Recombinant Rat Nitrilase homolog 1 (Nit1)

Shipped with Ice Packs
In Stock

Product Specs

Form
Lyophilized powder
Note: While we prioritize shipping the format currently in stock, please specify your format preference during order placement for customized preparation.
Lead Time
Delivery times vary depending on the purchase method and location. Please contact your local distributor for precise delivery estimates.
Note: All proteins are shipped with standard blue ice packs. Dry ice shipping requires advance notice and incurs additional charges.
Notes
Avoid repeated freeze-thaw cycles. Store working aliquots at 4°C for up to one week.
Reconstitution
Centrifuge the vial briefly before opening to collect the contents. Reconstitute the protein in sterile deionized water to a concentration of 0.1-1.0 mg/mL. For long-term storage, we recommend adding 5-50% glycerol (final concentration) and aliquoting at -20°C/-80°C. Our standard glycerol concentration is 50%, which can be used as a reference.
Shelf Life
Shelf life depends on various factors including storage conditions, buffer composition, temperature, and protein stability. Generally, liquid formulations have a 6-month shelf life at -20°C/-80°C, while lyophilized forms have a 12-month shelf life at -20°C/-80°C.
Storage Condition
Upon receipt, store at -20°C/-80°C. Aliquoting is recommended for multiple uses. Avoid repeated freeze-thaw cycles.
Tag Info
Tag type is determined during the manufacturing process.
The tag type is determined during production. If a specific tag type is required, please inform us, and we will prioritize its development.
Synonyms
Nit1; rCG_20134; Deaminated glutathione amidase; dGSH amidase; EC 3.5.1.128; Nitrilase homolog 1
Buffer Before Lyophilization
Tris/PBS-based buffer, 6% Trehalose.
Datasheet
Please contact us to get it.
Expression Region
1-292
Protein Length
full length protein
Purity
>85% (SDS-PAGE)
Species
Rattus norvegicus (Rat)
Target Names
Nit1
Target Protein Sequence
MAESSSTSWE LPLVAVCQVT STPNKQENFK TCAELVQEAT RLGACLAFLP EAFDFIARNP AETLLLSEPL DGDLLGQYSQ LARECGIWLS LGGFHERGQD WEQTQKIYNC HVLLNSKGSV VASYRKTHLC DVEIPGQGPM RESNYTMPGY ALEPPVKTPA GKVGLAICYD MRFPELSLKL AQAGAEILTY PSAFGSVTGP AHWEVLLRAR AIESQCYVIA AAQCGRHHET RASYGHSMVV DPWGTVVASC SEGPGLCLAR IDLHFLQQMR QHLPVFQHRR PDLYGSLGLP LS
Uniprot No.

Target Background

Function
Nitrilase homolog 1 (Nit1) catalyzes the hydrolysis of N-(4-oxoglutarate)-L-cysteinylglycine (deaminated glutathione), a crucial metabolite repair reaction that eliminates harmful deaminated glutathione. Nit1 plays a significant role in cell growth and apoptosis; reduced expression promotes cell growth, resistance to DNA damage, and increased susceptibility to NMBA-induced tumors. It exhibits tumor suppressor properties, enhancing apoptotic responsiveness in cancer cells, an effect additive to the tumor suppressor activity of FHIT. Furthermore, Nit1 acts as a negative regulator of primary T-cells.
Database Links
Protein Families
Carbon-nitrogen hydrolase superfamily, NIT1/NIT2 family
Subcellular Location
[Isoform 1]: Mitochondrion.; [Isoform 2]: Cytoplasm.

Q&A

What is the basic function and structure of Rat Nitrilase homolog 1?

Rat Nitrilase homolog 1 (Nit1) is a protein belonging to the nitrilase family, which are presumed to function as amidases based on sequence homology to known amidases and the presence of a Cys-Glu-Lys catalytic triad in the putative active site . While the precise substrate specificities remain partially characterized, Nit1 functions as a tumor suppressor that stimulates apoptosis in cancer cells . This protein is present in the cytoplasm and mitochondria but absent from nuclei of cells . Research findings show that Nit1 plays significant roles in cellular responses to replicative and oxidative stress, with its deficiency resulting in altered DNA damage responses, enhanced cell survival after exposure to damaging agents, and increased tumor susceptibility .

What cellular compartments contain Nit1 and how might this influence its function?

According to research data, Nit1 protein is primarily localized in the cytoplasm and mitochondria of rat cells, but is absent from nuclei . This subcellular localization pattern closely parallels that of Fhit, another tumor suppressor protein, suggesting potential functional relationships in these cellular compartments. The mitochondrial localization is particularly significant for understanding Nit1's potential role in apoptotic pathways, as mitochondria are central organelles in programmed cell death. Additionally, mitochondrial localization may relate to Nit1's role in oxidative stress responses, as mitochondria are major sites of reactive oxygen species (ROS) generation. The cytoplasmic presence of Nit1 indicates it likely interacts with signaling pathways and protein complexes in this compartment, potentially influencing cell cycle regulation and stress responses.

What evidence supports Nit1's role as a tumor suppressor?

Multiple lines of evidence establish Nit1 as a tumor suppressor. Knockout studies demonstrate that Nit1 deficiency in mice results in increased cell proliferation, enhanced survival of cells exposed to DNA-damaging agents, and an increased incidence of N-nitrosomethylbenzylamine (NMBA)-induced tumors . Conversely, overexpression of Nit1 leads to decreased cancer cell viability and increased caspase-3-dependent apoptosis, supporting its role in programmed cell death pathways . At the clinical level, loss of Nit1 expression has been observed in 48% of human esophageal adenocarcinomas, establishing its relevance to human cancers . The additive tumor susceptibility observed in Fhit−/−Nit1−/− double knockout mice compared to single gene knockouts further reinforces Nit1's independent role in tumor suppression . Additionally, Nit1-deficient cells show altered responses to replicative and oxidative stress, suggesting its involvement in DNA damage response pathways crucial for preventing genomic instability and malignant transformation.

How do Nit1 and Fhit deficiencies interact in tumor development models?

Research has clearly demonstrated that mice with combined deficiency of both Nit1 and Fhit (double knockout or DKO mice) develop more spontaneous and carcinogen-induced tumors than mice deficient in only Fhit . This additive effect on tumor susceptibility suggests that Nit1 and Fhit likely affect distinct signaling pathways in mammals, rather than operating in a single linear pathway . This finding is particularly interesting given the evolutionary history of these proteins—in flies and worms, they form a fusion protein encoded by the NitFhit gene, suggesting a closer functional relationship in these organisms . In carcinogen-induced tumor models using N-nitrosomethylbenzylamine (NMBA), the combined deficiency of Nit1 and Fhit leads to enhanced tumor development compared to single gene knockouts . This interaction indicates that both tumor suppressors provide complementary protection against carcinogenesis, suggesting that therapeutic approaches targeting both pathways might be more effective in certain cancer types.

How does Nit1 deficiency affect cellular responses to stress?

Nit1-deficient cells display dramatically altered responses to both replicative and oxidative stress. When treated with hydroxyurea, which induces replicative stress by inhibiting DNA synthesis, kidney-derived cells from Nit1-deficient mice fail to activate the pChk2 pathway . The Chk2 protein is a checkpoint kinase that normally becomes phosphorylated (pChk2) in response to DNA damage, leading to cell cycle arrest and DNA repair. The lack of pChk2 activation in Nit1-deficient cells suggests impaired ability to detect or respond to DNA damage caused by replicative stress. Similarly, when treated with hydrogen peroxide (H2O2) to induce oxidative stress, Nit1-deficient cells show little evidence of DNA damage compared to wild-type cells . This unusual response could indicate that Nit1 plays a role in pathways that recognize or process oxidative DNA damage. These altered stress responses may contribute to the increased survival of Nit1-deficient cells following exposure to DNA-damaging agents and ultimately to their increased susceptibility to tumor development.

How do environmental chemicals affect Nit1 expression and function?

Nit1 expression and function in rats are significantly modulated by exposure to various environmental chemicals. The Rat Genome Database documents numerous gene-chemical interaction annotations showing diverse effects on Nit1 . For example, bisphenol A demonstrates complex effects, including both decreasing and increasing Nit1 mRNA expression depending on experimental conditions, as well as increasing methylation of the Nit1 promoter . Diazinon, an organophosphate insecticide, increases methylation of the Nit1 gene while simultaneously decreasing Nit1 protein expression . Heavy metals show varied effects, with cadmium dichloride reported to both decrease and increase Nit1 mRNA expression . Arsenic compounds generally decrease Nit1 expression . These varied responses suggest complex regulatory mechanisms for Nit1 expression that are dependent on the specific chemical exposure, dose, duration, and experimental conditions. The sensitivity of Nit1 to environmental chemicals suggests it may serve as a mediator through which chemical exposures influence cancer risk.

What patterns emerge from chemical modulation of Nit1 expression?

Analysis of chemical interactions with Nit1 reveals several notable patterns. The following table summarizes key findings from the Rat Genome Database:

Chemical CategoryRepresentative CompoundsPredominant Effect on Nit1Evidence Type
Endocrine disruptorsBisphenol A, Bisphenol F, NonylphenolVariable expression changes; increased methylationExperimental and ISO
Heavy metalsCadmium dichloride, Arsenic compoundsPredominantly decreased expressionExperimental and ISO
Therapeutic drugsDoxorubicin, Gentamycin, OmeprazoleDecreased expressionExperimental and ISO
Environmental toxicantsDiazinon, Atrazine, ChloropreneVaried (increased expression/methylation)Experimental and ISO
Polycyclic compoundsBenzo[a]pyrene, Beta-lapachoneIncreased expressionISO

A significant pattern emerges showing that many environmental pollutants and toxicants affect Nit1 through epigenetic mechanisms, particularly DNA methylation . Additionally, compounds that induce oxidative stress frequently alter Nit1 expression, suggesting a potential link between Nit1 function and cellular redox status . These patterns may provide insights into how environmental exposures could modulate cancer risk through effects on tumor suppressor genes like Nit1.

What methodologies best reveal Nit1's role in tumor suppression?

Investigating Nit1's tumor suppressor function requires a multi-faceted methodological approach. In vivo tumor models using Nit1 knockout mice provide physiologically relevant systems for studying tumor development, particularly when challenged with carcinogens like NMBA . For mechanistic insights, cellular assays examining apoptosis (using Annexin V/PI staining, caspase activity assays), cell cycle progression (BrdU incorporation, flow cytometry), and DNA damage responses (γH2AX immunostaining, comet assays) in Nit1-deficient versus wild-type cells are valuable . Molecular interaction studies can identify Nit1's protein partners through techniques like co-immunoprecipitation or proximity labeling approaches. Substrate identification remains crucial, with metabolomic profiling of Nit1-deficient versus wild-type samples potentially revealing accumulating substrates. Clinical relevance can be established through analysis of Nit1 expression in human tumor samples using techniques like immunohistochemistry, which has already revealed loss of expression in 48% of esophageal adenocarcinomas . Combined, these methodological approaches provide a comprehensive understanding of Nit1's role in tumor suppression.

How can researchers effectively analyze Nit1 expression loss in cancers?

Assessing Nit1 expression loss in cancer tissues requires reliable and sensitive techniques. Immunohistochemistry (IHC) has proven effective for analyzing Nit1 protein expression in tissue sections, as demonstrated in studies of esophageal adenocarcinomas where 48% showed loss of expression . For reliable IHC analysis, careful validation of antibody specificity is essential, ideally using positive controls (normal tissue) and negative controls (tissue from Nit1 knockout models). Quantitative PCR (qPCR) provides sensitive detection of Nit1 mRNA levels in fresh or frozen tissue specimens, while digital droplet PCR (ddPCR) offers even greater sensitivity and precision for samples with low RNA quality. Western blotting can quantify Nit1 protein expression but requires tissue homogenization. For mechanistic insights into Nit1 loss, researchers should consider epigenetic analyses, particularly DNA methylation studies of the Nit1 promoter, given that chemical exposures like bisphenol A and diazinon have been shown to increase Nit1 methylation . Integration of these techniques can provide comprehensive characterization of Nit1 status in cancer tissues.

How do the enzymatic activities of Nit1 relate to its tumor suppressor function?

The relationship between Nit1's presumed amidase activity and its tumor suppressor function remains an area requiring further investigation. Nit1 contains a Cys-Glu-Lys catalytic triad typical of amidases , suggesting enzymatic function, but its natural substrates in mammalian cells are not fully characterized. Several research approaches could clarify this relationship: (1) Structure-function studies with site-directed mutagenesis of the catalytic triad to determine if enzymatic activity is essential for tumor suppression; (2) Substrate identification through metabolomic profiling comparing wild-type and Nit1-deficient samples; (3) Assessment of whether catalytically inactive Nit1 mutants can still suppress tumor development or promote apoptosis. Preliminary evidence suggests that Nit1's role in stress responses, particularly to oxidative and replicative stress, may connect its enzymatic function to tumor suppression . The identification of specific substrates could potentially reveal metabolic or signaling pathways through which Nit1 executes its tumor suppressor function, opening new avenues for therapeutic intervention in cancers with Nit1 deficiency.

What potential exists for therapeutic targeting of Nit1 pathways in cancer?

Understanding Nit1's role in tumor suppression offers several potential therapeutic avenues for cancer treatment. For cancers with reduced but not completely lost Nit1 expression, small molecule activators could enhance the tumor suppressor function of remaining Nit1 protein. Given that Nit1 deficiency results in altered responses to replicative and oxidative stress , synthetic lethality approaches targeting these altered stress responses could selectively kill Nit1-deficient cancer cells while sparing normal cells. The additive effects of Nit1 and Fhit deficiencies on tumor susceptibility suggest that cancers with deficiencies in both proteins might benefit from combination therapies targeting both pathways. For the 48% of esophageal adenocarcinomas showing loss of Nit1 expression , Nit1 status could serve as a biomarker for treatment selection or patient stratification. Drug discovery efforts could focus on compounds that specifically target cellular vulnerabilities created by Nit1 deficiency, such as altered DNA damage responses or apoptotic pathways, potentially leading to more personalized cancer treatment approaches.

How might Nit1 function in non-cancer pathological conditions?

While research has primarily focused on Nit1's role in cancer, its involvement in cellular stress responses suggests potential functions in non-cancer pathologies. Nit1's presence in mitochondria indicates possible roles in mitochondrial disorders or conditions involving oxidative stress. Neurodegenerative diseases often involve disruptions in proteostasis and stress responses, areas where Nit1's presumed amidase activity could be relevant. Similarly, inflammatory diseases feature oxidative stress components where Nit1 might play regulatory roles. The sensitivity of Nit1 expression to various environmental chemicals suggests it might function in toxicant-induced pathologies beyond cancer. The evolutionary conservation of Nit1, particularly its fusion with Fhit in invertebrates , hints at fundamental cellular functions that could be relevant across multiple disease contexts. Future research should explore Nit1 expression and function in various disease models, particularly those involving oxidative stress, DNA damage, or mitochondrial dysfunction, to determine if Nit1-targeted approaches might have therapeutic potential beyond cancer.

What are the critical controls for Nit1 knockout or knockdown experiments?

Designing rigorous Nit1 knockout or knockdown experiments requires several critical controls to ensure valid interpretation of results. For genetic knockouts using CRISPR/Cas9, researchers should include wild-type parental cells, clonal control cells that underwent the CRISPR process but maintain Nit1 expression, and ideally rescue experiments where Nit1 expression is restored in knockout cells. For knockdown approaches using siRNA or shRNA, appropriate controls include non-targeting siRNA/shRNA controls and rescue experiments with overexpression of an siRNA/shRNA-resistant Nit1 construct. Verification of Nit1 depletion should employ multiple methods, including Western blotting for protein levels, qPCR for mRNA expression, and potentially functional assays if suitable for Nit1 activity. Given Nit1's role in stress responses , experimental design should carefully control stress conditions that might influence results, including cell density, passage number, and media conditions. When examining phenotypes like apoptosis or DNA damage responses in Nit1-deficient models, researchers should test multiple stimuli and concentrations to fully characterize alterations in cellular responses. These controls ensure that observed phenotypes can be specifically attributed to Nit1 deficiency rather than off-target effects or experimental artifacts.

What purification strategies yield optimal recombinant rat Nit1 quality?

Purification of recombinant rat Nit1 requires strategies tailored to its biochemical properties. A multi-step purification protocol typically begins with affinity chromatography using tags incorporated into the recombinant construct, such as hexahistidine (His6) for immobilized metal affinity chromatography or GST for glutathione sepharose purification. Following initial capture, secondary purification steps should include ion exchange chromatography to separate Nit1 from proteins with different charge properties, and size exclusion chromatography as a final polishing step to separate monomeric Nit1 from aggregates and to perform buffer exchange. Throughout purification, maintaining reducing conditions is crucial to preserve the catalytic cysteine residue in Nit1's active site. Inclusion of protease inhibitors and working at reduced temperatures (4°C) helps prevent degradation during purification. Buffer optimization should consider Nit1's stability, with typical buffers including 20-50 mM Tris or phosphate at pH 7.5-8.0, 150-300 mM NaCl, and 1-5 mM reducing agent (DTT or β-mercaptoethanol). Validation of purified protein quality should include activity assays if suitable substrates are available, as well as biophysical characterization through techniques like circular dichroism or thermal shift assays to confirm proper folding.

Quick Inquiry

Personal Email Detected
Please use an institutional or corporate email address for inquiries. Personal email accounts ( such as Gmail, Yahoo, and Outlook) are not accepted. *
© Copyright 2025 TheBiotek. All Rights Reserved.