figure 9</b>). Starting at an initial concentration of 26 mM (with an expected value of 44 mM), creatinine degradation followed a similar trajectory to the other compounds. However, the degradation rate of creatinine was slower than that of sarcosine and creatine suggesting a lack of expression of the gene crnA as discussed in the precedent part. A creatine uptake rate of 3.58 mmol.h⁻¹.gDW⁻¹ was obtained, while the measured growth rate under this condition was 0.034 h⁻¹. To validate the predictability of our metabolic model, we also used the experimentally measured sarcosine uptake rate as an input parameter and the model predicted a growth rate of 0.014 h⁻¹, which is this time quite high in comparison with the experimentally observed rate of 0.034 h⁻¹.</p>
+
 
+
<div align="center">
+
        <figure class="normal mx-auto">   
+
            <img class="d-block"
+
            style="width:60%;"
+
            src="https://static.igem.wiki/teams/5108/lea/rmn-creatinine.png"><br><br>
+
            <figcaption class="normal"><span class="titre-image"><b>Figure 9: Uptake of creatinine by the transformed pSEVA438-Ptet-creA-crnA <i>Pseudomonas fluorescens</i> SBW25 strain on M9 minimal medium complemented with creatinine.</b> Samples of supernatants were taken every hour during the exponential phase and creatine concentration was analyzed by 500 MHz NMR spectrometry. Simulation of creatine uptake (dashed line) was done with Physiofit with the following characteristics. The shaded area indicates the 95% confidence interval. The growth rate on creatinine is 0.034 h⁻¹ (n=1).</span></figcaption>
+
        </figure>
+
</div>
+
 
+
<p>For the three experiments, we observed lower-than-expected initial substrate concentrations. It could be due to incomplete errors during preparation or measurement. While this raises questions about the accuracy of the initial concentrations, it does not undermine the validity of the results as long as the degradation rates and growth patterns are consistent across experiments and closely match model predictions. Further replicates or adjustments to preparation protocols could have helped us clarify this issue.</p>
+
 
+
<p>These results demonstrate that <i>P. fluorescens</i> carrying the pSEVA438-Ptet-creA-crnA plasmid can degrade sarcosine, creatine, and creatinine, and utilize it as a sole source of carbon and nitrogen to support its growth. The experimental data from NMR closely matched the simulations for sarcosine and creatine, confirming the functionality of our metabolic module for these compounds. Although creatinine degradation appeared less efficient, it was still observed, further supporting the module.</p>  
+
 
+
  
 +
<br>
 
<h2 style="color: blue;"><b>Conclusion and Perspectives</b></h2>
 
<h2 style="color: blue;"><b>Conclusion and Perspectives</b></h2>
  
<p>Further studies will be needed to optimize the metabolic pathway for creatinine and to investigate the potential causes of lower degradation efficiency.</p>
+
<p>In conclusion, our <i>creA-crnA</i> operon enables the metabolization of a urine compound by the plant growth-promoting rhizobacteria, <i>Pseudomonas fluorescens</i>. The cloning of these two genes into plasmid pSEVA438 and transformation into our chassis bacteria allowed it to use creatinine as sole carbon and nitrogen source.<br>  
  
 +
However, the slower growth rates on creatinine (0.06 h⁻¹) compared to sarcosine (0.10 h⁻¹) suggests that further optimization may be needed to improve the pathway efficiency. For instance, we suggest to increase the activity of creatinase (CreA) (<a href="https://2024.igem.wiki/toulouse-insa-ups/results">iGEM ToulouseINSA-UPS 2024</a>).</p>
  
 +
<br>
 
<h2 style="color: blue;"><b>References</b></h2>
 
<h2 style="color: blue;"><b>References</b></h2>
  
Line 206: Line 144:
 
</i>
 
</i>
 
</ol>
 
</ol>
 
 
</html>
 
</html>
 
 
 
 
 
 
 
  
  

Latest revision as of 09:23, 30 September 2024

RBS aprA CHA0 - creA - RBS hcnA CHA0 - crnA

P. fluorescens creatinine amidohydrolase and creatinase ORFs with RBS


    Contents
  1. Usage and Biology
  2. Sequence and Features
  3. Characterization and Measurements
    1. SDS-PAGE
    2. Growth analysis
    3. Consumption analysis of creatinine by NMR spectroscopy
  4. Conclusion and Perspectives
  5. References


Assembly Compatibility:
  • 10
    COMPATIBLE WITH RFC[10]
  • 12
    COMPATIBLE WITH RFC[12]
  • 21
    COMPATIBLE WITH RFC[21]
  • 23
    COMPATIBLE WITH RFC[23]
  • 25
    INCOMPATIBLE WITH RFC[25]
    Illegal NgoMIV site found at 1329
    Illegal NgoMIV site found at 1931
  • 1000
    INCOMPATIBLE WITH RFC[1000]
    Illegal BsaI site found at 249
    Illegal BsaI site found at 1036
    Illegal BsaI site found at 1467
    Illegal BsaI.rc site found at 1815


Usage and Biology

Creatinine is a urinary human waste, rich in carbon and nitrogen [1]. During recent years countless research has been done on the topic of waste-upcycling and revalorization. Creatinine is one of the few human waste products still to be valorized during space missions. In our project, we wanted to use it as a carbon and nitrogen source to support the growth of Pseudomonas fluorescens, which serves as a biostimulant for plants. Certain species of Pseudomonas like Pseudomonas putida can degrade creatinine and use it as carbon and nitrogen sources to sustain growth. There is no bibliography on the presence of this pathway in P. fluorescens. The two enzymes permitting creatinine metabolization are creatinine amidohydrolase (CrnA, EC 3.5.2.10) and creatinase (CreA, EC 3.5.3.3), both expressed from an operon. CrnA catalyzes the hydrolysis of creatinine into creatine. Then, CreA catalyzes the hydrolysis of creatine into sarcosine and urea. Finally, in P. putida - but this was not reported in P. fluorescens - sarcosine is degraded by a sarcosine oxidase to enter the glycine metabolism (figure 1).



Figure 1: Metabolic pathway of the creatinine degradation in Pseudomonas putida.

Sequence and Features

The part BBa_K5108009 permits the utilization of creatinine as sole carbon and nitrogen sources to sustain growth of P. fluorescens. This part is composed of the creatinase and creatinine amidohydrolase ORFs (creA BBa_K5108003, crnA BBa_K5108004) keeping the natural order found in P. putida in the form of an operon, and two RBS (BBa_K5108006, BBa_K5108007) enabling their expression in P. fluorescens. This part was expressed under control of the Pm promoter into the pSEVA438-Ptet vector (Figure 2). Repression by the XylS transcription factor can be lifted by addition of m-toluic acid.



Figure 2: Schematic of the cloning strategy for pSEVA438-Ptet-creA-crnA plasmid.

To create the expression plasmid containing the creA-crnA operon, we cloned of the PCR-amplified creA-crnA gBlocks into the linearized pSEVA438-Ptet vector by In-Fusion Assembly (Takara, France). Figure 3 shows the expected digestion pattern using EcoRI and HindIII enzymes (New England Biolabs, France, R3101S, R3104S). The construct was confirmed by Sanger sequencing (Genewyz, Germany).



Figure 3: Restriction digest of pSEVA438-Ptet-creA-crnA plasmid. The plasmid was digested with EcoRI and HindIII separately or in combination. The expected (left) and experimental (right) digestion patterns are shown.

Characterization and Measurements

SDS-PAGE

To assess protein production, the strain was grown on M9 minimal medium with glucose (28 mM), with or without 0.5 mM of m-toluic acid (as inducer of the Pm promoter). P. fluorescens transformed with a pSEVA438-MBPeGFP plasmid was used as positive control of Pm promoter's inducibility in P. fluorescens. This construct encodes a maltose-binding protein (MBP) fused to enhanced Green Fluorescent Protein (eGFP) under the control of the Pm promoter (gift from P. Vogeleer) [3].

The obtained SDS-PAGE is presented in Figure 4. Both soluble and insoluble fractions contain MBPeGFP, with the majority of protein being in the soluble fraction independently of the presence of the inducer. Although transcriptional leakage was clearly observed without the inducer, MBPeGFP was produced at higher yield when the Pm promoter was activated with 0.5 mM of m-toluic acid, confirming the functionality of the Pm promoter in P. fluorescens. The presence of insoluble MBPeGFP can be caused by its overexpression leading to protein aggregation.

SDS-PAGE analysis of the cell lysate derived from P. fluorescens transformed with pSEVA438-Ptet-creA-crnA revealed a clearly visible band at the expected size of CreA in both soluble and insoluble fractions when its expression was induced. As expected based on the leaky expression of MBPeGFP, CreA is also produced without the inducer in the soluble protein fraction. In contrast, there is no visible band at the expected size of CrnA, suggesting that the crnA gene is not or poorly expressed.



Figure 4: SDS-PAGE of soluble and insoluble protein fractions from cultures of Pseudomonas fluorescens transformed with pSEVA438-MBPeGFP or pSEVA438-Ptet-creA-crnA. P. fluorescens was cultured with and without inducer. Arrows show expected size of MBPeGFP, creatinine amidohydrolase (CrnA) and creatinase (CreA).

Growth analysis

WT P. fluorescens SBW25 was transformed with either the empty plasmid pSEVA438-Ptet as a control or the plasmid pSEVA438-Ptet-creA-crnA, or no plasmid as a second control. The three strains were cultured in M9 minimal medium without NH4Cl (as it would provide a nitrogen source) and supplemented either with creatinine (44 mM), creatine (38 mM), or sarcosine (67 mM). The inducer m-toluic acid was added at 0.5 mM. The growth curves for each condition are presented in Figure 5.



Figure 5: Growth of the engineered Pseudomonas fluorescens SBW25 strain with creA-crnA operon on M9 minimal medium complemented with creatinine, creatine or sarcosine. Optical density (OD) at 600 nm was measured every 30 minutes. The mean value of the biological and technical replicates for each condition is shown (n ≥ 3).

No growth was observed in the WT and the negative control strains when cultured with creatinine, confirming that our strain P. fluorescens SBW25 does not naturally metabolize this compound. In contrast, the engineered strain harboring the creA and crnA genes exhibited clear growth on creatinine, demonstrating that the introduced metabolic pathway is functional with a growth rate of 0.06 h⁻¹ (Figure 5). These physiology results indicate that the crnA gene is well expressed, although the yield of protein production was too low to be detected by SDS-PAGE (Figure 4).


Consumption analysis of creatinine by NMR spectroscopy

Supernatants from bacterial cultures were collected during the exponential growth phase and analyzed using 500 MHz NMR spectroscopy to monitor the concentrations of sarcosine, creatine, and creatinine over time. Growth rates and uptake rates were calculated using Physiofit.

The consumption of creatinine was analyzed (Figure 6). Starting at an initial concentration of 26 mM, the creatinine uptake rate was established at 3.58 mmol.h⁻¹.gDW⁻¹. These results demonstrate that P. fluorescens carrying the creA-crnA plasmid can degrade creatinine and utilize it as a sole source of carbon and nitrogen.



Figure 6: Uptake of creatinine by the transformed pSEVA438-Ptet-creA-crnA Pseudomonas fluorescens SBW25 strain on M9 minimal medium complemented with creatinine. Samples of supernatants were taken every hour during the exponential phase and creatine concentration was analyzed by 500 MHz NMR spectroscopy. Simulation of creatine uptake (dashed line) was done with Physiofit. The shaded area indicates the 95% confidence interval.

Similar results are obtained for the creatine and sarcosine consumption (iGEM ToulouseINSA-UPS 2024).


Conclusion and Perspectives

In conclusion, our creA-crnA operon enables the metabolization of a urine compound by the plant growth-promoting rhizobacteria, Pseudomonas fluorescens. The cloning of these two genes into plasmid pSEVA438 and transformation into our chassis bacteria allowed it to use creatinine as sole carbon and nitrogen source.
However, the slower growth rates on creatinine (0.06 h⁻¹) compared to sarcosine (0.10 h⁻¹) suggests that further optimization may be needed to improve the pathway efficiency. For instance, we suggest to increase the activity of creatinase (CreA) (iGEM ToulouseINSA-UPS 2024).


References

  1. Washington IM & Van Hoosier G (2012) Chapter 3 - Clinical Biochemistry and Hematology. In The Laboratory Rabbit, Guinea Pig, Hamster, and Other Rodents, Suckow MA Stevens KA & Wilson RP (eds) pp 57–116. Boston: Academic Press
  2. Tsuru D, Oka I & Yoshimoto T (1976) Creatinine Decomposing Enzymes in Pseudomonas putida. Agricultural and Biological Chemistry 40: 1011–1018
  3. Vogeleer P, Millard P, Arbulú A-SO, Pflüger-Grau K, Kremling A & Létisse F (2024) Metabolic impact of heterologous protein production in Pseudomonas putida: Insights into carbon and energy flux control. Metabolic Engineering 81: 26–37