Background: Mounting evidence underscores the significance of cellular diversity within the endocrine system and the intricate interplay between different cell types and tissues, essential for preserving physiological balance and influencing disease trajectories. The pituitary gland, a central player in the endocrine orchestra, exemplifies this complexity with its assortment of hormone-secreting and nonsecreting cells. Summary: The pituitary gland houses several types of cells responsible for hormone production, alongside nonsecretory cells like fibroblasts and endothelial cells, each playing a crucial role in the gland’s function and regulatory mechanisms. Despite the acknowledged importance of these cellular interactions, the detailed mechanisms by which they contribute to pituitary gland physiology and pathology remain largely uncharted. The last decade has seen the emergence of groundbreaking technologies such as single-cell RNA sequencing, offering unprecedented insights into cellular heterogeneity and interactions. However, the application of this advanced tool in exploring the pituitary gland’s complexities has been scant. This review provides an overview of this methodology, highlighting its strengths and limitations, and discusses future possibilities for employing it to deepen our understanding of the pituitary gland and its dysfunction in disease states. Key Message: Single-cell RNA sequencing technology offers an unprecedented means to study the heterogeneity and interactions of pituitary cells, though its application has been limited thus far. Further utilization of this tool will help uncover the complex physiological and pathological mechanisms of the pituitary, advancing research and treatment of pituitary diseases.

The intricacy of the human body is epitomized by its composition of approximately 30–40 trillion cells, encompassing over 200 distinct cell types [1, 2]. This cellular diversity underpins critical biological processes, including development, regeneration, and aging, by affecting intercellular signaling, metabolism, and cell division [3]. In the context of the pituitary gland – an endocrine beacon orchestrating a plethora of physiological processes – cellular heterogeneity assumes a paramount role. The pituitary gland’s multifaceted cell types contribute to its complex function, development, and pathophysiology [4]. Traditional research methodologies, while foundational, often fall short in dissecting this cellular complexity due to their inherent limitations in resolving the nuances of cell-to-cell interactions and gene expression changes.

The last decade has witnessed remarkable technological advancements, notably single-cell RNA sequencing (scRNA-seq), which has revolutionized our understanding of cellular diversity [5]. By enabling the detailed analysis of individual cells, scRNA-seq has emerged as a powerful tool for unveiling previously unrecognized cell subtypes, delineating intricate cellular interactions, and understanding the molecular mechanisms underpinning cellular functions [6]. However, the application of these technologies to endocrine organs, especially the pituitary gland, remains nascent.

This review aims to illuminate the pivotal role of scRNA-seq in advancing pituitary gland research. By unraveling the gland’s cellular landscape and molecular dynamics, scRNA-seq offers novel insights into the normal functioning of the pituitary, as well as the genesis and progression of pituitary disorders. It facilitates a granular understanding of cellular heterogeneity, which is crucial for the identification of therapeutic targets and the development of personalized medicine approaches. Moreover, in delving into the applications of scRNA-seq in pituitary research, this review not only highlights the current state of knowledge but also sets the stage for future investigations that could redefine endocrine research. By offering a deeper understanding of the pituitary gland’s cellular diversity and its implications for health and disease, scRNA-seq paves the way for innovative therapeutic strategies and enhances our ability to tackle complex pituitary disorders.

Before scRNA-seq, identifying cell surface markers without prior genetic or protein knowledge was challenging, often relying on methods like FACS or cytometry [7]. scRNA-seq offers an alternative which is ideal for analyzing cellular transcriptomes and cell-type compositions, even with limited samples [8]. Standard scRNA-seq workflow includes sample preparation, cell isolation, reverse transcription and amplification, sequencing, and bioinformatic analysis.

Experimental Process

Single-Cell Isolation

Single-cell isolation in scRNA-seq requires precise techniques [9]. Options vary based on experimental needs and include serial dilution, micropipette aspiration, FACS, LCM, and microfluidic systems [10, 11]. FACS sorts cells efficiently but can cause damage, while LCM preserves spatial data but is technically demanding [12, 13]. Microfluidic methods like Fluidigm C1 offer high throughput and automation [14]. Each method’s selection depends on specific experimental requirements, balancing precision, efficiency, and cost.

Reverse Transcription and Amplification

CDNA synthesis integrates unique molecular identifiers (UMIs), which are random sequences that act as barcodes to track transcripts [15]. Sequencing adapters are added for platform compatibility. PCR amplification, crucial for library construction, uses techniques like template switching and in vitro transcription to enhance the sequencing process [16].

Sequencing

Researchers have developed over 15 distinct scRNA-seq methodologies (shown in Fig. 1; Table 1), categorized into full-length and tag-based approaches. Full-length methods cover the entire gene body to facilitate gene expression analysis and detection of SNPs, splice variants, and mutations [17, 18]. Tag-based methods focus on sequencing the 5′ or 3′ ends, trading full-length coverage for improved transcript abundance estimation and enhanced cDNA synthesis multiplexing [19, 20]. Tang et al. [21, 22] initiated scRNA-seq innovation by isolating single cells for cDNA synthesis using oligo-dT primers with anchor sequences for PCR amplification, followed by enzymatic polyA tailing. Smart-seq by Ramsköld et al. [18] in 2012 and its iterations, Smart-seq2 and Smart-seq3, have advanced tumor cell research by identifying variable splicing and allele-specific expression, with Smart-seq3 integrating full-length coverage with a UMI strategy [23, 24]. Meanwhile, Quartz-seq simplifies the protocol in a single PCR tube without purification, enhancing sensitivity and reproducibility [25]. CEL-seq uses linear in vitro transcription with a barcode and T7 promoter for RNA amplification, with CEL-seq2 introducing UMIs to reduce costs and operational time [26]. The progression from CEL-seq to MARS-seq emphasizes automation with cell barcodes and UMIs for multiplexing, enhancing throughput and cost efficiency [27]. STRT-seq has adapted for use with microfluidic platforms, with STRT/C1 incorporating UMI tags for precision in library enrichment [28, 29]. Microfluidic methods like CytoSeq and BD Rhapsody, and droplet-based techniques like Drop-seq and inDrop, enable high-throughput cell sequencing with minimal reagent use and reduced costs [20, 30, 31]. Each scRNA-seq method offers unique benefits, with Smart-seq2 noted for detecting the highest number of genes per cell and microfluidic methods reducing amplification noise with UMIs [19, 20].

Fig. 1.

Timeline of publication and throughput of different scRNA-seq methods.

Fig. 1.

Timeline of publication and throughput of different scRNA-seq methods.

Close modal
Table 1.

List of critical scRNA-seq methods and platforms

MethodologyTranscript coverageIsolationYearLibrary amplification
STRT-seq 5′ FACS 2011 PCR 
CEL-seq 3′ Micromanipulation 2012 In vitro transcription 
SMART-seq Full length FACS 2012 PCR 
Quartz-seq Full length FACS 2013 PCR 
SMART-seq2 Full length FACS 2013 PCR 
MARS-seq 3′ FACS 2014 In vitro transcription 
Cyto-seq 3′ Micromanipulation 2015 PCR 
Drop-seq 3′ Droplet 2015 PCR 
inDrop 3′ Droplet 2015 In vitro transcription 
CEL-seq2 3′ FACS 2016 In vitro transcription 
10x Genomics 3′ or 5′ Droplet 2017 PCR 
MATQ-seq Full length Micromanipulation 2017 PCR 
Sci-RNA-seq 3′ In situ barcoding 2017 PCR 
Seq-Well 3′ Microwell platform 2017 PCR 
Microwell-seq 3′ FACS 2018 PCR 
Quartz-seq2 Full length Droplet 2018 PCR 
SPLit-seq 3′ In situ barcoding 2018 PCR 
MARS-seq2 3′ FACS 2019 In vitro transcription 
sci-Plex 3′ In situ barcoding 2019 PCR 
Sci-RNA-seq3 3′ In situ barcoding 2019 PCR 
MIcrowell-seq2 3′ FACS 2020 PCR 
SCAN-seq Full length Dilution 2020 PCR 
Seq-Well S3 3′ Microwell platform 2020 PCR 
SMART-seq3 Full length FACS 2020 PCR 
FLASH-seq Full length FACS 2022 PCR 
SMART-seq3xpress Full length FACS 2022 PCR 
VASA-seq Full length Plate-based formats and droplet microfluidics 2022 PCR 
SCAN-seq2 Full length FACS 2023 PCR 
MethodologyTranscript coverageIsolationYearLibrary amplification
STRT-seq 5′ FACS 2011 PCR 
CEL-seq 3′ Micromanipulation 2012 In vitro transcription 
SMART-seq Full length FACS 2012 PCR 
Quartz-seq Full length FACS 2013 PCR 
SMART-seq2 Full length FACS 2013 PCR 
MARS-seq 3′ FACS 2014 In vitro transcription 
Cyto-seq 3′ Micromanipulation 2015 PCR 
Drop-seq 3′ Droplet 2015 PCR 
inDrop 3′ Droplet 2015 In vitro transcription 
CEL-seq2 3′ FACS 2016 In vitro transcription 
10x Genomics 3′ or 5′ Droplet 2017 PCR 
MATQ-seq Full length Micromanipulation 2017 PCR 
Sci-RNA-seq 3′ In situ barcoding 2017 PCR 
Seq-Well 3′ Microwell platform 2017 PCR 
Microwell-seq 3′ FACS 2018 PCR 
Quartz-seq2 Full length Droplet 2018 PCR 
SPLit-seq 3′ In situ barcoding 2018 PCR 
MARS-seq2 3′ FACS 2019 In vitro transcription 
sci-Plex 3′ In situ barcoding 2019 PCR 
Sci-RNA-seq3 3′ In situ barcoding 2019 PCR 
MIcrowell-seq2 3′ FACS 2020 PCR 
SCAN-seq Full length Dilution 2020 PCR 
Seq-Well S3 3′ Microwell platform 2020 PCR 
SMART-seq3 Full length FACS 2020 PCR 
FLASH-seq Full length FACS 2022 PCR 
SMART-seq3xpress Full length FACS 2022 PCR 
VASA-seq Full length Plate-based formats and droplet microfluidics 2022 PCR 
SCAN-seq2 Full length FACS 2023 PCR 

Data Analysis

The analysis of scRNA-seq data is broadly segmented into two principal phases: preprocessing and downstream analysis [32].

Preprocessing

Quality control invariably marks the commencement of scRNA-seq data analysis [33, 34]. This critical step involves the removal of data from inferior-quality cells to minimize their adverse effects on the results of downstream analysis. A common practice involves evaluating the proportion of mitochondrial reads; a high prevalence of mitochondrial transcripts indicates cellular distress, leading to the exclusion of cells exceeding a specific mitochondrial transcript threshold [34, 35]. Similarly, cells with excessive ribosomal reads are also discarded despite ribosomal RNA depletion efforts, aligning with scRNA-seq’s focus on functional mRNA [36, 37]. “scRNA Batch QC” method aids in cross-dataset quality assessments to identify biases and outliers. Batch effects are addressed using tools like Seurat3, Liger, and Harmony, with Harmony noted for computational efficiency [38].

Normalization is crucial for comparing gene expression within or among samples [39]. While traditional RNA-seq normalization methods are common, SCONE and regularized negative binomial regression are specifically designed for scRNA-seq, addressing technical variances and preserving biological heterogeneity [40, 41]. A comparative analysis of seven distinct normalization methods for scRNA-seq data revealed that each method exhibits specific strengths for normalizing different data types [39].

Downstream Analysis

Following preprocessing, scRNA-seq data undergo further scrutiny, which outlines prevalent downstream analysis techniques [42] (shown in Fig. 2). Here, we delve into several pivotal methods essential for in-depth understanding.

Fig. 2.

Downstream data analysis of scRNA-seq methods.

Fig. 2.

Downstream data analysis of scRNA-seq methods.

Close modal

Cell Clustering

Identifying cell types within samples is paramount, aimed at unraveling tissue diversity and complexity [43]. The development of algorithms like SIMLR, SAFE, CIDR, CountClust, and others reflects ongoing efforts in this domain [44‒46]. Comprehensive evaluations of these clustering algorithms have been conducted [47, 48], alongside innovative methods such as DivBiclust [49], which utilizes biclustering, SAME for amalgamating solutions from various methods, and PARC [50], tailored for large dataset analysis. To circumvent manual cell-type labeling’s constraints, Shao et al. [51] introduced “scCATCH,” an automated, highly repeatable clustering-based labeling tool.

Cell Trajectory and Pseudotime Analysis

Reconstructing cell trajectories and pseudotime calculations is vital for simulating dynamic cellular processes, especially in understanding tumor cell state transitions [52]. Algorithms such as Monocle 2, Monocle 3, TSCAN, Slingshot, SLICE, p-Creode, and Waddington-OT have been pivotal [53‒55]. The surge in scRNA-seq data has led to “LISA,” an unsupervised method for large datasets [56]. p-Creode offers another unsupervised trajectory prediction [57], while Waddington-OT applies optimal transport techniques for ancestral and descendant cell fate determination [58]. Algorithms such as TSCAN, Monocle 2/3, and Slingshot are proficient in cell trajectory reconstruction and time estimation [59]. However, the use of pseudotime and RNA velocity analyses must be approached with an awareness of their inherent limitations. Notably, RNA velocity analysis heavily relies on the construction of a k-nearest neighbors graph, which can lead to substantial errors in estimating both the direction and speed of RNA velocities, especially when the k-nearest neighbor graph fails to accurately represent the true structure of the data. Additionally, the application of RNA velocity in validating the correctness of low-dimensional embeddings may result in circular reasoning, as the method can only confirm the embeddings to the extent that they support the velocity estimates. Overinterpretation of the expression dynamics inferred from RNA velocity could lead to misleading conclusions about the biological processes under study. Therefore, caution is advised when interpreting RNA velocity results, particularly regarding the estimation of gene expression changes' speed, which is often inaccurately assessed by RNA velocity methods [60].

Differential Expression and Gene Set Enrichment Analysis

Differentially expressed gene (DEG) analysis is a common scRNA-seq application [60], with zero expression levels indicating either “true” zero expression or “dropout” due to technical reasons [61]. “DEsingle,” developed by Miao et al. [62], accurately differentiates between these “zero expressions” [63]. DECENT, utilizing UMI, analyzes RNA molecule distribution pre-dropout. Addressing dropout and multimodal data distribution challenges, ZIAQ was the first to consider dropout rates and scRNA-seq data distribution for DEG identification [64, 65]. Data imputation is a method to reduce dropout noise in scRNA-seq, categorized into model-based, smoothing-based, deep learning-based, and low-rank matrix-based approaches. Xu et al. [66] found that imputation generally improves clustering and data visualization, but its effectiveness varies with dataset size. While imputation enhances the accuracy of analyses by reducing false positives and negatives, it can introduce biases if overused. Therefore, careful selection and application of imputation methods are essential to avoid misleading biological interpretations [67]. Gene set enrichment analysis methods such as PAGE, DAVID, and CAMERA, though primarily for bulk sequencing, are crucial for interpreting DEGs in biological processes [68]. Ma et al. [69]’s IDEA integrates differential expression analysis with gene set enrichment analysis, significantly enhancing outcomes.

Transcription Factor Analysis

Transcription factor (TF) analysis is predominantly characterized by their potential gene regulatory networks. The expression of target genes and other specific genes is driven by TFs, thus establishing profiles of gene expression [70]. Single-cell regulatory network inference and clustering (SCENIC) is a methodology to reconstruct gene regulatory networks and identify cell states based on co-expression and motif analysis in scRNA-seq data [71]. This method includes evaluating the expression of TFs and their target genes in individual cells and identifying significant cell clusters. Analyzing disparities in TF activity using scRNA-seq enhances our comprehension of cell heterogeneity, providing a robust means for exploring the important regulatory mechanisms of specific TFs [72].

Cell Communication

CellPhoneDB contains a database of ligands, receptors, and their interactions [73]. This database enables comprehensive analysis of molecules participating in intercellular communication, elucidating communication networks among various cell types. The database also considers the subunit structures of ligands and receptors, representing heterodimers accurately, which are crucial for precise analysis of cell communication mediated by multimeric complexes.

CellChat is able to identify cell-to-cell communication signals accurately, uncover novel communication networks between cells, and map within diverse tissues through systematic analysis [74]. CellChat computes the probability of cell-to-cell communication by combining known receptors, ligands, and their associated factors with expression profiles. Due to the capacity of this method to calculate the strength or quantity of ligand-receptor relationship and summarize the integrated cell-to-cell communication network, its predictive capabilities extend beyond classifying cell populations and establishing their lineages.

In a study, 16 cell-cell communication inference resources and 7 methods were compared, finding that scRNA-seq predominantly captures local cell signals, neglecting processes such as protein translation and only reflecting protein levels. Furthermore, these methods are constrained to single species and do not encompass interspecies communication [75].

Advantages and Disadvantages

scRNA-seq stands out as a powerful tool for delving into the complexities of individual cells, offering unparalleled high-throughput capabilities with minimal sample requirements [76]. This technique is adept at facilitating multi-omic investigations, covering the spectrum from genomics and transcriptomics to epigenomics, proteomics, and metabolomics [77]. It shines in its ability to uncover new cell types, delineate cell lineage trajectories, and chart pathways of cell development. When compared to conventional sequencing methods, scRNA-seq is superior in discovering new genes, pinpointing rare mutations, and accurately quantifying transcript levels. A distinctive benefit of this method is its capability to circumvent biases typically introduced during PCR amplification, by quantifying nucleic acids prior to amplification [78].

However, the application of scRNA-seq is not without its hurdles. Foremost among these is the critical need for high cell viability and quality, as the success of the technique hinges on this factor. Achieving a high-quality single-cell suspension can prove to be a significant challenge [79]. Additionally, a notable limitation of scRNA-seq is the induction of “artificial transcriptional stress responses” due to stress from tissue dissociation [80], necessitating careful optimization of the dissociation process to ensure accurate results [81]. It is also worth noting that scRNA-seq cannot provide information about cell morphology or the spatial distribution of cells within tissues. This technology only analyzes the gene expression patterns of cells at the molecular level, without addressing their actual structural morphology or their specific locations within the tissue. Due to this limitation, relying solely on gene expression signatures to identify cell types without considering histomorphological features can lead to incorrect assumptions about the cells in the sample. For example, certain gene expressions may be present in different cell types, and without considering their spatial relationships and morphological characteristics within the tissue, they might be erroneously classified as the same cell type. Therefore, to more accurately understand and interpret scRNA-seq data, it is recommended to use additional techniques, such as spatial transcriptomics or flow cytometry, to complement the limitations of scRNA-seq. This multimodal approach can provide more comprehensive information about cells and tissues, aiding in the correct identification and interpretation of cell types and their functional states [82]. Furthermore, scRNA-seq is subject to technical biases including gene dropout, library preparation biases, capture efficiency variations, batch effects, amplification biases, and issues related to sequencing depth and coverage. These biases can distort gene expression data, but can be mitigated through strategies such as the use of UMIs, advanced normalization techniques, batch effect correction methods, and continuous refinement of protocols. Acknowledging and addressing these biases are essential for improving the accuracy and reliability of scRNA-seq data, thereby enabling more robust biological insights [83]. Lastly, while scRNA-seq offers powerful insights into cellular heterogeneity, cell-type identification can be challenging, especially when distinguishing between different cell states within the same cell type. It is important to note that cell-type identification is not always robust due to inherent technical biases and variability in gene expression. Initial quality control and filtering steps, while necessary, can introduce biases that affect the accuracy of cell type identification. Therefore, to ensure the robustness of scRNA-seq findings, it is essential to validate these results using complementary techniques such as flow cytometry, immunohistochemistry, or spatial transcriptomics. These orthogonal methods can provide additional layers of verification, enhancing the reliability of cell type identification and offering a more comprehensive understanding of the biological context [84]. Despite these challenges, the growing standardization of single-cell analyses and the formation of dedicated analysis cores within research institutions are progressively alleviating these obstacles.

The Role and Necessity of Single-Cell RNA Sequencing in Pituitary Research

The pituitary gland, a central endocrine organ, plays a crucial role in maintaining physiological balance through a complex cellular composition. It coordinates various bodily functions such as growth, metabolism, and reproduction by different types of endocrine cells, including thyrotrophs, lactotrophs, and corticotrophs [4]. Therefore, accurately analyzing the types, developmental trajectories, and interactions of pituitary cells is essential for understanding both normal functions and pathological conditions of the gland.

Traditional gene expression analysis techniques, such as microarrays and bulk RNA sequencing, have been standard methods for studying cellular responses and functions over the past decades [85]. However, these methods provide only average signals from mixed cell populations, which presents several significant limitations [86]: First, traditional sequencing methods struggle to resolve specific gene expression differences among various cell types within the pituitary. For instance, distinguishing between diseased and normal cells in PitNETs is challenging, limiting comprehensive understanding of disease mechanisms. Second, pituitary cells exhibit significant dynamic changes in different physiological or pathological states. Traditional methods lack the resolution to detect these subtle changes, hindering the understanding of state transitions during disease progression. Lastly, pituitary research is often constrained by limited sample sizes, especially in clinical samples. Traditional techniques require larger sample volumes, posing substantial obstacles in studying rare cell types or small sample quantities.

The introduction of scRNA-seq has revolutionized pituitary research by offering a powerful tool for precise gene expression measurement at the single-cell level [5]. This technology reveals cellular heterogeneity, identifies new cell subtypes, traces cell fate decisions, and analyzes cell-to-cell interactions, providing valuable insights for developing new diagnostic and therapeutic strategies. Specific advantages include the following: Firstl, scRNA-seq enables high-resolution cell classification and phenotyping. Tools like Seurat and Scanpy facilitate clustering analysis, allowing researchers to identify different cell types and subtypes within the pituitary, unveiling their specific roles in gland function. For example, scRNA-seq can discover new cell types or functional subgroups that play crucial roles under specific physiological or pathological conditions. Second, scRNA-seq can construct developmental trajectories using tools like Monocle, mapping continuous state transitions, and differentiation pathways of cells [54]. Understanding how progenitor cells develop into mature hormone-secreting cells and how these pathways are disrupted in pathological conditions can provide insights into disease mechanisms. Additionally, scRNA-seq data allow exploration of gene regulatory networks within pituitary cells. Tools like SCENIC can identify TF networks regulating gene expression, revealing which genes and pathways are activated or repressed under specific physiological or pathological states [71]. This is crucial for understanding molecular mechanisms of pituitary diseases and identifying new therapeutic targets. While scRNA-seq itself does not provide spatial information, it can be combined with other techniques to reveal cell-to-cell interactions. Understanding how cells interact within the pituitary tissue and their precise locations helps elucidate the cooperative mechanisms in maintaining tissue function and responding to environmental changes.

Through these advanced analytical methods, scRNA-seq not only provides detailed molecular-level maps of pituitary cells but also reveals how cells respond to diseases or other physiological states. This offers new strategies and targets for diagnosing and treating pituitary-related diseases. The application of these technologies significantly advances the depth and breadth of pituitary research, demonstrating the powerful potential of single-cell techniques in biomedical research.

In summary, the importance of scRNA-seq in pituitary research lies not only in its precision and sensitivity but also in its ability to provide deep molecular insights into pituitary biology that traditional sequencing techniques cannot match. This technology holds promise for advancing early diagnosis, treatment, and prognosis evaluation of pituitary diseases, ultimately offering more precise medical solutions for patients.

Multi-Omics Technologies and Their Impact on Pituitary Research

While scRNA-seq has significantly advanced our understanding of the cellular heterogeneity within the pituitary gland, it is not without limitations. Complementary single-cell multi-omics technologies offer additional layers of information that can address these shortcomings and further propel pituitary research.

Single-Cell Genomics and Transcriptomics

Single-cell genomics-plus-transcriptomics methods, such as G&T-seq and SIDR-seq, allow for the simultaneous analysis of both the genome and transcriptome of individual cells [87, 88]. This dual analysis enables researchers to investigate the impact of genetic variations, such as mutations or copy number alterations, on gene expression within the same cell. Understanding how these genetic changes influence pituitary cell function can provide deeper insights into the pathogenesis of pituitary disorders.

Single-Cell Epigenomics and Transcriptomics

Advances in single-cell epigenomics-plus-transcriptomics methods, such as scM&T-seq and scNMT-seq, enable the parallel profiling of the epigenome (e.g., DNA methylation, chromatin accessibility) and the transcriptome [89, 90]. Additionally, single-cell ATAC-seq provides insights into chromatin accessibility at the single-cell level, revealing how DNA is packaged and regulated in the nucleus [91]. This technique is particularly valuable for identifying regulatory elements such as enhancers and promoters that are active in specific cell types. When combined with scRNA-seq, single-cell ATAC-seq allows researchers to correlate chromatin accessibility with gene expression, providing a comprehensive view of the regulatory landscape in pituitary cells [92]. These methods are crucial for understanding how epigenetic modifications regulate gene expression in pituitary cells. For instance, they can reveal how changes in DNA methylation or histone modifications influence cell differentiation and function, thereby providing insights into the regulatory mechanisms underlying pituitary development and disease.

Spatial Multi-Omics

Multi-omics approaches, such as spatial transcriptomics combined with protein profiling, provide spatially resolved data that highlight the physical and functional interactions between pituitary cells [93]. These methods enable researchers to map the precise locations and interactions of different cell types within the tissue, uncovering how these interactions contribute to pituitary function and pathology. Understanding cell-cell communication and the tissue microenvironment is particularly important for elucidating the mechanisms of disease progression and identifying potential therapeutic targets.

In summary, by integrating multiple layers of omics data, single-cell multi-omics approaches can mitigate some of the limitations inherent in scRNA-seq. For example, while scRNA-seq excels at capturing gene expression profiles, it does not provide direct information about the genomic context or the epigenetic landscape that influences these profiles. Multi-omics approaches bridge this gap, offering a more comprehensive view of the molecular mechanisms at play. Additionally, these methods can improve the accuracy of cell-type identification and functional annotation by incorporating genomic and epigenomic data, thereby reducing the biases introduced by single-modality analyses.

The Application of Multi-Omics Technologies in Pituitary Research

The application of multi-omics technologies in pituitary research holds great promise for advancing our understanding of this complex endocrine organ. These advanced methodologies enable researchers to provide high-resolution molecular profiles, which can identify previously unrecognized cell types and functional states within the pituitary [94]. This contributes to a more detailed cellular atlas of the gland, significantly enhancing our knowledge of its intricate cellular diversity [4].

Integrating transcriptomic and epigenomic data through these multi-omics approaches allows for the reconstruction of gene regulatory networks [92]. This integration highlights key TFs and regulatory elements that drive pituitary cell function and differentiation. Such detailed regulatory maps are crucial for understanding the molecular underpinnings of pituitary biology and its various functional states. Moreover, multi-omics data can elucidate the molecular alterations associated with pituitary diseases, such as PitNETs. By linking genetic and epigenetic changes to aberrant gene expression patterns and cellular behaviors, researchers can gain a comprehensive understanding of disease mechanisms [95]. This knowledge is vital for identifying potential biomarkers and therapeutic targets. Insights gained from multi-omics studies also inform the development of targeted therapies by identifying specific molecular pathways and cell types involved in disease processes. This precision in identifying therapeutic targets can lead to more effective and personalized treatment strategies for pituitary disorders.

In summary, the integration of single-cell multi-omics technologies provides a comprehensive toolkit for dissecting the complex biology of the pituitary gland. These approaches complement scRNA-seq by offering additional layers of molecular information, thereby addressing its limitations and opening new avenues for research into pituitary function and disease. This holistic view of pituitary biology and pathology is essential for advancing both basic and translational research in endocrinology.

Physiological Conditions

In the past decades, research has predominantly harnessed tissue- or organ-level aggregate data to elucidate cell population functionality and traits. The advent of scRNA-seq technology, however, has ushered in the capability to probe into previously indistinguishable cell populations, which were masked by analogous histological characteristics, genetic markers, and proximal tissue locations. This method affords a granular view, uncovering cellular heterogeneity and novel cellular identities of pituitary (shown in Fig. 3; Table 2).

Fig. 3.

Application of scRNA-seq methods in physiological conditions of pituitary.

Fig. 3.

Application of scRNA-seq methods in physiological conditions of pituitary.

Close modal
Table 2.

Summary illustrating single studies in pituitary research

Main discoverySpeciesMethodologyNumber of cells sequenced
Physiological conditions 
 Identifying novel markers for male pituitary stem and hormone-producing cells [96Mus musculus 10x Genomics 13,663 
 Unveiling sexual dimorphism and demand-induced cellular plasticity [97Mus musculus Drop-seq and 10x Genomics 21,185 
 Offering a detailed molecular and cellular overview of neurohypophyseal cell types [98Mus musculus 10x Genomics 528 
 Mapping a new developmental trajectory for thyrotropes and potential regulators [99Mus musculus 10x Genomics 4,648 
 Identifying a gene-specific, all-or-none transcript induction pattern triggered by rising GnRH concentrations [100Mus musculus 10x Genomics >1,000 (C1) and 6,332 (10x) 
 Marking the first comprehensive epigenetic and single-cell transcriptomic analysis of the LβT2 gonadotropic cell [101Mus musculus 10x Genomics 3,881 
 Offering new insights into regulatory networks and gene control relevant to pituitary physiology and disease [102Mus musculus 10x Genomics >69,000 
 Introducing CREMA that identify functional regulatory elements missed by traditional methods [103Mus musculus 
 Demonstrating that pioneer factors are limited to uniquely recognize heterochromatin and enable nonpioneer factor binding [104Mus musculus 10x Genomics 9,296 
 Showing IL-6 activates PSC s postdamage, with this effect decreasing in aging due to higher IL-6 levels [105Mus musculus 10x Genomics 26,115 
 Decoding the neonatal pituitary’s stem cell compartment, revealing an activated state in the maturing gland [106Mus musculus 10x Genomics 21,419 
 Offering a valuable system for examining how evolving states affect SC mobilization mechanisms [107Mus musculus 10x Genomics 1,993 
 Characterizing human stem cell lineages, uncovering varied mechanisms that regulate pivotal PSC genes and cell identity [108Homo sapiens and Mus musculus 10x Genomics 76,016 
 Defining cell substates and subtypes, showing TF dynamics in cell fate decisions [109Homo sapiens Illumina Hiseq 4,113 
 Identifying PI4KA as crucial for calcium secretion in pituitary lactotrophs [110Rattus norvegicus 10x Genomics 15,876 
 Challenging the belief that adenohypophyseal cells stem only from nonneural ectoderm, showing that interactions between neuro- and adenohypophyseal cells influence pituitary cell differentiation [111Danio rerio Illumina NextSeq 500 1,281 
Pathological conditions 
 Showing that male pituitary cells exhibit greater resistance to obesity-related oxidative stress compared to females [112Mus musculus 10x Genomics 10,000 
 Investigating how diet influences pituitary gland activity and gene expression, leading to hormonal imbalances in obesity [113Mus musculus 10x Genomics 23,887 
 Unveiling widespread interactions between the pituitary gland and immune system [114Mus musculus STRT-seq2 5,506 
 Suggesting targeting SCs could enhance antitumor strategies in specific contexts [115Mus musculus 10x Genomics 7,839 
 Identifying tumor immune microenvironment traits and discovering a novel, aggressive epithelial cell subpopulation [116Homo sapiens Illumina HiSeq 2,679 
 Identifying several features of the tumor immune microenvironments, and found a novel epithelial cell subpopulation with aggressive signatures across all the studied cases [117Homo sapiens 10x Genomics 16,533 
 Laying groundwork for translational studies on subgroup/lineage-specific therapies [118Homo sapiens 10x Genomics 2,311 
 Proteasomal inhibition rescues noxa and induces apoptosis in CD [119Homo sapiens 10x Genomics 27,594 
 Indicating a shared transcriptional reprogramming that disrupts POMC processing and triggers tumor invasion [120Homo sapiens 10x Genomics 56,458 
 Uncovering PA’s possible origin and differentiation, proposing and validating a new classification to predict recurrence [121Homo sapiens 10x Genomics 64,937 
 DA resistance in prolactinomas links to increased FA signaling, which is inhibited by Genistein to counteract tumor growth [122Homo sapiens 10x Genomics 58,612 
 Highlighting IFN-γ’s role in remodeling tumor-associated fibroblasts (TAFs) to combat tumor growth [123Homo sapiens 10x Genomics 30,374 
Main discoverySpeciesMethodologyNumber of cells sequenced
Physiological conditions 
 Identifying novel markers for male pituitary stem and hormone-producing cells [96Mus musculus 10x Genomics 13,663 
 Unveiling sexual dimorphism and demand-induced cellular plasticity [97Mus musculus Drop-seq and 10x Genomics 21,185 
 Offering a detailed molecular and cellular overview of neurohypophyseal cell types [98Mus musculus 10x Genomics 528 
 Mapping a new developmental trajectory for thyrotropes and potential regulators [99Mus musculus 10x Genomics 4,648 
 Identifying a gene-specific, all-or-none transcript induction pattern triggered by rising GnRH concentrations [100Mus musculus 10x Genomics >1,000 (C1) and 6,332 (10x) 
 Marking the first comprehensive epigenetic and single-cell transcriptomic analysis of the LβT2 gonadotropic cell [101Mus musculus 10x Genomics 3,881 
 Offering new insights into regulatory networks and gene control relevant to pituitary physiology and disease [102Mus musculus 10x Genomics >69,000 
 Introducing CREMA that identify functional regulatory elements missed by traditional methods [103Mus musculus 
 Demonstrating that pioneer factors are limited to uniquely recognize heterochromatin and enable nonpioneer factor binding [104Mus musculus 10x Genomics 9,296 
 Showing IL-6 activates PSC s postdamage, with this effect decreasing in aging due to higher IL-6 levels [105Mus musculus 10x Genomics 26,115 
 Decoding the neonatal pituitary’s stem cell compartment, revealing an activated state in the maturing gland [106Mus musculus 10x Genomics 21,419 
 Offering a valuable system for examining how evolving states affect SC mobilization mechanisms [107Mus musculus 10x Genomics 1,993 
 Characterizing human stem cell lineages, uncovering varied mechanisms that regulate pivotal PSC genes and cell identity [108Homo sapiens and Mus musculus 10x Genomics 76,016 
 Defining cell substates and subtypes, showing TF dynamics in cell fate decisions [109Homo sapiens Illumina Hiseq 4,113 
 Identifying PI4KA as crucial for calcium secretion in pituitary lactotrophs [110Rattus norvegicus 10x Genomics 15,876 
 Challenging the belief that adenohypophyseal cells stem only from nonneural ectoderm, showing that interactions between neuro- and adenohypophyseal cells influence pituitary cell differentiation [111Danio rerio Illumina NextSeq 500 1,281 
Pathological conditions 
 Showing that male pituitary cells exhibit greater resistance to obesity-related oxidative stress compared to females [112Mus musculus 10x Genomics 10,000 
 Investigating how diet influences pituitary gland activity and gene expression, leading to hormonal imbalances in obesity [113Mus musculus 10x Genomics 23,887 
 Unveiling widespread interactions between the pituitary gland and immune system [114Mus musculus STRT-seq2 5,506 
 Suggesting targeting SCs could enhance antitumor strategies in specific contexts [115Mus musculus 10x Genomics 7,839 
 Identifying tumor immune microenvironment traits and discovering a novel, aggressive epithelial cell subpopulation [116Homo sapiens Illumina HiSeq 2,679 
 Identifying several features of the tumor immune microenvironments, and found a novel epithelial cell subpopulation with aggressive signatures across all the studied cases [117Homo sapiens 10x Genomics 16,533 
 Laying groundwork for translational studies on subgroup/lineage-specific therapies [118Homo sapiens 10x Genomics 2,311 
 Proteasomal inhibition rescues noxa and induces apoptosis in CD [119Homo sapiens 10x Genomics 27,594 
 Indicating a shared transcriptional reprogramming that disrupts POMC processing and triggers tumor invasion [120Homo sapiens 10x Genomics 56,458 
 Uncovering PA’s possible origin and differentiation, proposing and validating a new classification to predict recurrence [121Homo sapiens 10x Genomics 64,937 
 DA resistance in prolactinomas links to increased FA signaling, which is inhibited by Genistein to counteract tumor growth [122Homo sapiens 10x Genomics 58,612 
 Highlighting IFN-γ’s role in remodeling tumor-associated fibroblasts (TAFs) to combat tumor growth [123Homo sapiens 10x Genomics 30,374 

In a pioneering study, Cheung et al. [96] implemented scRNA-seq on the pituitary glands of 7-week-old male mice, unveiling not only endocrine and stem cells in a digital format but also auxiliary cells including endothelial, connective tissue, and hematopoietic cells. Their research highlights scRNA-seq’s capacity to explore the pituitary gland’s cellular dynamics and functional evolution. Nonetheless, the exclusive focus on male specimens constituted a notable study limitation. Addressing this, Ho et al. [97] extended the application of scRNA-seq to delineate cell-type diversity and functional state heterogeneity within both male and female mouse pituitaries. Their analysis discerned sex-specific cellular architectures in physiological pituitary regulation and pinpointed cell clusters enriched with various hormones alongside nonhormonal interstitial and support cells. Remarkably, they discovered a Pou1f1-expressing cell group characterized by a distinctive hormonal gene expression profile. This discovery reveals the pituitary’s cellular complexity and functional plasticity, highlighting how multifunctional cell groups can challenge traditional views on pituitary cell lineages and suggest dynamic adaptations in physiological and pathological states. These insights underscore the adult pituitary’s complex cellular composition and adaptive capacity.

In addition to the anterior lobe, the posterior and intermediate lobes of the pituitary have garnered significant interest among researchers. A study by Chen et al. [98] employed scRNA-seq on the pituitaries of adult male mice, uncovering the cellular diversity encompassing seven unique cell types within the posterior and intermediate lobes. Notably, this investigation identified new markers including Col25a1, Scn7a, and Srebf1 for pituicytes that surpass the specificity of those previously known, thus offering indispensable resources for the exploration of distinct cellular functions and physiological processes.

Apart from profiling major cell lineages within the pituitary, we can also utilize scRNA-seq for an in-depth investigation of specific cell populations. Cheung et al. [99] applied scRNA-seq combined with lineage tracing to delineate and corroborate a previously unidentified endocrine subset in the juvenile mouse pituitary, elucidating a novel developmental pathway leading to thyrotropes. This discovery furnishes a foundation for further exploration into the biology of thyrotropes. The interest of the researchers has also extended to lactotropes. Through scRNA-seq of the rat pituitary, Kučka et al. [110] underscored the pivotal involvement of PI4KA in the coupling of calcium secretion in pituitary lactotropes, subsequent to voltage-gated and PI(4,5)P2-mediated calcium signaling. Furthermore, an insightful study by Chen et al. [111] challenged established views regarding the embryonic origins of neurohypophyseal and adenohypophyseal cells by demonstrating that neural plate progenitors contribute to both cell types, as evidenced through scRNA-seq analysis of the zebrafish pituitary.

Collectively, the aforementioned studies underscore the profound impact of single-cell sequencing technologies on the identification and characterization of cellular subsets, thereby enriching our understanding of cellular composition of the pituitary. While the current depiction of the pituitary gland’s cellular composition is comprehensive, forthcoming investigations should aim at uncovering finer details through more in-depth mechanistic studies. Potential directions include the discovery of novel cellular subsets influencing hormone release or the recognition of previously unknown roles of established cell types.

The evolution of scRNA-seq technology has revolutionized our capacity to attribute gene expression profiles to distinct cell populations, especially within intricately structured tissues such as the pituitary gland. Professor Ruf-Zamojski and her colleagues have embarked on a series of studies. In their 2018 investigation, they leveraged scRNA-seq on the LβT2 gonadotroph cell line to uncover a cell-specific, all-or-none transcript induction pattern in reaction to GnRH gradient [101]. This finding introduced the concept that immediate-early gene activation at the single-cell level might operate via binary switch-like gene bits. Subsequent research by the team refined this understanding, illustrating that LβT2 cell responses to GnRH are not linearly amplifying but rather follow a bimodal pattern [100]. In 2021, they unveiled transcriptional and chromatin accessibility profiles unique to each primary pituitary cell type, including sex-specific regulatory elements, by analyzing over 70,000 nuclei from adult mice pituitaries, underlining the pivotal influence of chromatin accessibility on cell identity transcriptional blueprints [102]. Advancing their research to human subjects, they identified critical regulatory domains and TFs correlating with gene expression in pituitary stem cells (PSCs) through a similar methodology [108]. Their latest innovation, CREMA, addresses the shortcomings of traditional approaches by enabling the identification of functional regulatory elements beyond conventional chromatin “peaks” [103]. Complementing their work, Mayran et al. [104] elucidated the necessity of both Pax7 and Tpit for the activation and chromatin accessibility of melanotrope-specific enhancers, underscoring the synergistic action of pioneer and non-pioneer factors in lineage-specific chromatin remodeling.

In summary, scRNA-seq presents an unparalleled lens through which to examine gene expression regulation within specific cellular subsets, offering profound insights into the biological intricacies of the pituitary gland. Future studies are likely to harness this technology for more in-depth research to uncover the dynamic changes and mechanisms of the pituitary gland under various physiological and pathological conditions. For example, such studies could track changes in gene expression patterns of the pituitary gland during different developmental stages, disease progression, or treatment responses, thereby providing new insights and targets for clinical intervention strategies.

Several studies have characterized PSC using scRNA-seq and these studies illustrate the potential role of PSC in the regenerative medicine. For instance, Laporte et al. [105] utilized scRNA-seq on the neonatal pituitary gland of mouse, revealing proliferative stem cell populations that displayed a hybrid epithelial/mesenchymal phenotype, characteristic of development-involved tissue stem cells. They also recapitulated the stem cells’ phenotype in organoid culturing. Interestingly, they found that the neonatal gland efficiently regenerated after local damage without the presence of upregulated IL-6, while Vennekens et al. had the opponent finding on the adult pituitary gland. They applied scRNA-seq on the adult pituitary, finding the acute PSC activation process with the presence of upregulated IL-6 upon targeted endocrine cell-ablation damage. The difference may be caused by the different status of pituitary gland, as the neonatal pituitary gland is in the already high stem cell activation status and the adult pituitary gland is in the dormant status. Notably, they found that administering IL-6 to young mice promptly triggered PSC proliferation upon local damage in vivo and in vitro using stem cell-derived organoids. Their study identified IL-6 as a PSC activator upon local damage. Another exciting finding is that they found the aging pituitary’s stem cells retain intrinsic activatability, resurfacing once released from their impeding tissue milieu. Taken together, above findings indicate that PSC plays a crucial role in regenerative medicine. However, it is well known that obtaining mature cell types from PSCs in vitro remains challenging, and further research is needed to overcome these difficulties and harness their full potential for therapeutic purposes.

One of the paramount benefits of scRNA-seq is its capability for pseudotime analysis. In a landmark study, Zhang et al. [109] conducted scRNA-seq on 4,113 cells from human fetal pituitaries, delineating varied developmental pathways and distinct transitional states across five hormone-secreting cell lineages. A significant aspect of their research was the delineation of PSC heterogeneity, identifying a unique hybrid epithelial/mesenchymal phase alongside an early-to-late state transition, which is in line with Laporte’s finding. Historically, delineating the roles of stem cell differentiation and the maintenance of their progeny in cell turnover has been challenging. Utilizing scRNA-seq coupled with lineage tracing, Rizzoti et al. [107] mapped the diversity and mobilization of PSCs, uncovering that PSC differentiation is more prevalent than previously believed. Under adaptive conditions, this differentiation is not only heightened but also more varied than lineage tracing alone has shown. Their thorough analysis of PSC progeny underlines the critical support role of the microenvironment in sustaining selected emerging cells, thereby influencing PSC output. A notable limitation of scRNA-seq is the requirement for fresh samples, which considerably limits its applicability. In response, advancements in single-nucleus sequencing technologies have emerged. Zhang et al. [108] utilized single-nucleus RNA-seq and ATAC-seq to map cell-type-specific gene expression and chromatin accessibility across all major pituitary cell lineages in postmortem samples from pediatric to aged individuals. Their work highlighted the presence of uncommitted PSCs, transitioning progenitor cells, and gender-based differences. Through linear modeling of multiome data, they pinpointed regulatory elements and TFs linked with PSC gene expression, also uncovering deterministic factors that account for variability in PSC marker expression. These insights provide a deeper understanding of the mechanisms regulating essential PSC genes and cell identity.

Taken together, scRNA-seq offers unprecedented depth in exploring the complex cellular ecosystems within the pituitary gland, yielding vital breakthroughs in understanding PSC biology. Such research paves the way for novel stem cell-based therapeutic strategies targeting the replenishment of lost or impaired endocrine cells in the pituitary gland, thereby offering potential treatments for conditions such as pituitary hormone deficiencies and tumors.

Pathological Conditions

Pituitary Neuroendocrine Tumor

Pituitary neuroendocrine tumors (PitNETs), which constitute common intracranial neoplasms accounting for an incidence rate of about 10%, are classified into various distinct categories, such as those aligning with PIT1, TPIT, and SF1 lineages, in addition to plurihormonal and null cell tumors [124]. The management of PitNETs typically involves a combination of surgical resection, pharmacological interventions, and radiotherapy. Despite these strategies, the patient faces significant challenges including resistance to pharmacological treatments, a scarcity of targeted therapeutic options, risks of tumor recurrence postsurgery, and a lack of comprehensive insights into the molecular drivers of these tumors [125]. Numerous researches have been committed to unraveling the complex molecular basis of PitNETs, yet the multifaceted gene interactions and the distinctive attributes of anterior pituitary cells within the tumor’s microenvironment have hindered a full elucidation of the disease’s pathogenesis [126‒128]. With the advent of recent technological advancements, the application of single-cell sequencing stands out as a transformative approach for delving into the molecular intricacies of PitNETs (shown in Fig. 4) [129, 130].

Fig. 4.

Application of scRNA-seq methods in PitNET.

Fig. 4.

Application of scRNA-seq methods in PitNET.

Close modal

In their groundbreaking study, Cui et al. [116] utilized scRNA-seq on 23 samples of PitNETs, revealing for the first time the transcriptomic profiles of both tumor and adjacent normal cells. This analysis laid the groundwork for the delineation of PitNET characteristics and heterogeneity, crucial for the discovery of novel biomarkers and therapeutic avenues. Similarly, Yan et al. [117] undertook scRNA-seq on 4 healthy and 24 PitNET specimens, elucidating the fundamental cell types within the healthy pituitary gland, the intrinsic heterogeneity of PitNETs, and characteristics of the tumor immune microenvironment. Intriguingly, they also identified an aggressive epithelial cell subset present in all cases examined. Furthermore, Batchu et al. [118]’s analysis of scRNA-seq data from 21 PitNET samples, sourced from an accessible public database, mapped out the metabolic profiles of different pituitary cell lineages. Their insights are pivotal for the formulation of targeted therapies specific to various PitNET lineages or subgroups.

In addition to showing the landscape of PitNETs, scRNA-seq facilitates an in-depth analysis of their pathogenesis. Asuzu et al. [119] utilized scRNA-seq on 27,594 cells from PitNETs and adjacent normal glands, revealing a previously unrecognized population of proliferating cells may drive tumorigenesis. A critical discovery was the identification of a pathway for apoptosis evasion through proteasome-mediated degradation of noxa, highlighting the therapeutic promise of proteasome inhibitors. In a focused study, Zhang et al. [120] analyzed scRNA-seq data from three silent ACTH-secreting tumors and five functioning ACTH-secreting tumors, revealing that silent ACTH-secreting tumors exhibit signs of epithelial-to-mesenchymal transition, characterized by enhanced mesenchymal gene expression and diminished transcripts involved in hormone synthesis and secretion. This study posited that a unified transcriptional reprogramming might simultaneously hinder POMC processing and foster tumor invasiveness. Additionally, another study by Zhang et al. [121] using scRNA-seq on 3 healthy and 21 PitNET samples, distinguished specific genes indicative of major subtypes within both well-differentiated and poorly differentiated PitNETs across each lineage, also confirming the prognostic utility of differentiation markers in an independent dataset.

The treatment of PitNETs is becoming increasingly complex, with more reports of PitNETs lacking effective treatments, especially refractory PitNETs. This underscores the urgency for innovative therapeutic approaches. Moncho-Amor et al. [115] utilized scRNA-seq on P27-null mouse, identifying the pivotal role of an SOX2-driven MAPK pathway in stem cells for tumorigenesis. This research suggests that targeting PSCs could be a viable antitumor strategy. In a related vein, Cheng et al. [122] explored the basis of dopamine agonist (DA) resistance by analyzing scRNA-seq data from both DA-resistant and DA-sensitive PitNETs, pinpointing the association of DA resistance with the focal adhesion signaling pathway and proposing Genistein as a potential therapeutic agent due to its ability to inhibit focal adhesion pathway expression. Furthermore, Lyu et al. [123] leveraged scRNA-seq to examine 4 PIT1-positive PitNETs, shedding light on the cellular and functional diversity of tumor-associated fibroblasts and immune cells. Their findings underscore the antitumor potential of IFN-γ through its role in modulating the tumor-promoting activities of tumor-associated fibroblasts.

In summary, scRNA-seq has emerged as a cornerstone technology in translational medical research, offering unparalleled insights into the complex landscape of cellular heterogeneity and dynamics within PitNETs. This cutting-edge approach allows for the detailed profiling of individual cells, uncovering rare cell types, defining various cell states, and mapping intricate cellular interactions with precision previously beyond reach. For instance, several studies have challenged the concept of prevailing mono-lineage PitNETs and reported frequent co-expression of TFs, particularly of PIT1 and SF1 [131‒133]. scRNA-seq provides a powerful tool to investigate these findings in greater detail. By analyzing cell-specific gene expression, scRNA-seq can identify cells that co-express PIT1 and SF1 within the same cell population. This enables researchers to confirm the presence of these co-expressing cells and study their prevalence and characteristics in pituitary tumors. Additionally, scRNA-seq data can construct developmental trajectories to track gene expression changes from precursor cells to mature tumor cells, providing insights into the origins of PIT1/SF1-co-expressing cells and their roles in tumor progression. The rich molecular insights gained from scRNA-seq provide critical resources for deciphering the pathogenesis of PitNETs, propelling forward the field of precision medicine. By facilitating the development of therapies specifically designed to target the distinct molecular characteristics of individual tumors, scRNA-seq heralds a new era in the diagnosis, treatment, and management of PitNETs, promising significant advancements in patient care and therapeutic outcomes.

Other Conditions

While the focus on PitNETs remains significant, the broader spectrum of pituitary gland disorders, such as obesity and systemic inflammation, has attracted increased scrutiny in recent research efforts. Miles et al. [112] utilized scRNA-seq on 10–15 weeks’ high-fat diet mouse, uncovering a notable resilience against obesity-induced oxidative stress in male pituitary cells compared to female ones, and pinpointing the pituitary cell populations most susceptible in females. Ruggiero-Ruff et al. [113] applied scRNA-seq on pituitary glands from male mice fed control and high-fat diet, emphasizing the diet-induced shifts in pituitary cell populations and gene expressions that contribute to the hormonal imbalances observed in obesity. Yan’s pioneering research delved into the pituitary gland’s response to systemic inflammation, utilizing scRNA-seq to demonstrate how all major pituitary hormone-producing cells engage in a cell type-specific response [114]. A key discovery was the upregulation of chemokine gene expression by hormone-producing cells, enhancing communication with immune cells and underscoring the pituitary’s complex role in modulating the body’s physiological response to inflammation.

The application of scRNA-seq in pituitary research represents a paradigm shift in our approach to understanding this crucial endocrine gland. Through detailed profiling of individual cells, scRNA-seq has illuminated the pituitary’s cellular diversity, revealing insights into the developmental trajectories, functional states, and pathological alterations of its constituent cells. Studies utilizing scRNA-seq have not only expanded our knowledge of PSC biology and the heterogeneity of PitNETs but have also highlighted the gland’s integral role in responding to systemic conditions. Despite the progress, challenges remain in translating these discoveries into clinical practice and developing targeted therapies for pituitary disorders. Future research, leveraging the depth and breadth of scRNA-seq, holds the promise of further unraveling the molecular underpinnings of pituitary function and disease, paving the way for precision medicine in endocrinology and oncology.

The authors have no conflicts of interest to declare.

This work was supported by the CAMS Innovation Fund for Medical Sciences (No. CIFMS 2021-I2M-1-003), the Beijing Municipal Natural Science Foundation (No. M22013), the Key-Area Research and Development Program of Guangdong Province (No. 2021B0101420005), the National High Level Hospital Clinical Research Funding (No. 2022-PUMCH-C-012), and the National Natural Science Foundation of China (82103302 to MC).

Shuangjian Yang conceived the review, collected and analyzed the literature, and completed the article. Congcong Deng, Changqin Pu, Xuexue Bai, and Chenxin Tian participated in the collection of literature and writing of the article. Mengqi Chang edited the language of the article and revised the manuscript. Ming Feng made the idea of the review, revised the manuscript, and took charge of the quality of the whole article.

Additional Information

Shuangjian Yang, Congcong Deng, and Changqin Pu contributed equally to this work.

1.
Bianconi
E
,
Piovesan
A
,
Facchin
F
,
Beraudi
A
,
Casadei
R
,
Frabetti
F
, et al
.
An estimation of the number of cells in the human body
.
Ann Hum Biol
.
2013
;
40
(
6
):
463
71
.
2.
Mostafa
HKK
.
Different cells of the human body: categories and morphological characters
.
J Microsc Ultrastruct
.
2022
;
10
(
2
):
40
6
.
3.
Abyzov
A
,
Vaccarino
FM
.
Cell lineage tracing and cellular diversity in humans
.
Annu Rev Genomics Hum Genet
.
2020
;
21
:
101
16
.
4.
Greenhill
C
.
Pituitary gland: understanding pituitary development
.
Nat Rev Endocrinol
.
2016
;
12
(
9
):
497
.
5.
Jovic
D
,
Liang
X
,
Zeng
H
,
Lin
L
,
Xu
F
,
Luo
Y
.
Single-cell RNA sequencing technologies and applications: a brief overview
.
Clin Transl Med
.
2022
;
12
(
3
):
e694
.
6.
Zhang
Y
,
Wang
D
,
Peng
M
,
Tang
L
,
Ouyang
J
,
Xiong
F
, et al
.
Single-cell RNA sequencing in cancer research
.
J Exp Clin Cancer Res
.
2021
;
40
(
1
):
81
.
7.
Papalexi
E
,
Satija
R
.
Single-cell RNA sequencing to explore immune cell heterogeneity
.
Nat Rev Immunol
.
2018
;
18
(
1
):
35
45
.
8.
Haque
A
,
Engel
J
,
Teichmann
SA
,
Lönnberg
T
.
A practical guide to single-cell RNA-sequencing for biomedical research and clinical applications
.
Genome Med
.
2017
;
9
(
1
):
75
.
9.
Gross
A
,
Schoendube
J
,
Zimmermann
S
,
Steeb
M
,
Zengerle
R
,
Koltay
P
.
Technologies for single-cell isolation
.
Int J Mol Sci
.
2015
;
16
(
8
):
16897
919
.
10.
Arsenio
J
.
Single-cell transcriptomics of immune cells: cell isolation and cDNA library generation for scRNA-seq
.
Methods Mol Biol
.
2020
;
2184
:
1
18
.
11.
Chen
G
,
Ning
B
,
Shi
T
.
Single-cell RNA-seq technologies and related computational data analysis
.
Front Genet
.
2019
;
10
:
317
.
12.
Hu
P
,
Zhang
W
,
Xin
H
,
Deng
G
.
Single cell isolation and analysis
.
Front Cell Dev Biol
.
2016
;
4
:
116
.
13.
Kamme
F
,
Salunga
R
,
Yu
J
,
Tran
DT
,
Zhu
J
,
Luo
L
, et al
.
Single-cell microarray analysis in hippocampus CA1: demonstration and validation of cellular heterogeneity
.
J Neurosci
.
2003
;
23
(
9
):
3607
15
.
14.
Kolodziejczyk
AA
,
Kim
JK
,
Svensson
V
,
Marioni
JC
,
Teichmann
SA
.
The technology and biology of single-cell RNA sequencing
.
Mol Cell
.
2015
;
58
(
4
):
610
20
.
15.
Kivioja
T
,
Vähärautio
A
,
Karlsson
K
,
Bonke
M
,
Enge
M
,
Linnarsson
S
, et al
.
Counting absolute numbers of molecules using unique molecular identifiers
.
Nat Methods
.
2011
;
9
(
1
):
72
4
.
16.
Olsen
TK
,
Baryawno
N
.
Introduction to single-cell RNA sequencing
.
Curr Protoc Mol Biol
.
2018
;
122
(
1
):
e57
.
17.
Picelli
S
,
Björklund
ÅK
,
Faridani
OR
,
Sagasser
S
,
Winberg
G
,
Sandberg
R
.
Smart-seq2 for sensitive full-length transcriptome profiling in single cells
.
Nat Methods
.
2013
;
10
(
11
):
1096
8
.
18.
Ramsköld
D
,
Luo
S
,
Wang
YC
,
Li
R
,
Deng
Q
,
Faridani
OR
, et al
.
Full-length mRNA-Seq from single-cell levels of RNA and individual circulating tumor cells
.
Nat Biotechnol
.
2012
;
30
(
8
):
777
82
.
19.
Hashimshony
T
,
Wagner
F
,
Sher
N
,
Yanai
I
.
CEL-Seq: single-cell RNA-Seq by multiplexed linear amplification
.
Cell Rep
.
2012
;
2
(
3
):
666
73
.
20.
Klein
AM
,
Mazutis
L
,
Akartuna
I
,
Tallapragada
N
,
Veres
A
,
Li
V
, et al
.
Droplet barcoding for single-cell transcriptomics applied to embryonic stem cells
.
Cell
.
2015
;
161
(
5
):
1187
201
.
21.
Tang
F
,
Barbacioru
C
,
Wang
Y
,
Nordman
E
,
Lee
C
,
Xu
N
, et al
.
mRNA-Seq whole-transcriptome analysis of a single cell
.
Nat Methods
.
2009
;
6
(
5
):
377
82
.
22.
Tang
F
,
Barbacioru
C
,
Nordman
E
,
Li
B
,
Xu
N
,
Bashkirov
VI
, et al
.
RNA-Seq analysis to capture the transcriptome landscape of a single cell
.
Nat Protoc
.
2010
;
5
(
3
):
516
35
.
23.
Picelli
S
,
Faridani
OR
,
Björklund
AK
,
Winberg
G
,
Sagasser
S
,
Sandberg
R
.
Full-length RNA-seq from single cells using Smart-seq2
.
Nat Protoc
.
2014
;
9
(
1
):
171
81
.
24.
Hagemann-Jensen
M
,
Ziegenhain
C
,
Chen
P
,
Ramsköld
D
,
Hendriks
GJ
,
Larsson
AJM
, et al
.
Single-cell RNA counting at allele and isoform resolution using Smart-seq3
.
Nat Biotechnol
.
2020
;
38
(
6
):
708
14
.
25.
Sasagawa
Y
,
Nikaido
I
,
Hayashi
T
,
Danno
H
,
Uno
KD
,
Imai
T
, et al
.
Quartz-Seq: a highly reproducible and sensitive single-cell RNA sequencing method, reveals non-genetic gene-expression heterogeneity
.
Genome Biol
.
2013
;
14
(
4
):
R31
.
26.
Hashimshony
T
,
Senderovich
N
,
Avital
G
,
Klochendler
A
,
de Leeuw
Y
,
Anavy
L
, et al
.
CEL-Seq2: sensitive highly-multiplexed single-cell RNA-Seq
.
Genome Biol
.
2016
;
17
:
77
.
27.
Keren-Shaul
H
,
Kenigsberg
E
,
Jaitin
DA
,
David
E
,
Paul
F
,
Tanay
A
, et al
.
MARS-seq2.0: an experimental and analytical pipeline for indexed sorting combined with single-cell RNA sequencing
.
Nat Protoc
.
2019
;
14
(
6
):
1841
62
.
28.
Natarajan
KN
.
Single-cell tagged reverse transcription (STRT-Seq)
.
Methods Mol Biol
.
2019
;
1979
:
133
53
.
29.
Gong
H
,
Do
D
,
Ramakrishnan
R
.
Single-cell mRNA-seq using the Fluidigm C1 system and integrated fluidics circuits
.
Methods Mol Biol
.
2018
;
1783
:
193
207
.
30.
Fan
HC
,
Fu
GK
,
Fodor
SPA
.
Expression profiling. Combinatorial labeling of single cells for gene expression cytometry
.
Science
.
2015
;
347
(
6222
):
1258367
.
31.
Zheng
GXY
,
Terry
JM
,
Belgrader
P
,
Ryvkin
P
,
Bent
ZW
,
Wilson
R
, et al
.
Massively parallel digital transcriptional profiling of single cells
.
Nat Commun
.
2017
;
8
:
14049
.
32.
Luecken
MD
,
Theis
FJ
.
Current best practices in single-cell RNA-seq analysis: a tutorial
.
Mol Syst Biol
.
2019
;
15
(
6
):
e8746
.
33.
Hsu
CW
,
Shahan
R
,
Nolan
TM
,
Benfey
PN
,
Ohler
U
.
Protocol for fast scRNA-seq raw data processing using scKB and non-arbitrary quality control with COPILOT
.
STAR Protoc
.
2022
;
3
(
4
):
101729
.
34.
Hippen
AA
,
Falco
MM
,
Weber
LM
,
Erkan
EP
,
Zhang
K
,
Doherty
JA
, et al
.
miQC: an adaptive probabilistic framework for quality control of single-cell RNA-sequencing data
.
PLoS Comput Biol
.
2021
;
17
(
8
):
e1009290
.
35.
Osorio
D
,
Cai
JJ
.
Systematic determination of the mitochondrial proportion in human and mice tissues for single-cell RNA-sequencing data quality control
.
Bioinformatics
.
2021
;
37
(
7
):
963
7
.
36.
Bacher
R
.
Normalization for single-cell RNA-seq data analysis
.
Methods Mol Biol
.
2019
;
1935
:
11
23
.
37.
Baran-Gale
J
,
Chandra
T
,
Kirschner
K
.
Experimental design for single-cell RNA sequencing
.
Brief Funct Genomics
.
2018
;
17
(
4
):
233
9
.
38.
Tran
HTN
,
Ang
KS
,
Chevrier
M
,
Zhang
X
,
Lee
NYS
,
Goh
M
, et al
.
A benchmark of batch-effect correction methods for single-cell RNA sequencing data
.
Genome Biol
.
2020
;
21
(
1
):
12
.
39.
Lytal
N
,
Ran
D
,
An
L
.
Normalization methods on single-cell RNA-seq data: an empirical survey
.
Front Genet
.
2020
;
11
:
41
.
40.
Cole
MB
,
Risso
D
,
Wagner
A
,
DeTomaso
D
,
Ngai
J
,
Purdom
E
, et al
.
Performance assessment and selection of normalization procedures for single-cell RNA-seq
.
Cell Syst
.
2019
;
8
(
4
):
315
28.e8
.
41.
Hafemeister
C
,
Satija
R
.
Normalization and variance stabilization of single-cell RNA-seq data using regularized negative binomial regression
.
Genome Biol
.
2019
;
20
(
1
):
296
.
42.
Zhang
Z
,
Cui
F
,
Lin
C
,
Zhao
L
,
Wang
C
,
Zou
Q
.
Critical downstream analysis steps for single-cell RNA sequencing data
.
Brief Bioinform
.
2021
;
22
(
5
):
bbab105
.
43.
Butler
A
,
Hoffman
P
,
Smibert
P
,
Papalexi
E
,
Satija
R
.
Integrating single-cell transcriptomic data across different conditions, technologies, and species
.
Nat Biotechnol
.
2018
;
36
(
5
):
411
20
.
44.
Kiselev
VY
,
Andrews
TS
,
Hemberg
M
.
Challenges in unsupervised clustering of single-cell RNA-seq data
.
Nat Rev Genet
.
2019
;
20
(
5
):
273
82
.
45.
Petegrosso
R
,
Li
Z
,
Kuang
R
.
Machine learning and statistical methods for clustering single-cell RNA-sequencing data
.
Brief Bioinform
.
2020
;
21
(
4
):
1209
23
.
46.
Qi
R
,
Ma
A
,
Ma
Q
,
Zou
Q
.
Clustering and classification methods for single-cell RNA-sequencing data
.
Brief Bioinform
.
2020
;
21
(
4
):
1196
208
.
47.
Wu
W
,
Ma
X
.
Joint learning dimension reduction and clustering of single-cell RNA-sequencing data
.
Bioinformatics
.
2020
;
36
(
12
):
3825
32
.
48.
Wu
W
,
Liu
Z
,
Ma
X
.
jSRC: a flexible and accurate joint learning algorithm for clustering of single-cell RNA-sequencing data
.
Brief Bioinform
.
2021
;
22
(
5
):
bbaa433
.
49.
Fang
Q
,
Su
D
,
Ng
W
,
Feng
J
.
An effective biclustering-based framework for identifying cell subpopulations from scRNA-seq data
.
IEEE/ACM Trans Comput Biol Bioinform
.
2021
;
18
(
6
):
2249
60
.
50.
Stassen
SV
,
Siu
DMD
,
Lee
KCM
,
Ho
JWK
,
So
HKH
,
Tsia
KK
.
PARC: ultrafast and accurate clustering of phenotypic data of millions of single cells
.
Bioinformatics
.
2020
;
36
(
9
):
2778
86
.
51.
Shao
X
,
Liao
J
,
Lu
X
,
Xue
R
,
Ai
N
,
Fan
X
.
scCATCH: automatic annotation on cell types of clusters from single-cell RNA sequencing data
.
iScience
.
2020
;
23
(
3
):
100882
.
52.
Herring
CA
,
Chen
B
,
McKinley
ET
,
Lau
KS
.
Single-cell computational strategies for lineage reconstruction in tissue systems
.
Cell Mol Gastroenterol Hepatol
.
2018
;
5
(
4
):
539
48
.
53.
Wei
J
,
Zhou
T
,
Zhang
X
,
Tian
T
.
DTFLOW: inference and visualization of single-cell pseudotime trajectory using diffusion propagation
.
Genomics Proteomics Bioinformatics
.
2021
;
19
(
2
):
306
18
.
54.
Ji
Z
,
Ji
H
.
TSCAN: pseudo-time reconstruction and evaluation in single-cell RNA-seq analysis
.
Nucleic Acids Res
.
2016
;
44
(
13
):
e117
.
55.
Liu
J
,
Fan
Z
,
Zhao
W
,
Zhou
X
.
Machine intelligence in single-cell data analysis: advances and new challenges
.
Front Genet
.
2021
;
12
:
655536
.
56.
Chen
Y
,
Zhang
Y
,
Ouyang
Z
.
LISA: accurate reconstruction of cell trajectory and pseudo-time for massive single cell RNA-seq data
.
Pac Symp Biocomput
.
2019
;
24
:
338
49
.
57.
Herring
CA
,
Banerjee
A
,
McKinley
ET
,
Simmons
AJ
,
Ping
J
,
Roland
JT
, et al
.
Unsupervised trajectory analysis of single-cell RNA-seq and imaging data reveals alternative tuft cell origins in the gut
.
Cell Syst
.
2018
;
6
(
1
):
37
51.e9
.
58.
Schiebinger
G
,
Shu
J
,
Tabaka
M
,
Cleary
B
,
Subramanian
V
,
Solomon
A
, et al
.
Optimal-transport analysis of single-cell gene expression identifies developmental trajectories in reprogramming
.
Cell
.
2019
;
176
(
4
):
928
43.e22
.
59.
Saelens
W
,
Cannoodt
R
,
Todorov
H
,
Saeys
Y
.
A comparison of single-cell trajectory inference methods
.
Nat Biotechnol
.
2019
;
37
(
5
):
547
54
.
60.
Zheng
SC
,
Stein-O’Brien
G
,
Boukas
L
,
Goff
LA
,
Hansen
KD
.
Pumping the brakes on RNA velocity by understanding and interpreting RNA velocity estimates
.
Genome Biol
.
2023
;
24
(
1
):
246
.
61.
Vieth
B
,
Parekh
S
,
Ziegenhain
C
,
Enard
W
,
Hellmann
I
.
A systematic evaluation of single cell RNA-seq analysis pipelines
.
Nat Commun
.
2019
;
10
(
1
):
4667
.
62.
Miao
Z
,
Deng
K
,
Wang
X
,
Zhang
X
.
DEsingle for detecting three types of differential expression in single-cell RNA-seq data
.
Bioinformatics
.
2018
;
34
(
18
):
3223
4
.
63.
Chen
HIH
,
Jin
Y
,
Huang
Y
,
Chen
Y
.
Detection of high variability in gene expression from single-cell RNA-seq profiling
.
BMC Genomics
.
2016
;
17
(
Suppl 7
):
508
.
64.
Ye
C
,
Speed
TP
,
Salim
A
.
DECENT: differential expression with capture efficiency adjustmeNT for single-cell RNA-seq data
.
Bioinformatics
.
2019
;
35
(
24
):
5155
62
.
65.
Zhang
W
,
Wei
Y
,
Zhang
D
,
Xu
EY
.
ZIAQ: a quantile regression method for differential expression analysis of single-cell RNA-seq data
.
Bioinformatics
.
2020
;
36
(
10
):
3124
30
.
66.
Xu
J
,
Cui
L
,
Zhuang
J
,
Meng
Y
,
Bing
P
,
He
B
, et al
.
Evaluating the performance of dropout imputation and clustering methods for single-cell RNA sequencing data
.
Comput Biol Med
.
2022
;
146
:
105697
.
67.
Jiang
R
,
Sun
T
,
Song
D
,
Li
JJ
.
Statistics or biology: the zero-inflation controversy about scRNA-seq data
.
Genome Biol
.
2022
;
23
(
1
):
31
.
68.
Geistlinger
L
,
Csaba
G
,
Santarelli
M
,
Ramos
M
,
Schiffer
L
,
Turaga
N
, et al
.
Toward a gold standard for benchmarking gene set enrichment analysis
.
Brief Bioinform
.
2021
;
22
(
1
):
545
56
.
69.
Ma
Y
,
Sun
S
,
Shang
X
,
Keller
ET
,
Chen
M
,
Zhou
X
.
Integrative differential expression and gene set enrichment analysis using summary statistics for scRNA-seq studies
.
Nat Commun
.
2020
;
11
(
1
):
1585
.
70.
Holland
CH
,
Tanevski
J
,
Perales-Patón
J
,
Gleixner
J
,
Kumar
MP
,
Mereu
E
, et al
.
Robustness and applicability of transcription factor and pathway analysis tools on single-cell RNA-seq data
.
Genome Biol
.
2020
;
21
(
1
):
36
.
71.
Aibar
S
,
González-Blas
CB
,
Moerman
T
,
Huynh-Thu
VA
,
Imrichova
H
,
Hulselmans
G
, et al
.
SCENIC: single-cell regulatory network inference and clustering
.
Nat Methods
.
2017
;
14
(
11
):
1083
6
.
72.
Ruohan
Z
,
Yicheng
B
,
Jingying
Z
,
Mei
H
,
Xinyan
Z
,
Min
Y
, et al
.
Advanced analysis and applications of single-cell transcriptome sequencing
.
Life
.
2023
;
16
(
1
):
2199140
.
73.
Efremova
M
,
Vento-Tormo
M
,
Teichmann
SA
,
Vento-Tormo
R
.
CellPhoneDB: inferring cell-cell communication from combined expression of multi-subunit ligand-receptor complexes
.
Nat Protoc
.
2020
;
15
(
4
):
1484
506
.
74.
Jin
S
,
Guerrero-Juarez
CF
,
Zhang
L
,
Chang
I
,
Ramos
R
,
Kuan
CH
, et al
.
Inference and analysis of cell-cell communication using CellChat
.
Nat Commun
.
2021
;
12
(
1
):
1088
.
75.
Dimitrov
D
,
Türei
D
,
Garrido-Rodriguez
M
,
Burmedi
PL
,
Nagai
JS
,
Boys
C
, et al
.
Comparison of methods and resources for cell-cell communication inference from single-cell RNA-Seq data
.
Nat Commun
.
2022
;
13
(
1
):
3224
.
76.
Zappia
L
,
Phipson
B
,
Oshlack
A
.
Exploring the single-cell RNA-seq analysis landscape with the scRNA-tools database
.
PLoS Comput Biol
.
2018
;
14
(
6
):
e1006245
.
77.
Baysoy
A
,
Bai
Z
,
Satija
R
,
Fan
R
.
The technological landscape and applications of single-cell multi-omics
.
Nat Rev Mol Cell Biol
.
2023
;
24
(
10
):
695
713
.
78.
Adil
A
,
Kumar
V
,
Jan
AT
,
Asger
M
.
Single-cell transcriptomics: current methods and challenges in data acquisition and analysis
.
Front Neurosci
.
2021
;
15
:
591122
.
79.
Denisenko
E
,
Guo
BB
,
Jones
M
,
Hou
R
,
de Kock
L
,
Lassmann
T
, et al
.
Systematic assessment of tissue dissociation and storage biases in single-cell and single-nucleus RNA-seq workflows
.
Genome Biol
.
2020
;
21
(
1
):
130
.
80.
Adam
M
,
Potter
AS
,
Potter
SS
.
Psychrophilic proteases dramatically reduce single-cell RNA-seq artifacts: a molecular atlas of kidney development
.
Development
.
2017
;
144
(
19
):
3625
32
.
81.
van den Brink
SC
,
Sage
F
,
Vértesy
Á
,
Spanjaard
B
,
Peterson-Maduro
J
,
Baron
CS
, et al
.
Single-cell sequencing reveals dissociation-induced gene expression in tissue subpopulations
.
Nat Methods
.
2017
;
14
(
10
):
935
6
.
82.
Sarkar
A
,
Stephens
M
.
Separating measurement and expression models clarifies confusion in single-cell RNA sequencing analysis
.
Nat Genet
.
2021
;
53
(
6
):
770
7
.
83.
Luo
Q
,
Zhang
H
.
Emergence of bias during the synthesis and amplification of cDNA for scRNA-seq
.
Adv Exp Med Biol
.
2018
;
1068
:
149
58
.
84.
Cheng
C
,
Chen
W
,
Jin
H
,
Chen
X
.
A review of single-cell RNA-seq annotation, integration, and cell-cell communication
.
Cells
.
2023
;
12
(
15
):
1970
.
85.
Shen
AJJ
,
King
J
,
Scott
H
,
Colman
P
,
Yates
CJ
.
Insights into pituitary tumorigenesis: from Sanger sequencing to next-generation sequencing and beyond
.
Expert Rev Endocrinol Metab
.
2019
;
14
(
6
):
399
418
.
86.
Kuksin
M
,
Morel
D
,
Aglave
M
,
Danlos
FX
,
Marabelle
A
,
Zinovyev
A
, et al
.
Applications of single-cell and bulk RNA sequencing in onco-immunology
.
Eur J Cancer
.
2021
;
149
:
193
210
.
87.
Macaulay
IC
,
Teng
MJ
,
Haerty
W
,
Kumar
P
,
Ponting
CP
,
Voet
T
.
Separation and parallel sequencing of the genomes and transcriptomes of single cells using G&T-seq
.
Nat Protoc
.
2016
;
11
(
11
):
2081
103
.
88.
SIDR
.
Simultaneous isolation and parallel sequencing of genomic DNA and total RNA from single cells
. Available from: https://pubmed.ncbi.nlm.nih.gov/29208629/ (Accessed June 29, 2024).
89.
Angermueller
C
,
Clark
SJ
,
Lee
HJ
,
Macaulay
IC
,
Teng
MJ
,
Hu
TX
, et al
.
Parallel single-cell sequencing links transcriptional and epigenetic heterogeneity
.
Nat Methods
.
2016
;
13
(
3
):
229
32
.
90.
Clark
SJ
,
Argelaguet
R
,
Kapourani
CA
,
Stubbs
TM
,
Lee
HJ
,
Alda-Catalinas
C
, et al
.
scNMT-seq enables joint profiling of chromatin accessibility DNA methylation and transcription in single cells
.
Nat Commun
.
2018
;
9
(
1
):
781
.
91.
Baek
S
,
Lee
I
.
Single-cell ATAC sequencing analysis: from data preprocessing to hypothesis generation
.
Comput Struct Biotechnol J
.
2020
;
18
:
1429
39
.
92.
Stuart
T
,
Butler
A
,
Hoffman
P
,
Hafemeister
C
,
Papalexi
E
,
Mauck
WM
3rd
, et al
.
Comprehensive integration of single-cell data
.
Cell
.
2019
;
177
(
7
):
1888
902.e21
.
93.
Liu
Y
,
DiStasio
M
,
Su
G
,
Asashima
,
H
,
Enninful
,
A
,
Qin
,
X
, et al
.
Spatial-CITE-seq: spatially resolved high-plex protein and whole transcriptome co-mapping
.
Res Sq
.
2022
:
rs.3.rs-1499315
.
94.
Vandereyken
K
,
Sifrim
A
,
Thienpont
B
,
Voet
T
.
Methods and applications for single-cell and spatial multi-omics
.
Nat Rev Genet
.
2023
;
24
(
8
):
494
515
.
95.
Zhang
K
,
Hocker
JD
,
Miller
M
,
Hou
X
,
Chiou
J
,
Poirion
OB
, et al
.
A single-cell atlas of chromatin accessibility in the human genome
.
Cell
.
2021
;
184
(
24
):
5985
6001.e19
.
96.
Cheung
LYM
,
George
AS
,
McGee
SR
,
Daly
AZ
,
Brinkmeier
ML
,
Ellsworth
BS
, et al
.
Single-cell RNA sequencing reveals novel markers of male pituitary stem cells and hormone-producing cell types
.
Endocrinology
.
2018
;
159
(
12
):
3910
24
.
97.
Ho
Y
,
Hu
P
,
Peel
MT
,
Chen
S
,
Camara
PG
,
Epstein
DJ
, et al
.
Single-cell transcriptomic analysis of adult mouse pituitary reveals sexual dimorphism and physiologic demand-induced cellular plasticity
.
Protein Cell
.
2020
;
11
(
8
):
565
83
.
98.
Chen
Q
,
Leshkowitz
D
,
Blechman
J
,
Levkowitz
G
.
Single-cell molecular and cellular architecture of the mouse neurohypophysis
.
eNeuro
.
2020
;
7
(
1
):
ENEURO.0345–19.2019
.
99.
Cheung
LYM
,
Menage
L
,
Rizzoti
K
,
Hamilton
G
,
Dumontet
T
,
Basham
K
, et al
.
Novel candidate regulators and developmental trajectory of pituitary thyrotropes
.
Endocrinology
.
2023
;
164
(
6
):
bqad076
.
100.
Ruf-Zamojski
F
,
Ge
Y
,
Nair
V
,
Zamojski
M
,
Pincas
H
,
Toufaily
C
, et al
.
Single-cell stabilization method identifies gonadotrope transcriptional dynamics and pituitary cell type heterogeneity
.
Nucleic Acids Res
.
2018
;
46
(
21
):
11370
80
.
101.
Ruf-Zamojski
F
,
Fribourg
M
,
Ge
Y
,
Nair
V
,
Pincas
H
,
Zaslavsky
E
, et al
.
Regulatory architecture of the LβT2 gonadotrope cell underlying the response to gonadotropin-releasing hormone
.
Front Endocrinol
.
2018
;
9
:
34
.
102.
Ruf-Zamojski
F
,
Zhang
Z
,
Zamojski
M
,
Smith
GR
,
Mendelev
N
,
Liu
H
, et al
.
Single nucleus multi-omics regulatory landscape of the murine pituitary
.
Nat Commun
.
2021
;
12
(
1
):
2677
.
103.
Zhang
Z
,
Ruf-Zamojski
F
,
Zamojski
M
,
Bernard
DJ
,
Chen
X
,
Troyanskaya
OG
, et al
.
Peak-agnostic high-resolution cis-regulatory circuitry mapping using single cell multiome data
.
Nucleic Acids Res
.
2024
;
52
(
2
):
572
82
.
104.
Mayran
A
,
Sochodolsky
K
,
Khetchoumian
K
,
Harris
J
,
Gauthier
Y
,
Bemmo
A
, et al
.
Pioneer and nonpioneer factor cooperation drives lineage specific chromatin opening
.
Nat Commun
.
2019
;
10
(
1
):
3807
.
105.
Vennekens
A
,
Laporte
E
,
Hermans
F
,
Cox
B
,
Modave
E
,
Janiszewski
A
, et al
.
Interleukin-6 is an activator of pituitary stem cells upon local damage, a competence quenched in the aging gland
.
Proc Natl Acad Sci U S A
.
2021
;
118
(
25
):
e2100052118
.
106.
Laporte
E
,
Hermans
F
,
De Vriendt
S
,
Vennekens
A
,
Lambrechts
D
,
Nys
C
, et al
.
Decoding the activated stem cell phenotype of the neonatally maturing pituitary
.
Elife
.
2022
;
11
:
e75742
.
107.
Rizzoti
K
,
Chakravarty
P
,
Sheridan
D
,
Lovell-Badge
R
.
SOX9-positive pituitary stem cells differ according to their position in the gland and maintenance of their progeny depends on context
.
Sci Adv
.
2023
;
9
(
40
):
eadf6911
.
108.
Zhang
Z
,
Zamojski
M
,
Smith
GR
,
Willis
TL
,
Yianni
V
,
Mendelev
N
, et al
.
Single nucleus transcriptome and chromatin accessibility of postmortem human pituitaries reveal diverse stem cell regulatory mechanisms
.
Cell Rep
.
2022
;
38
(
10
):
110467
.
109.
Zhang
S
,
Cui
Y
,
Ma
X
,
Yong
J
,
Yan
L
,
Yang
M
, et al
.
Single-cell transcriptomics identifies divergent developmental lineage trajectories during human pituitary development
.
Nat Commun
.
2020
;
11
(
1
):
5275
.
110.
Kučka
M
,
Gonzalez-Iglesias
AE
,
Tomić
M
,
Prévide
RM
,
Smiljanic
K
,
Sokanovic
SJ
, et al
.
Calcium-Prolactin secretion coupling in rat pituitary lactotrophs is controlled by PI4-kinase alpha
.
Front Endocrinol
.
2021
;
12
:
790441
.
111.
Chen
Q
,
Leshkowitz
D
,
Li
H
,
van Impel
A
,
Schulte-Merker
S
,
Amit
I
, et al
.
Neural plate progenitors give rise to both anterior and posterior pituitary cells
.
Dev Cell
.
2023
;
58
(
23
):
2652
65.e6
.
112.
Miles
TK
,
Odle
AK
,
Byrum
SD
,
Lagasse
A
,
Haney
A
,
Ortega
VG
, et al
.
Anterior pituitary transcriptomics following a high-fat diet: impact of oxidative stress on cell metabolism
.
Endocrinology
.
2023
;
165
(
2
):
bqad191
.
113.
Ruggiero-Ruff
RE
,
Le
BH
,
Villa
PA
,
Lainez
NM
,
Athul
SW
,
Das
P
, et al
.
Single-cell transcriptomics identifies pituitary gland changes in diet-induced obesity in male mice
.
Endocrinology
.
2024
;
165
(
3
):
bqad196
.
114.
Yan
T
,
Wang
R
,
Yao
J
,
Luo
M
.
Single-cell transcriptomic analysis reveals rich pituitary-Immune interactions under systemic inflammation
.
PLoS Biol
.
2023
;
21
(
12
):
e3002403
.
115.
Moncho-Amor
V
,
Chakravarty
P
,
Galichet
C
,
Matheu
A
,
Lovell-Badge
R
,
Rizzoti
K
.
SOX2 is required independently in both stem and differentiated cells for pituitary tumorigenesis in p27-null mice
.
Proc Natl Acad Sci U S A
.
2021
;
118
(
7
):
e2017115118
.
116.
Cui
Y
,
Li
C
,
Jiang
Z
,
Zhang
S
,
Li
Q
,
Liu
X
, et al
.
Single-cell transcriptome and genome analyses of pituitary neuroendocrine tumors
.
Neuro Oncol
.
2021
;
23
(
11
):
1859
71
.
117.
Yan
N
,
Xie
W
,
Wang
D
,
Fang
Q
,
Guo
J
,
Chen
Y
, et al
.
Single-cell transcriptomic analysis reveals tumor cell heterogeneity and immune microenvironment features of pituitary neuroendocrine tumors
.
Genome Med
.
2024
;
16
(
1
):
2
.
118.
Batchu
S
,
Diaz
MJ
,
Lin
K
,
Arya
N
,
Patel
K
,
Lucke-Wold
B
.
Single cell metabolic landscape of pituitary neuroendocrine tumor subgroups and lineages
.
OBM Neurobiol
.
2023
;
07
(
01
):
1
11
.
119.
Asuzu
DT
,
Alvarez
R
,
Fletcher
PA
,
Mandal
D
,
Johnson
K
,
Wu
W
, et al
.
Pituitary adenomas evade apoptosis via noxa deregulation in Cushing’s disease
.
Cell Rep
.
2022
;
40
(
8
):
111223
.
120.
Zhang
D
,
Hugo
W
,
Bergsneider
M
,
Wang
MB
,
Kim
W
,
Vinters
HV
, et al
.
Single-cell RNA sequencing in silent corticotroph tumors confirms impaired POMC processing and provides new insights into their invasive behavior
.
Eur J Endocrinol
.
2022
;
187
(
1
):
49
64
.
121.
Zhang
Q
,
Yao
B
,
Long
X
,
Chen
Z
,
He
M
,
Wu
Y
, et al
.
Single-cell sequencing identifies differentiation-related markers for molecular classification and recurrence prediction of PitNET
.
Cell Rep Med
.
2023
;
4
(
2
):
100934
.
122.
Cheng
J
,
Xie
W
,
Chen
Y
,
Sun
Y
,
Gong
L
,
Wang
H
, et al
.
Drug resistance mechanisms in dopamine agonist-resistant prolactin pituitary neuroendocrine tumors and exploration for new drugs
.
Drug Resist Updat
.
2024
;
73
:
101056
.
123.
Lyu
L
,
Jiang
Y
,
Ma
W
,
Li
H
,
Liu
X
,
Li
L
, et al
.
Single-cell sequencing of PIT1-positive pituitary adenoma highlights the pro-tumour microenvironment mediated by IFN-γ-induced tumour-associated fibroblasts remodelling
.
Br J Cancer
.
2023
;
128
(
6
):
1117
33
.
124.
Melmed
S
,
Kaiser
UB
,
Lopes
MB
,
Bertherat
J
,
Syro
LV
,
Raverot
G
, et al
.
Clinical biology of the pituitary adenoma
.
Endocr Rev
.
2022
;
43
(
6
):
1003
37
.
125.
Molitch
ME
.
Diagnosis and treatment of pituitary adenomas: a review
.
JAMA
.
2017
;
317
(
5
):
516
24
.
126.
Elsarrag
M
,
Patel
PD
,
Chatrath
A
,
Taylor
D
,
Jane
JA
.
Genomic and molecular characterization of pituitary adenoma pathogenesis: review and translational opportunities
.
Neurosurg Focus
.
2020
;
48
(
6
):
E11
.
127.
Spada
A
,
Mantovani
G
,
Lania
AG
,
Treppiedi
D
,
Mangili
F
,
Catalano
R
, et al
.
Pituitary tumors: genetic and molecular factors underlying pathogenesis and clinical behavior
.
Neuroendocrinology
.
2022
;
112
(
1
):
15
33
.
128.
Jiang
X
,
Zhang
X
.
The molecular pathogenesis of pituitary adenomas: an update
.
Endocrinol Metab
.
2013
;
28
(
4
):
245
54
.
129.
Aldridge
S
,
Teichmann
SA
.
Single cell transcriptomics comes of age
.
Nat Commun
.
2020
;
11
(
1
):
4307
.
130.
Svensson
V
,
Vento-Tormo
R
,
Teichmann
SA
.
Exponential scaling of single-cell RNA-seq in the past decade
.
Nat Protoc
.
2018
;
13
(
4
):
599
604
.
131.
Rymuza
J
,
Kober
P
,
Rusetska
N
,
Mossakowska
BJ
,
Maksymowicz
M
,
Nyc
A
, et al
.
Transcriptomic classification of pituitary neuroendocrine tumors causing acromegaly
.
Cells
.
2022
;
11
(
23
):
3846
.
132.
Asa
SL
,
Mete
O
,
Riddle
ND
,
Perry
A
.
Multilineage Pituitary Neuroendocrine Tumors (PitNETs) expressing PIT1 and SF1
.
Endocr Pathol
.
2023
;
34
(
3
):
273
8
.
133.
Dottermusch
M
,
Ryba
A
,
Ricklefs
FL
,
Flitsch
J
,
Schmid
S
,
Glatzel
M
, et al
.
Pituitary neuroendocrine tumors with PIT1/SF1 co-expression show distinct clinicopathological and molecular features
.
Acta Neuropathol
.
2024
;
147
(
1
):
16
.