These mice overexpress GH, often through transgenic insertion of GH genes (e.g., bovine GH under a metallothionein promoter). Key features include:
GH/IGF-1 Axis: Elevated GH and insulin-like growth factor-1 (IGF-1) drive excessive growth but accelerate aging-related pathologies .
Cancer Link: Preclinical studies suggest GH/IGF-1 signaling may promote tumor growth, though clinical evidence remains debated .
These mice lack functional GH receptors, mimicking human Laron Syndrome. Their phenotype contrasts sharply with GH transgenic mice:
Aging: GH receptor insensitivity correlates with delayed physical and cognitive aging, suggesting GH signaling may drive senescence .
Cancer Resistance: Reduced GH/IGF-1 signaling in GHR−/− mice is linked to lower cancer incidence, highlighting therapeutic potential .
The following table contrasts key physiological and pathological outcomes:
Parameter | GH Transgenic Mice | GHR−/− Mice |
---|---|---|
GH Levels | Elevated | Elevated (due to receptor insensitivity) |
IGF-1 Levels | Elevated | Low |
Growth Pattern | Gigantism | Dwarfism |
Lifespan | Shortened | Extended |
Cancer Risk | Increased | Reduced |
Metabolic Profile | Hyperinsulinemia, lean | Insulin sensitivity, obesity resistance |
These models have driven advancements in:
Aging Biology: GHR−/− mice are used to study mechanisms of longevity and age-related disease resistance .
Cancer Therapeutics: Pegvisomant, a GH receptor antagonist developed using GHR−/− mice, is under investigation for cancer treatment .
Metabolic Disorders: Insights into GH’s role in insulin resistance and obesity have informed diabetes research .
Tissue-Specific Effects: Liver-specific GHR knockout in mice causes hepatic steatosis, while muscle-specific deletion improves glucose metabolism .
Neurological Impact: GHR−/− mice exhibit delayed cognitive decline, suggesting GH signaling may influence brain aging .
Translational Challenges: Mouse findings must be cautiously extrapolated to humans due to species-specific differences in metabolism and disease progression .
Therapeutic Potential: Targeting GH/IGF-1 pathways (e.g., pegvisomant) remains a promising but controversial approach for cancer and aging-related diseases .
GH1, GH, GHN, GH-N, hGH-N, Pituitary GH, GH-1.
The primary GH mouse models used in research fall into two main categories: those with enhanced GH signaling and those with reduced or absent GH signaling. The enhanced GH signaling models include transgenic mice expressing various species of GH genes (bovine, human, rat, or ovine GH), with the bovine GH (bGH) transgenic mouse being one of the most extensively studied . These mice exhibit elevated levels of GH and IGF-1, resulting in a giant phenotype. The reduced GH signaling models include the GH receptor gene disrupted mouse (GHR−/−), which is completely resistant to GH action, resembling human Laron Syndrome . There are also tissue-specific GHR knockout models that allow for examination of GH's effects in specific organs or tissues. Each model provides unique insights into different aspects of GH physiology and pathophysiology, making them invaluable tools for understanding growth, metabolism, aging, and various disease states.
GH mouse models serve as crucial tools for elucidating the complex roles of growth hormone in human physiology and disease. The bGH transgenic mice with chronically elevated GH levels develop a phenotype that closely resembles human acromegaly, including accelerated somatic growth, organomegaly, hyperinsulinemia despite euglycemia, and increased cancer incidence . These models allow researchers to investigate the molecular mechanisms underlying these conditions without the ethical limitations of human experimentation. Conversely, GHR−/− mice with no GH action exhibit extended lifespan and protection from various age-related diseases including cancer and diabetes, providing valuable insights into the role of GH/IGF-1 signaling in aging and longevity . The phenotypes observed in these mouse models have led to important translational discoveries, such as the development of pegvisomant, a GH receptor antagonist approved for treating acromegaly in humans . Additionally, these models help identify potential therapeutic targets for conditions ranging from growth disorders to metabolic diseases and cancer, demonstrating their significant contribution to both basic science and clinical medicine.
GH transgenic and GH receptor knockout mice exhibit strikingly opposite physiological characteristics, providing complementary insights into GH action. GH transgenic mice display significantly higher body weight and length compared to wild-type controls, while GHR−/− mice are substantially smaller . The table below summarizes key differences:
Phenotype | GH transgenic mouse vs. control | GHR−/− mouse vs. control |
---|---|---|
Body weight and length | Higher | Lower |
IGF-I level | Higher | Lower |
Lifespan | Shorter | Longer |
Age-related diseases | Increased incidence of liver inflammation, tumors, glomerulosclerosis | Lower incidence of cancer |
Glucose level | Normal or lower | Lower (normal in older animals) |
Insulin level | Higher | Lower |
Insulin sensitivity | Lower | Higher in muscle and heart, mixed results for liver |
These contrasting phenotypes highlight GH's wide-ranging effects on growth, metabolism, and aging . GH transgenic mice typically live only 12-18 months, dying prematurely from heart, liver, and kidney complications, whereas GHR−/− mice enjoy extended lifespans with protection from various age-related diseases . The metabolic profiles also differ dramatically, with GH transgenic mice showing insulin resistance despite their lean phenotype, while GHR−/− mice demonstrate enhanced insulin sensitivity in certain tissues despite increased adiposity. These opposing physiological outcomes provide powerful experimental paradigms for investigating the complex roles of GH signaling in health and disease.
Measuring GH pulsatility in mice presents significant technical challenges due to their small body size and limited blood volume, making conventional serial sampling approaches difficult. Researchers have developed alternative methods to overcome these limitations and accurately assess endogenous GH secretion patterns . One innovative approach involves detecting physiological GH patterns from randomly obtained spot samples, which allows for routine application in mice without requiring complicated catheterization procedures . This method can determine individual GH secretory patterns while minimizing stress to the animals. Additionally, some researchers employ specialized micro-sampling techniques with ultra-sensitive assays that require minimal blood volume. For more detailed studies, implantable micro-pumps and automated blood sampling systems have been developed that can collect small blood volumes at frequent intervals. Advanced statistical methods such as deconvolution analysis and time-series analysis are then applied to characterize pulsatile secretion parameters. These methodological advances have greatly enhanced our ability to study the complex temporal dynamics of GH secretion in mouse models, which is essential for understanding how alterations in secretory patterns contribute to various physiological and pathological states.
Proper control selection is critical for generating valid experimental results when working with GH mouse models. For GH transgenic mice, the ideal controls are non-transgenic littermates from the same breeding colony, which share the same genetic background except for the transgene . This approach minimizes confounding variables related to strain differences or environmental factors. For studies involving GHR−/− mice, heterozygous (GHR+/−) and wild-type (GHR+/+) littermates serve as appropriate controls, with heterozygotes showing generally normal phenotypes despite reduced receptor expression . When comparing across different GH mouse models (e.g., transgenic vs. knockout), researchers should include separate control groups for each model rather than using a single control group. Age and sex matching is particularly important due to the significant sexual dimorphism in GH secretion patterns in rodents. Additionally, researchers should consider developing tissue-specific or inducible GH receptor knockout models when studying adult-onset GH resistance, as global GHR−/− mice have developmental effects that may confound interpretation of adult phenotypes. These careful control strategies help distinguish direct effects of altered GH signaling from secondary adaptations or strain-specific characteristics, ensuring robust and reproducible experimental outcomes.
Multiple complementary techniques offer robust analysis of GH-dependent gene expression changes, each providing unique insights into the molecular mechanisms of GH action. RNA sequencing (RNA-seq) provides comprehensive, unbiased transcriptome profiling and has largely replaced microarray approaches for identifying novel GH-regulated genes and pathways . Quantitative real-time PCR remains essential for validating expression changes of specific target genes with high sensitivity and specificity. For protein-level confirmation, Western blotting and proteomic approaches such as SYPRO Orange staining followed by mass spectrometry (MS and MS/MS using MALDI-TOF) have proven effective in identifying GH-regulated proteins . When analyzing tissue-specific effects, laser capture microdissection combined with RNA-seq allows for precise examination of GH action in specific cell populations. Chromatin immunoprecipitation sequencing (ChIP-seq) has become invaluable for mapping GH-activated transcription factor binding sites, particularly for STAT5, a primary mediator of GH signaling. Time-course experiments are critical for distinguishing between early (primary) and late (secondary) gene expression changes following GH stimulation. Computational approaches integrating these diverse datasets are increasingly important for constructing comprehensive models of GH-regulated gene networks. These methodological approaches should be selected based on the specific research question, considering factors such as sensitivity requirements, temporal dynamics, and the biological context of GH signaling being investigated.
Different species of GH transgenes produce varying phenotypes in mice, primarily due to differences in receptor binding specificity and expression levels. Human GH (hGH) is unique among the species variants because it can bind to both GH and prolactin receptors in mice, resulting in simultaneous activation of both signaling pathways . This dual-receptor activation leads to distinct phenotypic effects compared to other species' GH transgenes, particularly in mammary tissue development and reproductive function. In contrast, bovine, rat, and ovine GH transgenes bind exclusively to the GH receptor, making them more suitable for studying specific GH-mediated effects without prolactin receptor interference . The bovine GH (bGH) transgenic mouse is one of the most extensively characterized models, exhibiting dramatic increases in body size, organ-specific enlargement (particularly liver and kidneys), and metabolic alterations including hyperinsulinemia despite a lean phenotype . The expression level of the transgene also significantly impacts phenotype severity, with higher expression generally correlating with more pronounced effects on growth, metabolism, and pathology. Some transgenic lines exhibit more severe diabetogenic effects or cancer predisposition, which may relate to both the species of GH and its expression level. These species-specific differences provide researchers with valuable options for investigating particular aspects of GH biology and pathophysiology, allowing for more precise modeling of human conditions associated with GH excess.
GH transgenic mice exhibit profound metabolic alterations that provide valuable insights into human conditions associated with GH excess, such as acromegaly. Despite their lean phenotype with reduced fat mass, these mice display significant insulin resistance and hyperinsulinemia with normal or slightly lower blood glucose levels . This paradoxical combination of leanness and insulin resistance challenges conventional understanding of metabolic disease and offers a unique model for studying the direct effects of GH on insulin signaling pathways. The hyperinsulinemia observed in GH transgenic mice results from GH's diabetogenic effects, which include decreased insulin receptor signaling in liver and muscle tissues, altered insulin receptor substrate (IRS) phosphorylation patterns, and changes in downstream kinase activation . These mice also show altered lipid metabolism with decreased adiposity despite increased caloric intake, reflecting GH's powerful lipolytic actions. Hepatic metabolism is particularly affected, with increased gluconeogenesis, altered lipid handling, and changes in mitochondrial function. The liver enlargement (hepatomegaly) observed in these mice is associated with both hypertrophy and hyperplasia of hepatocytes, leading to dysregulation of multiple oncogenic pathways and increased tumor risk . These metabolic phenotypes make GH transgenic mice excellent models for studying the pathophysiology of acromegaly-associated metabolic dysfunction, GH-induced insulin resistance, and the connections between growth factor signaling and metabolic disease.
Managing premature mortality in GH transgenic mouse colonies requires multifaceted approaches targeting the underlying pathologies associated with chronic GH excess. Since these mice typically live only 12-18 months due to heart, liver, and kidney complications , researchers should implement regular health monitoring protocols, including non-invasive assessments of cardiovascular function (echocardiography), renal function (urine analysis), and liver health (serum enzyme measurements). Dietary interventions can help mitigate some metabolic complications; specifically, low-carbohydrate diets may improve insulin sensitivity and reduce hyperinsulinemia, while protein-restricted diets might alleviate renal stress. Housing conditions should be optimized to minimize additional stressors, with particular attention to temperature control, as these mice may have altered thermoregulation due to their metabolic phenotype. Breeding strategies should focus on maintaining heterozygous carriers rather than homozygous transgenic mice when possible, as heterozygotes often show intermediate phenotypes with less severe complications. For certain experiments, researchers might consider using inducible transgenic systems that allow temporal control of GH overexpression, thereby limiting chronic exposure effects. Tissue-specific GH transgenic models may also provide alternatives with less systemic toxicity while still allowing study of GH effects in target tissues. Finally, researchers should consider incorporating GH antagonist treatments (similar to pegvisomant used in human acromegaly) as experimental interventions to determine if pharmacological blockade of GH action can extend lifespan and improve health outcomes in these models . These strategies not only improve animal welfare but also enhance experimental utility by extending the window for longitudinal studies.
Tissue-specific GHR knockout models offer several distinct advantages over global GHR−/− mice for dissecting the complex, tissue-specific actions of GH. By selectively eliminating GH signaling in specific tissues while maintaining normal GH action elsewhere, these models allow researchers to distinguish direct from indirect effects of GH on particular organ systems . This approach overcomes a major limitation of global knockouts, where developmental adaptations and compensatory mechanisms can confound interpretation of phenotypes. Tissue-specific models enable investigation of adult-onset GH resistance without the developmental reprogramming that occurs in global knockouts from embryonic stages. For example, liver-specific GHR knockouts have revealed distinct roles of hepatic GH signaling in regulating systemic IGF-1 levels and metabolism that differ from effects observed in global knockouts . Fat-specific GHR knockout mice (FaGHRKO) demonstrate dramatically different body composition compared to global knockouts, with increased adiposity yet normal or increased body size, highlighting tissue-specific roles of GH in regulating fat versus lean mass . Muscle, bone, and pancreatic β-cell specific knockouts have similarly revealed unique aspects of GH action in these tissues that were not apparent from studies of global knockouts. These models also better recapitulate certain human conditions where GH resistance may develop in a tissue-specific manner, such as in metabolic syndrome or diabetes. The combined use of global and tissue-specific GHR knockout models provides a comprehensive approach to understanding the integrated physiological effects of GH across multiple organ systems.
The extended lifespan and remarkable cancer resistance of GHR−/− mice involve multiple interconnected mechanisms related to altered metabolism, reduced oxidative stress, and modified cellular signaling pathways. Lifespan extension in these mice is substantial, with a significant reduction in neoplastic disease burden – 83% of wild-type mice die from neoplastic disease compared to only 42% of GHR−/− mice, representing a 49% reduction . At the molecular level, reduced IGF-1 signaling appears central to these benefits, as IGF-1 promotes cell proliferation and inhibits apoptosis, two processes critically involved in cancer development and progression . GHR−/− mice also exhibit enhanced insulin sensitivity in specific tissues, particularly muscle and heart, which may contribute to their metabolic health despite increased adiposity . Reduced oxidative stress constitutes another key mechanism, with GHR−/− mice showing decreased production of reactive oxygen species and enhanced antioxidant defenses, protecting against DNA damage and cellular senescence. Proteomic studies have revealed altered expression of proteins involved in various metabolic pathways, stress responses, and detoxification systems, suggesting broad reprogramming of cellular maintenance systems . Additionally, these mice show evidence of altered autophagy and protein homeostasis, which may contribute to improved cellular maintenance and stress resistance. The metabolic shift toward enhanced fat metabolism rather than glucose utilization may also promote cellular resilience under various stress conditions. These multiple mechanisms highlight how reduced GH signaling triggers comprehensive physiological adaptations that collectively enhance longevity and disease resistance, providing valuable insights into the fundamental biology of aging and cancer.
GH mouse models have revolutionized our understanding of aging and longevity by establishing the GH/IGF-1 axis as a critical regulator of lifespan. GHR−/− mice with absent GH signaling consistently demonstrate extended lifespan compared to wild-type controls, while GH transgenic mice with chronic GH excess show dramatically shortened lifespans, dying prematurely from multiple organ complications . This inverse relationship between GH action and longevity provides compelling evidence that reduced growth factor signaling promotes longevity across species. The longevity benefit in GHR−/− mice involves multiple mechanisms including reduced oxidative stress, enhanced stress resistance, altered metabolism, and reduced cancer incidence (49% reduction compared to wild-type) . Notably, the extended lifespan phenotype despite increased adiposity in GHR−/− mice challenges conventional wisdom about obesity and aging, suggesting that metabolic health rather than adiposity per se determines longevity outcomes. Proteomic studies have identified numerous differentially expressed proteins in these models related to stress response, detoxification systems, and metabolic pathways that may contribute to their altered aging trajectories . These models have also helped establish important connections between early-life growth, adult metabolism, and longevity, supporting the concept that developmental programming influences aging outcomes. The parallels between findings in these mouse models and observations in long-lived human populations with GH/IGF-1 alterations, including Laron Syndrome patients, strengthen the translational significance of these discoveries. By establishing the GH/IGF-1 pathway as a conserved longevity-regulating mechanism, these mouse models have significantly influenced theories of aging and identified potential intervention targets for extending healthy lifespan.
GH mouse models offer powerful systems for investigating the complex relationship between growth hormone signaling and cancer development, progression, and treatment. The contrasting cancer phenotypes observed in GH transgenic versus GHR−/− mice provide particularly valuable insights: GH transgenic mice show significantly increased cancer incidence, especially liver tumors and mammary neoplasms, while GHR−/− mice exhibit remarkable cancer resistance with a 49% reduction in neoplastic disease mortality compared to wild-type mice . Researchers can utilize these models to investigate several key aspects of cancer biology. First, they can explore the molecular mechanisms through which GH/IGF-1 signaling promotes tumorigenesis, including effects on cell proliferation, apoptosis, angiogenesis, and DNA repair pathways. Second, tissue-specific GHR knockout models allow precise determination of whether GH's effects on cancer are direct actions on pre-neoplastic or tumor cells versus indirect effects mediated through altered metabolism or systemic environment. Third, these models are invaluable for testing GH antagonists, such as pegvisomant, as potential anti-cancer therapeutics, particularly for cancers where IGF-1 signaling is implicated . Fourth, by crossing GH mouse models with established cancer models (e.g., APC^Min/+^ for intestinal tumors or MMTV-PyMT for mammary tumors), researchers can directly assess how altered GH signaling modifies tumor initiation, progression, and metastasis in specific cancer types. Finally, these models allow investigation of the relationship between metabolism, aging, and cancer, helping to elucidate how GH-induced metabolic changes influence the tumor microenvironment. These diverse applications make GH mouse models essential tools for developing novel cancer prevention and treatment strategies targeted at the GH/IGF-1 axis.
Proteomic analyses of GH mouse models have yielded comprehensive molecular insights into the diverse physiological effects of altered GH signaling. Using advanced techniques such as SYPRO Orange staining followed by mass spectrometry (MS and MS/MS using MALDI-TOF and MALDI-TOF-TOF), researchers have identified approximately 160 mouse plasma protein spots with significant differences between bGH transgenic, GHR−/− mice, and their controls . These analyses have revealed altered expression of proteins involved in multiple pathways and processes. In GH transgenic mice, proteins associated with growth promotion, inflammation, oxidative stress, and tumor development show increased expression, correlating with their phenotype of accelerated growth, premature aging, and increased cancer risk . Conversely, GHR−/− mice exhibit differential expression of proteins involved in stress resistance, detoxification pathways, and alternative metabolic processing, which may contribute to their extended lifespan and cancer resistance . Liver proteome analyses have been particularly informative, revealing extensive remodeling of metabolic enzymes, acute phase proteins, and cellular maintenance systems across these models. Proteomic studies have also identified post-translational modifications affected by GH status, including phosphorylation patterns of insulin signaling components that help explain the paradoxical insulin sensitivity phenotypes observed in these models. Time-course proteomic analyses have distinguished between early and late protein expression changes following altered GH signaling, helping to separate primary from secondary effects. These comprehensive molecular profiles have significantly advanced our understanding of how GH signaling integrates growth, metabolism, and aging at the protein level, complementing transcriptomic studies and providing new biomarkers and therapeutic targets for conditions associated with altered GH action.
GH mouse models serve as powerful platforms for developing novel therapeutics targeting metabolic and age-related diseases through multiple complementary approaches. The discovery of the growth hormone receptor antagonist (GHA) and subsequent development of pegvisomant for treating acromegaly represents a prime example of successful therapeutic translation from these models . Researchers can leverage the extreme metabolic phenotypes in these mice to identify and validate new drug targets. For instance, the enhanced insulin sensitivity observed in GHR−/− mice despite increased adiposity has led to investigation of tissue-specific GH antagonists that could improve insulin action without affecting growth . The extended lifespan and reduced cancer incidence in GHR−/− mice also provide rationale for developing partial GH/IGF-1 inhibitors as potential anti-aging or cancer preventive agents . These models are excellent testbeds for evaluating therapeutic hypotheses before human trials, as demonstrated by studies showing that GHR−/− mice are protected from both chemically-induced diabetes and high-fat diet-induced insulin resistance . Conditional and inducible knockout models allow precise determination of whether interventions need to be initiated early in development or can be effective when started in adulthood. Proteomic and metabolomic profiling of these models has identified numerous biomarkers that can serve as pharmacodynamic endpoints in preclinical drug development . Additionally, these models can be used to investigate potential side effects of GH modulation therapies, such as effects on bone density, reproductive function, and cognitive outcomes. By providing systems with predictable, extreme alterations in GH signaling, these mouse models continue to be invaluable for developing targeted interventions for conditions ranging from diabetes and obesity to cancer and age-related diseases.
Breeding and maintaining GH mouse colonies presents several challenges that require specialized management strategies. For GH transgenic mice, reduced fertility is a significant obstacle, particularly in females, which often show irregular estrous cycles and reduced reproductive success . Males may also exhibit reduced fertility despite their larger size, necessitating careful breeding schemes that typically utilize heterozygous rather than homozygous transgenic mice. The shortened lifespan of GH transgenic mice (12-18 months) further complicates colony maintenance, requiring accelerated breeding schedules and careful health monitoring . GHR−/− mice present different challenges, with both males and females showing delayed sexual maturation and reduced fertility compared to wild-type littermates . These mice also typically have smaller litter sizes and sometimes exhibit poor maternal care behaviors, which may necessitate fostering pups to wild-type dams. For both models, genotyping is essential but can be complicated by the need to distinguish heterozygous from homozygous animals for some experimental purposes. Proper genetic background maintenance is critical, as background strains can significantly influence phenotype severity and variability. Health monitoring should be tailored to the specific complications each model is prone to develop: cardiovascular, renal, and liver complications for GH transgenic mice; and hypoglycemia, thermoregulatory issues, and developmental delays for GHR−/− mice. Nutritional management also differs, with GH transgenic mice requiring higher caloric intake while GHR−/− mice may benefit from specialized diets to address their altered metabolism. These challenges underscore the importance of experienced animal care staff and close collaboration between researchers and veterinarians to maintain healthy, productive colonies of these valuable but demanding mouse models.
Addressing data variability and ensuring reproducibility in GH mouse studies requires rigorous methodological approaches tailored to these unique models. Standardizing experimental conditions is critical, as GH secretion in mice is highly pulsatile and sensitive to environmental factors such as stress, feeding time, and light cycles . Researchers should conduct experiments at consistent times of day, preferably in the morning when endogenous GH pulses are more predictable in mice. Sample sizes should be appropriately powered to account for the inherent variability in GH-related parameters, with power calculations based on preliminary data from the specific model being used. Careful genetic background control is essential, as background strains significantly influence GH-related phenotypes; backcrossing for at least 8-10 generations onto a common background is recommended when comparing different GH models . Sex stratification in experimental design and analysis is particularly important given the pronounced sexual dimorphism in GH secretion patterns in rodents. Age standardization is equally critical, as the phenotypic expression of both GH excess and deficiency changes markedly with developmental stage and aging. For studies involving metabolic parameters, which are especially variable, overnight fasting protocols should be standardized and consistently applied. When measuring GH-dependent outcomes, consideration of the pulsatile nature of GH secretion requires either multiple sampling strategies or alternative approaches such as the random sampling method with statistical pattern recognition described in the literature . Finally, comprehensive reporting of methodological details, including specific substrain, housing conditions, diet, age, sex, sample collection timing, and assay characteristics is essential for reproducibility. These methodological considerations, while demanding, significantly enhance the reliability and translational value of findings from GH mouse models.
Translating findings from mouse GH models to human applications requires careful consideration of several important physiological differences between species. The most fundamental difference is in GH secretion patterns: while both species exhibit pulsatile GH release, humans show minimal sex differences in secretion patterns, whereas mice display profound sexual dimorphism, with males having low baseline levels punctuated by high-amplitude pulses and females showing more continuous secretion with smaller pulses . This sexual dimorphism in mice leads to sex-specific effects of GH on gene expression, particularly in the liver, which may not directly parallel human responses. Another key difference is receptor cross-reactivity: human GH can bind both GH and prolactin receptors in mice, whereas mouse GH binds only to the mouse GH receptor . This means that transgenic mice expressing human GH exhibit combined effects of both hormonal systems, potentially complicating interpretation. The metabolic effects of altered GH signaling also differ somewhat between species, particularly regarding glucose homeostasis and fat distribution patterns. While both GHR−/− mice and human Laron Syndrome patients show increased adiposity, the distribution patterns and metabolic consequences may vary . Additionally, developmental timing differences between mice and humans affect how alterations in GH signaling impact growth, puberty, and aging. Mice have proportionally longer developmental and shorter adult phases than humans, potentially amplifying developmental effects of GH alterations. Differences in lifespan and age-related pathology between the species also influence how GH-related interventions affect longevity and disease outcomes. Despite these differences, the fundamental mechanisms of GH signaling are highly conserved, and careful experimental design with appropriate controls can maximize the translational value of findings from these important model systems.
GH belongs to the somatotropin/prolactin family of hormones, which are essential for growth control . The gene encoding GH, along with four other related genes, is located at the growth hormone locus on chromosome 17. These genes share a high degree of sequence identity, which is believed to have evolved through a series of gene duplications . Alternative splicing of these genes generates multiple isoforms, adding to the diversity and specialization of growth hormones .
In mice, GH is primarily expressed in the pituitary gland and is not found in placental tissue, unlike some other members of the growth hormone family . GH exerts its effects by binding to the growth hormone receptor (GHR), which triggers a cascade of downstream signaling pathways that promote growth and metabolic processes .
Recombinant mouse GH is produced using an expression system in Escherichia coli (E. coli) . The recombinant protein is typically purified to a high degree of purity (>95%) and is biologically active, comparable to standard human growth hormone . The molecular weight of recombinant mouse GH is approximately 22 kDa, and it is often formulated in a lyophilized form for stability and ease of use .
Recombinant mouse GH is widely used in various research applications, including: