DIHEXA Synthetic is a single, non-glycosylated polypeptide chain containing 3 amino acids, having a molecular mass of 504.28 Dalton and a Molecular formula of C27H44N4O5.
Hexanoyl-Tyr-Ile-Ahx-NH2.
DIHEXA (N-hexanoic-Tyr-Ile-(6) aminohexanoic amide) is a hexapeptide derived from Angiotensin IV (AngIV). It functions as a small molecule activator of the hepatocyte growth factor (HGF)/c-Met system, enhancing HGF's activity while simultaneously reducing harmful chemical reactions in the body. This mechanism doubles the capacity of available growth factors to promote signaling cascades necessary for mitogenesis and neurogenesis . DIHEXA possesses three essential properties that make it particularly valuable for neurological research: it is orally active, metabolically stable, and capable of crossing the blood-brain barrier .
Methodological approach: When investigating DIHEXA's mechanism, researchers should employ co-immunoprecipitation assays to confirm HGF binding and Western blot analysis to measure c-Met phosphorylation levels. Quantitative PCR is recommended for monitoring downstream gene expression changes resulting from HGF/c-Met signaling activation.
Evidence suggests that APP/PS1 transgenic mice represent an optimal model for evaluating DIHEXA's cognitive effects. These mice exhibit Alzheimer's-like pathology and cognitive deficits that can be measured using standardized behavioral assays. The Morris Water Maze (MWM) has been effectively used to assess DIHEXA's impact on spatial learning and memory in this model .
Methodological approach: When designing cognitive testing protocols with APP/PS1 mice, researchers should implement a five-day acquisition training period (four trials per day) using a round black tub (diameter: 136 cm; depth: 60 cm) filled with water at 18–20°C and containing a small hidden platform (10 cm × 6.5 cm × 21.5 cm) placed 1 cm below the water surface. On the sixth day, conduct a probe trial with the platform removed to assess memory retention. Key measurements should include escape latency, platform crossings, and swimming speed .
DIHEXA demonstrates neurotrophic activity approximately seven times greater than brain-derived neurotrophic factor (BDNF), making it significantly more potent in promoting neuronal growth and synaptic connectivity . Unlike BDNF, which has limited blood-brain barrier penetration and a short half-life when administered peripherally, DIHEXA can cross the blood-brain barrier and has demonstrated sustained activity .
Methodological approach: When comparing DIHEXA with other neurotrophic factors, researchers should use equivalent molar concentrations rather than equivalent weights. Implement parallel experimental designs measuring dendritic spine formation, synaptogenesis, and neurite outgrowth using both in vitro neuronal cultures and in vivo models. Quantitative confocal microscopy with appropriate synaptic markers (PSD95, synaptophysin) enables direct comparison of synaptic density changes.
Studies using the Morris Water Maze demonstrated that DIHEXA administration (1.44 mg/kg and 2.88 mg/kg) to APP/PS1 mice significantly decreased escape latency compared to untreated APP/PS1 controls. During probe trials, DIHEXA-treated mice showed increased platform crossings, indicating improved spatial memory retention .
Parameter | Wild-Type | APP/PS1 Control | APP/PS1 + DIHEXA (1.44 mg/kg) | APP/PS1 + DIHEXA (2.88 mg/kg) |
---|---|---|---|---|
Escape Latency (Day 5) | Lowest | Significantly higher | Significantly lower than control | Significantly lower than control |
Platform Crossings | Highest | Significantly reduced | Significantly higher than control | Significantly higher than control |
Swimming Speed | No difference | No difference | No difference | No difference |
Methodological approach: When assessing DIHEXA's cognitive effects, researchers should include multiple behavioral paradigms beyond MWM, such as novel object recognition, Y-maze spontaneous alternation, and fear conditioning to comprehensively evaluate different aspects of cognition.
DIHEXA has a long half-life and demonstrates high metabolic stability, which contributes to its sustained therapeutic effects . It is orally active and efficiently crosses the blood-brain barrier, allowing for flexible administration routes in experimental settings .
Methodological approach: To characterize DIHEXA's pharmacokinetics, researchers should employ liquid chromatography-mass spectrometry (LC-MS/MS) for quantifying the compound in plasma and brain tissue at multiple time points after administration. Radiolabeled DIHEXA can be used to track tissue distribution through autoradiography and to determine blood-brain barrier penetration coefficients.
Research indicates that dosages of 1.44 mg/kg and 2.88 mg/kg administered over a three-month period produce significant cognitive improvements in APP/PS1 mice . These doses effectively increased AngIV levels and demonstrated protective effects in rescuing cognitive impairment.
Methodological approach: Researchers should implement dose-response studies using at least four different concentrations to establish the minimum effective dose and dose-dependent effects. Administration schedules should be systematically varied (daily, alternate days, weekly) to determine optimal dosing frequency. Both acute and chronic administration protocols should be compared for different outcome measures.
DIHEXA has been shown to augment synaptic connectivity through the formation of new functional synapses . This effect is central to its potential for treating neurodegenerative conditions.
Methodological approach: Researchers should employ multiple complementary techniques:
Golgi staining and dendritic spine quantification in fixed brain tissue
Electron microscopy to analyze ultrastructural synaptic features
Immunohistochemistry for pre- and post-synaptic markers (synaptophysin, PSD-95)
Electrophysiological recordings (field potentials, patch clamp) to assess functional synaptic changes
In vivo two-photon microscopy for longitudinal studies of spine dynamics in living animals
Several biomarkers can indicate DIHEXA's therapeutic efficacy in preclinical models:
Methodological approach: Researchers should systematically assess:
HGF and phosphorylated c-Met levels (Western blot/ELISA)
BDNF expression levels (ELISA/qPCR)
Synaptic protein markers including PSD-95, synaptophysin, and drebrin (Western blot/immunohistochemistry)
Neurogenesis markers such as DCX and BrdU incorporation
Inflammatory cytokines to assess neuroprotective effects
AngIV levels as DIHEXA has been shown to increase AngIV in treated APP/PS1 mice
DIHEXA enhances neurogenesis by activating the HGF/c-Met signaling pathway, which promotes neural stem cell proliferation, differentiation, and survival . The compound also supports synaptic plasticity by increasing spine formation and strengthening existing synaptic connections.
Methodological approach: To investigate these molecular mechanisms, researchers should:
Perform transcriptomic analysis (RNA-seq) of hippocampal tissue after DIHEXA treatment
Use fluorescent in situ hybridization to localize gene expression changes
Employ time-lapse imaging of fluorescently labeled neurons to track spine dynamics
Conduct proteomic analysis focusing on synaptic fraction proteins
Implement CRISPR-Cas9 mediated knockdown of c-Met to confirm pathway-specific effects
Methodological approach: For accurate quantification of DIHEXA in biological samples, researchers should:
Develop a validated LC-MS/MS method with appropriate internal standards
Optimize sample preparation protocols for different tissue types (brain, plasma, CSF)
Establish a standard curve using pure DIHEXA standards
Validate the method for parameters including selectivity, sensitivity, recovery, and stability
Consider developing an ELISA-based method for high-throughput screening when appropriate
There are theoretical concerns that DIHEXA, through its activation of the HGF/c-Met pathway, could promote tumorigenesis and cancer progression . The c-Met pathway is involved in cell proliferation, survival, and motility—processes that can contribute to cancer growth and metastasis when dysregulated.
Methodological approach: To address these concerns, researchers should:
Conduct long-term administration studies in cancer-prone animal models
Implement tissue-specific c-Met conditional knockout models to isolate effects
Perform comprehensive histopathological analyses of multiple organ systems after extended DIHEXA treatment
Use patient-derived xenograft models to assess effects on human tumors
Develop biomarker panels to monitor potential pro-oncogenic signaling during treatment
No studies in animals or humans have examined the long-term safety of DIHEXA, despite concerns about its potential tumorigenic effects .
Methodological approach: To establish a comprehensive safety profile, researchers should:
Design multi-generation toxicity studies in rodents with varied dosing regimens
Conduct carcinogenicity studies following established FDA/ICH guidelines
Implement toxicogenomic approaches to identify early molecular markers of toxicity
Develop tissue-specific conditional expression systems to isolate organ-specific effects
Establish biomarker panels for safety monitoring applicable to future clinical studies
Design specialized studies to assess effects on neural progenitor populations during development and in adulthood
DIHEXA's powerful effects on neuroplasticity require specialized experimental designs to fully characterize.
Methodological approach: Researchers should consider:
Using transgenic mice with fluorescent neurons for in vivo two-photon imaging of dendrites and spines
Implementing transcranial optogenetic stimulation protocols to assess activity-dependent plasticity
Conducting multi-electrode array recordings in hippocampal slices to assess network-level changes
Performing chronic in vivo electrophysiology before, during, and after DIHEXA treatment
Implementing designer receptor exclusively activated by designer drugs (DREADD) technology to modulate specific neural circuits and assess DIHEXA's interaction with targeted activation/inhibition
Developing computational models of neural network dynamics based on experimental data
The unique mechanism of DIHEXA may complement or interfere with other neurotherapeutic approaches.
Methodological approach: To investigate these interactions, researchers should:
Design factorial experiments testing DIHEXA in combination with established neuroprotective agents
Implement transcriptomic and proteomic analyses to identify convergent or divergent signaling pathways
Use in vitro and in vivo models to assess synergistic or antagonistic effects on neurogenesis and synaptic plasticity
Develop pharmacokinetic interaction studies to identify potential alterations in drug metabolism or distribution
Establish behavioral testing paradigms sensitive to multiple cognitive domains to detect interaction effects
The efficacy of DIHEXA may vary depending on age and disease progression, particularly in neurodegenerative conditions.
Methodological approach: Researchers should design experiments that:
Include multiple age cohorts in preclinical models (young, middle-aged, aged)
Initiate treatment at different disease stages in transgenic models
Assess cellular responses to DIHEXA across the lifespan using age-appropriate biomarkers
Implement longitudinal imaging studies to track disease progression during treatment
Develop computational models predicting age-dependent responses based on c-Met receptor expression and downstream signaling efficiency
Dihexa was developed by Joseph Harding and his team at Washington State University . The compound was later commercialized by M3 Biotechnology, a company founded to further develop and market Dihexa . The peptide was formally discovered in 2007, and its first scientific study in rats was published in 2012 .
Dihexa is derived from angiotensin IV, a peptide hormone that affects the central nervous system . It binds with high affinity to hepatocyte growth factor (HGF) and potentiates its activity at its receptor, c-Met . This interaction promotes brain-derived neurotrophic factor (BDNF) levels, which are crucial for neuron growth, differentiation, and overall synaptic plasticity . In an assay of neurotrophic activity, Dihexa was found to be seven orders of magnitude more potent than brain-derived neurotrophic factor .
Dihexa has shown promise in improving cognitive function in animal models of Alzheimer’s disease-like mental impairment . It is also being explored for its potential to treat other neurodegenerative diseases such as Parkinson’s disease, amyotrophic lateral sclerosis (ALS), and Huntington’s disease . Fosgonimeton, a phosphate pro-drug of Dihexa, is currently in clinical trials for the treatment of these diseases .
Short duration safety studies with Dihexa have uncovered no apparent toxicity . Of particular note is the lack of neoplastic induction, which is significant since c-Met is recognized as an oncogene . This suggests that Dihexa may be a safe option for long-term use in cognitive enhancement and neuroprotection.