Humanin

Humanin
Shipped with Ice Packs
In Stock

Description

Molecular Structure and Biosynthesis

Humanin exists in two forms:

  • 21-amino acid peptide: Produced within mitochondria due to differences in mitochondrial translation machinery .

  • 24-amino acid peptide: Synthesized in the cytosol from nuclear-encoded mitochondrial DNA pseudogenes .

Key structural features include:

  • A three-turn α-helix with a hydrophobic core (residues 5–18) .

  • Critical residues for function: Cys8, Leu9, Leu12, and Ser14 (mutations here abolish neuroprotection) .

  • A Ser14Gly (S14G) substitution enhances potency by >1,000-fold, forming the variant HNG .

Mechanisms of Action

Humanin interacts with multiple receptors and pathways:

Receptor/PathwayFunctionReference
FPRL1 (Formyl Peptide Receptor-Like 1)Activates ERK1/2 signaling for cell survival
CNTFR/WSX-1/gp130 complexTriggers JAK2/STAT3 phosphorylation, promoting antioxidant gene expression
IGFBP3 (Insulin-like Growth Factor Binding Protein 3)Binds IGFBP3 to inhibit apoptosis
BAX/BCL-2Blocks pro-apoptotic BAX activation

Additional mechanisms include:

  • Chaperone-mediated autophagy (CMA): Degrades oxidized proteins, reducing cellular stress .

  • Mitochondrial biogenesis: Enhances mitochondrial respiration and ATP production .

Neuroprotection

  • Alzheimer’s Disease (AD):

    • Binds amyloid-β (Aβ) oligomers, preventing neuronal toxicity .

    • In triple-transgenic AD mice, Humanin restored cognitive function and reduced Aβ plaques .

  • Parkinson’s Disease (PD):

    • Intranasal Humanin delivery rescued dopaminergic neurons in PD mouse models .

    • Plasma Humanin levels inversely correlate with PD severity in humans .

Aging and Longevity

  • Lifespan extension:

    • Transgenic C. elegans overexpressing Humanin showed a 15–20% lifespan increase .

    • Middle-aged mice treated with HNG exhibited improved glucose metabolism and reduced inflammation .

  • Age-related decline:

    • Circulating Humanin decreases with age in humans and mice, except in naked mole-rats .

    • Centenarians’ offspring have 40% higher plasma Humanin levels than age-matched controls .

Metabolic Regulation

Study ModelEffect of HumaninReference
Obese miceReduced fat mass, improved insulin sensitivity
High-fat diet ratsEnhanced glucose-stimulated insulin secretion
Human skeletal muscleAcute exercise increases Humanin levels by 30–50%

Neurodegenerative Diseases

  • Clinical correlations:

    • Low CSF Humanin levels predict AD progression (AUC = 0.87) .

    • A mitochondrial SNP (m.2158 T>C) linked to lower Humanin levels increases dementia risk .

Cardiovascular Health

  • Reduces oxidative stress in endothelial cells, improving vascular function .

  • In rat models, Humanin attenuated myocardial infarction damage by 60% .

Controversies and Gaps

  • Cancer: Conflicting data show both pro-survival (in normal cells) and potential pro-tumorigenic effects .

  • Dosing: Optimal therapeutic doses for humans remain undefined, though rodent studies use 1–5 mg/kg .

Future Directions

  • Clinical trials: Only preclinical data exist; phase I trials for neurodegenerative diseases are pending .

  • Biomarker potential: Plasma Humanin may serve as a non-invasive marker for mitochondrial health .

Product Specs

Description
Humanin Synthetic is a single, non-glycosylated polypeptide chain containing 24 amino acids with a molecular mass of 2687 Daltons and a molecular formula of C119H204N34O32S2.
Physical Appearance
Sterile Filtered White lyophilized (freeze-dried) powder.
Formulation
The protein was lyophilized without additives.
Solubility
Reconstitute the lyophilized Humanin in sterile 18MΩ-cm H2O at a concentration of at least 100 µg/ml. This solution can be further diluted into other aqueous solutions as needed.
Stability
Lyophilized Humanin is stable at room temperature for 3 weeks but should be stored desiccated below -18°C for extended periods. Upon reconstitution, Humanin should be stored at 4°C for 2-7 days. For future use, store below -18°C. For long-term storage, add a carrier protein (0.1% HSA or BSA). Avoid freeze-thaw cycles.
Purity
Greater than 97.0% as determined by RP-HPLC analysis.
Amino Acid Sequence

H-Met-Ala-Pro-Arg-Gly-Phe-Ser-Cys-Leu-Leu-Leu-Leu-Thr-Ser-Glu-Ile-Asp-Leu-Pro-Val-Lys-Arg-Arg-Ala-OH.

Q&A

What is Humanin and what are its primary functions?

Humanin is a 24-amino acid peptide derived from the mitochondrial genome, specifically within the 16S ribosomal RNA gene (MT-RNR2). It was first discovered in 2001 during a search for neurosurvival factors in unaffected areas of an Alzheimer's disease patient's brain . Humanin functions through multiple mechanisms:

  • Neuroprotection: Protects neurons from amyloid-beta (Aβ) toxicity associated with Alzheimer's disease

  • Anti-apoptotic effects: Counters cell death induced by the Bax protein by preventing its translocation to mitochondria

  • Mitochondrial protection: Prevents Aβ-induced increases in reactive oxygen species (ROS) production

  • Cellular defense: Protects against oxidative stress, serum starvation, hypoxia, and other stressors

  • Metabolic regulation: Influences glucose metabolism and insulin sensitivity

Humanin activates signaling pathways such as JAK/STAT to protect various cell types under stressful conditions, making it a multifunctional peptide with implications for numerous age-related conditions .

How do Humanin levels change throughout the lifespan?

Multiple studies demonstrate that humanin levels decline with age across species:

  • In mice, 13-month-old animals have significantly reduced circulating humanin compared to 2-month-old mice

  • In rats, 24-month-old animals show decreased levels of hypothalamic and skeletal muscle humanin compared to 3-month-old rats

  • In humans, circulating humanin levels gradually decline across age groups (45-65 vs. 66-80 vs. 81-100 years old)

This age-related decline suggests a potential role for humanin deficiency in age-associated diseases and cognitive impairment . The consistent observation across species highlights an evolutionarily conserved pattern that may be fundamental to aging processes.

What is the significance of the rs2854128 SNP in humanin research?

The SNP rs2854128 in the humanin-coding region of the mitochondrial genome has significant implications for humanin function:

  • It is associated with decreased circulating humanin levels in humans

  • In a large, independent cohort study, this SNP correlates with accelerated cognitive aging

  • The SNP has been separately associated with cardiovascular disease and cholesterol levels

  • Its prevalence differs across racial/ethnic groups, potentially explaining some ethnic-specific disease differences

This genetic variant represents an important consideration for personalized medicine approaches and highlights how mitochondrial genetics may influence age-related cognitive decline through alterations in humanin expression.

What experimental models are most effective for studying Humanin?

Based on published literature, researchers have successfully employed various models to study humanin:

In vitro models:

  • SH-SY5Y human neuroblastoma cells for studying neuroprotective effects against Aβ toxicity

  • Isolated neuronal mitochondria for direct assessment of ROS production and protection

In vivo models:

  • Aged C57BL/6N mice (18-28 months old) for cognitive assessments

  • Alzheimer's disease mouse models including triple-transgenic AD mice

  • Animal models of ischemia/reperfusion injury

Human studies:

  • Cross-sectional analyses comparing humanin levels across age groups

  • Genetic association studies examining the rs2854128 SNP

When selecting a model, researchers should consider the specific aspect of humanin biology they wish to investigate and the translational relevance of their chosen system.

What administration protocols have proven effective in animal studies?

Effective humanin administration protocols in mice include:

  • Compound: Humanin-S14G (HNG), a potent humanin analogue

  • Dose: 4 mg/kg body weight administered intraperitoneally (IP)

  • Frequency: Twice-weekly injections

  • Duration: Long-term studies from 18 months to 28 months of age (10 months treatment)

This regimen has demonstrated improvements in various cognitive measures including rotarod performance, Barnes maze navigation, and Y-maze spontaneous alternation behavior in aged mice .

Administration ParameterEffective Protocol
CompoundHumanin-S14G (HNG)
RouteIntraperitoneal (IP) injection
Dose4 mg/kg body weight
FrequencyTwice weekly
Treatment duration6-10 months (in mouse aging studies)
Starting age18 months (for aged mouse models)

What cognitive assessment protocols are most informative for Humanin studies?

The literature indicates several validated cognitive tests for assessing humanin's effects:

  • Accelerating Rotarod Test:

    • Initial speed: 4 rpm

    • Gradual increase to 40 rpm over 5 minutes

    • Measure: Time until fall

    • Assesses: Motor coordination and balance

    • Control for body weight differences

  • Y-maze Test:

    • Protocol: 8-minute free exploration

    • Measure: Spontaneous alternation behavior (proportion of arm choices different from previous two choices)

    • Assesses: Working memory and exploratory behavior

  • Barnes Maze Test:

    • Protocol: 30-second habituation followed by 2-minute exploration

    • Measure: Time to find escape box and search strategy

    • Strategy categorization:

      • Random: Localized searches with maze center crosses

      • Serial: Systematic search of consecutive holes

      • Spatial: Direct navigation to escape hole

    • Assesses: Spatial learning and memory

These tests provide complementary information about different cognitive domains affected by aging and potentially improved by humanin treatment.

Through what molecular mechanisms does Humanin protect against Alzheimer's pathology?

Humanin exerts neuroprotection against Alzheimer's disease through several complementary mechanisms:

  • Direct protection against Aβ toxicity:

    • Prevents amyloid-beta-induced neuronal death in vitro

    • Protects against memory deficits caused by intracerebroventricular injection of Aβ25-35 in vivo

    • Demonstrates efficacy in triple-transgenic AD mouse models

  • Mitochondrial protection:

    • Prevents Aβ-induced increases in reactive oxygen species (ROS) production

    • Preserves mitochondrial function during cellular stress

    • Demonstrates protection in isolated neuronal mitochondria

  • Anti-apoptotic actions:

    • Counters cell death induced by the Bax protein

    • Prevents the release of proteins that trigger apoptotic cascades

    • Protects against multiple forms of cellular stress

  • Receptor-mediated signaling:

    • Interacts with cell surface receptors to activate protective pathways

    • Induces protective signaling cascades through the JAK/STAT pathway

These mechanisms collectively contribute to humanin's capacity to mitigate both the cellular and behavioral manifestations of Alzheimer's disease.

What cellular assays best demonstrate Humanin's protective effects?

Several well-validated assays effectively demonstrate humanin's protective mechanisms:

  • Cell viability assays:

    • MTT assay: Measures cell metabolism/viability in response to Aβ with/without humanin treatment

    • Calcein staining: Visualizes viable cells protected by humanin

  • Mitochondrial function assays:

    • Direct measurement of ROS production in isolated neuronal mitochondria

    • Verification using Western blots for cytoplasmic (GAPDH) and mitochondrial (mtCOX2) markers

  • Apoptosis assessment:

    • Analysis of Bax translocation to mitochondria

    • Measurement of proteins that trigger cell death pathways

  • Stress response assays:

    • Protection against cobalt chloride (CoCl₂), a hypoxia mimetic

    • Assessment of cellular responses to serum starvation and oxidative stressors

These assays provide complementary data on different aspects of humanin's protective mechanisms, from cellular survival to specific molecular pathways affected.

How does Humanin interact with other mitochondrial-derived peptides?

Humanin was the first identified mitochondrial-derived peptide (MDP), but research has since discovered others with interrelated functions:

  • SHLP2, another MDP, shows population differences similar to humanin (lower in African Americans compared to Caucasian Americans)

  • Both humanin and SHLP2 may serve as biomarkers for disease risk and progression

  • The declining levels of these peptides with age suggest a coordinated role in aging processes

Current evidence suggests that MDPs may function as an ancient mitochondrial signaling mechanism crucial for regulating health and lifespan . The relationship between humanin and other MDPs represents an active area of research with implications for understanding coordinated mitochondrial regulation of aging processes.

How should researchers control for age-related variables in Humanin studies?

When designing studies to investigate age-related effects of humanin, researchers should implement the following controls:

  • Age-matched control groups:

    • Use animals/subjects of identical ages for treatment and control groups

    • Consider the timing of age-related humanin decline in the species studied

  • Genetic considerations:

    • Screen for the rs2854128 SNP associated with decreased humanin levels

    • Consider stratifying analyses by genetic variants

    • Account for population/ethnic differences in humanin expression

  • Environmental factors:

    • Control for or investigate exercise effects, which may naturally boost humanin production

    • Standardize housing conditions in animal studies

    • Document health status and comorbidities

  • Temporal considerations:

    • Design studies to capture age-related trajectories rather than single time points

    • Consider both longitudinal and cross-sectional approaches

    • Start interventions at appropriate ages (e.g., 18 months in mice)

Proper control of these variables is essential for distinguishing humanin's effects from other age-related processes and for ensuring reproducibility across studies.

What are the key considerations for translating Humanin research to human applications?

Translating humanin research from animals to humans requires careful consideration of several factors:

  • Dosing translation:

    • Allometric scaling of effective doses from mice to humans

    • Consideration of administration routes feasible in humans

    • Evaluation of pharmacokinetics across species

  • Target population selection:

    • Identification of individuals most likely to benefit (e.g., carriers of the rs2854128 SNP)

    • Age considerations, given the established age-related decline in endogenous humanin

    • Disease-specific targeting (Alzheimer's, cardiovascular disease)

  • Biomarker development:

    • Standardization of humanin measurement for clinical use

    • Establishment of age-specific reference ranges

    • Validation of humanin as a biomarker for disease risk or progression

  • Clinical trial design:

    • Selection of appropriate cognitive assessment tools that parallel those used in animal studies

    • Determination of treatment duration based on animal study timelines

    • Consideration of preventive versus therapeutic approaches

These considerations help bridge the gap between promising animal studies and potential human applications, maximizing the translational impact of humanin research.

How can contradictory findings in Humanin research be reconciled?

When faced with contradictory findings in humanin research, researchers should:

  • Examine methodological differences:

    • Different humanin analogues used (e.g., HNG versus native humanin)

    • Variations in dosing regimens and administration routes

    • Different experimental models (cell lines, animal models, human studies)

  • Consider context-specific effects:

    • Tissue-specific responses to humanin

    • Age-dependent variations in effectiveness

    • Disease stage-specific effects

  • Account for genetic factors:

    • Presence of the rs2854128 SNP or other genetic variations

    • Differences in mitochondrial haplogroups across populations

    • Potential nuclear-mitochondrial genetic interactions

  • Statistical considerations:

    • Sample size and power limitations

    • Appropriate statistical approaches for age-related analyses

    • Consideration of covariates that might influence outcomes

Systematic reviews and meta-analyses can help identify patterns across studies and provide a broader perspective on seemingly contradictory results.

What are promising approaches for developing Humanin-based therapeutics?

Several approaches show promise for developing humanin-based interventions:

  • Humanin analogues:

    • Development of more potent analogues like HNG

    • Exploration of modified peptides with improved stability and bioavailability

    • Investigation of tissue-specific targeting strategies

  • Combinatorial approaches:

    • Co-administration with other MDPs or complementary compounds

    • Integration with existing therapeutic approaches for neurodegenerative diseases

    • Personalized combinations based on genetic profiles (e.g., rs2854128 status)

  • Endogenous humanin upregulation:

    • Identification of compounds that increase endogenous humanin production

    • Investigation of exercise and other lifestyle interventions that may naturally boost humanin levels

    • Exploration of mitochondrial biogenesis enhancers as indirect humanin modulators

  • Delivery systems:

    • Development of specialized delivery systems to address blood-brain barrier penetration

    • Extended-release formulations to reduce administration frequency

    • Targeted delivery to specific tissues or cell types

These approaches represent promising avenues for translating the neuroprotective and anti-aging effects of humanin into clinical applications.

What omics approaches might enhance understanding of Humanin's mechanisms?

Multi-omics approaches can provide comprehensive insights into humanin's mechanisms of action:

  • Transcriptomics:

    • RNA-seq to identify gene expression changes in response to humanin administration

    • Analysis of age-related transcriptomic changes modified by humanin

    • Single-cell RNA-seq to identify cell type-specific responses

  • Proteomics:

    • Identification of proteins interacting with humanin

    • Analysis of post-translational modifications influenced by humanin

    • Quantitative proteomics to track cellular responses to humanin treatment

  • Metabolomics:

    • Measurement of metabolic changes associated with humanin treatment

    • Analysis of energy metabolism pathways affected by humanin

    • Identification of biomarkers associated with humanin response

  • Epigenomics:

    • Investigation of epigenetic changes influenced by humanin

    • Analysis of age-related epigenetic drift and potential modification by humanin

    • Identification of epigenetic mechanisms contributing to humanin expression

Integration of these approaches through systems biology frameworks could reveal the complex network of interactions through which humanin exerts its beneficial effects on aging and cognition.

Product Science Overview

Discovery and Origin

Humanin was first discovered in 2001 by Dr. Mori and colleagues while they were investigating genes related to Alzheimer’s disease. It is a mitochondrial-derived peptide (MDP), meaning it is encoded by the mitochondrial DNA rather than the nuclear DNA . Specifically, Humanin is encoded within the 16S ribosomal RNA gene (MT-RNR2) in the mitochondrial genome .

Structure and Function

Humanin is a small peptide consisting of 24 amino acids when produced outside the mitochondria and 21 amino acids when produced inside the mitochondria . Its structure includes a three-turn α-helix, which is crucial for its biological activity . Humanin has been shown to have cytoprotective effects, meaning it helps protect cells from various forms of stress and damage .

Biological Roles

Humanin plays several important roles in the body:

  • Neuroprotection: It has been shown to exert protective effects against neurodegenerative diseases such as Alzheimer’s and Parkinson’s .
  • Cardiovascular Health: Humanin helps reduce inflammation and oxidative stress, which are key factors in the development of cardiovascular diseases .
  • Metabolic Regulation: It improves glucose metabolism and insulin sensitivity, making it a potential therapeutic candidate for diabetes and obesity .
  • Longevity: Studies in animal models have shown that Humanin can increase both lifespan and health span by promoting autophagy and maintaining mitochondrial function .
Research and Therapeutic Potential

Humanin’s ability to protect cells and maintain mitochondrial function under stress has made it a subject of intense research. Its potential therapeutic applications are vast, ranging from treating age-related diseases to improving overall health and longevity .

Quick Inquiry

Personal Email Detected
Please use an institutional or corporate email address for inquiries. Personal email accounts ( such as Gmail, Yahoo, and Outlook) are not accepted. *
© Copyright 2024 Thebiotek. All Rights Reserved.