MGSSHHHHHH SSGLVPRGSH QSFGAVCKEP QEEVVPGGGR SKRDPDLYQL LQRLFKSHSS LEGLLKALSQ ASTDPKESTS PEKRDMHDFF VGLMGKRSVQ PDSPTDVNQE NVPSFGILKY PPRAE.
TAC3 belongs to the tachykinin family of neuropeptides distributed throughout the mammalian central and peripheral nervous systems. Tachykinins are characterized by a common C-terminal structure (Phe-Xaa-Gly-Leu-Met-NH2) and include TAC1-3 . TAC3 encodes neurokinin B (NKB), which functions primarily as a signaling molecule in the central nervous system.
From a molecular perspective, the TAC3 gene undergoes transcription and translation to produce the NKB precursor protein, which is then post-translationally modified to yield the active neuropeptide. This processing typically involves enzymatic cleavage at dibasic amino acid residues followed by C-terminal amidation, which is essential for biological activity .
TAC3 has been particularly recognized for its indispensable role in physiological development and reproductive function. It modulates gonadotropin-releasing hormone (GnRH) secretion through action on kisspeptin neurons in the hypothalamus, thereby influencing the entire hypothalamic-pituitary-gonadal axis . Beyond reproduction, emerging research indicates roles in neurotransmission, pain perception, and potentially in disease processes including cancer.
The TAC3 gene shows notable evolutionary conservation but with specific variations across species. Human TAC3 is located on chromosome 12, while the mouse counterpart is found on chromosome 6. Comparative genomic analysis reveals important structural and functional differences between species that researchers must consider when designing translational studies.
In humans, the TAC4 gene (related to TAC3) can generate four splice variants leading to four unique tachykinin peptides named endokinins A-D (EKA-D) . This represents a significant divergence from the mouse TAC4 gene, which encodes only a single transcript. The human genome contains an additional unique exon (exon 3) that provides the N-terminal dibasic residues for cleavage of the second human tachykinin from its precursor .
When comparing the tachykinin peptides themselves, there are notable sequence differences particularly at the N-termini. While the structures of substance P, neurokinin A, and neurokinin B are identical throughout mammals, there are unusual species differences between the hemokinin (HK) peptides . These distinctions necessitate careful consideration when extrapolating findings from animal models to human applications.
In the central nervous system, TAC3 is predominantly expressed in specific hypothalamic nuclei where it participates in reproductive function regulation. Outside the central nervous system, research has identified TAC3 expression in peripheral sensory nerves, including those in the mandible . Interestingly, while investigating oral squamous cell carcinoma, researchers found that TAC3 was not detected in tumor cells but was expressed in PGP-9.5-positive sensory nerves in the mandible .
The receptor TACR3 shows more widespread expression and has been identified in numerous tissues. In studies of oral squamous cell carcinoma, TACR3 was found to be highly elevated in tumor cells despite being absent in normal epithelium, with particularly intense signals observed at the invasive front of tumor cells migrating into mandible bone matrix . This differential expression pattern suggests potential roles in cancer progression that warrant further investigation.
The TAC3/TACR3 signaling system holds a central position in regulating the hypothalamic-pituitary-gonadal (HPG) axis. TAC3, which encodes neurokinin B (NKB), and its receptor TACR3 primarily function within the hypothalamus to modulate gonadotropin-releasing hormone (GnRH) secretion . This modulation occurs through direct action on kisspeptin-1 neurons, creating a sophisticated regulatory network that orchestrates reproductive hormone release .
Within the hypothalamus, NKB acts through TACR3, a G protein-coupled receptor, to stimulate kisspeptin neurons. These neurons subsequently promote GnRH release, which triggers luteinizing hormone (LH) and follicle-stimulating hormone (FSH) secretion from the anterior pituitary . This cascade ultimately controls gonadal development, steroidogenesis, and gametogenesis.
The critical nature of this pathway is demonstrated by the phenotypic consequences of TAC3/TACR3 dysfunction. Individuals with loss-of-function mutations in these genes typically present with normosmic isolated hypogonadotropic hypogonadism (IHH), characterized by failure of pubertal development and infertility . Clinical observations indicate that patients with TACR3 mutations often display a distinctive FSH/LH ratio (mean 2.40±1.33 in one study), which may serve as a diagnostic biomarker .
Mutations in TAC3 and TACR3 genes have profound impacts on reproductive function, primarily manifesting as normosmic isolated hypogonadotropic hypogonadism (IHH). This condition is characterized by the failure of sexual maturation, impaired gametogenesis, and infertility, despite normal olfactory function .
Clinical findings reveal distinctive phenotypic presentations. In one documented case, a 30-year-old male with a homozygous p.W275* variant in TACR3 presented with micropenis, small testes (< 2.0 × 1.5 cm), and pubic hair at Tanner stage III. Hormonal evaluation showed significantly reduced levels of basal gonadotropins (LH: < 0.6 U/L; FSH: < 1.0 U/L) and testosterone (35 ng/dL) . This case exemplifies the classic presentation of IHH resulting from TACR3 dysfunction.
The molecular mechanisms underlying these phenotypes involve disruption of the hypothalamic-pituitary-gonadal axis signaling cascade. When TAC3/TACR3 signaling is compromised, the pulsatile release of GnRH is altered, leading to insufficient gonadotropin production and consequently inadequate gonadal stimulation . The typical biochemical profile includes low levels of FSH, LH, and sex steroids, with a characteristic elevation in the FSH/LH ratio.
Importantly, beyond reproductive deficits, individuals with TAC3/TACR3 mutations may experience psychological consequences. Research indicates that hypogonadal men commonly experience depression and anxiety, attributed partially to low testosterone levels . These findings highlight the broader physiological and psychological impacts of TAC3/TACR3 dysfunction.
Research into TAC3 expression in reproductive tissues employs a multifaceted methodological approach that combines molecular, cellular, and physiological techniques. Understanding these methodologies is crucial for designing rigorous research protocols in this field.
Genetic and Transcriptomic Analysis:
Polymerase chain reaction (PCR) and RACE (Rapid Amplification of cDNA Ends) techniques are employed to amplify and identify TAC3 transcripts . In human studies, primers are designed to match regions on human clone AC027801 homologous to the mouse TAC4 gene .
For variant detection, the entire coding regions and intron-exon junctions of TAC3 and TACR3 genes are amplified by PCR using specific primers followed by automatic sequencing . This approach has successfully identified mutations associated with reproductive disorders.
Protein Expression Analysis:
Immunohistochemistry and immunofluorescence analysis are standard techniques for examining TAC3/TACR3 protein expression patterns in tissue samples . These methods allow for visualization of protein localization and semi-quantitative assessment of expression levels.
Western blotting provides quantitative protein expression data and can be used to compare TAC3/TACR3 levels across different tissues or experimental conditions.
Functional Assays:
Receptor binding assays measure the interaction between NKB and TACR3, providing insights into receptor functionality.
Second messenger signaling assays evaluate downstream pathway activation following receptor stimulation.
In vitro cell culture systems expressing TAC3/TACR3 allow for controlled studies of pathway dynamics.
Clinical Assessment:
Hormonal profiling through blood sampling measures LH, FSH, and sex steroid levels, providing functional readouts of the TAC3/TACR3 system in human subjects .
Physical examination parameters such as Tanner staging for pubertal development offer clinical correlates to molecular findings .
TAC3 and its receptor TACR3 exert significant influence on neural signaling throughout the central nervous system, extending well beyond their established roles in reproductive function. As a G protein-coupled receptor, TACR3 activates multiple intracellular signaling cascades that modulate neuronal excitability, synaptic transmission, and plasticity .
In the hippocampus, TACR3 activation affects synaptic plasticity through regulation of calcium-dependent pathways. Research indicates that TACR3 signaling modulates CaMKII (calcium/calmodulin-dependent protein kinase II) activation and AMPA receptor phosphorylation . These molecular changes directly impact synaptic strength and connectivity. Specifically, inhibition of TACR3 activity has been shown to provoke hyperactivation of CaMKII and enhanced AMPA receptor phosphorylation, associated with increased spine density .
Electrophysiological studies using multielectrode arrays have demonstrated that TACR3 inhibition leads to stronger cross-correlation of firing among neurons, indicating enhanced connectivity . This finding suggests that TACR3 normally functions to regulate synaptic strength and neural network dynamics. Furthermore, deficient TACR3 activity in rats results in impaired long-term potentiation (LTP) in the dentate gyrus, a critical mechanism underlying learning and memory .
The spatial distribution of TACR3 within neurons is also significant for its function. The receptor is predominantly expressed in the cell membrane, including the presynaptic compartment, positioning it to modulate neurotransmitter release . This strategic localization allows TACR3 to influence synaptic transmission directly.
The interrelationship between hippocampal TACR3 expression, testosterone levels, and anxiety represents a fascinating nexus of neuroendocrine function with significant implications for both basic neuroscience and clinical applications. Research has unveiled a complex bidirectional relationship where TACR3 influences testosterone production, while testosterone modulates TACR3 expression .
Studies show that downregulation of TACR3 in specific brain regions, such as the lateral habenula, is associated with anxiety-like behaviors in mice. Conversely, TACR3 overexpression in these same areas significantly reverses such behaviors . This relationship extends to clinical observations, as hypogonadal men with mutations in TAC3/TACR3 genes frequently experience depression and anxiety as comorbidities .
The molecular mechanism connecting these factors involves testosterone's influence on hippocampal TACR3 expression. Sexual development in male rats coincides with a substantial increase in hippocampal TACR3 expression, which correlates with elevated serum testosterone and reduced anxiety . This suggests a developmental programming where rising testosterone levels during puberty upregulate TACR3 expression, which in turn modulates anxiety-related neural circuits.
At the synaptic level, TACR3 dysfunction leads to altered spine morphology and synaptic connectivity. Aberrant expression of functional TACR3 in dendritic spines results in spine shrinkage and pruning, while expression of defective TACR3 increases spine density and size . These structural changes modify the firing patterns of hippocampal neurons, particularly in response to LTP induction. Remarkably, treatment with testosterone can rectify these abnormal firing patterns in neurons expressing defective TACR3 , demonstrating a direct therapeutic effect of testosterone on TACR3-mediated neural dysfunction.
Selecting appropriate experimental models is crucial for advancing our understanding of TAC3/TACR3 in neural functions. Several complementary approaches provide insights into different aspects of this signaling system, each with specific advantages and limitations.
In Vitro Cellular Models:
Primary neuronal cultures from hippocampus or hypothalamus allow for detailed investigation of TACR3 signaling mechanisms at the cellular level.
Neuronal cell lines expressing TACR3 provide a controlled system for studying receptor pharmacology and downstream pathways.
These models are particularly valuable for examining the molecular mechanisms of TACR3 signaling, including calcium dynamics, CaMKII activation, and AMPA receptor phosphorylation .
Ex Vivo Tissue Preparations:
Acute brain slices maintain neural circuitry while allowing access for electrophysiological recordings and pharmacological manipulations.
Multielectrode arrays enable assessment of neuronal network activity and connectivity following TACR3 modulation .
This approach has revealed that TACR3 inhibition leads to stronger cross-correlation of firing among neurons, indicating enhanced connectivity .
In Vivo Rodent Models:
Genetic models with TAC3/TACR3 mutations or conditional knockouts provide insights into developmental and functional roles.
Behavioral assays combined with in vivo electrophysiology allow correlation between TACR3 function and anxiety-like behaviors.
Studies in rats have demonstrated that deficient TACR3 activity leads to lower serum testosterone levels, increased spine density, and impaired long-term potentiation in the dentate gyrus .
Estrous Cycle Models:
Female rats exhibit fluctuations in TACR3 expression during the estrous cycle, making them valuable models for studying hormonal influences on TACR3 function .
This approach has revealed that TACR3 expression is sensitive to sex hormone levels, with implications for gender differences in TACR3-related phenotypes.
Human Clinical Studies:
Patients with identified TAC3/TACR3 mutations provide invaluable insights into the physiological relevance of findings from animal models .
Neuroimaging combined with hormonal and psychiatric assessment can establish correlations between TACR3 function, testosterone levels, and anxiety in humans.
Investigating the role of TAC3 in oral squamous cell carcinoma (OSCC) requires a sophisticated research approach that combines tissue analysis, molecular profiling, and functional studies. The discovery that TACR3 is highly elevated in OSCC tumor cells, despite being absent in normal epithelium, suggests its potential role in cancer progression .
Tissue Expression Analysis:
Researchers should employ immunohistochemistry and immunofluorescence to examine TAC3/TACR3 expression patterns in clinical OSCC samples. This approach has revealed that TACR3 shows particularly intense signals at the invasive front of tumor cells that have migrated into the mandible bone matrix . Simultaneous staining for PGP-9.5 (a neuronal marker) and TAC3 can identify TAC3-expressing sensory nerves in the mandible microenvironment .
Co-culture Systems:
To model the interaction between nerve-derived TAC3 and TACR3-expressing tumor cells, researchers can develop co-culture systems using:
Primary sensory neurons or neuronal cell lines expressing TAC3
OSCC cell lines expressing TACR3
Three-dimensional bone matrix mimetics to recreate the mandible microenvironment
This approach allows for controlled assessment of how nerve-derived TAC3 affects OSCC cell behavior, including invasion, migration, and proliferation.
Functional Assays:
Key functional readouts should include:
Migration and invasion assays to quantify the effect of TAC3/TACR3 signaling on tumor cell motility
Proliferation and survival assays to assess growth effects
Matrix metalloproteinase activity assays to evaluate bone matrix degradation
Calcium imaging to visualize TACR3-mediated signaling in real-time
Molecular Pathway Analysis:
RNA sequencing and phospho-proteomics can identify downstream effectors of TACR3 activation in OSCC cells, potentially revealing novel therapeutic targets. Special attention should be paid to pathways regulating epithelial-mesenchymal transition and bone invasion.
In Vivo Models:
Orthotopic OSCC models with TAC3/TACR3 manipulation (overexpression, knockdown, or pharmacological inhibition) can validate in vitro findings. Bioluminescence imaging allows for non-invasive monitoring of tumor progression over time.
Identifying mutations in TAC3/TACR3 genes requires a comprehensive genetic analysis approach that maximizes detection sensitivity while minimizing false positives. Several complementary techniques offer distinct advantages for different research contexts.
Targeted Sequencing:
Sanger sequencing of TAC3/TACR3 coding regions and intron-exon junctions remains the gold standard for confirming specific variants . This approach is particularly useful when examining candidate regions in patients with suspected TAC3/TACR3-related disorders. PCR amplification of the entire coding region of both genes followed by automatic sequencing has successfully identified variants in patients with pubertal disorders .
Next-Generation Sequencing Panels:
Custom gene panels including TAC3, TACR3, and related reproductive pathway genes provide higher throughput than Sanger sequencing while maintaining depth of coverage. This approach is cost-effective for screening larger patient cohorts and can detect both common and rare variants.
Whole Exome Sequencing (WES):
WES offers comprehensive coverage of all coding regions, enabling detection of variants in TAC3/TACR3 alongside potential modifier genes. This approach is particularly valuable for patients with complex phenotypes that may involve multiple genetic factors.
RNA Sequencing:
RNA-Seq can identify alternative splicing events and expression changes in TAC3/TACR3. This is especially relevant given that the human TAC4 gene (related to TAC3) generates four splice variants leading to four unique tachykinin peptides . SMART RACE (Rapid Amplification of cDNA Ends) techniques have been successfully employed to identify and characterize these splice variants .
Functional Validation:
Identifying variants is only the first step; functional validation is essential for establishing pathogenicity. This typically involves:
In silico prediction tools to assess potential functional impacts
In vitro expression studies to examine protein stability and subcellular localization
Receptor binding and signaling assays to evaluate functional consequences
CRISPR/Cas9-mediated genome editing to recapitulate mutations in cellular or animal models
Endokinins (EKA-D) represent an important expansion of the tachykinin family with significant implications for TAC3 research. These peptides are encoded by the human tachykinin precursor 4 gene (TAC4), which generates four mRNAs (α, β, γ, δ) through alternative splicing . Understanding the relationship between endokinins and TAC3 opens new avenues for research and potential therapeutic applications.
Expression Patterns:
Unlike the neurokinin B encoded by TAC3, which is predominantly expressed in the central nervous system, endokinins show widespread peripheral tissue distribution . This complementary expression pattern suggests that endokinins may serve as peripheral tachykinin signaling molecules in tissues where TAC3 expression is limited. Research indicates that EKA/B may function as "the peripheral SP-like endocrine/paracrine agonists where SP is not expressed" .
Evolutionary Significance:
Comparative genomic analysis reveals interesting evolutionary patterns. The mouse TAC4 gene encodes only a single transcript, while the human gene results in four splice variants . This divergence is attributed to an additional unique exon (exon 3) in humans that provides the N-terminal dibasic residues for cleavage of the second human tachykinin from its precursor . This evolutionary distinction highlights the importance of species-specific considerations in translational research.
Research Implications:
For comprehensive investigation of tachykinin signaling, researchers should consider the following approaches:
Simultaneous profiling of TAC3 and TAC4 expression in tissues of interest
Evaluation of receptor binding profiles for neurokinin B and endokinins
Assessment of potential functional redundancy or complementarity
Analysis of coordinated regulation of TAC3 and TAC4 expression
Accurate measurement of TAC3/TACR3 expression across diverse tissues requires selecting appropriate techniques based on research objectives, tissue characteristics, and desired level of quantification. A multi-technique approach often provides the most comprehensive assessment.
Nucleic Acid-Based Methods:
For transcript detection and quantification:
RT-PCR offers high sensitivity for qualitative detection of TAC3/TACR3 mRNA, with specific primers designed to match regions on human clone AC027801 for TAC3 gene amplification .
Quantitative RT-PCR provides precise quantification of transcript levels and can detect splice variants when properly designed. For normalization, GAPDH has been effectively used as a reference gene in TAC3/TACR3 studies .
RNA-Seq offers comprehensive transcriptome analysis, allowing simultaneous examination of TAC3/TACR3 along with related pathways. This approach has revealed tissue-specific expression patterns across different human and mouse tissues .
In situ hybridization enables localization of TAC3/TACR3 mRNA within complex tissues while preserving spatial context, particularly valuable for heterogeneous tissues like brain.
Protein Detection Methods:
For protein localization and quantification:
Immunohistochemistry is optimal for examining TAC3/TACR3 expression patterns in fixed tissue samples, as demonstrated in studies of oral squamous cell carcinoma .
Immunofluorescence, often combined with confocal microscopy, allows co-localization studies with other proteins. This approach has successfully shown that TAC3 is expressed in PGP-9.5-positive sensory nerves in the mandible .
Western blotting provides semi-quantitative assessment of total protein levels.
Proximity ligation assays can detect protein-protein interactions involving TAC3/TACR3.
Functional Readouts:
For assessing receptor activity:
Calcium imaging measures TACR3 activation through changes in intracellular calcium.
Phospho-specific antibodies detect activation of downstream signaling molecules such as CaMKII .
Electrophysiological recordings capture TACR3-mediated changes in neuronal activity .
Validation Strategies:
For ensuring specificity and accuracy:
Multiple antibodies targeting different epitopes
Genetic controls (knockout tissues or siRNA-treated cells)
Peptide competition assays to confirm antibody specificity
Cross-validation with complementary techniques
Conflicting data in TAC3/TACR3 research is not uncommon given the complex nature of neuropeptide signaling, diverse experimental approaches, and tissue-specific effects. Addressing such contradictions requires a systematic and rigorous approach to ensure scientific advancement rather than confusion.
Methodological Evaluation:
When confronting conflicting results, researchers should first scrutinize methodological differences:
Species differences: TAC3/TACR3 shows important species variations, with human and mouse TAC4 genes encoding different numbers of tachykinin peptides .
Tissue specificity: TACR3 expression varies significantly across tissues and may even show different patterns within regions of the same tissue, as seen in the hippocampus .
Technical variations: Different antibodies, PCR primers, or expression detection methods may yield varying results based on sensitivity and specificity.
Sex differences: TACR3 expression in female rats fluctuates during the estrous cycle, reflecting sensitivity to sex hormones .
Developmental stage: Sexual development in males is associated with substantial increases in hippocampal TACR3 expression .
Integration Strategies:
To reconcile contradictory findings, consider these approaches:
Perform comparative studies using standardized protocols across different model systems
Conduct meta-analyses of published data with particular attention to methodological variables
Design experiments specifically to test competing hypotheses
Employ multiple complementary techniques to examine the same biological question
Consider context-dependent effects where both contradictory findings may be valid under specific conditions
Specific TAC3/TACR3 Considerations:
Some recurring issues in this field include:
Differential effects of TACR3 activation versus inhibition on spine morphology and synaptic plasticity
Varying roles of TACR3 in different brain regions (hypothalamus versus hippocampus)
Seemingly contradictory effects of testosterone on TACR3 expression across developmental stages
Reporting Recommendations:
When publishing potentially contradictory findings:
Explicitly acknowledge contradictions with existing literature
Discuss potential methodological or biological explanations
Consider publishing detailed protocols to enhance reproducibility
Include negative results alongside positive findings
Transparently report all experimental conditions that might affect outcomes
The rapidly evolving landscape of biomedical research technologies offers exciting opportunities to advance our understanding of TAC3/TACR3 biology. These emerging approaches can address current limitations and open new avenues of investigation.
Single-Cell Technologies:
Single-cell RNA sequencing can reveal cell-type specific expression patterns of TAC3/TACR3 and identify previously unknown cell populations involved in tachykinin signaling.
Single-cell proteomics may eventually allow protein-level characterization of TAC3/TACR3 expression with cellular resolution.
Spatial transcriptomics preserves tissue architecture while providing transcriptomic data, offering insights into TAC3/TACR3 expression in relation to anatomical features.
Advanced Imaging Approaches:
Super-resolution microscopy enables visualization of TACR3 subcellular localization with unprecedented detail, potentially revealing compartmentalization within synapses.
In vivo calcium imaging with genetically encoded calcium indicators permits real-time monitoring of TACR3-mediated neuronal activity in behaving animals.
PET tracers for TACR3 could enable non-invasive imaging of receptor distribution in humans, bridging animal and clinical research.
Genetic Engineering Tools:
CRISPR/Cas9-based approaches allow precise manipulation of TAC3/TACR3 genes, including introduction of specific mutations identified in patients.
Conditional and cell-type specific knockout models can dissect the role of TAC3/TACR3 in specific tissues or developmental stages.
Optogenetic and chemogenetic tools coupled with TAC3/TACR3 manipulation permit temporal control of pathway activation.
Computational Approaches:
Machine learning algorithms applied to large datasets may identify novel patterns in TAC3/TACR3 expression or activity.
Systems biology modeling can integrate diverse experimental data to predict network-level effects of TAC3/TACR3 modulation.
Molecular dynamics simulations offer insights into TACR3 structural changes upon ligand binding, potentially informing drug development.
Translational Technologies:
Organ-on-chip systems incorporating TAC3-expressing neurons and TACR3-expressing target cells can model complex tissue interactions.
Patient-derived iPSCs differentiated into relevant cell types allow study of TAC3/TACR3 function in human genetic disease backgrounds.
High-throughput drug screening platforms targeting TACR3 may identify novel therapeutic compounds for conditions ranging from reproductive disorders to anxiety .
Tachykinin-3, also known as Neurokinin B, is a member of the tachykinin family of neuropeptides. These peptides are characterized by their ability to excite neurons, evoke behavioral responses, act as potent vasodilators and secretagogues, and contract various smooth muscles. Tachykinin-3 is encoded by the TAC3 gene in humans .
The TAC3 gene encodes a precursor protein that is proteolytically processed to generate the mature Tachykinin-3 peptide. This peptide is primarily expressed in the central and peripheral nervous systems and functions as a neurotransmitter. The mature peptide is the ligand for the neurokinin-3 receptor (NK3R), which is involved in various physiological processes .
Tachykinin-3 plays a critical role in the regulation of gonadal function. It is a central regulator of reproductive hormones and is involved in the modulation of the hypothalamic-pituitary-gonadal axis. This peptide is also expressed in the outer syncytiotrophoblast of the placenta and may be associated with pregnancy-induced hypertension and pre-eclampsia .
Mutations in the TAC3 gene are associated with normosmic hypogonadotropic hypogonadism, a condition characterized by delayed or absent puberty and impaired reproductive function. This condition can occur with or without anosmia (loss of the sense of smell). Tachykinin-3 is also being studied for its potential role in various other disorders, including infertility and certain types of cancer .
Human recombinant Tachykinin-3 is produced using recombinant DNA technology, which involves inserting the TAC3 gene into a suitable expression system, such as bacteria or yeast, to produce the peptide in large quantities. This recombinant form is used in research to study the physiological and pathological roles of Tachykinin-3 and to develop potential therapeutic applications .
Research on Tachykinin-3 has provided insights into its role in various physiological processes and its potential as a therapeutic target. For example, studies have shown that Tachykinin-3 and its receptor NK3R are involved in the regulation of mood, stress, and pain. Additionally, Tachykinin-3 analogs and antagonists are being explored for their potential use in treating conditions such as depression, anxiety, and chronic pain .